Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Neuroscience ; 451: 149-163, 2020 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-33039523

RESUMO

In the present study, we examined the neurobehavioral effects of a sensory functional food ingredient mainly based on Citrus sinensis extracts (D11399) using a battery of tests recapitulating various endophenotypes of depression such as anxiety in the open field (OF), the elevated plus-maze (EPM), and the novelty suppressed feeding (NSF), self-care in the splash test (ST), despair in the forced swimming task (FST) but also anhedonia in the sucrose preference test (SPT) in mice. A one-week oral administration of D11399 promoted anxiolytic- and antidepressant-like responses in naïve mice subjected to the NSF and FST. In a marked contrast, the administration of D11399 by oral gavage or the inhibition of olfaction by methimazole prevented such beneficial effects. We further investigated the neurobehavioral properties of a ten-week oral administration of D11399 in the corticosterone (CORT) mouse model of depression. Interestingly, D11399 also elicited anxiolytic- and antidepressant-like effects in various paradigms. To characterize the putative underpinning neurobiological mechanisms in CORT mice, we investigated whether cellular and molecular processes commonly associated with antidepressant responses such as monoaminergic neurotransmission and neuronal maturation in the hippocampus were impacted. Although D11399 did not modify the hippocampal extracellular levels of monoamines (i.e. serotonin and norepinephrine), it reversed the ability of CORT to decrease serotonin neurons firing rate in the dorsal raphe and neuronal maturation in the hippocampus. These findings suggest that the anxiolytic- and antidepressant-like effects of this sensory functional food ingredient are closely related with olfaction and likely a concomitant change in the activity of the central serotonergic system. Further experiments are warranted to precise the neuronal circuits linking sensorial and emotional modalities and identify innovative therapeutic strategies aimed to relieve depressive endophenotypes.


Assuntos
Citrus , Ingredientes de Alimentos , Animais , Antidepressivos/farmacologia , Ansiedade , Comportamento Animal , Depressão/tratamento farmacológico , Modelos Animais de Doenças , Camundongos , Olfato
2.
Br J Pharmacol ; 165(2): 424-35, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21718302

RESUMO

BACKGROUND AND PURPOSE Opiates remain the most effective compounds for alleviating severe pain across a wide range of conditions. However, their use is associated with significant side effects. Neuropeptide FF (NPFF) receptors have been implicated in several opiate-induced neuroadaptive changes including the development of tolerance. In this study, we investigated the consequences of NPFF receptor blockade on acute and chronic stimulation of opioid receptors in mice by using RF9, a potent and selective antagonist of NPFF receptors that can be administered systemically. EXPERIMENTAL APPROACH The effects of RF9 were investigated on opioid pharmacological responses including locomotor activity, antinociception, opioid-induced hyperalgesia, rewarding properties and physical dependence. KEY RESULTS RF9 had no effect on morphine-induced horizontal hyperlocomotion and slightly attenuated the decrease induced in vertical activity. Furthermore, RF9 dose-dependently blocked the long-lasting hyperalgesia produced by either acute fentanyl or chronic morphine administration. RF9 also potentiated opiate early analgesic effects and prevented the development of morphine tolerance. Finally, RF9 increased morphine-induced conditioned place preference without producing any rewarding effect by itself and decreased naltrexone-precipitated withdrawal syndrome following chronic morphine treatment. CONCLUSION AND IMPLICATIONS The NPFF system is involved in the development of two major undesirable effects: tolerance and dependence, which are clinically associated with prolonged exposure to opiates. Our findings suggest that NPFF receptors are interesting therapeutic targets to improve the analgesic efficacy of opiates by limiting the development of tolerance, and for the treatment of opioid dependence.


Assuntos
Adamantano/análogos & derivados , Analgésicos Opioides/farmacologia , Dipeptídeos/farmacologia , Tolerância a Medicamentos/fisiologia , Transtornos Relacionados ao Uso de Opioides/fisiopatologia , Receptores de Neuropeptídeos/antagonistas & inibidores , Adamantano/farmacologia , Animais , Comportamento Animal/efeitos dos fármacos , Condicionamento Clássico , Fentanila/farmacologia , Temperatura Alta , Hiperalgesia/induzido quimicamente , Hiperalgesia/tratamento farmacológico , Hiperalgesia/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Morfina/farmacologia , Atividade Motora/efeitos dos fármacos , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Transtornos Relacionados ao Uso de Opioides/tratamento farmacológico , Dor/tratamento farmacológico , Dor/fisiopatologia , Receptores de Neuropeptídeos/fisiologia , Síndrome de Abstinência a Substâncias/tratamento farmacológico , Síndrome de Abstinência a Substâncias/fisiopatologia
3.
Peptides ; 21(7): 893-900, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10998522

RESUMO

Nociceptin and the ORL1 receptor share high sequence similarity with opioid peptides, particularly dynorphin A, and their receptors. However, nociceptin and dynorphin A may use distinct molecular pathways to bind and activate their cognate receptors. Activation of the kappa-opioid receptor by dynorphin A is thought to require interactions of its N-terminal hydrophobic domain (Y(1)GGF) with the receptor opioid binding pocket, located within the transmembrane helix bundle, while activation of the ORL1 receptor appears to require interactions of the positively charged core (R(8)KSARK) of nociceptin with the negatively charged second extracellular receptor loop.


Assuntos
Receptores Opioides/química , Receptores Opioides/genética , Sequência de Aminoácidos , Animais , Membrana Celular/metabolismo , Clonagem Molecular , Dinorfinas/química , Humanos , Ligantes , Dados de Sequência Molecular , Peptídeos/química , Ligação Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Homologia de Sequência de Aminoácidos , Receptor de Nociceptina
4.
Peptides ; 21(7): 907-17, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10998524

RESUMO

The ORL1 receptor is a G protein-coupled receptor structurally related to the opioid receptors, whose endogenous ligand is the heptadecapeptide nociceptin/orphanin FQ. In this review, data which have contributed to the mapping of the anatomic distribution of the ORL1 receptor have been collated with an emphasis on their relation to physiological functions. The ORL1 receptor is widely expressed in the central nervous system, in particular in the forebrain (cortical areas, olfactory regions, limbic structures, thalamus), throughout the brainstem (central periaqueductal gray, substantia nigra, several sensory and motor nuclei), and in both the dorsal and ventral horns of the spinal cord. Regions almost devoid of ORL1 receptors are the caudate-putamen and the cerebellum. ORL1 mRNA and binding sites exhibit approximately the same distribution pattern, indicating that the ORL1 receptor is located on local neuronal circuits. The ORL1 receptor is also expressed at the periphery in smooth muscles, peripheral ganglia, and the immune system. The anatomic distribution of ORL1 receptor suggests a broad spectrum of action for the nociceptin/orphanin FQ system (sensory perception, memory process, emotional behavior, etc.).


Assuntos
Receptores Opioides/biossíntese , Animais , Sítios de Ligação , Tronco Encefálico/metabolismo , Tronco Encefálico/fisiologia , Humanos , Sistema Imunitário/metabolismo , Ligantes , Camundongos , Modelos Biológicos , Peptídeos Opioides/metabolismo , Peptídeos Opioides/fisiologia , RNA Mensageiro/metabolismo , Ratos , Receptores Opioides/fisiologia , Medula Espinal/metabolismo , Medula Espinal/fisiologia , Distribuição Tecidual , Vasodilatadores/metabolismo , Receptor de Nociceptina , Nociceptina
5.
Protein Eng ; 13(7): 477-90, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10906343

RESUMO

Evidence from statistical cluster analyses of a multiple sequence alignment of G protein-coupled receptor seven-helix folds supports the existence of structurally conserved transmembrane (TM) ligand binding sites in the opioid/opioid receptor-like (ORL1) and amine receptor families. Based on the expectation that functionally conserved regions in homologous proteins will display locally higher levels of sequence identity compared with global sequence similarities that pertain to the overall fold, this approach may have wider applications in functional genomics to annotate sequence data. Binding sites in models of the kappa-opioid receptor seven-helix bundle built from the rhodopsin templates of Baldwin et al. (1997) [J. Mol. Biol., 272, 144-164] and Herzyk and Hubbard (1998) [J. Mol. Biol., 281, 742-751] are compared. The Herzyk and Hubbard template is found to be in better accord with experimental studies of amine, opioid and rhodopsin receptors owing to the reduced physical separation of the extracellular parts of TM helices V and VI and differences in the rotational orientation of the N-terminal of helix V that reveal side chain accessibilities in the Baldwin et al. structure to be out of phase with relative alkylation rates of engineered cysteine residues in the TM binding site of the alpha(2A)-adrenergic receptor. TM helix V in the Baldwin et al. template has been remodelled with a different proline kink to satisfy experimental constraints. A recent proposal that rotation of helix V is associated with receptor activation is critically discussed.


Assuntos
Simulação por Computador , Modelos Moleculares , Receptores Opioides/química , Sítios de Ligação , Gráficos por Computador , Proteínas de Ligação ao GTP/fisiologia , Humanos , Família Multigênica , Mutagênese Sítio-Dirigida , Ligação Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Receptores Opioides/genética , Receptores Opioides kappa/química , Rodopsina/química , Alinhamento de Sequência , Moldes Genéticos , Receptor de Nociceptina
6.
J Biol Chem ; 275(38): 29268-74, 2000 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-10880520

RESUMO

The heptadecapeptide nociceptin, also known as orphanin FQ, is the endogenous agonist of the opioid receptor-like 1 (ORL1) G protein-coupled receptor. An affinity labeling approach has been implemented to probe the interactions of the neuropeptide with the receptor using the photolabile nociceptin derivative, [p-benzoyl-l-Phe(10),Tyr(14)]nociceptin ([Bpa(10),Tyr(14)]noc). In recombinant Chinese hamster ovary cells expressing the human ORL1 receptor, [Bpa(10),Tyr(14)]noc binds the receptor with high affinity (K(i) approximately 0.7 nm) and is as potent as nociceptin in the inhibition of forskolin-induced cAMP synthesis (EC(50) approximately 0.5 nm). UV irradiation at 365 nm of the complex formed by the ORL1 receptor and radioiodinated [Bpa(10),Tyr(14)]noc results in the irreversible labeling of a glycoprotein of approximately 65 kDa, determined by SDS-polyacrylamide gel electrophoresis. Complete digestion of the partially purified 65-kDa complex with kallikrein generates a single labeled fragment (approximately 6.5 kDa) that is readily cleaved by endoproteinase Glu-C to yield a labeled fragment of approximately 3.2 kDa. Kallikrein treatment of the photoaffinity cross-linked Glu(295) --> Asp mutant receptor also yields a single labeled fragment of approximately 6.5 kDa but is resistant to further cleavage by endoproteinase Glu-C. Based upon the expected proteolytic fingerprint of the labeled receptor, the photoreactive region can be identified as ORL1-(296-302; residues Thr-Ala-Val-Ala-Ile-Leu-Arg) spanning the C terminus of extracellular loop 3 and the N terminus of transmembrane helix VII. Molecular modeling of the ORL1 receptor complex with [Bpa(10)]noc suggests that reaction of the Bpa carbonyl group may occur with the side chain of Ile(300) within the experimentally identified photoreactive region.


Assuntos
Peptídeos Opioides/química , Receptores Opioides/química , Animais , Sítios de Ligação , Células CHO , Cricetinae , Humanos , Ligantes , Peptídeos Opioides/metabolismo , Marcadores de Fotoafinidade , Ligação Proteica , Receptores Opioides/metabolismo , Receptor de Nociceptina , Nociceptina
7.
Mol Pharmacol ; 57(3): 495-502, 2000 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10692489

RESUMO

A site-directed mutagenesis approach has been used to gain insight into the molecular events whereby the heptadecapeptide nociceptin binds and activates the opioid receptor-like 1 (ORL1) receptor, a G protein-coupled receptor. Alanine mutation, in the human ORL1 receptor, of transmembrane amino acid residues that are conserved in opioid receptors, Asp(130) and Tyr(131) in transmembrane segment (TM) III, Phe(220) and Phe(224) in TM V, and Trp(276) in TM VI, yields mutant receptors with reduced affinity, and proportionally decreased reactivity, toward nociceptin. Least to most deleterious in this respect are Ala substitutions of Phe(220) approximately W276A < Tyr(131) << Phe(224) 10,000 nM compared with 0.8 nM at the wild-type receptor). In all respects, this mutant receptor appears to be functionally inactive, indicating that residue Gln(286) may play a pivotal role in ORL1 receptor-mediated transduction of the nociceptin signal.


Assuntos
Alanina/metabolismo , Glutamina/metabolismo , Receptores Opioides/metabolismo , Alanina/genética , Substituição de Aminoácidos , Animais , Asparagina/genética , Asparagina/metabolismo , Células CHO , Cricetinae , Glutamina/genética , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Mutagênese Sítio-Dirigida , Fenilalanina/genética , Fenilalanina/metabolismo , Conformação Proteica , Receptores Opioides/química , Receptores Opioides/genética , Transdução de Sinais , Triptofano/genética , Triptofano/metabolismo , Tirosina/genética , Tirosina/metabolismo , Receptor de Nociceptina
8.
Neurochem Int ; 36(1): 7-18, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10566954

RESUMO

[125I]17alpha-hydroxy-20alpha-yohimban-16beta-(N-4-p6 hydroxyphenethyl)carboxamide or [125I]rauwolscine-OHPC, a new radioiodinated probe derived from rauwolscine was synthesized and its binding characteristics investigated on sections of the mouse caudate putamen. [125I]rauwolscine-OHPC binding was saturable and revealed interaction with a single class of binding sites (KD= 0.171 nM, Bmax = 3082 pCi/mg of tissue). The kinetically derived affinity was in close agreement with the affinity evaluated by saturation experiments: k(-1)/k(+1)(0.0403 min(-1)/114 10(6) M(-1) min(-1))=0.35 nM. Competition studies revealed interaction with one single class of binding sites for each of the twelve compounds tested. The rank of potency suggested an interaction with alpha2 adrenoceptors (atipamezole > or = RX 821002 > yohimbine > (-)epinephrine). Moreover, the good affinity of [125I] rauwolscine-OHPC binding sites for spiroxatrine, yohimbine, WB 4101, the relatively good affinity for prazosin (Ki =37.4 nM) and the affinity ratio prazosin/oxymetazoline (37.4/43.4=0.86) were consistent with an alpha2C selective labelling of [125I]rauwolscine-OHPC. The distribution of [125I]rauwolscine-OHPC binding sites in mouse brain was characterized by autoradiography. The density of binding sites was high in the islands of Calleja, accumbens nucleus, caudate putamen and olfactory tubercles, moderate in the hippocampus, amygdala and anterodorsal nucleus of the thalamus. These findings demonstrated that [125I]rauwolscine-OHPC is a useful radioiodinated probe to label alpha2C adrenoceptors in mouse brain.


Assuntos
Encéfalo/metabolismo , Receptores Adrenérgicos alfa 2/metabolismo , Ioimbina/análogos & derivados , Animais , Autorradiografia , Sítios de Ligação , Ligação Competitiva , Núcleo Caudado/metabolismo , Epinefrina/farmacologia , Radioisótopos do Iodo , Cinética , Masculino , Camundongos , Camundongos Endogâmicos , Estrutura Molecular , Especificidade de Órgãos , Oximetazolina/farmacologia , Prazosina/farmacologia , Putamen/metabolismo , Receptores Adrenérgicos alfa 2/análise , Ioimbina/síntese química , Ioimbina/farmacocinética , Ioimbina/farmacologia
9.
Mol Pharmacol ; 55(2): 324-31, 1999 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9927625

RESUMO

To understand how two structurally analogous ligand-receptor systems, the nociceptin/opioid receptor-like 1 (ORL1) and dynorphin A/kappa-opioid receptor 1 (KOR1) systems, achieve selectivity, receptor chimeras were generated and analyzed. Replacing discrete domains located between the N-terminus and top of the third transmembrane helix of the KOR1 by the homologous domains of the ORL1 receptor yields hybrid receptors, which, in comparison with the parent KOR1, display up to 300-fold increased affinity but low sensitivity toward nociceptin, and unaltered (high) affinity and sensitivity toward dynorphin A. These substitutions contribute elements for binding of nociceptin but do not suppress determinants necessary for binding and potency of dynorphin A. More importantly, further replacement in these chimeras of the second extracellular loop with that of the ORL1 receptor fully restores responsiveness to nociceptin without impairing responsiveness to dynorphin A. A bifunctional hybrid receptor has thus been identified that binds and responds to both nociceptin and dynorphin A as efficiently as the ORL1 receptor does to nociceptin and the KOR1 to dynorphin A. Together, these results suggest that distinct peptide activation mechanisms operate in the two receptor systems. In particular, the second extracellular receptor loop appears to be an absolute requirement for activation of the ORL1 receptor by nociceptin, but not for activation of the KOR1 by dynorphin A.


Assuntos
Dinorfinas/metabolismo , Peptídeos Opioides/metabolismo , Receptores Opioides kappa/metabolismo , Receptores Opioides/metabolismo , Animais , Sítios de Ligação , Ligação Competitiva/efeitos dos fármacos , Células CHO , Cricetinae , Dinorfinas/farmacologia , Peptídeos Opioides/farmacologia , Estrutura Secundária de Proteína , Receptores Opioides/química , Receptores Opioides/genética , Receptores Opioides kappa/genética , Proteínas Recombinantes de Fusão/efeitos dos fármacos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Relação Estrutura-Atividade , Receptor de Nociceptina , Nociceptina
10.
Protein Eng ; 11(12): 1163-79, 1998 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9930666

RESUMO

The opioid receptor like (ORL1) receptor is a G-protein coupled receptor superfamily, and regulates a plethora of neurophysiological functions. The structural requirements for receptor activation by its endogenous agonist, nociceptin (FGGFTGARKSARKLANQ), differ markedly from those of the kappa-opioid receptor and its putative peptide agonist, dynorphin A (YGGFLRRIRPKLKWDNQ). In order to probe the functional architecture of the ORL1 receptor, a molecular model of the receptor has been built, including the TM domain and the extra- and intracellular loops. An extended binding site able to accommodate nociceptin-(1-13), the shortest fully active analogue of nociceptin, has been characterized. The N-terminal FGGF tetrapeptide is proposed to bind in a highly conserved region, comprising two distinct hydrophobic pockets in a cavity formed by TM helices 3, 5, 6 and 7, capped by the acidic second extracellular (EL2) loop controlling access to the TM elements of the peptide binding site. The nociceptin conformation provides for the selective preference of the ORL1 receptor for nociceptin over dynorphin A, conferred by residue positions 5 and 6 (TG versus LR), and the favourable interaction of its highly positively charged core (residues 8-13) with the EL2 loop, thought to mediate receptor activation. The functional roles of the EL2 loop and the conserved N-terminal tetrapeptide opioid 'message' binding site are discussed in the context of the different structural requirements of the ORL1 and kappa-opioid receptors for activation.


Assuntos
Modelos Moleculares , Peptídeos Opioides/metabolismo , Receptores Opioides/química , Receptores Opioides/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Bovinos , Simulação por Computador , Sequência Conservada , Dinorfinas/metabolismo , Humanos , Dados de Sequência Molecular , Peptídeos Opioides/química , Conformação Proteica , Estrutura Secundária de Proteína , Receptores Opioides/agonistas , Alinhamento de Sequência , Receptor de Nociceptina , Nociceptina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...