Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 13.575
Filtrar
1.
Medicine (Baltimore) ; 103(29): e38837, 2024 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-39029082

RESUMO

Opioids exert analgesic effects by agonizing opioid receptors and activating signaling pathways coupled to receptors such as G-protein and/or ß-arrestin. Concomitant respiratory depression (RD) is a common clinical problem, and improvement of RD is usually achieved with specific antagonists such as naloxone; however, naloxone antagonizes opioid analgesia and may produce more unknown adverse effects. In recent years, researchers have used various methods to isolate opioid receptor-mediated analgesia and RD, with the aim of preserving opioid analgesia while attenuating RD. At present, the focus is mainly on the development of new opioids with weak respiratory inhibition or the use of non-opioid drugs to stimulate breathing. This review reports recent advances in novel opioid agents, such as mixed opioid receptor agonists, peripheral selective opioid receptor agonists, opioid receptor splice variant agonists, biased opioid receptor agonists, and allosteric modulators of opioid receptors, as well as in non-opioid agents, such as AMPA receptor modulators, 5-hydroxytryptamine receptor agonists, phosphodiesterase-4 inhibitors, and nicotinic acetylcholine receptor agonists.


Assuntos
Analgésicos Opioides , Insuficiência Respiratória , Humanos , Insuficiência Respiratória/induzido quimicamente , Insuficiência Respiratória/tratamento farmacológico , Analgésicos Opioides/efeitos adversos , Analgésicos Opioides/uso terapêutico , Receptores Opioides/efeitos dos fármacos , Receptores Opioides/metabolismo , Receptores Opioides/agonistas , Antagonistas de Entorpecentes/uso terapêutico , Antagonistas de Entorpecentes/farmacologia
2.
Chem Commun (Camb) ; 60(63): 8224-8227, 2024 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-39007214

RESUMO

The current opioid epidemic has incentivized the discovery of new non-addictive analgesics, a process that requires the screening of opioid receptor binding, traditionally performed using radiometric assays. Here we describe a label-free alternative based on high-throughput (1 Hz) ambient mass spectrometry for screening the receptor binding of new opioid analogues.


Assuntos
Ensaios de Triagem em Larga Escala , Receptores Opioides , Espectrometria de Massas por Ionização por Electrospray , Receptores Opioides/metabolismo , Receptores Opioides/química , Analgésicos Opioides/química , Analgésicos Opioides/metabolismo , Humanos , Ligação Proteica , Automação
3.
Adv Neurobiol ; 35: 9-26, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38874716

RESUMO

The function of endogenous opioids spans from initiating behaviors that are critical for survival, to responding to rapidly changing environmental conditions. A network of interconnected systems throughout the body characterizes the endogenous opioid system (EOS). EOS receptors for beta-endorphin, enkephalin, dynorphin, and endomorphin underpin the diverse functions of the EOS across biological systems. This chapter presents a succinct yet comprehensive summary of the structure of the EOS, EOS receptors, and their relationship to other biological systems.


Assuntos
Analgésicos Opioides , Receptores Opioides , Animais , Humanos , Analgésicos Opioides/metabolismo , beta-Endorfina/metabolismo , Dinorfinas/metabolismo , Encefalinas/metabolismo , Peptídeos Opioides/metabolismo , Receptores Opioides/metabolismo
4.
Adv Neurobiol ; 35: 27-43, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38874717

RESUMO

The endogenous opioid system, which consists of opioid receptors and their ligands, is widely expressed in the nervous system and also found in the immune system. As a part of the body's defense machinery, the immune system is heavily regulated by endogenous opioid peptides. Many types of immune cells, including macrophages, dendritic cells, neutrophils, and lymphocytes are influenced by endogenous opioids, which affect cell activation, differentiation, proliferation, apoptosis, phagocytosis, and cytokine production. Additionally, immune cells also synthesize and secrete endogenous opioid peptides and participate peripheral analgesia. This chapter is structured into two sections. Part one focuses on immunoregulatory functions of central endogenous opioids; and part two describes how opioid peptide-containing immune cells participate in local analgesia.


Assuntos
Sistema Imunitário , Peptídeos Opioides , Receptores Opioides , Animais , Humanos , Sistema Imunitário/metabolismo , Sistema Imunitário/imunologia , Peptídeos Opioides/metabolismo , Receptores Opioides/metabolismo , Receptores Opioides/imunologia
5.
Adv Neurobiol ; 35: 87-106, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38874719

RESUMO

Understanding the relationship between stress and breast cancer development is essential to preventing and alleviating the cancer. Recent research has shed light on the cognitive, physiological, cellular, and molecular underpinnings of how the endorphin pathway and stress pathway affect breast cancer. This chapter consists of two parts. Part 1 will discuss the role of endorphins in breast cancer development. This includes a discussion of three topics: (1) the neurophysiological effect of endorphins on breast tumor growth in vivo, along with further experiments that will deepen our knowledge of how ß-endorphin affects breast cancer; (2) how both the opioid receptor and somatostatin receptor classes alter intracellular signaling in breast cancer cells; and (3) genetic alleles in the opioid signaling pathway that are correlated with increased breast cancer risk. Part 2 will discuss the role of endorphins in recovery from breast cancer. This includes a discussion of three topics: (1) the relationship between breast cancer diagnosis and depression; (2) the effectiveness of cognitive behavioral therapy in reducing stress in breast cancer patients; and (3) the effect of psychotherapy and exercise on preserving telomere length in breast cancer patients.


Assuntos
Neoplasias da Mama , Estresse Psicológico , Animais , Feminino , Humanos , beta-Endorfina/metabolismo , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Terapia Cognitivo-Comportamental , Depressão/metabolismo , Endorfinas/metabolismo , Receptores Opioides/metabolismo , Transdução de Sinais , Estresse Psicológico/metabolismo
6.
Adv Neurobiol ; 35: 45-85, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38874718

RESUMO

It has become apparent that endogenous opioids act not only as neurotransmitters and neuromodulators, but have multiple functions in the body. Activation of the opioid system by opiate drugs is associated with a risk of cancer development through direct stimulation of tumor cell proliferation and through immunosuppression. In contrast, the endogenous peptide opioid [Met5]-enkephalin, now commonly referred to as Opioid Growth Factor (OGF), negatively regulates cell proliferation in a wide number of cells during development, homeostasis, and neoplasia. This action is mediated through the opioid growth factor receptor, originally designated the zeta (ζ) opioid receptor. Further, contrary to the traditional notion of opiates as immunosuppressive, endogenous OGF has been shown to possess a number of positive immunomodulatory properties and may provide a beneficial effect in cancer by augmenting the activity of cells involved in both innate and acquired immunity. Taken together, the evidence supports consideration of opioid peptides such as OGF as new strategies for cancer therapy.


Assuntos
Neoplasias , Receptores Opioides , Animais , Humanos , Proliferação de Células/efeitos dos fármacos , Encefalina Metionina/metabolismo , Neoplasias/imunologia , Neoplasias/metabolismo , Peptídeos Opioides/metabolismo , Receptores Opioides/metabolismo
7.
Adv Neurobiol ; 35: 241-250, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38874726

RESUMO

Pleasant emotions take a variety of forms and are a key part of the human experience. Although negative emotions have often been a focus of research, positive emotions, e.g., joy, pleasure, and love, have recently gained more attention. Each of these emotions is rich and complex in its own right. However, positive emotions appear to serve key evolutionary functions, which are mediated by complex biological substrates. This chapter summarizes key research and explores the biological underpinnings of positive emotions, with an emphasis on the roles that endogenous opioids play in the experience, expression, and development of positive emotions. The necessity of emphasizing positive emotions in research is also discussed.


Assuntos
Emoções , Peptídeos Opioides , Animais , Humanos , Encéfalo/metabolismo , Emoções/fisiologia , Peptídeos Opioides/metabolismo , Prazer/fisiologia , Receptores Opioides/metabolismo
8.
Adv Neurobiol ; 35: 381-395, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38874733

RESUMO

The opioid system involves opioid receptors (OPRs) and endogenous opioid peptides.This chapter will focus on the distribution of OPRs in the cardiovascular system, the expression pattern in the heart, the activation by opioid peptides, and the effects of OPRs activation with potential relevance in cardiovascular performance. In the heart, OPRs are co-expressed with beta adrenergic receptors (ß-ARs) in the G-protein-coupled receptor (GPCR) superfamily, functionally cross-talk with ß-Ars and modify catecholamine-induced effects. They are involved in cardiac contractility, energy metabolism, myocyte survival or death, vascular resistance. The effects of the opioid system in the regulation of systemic circulation at both the central and peripheral level are presented. The pathways are discussed under physiological (i.e., aging) and pathological conditions (atherosclerosis, heart failure, essential hypertension, ischemic stress). Stimulation of OPRs not only inhibits cardiac excitation-contraction coupling, but also protects the heart against hypoxic and ischemic injury. An enhanced sensitivity to opioids of endocrine organs and neuronal systems is operative in hypertensive patients. The opioid system can be pharmacologically engaged to selectively mimic these responses via cardiac and nervous signaling. The clinical opportunities for the use of cardioprotective effects of opioids require future investigations to provide more specific details of the impact on cardiac performance and electrophysiological properties.


Assuntos
Receptores Opioides , Animais , Humanos , Analgésicos Opioides/metabolismo , Cardiotônicos/farmacologia , Coração/efeitos dos fármacos , Peptídeos Opioides/metabolismo , Receptores Opioides/metabolismo
9.
Nat Commun ; 15(1): 5353, 2024 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-38918403

RESUMO

Nociceptin/orphanin-FQ (N/OFQ) is a recently appreciated critical opioid peptide with key regulatory functions in several central behavioral processes including motivation, stress, feeding, and sleep. The functional relevance of N/OFQ action in the mammalian brain remains unclear due to a lack of high-resolution approaches to detect this neuropeptide with appropriate spatial and temporal resolution. Here we develop and characterize NOPLight, a genetically encoded sensor that sensitively reports changes in endogenous N/OFQ release. We characterized the affinity, pharmacological profile, spectral properties, kinetics, ligand selectivity, and potential interaction with intracellular signal transducers of NOPLight in vitro. Its functionality was established in acute brain slices by exogeneous N/OFQ application and chemogenetic induction of endogenous N/OFQ release from PNOC neurons. In vivo studies with fibre photometry enabled direct recording of NOPLight binding to exogenous N/OFQ receptor ligands, as well as detection of endogenous N/OFQ release within the paranigral ventral tegmental area (pnVTA) during natural behaviors and chemogenetic activation of PNOC neurons. In summary, we show here that NOPLight can be used to detect N/OFQ opioid peptide signal dynamics in tissue and freely behaving animals.


Assuntos
Neurônios , Nociceptina , Peptídeos Opioides , Receptores Opioides , Animais , Peptídeos Opioides/metabolismo , Receptores Opioides/metabolismo , Receptores Opioides/genética , Neurônios/metabolismo , Humanos , Camundongos , Masculino , Área Tegmentar Ventral/metabolismo , Receptor de Nociceptina , Células HEK293 , Encéfalo/metabolismo , Camundongos Endogâmicos C57BL , Ligantes , Técnicas Biossensoriais/métodos
10.
Cell Rep ; 43(6): 114343, 2024 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-38865247

RESUMO

Activation of prepronociceptin (PNOC)-expressing neurons in the arcuate nucleus (ARC) promotes high-fat-diet (HFD)-induced hyperphagia. In turn, PNOCARC neurons can inhibit the anorexic response of proopiomelanocortin (POMC) neurons. Here, we validate the necessity of PNOCARC activity for HFD-induced inhibition of POMC neurons in mice and find that PNOCARC-neuron-dependent inhibition of POMC neurons is mediated by gamma-aminobutyric acid (GABA) release. When monitoring individual PNOCARC neuron activity via Ca2+ imaging, we find a subpopulation of PNOCARC neurons that is inhibited upon gastrointestinal calorie sensing and disinhibited upon HFD feeding. Combining retrograde rabies tracing and circuit mapping, we find that PNOC neurons from the bed nucleus of the stria terminalis (PNOCBNST) provide inhibitory input to PNOCARC neurons, and this inhibitory input is blunted upon HFD feeding. This work sheds light on how an increase in caloric content of the diet can rewire a neuronal circuit, paving the way to overconsumption and obesity development.


Assuntos
Dieta Hiperlipídica , Hiperfagia , Núcleos Septais , Animais , Hiperfagia/metabolismo , Camundongos , Núcleos Septais/metabolismo , Neurônios/metabolismo , Masculino , Ácido gama-Aminobutírico/metabolismo , Pró-Opiomelanocortina/metabolismo , Neurônios GABAérgicos/metabolismo , Núcleo Arqueado do Hipotálamo/metabolismo , Camundongos Endogâmicos C57BL , Precursores de Proteínas , Receptores Opioides
11.
Peptides ; 179: 171268, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38943841

RESUMO

This paper is the forty-sixth consecutive installment of the annual anthological review of research concerning the endogenous opioid system, summarizing articles published during 2023 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 and alcohol abuse (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
Peptídeos Opioides , Receptores Opioides , Humanos , Peptídeos Opioides/metabolismo , Animais , Receptores Opioides/metabolismo , Dor/tratamento farmacológico , Dor/metabolismo , Analgésicos Opioides/farmacologia , Comportamento/efeitos dos fármacos
12.
Eur J Pharmacol ; 975: 176648, 2024 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-38759706

RESUMO

Opioids are used for pain relief in patients suffering from acute myocardial ischemia or infarction. Clinical and laboratory studies demonstrate that morphine treated patients or the experimental animal model suffering acute myocardial ischemia and reperfusion, may worsen myocardial viability. As transient receptor potential vanilloid 1 (TRPV1) plays important roles in pain sensation and cardio-protection, we query whether opioids may exacerbate myocardial viability via interaction with TRPV1 activity in the pain relief. We found the co-expressions of TRPV1 and opioid µ, δ and κ receptors in adult rat cardiomyocytes. Intravenous injection of morphine (0.3 mg/kg) at 20 min after induction of myocardial ischemia, in the rat model of acute myocardial ischemia and reperfusion, induced significant reduction of phosphorylated TRPV1 (p-TRPV1) in the ventricular myocardium and increase in serum cardiac troponin I (cTnI), compared with the ischemia/reperfusion controls (all P < 0.05). The effects of morphine were completely reversed by selective opioid µ, δ and κ receptor antagonists. While significant upregulation of p-TRPV1 (P < 0.05) and improvement of ±dP/dt max (all P < 0.05) were detected in the animals giving the same dose of morphine before induction of myocardial ischemia. The changes in p-TRPV1 correlate with the alterations of cTnI (r = -0.5840, P = 0.0283) and ±dP/dt max (r = 0.8084, P = 0.0005 and r = -0.8133, P = 0.0004, respectively). The findings of this study may indicate that potentiation and attenuation of TRPV1 sensitivity correlate with the improvement of the cardiac performance and the aggravation of myocardial viability, respectively, by giving morphine before and during myocardial ischemia and reperfusion.


Assuntos
Morfina , Traumatismo por Reperfusão Miocárdica , Ratos Sprague-Dawley , Canais de Cátion TRPV , Animais , Canais de Cátion TRPV/metabolismo , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Morfina/farmacologia , Fosforilação/efeitos dos fármacos , Masculino , Ratos , Fatores de Tempo , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Analgésicos Opioides/farmacologia , Receptores Opioides/metabolismo , Troponina I/metabolismo , Troponina I/sangue , Miocárdio/metabolismo , Miocárdio/patologia
13.
Behav Brain Res ; 469: 115065, 2024 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-38782097

RESUMO

Acetaminophen (paracetamol) is one of the most popular analgesics for the management of fever and pain but few reports have investigated its antidepressant-like effect. Moreover, the role of the opioidergic pathway has been indicated in depression pathophysiology. This study aimed to examine the involvement of the opioid receptors in the antidepressant-like effect of acetaminophen after acute and sub-chronic administration using mice forced swimming test (FST). Our finding showed that administration of acetaminophen (50 and 100 mg/kg, i.p.) 30 min before the FST produced an antidepressant effect which was reduced by naloxone (1 mg/kg, i.p., a nonselective opioid receptor antagonist). Moreover, we observed that acetaminophen in higher doses (200 and 400 mg/kg) was ineffective. Also, the response of the non-effective dose of acetaminophen (25 mg/kg) was potentiated by the non-effective dose of morphine (0.1 mg/kg) in the FST that was antagonized by naloxone. Also, in contrast to morphine (10 mg/kg), acetaminophen (100 mg/kg, i.p.) induced neither tolerance to the anti-immobility behavior nor withdrawal syndrome after repeated administration. In addition, RT-PCR showed that hippocampal mu- and kappa-opioid receptor mRNA expression increased in mice after repeated administration of acetaminophen; however, morphine therapy for 6 days did not affect kappa-opioid receptor expression. Our findings demonstrated that acetaminophen in lower doses but not high doses revealed an antidepressant-like activity without inducing tolerance and withdrawal syndromes. Moreover, the observed effect of acetaminophen may be via altering the opioid system, particularly hippocampal mu- and kappa-receptors.


Assuntos
Acetaminofen , Antidepressivos , Relação Dose-Resposta a Droga , Naloxona , Antagonistas de Entorpecentes , Animais , Acetaminofen/farmacologia , Acetaminofen/administração & dosagem , Masculino , Camundongos , Antidepressivos/farmacologia , Antidepressivos/administração & dosagem , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Antagonistas de Entorpecentes/administração & dosagem , Natação , Depressão/tratamento farmacológico , Depressão/metabolismo , Morfina/farmacologia , Morfina/administração & dosagem , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Modelos Animais de Doenças , Analgésicos Opioides/farmacologia , Analgésicos Opioides/administração & dosagem , Analgésicos não Narcóticos/farmacologia , Analgésicos não Narcóticos/administração & dosagem , Receptores Opioides/metabolismo , Receptores Opioides/efeitos dos fármacos , Receptores Opioides mu/metabolismo , Receptores Opioides mu/efeitos dos fármacos
14.
Chem Commun (Camb) ; 60(47): 6007-6010, 2024 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-38787679

RESUMO

The enantioselective de novo synthesis of pharmacologically important 14-hydroxy-6-oxomorphinans is described. 4,5-Desoxynaltrexone and 4,5-desoxynaloxone were prepared using this route and their biological activities against the opioid receptors were measured.


Assuntos
Morfinanos , Estereoisomerismo , Morfinanos/química , Morfinanos/síntese química , Naltrexona/análogos & derivados , Naltrexona/química , Naltrexona/síntese química , Estrutura Molecular , Antagonistas de Entorpecentes/síntese química , Receptores Opioides/metabolismo
15.
Int J Mol Sci ; 25(7)2024 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-38612817

RESUMO

Diverse chemical and pharmacological strategies are currently being explored to minimize the unwanted side effects of currently used opioid analgesics while achieving effective pain relief. The use of multitarget ligands with activity at more than one receptor represents a promising therapeutic approach. We recently reported a bifunctional peptide-based hybrid LENART01 combining dermorphin and ranatensin pharmacophores, which displays activity to the mu-opioid receptor (MOR) and dopamine D2 receptor (D2R) in rat brains and spinal cords. In this study, we investigated the in vitro binding and functional activities to the human MOR and the in vivo pharmacology of LENART01 in mice after subcutaneous administration. In vitro binding assays showed LENART01 to bind and be selective to the human MOR over the other opioid receptor subtypes and delta, kappa and nociceptin receptors. In the [35S]GTPγS binding assay, LENART01 acted as a potent and full agonist to the human MOR. In mice, LENART01 produced dose-dependent antinociceptive effects in formalin-induced inflammatory pain, with increased potency than morphine. Antinociceptive effects were reversed by naloxone, indicating MOR activation in vivo. Behavioral studies also demonstrated LENART01's properties to induce less adverse effects without locomotor dysfunction and withdrawal syndrome compared to conventional opioid analgesics, such as morphine. LENART01 is the first peptide-based MOR-D2R ligand known to date and the first dual MOR-dopamine D2R ligand for which in vivo pharmacology is reported with antinociceptive efficacy and reduced opioid-related side effects. Our current findings may pave the way to new pain therapeutics with limited side effects in acute and chronic use.


Assuntos
Analgésicos Opioides , Oligopeptídeos , Ácido Pirrolidonocarboxílico/análogos & derivados , Receptores Opioides , Humanos , Ratos , Animais , Camundongos , Analgésicos Opioides/farmacologia , Ligantes , Morfina , Peptídeos Opioides/farmacologia , Dor/tratamento farmacológico
16.
Food Funct ; 15(9): 4773-4784, 2024 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-38469873

RESUMO

Raphanus sativus L. cv. Sango, commonly known as red radish, is widely consumed around the world as a vegetable, but its benefit in pain relief is not sufficiently investigated. This study aimed to evaluate the antinociceptive effects of R. sativus and a possible mechanism of action. An aqueous extract of R. sativus sprouts (AERSS) was investigated by parenteral (10, 30, and 100 mg kg-1, i.p.) and enteral (500 mg kg-1, p.o.) administration in the neurogenic and inflammatory phases of the formalin test, where gastric damage was also evaluated as a possible adverse effect. Ketorolac (5 mg kg-1, i.p.) was used as the reference drug. Endogenous opioid and 5-HT1A serotonin receptors, as well as the cAMP/NO-cGMP pathways, were explored in the study of a possible mechanism of action by using their corresponding antagonists: naloxone, 1 mg kg-1, i.p., WAY100635, 1 mg kg-1, i.p., and enzymatic activators or inhibitors, respectively. Sulforaphane (SFN), a known bioactive metabolite, was analyzed using electroencephalography (EEG) to evidence its central involvement. A significant and dose-dependent antinociceptive activity was observed with the AERSS resembling the antinociceptive effect of the reference drug, with an equivalent significant response with a dose of 500 mg kg-1, p.o. without causing gastric damage. The participation of the endogenous opioid and 5-HT1A serotonin receptors at central and peripheral levels was also observed, with a differential participation of cAMP/NO-cGMP. SFN as one metabolite produced significant changes in the EEG analysis, reinforcing its effects on the CNS. Our preclinical evidence supports the benefits of consuming Raphanus sativus cv. Sango sprouts for pain relief.


Assuntos
Analgésicos , Isotiocianatos , Extratos Vegetais , Raphanus , Transdução de Sinais , Animais , Humanos , Masculino , Camundongos , Analgésicos/farmacologia , AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Isotiocianatos/farmacologia , Dor/tratamento farmacológico , Extratos Vegetais/farmacologia , Raphanus/química , Receptor 5-HT1A de Serotonina/metabolismo , Receptores Opioides/metabolismo , Transdução de Sinais/efeitos dos fármacos , Sulfóxidos/farmacologia
17.
J Neural Transm (Vienna) ; 131(5): 491-494, 2024 05.
Artigo em Inglês | MEDLINE | ID: mdl-38436758

RESUMO

Synthetic and semi-synthetic opioids are prescribed for the management of severe pain conditions, but their long-term use is often leading to physical dependence and addiction disorders. Understanding the complex neurobiology of the opioid system in preclinical models will be essential for the development of safe and efficacious analgesics. With rising numbers of synthetic opioid users and overdose cases, a better understanding of the neuroanatomical and cellular pathways associated with physical dependence and addiction is expected to guide treatment approaches for opioid use disorders. In this commentary, we highlight the importance of advanced genetic mouse models for studying the regional effects of opioid receptors, and we discuss the need of genetic mouse models for the investigation of the regional, circuit and cell compartment-specific role of intracellular mediators of opioid actions.


Assuntos
Analgésicos Opioides , Modelos Animais de Doenças , Transtornos Relacionados ao Uso de Opioides , Receptores Opioides , Animais , Camundongos , Analgésicos Opioides/farmacologia , Transtornos Relacionados ao Uso de Opioides/genética , Receptores Opioides/genética , Receptores Opioides/metabolismo , Humanos
18.
Bull Exp Biol Med ; 176(4): 433-436, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38488963

RESUMO

Hypoxia (20 min) and reoxygenation (30 min) were simulated on isolated rat cardiomyocytes to evaluate the cytoprotective effect of selective δ2-opioid receptor agonist deltorphin II, opioid receptor antagonist naloxone methiodide, µ-opioid receptor antagonist CTAP, κ-opioid receptor antagonist nor-binaltorphimine, ε1-opioid receptor antagonist BNTX, and δ2-opioid receptors naltriben. Deltorphin II was administered 5 min before reoxygenation, antagonists were administered 10 min before reoxygenation. The cytoprotective effect of deltorphin II was assessed by the number of cardiomyocytes survived after hypoxia/reoxygenation, as well as by the lactate dehydrogenase content in the incubation medium. It has been established that the cytoprotective effect of deltorphin II occurs at a concentration of 64 nmol/liter and is associated with activation of δ2-opioid receptors.


Assuntos
Antagonistas de Entorpecentes , Receptores Opioides , Ratos , Animais , Antagonistas de Entorpecentes/farmacologia , Receptores Opioides delta/genética , Miócitos Cardíacos , Receptores Opioides mu , Hipóxia
19.
Clin Pharmacol Drug Dev ; 13(7): 790-800, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38476082

RESUMO

Sunobinop is an investigational, potent, selective partial agonist at the nociceptin/orphanin FQ peptide receptor in vitro. Three phase 1 studies were conducted to evaluate the safety, tolerability, and pharmacokinetics (PK) of escalating single- and multiple-dose administration of sunobinop in healthy participants. Study 1 was a randomized, double-blind, placebo-controlled, single-ascending dose study. Study 2 was a randomized, double-blind, placebo-controlled, multiple-ascending dose study. Study 3 was a randomized, open-label, single-dose, 4-way crossover study of oral and sublingual sunobinop comparing morning (AM) and bedtime (PM) administration. Seventy participants were included. Systemic exposure (peak plasma concentration [Cmax], area under the plasma concentration-time curve from time 0 to the time of last quantifiable concentration [AUC0-t], and area under the plasma concentration-time curve from time 0 extrapolated to infinity [AUCinf]) of sunobinop was characterized by dose proportionality from 0.6 to 2 mg and increased less than proportionally from 3 to 30 mg. The PKs of sunobinop were similar, regardless of AM or PM administration, for both the oral and sublingual formulations. The majority of absorbed sunobinop was excreted unchanged in the urine within 8 hours of dosing, thereby showing rapid elimination with no appreciable accumulation following 14 consecutive days of once-daily dosing and suggesting exclusive renal elimination. Most treatment-emergent adverse events (TEAEs) were mild in severity; 1 severe TEAE occurred and all TEAEs resolved by the end of the studies. Sunobinop was generally well-tolerated and safe across the range of doses evaluated and presents a clinical profile suitable for continued development.


Assuntos
Área Sob a Curva , Estudos Cross-Over , Voluntários Saudáveis , Humanos , Masculino , Adulto , Método Duplo-Cego , Feminino , Pessoa de Meia-Idade , Adulto Jovem , Administração Oral , Relação Dose-Resposta a Droga , Administração Sublingual , Esquema de Medicação , Receptor de Nociceptina , Receptores Opioides/metabolismo , Adolescente , Morfinanos/farmacocinética , Morfinanos/administração & dosagem , Morfinanos/efeitos adversos , Naltrexona/análogos & derivados
20.
Physiol Rep ; 12(5): e15965, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38444051

RESUMO

Intravenous (systemic) bolus injection of fentanyl (FNT) reportedly induces an immediate vagal-mediated apnea; however, the precise origin of vagal afferents responsible for this apnea remains unknown. We tested whether intralaryngeal (local) application of FNT would also trigger an apnea and whether the apneic response to both local and systemic administration of FNT was laryngeal afferent-mediated. Cardiorespiratory responses to FNT were recorded in anesthetized male adult rats with and without bilateral sectioning of the superior laryngeal nerve (SLNx) or peri-SLN capsaicin treatment (SLNcap) to block local C-fiber signal conduction. Opioid mu-receptor (MOR)-immunoreactivity was detected in laryngeal C- and myelinated neurons. We found that local and systemic administration of FNT elicited an immediate apnea. SLNx, rather than SLNcap, abolished the apneic response to local FNT application though MORs were abundantly expressed in both laryngeal C- and myelinated neurons. Importantly, SLNx failed to affect the apneic response to systemic FNT administration. These results lead to the conclusion that laryngeal afferents' MORs are responsible for the apneic response to local, but not systemic, administration of FNT.


Assuntos
Líquidos Corporais , Fentanila , Masculino , Animais , Ratos , Fentanila/farmacologia , Apneia/induzido quimicamente , Administração Cutânea , Administração Intravenosa , Receptores Opioides
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...