Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 164
Filter
1.
Nat Commun ; 13(1): 382, 2022 01 19.
Article in English | MEDLINE | ID: mdl-35046410

ABSTRACT

α-adrenergic receptors (αARs) are G protein-coupled receptors that regulate vital functions of the cardiovascular and nervous systems. The therapeutic potential of αARs, however, is largely unexploited and hampered by the scarcity of subtype-selective ligands. Moreover, several aminergic drugs either show off-target binding to αARs or fail to interact with the desired subtype. Here, we report the crystal structure of human α1BAR bound to the inverse agonist (+)-cyclazosin, enabled by the fusion to a DARPin crystallization chaperone. The α1BAR structure allows the identification of two unique secondary binding pockets. By structural comparison of α1BAR with α2ARs, and by constructing α1BAR-α2CAR chimeras, we identify residues 3.29 and 6.55 as key determinants of ligand selectivity. Our findings provide a basis for discovery of α1BAR-selective ligands and may guide the optimization of aminergic drugs to prevent off-target binding to αARs, or to elicit a selective interaction with the desired subtype.


Subject(s)
Crystallography, X-Ray , Receptors, Adrenergic, alpha-1/chemistry , Binding Sites , HEK293 Cells , Humans , Ligands , Lipids/chemistry , Models, Molecular , Quinazolines/chemistry , Quinazolines/metabolism , Quinoxalines/chemistry , Quinoxalines/metabolism , Receptors, Adrenergic, alpha-2/chemistry
2.
Int J Mol Sci ; 22(23)2021 Nov 25.
Article in English | MEDLINE | ID: mdl-34884552

ABSTRACT

Dexmedetomidine (DEX), a selective α2 adrenergic receptor (AR) agonist, is commonly used as a sedative drug during critical illness. In the present study, we explored a novel accelerative effect of DEX on cardiac fibroblast (CF) differentiation mediated by LPS and clarified its potential mechanism. LPS apparently increased the expression of α-SMA and collagen I/III and the phosphorylation of p38 and Smad-3 in the CFs of mice. These effects were significantly enhanced by DEX through increasing α2A-AR expression in CFs after LPS stimulation. The CFs from α2A-AR knockout mice were markedly less sensitive to DEX treatment than those of wild-type mice. Inhibition of protein kinase C (PKC) abolished the enhanced effects of DEX on LPS-induced differentiation of CFs. We also found that the α-SMA level in the second-passage CFs was much higher than that in the nonpassage and first-passage CFs. However, after LPS stimulation, the TNF-α released from the nonpassage CFs was much higher than that in the first- and second-passage CFs. DEX had no effect on LPS-induced release of TNF-α and IL-6 from CFs. Further investigation indicated that DEX promoted cardiac fibrosis and collagen I/III synthesis in mice exposed to LPS for four weeks. Our results demonstrated that DEX effectively accelerated LPS-induced differentiation of CFs to myofibroblasts through the PKC-p38-Smad2/3 signaling pathway by activating α2A-AR.


Subject(s)
Cell Differentiation , Collagen Type III/metabolism , Collagen Type I/metabolism , Dexmedetomidine/pharmacology , Gene Expression Regulation/drug effects , Lipopolysaccharides/pharmacology , Myofibroblasts/cytology , Receptors, Adrenergic, alpha-2/chemistry , Adrenergic alpha-2 Receptor Agonists/pharmacology , Animals , Male , Mice , Mice, Inbred C57BL , Myofibroblasts/drug effects , Myofibroblasts/metabolism , Protein Kinase C/genetics , Protein Kinase C/metabolism , Signal Transduction , Smad2 Protein/genetics , Smad2 Protein/metabolism , Smad3 Protein/genetics , Smad3 Protein/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
3.
ACS Chem Biol ; 16(2): 404-413, 2021 02 19.
Article in English | MEDLINE | ID: mdl-33543920

ABSTRACT

Identification of physiologically relevant targets for lead compounds emerging from drug discovery screens is often the rate-limiting step toward understanding their mechanism of action and potential for undesired off-target effects. To this end, we developed a streamlined chemical proteomic approach utilizing a single, photoreactive cleavable chloroalkane capture tag, which upon attachment to bioactive compounds facilitates selective isolation of their respective cellular targets for subsequent identification by mass spectrometry. When properly positioned, the tag does not significantly affect compound potency and membrane permeability, allowing for binding interactions with the tethered compound (probe) to be established within intact cells under physiological conditions. Subsequent UV-induced covalent photo-cross-linking "freezes" the interactions between the probe and its cellular targets and prevents their dissociation upon cell lysis. Targets cross-linked to the capture tag are then efficiently enriched through covalent capture onto HaloTag coated beads and subsequent selective chemical release from the solid support. The tag's built-in capability for selective enrichment eliminates the need for ligation of a capture tag, thereby simplifying the workflow and reducing variability introduced through additional operational steps. At the same time, the capacity for adequate cross-linking without structural optimization permits modular assembly of photoreactive chloroalkane probes, which reduces the burden of customized chemistry. Using three model compounds, we demonstrate the capability of this approach to identify known and novel cellular targets, including those with low affinity and/or low abundance as well as membrane targets with several transmembrane domains.


Subject(s)
Affinity Labels/chemistry , Azides/chemistry , Cross-Linking Reagents/chemistry , Diazomethane/analogs & derivatives , Hydrocarbons, Chlorinated/chemistry , Proteomics/methods , Affinity Labels/radiation effects , Azides/radiation effects , Chromatography, Liquid , Cross-Linking Reagents/radiation effects , Dasatinib/analogs & derivatives , Dasatinib/pharmacology , Dasatinib/radiation effects , Diazomethane/radiation effects , Histone Deacetylases/analysis , Histone Deacetylases/chemistry , Humans , Hydrocarbons, Chlorinated/radiation effects , Hydrolases/chemistry , K562 Cells , Mass Spectrometry , Propranolol/analogs & derivatives , Propranolol/pharmacology , Propranolol/radiation effects , Protein Kinases/analysis , Protein Kinases/chemistry , Receptors, Adrenergic, alpha-2/analysis , Receptors, Adrenergic, alpha-2/chemistry , Ultraviolet Rays , Vorinostat/analogs & derivatives , Vorinostat/pharmacology , Vorinostat/radiation effects
4.
Int J Mol Sci ; 21(21)2020 Oct 22.
Article in English | MEDLINE | ID: mdl-33105703

ABSTRACT

Quantitative Structure Activity Relationship (QSAR) models can inform on the correlation between activities and structure-based molecular descriptors. This information is important for the understanding of the factors that govern molecular properties and for designing new compounds with favorable properties. Due to the large number of calculate-able descriptors and consequently, the much larger number of descriptors combinations, the derivation of QSAR models could be treated as an optimization problem. For continuous responses, metrics which are typically being optimized in this process are related to model performances on the training set, for example, R2 and QCV2. Similar metrics, calculated on an external set of data (e.g., QF1/F2/F32), are used to evaluate the performances of the final models. A common theme of these metrics is that they are context -" ignorant". In this work we propose that QSAR models should be evaluated based on their intended usage. More specifically, we argue that QSAR models developed for Virtual Screening (VS) should be derived and evaluated using a virtual screening-aware metric, e.g., an enrichment-based metric. To demonstrate this point, we have developed 21 Multiple Linear Regression (MLR) models for seven targets (three models per target), evaluated them first on validation sets and subsequently tested their performances on two additional test sets constructed to mimic small-scale virtual screening campaigns. As expected, we found no correlation between model performances evaluated by "classical" metrics, e.g., R2 and QF1/F2/F32 and the number of active compounds picked by the models from within a pool of random compounds. In particular, in some cases models with favorable R2 and/or QF1/F2/F32 values were unable to pick a single active compound from within the pool whereas in other cases, models with poor R2 and/or QF1/F2/F32 values performed well in the context of virtual screening. We also found no significant correlation between the number of active compounds correctly identified by the models in the training, validation and test sets. Next, we have developed a new algorithm for the derivation of MLR models by optimizing an enrichment-based metric and tested its performances on the same datasets. We found that the best models derived in this manner showed, in most cases, much more consistent results across the training, validation and test sets and outperformed the corresponding MLR models in most virtual screening tests. Finally, we demonstrated that when tested as binary classifiers, models derived for the same targets by the new algorithm outperformed Random Forest (RF) and Support Vector Machine (SVM)-based models across training/validation/test sets, in most cases. We attribute the better performances of the Enrichment Optimizer Algorithm (EOA) models in VS to better handling of inactive random compounds. Optimizing an enrichment-based metric is therefore a promising strategy for the derivation of QSAR models for classification and virtual screening.


Subject(s)
Quantitative Structure-Activity Relationship , Algorithms , Databases, Pharmaceutical , Drug Evaluation, Preclinical/methods , ERG1 Potassium Channel/chemistry , Humans , Linear Models , Receptor, Muscarinic M3/chemistry , Receptor, Serotonin, 5-HT2C/chemistry , Receptors, Adrenergic, alpha-2/chemistry , Receptors, Dopamine D1/chemistry , Support Vector Machine
5.
Nat Chem Biol ; 16(5): 507-512, 2020 05.
Article in English | MEDLINE | ID: mdl-32152538

ABSTRACT

The α2 adrenergic receptors (α2ARs) are G protein-coupled receptors (GPCRs) that respond to adrenaline and noradrenaline and couple to the Gi/o family of G proteins. α2ARs play important roles in regulating the sympathetic nervous system. Dexmedetomidine is a highly selective α2AR agonist used in post-operative patients as an anxiety-reducing, sedative medicine that decreases the requirement for opioids. As is typical for selective αAR agonists, dexmedetomidine consists of an imidazole ring and a substituted benzene moiety lacking polar groups, which is in contrast to ßAR-selective agonists, which share an ethanolamine group and an aromatic system with polar, hydrogen-bonding substituents. To better understand the structural basis for the selectivity and efficacy of adrenergic agonists, we determined the structure of the α2BAR in complex with dexmedetomidine and Go at a resolution of 2.9 Å by single-particle cryo-EM. The structure reveals the mechanism of α2AR-selective activation and provides insights into Gi/o coupling specificity.


Subject(s)
Adrenergic alpha-2 Receptor Agonists/chemistry , Dexmedetomidine/chemistry , Receptors, Adrenergic, alpha-2/chemistry , Receptors, Adrenergic, alpha-2/metabolism , Adrenergic alpha-2 Receptor Agonists/pharmacology , Animals , Binding Sites , Cryoelectron Microscopy , Dexmedetomidine/metabolism , Dexmedetomidine/pharmacology , GTP-Binding Proteins/chemistry , GTP-Binding Proteins/metabolism , Insecta/cytology , Molecular Docking Simulation , Molecular Dynamics Simulation , Multiprotein Complexes/chemistry , Receptors, Adrenergic, alpha-2/genetics , Sympatholytics/chemistry , Sympatholytics/pharmacology
6.
Anal Chim Acta ; 1102: 53-62, 2020 Mar 15.
Article in English | MEDLINE | ID: mdl-32043996

ABSTRACT

Chemical cross-linking would conceivably cause structural disruption of a protein, but few cross-linkers have been fully evaluated in this aspect. Furthermore, integral membrane proteins may differ from soluble proteins in the selection of suitable cross-linkers, which has never been investigated. In this study, we systematically evaluated the impact of five conventional cross-linkers targeting Lys, Asp and Glu, and two Arg-reactive cross-linkers on the structural and functional integrity of two G protein-coupled receptors (GPCRs). Perturbation of the receptor structure and ligand-binding activity was observed, depending on the receptor and cross-linking conditions. In particular, our study demonstrated that the concentrations of PDH and KArGO need to be fine-tuned in order to minimize the structural and functional disturbance of specific GPCRs. A set of amenable cross-linkers was selected to acquire the most comprehensive cross-link maps for two GPCRs. Our in-depth cross-linking mass spectrometry (CXMS) analysis has revealed dynamic features of structural regions in GPCRs that are not observable in the crystal structures. Thus, CXMS analysis of GPCRs using the expanded toolkit would facilitate structural modeling of uncharacterized receptors and gain new insights into receptor-ligand interactions.


Subject(s)
Cross-Linking Reagents/chemistry , Glucagon-Like Peptide-1 Receptor/chemistry , Receptors, Adrenergic, alpha-2/chemistry , Chromatography, Gel , Chromatography, Liquid , Glucagon-Like Peptide-1 Receptor/metabolism , Ligands , Molecular Dynamics Simulation , Protein Conformation , Protein Stability , Receptors, Adrenergic, alpha-2/metabolism , Tandem Mass Spectrometry/methods
7.
Oxid Med Cell Longev ; 2019: 9426368, 2019.
Article in English | MEDLINE | ID: mdl-31827710

ABSTRACT

Patients with orthotopic liver transplantation (OLT) frequently develop acute gut injury (AGI), and dexmedetomidine (Dex) has been reported to exert a protective effect against AGI. We investigated whether Dex protects against AGI through antioxidative stress effects by the Nrf2/HO-1 antioxidative signaling pathway. Rats were randomly allocated into a sham group and six orthotopic autologous liver transplantation (OALT) groups receiving different doses of Dex together with/without α 2-adrenergic receptor (AR) blockers. Intestinal tissues were collected to visualize the barrier damage and to measure the indexes of oxidative stress. For in vitro studies, rat intestinal recess epithelial cells (IEC-6) underwent hypoxia/reoxygenation (H/R), and the protective role of Dex was evaluated after α 2A-AR siRNA silencing. OALT resulted in increased oxidative stress, significant intestinal injury, and barrier dysfunction. Dex attenuated OALT-induced oxidative stress and intestinal injury, which was abolished by the pretreatment with the nonspecific α 2A-AR siRNA blocker atipamezole and the specific α 2A-AR siRNA blocker BRL-44408, but not by the specific 2B/C-AR siRNA blocker ARC239. Silencing of α 2A-AR siRNA also attenuated the protective role of Dex on alleviating oxidative stress in IEC-6 cells subjected to H/R. Dex exerted its protective effects by activating Nrf2/HO-1 antioxidative signaling. Collectively, Dex attenuates OALT-induced AGI via α 2A-AR-dependent suppression of oxidative stress, which might be a novel potential therapeutic target for OALT-induced AGI.


Subject(s)
Adrenergic alpha-2 Receptor Agonists/pharmacology , Dexmedetomidine/pharmacology , Gastrointestinal Diseases/prevention & control , Liver Transplantation/adverse effects , Oxidative Stress/drug effects , Protective Agents/pharmacology , Receptors, Adrenergic, alpha-2/metabolism , Animals , Antioxidants/pharmacology , Gastrointestinal Diseases/etiology , Gastrointestinal Diseases/metabolism , Gastrointestinal Diseases/pathology , Male , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, alpha-2/chemistry , Receptors, Adrenergic, alpha-2/genetics
8.
J Chem Inf Model ; 59(5): 2479-2486, 2019 05 28.
Article in English | MEDLINE | ID: mdl-30879296

ABSTRACT

A theoretical study has been carried out at the M062X/6-311++G(d,p) computational level to search for a rationale on ligands' affinity toward α2-adrenoceptors by estimating the nature and strength of intramolecular hydrogen bonds potentially formed (by means of the QTAIM and NBO approaches) as well as the degree of deviation from planarity that could be observed in some of the compounds. Four different families have been studied: thiophen-2-yl, 3-carboxylatethiophen-2-yl esters, 3-cyanothiophen-2-yl, and 2-thiazolyl guanidinium derivatives. In the case of the thiophen-2-yl guanidines not substituted in the 3 position, nonplanarity was always observed, whereas in the thiazole series, intramolecular hydrogen bonds were identified between the guanidinium and the thiazole ring forcing the systems to planarity. Regarding the carboxylic esters, two different rotamers were found: quasi-planar and quasi-perpendicular systems with very similar energy. Both of these isomers can form different nets of intramolecular hydrogen bonds and other types of noncovalent interactions. Different physicochemical properties such as basicity, solubility, or lipophilicity were calculated for these systems, but no correlation to the degree of planarity was found. However, when comparing the α2-ARs affinity with the planarity of the molecules, a trend appears in the thiophen-2-yl guanidinium series indicating that lack of planarity seems to be optimal for α2-ARs engagement.


Subject(s)
Guanidine/chemistry , Guanidine/metabolism , Models, Molecular , Receptors, Adrenergic, alpha-2/metabolism , Electrons , Hydrophobic and Hydrophilic Interactions , Ligands , Molecular Conformation , Receptors, Adrenergic, alpha-2/chemistry , Solubility , Thermodynamics
9.
Am J Pathol ; 188(12): 2774-2785, 2018 12.
Article in English | MEDLINE | ID: mdl-30273604

ABSTRACT

α2-Adrenergic receptors (α2ARs) are G-protein-coupled receptors involved in catecholamine signaling by extracellular regulated protein kinase 1 and 2 (ERK1/2) pathways. We examined placental expression and function of α2AR subtypes in women with severe preeclampsia (sPE) with and without intrauterine growth restriction (IUGR). Placental biopsies were analyzed from 52 women with i) sPE (n = 8); ii) sPE + IUGR (n = 9); iii) idiopathic IUGR (n = 8); iv) idiopathic preterm birth (n = 16); and v) healthy term controls (n = 11). Expression of α2AR subtypes (α2A, α2B, α2C) and phospho-ERK1/2 (receptor activation marker) was investigated by immunohistochemistry and/or quantitative real-time RT-PCR. The effects of α2CAR knockdown on syncytialization (syncytin-1 and -2) and ß-human chorionic gonadotropin secretion were examined in BeWo cells stimulated with forskolin. The effects of α2AR agonist UK 14,304 and specific α2CAR antagonist were tested, using a trophoblast migration assay. All three α2ARs were expressed and functionally active in human placenta with site-specific localization. Highest α2BAR and α2CAR mRNA expression was identified in sPE + IUGR. α2CAR knockdown increased expression of syncytin-1 and -2 but decreased secretion of ß-human chorionic gonadotropin. UK 14,304 impaired trophoblast migration. The observed α2AR expression pattern suggests different function for each subtype. α2CAR modulates trophoblast syncytialization and migration and may carry pathogenic role in sPE + IUGR.


Subject(s)
Fetal Growth Retardation/pathology , Placenta/pathology , Pre-Eclampsia/pathology , Premature Birth/pathology , Receptors, Adrenergic, alpha-2/metabolism , Trophoblasts/pathology , Adrenergic alpha-2 Receptor Agonists/pharmacology , Brimonidine Tartrate/pharmacology , Case-Control Studies , Cells, Cultured , Chorionic Gonadotropin, beta Subunit, Human/metabolism , Female , Fetal Growth Retardation/metabolism , Humans , Infant, Newborn , Placenta/metabolism , Pre-Eclampsia/metabolism , Pregnancy , Premature Birth/metabolism , Receptors, Adrenergic, alpha-2/chemistry , Trophoblasts/metabolism
10.
Nature ; 559(7714): 423-427, 2018 07.
Article in English | MEDLINE | ID: mdl-29995853

ABSTRACT

G-protein-coupled receptors (GPCRs) are involved in many physiological processes and are therefore key drug targets1. Although detailed structural information is available for GPCRs, the effects of lipids on the receptors, and on downstream coupling of GPCRs to G proteins are largely unknown. Here we use native mass spectrometry to identify endogenous lipids bound to three class A GPCRs. We observed preferential binding of phosphatidylinositol-4,5-bisphosphate (PtdIns(4,5)P2) over related lipids and confirm that the intracellular surface of the receptors contain hotspots for PtdIns(4,5)P2 binding. Endogenous lipids were also observed bound directly to the trimeric Gαsßγ protein complex of the adenosine A2A receptor (A2AR) in the gas phase. Using engineered Gα subunits (mini-Gαs, mini-Gαi and mini-Gα12)2, we demonstrate that the complex of mini-Gαs with the ß1 adrenergic receptor (ß1AR) is stabilized by the binding of two PtdIns(4,5)P2 molecules. By contrast, PtdIns(4,5)P2 does not stabilize coupling between ß1AR and other Gα subunits (mini-Gαi or mini-Gα12) or a high-affinity nanobody. Other endogenous lipids that bind to these receptors have no effect on coupling, highlighting the specificity of PtdIns(4,5)P2. Calculations of potential of mean force and increased GTP turnover by the activated neurotensin receptor when coupled to trimeric Gαißγ complex in the presence of PtdIns(4,5)P2 provide further evidence for a specific effect of PtdIns(4,5)P2 on coupling. We identify key residues on cognate Gα subunits through which PtdIns(4,5)P2 forms bridging interactions with basic residues on class A GPCRs. These modulating effects of lipids on receptors suggest consequences for understanding function, G-protein selectivity and drug targeting of class A GPCRs.


Subject(s)
Heterotrimeric GTP-Binding Proteins/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism , Animals , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Protein alpha Subunits, Gs/metabolism , Humans , Molecular Dynamics Simulation , Protein Stability , Rats , Receptors, Adrenergic, alpha-2/chemistry , Receptors, Adrenergic, alpha-2/genetics , Receptors, Adrenergic, alpha-2/metabolism , Receptors, Adrenergic, beta-1/chemistry , Receptors, Adrenergic, beta-1/genetics , Receptors, Adrenergic, beta-1/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, Neurotensin/chemistry , Receptors, Neurotensin/genetics , Receptors, Neurotensin/metabolism , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/metabolism , Substrate Specificity , Turkeys
11.
J Physiol ; 596(13): 2507-2519, 2018 07.
Article in English | MEDLINE | ID: mdl-29708589

ABSTRACT

KEY POINTS: Contraction-mediated blunting of postjunctional α-adrenergic vasoconstriction (functional sympatholysis) is attenuated in skeletal muscle of ageing males, brought on by altered postjunctional α1 - and α2 -adrenergic receptor sensitivity. The extent to which postjunctional α-adrenergic vasoconstriction occurs in the forearms at rest and during exercise in postmenopausal women remains unknown. The novel findings indicate that contraction-mediated blunting of α1 - (via intra-arterial infusion of phenylephrine) but not α2 -adrenergic (via intra-arterial infusion of dexmedetomidine) vasoconstriction was attenuated in postmenopausal women compared to young women. Additional important findings revealed that postjunctional α-adrenergic vasoconstrictor responsiveness at rest does not appear to be affected by age in women. Collectively, these results contribute to our understanding of local neurovascular control at rest and during exercise with age in women. ABSTRACT: Contraction-mediated blunting of postjunctional α-adrenergic vasoconstriction (functional sympatholysis) is attenuated in older males; however, direct confirmation of this effect remains unknown in postmenopausal women (PMW). The present study examined whether PMW exhibit augmented postjunctional α-adrenergic receptor vasoconstriction at rest and during forearm exercise compared to young women (YW). Eight YW (24 ± 1 years) and eight PMW (65 ± 1 years) completed a series of randomized experimental trials: (1) at rest, (2) under high flow (adenosine infusion) conditions and (3) during 6 min of forearm exercise at relative (20% of maximum) and absolute (7 kg) intensities. Phenylephrine (α1 -agonist) or dexmedetomidine (α2 -agonist) was administered during the last 3 min of each trial to elicit α-adrenergic vasoconstriction. Forearm vascular conductance (FVC) was calculated from blood flow and blood pressure. Vasoconstrictor responsiveness was identified as the change in FVC (%) during α-adrenergic agonist infusions from baseline (resting trial) or from steady-state conditions (high flow and exercise trials). During resting and high flow trials, the %FVC during α1 - and α2 -agonist stimulation was similar between YW and PMW. During exercise, α1 -mediated vasoconstriction was blunted in YW vs. PMW at relative (-6 ± 2% vs. -15 ± 3%) and absolute (-4 ± 2% vs. -14 ± 5%) workloads, such that blood flow and FVC were lower in PMW (P < 0.05 for all). Conversely, α2 -mediated vasoconstriction was similar between YW and PMW at relative (-22 ± 3% vs. -22 ± 4%; P > 0.05) and absolute (-19 ± 3% vs. -18 ± 4%; P > 0.05) workloads. Collectively, these findings demonstrate that despite similar α-adrenergic vasoconstrictor responsiveness at rest, PMW have a decreased ability to attenuate α1 -adrenergic vasoconstriction in contracting skeletal muscle.


Subject(s)
Forearm/physiopathology , Muscle Contraction , Muscle, Skeletal/physiopathology , Postmenopause , Receptors, Adrenergic, alpha-1/chemistry , Receptors, Adrenergic, alpha-2/chemistry , Vasoconstriction/physiology , Adenosine Triphosphate/metabolism , Adrenergic alpha-1 Receptor Agonists/pharmacology , Adrenergic alpha-2 Receptor Agonists/pharmacology , Adult , Aged , Case-Control Studies , Dexmedetomidine/pharmacology , Female , Forearm/blood supply , Humans , Muscle, Skeletal/blood supply , Muscle, Skeletal/drug effects , Phenylephrine/pharmacology , Receptors, Adrenergic, alpha-1/metabolism , Receptors, Adrenergic, alpha-2/metabolism , Vasoconstriction/drug effects , Young Adult
12.
Sci Rep ; 8(1): 4631, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29545586

ABSTRACT

Influenza A viruses cause seasonal epidemics and occasional pandemics. The emergence of viruses resistant to neuraminidase (NA) inhibitors and M2 ion channel inhibitors underlines the need for alternate anti-influenza drugs with novel mechanisms of action. Here, we report the discovery of a host factor as a potential target of anti-influenza drugs. By using cell-based virus replication screening of a chemical library and several additional assays, we identified clonidine as a new anti-influenza agent in vitro. We found that clonidine, which is an agonist of the alpha2-adrenergic receptor (α2-AR), has an inhibitory effect on the replication of various influenza virus strains. α2-AR is a Gi-type G protein-coupled receptor that reduces intracellular cyclic AMP (cAMP) levels. In-depth analysis showed that stimulation of α2-ARs leads to impairment of influenza virus replication and that α2-AR agonists inhibit the virus assembly step, likely via a cAMP-mediated pathway. Although clonidine administration did not reduce lung virus titers or prevent body weight loss, it did suppress lung edema and improve survival in a murine lethal infection model. Clonidine may thus protect against lung damage caused by influenza virus infection. Our results identify α2-AR-mediated signaling as a key pathway to exploit in the development of anti-influenza agents.


Subject(s)
Adrenergic alpha-2 Receptor Agonists/pharmacology , Clonidine/pharmacology , Influenza A virus/drug effects , Orthomyxoviridae Infections/prevention & control , Receptors, Adrenergic, alpha-2/chemistry , Receptors, Adrenergic, alpha-2/metabolism , Virus Replication/drug effects , Animals , Dogs , Female , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred BALB C , Neuraminidase/metabolism , Orthomyxoviridae Infections/virology
13.
Mol Inform ; 36(12)2017 12.
Article in English | MEDLINE | ID: mdl-28815921

ABSTRACT

In drug and material design, the activity and property values of the designed chemical structures can be predicted by quantitative structure-activity and structure-property relationship (QSAR/QSPR) models. When a QSAR/QSPR model is applied to chemical structures, its applicability domain (AD) must be considered. The predicted activity/property values are only reliable for chemical structures inside the AD. Chemical structures outside the AD are usually neglected, as the predicted values are unreliable. The purpose of this study is to develop a methodology for obtaining novel chemical structures with the desired activity or property based on a QSAR/QSPR model by making use of the neglected structures. We propose a structure modification strategy for the AD that considers the activity and property simultaneously. The AD is defined by a one-class support vector machine and the structure modification is guided by a partial derivative of the AD model and matched molecular pairs analysis. Three proof-of-concept case studies generate novel chemical structures inside the AD that exhibit preferable activity/property values according to the QSAR/QSPR model.


Subject(s)
Quantitative Structure-Activity Relationship , Receptors, Adrenergic, alpha-2/chemistry , Receptors, Adrenergic, alpha-2/metabolism , Databases, Protein , Humans , Models, Molecular , Molecular Structure , Software
14.
J Chem Inf Model ; 56(10): 1885-1893, 2016 10 24.
Article in English | MEDLINE | ID: mdl-27632418

ABSTRACT

To discover drug compounds in chemical space containing an enormous number of compounds, a structure generator is required to produce virtual drug-like chemical structures. The de novo design algorithm for exploring chemical space (DAECS) visualizes the activity distribution on a two-dimensional plane corresponding to chemical space and generates structures in a target area on a plane selected by the user. In this study, we modify the DAECS to enable the user to select a target area to consider properties other than activity and improve the diversity of the generated structures by visualizing the drug-likeness distribution and the activity distribution, generating structures by substructure-based structural changes, including addition, deletion, and substitution of substructures, as well as the slight structural changes used in the DAECS. Through case studies using ligand data for the human adrenergic alpha2A receptor and the human histamine H1 receptor, the modified DAECS can generate high diversity drug-like structures, and the usefulness of the modification of the DAECS is verified.


Subject(s)
Algorithms , Drug Design , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Humans , Ligands , Molecular Docking Simulation , Receptors, Adrenergic, alpha-2/chemistry , Receptors, Adrenergic, alpha-2/metabolism , Receptors, Histamine H1/chemistry , Receptors, Histamine H1/metabolism
15.
PLoS One ; 11(7): e0158486, 2016.
Article in English | MEDLINE | ID: mdl-27384184

ABSTRACT

BACKGROUND: In human cirrhosis, adrenergic hyperfunction causes proximal tubular fluid retention and contributes to diuretic-resistant ascites, and clonidine, a sympatholytic drug, improves natriuresis in difficult-to-treat ascites. AIM: To compare clonidine (aspecific α2-adrenoceptor agonist) to SSP-002021R (prodrug of guanfacine, specific α2A-receptor agonist), both associated with diuretics, in experimental cirrhotic ascites. METHODS AND RESULTS: Six groups of 12 rats were studied: controls (G1); controls receiving furosemide and potassium canrenoate (G2); rats with ascitic cirrhosis due to 14-week CCl4 treatment (G3); cirrhotic rats treated (over the 11th-14th CCl4 weeks) with furosemide and canrenoate (G4), furosemide, canrenoate and clonidine (G5), or diuretics and SSP002021R (G6). Three rats of each group had their hormonal status and renal function assessed at the end of 11th, 12th, 13th, and 14th weeks of respective treatments.Cirrhotic rats in G3 and G4 gained weight over the 12th-14th CCl4 weeks. In G4, brief increase in sodium excretion over the 11th-12th weeks preceded worsening of inulin clearance and natriuresis (diuretic resistance). In comparison with G4, the addition of clonidine (G5) or guanfacine (G6) to diuretics improved, respectively, sodium excretion over the 11th-12th CCl4 weeks, or GFR and electrolytes excretion over the 13th-14th CCl4 weeks. Natriuretic responses in G5 and G6 were accompanied by reduced catecholamine serum levels. CONCLUSIONS: α2A-receptor agonists restore glomerular filtration rate and natriuresis, and delay diuretic-resistant ascites in experimental advanced cirrhosis. Clonidine ameliorates diuretic-dependent natriuresis just for a short time.


Subject(s)
Adrenergic Agonists/therapeutic use , Ascites/drug therapy , Clonidine/therapeutic use , Diuretics/therapeutic use , Fibrosis/drug therapy , Guanfacine/analogs & derivatives , Guanfacine/therapeutic use , Animals , Canrenoic Acid/administration & dosage , Clonidine/administration & dosage , Drug Synergism , Furosemide/administration & dosage , Guanfacine/chemistry , Male , Rats , Rats, Wistar , Receptors, Adrenergic, alpha-2/chemistry
16.
Br J Pharmacol ; 173(17): 2589-99, 2016 09.
Article in English | MEDLINE | ID: mdl-27273730

ABSTRACT

BACKGROUND AND PURPOSE: Agmatine, a putative neurotransmitter, plays a vital role in learning and memory. Although it is considered an endogenous ligand of imidazoline receptors, agmatine exhibits high affinity for α-adrenoceptors, NOS and NMDA receptors. These substrates within the locus coeruleus (LC) are critically involved in learning and memory processes. EXPERIMENTAL APPROACH: The hippocampus and LC of male Wistar rat were stereotaxically cannulated for injection. Effects of agmatine, given i.p. or intra-LC, on acquisition, consolidation and retrieval of inhibitory avoidance (IA) memory were measured. The NO donor S-nitrosoglutathione, non-specific (L-NAME) and specific NOS inhibitors (L-NIL, 7-NI, L-NIO), the α2 -adrenoceptor antagonist (yohimbine) or the corresponding agonist (clonidine) were injected intra-LC before agmatine. Intra-hippocampal injections of the NMDA antagonist, MK-801 (dizocilpine), were used to modify the memory enhancing effects of agmatine, SNG and yohimbine. Expression of tyrosine hydroxylase (TH) and eNOS in the LC was assessed immunohistochemically. KEY RESULTS: Agmatine (intra-LC or i.p.) facilitated memory retrieval in the IA test. S-nitrosoglutathione potentiated, while L-NAME and L-NIO decreased, these effects of agmatine. L-NIL and 7-NI did not alter the effects of agmatine. Yohimbine potentiated, whereas clonidine attenuated, effects of agmatine within the LC. The effects of agmatine, S-nitrosoglutathione and yohimbine were blocked by intra-hippocampal MK-801. Agmatine increased the population of TH- and eNOS-immunoreactive elements in the LC. CONCLUSIONS AND IMPLICATIONS: The facilitation of memory retrieval in the IA test by agmatine is probably mediated by interactions between eNOS, NO and noradrenergic pathways in the LC.


Subject(s)
Agmatine/pharmacology , Avoidance Learning/drug effects , Locus Coeruleus/drug effects , Locus Coeruleus/metabolism , Nitric Oxide/metabolism , Receptors, Adrenergic, alpha-2/metabolism , Agmatine/administration & dosage , Animals , Dose-Response Relationship, Drug , Male , Nitric Oxide/chemistry , Rats , Rats, Wistar , Receptors, Adrenergic, alpha-2/chemistry , Structure-Activity Relationship
17.
PLoS One ; 10(4): e0120788, 2015.
Article in English | MEDLINE | ID: mdl-25860977

ABSTRACT

The objective of this study was to clarify the effects of disease on neurally mediated syncope (NMS) during an acute stress reaction. We analyzed the mechanism of the molecular interaction and the polymorphisms of the alpha-2 adrenoreceptor (α2B-AR) gene as the potential psychiatric cause of incentive stress. We focused on the following three genotypes of the repeat polymorphism site at Glu 301-303 in the α2B-AR gene: Glu12/12, Glu12/9, and Glu9/9. On the basis of our clinical research, NMS is likely to occur in people with the Glu12/9 heterotype. To verify this, we assessed this relationship with the interaction of Gi protein and adenylate cyclase by in silico analysis of the Glu12/9 heterotype. By measuring the difference in the dissociation time of the Gi-α subunit twice, we found that the Glu12/9 heterotype suppressed the action of adenylate cyclase longer than the Glu homotypes. As this difference in the Glu repeat number effect is thought to be one of the causes of NMS, we investigated the evolutionary significance of the Glu repeat number. Glu8 was originally repeated in simians, while the Glu12 repeats occurred over time during the evolution of bipedalism in humans. Taken with the Glu12 numbers, NMS would likely become a defensive measure to prevent significant blood flow to the human brain.


Subject(s)
Receptors, Adrenergic, alpha-2/genetics , Syncope, Vasovagal/pathology , Alleles , Animals , Base Sequence , DNA/analysis , DNA/isolation & purification , DNA, Mitochondrial/analysis , DNA, Mitochondrial/classification , Epinephrine/blood , Evolution, Molecular , Female , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Gene Frequency , Genotype , Humans , Male , Norepinephrine/blood , Phylogeny , Polymorphism, Single Nucleotide , Primates/genetics , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Receptors, Adrenergic, alpha-2/chemistry , Receptors, Adrenergic, alpha-2/metabolism , Syncope, Vasovagal/metabolism , Thermodynamics
18.
Bioorg Med Chem ; 23(14): 3991-9, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-25648685

ABSTRACT

Fragment-based drug discovery has emerged as an alternative to conventional lead identification and optimization strategies generally supported by biophysical detection techniques. Membrane targets like G protein-coupled receptors (GPCRs), however, offer challenges in lack of generic immobilization or stabilization methods for the dynamic, membrane-bound supramolecular complexes. Also modeling of different functional states of GPCRs proved to be a challenging task. Here we report a functional cell-based high concentration screening campaign for the identification of adrenergic α2C receptor agonists compared with the virtual screening of the same ligand set against an active-like homology model of the α2C receptor. The conventional calcium mobilization-based assay identified active fragments with a similar incidence to several other reported fragment screens on GPCRs. 16 out of 3071 screened fragments turned out as specific ligands of α2C, two of which were identified by virtual screening as well and several of the hits possessed surprisingly high affinity and ligand efficiency. Our results indicate that in vitro biological assays can be utilized in the fragment hit identification process for GPCR targets.


Subject(s)
Adrenergic alpha-2 Receptor Agonists/pharmacology , Drug Evaluation, Preclinical/methods , Receptors, Adrenergic, alpha-2/metabolism , Adrenergic alpha-2 Receptor Agonists/chemistry , Animals , CHO Cells/drug effects , Cricetulus , Humans , Ligands , Protein Conformation , Receptors, Adrenergic, alpha-2/chemistry , Receptors, Adrenergic, alpha-2/genetics , Structure-Activity Relationship , User-Computer Interface
19.
Nat Chem Biol ; 11(4): 271-9, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25706338

ABSTRACT

Hypersecretion of norepinephrine (NE) and angiotensin II (AngII) is a hallmark of major prevalent cardiovascular diseases that contribute to cardiac pathophysiology and morbidity. Herein, we explore whether heterodimerization of presynaptic AngII AT1 receptor (AT1-R) and NE α2C-adrenergic receptor (α2C-AR) could underlie their functional cross-talk to control NE secretion. Multiple bioluminescence resonance energy transfer and protein complementation assays allowed us to accurately probe the structures and functions of the α2C-AR-AT1-R dimer promoted by ligand binding to individual protomers. We found that dual agonist occupancy resulted in a conformation of the heterodimer different from that induced by active individual protomers and triggered atypical Gs-cAMP-PKA signaling. This specific pharmacological signaling unit was identified in vivo to promote not only NE hypersecretion in sympathetic neurons but also sympathetic hyperactivity in mice. Thus, we uncovered a new process by which GPCR heterodimerization creates an original functional pharmacological entity and that could constitute a promising new target in cardiovascular therapeutics.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Receptor, Angiotensin, Type 1/agonists , Signal Transduction , Adrenergic alpha-Agonists/chemistry , Animals , Biophysics , Cardiovascular Diseases/metabolism , Cyclic AMP/metabolism , Dimerization , Drug Design , GTP-Binding Proteins/metabolism , HEK293 Cells , Humans , Ligands , Male , Mice , Mice, Inbred C57BL , Neurons/metabolism , Norepinephrine/chemistry , PC12 Cells , Phosphorylation , Protein Conformation , Rats , Receptors, Adrenergic, alpha-2/chemistry , Sympathetic Nervous System/drug effects
20.
PLoS One ; 9(8): e103099, 2014.
Article in English | MEDLINE | ID: mdl-25110951

ABSTRACT

Vascular smooth muscle α2C-adrenoceptors (α2C-ARs) mediate vasoconstriction of small blood vessels, especially arterioles. Studies of endogenous receptors in human arteriolar smooth muscle cells (referred to as microVSM) and transiently transfected receptors in heterologous HEK293 cells show that the α2C-ARs are perinuclear receptors that translocate to the cell surface under cellular stress and elicit a biological response. Recent studies in microVSM unraveled a crucial role of Rap1A-Rho-ROCK-F-actin pathways in receptor translocation, and identified protein-protein interaction of α2C-ARs with the actin binding protein filamin-2 as an essential step in the process. To better understand the molecular nature and specificity of this interaction, in this study, we constructed comparative models of human α2C-AR and human filamin-2 proteins. Finally, we performed in silico protein-protein docking to provide a structural platform for the investigation of human α2C-AR and filamin-2 interactions. We found that electrostatic interactions seem to play a key role in this complex formation which manifests in interactions between the C-terminal arginines of α2C-ARs (particularly R454 and R456) and negatively charged residues from filamin-2 region between residues 1979 and 2206. Phylogenetic and sequence analysis showed that these interactions have evolved in warm-blooded animals.


Subject(s)
Computer Simulation , Filamins/metabolism , Molecular Docking Simulation , Receptors, Adrenergic, alpha-2/metabolism , Amino Acid Sequence , Filamins/chemistry , Humans , Molecular Sequence Data , Phylogeny , Protein Binding , Protein Structure, Tertiary , Receptors, Adrenergic, alpha-2/chemistry , Sequence Alignment , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...