Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
Drug Alcohol Depend ; 231: 109255, 2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-34998256

RESUMO

RATIONALE AND OBJECTIVE: One objective of the National Institutes of Health Helping to End Addiction Long-term (HEAL) initiative is to accelerate research on safer and more effective medications for both pain and opioid use disorder. Ligands that activate the nociceptin opioid peptide receptor (NOP) constitute one class of candidate drugs for both applications. The present preclinical study determined the effectiveness of the NOP agonist Ro 64-6198 to produce antinociception in a pain-depressed behavior procedure and attenuate opioid self-administration in a heroin-vs-food choice procedure. METHODS: In Experiment 1, Adult Sprague-Dawley rats were equipped with microelectrodes and trained to respond for electrical brain stimulation in an intracranial self-stimulation (ICSS) procedure. The potency, time course, and receptor mechanism of effects produced by R0 64-6198 alone (0.32-3.2 mg/kg) on ICSS were examined, followed by evaluation of 0.32-1.0 mg/kg Ro 64-6198 effectiveness to block lactic acid-induced depression of ICSS. In Experiment 2, rats self-administered heroin under a heroin-vs-food choice procedure during a regimen of repeated, daily intraperitoneal administration of vehicle or Ro 64-6198 (1-3.2 mg/kg/day). RESULTS: Ro 64-6198 produced dose- and time-dependent ICSS depression that was blocked by the selective NOP antagonist SB612111 but not by naltrexone. Ro 64-6198 failed to block acid-induced depression of ICSS. Repeated Ro 64-6198 pretreatment also failed to attenuate heroin-vs-food choice up to doses that significantly decreased operant behavior. CONCLUSIONS: These results do not support the utility of Ro 64-6198 as a stand-alone medication for either acute pain or opioid use disorder.


Assuntos
Dor Aguda , Heroína , Imidazóis/farmacologia , Peptídeos Opioides/agonistas , Transtornos Relacionados ao Uso de Opioides/tratamento farmacológico , Compostos de Espiro/farmacologia , Dor Aguda/tratamento farmacológico , Animais , Relação Dose-Resposta a Droga , Peptídeos Opioides/farmacologia , Ratos , Ratos Sprague-Dawley , Nociceptina
2.
Int J Mol Sci ; 22(23)2021 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-34884757

RESUMO

Nociceptin/orphanin FQ (N/OFQ) is a 17-residue neuropeptide that binds the nociceptin opioid-like receptor (NOP). N/OFQ exhibits nucleotidic and aminoacidics sequence homology with the precursors of other opioid neuropeptides but it does not activate either MOP, KOP or DOP receptors. Furthermore, opioid neuropeptides do not activate the NOP receptor. Generally, activation of N/OFQ system exerts anti-opioids effects, for instance toward opioid-induced reward and analgesia. The NOP receptor is widely expressed throughout the brain, whereas N/OFQ localization is confined to brain nuclei that are involved in stress response such as amygdala, BNST and hypothalamus. Decades of studies have delineated the biological role of this system demonstrating its involvement in significant physiological processes such as pain, learning and memory, anxiety, depression, feeding, drug and alcohol dependence. This review discusses the role of this peptidergic system in the modulation of stress and stress-associated psychiatric disorders in particular drug addiction, mood, anxiety and food-related associated-disorders. Emerging preclinical evidence suggests that both NOP agonists and antagonists may represent a effective therapeutic approaches for substances use disorder. Moreover, the current literature suggests that NOP antagonists can be useful to treat depression and feeding-related diseases, such as obesity and binge eating behavior, whereas the activation of NOP receptor by agonists could be a promising tool for anxiety.


Assuntos
Peptídeos Opioides/fisiologia , Receptores Opioides/fisiologia , Estresse Fisiológico/fisiologia , Animais , Transtornos de Ansiedade/tratamento farmacológico , Transtornos de Ansiedade/fisiopatologia , Encéfalo/efeitos dos fármacos , Encéfalo/fisiopatologia , Transtornos da Alimentação e da Ingestão de Alimentos/tratamento farmacológico , Transtornos da Alimentação e da Ingestão de Alimentos/fisiopatologia , Humanos , Modelos Neurológicos , Transtornos do Humor/tratamento farmacológico , Transtornos do Humor/fisiopatologia , Peptídeos Opioides/agonistas , Peptídeos Opioides/antagonistas & inibidores , Recompensa , Estresse Fisiológico/efeitos dos fármacos , Transtornos Relacionados ao Uso de Substâncias/tratamento farmacológico , Transtornos Relacionados ao Uso de Substâncias/fisiopatologia , Receptor de Nociceptina , Nociceptina
3.
Peptides ; 132: 170348, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32574695

RESUMO

This paper is the forty-first consecutive installment of the annual anthological review of research concerning the endogenous opioid system, summarizing articles published during 2018 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 (2), the roles of these opioid peptides and receptors in pain and analgesia in animals (3) and humans (4), opioid-sensitive and opioid-insensitive effects of nonopioid analgesics (5), opioid peptide and receptor involvement in tolerance and dependence (6), stress and social status (7), learning and memory (8), eating and drinking (9), drug abuse and alcohol (10), sexual activity and hormones, pregnancy, development and endocrinology (11), mental illness and mood (12), seizures and neurologic disorders (13), electrical-related activity and neurophysiology (14), general activity and locomotion (15), gastrointestinal, renal and hepatic functions (16), cardiovascular responses (17), respiration and thermoregulation (18), and immunological responses (19).


Assuntos
Analgésicos Opioides/farmacologia , Comportamento/efeitos dos fármacos , Aprendizagem/fisiologia , Transtornos Mentais/tratamento farmacológico , Peptídeos Opioides/metabolismo , Dor/tratamento farmacológico , Receptores Opioides/agonistas , Animais , Humanos , Transtornos Mentais/metabolismo , Antagonistas de Entorpecentes/farmacologia , Peptídeos Opioides/agonistas , Peptídeos Opioides/antagonistas & inibidores , Peptídeos Opioides/farmacologia , Dor/metabolismo
4.
Proc Natl Acad Sci U S A ; 117(21): 11820-11828, 2020 05 26.
Artigo em Inglês | MEDLINE | ID: mdl-32393639

RESUMO

Opioids, such as morphine and fentanyl, are widely used for the treatment of severe pain; however, prolonged treatment with these drugs leads to the development of tolerance and can lead to opioid use disorder. The "Opioid Epidemic" has generated a drive for a deeper understanding of the fundamental signaling mechanisms of opioid receptors. It is generally thought that the three types of opioid receptors (µ, δ, κ) are activated by endogenous peptides derived from three different precursors: Proopiomelanocortin, proenkephalin, and prodynorphin. Posttranslational processing of these precursors generates >20 peptides with opioid receptor activity, leading to a long-standing question of the significance of this repertoire of peptides. Here, we address some aspects of this question using a technical tour de force approach to systematically evaluate ligand binding and signaling properties ([35S]GTPγS binding and ß-arrestin recruitment) of 22 peptides at each of the three opioid receptors. We show that nearly all tested peptides are able to activate the three opioid receptors, and many of them exhibit agonist-directed receptor signaling (functional selectivity). Our data also challenge the dogma that shorter forms of ß-endorphin do not exhibit receptor activity; we show that they exhibit robust signaling in cultured cells and in an acute brain slice preparation. Collectively, this information lays the groundwork for improved understanding of the endogenous opioid system that will help in developing more effective treatments for pain and addiction.


Assuntos
Peptídeos Opioides , Receptores Opioides/metabolismo , Transdução de Sinais/fisiologia , Animais , Linhagem Celular Tumoral , Humanos , Masculino , Peptídeos Opioides/agonistas , Peptídeos Opioides/metabolismo , Pró-Opiomelanocortina/metabolismo , Ligação Proteica , Ratos , Ratos Sprague-Dawley
5.
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
6.
Neuron ; 100(6): 1280-1282, 2018 12 19.
Artigo em Inglês | MEDLINE | ID: mdl-30571940

RESUMO

Nociceptin opioid peptide receptor agonists interact with mu-opioid receptor agonists for pain relief. A new study by Ding et al. (2018) examines a bifunctional nociceptin- and mu-opioid receptor agonist, AT-121, that provides analgesia without physiological side effects or abuse liability, offering a promising new hope toward better analgesics.


Assuntos
Analgésicos Opioides/uso terapêutico , Analgésicos/uso terapêutico , Dor/tratamento farmacológico , Animais , Humanos , Peptídeos Opioides/agonistas , Peptídeos Opioides/metabolismo , Receptores Opioides mu/metabolismo , Nociceptina
7.
Psychopharmacology (Berl) ; 235(11): 3093-3102, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30145654

RESUMO

RATIONALE: Nociceptin/orphanin FQ (N/OFQ) is the endogenous ligand of a Gi protein-coupled receptor named NOP. Both N/OFQ and NOP receptor are widely expressed in brain areas involved in the control of emotional processes. Clinical and preclinical studies support antidepressant effects due to the blockade of NOP receptor signaling. By contrast, NOP receptor activation did not evoke any change in behavioral despair tests. OBJECTIVES: The present study aimed to investigate the effects of the co-administration of NOP agonists and classic antidepressant drugs in the forced swimming test (FST) and learned helplessness model (LH) in mice. METHODS: Male Swiss mice were co-administered with NOP agonists (N/OFQ and Ro 65-6570) and antidepressants (nortriptyline, fluoxetine, and R-ketamine) or SB-612111 (NOP antagonist) and the behavioral effects were assessed in the FST and LH tests. RESULTS: Fluoxetine, nortriptyline, R-ketamine and the NOP antagonist SB-612111 displayed antidepressant-like effects in the FST. The administration of the NOP agonists N/OFQ and Ro 65-6570 did not induce any behavioral change. However, co-administration of NOP agonists blocked the antidepressant effects of SB-612111, fluoxetine, and nortriptyline, but not R-ketamine in the FST. Similarly, in the LH, the systemic injection of SB-612111, nortriptyline, and R-ketamine reversed helplessness. The co-administration of Ro 65-6570 blocked the antidepressant-like effects of SB-612111 and nortriptyline, but not R-ketamine. CONCLUSIONS: NOP receptor activation inhibits the acute antidepressant effects of nortriptyline and fluoxetine, but not R-ketamine. The present findings contribute to further understand the role played by the N/OFQ-NOP receptor system in regulating mood states.


Assuntos
Antidepressivos/farmacologia , Fluoxetina/farmacologia , Desamparo Aprendido , Ketamina/farmacologia , Nortriptilina/farmacologia , Peptídeos Opioides/agonistas , Anestésicos Dissociativos/farmacologia , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/fisiologia , Emoções/efeitos dos fármacos , Emoções/fisiologia , Fluoxetina/antagonistas & inibidores , Imidazóis/farmacologia , Masculino , Camundongos , Nortriptilina/antagonistas & inibidores , Peptídeos Opioides/metabolismo , Receptores Opioides/agonistas , Receptores Opioides/metabolismo , Compostos de Espiro/farmacologia , Natação/fisiologia , Natação/psicologia , Nociceptina
8.
Psychopharmacology (Berl) ; 234(9-10): 1371-1394, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28285326

RESUMO

BACKGROUND: Nicotine addiction continues to be a health challenge across the world. Despite several approved medications, smokers continue to relapse. Several human and animal studies have evaluated the role of the endogenous opioid system as a potential target for smoking cessation medications. METHODS: In this review, studies that have elucidated the role of the mu (MORs), delta (DORs), and kappa (KORs) opioid receptors in nicotine reward, nicotine withdrawal, and reinstatement of nicotine seeking will be discussed. Additionally, the review will discuss discrepancies in the literature and therapeutic potential of the endogenous opioid system, and suggest studies to address gaps in knowledge with respect to the role of the opioid receptors in nicotine dependence. RESULTS: Data available till date suggest that blockade of the MORs and DORs decreased the rewarding effects of nicotine, while activation of the MORs and DORs decreased nicotine withdrawal-induced aversive effects. In contrast, activation of the KORs decreased the rewarding effects of nicotine, while blockade of the KORs decreased nicotine withdrawal-induced aversive effects. Interestingly, blockade of the MORs and KORs attenuated reinstatement of nicotine seeking. In humans, MOR antagonists have shown benefits in select subpopulations of smokers and further investigation is required to realize their full therapeutic potential. CONCLUSION: Future work must assess the influence of polymorphisms in opioid receptor-linked genes in nicotine dependence, which will help in both identifying individuals vulnerable to nicotine addiction and the development of opioid-based smoking cessation medications. Overall, the endogenous opioid system continues to be a promising target for future smoking cessation medications.


Assuntos
Analgésicos Opioides/metabolismo , Sistemas de Liberação de Medicamentos/tendências , Receptores Opioides/metabolismo , Abandono do Hábito de Fumar , Tabagismo/metabolismo , Analgésicos Opioides/administração & dosagem , Animais , Sistemas de Liberação de Medicamentos/métodos , Previsões , Humanos , Nicotina/administração & dosagem , Peptídeos Opioides/agonistas , Peptídeos Opioides/antagonistas & inibidores , Peptídeos Opioides/metabolismo , Receptores Opioides/agonistas , Recompensa , Abandono do Hábito de Fumar/métodos , Síndrome de Abstinência a Substâncias/tratamento farmacológico , Síndrome de Abstinência a Substâncias/metabolismo , Tabagismo/tratamento farmacológico
9.
Pain Pract ; 17(8): 1032-1041, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28112482

RESUMO

Cebranopadol is a novel and highly potent analgesic acting via nociceptin/orphanin FQ peptide (NOP) and opioid receptors. Since NOP and opioid receptors are expressed in the central nervous system as well as in the periphery, this study addressed the question of where cebranopadol exerts its effects in animal models of chronic neuropathic pain. Mechanical hypersensitivity in streptozotocin (STZ)-treated diabetic rats, cold allodynia in the chronic constriction injury (CCI) model in rats, and heat hyperalgesia and nociception in STZ-treated diabetic and control mice was determined after intraplantar (i.pl.), intracerebroventricular (i.c.v.), or intrathecal (i.th.) administration. In STZ-treated rats, cebranopadol (i.pl.) reduced mechanical hypersensitivity in the ipsilateral paw, but had no effect at the contralateral paw. In CCI rats, cebranopadol (i.pl.) showed antiallodynic activity at the ipsilateral paw. After administration to the contralateral paw, cebranopadol also showed ipsilateral antiallodynic activity, but with reduced potency and delayed onset. In diabetic mice, cebranopadol i.th. and i.c.v. decreased heat hyperalgesia with full efficacy and similar potency for both routes. Cebranopadol also produced significant antinociception in nondiabetic controls. Thus, cebranopadol exerts potent and efficacious antihyperalgesic, antiallodynic, and antinociceptive effects after local/peripheral, spinal, and supraspinal administration. The contralateral effects after i.pl. administration were likely due to systemic redistribution. After central administration of cebranopadol, antihyperalgesic efficacy is reached at doses that are not yet antinociceptive. This study shows that cebranopadol is effective after peripheral as well as central administration in nociceptive and chronic neuropathic pain. Thus, it may be well-suited for the treatment of chronic pain conditions with a neuropathic component.


Assuntos
Analgésicos/farmacologia , Indóis/farmacologia , Neuralgia/tratamento farmacológico , Compostos de Espiro/farmacologia , Analgésicos Opioides/uso terapêutico , Animais , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Hiperalgesia/tratamento farmacológico , Injeções Intraventriculares , Injeções Espinhais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Peptídeos Opioides/agonistas , Ratos , Ratos Sprague-Dawley , Receptores Opioides/agonistas , Nociceptina
10.
Proc Natl Acad Sci U S A ; 113(37): E5511-8, 2016 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-27573832

RESUMO

Despite the critical need, no previous research has substantiated safe opioid analgesics without abuse liability in primates. Recent advances in medicinal chemistry have led to the development of ligands with mixed mu opioid peptide (MOP)/nociceptin-orphanin FQ peptide (NOP) receptor agonist activity to achieve this objective. BU08028 is a novel orvinol analog that displays a similar binding profile to buprenorphine with improved affinity and efficacy at NOP receptors. The aim of this preclinical study was to establish the functional profile of BU08028 in monkeys using clinically used MOP receptor agonists for side-by-side comparisons in various well-honed behavioral and physiological assays. Systemic BU08028 (0.001-0.01 mg/kg) produced potent long-lasting (i.e., >24 h) antinociceptive and antiallodynic effects, which were blocked by MOP or NOP receptor antagonists. More importantly, the reinforcing strength of BU08028 was significantly lower than that of cocaine, remifentanil, or buprenorphine in monkeys responding under a progressive-ratio schedule of drug self-administration. Unlike MOP receptor agonists, BU08028 at antinociceptive doses and ∼10- to 30-fold higher doses did not cause respiratory depression or cardiovascular adverse events as measured by telemetry devices. After repeated administration, the monkeys developed acute physical dependence on morphine, as manifested by precipitated withdrawal signs, such as increased respiratory rate, heart rate, and blood pressure. In contrast, monkeys did not show physical dependence on BU08028. These in vivo findings in primates not only document the efficacy and tolerability profile of bifunctional MOP/NOP receptor agonists, but also provide a means of translating such ligands into therapies as safe and potentially abuse-free opioid analgesics.


Assuntos
Analgésicos Opioides/administração & dosagem , Buprenorfina/análogos & derivados , Cocaína/toxicidade , Dor/tratamento farmacológico , Analgésicos Opioides/efeitos adversos , Analgésicos Opioides/agonistas , Analgésicos Opioides/antagonistas & inibidores , Animais , Buprenorfina/administração & dosagem , Buprenorfina/efeitos adversos , Buprenorfina/química , Buprenorfina/metabolismo , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/patologia , Humanos , Ligantes , Peptídeos Opioides/agonistas , Peptídeos Opioides/antagonistas & inibidores , Dor/patologia , Primatas , Receptores Opioides mu/agonistas , Receptores Opioides mu/antagonistas & inibidores , Insuficiência Respiratória/induzido quimicamente , Insuficiência Respiratória/patologia
11.
J Clin Psychiatry ; 77(9): 1182-1188, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27035058

RESUMO

OBJECTIVE: Posttraumatic stress disorder (PTSD), chronic pain, and substance use disorders are prevalent co-occurring conditions that are challenging to treat individually, and there is no evidence-based treatment for all 3. Buprenorphine, used to treat opioid use disorder and chronic pain, is a partial nociceptin opioid receptor agonist. In preclinical studies, a nociceptin opioid receptor agonist was shown to mitigate PTSD symptoms in acute trauma. We compared buprenorphine to other opioid medications in its impact on PTSD symptoms in patients with chronic pain and opioid and/or other substance use disorders. METHOD: We assembled a retrospective cohort of 382 Iraq and Afghanistan veterans in US Department of Veterans Affairs health care from October 1, 2007, to July 29, 2013, with ICD-9-CM diagnoses of PTSD, chronic pain, and substance use disorders. We used time-varying general estimating equation models to assess the primary outcome, which was change in PTSD symptoms (measured using the PTSD Checklist and the Primary Care PTSD Screen) among veterans initiated on sublingual buprenorphine versus those maintained on moderately high-dose opioid therapy. RESULTS: Twice as many veterans in the buprenorphine group (23.7%) compared to those in the opioid therapy group (11.7%) experienced improvement in PTSD symptoms (P = .001). Compared to veterans in the opioid therapy group, veterans receiving buprenorphine showed significant improvement in PTSD symptoms after 8 months, with increasing improvement up to 24 months (incidence rate ratio = 1.79; 95% CI, 1.16-2.77; P = .009). There were no differences in the longitudinal course of pain ratings between groups. CONCLUSIONS: This observational study is the first to report an incidental effect of buprenorphine compared to opioid therapy in improving PTSD symptoms in veterans.


Assuntos
Buprenorfina/farmacologia , Dor Crônica/tratamento farmacológico , Antagonistas de Entorpecentes/farmacologia , Tratamento de Substituição de Opiáceos/métodos , Peptídeos Opioides/agonistas , Transtornos Relacionados ao Uso de Opioides/tratamento farmacológico , Avaliação de Resultados em Cuidados de Saúde , Receptores Opioides/agonistas , Transtornos de Estresse Pós-Traumáticos/tratamento farmacológico , Veteranos , Adulto , Campanha Afegã de 2001- , Buprenorfina/administração & dosagem , Dor Crônica/epidemiologia , Comorbidade , Feminino , Humanos , Guerra do Iraque 2003-2011 , Masculino , Antagonistas de Entorpecentes/administração & dosagem , Tratamento de Substituição de Opiáceos/estatística & dados numéricos , Transtornos Relacionados ao Uso de Opioides/epidemiologia , Estudos Retrospectivos , Transtornos de Estresse Pós-Traumáticos/epidemiologia , Estados Unidos , Nociceptina
12.
Mol Pharmacol ; 88(2): 335-46, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26013541

RESUMO

Biased agonism is having a major impact on modern drug discovery, and describes the ability of distinct G protein-coupled receptor (GPCR) ligands to activate different cell signaling pathways, and to result in different physiologic outcomes. To date, most studies of biased agonism have focused on synthetic molecules targeting various GPCRs; however, many of these receptors have multiple endogenous ligands, suggesting that "natural" bias may be an unappreciated feature of these GPCRs. The µ-opioid receptor (MOP) is activated by numerous endogenous opioid peptides, remains an attractive therapeutic target for the treatment of pain, and exhibits biased agonism in response to synthetic opiates. The aim of this study was to rigorously assess the potential for biased agonism in the actions of endogenous opioids at the MOP in a common cellular background, and compare these to the effects of the agonist d-Ala2-N-MePhe4-Gly-ol enkephalin (DAMGO). We investigated activation of G proteins, inhibition of cAMP production, extracellular signal-regulated kinase 1 and 2 phosphorylation, ß-arrestin 1/2 recruitment, and MOP trafficking, and applied a novel analytical method to quantify biased agonism. Although many endogenous opioids displayed signaling profiles similar to that of DAMGO, α-neoendorphin, Met-enkephalin-Arg-Phe, and the putatively endogenous peptide endomorphin-1 displayed particularly distinct bias profiles. These may represent examples of natural bias if it can be shown that they have different signaling properties and physiologic effects in vivo compared with other endogenous opioids. Understanding how endogenous opioids control physiologic processes through biased agonism can reveal vital information required to enable the design of biased opioids with improved pharmacological profiles and treat diseases involving dysfunction of the endogenous opioid system.


Assuntos
Endorfinas/farmacologia , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Oligopeptídeos/farmacologia , Peptídeos Opioides/agonistas , Precursores de Proteínas/farmacologia , Receptores Opioides mu/metabolismo , Animais , Células CHO , Cricetulus , Proteínas de Ligação ao GTP/metabolismo , Análise de Componente Principal , Receptores Opioides mu/química , Transdução de Sinais/efeitos dos fármacos
13.
Gen Comp Endocrinol ; 210: 81-6, 2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-25307952

RESUMO

Endocannabinoids and their receptors are found throughout the brain of all vertebrates. By virtue of their wide distribution, endocannabinoids have the potential to affect many behaviors. Prior research has shown that cannabinoids inhibit courtship-clasping and mediate behavioral responses to stress in male rough-skinned newts, Taricha granulosa, and cannabinoid signaling is initiated by rapid actions of the steroid corticosterone (CORT) at its specific membrane receptor (mCR). This same mCR also recognizes κ-opioid receptor agonists and antagonists. Prior behavioral studies show that κ-opioid agonists suppress clasping behavior in a dose dependent manner. Combined, these studies suggest that κ-opioid agonists might suppress clasping behavior via the same pathway initiated by CORT: up-regulation of endocannabinoid signaling. We examined whether pretreatment with a CB1 antagonist, AM281, would block κ-opioid-mediated suppression of clasping. We found that the CB1 antagonist did not reverse κ-opioid-induced suppression of clasping, revealing that while endocannabinoids mediate CORT-induced suppression of clasping, endocannabinoids do not mediate the κ-opioid-induced suppression of clasping.


Assuntos
Corticosterona/farmacologia , Sistemas Neurossecretores/efeitos dos fármacos , Peptídeos Opioides/farmacologia , Salamandridae , Comportamento Sexual Animal/efeitos dos fármacos , (trans)-Isômero de 3,4-dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclo-hexil)-benzenoacetamida/farmacologia , Analgésicos Opioides/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Corte , Regulação para Baixo/efeitos dos fármacos , Endocanabinoides/farmacologia , Feminino , Masculino , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Sistemas Neurossecretores/fisiologia , Peptídeos Opioides/agonistas , Peptídeos Opioides/antagonistas & inibidores , Salamandridae/fisiologia , Comportamento Sexual Animal/fisiologia
14.
J Cereb Blood Flow Metab ; 34(10): 1604-12, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25005876

RESUMO

Understanding the cellular processes underpinning the changes in binding observed during positron emission tomography neurotransmitter release studies may aid translation of these methodologies to other neurotransmitter systems. We compared the sensitivities of opioid receptor radioligands, carfentanil, and diprenorphine, to amphetamine-induced endogenous opioid peptide (EOP) release and methadone administration in the rat. We also investigated whether agonist-induced internalization was involved in reductions in observed binding using subcellular fractionation and confocal microscopy. After radioligand administration, significant reductions in [(11)C]carfentanil, but not [(3)H]diprenorphine, uptake were observed after methadone and amphetamine pretreatment. Subcellular fractionation and in vitro radioligand binding studies showed that amphetamine pretreatment only decreased total [(11)C]carfentanil binding. In vitro saturation binding studies conducted in buffers representative of the internalization pathway suggested that µ-receptors are significantly less able to bind the radioligands in endosomal compared with extracellular compartments. Finally, a significant increase in µ-receptor-early endosome co-localization in the hypothalamus was observed after amphetamine and methadone treatment using double-labeling confocal microscopy, with no changes in δ- or κ-receptor co-localization. These data indicate carfentanil may be superior to diprenorphine when imaging EOP release in vivo, and that alterations in the ability to bind internalized receptors may be a predictor of ligand sensitivity to endogenous neurotransmitter release.


Assuntos
Anfetamina/farmacologia , Analgésicos Opioides/farmacologia , Encéfalo/efeitos dos fármacos , Estimulantes do Sistema Nervoso Central/farmacologia , Diprenorfina/metabolismo , Fentanila/análogos & derivados , Metadona/farmacologia , Peptídeos Opioides/metabolismo , Animais , Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo , Fentanila/metabolismo , Imunofluorescência , Masculino , Peptídeos Opioides/agonistas , Peptídeos Opioides/análise , Tomografia por Emissão de Pósitrons/métodos , Ensaio Radioligante , Ratos , Ratos Sprague-Dawley
15.
J Pharmacol Exp Ther ; 349(3): 535-48, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24713140

RESUMO

Cebranopadol (trans-6'-fluoro-4',9'-dihydro-N,N-dimethyl-4-phenyl-spiro[cyclohexane-1,1'(3'H)-pyrano[3,4-b]indol]-4-amine) is a novel analgesic nociceptin/orphanin FQ peptide (NOP) and opioid receptor agonist [Ki (nM)/EC50 (nM)/relative efficacy (%): human NOP receptor 0.9/13.0/89; human mu-opioid peptide (MOP) receptor 0.7/1.2/104; human kappa-opioid peptide receptor 2.6/17/67; human delta-opioid peptide receptor 18/110/105]. Cebranopadol exhibits highly potent and efficacious antinociceptive and antihypersensitive effects in several rat models of acute and chronic pain (tail-flick, rheumatoid arthritis, bone cancer, spinal nerve ligation, diabetic neuropathy) with ED50 values of 0.5-5.6 µg/kg after intravenous and 25.1 µg/kg after oral administration. In comparison with selective MOP receptor agonists, cebranopadol was more potent in models of chronic neuropathic than acute nociceptive pain. Cebranopadol's duration of action is long (up to 7 hours after intravenous 12 µg/kg; >9 hours after oral 55 µg/kg in the rat tail-flick test). The antihypersensitive activity of cebranopadol in the spinal nerve ligation model was partially reversed by pretreatment with the selective NOP receptor antagonist J-113397[1-[(3R,4R)-1-cyclooctylmethyl-3-hydroxymethyl-4-piperidyl]-3-ethyl-1,3-dihydro-2H-benzimidazol-2-one] or the opioid receptor antagonist naloxone, indicating that both NOP and opioid receptor agonism are involved in this activity. Development of analgesic tolerance in the chronic constriction injury model was clearly delayed compared with that from an equianalgesic dose of morphine (complete tolerance on day 26 versus day 11, respectively). Unlike morphine, cebranopadol did not disrupt motor coordination and respiration at doses within and exceeding the analgesic dose range. Cebranopadol, by its combination of agonism at NOP and opioid receptors, affords highly potent and efficacious analgesia in various pain models with a favorable side effect profile.


Assuntos
Analgésicos Opioides/uso terapêutico , Indóis/uso terapêutico , Peptídeos Opioides/agonistas , Dor/tratamento farmacológico , Receptores Opioides/agonistas , Compostos de Espiro/uso terapêutico , Analgésicos Opioides/administração & dosagem , Analgésicos Opioides/efeitos adversos , Analgésicos Opioides/farmacologia , Animais , Artrite Experimental/complicações , Artrite Experimental/tratamento farmacológico , Artrite Experimental/metabolismo , Comportamento Animal/efeitos dos fármacos , Neoplasias Ósseas/complicações , Neoplasias Ósseas/tratamento farmacológico , Neoplasias Ósseas/metabolismo , Células CHO , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Cricetinae , Cricetulus , Feminino , Indóis/administração & dosagem , Indóis/efeitos adversos , Indóis/farmacologia , Masculino , Dor/etiologia , Dor/metabolismo , Polineuropatias/complicações , Polineuropatias/tratamento farmacológico , Polineuropatias/metabolismo , Ligação Proteica , Ensaio Radioligante , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Teste de Desempenho do Rota-Rod , Compostos de Espiro/administração & dosagem , Compostos de Espiro/efeitos adversos , Compostos de Espiro/farmacologia , Nociceptina
16.
Br J Pharmacol ; 168(1): 151-62, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22827708

RESUMO

BACKGROUND AND PURPOSE: Intrathecally (i.t.) administered nociceptin/orphanin FQ (N/OFQ) evokes antinociceptive effects in rodents. Recent studies in monkeys demonstrated that i.t. co-application of N/OFQ and morphine elicits synergistic antinociceptive actions suggesting mixed N/OFQ peptide (NOP) and µ opioid receptor agonists as innovative spinal analgesics. Thus, novel N/OFQ related peptides were synthesized in order to identify and pharmacologically characterize a mixed NOP/ µ opioid receptor agonist. EXPERIMENTAL APPROACH: The following in vitro assays were used: calcium mobilization in cells expressing the human NOP or classical opioid receptors and chimeric G proteins, receptor and [(35)S]-GTPγS binding, [(35)S]-GTPγS binding in rat spinal cord membranes, guinea pig ileum bioassay. In vivo experiments were performed in monkeys using the tail withdrawal assay. KEY RESULTS: From calcium mobilization studies [Dmt(1)]N/OFQ(1-13)-NH(2) was selected as the most potent and least selective compound. The mixed NOP/opioid full agonist activity and high affinity of [Dmt(1)]N/OFQ(1-13)-NH(2) was confirmed at human recombinant receptors in receptor binding, calcium mobilization and/or [(35)S]-GTPγS binding studies, at rat spinal cord receptors in [(35)S]-GTPγS binding experiments, and at guinea pig receptors inhibiting neurogenic contractions in the ileum. In vivo in the tail withdrawal assay in monkeys i.t. [Dmt(1) ]N/OFQ(1-13)-NH(2) was able to elicit robust and long-lasting antinociceptive effects. CONCLUSIONS AND IMPLICATIONS: Collectively, these results demonstrate that [Dmt(1)]N/OFQ(1-13)-NH(2) behaves as NOP/opioid receptor universal agonist and substantiate the suggestion that such mixed ligands are worthy of development as innovative spinal analgesics.


Assuntos
Analgésicos/farmacologia , Cálcio/metabolismo , Íleo/efeitos dos fármacos , Íleo/metabolismo , Oligopeptídeos/farmacologia , Peptídeos Opioides/agonistas , Receptores Opioides/agonistas , Animais , Cricetinae , Cricetulus , Relação Dose-Resposta a Droga , Estimulação Elétrica , Feminino , Guanosina 5'-O-(3-Tiotrifosfato)/química , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Cobaias , Humanos , Técnicas In Vitro , Injeções Espinhais , Macaca mulatta , Masculino , Ligação Proteica , Ratos , Receptor de Nociceptina , Nociceptina
17.
J Neurosci ; 32(46): 16106-19, 2012 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-23152595

RESUMO

In the present study we investigated whether the neuropeptide nociceptin/orphanin FQ (N/OFQ), previously implicated in the pathogenesis of Parkinson's disease, also affects L-DOPA-induced dyskinesia. In striatal slices of naive rodents, N/OFQ (0.1-1 µm) prevented the increase of ERK phosphorylation and the loss of depotentiation of synaptic plasticity induced by the D1 receptor agonist SKF38393 in spiny neurons. In vivo, exogenous N/OFQ (0.03-1 nmol, i.c.v.) or a synthetic N/OFQ receptor agonist given systemically (0.01-1 mg/Kg) attenuated dyskinesias expression in 6-hydroxydopamine hemilesioned rats primed with L-DOPA, without causing primary hypolocomotive effects. Conversely, N/OFQ receptor antagonists worsened dyskinesia expression. In vivo microdialysis revealed that N/OFQ prevented dyskinesias simultaneously with its neurochemical correlates such as the surge of nigral GABA and glutamate, and the reduction of thalamic GABA. Regional microinjections revealed that N/OFQ attenuated dyskinesias more potently and effectively when microinjected in striatum than substantia nigra (SN) reticulata, whereas N/OFQ receptor antagonists were ineffective in striatum but worsened dyskinesias when given in SN. Quantitative autoradiography showed an increase in N/OFQ receptor binding in striatum and a reduction in SN of both unprimed and dyskinetic 6-hydroxydopamine rats, consistent with opposite adaptive changes of N/OFQ transmission. Finally, the N/OFQ receptor synthetic agonist also reduced dyskinesia expression in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated dyskinetic macaques without affecting the global parkinsonian score. We conclude that N/OFQ receptor agonists may represent a novel strategy to counteract L-DOPA-induced dyskinesias. Their action is possibly mediated by upregulated striatal N/OFQ receptors opposing the D1 receptor-mediated overactivation of the striatonigral direct pathway.


Assuntos
Antidiscinéticos , Antiparkinsonianos/efeitos adversos , Discinesia Induzida por Medicamentos/tratamento farmacológico , Levodopa/efeitos adversos , Peptídeos Opioides/agonistas , Animais , Autorradiografia , Comportamento Animal/efeitos dos fármacos , Fenômenos Eletrofisiológicos , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Ácido Glutâmico/metabolismo , Macaca , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microdiálise , Microinjeções , Peptídeos Opioides/antagonistas & inibidores , Peptídeos Opioides/genética , Oxidopamina/toxicidade , Equilíbrio Postural/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Ácido gama-Aminobutírico/metabolismo , Nociceptina
18.
J Pharmacol Exp Ther ; 343(1): 72-81, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22743574

RESUMO

Buprenorphine is known as a µ-opioid peptide (MOP) receptor agonist, but its antinociception is compromised by the activation of nociceptin/orphanin FQ peptide (NOP) receptors in rodents. The aim of this study was to investigate the roles of MOP and NOP receptors in regulating buprenorphine-induced physiological responses in primates (rhesus monkeys). The effects of MOP antagonist (naltrexone), NOP antagonist [(±)-1-[(3R*,4R*)-1-(cyclooctylmethyl)-3-(hydroxymethyl)-4-piperidinyl]-3-ethyl-1,3-dihydro-2H-benzimidazol-2-one (J-113397)], and NOP agonists [(1S,3aS)-8-(2,3,3a,4,5,6-hexahydro-1H-phenalen-1-yl)-1-phenyl-1,3,8-triaza-spiro[4.5] decan-4-one (Ro 64-6198) and 3-endo-8-[bis(2-methylphenyl)methyl]-3-phenyl-8-azabicyclo[3.2.1]octan-3-ol (SCH 221510)] on buprenorphine were studied in three functional assays for measuring analgesia, respiratory depression, and itch in primates. Over the dose range of 0.01 to 0.1 mg/kg, buprenorphine dose-dependently produced antinociception, respiratory depression, and itch/scratching responses, and there was a ceiling effect at higher doses (0.1-1 mg/kg). Naltrexone (0.03 mg/kg) produced similar degrees of rightward shifts of buprenorphine's dose-response curves for all three endpoints. Mean pK(B) values of naltrexone (8.1-8.3) confirmed that MOP receptors mediated mainly buprenorphine-induced antinociception, respiratory depression, and itch/scratching. In contrast, J-113397 (0.1 mg/kg) did not change buprenorphine-induced physiological responses, indicating that there were no functional NOP receptors in buprenorphine-induced effects. More importantly, both NOP agonists, Ro 64-6198 and SCH 221510, enhanced buprenorphine-induced antinociception without respiratory depression and itch/ scratching. The dose-addition analysis revealed that buprenorphine in combination with the NOP agonist synergistically produced antinociceptive effects. These findings provided functional evidence that the activation of NOP receptors did not attenuate buprenorphine-induced antinociception in primates; instead, the coactivation of MOP and NOP receptors produced synergistic antinociception without other side effects. This study strongly supports the therapeutic potential of mixed MOP/NOP agonists as innovative analgesics.


Assuntos
Buprenorfina/farmacologia , Peptídeos Opioides/fisiologia , Receptores Opioides mu/fisiologia , Receptores Opioides/fisiologia , Animais , Compostos Azabicíclicos/farmacologia , Feminino , Macaca mulatta , Masculino , Peptídeos Opioides/agonistas , Medição da Dor/efeitos dos fármacos , Medição da Dor/métodos , Receptores Opioides/agonistas , Receptores Opioides mu/agonistas , Receptor de Nociceptina , Nociceptina
19.
Mol Brain ; 5: 4, 2012 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-22289619

RESUMO

BACKGROUND: An important limiting factor in the development of centrally acting pharmaceuticals is the blood-brain barrier (BBB). Transport of therapeutic peptides through this highly protective physiological barrier remains a challenge for peptide drug delivery into the central nervous system (CNS). Because the most common strategy to treat moderate to severe pain consists of the activation of opioid receptors in the brain, the development of active opioid peptide analogues as potential analgesics requires compounds with a high resistance to enzymatic degradation and an ability to cross the BBB. RESULTS: Herein we report that tetrapeptide analogues of the type H-Dmt1-Xxx2-Yyy3-Gly4-NH2 are transported into the brain after intravenous and subcutaneous administration and are able to activate the µ- and δ opioid receptors more efficiently and over longer periods of time than morphine. Using the hot water tail flick test as the animal model for antinociception, a comparison in potency is presented between a side chain conformationally constrained analogue containing the benzazepine ring (BVD03, Yyy3: Aba), and a "ring opened" analogue (BVD02, Yyy3: Phe). The results show that in addition to the increased lipophilicity through amide bond N-methylation, the conformational constraint introduced at the level of the Phe3 side chain causes a prolonged antinociception. Further replacement of NMe-D-Ala2 by D-Arg2 in the tetrapeptide sequence led to an improved potency as demonstrated by a higher and maintained antinociception for AN81 (Xxx2: D-Arg) vs. BVD03 (Xxx2: NMe-D-Ala). A daily injection of the studied opioid ligands over a time period of 5 days did however result in a substantial decrease in antinociception on the fifth day of the experiment. The compact opioid agonist-NK1 antagonist hybrid SBCHM01 could not circumvent opioid induced tolerance. CONCLUSIONS: We demonstrated that the introduction of a conformational constraint has an important impact on opioid receptor activation and subsequent antinociception in vivo. Further amino acid substitution allowed to identify AN81 as an opioid ligand able to access the CNS and induce antinociception at very low doses (0.1 mg/kg) over a time period up to 7 hours. However, tolerance became apparent after repetitive i.v. administration of the investigated tetrapeptides. This side effect was also observed with the dual opioid agonist-NK1 receptor antagonist SBCHM01.


Assuntos
Antagonistas dos Receptores de Neurocinina-1 , Nociceptividade/efeitos dos fármacos , Oligopeptídeos/farmacologia , Peptídeos Opioides/agonistas , Receptores Opioides mu/agonistas , Proteínas Recombinantes/farmacologia , Analgésicos/farmacologia , Animais , Relação Dose-Resposta a Droga , Injeções Intravenosas , Ligantes , Camundongos , Camundongos Endogâmicos C57BL , Morfina/administração & dosagem , Morfina/farmacologia , Oligopeptídeos/química , Peptídeos Opioides/administração & dosagem , Peptídeos Opioides/farmacologia , Receptores da Neurocinina-1/metabolismo , Receptores Opioides mu/metabolismo , Proteínas Recombinantes/química , Fatores de Tempo
20.
Pharmacology ; 88(1-2): 50-4, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21757975

RESUMO

Cough continues to be one of the top reasons why patients seek medical attention from health care providers. The prescription antitussive market is dominated by opioids, such as codeine that produces inconsistent efficacy and is often accompanied by significant side effect liabilities. Consequently, cough represents an unmet medical need and an underserved market. Yet, against the backdrop of increasing cough research, the development of novel treatments has been exceptionally challenging with dextromethorphan being the last US drug approved for cough almost a half century ago. We support the position that an unambiguous and actionable 'road map' that clearly delineates the pathway forward for new cough suppressants from basic research to and beyond clinical proof-of-concept studies will be an important aspect for future success of this pharmacological class of drug. Pivotal to the establishment of such a road map will be the review of lessons learned from antitussive agents that have been recently progressed to proof-of-concept trials. In the present commentary, we briefly discuss observations and challenges pertaining to SCH 486757, a selective orally active NOP agonist that has recently advanced to human antitussive testing.


Assuntos
Antitussígenos/farmacologia , Compostos Azabicíclicos/farmacologia , Tosse/tratamento farmacológico , Pirimidinas/farmacologia , Animais , Antitussígenos/administração & dosagem , Antitussígenos/uso terapêutico , Compostos Azabicíclicos/administração & dosagem , Compostos Azabicíclicos/uso terapêutico , Ensaios Clínicos como Assunto , Codeína/administração & dosagem , Codeína/farmacologia , Dextrometorfano/administração & dosagem , Dextrometorfano/farmacologia , Modelos Animais de Doenças , Cães , Avaliação Pré-Clínica de Medicamentos , Cobaias , Humanos , Peptídeos Opioides/agonistas , Peptídeos Opioides/metabolismo , Pirimidinas/administração & dosagem , Pirimidinas/uso terapêutico , Ratos , Receptores Opioides/agonistas , Nociceptina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA