Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 129
Filter
1.
Circ Res ; 128(3): 386-400, 2021 02 05.
Article in English | MEDLINE | ID: mdl-33292062

ABSTRACT

RATIONALE: Current thrombolytic agents activate plasminogen to plasmin which triggers fibrinolysis to dissolve thrombi. Since plasmin is a nonspecific proteolytic enzyme, all of the current plasmin-dependent thrombolytics lead to serious hemorrhagic complications, demanding a new class of fibrinolytic enzymes independent from plasmin activation and undesirable side effects. We speculated that the mammalian version of bacterial heat-shock proteins could selectively degrade intravascular thrombi, a typical example of a highly aggregated protein mixture. OBJECTIVE: The objective of this study is to identify enzymes that can dissolve intravascular thrombi specifically without affecting fibrinogen and fibronectin so that the wound healing processes remain uninterrupted and tissues are not damaged. In this study, HtrA (high-temperature requirement A) proteins were tested for its specific proteolytic activity on intravascular thrombi independently from plasmin activation. METHODS AND RESULTS: HtrA1 and HtrA2/Omi proteins, collectively called as HtrAs, lysed ex vivo blood thrombi by degrading fibrin polymers. The thrombolysis by HtrAs was plasmin-independent and specific to vascular thrombi without causing the systemic activation of plasminogen and preventing nonspecific proteolysis of other proteins including fibrinogen and fibronectin. As expected, HtrAs did not disturb clotting and wound healing of excised wounds from mouse skin. It was further confirmed in a tail bleeding and a rebleeding assay that HtrAs allowed normal clotting and maintenance of clot stability in wounds, unlike other thrombolytics. Most importantly, HtrAs completely dissolved blood thrombi in tail thrombosis mice, and the intravenous injection of HtrAs to mice with pulmonary embolism completely dissolved intravascular thrombi and thus rescued thromboembolism. CONCLUSIONS: Here, we identified HtrA1 and HtrA2/Omi as plasmin-independent and highly specific thrombolytics that can dissolve intravascular thrombi specifically without bleeding risk. This work is the first report of a plasmin-independent thrombolytic pathway, providing HtrA1 and HtrA2/Omi as ideal therapeutic candidates for various thrombotic diseases without hemorrhagic complications.


Subject(s)
Fibrin/metabolism , Fibrinolysis/drug effects , Fibrinolytic Agents/pharmacology , High-Temperature Requirement A Serine Peptidase 1/pharmacology , High-Temperature Requirement A Serine Peptidase 2/pharmacology , Pulmonary Embolism/drug therapy , Thrombosis/drug therapy , Animals , Disease Models, Animal , Female , Fibrinolytic Agents/toxicity , Hemorrhage/chemically induced , High-Temperature Requirement A Serine Peptidase 1/toxicity , High-Temperature Requirement A Serine Peptidase 2/toxicity , Humans , Kinetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Pulmonary Embolism/blood , Pulmonary Embolism/enzymology , Recombinant Proteins/pharmacology , Thrombosis/blood , Thrombosis/enzymology , Wound Healing/drug effects
2.
Arterioscler Thromb Vasc Biol ; 41(2): 683-697, 2021 02.
Article in English | MEDLINE | ID: mdl-33267663

ABSTRACT

OBJECTIVE: Using 3KO (triple NOX [NADPH oxidase] knockout) mice (ie, NOX1-/-/NOX2-/-/NOX4-/-), we aimed to clarify the role of this family of enzymes in the regulation of platelets in vitro and hemostasis in vivo. Approach and Results: 3KO mice displayed significantly reduced platelet superoxide radical generation, which was associated with impaired platelet aggregation, adhesion, and thrombus formation in response to the key agonists collagen and thrombin. A comparison with single-gene knockouts suggested that the phenotype of 3KO platelets is the combination of the effects of the genetic deletion of NOX1 and NOX2, while NOX4 does not show any significant function in platelet regulation. 3KO platelets displayed significantly higher levels of cGMP-a negative platelet regulator that activates PKG (protein kinase G). The inhibition of PKG substantially but only partially rescued the defective phenotype of 3KO platelets, which are responsive to both collagen and thrombin in the presence of the PKG inhibitors KT5823 or Rp-8-pCPT-cGMPs, but not in the presence of the NOS (NO synthase) inhibitor L-NG-monomethyl arginine. In vivo, triple NOX deficiency protected against ferric chloride-driven carotid artery thrombosis and experimental pulmonary embolism, while hemostasis tested in a tail-tip transection assay was not affected. Procoagulatory activity of platelets (ie, phosphatidylserine surface exposure) and the coagulation cascade in platelet-free plasma were normal. CONCLUSIONS: This study indicates that inhibiting NOXs has strong antithrombotic effects partially caused by increased intracellular cGMP but spares hemostasis. NOXs are, therefore, pharmacotherapeutic targets to develop new antithrombotic drugs without bleeding side effects.


Subject(s)
Blood Coagulation , Blood Platelets/enzymology , Carotid Artery Thrombosis/enzymology , NADPH Oxidases/blood , Platelet Activation , Pulmonary Embolism/enzymology , Animals , Blood Coagulation/drug effects , Blood Platelets/drug effects , Carotid Artery Thrombosis/blood , Carotid Artery Thrombosis/genetics , Carotid Artery Thrombosis/prevention & control , Cyclic GMP/blood , Cyclic GMP-Dependent Protein Kinases/blood , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Female , Fibrinolytic Agents/pharmacology , Humans , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , NADPH Oxidase 1 , NADPH Oxidase 2 , NADPH Oxidase 4 , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/genetics , Platelet Activation/drug effects , Pulmonary Embolism/blood , Pulmonary Embolism/genetics , Pulmonary Embolism/prevention & control , Signal Transduction , Superoxides/blood
3.
J Biol Chem ; 294(33): 12547-12554, 2019 08 16.
Article in English | MEDLINE | ID: mdl-31266805

ABSTRACT

Protein tyrosine phosphatase nonreceptor type 7 (PTPN7), also called hematopoietic protein tyrosine phosphatase, controls extracellular signal-regulated protein kinase 1/2 (ERK1/2) and p38 mitogen-activated protein kinase in T lymphocytes. Because ERK1/2 plays an important role in regulating thromboxane A2 (TXA2) generation in platelets, we investigated the function of PTPN7 in these cells. Using immunoblot analysis, we detected PTPN7 in both human and mouse platelets but not in PTPN7-null mice. PTPN7 KO mouse platelets exhibited increased platelet functional responses, including aggregation, dense granule secretion, and TXA2 generation, compared with platelets from WT littermates, upon stimulation with both G protein-coupled receptor (GPCR) and glycoprotein VI (GPVI) agonists. Using the GPCR agonist AYPGKF in the presence of the COX inhibitor indomethacin, we found that PTPN7 KO mouse platelets aggregated and secreted to the same extent as WT platelets, suggesting that elevated TXA2 is responsible for the potentiation of platelet functional responses in PTPN7-KO platelets. Phosphorylation of ERK1/2 was also elevated in PTPN7 KO platelets. Stimulation of platelets with the GPVI agonist collagen-related peptide along with the COX inhibitor indomethacin did not result in phosphorylation of ERK1/2, indicating that GPVI-mediated ERK phosphorylation occurs through TXA2 Although bleeding times did not significantly differ between PTPN7-null and WT mice, time to death was significantly faster in PTPN7-null mice than in WT mice in a pulmonary thromboembolism model. We conclude that PTPN7 regulates platelet functional responses downstream of GPCR agonists, but not GPVI agonists, through inhibition of ERK activation and thromboxane generation.


Subject(s)
Blood Platelets/enzymology , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Pulmonary Embolism/enzymology , Animals , Blood Platelets/pathology , Disease Models, Animal , Enzyme Activation , Humans , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/genetics , Oligopeptides/pharmacology , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Pulmonary Embolism/genetics , Pulmonary Embolism/pathology
4.
Arterioscler Thromb Vasc Biol ; 38(4): 801-815, 2018 04.
Article in English | MEDLINE | ID: mdl-29301786

ABSTRACT

OBJECTIVE: One source of endogenous reverse transcriptase (eRT) activity in nucleated cells is the LINE-1/L1 (long interspersed nuclear element-1), a non-LTR retrotransposon that is implicated in the regulation of gene expression. Nevertheless, the presence and function of eRT activity and LINE-1 in human platelets, an anucleate cell, has not previously been determined. APPROACH AND RESULTS: We demonstrate that human and murine platelets possess robust eRT activity and identify the source as being LINE-1 ribonucleoprotein particles. Inhibition of eRT in vitro in isolated platelets from healthy individuals or in people with HIV treated with RT inhibitors enhanced global protein synthesis and platelet activation. If HIV patients were treated with reverse transcriptase inhibitor, we found that platelets from these patients had increased basal activation. We next discovered that eRT activity in platelets controlled the generation of RNA-DNA hybrids, which serve as translational repressors. Inhibition of platelet eRT lifted this RNA-DNA hybrid-induced translational block and was sufficient to increase protein expression of target RNAs identified by RNA-DNA hybrid immunoprecipitation. CONCLUSIONS: Thus, we provide the first evidence that platelets possess L1-encoded eRT activity. We also demonstrate that platelet eRT activity regulates platelet hyperreactivity and thrombosis and controls RNA-DNA hybrid formation and identify that RNA-DNA hybrids function as a novel translational control mechanism in human platelets.


Subject(s)
Blood Platelets/enzymology , DNA/blood , Long Interspersed Nucleotide Elements , Platelet Activation , Protein Biosynthesis , RNA-Directed DNA Polymerase/blood , RNA/blood , Thrombosis/blood , Animals , Blood Platelets/drug effects , Cell Line , DNA/genetics , Disease Models, Animal , Female , HIV Infections/blood , HIV Infections/drug therapy , HIV Infections/enzymology , HIV Infections/genetics , Humans , Male , Mice, Inbred C57BL , Platelet Activation/drug effects , Protein Biosynthesis/drug effects , Pulmonary Embolism/blood , Pulmonary Embolism/enzymology , Pulmonary Embolism/genetics , RNA/genetics , RNA-Directed DNA Polymerase/genetics , Reverse Transcriptase Inhibitors/therapeutic use , Thrombosis/enzymology , Thrombosis/genetics
5.
Arterioscler Thromb Vasc Biol ; 36(6): 1293-301, 2016 06.
Article in English | MEDLINE | ID: mdl-27102961

ABSTRACT

OBJECTIVE: The pathogenesis of chronic thromboembolic pulmonary hypertension (CTEPH) remains to be elucidated. Thrombin-activatable fibrinolysis inhibitor (TAFI) inhibits fibrinolysis. It remains to be elucidated whether TAFI is directly involved in the pathogenesis of CTEPH. We examined potential involvement of TAFI in the pathogenesis of CTEPH in humans. APPROACH AND RESULTS: We enrolled 68 consecutive patients undergoing right heart catheterization in our hospital, including those with CTEPH (n=27), those with pulmonary arterial hypertension (n=22), and controls (non-pulmonary hypertension, n=19). Whole blood clot lysis assay showed that the extent of clot remaining after 4 hours was significantly higher in CTEPH compared with pulmonary arterial hypertension or controls (41.9 versus 26.5 and 24.6%, both P<0.01). Moreover, plasma levels of TAFI were significantly higher in CTEPH than in pulmonary arterial hypertension or controls (19.4±4.2 versus 16.1±4.5 or 16.3±3.3 µg/mL, both P<0.05), which remained unchanged even after hemodynamic improvement by percutaneous transluminal pulmonary angioplasty. Furthermore, the extent of clot remaining after 4 hours was significantly improved with CPI-2KR (an inhibitor of activated TAFI) or prostaglandin E1 (an inhibitor of activation of platelets). Importantly, plasma levels of TAFI were significantly correlated with the extent of clot remaining after 4 hours. In addition, the extent of clot remaining after 4 hours was improved with an activated TAFI inhibitor. CONCLUSIONS: These results indicate that plasma levels of TAFI are elevated in patients with CTEPH and are correlated with resistance to clot lysis in those patients.


Subject(s)
Blood Platelets/enzymology , Carboxypeptidase B2/blood , Fibrinolysis , Hypertension, Pulmonary/blood , Pulmonary Embolism/blood , Adult , Aged , Biomarkers/blood , Blood Coagulation Tests , Blood Platelets/drug effects , Carboxypeptidase B2/antagonists & inhibitors , Carboxypeptidase B2/genetics , Cardiac Catheterization , Case-Control Studies , Chronic Disease , Female , Fibrinolysis/drug effects , Gene Frequency , Humans , Hypertension, Pulmonary/diagnosis , Hypertension, Pulmonary/enzymology , Hypertension, Pulmonary/etiology , Male , Middle Aged , Polymorphism, Single Nucleotide , Protease Inhibitors/pharmacology , Pulmonary Embolism/complications , Pulmonary Embolism/diagnosis , Pulmonary Embolism/enzymology , Time Factors , Up-Regulation
6.
Circulation ; 131(7): 656-68, 2015 Feb 17.
Article in English | MEDLINE | ID: mdl-25520375

ABSTRACT

BACKGROUND: A limitation of current antiplatelet therapies is their inability to separate thrombotic events from bleeding occurrences. A better understanding of the molecular mechanisms leading to platelet activation is important for the development of improved therapies. Recently, protein tyrosine phosphatases have emerged as critical regulators of platelet function. METHODS AND RESULTS: This is the first report implicating the dual-specificity phosphatase 3 (DUSP3) in platelet signaling and thrombosis. This phosphatase is highly expressed in human and mouse platelets. Platelets from DUSP3-deficient mice displayed a selective impairment of aggregation and granule secretion mediated by the collagen receptor glycoprotein VI and the C-type lectin-like receptor 2. DUSP3-deficient mice were more resistant to collagen- and epinephrine-induced thromboembolism compared with wild-type mice and showed severely impaired thrombus formation on ferric chloride-induced carotid artery injury. Intriguingly, bleeding times were not altered in DUSP3-deficient mice. At the molecular level, DUSP3 deficiency impaired Syk tyrosine phosphorylation, subsequently reducing phosphorylation of phospholipase Cγ2 and calcium fluxes. To investigate DUSP3 function in human platelets, a novel small-molecule inhibitor of DUSP3 was developed. This compound specifically inhibited collagen- and C-type lectin-like receptor 2-induced human platelet aggregation, thereby phenocopying the effect of DUSP3 deficiency in murine cells. CONCLUSIONS: DUSP3 plays a selective and essential role in collagen- and C-type lectin-like receptor 2-mediated platelet activation and thrombus formation in vivo. Inhibition of DUSP3 may prove therapeutic for arterial thrombosis. This is the first time a protein tyrosine phosphatase, implicated in platelet signaling, has been targeted with a small-molecule drug.


Subject(s)
Dual Specificity Phosphatase 3/antagonists & inhibitors , Dual Specificity Phosphatase 3/deficiency , Platelet Activation/physiology , Pulmonary Embolism/enzymology , Animals , Cells, Cultured , Enzyme Inhibitors/pharmacology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Platelet Activation/drug effects , Pulmonary Embolism/blood , Thrombosis/blood , Thrombosis/enzymology
7.
Vasa ; 43(6): 450-8, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25339163

ABSTRACT

BACKGROUND: We evaluated the diagnostic efficacy of tissue plasminogen activator (tPA), using an enzyme-linked immunosorbent assay (ELISA) and compared it with an ELISA D-dimer (VIDAS D-dimer) in acute pulmonary embolism (PE). PATIENTS AND METHODS: We studied 127 consecutive outpatients with clinically suspected PE. The diagnosis of PE was based on a clinical probability pretest for PE and a strict protocol of imaging studies. A plasma sample to measure the levels of tPA and D-dimer was obtained at enrollment. Diagnostic accuracy for tPA and D-dimer was determined by the area under the receiver operating characteristic (ROC) curve. Sensitivity, specificity, predictive values, and the diagnostic utility of tPA with a cutoff of 8.5 ng/mL and D-dimer with a cutoff of 500 ng/mL, were calculated for PE diagnosis. RESULTS: PE was confirmed in 41 patients (32 %). Areas under ROC curves were 0.86 for D-dimer and 0.71 for tPA. The sensitivity/negative predictive value for D-dimer using a cutoff of 500 ng/mL, and tPA using a cutoff of 8.5 ng/mL, were 95 % (95 % CI, 88-100 %)/95 % (95 % CI, 88-100 %) and 95 % (95 % CI, 88-100 %)/94 %), respectively. The diagnostic utility to exclude PE was 28.3 % (95 % CI, 21-37 %) for D-dimer and 24.4 % (95 % CI, 17-33 %) for tPA. CONCLUSIONS: The tPA with a cutoff of 8.5 ng/mL has a high sensitivity and negative predictive value for exclusion of PE, similar to those observed for the VIDAS D-dimer with a cutoff of 500 ng/mL, although the diagnostic utility was slightly higher for the D-dimer.


Subject(s)
Pulmonary Embolism/diagnosis , Tissue Plasminogen Activator/blood , Acute Disease , Adult , Aged , Aged, 80 and over , Area Under Curve , Biomarkers/blood , Diagnostic Imaging , Enzyme-Linked Immunosorbent Assay , Female , Fibrin Fibrinogen Degradation Products/analysis , Humans , Male , Middle Aged , Predictive Value of Tests , Prospective Studies , Pulmonary Embolism/blood , Pulmonary Embolism/enzymology , ROC Curve , Reproducibility of Results
8.
J Thromb Thrombolysis ; 36(3): 352-4, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23277116

ABSTRACT

Heparin-induced thrombocytopenia (HIT) is a rare complication of heparin treatment resulting in a severe acquired thrombophilic condition with an associated mortality of about 10 %. We report the first case of successful urgent liver transplantation (LT) in a patient with end-stage liver disease due to a Budd-Chiari syndrome, portal vein thrombosis and pulmonary embolism due to acquired thrombophilia associated to polycythemia vera carrying JAK2V617F gene mutation and HIT in the acute phase. Lepirudin was used to provide anticoagulation in the LT perioperative period that was performed without haemorrhagic and thrombotic complications despite the donor received heparin during liver explantation.


Subject(s)
Anticoagulants/adverse effects , Budd-Chiari Syndrome , Heparin/adverse effects , Janus Kinase 2/genetics , Liver Transplantation , Mutation, Missense , Polycythemia Vera , Pulmonary Embolism , Thrombocytopenia , Venous Thrombosis , Amino Acid Substitution , Anticoagulants/administration & dosage , Budd-Chiari Syndrome/complications , Budd-Chiari Syndrome/enzymology , Budd-Chiari Syndrome/genetics , Budd-Chiari Syndrome/surgery , Female , Heparin/administration & dosage , Humans , Middle Aged , Polycythemia Vera/complications , Polycythemia Vera/enzymology , Polycythemia Vera/genetics , Polycythemia Vera/surgery , Pulmonary Embolism/complications , Pulmonary Embolism/enzymology , Pulmonary Embolism/genetics , Pulmonary Embolism/surgery , Thrombocytopenia/chemically induced , Thrombocytopenia/enzymology , Thrombocytopenia/surgery , Venous Thrombosis/complications , Venous Thrombosis/enzymology , Venous Thrombosis/genetics , Venous Thrombosis/surgery
9.
Am J Emerg Med ; 30(6): 908-15, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22386346

ABSTRACT

BACKGROUND: Increased γ-glutamyl transferase (GGT) level is associated with increased oxidative stress, all-cause mortality, the development of cardiovascular disease, and metabolic syndrome. However, its role in acute pulmonary embolism (PE) is unknown. In this study, we aimed to investigate the relationship between GGT and early mortality in patients with acute PE. METHODS: A total of 127 consecutive patients with confirmed PE were evaluated. The optimal cutoff value of GGT to predict early mortality was measured as more than 55 IU/L with 94.4% sensitivity and 66.1% specificity. Patients with acute PE were categorized prospectively as having no increased (group I) or increased (group II) GGT based on a cutoff value. RESULTS: Of these 127 patients, 18 patients (14.2%) died during follow-up. Among these 18 patients, 1 (1.4%) patient was in group I, and 17 (30.9%) patients were in group II (P < .001). γ-Glutamyl transferase level on admission, presence of shock, heart rate, oxygen saturation, right ventricular dilatation/hypokinesia, main pulmonary artery involvement, troponin I, alanine aminotransferase, alkaline phosphatase, and creatinine levels were found to have prognostic significance in univariate analysis. In the multivariate Cox proportional hazards model, GGT level on admission (hazard ratio [HR], 1.015; P = .017), presence of shock (HR, 15.124; P = .005), age (HR, 1.107; P = .010), and heart rate (HR, 1.101; P = .032) remained associated with an increased risk of acute PE-related early mortality after the adjustment of other potential confounders. CONCLUSIONS: We have shown that a high GGT level is associated with worse hemodynamic parameters, and it seems that GGT helps risk stratification in patients with acute PE.


Subject(s)
Pulmonary Embolism/enzymology , gamma-Glutamyltransferase/blood , Aged , Chi-Square Distribution , Echocardiography , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Predictive Value of Tests , Proportional Hazards Models , Prospective Studies , Pulmonary Embolism/blood , Pulmonary Embolism/diagnostic imaging , Pulmonary Embolism/mortality , ROC Curve , Sensitivity and Specificity , Statistics, Nonparametric
10.
Biosci Rep ; 42(1)2022 01 28.
Article in English | MEDLINE | ID: mdl-34778900

ABSTRACT

BACKGROUND: Acute pulmonary embolism (APE) is a prevalent reason of cardiovascular morbidity and mortality. Recent studies have underscored the positive effects of microRNAs (miRNAs) on many diseases. The present study aimed to identify the critical miRNA with differential expressions and explore its role in APE. METHODS: The critical miRNA with its target gene was screened by bioinformatics analysis. Their binding relationship was analyzed by TargetScan, Dual-luciferase reporter and RNA pull-down assays. A rat model of APE was established by self-blood coagulum. Human pulmonary artery smooth muscle cells (PASMCs) were exposed to platelet-derived growth factor (PDGF-BB) for excessive proliferation, and transfected with miR-34a-3p mimic. Mean pulmonary arterial pressure (mPAP) of rat was measured, and the pulmonary tissues were used for the pathological observation by Hematoxylin-Eosin (H&E) staining. Cell viability and proliferation were detected by Cell Counting Kit-8 (CCK-8) and EdU assays. The expressions of miR-34a-3p with its target genes (including dual-specificity phosphatase-1 (DUSP1)), neuron-derived orphan receptor-1 (NOR-1) and proliferating cell nuclear antigen (PCNA) were determined by quantitative reverse transcription polymerase chain reaction (RT-qPCR) or/and Western blot. RESULTS: MiR-34a-3p expression was down-regulated in APE patients, which attenuated the increment of mPAP and thickening of the pulmonary arterial walls in APE rats, accompanied with regulation of NOR-1 and PCNA levels. MiR-34a-3p suppressed DUSP1 expression by directly binding to its 3'-untranslated region (UTR), and attenuated cell viability, proliferation, and the expressions of NOR-1 and PCNA in PDGF-BB-induced PASMCs by inhibiting DUSP1 expression. CONCLUSION: Up-regulated miR-34a-3p negatively regulates DUSP1 expression to inhibit PASMC proliferation, which, thus, may act on APE treatment by negatively regulating pulmonary vascular proliferation.


Subject(s)
Cell Proliferation , Dual Specificity Phosphatase 1/metabolism , MicroRNAs/metabolism , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , Pulmonary Embolism/enzymology , Animals , Case-Control Studies , Cells, Cultured , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Disease Models, Animal , Dual Specificity Phosphatase 1/genetics , Gene Expression Regulation, Enzymologic , Male , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , MicroRNAs/genetics , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/metabolism , Pulmonary Artery/enzymology , Pulmonary Artery/pathology , Pulmonary Embolism/genetics , Pulmonary Embolism/pathology , Rats, Sprague-Dawley , Signal Transduction , Vascular Remodeling
11.
Pulm Pharmacol Ther ; 24(4): 407-13, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21281730

ABSTRACT

Pulmonary embolism (PE) causes pulmonary hypertension by mechanical obstruction and constriction of non-obstructed vasculature. We tested if experimental PE impairs pulmonary vascular endothelium-dependent dilation via activation of arginase II. Experimental PE was induced in male Sprague-Dawley rats by infusing 25 µm microspheres in the right jugular vein, producing moderate pulmonary hypertension. Shams received vehicle injection. Pulmonary arterial rings were isolated after 18 h and isometric tensions were determined. Dilations were induced with acetylcholine, calcium ionophore A23187 or nitroglycerin (NTG) in pre-contracted rings (phenylephrine). Protein expression was assessed by Western blot and immunohistochemistry. Arginase activity was inhibited by intravenous infusion of N(w)-hydroxy-nor-l-arginine (nor-NOHA). l-Arginine supplementation was also given. Endothelium-dependent dilation responses were significantly reduced in PE vs. vehicle-treated animals (ACh: 50 ± 9% vs. 93 ± 3%; A23187: 19 ± 7% vs. 85 ± 7%, p < 0.05), while endothelium-independent dilations (NTG) were unchanged. Endothelial nitric oxide synthase (eNOS) protein content was unchanged by PE. Expression of arginase II increased 4.5-fold and immunohistochemistry revealed increased arginase II staining. Nor-NOHA treatment and l-arginine supplementation significantly improved pulmonary artery ring endothelium-dependent dilation in PE (ACh: 58 ± 6% PE, 88 ± 6% PE + nor-NOHA, 84 ± 4% PE + l-arginine). Experimental PE impairs endothelium-dependent pulmonary artery dilation, while endothelium-independent dilation remains unchanged. The data support the conclusion that up-regulation of arginase II protein expression contributes to pulmonary artery endothelial dysfunction in this model of experimental PE.


Subject(s)
Arginase/physiology , Endothelial Cells/physiology , Pulmonary Artery/physiopathology , Pulmonary Embolism/physiopathology , Animals , Arginine/pharmacology , Male , Nitric Oxide Synthase Type III/analysis , Pulmonary Embolism/enzymology , Rats , Rats, Sprague-Dawley , Up-Regulation , Vasodilation
12.
Ter Arkh ; 83(12): 43-8, 2011.
Article in Russian | MEDLINE | ID: mdl-22416444

ABSTRACT

AIM: To study effects of thrombin-activated fibrinolysis inhibitor (TAFI) on efficacy and safety of long-term anti-coagulant treatment in patients with venous thromboembolic complications (VTEC). MATERIAL AND METHODS: A total of 111 patients with a history of an episode of deep vein thrombosis (DVT) and/or pulmonary artery thromboembolism (PATE) entered the study. All the patients received unfractionated or low-molecular heparin for at least 5 days than switch on warfarin (target values of INR 2.0-3.0). Baseline blood levels of TAFI were measured. The patients were followed up for 18 months. Recurrent (DVT/TAFI and hemorrhagic complications (HC) were endpoints. Also, frequency of complete lysis of deep vein thrombi was assessed after 12 months of treatment. RESULTS: A TAFI level varied from 50 to 217% (median 106%, interquartile rage 90-133%). TAFI concentration positively correlated with fibrinogen and thromb size. The patients were divided into two groups depending on TAFI content: group 1 patients had low TAFI (under 25th percentile; < 90%); patients of group 2 had high TAFI (above 25th percentile; > 90%). Group 1 patients were characterized by less stable anticoagulation. This association did not depend on genetic characteristics which determine sensitivity to warfarin (CYP2C9 and VKORC1). Low TAFI was associated with reduced risk of DVT for 18 months and higher probability of complete lysis of the thrombi after 12 months of anticoagulant therapy compared to VTEC patients with high TAFI. No differences were found by TAFI level in patients with HC and without HC, but in HC patients low TAFI was associated with spontaneous hemorrhages and bleeding in therapeutic INR values. CONCLUSION: The results of this pilot study evidence that a TAFI level can be one of the factors influencing efficacy and safety of long-term anticoagulant therapy in patients with VTEC on warfarin treatment.


Subject(s)
Anticoagulants/therapeutic use , Carboxypeptidase B2/blood , Pulmonary Embolism/drug therapy , Venous Thrombosis/drug therapy , Warfarin/therapeutic use , Adolescent , Adult , Aged , Anticoagulants/administration & dosage , Anticoagulants/adverse effects , Dose-Response Relationship, Drug , Female , Hemorrhage/chemically induced , Hemorrhage/enzymology , Humans , Male , Middle Aged , Multivariate Analysis , Pilot Projects , Prospective Studies , Pulmonary Embolism/blood , Pulmonary Embolism/enzymology , Regression Analysis , Risk , Time Factors , Venous Thrombosis/blood , Venous Thrombosis/enzymology , Warfarin/administration & dosage , Warfarin/adverse effects , Young Adult
13.
Am J Physiol Lung Cell Mol Physiol ; 298(3): L297-303, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19915157

ABSTRACT

Acute pulmonary embolism (PE) is a life-threatening disease, and several vasoconstrictors, including endothelin-1 (ET-1), play a key role in vasoconstriction and hypoxemia during the development of PE. Rho kinase is activated by various vasoconstrictors resulting in vascular contraction and remodeling. Recent evidence has revealed an important role of Rho kinase in the pathogenesis of systemic and pulmonary vascular diseases. However, contribution of Rho kinase in PE remains unclear. We thus investigated the role of Rho kinase in the PE rat model induced by intrajugular administration of polystyrene microspheres (mean diameter, 26 microm). At 6 h following the administration of microspheres (1.5 ml/kg), right ventricular systolic pressure (RVSP) was higher in the PE than in the control rats (15.8 +/- 1.6 vs. 32.9 +/- 7.5 mmHg). Arterial oxygen tension was lower (92.3 +/- 12.5 vs. 66.0 +/- 17.7 Torr), and alveolar-arterial difference in oxygen partial pressure was higher (3.9 +/- 3.8 vs. 36.5 +/- 26.9 Torr) in the PE rats. Western blotting analysis revealed upregulation and downregulation in expression of vascular cell adhesion molecule-1 and endothelial nitric oxide synthase in lungs from the PE rats, respectively, and radioimmunoassay demonstrated an increase in plasma ET-1 levels. Lung Rho kinase alpha expression was greater in the PE rats. At 5 h following administration of microspheres (0.75 ml/kg), intravenous Rho kinase inhibitors HA1077 and Y27632 (3 mg/kg each) attenuated elevation of RVSP (22.0 +/- 3.7, 17.1 +/- 3.2, 14.3 +/- 2.6 mmHg, PE, PE+HA1077, PE+Y27632) and the severity of hypoxemia (66.3 +/- 16.2, 94.9 +/- 23.0, 89.1 +/- 8.5 Torr, PE, PE+HA1077, PE+Y27632) in the PE rats. These results suggest that pulmonary endothelial dysfunction and activation of Rho kinase may contribute to the potentiation of vasoconstriction and hypoxemia in the PE rats.


Subject(s)
Microspheres , Pulmonary Embolism/enzymology , Pulmonary Embolism/etiology , rho-Associated Kinases/metabolism , Acute Disease , Animals , Blood Gas Analysis , Blood Pressure/drug effects , Endothelin-1/blood , Hemodynamics/drug effects , Injections, Intravenous , Lung/drug effects , Lung/enzymology , Lung/pathology , Male , Nitric Oxide Synthase Type III/metabolism , Polystyrenes , Protein Kinase Inhibitors/pharmacology , Pulmonary Artery/drug effects , Pulmonary Artery/pathology , Pulmonary Embolism/blood , Pulmonary Embolism/chemically induced , Rats , Rats, Sprague-Dawley , Vascular Cell Adhesion Molecule-1/metabolism , rho-Associated Kinases/antagonists & inhibitors
14.
J Appl Genet ; 50(4): 399-403, 2009.
Article in English | MEDLINE | ID: mdl-19875892

ABSTRACT

A daily dose of vitamin K antagonists (VKAs) may vary and its range depends on various interrelated factors. Low responsiveness to VKA (defined as a failure to achieve a target international normalized ratio [INR]) is associated with polymorphisms of the vitamin K epoxide reductase-oxidase complex gene (VKORC1). A highly prevalent promoter single-nucleotide polymorphism (VKORC1-1639 G>A, rs17878363) impairs VKORC1 expression and determines the interindividual variability of the target INR. We studied 57 patients receiving oral anticoagulation, including 50 subjects treated with acenocoumarol (mean dose: 5.7+/-2.3 mg/day) and 7 treated with warfarin (mean dose: 9.6+/-4.2 mg/day). The indications for the use of oral anticoagulant therapy were as follows: deep-vein thrombosis (N = 23); pulmonary embolism (N = 20); arterial thrombosis (N = 5); stroke (N = 4); atrial fibrillation with transient ischemic attacks (N = 2), and history of multiple thromboembolic events (N = 3). Identification of the VKORC1 genomic variation was performed using DNA sequencing methods. The prevalence of the mutated allele (VKORC1 -1639A) was 41%. The VKORC1 -1639G allele carriers required a higher daily dose of acenocoumarol (5.9+/-1.9 mg) than the noncarriers (4.1+/-3.3 mg; P < 0.001). All of 5 low responders (who failed to achieve a target INR using standard dose requirements of VKAs) were homozygous for the 1639G allele. Low responders did not differ from good responders with respect to age, gender, and body mass index. Our findings suggest the potential benefits from pharmacogenetic testing, and provide evidence that the VKORC1 -1639 G>A gene polymorphism may explain at least in part the low responsiveness to acenocoumarol.


Subject(s)
Anticoagulants/administration & dosage , Mixed Function Oxygenases/genetics , Polymorphism, Single Nucleotide , Vitamin K/antagonists & inhibitors , Acenocoumarol/administration & dosage , Adult , Alleles , Atrial Fibrillation/drug therapy , Atrial Fibrillation/enzymology , Atrial Fibrillation/genetics , Base Sequence , DNA Primers/genetics , Dose-Response Relationship, Drug , Drug Resistance/genetics , Female , Gene Frequency , Humans , International Normalized Ratio , Male , Middle Aged , Poland , Promoter Regions, Genetic , Pulmonary Embolism/drug therapy , Pulmonary Embolism/enzymology , Pulmonary Embolism/genetics , Venous Thrombosis/drug therapy , Venous Thrombosis/enzymology , Venous Thrombosis/genetics , Vitamin K Epoxide Reductases , Warfarin/administration & dosage
15.
Eur J Prev Cardiol ; 26(2): 120-134, 2019 01.
Article in English | MEDLINE | ID: mdl-30466296

ABSTRACT

We performed this meta-analysis to better assess the relationship between methylenetetrahydrofolate reductase gene ( MTHFR) polymorphisms and the risk of venous thromboembolism. Eligible studies were searched in PubMed, Medline, Embase, and Web of Science. Odds ratios with 95% confidence intervals were used to assess associations of MTHFR polymorphisms with venous thromboembolism. A total of 99 genetic association studies were enrolled for analyses. Although no positive results were detected in overall analyses for the rs1801131 polymorphism. Further subgroup analyses according to ethnicity of participants and type of disease revealed that the rs1801131 polymorphism was significantly correlated with the risk of pulmonary embolism. For the rs1801133 polymorphism, significant association with the risk of venous thromboembolism was found in the dominant, recessive, and allele models. Further subgroup analyses according to ethnicity of participants revealed that the rs1801133 polymorphism was significantly associated with the risk of venous thromboembolism in Caucasians, East Asians, and West Asians. When we stratified available data according to type of disease, we found that the rs1801133 polymorphism was also significantly correlated with the risk of deep vein thrombosis and pulmonary embolism. In conclusion, our findings indicate that the MTHFR rs1801133 polymorphism may serve as a potential biological marker for venous thromboembolism in Caucasians, East Asians, and West Asians. Moreover, the MTHFR rs1801133 polymorphism may be implicated in the development of deep vein thrombosis and pulmonary embolism, while the MTHFR rs1801131 polymorphism may contribute to the development of pulmonary embolism.


Subject(s)
Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Polymorphism, Single Nucleotide , Pulmonary Embolism/genetics , Venous Thromboembolism/genetics , Venous Thrombosis/genetics , Case-Control Studies , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Phenotype , Pulmonary Embolism/diagnosis , Pulmonary Embolism/enzymology , Pulmonary Embolism/ethnology , Risk Assessment , Risk Factors , Venous Thromboembolism/diagnosis , Venous Thromboembolism/enzymology , Venous Thromboembolism/ethnology , Venous Thrombosis/diagnosis , Venous Thrombosis/enzymology , Venous Thrombosis/ethnology
16.
Crit Care Med ; 36(2): 565-71, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18216605

ABSTRACT

OBJECTIVES: Fat embolism syndrome is a clinical issue in subjects with long-bone fracture. It may lead to acute lung injury. The mechanisms and therapeutic regimen remain unclear. The present study was designed to investigate the pathologic and biochemical changes after fat embolization in isolated rat lungs, and to test the effects of posttreatment with N-acetylcysteine (NAC). DESIGN: Prospective, randomized, controlled animal study. SETTING: University research laboratory. SUBJECTS: A total of 36 perfused lungs isolated from Sprague-Dawley rats. INTERVENTIONS: The isolated lungs were randomly assigned to receive physiologic saline solution (vehicle group), fat embolism (FE group), or FE with NAC posttreatment (FE + NAC group). There were 12 isolated lungs in each group. FE was produced by introduction of corn oil micelles. NAC at a dose 150 mg/kg was given 10 mins after FE. MEASUREMENTS AND MAIN RESULTS: The extent of acute lung injury was evaluated by lung weight change, protein concentration in bronchoalveolar lavage, and exhaled nitric oxide. We also measured the pulmonary arterial pressure and capillary filtration coefficient and determined the nitrate/nitrite, methylguanidine, tumor necrosis factor-alpha, and interleukin-1beta in lung perfusate. Histopathologic changes of the lung were examined and quantified. The levels of neutrophil elastase and myeloperoxidase were determined. The expression of inducible nitric oxide synthase was detected. FE caused acute lung injury as evidenced by the lung weight changes, increases in exhaled nitric oxide and protein concentration in bronchoalveolar lavage, pulmonary hypertension, increased capillary filtration coefficient, and lung pathology. The insult also increased nitrate/nitrite, methylguanidine, tumor necrosis factor-alpha, and interleukin-1beta in lung perfusate, increased neutrophil elastase and myeloperoxidase levels, and upregulated inducible nitric oxide synthase expression. Posttreatment with NAC abrogated these changes induced by FE. CONCLUSION: FE caused acute lung injury and associated biochemical changes. Posttreatment with NAC was effective to alleviate the pathologic and biochemical changes caused by FE.


Subject(s)
Acetylcysteine/therapeutic use , Embolism, Fat/drug therapy , Expectorants/therapeutic use , Pulmonary Embolism/drug therapy , Respiratory Distress Syndrome/prevention & control , Animals , Embolism, Fat/complications , Embolism, Fat/enzymology , Leukocyte Elastase/metabolism , Male , Nitric Oxide Synthase Type II/metabolism , Peroxidase/metabolism , Pulmonary Embolism/complications , Pulmonary Embolism/enzymology , Rats , Rats, Sprague-Dawley , Respiratory Distress Syndrome/enzymology , Respiratory Distress Syndrome/etiology
17.
Pathobiology ; 75(5): 295-305, 2008.
Article in English | MEDLINE | ID: mdl-18931532

ABSTRACT

We previously reported that during total knee arthroplasty in rheumatoid arthritis (RA) patients, the use of tourniquet might promote local release of neutrophil elastase (NE) from neutrophils, which may contribute to the development of pulmonary thromboembolism (PTE) and tissue injury. The aim of this study was to develop PTE by the use of NE in a mouse model of collagen-induced arthritis (CIA) and investigate the relationship between thrombus and endothelial cells as well as the effect of recombinant human soluble thrombomodulin (rhs-TM) in reducing the risk of PTE. Male DBA/1J mice were injected intracutaneously at several sites with an emulsion containing bovine collagen and later a booster shot to produce CIA mice. Subsequently, NE was injected intravenously 2 times a day for 3 days and after a further 4 days, mice were sacrificed. A group of mice received rhs-TM injections prior to NE injections. We divided the mice into four groups of normal, CIA control, CIA + NE, and CIA + rhs-TM + NE mice and evaluated thrombus formation status. All CIA + NE mice developed PTE. In contrast, no thrombosis was found in normal control, CIA control and CIA + rhs-TM + NE mice. Plasma thrombin level, fibrinogen expression and neutrophil count were increased in CIA + NE mice. Double staining for anticoagulant TM and procoagulant von Willebrand factor (vWF) in pulmonary endothelial cells in normal mice showed a TM-dominant expression while in both CIA control and CIA + NE mice a vWF-dominant expression compatible with coagulant status was observed. Injection of rhs-TM into CIA + NE mice resulted in a phenotypic conversion of endothelial cells from vWF-dominant to TM-dominant expression and a reduction in fibrinogen deposition. These findings demonstrate that by repeated use of NE in CIA mice, it is feasible to produce PTE and to study its pathogenesis and that rhs-TM reduces the risk of PTE. We suggest that in surgical operations of upper and lower extremities in RA patients, the use of a tourniquet should be avoided as it may trigger NE release.


Subject(s)
Arthritis, Experimental/enzymology , Leukocyte Elastase/metabolism , Pulmonary Embolism/prevention & control , Recombinant Proteins/therapeutic use , Thrombomodulin/therapeutic use , Animals , Arthritis, Experimental/surgery , Cattle , Endothelial Cells/metabolism , Endothelial Cells/pathology , Humans , Injections, Intravenous , Leukocyte Elastase/toxicity , Lung/metabolism , Lung/pathology , Male , Mice , Mice, Inbred DBA , Pulmonary Embolism/enzymology , Pulmonary Embolism/etiology , Recombinant Proteins/administration & dosage , Thrombomodulin/administration & dosage , Tourniquets/adverse effects , von Willebrand Factor/metabolism
18.
Clin Appl Thromb Hemost ; 14(3): 365-8, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18586684

ABSTRACT

Pulmonary thromboembolism is a life-threatening condition resulting mostly from lower extremity deep-vein or pelvic-vein thrombosis. A 46-year-old woman was admitted to hospital with pain on the right side of the chest and hemoptysis. On laboratory analysis, D-dimer level was elevated. Computed tomographic pulmonary angiography revealed intravascular filling defects due to thrombi in right lower lobe pulmonary segmental arteries. Screening for thrombophilic states was normal except for heterozygous mutations of both prothrombin and methylene tetrahydrofolate reductase (MTHFR 677) genes. Homocysteine level was high, and vitamin B12 level and serum ferritin level were reduced. Serum antiparietal antibody was positive, and therefore, pernicious anemia was diagnosed along with iron-deficiency anemia. After the diagnoses were established, enoxaparin followed by warfarin was started in addition to oral vitamin B12, pyridoxine, thiamine, folic acid, and ferroglycine sulfate supplementation. At the end of 8 weeks of the replacement therapy, vitamin B12, folate, and homocysteine levels and red cell volume were found to be normal, with complete resolution of the thrombus confirmed by repeat computed tomographic pulmonary angiography. We conclude that hyperhomocysteinemia due to vitamin B12 deficiency associated with pernicious anemia might have decreased the threshold for thrombosis. In addition, the presence of heterozygous prothrombin and methylene tetrahydrofolate reductase mutations might serve as synergistic cofactors triggering pulmonary thromboembolism.


Subject(s)
Anemia, Pernicious/complications , Hyperhomocysteinemia/etiology , Pulmonary Embolism/etiology , Anemia, Pernicious/blood , Anemia, Pernicious/enzymology , Anemia, Pernicious/genetics , Female , Heterozygote , Humans , Hyperhomocysteinemia/blood , Hyperhomocysteinemia/complications , Hyperhomocysteinemia/genetics , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Middle Aged , Mutation , Prothrombin/genetics , Pulmonary Embolism/blood , Pulmonary Embolism/enzymology , Pulmonary Embolism/genetics
19.
Orv Hetil ; 149(20): 935-48, 2008 May 18.
Article in Hungarian | MEDLINE | ID: mdl-18467263

ABSTRACT

UNLABELLED: Acute pulmonary embolism is the third most common cause of cardiovascular mortality. Thrombolytic treatment of massive pulmonary embolism can be complicated with haemorrhage, re-thrombosis and oxidative stress. AIMS: The purpose of this study was to evaluate the changes in platelet aggregation, haemostatic, leukocyte function parameters and oxidative stress in patients with acute pulmonary embolism treated with thrombolytics. METHODS: Fifteen patients undergoing thrombolysis with ultra-high dose streptokinase ( n = 8), or alteplase ( n = 7) treatment were studied. Arterial blood samples were taken before (baseline) and after thrombolysis between the 4th and 24th hour at every four hours, on the second day twice a day and daily on the 3rd, 4th, 5th and 30th day. Platelet aggregation was examined as spontaneous and induced aggregation with adrenaline, collagen and adenosine diphosphate. D-dimer and fibrinogen were measured 8 hourly on the first day and later at the same time intervals as above. To analyse oxidative stress, blood samples were collected prior to thrombolysis, and then 8 hours, 1, 3, 5 and 30 days after treatment. Malondialdehyde, reduced glutathion, plasma sulphydryl groups levels, superoxide dismutase and myeloperoxidase enzyme activities were measured in plasma or whole blood for monitoring of the oxidative stress markers. Production of reactive oxygen species in whole blood was measured by luminol dependent chemiluminescence. Flow cytometry was used to determine CD11a, CD18, and CD97 surface antigen expression on leukocytes. RESULTS: In streptokinase group, adrenaline induced platelet aggregation decreased at the 4th and 8th hour ( p < 0.03) and was significantly lower than in the alteplase group at the 36th hour and on the 3rd day. Platelet aggregation induced by adenosine diphosphate was lower at the 4th hour than at baseline in streptokinase group ( p < 0.05). Collagen induced platelet aggregation was lower at the 4th and 8th hour than at baseline ( p < 0.05) in streptokinase group. Compared to baseline, fibrinogen levels decreased in both groups after thrombolysis. D-dimer levels elevated significantly in both therapeutic groups at the 8th hour. Spontaneous platelet aggregation was not detectable and major bleeding or re-embolism was not documented. The elevated malondialdehyde, reactive oxygen species and myeloperoxidase, decreased reduced glutathion and plasma sulphydryl levels indicated the presence of oxidative stress in patients with pulmonary embolism. Malondialdehyde significantly increased, reduced glutathion significantly decreased following thrombolysis. Reactive oxygen species production peaked on the 3rd and 5th days. Thrombolysis was accompanied by significant decrease in granulocyte and monocyte CD11a and CD18 as well as in granulocyte CD97 expression ( p < 0.05). CONCLUSION: Massive/submassive pulmonary embolism and thrombolysis injures inducible platelet aggregation. The changes in fibrinogen levels correlate significantly with the improvement of pulmonary perfusion which shows the effect of thrombolysis. Pulmonary embolism induced oxidative stress was detected on patients before thrombolysis. Thrombolytic treatment of pulmonary embolism augmented the increase of oxidative stress response and leukocyte activation following reperfusion, and these parameters normalised only on the 30th day.


Subject(s)
Oxidative Stress/drug effects , Platelet Aggregation/drug effects , Pulmonary Embolism/blood , Pulmonary Embolism/drug therapy , Streptokinase/administration & dosage , Thrombolytic Therapy , Tissue Plasminogen Activator/administration & dosage , Acute Disease , Adult , Aged , Antigens, CD/blood , Biomarkers/blood , CD11a Antigen/blood , CD18 Antigens/blood , Female , Fibrin Fibrinogen Degradation Products/metabolism , Fibrinogen/metabolism , Glutathione/blood , Hemoglobins/metabolism , Hemostasis/drug effects , Humans , Leukocytes/drug effects , Male , Malondialdehyde/blood , Membrane Glycoproteins/blood , Middle Aged , Patient Selection , Peroxidase/blood , Pulmonary Embolism/enzymology , Reactive Oxygen Species/blood , Receptors, G-Protein-Coupled , Superoxide Dismutase/blood , Thrombolytic Therapy/methods , Time Factors
20.
J Thromb Haemost ; 16(6): 1211-1225, 2018 06.
Article in English | MEDLINE | ID: mdl-29575487

ABSTRACT

Essentials Phosphoinositide 3-kinase and MAPK pathways crosstalk via PDK1. PDK1 is required for adenosine diphosphate-induced platelet activation and thromboxane generation. PDK1 regulates RAF proto-oncogene Ser/Thr kinase (Raf1) activation in the MAPK pathway. Genetic ablation of PDK1 protects against platelet-dependent thrombosis in vivo. SUMMARY: Background Platelets are dynamic effector cells with functions that span hemostatic, thrombotic and inflammatory continua. Phosphoinositide-dependent protein kinase 1 (PDK1) regulates protease-activated receptor 4-induced platelet activation and thrombus formation through glycogen synthase kinase3ß. However, whether PDK1 also signals through the ADP receptor and its functional importance in vivo remain unknown. Objective To establish the mechanism of PDK1 in ADP-induced platelet activation and thrombosis. Methods We assessed the role of PDK1 on 2MeSADP-induced platelet activation by measuring aggregation, thromboxane generation and phosphorylation events in the presence of BX-795, which inhibits PDK1, or by using platelet-specific PDK1 knockout mice and performing western blot analysis. PDK1 function in thrombus formation was assessed with an in vivo pulmonary embolism model. Results PDK1 inhibition with BX-795 reduced 2-methylthio-ADP (2MeSADP)-induced aggregation of human and murine platelets by abolishing thromboxane generation. Similar results were observed in pdk1-/- mice. PDK1 was also necessary for the phosphorylation of mitogen-activated protein kinase kinase 1/2 (MEK1/2), extracellular signal-regulated kinase 1/2, and cytosolic phospholipase A2, indicating that PDK1 regulates an upstream kinase in the mitogen-activated protein kinase (MAPK) pathway. We next determined that this upstream kinase is Raf-1, a serine/threonine kinase that is necessary for the phosphorylation of MEK1/2, as pharmacological inhibition and genetic ablation of PDK1 were sufficient to prevent Raf1 phosphorylation. Furthermore, in vivo inhibition or genetic ablation of PDK1 protected mice from collagen/epinephrine-induced pulmonary embolism. Conclusion PDK1 governs thromboxane generation and thrombosis in platelets that are stimulated with 2MeSADP by regulating activation of the MAPK pathway.


Subject(s)
3-Phosphoinositide-Dependent Protein Kinases/metabolism , Blood Platelets/enzymology , Mitogen-Activated Protein Kinases/blood , Platelet Aggregation/drug effects , Proto-Oncogene Proteins c-raf/blood , Pulmonary Embolism/enzymology , Thrombosis/enzymology , Thromboxanes/blood , 3-Phosphoinositide-Dependent Protein Kinases/antagonists & inhibitors , 3-Phosphoinositide-Dependent Protein Kinases/blood , 3-Phosphoinositide-Dependent Protein Kinases/deficiency , 3-Phosphoinositide-Dependent Protein Kinases/genetics , Animals , Blood Platelets/drug effects , Disease Models, Animal , Humans , Mice, Knockout , Phosphorylation , Platelet Aggregation Inhibitors/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Mas , Pulmonary Embolism/blood , Pulmonary Embolism/genetics , Pulmonary Embolism/prevention & control , Pyrimidines/pharmacology , Signal Transduction , Thiophenes/pharmacology , Thrombosis/blood , Thrombosis/genetics , Thrombosis/prevention & control
SELECTION OF CITATIONS
SEARCH DETAIL