Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Proteins ; 89(10): 1386-1393, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34152652

RESUMO

We have shown that water-soluble variants of the human mu opioid receptor (wsMOR) containing a reduced number of hydrophobic residues at the lipid-facing residues of the transmembrane (TM) helices can be expressed in E. coli. In this study, we tested the consequences of increasing the number of mutations on the surface of the transmembrane domain on the receptor's aqueous solubility and ligand binding properties, along with mutation of 11 cysteine residues regardless of their solvent exposure value and location in the protein. We computationally engineered 10 different variants of MOR, and tested four of them for expression in E. coli. We found that all four variants were successfully expressed and could be purified in high quantities. The variants have alpha helical structural content similar to that of the native MOR, and they also display binding affinities for the MOR antagonist (naltrexone) similar to the wsMOR variants we engineered previously that contained many fewer mutations. Furthermore, for these full-length variants, the helical content remains unchanged over a wide range of pH values (pH 6 ~ 9). This study demonstrates the flexibility and robustness of the water-soluble MOR variants with respect to additional designed mutations in the TM domain and changes in pH, whereupon the protein's structural integrity and its ligand binding affinity are maintained. These variants of the full-length MOR with less hydrophobic surface residues and less cysteines can be obtained in large amounts from expression in E. coli and can serve as novel tools to investigate structure-function relationships of the receptor.


Assuntos
Receptores Opioides mu/química , Escherichia coli/metabolismo , Humanos , Mutação , Receptores Opioides mu/genética , Solubilidade , Água
2.
Int J Mol Sci ; 21(17)2020 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-32872204

RESUMO

Opioids and their antagonists alter vitamin C metabolism. Morphine binds to glutathione (l-γ-glutamyl-l-cysteinyl-glycine), an intracellular ascorbic acid recycling molecule with a wide range of additional activities. The morphine metabolite morphinone reacts with glutathione to form a covalent adduct that is then excreted in urine. Morphine also binds to adrenergic and histaminergic receptors in their extracellular loop regions, enhancing aminergic agonist activity. The first and second extracellular loops of adrenergic and histaminergic receptors are, like glutathione, characterized by the presence of cysteines and/or methionines, and recycle ascorbic acid with similar efficiency. Conversely, adrenergic drugs bind to extracellular loops of opioid receptors, enhancing their activity. These observations suggest functional interactions among opioids and amines, their receptors, and glutathione. We therefore explored the relative binding affinities of ascorbic acid, dehydroascorbic acid, opioid and adrenergic compounds, as well as various control compounds, to glutathione and glutathione-like peptides derived from the extracellular loop regions of the human beta 2-adrenergic, dopamine D1, histamine H1, and mu opioid receptors, as well as controls. Some cysteine-containing peptides derived from these receptors do bind ascorbic acid and/or dehydroascorbic acid and the same peptides generally bind opioid compounds. Glutathione binds not only morphine but also naloxone, methadone, and methionine enkephalin. Some adrenergic drugs also bind to glutathione and glutathione-like receptor regions. These sets of interactions provide a novel basis for understanding some ways that adrenergic, opioid and antioxidant systems interact during anesthesia and drug abuse and may have utility for understanding drug interactions.


Assuntos
Analgésicos Opioides/farmacologia , Ácido Ascórbico/farmacologia , Glutationa/metabolismo , Peptídeos/farmacologia , Ácido Desidroascórbico/farmacologia , Encefalina Metionina/farmacologia , Humanos , Hidromorfona/análogos & derivados , Hidromorfona/urina , Metadona/farmacologia , Morfina/farmacologia , Naloxona/farmacologia , Receptores Adrenérgicos beta 2/química , Receptores de Dopamina D1/química , Receptores Histamínicos H1/química , Receptores Opioides/química , Receptores Opioides mu/química
3.
Cell Mol Biol (Noisy-le-grand) ; 66(4): 243-249, 2020 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-32583783

RESUMO

Phytol (PHY), a chlorophyll-derived diterpenoid, exhibits numerous pharmacological properties, including antioxidant, antimicrobial, and anticancer activities. This study evaluates the anti-diarrheal effect of phytol (PHY) along with its possible mechanism of action through in-vivo and in-silico models. The effect of PHY was investigated on castor oil-induced diarrhea in Swiss mice by using prazosin, propranolol, loperamide, and nifedipine as standards with or without PHY. PHY at 50 mg/kg (p.o.) and all other standards exhibit significant (p < 0.05) anti-diarrheal effect in mice. The effect was prominent in the loperamide and propranolol groups. PHY co-treated with prazosin and propranolol was found to increase in latent periods along with a significant reduction in diarrheal section during the observation period than other individual or combined groups. Furthermore, molecular docking studies also suggested that PHY showed better interactions with the α- and ß-adrenergic receptors, especially with α-ADR1a and ß-ADR1. In the former case, PHY showed interaction with hydroxyl group of Ser192 at a distance of 2.91Å, while in the latter it showed hydrogen bond interactions with Thr170 and Lys297 with a distance of 2.65 and 2.72Å, respectively. PHY exerted significant anti-diarrheal effect in Swiss mice, possibly through blocking α- and ß-adrenergic receptors.


Assuntos
Simulação por Computador , Diarreia/tratamento farmacológico , Modelos Biológicos , Fitol/uso terapêutico , Sequência de Aminoácidos , Animais , Óleo de Rícino , Modelos Animais de Doenças , Jejum , Humanos , Canais Iônicos/química , Canais Iônicos/metabolismo , Masculino , Camundongos , Simulação de Acoplamento Molecular , Fitol/farmacologia , Receptores Adrenérgicos alfa 1/química , Receptores Adrenérgicos alfa 1/metabolismo , Receptores Adrenérgicos beta/química , Receptores Adrenérgicos beta/metabolismo , Receptores Opioides mu/química , Receptores Opioides mu/metabolismo , Homologia de Sequência de Aminoácidos
4.
Molecules ; 25(9)2020 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-32365707

RESUMO

The mu opioid receptor (MOR) is the primary target for analgesia of endogenous opioid peptides, alkaloids, synthetic small molecules with diverse scaffolds, and peptidomimetics. Peptide-based opioids are viewed as potential analgesics with reduced side effects and have received constant scientific interest over the years. This study focuses on three potent peptide and peptidomimetic MOR agonists, DALDA, [Dmt1]DALDA, and KGOP01, and the prototypical peptide MOR agonist DAMGO. We present the first molecular modeling study and structure-activity relationships aided by in vitro assays and molecular docking of the opioid peptide analogues, in order to gain insight into their mode of binding to the MOR. In vitro binding and functional assays revealed the same rank order with KGOP01 > [Dmt1]DALDA > DAMGO > DALDA for both binding and MOR activation. Using molecular docking at the MOR and three-dimensional interaction pattern analysis, we have rationalized the experimental outcomes and highlighted key amino acid residues responsible for agonist binding to the MOR. The Dmt (2',6'-dimethyl-L-Tyr) moiety of [Dmt1]DALDA and KGOP01 was found to represent the driving force for their high potency and agonist activity at the MOR. These findings contribute to a deeper understanding of MOR function and flexible peptide ligand-MOR interactions, that are of significant relevance for the future design of opioid peptide-based analgesics.


Assuntos
Oligopeptídeos/química , Oligopeptídeos/metabolismo , Peptídeos/química , Peptídeos/metabolismo , Receptores Opioides mu/química , Receptores Opioides mu/metabolismo , Animais , Sítios de Ligação , Células CHO , Cricetulus , Humanos , Cinética , Camundongos , Modelos Moleculares , Ligação Proteica , Conformação Proteica , Relação Estrutura-Atividade
5.
Sci Adv ; 5(11): eaax9115, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31807708

RESUMO

Selective activation of the δ-opioid receptor (DOP) has great potential for the treatment of chronic pain, benefitting from ancillary anxiolytic and antidepressant-like effects. Moreover, DOP agonists show reduced adverse effects as compared to µ-opioid receptor (MOP) agonists that are in the spotlight of the current "opioid crisis." Here, we report the first crystal structures of the DOP in an activated state, in complex with two relevant and structurally diverse agonists: the potent opioid agonist peptide KGCHM07 and the small-molecule agonist DPI-287 at 2.8 and 3.3 Å resolution, respectively. Our study identifies key determinants for agonist recognition, receptor activation, and DOP selectivity, revealing crucial differences between both agonist scaffolds. Our findings provide the first investigation into atomic-scale agonist binding at the DOP, supported by site-directed mutagenesis and pharmacological characterization. These structures will underpin the future structure-based development of DOP agonists for an improved pain treatment with fewer adverse effects.


Assuntos
Simulação de Acoplamento Molecular , Peptídeos/química , Receptores Opioides delta/agonistas , Receptores Opioides delta/química , Animais , Cristalografia por Raios X , Humanos , Domínios Proteicos , Receptores Opioides mu/agonistas , Receptores Opioides mu/química , Células Sf9 , Spodoptera
6.
Proc Natl Acad Sci U S A ; 116(44): 22353-22358, 2019 10 29.
Artigo em Inglês | MEDLINE | ID: mdl-31611414

RESUMO

An Australian estuarine isolate of Penicillium sp. MST-MF667 yielded 3 tetrapeptides named the bilaids with an unusual alternating LDLD chirality. Given their resemblance to known short peptide opioid agonists, we elucidated that they were weak (Ki low micromolar) µ-opioid agonists, which led to the design of bilorphin, a potent and selective µ-opioid receptor (MOPr) agonist (Ki 1.1 nM). In sharp contrast to all-natural product opioid peptides that efficaciously recruit ß-arrestin, bilorphin is G protein biased, weakly phosphorylating the MOPr and marginally recruiting ß-arrestin, with no receptor internalization. Importantly, bilorphin exhibits a similar G protein bias to oliceridine, a small nonpeptide with improved overdose safety. Molecular dynamics simulations of bilorphin and the strongly arrestin-biased endomorphin-2 with the MOPr indicate distinct receptor interactions and receptor conformations that could underlie their large differences in bias. Whereas bilorphin is systemically inactive, a glycosylated analog, bilactorphin, is orally active with similar in vivo potency to morphine. Bilorphin is both a unique molecular tool that enhances understanding of MOPr biased signaling and a promising lead in the development of next generation analgesics.


Assuntos
Analgésicos Opioides/farmacologia , Proteínas Fúngicas/farmacologia , Oligopeptídeos/farmacologia , Penicillium/química , Receptores Opioides mu/agonistas , Analgésicos Opioides/química , Animais , Sítios de Ligação , Linhagem Celular Tumoral , Proteínas Fúngicas/química , Células HEK293 , Humanos , Camundongos , Simulação de Acoplamento Molecular , Oligopeptídeos/química , Ligação Proteica , Receptores Opioides mu/química , Receptores Opioides mu/metabolismo
7.
Pharmacol Res Perspect ; 7(5): e00511, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31463067

RESUMO

Opioids are powerful analgesics acting via the human µ-opiate receptor (hMOR). Opioid use is associated with adverse effects such as tolerance, addiction, respiratory depression, and constipation. Two synthetic opioids, AH-7921 and U-47700 that were developed in the 1970s but never marketed, have recently appeared on the illegal drug market and in forensic toxicology reports. These agents were initially characterized for their analgesic activity in rodents; however, their pharmacology at hMOR has not been delineated. Thus, we synthesized over 50 chemical analogs based on core AH-7921 and U-47700 structures to assess for their ability to couple to Gαi signaling and induce hMOR internalization. For both the AH-7921 and U-47700 analogs, the 3,4-dichlorobenzoyl substituents were the most potent with comparable EC50 values for inhibition of cAMP accumulation; 26.49 ± 11.2 nmol L-1 and 8.8 ± 4.9 nmol L-1, respectively. Despite similar potencies for Gαi coupling, these two compounds had strikingly different hMOR internalization efficacies: U-47700 (10 µmol L-1) induced ~25% hMOR internalization similar to DAMGO while AH-7921 (10 µmol L-1) induced ~5% hMOR internalization similar to morphine. In addition, the R, R enantiomer of U-47700 is significantly more potent than the S, S enantiomer at hMOR. In conclusion, these data suggest that U-47700 and AH-7921 analogs have high analgesic potential in humans, but with divergent receptor internalization profiles, suggesting that they may exhibit differences in clinical utility or abuse potential.


Assuntos
Analgésicos Opioides/síntese química , Etilenodiaminas/síntese química , Receptores Opioides mu/metabolismo , Analgésicos Opioides/química , Analgésicos Opioides/farmacologia , Linhagem Celular , AMP Cíclico/metabolismo , Etilenodiaminas/química , Etilenodiaminas/farmacologia , Humanos , Estrutura Molecular , Receptores Opioides mu/química
8.
J Med Chem ; 62(8): 4204-4217, 2019 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-30939014

RESUMO

σ1 and/or σ2 receptors play a crucial role in pathological conditions such as pain, neurodegenerative disorders, and cancer. A set of spirocyclic cyclohexanes with diverse O-heterocycles and amino moieties (general structure III) was prepared and pharmacologically evaluated. In structure-activity relationships studies, the σ1 receptor affinity and σ1:σ2 selectivity were correlated with the stereochemistry, the kind and substitution pattern of the O-heterocycle, and the substituents at the exocyclic amino moiety. cis-configured 2-benzopyran cis-11b bearing a methoxy group and a tertiary cyclohexylmethylamino moiety showed the highest σ1 affinity ( Ki = 1.9 nM) of this series of compounds. In a Ca2+ influx assay, cis-11b behaved as a σ1 antagonist. cis-11b reveals high selectivity over σ2 and opioid receptors. The interactions of the novel σ1 ligands were analyzed on the molecular level using the recently reported X-ray crystal structure of the σ1 receptor protein. The protonated amino moiety forms a persistent salt bridge with E172. The spiro[benzopyran-1,1'-cyclohexane] scaffold and the cyclohexylmethyl moiety occupy two hydrophobic pockets. Exchange of the N-cyclohexylmethyl moiety by a benzyl group led unexpectedly to potent and selective µ-opioid receptor ligands.


Assuntos
Benzopiranos/química , Compostos de Espiro/química , Benzopiranos/síntese química , Benzopiranos/metabolismo , Sítios de Ligação , Cristalografia por Raios X , Humanos , Interações Hidrofóbicas e Hidrofílicas , Ligantes , Simulação de Dinâmica Molecular , Estrutura Terciária de Proteína , Receptores Opioides mu/química , Receptores Opioides mu/metabolismo , Receptores sigma/química , Receptores sigma/metabolismo , Relação Estrutura-Atividade , Receptor Sigma-1
9.
Peptides ; 116: 63-67, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31014958

RESUMO

The current opioid crisis has created a tragic problem in medicine and society. Pain is the most ubiquitous and costly disease in society and yet all of our "treatments" have toxicities, especially for prolonged use. However, there are several alternatives that have been discovered in the past fifteen years that have been demonstrated in animals to have none of the toxicities of current drugs. Many of the compounds are multivalent and have novel biological activity profiles. Unfortunately, none of these have been in clinical trials in humans, perhaps because they were discovered in academic laboratories. A review of these novel chemicals are given in this paper.


Assuntos
Analgésicos Opioides/uso terapêutico , Dor/tratamento farmacológico , Peptídeos/uso terapêutico , Peptidomiméticos/uso terapêutico , Animais , Humanos , Ligantes , Peptídeos Opioides/química , Peptídeos Opioides/uso terapêutico , Dor/patologia , Manejo da Dor , Peptídeos/efeitos adversos , Peptidomiméticos/efeitos adversos , Receptores Opioides/química , Receptores Opioides/uso terapêutico , Receptores Opioides delta/química , Receptores Opioides delta/genética , Receptores Opioides mu/química , Receptores Opioides mu/genética
10.
J Mol Neurosci ; 65(4): 472-479, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-30033503

RESUMO

Single nucleotide polymorphism in OPRM1 gene is associated with hedonic and reinforcing consequences of opioids. Risk and protective alleles may vary in different populations. One hundred healthy controls and 100 opioids (predominantly heroin) addicts from Pakistani origin were genotyped for A118G (N40D) polymorphism in OPRM1. Structural and functional impact of the polymorphism on encoded protein was predicted by in silico analysis. Results show significant association between homozygous GG genotype and opioid addiction in Pakistani population (p value = 0.016). In silico analysis by SIFT (TI = 0.61), PolyPhen (PISC = 0.227), PANTHER (subPSEC = -1.7171), and SNP effect predicted this SNP benign for encoded protein. Superimposing wild-type and mutated proteins by MODELLER shows no change (RMSD = 0.1) in extracellular ligand binding domain of µ-opioid receptor. However, Haploreg and RegulomeDB predicted OPRM1 gene repression by chromatin condensation and increased binding affinity of RXRA transcription factor that may reduce protein translation and hence the number of available receptors to bind with drugs, which may trigger underlying mechanisms for opioids addiction. Thus, this study outlines causal relationship between opioids addiction and genetic predisposition in Pakistani population.


Assuntos
Transtornos Relacionados ao Uso de Opioides/genética , Polimorfismo de Nucleotídeo Único , Receptores Opioides mu/genética , Adulto , Estudos de Casos e Controles , Feminino , Humanos , Masculino , Mutação de Sentido Incorreto , Paquistão , Domínios Proteicos , Receptores Opioides mu/química
11.
J Med Chem ; 61(14): 6075-6086, 2018 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-29939746

RESUMO

A major limitation in the study of the mu-delta opioid receptor heterodimer (MDOR) is that few selective pharmacological tools exist and no heteromer-selective antagonists. We thus designed a series of variable-length (15-41 atoms) bivalent linked peptides with selective but moderate/low-affinity pharmacophores for the mu and delta opioid receptors. We observed a U-shaped MDOR potency/affinity profile in vitro, with the 24-atom spacer length (D24M) producing the highest MDOR potency/affinity (<1 nM) and selectivity (≥89-fold). We further evaluated D24M in mice and observed that D24M dose-dependently antagonized tail flick antinociception produced by the MDOR agonists CYM51010 and Deltorphin-II, without antagonizing the monomer agonists DAMGO and DSLET. We also observed that D24M sharply reduced withdrawal behavior in models of acute and chronic morphine dependence. These findings suggest that D24M is a first-in-class high-potency MDOR-selective antagonist both in vitro and in vivo.


Assuntos
Morfina/farmacologia , Peptídeos/farmacologia , Multimerização Proteica/efeitos dos fármacos , Receptores Opioides delta/química , Receptores Opioides mu/química , Síndrome de Abstinência a Substâncias/tratamento farmacológico , Animais , Células CHO , Técnicas de Química Sintética , Cricetulus , Relação Dose-Resposta a Droga , Interações Hidrofóbicas e Hidrofílicas , Camundongos , Peptídeos/síntese química , Peptídeos/química , Peptídeos/uso terapêutico , Estrutura Quaternária de Proteína
12.
Mol Pharmacol ; 93(2): 157-167, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29233847

RESUMO

Allosteric modulators of G protein-coupled receptors, including opioid receptors, have been proposed as possible therapeutic agents with enhanced selectivity. BMS-986122 is a positive allosteric modulator (PAM) of the µ-opioid receptor (µ-OR). BMS-986187 is a structurally distinct PAM for the δ-opioid receptor (δ-OR) that has been reported to exhibit 100-fold selectivity in promoting δ-OR over µ-OR agonism. We used ligand binding and second-messenger assays to show that BMS-986187 is an effective PAM at the µ-OR and at the κ-opioid receptor (κ-OR), but it is ineffective at the nociceptin receptor. The affinity of BMS-986187 for δ-ORs and κ-ORs is approximately 20- to 30-fold higher than for µ-ORs, determined using an allosteric ternary complex model. Moreover, we provide evidence, using a silent allosteric modulator as an allosteric antagonist, that BMS-986187 and BMS-986122 bind to a similar region on all three traditional opioid receptor types (µ-OR, δ-OR, and κ-OR). In contrast to the dogma surrounding allosteric modulators, the results indicate a possible conserved allosteric binding site across the opioid receptor family that can accommodate structurally diverse molecules. These findings have implications for the development of selective allosteric modulators.


Assuntos
Receptores Opioides delta/metabolismo , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/metabolismo , Regulação Alostérica/efeitos dos fármacos , Sítio Alostérico , Animais , Células CHO , Linhagem Celular Tumoral , Cricetulus , Células HEK293 , Humanos , Antagonistas de Entorpecentes/farmacologia , Ensaio Radioligante , Ratos , Receptores Opioides delta/química , Receptores Opioides delta/efeitos dos fármacos , Receptores Opioides kappa/química , Receptores Opioides kappa/efeitos dos fármacos , Receptores Opioides mu/química , Receptores Opioides mu/efeitos dos fármacos , Sódio/metabolismo , Sulfonas/farmacologia , Xantonas/farmacologia
13.
Mol Pharmacol ; 93(2): 36-48, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29237725

RESUMO

The µ-opioid receptor (MOR) is a Gi/o protein-coupled receptor that mediates analgesic, euphoric, and reward effects. Using a bacterial two-hybrid screen, we reported that the carboxyl tail of the rat MOR associates with A20-binding inhibitor of nuclear factor κB (ABIN-1). This interaction was confirmed by direct protein-protein binding and coimmunoprecipitation of MOR and ABIN-1 proteins in cell lysates. Saturation binding studies showed that ABIN-1 had no effect on MOR binding. However, the interaction of ABIN-1 and MOR inhibited the activation of G proteins induced by DAMGO ([d-Ala2,N-Me-Phe4,Gly5-ol]-Enkephalin). MOR phosphorylation, ubiquitination, and internalization induced by DAMGO were decreased in Chinese hamster ovary cells that coexpressed MOR and ABIN-1. The suppression of forskolin-stimulated adenylyl cyclase by DAMGO was also inhibited by the interaction of ABIN-1 with MOR. In addition, extracellular signal-regulated kinase activation was also negatively regulated by overexpression of ABIN-1. These data suggest that ABIN-1 is a negative coregulator of MOR activation, phosphorylation, and internalization in vitro. ABIN-1 also inhibited morphine-induced hyperlocomotion in zebrafish larvae (AB strain). By utilization of an antisense morpholino oligonucleotide (MO) gene knockdown technology, the ABIN-1 MO-injected zebrafish larvae showed a significant increase (approximately 60%) in distance moved compared with control MO-injected larvae after acute morphine treatment (P < 0.01). Taken together, ABIN-1 negatively regulates MOR function in vitro and in vivo.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Receptores Opioides mu/fisiologia , Analgésicos Opioides/farmacologia , Animais , Sítios de Ligação , Células CHO , Linhagem Celular Tumoral , Cricetulus , AMP Cíclico/metabolismo , Proteínas de Ligação a DNA/metabolismo , Endocitose , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Humanos , Larva , Ligantes , Proteínas Luminescentes , Fosforilação , Ratos , Receptores Opioides mu/agonistas , Receptores Opioides mu/química , Receptores Opioides mu/metabolismo , Transdução de Sinais , Radioisótopos de Enxofre/metabolismo , Ubiquitinação , Peixe-Zebra/crescimento & desenvolvimento
14.
Trends Pharmacol Sci ; 38(11): 1016-1037, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28935293

RESUMO

Pain is both a major clinical and economic problem, affecting more people than diabetes, heart disease, and cancer combined. While a variety of prescribed or over-the-counter (OTC) medications are available for pain management, opioid medications, especially those acting on the µ-opioid receptor (µOR) and related pathways, have proven to be the most effective, despite some serious side effects including respiration depression, pruritus, dependence, and constipation. It is therefore imperative that both academia and industry develop novel µOR analgesics which retain their opioid analgesic properties but with fewer or no adverse effects. In this review we outline recent progress towards the discovery of safer opioid analgesics.


Assuntos
Analgésicos Opioides/química , Analgésicos Opioides/farmacologia , Receptores Opioides mu/antagonistas & inibidores , Animais , Desenho de Fármacos , Humanos , Receptores Opioides mu/química , Receptores Opioides mu/metabolismo
15.
Science ; 355(6328): 966-969, 2017 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-28254944

RESUMO

Indiscriminate activation of opioid receptors provides pain relief but also severe central and intestinal side effects. We hypothesized that exploiting pathological (rather than physiological) conformation dynamics of opioid receptor-ligand interactions might yield ligands without adverse actions. By computer simulations at low pH, a hallmark of injured tissue, we designed an agonist that, because of its low acid dissociation constant, selectively activates peripheral µ-opioid receptors at the source of pain generation. Unlike the conventional opioid fentanyl, this agonist showed pH-sensitive binding, heterotrimeric guanine nucleotide-binding protein (G protein) subunit dissociation by fluorescence resonance energy transfer, and adenosine 3',5'-monophosphate inhibition in vitro. It produced injury-restricted analgesia in rats with different types of inflammatory pain without exhibiting respiratory depression, sedation, constipation, or addiction potential.


Assuntos
Dor Aguda/tratamento farmacológico , Analgésicos Opioides/química , Analgésicos Opioides/farmacologia , Desenho de Fármacos , Fentanila/análogos & derivados , Piperidinas/química , Piperidinas/farmacologia , Receptores Opioides mu/agonistas , Receptores Opioides mu/química , Monofosfato de Adenosina/antagonistas & inibidores , Analgesia , Analgésicos Opioides/efeitos adversos , Animais , Simulação por Computador , Constipação Intestinal/induzido quimicamente , Fentanila/efeitos adversos , Transferência Ressonante de Energia de Fluorescência , Subunidades alfa de Proteínas de Ligação ao GTP/metabolismo , Células HEK293 , Humanos , Concentração de Íons de Hidrogênio , Ligantes , Modelos Moleculares , Manejo da Dor , Piperidinas/efeitos adversos , Ligação Proteica , Conformação Proteica , Ratos , Receptores Opioides mu/genética , Insuficiência Respiratória/induzido quimicamente , Transfecção
16.
Neuropharmacology ; 118: 46-58, 2017 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-28283391

RESUMO

The development of G protein-biased agonists for the µ-opioid receptor (MOR) offers a clear drug discovery rationale for improved analgesia and reduced side-effects of opiate pharmacotherapy. However, our understanding of the molecular mechanisms governing ligand bias is limited, which hinders our ability to rationally design biased compounds. We have investigated the role of MOR binding site residues W320 and Y328 in controlling bias, by receptor mutagenesis. The pharmacology of a panel of ligands in a cAMP and a ß-arrestin2 assay were compared between the wildtype and mutated receptors, with bias factors calculated by operational analysis using ΔΔlog(τ/KA) values. [3H]diprenorphine competition binding was used to estimate affinity changes. Introducing the mutations W320A and Y328F caused changes in pathway bias, with different patterns of change between ligands. For example, DAMGO increased relative ß-arrestin2 activity at the W320A mutant, whilst its ß-arrestin2 response was completely lost at Y328F. In contrast, endomorphin-1 gained activity with Y328F but lost activity at W320A, in both pathways. For endomorphin-2 there was a directional shift from cAMP bias at the wildtype towards more ß-arrestin2 bias at W320A. We also observe clear uncoupling between mutation-driven changes in function and binding affinity. These findings suggest that the mutations influenced the balance of pathway activation in a ligand-specific manner, thus identifying residues in the MOR binding pocket that govern ligand bias. This increases our understanding of how ligand/receptor binding interactions can be translated into agonist-specific pathway activation.


Assuntos
Mutação/genética , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo , Triptofano/genética , Tirosina/genética , Analgésicos Opioides/farmacologia , Sítios de Ligação/efeitos dos fármacos , Sítios de Ligação/genética , AMP Cíclico/metabolismo , Diprenorfina/farmacocinética , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Quinase 2 de Receptor Acoplado a Proteína G/genética , Quinase 2 de Receptor Acoplado a Proteína G/metabolismo , Células HEK293 , Humanos , Modelos Moleculares , Mutagênese , Antagonistas de Entorpecentes/farmacocinética , Oligopeptídeos/farmacologia , Receptores Opioides mu/química , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Transfecção , Trítio/farmacocinética , Triptofano/metabolismo , Tirosina/metabolismo , beta-Arrestinas/metabolismo
17.
PLoS One ; 11(6): e0156897, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27272042

RESUMO

INTRODUCTION: Opioid receptors are currently classified as Mu (µ), Delta (δ), Kappa (κ) plus the opioid related nociceptin/orphanin FQ (N/OFQ) peptide receptor (NOP). Despite compelling evidence for interactions and benefits of targeting more than one receptor type in producing analgesia, clinical ligands are Mu agonists. In this study we have designed a Mu-NOP agonist named DeNo. The Mu agonist component is provided by dermorphin, a peptide isolated from the skin of Phyllomedusa frogs and the NOP component by the endogenous agonist N/OFQ. METHODS: We have assessed receptor binding profile of DeNo and compared with dermorphin and N/OFQ. In a series of functional screens we have assessed the ability to (i) increase Ca2+ in cells coexpressing recombinant receptors and a the chimeric protein Gαqi5, (ii) stimulate the binding of GTPγ[35S], (iii) inhibit cAMP formation, (iv) activate MAPKinase, (v) stimulate receptor-G protein and arrestin interaction using BRET, (vi) electrically stimulated guinea pig ileum (gpI) assay and (vii) ability to produce analgesia via the intrathecal route in rats. RESULTS: DeNo bound to Mu (pKi; 9.55) and NOP (pKi; 10.22) and with reasonable selectivity. This translated to increased Ca2+ in Gαqi5 expressing cells (pEC50 Mu 7.17; NOP 9.69), increased binding of GTPγ[35S] (pEC50 Mu 7.70; NOP 9.50) and receptor-G protein interaction in BRET (pEC50 Mu 8.01; NOP 9.02). cAMP formation was inhibited and arrestin was activated (pEC50 Mu 6.36; NOP 8.19). For MAPK DeNo activated p38 and ERK1/2 at Mu but only ERK1/2 at NOP. In the gpI DeNO inhibited electrically-evoked contractions (pEC50 8.63) that was sensitive to both Mu and NOP antagonists. DeNo was antinociceptive in rats. CONCLUSION: Collectively these data validate the strategy used to create a novel bivalent Mu-NOP peptide agonist by combining dermorphin (Mu) and N/OFQ (NOP). This molecule behaves essentially as the parent compounds in vitro. In the antonocicoeptive assays employed in this study DeNo displays only weak antinociceptive properties.


Assuntos
Peptídeos Opioides/química , Peptídeos/síntese química , Receptores Opioides mu/agonistas , Receptores Opioides/agonistas , Animais , Células CHO , Cálcio/metabolismo , Cricetulus , Cobaias , Células HEK293 , Humanos , Masculino , Peptídeos/química , Peptídeos/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores Opioides/química , Receptores Opioides/metabolismo , Receptores Opioides mu/química , Receptores Opioides mu/metabolismo , Receptor de Nociceptina
18.
Chem Biol Interact ; 254: 54-62, 2016 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-27231091

RESUMO

The anti-hyperalgesic effect of the complex containing α-terpineol (αTPN) and ß-cyclodextrin (ßCD) was analyzed in a non-inflammatory chronic muscle pain model, as well as its mechanism of action through docking study for a possible interaction with receptors. The αTPN-ßCD complex was prepared and characterized through the thermogravimetry/derivate thermogravimetry (TG/DTG), Fourier transform infrared spectroscopy (FTIR) and scanning electron microscope (SEM). The model of chronic muscle pain was induced by two injections of pH 4.0 saline (20 µl) into the left gastrocnemius 5 days apart. After confirming hyperalgesia, male mice were treated with αTPN-ßCD (25, 50 or 100 mg/kg; p.o.) or vehicle (saline 0.9%, p.o.) daily for 10 days. 1 h after the mechanical hyperalgesia, motor performance was evaluated. In addition, the systemic administration of naloxone and ondansetron tested the analgesic effect on the active opioid and serotonin receptors, respectively. The characterization tests indicated that αTPN was efficiently incorporated into ßCD. The oral treatment with αTPN-ßCD, at all doses tested, produced a significant (p < 0.001) decrease in the mechanical hyperalgesia, without causing any alteration in the force and in motor performance. This analgesic effect was reversed by the systemic administration of naloxone or ondansetron. These findings are corroborated by the docking study described in the present study, which verified a possible interaction of αTPN-ßCD with opioid (MU, Kappa, Delta) and 5-HT receptors. Thus, it can be concluded that αTPN-ßCD reduced the hyperalgesia followed by the chronic muscle pain model, probably evoked by the descending inhibitory pain system, specifically by opioid and serotoninergic receptors.


Assuntos
Cicloexenos/química , Cicloexenos/farmacologia , Monoterpenos/química , Monoterpenos/farmacologia , Receptores Opioides delta/química , Receptores Opioides kappa/química , Receptores Opioides mu/química , beta-Ciclodextrinas/química , Analgésicos/farmacologia , Animais , Comportamento Animal/efeitos dos fármacos , Sítios de Ligação , Monoterpenos Cicloexânicos , Cicloexenos/uso terapêutico , Modelos Animais de Doenças , Fibromialgia/tratamento farmacológico , Fibromialgia/metabolismo , Fibromialgia/patologia , Hiperalgesia/induzido quimicamente , Hiperalgesia/tratamento farmacológico , Hiperalgesia/patologia , Masculino , Camundongos , Simulação de Acoplamento Molecular , Monoterpenos/uso terapêutico , Naloxona/farmacologia , Ondansetron/farmacologia , Estrutura Terciária de Proteína , Receptores Opioides delta/metabolismo , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/metabolismo
19.
Sci Rep ; 5: 18198, 2015 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-26657998

RESUMO

The primary molecular target for clinically used opioids is the µ-opioid receptor (MOR). Besides the major seven-transmembrane (7TM) receptors, the MOR gene codes for alternatively spliced six-transmembrane (6TM) isoforms, the biological and clinical significance of which remains unclear. Here, we show that the otherwise exclusively intracellular localized 6TM-MOR translocates to the plasma membrane upon coexpression with ß2-adrenergic receptors (ß2-ARs) through an interaction with the fifth and sixth helices of ß2-AR. Coexpression of the two receptors in BE(2)-C neuroblastoma cells potentiates calcium responses to a 6TM-MOR ligand, and this calcium response is completely blocked by a selective ß2-antagonist in BE(2)-C cells, and in trigeminal and dorsal root ganglia. Co-administration of 6TM-MOR and ß2-AR ligands leads to substantial analgesic synergy and completely reverses opioid-induced hyperalgesia in rodent behavioral models. Together, our results provide evidence that the heterodimerization of 6TM-MOR with ß2-AR underlies a molecular mechanism for 6TM cellular signaling, presenting a unique functional responses to opioids. This signaling pathway may contribute to the hyperalgesic effects of opioids that can be efficiently blocked by ß2-AR antagonists, providing a new avenue for opioid therapy.


Assuntos
Analgésicos Opioides/metabolismo , Receptores Adrenérgicos beta 2/química , Receptores Adrenérgicos beta 2/metabolismo , Receptores Opioides mu/química , Receptores Opioides mu/metabolismo , Transdução de Sinais , Animais , Cálcio/metabolismo , Gânglios/metabolismo , Expressão Gênica , Humanos , Ligantes , Camundongos , Modelos Moleculares , Conformação Molecular , Neurônios/metabolismo , Ligação Proteica , Receptores Opioides mu/genética , Relação Estrutura-Atividade
20.
J Proteome Res ; 14(8): 3162-73, 2015 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-26090583

RESUMO

Human µ-opioid receptor (hMOR) is a class-A G-protein-coupled receptor (GPCR), a prime therapeutic target for the management of moderate and severe pain. A chimeric form of the receptor has been cocrystallized with an opioid antagonist and resolved by X-ray diffraction; however, further direct structural analysis is still required to identify the active form of the receptor to facilitate the rational design of hMOR-selective agonist and antagonists with therapeutic potential. Toward this goal and in spite of the intrinsic difficulties posed by the highly hydrophobic transmembrane motives of hMOR, we have comprehensively characterized by mass spectrometry (MS) analysis the primary sequence of the functional hMOR. Recombinant hMOR was overexpressed as a C-terminal c-myc and 6-his tagged protein using an optimized expression procedure in Pichia pastoris cells. After membrane solubilization and metal-affinity chromatography purification, a procedure was devised to enhance the concentration of the receptor. Subsequent combinations of in-solution and in-gel digestions using either trypsin, chymotrypsin, or proteinase K, followed by matrix-assisted laser desorption ionization time-of-flight MS or nanoliquid chromatography coupled with tandem MS analyses afforded an overall sequence coverage of up to >80%, a level of description first attained for an opioid receptor and one of the six such high-coverage MS-based analyses of any GPCR.


Assuntos
Cromatografia Líquida/métodos , Receptores Opioides mu/química , Proteínas Recombinantes de Fusão/química , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz/métodos , Espectrometria de Massas em Tandem/métodos , Sequência de Aminoácidos , Western Blotting , Quimotripsina/metabolismo , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Peptídeos/metabolismo , Pichia/genética , Estrutura Secundária de Proteína , Proteômica/instrumentação , Proteômica/métodos , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Tripsina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA