Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Br J Pharmacol ; 181(1): 21-35, 2024 01.
Article in English | MEDLINE | ID: mdl-37530222

ABSTRACT

BACKGROUND AND PURPOSE: Ticagrelor is labelled as a reversible, direct-acting platelet P2Y12 receptor (P2Y12 R) antagonist that is indicated clinically for the prevention of thrombotic events in patients with acute coronary syndrome (ACS). As with many antiplatelet drugs, ticagrelor therapy increases bleeding risk in patients, which may require platelet transfusion in emergency situations. The aim of this study was to further examine the reversibility of ticagrelor at the P2Y12 R. EXPERIMENTAL APPROACH: Studies were performed in human platelets, with P2Y12 R-stimulated GTPase activity and platelet aggregation assessed. Cell-based bioluminescence resonance energy transfer (BRET) assays were undertaken to assess G protein-subunit activation downstream of P2Y12 R activation. KEY RESULTS: Initial studies revealed that a range of P2Y12 R ligands, including ticagrelor, displayed inverse agonist activity at P2Y12 R. Only ticagrelor was resistant to washout and, in human platelet and cell-based assays, washing failed to reverse ticagrelor-dependent inhibition of ADP-stimulated P2Y12 R function. The P2Y12 R agonist 2MeSADP, which was also resistant to washout, was able to effectively compete with ticagrelor. In silico docking revealed that ticagrelor and 2MeSADP penetrated more deeply into the orthosteric binding pocket of the P2Y12 R than other P2Y12 R ligands. CONCLUSION AND IMPLICATIONS: Ticagrelor binding to P2Y12 R is prolonged and more akin to that of an irreversible antagonist, especially versus the endogenous P2Y12 R agonist ADP. This study highlights the potential clinical need for novel ticagrelor reversal strategies in patients with spontaneous major bleeding, and for bleeding associated with urgent invasive procedures.


Subject(s)
Acute Coronary Syndrome , Diphosphates , Humans , Ticagrelor/pharmacology , Ticagrelor/metabolism , Ticagrelor/therapeutic use , Diphosphates/metabolism , Diphosphates/pharmacology , Diphosphates/therapeutic use , Adenosine/pharmacology , Drug Inverse Agonism , Purinergic P2Y Receptor Antagonists/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Adenosine Diphosphate/pharmacology , Adenosine Diphosphate/metabolism , Blood Platelets , Acute Coronary Syndrome/drug therapy , Acute Coronary Syndrome/complications , Receptors, Purinergic P2Y12/metabolism
2.
J Thromb Haemost ; 21(5): 1307-1321, 2023 05.
Article in English | MEDLINE | ID: mdl-36716966

ABSTRACT

BACKGROUND: Patients with COVID-19 are at increased risk of thrombosis, which is associated with altered platelet function and coagulopathy, contributing to excess mortality. OBJECTIVES: To characterize the mechanism of altered platelet function in COVID-19 patients. METHODS: The platelet proteome, platelet functional responses, and platelet-neutrophil aggregates were compared between patients hospitalized with COVID-19 and healthy control subjects using tandem mass tag proteomic analysis, Western blotting, and flow cytometry. RESULTS: COVID-19 patients showed a different profile of platelet protein expression (858 altered of the 5773 quantified). Levels of COVID-19 plasma markers were enhanced in the platelets of COVID-19 patients. Gene ontology pathway analysis demonstrated that the levels of granule secretory proteins were raised, whereas those of platelet activation proteins, such as the thrombopoietin receptor and protein kinase Cα, were lowered. Basally, platelets of COVID-19 patients showed enhanced phosphatidylserine exposure, with unaltered integrin αIIbß3 activation and P-selectin expression. Agonist-stimulated integrin αIIbß3 activation and phosphatidylserine exposure, but not P-selectin expression, were decreased in COVID-19 patients. COVID-19 patients had high levels of platelet-neutrophil aggregates, even under basal conditions, compared to controls. This association was disrupted by blocking P-selectin, demonstrating that platelet P-selectin is critical for the interaction. CONCLUSIONS: Overall, our data suggest the presence of 2 platelet populations in patients with COVID-19: one of circulating platelets with an altered proteome and reduced functional responses and another of P-selectin-expressing neutrophil-associated platelets. Platelet-driven thromboinflammation may therefore be one of the key factors enhancing the risk of thrombosis in COVID-19 patients.


Subject(s)
COVID-19 , Thrombosis , Humans , Proteome/metabolism , COVID-19/complications , Proteomics , Phosphatidylserines/metabolism , Inflammation/metabolism , Thrombosis/etiology , Blood Platelets/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Platelet Activation , Selectins/metabolism
3.
Blood Adv ; 5(7): 1884-1898, 2021 04 13.
Article in English | MEDLINE | ID: mdl-33792632

ABSTRACT

The reactivity of platelets, which play a key role in the pathogenesis of atherothrombosis, is tightly regulated. The integral membrane protein tetherin/bone marrow stromal antigen-2 (BST-2) regulates membrane organization, altering both lipid and protein distribution within the plasma membrane. Because membrane microdomains have an established role in platelet receptor biology, we sought to characterize the physiological relevance of tetherin/BST-2 in those cells. To characterize the potential importance of tetherin/BST-2 to platelet function, we used tetherin/BST-2-/- murine platelets. In the mice, we found enhanced function and signaling downstream of a subset of membrane microdomain-expressing receptors, including the P2Y12, TP thromboxane, thrombin, and GPVI receptors. Preliminary studies in humans have revealed that treatment with interferon-α (IFN-α), which upregulates platelet tetherin/BST-2 expression, also reduces adenosine diphosphate-stimulated platelet receptor function and reactivity. A more comprehensive understanding of how tetherin/BST-2 negatively regulates receptor function was provided in cell line experiments, where we focused on the therapeutically relevant P2Y12 receptor (P2Y12R). Tetherin/BST-2 expression reduced both P2Y12R activation and trafficking, which was accompanied by reduced receptor lateral mobility specifically within membrane microdomains. In fluorescence lifetime imaging-Förster resonance energy transfer (FLIM-FRET)-based experiments, agonist stimulation reduced basal association between P2Y12R and tetherin/BST-2. Notably, the glycosylphosphatidylinositol (GPI) anchor of tetherin/BST-2 was required for both receptor interaction and observed functional effects. In summary, we established, for the first time, a fundamental role of the ubiquitously expressed protein tetherin/BST-2 in negatively regulating membrane microdomain-expressed platelet receptor function.


Subject(s)
Antigens, CD , Bone Marrow Stromal Antigen 2 , Animals , Antigens, CD/genetics , Blood Platelets , Cell Line , GPI-Linked Proteins/genetics , Mice
5.
Haematologica ; 106(7): 1968-1978, 2021 07 01.
Article in English | MEDLINE | ID: mdl-32467143

ABSTRACT

Pim kinases are upregulated in several forms of cancer, contributing to cell survival and tumour development, but their role in platelet function and thrombotic disease has not been explored. We report for the first time that Pim-1 is expressed in human and mouse platelets. Genetic deletion or pharmacological inhibition of Pim kinase results in reduced thrombus formation but is not associated with impaired haemostasis. Attenuation of thrombus formation was found to be due to inhibition of the thromboxane A2 receptor as effects on platelet function was non-additive to inhibition caused by the cyclooxygenase inhibitor indomethacin or thromboxane A2 receptor antagonist GR32191. Treatment with Pim kinase inhibitors caused reduced surface expression of the thromboxane A2 receptor and resulted in reduced responses to thromboxane A2 receptor agonists, indicating a role for Pim kinase in the regulation of thromboxane A2 receptor function. Our research identifies a novel, Pim kinase dependent regulatory mechanism for the thromboxane A2 receptor and represents a new targeting strategy that is independent of COX-1 inhibition or direct antagonism of the thromboxane A2 receptor that whilst attenuating thrombosis does not increase bleeding.


Subject(s)
Receptors, Thromboxane A2, Prostaglandin H2 , Thrombosis , Blood Platelets , Humans , Platelet Aggregation , Proto-Oncogene Proteins c-pim-1/genetics , Receptors, Thromboxane A2, Prostaglandin H2/genetics , Thrombosis/drug therapy
6.
J Am Heart Assoc ; 9(24): e016495, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33305660

ABSTRACT

Background To assess differences in platelet inhibition during ticagrelor monotherapy (TIC) or dual therapy with ticagrelor and aspirin (TIC+ASP) in patients after percutaneous coronary intervention using a comprehensive panel of functional tests. Methods and Results In a single-center parallel group, open label, randomized controlled trial, 110 participants were randomized to receive either TIC (n=55) or TIC+ASP (n=55) for 4 weeks. The primary outcome was the platelet aggregation response with 10 µmol/L thrombin receptor activation peptide-6 (TRAP-6). The secondary outcomes were platelet aggregation responses and binding of surface activation markers with a panel of other activators. The mean percentage aggregation for 10 µmol/L TRAP-6 was similar for the TIC and TIC+ASP groups (mean difference+4.29; 95% CI, -0.87 to +9.46). Aggregation was higher in the TIC group compared with the TIC+ASP group with 1 µg/mL (+6.47; +2.04 to +10.90) and 0.5 µg/mL (+14.00; +7.63 to +20.39) collagen related peptide. Aggregation responses with 5 µmol/L TRAP-6, 5 µmol/L or 2.5 µmol/L thromboxane A2 receptor agonist and surface activation marker binding with 5 µmol/L TRAP-6 or 0.5 µg/mL collagen related peptide were the same between the treatment groups. Conclusions Patients with PCI show similar levels of inhibition of most platelet activation pathways with TIC compared with dual therapy with TIC + ASP. However, the greater aggregation response with collagen related peptide during TIC indicates incomplete inhibition of glycoprotein VI (collagen) receptor-mediated platelet activation. This difference in pharmacodynamic response to anti-platelet medication may contribute to the lower bleeding rates observed with TIC compared with dual antiplatelet therapy in recent clinical trials. Registration Information URL: https://www.isrctn.com; Unique Identifier ISRCTN84335288.


Subject(s)
Acute Coronary Syndrome/drug therapy , Drug Therapy, Combination/adverse effects , Percutaneous Coronary Intervention/adverse effects , Purinergic P2Y Receptor Antagonists/pharmacology , Ticagrelor/pharmacology , Acute Coronary Syndrome/blood , Aged , Arachidonic Acid/blood , Aspirin/therapeutic use , Drug Therapy, Combination/methods , Dual Anti-Platelet Therapy/adverse effects , Dual Anti-Platelet Therapy/methods , Female , Humans , Male , Middle Aged , Peptide Fragments/drug effects , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/therapeutic use , Platelet Function Tests/methods , Purinergic P2Y Receptor Antagonists/administration & dosage , Purinergic P2Y Receptor Antagonists/therapeutic use , Receptors, Thromboxane A2, Prostaglandin H2/agonists , Ticagrelor/administration & dosage , Ticagrelor/therapeutic use
7.
Platelets ; 31(5): 641-645, 2020 Jul 03.
Article in English | MEDLINE | ID: mdl-31684789

ABSTRACT

The principal demonstrated role of the nonvisual arrestins in vivo is to limit G protein-coupled receptor (GPCR) signaling. Nonetheless, a direct demonstration of this fundamental ability in platelets remains lacking, despite the prominent role played by GPCRs in platelet activation. This paper describes the basic characterization of the activatory responses of platelets from mice lacking arrestin-3 (arr3-/-), revealing pleiotropic roles dependent on GPCR ligand. Functionally, arrestin-3 acts as a brake on platelet aggregation regardless of ligand tested. Downstream of P2Y receptors, arr3-/- mice show increased secretion and integrin activation mirrored by enhanced intracellular calcium signaling and global PKC-dependent phosphorylation. Furthermore, P2Y12 receptor (P2Y12R) activity as assessed by ADP-mediated reduction of VASP phosphorylation is enhanced in arr3-/-mice. Downstream of PAR receptors there are similar increases in secretion and integrin activation in arr3-/- mice, together with enhanced PKC activity. Last, in arr3-/- mice the TP receptor displays unaltered PKC activity but markedly reduced calcium responses, which together with the kinetics of the aggregation response suggested a unique positive regulatory role for arrestin-3 in TP signaling. Overall, this paper reveals pleiotropic roles for arrestin-3 dependent on GPCR ligand describing for the first time a negative regulatory function for arrestin-3 in platelets.


Subject(s)
Arrestins/metabolism , Blood Platelets/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Blood Platelets/cytology , Humans , Mice
8.
Mol Cell Endocrinol ; 449: 74-81, 2017 07 05.
Article in English | MEDLINE | ID: mdl-28212842

ABSTRACT

Genetic variations in G protein-coupled receptor (GPCR) genes can disrupt receptor function in a wide variety of human genetic diseases, including platelet bleeding disorders. Platelets are critical for haemostasis with inappropriate platelet activation leading to the development of arterial thrombosis, which can result in heart attack and stroke whilst decreased platelet activity is associated with an increased risk of bleeding. GPCRs expressed on the surface of platelets play key roles in regulating platelet activity and therefore function. Receptors include purinergic receptors (P2Y1 and P2Y12), proteinase-activated receptor (PAR1 and PAR4) and thromboxane receptors (TPα), among others. Pharmacological blockade of these receptors forms a powerful therapeutic tool in the treatment and prevention of arterial thrombosis. With the advance of genomic technologies, there has been a substantial increase in the identification of naturally occurring rare and common GPCR variants. These variants include single-nucleotide polymorphisms (SNPs) and insertion or deletions that have the potential to alter GPCR expression or function. A number of defects in platelet GPCRs that disrupt receptor function have now been characterized in patients with mild bleeding disorders. This review will focus on rare, function-disrupting variants of platelet GPCRs with particular emphasis upon mutations in the P2Y12 receptor gene that affect receptor traffic to modulate platelet function. Further this review will outline how the identification and characterization of function-disrupting GPCR mutations provides an essential link in translating our detailed understanding of receptor traffic and function in cell line studies into relevant human biological systems.


Subject(s)
Blood Platelets/metabolism , Receptors, Purinergic P2Y12/metabolism , Animals , Disease , Genetic Variation , Humans , Models, Biological , Platelet Activation , Receptors, Purinergic P2Y12/chemistry , Receptors, Purinergic P2Y12/genetics
9.
Biochem Pharmacol ; 124: 43-56, 2017 Jan 15.
Article in English | MEDLINE | ID: mdl-27845050

ABSTRACT

Thromboxane A2 is a potent mediator of inflammation and platelet aggregation exerting its effects through the activation of a G protein-coupled receptor (GPCR), termed TP. Although the existence of dimers/oligomers in Class A GPCRs is widely accepted, their functional significance still remains controversial. Recently, we have shown that TPα and TPß homo-/hetero-dimers interact through an interface of residues in transmembrane domain 1 (TM1) whose disruption impairs dimer formation. Here, biochemical and pharmacological characterization of this dimer deficient mutant (DDM) in living cells indicates a significant impairment in its response to agonists. Interestingly, two single loss-of-function TPα variants, namely W29C and N42S recently identified in two heterozygous patients affected by bleeding disorders, match some of the residues mutated in our DDM. These two naturally occurring variants display a reduced potency to TP agonists and are characterized by impaired dimer formation in transfected HEK-293T cells. These findings provide proofs that lack of homo-dimer formation is a crucial process for reduced TPα function in vivo, and might represent one molecular mechanism through which platelet TPα receptor dysfunction affects the patient(s) carrying these mutations.


Subject(s)
Blood Platelets/physiology , Receptors, Thromboxane/metabolism , Signal Transduction , Dimerization , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Ligands , Mutation , Receptors, Thromboxane/agonists , Receptors, Thromboxane/antagonists & inhibitors , Receptors, Thromboxane/genetics
10.
Blood ; 128(23): 2717-2728, 2016 12 08.
Article in English | MEDLINE | ID: mdl-27694321

ABSTRACT

Ticagrelor is a potent antagonist of the P2Y12 receptor (P2Y12R) and consequently an inhibitor of platelet activity effective in the treatment of atherothrombosis. Here, we sought to further characterize its molecular mechanism of action. Initial studies showed that ticagrelor promoted a greater inhibition of adenosine 5'-diphosphate (ADP)-induced Ca2+ release in washed platelets vs other P2Y12R antagonists. This additional effect of ticagrelor beyond P2Y12R antagonism was in part as a consequence of ticagrelor inhibiting the equilibrative nucleoside transporter 1 (ENT1) on platelets, leading to accumulation of extracellular adenosine and activation of Gs-coupled adenosine A2A receptors. This contributed to an increase in basal cyclic adenosine monophosphate (cAMP) and vasodilator-stimulated phosphoprotein phosphorylation (VASP-P). In addition, ticagrelor increased platelet cAMP and VASP-P in the absence of ADP in an adenosine receptor-independent manner. We hypothesized that this increase originated from a direct effect on basal agonist-independent P2Y12R signaling, and this was validated in 1321N1 cells stably transfected with human P2Y12R. In these cells, ticagrelor blocked the constitutive agonist-independent activity of the P2Y12R, limiting basal Gi-coupled signaling and thereby increasing cAMP levels. These data suggest that ticagrelor has the pharmacological profile of an inverse agonist. Based on our results showing insurmountable inhibition of ADP-induced Ca2+ release and forskolin-induced cAMP, the mode of antagonism of ticagrelor also appears noncompetitive, at least functionally. In summary, our studies describe 2 novel modes of action of ticagrelor, inhibition of platelet ENT1 and inverse agonism at the P2Y12R that contribute to its effective inhibition of platelet activation.


Subject(s)
Adenosine/analogs & derivatives , Blood Platelets/metabolism , Equilibrative Nucleoside Transporter 1/antagonists & inhibitors , Platelet Activation/drug effects , Receptors, Purinergic P2Y12/metabolism , Adenosine/pharmacology , Adenosine Diphosphate/pharmacology , Blood Platelets/cytology , Calcium/metabolism , Calcium Signaling/drug effects , Cell Adhesion Molecules/metabolism , Colforsin/pharmacology , Cyclic AMP/metabolism , Equilibrative Nucleoside Transporter 1/metabolism , Female , Humans , Male , Microfilament Proteins/metabolism , Phosphoproteins/metabolism , Phosphorylation/drug effects , Ticagrelor
11.
PLoS One ; 11(10): e0163006, 2016.
Article in English | MEDLINE | ID: mdl-27716777

ABSTRACT

Thioredoxin (Trx) is an oxidoreductase with important physiological function. Imbalances in the NADPH/thioredoxin reductase/thioredoxin system are associated with a number of pathologies, particularly cancer, and a number of clinical trials for thioredoxin and thioredoxin reductase inhibitors have been carried out or are underway. Due to the emerging role and importance of oxidoreductases for haemostasis and the current interest in developing inhibitors for clinical use, we thought it pertinent to assess whether inhibition of the NADPH/thioredoxin reductase/thioredoxin system affects platelet function and thrombosis. We used small molecule inhibitors of Trx (PMX 464 and PX-12) to determine whether Trx activity influences platelet function, as well as an unbiased proteomics approach to identify potential Trx substrates on the surface of platelets that might contribute to platelet reactivity and function. Using LC-MS/MS we found that PMX 464 and PX-12 affected the oxidation state of thiols in a number of cell surface proteins. Key surface receptors for platelet adhesion and activation were affected, including the collagen receptor GPVI and the von Willebrand factor receptor, GPIb. To experimentally validate these findings we assessed platelet function in the presence of PMX 464, PX-12, and rutin (a selective inhibitor of the related protein disulphide isomerase). In agreement with the proteomics data, small molecule inhibitors of thioredoxin selectively inhibited GPVI-mediated platelet activation, and attenuated ristocetin-induced GPIb-vWF-mediated platelet agglutination, thus validating the findings of the proteomics study. These data reveal a novel role for thioredoxin in regulating platelet reactivity via proteins required for early platelet responses at sites of vessel injury (GPVI and GPIb). This work also highlights a potential opportunity for repurposing of PMX 464 and PX-12 as antiplatelet agents.


Subject(s)
Blood Platelets/drug effects , Platelet Aggregation Inhibitors/pharmacology , Thioredoxins/pharmacology , Thrombosis/drug therapy , Benzothiazoles/pharmacology , Blood Coagulation Tests/methods , Blood Platelets/metabolism , Disulfides/pharmacology , Humans , Hydroquinones/pharmacology , Imidazoles/pharmacology , Platelet Activation/drug effects , Platelet Adhesiveness/drug effects , Platelet Aggregation/drug effects , Platelet Function Tests/methods , Platelet Membrane Glycoproteins/metabolism , Receptors, Collagen/metabolism , Ristocetin/pharmacology , Thrombosis/metabolism , von Willebrand Factor/metabolism
12.
Arterioscler Thromb Vasc Biol ; 36(5): 952-60, 2016 05.
Article in English | MEDLINE | ID: mdl-26966273

ABSTRACT

OBJECTIVE: Protease-activated receptor 4 (PAR4) is a key regulator of platelet reactivity and is encoded by F2RL3, which has abundant rare missense variants. We aimed to provide proof of principle that rare F2LR3 variants potentially affect platelet reactivity and responsiveness to PAR1 antagonist drugs and to explore underlying molecular mechanisms. APPROACH AND RESULTS: We identified 6 rare F2RL3 missense variants in 236 cardiac patients, of which the variant causing a tyrosine 157 to cysteine substitution (Y157C) was predicted computationally to have the greatest effect on PAR4 structure. Y157C platelets from 3 cases showed reduced responses to PAR4-activating peptide and to α-thrombin compared with controls, but no reduction in responses to PAR1-activating peptide. Pretreatment with the PAR1 antagonist vorapaxar caused lower residual α-thrombin responses in Y157C platelets than in controls, indicating greater platelet inhibition. HEK293 cells transfected with a PAR4 Y157C expression construct had reduced PAR4 functional responses, unchanged total PAR4 expression but reduced surface expression. PAR4 Y157C was partially retained in the endoplasmic reticulum and displayed an expression pattern consistent with defective N-glycosylation. Mutagenesis of Y322, which is the putative hydrogen bond partner of Y157, also reduced PAR4 surface expression in HEK293 cells. CONCLUSIONS: Reduced PAR4 responses associated with Y157C result from aberrant anterograde surface receptor trafficking, in part, because of disrupted intramolecular hydrogen bonding. Characterization of PAR4 Y157C establishes that rare F2RL3 variants have the potential to markedly alter platelet PAR4 reactivity particularly after exposure to therapeutic PAR1 antagonists.


Subject(s)
Blood Platelets/metabolism , Platelet Activation , Receptors, Thrombin/metabolism , Aged , Blood Platelets/drug effects , Case-Control Studies , Computer Simulation , Dose-Response Relationship, Drug , Endoplasmic Reticulum/metabolism , England , Female , Genotype , Glycosylation , HEK293 Cells , Humans , Hydrogen Bonding , Lactones/pharmacology , Male , Models, Molecular , Mutation, Missense , Peptides/pharmacology , Phenotype , Platelet Activation/drug effects , Platelet Aggregation , Platelet Aggregation Inhibitors/pharmacology , Polymorphism, Single Nucleotide , Protein Conformation , Protein Transport , Pyridines/pharmacology , Receptor, PAR-1/drug effects , Receptor, PAR-1/metabolism , Receptors, Thrombin/chemistry , Receptors, Thrombin/drug effects , Receptors, Thrombin/genetics , Structure-Activity Relationship , Thrombin/pharmacology , Transfection
13.
PLoS One ; 10(12): e0143913, 2015.
Article in English | MEDLINE | ID: mdl-26630678

ABSTRACT

The clinical expression of type 1 von Willebrand disease may be modified by co-inheritance of other mild bleeding diatheses. We previously showed that mutations in the platelet P2Y12 ADP receptor gene (P2RY12) could contribute to the bleeding phenotype in patients with type 1 von Willebrand disease. Here we investigated whether variations in platelet G protein-coupled receptor genes other than P2RY12 also contributed to the bleeding phenotype. Platelet G protein-coupled receptor genes P2RY1, F2R, F2RL3, TBXA2R and PTGIR were sequenced in 146 index cases with type 1 von Willebrand disease and the potential effects of identified single nucleotide variations were assessed using in silico methods and heterologous expression analysis. Seven heterozygous single nucleotide variations were identified in 8 index cases. Two single nucleotide variations were detected in F2R; a novel c.-67G>C transversion which reduced F2R transcriptional activity and a rare c.1063C>T transition predicting a p.L355F substitution which did not interfere with PAR1 expression or signalling. Two synonymous single nucleotide variations were identified in F2RL3 (c.402C>G, p.A134 =; c.1029 G>C p.V343 =), both of which introduced less commonly used codons and were predicted to be deleterious, though neither of them affected PAR4 receptor expression. A third single nucleotide variation in F2RL3 (c.65 C>A; p.T22N) was co-inherited with a synonymous single nucleotide variation in TBXA2R (c.6680 C>T, p.S218 =). Expression and signalling of the p.T22N PAR4 variant was similar to wild-type, while the TBXA2R variation introduced a cryptic splice site that was predicted to cause premature termination of protein translation. The enrichment of single nucleotide variations in G protein-coupled receptor genes among type 1 von Willebrand disease patients supports the view of type 1 von Willebrand disease as a polygenic disorder.


Subject(s)
Blood Platelets/metabolism , Receptors, G-Protein-Coupled/genetics , von Willebrand Diseases/genetics , von Willebrand Factor/genetics , 5' Untranslated Regions , Animals , Base Sequence , HEK293 Cells , Hemorrhage/physiopathology , Humans , Molecular Sequence Data , Polymorphism, Single Nucleotide , Sequence Homology, Nucleic Acid , Transcription, Genetic
14.
Thromb Haemost ; 113(4): 826-37, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25567036

ABSTRACT

Platelet responses to activating agonists are influenced by common population variants within or near G protein-coupled receptor (GPCR) genes that affect receptor activity. However, the impact of rare GPCR gene variants is unknown. We describe the rare single nucleotide variants (SNVs) in the coding and splice regions of 18 GPCR genes in 7,595 exomes from the 1,000-genomes and Exome Sequencing Project databases and in 31 cases with inherited platelet function disorders (IPFDs). In the population databases, the GPCR gene target regions contained 740 SNVs (318 synonymous, 410 missense, 7 stop gain and 6 splice region) of which 70 % had global minor allele frequency (MAF) < 0.05 %. Functional annotation using six computational algorithms, experimental evidence and structural data identified 156/740 (21 %) SNVs as potentially damaging to GPCR function, most commonly in regions encoding the transmembrane and C-terminal intracellular receptor domains. In 31 index cases with IPFDs (Gi-pathway defect n=15; secretion defect n=11; thromboxane pathway defect n=3 and complex defect n=2) there were 256 SNVs in the target regions of 15 stimulatory platelet GPCRs (34 unique; 12 with MAF< 1 % and 22 with MAF≥ 1 %). These included rare variants predicting R122H, P258T and V207A substitutions in the P2Y12 receptor that were annotated as potentially damaging, but only partially explained the platelet function defects in each case. Our data highlight that potentially damaging variants in platelet GPCR genes have low individual frequencies, but are collectively abundant in the population. Potentially damaging variants are also present in pedigrees with IPFDs and may contribute to complex laboratory phenotypes.


Subject(s)
Blood Platelet Disorders/blood , Blood Platelet Disorders/genetics , Blood Platelets/metabolism , Polymorphism, Single Nucleotide , Receptors, G-Protein-Coupled/blood , Receptors, G-Protein-Coupled/genetics , Computational Biology , Databases, Genetic , Exome , Female , Gene Frequency , Genetic Predisposition to Disease , HEK293 Cells , Heredity , Humans , Male , Pedigree , Phenotype , Platelet Function Tests , Receptors, Purinergic P2Y12/blood , Receptors, Purinergic P2Y12/genetics , Transfection
15.
Psychopharmacology (Berl) ; 232(1): 305-14, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24973897

ABSTRACT

RATIONALE: The novel opioid receptor antagonist, GSK1421498, has been shown to attenuate reward-driven compulsive behaviours, such as stimulant drug seeking or binge eating, in animals and humans. Here, we report new data on the receptor pharmacology of GSK121498, in comparison to naltrexone, naloxone, 6-ß-naltrexol and nalmefene. OBJECTIVES: To determine whether the novel opioid antagonist, GSK1521498, is an orthosteric or allosteric antagonist at the µ opioid receptor (MOPr) and whether it has neutral antagonist or inverse agonist properties. METHODS: A combination of radioligand binding assays and [(35)S]GTPγS binding assays was employed. RESULTS: GSK1521498 completely displaced [(3)H]naloxone binding to MOPr and did not alter the rate of [(3)H]naloxone dissociation from MOPr observations compatible with it binding to the orthosteric site on MOPr. GSK1521498 exhibited inverse agonism when MOPr was overexpressed but not when the level of MOPr expression was low. In parallel studies under conditions of high receptor expression density, naloxone, naltrexone, 6-ß-naltrexol and nalmefene exhibited partial agonism, not inverse agonism as has been reported previously for naloxone and naltrexone. In brain tissue from mice receiving a prolonged morphine pre-treatment, GSK1521498 exhibited slight inverse agonism. CONCLUSIONS: Differences between GSK1521498 and naltrexone in their effects on compulsive reward seeking are arguably linked to the more selective and complete MOPr antagonism of GSK1521498 versus the partial MOPr agonism of naltrexone. GSK1521498 is also pharmacologically differentiated by its inverse agonist efficacy at high levels of MOPr expression, but this may be less likely to contribute to behavioural differentiation at patho-physiological levels of expression.


Subject(s)
Compulsive Behavior/metabolism , Indans/pharmacology , Receptors, Opioid, mu/antagonists & inhibitors , Reward , Triazoles/pharmacology , Analgesics, Opioid/metabolism , Analgesics, Opioid/pharmacology , Analgesics, Opioid/therapeutic use , Animals , CHO Cells , Compulsive Behavior/drug therapy , Cricetinae , Cricetulus , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , HEK293 Cells , Humans , Indans/metabolism , Indans/therapeutic use , Ligands , Male , Mice , Morphine/metabolism , Morphine/pharmacology , Morphine/therapeutic use , Naloxone/metabolism , Naloxone/pharmacology , Naloxone/therapeutic use , Naltrexone/analogs & derivatives , Naltrexone/metabolism , Naltrexone/pharmacology , Naltrexone/therapeutic use , Narcotic Antagonists/metabolism , Narcotic Antagonists/pharmacology , Narcotic Antagonists/therapeutic use , Protein Binding/physiology , Receptors, Opioid, mu/metabolism , Treatment Outcome , Triazoles/metabolism , Triazoles/therapeutic use
16.
Thromb Haemost ; 111(5): 923-32, 2014 May 05.
Article in English | MEDLINE | ID: mdl-24452735

ABSTRACT

A small number of thromboxane receptor variants have been described in patients with a bleeding history that result in platelet dysfunction. We have identified a patient with a history of significant bleeding, who expresses a novel heterozygous thromboxane receptor variant that predicts an asparagine to serine substitution (N42S). This asparagine is conserved across all class A GPCRs, suggesting a vital role for receptor structure and function.We investigated the functional consequences of the TP receptor heterozygous N42S substitution by performing platelet function studies on platelet-rich plasma taken from the patient and healthy controls. We investigated the N42S mutation by expressing the wild-type (WT) and mutant receptor in human embryonic kidney (HEK) cells. Aggregation studies showed an ablation of arachidonic acid responses in the patient, whilst there was right-ward shift of the U46619 concentration response curve (CRC). Thromboxane generation was unaffected. Calcium mobilisation studies in cells lines showed a rightward shift of the U46619 CRC in N42S-expressing cells compared to WT. Radioligand binding studies revealed a reduction in BMax in platelets taken from the patient and in N42S-expressing cells, whilst cell studies confirmed poor surface expression. We have identified a novel thromboxane receptor variant, N42S, which results in platelet dysfunction due to reduced surface expression. It is associated with a significant bleeding history in the patient in whom it was identified. This is the first description of a naturally occurring variant that results in the substitution of this highly conserved residue and confirms the importance of this residue for correct GPCR function.


Subject(s)
Blood Platelets/physiology , Hemorrhage/genetics , Receptors, Thromboxane A2, Prostaglandin H2/metabolism , Asparagine/genetics , Conserved Sequence/genetics , Down-Regulation/genetics , Female , Genetic Variation , HEK293 Cells , Hemorrhage/blood , Heterozygote , Humans , Middle Aged , Mutation/genetics , Platelet Activation/genetics , Platelet Function Tests , Radioligand Assay , Receptors, G-Protein-Coupled/genetics , Receptors, Thromboxane A2, Prostaglandin H2/genetics
17.
Blood ; 123(8): e11-22, 2014 Feb 20.
Article in English | MEDLINE | ID: mdl-24408324

ABSTRACT

Up to 1% of the population have mild bleeding disorders, but these remain poorly characterized, particularly with regard to the roles of platelets. We have compared the usefulness of Optimul, a 96-well plate-based assay of 7 distinct pathways of platelet activation to characterize inherited platelet defects in comparison with light transmission aggregometry (LTA). Using Optimul and LTA, concentration-response curves were generated for arachidonic acid, ADP, collagen, epinephrine, Thrombin receptor activating-peptide, U46619, and ristocetin in samples from (1) healthy volunteers (n = 50), (2) healthy volunteers treated with antiplatelet agents in vitro (n = 10), and (3) patients with bleeding of unknown origin (n = 65). The assays gave concordant results in 82% of cases (κ = 0.62, P < .0001). Normal platelet function results were particularly predictive (sensitivity, 94%; negative predictive value, 91%), whereas a positive result was not always substantiated by LTA (specificity, 67%; positive predictive value, 77%). The Optimul assay was significantly more sensitive at characterizing defects in the thromboxane pathway, which presented with normal responses with LTA. The Optimul assay is sensitive to mild platelet defects, could be used as a rapid screening assay in patients presenting with bleeding symptoms, and detects changes in platelet function more readily than LTA. This trial was registered at www.isrctn.org as #ISRCTN 77951167.


Subject(s)
Blood Platelet Disorders/diagnosis , Drug Monitoring/methods , Hemorrhage/diagnosis , High-Throughput Screening Assays/methods , Platelet Activation/physiology , Platelet Aggregation Inhibitors/pharmacology , Adult , Blood Platelet Disorders/blood , Blood Platelet Disorders/genetics , Blood Platelets/drug effects , Blood Platelets/physiology , Female , Genetic Association Studies , Healthy Volunteers , Hemorrhage/blood , Hemorrhage/physiopathology , Humans , Male , Platelet Activation/drug effects , Predictive Value of Tests , Receptors, Thromboxane A2, Prostaglandin H2/genetics , Sensitivity and Specificity , Young Adult
18.
J Biol Chem ; 288(47): 34217-34229, 2013 Nov 22.
Article in English | MEDLINE | ID: mdl-24106270

ABSTRACT

Rho GTPases such as Rac, RhoA, and Cdc42 are vital for normal platelet function, but the role of RhoG in platelets has not been studied. In other cells, RhoG orchestrates processes integral to platelet function, including actin cytoskeletal rearrangement and membrane trafficking. We therefore hypothesized that RhoG would play a critical role in platelets. Here, we show that RhoG is expressed in human and mouse platelets and is activated by both collagen-related peptide (CRP) and thrombin stimulation. We used RhoG(-/-) mice to study the function of RhoG in platelets. Integrin activation and aggregation were reduced in RhoG(-/-) platelets stimulated by CRP, but responses to thrombin were normal. The central defect in RhoG(-/-) platelets was reduced secretion from α-granules, dense granules, and lysosomes following CRP stimulation. The integrin activation and aggregation defects could be rescued by ADP co-stimulation, indicating that they are a consequence of diminished dense granule secretion. Defective dense granule secretion in RhoG(-/-) platelets limited recruitment of additional platelets to growing thrombi in flowing blood in vitro and translated into reduced thrombus formation in vivo. Interestingly, tail bleeding times were normal in RhoG(-/-) mice, suggesting that the functions of RhoG in platelets are particularly relevant to thrombotic disorders.


Subject(s)
Blood Coagulation , Blood Platelets/enzymology , GTP Phosphohydrolases/metabolism , Secretory Vesicles/metabolism , Thrombosis/enzymology , Adenosine Diphosphate/pharmacology , Animals , Blood Platelets/pathology , Carrier Proteins/pharmacology , Female , GTP Phosphohydrolases/genetics , Hemostatics/pharmacology , Humans , Male , Mice , Mice, Knockout , Peptides/pharmacology , Platelet Activation/drug effects , Platelet Activation/genetics , Secretory Vesicles/genetics , Thrombin/metabolism , Thrombin/pharmacology , Thrombosis/genetics , Thrombosis/pathology , rho GTP-Binding Proteins
19.
Traffic ; 14(5): 585-98, 2013 May.
Article in English | MEDLINE | ID: mdl-23387322

ABSTRACT

P2Y12 receptor internalization and recycling play an essential role in ADP-induced platelet activation. Recently, we identified a patient with a mild bleeding disorder carrying a heterozygous mutation of P2Y12 (P341A) whose P2Y12 receptor recycling was significantly compromised. Using human cell line models, we identified key proteins regulating wild-type (WT) P2Y12 recycling and investigated P2Y12 -P341A receptor traffic. Treatment with ADP resulted in delayed Rab5-dependent internalization of P341A when compared with WT P2Y12 . While WT P2Y12 rapidly recycled back to the membrane via Rab4 and Rab11 recycling pathways, limited P341A recycling was observed, which relied upon Rab11 activity. Although minimal receptor degradation was evident, P341A was localized in Rab7-positive endosomes with considerable agonist-dependent accumulation in the trans-Golgi network (TGN). Rab7 activity is known to facilitate recruitment of retromer complex proteins to endosomes to transport cargo to the TGN. Here, we identified that P341A colocalized with Vps26; depletion of which blocked limited recycling and promoted receptor degradation. This study has identified key points of divergence in the endocytic traffic of P341A versus WT-P2Y12 . Given that these pathways are retained in human platelets, this research helps define the molecular mechanisms regulating P2Y12 receptor traffic and explain the compromised receptor function in the platelets of the P2Y12 -P341A-expressing patient.


Subject(s)
Endosomes/metabolism , Gene Expression Regulation , Receptors, Purinergic P2Y12/metabolism , rab5 GTP-Binding Proteins/metabolism , Adenosine Diphosphate/metabolism , Biological Transport , Blood Platelets/metabolism , Cell Line , Endocytosis , HEK293 Cells , Humans , Ligands , Mutant Proteins/metabolism , Mutation , Plasmids/metabolism , Protein Structure, Tertiary , rab GTP-Binding Proteins/metabolism
20.
Biochem Soc Trans ; 41(1): 225-30, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23356287

ABSTRACT

Platelets are critical for haemostasis, however inappropriate activation can lead to the development of arterial thrombosis, which can result in heart attack and stroke. ADP is a key platelet agonist that exerts its actions via stimulation of two surface GPCRs (G-protein-coupled receptors), P2Y(1) and P2Y(12). Similar to most GPCRs, P2Y receptor activity is tightly regulated by a number of complex mechanisms including receptor desensitization, internalization and recycling. In the present article, we review the molecular mechanisms that underlie P2Y(1) and P2Y(12) receptor regulation, with particular emphasis on the structural motifs within the P2Y(12) receptor, which are required to maintain regulatory protein interaction. The implications of these findings for platelet responsiveness are also discussed.


Subject(s)
Blood Platelets/metabolism , Receptors, Purinergic P2Y12/physiology , Amino Acid Sequence , Endocytosis , Humans , Molecular Sequence Data , Receptors, Purinergic P2Y12/chemistry , Receptors, Purinergic P2Y12/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...