Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 256
Filtrar
1.
Nature ; 631(8021): 686-693, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38961287

RESUMO

The µ-opioid receptor (µOR) is a well-established target for analgesia1, yet conventional opioid receptor agonists cause serious adverse effects, notably addiction and respiratory depression. These factors have contributed to the current opioid overdose epidemic driven by fentanyl2, a highly potent synthetic opioid. µOR negative allosteric modulators (NAMs) may serve as useful tools in preventing opioid overdose deaths, but promising chemical scaffolds remain elusive. Here we screened a large DNA-encoded chemical library against inactive µOR, counter-screening with active, G-protein and agonist-bound receptor to 'steer' hits towards conformationally selective modulators. We discovered a NAM compound with high and selective enrichment to inactive µOR that enhances the affinity of the key opioid overdose reversal molecule, naloxone. The NAM works cooperatively with naloxone to potently block opioid agonist signalling. Using cryogenic electron microscopy, we demonstrate that the NAM accomplishes this effect by binding a site on the extracellular vestibule in direct contact with naloxone while stabilizing a distinct inactive conformation of the extracellular portions of the second and seventh transmembrane helices. The NAM alters orthosteric ligand kinetics in therapeutically desirable ways and works cooperatively with low doses of naloxone to effectively inhibit various morphine-induced and fentanyl-induced behavioural effects in vivo while minimizing withdrawal behaviours. Our results provide detailed structural insights into the mechanism of negative allosteric modulation of the µOR and demonstrate how this can be exploited in vivo.


Assuntos
Microscopia Crioeletrônica , Morfina , Naloxona , Receptores Opioides mu , Receptores Opioides mu/metabolismo , Receptores Opioides mu/agonistas , Receptores Opioides mu/química , Naloxona/farmacologia , Animais , Camundongos , Regulação Alostérica/efeitos dos fármacos , Humanos , Morfina/farmacologia , Morfina/química , Masculino , Modelos Moleculares , Analgésicos Opioides/química , Analgésicos Opioides/farmacologia , Analgésicos Opioides/metabolismo , Antagonistas de Entorpecentes/farmacologia , Antagonistas de Entorpecentes/química , Ligantes , Feminino , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia , Overdose de Opiáceos/tratamento farmacológico , Cinética , Fentanila/química , Fentanila/farmacologia , Fentanila/análogos & derivados
2.
Molecules ; 29(11)2024 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-38893511

RESUMO

The opioid crisis in the United States is a significant public health issue, with a nearly threefold increase in opioid-related fatalities between 1999 and 2014. In response to this crisis, society has made numerous efforts to mitigate its impact. Recent advancements in understanding the structural intricacies of the κ opioid receptor (KOR) have improved our knowledge of how opioids interact with their receptors, triggering downstream signaling pathways that lead to pain relief. This review concentrates on the KOR, offering crucial structural insights into the binding mechanisms of both agonists and antagonists to the receptor. Through comparative analysis of the atomic details of the binding site, distinct interactions specific to agonists and antagonists have been identified. These insights not only enhance our understanding of ligand binding mechanisms but also shed light on potential pathways for developing new opioid analgesics with an improved risk-benefit profile.


Assuntos
Analgésicos Opioides , Receptores Opioides kappa , Receptores Opioides kappa/metabolismo , Receptores Opioides kappa/química , Humanos , Analgésicos Opioides/química , Analgésicos Opioides/farmacologia , Animais , Sítios de Ligação , Ligantes , Transdução de Sinais/efeitos dos fármacos , Ligação Proteica , Relação Estrutura-Atividade , Antagonistas de Entorpecentes/química , Dor/tratamento farmacológico , Dor/metabolismo
3.
J Med Chem ; 67(12): 10447-10463, 2024 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-38869493

RESUMO

In recent years, synthetic opioids have emerged as a predominant cause of drug-overdose-related fatalities, causing the "opioid crisis." To design safer therapeutic agents, we accidentally discovered µ-opioid receptor (MOR) antagonists based on fentanyl with a relatively uncomplicated chemical composition that potentiates structural modifications. Here, we showed the development of novel atropisomeric fentanyl analogues that exhibit more potent antagonistic activity against MOR than naloxone, a morphinan MOR antagonist. Derivatives displaying stable axial chirality were synthesized based on the amide structure of fentanyl. The aS- and aR-enantiomers exerted antagonistic and agonistic effects on the MOR, respectively, and each atropisomer interacted with the MOR by assuming a distinct binding mode through molecular docking. These findings suggest that introducing atropisomerism into fentanyl may serve as a key feature in the molecular design of future MOR antagonists to help mitigate the opioid crisis.


Assuntos
Fentanila , Receptores Opioides mu , Receptores Opioides mu/antagonistas & inibidores , Receptores Opioides mu/metabolismo , Fentanila/farmacologia , Fentanila/análogos & derivados , Fentanila/química , Estereoisomerismo , Humanos , Simulação de Acoplamento Molecular , Relação Estrutura-Atividade , Animais , Antagonistas de Entorpecentes/química , Antagonistas de Entorpecentes/farmacologia , Conformação Molecular , Analgésicos Opioides/farmacologia , Analgésicos Opioides/química , Analgésicos Opioides/síntese química , Células CHO , Cricetulus
4.
Bioorg Chem ; 148: 107489, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38797065

RESUMO

The number of opioid-related overdose deaths and individuals that have suffered from opioid use disorders have significantly increased over the last 30 years. FDA approved maintenance therapies to treat opioid use disorder may successfully curb drug craving and prevent relapse but harbor adverse effects that reduce patient compliance. This has created a need for new chemical entities with improved patient experience. Previously our group reported a novel lead compound, NAT, a mu-opioid receptor antagonist that potently antagonized the antinociception of morphine and showed significant blood-brain barrier permeability. However, NAT belongs to thiophene containing compounds which are known structural alerts for potential oxidative metabolism. To overcome this, 15 NAT derivatives with various substituents at the 5'-position of the thiophene ring were designed and their structure-activity relationships were studied. These derivatives were characterized for their binding affinity, selectivity, and functional activity at the mu opioid receptor and assessed for their ability to antagonize the antinociceptive effects of morphine in vivo. Compound 12 showed retention of the basic pharmacological attributes of NAT while improving the withdrawal effects that were experienced in opioid-dependent mice. Further studies will be conducted to fully characterize compound 12 to examine whether it would serve as a new lead for opioid use disorder treatment and management.


Assuntos
Receptores Opioides mu , Animais , Relação Estrutura-Atividade , Camundongos , Receptores Opioides mu/metabolismo , Receptores Opioides mu/antagonistas & inibidores , Humanos , Estrutura Molecular , Tiofenos/química , Tiofenos/farmacologia , Tiofenos/síntese química , Tiofenos/uso terapêutico , Masculino , Relação Dose-Resposta a Droga , Analgésicos Opioides/farmacologia , Analgésicos Opioides/química , Antagonistas de Entorpecentes/farmacologia , Antagonistas de Entorpecentes/química , Morfina/farmacologia
5.
Bioorg Chem ; 130: 106257, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36375349

RESUMO

Ten new indole alkaloids (1-10) as well as eleven known analogs (11-21) were isolated from the stems and hooks of Uncaria rhynchophylla. Their structure elucidation was based on extensive NMR studies, MS and ECD data, with the essential aid of DFT prediction of ECD spectra. Compound 1 was determined as a 17,19-seco-cadambine-type alkaloid, and compound 3 was confirmed to be a 3,4-seco-tricyclic monoterpene indole alkaloid, which are the first seco-alkaloids possessing such cleavage positions from U. rhynchophylla. All the isolated compounds were evaluated for their bioactivities on dopamine D2 and Mu opioid receptors for discovering natural therapeutic drugs targeting central nervous system (CNS) diseases. Compounds 1, 2, 4, 5, 20 and 21 showed antagonistic bioactivities on the D2 receptor (IC50 0.678-15.200 µM), and compounds 1, 3, 6, 9, 10, 13, 18, 19 and 21 exhibited antagonistic effects on the Mu receptor (IC50 2.243-32.200 µM). Among them, compounds 1 and 21 displayed dual-target activities. Compound 1 showed conspicuous antagonistic activity on D2 and Mu receptors with the IC50 values of 0.678 ± 0.182 µM and 13.520 ± 2.480 µM, respectively. Compound 21 displayed moderate antagonistic activity on the two receptors with the IC50 values at 15.200 ± 1.764 µM and 32.200 ± 5.695 µM, respectively.


Assuntos
Antagonistas dos Receptores de Dopamina D2 , Alcaloides Indólicos , Uncaria , Alcaloides/química , Alcaloides/farmacologia , Dopamina/metabolismo , Alcaloides Indólicos/farmacologia , Alcaloides Indólicos/química , Receptores Opioides mu/antagonistas & inibidores , Uncaria/química , Antagonistas dos Receptores de Dopamina D2/química , Antagonistas dos Receptores de Dopamina D2/farmacologia , Antagonistas de Entorpecentes/química , Antagonistas de Entorpecentes/farmacologia
6.
Arch Pharm (Weinheim) ; 356(1): e2200432, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36328777

RESUMO

The development of novel µ-opioid receptor (MOR) antagonists is one of the main objectives of drug discovery and development. Based on a simplified version of the morphinan scaffold, 3-[3-(phenalkylamino)cyclohexyl]phenol analogs were designed, synthesized, and evaluated for their MOR antagonist activity in vitro and in silico. At the highest concentrations, the compounds decreased by 52% to 75% DAMGO-induced GTPγS stimulation, suggesting that they acted as antagonists. Moreover, Extra-Precision Glide and Generalized-Born Surface Area experiments provided useful information on the nature of the ligand-receptor interactions, indicating a peculiar combination of C-1 stereochemistry and N-substitutions as feasibly essential for MOR-ligand complex stability. Interestingly, compound 9 showed the best experimental binding affinity, the highest antagonist activity, and the finest MOR-ligand complex stability. In silico experiments also revealed that the most promising stereoisomer (1R, 3R, 5S) 9 retained 1,3-cis configuration with phenol ring equatorial oriented. Further studies are needed to better characterize the pharmacodynamics and pharmacokinetic properties of these compounds.


Assuntos
Naltrexona , Antagonistas de Entorpecentes , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Antagonistas de Entorpecentes/química , Ligantes , Fenóis/farmacologia , Relação Estrutura-Atividade , Receptores Opioides mu/química , Receptores Opioides mu/metabolismo
7.
Hum Vaccin Immunother ; 18(6): 2122507, 2022 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-36194773

RESUMO

Innovative therapies to complement current treatments are needed to curb the growing incidence of fatal overdoses related to synthetic opioids. Murine and chimeric monoclonal antibodies (mAb) specific for fentanyl and its analogs have demonstrated pre-clinical efficacy in preventing and reversing drug-induced toxicity in rodent models. However, mAb-based therapeutics require extensive engineering as well as in vitro and in vivo characterization to advance to first-in-human clinical trials. Here, novel murine anti-fentanyl mAbs were selected for development based on affinity for fentanyl, and efficacy in counteracting the pharmacological effects of fentanyl in mice. Humanization and evaluation of mutations designed to eliminate predicted post-translational modifications resulted in two humanized mAbs that were effective at preventing fentanyl-induced pharmacological effects in rats. These humanized mAbs showed favorable biophysical properties with respect to aggregation and hydrophobicity by chromatography-based assays, and thermostability by dynamic scanning fluorimetry. These results collectively support that the humanized anti-fentanyl mAbs developed herein warrant further clinical development for treatment of fentanyl toxicity.


Assuntos
Anticorpos Monoclonais Humanizados , Fentanila , Antagonistas de Entorpecentes , Animais , Humanos , Camundongos , Ratos , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados/uso terapêutico , Proteínas do Sistema Complemento , Fentanila/imunologia , Fentanila/toxicidade , Antagonistas de Entorpecentes/química , Antagonistas de Entorpecentes/imunologia
8.
ACS Chem Neurosci ; 13(8): 1315-1332, 2022 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-35410469

RESUMO

Focused modification of a sulfonamide-based kappa opioid receptor (KOR) antagonist series previously reported by this laboratory was investigated. A total of 32 analogues were prepared to explore linker replacement, constraint manipulation, and aryl group or amine substitution. All analogues were assayed for KOR antagonist activity, and the initial lead compound was assessed for in vivo CNS penetration. The most improved analogue possessed a 4-fold increase of potency (IC50 = 18.9 ± 4.4 nM) compared with the lead compound (IC50 = 83.5 ± 20 nM) from an earlier work. The initial lead compound was found to attain suitable brain levels and to possess a shorter clearance time than canonical KOR antagonists such as JDTic.


Assuntos
Receptores Opioides kappa , Tetra-Hidroisoquinolinas , Antagonistas de Entorpecentes/química , Antagonistas de Entorpecentes/farmacologia , Sulfonamidas/farmacologia , Tetra-Hidroisoquinolinas/química
9.
J Med Chem ; 64(14): 10139-10154, 2021 07 22.
Artigo em Inglês | MEDLINE | ID: mdl-34236190

RESUMO

A new series of propionamide derivatives was developed as dual µ-opioid receptor agonists and σ1 receptor antagonists. Modification of a high-throughput screening hit originated a series of piperazinylcycloalkylmethyl propionamides, which were explored to overcome the challenge of achieving balanced dual activity and convenient drug-like properties. The lead compound identified, 18g, showed good analgesic effects in several animal models of both acute (paw pressure) and chronic (partial sciatic nerve ligation) pain, with reduced gastrointestinal effects in comparison with oxycodone.


Assuntos
Amidas/farmacologia , Analgésicos Opioides/farmacologia , Antagonistas de Entorpecentes/farmacologia , Dor/tratamento farmacológico , Receptores Opioides mu/agonistas , Receptores sigma/antagonistas & inibidores , Amidas/síntese química , Amidas/química , Analgésicos Opioides/síntese química , Analgésicos Opioides/química , Animais , Relação Dose-Resposta a Droga , Humanos , Camundongos , Estrutura Molecular , Antagonistas de Entorpecentes/síntese química , Antagonistas de Entorpecentes/química , Relação Estrutura-Atividade , Receptor Sigma-1
10.
Phys Chem Chem Phys ; 23(21): 12260-12269, 2021 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-34013938

RESUMO

Nalmefene is an opiate derivative having a similar structure to naltrexone. Recent evidence suggests that nalmefene, acting as the innate immune protein toll-like receptor 4 (TLR4) antagonist, effectively reduces the injury of lung ischemia-reperfusion and prevents neuroinflammation. However, the molecular recognition mechanism, especially the enantioselectivity, of nalmefene by the innate immune receptor is not well understood. Herein in vitro assays and in silico simulations were performed to dissect the innate immune recognition of nalmefene at the atomic, molecular, and cellular levels. Biophysical binding experiments and molecular dynamic simulations provide direct evidence that (-)-nalmefene and (+)-nalmefene bind to the hydrophobic cavity of myeloid differentiation protein 2 (MD-2) and behave similarly, which is primarily driven by hydrophobic interactions. The inhibition activity and the calculated binding free energies show that no enantioselectivity was observed during the interaction of nalmefene with MD-2 as well as the inhibition of TLR4 signaling. Interestingly, nalmefene showed ∼6 times better TLR4 antagonisic activity than naltrexone, indicating that the bioisosteric replacement with the methylene group is critical for the molecular recognition of nalmefene by MD-2. In all, this study provides molecular insight into the innate immune recognition of nalmefene, which demonstrates that nalmefene is non-enantioselectively sensed by MD-2.


Assuntos
Antígeno 96 de Linfócito/antagonistas & inibidores , Naltrexona/análogos & derivados , Antagonistas de Entorpecentes/farmacologia , Receptor 4 Toll-Like/antagonistas & inibidores , Animais , Células Cultivadas , Humanos , Interações Hidrofóbicas e Hidrofílicas , Antígeno 96 de Linfócito/genética , Antígeno 96 de Linfócito/isolamento & purificação , Camundongos , Conformação Molecular , Simulação de Dinâmica Molecular , Naltrexona/química , Naltrexona/farmacologia , Antagonistas de Entorpecentes/química , Transdução de Sinais/efeitos dos fármacos , Estereoisomerismo , Termodinâmica
11.
Bull Exp Biol Med ; 170(6): 710-713, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33893949

RESUMO

We performed a comparative analysis of infarction-limiting activity of analogues of opioid receptor agonist U-50488 under conditions of heart reperfusion in rats. Derivatives of amide N-methyl-2-(pyrrolidin-1-yl)cyclohexyl-1-amine were administered 5 min before reperfusion in a dose of 1 mg/kg, derivative II (opicor) was additionally used in a dose of 2 mg/kg. In a dose of 1 mg/kg, all derivatives of opioid U-50488 were ineffective and produced no infarction-limiting effect. Opicor in a dose of 2 mg/kg reduced the infarction size/area at risk ratio and improved the contractility parameters of the isolated heart. Opioid receptor antagonist naltrexone (5 mg/kg) abolished the infarction-limiting effect of opicor. Hence, the infarction-reducing effect of opicor is associated with activation of opioid receptors. We also demonstrated that the opioid (opicor) can improve cardiac contractility during the reperfusion period.


Assuntos
Amidas/química , Aminas/química , Aminas/uso terapêutico , Analgésicos Opioides/uso terapêutico , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Animais , Cardiotônicos/química , Cardiotônicos/uso terapêutico , Coração/efeitos dos fármacos , Masculino , Antagonistas de Entorpecentes/química , Antagonistas de Entorpecentes/uso terapêutico , Ratos , Ratos Wistar , Receptores Opioides/metabolismo
12.
ChemMedChem ; 16(12): 1917-1926, 2021 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-33599108

RESUMO

The synthesis of 5-formyl-6-aryl-6H-indolo[3,2,1-de][1,5] naphthyridine-2-carboxylates by reaction between 1-formyl-9H-ß-carbolines and cinnamaldehydes in the presence of pyrrolidine in water with microwave irradiation is described. Pharmacophoric modification of the formyl group offered several new fused ß-carboline derivatives, which were investigated for their κ-opioid receptor (KOR) agonistic activity. Two compounds 4 a and 4 c produced appreciable agonist activity on KOR with EC50 values of 46±19 and 134±9 nM, respectively. Moreover, compound-induced KOR signaling studies suggested both compounds to be extremely G-protein-biased agonists. The analgesic effect of 4 a was validated by the increase in tail flick latency in mice in a time-dependent manner, which was completely blocked by the KOR-selective antagonist norBNI. Moreover, unlike U50488, an unbiased full KOR agonist, 4 a did not induce sedation. The docking of 4 a with the human KOR was studied to rationalize the result.


Assuntos
Analgésicos/farmacologia , Carbolinas/farmacologia , Antagonistas de Entorpecentes/farmacologia , Dor/tratamento farmacológico , Receptores Opioides kappa/agonistas , Analgésicos/síntese química , Analgésicos/química , Animais , Carbolinas/síntese química , Carbolinas/química , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Estrutura Molecular , Antagonistas de Entorpecentes/síntese química , Antagonistas de Entorpecentes/química
13.
J Enzyme Inhib Med Chem ; 36(1): 183-187, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33401948

RESUMO

We have applied a recently developed HPLC-MS enzymatic assay to investigate the cryptic peptides generated by the action of the insulin-degrading enzyme (IDE) on some neuropeptides (NPs) involved in the development of tolerance and dependence to opioids. Particularly, the tested NPs are generated from the NPFF precursor (pro-NPFF (A)): NPFF (FLFQPQRF) and NPAF (AGEGLSSPFWSLAAPQRF). The results show that IDE is able to cleave NPFF and NPAF, generating specific cryptic peptides. As IDE is also responsible for the processing of many other peptides in the brain (amyloid beta protein among the others), we have also performed competitive degradation assays using mixtures of insulin and the above mentioned NPs. Data show that insulin is able to slow down the degradation of both NPs tested, whereas, surprisingly, NPAF is able to accelerate insulin degradation, hinting IDE as the possible link responsible of the mutual influence between insulin and NPs metabolism.


Assuntos
Insulina/química , Insulisina/química , Antagonistas de Entorpecentes/química , Neuropeptídeos/química , Oligopeptídeos/química , Fragmentos de Peptídeos/química , Sequência de Aminoácidos , Analgésicos Opioides/efeitos adversos , Animais , Cromatografia Líquida de Alta Pressão , Tolerância a Medicamentos , Humanos , Insulina/metabolismo , Espectrometria de Massas , Neuropeptídeos/metabolismo , Fragmentos de Peptídeos/análise , Proteólise , Ratos , Proteínas Recombinantes/química , Soluções
14.
Anesth Analg ; 132(2): 406-419, 2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-33332902

RESUMO

A serious adverse effect of prescription opioid analgesics is addiction, both to these analgesics and to illicit drugs like heroin that also activate the µ-opioid receptor (MOR). Opioid use disorder (OUD) and opioid overdose deaths represent a current American health crisis, and the prescription of opioid analgesics has contributed significantly to this crisis. While prescription opioids are highly effective analgesics, there currently exists no facile way to use them for extended periods without the risk of addiction. If addiction caused by MOR-targeting analgesics could be blocked by blending in a new "antiaddiction" ingredient that does not diminish analgesia and does not introduce its own therapeutically limiting side effects, then continued clinical use of prescription opioids for treating pain could be maintained (or even enhanced) instead of curtailed. In this narrative review, we contextualize this hypothesis, first with a brief overview of the current American opioid addiction crisis. The neurobiology of 2 key receptors in OUD development, MOR and the κ-opioid receptor (KOR), is then discussed to highlight the neuroanatomical features and circuitry in which signal transduction from these receptors lie in opposition-creating opportunities for pharmacological intervention in curtailing the addictive potential of MOR agonism. Prior findings with mixed MOR/KOR agonists are considered before exploring new potential avenues such as biased KOR agonists. New preclinical data are highlighted, demonstrating that the G protein-biased KOR agonist nalfurafine reduces the rewarding properties of MOR-targeting analgesics and enhances MOR-targeting analgesic-induced antinociception. Finally, we discuss the recent discovery that a regulator of G protein signaling (namely, RGS12) is a key component of signaling bias at KOR, presenting another drug discovery target toward identifying a single agent or adjuvant to be added to traditional opioid analgesics that could reduce or eliminate the addictive potential of the latter drug.


Assuntos
Desenho de Fármacos , Antagonistas de Entorpecentes/farmacologia , Nociceptividade/efeitos dos fármacos , Dor Nociceptiva/tratamento farmacológico , Transtornos Relacionados ao Uso de Opioides/prevenção & controle , Receptores Opioides kappa/agonistas , Receptores Opioides mu/agonistas , Animais , Humanos , Estrutura Molecular , Antagonistas de Entorpecentes/efeitos adversos , Antagonistas de Entorpecentes/química , Dor Nociceptiva/metabolismo , Dor Nociceptiva/fisiopatologia , Dor Nociceptiva/psicologia , Transtornos Relacionados ao Uso de Opioides/etiologia , Proteínas RGS/metabolismo , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/metabolismo , Transdução de Sinais , Relação Estrutura-Atividade
15.
Chem Biodivers ; 18(1): e2000871, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33351271

RESUMO

Nociceptin receptor (NOP) belongs to the family of opioid receptors but was discovered and characterized much later than the so called classical opioid receptors, µ, δ and κ (or MOP, DOP and KOP, resp.). Nociceptin/orphanin FQ (N/OFQ) is the endogenous ligand of this receptor and it controls numerous important functions in the central nervous system and in the periphery, so its analogs may be developed as innovative drugs for the treatment of a variety of conditions and pathological states. Availability of potent and selective ligands with high affinity to NOP receptor is essential to fully understand the role of NOP-N/OFQ system in the body, which in turn may lead to designing novel therapeutics. Here, we have focused on reviewing the structure of potent peptide-based agonists, antagonists, biased analogs and bivalent ligands that target NOP receptor.


Assuntos
Descoberta de Drogas , Peptídeos Opioides/química , Receptores Opioides/metabolismo , Sequência de Aminoácidos , Humanos , Ligantes , Antagonistas de Entorpecentes/química , Antagonistas de Entorpecentes/metabolismo , Peptídeos Opioides/metabolismo , Receptores Opioides/agonistas , Receptores Opioides/química , Relação Estrutura-Atividade , Receptor de Nociceptina , Nociceptina
16.
Cell Mol Neurobiol ; 41(5): 961-975, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32323150

RESUMO

Compound 511 (511) is specially developed for opioid addiction treatment based on the Ancient Chinese drug rehabilitation literature, and its composition has profound effects in the treatment of drug addiction in various clinical trials and animal experiments. The effect of 511 on the rewarding properties of morphine and craving responses and its potential mechanisms remain unclear. Here, we have applied a conditioned place preference (CPP) paradigm in mice to measure morphine-induced rewarding effects under the treatment of 511. Then we used the RNA sequencing strategy to screen its potential mechanisms. In our research, firstly, we found 511 could decrease CPP score, locomotor activity, self-administration, jumping behavior, weight loss, wet-dog shakes, and stereotyped behavior. Then the brain VTA region tissues were performed mRNA sequencing to detect potential mechanisms. We found the brain-derived neurotrophic factor (BDNF) and tropomyosin-related kinase B (TrkB) were downregulated in morphine-induced CPP, whereas the decreased BDNF and TrkB were reversed after 511 treatment. We retested the levels of BDNF and TrkB using qRT-PCR and Western blot and found the similar results to mRNA sequencing. It has been widely reported that BDNF-TrkB signaling in the VTA is involved in multiple facets of addiction, including reward and motivation, so we focused on the BDNF-TrkB signaling to investigate the anti-addiction mechanisms of 511 in morphine addiction mice. We studied the downstream pathway of BDNF-TrkB and the soma size of dopaminergic neurons. The results showed 511 could increase the phosphorylation levels of PI3K and AKT, which were decreased in morphine-induced CPP. Simultaneously, 511 could decrease the level of PLCγ1 and the phosphorylation levels of ERK and S6K, which were increased in morphine-induced CPP. In addition, 511 also enlarged the soma size of VTA dopaminergic neurons, which was reduced in morphine-induced CPP. Hence, our research indicated 511 maybe mediate the BDNF-TrkB signaling in VTA to improve morphine addiction behavior.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/metabolismo , Condicionamento Clássico/fisiologia , Medicamentos de Ervas Chinesas/farmacologia , Glicoproteínas de Membrana/metabolismo , Morfina/administração & dosagem , Proteínas Tirosina Quinases/metabolismo , Área Tegmentar Ventral/metabolismo , Animais , Fator Neurotrófico Derivado do Encéfalo/antagonistas & inibidores , Condicionamento Clássico/efeitos dos fármacos , Medicamentos de Ervas Chinesas/química , Masculino , Glicoproteínas de Membrana/antagonistas & inibidores , Camundongos , Camundongos Endogâmicos C57BL , Antagonistas de Entorpecentes/química , Antagonistas de Entorpecentes/farmacologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Recompensa , Autoadministração , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Área Tegmentar Ventral/efeitos dos fármacos
17.
Biomolecules ; 10(10)2020 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-32998249

RESUMO

Opioid peptides and receptors are broadly expressed throughout peripheral and central nervous systems and have been the subject of intense long-term investigations. Such studies indicate that some endogenous neuropeptides, called anti-opioids, participate in a homeostatic system that tends to reduce the effects of endogenous and exogenous opioids. Anti-opioid properties have been attributed to various peptides, including melanocyte inhibiting factor (MIF)-related peptides, cholecystokinin (CCK), nociceptin/orphanin FQ (N/OFQ), and neuropeptide FF (NPFF). These peptides counteract some of the acute effects of opioids, and therefore, they are involved in the development of opioid tolerance and addiction. In this work, the anti-opioid profile of endogenous peptides was described, mainly taking into account their inhibitory influence on opioid-induced effects. However, the anti-opioid peptides demonstrated complex properties and could show opioid-like as well as anti-opioid effects. The aim of this review is to detail the phenomenon of crosstalk taking place between opioid and anti-opioid systems at the in vivo pharmacological level and to propose a cellular and molecular basis for these interactions. A better knowledge of these mechanisms has potential therapeutic interest for the control of opioid functions, notably for alleviating pain and/or for the treatment of opioid abuse.


Assuntos
Antagonistas de Entorpecentes/metabolismo , Receptores Opioides/metabolismo , Analgésicos Opioides/uso terapêutico , Colecistocinina/metabolismo , Tolerância a Medicamentos , Humanos , Antagonistas de Entorpecentes/química , Antagonistas de Entorpecentes/uso terapêutico , Oligopeptídeos/metabolismo , Peptídeos Opioides/metabolismo , Dor/tratamento farmacológico , Dor/metabolismo , Dor/patologia , Transtornos Relacionados ao Uso de Substâncias/tratamento farmacológico , Transtornos Relacionados ao Uso de Substâncias/metabolismo , Transtornos Relacionados ao Uso de Substâncias/patologia , Nociceptina
18.
Drug Des Devel Ther ; 14: 4179-4187, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33116406

RESUMO

BACKGROUND AND OBJECTIVE: The combination of naloxone hydrochloride (NH) and fentanyl citrate (FC) in patient-controlled analgesia (PCA) is examined to reduce the risk of opioid-induced nausea and vomiting. However, there are no such commercially available drug mixtures, and there is also no published evidence on the compatibility and stability of NH and FC. Thus, the primary purpose of the current research is to investigate the physical compatibility and chemical stability of NH when mixed with FC over a 72-h period in a 0.9% sodium chloride injection solution for PCA administration under storage at 4°C and 25°C. METHODS: Test solutions of 20 µg/mL FC and 4 µg/mL NH were prepared and stored in polyvinyl chloride (PVC) bags or glass bottles with a 0.9% sodium chloride injection solution as the diluent. During the 72-h storage period at 4°C or 25°C without light protection, the concentrations of the test drugs were assayed via high-performance liquid chromatography (HPLC), and the physical compatibility was determined with the naked eye. Furthermore, pH measurement of each sample was also performed with a pH meter. RESULTS: The percentages of the initial concentrations of FC and NH in the various solutions were maintained at a minimum of 98% over the 72-h study period. All of the mixtures remained clear and colourless throughout the observation period, and no precipitation or turbidity was observed in any of the batches. CONCLUSION: The 20 µg/mL FC test solution was physically compatible and chemically stable with the 4 µg/mL NH test solution when stored at 4°C or 25°C in PVC bags or glass bottles containing the 0.9% sodium chloride injection solution.


Assuntos
Analgesia Controlada pelo Paciente/métodos , Analgésicos Opioides/química , Fentanila/química , Naloxona/química , Antagonistas de Entorpecentes/química , Analgésicos Opioides/administração & dosagem , Cromatografia Líquida de Alta Pressão , Incompatibilidade de Medicamentos , Embalagem de Medicamentos , Estabilidade de Medicamentos , Fentanila/administração & dosagem , Injeções Intravenosas , Naloxona/administração & dosagem , Antagonistas de Entorpecentes/administração & dosagem , Padrões de Referência , Reprodutibilidade dos Testes , Solução Salina
19.
Molecules ; 25(18)2020 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-32957550

RESUMO

The opioid receptors are members of the G-protein-coupled receptor (GPCR) family and are known to modulate a variety of biological functions, including pain perception. Despite considerable advances, the mechanisms by which opioid agonists and antagonists interact with their receptors and exert their effect are still not completely understood. In this report, six new hybrids of the Dmt-Tic pharmacophore and cyclic peptides, which were shown before to have a high affinity for the µ-opioid receptor (MOR) were synthesized and characterized pharmacologically in calcium mobilization functional assays. All obtained ligands turned out to be selective antagonists of the δ-opioid receptor (DOR) and did not activate or block the MOR. The three-dimensional structural determinants responsible for the DOR antagonist properties of these analogs were further investigated by docking studies. The results indicate that these compounds attach to the DOR in a slightly different orientation with respect to the Dmt-Tic pharmacophore than Dmt-TicΨ[CH2-NH]Phe-Phe-NH2 (DIPP-NH2[Ψ]), a prototypical DOR antagonist peptide. Key pharmacophoric contacts between the DOR and the ligands were maintained through an analogous spatial arrangement of pharmacophores, which could provide an explanation for the predicted high-affinity binding and the experimentally observed functional properties of the novel synthetic ligands.


Assuntos
Dipeptídeos/metabolismo , Peptídeos Opioides/metabolismo , Peptídeos Cíclicos/metabolismo , Receptores Opioides delta/metabolismo , Tetra-Hidroisoquinolinas/metabolismo , Analgésicos Opioides/antagonistas & inibidores , Animais , Humanos , Ligantes , Simulação de Acoplamento Molecular , Antagonistas de Entorpecentes/química , Antagonistas de Entorpecentes/metabolismo , Peptídeos Opioides/síntese química , Peptídeos Cíclicos/síntese química , Receptores Opioides/química , Relação Estrutura-Atividade
20.
Org Lett ; 22(17): 6891-6896, 2020 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-32790319

RESUMO

The most challenging step in the preparation of many opioid antagonists is the selective N-demethylation of a 14-hydroxymorphinan precursor. This process is carried out on a large scale using stoichiometric amounts of hazardous chemicals like cyanogen bromide or chloroformates. We have developed a mild reagent- and catalyst-free procedure for the N-demethylation step based on the anodic oxidation of the tertiary amine. The ensuing intermediates can be readily hydrolyzed to the target nor-opioids in very good yields.


Assuntos
Analgésicos Opioides/química , Morfinanos/química , Antagonistas de Entorpecentes/química , Analgésicos Opioides/análise , Catálise , Desmetilação , Hidrólise , Estrutura Molecular , Oxirredução
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...