Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Int J Mol Sci ; 22(21)2021 Oct 20.
Article in English | MEDLINE | ID: mdl-34768772

ABSTRACT

Current cytoreductive and antithrombotic strategies in MPNs are mostly based on cell counts and on patient's demographic and clinical history. Despite the numerous studies conducted on platelet function and on the role of plasma factors, an accurate and reliable method to dynamically quantify the hypercoagulability states of these conditions is not yet part of clinical practice. Starting from our experience, and after having sifted through the literature, we propose an in-depth narrative report on the contribution of the clonal platelets of MPNs-rich in tissue factor (TF)-in promoting a perpetual procoagulant mechanism. The whole process results in an unbalanced generation of thrombin and is self-maintained by Protease Activated Receptors (PARs). We chose to define this model as a "circulating wound", as it indisputably links the coagulation, inflammation, and fibrotic progression of the disease, in analogy with what happens in some solid tumours. The platelet contribution to thrombin generation results in triggering a vicious circle supported by the PARs/TGF-beta axis. PAR antagonists could therefore be a good option for target therapy, both to contain the risk of vascular events and to slow the progression of the disease towards end-stage forms. Both the new and old strategies, however, will require tools capable of measuring procoagulant or prohaemorrhagic states in a more extensive and dynamic way to favour a less empirical management of MPNs and their potential clinical complications.


Subject(s)
Blood Platelets/metabolism , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/metabolism , Thrombin/biosynthesis , Animals , Biological Assay , Humans , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/drug therapy , Models, Biological , Receptors, Fibrinogen/metabolism , Thrombin/antagonists & inhibitors , Thrombophilia/physiopathology
2.
Vascul Pharmacol ; 138: 106830, 2021 06.
Article in English | MEDLINE | ID: mdl-33422688

ABSTRACT

INTRODUCTION: Although platelets contain a full proteasome system, its role in platelet function is not completely understood yet. Since the proteasome system may be involved in time-delayed processes, platelet responsiveness was investigated after long-term, bortezomib-mediated proteasome inhibition. MATERIALS AND METHODS: Citrate-anticoagulated whole blood was stored with 5 nM and 1 µM bortezomib for 24 h. Consecutively, aggregation was measured by light transmission in platelet-rich-plasma (PRP). Flow cytometry was performed to determine phosphorylation levels of the vasodilator-stimulated phosphoprotein (VASP), fibrinogen binding, PAC1-antibody binding and purinergic receptor expression in PRP, P2Y12 activity or glycoprotein (GP) Ib and IIb expression in whole blood. P2Y1 and P2X1 activities were assessed by calcium flux-induced fluorescence in washed platelets. Using PRP, adherent platelets on fibrinogen-, collagen- and ristocetin-coated surfaces were visualized and quantified by immunostaining. RESULTS: Under bortezomib, VASP phosphorylation was less inducible and nitric oxide-induced inhibition of fibrinogen binding was slightly reduced. Proteasome inhibition did not tamper adenosine diphosphate-mediated aggregation or purinergic receptor expression and activity. Induced expression of activated fibrinogen receptors and fibrinogen binding were not significantly influenced by incubation with bortezomib for 24 h. Aggregation values with threshold agonist concentrations were increased under bortezomib. Despite unchanged GPIb expression, bortezomib-treated platelets showed enhanced adhesion on coated surfaces. CONCLUSIONS: In platelets incubated for 24 h, bortezomib mediates a slight attenuation of inhibitory signaling, associated with facilitated platelet aggregation using threshold agonist concentrations and enhanced adhesion on agonist-coated surfaces.


Subject(s)
Blood Platelets/drug effects , Bortezomib/pharmacology , Platelet Adhesiveness/drug effects , Platelet Aggregation/drug effects , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/pharmacology , Blood Platelets/enzymology , Cell Adhesion Molecules/metabolism , Humans , Microfilament Proteins/metabolism , Nitric Oxide/metabolism , Phosphoproteins/metabolism , Phosphorylation , Platelet Glycoprotein GPIb-IX Complex/metabolism , Receptors, Fibrinogen/metabolism , Receptors, Purinergic P2/metabolism , Signal Transduction , Time Factors
3.
J Trauma Acute Care Surg ; 87(5): 1052-1060, 2019 11.
Article in English | MEDLINE | ID: mdl-31162329

ABSTRACT

BACKGROUND: Females are hypercoagulable and have survival benefit in trauma-induced coagulopathy (TIC). The mechanism for this sex-specific hypercoagulability is unknown. Platelets and platelet function are central in providing hemostatic potential and are the largest contributor to clot strength. Ligands (adenosine diphosphate [ADP] and platelet-activating factor [PAF]) bind distinct platelet receptors to potentiate activation and aggregation. We hypothesize that female platelets have a differential response to ADP and PAF, resulting in greater aggregation and activation compared to males, and that estradiol pretreatment of male or female platelets enhances this activity. METHODS: Platelets were collected from healthy volunteers: premenopausal/postmenopausal females (≤54 years, >54 years) and similarly aged males. Platelet aggregometry and flow cytometry (fibrinogen binding capacity) were examined. After treatment with ADP or PAF, platelet aggregation was assessed with Chronolog and activation assessed by CD41 receptor surface expression using flow cytometry. Aggregation and activation were again assessed after platelet pretreatment with estradiol. RESULTS: Healthy volunteers included 12 premenopausal and 13 postmenopausal females and 18 similarly aged males. Female platelets (combined premenopausal and postmenopausal) had increased aggregation with ADP stimulation, as compared to male platelets. Male and female platelets had differential fibrinogen receptor expression, with female platelets (combined premenopausal and postmenopausal) demonstrating robust activation with ADP versus male platelets with PAF. In the presence of estradiol incubation, male platelets' activation with PAF approximated that of females (combined premenopausal and postmenopausal) and activation with PAF was enhanced in both male and female platelets. CONCLUSION: Male and female platelets have differential response to stimuli, suggesting sex-dependent signaling and cellular activation. Female platelets have both increased aggregation and activation potential, and estradiol pretreatment feminizes male platelets to approximate female platelet activation with PAF. These findings offer potential explanation for sex-based differences in hemostatic potential in TIC and question whether donor sex of transfused platelets should be considered in resuscitation. Estradiol may also serve as a novel therapeutic adjunct in TIC.


Subject(s)
Blood Coagulation/physiology , Blood Platelets/metabolism , Platelet Aggregation/physiology , Adenosine Diphosphate/metabolism , Adult , Aged , Blood Coagulation Disorders/etiology , Blood Coagulation Disorders/physiopathology , Estradiol/metabolism , Female , Healthy Volunteers , Humans , Male , Middle Aged , Platelet Activating Factor/metabolism , Platelet Activation/physiology , Platelet Function Tests , Postmenopause/blood , Premenopause/blood , Receptors, Fibrinogen/metabolism , Sex Factors , Wounds and Injuries/complications , Young Adult
4.
Int J Nanomedicine ; 13: 1985-1992, 2018.
Article in English | MEDLINE | ID: mdl-29662311

ABSTRACT

BACKGROUND: Erythrocyte aggregation, a cardiovascular risk factor, is increased by high plasma fibrinogen levels. Here, the effect of different fibrinogen mutations on binding to its human erythrocyte receptor was assessed in order to identify the interaction sites. METHODS: Three fibrinogen variants were tested, specifically mutated in their putative integrin recognition sites on the Aα chain (mutants D97E, D574E and D97E/D574E) and compared with wild-type fibrinogen. RESULTS: Atomic force microscopy-based force spectroscopy measurements showed a significant decrease both on the fibrinogen-erythrocyte binding force and on its frequency for fibrinogen with the D97E mutation, indicating that the corresponding arginine-glycine-aspartate sequence (residues 95-97) is involved in this interaction, and supporting that the fibrinogen receptor on erythrocytes has a ß3 subunit. Changes in the fibrin clot network structure obtained with the D97E mutant were observed by scanning electron microscopy. CONCLUSION: These findings may lead to innovative perspectives on the development of new therapeutic approaches to overcome the risks of fibrinogen-driven erythrocyte hyperaggregation.


Subject(s)
Erythrocytes/metabolism , Fibrinogen/metabolism , Receptors, Fibrinogen/metabolism , Binding Sites , Fibrin/metabolism , Fibrinogen/genetics , Humans , Integrins/metabolism , Microscopy, Atomic Force , Mutation , Protein Binding , Thrombosis/metabolism
5.
Int J Nanomedicine ; 12: 3711-3720, 2017.
Article in English | MEDLINE | ID: mdl-28553109

ABSTRACT

Thromboembolic events (TEE) underwrite key causes of death in developed countries. While advanced imaging technologies such as computed tomography scans serve to diagnose blood clots during acute cardiovascular events, no such technology is available in routine primary care for TEE risk assessment. Here, we describe an imaging platform technology based on bioengineered fluorescent nanodiamond particles (F-NDPs) functionalized with bitistatin (Bit), a disintegrin that specifically binds to the αIIbß3 integrin, platelet fibrinogen receptor (PFR) on activated platelets. Covalent linkage of purified Bit to F-NDP was concentration-dependent and saturable, as validated by enzyme-linked immunosorbent assay using specific anti-Bit antibodies. F-NDP-Bit interacted with purified PFR, either in immobilized or soluble form. Lotrafiban, a nonpeptide, αIIbß3 receptor antagonist, specifically blocked F-NDP-Bit-PFR complex formation. Moreover, F-NDP-Bit specifically binds to activated platelets incorporated into a clot generated by thrombin-activated rat platelet-rich plasma (PRP). Our results suggest that engineered F-NDP-Bit particles could serve as noninvasive, "real-time" optical diagnostics for clots present in blood vessels.


Subject(s)
Nanodiamonds/chemistry , Peptides/chemistry , Platelet Activation/drug effects , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Receptors, Fibrinogen/metabolism , Animals , Benzodiazepines/pharmacology , Blood Platelets/drug effects , Fibrinogen/metabolism , Humans , Peptides/pharmacology , Piperidines/pharmacology , Rats, Inbred F344 , Receptors, Fibrinogen/chemistry , Snake Venoms , Thrombin/metabolism , Thrombosis/diagnostic imaging
6.
Thromb Haemost ; 110(5): 876-87, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23702580

ABSTRACT

Platelets are critically involved in atherosclerosis and acute thrombosis. The platelet phenotype shows a wide variability documented by the inherited difference of platelet reactivity, platelet volume and count and function of platelet surface receptors. Several candidate genes have been put into focus and investigated for their functional and prognostic role in healthy individuals and patients with cardiovascular (CV) disease treated with antiplatelet agents. In addition to genetic variation, other clinical, disease-related and demographic factors are important so-called non-genetic factors. Due to the small effect sizes of single nucleotide polymorphisms (SNP) in candidate genes and due to the low allele frequencies of functional relevant candidate SNPs, the identification of genetic risk factors with high predictive values generally depends on the sample size of study cohorts. In the post-genome era new array and bioinformatic technologies facilitate high throughput genome-wide association studies (GWAS) for the identification of novel candidate genes in large cardiovascular cohorts. One of the crucial aspects of platelet genomic studies is the precise definition of a specific clinical phenotype (e.g. stent thrombosis) as this will impact importantly the findings of genomic studies like GWAS. Here, we provide an update on genetic variation of platelet receptors and drug metabolising enzymes under specific consideration of data derived by GWAS. The potential impact of this information and the role in personalised therapeutic concepts will be discussed.


Subject(s)
Blood Platelets/physiology , Cardiovascular Diseases/diagnosis , Cardiovascular Diseases/drug therapy , Platelet Aggregation Inhibitors/therapeutic use , Receptors, Collagen/genetics , Receptors, Fibrinogen/genetics , Receptors, Purinergic P2Y/genetics , Animals , Biomarkers, Pharmacological/metabolism , Cardiovascular Diseases/genetics , Genetic Predisposition to Disease , Genome-Wide Association Study , High-Throughput Nucleotide Sequencing , Humans , Platelet Aggregation Inhibitors/pharmacology , Polymorphism, Single Nucleotide , Prognosis , Receptors, Collagen/metabolism , Receptors, Fibrinogen/metabolism , Receptors, Purinergic P2Y/metabolism , Risk
7.
Platelets ; 24(6): 428-34, 2013.
Article in English | MEDLINE | ID: mdl-22916829

ABSTRACT

Dopamine (DA) is a co-agonist for platelet activation; yet, donor DA treatment is associated with improved transplantation outcome in renal and heart recipients. Recently, N-octanoyl-dopamine (NOD) was developed which displays superior effects compared to DA in terms of graft protecting properties. Whereas DA is a known platelet co-agonist, the effect of NOD on platelet function is unknown. This is a hypothesis generating study with the aim to assess the effects and molecular mechanisms of NOD and NOD-like compounds on platelet function. The influence of DA, NOD, and NOD-like compounds on platelet responses to classical agonists (adenosine 5'-diphosphate (ADP), U46619) was investigated in six healthy donors by applying whole blood aggregometry (Multiplate®) and flow cytometry for Pac-1, CD62P, and CD63 expression. Changes in platelet cAMP concentrations were assessed by ELISA. While DA showed synergy in platelet activation by ADP and U46619, NOD caused significant inhibition of platelet function both in whole blood aggregometry and flow cytometry. The inhibitory effect of NOD was not mediated via cAMP levels. The nonredox-active NOD-analog N-octanoyl-tyramine had no effects on platelet function. Acetylated NOD conferred to NOD by intracellular esterases showed similar inhibitory effects as NOD. In contrast to DA, NOD is a potent inhibitor of platelet function most likely through intracellular redox-active processes. This adds to the overall protective effect of NOD on pre-transplantation injury and makes NOD an attractive candidate compound for donor or organ conditioning prior to transplantation.


Subject(s)
Blood Platelets/drug effects , Blood Platelets/physiology , Dopamine/analogs & derivatives , Cell Degranulation/drug effects , Cyclic AMP/biosynthesis , Dopamine/pharmacology , Humans , Platelet Aggregation/drug effects , Receptors, Fibrinogen/agonists , Receptors, Fibrinogen/metabolism
8.
IUBMB Life ; 64(6): 465-72, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22550017

ABSTRACT

The use of atomic force microscopy (AFM) applied to biological systems to generate high resolution images is gaining a wider acceptance. However, the most remarkable advances are being achieved on the use of the AFM to measure inter- and intramolecular interaction forces with piconewton resolution, not only to demonstrate this ability but also actually to solve biological and biomedical relevant questions. Single-molecule force spectroscopy recognition studies enable the detection of specific interaction forces, based on the AFM sensitivity and the possibility of manipulating individual molecules. In this review, we describe the basic principles of this methodology and some of the practical aspects involved. The ability to measure interactions at the single-molecule level is illustrated by some relevant examples. A special focus is given to the study of the fibrinogen-erythrocyte binding and its relevance as a cardiovascular risk factor. An approach to the latter problem by single-molecule force spectroscopy allowed the molecular recognition, characterization, and partial identification of a previously unknown receptor for fibrinogen on human erythrocytes.


Subject(s)
Microscopy, Atomic Force , Biomechanical Phenomena , Blood Platelets/physiology , Cell Shape , Erythrocytes/metabolism , Erythrocytes/physiology , Fibrinogen/chemistry , Fibrinogen/metabolism , Humans , Immobilized Proteins/chemistry , Immobilized Proteins/metabolism , Microscopy, Atomic Force/methods , Protein Binding , Receptors, Fibrinogen/chemistry , Receptors, Fibrinogen/metabolism , Single-Cell Analysis/methods , Surface Properties
9.
Bioorg Med Chem Lett ; 21(19): 5971-4, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-21852128

ABSTRACT

The novel RGD mimetics with phthalimidine central fragment were synthesized with the use of 4-piperidine-4-yl-butyric, 4-piperidine-4-yl-benzoic, 4-piperazine-4-yl-benzoic and 1,2,3,4-tetrahydroisoquinoline-7-carboxylic acids as surrogates of Arg motif. The synthesized compounds potently inhibited platelet aggregation in vitro and blocked FITC-Fg binding to α(IIb)ß(3) integrin in a suspension of washed human platelets. The key α(IIb)ß(3) protein-ligand interactions were determined in docking experiments.


Subject(s)
Drug Design , Phthalimides/chemical synthesis , Platelet Aggregation Inhibitors/chemical synthesis , Platelet Aggregation Inhibitors/pharmacology , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Receptors, Fibrinogen/antagonists & inhibitors , Arginine/analogs & derivatives , Arginine/metabolism , Blood Platelets/metabolism , Drug Evaluation, Preclinical , Fibrinogen/metabolism , Fluorescein-5-isothiocyanate/metabolism , Humans , Isoquinolines/chemistry , Isoquinolines/metabolism , Ligands , Oligopeptides/chemistry , Oligopeptides/metabolism , Platelet Aggregation Inhibitors/chemistry , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Protein Binding , Receptors, Fibrinogen/metabolism , Software , Stereoisomerism , Structure-Activity Relationship , Tirofiban , Tyrosine/analogs & derivatives , Tyrosine/chemistry , Tyrosine/metabolism
10.
Arterioscler Thromb Vasc Biol ; 31(10): 2306-13, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21737785

ABSTRACT

OBJECTIVE: Arterial thrombi contain variable amounts of red blood cells (RBCs), which interact with fibrinogen through an eptifibatide-sensitive receptor and modify the structure of fibrin. In this study, we evaluated the modulator role of RBCs in the lytic susceptibility of fibrin. METHODS AND RESULTS: If fibrin is formed at increasing RBC counts, scanning electron microscopy evidenced a decrease in fiber diameter from 150 to 96 nm at 40% (v/v) RBCs, an effect susceptible to eptifibatide inhibition (restoring 140 nm diameter). RBCs prolonged the lysis time in a homogeneous-phase fibrinolytic assay with tissue plasminogen activator (tPA) by up to 22.7±1.6%, but not in the presence of eptifibatide. Confocal laser microscopy using green fluorescent protein-labeled tPA and orange fluorescent fibrin showed that 20% to 40% (v/v) RBCs significantly slowed down the dissolution of the clots. The fluorescent tPA variant did not accumulate on the surface of fibrin containing RBCs at any cell count above 10%. The presence of RBCs in the clot suppressed the tPA-induced plasminogen activation, resulting in 45% less plasmin generated after 30 minutes of activation at 40% (v/v) RBCs. CONCLUSIONS: RBCs confer lytic resistance to fibrin resulting from modified fibrin structure and impaired plasminogen activation through a mechanism that involves eptifibatide-sensitive fibrinogen-RBC interactions.


Subject(s)
Erythrocytes/metabolism , Fibrin/metabolism , Fibrinolysis , Thrombosis/blood , Eptifibatide , Erythrocytes/drug effects , Fibrin/ultrastructure , Fibrinolysin/metabolism , Fibrinolysis/drug effects , Fibrinolytic Agents/pharmacology , Humans , Kinetics , Microscopy, Atomic Force , Microscopy, Confocal , Peptides/pharmacology , Plasminogen/metabolism , Receptors, Fibrinogen/drug effects , Receptors, Fibrinogen/metabolism , Recombinant Fusion Proteins/metabolism , Tissue Plasminogen Activator/metabolism
11.
Protein J ; 30(3): 159-66, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21380721

ABSTRACT

Streptococcus agalactiae is a leading cause of bacterial sepsis and meningitis in neonates. FbsA, a fibrinogen receptor of S. agalactiae is highly repetitive protein with each repeat containing 16 amino acids. The protein sequence of FbsA shows no homology to any known fibrinogen binding protein from other bacterial species, making it a unique fibrinogen receptor. FbsA is cloned, expressed in E. coli and purified. The recombinant protein shows a laddering pattern in SDS-PAGE gel because of its poor stability in solution. The instability of the protein is probably because of the presence Gln-Gly dipeptide in each repeat. The circular dichroism study of FbsA has shown that the protein is composed of alpha helices predominantly and random coils to a lesser extent, which agrees with the predicted secondary structure. Ab initio modeling of a single repeat shows that FbsA is made up of mainly alpha helix and the structural model of multiple repeats (3 or 4) suggests that the protein might adopt some form of a repeating helical structure and the overall conformation of the molecule might change depending on the number of repeats.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/isolation & purification , Carrier Proteins/chemistry , Carrier Proteins/isolation & purification , Gene Expression , Receptors, Fibrinogen/chemistry , Receptors, Fibrinogen/isolation & purification , Streptococcus agalactiae/metabolism , Amino Acid Sequence , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Carrier Proteins/genetics , Carrier Proteins/metabolism , Molecular Sequence Data , Protein Structure, Secondary , Receptors, Fibrinogen/genetics , Receptors, Fibrinogen/metabolism , Sequence Alignment , Streptococcus agalactiae/chemistry , Streptococcus agalactiae/genetics
12.
ACS Nano ; 4(8): 4609-20, 2010 Aug 24.
Article in English | MEDLINE | ID: mdl-20731444

ABSTRACT

The established hypothesis for the increase on erythrocyte aggregation associated with a higher incidence of cardiovascular pathologies is based on an increase on plasma adhesion proteins concentration, particularly fibrinogen. Fibrinogen-induced erythrocyte aggregation has been considered to be caused by its nonspecific binding to erythrocyte membranes. In contrast, platelets are known to have a fibrinogen integrin receptor expressed on the membrane surface (the membrane glycoprotein complex alpha(IIb)beta(3)). We demonstrate, by force spectroscopy measurements using an atomic force microscope (AFM), the existence of a single molecule interaction between fibrinogen and an unknown receptor on the erythrocyte membrane, with a lower but comparable affinity relative to platelet binding (average fibrinogen--erythrocyte and --platelet average (un)binding forces were 79 and 97 pN, respectively). This receptor is not as strongly influenced by calcium and eptifibatide (an alpha(IIb)beta(3) specific inhibitor) as the platelet receptor. However, its inhibition by eptifibatide indicates that it is an alpha(IIb)beta(3)-related integrin. Results obtained for a Glanzmann thrombastenia (a rare hereditary bleeding disease caused by alpha(IIb)beta(3) deficiency) patient show (for the first time) an impaired fibrinogen--erythrocyte binding. Correlation with genetic sequencing data demonstrates that one of the units of the fibrinogen receptor on erythrocytes is a product of the expression of the beta(3) gene, found to be mutated in this patient. This work demonstrates and validates the applicability of AFM-based force spectroscopy as a highly sensitive, rapid and low operation cost nanotool for the diagnostic of genetic mutations resulting in hematological diseases, with an unbiased functional evaluation of their severity.


Subject(s)
Erythrocytes/metabolism , Microscopy, Atomic Force/methods , Receptors, Fibrinogen/metabolism , Blood Platelets/metabolism , Calcium/metabolism , Eptifibatide , Erythrocyte Membrane/metabolism , Female , Fibrinogen/metabolism , Humans , Mutation , Peptides/pharmacology , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Protein Binding , Reproducibility of Results , Substrate Specificity , Thrombasthenia/genetics , Thrombasthenia/metabolism
13.
Methods Mol Biol ; 663: 1-28, 2010.
Article in English | MEDLINE | ID: mdl-20617413

ABSTRACT

Platelets play a crucial role in primary hemostasis by forming hemostatic plugs at sites of vascular injury. There is abundant evidence that platelets also play a pivotal role in the pathogenesis of arterial thrombotic disorders, including unstable angina (UA), myocardial infarction (MI), and stroke. The underlying pathophysiological mechanism of these processes has been recognized as the disruption or erosion of a vulnerable atherosclerotic plaque, leading to local platelet adhesion and subsequent formation of partially or completely occlusive platelet thrombi. A variety of methods have been used to assess platelet aggregation, blood coagulation, and the ex vivo and/or in vitro efficacy of platelet antagonists, anticoagulants, and thrombolytics.


Subject(s)
Drug Evaluation, Preclinical/methods , Fibrinolytic Agents/pharmacology , Animals , Blood Coagulation/drug effects , Blood Platelets/cytology , Blood Platelets/drug effects , Cell Adhesion/drug effects , Chromatography, Gel , Collagen/metabolism , Erythrocytes/cytology , Erythrocytes/metabolism , Fibrinolytic Agents/therapeutic use , Flow Cytometry , Humans , Lasers , Male , Platelet Aggregation/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Fibrinogen/metabolism , Rheology , Thrombelastography/drug effects , Thrombosis/drug therapy , Thrombosis/physiopathology , Viscosity
14.
Med Chem ; 5(2): 158-64, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19275714

ABSTRACT

It has been proposed a novel method for obtaining of 1,2,3,4-tetrahydroisoquinoline-7-carboxylic acid as Arg-mimetic within the framework of search for novel fibrinogen receptor antagonists. New alpha (IIb)beta(3) antagonists were prepared on a base of 1,2,3,4-tetrahydroisoquinoline-7-carboxylic acid. Their high antiaggregatory activities in a human platelet rich plasma and ability to block FITC-Fg binding to alpha (IIb)beta(3) on washed human platelets were estimated.


Subject(s)
Receptors, Fibrinogen/antagonists & inhibitors , Tetrahydroisoquinolines/chemical synthesis , Tetrahydroisoquinolines/pharmacology , Blood Platelets/drug effects , Blood Platelets/metabolism , Fibrinogen/metabolism , Humans , Magnetic Resonance Spectroscopy , Molecular Structure , Oligopeptides/antagonists & inhibitors , Platelet Aggregation/drug effects , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Platelet-Rich Plasma/drug effects , Protein Binding/drug effects , Receptors, Fibrinogen/metabolism , Tetrahydroisoquinolines/chemistry
15.
Acc Chem Res ; 39(11): 831-40, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17115723

ABSTRACT

How does one go about discovering new drugs? This question is addressed by descriptions of drug discovery research in three project areas that pertain to antagonist ligands for cell-surface receptors. The molecular targets of interest are protease-activated receptor-1 (PAR-1), vasopressin receptors (V1a and V2 subtypes), and the fibrinogen receptor (GPIIb/IIIa). I present different approaches to the identification of high-affinity ligands for these receptors, en route to drug candidates. The PAR-1 project resulted in a pharmacological tool compound that facilitated in vivo proof-of-principle studies, whereas the vasopressin and fibrinogen receptor projects resulted in several preclinical development compounds, three of which advanced into human clinical trials.


Subject(s)
Drug Design , Receptor, PAR-1/metabolism , Receptors, Fibrinogen/metabolism , Receptors, Vasopressin/metabolism , Animals , Humans , Ligands , Molecular Structure , Receptors, Cell Surface/metabolism
16.
J Thromb Haemost ; 4(2): 436-42, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16420577

ABSTRACT

BACKGROUND: The platelet fibrinogen receptor, a heterodimer consisting of integrin subunits alpha(IIb) and beta(3), is required for platelet aggregation, spreading, and hemostasis. Platelet agonists such as thrombin and adenosine diphosphate (ADP) lead to the activation of alpha(IIb)beta(3), thereby enhancing its affinity and avidity for binding fibrinogen (inside-out signaling). Furthermore, fibrinogen binding to alpha(IIb)beta(3) triggers cytoskeletal changes and granule release (outside-in signaling). AIM: Genetic approaches to characterize the molecular pathways involved in alpha(IIb)beta(3) signaling are not possible with anucleate blood platelets. Therefore, we have established an OP9 stromal cell co-culture system to generate megakaryocytes from human embryonic stem cells (hESCs). RESULTS: alpha(IIb)beta(3) activation, measured by soluble fibrinogen binding to hESC-derived megakaryocytes, /GPIbalpha(+) cells, is readily detectable following stimulation with known platelet agonists. Dose-response curves for peptide agonists specific for the two platelet thrombin receptors, protease-activated receptor 1 (PAR1) and PAR4, show a relative responsiveness that mirrors that of human platelets, and sub-maximal ADP responses are augmented by epinephrine. Moreover, hESC-derived megakaryocytes undergo lamellipodia formation, actin filament assembly, and vinculin localization at focal adhesions when plated on a fibrinogen-coated surface, characteristic of alpha(IIb)beta(3) outside-in signaling. Undifferentiated hESCs genetically modified by lentiviral infection can be cloned and maintained in an undifferentiated state and then differentiated into megakaryocytes capable of alpha(IIb)beta(3) activation. CONCLUSION: Using hESCs, we have developed a renewable source of human megakaryocytes, and a genetically tractable system for studying megakaryocytopoiesis and alpha(IIb)beta(3) signaling in the native cellular environment.


Subject(s)
Integrins/metabolism , Megakaryocytes/cytology , Megakaryocytes/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Thrombopoiesis/physiology , Cell Line , DNA/genetics , Embryo, Mammalian , Gene Expression , Genetic Vectors/genetics , Green Fluorescent Proteins/genetics , Humans , Lentivirus/genetics , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Ploidies , Receptors, Fibrinogen/metabolism , Recombinant Proteins/genetics , Signal Transduction , Thrombopoiesis/genetics
17.
Platelets ; 17(1): 49-55, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16308187

ABSTRACT

Platelet function can be studied using many different methods why it is of interest to understand how data from different assays relate to each other. In the present study we compare two methods suitable for screening purposes with two established although laborious methods, impedance aggregometry and platelet-rich plasma (PRP) aggregation. The alternative assays tested were: (i) exposure of active alphaIIbbeta3, in diluted whole blood and (ii) whole blood aggregation assessed by residual platelet counting. The fibrinogen receptor activation assay was found to have the lower variability, higher sensitivity to ADP, and higher signal to noise ratio compared with residual platelet counting. The sensitivity and response profile of the fibrinogen receptor activation assay and residual platelet counting were more similar to PRP aggregation than to impedance aggregometry, whereas impedance aggregometry displayed lower sensitivity to ADP. The two alternative assays correlated well with PRP aggregation as well as with each other. The fibrinogen receptor activation assay displayed the highest potency for AR-C69931MX, possibly due to a lower protein content compared with residual platelet counting. The two studied assays compare well with the more established assays, and are thus both good alternatives for platelet function testing and evaluation of new potential platelet antagonists.


Subject(s)
Adenosine Monophosphate/analogs & derivatives , Blood Platelets/physiology , Platelet Activation/physiology , Platelet Function Tests/methods , Adenosine Diphosphate/pharmacology , Adenosine Monophosphate/pharmacology , Blood Platelets/drug effects , Dose-Response Relationship, Drug , Drug Monitoring/methods , Humans , Platelet Activation/drug effects , Platelet Aggregation/drug effects , Platelet Aggregation/physiology , Platelet Count , Platelet Glycoprotein GPIIb-IIIa Complex/pharmacology , Receptors, Fibrinogen/drug effects , Receptors, Fibrinogen/metabolism , Reference Values , Regression Analysis , Sensitivity and Specificity , Time Factors
18.
Platelets ; 17(1): 61-6, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16308189

ABSTRACT

Platelet-leukocyte aggregates are considered to play a significant role in blood coagulation and inflammatory processes. We hypothesized that hormonal changes during the menstrual cycle affect the formation of heterotypic aggregates and therefore may constitute cycle-dependent variations of the susceptibility for thromboembolic events and inflammatory disease. We therefore measured platelet-leukocyte interaction by the determination of platelet-leukocyte aggregates (PLA), platelet P-Selectin expression, and platelet fibrinogen receptor activation by PAC-1 binding in 20 healthy women during their menstrual cycle by flow cytometry. The number of platelet-granulocyte aggregates (PGA) and platelet-monocyte aggregates (PMA) was higher at ovulation compared to any other time-point of the menstrual cycle (p = 0.005, p = 0.022, respectively). Likewise, P-Selectin expression peaked on day 14 (p = 0.040). The course of PLA formation during the menstrual cycle followed the course of estrogen levels, strongly suggesting direct effects of estrogen on platelet-leukocyte interaction. The susceptibility to form platelet-leukocyte aggregates that are inducible in vitro by a suboptimal concentration of thrombin receptor activating peptide-6 decreased slightly during the transition from day 1 to 14 (p = 0.040). These data indicate that platelet function varies during particular phases of the normal menstrual cycle.


Subject(s)
Blood Platelets/physiology , Leukocytes/physiology , Menstrual Cycle/physiology , Platelet Aggregation/physiology , Adult , Blood Platelets/cytology , Cell Adhesion/physiology , Dual Specificity Phosphatase 2 , Estrogens/analysis , Female , Fibrinogen/analysis , Flow Cytometry/methods , Humans , Leukocytes/cytology , P-Selectin/biosynthesis , Platelet Activation/physiology , Platelet Count , Protein Phosphatase 2 , Protein Tyrosine Phosphatases/metabolism , Receptors, Fibrinogen/metabolism , Reference Values
19.
Arterioscler Thromb Vasc Biol ; 25(10): 2222-7, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16123330

ABSTRACT

OBJECTIVE: The purpose of this study was to investigate the receptor requirements for enhanced IL-1beta-induced secretion of nitric oxide (NO) by endothelial cells (ECs) in the presence of fibrinogen. METHODS AND RESULTS: ECs were exposed to IL-1beta with or without fibrinogen and NO was measured as nitrite. NO production by EC exposed to fibrinogen (0.3+/-0.1 micromol/L) was comparable concentration to control (0.2+/-0.1 micromol/L), but IL-1beta significantly increased NO production (0.8+/-0.1 micromol/L), and the combination of both fibrinogen and IL-1beta resulted in a further increase to 2.2+/-0.2 micromol/L (P<0.002). 7E3 or LM609, antibodies to alphavbeta3, inhibited NO production stimulated by fibrinogen-bound IL-1beta to 0.2+/-0.1 micromol/L (P<0.001) or 0.2+/-0.03 micromol/L (P<0.0001), respectively. These levels were comparable to control and significantly less than with IL-1beta (P<0.002). EC or fibroblasts exposed to both fibrinogen and IL-1beta, but not vitronectin and IL-1beta, demonstrated positive Western blotting for alphavbeta3 after immunopurification with anti- IL-1R, indicating specific association between alphavbeta3 and IL-1R. Dual immunofluorescence also revealed colocalization of alphavbeta3 and IL-1R only when the cells were exposed to both fibrinogen and IL-1beta. CONCLUSIONS: The enhanced NO production by ECs in the presence of fibrinogen-bound IL-1beta requires the coordinated effects of colocalized alphavbeta3 and IL-1R.


Subject(s)
Endothelium, Vascular/drug effects , Fibrinogen/pharmacology , Integrin alphaVbeta3/metabolism , Interleukin-1/pharmacology , Receptors, Fibrinogen/metabolism , Thrombosis/metabolism , Antibodies, Monoclonal/pharmacology , Cells, Cultured , Drug Interactions , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Fibrinogen/metabolism , Humans , Integrin alphaVbeta3/immunology , Interleukin-1/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Receptors, Fibrinogen/immunology , Receptors, Interleukin-1/immunology , Receptors, Interleukin-1/metabolism , Umbilical Veins/cytology
20.
Blood ; 106(10): 3410-4, 2005 Nov 15.
Article in English | MEDLINE | ID: mdl-16020504

ABSTRACT

The binding of von Willebrand factor (VWF) to the platelet membrane glycoprotein Ib-IX (GPIb-IX) results in platelet activation. In this study, we sought to clarify previous conflicting reports and to elucidate the mechanism of activation and the precise role of extracellular signal-regulated kinase (Erk) in VWF-induced platelet activation. Erk2 is activated in platelets on stimulation with VWF/ristocetin in a time-dependent manner. VWF-induced Erk2 phosphorylation and thromboxane A2 (TXA2) release were completely blocked by PP2, an Src family kinase inhibitor, suggesting that Erk is downstream of Src family kinases. U73122, a phospholipase C inhibitor, also abolished TXA2 generation and Erk phosphorylation. Although VWF fostered the agglutination of platelets regardless of any additional treatment, the inhibition of mitogen-activated protein kinase kinase (MEK) with U0126 abolished VWF-induced platelet aggregation and thromboxane production in non-aspirin-treated washed platelets. However, in platelets treated with aspirin, VWF failed to cause any aggregation. Thus, we conclude that VWF stimulation of platelets results in phospholipase A2 activation through Erk stimulation and that Src family kinases and phospholipase C play essential roles in this event. We further conclude that VWF-induced platelet aggregation does not directly depend on Erk activation but has an absolute requirement for Src/Erk-mediated TXA2 generation.


Subject(s)
MAP Kinase Signaling System/physiology , Platelet Aggregation/physiology , Platelet Glycoprotein GPIb-IX Complex/metabolism , Receptors, Fibrinogen/metabolism , Thromboxane A2/metabolism , von Willebrand Factor/metabolism , Blood Platelets/metabolism , Cells, Cultured , Enzyme Activation/drug effects , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , MAP Kinase Signaling System/drug effects , Male , Phosphorylation/drug effects , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/pharmacology , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...