Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 244
Filtrar
1.
Br J Pharmacol ; 180(17): 2298-2314, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37021779

RESUMO

BACKGROUND AND PURPOSE: In animal models of sepsis, increased activation of the Nociceptin/Orphanin FQ (N/OFQ) receptor NOP is associated with mortality and NOP antagonists improved survival. We have explored the role of the N/OFQ-NOP system in freshly isolated volunteer human B- and T-cells incubated with lipopolysaccharide (LPS) and peptidoglycan G (PepG) as a model of in vitro sepsis. EXPERIMENTAL APPROACH: B- and T-cell NOP expression was measured using the NOP fluorescent probe N/OFQATTO594 , N/OFQ content was measured using immunofluorescence, N/OFQ release was tracked using a CHOhNOPGαiq5 biosensor assay and NOP function was measured using transwell migration and cytokine/chemokine release using a 25-plex assay format. Cells were challenged with LPS/PepG. KEY RESULTS: CD19-positive B-cells bound N/OFQATTO594 ; they also contain N/OFQ. Stimulation with CXCL13/IL-4 increased N/OFQ release. N/OFQ trended to reduced migration to CXCL13/IL-4. Surface NOP expression was unaffected by LPS/PepG, but this treatment increased GM-CSF release in an N/OFQ sensitive manner. CD3-positive T-cells did not bind N/OFQATTO594 ; they did contain N/OFQ. Stimulation with CXCL12/IL-6 increased N/OFQ release. When incubated with LPS/PepG, NOP surface expression was induced leading to N/OFQATTO594 binding. In LPS/PepG-treated cells, N/OFQ reduced migration to CXCL12/IL-6. LPS/PepG increased GM-CSF release in an N/OFQ sensitive manner. CONCLUSIONS AND IMPLICATIONS: We suggest both a constitutive and sepsis-inducible N/OFQ-NOP receptor autocrine regulation of B- and T-cell function, respectively. These NOP receptors variably inhibit migration and reduce GM-CSF release. These data provide mechanistic insights to the detrimental role for increased N/OFQ signalling in sepsis and suggest a potential role for NOP antagonists as treatments.


Assuntos
Receptores Opioides , Sepse , Animais , Humanos , Receptores Opioides/metabolismo , Receptor de Nociceptina , Fator Estimulador de Colônias de Granulócitos e Macrófagos , Lipopolissacarídeos/farmacologia , Interleucina-4 , Interleucina-6 , Peptídeos Opioides/fisiologia , Sepse/tratamento farmacológico , Nociceptina
2.
Peptides ; 161: 170943, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36621672

RESUMO

The most commonly used opioid analgesics are limited by their severe side-effects in the clinical treatment of pain. Preliminary reports indicate that the combination of classical opioids and N/OFQ receptor (NOP) ligands may be an effective strategy to reduce unwanted side-effects and improve antinociception. But the interaction of these two receptor ligands in pain regulation at the peripheral level remains unclear. In this study, the antinociception of a designed amide analogue of the mu opioid receptor (MOP) peptide agonist DAMGO, DAMGO-NH2, and its antinociceptive interaction with the peripherally limited NOP peptide agonist NOP01 was investigated in two mouse models of formalin pain. Our results showed that DAMGO-NH2 acted as a MOP agonist in in vitro functional assays. Moreover, local subcutaneous or intraplantar injection of DAMGO-NH2 exerted dose-related antinociception in both phases of the formalin orofacial and intraplantar pain, which could be mediated by the classical opioid receptor. Peripheral but not central pretreatment with the peripherally restricted opioid antagonist naloxone methiodide inhibited local DAMGO-NH2-induced antinociception, supporting the involvement of the peripheral opioid receptor in local DAMGO-NH2-induced antinociception. Furthermore, co-administration of the inactive doses of DAMGO-NH2 and NOP01 produced effective antinociception. More importantly, isobolographic analysis indicates that the combination of DAMGO-NH2 and NOP01 elicited supra-additive antinociception in these two models of formalin pain. In addition, the combination of DAMGO-NH2 and NOP01 did not change motor function of mice in rotarod test. In conclusion, these data suggest that peripheral DAMGO-NH2 and particularly its combination therapy with NOP01 may be effective for pain management.


Assuntos
Analgésicos Opioides , Dor , Receptores Opioides , Animais , Camundongos , Analgésicos/farmacologia , Analgésicos/uso terapêutico , Analgésicos Opioides/farmacologia , Analgésicos Opioides/uso terapêutico , Relação Dose-Resposta a Droga , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Ala(2)-MePhe(4)-Gly(5)-Encefalina/uso terapêutico , Ligantes , Receptor de Nociceptina , Dor/tratamento farmacológico , Peptídeos/uso terapêutico , Receptores Opioides/agonistas , Receptores Opioides mu/agonistas , Interações Medicamentosas
3.
Br J Pharmacol ; 179(2): 287-300, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34705263

RESUMO

BACKGROUND AND PURPOSE: Partial agonists of the nociceptin opioid peptide (NOP) receptor have potential therapeutic use as antihypertensive and water diuretics (aquaretics). To date, peptide NOP receptor ligands have failed to progress in clinical trials due to poor pharmacokinetics and adverse effects. Nonpeptide, small-molecule NOP receptor ligands may be more suitable as therapeutic agents. This study investigated the cardiovascular and renal responses produced by the novel nonpeptide NOP agonists AT-403, AT-090, AT-127, and AT-039. EXPERIMENTAL APPROACH: Changes in mean arterial pressure (MAP), heart rate (HR), renal excretory function and occurrence of sedation and hyperphagia were determined before and after i.v. bolus injection or infusion of the NOP agonists in conscious Sprague-Dawley rats. Additional studies involving (i) measurement of renal sympathetic nerve activity (RSNA) and (ii) renal denervation were conducted to investigate the role of the renal nerves in the cardiorenal responses to AT-039. KEY RESULTS: Bolus i.v. injection of AT-403, AT-090, AT-127 and AT-039 produced significant decreases in MAP and HR and a sodium-sparing diuresis. AT-403, AT-090, and AT-127, but not AT-039, induced sedation and hyperphagia at all doses tested. Infusion i.v. of AT-039 produced hypotension and aquaresis without adverse central nervous system effects or change in HR, responses that were also observed in renal denervated rats. CONCLUSIONS AND IMPLICATIONS: Nonpeptide NOP agonists decrease blood pressure and produce aquaresis in conscious rodents. Due to lack of sedation and hyperphagia, AT-039 represents a novel NOP agonist that may be useful for treatment of hypertension and/or volume overload/hyponatraemic states.


Assuntos
Analgésicos Opioides , Receptores Opioides , Analgésicos Opioides/farmacologia , Animais , Hiperfagia , Hipertensão/tratamento farmacológico , Hipertensão/metabolismo , Ligantes , Ratos , Ratos Sprague-Dawley , Receptores Opioides/agonistas , Receptor de Nociceptina
4.
Br J Pharmacol ; 179(11): 2647-2658, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-34854073

RESUMO

BACKGROUND AND PURPOSE: The nociceptin/orphanin FQ (N/OFQ)-nociceptin opioid-like peptide (NOP) receptor system is widely distributed in the brain and pharmacological activation of this system revealed therapeutic potential in animal models of substance use disorder. Studies also showed that genetic deletion or pharmacological blockade of NOP receptors confer resistance to the development of alcohol abuse. Here, we have used a genetic and pharmacological approach to evaluate the therapeutic potential of NOP antagonism in smoking cessation. EXPERIMENTAL APPROACH: Constitutive NOP receptor knockout rats (NOP-/- ) and their wild-type counterparts (NOP+/+ ) were tested over a range of behaviours to characterize their motivation for nicotine. We next explored the effects of systemic administration of the NOP receptor antagonist LY2817412 (1.0 & 3.0 mg·kg-1 ) on nicotine self-administration. NOP receptor blockade was further evaluated at the brain circuitry level, by microinjecting LY2817412 (3.0 & 6.0 µg·µl-1 ) into the ventral tegmental area (VTA), nucleus accumbens (NAc) and central amygdala (CeA). KEY RESULTS: Genetic NOP receptor deletion resulted in decreased nicotine intake, decreased motivation to self-administer and attenuation of cue-induced nicotine reinstatement. LY2817412 reduced nicotine intake in NOP+/+ but not in NOP-/- rats, confirming that its effect is mediated by inhibition of NOP transmission. Finally, injection of LY2817412 into the VTA but not into the NAc or CeA decreased nicotine self-administration. CONCLUSIONS AND IMPLICATIONS: These findings indicate that inhibition of NOP transmission attenuates the motivation for nicotine through mechanisms involving the VTA and suggest that NOP receptor antagonism may represent a potential treatment for smoking cessation.


Assuntos
Nicotina , Área Tegmentar Ventral , Animais , Nicotina/farmacologia , Peptídeos Opioides/metabolismo , Ratos , Receptores Opioides , Área Tegmentar Ventral/metabolismo , Receptor de Nociceptina , Nociceptina
5.
Int J Mol Sci ; 22(23)2021 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-34884858

RESUMO

The melanocortin system is a major regulator of stress responses in the skin and is responsible for the induction of melanin synthesis through activation of melanogenesis enzymes. The expression of both melanocortin system genes and melanogenesis enzyme genes is altered in psoriasis, and the focus here was on twelve genes related to the signal transduction between them. Additionally, five endogenous opioid system genes that are involved in cutaneous inflammation were examined. Quantitative real-time-PCR was utilized to measure mRNA expression in punch biopsies from lesional and non-lesional skin of psoriasis patients and from the skin of healthy control subjects. Most of the genes related to melanogenesis were down-regulated in patients (CREB1, MITF, LEF1, USF1, MAPK14, ICAM1, PIK3CB, RPS6KB1, KIT, and ATRN). Conversely, an up-regulation occurred in the case of opioids (PENK, PDYN, and PNOC). The suppression of genes related to melanogenesis is in agreement with the reported reduction in pigmentation signaling in psoriatic skin and potentially results from the pro-inflammatory environment. The increase in endogenous opioids can be associated with their involvement in inflammatory dysregulation in psoriasis.


Assuntos
Psoríase/genética , Psoríase/patologia , Pigmentação da Pele/genética , Adolescente , Adulto , Analgésicos Opioides/metabolismo , Biópsia , Estudos de Casos e Controles , Classe I de Fosfatidilinositol 3-Quinases/genética , Encefalinas/genética , Feminino , Perfilação da Expressão Gênica , Humanos , Masculino , Fator de Transcrição Associado à Microftalmia/genética , Precursores de Proteínas/genética , Receptores Opioides/genética , Pele/patologia , Adulto Jovem , Receptor de Nociceptina
6.
J Med Chem ; 64(10): 6656-6669, 2021 05 27.
Artigo em Inglês | MEDLINE | ID: mdl-33998786

RESUMO

The nociceptin/orphanin FQ (N/OFQ)/N/OFQ receptor (NOP) system controls different biological functions including pain and cough reflex. Mixed NOP/opioid receptor agonists elicit similar effects to strong opioids but with reduced side effects. In this work, 31 peptides with the general sequence [Tyr/Dmt1,Xaa5]N/OFQ(1-13)-NH2 were synthesized and pharmacologically characterized for their action at human recombinant NOP/opioid receptors. The best results in terms of NOP versus mu opioid receptor potency were obtained by substituting both Tyr1 and Thr5 at the N-terminal portion of N/OFQ(1-13)-NH2 with the noncanonical amino acid Dmt. [Dmt1,5]N/OFQ(1-13)-NH2 has been identified as the most potent dual NOP/mu receptor peptide agonist so far described. Experimental data have been complemented by in silico studies to shed light on the molecular mechanisms by which the peptide binds the active form of the mu receptor. Finally, the compound exerted antitussive effects in an in vivo model of cough.


Assuntos
Peptídeos/química , Receptores Opioides mu/agonistas , Receptores Opioides/agonistas , Animais , Sítios de Ligação , Tosse/induzido quimicamente , Tosse/tratamento farmacológico , Modelos Animais de Doenças , Cobaias , Humanos , Interações Hidrofóbicas e Hidrofílicas , Masculino , Simulação de Dinâmica Molecular , Peptídeos/metabolismo , Peptídeos/uso terapêutico , Receptores Opioides/metabolismo , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/isolamento & purificação , Relação Estrutura-Atividade , Receptor de Nociceptina
7.
Cell Mol Neurobiol ; 41(5): 1039-1055, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33010019

RESUMO

The opioid receptor (OPR) family comprises the mu-, delta-, and kappa-opioid, and nociceptin receptors that belong to the superfamily of 7-transmembrane spanning G protein-coupled receptors (GPCRs). The mu-opioid receptor is the main target for clinically used opioid analgesics, and its biology has been extensively studied. The N-terminally truncated 6TM receptors isoform produced through alternative splicing of the OPRM1 gene displays unique signaling and analgesic properties, but it is unclear if other OPRs have the same ability. In this study, we have built a comprehensive map of alternative splicing events that produce 6TM receptor variants in all the OPRs and demonstrated their evolutionary conservation. We then obtained evidence for their translation through ribosomal footprint analysis. We discovered that N-terminally truncated 6TM GPCRs are rare in the human genome and OPRs are overrepresented in this group. Finally, we also observed a significant enrichment of 6TM GPCR genes among genes associated with pain, psychiatric disorders, and addiction. Understanding the biology of 6TM receptors and leveraging this knowledge for drug development should pave the way for novel therapies.


Assuntos
Processamento Alternativo/genética , Sequência Conservada/genética , Receptores Opioides delta/genética , Receptores Opioides kappa/genética , Receptores Opioides mu/genética , Receptores Opioides/genética , Animais , Linhagem Celular Tumoral , Bases de Dados Genéticas , Variação Genética/genética , Humanos , Macaca , Camundongos , Especificidade da Espécie , Receptor de Nociceptina
8.
Br J Pharmacol ; 178(9): 1995-2007, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-31724155

RESUMO

BACKGROUND AND PURPOSE: Cancer-induced bone pain remains inadequately controlled, and current standard of care analgesics is accompanied by several side effects. Nociceptin/orphanin FQ peptide (NOP) receptor agonists have demonstrated broad analgesic properties in rodent neuropathic and inflammatory pain models. Here, we investigate the analgesic potential of NOP receptor activation in a rodent cancer-induced bone pain model. EXPERIMENTAL APPROACH: Model validation by intratibial inoculation in male Sprague Dawley rats was performed with varying MRMT-1/Luc2 cell quantities (0.5-1.5 × 106 ·ml-1 ) and a behavioural battery (>14 days post-surgery) including evoked and non-evoked readouts: paw pressure test, cold plate, von Frey, open field, and weight distribution. Anti-allodynic potential of the endogenous NOP receptor ligand nociceptin (i.t.) and NOP receptor agonist Ro65-6570 ( i.p.) was tested using von Frey filaments, followed by a combination experiment with Ro65-6570 and the NOP receptor antagonist J-113397 (i.p.). Plasma cytokine levels and NOP receptor gene expression in dorsal root ganglion (DRG, L4-L6) and bone marrow were examined. KEY RESULTS: Inoculation with 1.5 × 106 ·ml-1 of MRMT-1/Luc2 cells resulted in a robust and progressive pain-related phenotype. Nociceptin and Ro65-6570 treatment inhibited cancer-induced mechanical allodynia. J-113397 selectively antagonized the effect of Ro65-6570. MRMT-1/Luc2-bearing animals demonstrated elevated plasma cytokine levels of IL-4, IL-5, IL-6 and IL-10 plus unaltered NOP-r gene expression in DRG and reduced expression in bone marrow. CONCLUSION AND IMPLICATIONS: Nociceptin and Ro65-6570 selectively and dose-dependently reversed cancer-induced bone pain-like behaviour. The NOP receptor system may be a potential target for cancer-induced bone pain treatment. LINKED ARTICLES: This article is part of a themed issue on The molecular pharmacology of bone and cancer-elated bone diseases. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.9/issuetoc.


Assuntos
Doenças Ósseas , Neoplasias , Animais , Masculino , Peptídeos Opioides , Dor/tratamento farmacológico , Ratos , Ratos Sprague-Dawley , Receptores Opioides , Receptor de Nociceptina , Nociceptina
9.
Eur J Pharmacol ; 883: 173306, 2020 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-32603693

RESUMO

Chemotherapy-induced peripheral neuropathy is a serious adverse effect of chemotherapeutic agents such as paclitaxel. JTC-801, a nociceptin/orphanin FQ opioid peptide (NOP) receptor antagonist, has been reported to attenuate neuropathic pain in several pain models. However, the therapeutic significance and function of JTC-801 in chemotherapy-induced peripheral neuropathy remain unclear. In this study, we determined the effect of JTC-801 on neuropathic pain induced by paclitaxel, and we explored the potential mechanism in the dorsal root ganglion (DRG). The behavioral test showed that single or multiple systemic administrations of JTC-801 significantly alleviated mechanical allodynia in paclitaxel-treated rats. Using Western blot analysis and immunohistochemistry, we found that paclitaxel increased the expression of phosphatidylinositol 3-kinase (PI3K) and phospho-Akt (p-Akt) in the DRG. Double immunofluorescence staining indicated that p-Akt was expressed in neurons in the DRG. Multiple injections of JTC-801 significantly inhibited the activation of Akt and decreased the expression of inflammatory cytokines. The data suggest that JTC-801 alleviates mechanical allodynia associated with paclitaxel-induced neuropathic pain via the PI3K/Akt pathway.


Assuntos
Aminoquinolinas/farmacologia , Analgésicos/farmacologia , Benzamidas/farmacologia , Gânglios Espinais/efeitos dos fármacos , Hiperalgesia/prevenção & controle , Antagonistas de Entorpecentes/farmacologia , Neuralgia/prevenção & controle , Limiar da Dor/efeitos dos fármacos , Fosfatidilinositol 3-Quinase/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Comportamento Animal/efeitos dos fármacos , Modelos Animais de Doenças , Gânglios Espinais/enzimologia , Gânglios Espinais/fisiopatologia , Hiperalgesia/induzido quimicamente , Hiperalgesia/enzimologia , Hiperalgesia/fisiopatologia , Mediadores da Inflamação/metabolismo , Interleucina-1beta/metabolismo , Masculino , Neuralgia/induzido quimicamente , Neuralgia/enzimologia , Neuralgia/fisiopatologia , Paclitaxel , Fosforilação , Ratos Sprague-Dawley , Receptores Opioides/efeitos dos fármacos , Receptores Opioides/metabolismo , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo , Receptor de Nociceptina
10.
Medicine (Baltimore) ; 99(22): e20429, 2020 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-32481444

RESUMO

Determining the clinically optimal dose in methadone maintenance therapy (MMT) is a time-consuming procedure, which considers clinical signs and symptoms.To perform a quantitative trait locus association for identifying genetic variants for MMT dosage that underlie heroin addiction and methadone metabolism and then integrate several genotypic and phenotypic factors are potential predictors for clinically optimal MMT dose for personalized prescription.In total, 316 heroin-dependent patients undergoing MMT were recruited at the Addiction Center of the China Medical University Hospital. A multinomial logistic regression model was used to assess associations between genetic polymorphisms and MMT dosing. The data were randomly separated into training and testing sets. In order to enhance the prediction accuracy and the reliability of the prediction model, we used areas under the receiver operating characteristic curves to evaluate optimal MMT dose in both training and testing sets.Four single nucleotide polymorphisms, namely rs806368 in CNR1, s1386493 in TPH2, s16974799 in CYP2B6, and rs2229205 in OPRL1, were significantly associated with the maximum MMT dose (P < .05). The genetic risk score (GRS) was associated with maximum MMT dose, and after adjustments for age, sex, and body mass index, the GRS remained independently associated with the maximum MMT dose. The area under the receiver operating characteristic curve of the combined GRS and craving score was 0.77 for maximum MMT dose, with 75% sensitivity and 60% specificity.Integrating the GRS and craving scores may be useful in the evaluation of individual MMT dose requirements at treatment initiation. Optimal dose prediction allows clinicians to tailor MMT to each patient's needs.


Assuntos
Fissura , Dependência de Heroína/tratamento farmacológico , Dependência de Heroína/genética , Metadona/administração & dosagem , Entorpecentes/administração & dosagem , Medicina de Precisão , Adulto , Citocromo P-450 CYP2B6/genética , Feminino , Estudos de Associação Genética , Marcadores Genéticos , Humanos , Masculino , Tratamento de Substituição de Opiáceos , Polimorfismo de Nucleotídeo Único , Locos de Características Quantitativas , Receptor CB1 de Canabinoide/genética , Receptores Opioides/genética , Sensibilidade e Especificidade , Índice de Gravidade de Doença , Triptofano Hidroxilase/genética , Receptor de Nociceptina
11.
Front Immunol ; 11: 300, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32194554

RESUMO

Opioid receptors comprise µ (MOP), δ (DOP), κ (KOP), and nociceptin/orphanin FQ (NOP) receptors. Opioids are agonists of MOP, DOP, and KOP receptors, whereas nociceptin/orphanin FQ (N/OFQ) is an agonist of NOP receptors. Activation of all four opioid receptors in neurons can induce analgesia in animal models, but the most clinically relevant are MOP receptor agonists (e.g., morphine, fentanyl). Opioids can also affect the function of immune cells, and their actions in relation to immunosuppression and infections have been widely discussed. Here, we analyze the expression and the role of opioid receptors in peripheral immune cells and glia in the modulation of pain. All four opioid receptors have been identified at the mRNA and protein levels in immune cells (lymphocytes, granulocytes, monocytes, macrophages) in humans, rhesus monkeys, rats or mice. Activation of leukocyte MOP, DOP, and KOP receptors was recently reported to attenuate pain after nerve injury in mice. This involved intracellular Ca2+-regulated release of opioid peptides from immune cells, which subsequently activated MOP, DOP, and KOP receptors on peripheral neurons. There is no evidence of pain modulation by leukocyte NOP receptors. More good quality studies are needed to verify the presence of DOP, KOP, and NOP receptors in native glia. Although still questioned, MOP receptors might be expressed in brain or spinal cord microglia and astrocytes in humans, mice, and rats. Morphine acting at spinal cord microglia is often reported to induce hyperalgesia in rodents. However, most studies used animals without pathological pain and/or unconventional paradigms (e.g., high or ultra-low doses, pain assessment after abrupt discontinuation of chronic morphine treatment). Therefore, the opioid-induced hyperalgesia can be viewed in the context of dependence/withdrawal rather than pain management, in line with clinical reports. There is convincing evidence of analgesic effects mediated by immune cell-derived opioid peptides in animal models and in humans. Together, MOP, DOP, and KOP receptors, and opioid peptides in immune cells can ameliorate pathological pain. The relevance of NOP receptors and N/OFQ in leukocytes, and of all opioid receptors, opioid peptides and N/OFQ in native glia for pain control is yet to be clarified.


Assuntos
Leucócitos/fisiologia , Macrófagos/fisiologia , Neuroglia/fisiologia , Dor/fisiopatologia , Receptores Opioides/fisiologia , Animais , Humanos , Receptor de Nociceptina
12.
Peptides ; 124: 170223, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31805297

RESUMO

This paper is the fortieth consecutive installment of the annual anthological review of research concerning the endogenous opioid system, summarizing articles published during 2017 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides and receptors as well as effects of opioid/opiate agonists and antagonists. The review is subdivided into the following specific topics: molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors (1), the roles of these opioid peptides and receptors in pain and analgesia in animals (2) and humans (3), opioid-sensitive and opioid-insensitive effects of nonopioid analgesics (4), opioid peptide and receptor involvement in tolerance and dependence (5), stress and social status (6), learning and memory (7), eating and drinking (8), drug abuse and alcohol (9), sexual activity and hormones, pregnancy, development and endocrinology (10), mental illness and mood (11), seizures and neurologic disorders (12), electrical-related activity and neurophysiology (13), general activity and locomotion (14), gastrointestinal, renal and hepatic functions (15), cardiovascular responses (16), respiration and thermoregulation (17), and immunological responses (18).


Assuntos
Analgésicos Opioides/farmacologia , Peptídeos Opioides/farmacologia , Peptídeos Opioides/fisiologia , Receptores Opioides/metabolismo , Transtornos Relacionados ao Uso de Substâncias/etiologia , Animais , Dor do Câncer/tratamento farmacológico , Dor Crônica/tratamento farmacológico , Dor Crônica/genética , Ingestão de Alimentos/efeitos dos fármacos , Emoções , Feminino , Humanos , Aprendizagem/efeitos dos fármacos , Masculino , Memória/efeitos dos fármacos , Peptídeos Opioides/agonistas , Peptídeos Opioides/antagonistas & inibidores , Transtornos Relacionados ao Uso de Opioides/etiologia , Transtornos Relacionados ao Uso de Opioides/prevenção & controle , Gravidez , Estresse Fisiológico , Receptor de Nociceptina
13.
Neuropharmacology ; 166: 107678, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31278929

RESUMO

There is unmet need to design an analgesic with fewer side effects for severe pain management. Although traditional opioids are the most effective painkillers, they are accompanied by severe adverse responses, such as respiratory depression, constipation symptoms, tolerance, withdrawal, and addiction. We indicated BPR1M97 as a dual mu opioid receptor (MOP)/nociceptin-orphanin FQ peptide (NOP) receptor full agonist and investigated the pharmacology of BPR1M97 in multiple animal models. In vitro studies on BPR1M97 were assessed using cyclic-adenosine monophosphate production, ß-arrestin, internalization, and membrane potential assays. In vivo studies were characterized using the tail-flick, tail-clip, lung functional, heart functional, acetone drop, von Frey hair, charcoal meal, glass bead, locomotor activity, conditioned place preference (CPP) and naloxone precipitation tests. BPR1M97 elicited full agonist properties for all cell-based assays tested in MOP-expressing cells. However, it acted as a G protein-biased agonist for NOP. BPR1M97 initiated faster antinociceptive effects at 10 min after subcutaneous injection and elicited better analgesia in cancer-induced pain than morphine. Unlike morphine, BPR1M97 caused less respiratory, cardiovascular, and gastrointestinal dysfunction. In addition, BPR1M97 decreased global activity and induced less withdrawal jumping precipitated by naloxone. Thus, BPR1M97 could serve as a novel small molecule dual receptor agonist for antinociception with fewer side effects than morphine. This article is part of the Special Issue entitled 'New Vistas in Opioid Pharmacology'.


Assuntos
Analgésicos Opioides/uso terapêutico , Analgésicos/uso terapêutico , Morfina/uso terapêutico , Medição da Dor/efeitos dos fármacos , Receptores Opioides mu/agonistas , Receptores Opioides/agonistas , Analgésicos/farmacologia , Analgésicos Opioides/farmacologia , Animais , Células CHO , Dor do Câncer/tratamento farmacológico , Dor do Câncer/patologia , Cricetulus , Relação Dose-Resposta a Droga , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Morfina/farmacologia , Medição da Dor/métodos , Resultado do Tratamento , Receptor de Nociceptina
14.
Bioconjug Chem ; 30(9): 2444-2451, 2019 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-31461266

RESUMO

The general aim of the work was the validation of a new synthetic methodology designed for obtaining bifunctional heterotetrabranched peptide ligands. Applying an easily accessible synthetic route, we provided a small series of heteromultimeric peptide conjugates targeting the nociceptin/orphanin FQ (N/OFQ) peptide receptors (NOP) and mu opioid receptors. Among these, H-PWT1-N/OFQ-[Dmt1]dermorphin demonstrated a similar and high agonist potency at the NOP and mu receptors. The achieved results confirmed the robustness of the approach that is extremely versatile and virtually applicable to different peptide sequences whose pharmacological activity can be combined for generating dual acting multimeric compounds. These innovative pharmacological tools will be extremely helpful for investigating the consequences of the simultaneous activation and/or blockage of different peptidergic receptors.


Assuntos
Peptídeos/química , Peptídeos/farmacologia , Receptores Opioides mu/agonistas , Receptores Opioides/agonistas , Animais , Benzaldeídos/química , Células CHO , Cricetulus , Humanos , Maleimidas/química , Receptor de Nociceptina
15.
Am J Psychiatry ; 176(6): 468-476, 2019 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-31055968

RESUMO

OBJECTIVE: Nociceptin/orphanin FQ (N/OFQ) is an antistress neuropeptide transmitter in the brain that counteracts corticotropin-releasing factor (CRF)-mediated stress and anxiety symptoms during drug and alcohol withdrawal. It also inhibits the release of a wide array of neurotransmitters, including dopamine and glutamate, which allows for it to block the rewarding properties of cocaine. Chronic cocaine administration in rodents has been shown to decrease N/OFQ and increase nociceptive opioid peptide (NOP) receptors in the nucleus accumbens. No previous studies have reported on the in vivo status of NOP in chronic cocaine-abusing humans. METHODS: [11C]NOP-1A and positron emission tomography (PET) were used to measure in vivo NOP binding in 24 individuals with cocaine use disorder and 26 healthy control subjects matched for age, sex, and smoking status. Participants with cocaine use disorder with no comorbid psychiatric or medical disorders were scanned after 2 weeks of outpatient-monitored abstinence. [11C]NOP-1A distribution volume (VT) was measured with kinetic analysis using the arterial input function in brain regions that mediate reward and stress behaviors. Participants with cocaine use disorder were followed up for 12 weeks after PET scanning to document relapse and relate it to VT. RESULTS: A significant increase in [11C]NOP-1A VT was observed in the cocaine use disorder group compared with the healthy control group. This increase, which was generalized across all regions of interest (approximately 10%), was most prominent in the midbrain, ventral striatum, and cerebellum. However, increased VT in these regions did not predict relapse. CONCLUSIONS: Increased NOP in cocaine use disorder suggests an adaptive response to decreased N/OFQ, or increased CRF transmission, or both. Future studies should examine the interactions between CRF and NOP to elucidate their role in negative reinforcement and relapse. NOP agonist medications to enhance N/OFQ should be explored as a therapeutic to treat cocaine use disorder.


Assuntos
Encéfalo/metabolismo , Transtornos Relacionados ao Uso de Cocaína/metabolismo , Peptídeos Opioides/metabolismo , Receptores Opioides/metabolismo , Adulto , Encéfalo/diagnóstico por imagem , Compostos Bicíclicos Heterocíclicos com Pontes , Estudos de Casos e Controles , Cerebelo/metabolismo , Transtornos Relacionados ao Uso de Cocaína/diagnóstico por imagem , Dopamina/metabolismo , Feminino , Ácido Glutâmico/metabolismo , Humanos , Masculino , Mesencéfalo/metabolismo , Pessoa de Meia-Idade , Tomografia por Emissão de Pósitrons , Compostos de Espiro , Estriado Ventral/metabolismo , Receptor de Nociceptina , Nociceptina
16.
Horm Behav ; 112: 89-99, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30981690

RESUMO

In estradiol-primed nonreceptive ovariectomized rats, activation of G protein-coupled estrogen receptor 1 (GPER) in the arcuate nucleus of the hypothalamus (ARH) rapidly facilitates sexual receptivity (lordosis). Estradiol priming activates ARH ß-endorphin (ß-END) neurons that then activate medial preoptic (MPN) µ-opioid receptors (MOP) to inhibit lordosis. ARH infusion of non-esterified 17ß-estradiol (E2) 47.5 h after 17ß-estradiol benzoate (2 µg EB) priming deactivates MPN MOP and rapidly facilitates lordosis within 30 min via activation of GPER. Since it was unclear where GPERs were located in the neuron, we tested the hypothesis that GPER signaling is initiated at the plasma membrane. Membrane impermeable estradiol (17ß-estradiol conjugated to biotin; E-Biotin) infused into the ARH of EB primed rats facilitated lordosis within 30 min, and MPN MOP was deactivated. These actions were blocked by pretreating with GPER antagonist, G-15. Further, we used cell fractionation and western blot techniques to demonstrate that GPER is expressed both in plasma membrane and cytosolic ARH fractions. In previous studies, the orphanin FQ/nociceptin-opioid receptor-like receptor-1 (OFQ/N-ORL-1) system mediated estradiol-only facilitation of lordosis. Therefore, we tested whether the OFQ/N-ORL-1 system mediates E-Biotin-GPER facilitation of lordosis. Pretreatment of UFP-101, an ORL-1 selective antagonist, blocked the facilitation of lordosis and deactivation of MPN MOP by ARH infusion of E-Biotin. Double-label immunohistochemistry revealed that GPER is expressed within approximately 70% of OFQ/N neurons. These data indicate that membrane GPER mediates the E2/E-Biotin facilitation of lordosis by inducing OFQ/N neurotransmission, which inhibits ß-END neurotransmission to reduce MPN MOP activation.


Assuntos
Estradiol/farmacologia , Peptídeos Opioides/metabolismo , Postura/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Receptores Opioides/metabolismo , Comportamento Sexual Animal/efeitos dos fármacos , Animais , Estradiol/metabolismo , Feminino , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Masculino , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Ratos , Ratos Long-Evans , Comportamento Sexual Animal/fisiologia , Transdução de Sinais/efeitos dos fármacos , Receptor de Nociceptina , Nociceptina
17.
Pain ; 160(6): 1383-1401, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30720581

RESUMO

Generalized pain and fatigue are both hallmarks of fibromyalgia, a syndrome with an indefinite etiology. The treatment options for fibromyalgia are currently limited, probably because of its intricate pathophysiology. Thus, further basic and clinical research on this condition is currently needed. This study investigated the effects of nociceptin/orphanin FQ (N/OFQ) receptor (NOPr) ligands and the modulation of the NOP system in the preclinical mouse model of reserpine-induced fibromyalgia. The effects of administration of the natural agonist N/OFQ and the selective NOPr antagonists (UFP-101 and SB-612111) were evaluated in fibromyalgia-related symptoms in reserpine-treated mice. The expression of prepronociceptin/orphanin FQ and NOPr was assessed in central and peripheral sites at different time points after reserpine administration. Nociceptin/orphanin FQ displayed dual effects in the behavioral changes in the reserpine-elicited fibromyalgia model. The peptide NOPr antagonist UFP-101 produced analgesic and antifatigue effects, by preventing alterations in brain activity and skeletal muscle metabolism, secondary to fibromyalgia induction. The nonpeptide NOPr antagonist SB-612111 mirrored the favorable effects of UFP-101 in painful and fatigue alterations induced by reserpine. A time-related up- or downregulation of prepronociceptin/orphanin FQ and NOPr was observed in supraspinal, spinal, and peripheral sites of reserpine-treated mice. Our data shed new lights on the mechanisms underlying the fibromyalgia pathogenesis, supporting a role for N/OFQ-NOP receptor system in this syndrome.


Assuntos
Analgésicos/farmacologia , Fadiga/tratamento farmacológico , Fibromialgia/tratamento farmacológico , Peptídeos Opioides/farmacologia , Animais , Modelos Animais de Doenças , Feminino , Masculino , Camundongos , Antagonistas de Entorpecentes/farmacologia , Dor/tratamento farmacológico , Precursores de Proteínas/farmacologia , Receptores Opioides/efeitos dos fármacos , Receptor de Nociceptina , Nociceptina
18.
Phytomedicine ; 50: 285-299, 2018 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-30466988

RESUMO

BACKGROUND: Curcumin (CC) exerts polyvalent pharmacological actions and multi-target effects, including pain relief and anti-nociceptive activity. In combination with Boswellia serrata extract (BS), curcumin shows greater efficacy in knee osteoarthritis management, presumably due to synergistic interaction of the ingredients. AIM: To elucidate the molecular mechanisms underlying the analgesic activity of curcumin and its synergistic interaction with BS. METHODS: We performed gene expression profiling by transcriptome-wide mRNA sequencing in human T98G neuroglia cells treated with CC (Curamed), BS, and the combination of CC and BS (CC-BS; Curamin), followed by interactive pathways analysis of the regulated genes. RESULTS: Treatment with CC and with CC-BS selectively downregulated opioid-related nociceptin receptor 1 gene (OPRL1) expression by 5.9-fold and 7.2-fold, respectively. No changes were detected in the other canonical opioid receptor genes: OPRK1, OPRD1, and OPRM1. Nociceptin reportedly increases the sensation of pain in supra-spinal pain transduction pathways. Thus, CC and CC-BS may downregulate OPRL1, consequently inhibiting production of the nociception receptor NOP, leading to pain relief. In neuroglia cells, CC and CC-BS inhibited signaling pathways related to opioids, neuropathic pain, neuroinflammation, osteoarthritis, and rheumatoid diseases. CC and CC-BS also downregulated ADAM metallopeptidase gene ADAMTS5 expression by 11.2-fold and 13.5-fold, respectively. ADAMTS5 encodes a peptidase that plays a crucial role in osteoarthritis development via inhibition of a corresponding signaling pathway. CONCLUSION: Here, we report for the first time that CC and CC-BS act as nociceptin receptor antagonists, selectively downregulating opioid-related nociceptin receptor 1 gene (OPRL1) expression, which is associated with pain relief. BS alone did not affect OPRL1 expression, but rather appears to potentiate the effects of CC via multiple mechanisms, including synergistic interactions of molecular networks.


Assuntos
Analgésicos/farmacologia , Curcumina/farmacologia , Neuroglia/efeitos dos fármacos , Extratos Vegetais/farmacologia , Receptores Opioides/metabolismo , Proteína ADAMTS5/metabolismo , Analgésicos Opioides , Boswellia/química , Linhagem Celular Tumoral , Regulação para Baixo , Humanos , Antagonistas de Entorpecentes/farmacologia , Receptor de Nociceptina
19.
Br J Anaesth ; 121(5): 1105-1114, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30336855

RESUMO

Opioids are characterised as classical (mu, delta, and kappa) along with the non-classical nociceptin/orphanin FQ (N/OFQ) receptor or NOP. Targeting NOP has therapeutic indications in control of the cardiovascular and respiratory systems and micturition, and a profile as an antidepressant. For all of these indications, there are translational human data. Opioids such as morphine and fentanyl (activating the mu receptor) are the mainstay of pain treatment in the perioperative period, despite a challenging side-effect profile. Opioids in general have poor efficacy in neuropathic pain. Moreover, longer term use is associated with tolerance. There is good evidence interactions between opioid receptors, and receptor co-activation can reduce side-effects without compromising analgesia; this is particularly true for mu and NOP co-activation. Recent pharmaceutical development has produced a mixed opioid/NOP agonist, cebranopadol. This new chemical entity is effective in animal models of nociceptive and neuropathic pain with greater efficacy in the latter. In animal models, there is little evidence for respiratory depression, and tolerance (compared with morphine) only develops after long treatment periods. There is now early phase clinical development in diabetic neuropathy, cancer pain, and low back pain where cebranopadol displays significant efficacy. In 1996, N/OFQ was formally identified with an innovative analgesic profile. Approximately 20 yr later, cebranopadol as a clinical ligand is advancing through the human trials process.


Assuntos
Analgésicos não Narcóticos/farmacologia , Indóis/farmacologia , Receptores Opioides/agonistas , Compostos de Espiro/farmacologia , Analgésicos não Narcóticos/efeitos adversos , Analgésicos não Narcóticos/uso terapêutico , Ensaios Clínicos como Assunto , Humanos , Indóis/efeitos adversos , Indóis/uso terapêutico , Receptores Opioides mu/agonistas , Compostos de Espiro/efeitos adversos , Compostos de Espiro/uso terapêutico , Receptor de Nociceptina
20.
J Mol Neurosci ; 66(1): 10-16, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30074175

RESUMO

Opioid receptor like 1 (ORL1) receptor activation displayed an anti-nociceptive effect at spinal level for acute and neuropathic pain. SCH221510, an orally active non-peptide ORL1 agonist, was reported to be effective in treating neuropathic pain. The present study used ORL1 antagonist and siRNA to investigate that ORL1 activation mediates intrathecal SCH221510 analgesia in neuropathic pain induced by chronic constrictive injury (CCI) to rat sciatic nerve. Paw withdrawal latency and 50% mechanical threshold were measured for thermal and mechanical hypersensitivity in rats. CCI significantly decreased paw withdrawal latency and mechanical threshold. SCH221510 (3, 10, 30 µg) or ORL1 antagonist ([Nphe1]nociceptin(1-13)NH2, 10 µg) was intrathecally injected to test the behavioral effects on neuropathic pain. Intrathecal siRNA was started on 1 day before CCI surgery and maintained for 7 days. L4-L5 spinal cord ORL1 mRNA and protein were measured by real-time PCR and Western blot. The effect of intrathecal siRNA on SCH2210510 was tested in CCI rats on day 7. Intrathecal SCH221510 dose-dependently reduced thermal and mechanical hypersensitivity induced by CCI. [Nphe1]nociceptin(1-13)NH2 blocked SCH221510 analgesia in CCI rats. Intrathecal siRNA blocked ORL1 mRNA and protein increase induced by CCI. Intrathecal ORL1 siRNA did not change thermal and mechanical hypersensitivity induced by nerve injury. Intrathecal siRNA blocked SCH221510 analgesia in neuropathic pain at spinal level. Conclusively, ORL1 activation mediates SCH221510 analgesia in neuropathic pain at spinal level. The results warrant a potential clinically applicable drug in treating neuropathic pain.


Assuntos
Analgésicos/farmacologia , Compostos Azabicíclicos/farmacologia , Neuralgia/metabolismo , Receptores Opioides/metabolismo , Analgésicos/uso terapêutico , Animais , Compostos Azabicíclicos/uso terapêutico , Masculino , Neuralgia/tratamento farmacológico , Peptídeos Opioides/farmacologia , Fragmentos de Peptídeos/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores Opioides/agonistas , Receptores Opioides/genética , Nervo Isquiático/efeitos dos fármacos , Nervo Isquiático/lesões , Receptor de Nociceptina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA