Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 315
Filter
1.
J Thromb Haemost ; 21(7): 1891-1902, 2023 07.
Article in English | MEDLINE | ID: mdl-36958516

ABSTRACT

BACKGROUND: The hemostatic plug formation at sites of vascular injury is strongly dependent on rapid platelet activation and integrin-mediated adhesion and aggregation. However, to prevent thrombotic complications, platelet aggregate formation must be a self-limiting process. The second-wave mediator adenosine diphosphate (ADP) activates platelets via Gq-coupled P2Y1 and Gi-coupled P2Y12 receptors. After ADP exposure, the P2Y1 receptor undergoes rapid phosphorylation-induced desensitization, a negative feedback mechanism believed to be critical for limiting thrombus growth. OBJECTIVE: The objective of this study was to examine the role of rapid P2Y1 receptor desensitization on platelet function and thrombus formation in vivo. METHODS: We analyzed a novel knock-in mouse strain expressing a P2Y1 receptor variant that cannot be phosphorylated beyond residue 340 (P2Y1340-0P), thereby preventing the desensitization of the receptor. RESULTS: P2Y1340-0P mice followed a Mendelian inheritance pattern, and peripheral platelet counts were comparable between P2Y1340-0P/340-0P and control mice. In vitro, P2Y1340-0P/340-0P platelets were hyperreactive to ADP, showed a robust activation response to the P2Y1 receptor-selective agonist, MRS2365, and did not desensitize in response to repeated ADP challenge. We observed increased calcium mobilization, protein kinase C substrate phosphorylation, alpha granule release, activation of the small GTPase Rap1, and integrin inside-out activation/aggregation. This hyperreactivity, however, did not lead to increased platelet adhesion or excessive plug formation under physiological shear conditions. CONCLUSION: Our studies demonstrate that receptor phosphorylation at the C-terminus is critical for P2Y1 receptor desensitization in platelets and that impaired desensitization leads to increased P2Y1 receptor signaling in vitro. Surprisingly, desensitization of the P2Y1 receptor is not required for limiting platelet adhesion/aggregation at sites of vascular injury, likely because ADP is degraded quickly or washed away in the bloodstream.


Subject(s)
Thrombosis , Vascular System Injuries , Mice , Animals , Platelet Aggregation , Blood Platelets/metabolism , Hemostasis , Thrombosis/genetics , Thrombosis/prevention & control , Thrombosis/metabolism , Adenosine Diphosphate/pharmacology , Integrins/metabolism , Receptors, Purinergic P2Y1/genetics , Receptors, Purinergic P2Y1/metabolism , Receptors, Purinergic P2Y12/genetics , Receptors, Purinergic P2Y12/metabolism
2.
Elife ; 82019 12 31.
Article in English | MEDLINE | ID: mdl-31889510

ABSTRACT

Direct activation of the human phospholipase C-γ isozymes (PLC-γ1, -γ2) by tyrosine phosphorylation is fundamental to the control of diverse biological processes, including chemotaxis, platelet aggregation, and adaptive immunity. In turn, aberrant activation of PLC-γ1 and PLC-γ2 is implicated in inflammation, autoimmunity, and cancer. Although structures of isolated domains from PLC-γ isozymes are available, these structures are insufficient to define how release of basal autoinhibition is coupled to phosphorylation-dependent enzyme activation. Here, we describe the first high-resolution structure of a full-length PLC-γ isozyme and use it to underpin a detailed model of their membrane-dependent regulation. Notably, an interlinked set of regulatory domains integrates basal autoinhibition, tyrosine kinase engagement, and additional scaffolding functions with the phosphorylation-dependent, allosteric control of phospholipase activation. The model also explains why mutant forms of the PLC-γ isozymes found in several cancers have a wide spectrum of activities, and highlights how these activities are tuned during disease.


Many enzymes are poised to receive signals from the surrounding environment and translate them into responses inside the cell. One such enzyme is phospholipase C-γ1 (PLC-γ1), which controls how cells grow, divide and migrate.When activating signals are absent, PLC-γ1 usually inhibits its own activity, a mechanism called autoinhibition. This prevents the enzyme from binding to its targets, which are fat molecules known as lipids. When activating signals are present, a phosphate group serves as a 'chemical tag' and is added onto PLC-γ1, allowing the enzyme to bind to lipids.Failure in the regulation of PLC-γ1 or other closely related enzymes may lead to conditions such as cancer, arthritis and Alzheimer's disease. However, it remains unclear how autoinhibition suppresses the activity of the enzyme, and how it is stopped by the addition of the phosphate group.Here, Hajicek et al. determine in great detail the three-dimensional structure of the autoinhibited form of the enzyme using a method known as X-ray crystallography. This reveals that PLC-γ1 has two major lobes: one contains the active site that modifies lipids, and the other sits on top of the active site to prevent lipids from reaching it. The findings suggest that when the phosphate group attaches to PLC-γ1, it triggers a large shape change that shifts the second lobe away from the active site to allow lipids to bind.The three-dimensional structure also helps to understand how mutations identified in certain cancers may activate PLC-γ1. In particular, these mutations disrupt the interactions between elements that usually hold the two lobes together, causing the enzyme to activate more easily.The work by Hajicek et al. provides a framework to understand how cells control PLC-γ1. It is a first step toward designing new drugs that alter the activity of this enzyme, which may ultimately be useful to treat cancer and other diseases.


Subject(s)
Enzyme Activation/genetics , Isoenzymes/ultrastructure , Phospholipase C gamma/ultrastructure , Protein Conformation , Crystallography, X-Ray , Humans , Isoenzymes/chemistry , Isoenzymes/genetics , Mutation/genetics , Neoplasms/genetics , Neoplasms/pathology , Phospholipase C gamma/chemistry , Phospholipase C gamma/genetics , Phosphorylation/genetics , Protein Domains/genetics , Tyrosine/genetics
3.
J Biol Chem ; 293(5): 1728-1735, 2018 02 02.
Article in English | MEDLINE | ID: mdl-29263090

ABSTRACT

A diverse group of cell-surface receptors, including many G protein-coupled receptors and receptor tyrosine kinases, activate phospholipase C (PLC) isozymes to hydrolyze phosphatidylinositol 4,5-bisphosphate into the second messengers diacylglycerol and 1,4,5-inositol trisphosphate. Consequently, PLCs control various cellular processes, and their aberrant regulation contributes to many diseases, including cancer, atherosclerosis, and rheumatoid arthritis. Despite the widespread importance of PLCs in human biology and disease, it has been impossible to directly monitor the real-time activation of these enzymes at membranes. To overcome this limitation, here we describe XY-69, a fluorogenic reporter that preferentially partitions into membranes and provides a selective tool for measuring the real-time activity of PLCs as either purified enzymes or in cellular lysates. Indeed, XY-69 faithfully reported the membrane-dependent activation of PLC-ß3 by Gαq Therefore, XY-69 can replace radioactive phosphatidylinositol 4,5-bisphosphate used in conventional PLC assays and will enable high-throughput screens to identify both orthosteric and allosteric PLC inhibitors. In the future, cell-permeable variants of XY-69 represent promising candidates for reporting the activation of PLCs in live cells with high spatiotemporal resolution.


Subject(s)
Cell Membrane/enzymology , Fluorescence , Genes, Reporter , Phospholipase C beta/metabolism , Cell Membrane/genetics , GTP-Binding Protein alpha Subunits/genetics , GTP-Binding Protein alpha Subunits/metabolism , GTP-Binding Protein beta Subunits/genetics , GTP-Binding Protein beta Subunits/metabolism , HEK293 Cells , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Phosphatidylinositol Phosphates/genetics , Phosphatidylinositol Phosphates/metabolism , Phospholipase C beta/genetics
4.
J Biol Chem ; 291(49): 25608-25616, 2016 Dec 02.
Article in English | MEDLINE | ID: mdl-27742837

ABSTRACT

In contrast to G protein-coupled receptors, for which chemical and peptidic inhibitors have been extensively explored, few compounds are available that directly modulate heterotrimeric G proteins. Active Gαq binds its two major classes of effectors, the phospholipase C (PLC)-ß isozymes and Rho guanine nucleotide exchange factors (RhoGEFs) related to Trio, in a strikingly similar fashion: a continuous helix-turn-helix of the effectors engages Gαq within its canonical binding site consisting of a groove formed between switch II and helix α3. This information was exploited to synthesize peptides that bound active Gαq in vitro with affinities similar to full-length effectors and directly competed with effectors for engagement of Gαq A representative peptide was specific for active Gαq because it did not bind inactive Gαq or other classes of active Gα subunits and did not inhibit the activation of PLC-ß3 by Gß1γ2 In contrast, the peptide robustly prevented activation of PLC-ß3 or p63RhoGEF by Gαq; it also prevented G protein-coupled receptor-promoted neuronal depolarization downstream of Gαq in the mouse prefrontal cortex. Moreover, a genetically encoded form of this peptide flanked by fluorescent proteins inhibited Gαq-dependent activation of PLC-ß3 at least as effectively as a dominant-negative form of full-length PLC-ß3. These attributes suggest that related, cell-penetrating peptides should effectively inhibit active Gαq in cells and that these and genetically encoded sequences may find application as molecular probes, drug leads, and biosensors to monitor the spatiotemporal activation of Gαq in cells.


Subject(s)
Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/pharmacology , GTP-Binding Protein alpha Subunits, Gq-G11/antagonists & inhibitors , Prefrontal Cortex/metabolism , Animals , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/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 , HEK293 Cells , Humans , Mice , Phospholipase C beta/genetics , Phospholipase C beta/metabolism , Protein Structure, Secondary
5.
Purinergic Signal ; 12(4): 627-635, 2016 12.
Article in English | MEDLINE | ID: mdl-27421735

ABSTRACT

In addition to their role in glycosylation reactions, UDP-sugars are released from cells and activate widely distributed cell surface P2Y14 receptors (P2Y14R). However, the physiological/pathophysiological consequences of UDP-sugar release are incompletely defined. Here, we report that UDP-glucose levels are abnormally elevated in lung secretions from patients with cystic fibrosis (CF) as well as in a mouse model of CF-like disease, the ßENaC transgenic (Tg) mouse. Instillation of UDP-glucose into wild-type mouse tracheas resulted in enhanced neutrophil lung recruitment, and this effect was nearly abolished when UDP-glucose was co-instilled with the P2Y14R antagonist PPTN [4-(piperidin-4-yl)-phenyl)-7-(4-(trifluoromethyl)-phenyl-2-naphthoic acid]. Importantly, administration of PPTN to ßENaC-Tg mice reduced neutrophil lung inflammation. These results suggest that UDP-glucose released into the airways acts as a local mediator of neutrophil inflammation.


Subject(s)
Cystic Fibrosis/metabolism , Lung/drug effects , Neutrophil Infiltration/drug effects , Neutrophils/drug effects , Uridine Diphosphate Glucose/pharmacology , Adenosine Triphosphate/metabolism , Adult , Animals , Cystic Fibrosis/immunology , Cytokines/metabolism , Disease Models, Animal , Female , Humans , Lung/immunology , Lung/metabolism , Male , Mice , Neutrophils/immunology , Neutrophils/metabolism , Sputum/immunology , Sputum/metabolism , Trachea/drug effects , Trachea/immunology , Uridine Diphosphate Glucose/metabolism , Young Adult
6.
J Med Chem ; 59(13): 6149-68, 2016 07 14.
Article in English | MEDLINE | ID: mdl-27331270

ABSTRACT

UDP and UDP-glucose activate the P2Y14 receptor (P2Y14R) to modulate processes related to inflammation, diabetes, and asthma. A computational pipeline suggested alternatives to naphthalene of a previously reported P2Y14R antagonist (3, PPTN) using docking and molecular dynamics simulations on a hP2Y14R homology model based on P2Y12R structures. By reevaluating the binding of 3 to P2Y14R computationally, two alternatives, i.e., alkynyl and triazolyl derivatives, were identified. Improved synthesis of fluorescent antagonist 4 enabled affinity quantification (IC50s, nM) using flow cytometry of P2Y14R-expressing CHO cells. p-F3C-phenyl-triazole 65 (32) was more potent than a corresponding alkyne 11. Thus, additional triazolyl derivatives were prepared, as guided by docking simulations, with nonpolar aryl substituents favored. Although triazoles were less potent than 3 (6), simpler synthesis facilitated further structural optimization. Additionally, relative P2Y14R affinities agreed with predicted binding of alkynyl and triazole analogues. These triazoles, designed through a structure-based approach, can be assessed in disease models.


Subject(s)
Drug Design , Purinergic P2 Receptor Antagonists/chemistry , Purinergic P2 Receptor Antagonists/pharmacology , Receptors, Purinergic P2/metabolism , Triazoles/chemistry , Triazoles/pharmacology , Animals , Biphenyl Compounds/chemistry , Biphenyl Compounds/pharmacology , CHO Cells , Cricetulus , Humans , Molecular Docking Simulation , Molecular Dynamics Simulation , Structure-Activity Relationship
7.
Nat Commun ; 6: 10156, 2015 Dec 14.
Article in English | MEDLINE | ID: mdl-26658454

ABSTRACT

Despite the discovery of heterotrimeric αßγ G proteins ∼25 years ago, their selective perturbation by cell-permeable inhibitors remains a fundamental challenge. Here we report that the plant-derived depsipeptide FR900359 (FR) is ideally suited to this task. Using a multifaceted approach we systematically characterize FR as a selective inhibitor of Gq/11/14 over all other mammalian Gα isoforms and elaborate its molecular mechanism of action. We also use FR to investigate whether inhibition of Gq proteins is an effective post-receptor strategy to target oncogenic signalling, using melanoma as a model system. FR suppresses many of the hallmark features that are central to the malignancy of melanoma cells, thereby providing new opportunities for therapeutic intervention. Just as pertussis toxin is used extensively to probe and inhibit the signalling of Gi/o proteins, we anticipate that FR will at least be its equivalent for investigating the biological relevance of Gq.


Subject(s)
Depsipeptides/pharmacology , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Animals , Ardisia/chemistry , Cell Line, Tumor , Depsipeptides/chemistry , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , Humans , Melanoma/metabolism , Mice , Models, Molecular , Molecular Structure , Protein Conformation , Protein Isoforms , Signal Transduction , Tail/blood supply , Vasoconstriction/drug effects
8.
Bioorg Med Chem Lett ; 25(21): 4733-4739, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26303895

ABSTRACT

The P2Y14R is a G(i/o)-coupled receptor of the P2Y family of purinergic receptors that is activated by extracellular UDP and UDP-glucose (UDPG). In an earlier report we described a P2Y14R fluorescent probe, MRS4174, based on the potent and selective antagonist PPTN, a naphthoic acid derivative. Here, we report the design, preparation, and activity of an agonist-based fluorescent probe MRS4183 (11) and a shorter P2Y14R agonist congener, which contain a UDP-glucuronic acid pharmacophore and BODIPY fluorophores conjugated through diaminoalkyl linkers. The design relied on both docking in a P2Y14R homology model and established structure activity relationship (SAR) of nucleotide analogs. 11 retained P2Y14R potency with EC50 value of 0.96 nM (inhibition of adenylyl cyclase), compared to parent UDPG (EC50 47 nM) and served as a tracer for microscopy and flow cytometry, displaying minimal nonspecific binding. Binding saturation analysis gave an apparent binding constant for 11 in whole cells of 21.4±1.1 nM, with a t1/2 of association at 50 nM 11 of 23.9 min. Known P2Y14R agonists and PPTN inhibited cell binding of 11 with the expected rank order of potency. The success in the identification of a new P2Y14R fluorescent agonist with low nonspecific binding illustrates the advantages of rational design based on recently determined GPCR X-ray structures. Such conjugates will be useful tools in expanding the SAR of this receptor, which still lacks chemical diversity in its collective ligands.


Subject(s)
Drug Design , Fluorescent Dyes/pharmacology , Heterocyclic Compounds, 3-Ring/pharmacology , Pyrans/pharmacology , Receptors, Purinergic P2/metabolism , Dose-Response Relationship, Drug , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/chemistry , Heterocyclic Compounds, 3-Ring/chemical synthesis , Heterocyclic Compounds, 3-Ring/chemistry , Humans , Models, Molecular , Molecular Structure , Pyrans/chemical synthesis , Pyrans/chemistry , Structure-Activity Relationship
9.
Mol Pharmacol ; 88(1): 139-40, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25943115

ABSTRACT

In this Perspective, former and current editors of Molecular Pharmacology, together with the guest editors for this 50th Anniversary Issue, provide a historical overview of the journal since its founding in 1965. The substantial impact that Molecular Pharmacology has had on the field of pharmacology as well as on biomedical science is discussed, as is the broad scope of the journal. The authors conclude that, true to the original goals for the journal, Molecular Pharmacology today remains an outstanding venue for work that provides a mechanistic understanding of drugs, molecular probes, and their biologic targets.


Subject(s)
Periodicals as Topic/trends , Pharmacogenetics/history , Animals , Drug Delivery Systems , History, 20th Century , Humans , Pharmaceutical Preparations/chemistry
10.
Mol Pharmacol ; 88(1): 151-60, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25829059

ABSTRACT

UDP-sugars, which are indispensable for protein glycosylation reactions in cellular secretory pathways, also act as important extracellular signaling molecules. We discuss here the broadly expressed P2Y14 receptor, a G-protein-coupled receptor targeted by UDP sugars, and the increasingly diverse set of physiologic responses discovered recently functioning downstream of this receptor in many epithelia as well as in immune, inflammatory, and other cells.


Subject(s)
Receptors, Purinergic P2Y/metabolism , Signal Transduction , Uridine Diphosphate Sugars/metabolism , Animals , Cell Differentiation , Humans , Immunity, Innate , Inflammation/metabolism
11.
ACS Chem Biol ; 9(12): 2833-42, 2014 Dec 19.
Article in English | MEDLINE | ID: mdl-25299434

ABSTRACT

The P2Y14 receptor (P2Y14R), one of eight P2Y G protein-coupled receptors (GPCR), is involved in inflammatory, endocrine, and hypoxic processes and is an attractive pharmaceutical target. The goal of this research is to develop high-affinity P2Y14R fluorescent probes based on the potent and highly selective antagonist 4-(4-(piperidin-4-yl)-phenyl)-7-(4-(trifluoromethyl)-phenyl)-2-naphthoic acid (6, PPTN). A model of hP2Y14R based on recent hP2Y12R X-ray structures together with simulated antagonist docking suggested that the piperidine ring is suitable for fluorophore conjugation while preserving affinity. Chain-elongated alkynyl or amino derivatives of 6 for click or amide coupling were synthesized, and their antagonist activities were measured in hP2Y14R-expressing CHO cells. Moreover, a new Alexa Fluor 488 (AF488) containing derivative 30 (MRS4174, Ki = 80 pM) exhibited exceptionally high affinity, as compared to 13 nM for the alkyne precursor 22. A flow cytometry assay employing 30 as a fluorescent probe was used to quantify specific binding to P2Y14R. Known P2Y receptor ligands inhibited binding of 30 with properties consistent with their previously established receptor selectivities and affinities. These results illustrate that potency in this series of 2-naphthoic acid derivatives can be preserved by chain functionalization, leading to highly potent fluorescent molecular probes for P2Y14R. Such conjugates will be useful tools in expanding the SAR of this receptor, which still lacks chemical diversity in its collective ligands. This approach demonstrates the predictive power of GPCR homology modeling and the relevance of newly determined X-ray structures to GPCR medicinal chemistry.


Subject(s)
Fluorescent Dyes/chemistry , Molecular Probes/chemistry , Naphthalenes/chemistry , Purinergic P2 Receptor Antagonists/chemistry , Receptors, Purinergic P2/chemistry , Animals , CHO Cells , Click Chemistry , Coumarins/chemistry , Cricetulus , Fluorescent Dyes/chemical synthesis , Humans , Ligands , Molecular Docking Simulation , Molecular Probes/chemical synthesis , Piperidines/chemistry , Purinergic P2 Receptor Antagonists/chemical synthesis , Rhodamines/chemistry , Structural Homology, Protein , Structure-Activity Relationship , Xanthenes/chemistry
12.
J Biol Chem ; 289(43): 29545-57, 2014 Oct 24.
Article in English | MEDLINE | ID: mdl-25193662

ABSTRACT

All peripheral membrane proteins must negotiate unique constraints intrinsic to the biological interface of lipid bilayers and the cytosol. Phospholipase C-ß (PLC-ß) isozymes hydrolyze the membrane lipid phosphatidylinositol 4,5-bisphosphate (PIP2) to propagate diverse intracellular responses that underlie the physiological action of many hormones, neurotransmitters, and growth factors. PLC-ß isozymes are autoinhibited, and several proteins, including Gαq, Gßγ, and Rac1, directly engage distinct regions of these phospholipases to release autoinhibition. To understand this process, we used a novel, soluble analog of PIP2 that increases in fluorescence upon cleavage to monitor phospholipase activity in real time in the absence of membranes or detergents. High concentrations of Gαq or Gß1γ2 did not activate purified PLC-ß3 under these conditions despite their robust capacity to activate PLC-ß3 at membranes. In addition, mutants of PLC-ß3 with crippled autoinhibition dramatically accelerated the hydrolysis of PIP2 in membranes without an equivalent acceleration in the hydrolysis of the soluble analog. Our results illustrate that membranes are integral for the activation of PLC-ß isozymes by diverse modulators, and we propose a model describing membrane-mediated allosterism within PLC-ß isozymes.


Subject(s)
Cell Membrane/enzymology , Phospholipase C beta/metabolism , Allosteric Regulation , Animals , Biocatalysis , COS Cells , Chlorocebus aethiops , Fluorescent Dyes/chemistry , Fluorescent Dyes/metabolism , Genes, Reporter , Heterotrimeric GTP-Binding Proteins/metabolism , Humans , Hydrolysis , Isoenzymes/chemistry , Isoenzymes/isolation & purification , Isoenzymes/metabolism , Models, Biological , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phospholipase C beta/chemistry , Phospholipase C beta/isolation & purification , Protein Structure, Secondary , Protein Structure, Tertiary , Solubility
13.
Chem Biol ; 21(7): 890-902, 2014 Jul 17.
Article in English | MEDLINE | ID: mdl-25036778

ABSTRACT

In spite of the crucial role of heterotrimeric G proteins as molecular switches transmitting signals from G protein-coupled receptors, their selective manipulation with small molecule, cell-permeable inhibitors still remains an unmet challenge. Here, we report that the small molecule BIM-46187, previously classified as pan-G protein inhibitor, preferentially silences Gαq signaling in a cellular context-dependent manner. Investigations into its mode of action reveal that BIM traps Gαq in the empty pocket conformation by permitting GDP exit but interdicting GTP entry, a molecular mechanism not yet assigned to any other small molecule Gα inhibitor to date. Our data show that Gα proteins may be "frozen" pharmacologically in an intermediate conformation along their activation pathway and propose a pharmacological strategy to specifically silence Gα subclasses with cell-permeable inhibitors.


Subject(s)
Cyclohexanes/metabolism , Cyclohexanes/pharmacology , GTP-Binding Protein alpha Subunits, Gq-G11/antagonists & inhibitors , GTP-Binding Protein alpha Subunits, Gq-G11/chemistry , Pyrazines/metabolism , Pyrazines/pharmacology , Animals , Cell Line , Cell Proliferation/drug effects , Cyclohexanes/chemistry , Dimerization , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Humans , Models, Molecular , Permeability , Protein Conformation/drug effects , Pyrazines/chemistry , Signal Transduction/drug effects
14.
J Med Chem ; 57(9): 3874-83, 2014 May 08.
Article in English | MEDLINE | ID: mdl-24712832

ABSTRACT

Extended N(4)-(3-arylpropyl)oxy derivatives of uridine-5'-triphosphate were synthesized and potently stimulated phospholipase C stimulation in astrocytoma cells expressing G protein-coupled human (h) P2Y receptors (P2YRs) activated by UTP (P2Y2/4R) or UDP (P2Y6R). The potent P2Y4R-selective N(4)-(3-phenylpropyl)oxy agonist was phenyl ring-substituted or replaced with terminal heterocyclic or naphthyl rings with retention of P2YR potency. This broad tolerance for steric bulk in a distal region was not observed for dinucleoside tetraphosphate agonists with both nucleobases substituted. The potent N(4)-(3-(4-methoxyphenyl)-propyl)oxy analogue 19 (EC50: P2Y2R, 47 nM; P2Y4R, 23 nM) was functionalized for chain extension using click tethering of fluorophores as prosthetic groups. The BODIPY 630/650 conjugate 28 (MRS4162) exhibited EC50 values of 70, 66, and 23 nM at the hP2Y2/4/6Rs, respectively, and specifically labeled cells expressing the P2Y6R. Thus, an extended N(4)-(3-arylpropyl)oxy group accessed a structurally permissive region on three Gq-coupled P2YRs, and potency and selectivity were modulated by distal structural changes. This freedom of substitution was utilized to design of a pan-agonist fluorescent probe of a subset of uracil nucleotide-activated hP2YRs.


Subject(s)
Imines/chemistry , Molecular Probes , Purinergic P2 Receptor Agonists/chemistry , Receptors, Purinergic P2Y2/drug effects , Uridine Triphosphate/chemistry , Fluorescent Dyes/chemistry , Humans , Microscopy, Fluorescence , Purinergic P2 Receptor Agonists/pharmacology , Receptors, Purinergic P2Y2/chemistry , Receptors, Purinergic P2Y2/classification
15.
Molecules ; 19(4): 4313-25, 2014 Apr 04.
Article in English | MEDLINE | ID: mdl-24714193

ABSTRACT

In this study we report the synthesis of C5/C6-fused uridine phosphonates that are structurally related to earlier reported allosteric P2Y2 receptor ligands. A silyl-Hilbert-Johnson reaction of six quinazoline-2,4-(1H,3H)-dione-like base moieties with a suitable ribofuranosephosphonate afforded the desired analogues after full deprotection. In contrast to the parent 5-(4-fluoropheny)uridine phosphonate, the present extended-base uridine phosphonates essentially failed to modulate the P2Y2 receptor.


Subject(s)
Organophosphonates/chemical synthesis , Purinergic P2Y Receptor Agonists/chemical synthesis , Receptors, Purinergic P2Y2/metabolism , Uridine/chemical synthesis , Allosteric Regulation , Astrocytes/cytology , Astrocytes/drug effects , Astrocytes/metabolism , Cell Line, Tumor , Humans , Ligands , Organophosphonates/pharmacology , Purinergic P2Y Receptor Agonists/pharmacology , Quinazolines/chemistry , Uridine/analogs & derivatives , Uridine/pharmacology
16.
Sci Signal ; 6(298): pe34, 2013 Oct 22.
Article in English | MEDLINE | ID: mdl-24150253

ABSTRACT

GPR17 is an orphan G protein-coupled receptor involved in orchestration of oligodendrocyte differentiation and myelination in the central nervous system. In this issue of Science Signaling, Hennen et al. used a signaling pathway-unbiased screen to identify two small molecule activators of this receptor. One of these, MDL29951, was carried forward to illustrate GPR17-dependent activation of Gαi- and Gαq-promoted signaling pathways in cell lines expressing recombinant GPR17, whereas no effect was observed with previously proposed but dubitable agonists (uracil nucleotides and cysteinyl leukotrienes) of this receptor. Conversely, MDL29951 did not activate any of the known uracil or adenine nucleotide-activated P2Y receptors or cysteinyl leukotriene receptors. Gαi- and Gαq-dependent signaling responses also were observed in primary rat oligodendrocytes in the presence of MDL29951. Moreover, MDL29951 diminished myelination in primary oligodendrocytes isolated from heterozygous mice but had no effect on myelination in oligodendrocytes from GPR17 knockout mice. Effects of a small-molecule GPR17 agonist observed during oligodendrocyte differentiation support the idea that development of antagonists of GPR17 is a rational goal for elaboration of pharmacotherapies in demyelinating diseases.


Subject(s)
Receptors, G-Protein-Coupled/agonists , Signal Transduction/drug effects , Small Molecule Libraries/pharmacology , Animals , Humans
17.
J Pharmacol Exp Ther ; 347(1): 38-46, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23908386

ABSTRACT

The orphan receptor GPR17 has been reported to be activated by UDP, UDP-sugars, and cysteinyl leukotrienes, and coupled to intracellular Ca(2+) mobilization and inhibition of cAMP accumulation, but other studies have reported either a different agonist profile or lack of agonist activity altogether. To determine if GPR17 is activated by uracil nucleotides and leukotrienes, the hemagglutinin-tagged receptor was expressed in five different cell lines and the signaling properties of the receptor were investigated. In C6, 1321N1, or Chinese hamster ovary (CHO) cells stably expressing GPR17, UDP, UDP-glucose, UDP-galactose, and cysteinyl leukotriene C4 (LTC4) all failed to promote inhibition of forskolin-stimulated cAMP accumulation, whereas both UDP and UDP-glucose promoted marked inhibition (>80%) of forskolin-stimulated cAMP accumulation in C6 and CHO cells expressing the P2Y14 receptor. Likewise, none of these compounds promoted accumulation of inositol phosphates in COS-7 or human embryonic kidney 293 cells transiently transfected with GPR17 alone or cotransfected with Gαq/i5, which links Gi-coupled receptors to the Gq-regulated phospholipase C (PLC) signaling pathway, or PLCε, which is activated by the Gα12/13 signaling pathway. Moreover, none of these compounds promoted internalization of GPR17 in 1321N1-GPR17 cells. Consistent with previous reports, coexpression experiments of GPR17 with cysteinyl leukotriene receptor 1 (CysLTR1) suggested that GPR17 acts as a negative regulator of CysLTR1. Taken together, these data suggest that UDP, UDP-glucose, UDP-galactose, and LTC4 are not the cognate ligands of GPR17.


Subject(s)
Cysteine/metabolism , Leukotrienes/metabolism , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/metabolism , Receptors, Purinergic P2Y/metabolism , Uracil Nucleotides/metabolism , Animals , CHO Cells , COS Cells , Chlorocebus aethiops , Cricetinae , Cricetulus , Cysteine/pharmacology , HEK293 Cells , Humans , Leukotrienes/pharmacology , Uracil Nucleotides/pharmacology , Uridine Diphosphate Glucose/metabolism , Uridine Diphosphate Glucose/pharmacology
18.
Biochemistry ; 52(28): 4810-9, 2013 Jul 16.
Article in English | MEDLINE | ID: mdl-23777354

ABSTRACT

Multiple extracellular stimuli, such as growth factors and antigens, initiate signaling cascades through tyrosine phosphorylation and activation of phospholipase C-γ (PLC-γ) isozymes. Like most other PLCs, PLC-γ1 is basally autoinhibited by its X-Y linker, which separates the X- and Y-boxes of the catalytic core. The C-terminal SH2 (cSH2) domain within the X-Y linker is the critical determinant for autoinhibition of phospholipase activity. Release of autoinhibition requires an intramolecular interaction between the cSH2 domain and a phosphorylated tyrosine, Tyr783, also located within the X-Y linker. The molecular mechanisms that mediate autoinhibition and phosphorylation-induced activation have not been defined. Here, we describe structures of the cSH2 domain both alone and bound to a PLC-γ1 peptide encompassing phosphorylated Tyr783. The cSH2 domain remains largely unaltered by peptide engagement. Point mutations in the cSH2 domain located at the interface with the peptide were sufficient to constitutively activate PLC-γ1, suggesting that peptide engagement directly interferes with the capacity of the cSH2 domain to block the lipase active site. This idea is supported by mutations in a complementary surface of the catalytic core that also enhanced phospholipase activity.


Subject(s)
Isoenzymes/metabolism , Phospholipase C gamma/metabolism , Amino Acid Sequence , Crystallography, X-Ray , Enzyme Activation , Isoenzymes/antagonists & inhibitors , Isoenzymes/chemistry , Models, Molecular , Molecular Sequence Data , Phospholipase C gamma/antagonists & inhibitors , Phospholipase C gamma/chemistry , Phosphorylation , Sequence Homology, Amino Acid , src Homology Domains
19.
Mol Pharmacol ; 84(1): 41-9, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23592514

ABSTRACT

The nucleotide-sugar-activated P2Y14 receptor (P2Y14-R) is highly expressed in hematopoietic cells. Although the physiologic functions of this receptor remain undefined, it has been strongly implicated recently in immune and inflammatory responses. Lack of availability of receptor-selective high-affinity antagonists has impeded progress in studies of this and most of the eight nucleotide-activated P2Y receptors. A series of molecules recently were identified by Gauthier et al. (Gauthier et al., 2011) that exhibited antagonist activity at the P2Y14-R. We synthesized one of these molecules, a 4,7-disubstituted 2-naphthoic acid derivative (PPTN), and studied its pharmacological properties in detail. The concentration-effect curve of UDP-glucose for promoting inhibition of adenylyl cyclase in C6 glioma cells stably expressing the P2Y14-R was shifted to the right in a concentration-dependent manner by PPTN. Schild analyses revealed that PPTN-mediated inhibition followed competitive kinetics, with a KB of 434 pM observed. In contrast, 1 µM PPTN exhibited no agonist or antagonist effect at the P2Y1, P2Y2, P2Y4, P2Y6, P2Y11, P2Y12, or P2Y13 receptors. UDP-glucose-promoted chemotaxis of differentiated HL-60 human promyelocytic leukemia cells was blocked by PPTN with a concentration dependence consistent with the KB determined with recombinant P2Y14-R. In contrast, the chemotactic response evoked by the chemoattractant peptide fMetLeuPhe was unaffected by PPTN. UDP-glucose-promoted chemotaxis of freshly isolated human neutrophils also was blocked by PPTN. In summary, this work establishes PPTN as a highly selective high-affinity antagonist of the P2Y14-R that is useful for interrogating the action of this receptor in physiologic systems.


Subject(s)
Chemotaxis/drug effects , Neutrophils/drug effects , Purinergic P2 Receptor Antagonists/pharmacology , Receptors, Purinergic P2/metabolism , Uridine Diphosphate Glucose/metabolism , Adenylyl Cyclase Inhibitors , Adenylyl Cyclases/metabolism , Animals , CHO Cells , Cell Line, Tumor , Cricetinae , Glioma/metabolism , HL-60 Cells , Humans , Leukemia, Promyelocytic, Acute/metabolism , Neutrophils/metabolism , Purinergic P2 Receptor Agonists/pharmacology , Purinergic P2 Receptor Antagonists/chemical synthesis , Rats
20.
J Biol Chem ; 288(8): 5840-8, 2013 Feb 22.
Article in English | MEDLINE | ID: mdl-23297405

ABSTRACT

Phospholipase C (PLC) isozymes are important signaling molecules, but few small molecule modulators are available to pharmacologically regulate their function. With the goal of developing a general approach for identification of novel PLC inhibitors, we developed a high-throughput assay based on the fluorogenic substrate reporter WH-15. The assay is highly sensitive and reproducible: screening a chemical library of 6280 compounds identified three novel PLC inhibitors that exhibited potent activities in two separate assay formats with purified PLC isozymes in vitro. Two of the three inhibitors also inhibited G protein-coupled receptor-stimulated PLC activity in intact cell systems. These results demonstrate the power of the high-throughput assay for screening large collections of small molecules to identify novel PLC modulators. Potent and selective modulators of PLCs will ultimately be useful for dissecting the roles of PLCs in cellular processes, as well as provide lead compounds for the development of drugs to treat diseases arising from aberrant phospholipase activity.


Subject(s)
Enzyme Inhibitors/pharmacology , Type C Phospholipases/antagonists & inhibitors , Biological Assay/methods , Chemistry, Pharmaceutical/methods , Cyclic AMP/metabolism , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , HEK293 Cells , Humans , Isoenzymes , Models, Biological , Models, Chemical , Phospholipases/chemistry , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Small Molecule Libraries , Substrate Specificity , Type C Phospholipases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...