Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Front Pharmacol ; 12: 702677, 2021.
Article in English | MEDLINE | ID: mdl-34566635

ABSTRACT

Neutrophilic inflammation is a key determinant of cystic fibrosis (CF) lung disease. Neutrophil-derived free DNA, released in the form of extracellular traps (NETs), significantly correlates with impaired lung function in patients with CF, underlying their pathogenetic role in CF lung disease. Thus, specific approaches to control NETosis of neutrophils migrated into the lungs may be clinically relevant in CF. We investigated the efficacy of phosphodiesterase (PDE) type-4 inhibitors, in vitro, on NET release by neutrophils from healthy volunteers and individuals with CF, and in vivo, on NET accumulation and lung inflammation in mice infected with Pseudomonas aeruginosa. PDE4 blockade curbed endotoxin-induced NET production and preserved cellular integrity and apoptosis in neutrophils, from healthy subjects and patients with CF, challenged with endotoxin, in vitro. The pharmacological effects of PDE4 inhibitors were significantly more evident on CF neutrophils. In a mouse model of Pseudomonas aeruginosa chronic infection, aerosol treatment with roflumilast, a selective PDE4 inhibitor, gave a significant reduction in free DNA in the BALF. This was accompanied by reduced citrullination of histone H3 in neutrophils migrated into the airways. Roflumilast-treated mice showed a significant improvement in weight recovery. Our study provides the first evidence that PDE4 blockade controls NETosis in vitro and in vivo, in CF-relevant models. Since selective PDE4 inhibitors have been recently approved for the treatment of COPD and psoriasis, our present results encourage clinical trials to test the efficacy of this class of drugs in CF.

2.
Clin Pharmacol Ther ; 104(1): 111-119, 2018 07.
Article in English | MEDLINE | ID: mdl-29574792

ABSTRACT

We studied the influence of cardiovascular (CV) risk factors, previous CV events, and cotreatments with preventive medicines, on residual platelet thromboxane (TX)B2 production in 182 patients chronically treated with enteric coated (EC)-aspirin (100 mg/day). The response to aspirin was also verified by assessing arachidonic acid-induced platelet aggregation and urinary 11-dehydro-TXB2 levels. Residual serum TXB2 levels exceeded the upper limit value for an adequate aspirin response in 14% of individuals. This phenomenon was detected at 12 hours after dosing with aspirin. The coadministration of statins (mostly atorvastatin) was an independent predictor of residual serum TXB2 levels, and the percentage of patients with enhanced values was significantly lower in statin users vs. nonusers. We provide evidence in vitro that atorvastatin reduced residual TXB2 generation by increasing the extent of acetylation of platelet COX-1 by aspirin. In conclusion, the coadministration of statins may counter the mechanisms associated with reduced bioavailability of aspirin detected in some individuals with CV disease.


Subject(s)
Aspirin/therapeutic use , Atorvastatin/therapeutic use , Blood Platelets/metabolism , Cardiovascular Diseases/prevention & control , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Platelet Aggregation Inhibitors/therapeutic use , Thromboxane B2/biosynthesis , Acetylation/drug effects , Aged , Aspirin/pharmacology , Atorvastatin/pharmacology , Biological Availability , Cardiovascular Diseases/epidemiology , Cyclooxygenase 1/drug effects , Cyclooxygenase 1/metabolism , Drug Therapy, Combination , Female , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , In Vitro Techniques , Male , Middle Aged , Platelet Aggregation Inhibitors/pharmacology , Primary Prevention , Risk Factors , Secondary Prevention , Tablets, Enteric-Coated , Thromboxane B2/analogs & derivatives , Thromboxane B2/urine
3.
Biochim Biophys Acta Mol Basis Dis ; 1863(12): 3243-3253, 2017 12.
Article in English | MEDLINE | ID: mdl-28847515

ABSTRACT

Although cystic fibrosis (CF) patients exhibit signs of endothelial perturbation, the functions of the cystic fibrosis conductance regulator (CFTR) in vascular endothelial cells (EC) are poorly defined. We sought to uncover biological activities of endothelial CFTR, relevant for vascular homeostasis and inflammation. We examined cells from human umbilical cords (HUVEC) and pulmonary artery isolated from non-cystic fibrosis (PAEC) and CF human lungs (CF-PAEC), under static conditions or physiological shear. CFTR activity, clearly detected in HUVEC and PAEC, was markedly reduced in CF-PAEC. CFTR blockade increased endothelial permeability to macromolecules and reduced trans­endothelial electrical resistance (TEER). Consistent with this, CF-PAEC displayed lower TEER compared to PAEC. Under shear, CFTR blockade reduced VE-cadherin and p120 catenin membrane expression and triggered the formation of paxillin- and vinculin-enriched membrane blebs that evolved in shrinking of the cell body and disruption of cell-cell contacts. These changes were accompanied by enhanced release of microvesicles, which displayed reduced capability to stimulate proliferation in recipient EC. CFTR blockade also suppressed insulin-induced NO generation by EC, likely by inhibiting eNOS and AKT phosphorylation, whereas it enhanced IL-8 release. Remarkably, phosphodiesterase inhibitors in combination with a ß2 adrenergic receptor agonist corrected functional and morphological changes triggered by CFTR dysfunction in EC. Our results uncover regulatory functions of CFTR in EC, suggesting a physiological role of CFTR in the maintenance EC homeostasis and its involvement in pathogenetic aspects of CF. Moreover, our findings open avenues for novel pharmacology to control endothelial dysfunction and its consequences in CF.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Cystic Fibrosis/pathology , Endothelial Cells/pathology , Antigens, CD/metabolism , Cadherins/metabolism , Cell Proliferation/physiology , Cyclic AMP/metabolism , Cystic Fibrosis/metabolism , Cytokines/metabolism , Endothelial Cells/metabolism , Homeostasis/physiology , Human Umbilical Vein Endothelial Cells , Humans , Insulin/pharmacology , Interleukin-8/metabolism , Nitric Oxide Synthase Type III/metabolism , Nitrogen Oxides/metabolism , Phosphorylation , Pulmonary Artery/metabolism , Pulmonary Artery/pathology , beta-Arrestin 2/metabolism
4.
J Biol Chem ; 292(31): 12828-12841, 2017 08 04.
Article in English | MEDLINE | ID: mdl-28600357

ABSTRACT

Inflammatory responses are elicited through lipid products of phospholipase A2 activity that acts on the membrane phospholipids, including the phosphoinositides, to form the proinflammatory arachidonic acid and, in parallel, the glycerophosphoinositols. Here, we investigate the role of the glycerophosphoinositol in the inflammatory response. We show that it is part of a negative feedback loop that limits proinflammatory and prothrombotic responses in human monocytes stimulated with lipopolysaccharide. This inhibition is exerted both on the signaling cascade initiated by the lipopolysaccharide with the glycerophosphoinositol-dependent decrease in IκB kinase α/ß, p38, JNK, and Erk1/2 kinase phosphorylation and at the nuclear level with decreased NF-κB translocation and binding to inflammatory gene promoters. In a model of endotoxemia in the mouse, treatment with glycerophosphoinositol reduced TNF-α synthesis, which supports the concept that glycerophosphoinositol inhibits the de novo synthesis of proinflammatory and prothrombotic compounds and might thus have a role as an endogenous mediator in the resolution of inflammation. As indicated, this effect of glycerophosphoinositol can also be exploited in the treatment of manifestations of severe inflammation by exogenous administration of the compound.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Blood Coagulation/drug effects , Endotoxemia/drug therapy , Gene Expression Regulation/drug effects , Inositol Phosphates/therapeutic use , Monocytes/drug effects , Protein Processing, Post-Translational/drug effects , Active Transport, Cell Nucleus/drug effects , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anticoagulants/pharmacology , Anticoagulants/therapeutic use , Biomarkers/blood , Biomarkers/metabolism , Cells, Cultured , Chromatin Immunoprecipitation , Dose-Response Relationship, Drug , Endotoxemia/immunology , Endotoxemia/metabolism , HeLa Cells , Humans , Inositol Phosphates/pharmacology , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/toxicity , Male , Mice, Inbred C57BL , Microscopy, Confocal , Monocytes/cytology , Monocytes/immunology , Monocytes/metabolism , NF-kappa B/antagonists & inhibitors , NF-kappa B/blood , NF-kappa B/metabolism , Phosphorylation/drug effects
6.
J Thromb Thrombolysis ; 39(2): 186-95, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25307674

ABSTRACT

In patients with acute coronary syndromes (ACS), early therapy with high-dose statins may reduce short-term adverse clinical outcomes. The mechanisms responsible are not known but could involve anti-inflammatory or anti-thrombotic effects. Compelling evidence from experimental models and clinical studies suggests that the interplay between inflammatory and thrombotic systems, typified by platelet-monocyte and platelet-neutrophil interactions, might be a key regulator of ischemic vascular events. The study sought to determine if early, high-dose administration of the HMG-CoA reductase inhibitor rosuvastatin in the setting of ACS exerts beneficial vascular effects by reducing, and inhibiting biomarkers of thromboinflammation, such as platelet-monocyte and platelet-neutrophil interactions, and biomarkers of myocardial necrosis. A total of 54 patients presenting with ACS within 8 h of symptom onset were randomized to rosuvastatin 40 mg or placebo. Rosuvastatin significantly reduced interactions between platelets and circulating neutrophils (P = 0.015) and monocytes (P = 0.009) within 24 h. No significant effects were observed on platelet aggregation or plasma levels of PF4, sP-selectin, or sCD40L, whereas significant reductions of RANTES occurred over time in both treatment groups. Plasma levels of myeloperoxidase (MPO) declined more rapidly with rosuvastatin therapy than placebo. In a subset of patients with normal cardiac necrosis biomarkers at randomization, rosuvastatin therapy was associated with less myocardial damage as measured by troponin-I or CK-MB. Early administration of high-dose statin therapy in patients with ACS appears to improve biomarkers of inflammation within 8 h, which may translate into fewer ischemic events.


Subject(s)
Acute Coronary Syndrome , Cell Communication/drug effects , Creatine Kinase, MB Form/blood , Peroxidase/blood , Rosuvastatin Calcium/administration & dosage , Troponin I/blood , Acute Coronary Syndrome/blood , Acute Coronary Syndrome/drug therapy , Acute Coronary Syndrome/physiopathology , Adult , Aged , Biomarkers , Blood Platelets , CD40 Ligand/blood , Dose-Response Relationship, Drug , Early Medical Intervention , Female , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Inflammation/blood , Male , Middle Aged , Monocytes , Neutrophils , P-Selectin/blood , Platelet Factor 4/blood , Thrombosis/blood , Treatment Outcome
7.
Arterioscler Thromb Vasc Biol ; 34(8): 1689-96, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24925970

ABSTRACT

OBJECTIVE: Platelet-neutrophil interactions play a key role in cardiovascular disease and inflammatory processes. Src family kinases mediate P-selectin glycoprotein ligand-1-Mac-1 cross talk necessary for firm platelet-neutrophil adhesion. Because Src family kinase activity can be regulated by cAMP-dependent pathways, in this work, we evaluated the role of phosphodiesterases in the signaling events that are required to sustain platelet-neutrophil interactions and neutrophil recruitment at the site of vascular injury. APPROACH AND RESULTS: In neutrophils exposed to P-selectin, selective phosphodiesterase 4 (PDE4) inhibition prevented Src family kinase-mediated phosphorylation of the proline-rich tyrosine kinase 2 on Tyr579/Tyr580. The effects of PDE4 inhibition required protein kinase A, likely through protein kinase A-mediated activation of COOH-terminal Src kinase, a major negative regulator of Src family kinases. PDE4, but not other phosphodiesterase inhibitors, reduced platelet-neutrophil conjugates as well as neutrophil firm adhesion on spread platelets under flow conditions. The effect of PDE4 inhibition on neutrophil adhesion was primarily mediated by downregulation of P-selectin-induced activation of Mac-1. In a murine model of endovascular injury, selective inhibition of PDE4 significantly reduced neutrophil recruitment at the site of vascular damage. CONCLUSIONS: This study identifies PDE4 as a central node in the signaling network that mediates platelet-neutrophil adhesion and suggests that pharmacological inhibition of PDE4 may represent a novel therapeutic avenue for the treatment of cardiovascular disease.


Subject(s)
Blood Platelets/drug effects , Femoral Artery/drug effects , Neutrophil Infiltration/drug effects , Neutrophils/drug effects , P-Selectin/metabolism , Phosphodiesterase 4 Inhibitors/pharmacology , Platelet Adhesiveness/drug effects , Vascular System Injuries/drug therapy , 4-(3-Butoxy-4-methoxybenzyl)-2-imidazolidinone/pharmacology , Animals , Blood Platelets/enzymology , Cyclic AMP-Dependent Protein Kinases/metabolism , Disease Models, Animal , Femoral Artery/enzymology , Femoral Artery/injuries , Focal Adhesion Kinase 2/metabolism , Humans , Macrophage-1 Antigen/genetics , Macrophage-1 Antigen/metabolism , Mice , Mice, Knockout , Neutrophils/enzymology , P-Selectin/genetics , Phosphorylation , Rolipram/pharmacology , Signal Transduction/drug effects , Time Factors , Vascular System Injuries/blood , Vascular System Injuries/enzymology , src-Family Kinases/metabolism
8.
Adv Ther ; 30(2): 190-202, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23359066

ABSTRACT

INTRODUCTION: Besides its critical role in metabolic homeostasis, peroxisome proliferator-activated receptor (PPAR)-γ modulates several cellular responses involved in atherothrombosis. This multicenter, double-blind, randomized study investigated the effects of two oral hypoglycemic agents on markers of inflammation, platelet activation, thrombogenesis, and oxidative stress in patients with type 2 diabetes. METHODS AND RESULTS: The primary objective of this study was to evaluate the effect on C-reactive protein (CRP) after a 16-week treatment period with either pioglitazone or metformin. Additionally, markers of vascular inflammatory response, platelet activation, thrombogenesis, oxidative stress, glucose, and lipid metabolism, as well as liver function, were measured. In total, 50 patients completed the study. Pioglitazone-treated patients were found to have statistically significantly larger decreases in mean CRP levels (-0.4 mg/dL) compared to those treated with metformin (-0.2 mg/dL) (P=0.04), as well as greater reductions in levels of mean fasting plasma glucose (-27 vs. -9 mg/dL; P=0.01), serum insulin (-2 vs. -1.9 mU/L; P=0.014), homeostatic model assessment (HOMA) (-1.2 vs. -0.9; P=0.015), and E-selectin (-12.4 vs. +3.4 µg/mL; P=0.01). Mean glycated hemoglobin (HbA1c) levels decreased in both treatment groups from baseline to week 16 (-0.4% in the pioglitazone group, -0.2% in the metformin group; P=0.36). Pioglitazone treatment was also found to be associated with a statistically significant increase in total cholesterol levels (+10 mg/dL in the pioglitazone arm, -3 mg/dL in the metformin arm; P=0.05) and a decrease in liver enzyme levels. CONCLUSIONS: The favorable changes in markers of systemic and vascular inflammatory response with pioglitazone suggest that it may positively influence the atherothrombotic process in type 2 diabetes.


Subject(s)
C-Reactive Protein/metabolism , Cardiovascular Diseases/prevention & control , Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/therapeutic use , Metformin/therapeutic use , Thiazolidinediones/therapeutic use , Aged , Biomarkers/metabolism , Cardiovascular Diseases/etiology , Cardiovascular Diseases/metabolism , Cell Adhesion Molecules/metabolism , Diabetes Mellitus, Type 2/complications , Double-Blind Method , Female , Humans , Inflammation/metabolism , Male , Middle Aged , Oxidative Stress , Pioglitazone , Platelet Activation , Treatment Outcome
9.
PLoS One ; 7(6): e39484, 2012.
Article in English | MEDLINE | ID: mdl-22761804

ABSTRACT

BACKGROUND: Neutrophils are involved in thrombus formation. We investigated whether specific features of neutrophil activation characterize patients with acute coronary syndromes (ACS) compared to stable angina and to systemic inflammatory diseases. METHODS AND FINDINGS: The myeloperoxidase (MPO) content of circulating neutrophils was determined by flow cytometry in 330 subjects: 69 consecutive patients with acute coronary syndromes (ACS), 69 with chronic stable angina (CSA), 50 with inflammation due to either non-infectious (acute bone fracture), infectious (sepsis) or autoimmune diseases (small and large vessel systemic vasculitis, rheumatoid arthritis). Four patients have also been studied before and after sterile acute injury of the myocardium (septal alcoholization). One hundred thirty-eight healthy donors were studied in parallel. Neutrophils with normal MPO content were 96% in controls, >92% in patients undergoing septal alcoholization, 91% in CSA patients, but only 35 and 30% in unstable angina and AMI (STEMI and NSTEMI) patients, compared to 80%, 75% and 2% of patients with giant cell arteritis, acute bone fracture and severe sepsis. In addition, in 32/33 STEMI and 9/21 NSTEMI patients respectively, 20% and 12% of neutrophils had complete MPO depletion during the first 4 hours after the onset of symptoms, a feature not observed in any other group of patients. MPO depletion was associated with platelet activation, indicated by P-selectin expression, activation and transactivation of leukocyte ß2-integrins and formation of platelet neutrophil and -monocyte aggregates. The injection of activated platelets in mice produced transient, P-selectin dependent, complete MPO depletion in about 50% of neutrophils. CONCLUSIONS: ACS are characterized by intense neutrophil activation, like other systemic inflammatory syndromes. In the very early phase of acute myocardial infarction only a subpopulation of neutrophils is massively activated, possibly via platelet-P selectin interactions. This paroxysmal activation could contribute to occlusive thrombosis.


Subject(s)
Angina, Unstable/immunology , Autoimmune Diseases/immunology , Myocardial Infarction/immunology , Neutrophil Activation/immunology , Neutrophils/immunology , Adult , Aged , Angina, Unstable/metabolism , Animals , Autoimmune Diseases/metabolism , Disease Models, Animal , Female , Humans , Male , Mice , Middle Aged , Myocardial Infarction/metabolism , Neutrophils/metabolism , P-Selectin/metabolism , Peroxidase/metabolism
10.
Thromb Haemost ; 107(6): 1130-40, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22436970

ABSTRACT

Prasugrel, through its active metabolite, reduces atherothrombosis and its clinical manifestations by inhibiting platelet activation and aggregation. Platelets also contribute to inflammation through interaction with different classes of leukocytes. We investigated whether the inhibitory effect of prasugrel on platelets also counteract inflammatory responses. The effect of prasugrel active metabolite, R-138727, was investigated on platelet P-selectin expression, platelet adhesion to polymorphonuclear leukocytes (PMN) and monocytes (MN) and Mac-1 expression in PMN and MN, in vitro, in human cells. The ex vivo effect of prasugrel administration on P-selectin, thromboxane (TXB)2 formation, platelet-PMN conjugates and Mac-1 expression in PMN triggered by PAR-4 agonist peptide was examined in whole blood from healthy mice as well as from mice in which an acute inflammatory reaction was induced by treatment with endotoxin. The effect of prasugrel on inflammatory markers in endotoxin-treated animals was also tested in vivo. R-138727 inhibited agonist-stimulated expression of platelet P-selectin, platelet-PMN and platelet-MN adhesion and platelet-dependent Mac-1 expression in leukocytes. Addition of aspirin did not modify the inhibitory effect elicited by R-138727. Treatment of mice with prasugrel resulted in a profound inhibition of platelet P-selectin expression, TXB2 production, platelet-PMN adhesion and Mac-1 expression in PMN induced by ex vivo stimulation with PAR-4 agonist peptide of whole blood from healthy or endotoxin-treated mice. Measurement of markers revealed that prasugrel reduced TXB2 and tumour necrosis factor-α synthesis and increased nitric oxide metabolites in endotoxin-treated mice in vivo. In conclusion, prasugrel reduces platelet interactions with PMN and MN. Through these effects prasugrel may curb platelet-mediated inflammatory responses.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Blood Platelets/drug effects , Inflammation Mediators/blood , Leukocytes/drug effects , Piperazines/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Purinergic P2Y Receptor Antagonists/pharmacology , Shock, Septic/drug therapy , Thiophenes/pharmacology , Animals , Aspirin/pharmacology , Biomarkers/blood , Blood Platelets/immunology , Disease Models, Animal , Down-Regulation , Humans , Leukocytes/immunology , Macrophage-1 Antigen/blood , Male , Mice , Mice, Inbred C57BL , Nitric Oxide/blood , Platelet Adhesiveness/drug effects , Platelet Function Tests , Prasugrel Hydrochloride , Selenoprotein P/blood , Shock, Septic/blood , Shock, Septic/immunology , Thromboxane B2/blood , Tumor Necrosis Factor-alpha/blood
11.
Prostaglandins Other Lipid Mediat ; 98(3-4): 122-8, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22330859

ABSTRACT

We studied the effects of 17ß-estradiol (E2) (10, 40 nM) on 2 vasoprotective pathways, i.e. cyclooxygenase-2 (COX-2)-dependent prostanoids and the antioxidant heme oxygenase-1 (HO-1), in human umbilical vein endothelial cells (HUVEC) exposed for 6h to steady laminar shear stress (LSS, 10 dyn/cm²), characteristic of atherosclerotic lesion-protected areas. COX-2 was induced by LSS versus static condition (SC). E2 did not significantly affect COX-2 expression in HUVEC cultured in SC or exposed to LSS. Prostacyclin (PGI2) and prostaglandin (PG)E2 were induced while PGF(2α) was reduced by LSS. E2 caused no effect or a small reduction of prostanoid biosynthesis. In HUVEC cultured in SC or exposed to LSS, E2 10 nM caused a comparable HO-1 induction (35-45%) while E2 40 nM was 5-fold more potent in LSS-exposed HUVEC than in SC (290% and 58%, respectively). PGI2 receptor antagonist RO3244794 did not affect HO-1 induction by E2. In conclusion, E2 may restrain oxidant stress in the endothelium through HO-1 induction by a mechanism independent on PGI2 signaling.


Subject(s)
Cyclooxygenase 2/metabolism , Estrogens/pharmacology , Heme Oxygenase-1/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/enzymology , Prostaglandins/biosynthesis , Cyclooxygenase 2/genetics , Gene Expression Regulation, Enzymologic/drug effects , Humans , RNA, Messenger/genetics , RNA, Messenger/metabolism , Stress, Mechanical
12.
J Clin Invest ; 121(11): 4462-76, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22005299

ABSTRACT

Diabetes mellitus is associated with platelet hyperactivity, which leads to increased morbidity and mortality from cardiovascular disease. This is coupled with enhanced levels of thromboxane (TX), an eicosanoid that facilitates platelet aggregation. Although intensely studied, the mechanism underlying the relationship among hyperglycemia, TX generation, and platelet hyperactivity remains unclear. We sought to identify key signaling components that connect high levels of glucose to TX generation and to examine their clinical relevance. In human platelets, aldose reductase synergistically modulated platelet response to both hyperglycemia and collagen exposure through a pathway involving ROS/PLCγ2/PKC/p38α MAPK. In clinical patients with platelet activation (deep vein thrombosis; saphenous vein graft occlusion after coronary bypass surgery), and particularly those with diabetes, urinary levels of a major enzymatic metabolite of TX (11-dehydro-TXB2 [TX-M]) were substantially increased. Elevated TX-M persisted in diabetic patients taking low-dose aspirin (acetylsalicylic acid, ASA), suggesting that such patients may have underlying endothelial damage, collagen exposure, and thrombovascular disease. Thus, our study has identified multiple potential signaling targets for designing combination chemotherapies that could inhibit the synergistic activation of platelets by hyperglycemia and collagen exposure.


Subject(s)
Aldehyde Reductase/blood , Blood Glucose/metabolism , Collagen/pharmacology , Platelet Activation/drug effects , Platelet Activation/physiology , Thromboxanes/blood , Adult , Aged , Aged, 80 and over , Aldehyde Reductase/antagonists & inhibitors , Aspirin/administration & dosage , Case-Control Studies , Diabetes Mellitus/blood , Enzyme Inhibitors/pharmacology , Female , Humans , In Vitro Techniques , Male , Middle Aged , Mitogen-Activated Protein Kinase 14/blood , Models, Biological , Oxidative Stress , Phospholipase C gamma/blood , Platelet Aggregation/drug effects , Platelet Aggregation/physiology , Platelet Aggregation Inhibitors/pharmacology , Protein Kinase C/blood , Reactive Oxygen Species/blood , Signal Transduction , Venous Thrombosis/blood
13.
Arterioscler Thromb Vasc Biol ; 30(12): 2357-61, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21071701

ABSTRACT

Platelet-leukocyte interactions define a basic cell process that is characterized by the exchange of signals between platelets and different types of leukocytes and that bridges 2 fundamental pathophysiological events: atherothrombosis and inflammatory immune reactions. When this process takes place at the site of atherosclerotic plaque development or at the site of endothelial injury, platelet-dependent leukocyte recruitment and activation contributes to the inflammatory reaction of the vessel wall, which accounts for the exacerbation of atherosclerosis and for intimal hyperplasia and plaque instability. Moreover, platelet-leukocyte interactions may have a key role in modulating a wide array of responses of both the innate and adaptive immune systems, thus contributing to the pathogenesis of inflammatory diseases and tissue damage, as well as to host defense.


Subject(s)
Blood Platelets/immunology , Cardiovascular Diseases/immunology , Inflammation/immunology , Leukocytes/immunology , Platelet Adhesiveness , Signal Transduction , Adaptive Immunity , Anti-Inflammatory Agents/therapeutic use , Blood Platelets/drug effects , Cardiovascular Diseases/blood , Cardiovascular Diseases/drug therapy , Humans , Immunity, Innate , Inflammation/blood , Inflammation/drug therapy , Inflammation Mediators/blood , Leukocytes/drug effects , Platelet Adhesiveness/drug effects , Signal Transduction/drug effects
14.
FASEB J ; 24(10): 3970-80, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20530751

ABSTRACT

Inflammatory lung disease is a primary cause of morbidity and mortality in cystic fibrosis (CF). Mechanisms of unresolved acute inflammation in CF are not completely known, although the involvement of cystic fibrosis transmembrane conductance regulator (CFTR) in nonrespiratory cells is emerging. Here we examined CFTR expression and function in human platelets (PLTs) and found that they express a biologically active CFTR. CFTR blockade gave an ∼50% reduction in lipoxin A(4) (LXA(4)) formation during PLT/polymorphonuclear leukocytes (PMN) coincubations by inhibiting the lipoxin synthase activity of PLT 12-lipoxygenase. PLTs from CF patients generated ∼40% less LXA(4) compared to healthy subject PLTs. CFTR inhibition increased PLT-dependent PMN viability (33.0±5.7 vs. 61.2±8.2%; P=0.033), suppressed nitric oxide generation (0.23±0.04 vs. 0.11±0.002 pmol/10(8) PLTs; P=0.004), while reducing AKT (1.02±0.12 vs. 0.71±0.007 U; P=0.04), and increasing p38 MAPK phosphorylation (0.650±0.09 vs. 1.04±0.24 U; P=0.03). Taken together, these findings indicate that PLTs from CF patients are affected by the molecular defect of CFTR. Moreover, this CF PLT abnormality may explain the failure of resolution in CF.


Subject(s)
Blood Platelets/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/blood , Inflammation Mediators/physiology , Apoptosis , Cell Line , Female , Humans , Immunohistochemistry , Male , Microscopy, Electron, Transmission , Phosphorylation , Protein Kinases/metabolism , Reverse Transcriptase Polymerase Chain Reaction
15.
Nat Immunol ; 11(4): 328-34, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20208538

ABSTRACT

Pentraxins are a superfamily of conserved proteins involved in the acute-phase response and innate immunity. Pentraxin 3 (PTX3), a prototypical member of the long pentraxin subfamily, is a key component of the humoral arm of innate immunity that is essential for resistance to certain pathogens. A regulatory role for pentraxins in inflammation has long been recognized, but the underlying mechanisms remain unclear. Here we report that PTX3 bound P-selectin and attenuated neutrophil recruitment at sites of inflammation. PTX3 released from activated leukocytes functioned locally to dampen neutrophil recruitment and regulate inflammation. Antibodies have glycosylation-dependent regulatory effect on inflammation. Therefore, PTX3, which is an essential component of humoral innate immunity, and immunoglobulins share functional outputs, including complement activation, opsonization and, as shown here, glycosylation-dependent regulation of inflammation.


Subject(s)
C-Reactive Protein/immunology , Inflammation/immunology , Leukocyte Rolling/immunology , Neutrophil Infiltration/immunology , Serum Amyloid P-Component/immunology , Acute Lung Injury/immunology , Animals , CHO Cells , Cell Separation , Cricetinae , Cricetulus , Electrophoresis, Polyacrylamide Gel , Flow Cytometry , Humans , Immunity, Humoral/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Recombinant Proteins/immunology
16.
Blood ; 113(21): 5254-65, 2009 May 21.
Article in English | MEDLINE | ID: mdl-19264679

ABSTRACT

Activated platelets express ligands, which are recognized by counterreceptors on neutrophils. Here, we show that the ensuing cell-to-cell interaction programs neutrophil phagocytic function, resulting in activated platelet clearance. Neutrophils that have internalized platelets circulate in the blood of patients with acute myocardial infarction, and the extent of platelet clearance correlates with expression of platelet activation, including P-selectin. Activated platelets injected intravenously in experimental animals are detectable in circulating neutrophils 60 minutes after, and within 3 hours, more than 70% circulating neutrophils have internalized platelets. Platelet clearance comprises 2 events: adhesion to neutrophils, which requires divalent cations and depends on P-selectin, on the P-selectin glycoprotein ligand-1 (PSGL-1), and on the CD11b/CD18 beta2 integrin; and internalization, which is abrogated by the phosphatidylserine-binding protein annexin A5. Adhesion to platelets causes neutrophil degranulation and is blocked by antibodies specific for P-selectin and PSGL-1, either in a synthetic medium in vitro or in the whole blood, therefore in the presence of a physiologic array of plasma cofactors and opsonins. The data suggest that the interaction between circulating platelets and neutrophils influences innate immune functions, possibly contributing to regulate vascular inflammation.


Subject(s)
Blood Platelets/immunology , CD18 Antigens/immunology , Neutrophils/immunology , P-Selectin/immunology , Phagocytosis , Phosphatidylserines/immunology , Cell Communication/immunology , Cell Degranulation , Humans , Immunity, Innate , Membrane Glycoproteins , Myocardial Infarction/blood , Platelet Activation/immunology
17.
Circ Res ; 104(4): 506-13, 2009 Feb 27.
Article in English | MEDLINE | ID: mdl-19122175

ABSTRACT

Cyclooxygenase (COX)-2 is among the endothelial genes upregulated by uniform laminar shear stress (LSS), characteristically associated with atherosclerotic lesion-protected areas. We have addressed whether the induction of COX-2-dependent prostanoids in endothelial cells by LSS plays a role in restraining endothelial tumor necrosis factor (TNF)-alpha generation, a proatherogenic cytokine, through the induction of heme oxygenase-1 (HO)-1, an antioxidant enzyme. In human umbilical vein endothelial cells (HUVECs) exposed to steady LSS of 10 dyn/cm(2) for 6 hours, COX-2 protein was significantly induced, whereas COX-1 and the downstream synthases were not significantly modulated. This was associated with significant (P<0.05) increase of 6-keto-prostaglandin (PG)F(1alpha) (the hydrolysis product of prostacyclin), PGE(2), and PGD(2). In contrast, TNF-alpha released in the medium in 6 hours (3633+/-882 pg) or detected in cells lysates (1091+/-270 pg) was significantly (P<0.05) reduced versus static condition (9100+/-2158 and 2208+/-300 pg, respectively). Coincident induction of HO-1 was detected. The finding that LSS-dependent reduction of TNF-alpha generation and HO-1 induction were abrogated by the selective inhibitor of COX-2 NS-398, the nonselective COX inhibitor aspirin, or the specific prostacyclin receptor (IP) antagonist RO3244794 illuminates the central role played by LSS-induced COX-2-dependent prostacyclin in restraining endothelial inflammation. Carbacyclin, an agonist of IP, induced HO-1. Similarly to inhibition of prostacyclin biosynthesis or activity, the novel imidazole-based HO-1 inhibitor QC15 reversed TNF-alpha reduction by LSS. These findings suggest that inhibition of COX-2-dependent prostacyclin might contribute to acceleration of atherogenesis in patients taking traditional nonsteroidal antiinflammatory drugs (NSAIDs) and NSAIDs selective for COX-2 through downregulation of HO-1, which halts TNF-alpha generation in human endothelial cells.


Subject(s)
Atherosclerosis/enzymology , Cyclooxygenase 2/metabolism , Endothelial Cells/enzymology , Epoprostenol/metabolism , Heme Oxygenase-1/metabolism , Inflammation/enzymology , Tumor Necrosis Factor-alpha/biosynthesis , 6-Ketoprostaglandin F1 alpha , Aspirin/adverse effects , Aspirin/pharmacology , Atherosclerosis/chemically induced , Benzofurans/pharmacology , Cells, Cultured , Cyclooxygenase 1/metabolism , Cyclooxygenase Inhibitors/adverse effects , Cyclooxygenase Inhibitors/pharmacology , Dinoprost/metabolism , Dinoprostone/metabolism , Down-Regulation , Endothelial Cells/drug effects , Epoprostenol/analogs & derivatives , Epoprostenol/pharmacology , Humans , Inflammation/chemically induced , Nitrobenzenes/adverse effects , Nitrobenzenes/pharmacology , Perfusion , Propionates/pharmacology , Prostaglandin D2/metabolism , Receptors, Epoprostenol , Receptors, Prostaglandin/drug effects , Receptors, Prostaglandin/metabolism , Stress, Mechanical , Sulfonamides/adverse effects , Sulfonamides/pharmacology , Up-Regulation
18.
Thromb Haemost ; 99(3): 594-601, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18327409

ABSTRACT

NAD(P)H oxidase is a prominent source of reactive oxygen species in the vasculature. Vascular NAD(P)H oxidase is comprised of several subunits, one of which, p22phox, is encoded by a gene exhibiting several allelic variants. Here the C(242)T nucleotide transition has been found to alter superoxide anion production and associated with an altered risk of coronary artery disease (CAD). We assessed the role of this variant in two case-control studies, and performed a meta-analysis of previously reported investigations relating it to vascular risk. Population I was comprised of 492 subjects with type 2 diabetes, with or without macrovascular disease, matched for age, sex, and duration of diabetes. Population II was comprised of 158 subjects with or without either CAD or cerebro-vascular disease, and matched for age, sex, smoking status, weight category and the presence of hypertension, dyslipidemia, and diabetes. Our findings were meta-analyzed together with additional studies retrieved from the literature. The C(242)T polymorphism distribution did not differ between cases and controls in populations I and II both at univariate and multivariate analyses, and this was confirmed in a meta-analysis with 11 previously published populations. The meta-analysis, however, suggested a protective role of the T allele on CAD as an end point in Asian populations. In conclusion, these data suggest a significant heterogeneity for a modulating role of the T allele in the C(242)T polymorphism of p22-phox for the occurrence of CAD across ethnicities, with the absence of a significant effect in Caucasians.


Subject(s)
Cardiovascular Diseases/genetics , Coronary Artery Disease/genetics , NADPH Oxidases/genetics , Polymorphism, Genetic , Aged , Asian People/genetics , Cardiovascular Diseases/enzymology , Cardiovascular Diseases/ethnology , Case-Control Studies , Coronary Artery Disease/enzymology , Coronary Artery Disease/ethnology , Female , Gene Frequency , Genetic Predisposition to Disease , Humans , Italy , Male , Middle Aged , NADPH Oxidases/metabolism , Odds Ratio , Reactive Oxygen Species/metabolism , Risk Assessment , Risk Factors , White People/genetics
19.
Toxicol Appl Pharmacol ; 229(2): 227-31, 2008 Jun 01.
Article in English | MEDLINE | ID: mdl-18289624

ABSTRACT

The mycotoxin ochratoxin A (OTA), an ubiquitous contaminant of food products endowed with a wide spectrum of toxicity, affects several functions of mononuclear leukocytes. Monocytes/macrophages play a major role in fibrin accumulation associated with immune-inflammatory processes through the production of tissue factor (TF) and plasminogen activator inhibitor 2 (PAI-2). We studied the effect of OTA on TF and PAI-2 production by human blood mononuclear cells (MNC). The cells were incubated for 3 or 18 h at 37 degrees C with non toxic OTA concentrations in the absence and in the presence of lipopolysaccharide (LPS) or other inflammatory agents. TF activity was measured by a one-stage clotting test. Antigen assays were performed by specific ELISAs in cell extracts or conditioned media and specific mRNAs were assessed by RT-PCR. OTA had no direct effect on TF and PAI-2 production by MNC. However, OTA caused a dose-dependent reduction in LPS-induced TF (activity, antigen and mRNA) and PAI-2 (antigen and mRNA) production with >85% inhibition at 1 mug/ml. Similar results were obtained when monocyte-enriched preparations were used instead of MNC. TF production was also impaired by OTA (1 mug/ml) when MNC were stimulated with phorbol myristate acetate (98% inhibition), IL-1beta (83%) or TNF-alpha (62%). The inhibition of TF and PAI-2 induction might represent a hitherto unrecognized mechanism whereby OTA exerts immunosuppressant activity.


Subject(s)
Monocytes/drug effects , Mycotoxins/toxicity , Plasminogen Activator Inhibitor 2/biosynthesis , Thromboplastin/biosynthesis , Base Sequence , DNA Primers , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Monocytes/immunology , Ochratoxins/toxicity , Reverse Transcriptase Polymerase Chain Reaction
20.
Br J Nutr ; 99(2): 240-7, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17845735

ABSTRACT

Lower CVD incidence is reported in Asian populations consuming soya-containing food. As polymorphonuclear leukocytes (PMN) are involved in the risk of CVD, we investigated the modulatory effect of soya isoflavones on several PMN functions and their molecular mechanisms in vitro. PMN, isolated from blood from healthy subjects, were tested upon activation with 1 microm- n-formyl-methyl-leucyl-phenylalanine (fMLP) for superoxide anion production (ferric cytochrome c reduction) and released elastase (chromogenic test). PMN homotypic aggregates stimulated by fMLP or P-selectin in dynamic conditions were detected by optical microscopy. PMN, mixed with thrombin-activated, washed platelets, formed cell aggregates, measured by flow cytometry. Phosphorylation of Pyk2, a focal adhesion kinase, was studied by immunoprecipitation and immunoblotting with specific antibodies. Genistein, daidzein and equol inhibited superoxide anion production (IC50 0.25 (sem 0.1), 21.0 (sem 4.2) and 13.0 (sem 2.8) microm, respectively); the release of elastase was prevented by genistein (IC50 63 (sem 17) microm). PMN homotypic aggregates, stimulated by fMLP, were significantly reduced (24 (sem 12) and 51 (sem 14) % of control) by 100 microm genistein and equol. P-selectin-induced aggregates were reduced to 19 (sem 6), 44 (sem 10) and 28 (sem 9) % of control by 100 microm genistein, daidzein and equol, respectively. Genistein, daidzein and equol also significantly reduced mixed platelet-PMN aggregates (IC50 4.0 (sem 0.9), 57 (sem 6) and 66 (sem 23) microm, respectively). In PMN challenged by fMLP or P-selectin, activation of Pyk2 was prevented by isoflavones. The cardioprotective effect of soya-containing food might be linked to reduction of PMN activation and PMN-platelet interaction, novel targets for the biological effects of soya isoflavones.


Subject(s)
Glycine max/chemistry , Isoflavones/pharmacology , Neutrophils/drug effects , Cell Adhesion/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Equol , Focal Adhesion Kinase 2/metabolism , Genistein/pharmacology , Humans , Macrophage-1 Antigen/metabolism , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neutrophils/physiology , P-Selectin/pharmacology , Pancreatic Elastase/metabolism , Phosphorylation , Platelet Adhesiveness/drug effects , Superoxides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...