Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 130
Filter
1.
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
2.
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
3.
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
4.
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
5.
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
6.
Vascul Pharmacol ; 109: 45-55, 2018 10.
Article in English | MEDLINE | ID: mdl-29890296

ABSTRACT

BACKGROUND & PURPOSE: Ginseng (Panax ginseng C.A. Mayer) contains saponin fractions called ginsenosides, which are thought to be the main components responsible for its various pharmacological activities. Ginsenosides have cardioprotective and antiplatelet effects. In the present study, we evaluated the effects of ginsenoside Rp3 (G-Rp3) on platelet function. METHODS: The in vitro effects of G-Rp3 were evaluated on agonist-induced human and rat platelet aggregation, while [Ca2+]i mobilization, granule secretion, integrin αIIbß3 activation, and clot retraction were assessed in rat platelets. Its effects on vasodilator-stimulated phosphoprotein (VASP) expression, phosphorylation of MAPK signaling molecules, and PI3K/Akt activation were also studied. Moreover, the tyrosine phosphorylation of components of the P2Y12 receptor downstream signaling pathway was also examined. The in vivo effects of G-Rp3 were studied using an acute pulmonary thromboembolism model and lung histopathology. KEY RESULTS: G-Rp3 significantly inhibited collagen, ADP, and thrombin-induced platelet aggregation. G-Rp3 elevated cAMP levels and VASP phosphorylation and suppressed agonist-induced [Ca2+]i mobilization, ATP release, and P-selectin expression along with fibrinogen binding to integrin αIIbß3, fibronectin adhesion, and clot retraction. G-Rp3 also attenuated the phosphorylation of MAPK, Src, and PLCγ2 as well as PI3K/Akt activation. Furthermore, it inhibited tyrosine phosphorylation of the Src family kinases (Src, Fyn, and Lyn) and PLCγ2 and protected mice from thrombosis. CONCLUSION AND IMPLICATION: G-Rp3 modulates agonist-induced platelet activation and thrombus formation by inhibiting granule secretion, integrin αIIbß3 activation, MAPK signaling, and Src, PLCγ2, and PI3K/Akt activation, and VASP stimulation. Our data suggest that G-Rp3 has therapeutic potential as a treatment for platelet-related cardiovascular disorders.


Subject(s)
Blood Platelets/drug effects , Fibrinolytic Agents/pharmacology , Ginsenosides/pharmacology , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinases/metabolism , Platelet Activation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Pulmonary Embolism/prevention & control , Second Messenger Systems/drug effects , Animals , Blood Platelets/enzymology , Calcium Signaling/drug effects , Cell Adhesion Molecules/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Humans , Male , Mice, Inbred C57BL , Microfilament Proteins/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Phospholipase C gamma/metabolism , Phosphoproteins/metabolism , Phosphorylation , Platelet Aggregation/drug effects , Platelet Glycoprotein GPIIb-IIIa Complex/drug effects , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Pulmonary Embolism/blood , Pulmonary Embolism/enzymology , Rats, Sprague-Dawley , Time Factors , src-Family Kinases/metabolism
7.
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
8.
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
9.
J Thromb Haemost ; 16(2): 352-363, 2018 02.
Article in English | MEDLINE | ID: mdl-29045015

ABSTRACT

Essentials Signaling by Gas6 through Tyro3/Axl/Mer receptors is essential for stable platelet aggregation. UNC2025 is a small molecule inhibitor of the Mer tyrosine kinase. UNC2025 decreases platelet activation in vitro and thrombus formation in vivo. UNC2025's anti-platelet effect is synergistic with inhibition of the ADP receptor, P2Y12 . SUMMARY: Background Growth arrest-specific protein 6 signals through the TAM (TYRO-3-AXL-MERTK) receptor family, mediating platelet activation and thrombus formation via activation of the aggregate-stabilizing αIIb ß3 integrin. Objective To describe the antithrombotic effects mediated by UNC2025, a small-molecule MERTK tyrosine kinase inhibitor. Methods MERTK phosphorylation and downstream signaling were assessed by immunoblotting. Light transmission aggregometry, flow cytometry and microfluidic analysis were used to evaluate the impact of MERTK inhibition on platelet activation and stability of aggregates in vitro. The effects of MERTK inhibition on arterial and venous thrombosis, platelet accumulation at microvascular injury sites and tail bleeding times were determined with murine models. The effects of combined treatment with ADP-P2Y1&12 pathway antagonists and UNC2025 were also evaluated. Results and Conclusions Treatment with UNC2025 inhibited MERTK phosphorylation and downstream activation of AKT and SRC, decreased platelet activation, and protected animals from pulmonary embolism and arterial thrombosis without increasing bleeding times. The antiplatelet effect of UNC2025 was enhanced in combination with ADP-P2Y1&12 pathway antagonists, and a greater than additive effect was observed when these two agents with different mechanisms of inhibition were coadministered. TAM kinase signaling represents a potential therapeutic target, as inhibition of this axis, especially in combination with ADP-P2Y pathway antagonism, mediates decreased platelet activation, aggregate stability, and thrombus formation, with less hemorrhagic potential than current treatment strategies. The data presented here also demonstrate antithrombotic activity mediated by UNC2025, a novel translational agent, and support the development of TAM kinase inhibitors for clinical applications.


Subject(s)
Adenine/analogs & derivatives , Blood Platelets/drug effects , Piperazines/pharmacology , Platelet Activation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Protein Kinase Inhibitors/pharmacology , Pulmonary Embolism/prevention & control , Thrombosis/prevention & control , c-Mer Tyrosine Kinase/antagonists & inhibitors , Adenine/pharmacokinetics , Adenine/pharmacology , Animals , Blood Platelets/enzymology , Disease Models, Animal , Drug Synergism , Drug Therapy, Combination , Female , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Male , Mice, Inbred C57BL , Phosphorylation , Piperazines/pharmacokinetics , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/pharmacokinetics , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Protein Kinase Inhibitors/pharmacokinetics , Proto-Oncogene Proteins/metabolism , Pulmonary Embolism/blood , Pulmonary Embolism/enzymology , Purinergic P2Y Receptor Antagonists/pharmacology , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction/drug effects , Thrombosis/blood , Thrombosis/enzymology , c-Mer Tyrosine Kinase/metabolism , Axl Receptor Tyrosine Kinase
10.
Eur Rev Med Pharmacol Sci ; 21(4): 827-836, 2017 02.
Article in English | MEDLINE | ID: mdl-28272699

ABSTRACT

OBJECTIVE: This study sought to explore endothelial nitric oxide synthase (eNOS) expression in acute pulmonary thromboembolism (APE). MATERIALS AND METHODS: eNOS expression in lung tissue and bone marrow-derived endothelial progenitor cells (BM-EPCs) from APE mouse models was assessed by immunohistochemistry and real-time PCR. A gene expression profile meta-analysis was performed on human venous thromboembolism (VTE) whole blood samples recorded in the Gene Expression Omnibus (GEO) repository. Significantly expressed genes were determined from the microarray data by unsupervised clustering and supervised classification. Selected sample data with significantly expressed genes were further analyzed by principal component analysis (PCA), followed by Bayesian probit regression. Key discriminate genes were further grouped and annotated using functional annotations and gene enrichments using the online Database for Annotation, Visualization and Integrated Discovery (DAVID) software (v. 6.7). RESULTS: While eNOS expression was significantly higher, serum nitric oxide levels were significantly lower in APE mice (20.42 ± 2.15 µM) compared to controls (53.50 ± 5.69 µM, p<0.001). eNOS mRNA and protein levels were significantly upregulated in BM-EPCs from APE mice. GEO repository data reported 3,397 upregulated and 4,173 downregulated genes (including eNOS) in VTE patients. In this regression analysis, the significant principal component PC1 and PC2 (p<0.05) were useful in distinguishing the VTE classification. The coefficient value of eNOS was -0.47707 in PC1 and -0.08429 in PC2, which did have some proportions on these significantly discriminated components but did not contribute significantly to the VTE classification. Functional enrichment in terms of acetylation and phosphoproteins were high. CONCLUSIONS: Our findings, therefore, suggest that expression of eNOS is significantly altered in APE and may be a potential peripheral blood biomarker. Modulation of eNOS expression may be used for APE treatment.


Subject(s)
Nitric Oxide Synthase Type III/metabolism , Pulmonary Embolism/enzymology , Animals , Bayes Theorem , Computational Biology , Humans , Mice , Nitric Oxide/blood , Nitric Oxide Synthase Type III/genetics , Oligonucleotide Array Sequence Analysis , Pulmonary Embolism/genetics , Stem Cells/metabolism , Transcriptome
11.
Naunyn Schmiedebergs Arch Pharmacol ; 389(11): 1159-1169, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27449068

ABSTRACT

Acute pulmonary embolism (APE) has a very high mortality rate, especially at cardiac arrest and even after the return of spontaneous circulation (ROSC). This study investigated the protective effect of the angiotensin-converting enzyme (ACE) inhibitor captopril on postresuscitation hemodynamics, in a porcine model of cardiac arrest established by APE. Twenty-nine Beijing Landrace pigs were infused with an autologous thrombus leading to cardiac arrest and subjected to standard cardiopulmonary resuscitation and thrombolysis. Ten resuscitated pigs were randomly and equally apportioned to receive either captopril (22.22 mg/kg) infusion or the same volume saline, 30 min after ROSC. Hemodynamic changes and ACE-Ang II-angiotensin II type 1 receptor (AT1R) and ACE2/Ang-(1-7)/Mas receptor axis levels were determined. APE was associated with a decline in mean arterial pressure and a dramatic increase in pulmonary artery pressure and mean right ventricular pressure. After ROSC, captopril infusion was associated with significantly lower mean right ventricular pressure and systemic and pulmonary vascular resistance, faster heart rate, and higher Ang-(1-7) levels, ACE2/ACE, and Ang-(1-7)/Ang II, compared with the saline infusion. The ACE2/Ang-(1-7)/Mas pathway correlated negatively with external vascular lung water and pulmonary vascular permeability and positively with the right cardiac index. In conclusion, in a pig model of APE leading to cardiac arrest, captopril infusion was associated with less mean right ventricular pressure overload after resuscitation, compared with saline infusion. The reduction in systemic and pulmonary vascular resistance associated with captopril may be by inhibiting the ACE-Ang II-AT1R axis and activating the ACE2/Ang-(1-7)/Mas axis.


Subject(s)
Angiotensin I/metabolism , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Captopril/pharmacology , Cardiopulmonary Resuscitation , Heart Arrest/therapy , Hemodynamics/drug effects , Peptide Fragments/metabolism , Peptidyl-Dipeptidase A/metabolism , Proto-Oncogene Proteins/metabolism , Pulmonary Embolism/therapy , Receptors, G-Protein-Coupled/metabolism , Angiotensin-Converting Enzyme 2 , Animals , Arterial Pressure/drug effects , Biomarkers/blood , Capillary Permeability/drug effects , Disease Models, Animal , Enzyme Activation , Female , Heart Arrest/blood , Heart Arrest/enzymology , Heart Arrest/physiopathology , Male , Proto-Oncogene Mas , Pulmonary Artery/drug effects , Pulmonary Artery/physiopathology , Pulmonary Edema/enzymology , Pulmonary Edema/physiopathology , Pulmonary Edema/prevention & control , Pulmonary Embolism/blood , Pulmonary Embolism/enzymology , Pulmonary Embolism/physiopathology , Renin-Angiotensin System/drug effects , Signal Transduction/drug effects , Sus scrofa , Thrombolytic Therapy , Time Factors , Vascular Resistance/drug effects , Ventricular Function, Right/drug effects , Ventricular Pressure/drug effects
12.
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
13.
Sao Paulo Med J ; 133(6): 488-94, 2015.
Article in English | MEDLINE | ID: mdl-26648276

ABSTRACT

CONTEXT AND OBJECTIVE: The location of embolism is associated with clinical findings and disease severity in cases of acute pulmonary embolism. The level of gamma-glutamyl transferase increases under oxidative stress-related conditions. In this study, we investigated whether gamma-glutamyl transferase levels could predict the location of pulmonary embolism. DESIGN AND SETTING: Hospital-based cross-sectional study at Cumhuriyet University, Sivas, Turkey. METHODS: 120 patients who were diagnosed with acute pulmonary embolism through computed tomography-assisted pulmonary angiography were evaluated. They were divided into two main groups (proximally and distally located), and subsequently into subgroups according to thrombus localization as follows: first group (thrombus in main pulmonary artery; n = 9); second group (thrombus in main pulmonary artery branches; n = 71); third group (thrombus in pulmonary artery segmental branches; n = 34); and fourth group (thrombus in pulmonary artery subsegmental branches; n = 8). RESULTS: Gamma-glutamyl transferase levels on admission, heart rate, oxygen saturation, right ventricular dilatation/hypokinesia, pulmonary artery systolic pressure and cardiopulmonary resuscitation requirement showed prognostic significance in univariate analysis. The multivariate logistic regression model showed that gamma-glutamyl transferase level on admission (odds ratio, OR = 1.044; 95% confidence interval, CI: 1.011-1.079; P = 0.009) and pulmonary artery systolic pressure (OR = 1.063; 95% CI: 1.005-1.124; P = 0.033) remained independently associated with proximally localized thrombus in pulmonary artery. CONCLUSIONS: The findings revealed a significant association between increased existing embolism load in the pulmonary artery and increased serum gamma-glutamyl transferase levels.


Subject(s)
Pulmonary Embolism/enzymology , gamma-Glutamyltransferase/blood , Acute Disease , Aged , Aged, 80 and over , Biomarkers/blood , Coronary Angiography , Cross-Sectional Studies , Female , Humans , Male , Middle Aged , Prognosis , Pulmonary Artery/pathology , Pulmonary Embolism/blood , Pulmonary Embolism/pathology , ROC Curve , Reference Values , Severity of Illness Index , Statistics, Nonparametric , Tomography, X-Ray Computed
14.
São Paulo med. j ; 133(6): 488-494, Nov.-Dec. 2015. tab, graf
Article in English | LILACS | ID: lil-770152

ABSTRACT

ABSTRACT CONTEXT AND OBJECTIVE: The location of embolism is associated with clinical findings and disease severity in cases of acute pulmonary embolism. The level of gamma-glutamyl transferase increases under oxidative stress-related conditions. In this study, we investigated whether gamma-glutamyl transferase levels could predict the location of pulmonary embolism. DESIGN AND SETTING: Hospital-based cross-sectional study at Cumhuriyet University, Sivas, Turkey. METHODS : 120 patients who were diagnosed with acute pulmonary embolism through computed tomography-assisted pulmonary angiography were evaluated. They were divided into two main groups (proximally and distally located), and subsequently into subgroups according to thrombus localization as follows: first group (thrombus in main pulmonary artery; n = 9); second group (thrombus in main pulmonary artery branches; n = 71); third group (thrombus in pulmonary artery segmental branches; n = 34); and fourth group (thrombus in pulmonary artery subsegmental branches; n = 8). RESULTS : Gamma-glutamyl transferase levels on admission, heart rate, oxygen saturation, right ventricular dilatation/hypokinesia, pulmonary artery systolic pressure and cardiopulmonary resuscitation requirement showed prognostic significance in univariate analysis. The multivariate logistic regression model showed that gamma-glutamyl transferase level on admission (odds ratio, OR = 1.044; 95% confidence interval, CI: 1.011-1.079; P = 0.009) and pulmonary artery systolic pressure (OR = 1.063; 95% CI: 1.005-1.124; P = 0.033) remained independently associated with proximally localized thrombus in pulmonary artery. CONCLUSIONS : The findings revealed a significant association between increased existing embolism load in the pulmonary artery and increased serum gamma-glutamyl transferase levels.


RESUMO CONTEXTO E OBJETIVO : A localização da embolia está associada com os resultados clínicos e a gravidade da doença do embolismo pulmonar agudo (EPA). O nível de gama-glutamil transferase (GGT) aumenta em condições relacionadas com estresse oxidativo. Investigou-se se os níveis de GGT podem prever a localização do EPA. TIPO DE ESTUDO E LOCAL : Estudo observacional transversal na Universidade Cumhuriyet, Sivas, Turquia. MÉTODOS : Avaliamos 120 pacientes diagnosticados com EPA após a realização de angiografia pulmonar assistida por tomografia computadorizada. Eles foram divididos em dois grupos principais (localização proximal e distal) e depois em subgrupos de acordo com a localização do trombo da seguinte forma: primeiro grupo (trombo na artéria pulmonar [AP] principal, n = 9); segundo (trombo no ramo da AP principal; n = 71); terceiro grupo (trombo na segmentar da AP; n = 34); quarto grupo (trombo na subsegmentar da AP; n = 8). RESULTADOS : Na análise univariada, os níveis de GGT tiveram significado prognóstico em relação à admissão, pulsação arterial, saturação de oxigênio, dilatação do ventrículo direito/hipocinesia, pressão sistólica da artéria pulmonar (PSAP) e necessidade de ressuscitação cardiopulmonar. O modelo de regressão logística multivariada demonstrou que o nível de GGT na admissão (razão de possibilidades, OR: 1,044; 95% intervalo de confiança, CI: 1,011-1,079; P = 0,009) e PSAP (OR: 1,063, 95% CI: 1,005-1,124; P = 0,033) permaneceram independentemente associados com trombo localizado proximalmente na AP. CONCLUSÕES : Os resultados demonstraram associação significativa entre aumento da carga existente de embolia da AP e aumento dos níveis séricos da GGT.


Subject(s)
Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Pulmonary Embolism/enzymology , gamma-Glutamyltransferase/blood , Acute Disease , Biomarkers/blood , Coronary Angiography , Cross-Sectional Studies , Prognosis , Pulmonary Artery/pathology , Pulmonary Embolism/blood , Pulmonary Embolism/pathology , ROC Curve , Reference Values , Severity of Illness Index , Statistics, Nonparametric , Tomography, X-Ray Computed
15.
Thromb Res ; 136(4): 769-74, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26337933

ABSTRACT

OBJECTIVE: Pulmonary embolism (PE) can cause intracardiac hemolysis and increased plasma hemoglobin and arginase-1, which can worsen pulmonary vasoconstriction. We test the hypothesis that patients with PE that causes tricuspid regurgitation (TR), indicative of higher pulmonary arterial pressures, have decreased leukocyte expression of hmox-1 compared with patients with PE and no TR and patients without PE. DESIGN: Prospective, noninterventional study. PATIENTS: Normotensive patients with suspected PE (n=87) who underwent CT pulmonary angiography and transthoracic Doppler-echocardiography. MEASUREMENTS: Significant TR was defined as a jet velocity >2.7m/s. Leukocyte expression of hmox-1, haptoglobin, haptoglobin related gene, the haptoglobin receptor, CD163 and cox-2 genes were assessed by quantitative rtPCR, and the hmox-1 promoter was examined for the -413 A→T SNP and GT repeat polymorphisms. RESULTS: Of the 44 (50%) with PE+, 22 had TR+, and their mean pulmonary vascular occlusion (39±32%) did not differ significantly from patients who were TR- (28±26%, P=0.15). Patients with PE+ and TR+ had significantly lower expression of hmox-1 and haptoglobin genes than patients without PE+ and no TR. Expression of hmox-1 varied inversely with TR velocity (r(2)=0.45, P<0.001) for PE+ (n=22) but not patients without PE. Hmox-1 expression did not vary significantly with genotype. Cox-2 did not differ between groups and had no correlation with TR. CONCLUSIONS: Severity of TR varied inversely with hmox-1 expression, suggesting that hmox-1 expression affects pulmonary vascular reactivity after PE.


Subject(s)
Heme Oxygenase-1/biosynthesis , Pulmonary Embolism/enzymology , Tricuspid Valve Insufficiency/enzymology , Acute Disease , Female , Fibrinolysis , Haptoglobins/metabolism , Heme Oxygenase-1/blood , Heme Oxygenase-1/genetics , Heme Oxygenase-1/immunology , Hemolysis , Humans , Male , Middle Aged , Prospective Studies , Pulmonary Embolism/blood , Pulmonary Embolism/genetics , Tricuspid Valve Insufficiency/blood , Tricuspid Valve Insufficiency/genetics , Tricuspid Valve Insufficiency/immunology
16.
Article in English | MEDLINE | ID: mdl-26005342

ABSTRACT

Deficiency in the serine protease inhibitor, alpha-1 antitrypsin (AAT), is known to cause emphysema and liver disease. Other manifestations, including airway disease or skin disorders, have also been described. A 44-year-old woman presented to our emergency department with dyspnea and respiratory insufficiency. She had never smoked, and had been diagnosed with COPD 9 years earlier. Three months previously, she had suffered a pulmonary embolism. Chest computed tomography scan revealed severe cystic bronchiectasis with destruction of the lung parenchyma. The sweat test was normal and there was no evidence of the cystic fibrosis transmembrane conductance regulator (CFTR) mutation. Capillary zone electrophoresis showed a decrease of alpha-1 globin band and AAT levels were below the quantification limit (<25 mg/dL). No S or Z mutation was identified, but sequencing analysis found a homozygous cytosine and adenine (CA) insertion in exon 2 of the SERPINA-1 gene, probably leading to a dysfunctional protein (PI Null/Null). This mutation has not been previously identified. The atypical presentation of the patient, with severe cystic bronchiectasis, highlights AAT deficiency as a differential diagnosis in bronchiectasis. Further, awareness should be raised regarding a possible increased risk of thromboembolism associated with AAT deficiency.


Subject(s)
Bronchiectasis/etiology , Mutagenesis, Insertional , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Embolism/etiology , alpha 1-Antitrypsin Deficiency/genetics , alpha 1-Antitrypsin/genetics , Adult , Bronchiectasis/diagnosis , Bronchiectasis/enzymology , Bronchiectasis/therapy , DNA Mutational Analysis , Electrophoresis, Capillary , Exons , Female , Genetic Predisposition to Disease , Homozygote , Humans , Phenotype , Predictive Value of Tests , Pulmonary Disease, Chronic Obstructive/diagnosis , Pulmonary Disease, Chronic Obstructive/enzymology , Pulmonary Disease, Chronic Obstructive/therapy , Pulmonary Embolism/diagnosis , Pulmonary Embolism/enzymology , Pulmonary Embolism/therapy , Risk Factors , Severity of Illness Index , Tomography, X-Ray Computed , alpha 1-Antitrypsin/metabolism , alpha 1-Antitrypsin Deficiency/complications , alpha 1-Antitrypsin Deficiency/diagnosis , alpha 1-Antitrypsin Deficiency/enzymology , alpha 1-Antitrypsin Deficiency/therapy
17.
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
18.
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
19.
Blood Coagul Fibrinolysis ; 25(6): 621-4, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24553061

ABSTRACT

We present the case of a patient with left atrial myxoma that presented with pulmonary embolism. The patient did not have any intracardiac communication between right and left sides of the heart. Using thrombelastography, the patient was determined to have an abnormally large velocity of plasma thrombus growth and strength with reduced vulnerability to lysis. Critically, increased carboxyhemoglobin concentrations were present, likely secondary to hemolysis from the tumor and engagement of systemic heme oxygenase-1. It was determined that the patient's plasmatic hypercoagulability was in part due to carboxyhemefibrinogen formation via a thrombelastographic method. In addition to circulating hypercoagulability, the patient also had an area of chronic venous stasis in his left ankle that had not changed for over a decade prior to this thrombophilic episode. In conclusion, we present the first case of paradoxical pulmonary embolism in the presence of a left atrial myxoma, potentially secondary to a combination of hemolysis, heme oxygenase-1 up-regulation, systemic hypercoagulability/hypofibrinolysis, and regional venous stasis.


Subject(s)
Heart Neoplasms/enzymology , Heme Oxygenase-1/blood , Myxoma/enzymology , Pulmonary Embolism/enzymology , Thrombophilia/enzymology , Aged , Carboxyhemoglobin/metabolism , Heart Atria/enzymology , Heart Atria/pathology , Heart Neoplasms/complications , Heart Neoplasms/diagnosis , Heart Neoplasms/pathology , Heme Oxygenase-1/genetics , Humans , Male , Myxoma/complications , Myxoma/diagnosis , Myxoma/pathology , Pulmonary Embolism/complications , Pulmonary Embolism/diagnosis , Pulmonary Embolism/pathology , Thrombelastography , Thrombophilia/complications , Thrombophilia/diagnosis , Thrombophilia/pathology , Up-Regulation
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...