Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 203
Filter
1.
Platelets ; 33(3): 404-415, 2022 Apr 03.
Article in English | MEDLINE | ID: mdl-34097573

ABSTRACT

Several Janus kinase (JAK) inhibitors (jakinibs) have recently been approved to treat inflammatory, autoimmune and hematological conditions. Despite emerging roles for JAKs and downstream signal transducer and activator of transcription (STAT) proteins in platelets, it remains unknown whether jakinibs affect platelet function. Here, we profile platelet biochemical and physiological responses in vitro in the presence of five different clinically relevant jakinibs, including ruxolitinib, upadacitinib, oclacitinib, baricitinib and tofacitinib. Flow cytometry, microscopy and other assays found that potent JAK1/2 inhibitors baricitinib and ruxolitinib reduced platelet adhesion to collagen, as well as platelet aggregation, secretion and integrin αIIbß3 activation in response to the glycoprotein VI (GPVI) agonist collagen-related peptide (CRP-XL). Western blot analysis demonstrated that jakinibs reduced Akt phosphorylation and activation following GPVI activation, where ruxolitinib and baricitinib prevented DAPP1 phosphorylation. In contrast, jakinibs had no effects on platelet responses to thrombin. Inhibitors of GPVI and JAK signaling also abrogated platelet STAT5 phosphorylation following CRP-XL stimulation. Additional pharmacologic experiments supported roles for STAT5 in platelet secretion, integrin activation and cytoskeletal responses. Together, our results demonstrate that ruxolitinib and baricitinib have inhibitory effects on platelet function in vitro and support roles for JAK/STAT5 pathways in GPVI/ITAM mediated platelet function.


Subject(s)
Azetidines/therapeutic use , Blood Platelets/metabolism , Janus Kinase Inhibitors/therapeutic use , Nitriles/therapeutic use , Platelet Activation/drug effects , Platelet Adhesiveness/drug effects , Platelet Membrane Glycoproteins/drug effects , Purines/therapeutic use , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use , Sulfonamides/therapeutic use , Azetidines/pharmacology , Humans , Janus Kinase Inhibitors/pharmacology , Nitriles/pharmacology , Platelet Membrane Glycoproteins/metabolism , Purines/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Sulfonamides/pharmacology
2.
Platelets ; 32(1): 29-41, 2021 Jan 02.
Article in English | MEDLINE | ID: mdl-33307909

ABSTRACT

Antiplatelet medications comprise the cornerstone of treatment for diseases that involve arterial thrombosis, including acute coronary syndromes (ACS), stroke and peripheral arterial disease. However, antiplatelet medications may cause bleeding and, furthermore, thrombotic events may still recur despite treatment. The interaction of collagen with GPVI receptors on the surface of platelets has been identified as one of the major players in the pathophysiology of arterial thrombosis that occurs following atherosclerotic plaque rupture. Promisingly, GPVI deficiency in humans appears to have a minimal impact on bleeding. These findings together suggest that targeting platelet GPVI may provide a novel treatment strategy that provides additional antithrombotic efficacy with minimal disruption of normal hemostasis compared to conventional antiplatelet medications. CLEC-2 is gaining interest as a therapeutic target for a variety of thrombo-inflammatory disorders including deep vein thrombosis (DVT) with treatment also predicted to cause minimal disruption to hemostasis. GPVI and CLEC-2 signal through Src, Syk and Tec family tyrosine kinases, providing additional strategies for inhibiting both receptors. In this review, we summarize the evidence regarding GPVI and CLEC-2 and strategies for inhibiting these receptors to inhibit platelet recruitment and activation in thrombotic diseases.


Subject(s)
Lectins, C-Type/drug effects , Membrane Glycoproteins/drug effects , Platelet Aggregation Inhibitors/therapeutic use , Platelet Membrane Glycoproteins/drug effects , Protein-Tyrosine Kinases/drug effects , Humans , Platelet Aggregation Inhibitors/pharmacology
3.
Blood Adv ; 3(7): 1154-1166, 2019 04 09.
Article in English | MEDLINE | ID: mdl-30967391

ABSTRACT

Diacylglycerol kinases (DGKs) are a family of enzymes that convert diacylglycerol (DAG) into phosphatidic acid (PA). The ζ isoform of DGK (DGKζ) has been reported to inhibit T-cell responsiveness by downregulating intracellular levels of DAG. However, its role in platelet function remains undefined. In this study, we show that DGKζ was expressed at significant levels in both platelets and megakaryocytes and that DGKζ-knockout (DGKζ-KO) mouse platelets were hyperreactive to glycoprotein VI (GPVI) agonists, as assessed by aggregation, spreading, granule secretion, and activation of relevant signal transduction molecules. In contrast, they were less responsive to thrombin. Platelets from DGKζ-KO mice accumulated faster on collagen-coated microfluidic surfaces under conditions of arterial shear and stopped blood flow faster after ferric chloride-induced carotid artery injury. Other measures of hemostasis, as measured by tail bleeding time and rotational thromboelastometry analysis, were normal. Interestingly, DGKζ deficiency led to increased GPVI expression on the platelet and megakaryocyte surfaces without affecting the expression of other platelet surface receptors. These results implicate DGKζ as a novel negative regulator of GPVI-mediated platelet activation that plays an important role in regulating thrombus formation in vivo.


Subject(s)
Diacylglycerol Kinase/pharmacology , Platelet Activation/drug effects , Platelet Membrane Glycoproteins/pharmacology , Animals , Blood Platelets/metabolism , Diacylglycerol Kinase/deficiency , Diacylglycerol Kinase/genetics , Diacylglycerol Kinase/metabolism , Hemostasis , Humans , Megakaryocytes/metabolism , Mice , Mice, Knockout , Platelet Membrane Glycoproteins/drug effects , Thrombosis/etiology
4.
Chin J Nat Med ; 13(1): 30-40, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25660286

ABSTRACT

The present study was designed to target fish for potential bioactive components contained in a Huang Lian Jie Du decoction (HLJDD) and identify the underlying mechanisms of action for the treatment of sepsis at the molecular level. he bioactive components database of HLJDD was constructed and the sepsis-associated targets were comprehensively investigated. The 3D structures of the PAFR and TXA2R proteins were established using the homology modelling (HM) method, and the molecular effects for sepsis treatment were analysed by comparing the bioactive components database and the sepsis targets using computational biology methods. The results of the screening were validated with biological testing against the human oral epidermal carcinoma cell line KB in vitro. We found that multiple bioactive compounds contained in the HLJDD interacted with multiple targets. We also predicted the promising compound leads for sepsis treatment, and the first 28 compounds were characterized. Several compounds, such as berberine, berberrubine and epiberberine, dose-dependently inhibited PGE2 production in human KB cells, and the effects were similar in the presence or absence of TPA. This study demonstrates a novel approach to identifying natural chemical compounds as new leads for the treatment of sepsis.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Berberine/pharmacokinetics , Drugs, Chinese Herbal/pharmacokinetics , Sepsis/drug therapy , Berberine/analogs & derivatives , Dinoprostone/biosynthesis , Drugs, Chinese Herbal/chemistry , Humans , KB Cells , Platelet Membrane Glycoproteins/drug effects , Protein Transport , Receptors, G-Protein-Coupled/drug effects , Receptors, Thromboxane A2, Prostaglandin H2/drug effects , Sepsis/metabolism , Tetradecanoylphorbol Acetate/pharmacokinetics
5.
Article in English | MEDLINE | ID: mdl-25143722

ABSTRACT

BACKGROUND: We recently published that platelet-activating factor receptor (PAFr) is upregulated on the epithelium of the proximal airways of current smokers and also in bronchial epithelial cells exposed to cigarette smoke extract. These treated cells also showed upregulation of Streptococcus pneumoniae adhesion. Bacterial wall phosphorylcholine specifically binds to PAFr expressed on airway epithelium, thus facilitating adherence and tissue invasion, which may be relevant to chronic obstructive pulmonary disease (COPD). Moreover, the use of inhaled corticosteroids (ICS) in COPD patients is associated with an increased risk of invasive respiratory pneumococcal infections. OBJECTIVE: In this study, we have investigated whether PAFr expression is especially upregulated in airway epithelium in COPD patients and whether this expression may be modulated by ICS therapy. METHODS: We cross-sectionally evaluated PAFr expression in bronchial biopsies from 15 COPD patients who were current smokers (COPD-smokers) and 12 COPD-ex-smokers, and we compared these to biopsies from 16 smokers with normal lung function. We assessed immunostaining with anti-PAFr monoclonal antibody. We also used material from a previous double-blinded randomized placebo-controlled 6-month ICS intervention study in COPD patients to explore the effect of ICS on PAFr expression. We employed computer-aided image analysis to quantify the percentage of epithelium stained for PAFr. RESULTS: Markedly enhanced expression of PAFr was found in both COPD-smokers (P<0.005) and COPD-ex-smokers (P<0.002) compared to smokers with normal lung function. There was little evidence that PAFr expression was affected by ICS therapy over 6 months. CONCLUSION: Epithelial PAFr expression is upregulated in smokers, especially in those with COPD, and is not obviously affected by ICS therapy.


Subject(s)
Bronchi/metabolism , Epithelial Cells/metabolism , Platelet Membrane Glycoproteins/metabolism , Pulmonary Disease, Chronic Obstructive/metabolism , Receptors, G-Protein-Coupled/metabolism , Administration, Inhalation , Adrenal Cortex Hormones/administration & dosage , Adult , Aged , Androstadienes/administration & dosage , Biopsy , Bronchi/drug effects , Cross-Sectional Studies , Epithelial Cells/drug effects , Female , Fluticasone , Humans , Image Interpretation, Computer-Assisted , Immunohistochemistry , Male , Middle Aged , Platelet Membrane Glycoproteins/drug effects , Pulmonary Disease, Chronic Obstructive/diagnosis , Pulmonary Disease, Chronic Obstructive/drug therapy , Receptors, G-Protein-Coupled/drug effects , Smoking/adverse effects , Smoking/metabolism , Smoking Cessation , Smoking Prevention , Time Factors , Treatment Outcome , Up-Regulation , Young Adult
6.
J Thromb Haemost ; 10(12): 2418-27, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23020554

ABSTRACT

The treatment of acute coronary syndromes has been considerably improved in recent years with the introduction of highly efficient antiplatelet drugs. However, there are still significant limitations: the recurrence of adverse vascular events remains a problem, and the improvement in efficacy is counterbalanced by an increased risk of bleeding, which is of particular importance in patients at risk of stroke. One of the most attractive targets for the development of new molecules with potential antithrombotic activity is platelet glycoprotein (GP)VI, because its blockade appears to ideally combine efficacy and safety. This review summarizes current knowledge on GPVI regarding its structure, its function, and its role in physiologic hemostasis and thrombosis. Strategies for inhibiting GPVI are presented, and evidence of the antithrombotic efficacy and safety of GPVI antagonists is provided.


Subject(s)
Antithrombins/pharmacology , Platelet Membrane Glycoproteins/drug effects , Acute Coronary Syndrome/drug therapy , Antithrombins/therapeutic use , Hemostasis , Humans , Platelet Membrane Glycoproteins/metabolism
7.
Curr Pharm Des ; 18(33): 5294-308, 2012.
Article in English | MEDLINE | ID: mdl-22724416

ABSTRACT

The concept of "pharmacogenetics" addresses genetically determined variation in how individuals respond to drugs. Accordingly, specific genetic variants have been suggested as contributors to a reduced response to various antiplatelet drugs. Aspirin is a cornerstone in secondary cardiovascular prevention and has been thoroughly investigated. The efficacy of aspirin is well documented, although with considerable interindividual variation. According to meta-analyses, a reduced antiplatelet effect of aspirin confers an increased risk of cardiovascular events. The platelet response to aspirin is assessed by in vitro evaluation of thromboxane-dependent platelet function. A reduced antiplatelet effect of aspirin can be explained by several mechanisms, which are largely determined by clinical, pharmacodynamic, biological and genetic factors. During the past decade, numerous studies have identified genetic polymorphisms significantly associated with cardiovascular events and modulating the antiplatelet effect of aspirin. However, results have been contradictory allowing only few firm conclusions to be drawn. Polymorphisms in genes encoding glycoproteins (IIb/IIIa, Ia/IIa, VI and Ibα), cyclooxygenases (1 and 2), adenosine diphosphate receptors (P2Y1 and P2Y12) and proteins of importance for haemostasis (thromboxane A2 receptor, coagulation factor XIII, etc.) have been investigated. In particular, a polymorphism in the gene encoding glycoprotein IIb/IIIa has been associated with a reduced antiplatelet effect of aspirin. The additive value of an individual's genetic makeup in predicting the antiplatelet effect of aspirin and the risk of cardiovascular events remains controversial. The present review outlines the pharmacology of aspirin and provides an overview of specific genetic variations considered to influence the antiplatelet effect of aspirin.


Subject(s)
Aspirin/therapeutic use , Blood Platelets/drug effects , Cyclooxygenase Inhibitors/therapeutic use , Drug Resistance/genetics , Pharmacogenetics , Platelet Aggregation Inhibitors/therapeutic use , Polymorphism, Genetic , Purinergic P2Y Receptor Antagonists/therapeutic use , Animals , Aspirin/adverse effects , Blood Platelets/metabolism , Cyclooxygenase Inhibitors/adverse effects , Genotype , Hemorrhage/chemically induced , Hemorrhage/genetics , Humans , Phenotype , Platelet Aggregation Inhibitors/adverse effects , Platelet Function Tests , Platelet Membrane Glycoproteins/drug effects , Platelet Membrane Glycoproteins/genetics , Platelet Membrane Glycoproteins/metabolism , Prostaglandin-Endoperoxide Synthases/blood , Prostaglandin-Endoperoxide Synthases/genetics , Purinergic P2Y Receptor Antagonists/adverse effects , Receptors, Purinergic P2Y/blood , Receptors, Purinergic P2Y/drug effects , Receptors, Purinergic P2Y/genetics , Risk Factors , Treatment Outcome
8.
Phytother Res ; 26(5): 687-91, 2012 May.
Article in English | MEDLINE | ID: mdl-22002630

ABSTRACT

Phytochemical investigation on the bark of Goniothalamus tapis Miq. and G. uvaroides King has resulted in the isolation of eight styryl-lactones, (-)-cryptomeridiol, liriodenine, 3-methyl-1H-benz[f]indole-4,9-dione, (-)-stigmasterol and dimethyl terephthalate. The structures of the compounds were elucidated by spectroscopic techniques. The compounds were evaluated for their effect on platelet-activating factor (PAF) receptor binding on rabbit platelets using (3) H-PAF as a ligand. Among the compounds tested, (-)-cryptomeridiol, (+)-goniothalamin and (+)-isoaltholactone exhibited a significant and concentration-dependent inhibitory effect on PAF receptor binding, with inhibitory concentration (IC)(50) values of 17.5, 19.7 and 46.5 µm, respectively. The inhibitory effects of the first two compounds were comparable to that obtained from the positive control, cedrol. The results indicated that these compounds were strong PAF receptor binding inhibitors.


Subject(s)
Goniothalamus/chemistry , Lactones/pharmacology , Plant Extracts/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Platelet Membrane Glycoproteins/antagonists & inhibitors , Receptors, G-Protein-Coupled/antagonists & inhibitors , Animals , Blood Platelets/drug effects , Blood Platelets/metabolism , Dose-Response Relationship, Drug , Furans/chemistry , Furans/isolation & purification , Furans/pharmacology , Inhibitory Concentration 50 , Lactones/chemistry , Lactones/isolation & purification , Naphthalenes/chemistry , Naphthalenes/isolation & purification , Naphthalenes/pharmacology , Plant Bark/chemistry , Plant Extracts/chemistry , Plant Extracts/isolation & purification , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/chemistry , Platelet Aggregation Inhibitors/isolation & purification , Platelet Membrane Glycoproteins/chemistry , Platelet Membrane Glycoproteins/drug effects , Platelet Membrane Glycoproteins/isolation & purification , Protein Binding/drug effects , Pyrones/chemistry , Pyrones/isolation & purification , Pyrones/pharmacology , Rabbits , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/drug effects , Receptors, G-Protein-Coupled/isolation & purification
9.
J Med Microbiol ; 61(Pt 1): 101-108, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21890513

ABSTRACT

Streptococcus pneumoniae is a major pathogen of respiratory infections that utilizes platelet-activating factor receptor (PAFR) for firm adherence to host cells. The mucolytic agent S-carboxymethylcysteine (S-CMC) has been shown to exert inhibitory effects against infection by several respiratory pathogens including S. pneumoniae in vitro and in vivo. Moreover, clinical studies have implicated the benefits of S-CMC in preventing exacerbation of chronic obstructive pulmonary disease, which is considered to be related to respiratory infections. In this study, to assess whether the potency of S-CMC is attributable to inhibition of pneumococcal adherence to host cells, an alveolar epithelial cell line stimulated with interleukin-1α was used as a model of inflamed epithelial cells. Despite upregulation of PAFR by inflammatory activation, treatment with S-CMC efficiently inhibited pneumococcal adherence to host epithelial cells. In order to gain insight into the inhibitory mechanism, the effects of S-CMC on PAFR expression were also investigated. Following treatment with S-CMC, PAFR expression was reduced at both mRNA and post-transcriptional levels. Interestingly, S-CMC was also effective in inhibiting pneumococcal adherence to cells transfected with PAFR small interfering RNAs. These results indicate S-CMC as a probable inhibitor targeting numerous epithelial receptors that interact with S. pneumoniae.


Subject(s)
Alveolar Epithelial Cells/microbiology , Anti-Infective Agents, Local/pharmacology , Bacterial Adhesion/drug effects , Carbocysteine/pharmacology , Streptococcus pneumoniae/drug effects , Cell Line, Tumor , Epithelial Cells/microbiology , Humans , Platelet Membrane Glycoproteins/drug effects , Platelet Membrane Glycoproteins/metabolism , Pulmonary Alveoli/cytology , Pulmonary Alveoli/microbiology , Receptors, G-Protein-Coupled/drug effects , Receptors, G-Protein-Coupled/metabolism , Streptococcus pneumoniae/physiology
10.
Am J Physiol Heart Circ Physiol ; 301(6): H2395-401, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21949111

ABSTRACT

Sodium-hydrogen exchanger (NHE), the principal sarcolemmal acid extruder in ventricular myocytes, is stimulated by a variety of autocrine/paracrine factors and contributes to myocardial injury and arrhythmias during ischemia-reperfusion. Platelet-activating factor (PAF; 1-o-alkyl-2-acetyl-sn-glycero-3-phosphocholine) is a potent proinflammatory phospholipid that is released in the heart in response to oxidative stress and promotes myocardial ischemia-reperfusion injury. PAF stimulates NHE in neutrophils and platelets, but its effect on cardiac NHE (NHE1) is unresolved. We utilized quiescent guinea pig ventricular myocytes bathed in bicarbonate-free solutions and epifluorescence to measure intracellular pH (pH(i)). Methylcarbamyl-PAF (C-PAF; 200 nM), a metabolically stable analog of PAF, significantly increased steady-state pH(i). The alkalosis was completely blocked by the NHE inhibitor, cariporide, and by sodium-free bathing solutions, indicating it was mediated by NHE activation. C-PAF also significantly increased the rate of acid extrusion induced by intracellular acidosis. The ability of C-PAF to increase steady-state pH(i) was completely blocked by the PAF receptor inhibitor WEB 2086 (10 µM), indicating the PAF receptor is required. A MEK inhibitor (PD98059; 25 µM) also completely blocked the rise in pH(i) induced by C-PAF, suggesting participation of the MAP kinase signaling cascade downstream of the PAF receptor. Inhibition of PKC with GF109203X (1 µM) and chelerythrine (2 µM) did not significantly affect the alkalosis induced by C-PAF. In summary, these results provide evidence that PAF stimulates cardiac NHE1, the effect occurs via the PAF receptor, and signal relay requires participation of the MAP kinase cascade.


Subject(s)
Heart Ventricles/metabolism , Myocytes, Cardiac/metabolism , Platelet Activating Factor/metabolism , Sodium-Hydrogen Exchangers/metabolism , Sodium/metabolism , Acidosis/metabolism , Alkalosis/metabolism , Animals , Azepines/pharmacology , Benzophenanthridines/pharmacology , Dose-Response Relationship, Drug , Flavonoids/pharmacology , Guanidines/pharmacology , Guinea Pigs , Heart Ventricles/drug effects , Hydrogen-Ion Concentration , Indoles/pharmacology , Ion Transport , Maleimides/pharmacology , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism , Myocytes, Cardiac/drug effects , Phospholipid Ethers/pharmacology , Platelet Membrane Glycoproteins/drug effects , Platelet Membrane Glycoproteins/metabolism , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Protein Kinase Inhibitors/pharmacology , Receptors, G-Protein-Coupled/drug effects , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Spectrometry, Fluorescence , Sulfones/pharmacology , Time Factors , Triazoles/pharmacology
11.
Arterioscler Thromb Vasc Biol ; 30(12): 2372-84, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21084706

ABSTRACT

Understanding genetic contributions to platelet function could have profound clinical ramifications for personalizing platelet-directed pharmacotherapy, by providing insight into the risks and possible benefits associated with specific genotypes. This article represents an integrated summary of presentations related to genetic regulation of platelet receptor expression and function given at the Fifth Annual Platelet Colloquium in January 2010. It is supplemented with additional highlights from the literature covering (1) approaches to determining and evidence for the associations of genetic variants with platelet hypo- and hyperresponsive phenotypes, (2) the ramifications of these polymorphisms with regard to clinical responses to antiplatelet therapies, and (3) the role of platelet function/genetic testing in guiding antiplatelet therapy.


Subject(s)
Blood Platelets/metabolism , Drug Design , Platelet Aggregation Inhibitors/therapeutic use , Platelet Membrane Glycoproteins/genetics , Animals , Blood Platelets/drug effects , Drug Resistance/genetics , Gene Expression Profiling , Gene Expression Regulation/drug effects , Genome-Wide Association Study , Genotype , Humans , Phenotype , Platelet Aggregation Inhibitors/chemistry , Platelet Membrane Glycoproteins/drug effects , Platelet Membrane Glycoproteins/metabolism , Polymorphism, Genetic
12.
J Clin Invest ; 120(2): 627-35, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20093777

ABSTRACT

Sickle cell disease (SCD) is characterized by intravascular hemolysis and inflammation coupled to a 400-fold greater incidence of invasive pneumococcal infection resulting in fulminant, lethal pneumococcal sepsis. Mechanistically, invasive infection is facilitated by a proinflammatory state that enhances receptor-mediated endocytosis of pneumococci into epithelial and endothelial cells. As statins reduce chronic inflammation, in addition to their serum cholesterol-lowering effects, we hypothesized that statin therapy might improve the outcome of pneumococcal infection in SCD. In this study, we tested this hypothesis in an experimental SCD mouse model and found that statin therapy prolonged survival following pneumococcal challenge. The protective effect resulted in part from decreased platelet-activating factor receptor expression on endothelia and epithelia, which led to reduced bacterial invasion. An additional protective effect resulted from inhibition of host cell lysis by pneumococcal cholesterol-dependent cytotoxins (CDCs), including pneumolysin. We conclude therefore that statins may be of prophylactic benefit against invasive pneumococcal disease in patients with SCD and, more broadly, in settings of bacterial pathogenesis driven by receptor-mediated endocytosis and the CDC class of toxins produced by Gram-positive invasive bacteria.


Subject(s)
Anemia, Sickle Cell/microbiology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Pneumococcal Infections/prevention & control , Anemia, Sickle Cell/pathology , Animals , Anti-Inflammatory Agents/therapeutic use , Bacterial Proteins/therapeutic use , Cytotoxins/antagonists & inhibitors , Cytotoxins/toxicity , Disease Models, Animal , Lung/drug effects , Lung/pathology , Mice , Mice, Knockout , Mice, Transgenic , Platelet Membrane Glycoproteins/drug effects , Platelet Membrane Glycoproteins/physiology , Pneumococcal Infections/pathology , Pneumococcal Infections/physiopathology , Receptors, G-Protein-Coupled/drug effects , Receptors, G-Protein-Coupled/physiology , Streptolysins/therapeutic use
13.
Inflamm Res ; 59 Suppl 2: S193-5, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20013029

ABSTRACT

BACKGROUND: Human leukocyte activation induced by specific and non-specific stimuli is characterized by the formation of lipid rafts defined as lipid-ordered domains that are more tightly packed than the surrounding non-raft phase of the bilayer. These lipid rafts are formed in parallel with profound membrane reorganization. OBJECTIVES: Analyse the rafting and non-rafting proteins present on the activated and resting basophil membrane and study their interest for the flow cytometric analysis of basophil activation. METHODS: Human basophils obtained from samples used for diagnostic cellular tests such as basophil or lymphocyte activation tests were stimulated either by the formyl-methionyl-leucyl-phenylalanine peptide (fMLP), by an anti-IgE or by an allergen. After 40 min at 37 degrees C, they were labelled by different antibodies conjugated to fluorescent dyes as an anti-IgE FITC, an anti-CCR3 PE, an anti-CD63, an anti-CD203c PE, an anti-11b, annexin V FITC or by cholera toxin FITC. Moreover, several experiments were analysed using an Amnis cytometer, allowing one to obtain the picture of the analysed cells. RESULTS: Anti-IgE or specific allergen elicits a membrane neo expression of CD63 at a high density and is poorly represented on resting basophil membrane. Upon an IgE-dependant activation some of the markers already present on resting basophil membrane, as CD203c, are up regulated and others, such as the IgE/IgE FcepsilonRI receptor and CCR3 are down regulated and submitted to the formation of clusters demonstrated by the pictures taken with the Amnis cytometer. For non-IgE dependant activators, such as fMLP, the picture was different since IgE was not down regulated, whereas CCR3 was down regulated. As demonstrated using annexin V or the cholera toxin used for analysing apoptosis, these phenomenon were paralleled by the formation of lipid rafts, gangliosides domains, such as GM1, which is accessible from the extra cellular medium. CONCLUSIONS: Basophil activation leads to membrane events close to the apoptosis phenomenon. The flow cytometric analysis of these membrane events may lead to protocols for allergen-induced activation and, may significantly increase cellular test sensitivity, particularly for drugs allergy diagnosis for which the usual protocols, such as those using CD63 alone, are insufficiently sensitive.


Subject(s)
Basophils/physiology , Macrophage Activation/physiology , Membrane Microdomains/chemistry , Annexin A5/pharmacology , Antigens, CD/drug effects , Cholera Toxin/pharmacology , Flow Cytometry , Humans , Image Processing, Computer-Assisted , Immunoglobulin E/pharmacology , In Vitro Techniques , Microscopy, Fluorescence , Microscopy, Video , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Platelet Membrane Glycoproteins/drug effects , Receptors, CCR3/antagonists & inhibitors , Receptors, IgE/drug effects , Tetraspanin 30
14.
FASEB J ; 23(11): 3957-66, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19608627

ABSTRACT

MicroRNAs (miRs) are 21- to 23-nucleotide RNA molecules that regulate the stability or translational efficiency of target messenger RNAs of proteins involved in cell growth and apoptosis. miR-92 is part of the mir-17-92 cluster, which comprises members with an effect on cell proliferation. However, the role of miR-92 is unknown, and its targets have not been identified. Here, we describe a mechanism through which miR-92 contributes to regulate cell proliferation. Using a miR-92 synthetic double-strand oligonucleotide, we demonstrate that miR-92 increases 32D myeloid cell proliferation and 5-bromo-2-deoxyuridine (BrdU) incorporation and inhibits cell death. The effect is miR-92 specific since the miR-92 antagomir inhibits cell proliferation. Moreover, we show that miR-92 acts by modulating p63-isoform abundance through down-regulatation of endogenous DeltaNp63beta. Using luciferase reporters containing p63 3'UTR fragments with wild-type or mutant miR-92 complementary sites, we demonstrate that the wild-type 3'UTR is a direct target of miR-92. Finally, we observed that a miR-92-resistant DeltaNp63beta isoform (without 3'UTR) inhibits cell proliferation and parallels the effect of the antagomir. We conclude that one of the molecular mechanisms through which miR-92 increases cell proliferation is by negative regulation of an isoform of the cell-cycle regulator p63.


Subject(s)
Antigens, CD/metabolism , MicroRNAs/physiology , Myeloid Cells/drug effects , Platelet Membrane Glycoproteins/metabolism , 3' Untranslated Regions , Antigens, CD/drug effects , Cell Proliferation/drug effects , Down-Regulation , Gene Knockdown Techniques , HCT116 Cells , Humans , Platelet Membrane Glycoproteins/drug effects , Protein Isoforms/metabolism , Tetraspanin 30
15.
Arterioscler Thromb Vasc Biol ; 29(3): 363-71, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19112165

ABSTRACT

OBJECTIVE: Mitochondrial depolarization aids platelet activation. Oxidized LDL (oxLDL) contains the medium length oxidatively truncated phospholipid hexadecyl azelaoyl-lysoPAF (HAz-LPAF) that disrupts mitochondrial function in nucleated cells, so oxLDL may augment platelet activation. METHODS AND RESULTS: Flow cytometry showed intact oxLDL particles synergized with subthreshold amounts of soluble agonists to increase intracellular Ca2+, and initiate platelet aggregation and surface expression of activated gpIIb/IIIa and P-selectin. oxLDL also induced aggregation and increased intracellular Ca2+ in FURA2-labeled cells by itself at low, although not higher, concentrations. HAz-LPAF, alone and in combination with substimulatory amounts of thrombin, rapidly increased cytoplasmic Ca2+ and initiated aggregation. HAz-LPAF depolarized mitochondria in intact platelets, but this required concentrations beyond those that directly activated platelets. An unexpectedly large series of chemically pure truncated phospholipids generated by oxidative fragmentation of arachidonoyl-, docosahexaneoyl-, or linoleoyl alkyl phospholipids were platelet agonists. The PAF receptor, thought to effectively recognize only phospholipids with very short sn-2 residues, was essential for platelet activation because PAF receptor agonists blocked signaling by all these medium length phospholipids and oxLDL. CONCLUSIONS: Intact oxLDL particles activate platelets through the PAF receptor, and the PAF receptor responds to a far wider range of oxidized phospholipids in oxLDL than anticipated.


Subject(s)
Blood Platelets/metabolism , Calcium Signaling , Lipoproteins, LDL/metabolism , Phospholipids/metabolism , Platelet Aggregation , Platelet Membrane Glycoproteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Adenosine Diphosphate/metabolism , Azepines/pharmacology , Blood Platelets/drug effects , Calcium Signaling/drug effects , Collagen/metabolism , Humans , Ligands , Membrane Potential, Mitochondrial , Mitochondria/metabolism , P-Selectin/metabolism , Phospholipid Ethers/pharmacology , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Platelet Membrane Glycoproteins/drug effects , Receptors, G-Protein-Coupled/drug effects , Thrombin/metabolism , Time Factors , Triazoles/pharmacology
16.
Blood ; 111(4): 2007-14, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-18056842

ABSTRACT

During blood feeding, mosquitoes inject saliva containing a mixture of molecules that inactivate or inhibit various components of the hemostatic response to the bite injury as well as the inflammatory reactions produced by the bite, to facilitate the ingestion of blood. However, the molecular functions of the individual saliva components remain largely unknown. Here, we describe anopheline antiplatelet protein (AAPP) isolated from the saliva of Anopheles stephensi, a human malaria vector mosquito. AAPP exhibited a strong and specific inhibitory activity toward collagen-induced platelet aggregation. The inhibitory mechanism involves direct binding of AAPP to collagen, which blocks platelet adhesion to collagen and inhibits the subsequent increase in intracellular Ca(2+) concentration ([Ca(2+)]i). The binding of AAPP to collagen effectively blocked platelet adhesion via glycoprotein VI (GPVI) and integrin alpha(2)beta(1). Cell adhesion assay showed that AAPP inhibited the binding of GPVI to collagen type I and III without direct effect on GPVI. Moreover, intravenously administered recombinant AAPP strongly inhibited collagen-induced platelet aggregation ex vivo in rats. In summary, AAPP is a malaria vector mosquito-derived specific antagonist of receptors that mediate the adhesion of platelets to collagen. Our study may provide important insights for elucidating the effects of mosquito blood feeding against host hemostasis.


Subject(s)
Collagen/pharmacology , Insect Proteins/pharmacology , Platelet Adhesiveness/drug effects , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/physiology , Platelet Membrane Glycoproteins/physiology , Salivary Proteins and Peptides/pharmacology , Animals , Anopheles , Blood Platelets/drug effects , Blood Platelets/physiology , Insect Proteins/genetics , Platelet Aggregation/drug effects , Platelet Membrane Glycoproteins/drug effects , Protein Binding , Recombinant Proteins/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Salivary Glands , Salivary Proteins and Peptides/genetics
17.
Thromb Haemost ; 98(6): 1285-90, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18064326

ABSTRACT

Glycoprotein (GP)VI, that binds collagen, together with GPIb-IX-V which binds vonWillebrand factor, forms an adheso-signalling complex on platelets that initiates thrombus formation in haemostasis and thrombosis. In this study, we show that two snake venom metalloproteinases, crotarhagin and alborhagin, induce ectodomain shedding of GPVI by a mechanism that involves activation of endogenous platelet metalloproteinases. Alborhagin is a viper venom metalloproteinase from Trimeresurus albolabris, while crotarhagin is a previously undescribed toxin from the rattlesnake Crotalus horridus horridus ( approximately 60-kDa non-reduced and reduced). Like alborhagin, crotarhagin induces aggregation in human platelet-rich plasma (maximal activity, approximately 0.3 microg/ml). Aggregation of washed platelets was inhibited by soluble GPVI ectodomain expressed as an Fc-fusion protein, confirming crotarhagin targeted GPVI. Treating washed platelets with crotarhagin or alborhagin resulted in time-dependent loss of surface GPVI and the appearance of an approximately 55-kDa soluble GPVI fragment in supernatants. Crotarhagin also induced shedding in GPVItransfected RBL-2H3 cells. Crotarhagin-induced shedding was metalloproteinase-dependent (inhibited by EDTA), but also blocked by inhibitors of GPVI signalling (Src kinase inhibitors, PP1 or PP2, or Syk inhibitor, piceatannol), indicating shedding required GPVI-dependent platelet activation. Together, the data suggest that the rattlesnake metalloproteinase, crotarhagin, and the viper toxin alborhagin, induce GPVI shedding by a mechanism involving activation of endogenous platelet metalloproteinases rather than direct cleavage of GPVI.


Subject(s)
Blood Platelets/drug effects , Crotalid Venoms/toxicity , Metalloendopeptidases/toxicity , Metalloproteases/toxicity , Platelet Aggregation/drug effects , Platelet Membrane Glycoproteins/drug effects , Receptors, Collagen/drug effects , Viper Venoms/toxicity , Animals , Blood Platelets/enzymology , Blood Platelets/metabolism , Cell Line , Dose-Response Relationship, Drug , Edetic Acid/pharmacology , Enzyme Activation , Humans , In Vitro Techniques , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/metabolism , Peptide Fragments/metabolism , Platelet Membrane Glycoproteins/chemistry , Platelet Membrane Glycoproteins/genetics , Platelet Membrane Glycoproteins/metabolism , Protease Inhibitors/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Structure, Tertiary , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Rats , Receptors, Collagen/chemistry , Receptors, Collagen/genetics , Receptors, Collagen/metabolism , Recombinant Fusion Proteins/metabolism , Signal Transduction/drug effects , Stilbenes/pharmacology , Syk Kinase , Time Factors , Transfection , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism
18.
J Neurochem ; 103(1): 88-97, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17877634

ABSTRACT

The bioactive lipid mediator platelet activating factor (PAF) is recognized as a key effecter of neuronal apoptosis, yet it is not clear whether its G-protein coupled receptor (PAFR) initiates or prevents PAF neurotoxicity. Using PAFR-/- and congenic wild-type mice, we show that PAF triggers caspase-3/7 activity and neuronal death in PAFR-/- but not PAFR+/+ cerebellar granule neurons. Restoring receptor expression by recombinant adenoviral infection protected cells from PAF challenge. Neuronal death was not mediated by nitric oxide or N-methyl-d-aspartate receptor signaling given that N-nitro-l-arginine methyl ester and MK-801 did not inhibit PAF-induced neuronal loss in PAFR-/- neurons. To intervene in PAFR-independent neurotoxicity, the anti-apoptotic actions of three structurally distinct PAF antagonists were compared to a panel of plant and fungal benzoic acid derivatives. We found that the PAF antagonist BN 52021 but not FR 49175 or CV 3988 inhibited PAFR-independent neurotoxicity. Orsellinic acid, a fungal-derived benzoic acid, blocked PAF-mediated neuronal apoptosis without affecting PAFR-mediated neuroprotection. These findings demonstrate that PAF can transduce apoptotic death in primary neurons independently of its G-protein coupled receptor, that PAFR activation is neuroprotective, and that orsellinic acid effectively attenuates PAFR-independent neuronal apoptosis.


Subject(s)
Apoptosis/drug effects , Neurons/drug effects , Platelet Activating Factor/toxicity , Platelet Membrane Glycoproteins/drug effects , Receptors, G-Protein-Coupled/drug effects , Resorcinols/pharmacology , Animals , Benzoates/pharmacology , Caspase Inhibitors , Cells, Cultured , Crosses, Genetic , Enzyme Inhibitors/pharmacology , Gene Transfer Techniques , Mice , Mice, Knockout , Neurons/cytology , Neurons/physiology , Platelet Activating Factor/antagonists & inhibitors , Platelet Membrane Glycoproteins/genetics , Platelet Membrane Glycoproteins/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects
19.
J Ethnopharmacol ; 111(1): 50-5, 2007 Apr 20.
Article in English | MEDLINE | ID: mdl-17150321

ABSTRACT

We investigated the vascular effects of a crude aqueous extract (AEEG) of Echinodorus grandiflorus (Alismataceae) using the in vitro experimental models of the rabbit isolated aorta and perfused kidney. Echinodorus grandiflorus, a native semi-aquatic plant widely distributed in Brazil, has been extensively used in Brazilian folk medicine for the treatment of high blood pressure and inflammatory diseases. The bolus injection of AEEG (0.1-10 mg) into the rabbit renal circulation pre-contracted with norepinephrine induced marked and dose-dependent vasodilator responses (maximum of 37+/-4%; n=6; P<0.001), which was similar to that induced by injection of 10 mmol acetylcholine (41+/-3%). Moreover, AEEG elicited a significant and concentration-dependent relaxation in the endothelium-intact, but not endothelium-denuded aortic rings, reaching the maximum of 81+/-5% (n=7, P<0.001). Inhibition of the nitric oxide-cGMP pathway with L-NAME (100 microM) or Methylene Blue (20 microM) reduced maximum relaxation induced by AEEG from 81+/-5% to 46+/-3 and 45+/-3%, respectively (n=7, P<0.001). A similar reduction was obtained with the incubation of the aortic rings with the selective PAF receptor antagonist WEB 2086 (10 microM) (from 81+/-5% to 55+/-3%; n=7; P<0.01). Conversely, blockade of muscarinic receptors with atropine (10 microM) did not affect the vasodilator effects of AEEG, while inhibition of the enzyme cyclooxigenase not only did not block, but rather potentiated vasodilation induced by AEEG (n=7, P<0.001). Finally, blockade of Ca(2+)- and ATP-activated K(+) channels using the specific blockers charydbotoxin (100 nM) and glibenclamide (3 microM), respectively, did not modify aortic relaxation induced by AEEG. We conclude that water-soluble extracts from leaves of Echinodorus grandiflorus elicit an endothelium-dependent, nitric oxide and PAF receptor-mediated vasodilation in rabbit aortic rings, which does not appear to involve the generation of vasodilating prostaglandins or the activation of K(+) channels. This potent vasodilator effect of the extracts was confirmed in the isolated rabbit renal circulation.


Subject(s)
Alismataceae , Aorta/drug effects , Kidney/blood supply , Renal Circulation/drug effects , Vasodilation/drug effects , Vasodilator Agents/pharmacology , Animals , Aorta/metabolism , Azepines/pharmacology , Brazil , Cyclic GMP/metabolism , Dose-Response Relationship, Drug , Endothelium, Vascular/drug effects , Enzyme Inhibitors/pharmacology , Guanylate Cyclase/antagonists & inhibitors , Guanylate Cyclase/metabolism , In Vitro Techniques , Kidney/metabolism , Methylene Blue/pharmacology , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Plant Extracts/pharmacology , Plant Leaves , Platelet Membrane Glycoproteins/drug effects , Platelet Membrane Glycoproteins/metabolism , Rabbits , Receptors, G-Protein-Coupled/drug effects , Receptors, G-Protein-Coupled/metabolism , Triazoles/pharmacology
20.
J Biol Chem ; 282(5): 3165-72, 2007 Feb 02.
Article in English | MEDLINE | ID: mdl-17132623

ABSTRACT

The human C-type lectin-like molecule CLEC-2 is expressed on the surface of platelets and signaling through CLEC-2 causes platelet activation and aggregation. CLEC-2 is a receptor for the platelet-aggregating snake venom protein rhodocytin. It is also a newly identified co-receptor for human immunodeficiency virus type 1 (HIV-1). An endogenous ligand has not yet been identified. We have solved the crystal structure of the extracellular domain of CLEC-2 to 1.6-A resolution, and identified the key structural features involved in ligand binding. A semi-helical loop region and flanking residues dominate the surface that is available for ligand binding. The precise distribution of hydrophobic and electrostatic features in this loop will determine the nature of any endogenous ligand with which it can interact. Major ligand-induced conformational change in CLEC-2 is unlikely as its overall fold is compact and robust. However, ligand binding could induce a tilt of a 3-10 helical portion of the long loop region. Mutational analysis and surface plasmon resonance binding studies support these observations. This study provides a framework for understanding the effects of rhodocytin venom binding on CLEC-2 and for understanding the nature of likely endogenous ligands and will provide a basis for rational design of drugs to block ligand binding.


Subject(s)
Lectins, C-Type/chemistry , Membrane Glycoproteins/chemistry , Platelet Membrane Glycoproteins/chemistry , Receptors, G-Protein-Coupled/chemistry , Crystallization , Crystallography, X-Ray , Humans , Lectins, C-Type/genetics , Membrane Glycoproteins/genetics , Models, Molecular , Mutagenesis , Platelet Membrane Glycoproteins/drug effects , Platelet Membrane Glycoproteins/genetics , Protein Conformation , Protein Structure, Secondary , Receptors, G-Protein-Coupled/drug effects , Receptors, G-Protein-Coupled/genetics , Recombinant Proteins/chemistry , Viper Venoms/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...