Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 212
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Anal Chem ; 95(5): 2848-2856, 2023 02 07.
Article in English | MEDLINE | ID: mdl-36700797

ABSTRACT

For the precise visualization of GPCR, subtype selectivity of turn-on fluorescent ligands is of major relevance. Although there are many thriving ß-adrenergic receptors (ß-ARs) probes, none of them are selective to the ß3-subtype, which severely limits the development of ß3-AR investigations. Using a polyethylene glycol (PEG) chain to conjugate the Py-5 fluorophore with mirabegron, we present here a highly selective fluorescent ligand, H2, for ß3-AR. It was established by the radioligand and NanoLuc-based bioluminescence resonance energy transfer (NanoBRET) binding experiments that molecule H2 has a substantially higher affinity for ß3-AR than the other two subtypes (1/3, 45-fold; 2/3, 16-fold). More crucially, when molecule H2 was incubated with ß3-AR, the turn-on fluorescent signals could be quickly released. The subsequent investigations, which included cell imaging, tissue imaging, and flow-cytometry analysis, proved that molecule H2 may make it possible to quickly and accurately fluorescently identify ß3-AR at different levels. We offer a prospective fluorescent turn-on ligand with exceptional selectivity for ß3-AR as a result of our combined efforts.


Subject(s)
Adrenergic beta-Agonists , Receptors, Adrenergic, beta , Ligands , Prospective Studies , Receptors, Adrenergic, beta/chemistry , Receptors, Adrenergic, beta/metabolism
2.
Arch Insect Biochem Physiol ; 106(4): e21772, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33719088

ABSTRACT

The ß-adrenergic-like octopamine receptor (OA2B2), which binds the biogenic amine octopamine, belongs to the class of G-protein coupled receptors and significantly regulates many physiological and behavioral processes in insects. In this study, the putative open reading frame sequence of the MsOA2B2 gene in Mythimna separata was cloned, the full-length complementary DNA was 1191 bp and it encoded a 396-amino acid protein (GenBank accession number MN822800). Orthologous sequence alignment, phylogenetic tree analysis, and protein sequence analysis all showed that the cloned receptor belongs to the OA2B2 protein family. Real-time quantitative polymerase chain reaction of spatial and temporal expression analysis revealed that the MsOAB2 gene was expressed in all developmental stages of M. separata and was most abundant in egg stages and second and fourth instars compared with other developmental stages, while the expression level during the pupal stage was much lower than that at the other stages. Further analysis with sixth instar M. separata larvae showed that the MsOA2B2 gene was expressed 1.81 times higher in the head than in integument and gut tissues. Dietary ingestion of dsMsOA2B2 significantly reduced the messenger RNA level of MsOA2B2 and decreased mortality following amitraz treatment. This study provides both a pharmacological characterization and the gene expression patterns of OA2B2 in M. separata, facilitating further research for insecticides using MsOA2B2 as a target.


Subject(s)
Moths/genetics , Receptors, Biogenic Amine , Animals , Gene Expression/drug effects , Genes, Insect , Insect Control , Insect Proteins/chemistry , Insect Proteins/genetics , Insect Proteins/metabolism , Insecticides/pharmacology , Larva/genetics , Larva/metabolism , Moths/metabolism , Phylogeny , Pupa/genetics , Pupa/metabolism , Receptors, Adrenergic, beta/chemistry , Receptors, Adrenergic, beta/drug effects , Receptors, Adrenergic, beta/genetics , Receptors, Adrenergic, beta/metabolism , Receptors, Biogenic Amine/chemistry , Receptors, Biogenic Amine/drug effects , Receptors, Biogenic Amine/genetics , Receptors, Biogenic Amine/metabolism , Toluidines/pharmacology
3.
J Recept Signal Transduct Res ; 40(5): 395-409, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32456520

ABSTRACT

ß adrenergic receptors mediate effects via activation of G proteins, transactivation of membrane growth factor receptors, or ß adrenergic receptor-ß arrestin-facilitated scaffold-mediated signaling. Agonist occupancy of the ß adrenergic receptor induces desensitization by promoting ß adrenergic receptor kinase phosphorylation of the carboxyl terminal domain, facilitating binding of the amino terminal of the ß arrestin, which sterically inhibits interactions between ß adrenergic receptors and G proteins and induces clathrin-coated pit-mediated receptor endocytosis. Scaffold formation promoted by ß arrestin binding to the ß adrenergic receptor activates extracellular regulated kinase 1/2 in a manner which elicits cytosolic retention of, and prevents promotion of nuclear transcriptional activity by, mitogen-activated protein kinase. The ß adrenergic receptor kinase also interacts with a yet to be determined microsomal membrane protein via high-affinity electrostatic interactions. We evaluate ß adrenergic receptor structure, function, and downstream signaling and ß arrestin-mediated desensitization, receptor endocytosis, and scaffold-facilitated signal transduction in order to illumine therapeutic strategies designed to modulate these pathways. We trust these approaches may arm us with the capacity to selectively modulate signal transduction pathways regulating cellular proliferation, immunogenicity, angiogenesis, and invasive and metastatic potential implicated in cancer initiation, promotion, and progression.


Subject(s)
GTP-Binding Proteins/genetics , Neoplasms/genetics , Receptors, Adrenergic, beta/genetics , beta-Arrestins/genetics , Adrenergic beta-Agonists/therapeutic use , Clathrin-Coated Vesicles/genetics , Endocytosis/genetics , Humans , Molecular Targeted Therapy/trends , Neoplasms/drug therapy , Neoplasms/pathology , Receptors, Adrenergic, beta/chemistry , Receptors, Adrenergic, beta/ultrastructure , Structure-Activity Relationship , beta-Arrestins/antagonists & inhibitors
4.
Cell Mol Biol (Noisy-le-grand) ; 66(4): 243-249, 2020 Jun 25.
Article in English | MEDLINE | ID: mdl-32583783

ABSTRACT

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


Subject(s)
Computer Simulation , Diarrhea/drug therapy , Models, Biological , Phytol/therapeutic use , Amino Acid Sequence , Animals , Castor Oil , Disease Models, Animal , Fasting , Humans , Ion Channels/chemistry , Ion Channels/metabolism , Male , Mice , Molecular Docking Simulation , Phytol/pharmacology , Receptors, Adrenergic, alpha-1/chemistry , Receptors, Adrenergic, alpha-1/metabolism , Receptors, Adrenergic, beta/chemistry , Receptors, Adrenergic, beta/metabolism , Receptors, Opioid, mu/chemistry , Receptors, Opioid, mu/metabolism , Sequence Homology, Amino Acid
5.
Nanomedicine ; 27: 102199, 2020 07.
Article in English | MEDLINE | ID: mdl-32275957

ABSTRACT

Carvedilol (CAR), a ß-adrenoceptor and α1-receptor blocker, has pH-dependent solubility, which greatly limits its oral bioavailability. In this work, a precipitation inhibitor-based self-nanoemulsifying drug delivery system (PI-SNEDDS) was developed by employing Soluplus and Poloxamer 407 to improve drug dissolution and to inhibit drug precipitation in the gastrointestinal tract. In vitro phase distribution and in vivo dissolution studies indicated that PI-SNEDDS significantly increased drug content in the oil phase of the nanoemulsions in the stomach and greatly inhibited the subsequent precipitation of CAR in the intestine compared with the carvedilol self-nanoemulsifying drug delivery system (CAR SNEDDS) and the carvedilol tablets. Moreover, a 1.56-fold increase in the relative bioavailability of CAR was observed for the CAR PI-SNEDDS (397.41%) compared to a CAR SNEDDS (254.09%) with commercial capsules as a reference. Therefore, our developed PI-SNEDDS is a promising vehicle for improving the dissolution and bioavailability of poorly soluble drugs with pH-dependent solubility.


Subject(s)
Carvedilol/pharmacology , Drug Delivery Systems , Gastrointestinal Tract/drug effects , Nanoparticles/chemistry , Receptors, Adrenergic, beta/genetics , Administration, Oral , Animals , Biological Availability , Carvedilol/chemistry , Gastrointestinal Tract/pathology , Humans , Poloxamer/chemistry , Poloxamer/pharmacology , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacology , Polyvinyls/chemistry , Polyvinyls/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, alpha-1/genetics , Receptors, Adrenergic, beta/chemistry
6.
J Physiol ; 596(2): 231-252, 2018 01 15.
Article in English | MEDLINE | ID: mdl-28983994

ABSTRACT

KEY POINTS: While several studies have investigated the effects of exercise training in human skeletal muscle and the chronic effect of ß2 -agonist treatment in rodent muscle, their effects on muscle proteome signature with related functional measures in humans are still incompletely understood. Herein we show that daily ß2 -agonist treatment attenuates training-induced enhancements in exercise performance and maximal oxygen consumption, and alters muscle proteome signature and phenotype in trained young men. Daily ß2 -agonist treatment abolished several of the training-induced enhancements in muscle oxidative capacity and caused a repression of muscle metabolic pathways; furthermore, ß2 -agonist treatment induced a slow-to-fast twitch muscle phenotype transition. The present study indicates that chronic ß2 -agonist treatment confounds the positive effect of high intensity training on exercise performance and oxidative capacity, which is of interest for the large proportion of persons using inhaled ß2 -agonists on a daily basis, including athletes. ABSTRACT: Although the effects of training have been studied for decades, data on muscle proteome signature remodelling induced by high intensity training in relation to functional changes in humans remains incomplete. Likewise, ß2 -agonists are frequently used to counteract exercise-induced bronchoconstriction, but the effects ß2 -agonist treatment on muscle remodelling and adaptations to training are unknown. In a placebo-controlled parallel study, we randomly assigned 21 trained men to 4 weeks of high intensity training with (HIT+ß2 A) or without (HIT) daily inhalation of ß2 -agonist (terbutaline, 4 mg dose-1 ). Of 486 proteins identified by mass-spectrometry proteomics of muscle biopsies sampled before and after the intervention, 32 and 85 were changing (false discovery rate (FDR) ≤5%) with the intervention in HIT and HIT+ß2 A, respectively. Proteome signature changes were different in HIT and HIT+ß2 A (P = 0.005), wherein ß2 -agonist caused a repression of 25 proteins in HIT+ß2 A compared to HIT, and an upregulation of 7 proteins compared to HIT. ß2 -Agonist repressed or even downregulated training-induced enrichment of pathways related to oxidative phosphorylation and glycogen metabolism, but upregulated pathways related to histone trimethylation and the nucleosome. Muscle contractile phenotype changed differently in HIT and HIT+ß2 A (P ≤ 0.001), with a fast-to-slow twitch transition in HIT and a slow-to-fast twitch transition in HIT+ß2 A. ß2 -Agonist attenuated training-induced enhancements in maximal oxygen consumption (P ≤ 0.01) and exercise performance (6.1 vs. 11.6%, P ≤ 0.05) in HIT+ß2 A compared to HIT. These findings indicate that daily ß2 -agonist treatment attenuates the beneficial effects of high intensity training on exercise performance and oxidative capacity, and causes remodelling of muscle proteome signature towards a fast-twitch phenotype.


Subject(s)
Adaptation, Physiological , Adrenergic beta-2 Receptor Agonists/pharmacology , Exercise , Muscle, Skeletal/physiology , Proteome/metabolism , Receptors, Adrenergic, beta/chemistry , Terbutaline/pharmacology , Adolescent , Adult , Humans , Male , Muscle Contraction , Muscle, Skeletal/drug effects , Oxygen Consumption , Physical Endurance , Proteome/drug effects , Young Adult
7.
J Biol Chem ; 292(28): 11815-11828, 2017 07 14.
Article in English | MEDLINE | ID: mdl-28515323

ABSTRACT

In liver steatosis (i.e. fatty liver), hepatocytes accumulate many large neutral lipid storage organelles known as lipid droplets (LDs). LDs are important in the maintenance of energy homeostasis, but the signaling mechanisms that stimulate LD metabolism in hepatocytes are poorly defined. In adipocytes, catecholamines target the ß-adrenergic (ß-AR)/cAMP pathway to activate cytosolic lipases and induce their recruitment to the LD surface. Therefore, the goal of this study was to determine whether hepatocytes, like adipocytes, also undergo cAMP-mediated lipolysis in response to ß-AR stimulation. Using primary rat hepatocytes and human hepatoma cells, we found that treatment with the ß-AR agent isoproterenol caused substantial LD loss via activation of cytosolic lipases adipose triglyceride lipase (ATGL) and hormone-sensitive lipase (HSL). ß-Adrenergic stimulation rapidly activated PKA, which led to the phosphorylation of ATGL and HSL and their recruitment to the LD surface. To test whether this ß-AR-dependent lipolysis pathway was altered in a model of alcoholic fatty liver, primary hepatocytes from rats fed a 6-week EtOH-containing Lieber-DeCarli diet were treated with cAMP agonists. Compared with controls, EtOH-exposed hepatocytes showed a drastic inhibition in ß-AR/cAMP-induced LD breakdown and the phosphorylation of PKA substrates, including HSL. This observation was supported in VA-13 cells, an EtOH-metabolizing human hepatoma cell line, which displayed marked defects in both PKA activation and isoproterenol-induced ATGL translocation to the LD periphery. In summary, these findings suggest that ß-AR stimulation mobilizes cytosolic lipases for LD breakdown in hepatocytes, and perturbation of this pathway could be a major consequence of chronic EtOH insult leading to fatty liver.


Subject(s)
Adrenergic beta-Agonists/pharmacology , Cyclic AMP/agonists , Fatty Liver, Alcoholic/metabolism , Hepatocytes/drug effects , Lipolysis/drug effects , Receptors, Adrenergic, beta/metabolism , Second Messenger Systems/drug effects , Animals , Cell Line, Tumor , Cells, Cultured , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/chemistry , Cyclic AMP-Dependent Protein Kinases/metabolism , Enzyme Activation/drug effects , Fatty Liver, Alcoholic/pathology , Female , Hepatocytes/cytology , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Lipase/chemistry , Lipase/metabolism , Lipid Droplets/drug effects , Lipid Droplets/metabolism , Lipid Droplets/pathology , Male , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Protein Transport/drug effects , Rats , Receptors, Adrenergic, beta/chemistry , Sterol Esterase/chemistry , Sterol Esterase/metabolism
8.
Bioorg Med Chem ; 26(13): 3773-3784, 2018 07 30.
Article in English | MEDLINE | ID: mdl-29706529

ABSTRACT

A series of aminoisopropanoloxy derivatives of xanthone has been synthesized and their pharmacological properties regarding the cardiovascular system has been evaluated. Radioligand binding and functional studies in isolated organs revealed that title compounds present high affinity and antagonistic potency for α1-(compound 2 and 8), ß-(compounds 1, 3, 4, 7), α1/ß-(compounds 5 and 6) adrenoceptors. Furthermore, compound 7, the structural analogue of verapamil, possesses calcium entry blocking activity. The title compounds showed hypotensive and antiarrhythmic properties due to their adrenoceptor blocking effect. Moreover, they did not affect QRS and QT intervals, and they did not have proarrhythmic potential at tested doses. In addition they exerted anti-aggregation effect. The results of this study suggest that new compounds with multidirectional activity in cardiovascular system might be found in the group of xanthone derivatives.


Subject(s)
Adrenergic Antagonists/chemical synthesis , Drug Design , Xanthones/chemistry , Adrenergic Antagonists/metabolism , Adrenergic Antagonists/pharmacology , Animals , Blood Pressure/drug effects , Calcium Channel Blockers/chemical synthesis , Calcium Channel Blockers/metabolism , Calcium Channel Blockers/pharmacology , Calcium Channels/chemistry , Calcium Channels/metabolism , Heart Rate/drug effects , Inhibitory Concentration 50 , Male , Platelet Aggregation/drug effects , Radioligand Assay , Rats , Rats, Wistar , Receptors, Adrenergic, alpha/chemistry , Receptors, Adrenergic, alpha/metabolism , Receptors, Adrenergic, beta/chemistry , Receptors, Adrenergic, beta/metabolism , Structure-Activity Relationship , Verapamil/chemistry , Xanthones/metabolism , Xanthones/pharmacology
9.
Biophys J ; 113(1): 60-72, 2017 Jul 11.
Article in English | MEDLINE | ID: mdl-28700926

ABSTRACT

The visual photoreceptor rhodopsin is a prototypical G-protein-coupled receptor (GPCR) that stabilizes its inverse agonist ligand, 11-cis-retinal (11CR), by a covalent, protonated Schiff base linkage. In the visual dark adaptation, the fundamental molecular event after photobleaching of rhodopsin is the recombination reaction between its apoprotein opsin and 11CR. Here we present a detailed analysis of the kinetics and thermodynamics of this reaction, also known as the "regeneration reaction". We compared the regeneration of purified rhodopsin reconstituted into phospholipid/detergent bicelles with rhodopsin reconstituted into detergent micelles. We found that the lipid bilayer of bicelles stabilized the chromophore-free opsin over the long timescale required for the regeneration experiments, and also facilitated the ligand reuptake binding reaction. We utilized genetic code expansion and site-specific bioorthogonal labeling of rhodopsin with Alexa488 to enable, to our knowledge, a novel fluorescence resonance energy transfer-based measurement of the binding kinetics between opsin and 11CR. Based on these results, we report a complete energy diagram for the regeneration reaction of rhodopsin. We show that the dissociation reaction of rhodopsin to 11CR and opsin has a 25-pM equilibrium dissociation constant, which corresponds to only 0.3 kcal/mol stabilization compared to the noncovalent, tightly bound antagonist-GPCR complex of iodopindolol and ß-adrenergic receptor. However, 11CR dissociates four orders-of-magnitude slower than iodopindolol, which corresponds to a 6-kcal/mol higher dissociation free energy barrier. We further used isothermal titration calorimetry to show that ligand binding in rhodopsin is enthalpy driven with -22 kcal/mol, which is 12 kcal/mol more stable than the antagonist-GPCR complex. Our data provide insights into the ligand-receptor binding reaction for rhodopsin in particular, and for GPCRs more broadly.


Subject(s)
Retinaldehyde/metabolism , Rhodopsin/metabolism , Animals , Calorimetry , Cattle , Cholic Acids/chemistry , Dynamic Light Scattering , Fluorescence Resonance Energy Transfer , Hydrodynamics , Kinetics , Lipid Bilayers/chemistry , Micelles , Phosphatidylcholines/chemistry , Photobleaching , Protein Binding , Protein Stability , Receptors, Adrenergic, beta/chemistry , Receptors, Adrenergic, beta/metabolism , Retinaldehyde/chemistry , Rhodopsin/agonists , Rhodopsin/chemistry , Thermodynamics , Water/chemistry
10.
Magn Reson Med ; 77(5): 1810-1817, 2017 05.
Article in English | MEDLINE | ID: mdl-27173806

ABSTRACT

PURPOSE: To demonstrate the feasibility of mapping intracellular pH within the in vivo rodent heart. Alterations in cardiac acid-base balance can lead to acute contractile depression and alterations in Ca2+ signaling. The transient reduction in adenosine triphosphate (ATP) consumption and cardiac contractility may be initially beneficial; however, sustained pH changes can be maladaptive, leading to myocardial damage and electrical arrhythmias. METHODS: Spectrally selective radiofrequency (RF) pulses were used to excite the HCO3- and CO2 resonances individually while preserving signal from the injected hyperpolarized [1-13 C]pyruvate. The large flip angle pulses were placed within a three-dimensional (3D) imaging acquisition, which exploited CA-mediated label exchange between HCO3- and CO2 . Images at 4.5 × 4.5 × 5 mm3 resolution were obtained in the in vivo rodent heart. The technique was evaluated in healthy rodents scanned at baseline and during high cardiac workload induced by dobutamine infusion. RESULTS: The intracellular pH was measured to be 7.15 ± 0.04 at baseline, and decreased to 6.90 ± 0.06 following 15 min of continuous ß-adrenergic stimulation. CONCLUSIONS: Volumetric maps of intracellular pH can be obtained following an injection of hyperpolarized [1-13 C]pyruvate. The new method is anticipated to enable assessment of stress-inducible ischemia and potential ventricular arrythmogenic substrates within the ischemic heart. Magn Reson Med 77:1810-1817, 2017. © 2016 The Authors Magnetic Resonance in Medicine published by Wiley Periodicals, Inc. on behalf of International Society for Magnetic Resonance in Medicine. This is an open access article under the terms of the Creative Commons Attribution License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.


Subject(s)
Carbon Isotopes/chemistry , Myocardium/pathology , Pyruvic Acid/chemistry , Adenosine Triphosphate/chemistry , Animals , Arrhythmias, Cardiac/pathology , Calcium Signaling , Carbon Dioxide/chemistry , Carbonic Anhydrases/chemistry , Cytosol/metabolism , Dobutamine/chemistry , Heart/physiology , Hydrogen-Ion Concentration , Image Processing, Computer-Assisted , Imaging, Three-Dimensional , Magnetic Resonance Imaging , Male , Myocardial Ischemia/pathology , Radio Waves , Rats , Rats, Wistar , Receptors, Adrenergic, beta/chemistry
11.
J Biol Chem ; 290(1): 272-83, 2015 Jan 02.
Article in English | MEDLINE | ID: mdl-25414258

ABSTRACT

G protein-coupled receptors (GPCRs) relay extracellular signals mainly to heterotrimeric G-proteins (Gαßγ) and they are the most successful drug targets. The mechanisms of G-protein activation by GPCRs are not well understood. Previous studies have revealed a signal relay route from a GPCR via the C-terminal α5-helix of Gα to the guanine nucleotide-binding pocket. Recent structural and biophysical studies uncover a role for the opening or rotating of the α-helical domain of Gα during the activation of Gα by a GPCR. Here we show that ß-adrenergic receptors activate eight Gαs mutant proteins (from a screen of 66 Gαs mutants) that are unable to bind Gßγ subunits in cells. Five of these eight mutants are in the αF/Linker 2/ß2 hinge region (extended Linker 2) that connects the Ras-like GTPase domain and the α-helical domain of Gαs. This extended Linker 2 is the target site of a natural product inhibitor of Gq. Our data show that the extended Linker 2 is critical for Gα activation by GPCRs. We propose that a GPCR via its intracellular loop 2 directly interacts with the ß2/ß3 loop of Gα to communicate to Linker 2, resulting in the opening and closing of the α-helical domain and the release of GDP during G-protein activation.


Subject(s)
GTP-Binding Protein alpha Subunits, Gs/chemistry , GTP-Binding Protein beta Subunits/chemistry , GTP-Binding Protein gamma Subunits/chemistry , Receptors, Adrenergic, beta/chemistry , Amino Acid Sequence , Binding Sites , Escherichia coli/genetics , Escherichia coli/metabolism , GTP-Binding Protein alpha Subunits, Gs/genetics , GTP-Binding Protein alpha Subunits, Gs/metabolism , GTP-Binding Protein beta Subunits/genetics , GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein gamma Subunits/genetics , GTP-Binding Protein gamma Subunits/metabolism , Gene Expression , Gene Expression Regulation , HEK293 Cells , Humans , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Molecular Sequence Data , Mutation , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Receptors, Adrenergic, beta/genetics , Receptors, Adrenergic, beta/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction
13.
Eur Biophys J ; 44(3): 149-63, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25726162

ABSTRACT

The ß2-adrenergic receptor (ß2-AR), a G protein-coupled receptor (GPCR), is a physiologically important transmembrane protein that is a target for drugs used for treatment of asthma and cardiovascular diseases. Study of the first steps of ligand recognition and the molecular basis of ligand binding to the orthosteric site is essential for understanding the pharmacological properties of the receptor. In this work we investigated the characteristic features of the agonist association-dissociation process to and from the different conformational forms of ß2-AR by use of advanced molecular modeling techniques. The investigation was focused on estimating the free energy profiles (FEPs) corresponding to the process of a full agonist ((R,R)-fenoterol) and an inverse agonist (carazolol) binding and unbinding to and from ß2-AR. The two different conformational forms of ß2-AR, i.e. active ß2-AR-PDB: 3P0G and inactive ß2-AR-PDB: 2RH1 were included in this stage of the study. We revealed several significant qualitative differences between FEPs characteristic of both conformational forms. Both FEPs suggest the existence of three transient binding sites in the extracellular domain of ß2-AR. Comparison of the residues surrounding these transient binding sites in both ß2-AR states revealed the importance of the aromatic residues F194, H93(2.64), H296(6.58), and H178 (extracellular part of ß2-AR) in the early stages of the binding process. In addition, slightly different exit and entry paths are preferred by the ligand molecule in the extracellular part of ß2-AR, depending on the conformation of the receptor.


Subject(s)
Adrenergic beta-Agonists/pharmacology , Fenoterol/pharmacology , Molecular Docking Simulation , Propanolamines/pharmacology , Receptors, Adrenergic, beta/chemistry , Adrenergic beta-Agonists/chemistry , Amino Acid Sequence , Binding Sites , Fenoterol/chemistry , Humans , Molecular Sequence Data , Propanolamines/chemistry , Protein Binding , Receptors, Adrenergic, beta/metabolism
14.
Bioorg Med Chem ; 23(14): 3880-906, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-25638496

ABSTRACT

Recent breakthroughs in GPCR structural biology have significantly increased our understanding of drug action at these therapeutically relevant receptors, and this will undoubtedly lead to the design of better therapeutics. In recent years, crystal structures of GPCRs from classes A, B, C and F have been solved, unveiling a precise snapshot of ligand-receptor interactions. Furthermore, some receptors have been crystallized in different functional states in complex with antagonists, partial agonists, full agonists, biased agonists and allosteric modulators, providing further insight into the mechanisms of ligand-induced GPCR activation. It is now obvious that there is enormous diversity in the size, shape and position of the ligand binding pockets in GPCRs. In this review, we summarise the current state of solved GPCR structures, with a particular focus on ligand-receptor interactions in the binding pocket, and how this can contribute to the design of GPCR ligands with better affinity, subtype selectivity or efficacy.


Subject(s)
Drug Design , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism , Chemistry, Pharmaceutical/methods , Crystallography, X-Ray , Humans , Ligands , Protein Conformation , Receptors, Adrenergic, beta/chemistry , Receptors, Adrenergic, beta/metabolism
15.
J Chem Phys ; 141(9): 090901, 2014 Sep 07.
Article in English | MEDLINE | ID: mdl-25194354

ABSTRACT

Molecular dynamics simulations have the potential to provide atomic-level detail and insight to important questions in chemical physics that cannot be observed in typical experiments. However, simply generating a long trajectory is insufficient, as researchers must be able to transform the data in a simulation trajectory into specific scientific insights. Although this analysis step has often been taken for granted, it deserves further attention as large-scale simulations become increasingly routine. In this perspective, we discuss the application of Markov models to the analysis of large-scale biomolecular simulations. We draw attention to recent improvements in the construction of these models as well as several important open issues. In addition, we highlight recent theoretical advances that pave the way for a new generation of models of molecular kinetics.


Subject(s)
Markov Chains , Molecular Dynamics Simulation , Proteins/chemistry , Protein Conformation , Protein Folding , Receptors, Adrenergic, beta/chemistry , beta-Lactamases/chemistry
16.
Molecules ; 19(3): 3417-35, 2014 Mar 20.
Article in English | MEDLINE | ID: mdl-24658567

ABSTRACT

A new series of oxime ethers 4a-z was designed and synthesized to test the blocking activity against ß1 and ß2-adrenergic receptors. Docking of these ether derivatives into the active site of the identified 3D structures of ß1 and ß2-adrenergic receptors showed MolDock scores comparable to those of reference compounds. Biological results revealed that the inhibition effects on the heart rate and contractility are less than those of propranolol. Nevertheless, the two compounds 4p and 4q that displayed the highest negative MolDock score with ß2-adrenergic receptors showed ß2-antagonistic activity by decreasing salbutamol relaxation of precontracted tracheal strips, which indicates the importance of a chlorothiophene moiety in the hydrophobic region for best complementarity with ß2 receptors. On other hand, the presence of a homoveratryl moiety increases the MolDock score of the tested compounds with the ß1 receptor.


Subject(s)
Adrenergic beta-Antagonists/chemistry , Ethers/chemistry , Molecular Docking Simulation , Oximes/chemistry , Receptors, Adrenergic, beta/chemistry , Adrenergic beta-Antagonists/chemical synthesis , Adrenergic beta-Antagonists/pharmacology , Animals , Ethers/chemical synthesis , Ethers/pharmacology , Guinea Pigs , Heart Atria/drug effects , Heart Atria/metabolism , Male , Molecular Conformation , Receptors, Adrenergic, beta/metabolism , Receptors, Adrenergic, beta-1/chemistry , Receptors, Adrenergic, beta-1/metabolism , Receptors, Adrenergic, beta-2/chemistry , Receptors, Adrenergic, beta-2/metabolism , Trachea/drug effects , Trachea/metabolism
17.
Exp Mol Med ; 56(9): 1952-1966, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39218975

ABSTRACT

Agonists targeting α2-adrenergic receptors (ARs) are used to treat diverse conditions, including hypertension, attention-deficit/hyperactivity disorder, pain, panic disorders, opioid and alcohol withdrawal symptoms, and cigarette cravings. These receptors transduce signals through heterotrimeric Gi proteins. Here, we elucidated cryo-EM structures that depict α2A-AR in complex with Gi proteins, along with the endogenous agonist epinephrine or the synthetic agonist dexmedetomidine. Molecular dynamics simulations and functional studies reinforce the results of the structural revelations. Our investigation revealed that epinephrine exhibits different conformations when engaging with α-ARs and ß-ARs. Furthermore, α2A-AR and ß1-AR (primarily coupled to Gs, with secondary associations to Gi) were compared and found to exhibit different interactions with Gi proteins. Notably, the stability of the epinephrine-α2A-AR-Gi complex is greater than that of the dexmedetomidine-α2A-AR-Gi complex. These findings substantiate and improve our knowledge on the intricate signaling mechanisms orchestrated by ARs and concurrently shed light on the regulation of α-ARs and ß-ARs by epinephrine.


Subject(s)
Epinephrine , Molecular Dynamics Simulation , Protein Binding , Epinephrine/metabolism , Epinephrine/chemistry , Humans , Dexmedetomidine/chemistry , Dexmedetomidine/pharmacology , Dexmedetomidine/metabolism , Receptors, Adrenergic, alpha-2/metabolism , Receptors, Adrenergic, alpha-2/chemistry , Protein Conformation , Cryoelectron Microscopy , Receptors, Adrenergic, beta/metabolism , Receptors, Adrenergic, beta/chemistry
18.
Cell Physiol Biochem ; 31(1): 25-36, 2013.
Article in English | MEDLINE | ID: mdl-23343624

ABSTRACT

BACKGROUND/AIMS: In diabetic ventricular myocytes, transient outward potassium current (Ito) amplitude is severely reduced because of the impaired catecholamine release that characterizes diabetic autonomic neuropathy. Sympathetic nervous system exhibits a trophic effect on Ito since incubation of myocytes with noradrenaline restores current amplitude via beta-adrenoceptor (ßAR) stimulation. Here, we investigate the intracellular signalling pathway though which incubation of diabetic cardiomyocytes with the ßAR agonist isoproterenol recovers Ito amplitude to normal values. METHODS: Experiments were performed in ventricular myocytes isolated from streptozotocin-diabetic rats. Ito current was recorded by using the patch-clamp technique. Kv4 channel expression was determined by immunofluorescence. Protein-protein interaction was determined by coimmunoprecipitation. RESULTS: Stimulation of ßAR activates first a Gαs protein, adenylyl cyclase and Protein Kinase A. PKA-phosphorylated receptor then switches to the Gαi protein. This leads to the activation of the ßAR-Kinase-1 and further receptor phosphorylation and arrestin dependent internalization. The internalized receptor-arrestin complex recruits and activates cSrc and the MAPK cascade, where Ras, c-Raf1 and finally ERK1/2 mediate the increase in Kv4.2 and Kv4.3 protein abundance in the plasma membrane. CONCLUSION: ß2AR stimulation activates a Gαs and Gαi protein dependent pathway where the ERK1/2 modulates the Ito current amplitude and the density of the Kv4.2 and Kv4.2 channels in the plasma membrane upon sympathetic stimulation in diabetic heart.


Subject(s)
Membrane Potentials/physiology , Myocytes, Cardiac/metabolism , Receptors, Adrenergic, beta/metabolism , Adenylyl Cyclases/metabolism , Adrenergic beta-Agonists/pharmacology , Animals , Arrestin/metabolism , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Protein alpha Subunits, Gs/metabolism , Isoproterenol/pharmacology , Membrane Potentials/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Patch-Clamp Techniques , Phosphorylation , Potassium/metabolism , Protein Interaction Maps/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, beta/chemistry , Shal Potassium Channels/metabolism , Signal Transduction
19.
Biochem Soc Trans ; 41(1): 159-65, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23356277

ABSTRACT

Structures of the inactive state of the thermostabilized ß1-adrenoceptor have been determined bound to eight different ligands, including full agonists, partial agonists, inverse agonists and biased agonists. Comparison of the structures shows distinct differences within the binding pocket that correlate with the pharmacological properties of the ligands. These data suggest that full agonists stabilize a structure with a contracted binding pocket and a rotamer change of serine (5.46) compared with when antagonists are bound. Inverse agonists may prevent both of these occurrences, whereas partial agonists stabilize a contraction of the binding pocket but not the rotamer change of serine (5.46). It is likely that subtle changes in the interactions between transmembrane helix 5 (H5) and H3/H4 on agonist binding promote the formation of the activated state.


Subject(s)
Receptors, Adrenergic, beta/metabolism , Arrestins/metabolism , Ligands , Protein Binding , Protein Conformation , Receptors, Adrenergic, beta/chemistry
20.
Eur J Med Chem ; 246: 114961, 2023 Jan 15.
Article in English | MEDLINE | ID: mdl-36495629

ABSTRACT

Biomedical applications of molecules that are able to modulate ß-adrenergic signaling have become increasingly attractive over the last decade, revealing that ß-adrenergic receptors (ß-ARs) are key targets for a plethora of therapeutic interventions, including cancer. Despite successes in ß-AR drug discovery, identification of ß-AR ligands that are useful as selective chemical tools in pharmacological studies of the three ß-AR subtypes, or lead compounds for drug development is still a highly challenging task. This is mainly due to the intrinsic plasticity of ß-ARs as G protein-coupled receptors in conjunction with the requirement for functional receptor subtype selectivity, tissue specificity and minimal off-target effects. With the aim to provide insight into structure-activity relationships for the three ß-AR subtypes, we have synthesized and obtained the pharmacological profile of a series of structurally diverse compounds (named MC) that were designed based on the aryloxy-propanolamine scaffold of SR59230A. Comparative analysis of their predicted binding mode within the active and inactive states of the receptors in combination with their pharmacological profile revealed key structural elements that control their activity as agonists or antagonists, in addition to clues about substituents that mediate selectivity for one receptor subtype over the others. We anticipate that these results will facilitate selective ß-AR drug development efforts.


Subject(s)
Receptors, Adrenergic, beta , Receptors, G-Protein-Coupled , Humans , Receptors, Adrenergic, beta/chemistry , Receptors, Adrenergic, beta/metabolism , Ligands , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL