Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
J Med Chem ; 66(15): 10304-10341, 2023 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-37467430

RESUMEN

A new generation of dual-target µ opioid receptor (MOR) agonist/dopamine D3 receptor (D3R) antagonist/partial agonists with optimized physicochemical properties was designed and synthesized. Combining in vitro cell-based on-target/off-target affinity screening, in silico computer-aided drug design, and BRET functional assays, we identified new structural scaffolds that achieved high affinity and agonist/antagonist potencies for MOR and D3R, respectively, improving the dopamine receptor subtype selectivity (e.g., D3R over D2R) and significantly enhancing central nervous system multiparameter optimization scores for predicted blood-brain barrier permeability. We identified the substituted trans-(2S,4R)-pyrrolidine and trans-phenylcyclopropyl amine as key dopaminergic moieties and tethered these to different opioid scaffolds, derived from the MOR agonists TRV130 (3) or loperamide (6). The lead compounds 46, 84, 114, and 121 have the potential of producing analgesic effects through MOR partial agonism with reduced opioid-misuse liability via D3R antagonism. Moreover, the peripherally limited derivatives could have therapeutic indications for inflammation and neuropathic pain.


Asunto(s)
Analgésicos Opioides , Trastornos Relacionados con Opioides , Humanos , Analgésicos Opioides/farmacología , Analgésicos Opioides/química , Dopamina , Ligandos , Analgésicos/farmacología , Receptores de Dopamina D3/agonistas , Receptores Opioides mu/agonistas
2.
EMBO J ; 42(11): e112940, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-37038975

RESUMEN

The peptide hormone angiotensin II regulates blood pressure mainly through the type 1 angiotensin II receptor AT1 R and its downstream signaling proteins Gq and ß-arrestin. AT1 R blockers, clinically used as antihypertensive drugs, inhibit both signaling pathways, whereas AT1 R ß-arrestin-biased agonists have shown great potential for the treatment of acute heart failure. Here, we present a cryo-electron microscopy (cryo-EM) structure of the human AT1 R in complex with a balanced agonist, Sar1 -AngII, and Gq protein at 2.9 Å resolution. This structure, together with extensive functional assays and computational modeling, reveals the molecular mechanisms for AT1 R signaling modulation and suggests that a major hydrogen bond network (MHN) inside the receptor serves as a key regulator of AT1 R signal transduction from the ligand-binding pocket to both Gq and ß-arrestin pathways. Specifically, we found that the MHN mutations N1113.35 A and N2947.45 A induce biased signaling to Gq and ß-arrestin, respectively. These insights should facilitate AT1 R structure-based drug discovery for the treatment of cardiovascular diseases.


Asunto(s)
Angiotensina II , Transducción de Señal , Humanos , Microscopía por Crioelectrón , Transducción de Señal/fisiología , beta-Arrestinas/metabolismo , Angiotensina II/química , Angiotensina II/metabolismo , Angiotensina II/farmacología , Receptores de Angiotensina/metabolismo
3.
Nature ; 613(7945): 767-774, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36450356

RESUMEN

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.


Asunto(s)
Diseño de Fármacos , Fentanilo , Morfinanos , Receptores Opioides mu , Animales , Ratones , Analgésicos Opioides/química , Analgésicos Opioides/metabolismo , Arrestinas/metabolismo , Microscopía por Crioelectrón , Fentanilo/análogos & derivados , Fentanilo/química , Fentanilo/metabolismo , Ligandos , Morfinanos/química , Morfinanos/metabolismo , Receptores Opioides mu/agonistas , Receptores Opioides mu/química , Receptores Opioides mu/metabolismo , Receptores Opioides mu/ultraestructura , Sitios de Unión , Nocicepción
4.
Handb Exp Pharmacol ; 271: 41-64, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-33945028

RESUMEN

The structure of the human kappa opioid receptor (KOR) in complex with the long-acting antagonist JDTic was solved crystallographically in 2012 and, along with structures of other opioid receptors, revolutionized our understanding of opioid system function and pharmacology. More recently, active state KOR structure was also determined, giving important insights into activation mechanisms of the receptor. In this review, we will discuss how the understanding of atomistic structures of KOR established a key platform for deciphering details of subtype and functional selectivity of KOR-targeting ligands and for discovery of new chemical probes with potentially beneficial pharmacological profiles.


Asunto(s)
Descubrimiento de Drogas , Receptores Opioides kappa , Analgésicos Opioides/farmacología , Humanos , Ligandos , Receptores Opioides
5.
J Med Chem ; 64(21): 16088-16105, 2021 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-34699207

RESUMEN

Linkers are emerging as a key component in regulating the pharmacology of bitopic ligands directed toward G-protein coupled receptors (GPCRs). In this study, the role of regio- and stereochemistry in cyclic aliphatic linkers tethering well-characterized primary and secondary pharmacophores targeting dopamine D2 and D3 receptor subtypes (D2R and D3R, respectively) is described. We introduce several potent and selective D2R (rel-trans-16b; D2R Ki = 4.58 nM) and D3R (rel-cis-14a; D3R Ki = 5.72 nM) agonists while modulating subtype selectivity in a stereospecific fashion, transferring D2R selectivity toward D3R via inversion of the stereochemistry around these cyclic aliphatic linkers [e.g., (-)-(1S,2R)-43 and (+)-(1R,2S)-42]. Pharmacological observations were supported with extensive molecular docking studies. Thus, not only is it an innovative approach to modulate the pharmacology of dopaminergic ligands described, but a new class of optically active cyclic linkers are also introduced, which can be used to expand the bitopic drug design approach toward other GPCRs.


Asunto(s)
Agonistas de Dopamina/farmacología , Receptores de Dopamina D2/efectos de los fármacos , Receptores de Dopamina D3/efectos de los fármacos , Agonistas de Dopamina/química , Células HEK293 , Humanos , Ligandos , Simulación del Acoplamiento Molecular , Ensayo de Unión Radioligante , Estereoisomerismo
6.
J Med Chem ; 64(11): 7778-7808, 2021 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-34011153

RESUMEN

The need for safer pain-management therapies with decreased abuse liability inspired a novel drug design that retains µ-opioid receptor (MOR)-mediated analgesia, while minimizing addictive liability. We recently demonstrated that targeting the dopamine D3 receptor (D3R) with highly selective antagonists/partial agonists can reduce opioid self-administration and reinstatement to drug seeking in rodent models without diminishing antinociceptive effects. The identification of the D3R as a target for the treatment of opioid use disorders prompted the idea of generating a class of ligands presenting bitopic or bivalent structures, allowing the dual-target binding of the MOR and D3R. Structure-activity relationship studies using computationally aided drug design and in vitro binding assays led to the identification of potent dual-target leads (23, 28, and 40), based on different structural templates and scaffolds, with moderate (sub-micromolar) to high (low nanomolar/sub-nanomolar) binding affinities. Bioluminescence resonance energy transfer-based functional studies revealed MOR agonist-D3R antagonist/partial agonist efficacies that suggest potential for maintaining analgesia with reduced opioid-abuse liability.


Asunto(s)
Antagonistas de Dopamina/química , Ligandos , Receptores de Dopamina D3/metabolismo , Receptores Opioides mu/metabolismo , Analgésicos Opioides/uso terapéutico , Animales , Sitios de Unión , Compuestos de Bifenilo/química , Compuestos de Bifenilo/metabolismo , Compuestos de Bifenilo/uso terapéutico , Modelos Animales de Enfermedad , Antagonistas de Dopamina/metabolismo , Antagonistas de Dopamina/uso terapéutico , Diseño de Fármacos , Transferencia Resonante de Energía de Fluorescencia , Ratones , Simulación del Acoplamiento Molecular , Trastornos Relacionados con Opioides/tratamiento farmacológico , Dolor/tratamiento farmacológico , Manejo del Dolor , Receptores de Dopamina D3/agonistas , Receptores de Dopamina D3/antagonistas & inhibidores , Receptores Opioides mu/agonistas , Relación Estructura-Actividad
7.
Elife ; 102021 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-33555255

RESUMEN

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.


Asunto(s)
Morfinanos/química , Receptores Opioides kappa/química , Receptores Opioides mu/química , Secuencias de Aminoácidos , Analgésicos/química , Analgésicos/metabolismo , Animales , Sitios de Unión , Ligandos , Masculino , Ratones , Ratones Endogámicos C57BL , Simulación del Acoplamiento Molecular , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/metabolismo , Relación Estructura-Actividad
8.
Endocrinology ; 162(2)2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33242090

RESUMEN

LA-PTH is a long-acting parathyroid hormone (PTH) peptide analogue in preclinical development for hypoparathyroidism (HP). Like native PTH, LA-PTH contains a methionine at position 8 (Met8) that is predicted to be critical for function. We assessed the impact of Met oxidation on the functional properties of LA-PTH and control PTH ligands. Oxidation of PTH(1-34) resulted in marked (~20-fold) reductions in binding affinity on the PTH receptor-1 (PTHR1) in cell membranes, similarly diminished potency for 3',5'-cyclic AMP signaling in osteoblastic cell lines (SaOS-2 and UMR106), and impaired efficacy for raising blood calcium in mice. Surprisingly, oxidation of LA-PTH resulted in little or no change in these functional responses. The signaling potency of oxidized-LA-PTH was, however, reduced approximately 40-fold compared to LA-PTH in cells expressing a PTHR1 construct that lacks the N-terminal extracellular domain (ECD). Molecular modeling revealed that while Met8 of both LA-PTH and PTH(1-34) is situated within the orthosteric ligand-binding pocket of the receptor's transmembrane domain bundle (TMD), the Met8 sidechain position is shifted for the 2 ligands so that on Met8 oxidation of PTH(1-34), steric clashes occur that are not seen with oxidized LA-PTH. The findings suggest that LA-PTH and PTH(1-34) engage the receptor differently in the Met8-interaction environment of the TMD bundle, and that this interaction environment can be allosterically influenced by the ECD component of the ligand-receptor complex. The findings should be useful for the future development of novel PTH-based peptide therapeutics for diseases of bone and mineral ion metabolism.


Asunto(s)
Hipoparatiroidismo/tratamiento farmacológico , Hormona Paratiroidea/análogos & derivados , Receptor de Hormona Paratiroídea Tipo 1/agonistas , Animales , Calcio/sangre , Línea Celular Tumoral , Evaluación Preclínica de Medicamentos , Femenino , Células HEK293 , Humanos , Metionina/metabolismo , Ratones , Modelos Moleculares , Norleucina , Oxidación-Reducción , Hormona Paratiroidea/metabolismo , Hormona Paratiroidea/farmacología , Hormona Paratiroidea/uso terapéutico , Ratas , Receptor de Hormona Paratiroídea Tipo 1/metabolismo
9.
Pharmacol Rev ; 71(4): 571-595, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31551350

RESUMEN

Endogenous ions play important roles in the function and pharmacology of G-protein coupled receptors (GPCRs). Historically the evidence for ionic modulation of GPCR function dates to 1973 with studies of opioid receptors, where it was demonstrated that physiologic concentrations of sodium allosterically attenuated agonist binding. This Na+-selective effect was distinct from effects of other monovalent and divalent cations, with the latter usually counteracting sodium's negative allosteric modulation of binding. Since then, numerous studies documenting the effects of mono- and divalent ions on GPCR function have been published. While ions can act selectively and nonselectively at many sites in different receptors, the discovery of the conserved sodium ion site in class A GPCR structures in 2012 revealed the unique nature of Na+ site, which has emerged as a near-universal site for allosteric modulation of class A GPCR structure and function. In this review, we synthesize and highlight recent advances in the functional, biophysical, and structural characterization of ions bound to GPCRs. Taken together, these findings provide a molecular understanding of the unique roles of Na+ and other ions as GPCR allosteric modulators. We will also discuss how this knowledge can be applied to the redesign of receptors and ligand probes for desired functional and pharmacological profiles. SIGNIFICANCE STATEMENT: The function and pharmacology of GPCRs strongly depend on the presence of mono and divalent ions in experimental assays and in living organisms. Recent insights into the molecular mechanism of this ion-dependent allosterism from structural, biophysical, biochemical, and computational studies provide quantitative understandings of the pharmacological effects of drugs in vitro and in vivo and open new avenues for the rational design of chemical probes and drug candidates with improved properties.


Asunto(s)
Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/metabolismo , Sodio/metabolismo , Sitio Alostérico , Aniones/química , Aniones/metabolismo , Sitios de Unión , Cationes Bivalentes/química , Cationes Bivalentes/metabolismo , Cationes Monovalentes/química , Cationes Monovalentes/metabolismo , Cloruros/química , Cloruros/metabolismo , Cristalografía por Rayos X , Humanos , Ligandos , Conformación Proteica , Receptores Acoplados a Proteínas G/química , Sodio/química , Relación Estructura-Actividad , Zinc/química , Zinc/metabolismo
10.
Nature ; 560(7720): 666-670, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30135577

RESUMEN

Frizzled receptors (FZDs) are class-F G-protein-coupled receptors (GPCRs) that function in Wnt signalling and are essential for developing and adult organisms1,2. As central mediators in this complex signalling pathway, FZDs serve as gatekeeping proteins both for drug intervention and for the development of probes in basic and in therapeutic research. Here we present an atomic-resolution structure of the human Frizzled 4 receptor (FZD4) transmembrane domain in the absence of a bound ligand. The structure reveals an unusual transmembrane architecture in which helix VI is short and tightly packed, and is distinct from all other GPCR structures reported so far. Within this unique transmembrane fold is an extremely narrow and highly hydrophilic pocket that is not amenable to the binding of traditional GPCR ligands. We show that such a pocket is conserved across all FZDs, which may explain the long-standing difficulties in the development of ligands for these receptors. Molecular dynamics simulations on the microsecond timescale and mutational analysis uncovered two coupled, dynamic kinks located at helix VII that are involved in FZD4 activation. The stability of the structure in its ligand-free form, an unfavourable pocket for ligand binding and the two unusual kinks on helix VII suggest that FZDs may have evolved a novel ligand-recognition and activation mechanism that is distinct from that of other GPCRs.


Asunto(s)
Receptores Frizzled/química , Sitios de Unión , Cristalografía por Rayos X , Cisteína/metabolismo , Proteínas Dishevelled/metabolismo , Receptores Frizzled/genética , Humanos , Ligandos , Modelos Moleculares , Simulación de Dinámica Molecular , Dominios Proteicos , Vía de Señalización Wnt
11.
Cell ; 172(1-2): 55-67.e15, 2018 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-29307491

RESUMEN

The κ-opioid receptor (KOP) mediates the actions of opioids with hallucinogenic, dysphoric, and analgesic activities. The design of KOP analgesics devoid of hallucinatory and dysphoric effects has been hindered by an incomplete structural and mechanistic understanding of KOP agonist actions. Here, we provide a crystal structure of human KOP in complex with the potent epoxymorphinan opioid agonist MP1104 and an active-state-stabilizing nanobody. Comparisons between inactive- and active-state opioid receptor structures reveal substantial conformational changes in the binding pocket and intracellular and extracellular regions. Extensive structural analysis and experimental validation illuminate key residues that propagate larger-scale structural rearrangements and transducer binding that, collectively, elucidate the structural determinants of KOP pharmacology, function, and biased signaling. These molecular insights promise to accelerate the structure-guided design of safer and more effective κ-opioid receptor therapeutics.


Asunto(s)
Simulación del Acoplamiento Molecular , Receptores Opioides kappa/química , Analgésicos/química , Analgésicos/farmacología , Animales , Sitios de Unión , Células HEK293 , Humanos , Simulación de Dinámica Molecular , Morfinanos/química , Morfinanos/farmacología , Unión Proteica , Estabilidad Proteica , Receptores Opioides kappa/agonistas , Receptores Opioides kappa/metabolismo , Células Sf9 , Spodoptera
12.
Structure ; 26(1): 171-180.e2, 2018 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-29249607

RESUMEN

Playing a central role in cell signaling, G-protein-coupled receptors (GPCRs) are the largest superfamily of membrane proteins and form the majority of drug targets in humans. How extracellular agonist binding triggers the activation of GPCRs and associated intracellular effector proteins remains, however, poorly understood. Structural studies have revealed that inactive class A GPCRs harbor a conserved binding site for Na+ ions in the center of their transmembrane domain, accessible from the extracellular space. Here, we show that the opening of a conserved hydrated channel in the activated state receptors allows the Na+ ion to egress from its binding site into the cytosol. Coupled with protonation changes, this ion movement occurs without significant energy barriers, and can be driven by physiological transmembrane ion and voltage gradients. We propose that Na+ ion exchange with the cytosol is a key step in GPCR activation. Further, we hypothesize that this transition locks receptors in long-lived active-state conformations.


Asunto(s)
Carbacol/química , Fosfatidilcolinas/química , Receptor Muscarínico M2/química , Sodio/química , Secuencias de Aminoácidos , Sitios de Unión , Carbacol/metabolismo , Cationes Monovalentes , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Activación del Canal Iónico , Transporte Iónico , Cinética , Ligandos , Simulación de Dinámica Molecular , Fosfatidilcolinas/metabolismo , Unión Proteica , Conformación Proteica en Hélice alfa , Dominios y Motivos de Interacción de Proteínas , Receptor Muscarínico M2/metabolismo , Sodio/metabolismo , Electricidad Estática , Termodinámica
13.
J Mol Graph Model ; 77: 189-199, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28869863

RESUMEN

Scavenger receptor A (SRA), as an immune regulator, has been shown to play important roles in lipid metabolism, cardiovascular diseases, and pathogen recognition. Several natural product inhibitors of SRA have been studied for their potential application in modulating SRA functions. To understand the binding mode of these inhibitors on SRA, we conducted systematic molecular modeling studies in order to identify putative binding domain(s) that may be responsible for their recognition to the receptor as well as their inhibitory activity. Treatment of SRA with one of the natural product inhibitors, rhein, led to significant dissociation of SRA oligomers to its trimer and dimer forms, which further supported our hypothesis on their putative mechanism of action. Such information is believed to shed light on design of more potent inhibitors for the receptor in order to develop potential therapeutics through immune system modulation.


Asunto(s)
Antraquinonas/química , Modelos Moleculares , Receptores Depuradores/química , Antraquinonas/farmacología , Sitios de Unión , Humanos , Unión Proteica , Multimerización de Proteína/efectos de los fármacos , Receptores Depuradores/antagonistas & inhibidores
14.
Elife ; 62017 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-28771403

RESUMEN

The activation mechanism of class B G-protein-coupled receptors (GPCRs) remains largely unknown. To characterize conformational changes induced by peptide hormones, we investigated interactions of the class B corticotropin-releasing factor receptor type 1 (CRF1R) with two peptide agonists and three peptide antagonists obtained by N-truncation of the agonists. Surface mapping with genetically encoded photo-crosslinkers and pair-wise crosslinking revealed distinct footprints of agonists and antagonists on the transmembrane domain (TMD) of CRF1R and identified numerous ligand-receptor contact sites, directly from the intact receptor in live human cells. The data enabled generating atomistic models of CRF- and CRF(12-41)-bound CRF1R, further explored by molecular dynamics simulations. We show that bound agonist and antagonist adopt different folds and stabilize distinct TMD conformations, which involves bending of helices VI and VII around flexible glycine hinges. Conservation of these glycine hinges among all class B GPCRs suggests their general role in activation of these receptors.


Asunto(s)
Hormona Liberadora de Corticotropina/metabolismo , Receptores de Hormona Liberadora de Corticotropina/química , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Sitios de Unión , Reactivos de Enlaces Cruzados/metabolismo , Células HEK293 , Humanos , Ligandos , Modelos Moleculares , Simulación de Dinámica Molecular , Unión Proteica , Dominios Proteicos , Estructura Secundaria de Proteína
15.
Bioorg Med Chem ; 25(8): 2463-2471, 2017 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-28302509

RESUMEN

Mu opioid receptor selective antagonists are highly desirable because of their utility as pharmacological probes for receptor characterization and functional studies. Furthermore, the mu opioid receptors act as an important target in drug abuse and addiction treatment. Previously, we reported NAP as a novel lead compound with high selectivity and affinity towards the mu opioid receptor. Based on NAP, we have synthesized its derivatives and further characterized their binding affinities and selectivity towards the receptor. NMP and NGP were identified as the two most selective MOR ligands among NAP derivatives. In the present study, molecular modeling methods were applied to assess the dual binding modes of NAP derivatives, particularly on NMP and NGP, in three opioid receptors, in order to analyze the effects of structural modifications on the pyridyl ring of NAP on the binding affinity and selectivity. The results indicated that the steric hindrance, electrostatic, and hydrophobic effects caused by the substituents on the pyridyl ring of NAP contributed complimentarily on the binding affinity and selectivity of NAP derivatives to three opioid receptors. Analyses of these contributions provided insights on future design of more potent and selective mu opioid receptor ligands.


Asunto(s)
Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Receptores Opioides mu/metabolismo , Interacciones Hidrofóbicas e Hidrofílicas , Ligandos , Electricidad Estática
16.
J Med Chem ; 60(7): 3070-3081, 2017 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-28339199

RESUMEN

The ongoing epidemics of opioid overdose raises an urgent need for effective antiaddiction therapies and addiction-free painkillers. The κ-opioid receptor (KOR) has emerged as a promising target for both indications, raising demand for new chemotypes of KOR antagonists as well as G-protein-biased agonists. We employed the crystal structure of the KOR-JDTic complex and ligand-optimized structural templates to perform virtual screening of available compound libraries for new KOR ligands. The prospective virtual screening campaign yielded a high 32% hit rate, identifying novel fragment-like and lead-like chemotypes of KOR ligands. A round of optimization resulted in 11 new submicromolar KOR binders (best Ki = 90 nM). Functional assessment confirmed at least two compounds as potent KOR antagonists, while compound 81 was identified as a potent Gi biased agonist for KOR with minimal ß-arrestin recruitment. These results support virtual screening as an effective tool for discovery of new lead chemotypes with therapeutically relevant functional profiles.


Asunto(s)
Diseño de Fármacos , Antagonistas de Narcóticos/química , Antagonistas de Narcóticos/farmacología , Receptores Opioides kappa/agonistas , Receptores Opioides kappa/antagonistas & inhibidores , Animales , Células HEK293 , Humanos , Simulación del Acoplamiento Molecular , Estudios Prospectivos , Receptores Opioides kappa/metabolismo , Relación Estructura-Actividad
17.
ACS Chem Neurosci ; 7(8): 1120-9, 2016 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-27269866

RESUMEN

The 6ß-N-heterocyclic naltrexamine derivative, NAP, has been demonstrated to be a peripherally selective mu opioid receptor modulator. To further improve peripheral selectivity of this highly potent ligand, its pyridal ring was quaterinized with benzyl bromide to produce BNAP. In radioligand binding assay, the Ki of BNAP for MOR was 0.76 ± 0.09 nM and was >900-fold more selective for MOR than DOR. The Ki for KOR was 3.46 ± 0.05 nM. In [(35)S]GTPγS ligand stimulated assay, BNAP showed low agonist efficacy with 14.6% of the maximum response of DAMGO with an EC50 of 4.84 ± 0.6 nM. However, unlike its parent compound NAP, BNAP displayed partial agonist activity at KOR with % maximum response at 45.9 ± 1.7% of U50,488H. BNAP did not reverse morphine-induced antinociception when administered subcutaneously but did antagonize when administered intracerebroventricularly. BNAP antagonized morphine-induced contractions of the circular muscle in mice colon. BNAP inhibition of field-stimulated contractions in longitudinal muscle strips for the guinea-pig ileum were also blocked by nor-BNI, a kappa opioid receptor antagonist. BNAP induced inhibition of acetic acid induced abdominal stretching in chronic morphine treated mice. These findings suggest that BNAP is a dual MOR antagonist/KOR agonist and may have functional use in irritable bowel patients.


Asunto(s)
Músculo Esquelético/efectos de los fármacos , Músculo Liso/efectos de los fármacos , Naltrexona/análogos & derivados , Naltrexona/farmacología , Receptores Opioides kappa/antagonistas & inhibidores , Receptores Opioides mu/antagonistas & inhibidores , Animales , Unión Competitiva/efectos de los fármacos , Células CHO , Cricetulus , Modelos Animales de Enfermedad , Motilidad Gastrointestinal/efectos de los fármacos , Guanosina 5'-O-(3-Tiotrifosfato)/farmacocinética , Cobayas , Íleon/anatomía & histología , Técnicas In Vitro , Ligandos , Masculino , Ratones , Contracción Muscular/efectos de los fármacos , Naltrexona/síntesis química , Naltrexona/química , Neuronas/efectos de los fármacos , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/metabolismo , Relación Estructura-Actividad
18.
ACS Chem Neurosci ; 7(3): 297-304, 2016 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-26716358

RESUMEN

Mounting evidence has suggested that G protein-coupled receptors can be stabilized in multiple conformations in response to distinct ligands, which exert discrete functions through selective activation of various downstream signaling events. In accordance with this concept, we report biased signaling of one C6-heterocyclic substituted naltrexamine derivative, namely, 17-cyclopropylmethyl-3,14ß-dihydroxy-4,5α-epoxy-6ß-(4'-pyridylcarboxamido)morphinan (NAP) at the mu opioid receptor (MOR). NAP acted as a low efficacy MOR partial agonist in the G protein-mediated [(35)S]GTPγS binding assay, whereas it did not significantly induce calcium flux or ß-arrestin2 recruitment. In contrast, it potently blocked MOR full agonist-induced ß-arrestin2 recruitment and translocation. Additionally, NAP dose-dependently antagonized MOR full agonist-induced intracellular calcium flux and ß-arrestin2 recruitment. Further results in an isolated organ bath preparation confirmed that NAP reversed the morphine-induced reduction in colon motility. Ligand docking and dynamics simulation studies of NAP at the MOR provided more supporting evidence for biased signaling of NAP at an atomic level. Due to the fact that NAP is MOR selective and preferentially distributed peripherally upon systemic administration while ß-arrestin2 is reportedly required for impairment of intestinal motility by morphine, biased antagonism of ß-arrestin2 recruitment by NAP further supports its utility as a treatment for opioid-induced constipation.


Asunto(s)
Analgésicos Opioides/farmacología , Modelos Moleculares , Receptores Opioides mu/agonistas , Analgésicos Opioides/química , Animales , Células CHO , Línea Celular , Cricetulus , Motilidad Gastrointestinal/efectos de los fármacos , Humanos , Masculino , Ratones , Microscopía Confocal , Morfinanos/química , Morfinanos/farmacología , Receptores Opioides mu/metabolismo
19.
J Nat Prod ; 78(8): 1859-67, 2015 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-26270954

RESUMEN

The involvement of the neurotransmitter serotonin (5-HT) in numerous physiological functions is often attributed to the diversity of receptors with which it interacts. Ligands targeting serotonin receptor 2B (5-HT2B) have received renewed interest for their potential to help understand the role of 5-HT2B in migraines, drug abuse, neurodegenerative diseases, and irritable bowel syndrome. To date, most of the ligands targeting 5-HT2B have been nitrogen-containing compounds. The natural product 5-hydroxy-2-(2-phenylethyl)chromone (5-HPEC, 5) has been shown previously to act as a non-nitrogenous antagonist for the 5-HT2B receptor (pKi = 5.6). This report describes further progress on the study of the structure-activity relationship of both naturally occurring and synthetic compounds bearing the 2-(2-phenylethyl)chromone scaffold at the 5-HT2B receptor. The inhibitory activity of the newly synthesized compounds (at 10 µM) was tested against each of the 5-HT2 receptors. Following this assay, the binding affinity and antagonism of the most promising compounds were then evaluated at 5-HT2B. Among all the analogues, 5-hydroxy-2-(2-phenylpropyl)chromone (5-HPPC, 22h) emerged as a new lead compound, showing a 10-fold improvement in affinity (pKi = 6.6) over 5-HPEC with reasonable antagonist properties at 5-HT2B. Additionally, ligand docking studies have identified a putative binding pocket for 5-HPPC and have helped understand its improved affinity.


Asunto(s)
Cromonas/química , Cromonas/síntesis química , Flavonoides/química , Receptor de Serotonina 5-HT2B/metabolismo , Serotonina/metabolismo , Cromonas/farmacología , Técnicas Químicas Combinatorias , Humanos , Ligandos , Modelos Moleculares , Estructura Molecular , Relación Estructura-Actividad
20.
Bioorg Med Chem Lett ; 25(17): 3721-5, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26096680

RESUMEN

Prostate cancer is one of the leading causes of death among males in the world. Prostate cancer cells have been shown to express upregulated chemokine receptor CCR5, a G protein-coupled receptor (GPCR) that relates to the inflammation process. Anibamine, a natural product containing a pyridine ring and two aliphatic side chains, was shown to carry a binding affinity of 1 µM at CCR5 as an antagonist with potential anti-cancer activity. However, it is not drug-like according to the Lipinski's rule of five mainly due to its two long aliphatic side chains. In our effort to improve its drug-like property, a series of anibamine derivatives were designed and synthesized by placement of aromatic side chains through an amide linkage to the pyridine ring. The newly synthesized compounds were tested for their CCR5 affinity and antagonism, and potential anti-proliferation activity against prostate cancer cell lines. Basal cytotoxicity was finally studied for compounds showing potent anti-proliferation activity. It was found that compounds with hydrophobic substitutions on the aromatic systems seemed to carry more promising CCR5 binding and prostate cancer cell proliferation inhibition activities.


Asunto(s)
Antineoplásicos/farmacología , Antagonistas de los Receptores CCR5/química , Antagonistas de los Receptores CCR5/farmacología , Neoplasias de la Próstata/tratamiento farmacológico , Piridinas/química , Animales , Antineoplásicos/química , Línea Celular Tumoral/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Técnicas de Química Sintética , Ensayos de Selección de Medicamentos Antitumorales/métodos , Humanos , Masculino , Ratones , Simulación del Acoplamiento Molecular , Células 3T3 NIH/efectos de los fármacos , Neoplasias de la Próstata/patología , Receptores CCR5/química , Receptores CCR5/metabolismo , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA