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










Base de dados
Intervalo de ano de publicação
1.
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
2.
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
3.
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
4.
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
5.
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
6.
Eur J Pharmacol ; 769: 177-84, 2015 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-26593425

RESUMO

The aim of the study was to investigate the roles of α1-adrenoceptor subtypes in the last-day pregnant rat uterus in vitro by the administration of subtype-specific antagonists (the α1A-adrenoceptor antagonist WB 4101 and the α1D-adrenoceptor antagonist BMY 7378) after 17ß-estradiol or progesterone pretreatment. In isolated organ bath studies, contractions were elicited with (-)-noradrenaline (10(-8)-10(-5)M) in the presence of propranolol (10(-5)M) and yohimbine (10(-6)M) in order to avoid ß-, and α2-adrenergic action. The myometrial expressions of the α1-adrenoceptor subtypes were determined by means of the real time reverse transcription-polymerase chain reaction (RT-PCR) and Western blotting techniques. The activated G protein levels were investigated through radiolabelled GTP binding assays. Both 17ß-estradiol and progesterone pretreatment changed the myometrial contracting effect of (-)-noradrenaline. In the presence of WB 4101, progesterone pretreatment decreased the (-)-noradrenaline-induced myometrial contraction. In the presence of BMY 7378, both the 17ß-estradiol and the progesterone pretreatment reduced the effect of (-)-noradrenaline. The mRNA and protein expressions of the α1A-adrenoceptors were decreased after 17ß-estradiol pretreatment. (-)-Noradrenaline increased the [(35)S]GTPγS binding of the α1-adrenoceptors, which was most markedly elevated by progesterone. Pertussis toxin inhibited the [(35)S]GTPγS binding-stimulating effect of (-)-noradrenaline, indicating the role of Gi proteins in the signal mechanisms. 17ß-estradiol pretreatment blocks the expression of the α1A-adrenoceptors, whereas it does not influence the expression of the α1D-adrenoceptors. Progesterone pretreatment does not have any effect on the myometrial mRNA and protein expressions of the α1-adrenoceptors, but it alters the G protein coupling of these receptors, promoting a Gi-dependent pathway.


Assuntos
Estradiol/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Miométrio/efeitos dos fármacos , Miométrio/metabolismo , Progesterona/farmacologia , Receptores Adrenérgicos alfa 1/metabolismo , Animais , Dioxanos/farmacologia , Feminino , Piperazinas/farmacologia , Gravidez , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores Adrenérgicos alfa 1/genética
7.
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
8.
Neuropharmacology ; 89: 298-307, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25446673

RESUMO

WHAT IS KNOWN: There is an increasing number of studies demonstrating the direct effect of the cannabinoid receptor 1 (CB1) antagonist/inverse agonist rimonabant on the opioid system. The kappa opioid receptors (KORs) are well known to mediate depression- and anxiety-like behavior. Clinical studies on chronic rimonabant administration have revealed that rimonabant leads to a very similar pathophysiology, suggesting a potential impact of rimonabant on KORs. OBJECTIVES: Our objectives were to examine the putative effects of rimonabant on KOR ligand binding, G-protein activity, protein expression and how all these contribute to the development of depression- and anxiety-like behavior. RESULTS: In Chinese hamster ovary (CHO) cell membranes transfected with rat KOR (CHO-rKOR) rimonabant inhibited KOR agonist [3H]U69593 binding in the micromolar range in competition binding experiments and specifically reduced KOR basal activity at lower micromolar concentrations in [35S]GTPγS binding assays. Rimonabant significantly inhibited dynorphin (1-11)-induced [35S]GTPγS binding in micromolar range in CHO-rKOR cells, CB1 knockout (CB1 K.O.) and CB1/CB2 double knockout mouse forebrain membranes. A single dose of i.p. 0.1 mg/kg rimonabant significantly reduced dynorphin (1-11)-induced KOR G-protein activity and KOR protein expression levels 24 h following the administration in both wild type and CB1 K.O. mice forebrain. Furthermore, in elevated plus maze mice showed an anxiolytic-like effect upon rimonabant injection that could be reversed by 1 mg/kg KOR antagonist norbinaltorphimine. The anxiolytic-like effects were further confirmed with the light­dark box test. CONCLUSION: Rimonabant reduced KOR ligand binding, receptor mediated G-protein activity and protein expression level, which overall leads to altered anxiety-like behavior.


Assuntos
Ansiedade/tratamento farmacológico , Antagonistas de Receptores de Canabinoides/uso terapêutico , Piperidinas/uso terapêutico , Pirazóis/uso terapêutico , Receptor CB1 de Canabinoide/metabolismo , Receptor CB2 de Canabinoide/metabolismo , Receptores Opioides kappa/metabolismo , Adaptação Ocular/efeitos dos fármacos , Adaptação Ocular/genética , Analgésicos Opioides/farmacologia , Animais , Células CHO , Antagonistas de Receptores de Canabinoides/farmacologia , Cricetulus , Modelos Animais de Doenças , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Antagonistas de Entorpecentes/farmacologia , Piperidinas/farmacologia , Prosencéfalo/efeitos dos fármacos , Prosencéfalo/metabolismo , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/genética , Pirazóis/farmacologia , Receptor CB1 de Canabinoide/genética , Receptor CB2 de Canabinoide/genética , Rimonabanto , Natação/psicologia
9.
Chem Biol Drug Des ; 86(2): 156-62, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25393330

RESUMO

N-type voltage-dependent Ca(2+) channels (CaV 2.2) are located at nerve endings in the central and peripheral nervous systems and are strongly associated with the pathological processes of cerebral ischaemia and neuropathic pain. CaV 2.2 blockers such as the ω-conotoxin MVIIA (Prialt) are analgesic and have opioid-sparing effects. With the aim to develop new multitarget analgesic compounds, we designed the first ω-conotoxin/opioid peptidomimetics based on the enkephalin-like sequence Tyr-D-Ala-Gly-Phe (for the opioid portion) and two fragments derived from the loop-2 pharmacophore of ω-conotoxin MVIIA. Antinociceptive activity evaluated in vitro and in vivo revealed differential affinity for CaV 2.2 and opioid receptors and no significant synergistic activity.


Assuntos
Analgésicos Opioides/síntese química , Analgésicos Opioides/farmacologia , Bloqueadores dos Canais de Cálcio/síntese química , Bloqueadores dos Canais de Cálcio/farmacologia , Oligopeptídeos/química , Oligopeptídeos/farmacologia , Sequência de Aminoácidos , Animais , Canais de Cálcio/metabolismo , Canais de Cálcio Tipo N/metabolismo , Conotoxinas/química , Desenho de Fármacos , Ligantes , Camundongos , Dor/tratamento farmacológico , ômega-Conotoxinas/química , ômega-Conotoxinas/farmacologia
10.
CNS Neurol Disord Drug Targets ; 13(9): 1520-9, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25478797

RESUMO

There is an increasing number of evidence showing analgesic properties of the kynurenic acid (KYNA), and also some studies demonstrate that kynurenine might interact with the opioid system. Therefore in this study, for the first time we investigated the direct binding affinity of KYNA and its structural analog KYNA-1 towards mu, kappa and delta opioid receptor in competition binding experiments applying opioid receptor specific radioligands. The binding affinity measurements were performed in Chinese hamster ovary cell lines overexpressing the corresponding opioid receptor (mu and kappa opioid receptor were rat, delta opioid receptor were mouse sequence). Additionally we also examined the chronic effect of these compounds on mu, kappa and delta opioid receptor and also nociceptin peptide receptor mediated G-protein activity in [(35)S]GTPγS binding assays performed in mouse cortex and striatum membranes. Our results showed that KYNA and KYNA-1 had no affinity towards any of the three classic opioid receptors. On the other hand the compounds significantly decreased opioid and nociceptin receptor mediated G-protein activity or in some cases enhanced the potency of the activating ligand. Moreover, the alterations were receptor and brain region specific. Accordingly, we conclude that KYNA and KYNA-1 do not interact directly with the opioid receptors, but more likely alter the receptor functions intracellularly.


Assuntos
Encéfalo/efeitos dos fármacos , Antagonistas de Aminoácidos Excitatórios/farmacologia , Proteínas de Ligação ao GTP/metabolismo , Ácido Cinurênico/análogos & derivados , Ácido Cinurênico/farmacologia , Receptores Opioides/metabolismo , Analgésicos Opioides/farmacologia , Animais , Autorradiografia , Células CHO , Cricetinae , Cricetulus , Relação Dose-Resposta a Droga , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Feminino , Guanosina 5'-O-(3-Tiotrifosfato)/farmacocinética , Camundongos , Camundongos Endogâmicos C57BL , Ligação Proteica/efeitos dos fármacos , Isótopos de Enxofre/farmacocinética , Transfecção
11.
Biomed Res Int ; 2014: 762930, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25276817

RESUMO

Nociceptin/orphanin FQ (N/OFQ) antinociception, which is mediated selectively by the N/OFQ peptide receptor (NOP), was demonstrated in pain models. In this study, we determine the role of activated microglia on the analgesic effects of N/OFQ in a rat model of neuropathic pain induced by chronic constriction injury (CCI) to the sciatic nerve. Repeated 7-day administration of minocycline (30 mg/kg i.p.), a drug that affects microglial activation, significantly reduced pain in CCI-exposed rats and it potentiates the analgesic effects of administered N/OFQ (2.5-5 µg i.t.). Minocycline also downregulates the nerve injury-induced upregulation of NOP protein in the dorsal lumbar spinal cord. Our in vitro study showed that minocycline reduced NOP mRNA, but not protein, level in rat primary microglial cell cultures. In [(35)S]GTPγS binding assays we have shown that minocycline increases the spinal N/OFQ-stimulated NOP signaling. We suggest that the modulation of the N/OFQ system by minocycline is due to the potentiation of its neuronal antinociceptive activity and weakening of the microglial cell activation. This effect is beneficial for pain relief, and these results suggest new targets for the development of drugs that are effective against neuropathic pain.


Assuntos
Minociclina/uso terapêutico , Neuralgia/tratamento farmacológico , Peptídeos Opioides/uso terapêutico , Animais , Células Cultivadas , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , Microglia/efeitos dos fármacos , Microglia/patologia , Minociclina/administração & dosagem , Minociclina/farmacologia , Modelos Biológicos , Neuralgia/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos Wistar , Receptores Opioides/genética , Receptores Opioides/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Medula Espinal/efeitos dos fármacos , Medula Espinal/patologia , Resultado do Tratamento , Receptor de Nociceptina , Nociceptina
12.
ACS Med Chem Lett ; 5(9): 1032-6, 2014 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-25221662

RESUMO

Two novel opioid analogues have been designed by substituting the native d-Ala residues in position 2,2' of biphalin with two residues of d-penicillamine or l-penicillamine and by forming a disulfide bond between the thiol groups. The so-obtained compound 9 containing d-penicillamines showed excellent µ/δ mixed receptor affinities (K i (δ) = 5.2 nM; K i (µ) = 1.9 nM), together with an efficacious capacity to trigger the second messenger and a very good in vivo antinociceptive activity, whereas product 10 was scarcely active. An explanation of the two different pharmacological behaviors of products 9 and 10 was found by studying their conformational properties.

13.
Eur J Pharmacol ; 731: 1-7, 2014 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-24657279

RESUMO

Several methodological approaches suggest that receptor heteromers exist in cell systems, but their presence in physiological tissue is widely contentious. We describe a novel method to determine if heterodimers exist in brain tissue sections using autoradiographic binding comparisons from single and double gene knockout mice, where tissues either have a full receptor complement and can form heterodimers, or are incapable of making heterodimers. We have tested this model, which we have named Knockout Subtraction Autoradiography, to determine if heterodimerisation of the kappa (KOP) and delta opioid (DOP) receptors occurs, as evidence from binding studies in cell systems suggest they are present in the brain. Using labeling of putative KOP receptor/DOP receptor heterodimers with either [(3)H]bremazocine or with [(3)H]naltrindole, two ligands which were used to provide evidence suggesting that these opioid receptor subtypes heterodimerize, we have applied a subtraction equation model based on the principle that receptor gene double knockout of either MOP receptor/KOP receptor (DOP receptor expression only) or MOP receptor/DOP receptor (KOP receptor expression only) produces tissue incapable of making the KOP receptor/DOP receptor heterodimer. We have shown in most brain regions that the labeling fits a simple additive model of monomer labeling, but that in a few brain regions opioid receptor heterodimerization does occur. The data does not support the conclusion that KOP receptor/DOP receptor heterodimerisation is widespread in the central nervous system, but does indicate that this novel methodology can detect heterodimerisation, when ligands with distinct binding affinities for monomer and heterodimer forms exist.


Assuntos
Autorradiografia/métodos , Encéfalo/metabolismo , Técnicas de Inativação de Genes , Multimerização Proteica , Receptores Opioides delta/química , Receptores Opioides kappa/química , Técnica de Subtração , Animais , Benzomorfanos/metabolismo , Masculino , Camundongos , Camundongos Knockout , Naltrexona/análogos & derivados , Naltrexona/metabolismo , Estrutura Quaternária de Proteína , Receptores Opioides delta/deficiência , Receptores Opioides delta/genética , Receptores Opioides kappa/deficiência , Receptores Opioides kappa/genética
14.
Neurochem Int ; 67: 14-22, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24508403

RESUMO

WHAT IS KNOWN: There is a growing number of evidence showing, that the cannabinoid receptor 1 (CB1) antagonist rimonabant has many non-cannabimimetic actions, such as affecting the opioid system. The direct effect of rimonabant on opioid receptors has been studied so far mainly on µ-opioid receptors. However recently the δ-opioid receptor (DOR) receives much more attention as before, due to its potential therapeutic applications, such as nociception or treatment for psychiatric disorders. OBJECTIVES: To investigate the direct effect of rimonabant on DOR specific ligand binding and on the DOR mediated G-protein activation. RESULTS: Micromolar concentrations of rimonabant directly inhibited the DOR specific agonist binding in radioligand competition binding experiments using Chinese hamster ovary cells stably transfected with mouse DOR (CHO-mDOR). However the inhibition occurred also in the subnanomolar range during DOR specific antagonist binding in similar experimental conditions. In functional [(35)S]GTPγS binding assays rimonabant significantly decreased the basal receptor activity in CHO-mDOR but also in parental CHO cell membranes. During DOR agonist stimulation, micromolar concentration of rimonabant attenuated the DOR G-protein activation and the potency of the activator ligand in [(35)S]GTPγS binding assays performed in CHO-mDOR, in wild type and also in CB1/CB2 double knock-out mouse forebrain membranes. Yet again this inhibitory action was DOR specific, since it did not occur during other specific GPCR agonist mediated G-protein activation. CONCLUSION: Rimonabant directly inhibited DOR function in the micromolar concentrations. The inhibitory actions indicate an antagonistic behavior towards DOR which was established by the followings: (i) rimonabant inhibited DOR antagonist binding more effectively than agonist binding, (ii) the inverse agonistic, agonistic effect of the compound can be excluded, and (iii) additionally according to previous findings the allosteric mechanism can also be foreclosed.


Assuntos
Proteínas de Ligação ao GTP/metabolismo , Piperidinas/farmacologia , Pirazóis/farmacologia , Receptores Opioides delta/antagonistas & inibidores , Animais , Células CHO , Cricetinae , Cricetulus , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Ligantes , Camundongos , Ligação Proteica , Receptor CB1 de Canabinoide/genética , Receptor CB2 de Canabinoide/genética , Receptores Opioides delta/agonistas , Receptores Opioides delta/metabolismo , Rimonabanto
15.
Eur J Med Chem ; 68: 167-77, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23974016

RESUMO

Endomorphin-2 [Tyr-Pro-Phe-Phe-NH2] and DAMGO [Tyr-D-Ala-Gly-(N-Me)Phe-Gly-ol] are natural (EM2) and synthetic (DAMGO) opioid peptides both selective for µ opioid receptor with high analgesic activity. In this work we report synthesis, in vitro and in vivo biological evaluation of five new hybrid EM2/DAMGO analogues, with the aim to obtain compounds with high affinity at µ-opioid receptor, high activity in animal nociception tests (hot plate and tail flick) and improved enzymatic stability. Double N-methylation on both Phe residues and C-terminal ethanolamide led to analogue 6e, which possesses a good in vitro µ affinity (Kiµ=34 nM), combined with a remarkable in vivo antinociceptive activity.


Assuntos
Ala(2)-MePhe(4)-Gly(5)-Encefalina/síntese química , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Oligopeptídeos/síntese química , Oligopeptídeos/farmacologia , Receptores Opioides mu/agonistas , Analgésicos Opioides/síntese química , Analgésicos Opioides/química , Analgésicos Opioides/farmacologia , Animais , Ala(2)-MePhe(4)-Gly(5)-Encefalina/química , Estabilidade Enzimática/efeitos dos fármacos , Cobaias , Humanos , Masculino , Estrutura Molecular , Oligopeptídeos/química , Ratos , Ratos Wistar
16.
Epilepsy Res ; 105(1-2): 13-9, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23337899

RESUMO

The opioid/nociceptin receptors are involved in many neurological disorders such as Alzheimer's disease, Parkinson's disease and epilepsy. Kainic acid (KA) is an analog of the excitatory amino acid transmitter glutamate and the systemic administration of KA induces status epilepticus (SE) in rodents. In this study, we examined the alterations in the G-protein activity and the gene expression levels of mu, kappa, delta opioid and nociceptin receptors (MOPr, KOPr, DOPr and NOPr) as well as PNOC, the precursor polypeptide of nociceptin-OFQ (N/OFQ) in KA-induced seizures in the rat brain cortex. KA was used to create seizures with the dose of 10 mg/kg body weight i.p. Following the KA administration, the rats were observed for 3 h to assess seizure activity. Seizures occurred approximately 45 min after the KA injection. Only rats exhibiting full limbic seizures, forelimb clonus with rearing, were used in this study. All animals were decapitated 4 h after the administration of KA. Our [(35)S]GTPγS binding results showed that there was a significant difference in both the affinity and efficacy particularly one of NOPr stimulation following KA treatment. Slight, but significant increase was observed for MOPr. Moreover PNOC, NOPr and MOPr mRNA levels were increased by KA treatment but there were no significant changes in the levels of DOPr and KOPr mRNAs. These results show that the activities of opioid/nociceptin receptors can be modified by KA-treatment, and MOPr, PNOC and NOPr are the most responsive to KA-induced seizures in the rat brain cortex.


Assuntos
Córtex Cerebral/metabolismo , Proteínas de Ligação ao GTP/biossíntese , Ácido Caínico/toxicidade , RNA Mensageiro/biossíntese , Receptores Opioides/biossíntese , Convulsões/metabolismo , Animais , Córtex Cerebral/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ala(2)-MePhe(4)-Gly(5)-Encefalina/metabolismo , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Regulação da Expressão Gênica , Masculino , Peptídeos Opioides/metabolismo , Peptídeos Opioides/farmacologia , Ligação Proteica/fisiologia , Ratos , Ratos Sprague-Dawley , Convulsões/induzido quimicamente , Receptor de Nociceptina
17.
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
18.
Eur J Pharmacol ; 698(1-3): 220-7, 2013 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-23142371

RESUMO

Endomorphin-1 (EM-1) and endomorphin-2 (EM-2) are two endogenous tetrapeptides with very high affinities for the µ-opioid receptor. Until recently, the precise neuroanatomical localization of the binding sites for these peptides was unknown. However, the recent synthesis of tritiated forms of these molecules has permitted these binding sites to be analysed with a very high degree of neuroanatomical specificity. Preliminary studies demonstrated a superior binding profile for EM-2, with less non-specific binding than EM-1. As the endogenous cannabinoid and opioid systems interact at several levels, we investigated how deletion of the CNR1 gene, which encodes the cannabinoid receptor 1 (CB(1)R) protein, affects the brain distribution of EM-2 binding sites. Our results revealed no differences in the average density of EM-2 binding sites in CB(1) receptor knockout (CB(1)R KO) and WT mice. However, when both hemispheres were analysed separately, we detected specific alterations in the distribution of EM-2 binding sites in the right hemisphere of CB(1)R KO mice. While, the density of EM-2 binding sites in CB(1)R KO mice was higher in the CA3 hippocampal field and in the pontine tegmental nuclei, it was lower in the superior colliculus and ventral tegmental area than in WT controls. No differences were observed in the left hemisphere for any of the regions analysed. For the first time these findings demonstrate a lateralization effect on cerebral opioid binding sites that may be mediated by the central cannabinoid system.


Assuntos
Cérebro/metabolismo , Deleção de Genes , Oligopeptídeos/química , Oligopeptídeos/metabolismo , Receptor CB1 de Canabinoide/deficiência , Receptor CB1 de Canabinoide/genética , Animais , Sítios de Ligação , Masculino , Camundongos
19.
Neurochem Int ; 60(6): 555-64, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22382076

RESUMO

Excitotoxicity is a contributing factor to the pathogenesis of acute or chronic neurodegenerative disease states. Kainic acid (KA) is an excitotoxic substance and the administration of it to rodents induces seizure activity (status epilepticus, SE) and leads to neurodegeneration. In this study the effect of KA-induced excitotoxicity on the G-protein activations and the gene expression levels of the opioid/nociceptin system receptors as MOPr, KOPr, DOPr, ORL-1, and PNOC (N/OFQ) were investigated, and the regulator effect of naloxone (Nal) on the gene expressions of the opioid system receptors against KA-induced seizures in the rat hippocampus was tested. In addition, the expression levels of stress-toxicity genes were assessed in the hippocampus following KA-induced excitotoxicity in order to determine the potential genetic targets which can be helpful for neuroprotective interventions. Our results indicate that the KA-induced excitotoxicity increased the mRNA levels of MOPr, DOPr, KOPr, PNOC, and ORL-1. However, G-protein activations of MOPr, DOPr, and KOPr remained relatively unchanged while both the potency and efficacy of N/OFQ were significantly increased. The PCR array data showed that KA-induced excitotoxicity altered the expression levels of genes in the cellular stress or toxicity pathways. Our data suggests that the induction of the opioid/nociceptin system may be involved in the cellular stress response following a neurodegenerative insult and that the genes modulated by the KA-treatment in the stress-toxicity pathways may be evaluated as targets of potential neuroprotective interventions.


Assuntos
Hipocampo/efeitos dos fármacos , Ácido Caínico/toxicidade , Peptídeos Opioides/fisiologia , Receptores Opioides/fisiologia , Estado Epiléptico/induzido quimicamente , Estado Epiléptico/fisiopatologia , Animais , Agonistas de Aminoácidos Excitatórios/toxicidade , Hipocampo/fisiologia , Masculino , Peptídeos Opioides/genética , Peptídeos Opioides/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores Opioides/genética , Estado Epiléptico/tratamento farmacológico , Nociceptina
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...