Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 423
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Nature ; 613(7945): 767-774, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36450356

RESUMO

Mu-opioid receptor (µOR) agonists such as fentanyl have long been used for pain management, but are considered a major public health concern owing to their adverse side effects, including lethal overdose1. Here, in an effort to design safer therapeutic agents, we report an approach targeting a conserved sodium ion-binding site2 found in µOR3 and many other class A G-protein-coupled receptors with bitopic fentanyl derivatives that are functionalized via a linker with a positively charged guanidino group. Cryo-electron microscopy structures of the most potent bitopic ligands in complex with µOR highlight the key interactions between the guanidine of the ligands and the key Asp2.50 residue in the Na+ site. Two bitopics (C5 and C6 guano) maintain nanomolar potency and high efficacy at Gi subtypes and show strongly reduced arrestin recruitment-one (C6 guano) also shows the lowest Gz efficacy among the panel of µOR agonists, including partial and biased morphinan and fentanyl analogues. In mice, C6 guano displayed µOR-dependent antinociception with attenuated adverse effects, supporting the µOR sodium ion-binding site as a potential target for the design of safer analgesics. In general, our study suggests that bitopic ligands that engage the sodium ion-binding pocket in class A G-protein-coupled receptors can be designed to control their efficacy and functional selectivity profiles for Gi, Go and Gz subtypes and arrestins, thus modulating their in vivo pharmacology.


Assuntos
Desenho de Fármacos , Fentanila , Morfinanos , Receptores Opioides mu , Animais , Camundongos , Analgésicos Opioides/química , Analgésicos Opioides/metabolismo , Arrestinas/metabolismo , Microscopia Crioeletrônica , Fentanila/análogos & derivados , Fentanila/química , Fentanila/metabolismo , Ligantes , Morfinanos/química , Morfinanos/metabolismo , Receptores Opioides mu/agonistas , Receptores Opioides mu/química , Receptores Opioides mu/metabolismo , Receptores Opioides mu/ultraestrutura , Sítios de Ligação , Nociceptividade
2.
Nature ; 558(7711): 547-552, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29899455

RESUMO

The µ-opioid receptor (µOR) is a G-protein-coupled receptor (GPCR) and the target of most clinically and recreationally used opioids. The induced positive effects of analgesia and euphoria are mediated by µOR signalling through the adenylyl cyclase-inhibiting heterotrimeric G protein Gi. Here we present the 3.5 Å resolution cryo-electron microscopy structure of the µOR bound to the agonist peptide DAMGO and nucleotide-free Gi. DAMGO occupies the morphinan ligand pocket, with its N terminus interacting with conserved receptor residues and its C terminus engaging regions important for opioid-ligand selectivity. Comparison of the µOR-Gi complex to previously determined structures of other GPCRs bound to the stimulatory G protein Gs reveals differences in the position of transmembrane receptor helix 6 and in the interactions between the G protein α-subunit and the receptor core. Together, these results shed light on the structural features that contribute to the Gi protein-coupling specificity of the µOR.


Assuntos
Microscopia Crioeletrônica , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/ultraestrutura , Receptores Opioides mu/metabolismo , Receptores Opioides mu/ultraestrutura , Animais , Sítios de Ligação , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Feminino , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/química , Subunidades alfa Gs de Proteínas de Ligação ao GTP/química , Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Humanos , Ligantes , Camundongos , Camundongos Endogâmicos BALB C , Simulação de Dinâmica Molecular , Morfinanos/química , Morfinanos/metabolismo , Estabilidade Proteica/efeitos dos fármacos , Receptores Adrenérgicos beta 2/química , Receptores Adrenérgicos beta 2/metabolismo , Receptores Opioides mu/agonistas , Receptores Opioides mu/química , Especificidade por Substrato
3.
J Sep Sci ; 47(19): e202400486, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39375918

RESUMO

Sinomenium acutum (Thunb.) Rehd. et Wils is widely used in the treatment of rheumatoid arthritis, with its alkaloid compound sinomenine (SIN) being renowned for its significant anti-inflammatory properties. However, despite its widespread application, the in vivo anti-inflammatory mechanisms and metabolic pathways of SIN remain incompletely understood. This study established a rapid and reliable method based on an ultra-high-performance liquid chromatography method coupled with Quadrupole-Exactive Orbitrap mass spectrometry and molecular docking to identify and characterize SIN and 69 metabolites in rat plasma, urine, and feces, revealing primary metabolic pathways of hydroxylation, demethylation, sulfation, and glucuronidation. Molecular docking results revealed that phase I reactions, including dedimethylation, demethylation, dehydrogenation, and dihydroxylation, along with their composite reactions, were pivotal in influencing SIN's in vivo anti-inflammatory activity. M28, M36, and M59 are potentially the most anti-inflammatory active metabolites of SIN in vivo. This comprehensive analysis unveils SIN's metabolic pathways, offering insights into its biological processes and suggesting a novel approach for exploring active drug constituents. These findings pave the way for further understanding SIN's anti-inflammatory mechanisms, contributing significantly to the development of new therapeutic strategies.


Assuntos
Espectrometria de Massas , Simulação de Acoplamento Molecular , Morfinanos , Animais , Morfinanos/metabolismo , Morfinanos/química , Cromatografia Líquida de Alta Pressão/métodos , Ratos , Masculino , Ratos Sprague-Dawley , Anti-Inflamatórios/química , Anti-Inflamatórios/metabolismo , Sinomenium/química , Estrutura Molecular
4.
Nature ; 524(7565): 375-8, 2015 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-26245377

RESUMO

µ-Opioid receptors (µORs) are G-protein-coupled receptors that are activated by a structurally diverse spectrum of natural and synthetic agonists including endogenous endorphin peptides, morphine and methadone. The recent structures of the µOR in inactive and agonist-induced active states (Huang et al., ref. 2) provide snapshots of the receptor at the beginning and end of a signalling event, but little is known about the dynamic sequence of events that span these two states. Here we use solution-state NMR to examine the process of µOR activation using a purified receptor (mouse sequence) preparation in an amphiphile membrane-like environment. We obtain spectra of the µOR in the absence of ligand, and in the presence of the high-affinity agonist BU72 alone, or with BU72 and a G protein mimetic nanobody. Our results show that conformational changes in transmembrane segments 5 and 6 (TM5 and TM6), which are required for the full engagement of a G protein, are almost completely dependent on the presence of both the agonist and the G protein mimetic nanobody, revealing a weak allosteric coupling between the agonist-binding pocket and the G-protein-coupling interface (TM5 and TM6), similar to that observed for the ß2-adrenergic receptor. Unexpectedly, in the presence of agonist alone, we find larger spectral changes involving intracellular loop 1 and helix 8 compared to changes in TM5 and TM6. These results suggest that one or both of these domains may play a role in the initial interaction with the G protein, and that TM5 and TM6 are only engaged later in the process of complex formation. The initial interactions between the G protein and intracellular loop 1 and/or helix 8 may be involved in G-protein coupling specificity, as has been suggested for other family A G-protein-coupled receptors.


Assuntos
Receptores Opioides mu/química , Receptores Opioides mu/metabolismo , Regulação Alostérica , Animais , Sítios de Ligação , Proteínas Heterotriméricas de Ligação ao GTP/metabolismo , Lisina/metabolismo , Camundongos , Modelos Moleculares , Morfinanos/química , Morfinanos/metabolismo , Morfinanos/farmacologia , Ressonância Magnética Nuclear Biomolecular , Ligação Proteica , Conformação Proteica/efeitos dos fármacos , Pirróis/química , Pirróis/metabolismo , Pirróis/farmacologia , Receptores Adrenérgicos beta 2/química , Anticorpos de Cadeia Única/química , Anticorpos de Cadeia Única/metabolismo , Anticorpos de Cadeia Única/farmacologia , Relação Estrutura-Atividade , Especificidade por Substrato
5.
Nature ; 524(7565): 315-21, 2015 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-26245379

RESUMO

Activation of the µ-opioid receptor (µOR) is responsible for the efficacy of the most effective analgesics. To shed light on the structural basis for µOR activation, here we report a 2.1 Å X-ray crystal structure of the murine µOR bound to the morphinan agonist BU72 and a G protein mimetic camelid antibody fragment. The BU72-stabilized changes in the µOR binding pocket are subtle and differ from those observed for agonist-bound structures of the ß2-adrenergic receptor (ß2AR) and the M2 muscarinic receptor. Comparison with active ß2AR reveals a common rearrangement in the packing of three conserved amino acids in the core of the µOR, and molecular dynamics simulations illustrate how the ligand-binding pocket is conformationally linked to this conserved triad. Additionally, an extensive polar network between the ligand-binding pocket and the cytoplasmic domains appears to play a similar role in signal propagation for all three G-protein-coupled receptors.


Assuntos
Receptores Opioides mu/química , Receptores Opioides mu/metabolismo , Regulação Alostérica , Animais , Sítios de Ligação , Cristalografia por Raios X , Proteínas Heterotriméricas de Ligação ao GTP/química , Proteínas Heterotriméricas de Ligação ao GTP/metabolismo , Camundongos , Modelos Moleculares , Simulação de Dinâmica Molecular , Morfinanos/química , Morfinanos/metabolismo , Morfinanos/farmacologia , Estabilidade Proteica/efeitos dos fármacos , Estrutura Terciária de Proteína , Pirróis/química , Pirróis/metabolismo , Pirróis/farmacologia , Receptor Muscarínico M2/química , Receptores Adrenérgicos beta 2/química , Receptores Opioides mu/agonistas , Anticorpos de Cadeia Única/química , Anticorpos de Cadeia Única/farmacologia , Relação Estrutura-Atividade
6.
Drug Metab Dispos ; 48(2): 106-115, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31727673

RESUMO

Oxycodone is used as a potent analgesic medication. Oxycodone is extensively metabolized. To fully describe its metabolism, the oxygenation of oxycodone to oxycodone N-oxide was investigated in hepatic preparations. The hypothesis tested was that oxycodone N-oxygenation was enzymatic and the amount of N-oxide detected was a consequence of both oxygenation and retro-reduction. Methods for testing the hypothesis included both in vitro and in vivo studies. Results indicated that oxycodone was N-oxygenated by the flavin-containing monooxygenase. Oxycodone N-oxide is chemically quite stable but in the presence of hepatic preparations and NADPH was retro-reduced to its parent compound oxycodone. Subsequently, oxycodone was metabolized to other metabolites including noroxycodone, noroxymorphone, and oxymorphone via cytochrome P-450. Retro-reduction of oxycodone N-oxide to oxycodone was facilitated by quinone reductase, aldehyde oxidase, and hemoglobin but not to a great extent by cytochrome P-450 or the flavin-containing monooxygenase. To confirm the in vitro observations, oxycodone was administered to rats and humans. In good agreement with in vitro results, substantial oxycodone N-oxide was observed in urine after oxycodone administration to rats and humans. Administration of oxycodone N-oxide to rats showed substantial amount of recovered oxycodone N-oxide. In vivo, noroxycodone was formed as a major rat urinary metabolite from oxycodone N-oxide presumably after retro-reduction to oxycodone and oxidative N-demethylation. To a lesser extent, oxycodone, noroxymorphone, and oxymorphone were observed as urinary metabolites. SIGNIFICANCE STATEMENT: This manuscript describes the N-oxygenation of oxycodone in vitro as well as in small animals and humans. A new metabolite was quantified as oxycodone N-oxide. Oxycodone N-oxide undergoes extensive retro-reduction to oxycodone. This re-establishes the metabolic profile of oxycodone and introduces new concepts about a metabolic futile cycle related to oxycodone metabolism.


Assuntos
Óxidos/metabolismo , Oxicodona/metabolismo , Analgésicos Opioides/metabolismo , Animais , Sistema Enzimático do Citocromo P-450/metabolismo , Feminino , Hemoglobinas/metabolismo , Humanos , Masculino , Oxigenases de Função Mista/metabolismo , Morfinanos/metabolismo , NADP/metabolismo , Oximorfona/metabolismo , Ratos
7.
Behav Pharmacol ; 31(2&3): 174-178, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32168026

RESUMO

Pain management is a challenging and unmet medical need. Despite their demonstrated efficacy, currently used opioid drugs and nonsteroidal anti-inflammatory drugs are frequently associated with several adverse events. The identification of new and safe analgesics is therefore needed. MP1104, an analogue of 3'-iodobenzoyl naltrexamine, is a potent nonselective full agonist at mu (MOR), kappa (KOR), and delta (DOR) opioid receptors, respectively. It was shown to possess potent antinociceptive effects in acute thermal pain assays without aversion in mice. In this study, we investigated MP1104 in the formalin test, a model of tonic pain. MP1104 (0.05, 0.1, and 1.0 mg/kg) reduced pain-like behaviors in phases I and II of the formalin test in male and female ICR mice. Pretreatment with KOR antagonist (norbinaltorphimine 10 mg/kg) and DOR antagonist (naltrindole 10 mg/kg) abolished the antinociceptive effects of MP1104 in the formalin test. These findings support the development of MP1104 for further testing in other pain models.


Assuntos
Morfinanos/farmacologia , Receptores Opioides delta/metabolismo , Receptores Opioides kappa/metabolismo , Analgésicos/farmacologia , Analgésicos Opioides/farmacologia , Animais , Feminino , Masculino , Camundongos , Camundongos Endogâmicos ICR , Morfinanos/metabolismo , Antagonistas de Entorpecentes/farmacologia , Dor/tratamento farmacológico , Manejo da Dor/métodos , Medição da Dor/efeitos dos fármacos , Receptores Opioides/agonistas , Receptores Opioides delta/agonistas , Receptores Opioides kappa/agonistas
8.
Behav Pharmacol ; 31(8): 792-797, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32804774

RESUMO

Mu-opioid receptor (MOR) agonists are highly efficacious for the treatment of pain but have significant abuse liability. Recently, we reported that nalfurafine, when combined with oxycodone at a certain ratio, reduced the reinforcing effects of oxycodone in rats while producing additive antinociceptive effects. Questions remain, however, including if the combination will function as a reinforcer in drug-naïve rats, and if the combination produces aversive effects that could explain nalfurafine's ability to reduce oxycodone self-administration? In the present study, we investigated nalfurafine's ability to reduce acquisition of oxycodone self-administration when the two were self-administered as a mixture in drug-naïve rats and nalfurafine's ability to attenuate a conditioned place preference (CPP) induced by oxycodone. In the self-administration study, male Sprague-Dawley rats self-administered intravenous injections of oxycodone (0.056 mg/kg/injection), an oxycodone/nalfurafine combination (0.056/0.0032 mg/kg/injection), or saline under fixed-ratio schedules of reinforcement for 20 days to compare rates of acquisition of drug taking. In the CPP assay, male Sprague-Dawley rats received subcutaneous injections of either saline, oxycodone (3.2 mg/kg), nalfurafine (0.18 mg/kg), or an oxycodone/nalfurafine combination at the same ratio used in the self-administration study (3.2 mg/kg/0.18 mg/kg). All subjects self-administering oxycodone alone met acquisition criteria. However, only 13% of subjects self-administering oxycodone/nalfurafine met criteria, and no subjects acquired self-administration of saline. Oxycodone, but not nalfurafine alone or the oxycodone/nalfurafine combination, produced rewarding effects in rats in the CPP test. These findings suggest that the combination of oxycodone and nalfurafine will be less habit forming in opioid-naïve patients than oxycodone alone.


Assuntos
Morfinanos/farmacologia , Transtornos Relacionados ao Uso de Opioides/tratamento farmacológico , Receptores Opioides kappa/agonistas , Compostos de Espiro/farmacologia , Analgésicos Opioides/farmacologia , Animais , Condicionamento Clássico/efeitos dos fármacos , Relação Dose-Resposta a Droga , Masculino , Morfinanos/metabolismo , Transtornos Relacionados ao Uso de Opioides/prevenção & controle , Oxicodona/efeitos adversos , Oxicodona/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores Opioides/agonistas , Receptores Opioides/metabolismo , Receptores Opioides kappa/metabolismo , Reforço Psicológico , Recompensa , Autoadministração , Compostos de Espiro/metabolismo , Transtornos Relacionados ao Uso de Substâncias/tratamento farmacológico , Transtornos Relacionados ao Uso de Substâncias/prevenção & controle
9.
Bioorg Med Chem Lett ; 30(3): 126893, 2020 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-31879208

RESUMO

The morphinan-type orexin 1 receptor (OX1R) antagonists such as YNT-707 (2) and YNT-1310 (3) show potent and extremely high selective antagonistic activity against OX1R. In the course of our studies of the essential structure of 2, we identified new scaffolds by simplification of the morphinan skeleton. However, the new chemical entities carrying the D-ring removed scaffold showed insufficient activity. To improve the activity of these derivatives, we investigated the effect of substituents mainly focused on the 17-nitrogen group. The 17-N-substituted derivatives, as well as the cyclic derivatives, were synthesized and examined the OX1R antagonistic activity. The assay results showed the interesting relationship between the OX1R antagonistic activity and the substituents on the 17-nitrogen: the antagonistic activity was increased as the bulkiness of 17-substituents increased. Finally, the 17-N-Boc derivative 14a showed the most potent OX1R antagonistic activity (Ki = 14.8 nM).


Assuntos
Morfinanos/química , Antagonistas dos Receptores de Orexina/química , Receptores de Orexina/química , Sulfonamidas/química , Aminas/química , Humanos , Cinética , Morfinanos/metabolismo , Antagonistas dos Receptores de Orexina/síntese química , Antagonistas dos Receptores de Orexina/metabolismo , Receptores de Orexina/metabolismo , Relação Estrutura-Atividade , Sulfonamidas/metabolismo
10.
Bioorg Med Chem Lett ; 30(17): 127360, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32738987

RESUMO

The D-nor-nalfurafine derivatives, which were synthesized by contraction of the six-membered D-ring in nalfurafine (1), had no affinity for orexin 1 receptors (OX1Rs). The 17N-lone electron pair in 1 oriented toward the axial direction, while that of D-nor-derivatives was directed in the equatorial configuration. The axial lone electron pair can form a hydrogen bond with the 14-hydroxy group, which could push the 6-amide side chain toward the downward direction with respect to the C-ring. The resulting conformation would be an active conformation for binding with OX1R. The dual affinities of 1 for OX1R and κ opioid receptor (KOR) led us to elucidate the mechanism by which only 1 showed no aversion but U-50488H. Actually, 1 selectively induced severe aversion in OX1R knockout mice, but not in wild-type mice. These results well support that OX1R suppresses the aversion of 1. This is the elucidation of long period puzzle which 1 showed no aversion in KOR.


Assuntos
Morfinanos/química , Antagonistas dos Receptores de Orexina/síntese química , Receptores de Orexina/metabolismo , Compostos de Espiro/química , Animais , Aprendizagem da Esquiva/efeitos dos fármacos , Sítios de Ligação , Camundongos , Camundongos Knockout , Conformação Molecular , Simulação de Acoplamento Molecular , Morfinanos/metabolismo , Morfinanos/farmacologia , Antagonistas dos Receptores de Orexina/metabolismo , Antagonistas dos Receptores de Orexina/farmacologia , Receptores de Orexina/química , Receptores de Orexina/genética , Receptores Opioides kappa/agonistas , Receptores Opioides kappa/metabolismo , Compostos de Espiro/metabolismo , Compostos de Espiro/farmacologia
11.
Anal Chem ; 91(16): 10582-10588, 2019 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-31314489

RESUMO

The opioid epidemic continues in the United States. Many have been impacted by this epidemic, including neonates who exhibit Neonatal Abstinence Syndrome (NAS). Opioid diagnosis and NAS can be negatively impacted by limited testing options outside the hospital, due to poor assay performance, false-negatives, rapid drug clearance rates, and difficulty in obtaining enough specimen for testing. Here we report a small volume urine assay for oxycodone, hydrocodone, fentanyl, noroxycodone, norhydrocodone, and norfentanyl with excellent LODs and LOQs. The free-solution assay (FSA), coupled with high affinity DNA aptamer probes and a compensated interferometric reader (CIR), represents a potential solution for quantifying opioids rapidly, at high sensitivity, and noninvasively on small sample volumes. The mix-and-read test is 5- to 275-fold and 50- to 1250-fold more sensitive than LC-MS/MS and immunoassays, respectively. Using FSA, oxycodone, hydrocodone, fentanyl, and their urinary metabolites were quantified using 10 µL of urine at 28-81 pg/mL, with >95% specificity and excellent accuracy in ∼1 h. The assay sensitivity, small sample size requirement, and speed could enable opioid screening, particularly for neonates, and points to the potential for pharmacokinetic tracking.


Assuntos
Analgésicos Opioides/urina , Aptâmeros de Nucleotídeos/química , Analgésicos Opioides/metabolismo , Fentanila/metabolismo , Fentanila/urina , Humanos , Hidrocodona/análogos & derivados , Hidrocodona/metabolismo , Hidrocodona/urina , Estrutura Molecular , Morfinanos/metabolismo , Morfinanos/urina , Oxicodona/metabolismo , Oxicodona/urina
12.
Behav Pharmacol ; 29(4): 306-315, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29035920

RESUMO

Sinomenine is a bioactive alkaloid extracted from Sinomenium acutum. Here, we investigated the antidepressant effects of sinomenine in mice. The antidepressant actions of sinomenine were first examined in the forced-swim test and the tail-suspension test, and then assessed in the chronic social defeat stress (CSDS) model of depression. Changes in the brain-derived neurotrophic factor (BDNF) signaling pathway after CSDS and sinomenine treatment were also investigated. A tryptophan hydroxylase inhibitor and a BDNF signaling inhibitor were also used to determine the pharmacological mechanisms of sinomenine. It was found that sinomenine induced antidepressant-like effects in the forced-swim test and tail-suspension test without affecting the locomotor activity of mice. Sinomenine also prevented the CSDS-induced depressive-like symptoms. Moreover, sinomenine fully restored the CSDS-induced decrease in the hippocampal BDNF signaling pathway, whereas a BDNF signaling inhibitor, but not a tryptophan hydroxylase inhibitor, blocked the antidepressant effects of sinomenine. In conclusion, sinomenine exerts antidepressant effects in mice by promoting the hippocampal BDNF signaling pathway.


Assuntos
Depressão/tratamento farmacológico , Morfinanos/farmacologia , Animais , Antidepressivos/farmacologia , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Transtorno Depressivo/tratamento farmacológico , Modelos Animais de Doenças , Hipocampo/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Morfinanos/metabolismo , Neurogênese/efeitos dos fármacos , Serotonina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Estresse Psicológico/tratamento farmacológico
13.
Nature ; 485(7398): 321-6, 2012 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-22437502

RESUMO

Opium is one of the world's oldest drugs, and its derivatives morphine and codeine are among the most used clinical drugs to relieve severe pain. These prototypical opioids produce analgesia as well as many undesirable side effects (sedation, apnoea and dependence) by binding to and activating the G-protein-coupled µ-opioid receptor (µ-OR) in the central nervous system. Here we describe the 2.8 Å crystal structure of the mouse µ-OR in complex with an irreversible morphinan antagonist. Compared to the buried binding pocket observed in most G-protein-coupled receptors published so far, the morphinan ligand binds deeply within a large solvent-exposed pocket. Of particular interest, the µ-OR crystallizes as a two-fold symmetrical dimer through a four-helix bundle motif formed by transmembrane segments 5 and 6. These high-resolution insights into opioid receptor structure will enable the application of structure-based approaches to develop better drugs for the management of pain and addiction.


Assuntos
Morfinanos/química , Receptores Opioides mu/antagonistas & inibidores , Receptores Opioides mu/química , Animais , Sítios de Ligação , Cristalografia por Raios X , Ligantes , Camundongos , Modelos Moleculares , Morfinanos/metabolismo , Morfinanos/farmacologia , Conformação Proteica , Multimerização Proteica , Receptores Opioides mu/metabolismo , Solventes/química
14.
Acta Pharmacol Sin ; 39(1): 154-164, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29188799

RESUMO

The µ opioid receptor (OR), a member of the class A subfamily of G-protein coupled receptors (GPCRs), is a major target for the treatment of pain. G-protein biased µ-OR agonists promise to be developed as analgesics. Thus, TRV130, the first representative µ-OR ligand with G-protein bias, has entered into phase III clinical trials. To identify the detailed G-protein-biased activation and inactivation mechanisms of the µ-OR, we constructed five µ-OR systems that were in complexes with the G-protein-biased agonists TRV130 and BU72, the antagonists ß-FNA and naltrexone, as well as the free receptor. We performed a series of conventional molecular dynamics simulations and analyses of G-protein-biased activation and inactivation mechanisms of µ-OR. Our results, together with previously reported mutation results, revealed the operating mode of the activation switch composed of residues W6.48 and Y7.43 (Ballesteros/Weinstein numbering), the activity of which was responsible for down- and up-regulation, respectively, of the ß-arrestin signaling, which in turn affected G-protein-biased activation of µ-OR. TRV130 was found to stabilize W6.48 by interacting with Y7.43. In addition, we obtained useful information regarding µ-OR-biased activation, such as strong stabilization of W7.35 through a hydrophobic ring interaction in the TRV130 system. These findings may facilitate understanding of µ-OR biased activation and the design of new biased ligands for GPCRs.


Assuntos
Proteínas de Ligação ao GTP/metabolismo , Morfinanos/metabolismo , Pirróis/metabolismo , Receptores Opioides mu/metabolismo , Compostos de Espiro/metabolismo , Tiofenos/metabolismo , Animais , Proteínas de Ligação ao GTP/química , Ligantes , Camundongos , Simulação de Dinâmica Molecular , Morfinanos/química , Naltrexona/análogos & derivados , Naltrexona/química , Naltrexona/metabolismo , Ligação Proteica , Conformação Proteica , Pirróis/química , Receptores Opioides mu/agonistas , Receptores Opioides mu/antagonistas & inibidores , Receptores Opioides mu/química , Compostos de Espiro/química , Tiofenos/química , Ureia/análogos & derivados , Ureia/química , Ureia/metabolismo , Água/química
15.
Med Sci Monit ; 24: 3631-3636, 2018 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-29849019

RESUMO

BACKGROUND This study aimed to investigate the effect and underlying molecular mechanism of sinomenine (SIN) on ankylosing spondylitis (AS). MATERIAL AND METHODS To study the potential role of SIN in the pathogenesis of AS, an AS mouse model was established and mice were treated with different concentrations of SIN (10, 30, and 50 mg/kg, administered intraperitoneally). Markers of inflammation and oxidative stress were determined by ELISA assay. Western blot analysis and qRT-PCR were used to quantify the levels of related proteins and gene mRNA expression. RESULTS The results suggest that AS mice has higher levels of TNF-α, IL-1ß, and IL-6 (p<0.01 for all), and lower levels of superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GSH-PX) (p<0.01 for all). SIN treatment reduced the level of TNF-α, IL-1ß, and IL-6 in a dose-dependent manner, and the levels of SOD, CAT, and GSH-PX were dose-dependently increased (p<0.05 for all). The results also revealed that NF-κBp65 expression decreased, while the level of IkB increased, in a dose-dependent manner, after SIN treatment in AS mice (p<0.05 for all). The level of p-p38 was dose-dependently reduced in AS mice by SIN treatment (p<0.05). Moreover, SIN inhibited Cox-2 expression in AS mice in a dose-dependent manner (p<0.05). CONCLUSIONS SIN has a beneficial role in AS through suppressing inflammatory mediators and by down-regulating oxidative stress via inhibiting the MAPKp38/NF-kB pathway and Cox-2 expression.


Assuntos
Morfinanos/farmacologia , Espondilite Anquilosante/tratamento farmacológico , Animais , Antioxidantes/farmacologia , Catalase/efeitos dos fármacos , Modelos Animais de Doenças , Glutationa Peroxidase/efeitos dos fármacos , Inflamação/tratamento farmacológico , Interleucina-1beta/efeitos dos fármacos , Interleucina-6/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Morfinanos/metabolismo , NF-kappa B/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Superóxido Dismutase/efeitos dos fármacos , Fator de Necrose Tumoral alfa/efeitos dos fármacos
16.
Biomed Chromatogr ; 32(2)2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28801971

RESUMO

The aim of this study was to elucidate the pharmacokinetics of olerciamide A in rats after oral and intravenous administration of Portulaca oleracea L. extract by a simple and rapid ultra high-performance liquid chromatography method with bergapten as internal standard. The pharmacokinetic results indicated that olerciamide A was rapidly distributed with a time to peak concentration of 30 min after oral administration and presented a low oral absolute bioavailability of 4.57%. The metabolism of olerciamide A in rats was also investigated using ultra-high-performance liquid chromatography electrospray coupled with quadrupole-time of flight mass spectrometry to elucidate the reason for the low absolute bioavailability of olerciamide A and seven metabolites of oleraciamide A were found in rat plasma and urine.


Assuntos
Alcaloides , Cromatografia Líquida de Alta Pressão/métodos , Morfinanos , Portulaca/química , Espectrometria de Massas por Ionização por Electrospray/métodos , Alcaloides/sangue , Alcaloides/metabolismo , Alcaloides/farmacocinética , Alcaloides/urina , Animais , Glucuronídeos/metabolismo , Glutationa/metabolismo , Limite de Detecção , Modelos Lineares , Masculino , Morfinanos/sangue , Morfinanos/metabolismo , Morfinanos/farmacocinética , Morfinanos/urina , Ratos , Ratos Wistar , Reprodutibilidade dos Testes , Sulfatos/metabolismo
17.
Int J Mol Sci ; 20(1)2018 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-30586944

RESUMO

Sinomenine (SIN) is widely used in China to treat a variety of rheumatic diseases (RA), and has various pharmacological effects such as anti-inflammatory, analgesic, and anti-tumor effects. However, due to the histamine release characteristics of SIN, its adverse reactions such as allergic reactions, gastrointestinal reactions, and circulatory systemic reactions have been drawing increasing attention. We present here a systematic review of the chemical structure, pharmacological effects, clinical application, and adverse reactions of SIN, a detailed discussion on the relationship between histamine/histamine receptor and mechanism of action of SIN. In addition, we simulated the binding of SIN to four histamine receptors by using a virtual molecular docking method and found that the bonding intensity between SIN and receptors varied in the order shown as follows: H1R > H2R ~ H3R > H4R. The docking results suggested that SIN might exhibit dual regulatory effects in many processes such as cyclooxygenase-2 (COX-2) expression, NF-κB pathway activation, and degranulation of mast cells to release histamine, thereby exhibiting pro-inflammatory (adverse reactions)/anti-inflammatory effects. This study provides a theoretical basis for the clinical treatment of inflammations seen such as in RA using SIN, and also suggests that SIN has great potential in the field of cancer treatment and will have very important social and economic significance.


Assuntos
Imunossupressores/metabolismo , Morfinanos/metabolismo , Receptores Histamínicos/metabolismo , Ciclo-Oxigenase 2/metabolismo , Doenças Hematológicas/etiologia , Liberação de Histamina , Humanos , Imunossupressores/efeitos adversos , Imunossupressores/uso terapêutico , Simulação de Acoplamento Molecular , Morfinanos/química , Morfinanos/uso terapêutico , Ligação Proteica , Receptores Histamínicos/química , Doenças Reumáticas/tratamento farmacológico
18.
J Pharmacol Exp Ther ; 363(1): 104-113, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28778859

RESUMO

The increasing availability of prescription opioid analgesics for the treatment of pain has been paralleled by an epidemic of opioid misuse, diversion, and overdose. The development of abuse-deterrent formulations (ADFs) of conventional opioids such as oxycodone and morphine represents an advance in the field and has had a positive but insufficient impact, as most opioids are still prescribed in highly abusable, non-ADF forms, and abusers can tamper with ADF medications to liberate the abusable opioid within. The abuse liability of mu-opioid agonists appears to be dependent on their rapid rate of entry into the central nervous system (CNS), whereas analgesic activity appears to be a function of CNS exposure alone, suggesting that a new opioid agonist with an inherently low rate of influx across the blood-brain barrier could mediate analgesia with low abuse liability, regardless of formulation or route of administration. NKTR-181 is a novel, long-acting, selective mu-opioid agonist with structural properties that reduce its rate of entry across the blood-brain barrier compared with traditional mu-opioid agonists. NKTR-181 demonstrated maximum analgesic activity comparable to that of oxycodone in hot-plate latency and acetic-acid writhing models. NKTR-181 was distinguishable from oxycodone by its reduced abuse potential in self-administration and progressive-ratio break point models, with behavioral effects similar to those of saline, as well as reduced CNS side effects as measured by the modified Irwin test. The in vitro and in vivo studies presented here demonstrate that NKTR-181 is the first selective mu-opioid agonist to combine analgesic efficacy and reduced abuse liability through the alteration of brain-entry kinetics.


Assuntos
Analgésicos Opioides/farmacologia , Morfinanos/farmacologia , Transtornos Relacionados ao Uso de Substâncias/prevenção & controle , Analgésicos Opioides/química , Analgésicos Opioides/metabolismo , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Células CACO-2 , Relação Dose-Resposta a Droga , Composição de Medicamentos , Humanos , Masculino , Morfinanos/química , Morfinanos/metabolismo , Permeabilidade , Ratos , Ratos Sprague-Dawley , Receptores Opioides mu/metabolismo , Fatores de Tempo
19.
Toxicol Appl Pharmacol ; 329: 85-95, 2017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28554659

RESUMO

Naloxegol is an opioid antagonist which has been developed for the treatment of patients with opioid induced constipation. In the nonclinical safety program naloxegol was shown to have a very benign toxicity profile. In the rat, but not the mouse, 2-year carcinogenicity study a change in tumour pattern with an increase in testicular Leydig cell tumours (LCT) was observed after dosing at high (supra-pharmacological) concentrations. To establish the basis of the increase in LCT and to assess its potential relevance to humans, studies to exclude and potentially identify mode-of-action (MoA) were performed. A genotoxic mechanism was ruled out following negative results in the Ames, mouse lymphoma, and micronucleus assays. An effect on androgen metabolism was excluded since the treatment of rats with naloxegol for 14days did not result in any induction of CYP protein levels. It was demonstrated that administration of centrally restricted opioid antagonists naloxegol or methylnaltrexone at high doses induced an increase in LH release with no clear increase in testosterone, in contrast to the centrally acting opioid antagonist naloxone, which showed marked increases in both LH and testosterone. LCT due to increased LH stimulation is common in rats but not documented in humans. Collectively, the lack of genotoxicity signal, the lack of androgen effect, the increase in LH secretion in rats, which is no considered to be relevant for LCT formation in humans, and high margins to clinical exposures, the observed increase in LCT in the rat is not expected to be clinically relevant.


Assuntos
Barreira Hematoencefálica/metabolismo , Tumor de Células de Leydig/induzido quimicamente , Hormônio Luteinizante/sangue , Morfinanos/toxicidade , Antagonistas de Entorpecentes/toxicidade , Polietilenoglicóis/toxicidade , Neoplasias Testiculares/induzido quimicamente , Animais , Biomarcadores/sangue , Permeabilidade Capilar , Cães , Feminino , Humanos , Tumor de Células de Leydig/patologia , Masculino , Camundongos , Morfinanos/metabolismo , Naltrexona/análogos & derivados , Naltrexona/metabolismo , Naltrexona/toxicidade , Antagonistas de Entorpecentes/metabolismo , Polietilenoglicóis/metabolismo , Compostos de Amônio Quaternário/metabolismo , Compostos de Amônio Quaternário/toxicidade , Coelhos , Ratos Sprague-Dawley , Medição de Risco , Especificidade da Espécie , Neoplasias Testiculares/patologia , Testosterona/sangue , Fatores de Tempo , Testes de Toxicidade/métodos , Regulação para Cima
20.
Nat Chem Biol ; 11(9): 728-32, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26147354

RESUMO

The gateway to morphine biosynthesis in opium poppy (Papaver somniferum) is the stereochemical inversion of (S)-reticuline since the enzyme yielding the first committed intermediate salutaridine is specific for (R)-reticuline. A fusion between a cytochrome P450 (CYP) and an aldo-keto reductase (AKR) catalyzes the S-to-R epimerization of reticuline via 1,2-dehydroreticuline. The reticuline epimerase (REPI) fusion was detected in opium poppy and in Papaver bracteatum, which accumulates thebaine. In contrast, orthologs encoding independent CYP and AKR enzymes catalyzing the respective synthesis and reduction of 1,2-dehydroreticuline were isolated from Papaver rhoeas, which does not accumulate morphinan alkaloids. An ancestral relationship between these enzymes is supported by a conservation of introns in the gene fusions and independent orthologs. Suppression of REPI transcripts using virus-induced gene silencing in opium poppy reduced levels of (R)-reticuline and morphinan alkaloids and increased the overall abundance of (S)-reticuline and its O-methylated derivatives. Discovery of REPI completes the isolation of genes responsible for known steps of morphine biosynthesis.


Assuntos
Aldeído Redutase/metabolismo , Carboidratos Epimerases/metabolismo , Sistema Enzimático do Citocromo P-450/metabolismo , Regulação da Expressão Gênica de Plantas , Morfina/biossíntese , Papaver/metabolismo , Proteínas de Plantas/metabolismo , Aldeído Redutase/genética , Aldo-Ceto Redutases , Alcaloides/biossíntese , Alcaloides/química , Sequência de Bases , Benzilisoquinolinas/química , Benzilisoquinolinas/metabolismo , Bromoviridae/genética , Bromoviridae/metabolismo , Carboidratos Epimerases/antagonistas & inibidores , Carboidratos Epimerases/genética , Sistema Enzimático do Citocromo P-450/genética , Escherichia coli/genética , Escherichia coli/metabolismo , Éxons , Fusão Gênica , Íntrons , Ligases/genética , Ligases/metabolismo , Dados de Sequência Molecular , Morfinanos/química , Morfinanos/metabolismo , Morfina/química , Fases de Leitura Aberta , Ópio/química , Ópio/metabolismo , Oxirredução , Papaver/genética , Proteínas de Plantas/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Estereoisomerismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA