Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 7 de 7
1.
J Med Chem ; 65(4): 3539-3562, 2022 02 24.
Article En | MEDLINE | ID: mdl-35077170

The sphingosine-1-phosphate-1 (S1P1) receptor agonists have great potential for the treatment of multiple sclerosis (MS) because they can inhibit lymphocyte egress through receptor internalization. We designed and synthesized triazole and isoxazoline derivatives to discover a novel S1P1 agonist for MS treatment. Of the two scaffolds, the isoxazoline derivative was determined to have excellent in vitro efficacy and drug-like properties. Among them, compound 21l was found to have superior drug-like properties as well as excellent in vitro efficacies (EC50 = 7.03 nM in ß-arrestin recruitment and EC50 = 11.8 nM in internalization). We also confirmed that 21l effectively inhibited lymphocyte egress in the peripheral lymphocyte count test and significantly improved the clinical score in the experimental autoimmune encephalitis MS mouse model.


Multiple Sclerosis/drug therapy , Sphingosine-1-Phosphate Receptors/antagonists & inhibitors , Animals , Dogs , Drug Design , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Heart Rate/drug effects , Humans , Isoxazoles/chemical synthesis , Isoxazoles/pharmacokinetics , Isoxazoles/pharmacology , Lymphocyte Count , Lymphocytes/drug effects , Male , Mice , Rats , Structure-Activity Relationship , Triazoles/chemical synthesis , Triazoles/pharmacokinetics , Triazoles/pharmacology , beta-Arrestins/drug effects
2.
Commun Biol ; 4(1): 569, 2021 05 12.
Article En | MEDLINE | ID: mdl-33980979

Following the FDA-approval of the hematopoietic stem cell (HSC) mobilizer plerixafor, orally available and potent CXCR4 antagonists were pursued. One such proposition was AMD11070, which was orally active and had superior antagonism in vitro; however, it did not appear as effective for HSC mobilization in vivo. Here we show that while AMD11070 acts as a full antagonist, plerixafor acts biased by stimulating ß-arrestin recruitment while fully antagonizing G protein. Consequently, while AMD11070 prevents the constitutive receptor internalization, plerixafor allows it and thereby decreases receptor expression. These findings are confirmed by the successful transfer of both ligands' binding sites and action to the related CXCR3 receptor. In vivo, plerixafor exhibits superior HSC mobilization associated with a dramatic reversal of the CXCL12 gradient across the bone marrow endothelium, which is not seen for AMD11070. We propose that the biased action of plerixafor is central for its superior therapeutic effect in HSC mobilization.


Benzylamines/pharmacology , Cyclams/pharmacology , Hematopoietic Stem Cell Mobilization/methods , Receptors, CXCR4/metabolism , Aminoquinolines/metabolism , Aminoquinolines/pharmacology , Animals , Benzimidazoles/metabolism , Benzimidazoles/pharmacology , Benzylamines/metabolism , Butylamines/metabolism , Butylamines/pharmacology , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Cyclams/metabolism , Drug Delivery Systems/methods , Female , Granulocyte Colony-Stimulating Factor , HEK293 Cells , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Humans , Mice , Mice, Inbred C57BL , Pharmaceutical Preparations/metabolism , Receptors, CXCR3/drug effects , Receptors, CXCR3/metabolism , Receptors, CXCR4/drug effects , beta-Arrestins/drug effects , beta-Arrestins/metabolism
3.
Circ Heart Fail ; 14(3): e007351, 2021 03.
Article En | MEDLINE | ID: mdl-33663236

BACKGROUND: New heart failure therapies that safely augment cardiac contractility and output are needed. Previous apelin peptide studies have highlighted the potential for APJ (apelin receptor) agonism to enhance cardiac function in heart failure. However, apelin's short half-life limits its therapeutic utility. Here, we describe the preclinical characterization of a novel, orally bioavailable APJ agonist, BMS-986224. METHODS: BMS-986224 pharmacology was compared with (Pyr1) apelin-13 using radio ligand binding and signaling pathway assays downstream of APJ (cAMP, phosphorylated ERK [extracellular signal-regulated kinase], bioluminescence resonance energy transfer-based G-protein assays, ß-arrestin recruitment, and receptor internalization). Acute effects on cardiac function were studied in anesthetized instrumented rats. Chronic effects of BMS-986224 were assessed echocardiographically in the RHR (renal hypertensive rat) model of cardiac hypertrophy and decreased cardiac output. RESULTS: BMS-986224 was a potent (Kd=0.3 nmol/L) and selective APJ agonist, exhibiting similar receptor binding and signaling profile to (Pyr1) apelin-13. G-protein signaling assays in human embryonic kidney 293 cells and human cardiomyocytes confirmed this and demonstrated a lack of signaling bias relative to (Pyr1) apelin-13. In anesthetized instrumented rats, short-term BMS-986224 infusion increased cardiac output (10%-15%) without affecting heart rate, which was similar to (Pyr1) apelin-13 but differentiated from dobutamine. Subcutaneous and oral BMS-986224 administration in the RHR model increased stroke volume and cardiac output to levels seen in healthy animals but without preventing cardiac hypertrophy and fibrosis, effects differentiated from enalapril. CONCLUSIONS: We identify a novel, potent, and orally bioavailable nonpeptidic APJ agonist that closely recapitulates the signaling properties of (Pyr1) apelin-13. We show that oral APJ agonist administration induces a sustained increase in cardiac output in the cardiac disease setting and exhibits a differentiated profile from the renin-angiotensin system inhibitor enalapril, supporting further clinical evaluation of BMS-986224 in heart failure.


Apelin Receptors/agonists , Cardiac Output/drug effects , Heart Failure/physiopathology , Intercellular Signaling Peptides and Proteins/pharmacology , Stroke Volume/drug effects , Animals , Bioluminescence Resonance Energy Transfer Techniques , CHO Cells , Cricetulus , Dogs , Extracellular Signal-Regulated MAP Kinases/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , HEK293 Cells , Haplorhini , Humans , In Vitro Techniques , MAP Kinase Signaling System/drug effects , Phosphorylation , Radioligand Assay , Rats , Tritium , Ventricular Pressure/drug effects , beta-Arrestins/drug effects , beta-Arrestins/metabolism
4.
Cardiovasc Drugs Ther ; 35(6): 1095-1110, 2021 12.
Article En | MEDLINE | ID: mdl-32474680

PURPOSE: This study tested if the protective anti-remodeling effect of GLP-1 agonist Exendin-4 after an acute myocardial infarction (MI) in rats involves inhibition of the Wnt1/ß-catenin signaling pathway. METHODS: Rats were divided into sham, sham + Exendin-4 (10 µg/day, i.p), MI, and MI + Exendin-4. MI was introduced to rats by permanent left anterior descending coronary artery (LAD) ligation. RESULTS: On day 7 post-infraction, MI rats showed LV dysfunction with higher serum levels of cardiac markers. Their remote myocardia showed increased mRNA and protein levels of collagen I/III with higher levels of reactive oxygen species (ROS) and inflammatory cytokines, as well as protein levels of Wnt1, phospho-Akt, transforming growth factor (TGF-ß1), Smad, phospho-Smad3, α-SMA, caspase-3, and Bax. They also showed higher protein levels of phospho-glycogen synthase kinase-3ß (p-GSK3ß), as well as total, phosphorylated, and nuclear ß-catenin with a concomitant decrease in the levels of cyclic adenosine monophosphate (cAMP), mRNA of manganese superoxide dismutase (MnSOD), and protein levels of Bcl-2, ß-arrestin-2, and protein phosphatase-2 (PP2A). Administration of Exendin-4 to MI rats reduced the infarct size and reversed the aforementioned signaling molecules without altering protein levels of TGF-1ß and Wnt1 or Akt activation. Interestingly, Exendin-4 increased mRNA levels of MnSOD, protein levels of ß-arrestin-2 and PP2A, and ß-catenin phosphorylation but reduced the phosphorylation of GSK3ß and Smad3, and total ß-catenin levels in the LV of control rats. CONCLUSION: Exendin-4 inhibits the remodeling in the remote myocardium of rats following acute MI by attenuating ß-catenin activation and activating ß-arrestin-2, PP2A, and GSK3ß. Graphical Abstract A graphical abstract that illustrates the mechanisms by which Exendin-4 inhibits cardiac remodeling in remote myocardium of left ventricle MI-induced rats. Mechanisms are assumed to occur in the cardiomyocytes and/or other resident cells such as fibroblast. Β-catenin activation and nuclear translocation are associated with increased synthesis of inflammatory cytokines and transforming growth factor ß-1 (TGF-ß1). GSK3ß is inhibited by phosphorylation at Ser9. Under normal conditions, ß-catenin is degraded in the cytoplasm by the active GSK3ß-dependent degradation complex (un-phosphorylated) which usually phosphorylates ß-catenin at Ser33/37/Thr41. After MI, TGF-ß1, and Wnt 1 levels are significantly increased, the overproduction of Wnt1 induces ß-catenin stabilization and nuclear translocation through increasing the phosphorylation of disheveled (DVL) protein which in turn phosphorylates and inhibits GSK3ß. TGF-ß1 stimulates the phosphorylation of Smad-3 and subsequent nuclear translocation to activate the transcription of collage 1/III and α-smooth muscle actin (α-SMA). Besides, TGF-ß1 stabilizes cytoplasmic ß-catenin levels indirectly by phosphorylation of Akt at Thr308-induced inhibition of GSK3ß by increasing phosphorylation of Ser9. Exendin-4, and possibly through G protein-coupled receptors (GPCRs), increases levels of cAMP and upregulates ß-arrestin-2 levels. Both can result in a positive inotropic effect. Besides, ß-arrestin-2 can stimulate PP2A to dephosphorylation Smad3 (inhibition) and GSK3ß (activation), thus reduces fibrosis and prevents the activation of ß-catenin and collagen deposition.


Exenatide/pharmacology , Glycogen Synthase Kinase 3/drug effects , Myocardial Infarction/physiopathology , Protein Phosphatase 2/drug effects , beta Catenin/drug effects , beta-Arrestins/drug effects , Animals , Hemodynamics/drug effects , Male , Phosphorylation , Rats , Rats, Wistar , Wnt1 Protein/drug effects
5.
J Psychopharmacol ; 34(12): 1431-1442, 2020 12.
Article En | MEDLINE | ID: mdl-33103555

BACKGROUND: Our previous studies showed that xanthone derivatives with N-(2-methoxyphenyl)piperazine fragment have an affinity to the 5-HT1A receptor and show antidepressant-like properties in rodents. In this study, we tested three xanthone derivatives, HBK-1 (R, S) and its enantiomers, in which we increased the distance between the piperazine and xanthone fragments by using a hydroxypropoxy linker. We hypothesized that this would increase the binding to the 5-HT1A receptor and consequently, pharmacological activity. AIMS: We aimed to assess the in vitro and in vivo pharmacological activity of the xanthone derivatives. METHODS: We evaluated the in vitro affinity for serotonin 5-HT1A and 5-HT2A receptors and serotonin transporter. We also determined the intrinsic activity at the 5-HT1A receptor. We investigated the antidepressant-like properties and safety after acute administration (dose range: 1.25-20 mg/kg) using the forced swim, tail suspension, locomotor activity, rotarod and chimney tests in mice. We also evaluated the basic pharmacokinetic parameters. RESULTS: Our results indicated that the compounds showed a high affinity for the 5-HT1A receptor but very weak antagonistic properties in the Ca2+ mobilization assay; however, they showed significant agonistic properties in the ß-arrestin recruitment assay. In both behavioural tests the studied xanthone derivatives showed antidepressant-like activity. Pre-treatment with p-chlorophenylalanine or WAY-100635 abolished their antidepressant-like activity. None of the compounds caused motor impairments at antidepressant-like doses. The racemate penetrated the blood-brain barrier and had a relatively high bioavailability after intraperitoneal administration. CONCLUSIONS: Xanthone derivatives with N-(2-methoxyphenyl)piperazine fragment and hydroxypropoxy linker show increased binding to the 5-HT1A receptor and may represent an attractive putative treatment candidate for depression.


Antidepressive Agents/pharmacology , Receptor, Serotonin, 5-HT1A/drug effects , Signal Transduction/drug effects , Xanthones/pharmacology , beta-Arrestins/drug effects , Animals , Antidepressive Agents/administration & dosage , Antidepressive Agents/pharmacokinetics , Behavior, Animal/drug effects , Male , Mice , Xanthones/administration & dosage , Xanthones/pharmacokinetics
6.
Physiol Rep ; 7(19): e14225, 2019 10.
Article En | MEDLINE | ID: mdl-31565870

Teriparatide and abaloparatide are parathyroid hormone receptor 1 (PTHR1) analogs with unexplained differential efficacy for the treatment of osteoporosis. Therefore, we compared the effects of abaloparatide and teriparatide on bone structure, turnover, and levels of receptor activator of nuclear factor-kappa B ligand (RANKL) and osteoprotegerin (OPG). Wild-type (WT) female mice were injected daily with vehicle or 20-80 µg/kg/day of teriparatide or abaloparatide for 30 days. Femurs and spines were examined by microcomputed tomography scanning and serum levels of bone turnover markers, RANKL, and OPG, were measured by ELISA. Both analogs similarly increased the distal femoral fractional trabecular bone volume, connectivity, and number, and reduced the structure model index (SMI) at 20-80 µg/kg/day doses. However, only abaloparatide exhibited a significant increase (13%) in trabecular thickness at 20 µg/kg/day dose. Femoral cortical evaluation showed that abaloparatide caused a greater dose-dependent increase in cortical thickness than teriparatide. Both teriparatide and abaloparatide increased lumbar 5 vertebral trabecular connectivity but had no or modest effect on other indices. Biochemical analysis demonstrated that abaloparatide promoted greater elevation of procollagen type 1 intact N-terminal propeptide, a bone formation marker, and tartrate-resistant acid phosphatase 5b levels, a bone resorption marker, and lowered the RANKL/OPG ratio. Furthermore, PTHR1 signaling was compared in cells treated with 0-100 nmol/L analog. Interestingly, abaloparatide had a markedly lower EC50 for cAMP formation (2.3-fold) and ß-arrestin recruitment (1.6-fold) than teriparatide. Therefore, abaloparatide-improved efficacy can be attributed to enhanced bone formation and cortical structure, reduced RANKL/OPG ratio, and amplified Gs-cAMP and ß-arrestin signaling.


Bone Density Conservation Agents/pharmacology , Bone Remodeling/drug effects , Bone and Bones/drug effects , Parathyroid Hormone-Related Protein/pharmacology , Teriparatide/pharmacology , Animals , Bone Density/drug effects , Cyclic AMP/metabolism , Female , Mice , Mice, Inbred C57BL , beta-Arrestins/drug effects , beta-Arrestins/metabolism
7.
J Med Chem ; 62(10): 5111-5131, 2019 05 23.
Article En | MEDLINE | ID: mdl-31042379

Starting from the ß-adrenoceptor agonist isoprenaline and beta-blocker carvedilol, we designed and synthesized three different chemotypes of agonist/antagonist hybrids. Investigations of ligand-mediated receptor activation using bioluminescence resonance energy transfer biosensors revealed a predominant effect of the aromatic head group on the intrinsic activity of our ligands, as ligands with a carvedilol head group were devoid of agonistic activity. Ligands composed of a catechol head group and an antagonist-like oxypropylene spacer possess significant intrinsic activity for the activation of Gαs, while they only show weak or even no ß-arrestin-2 recruitment at both ß1- and ß2-AR. Molecular dynamics simulations suggest that the difference in G protein efficacy and ß-arrestin recruitment of the hybrid ( S)-22, the full agonist epinephrine, and the ß2-selective, G protein-biased partial agonist salmeterol depends on specific hydrogen bonding between Ser5.46 and Asn6.55, and the aromatic head group of the ligands.


Adrenergic beta-Agonists/chemistry , Adrenergic beta-Antagonists/chemistry , GTP-Binding Proteins/drug effects , Adrenergic beta-Agonists/pharmacology , Adrenergic beta-Antagonists/pharmacology , Animals , Carvedilol/chemical synthesis , Carvedilol/chemistry , Catechols/chemistry , Drug Design , Humans , Hydrogen Bonding , Indicators and Reagents , Isoproterenol/chemical synthesis , Isoproterenol/chemistry , Ligands , Mice , Models, Molecular , Molecular Dynamics Simulation , Salmeterol Xinafoate/pharmacology , beta-Arrestins/drug effects , beta-Arrestins/metabolism
...