Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 169
Filter
1.
Molecules ; 29(11)2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38893511

ABSTRACT

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.


Subject(s)
Analgesics, Opioid , Receptors, Opioid, kappa , Receptors, Opioid, kappa/metabolism , Receptors, Opioid, kappa/chemistry , Humans , Analgesics, Opioid/chemistry , Analgesics, Opioid/pharmacology , Animals , Binding Sites , Ligands , Signal Transduction/drug effects , Protein Binding , Structure-Activity Relationship , Narcotic Antagonists/chemistry , Pain/drug therapy , Pain/metabolism
2.
FEBS J ; 291(13): 2784-2791, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38151714

ABSTRACT

Receptor-G protein promiscuity is frequently observed in class A G protein-coupled receptors (GPCRs). In particular, GPCRs can couple with G proteins from different families (Gαs, Gαq/11, Gαi/o, and Gα12/13) or the same family subtypes. The molecular basis underlying the selectivity/promiscuity is not fully revealed. We recently reported the structures of kappa opioid receptor (KOR) in complex with the Gi/o family subtypes [Gαi1, GαoA, Gαz, and Gustducin (Gαg)] determined by cryo-electron microscopy (cryo-EM). The structural analysis, in combination with pharmacological studies, provides insights into Gi/o subtype selectivity. Given the conserved sequence identity and activation mechanism between different G protein families, the findings within Gi/o subtypes could be likely extended to other families. Understanding the KOR-Gi/o or GPCR-G protein selectivity will facilitate the development of more precise therapeutics targeting a specific G protein subtype.


Subject(s)
Cryoelectron Microscopy , Receptors, G-Protein-Coupled , Receptors, Opioid, kappa , Humans , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/chemistry , Receptors, Opioid, kappa/metabolism , Receptors, Opioid, kappa/chemistry , Receptors, Opioid, kappa/agonists , Receptors, Opioid, kappa/genetics , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/chemistry , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , GTP-Binding Proteins/metabolism , GTP-Binding Proteins/chemistry , GTP-Binding Proteins/genetics , Protein Binding , Animals , Protein Conformation , Models, Molecular
3.
Arch Pharm (Weinheim) ; 356(9): e2300256, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37452407

ABSTRACT

The increasing misuse of novel synthetic opioids (NSOs) represents a serious public health concern. In this regard, U-47700 (trans-3,4-dichloro-N-[2-(dimethylamino)cyclohexyl]-N-methylbenzamide) and related "U-compounds" emerged on recreational drug markets as synthetic substitutes for illicit heroin and constituents of counterfeit pain medications. While the pharmacology of U-compounds has been investigated using in vitro and in vivo methods, there is still a lack of understanding about the details of ligand-receptor interactions at the molecular level. To this end, we have developed a molecular modeling protocol based on docking and molecular dynamics simulations to assess the nature of ligand-receptor interactions for U-47700, N,N-didesmethyl U-47700, and U-50488 at the mu-opioid receptor (MOR) and kappa-opioid receptor (KOR). The evaluation of ligand-receptor and ligand-receptor-membrane interaction energies enabled the identification of subtle conformational shifts in the receptors induced by ligand binding. Interestingly, the removal of two key methyl groups from U-47700, to form N,N-didesmethyl U-47700, caused a loss of hydrogen bond contact with tryptophan (Trp)229, which may underlie the lower interaction energy and reduced MOR affinity for the compound. Taken together, our results are consistent with the reported biological findings for U-compounds and provide a molecular basis for the MOR selectivity of U-47700 and KOR selectivity of U-50488.


Subject(s)
Receptors, Opioid, kappa , Receptors, Opioid, mu , Receptors, Opioid, kappa/chemistry , Receptors, Opioid, kappa/metabolism , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology , Ligands , Structure-Activity Relationship , Receptors, Opioid, mu/metabolism , Analgesics, Opioid/pharmacology , Analgesics, Opioid/chemistry
4.
Nature ; 617(7960): 417-425, 2023 05.
Article in English | MEDLINE | ID: mdl-37138078

ABSTRACT

The κ-opioid receptor (KOR) represents a highly desirable therapeutic target for treating not only pain but also addiction and affective disorders1. However, the development of KOR analgesics has been hindered by the associated hallucinogenic side effects2. The initiation of KOR signalling requires the Gi/o-family proteins including the conventional (Gi1, Gi2, Gi3, GoA and GoB) and nonconventional (Gz and Gg) subtypes. How hallucinogens exert their actions through KOR and how KOR determines G-protein subtype selectivity are not well understood. Here we determined the active-state structures of KOR in a complex with multiple G-protein heterotrimers-Gi1, GoA, Gz and Gg-using cryo-electron microscopy. The KOR-G-protein complexes are bound to hallucinogenic salvinorins or highly selective KOR agonists. Comparisons of these structures reveal molecular determinants critical for KOR-G-protein interactions as well as key elements governing Gi/o-family subtype selectivity and KOR ligand selectivity. Furthermore, the four G-protein subtypes display an intrinsically different binding affinity and allosteric activity on agonist binding at KOR. These results provide insights into the actions of opioids and G-protein-coupling specificity at KOR and establish a foundation to examine the therapeutic potential of pathway-selective agonists of KOR.


Subject(s)
Cryoelectron Microscopy , Heterotrimeric GTP-Binding Proteins , Ligands , Receptors, Opioid, kappa , Analgesics, Opioid/metabolism , Analgesics, Opioid/pharmacology , Receptors, Opioid, kappa/chemistry , Receptors, Opioid, kappa/metabolism , Receptors, Opioid, kappa/ultrastructure , Signal Transduction , Heterotrimeric GTP-Binding Proteins/chemistry , Heterotrimeric GTP-Binding Proteins/metabolism , Heterotrimeric GTP-Binding Proteins/ultrastructure , Substrate Specificity , Allosteric Regulation/drug effects , Hallucinogens/metabolism , Hallucinogens/pharmacology
5.
Molecules ; 27(3)2022 Jan 18.
Article in English | MEDLINE | ID: mdl-35163865

ABSTRACT

Most contemporary drug discovery projects start with a 'hit discovery' phase where small chemicals are identified that have the capacity to interact, in a chemical sense, with a protein target involved in a given disease. To assist and accelerate this initial drug discovery process, 'virtual docking calculations' are routinely performed, where computational models of proteins and computational models of small chemicals are evaluated for their capacities to bind together. In cutting-edge, contemporary implementations of this process, several conformations of protein targets are independently assayed in parallel 'ensemble docking' calculations. Some of these protein conformations, a minority of them, will be capable of binding many chemicals, while other protein conformations, the majority of them, will not be able to do so. This fact that only some of the conformations accessible to a protein will be 'selected' by chemicals is known as 'conformational selection' process in biology. This work describes a machine learning approach to characterize and identify the properties of protein conformations that will be selected (i.e., bind to) chemicals, and classified as potential binding drug candidates, unlike the remaining non-binding drug candidate protein conformations. This work also addresses the class imbalance problem through advanced machine learning techniques that maximize the prediction rate of potential protein molecular conformations for the test case proteins ADORA2A (Adenosine A2a Receptor) and OPRK1 (Opioid Receptor Kappa 1), and subsequently reduces the failure rates and hastens the drug discovery process.


Subject(s)
Algorithms , Big Data , Drug Discovery , Machine Learning , Receptor, Adenosine A2A/metabolism , Receptors, Opioid, kappa/metabolism , Computer Simulation , Humans , Ligands , Protein Binding , Protein Conformation , Receptor, Adenosine A2A/chemistry , Receptors, Opioid, kappa/chemistry
6.
Mol Med Rep ; 25(2)2022 Feb.
Article in English | MEDLINE | ID: mdl-34878160

ABSTRACT

The κ­opioid receptor (KOR) is one of the primary receptors of opioids and serves a vital role in the regulation of pain, anesthesia, addiction and other pathological and physiological processes. KOR is associated with several types of cancer and may influence cancer progression. It has been proposed that KOR may represent a new tumor molecular marker and provide a novel basis for molecular targeted therapies for cancer. However, the association between KOR and cancer remains to be explored comprehensively. The present review introduces KOR and its association with different types of cancer. Improved understanding of KOR may facilitate development of novel antitumor therapies.


Subject(s)
Analgesics, Opioid/metabolism , Hypoxia/metabolism , Inflammation/metabolism , Neoplasms/metabolism , Neovascularization, Pathologic , Receptors, Opioid, kappa/physiology , Animals , Disease Progression , Humans , Receptors, Opioid, kappa/chemistry
7.
Molecules ; 26(16)2021 Aug 06.
Article in English | MEDLINE | ID: mdl-34443366

ABSTRACT

The kappa opioid receptor (KOR) represents an attractive target for the development of drugs as potential antidepressants, anxiolytics and analgesics. A robust computational approach may guarantee a reduction in costs in the initial stages of drug discovery, novelty and accurate results. In this work, a virtual screening workflow of a library consisting of ~6 million molecules was set up, with the aim to find potential lead compounds that could manifest activity on the KOR. This in silico study provides a significant contribution in the identification of compounds capable of interacting with a specific molecular target. The main computational techniques adopted in this experimental work include: (i) virtual screening; (ii) drug design and leads optimization; (iii) molecular dynamics. The best hits are tripeptides prepared via solution phase peptide synthesis. These were tested in vivo, revealing a good antinociceptive effect after subcutaneous administration. However, further work is due to delineate their full pharmacological profile, in order to verify the features predicted by the in silico outcomes.


Subject(s)
Computer Simulation , Drug Design , Oligopeptides/chemistry , Oligopeptides/metabolism , Receptors, Opioid, kappa/metabolism , Ligands , Molecular Dynamics Simulation , Protein Conformation , Receptors, Opioid, kappa/chemistry
9.
Elife ; 102021 02 08.
Article in English | MEDLINE | ID: mdl-33555255

ABSTRACT

Controlling receptor functional selectivity profiles for opioid receptors is a promising approach for discovering safer analgesics; however, the structural determinants conferring functional selectivity are not well understood. Here, we used crystal structures of opioid receptors, including the recently solved active state kappa opioid complex with MP1104, to rationally design novel mixed mu (MOR) and kappa (KOR) opioid receptor agonists with reduced arrestin signaling. Analysis of structure-activity relationships for new MP1104 analogs points to a region between transmembrane 5 (TM5) and extracellular loop (ECL2) as key for modulation of arrestin recruitment to both MOR and KOR. The lead compounds, MP1207 and MP1208, displayed MOR/KOR Gi-partial agonism with diminished arrestin signaling, showed efficient analgesia with attenuated liabilities, including respiratory depression and conditioned place preference and aversion in mice. The findings validate a novel structure-inspired paradigm for achieving beneficial in vivo profiles for analgesia through different mechanisms that include bias, partial agonism, and dual MOR/KOR agonism.


Subject(s)
Morphinans/chemistry , Receptors, Opioid, kappa/chemistry , Receptors, Opioid, mu/chemistry , Amino Acid Motifs , Analgesics/chemistry , Analgesics/metabolism , Animals , Binding Sites , Ligands , Male , Mice , Mice, Inbred C57BL , Molecular Docking Simulation , Receptors, Opioid, kappa/metabolism , Receptors, Opioid, mu/metabolism , Structure-Activity Relationship
10.
Future Med Chem ; 13(6): 551-573, 2021 03.
Article in English | MEDLINE | ID: mdl-33590767

ABSTRACT

The modulation and selectivity mechanisms of seven mixed-action kappa opioid receptor (KOR)/mu opioid receptor (MOR) bitopic modulators were explored. Molecular modeling results indicated that the 'message' moiety of seven bitopic modulators shared the same binding mode with the orthosteric site of the KOR and MOR, whereas the 'address' moiety bound with different subdomains of the allosteric site of the KOR and MOR. The 'address' moiety of seven bitopic modulators bound to different subdomains of the allosteric site of the KOR and MOR may exhibit distinguishable allosteric modulations to the binding affinity and/or efficacy of the 'message' moiety. Moreover, the 3-hydroxy group on the phenolic moiety of the seven bitopic modulators induced selectivity to the KOR over the MOR.


Subject(s)
Receptors, Opioid, kappa/metabolism , Receptors, Opioid, mu/metabolism , Allosteric Regulation , Allosteric Site , Binding Sites , Humans , Ligands , Molecular Docking Simulation , Morphinans/chemistry , Morphinans/metabolism , Naltrexone/analogs & derivatives , Naltrexone/chemistry , Naltrexone/metabolism , Protein Binding , Receptors, Opioid, kappa/chemistry , Receptors, Opioid, mu/chemistry , Spiro Compounds/chemistry , Spiro Compounds/metabolism , Thermodynamics
11.
Molecules ; 25(21)2020 Nov 02.
Article in English | MEDLINE | ID: mdl-33147885

ABSTRACT

Among the opioid receptors, the kappa opioid receptor (KOR) has been gaining substantial attention as a promising molecular target for the treatment of numerous human disorders, including pain, pruritus, affective disorders (i.e., depression and anxiety), drug addiction, and neurological diseases (i.e., epilepsy). Particularly, the knowledge that activation of the KOR, opposite to the mu opioid receptor (MOR), does not produce euphoria or leads to respiratory depression or overdose, has stimulated the interest in discovering ligands targeting the KOR as novel pharmacotherapeutics. However, the KOR mediates the negative side effects of dysphoria/aversion, sedation, and psychotomimesis, with the therapeutic promise of biased agonism (i.e., selective activation of beneficial over deleterious signaling pathways) for designing safer KOR therapeutics without the liabilities of conventional KOR agonists. In this review, the development of new KOR ligands from the class of diphenethylamines is presented. Specifically, we describe the design strategies, synthesis, and pharmacological activities of differently substituted diphenethylamines, where structure-activity relationships have been extensively studied. Ligands with distinct profiles as potent and selective agonists, G protein-biased agonists, and selective antagonists, and their potential use as therapeutic agents (i.e., pain treatment) and research tools are described.


Subject(s)
Analgesics, Opioid , Pain , Receptors, Opioid, kappa , Analgesics, Opioid/chemistry , Analgesics, Opioid/therapeutic use , Humans , Ligands , Pain/drug therapy , Pain/metabolism , Receptors, Opioid, kappa/agonists , Receptors, Opioid, kappa/chemistry , Receptors, Opioid, kappa/metabolism , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/chemistry , Receptors, Opioid, mu/metabolism , Structure-Activity Relationship
12.
Biochem Pharmacol ; 182: 114293, 2020 12.
Article in English | MEDLINE | ID: mdl-33091380

ABSTRACT

Substituted fentanyls are abused and cause rapid fatal overdose. As their pharmacology is not well characterized, we examined in vitro pharmacology and structure-activity relationships of 22 substituted fentanyls with modifications of the fentanyl propyl group, and conducted in silico receptor/ligand modeling. Affinities for mu, kappa, and delta opioid receptors (MOR, KOR, and DOR, respectively) heterologously expressed in mammalian cells were assessed in agonist radioligand binding assays. At MOR, furanyl fentanyl had higher affinity than fentanyl, while acryl, isobutyryl and cyclopropyl fentanyls had similar affinities. Comparing affinities, thiophene and methoxyacetyl fentanyls had highest selectivity for MOR (2520- and 2730-fold compared to KOR and DOR, respectively). Functional activities were assessed using [35S]GTPγS binding assays. At MOR, furanyl fentanyl had higher potency and 11 substituted fentanyls had similar high potencies compared to fentanyl. Eight compounds were full agonists of MOR and twelve compounds were partial agonists, with efficacies from 8.8% (phenyl fentanyl) to 60.2% (butyryl fentanyl). All efficacious compounds had selective functional potency for MOR. The predicted binding poses of flexible fentanyl and rigid morphine against MOR show partially overlapping binding pockets, with fentanyl maintaining additional interaction with the transmembrane (TM) 2 helix. Subsequent molecular dynamics simulations revealed a predominant fentanyl binding pose involving various TM interactions. The piperidine nitrogen of substituted fentanyls establishes a salt-bridge with the conserved D-1473.32 residue and the propanamide carbonyl group establishes a hydrogen bond with the indole side-chain (-NH) of W-3187.35. The simulation suggests theN-linked phenethyl group may regulate the rotameric switch of W-2936.48. The predicted binding pose, in conjunction with in vitro binding affinity, clarified the molecular basis of the binding/selectivity profile of furanyl fentanyl and other derivatives at the sequence level. In summary, substituted fentanyls with high MOR potencies, selectivities, and efficacies are likely to have abuse and overdose potential. The work presented here is a prototype to investigate fentanyl derivatives and their abuse potential.


Subject(s)
Analgesics, Opioid/metabolism , Fentanyl/metabolism , Models, Molecular , Molecular Docking Simulation/methods , Receptors, Opioid, kappa/metabolism , Analgesics, Opioid/chemistry , Analgesics, Opioid/pharmacology , Animals , CHO Cells , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Fentanyl/analogs & derivatives , Fentanyl/chemistry , Fentanyl/pharmacology , Furans/chemistry , Furans/metabolism , Furans/pharmacology , Humans , Protein Binding/drug effects , Protein Binding/physiology , Protein Structure, Secondary , Receptors, Opioid, kappa/agonists , Receptors, Opioid, kappa/chemistry , Structure-Activity Relationship , Treatment Outcome
13.
Molecules ; 25(17)2020 Aug 25.
Article in English | MEDLINE | ID: mdl-32854311

ABSTRACT

Analgesic and anti-inflammatory properties mediated by the κ opioid receptor (KOR) have been reported for oxadiazole imidazodiazepines. Affinities determined by radioligand competition assays of more than seventy imidazodiazepines using cell homogenates from HEK293 cells that overexpress KOR, µ opioid receptor (MOR), and δ opioid receptor (DOR) are presented. Affinities to synaptic, benzodiazepine-sensitive receptors (BZR) were determined with rat brain extract. The highest affinity for KOR was recorded for GL-I-30 (Ki of 27 nM) and G-protein recruitment was observed with an EC50 of 32 nM. Affinities for MOR and DOR were weak for all compounds. Ester and amide imidazodiazepines were among the most active KOR ligands but also competed with 3H-flunitrazepam for brain extract binding, which is mediated predominately by gamma aminobutyric acid type A receptors (GABAAR) of the α1-3ß2-3γ1-2 subtypes. Imidazodiazepines with carboxylic acid and primary amide groups did not bind KOR but interacted strongly with GABAARs. Pyridine substitution reduced KOR affinity. Oxadiazole imidazodiazepines exhibited good KOR binding and interacted weakly with BZR, whereas oxazole imidazodiazepines were more selective towards BZR. Compounds that lack the imidazole moiety, the pendent phenyl, or pyridine substitutions exhibited insignificant KOR affinities. It can be concluded that a subset of imidazodiazepines represents novel KOR ligands with high selectivity among opioid receptors.


Subject(s)
Azepines , GABA-A Receptor Agonists , Receptors, GABA-A , Receptors, Opioid, delta , Receptors, Opioid, kappa , Receptors, Opioid, mu , Animals , Azepines/chemistry , Azepines/pharmacology , GABA-A Receptor Agonists/chemistry , GABA-A Receptor Agonists/pharmacology , HEK293 Cells , Humans , Protein Binding , Receptors, GABA-A/chemistry , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Receptors, Opioid, delta/agonists , Receptors, Opioid, delta/chemistry , Receptors, Opioid, delta/genetics , Receptors, Opioid, delta/metabolism , Receptors, Opioid, kappa/agonists , Receptors, Opioid, kappa/chemistry , Receptors, Opioid, kappa/genetics , Receptors, Opioid, kappa/metabolism , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/chemistry , Receptors, Opioid, mu/genetics , Receptors, Opioid, mu/metabolism , Structure-Activity Relationship
14.
Anesth Analg ; 131(3): 935-942, 2020 09.
Article in English | MEDLINE | ID: mdl-32701545

ABSTRACT

BACKGROUND: The misuse of opioids stems, in part, from inadequate knowledge of molecular interactions between opioids and opioid receptors. It is still unclear why some opioids are far more addictive than others. The κ-opioid receptor (KOR) plays a critical role in modulating pain, addiction, and many other physiological and pathological processes. Butorphanol, an opioid analgesic, is a less addictive opioid with unique pharmacological profiles. In this study, we investigated the interaction between butorphanol and KOR to obtain insights into the safe usage of this medication. METHODS: We determined the binding affinity of butorphanol to KOR with a naltrexone competition study. Recombinant KORs expressed in mammalian cell membranes (Chem-1) were used for G-protein activation studies, and a human embryonic kidney-293 (HEK-293) cell line stably transfected with the human KOR was used for ß-arrestin study as previously described in the literature. The effects of butorphanol on KOR internalization were investigated using mouse neuroblastoma Neuro2A cells stably transfected with mKOR-tdTomato fusion protein (N2A-mKOR-tdT) cells overexpressing KOR. The active-state KOR crystal structure was used for docking calculation of butorphanol to characterize the ligand binding site. Salvinorin A, a full KOR agonist, was used as a control for comparison. RESULTS: The affinity of KOR for butorphanol is characterized by Kd of 0.1 ± 0.02 nM, about 20-fold higher compared with that of the µ-opioid receptor (MOR; 2.4 ± 1.2 nM). Our data indicate that butorphanol is more potent on KOR than on MOR. In addition, butorphanol acts as a partial agonist of KOR in the G-protein activation pathway and is a full agonist on the ß-arrestin recruitment pathway, similar to that of salvinorin A. The activation of the ß-arrestin pathway is further confirmed by KOR internalization. The in silico docking model indicates that both salvinorin A and butorphanol share the same binding cavity with the KOR full agonist MP1104. This cavity plays an important role in determining either agonist or antagonist effects of the ligand. CONCLUSIONS: In conclusion, butorphanol is a partial KOR agonist in the G-protein activation pathway and a potent KOR full agonist in the ß-arrestin recruitment pathway. The structure analysis offers insights into the molecular mechanism of KOR interaction and activation by butorphanol.


Subject(s)
Analgesics, Opioid/pharmacology , Butorphanol/pharmacology , Neurons/drug effects , Receptors, Opioid, kappa/agonists , Analgesics, Opioid/chemistry , Analgesics, Opioid/metabolism , Analgesics, Opioid/toxicity , Animals , Butorphanol/chemistry , Butorphanol/metabolism , Butorphanol/toxicity , Cell Line, Tumor , Drug Partial Agonism , HEK293 Cells , Humans , Mice , Molecular Docking Simulation , Neurons/metabolism , Protein Binding , Protein Conformation , Receptors, Opioid, kappa/chemistry , Receptors, Opioid, kappa/metabolism , Signal Transduction , Structure-Activity Relationship , beta-Arrestins/metabolism
15.
Biochem Pharmacol ; 177: 114014, 2020 07.
Article in English | MEDLINE | ID: mdl-32387457

ABSTRACT

Developing new drugs for killing colorectal cancer (CRC) cells is urgently needed. Here, we explored the antitumor effects of toosendanin (TSN) in CRC, as well as explored its antitumor mechanisms and direct targets. Cell proliferation and apoptosis were analyzed by CCK8, colony formation, real-time cell impedance and flow cytometry. The signaling pathway and Wnt activity were analyzed by Wnt luciferase activity assay, quantitative real-time PCR and western blot. The interaction between TSN and the κ-opioid receptor was analyzed by a molecular docking simulation. BALB/c nude mice were used to detect the effects of TSN on tumor growth in vivo. We found that TSN inhibited proliferation, induced G1 phase arrest and caused caspase-dependent apoptosis in both 5-FU-sensitive and 5-FU-resistant CRC cells. Moreover, TSN effectively inhibited CRC growth in vivo. In terms of the mechanism, TSN inhibited Wnt/ß-catenin signaling in CRC cells, and the molecular docking results showed that TSN could bind to κ-opioid receptors directly. Additionally, TSN-induced apoptosis and ß-catenin decline were both reversed by the selective κ-opioid receptor agonist U50,488H. Our data demonstrate that TSN-induced apoptosis in CRC cells is associated with the κ-opioid receptor/ß-catenin signaling axis, and TSN has promising potential as an antitumor agent for CRC treatment.


Subject(s)
Apoptosis/drug effects , Colorectal Neoplasms/drug therapy , Drugs, Chinese Herbal/pharmacology , Receptors, Opioid, kappa/metabolism , beta Catenin/metabolism , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/physiology , Cell Line, Tumor , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Drug Resistance, Neoplasm/drug effects , Drugs, Chinese Herbal/chemistry , Female , Fluorouracil/pharmacology , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , Mice, Inbred BALB C , Molecular Docking Simulation , Receptors, Opioid, kappa/agonists , Receptors, Opioid, kappa/chemistry , Wnt Signaling Pathway/drug effects , Xenograft Model Antitumor Assays , beta Catenin/genetics
16.
Proc Natl Acad Sci U S A ; 117(11): 5836-5843, 2020 03 17.
Article in English | MEDLINE | ID: mdl-32127473

ABSTRACT

The kappa opioid receptor (κOR) is an important target for pain therapeutics to reduce depression and other harmful side effects of existing medications. The analgesic activity is mediated by κOR signaling through the adenylyl cyclase-inhibitory family of Gi protein. Here, we report the three-dimensional (3D) structure for the active state of human κOR complexed with both heterotrimeric Gi protein and MP1104 agonist. This structure resulted from long molecular dynamics (MD) and metadynamics (metaMD) simulations starting from the 3.1-Å X-ray structure of κOR-MP1104 after replacing the nanobody with the activated Gi protein and from the 3.5-Å cryo-EM structure of µOR-Gi complex after replacing the 168 missing residues. Using MD and metaMD we discovered interactions to the Gi protein with strong anchors to two intracellular loops and transmembrane helix 6 of the κOR. These anchors strengthen the binding, contributing to a contraction in the binding pocket but an expansion in the cytoplasmic region of κOR to accommodate G protein. These remarkable changes in κOR structure reveal that the anchors are essential for activation.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go/chemistry , Morphinans/chemistry , Receptors, Opioid, kappa/chemistry , Analgesics , Binding Sites , Biophysical Phenomena , Humans , Models, Molecular , Molecular Dynamics Simulation , Protein Conformation
17.
Nat Commun ; 11(1): 1145, 2020 03 02.
Article in English | MEDLINE | ID: mdl-32123179

ABSTRACT

Recent studies show that GPCRs rapidly interconvert between multiple states although our ability to interrogate, monitor and visualize them is limited by a relative lack of suitable tools. We previously reported two nanobodies (Nb39 and Nb6) that stabilize distinct ligand- and efficacy-delimited conformations of the kappa opioid receptor. Here, we demonstrate via X-ray crystallography a nanobody-targeted allosteric binding site by which Nb6 stabilizes a ligand-dependent inactive state. As Nb39 stabilizes an active-like state, we show how these two state-dependent nanobodies can provide real-time reporting of ligand stabilized states in cells in situ. Significantly, we demonstrate that chimeric GPCRs can be created with engineered nanobody binding sites to report ligand-stabilized states. Our results provide both insights regarding potential mechanisms for allosterically modulating KOR with nanobodies and a tool for reporting the real-time, in situ dynamic range of GPCR activity.


Subject(s)
Receptors, Opioid, kappa/chemistry , Receptors, Opioid, kappa/metabolism , Single-Domain Antibodies/chemistry , Allosteric Site , Binding Sites , Biosensing Techniques , Crystallography, X-Ray , Cyclic AMP/metabolism , Dynorphins/chemistry , Dynorphins/pharmacology , HEK293 Cells , Humans , Luminescent Measurements/methods , Piperazines/chemistry , Piperazines/pharmacology , Piperidines/chemistry , Piperidines/pharmacology , Protein Conformation , Pyrrolidines/chemistry , Pyrrolidines/pharmacology , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism , Receptors, Opioid, kappa/agonists , Receptors, Opioid, kappa/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Single-Domain Antibodies/metabolism , Tetrahydroisoquinolines/chemistry , Tetrahydroisoquinolines/pharmacology
18.
Dalton Trans ; 48(44): 16476-16492, 2019 Nov 12.
Article in English | MEDLINE | ID: mdl-31599913

ABSTRACT

Bispidones (3,7-diazabicyclo[3.3.1]nonan-9-one) are bicyclic analogues of the natural antiarrhythmic agent, spartein. They can straightforwardly be obtained from two successive Mannich reactions. Reduction of the ketone gives the corresponding bispidol. Substituted bispidones and bispidols offer a large playground by varying the substituents, the configuration of the carbon atoms in position 2 and 4 as well as the conformation of the bicycle. While chair-boat conformers display a strong affinity for κ-opioid receptors, chair-chair bispidines provide adaptable coordination spheres for transition metal and rare-earth ions. Because of their very rich coordination chemistry, substituted bispidines have emerged in various applications of coordination chemistry, such as catalysis, magnetism and medical imaging.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic/chemistry , Bridged Bicyclo Compounds, Heterocyclic/metabolism , Coordination Complexes/chemistry , Receptors, Opioid, kappa/chemistry , Receptors, Opioid, kappa/metabolism , Animals , Coordination Complexes/metabolism , Humans , Metals, Rare Earth/chemistry , Molecular Conformation , Radiotherapy/methods , Transition Elements/chemistry
19.
Sci Rep ; 9(1): 5550, 2019 04 03.
Article in English | MEDLINE | ID: mdl-30944388

ABSTRACT

The tumour suppressor protein RASSF1A is phosphorylated by Aurora A kinase, thereby impairing its tumour suppressor function. Consequently, inhibiting the interaction between Aurora A and RASSF1A may be used for anti-tumour therapy. We used recombinant variants of RASSF1A to map the sites of interaction with Aurora A. The phosphorylation kinetics of three truncated RASSF1A variants has been analysed. Compared to the RASSF1A form lacking the 120 residue long N-terminal part, the Km value of the phosphorylation is increased from 10 to 45 µM upon additional deletion of the C-terminal SARAH domain. On the other hand, deletion of the flexible loop (Δ177-197) that precedes the phosphorylation site/s (T202/S203) results in a reduction of the kcat value from about 40 to 7 min-1. Direct physical interaction between the isolated SARAH domain and Aurora A was revealed by SPR. These data demonstrate that the SARAH domain of RASSF1A is involved in the binding to Aurora A kinase. Structural modelling confirms that a novel complex is feasible between the SARAH domain and the kinase domain of Aurora A. In addition, a regulatory role of the loop in the catalytic phosphorylation reaction has been demonstrated both experimentally and by structural modelling.


Subject(s)
Aurora Kinase A/metabolism , Protein Interaction Domains and Motifs , Receptors, Opioid, kappa/metabolism , Aurora Kinase A/chemistry , Aurora Kinase A/genetics , Binding Sites , Chromatography, Gel , Models, Molecular , Mutation , Phosphorylation , Protein Multimerization , Receptors, Opioid, kappa/chemistry , Receptors, Opioid, kappa/genetics , Surface Plasmon Resonance
20.
J Med Chem ; 61(13): 5751-5757, 2018 07 12.
Article in English | MEDLINE | ID: mdl-29901392

ABSTRACT

Herein we present the expedient synthesis of endomorphin-1 analogues containing stereoisomeric ß2-homo-Freidinger lactam-like scaffolds ([Amo2]EM), and we discuss opioid receptor (OR) affinity, enzymatic stability, functional activity, in vivo antinociceptive effects, and conformational and molecular docking analysis. Hence, H-Tyr-Amo-Trp-PheNH2 resulted to be a new chemotype of highly stable, selective, partial KOR agonist inducing analgesia, therefore displaying great potential interest as a painkiller possibly with reduced harmful side effects.


Subject(s)
Analgesics/chemistry , Analgesics/pharmacology , Dipeptides/chemistry , Heterocyclic Compounds/chemistry , Oligopeptides/chemistry , Oligopeptides/pharmacology , Receptors, Opioid, kappa/agonists , Analgesics/chemical synthesis , Analgesics/metabolism , Animals , Mice , Molecular Docking Simulation , Oligopeptides/chemical synthesis , Oligopeptides/metabolism , Protein Conformation , Receptors, Opioid, kappa/chemistry , Receptors, Opioid, kappa/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL