1
|
Napolitano F, Montuori N. Role of Plasminogen Activation System in Platelet Pathophysiology: Emerging Concepts for Translational Applications. Int J Mol Sci 2022; 23:ijms23116065. [PMID: 35682744 PMCID: PMC9181697 DOI: 10.3390/ijms23116065] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 05/25/2022] [Accepted: 05/26/2022] [Indexed: 12/13/2022] Open
Abstract
Traditionally, platelets have been exclusively considered for their procoagulant and antifibrinolytic effects during normal activation of hemostasis. Effectively, activated platelets secrete coagulation factors, expose phosphatidylserine, and promote thrombin and fibrin production. In addition to procoagulant activities, platelets confer resistance of thrombi to fibrinolysis by inducing clot retraction of the fibrin network and release of huge amounts of plasminogen activator inhibitor-1, which is the major physiologic inhibitor of the fibrinolytic cascade. However, the discovery of multiple relations with the fibrinolytic system, also termed Plasminogen Activation System (PAS), has introduced new perspectives on the platelet role in fibrinolysis. Indeed, the activated membrane surface of platelets provides binding sites on which fibrinolytic enzymes can be activated. This review discusses the evidence of the profibrinolytic properties of platelets through the description of PAS components and related proteins that are contained in or bind to platelets. Our analyses of literature data lead to the conclusion that in the initial phase of the hemostatic process, antifibrinolytic effects prevail over profibrinolytic activity, but at later stages, platelets might enhance fibrinolysis through the engagement of PAS components. A better understanding of spatial and temporal characteristics of platelet-mediated fibrinolysis during normal hemostasis could improve therapeutic options for bleeding and thrombotic disorders.
Collapse
|
2
|
Lisman T, Adelmeijer J, Huskens D, Meijers JCM. Aprotinin Inhibits Thrombin Generation by Inhibition of the Intrinsic Pathway, but is not a Direct Thrombin Inhibitor. TH OPEN 2021; 5:e363-e375. [PMID: 34485811 PMCID: PMC8407936 DOI: 10.1055/s-0041-1735154] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 06/29/2021] [Indexed: 12/01/2022] Open
Abstract
Background
Aprotinin is a broad-acting serine protease inhibitor that has been clinically used to prevent blood loss during major surgical procedures including cardiac surgery and liver transplantation. The prohemostatic properties of aprotinin likely are related to its antifibrinolytic effects, but other mechanisms including preservation of platelet function have been proposed.
Aim
Here we assessed effects of aprotinin on various hemostatic pathways in vitro, and compared effects to tranexamic acid(TXA), which is an antifibrinolytic but not a serine protease inhibitor.
Methods
We used plasma-based clot lysis assays, clotting assays in whole blood, plasma, and using purified proteins, and platelet activation assays to which aprotinin or TXA were added in pharmacological concentrations.
Results
Aprotinin and TXA dose-dependently inhibited fibrinolysis in plasma. Aprotinin inhibited clot formation and thrombin generation initiated via the intrinsic pathway, but had no effect on reactions initiated by tissue factor. However, in the presence of thrombomodulin, aprotinin enhanced thrombin generation in reactions started by tissue factor. TXA had no effect on coagulation. Aprotinin did not inhibit thrombin, only weakly inhibited the TF-VIIa complex and had no effect on platelet activation and aggregation by various agonists including thrombin. Aprotinin and TXA inhibited plasmin-induced platelet activation.
Conclusion
Pharmacologically relevant concentrations of aprotinin inhibit coagulation initiated via the intrinsic pathway. The antifibrinolytic activity of aprotinin likely explains the prohemostatic effects of aprotinin during surgical procedures. The anticoagulant properties may be beneficial during surgical procedures in which pathological activation of the intrinsic pathway, for example by extracorporeal circuits, occurs.
Collapse
Affiliation(s)
- Ton Lisman
- Surgical Research Laboratory, Department of Surgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jelle Adelmeijer
- Surgical Research Laboratory, Department of Surgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Dana Huskens
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Synapse Research Institute, Maastricht, The Netherlands
| | - Joost C M Meijers
- Department of Molecular Hematology, Sanquin Research, Amsterdam, The Netherlands.,Department of Experimental Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
3
|
Pielsticker C, Brodde MF, Raum L, Jurk K, Kehrel BE. Plasmin-Induced Activation of Human Platelets Is Modulated by Thrombospondin-1, Bona Fide Misfolded Proteins and Thiol Isomerases. Int J Mol Sci 2020; 21:ijms21228851. [PMID: 33238433 PMCID: PMC7700677 DOI: 10.3390/ijms21228851] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 11/10/2020] [Accepted: 11/18/2020] [Indexed: 02/06/2023] Open
Abstract
Inflammatory processes are triggered by the fibrinolytic enzyme plasmin. Tissue-type plasminogen activator, which cleaves plasminogen to plasmin, can be activated by the cross-β-structure of misfolded proteins. Misfolded protein aggregates also represent substrates for plasmin, promoting their degradation, and are potent platelet agonists. However, the regulation of plasmin-mediated platelet activation by misfolded proteins and vice versa is incompletely understood. In this study, we hypothesize that plasmin acts as potent agonist of human platelets in vitro after short-term incubation at room temperature, and that the response to thrombospondin-1 and the bona fide misfolded proteins Eap and SCN--denatured IgG interfere with plasmin, thereby modulating platelet activation. Plasmin dose-dependently induced CD62P surface expression on, and binding of fibrinogen to, human platelets in the absence/presence of plasma and in citrated whole blood, as analyzed by flow cytometry. Thrombospondin-1 pre-incubated with plasmin enhanced these plasmin-induced platelet responses at low concentration and diminished them at higher dose. Platelet fibrinogen binding was dose-dependently induced by the C-terminal thrombospondin-1 peptide RFYVVMWK, Eap or NaSCN-treated IgG, but diminished in the presence of plasmin. Blocking enzymatically catalyzed thiol-isomerization decreased plasmin-induced platelet responses, suggesting that plasmin activates platelets in a thiol-dependent manner. Thrombospondin-1, depending on the concentration, may act as cofactor or inhibitor of plasmin-induced platelet activation, and plasmin blocks platelet activation induced by misfolded proteins and vice versa, which might be of clinical relevance.
Collapse
Affiliation(s)
- Claudia Pielsticker
- Department of Anaesthesiology, Intensive Care and Pain Medicine, Experimental and Clinical Haemostasis, University of Muenster, 48149 Muenster, Germany; (C.P.); (L.R.)
| | | | - Lisa Raum
- Department of Anaesthesiology, Intensive Care and Pain Medicine, Experimental and Clinical Haemostasis, University of Muenster, 48149 Muenster, Germany; (C.P.); (L.R.)
| | - Kerstin Jurk
- Department of Anaesthesiology, Intensive Care and Pain Medicine, Experimental and Clinical Haemostasis, University of Muenster, 48149 Muenster, Germany; (C.P.); (L.R.)
- Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
- Correspondence: (K.J.); (B.E.K.); Tel.: +49-6131178278 (K.J.); +49-2518356725 (B.E.K.)
| | - Beate E. Kehrel
- Department of Anaesthesiology, Intensive Care and Pain Medicine, Experimental and Clinical Haemostasis, University of Muenster, 48149 Muenster, Germany; (C.P.); (L.R.)
- OxProtect GmbH, 48149 Muenster, Germany;
- Correspondence: (K.J.); (B.E.K.); Tel.: +49-6131178278 (K.J.); +49-2518356725 (B.E.K.)
| |
Collapse
|
4
|
Letson HL, Morris J, Biros E, Dobson GP. Conventional and Specific-Pathogen Free Rats Respond Differently to Anesthesia and Surgical Trauma. Sci Rep 2019; 9:9399. [PMID: 31253875 PMCID: PMC6599031 DOI: 10.1038/s41598-019-45871-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 06/12/2019] [Indexed: 02/08/2023] Open
Abstract
Specific-pathogen free (SPF) animals were introduced in the 1960s to minimize disease and infection as variables in biomedical research. Our aim was to examine differences in physiological response in rat colonies bred and housed in a conventional versus SPF facility, and implications for research. Sprague-Dawley rats were anesthetized and catheterized for blood and pressure monitoring, and electrocardiogram (ECG) leads implanted. Hematology was assessed, and coagulation profile using rotational thromboelastometry. Health screening was outsourced to Cerberus Sciences. SPF rats had significantly lower pulse pressure (38% decrease), arrhythmias and prolonged QTc (27% increase) compared to conventional rats. No arrhythmias were found in conventional rats. SPF rats had significantly higher white cell, monocyte, neutrophil and lymphocyte counts, and were hyperfibrinolytic, indicated by EXTEM maximum lysis >15%. Independent assessment revealed similar pathogen exclusion between colonies, with the exception of Proteus in SPF animals. Returning to a conventional facility restored normal host physiology. We conclude that SPF animals displayed an abnormal hemodynamic, hematological and hemostatic phenotype in response to anesthesia and surgery, and provide a number of recommendations to help standardize research outcomes and translation.
Collapse
Affiliation(s)
- Hayley L Letson
- Heart, Trauma and Sepsis Research Laboratory, College of Medicine and Dentistry, James Cook University, Queensland, 4811, Australia
| | - Jodie Morris
- Heart, Trauma and Sepsis Research Laboratory, College of Medicine and Dentistry, James Cook University, Queensland, 4811, Australia
| | - Erik Biros
- Heart, Trauma and Sepsis Research Laboratory, College of Medicine and Dentistry, James Cook University, Queensland, 4811, Australia
| | - Geoffrey P Dobson
- Heart, Trauma and Sepsis Research Laboratory, College of Medicine and Dentistry, James Cook University, Queensland, 4811, Australia.
| |
Collapse
|
5
|
Letson HL, Dobson GP. 7.5% NaCl Resuscitation Leads to Abnormal Clot Fibrinolysis after Severe Hemorrhagic Shock and its Correction with 7.5% NaCl Adenosine, Lidocaine, and Mg 2. J Emerg Trauma Shock 2018; 11:15-24. [PMID: 29628664 PMCID: PMC5852910 DOI: 10.4103/jets.jets_84_17] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Background Hyperfibrinolysis is a common complication of hemorrhagic shock. Our aim was to examine the effect of small-volume 7.5% NaCl adenosine, lidocaine, and Mg2+ (ALM) on fibrinolysis in the rat model of hemorrhagic shock. Methods Rats were anesthetized and randomly assigned to one of four groups: (1) baseline, (2) shock, (3) 7.5% NaCl controls, and (4) 7.5% NaCl ALM. Animals were bled for 20 min (42% blood loss) and left in shock for 60 min before resuscitation with 0.3 ml intravenous bolus 7.5% NaCl ± ALM. Rats were sacrificed at 5, 10, 15, 30, and 60 min for rotation thromboelastometry and 15 and 60 min for ELISA analyses. Results After hemorrhagic shock, 7.5% NaCl failed to resuscitate and exacerbated coagulopathy and fibrinolysis. At 15 and 60 min, the activation as extrinsically-activated test using tissue factor (EXTEM) with aprotinin to inhibit fibrinolysis (APTEM) test showed little or no correction of fibrinolysis, indicating a plasmin-independent fibrinolysis. Clots also had ~ 60% lower fibrinogen (fibrin-based EXTEM activated test with platelet inhibitor cytochalasin D A10) and 36%-50% reduced fibrinogen-to-platelet ratio (11%-14% vs. 22% baseline). In contrast, 7.5% NaCl ALM resuscitated mean arterial pressure and attenuated hyperfibrinolysis and coagulopathy by 15 min. Correction was associated with lower plasma tissue factor, higher plasminogen activator inhibitor-1, and lower D-dimers (5% of controls at 60 min). Platelet selectin fell to undetectable levels in ALM animals, which may imply improved endothelial and platelet function during resuscitation. Conclusions Small-volume 7.5% NaCl resuscitation exacerbated coagulopathy and fibrinolysis that was not corrected by APTEM test. Fibrinolysis appears to be associated with altered fibrin structure during early clot formation and elongation. In contrast, 7.5% NaCl ALM rapidly corrected both coagulopathy and hyperfibrinolysis.
Collapse
Affiliation(s)
- Hayley Louise Letson
- Heart, Trauma and Sepsis Research Laboratory, College of Medicine and Dentistry, James Cook University, Queensland 4811, Australia
| | - Geoffrey Phillip Dobson
- Heart, Trauma and Sepsis Research Laboratory, College of Medicine and Dentistry, James Cook University, Queensland 4811, Australia
| |
Collapse
|
6
|
Mossad EB, Machado S, Apostolakis J. Bleeding Following Deep Hypothermia and Circulatory Arrest in Children. Semin Cardiothorac Vasc Anesth 2016; 11:34-46. [PMID: 17484172 DOI: 10.1177/1089253206297413] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Deep hypothermic circulatory arrest (DHCA) is a technique of extracorporeal circulation commonly used in children with complex congenital heart defects undergoing surgical repairs. The use of profound cooling (20°C) and complete cessation of circulation allow adequate exposure and correction of these complex lesions, with enhanced cerebral protection. However, the profound physiologic state of DHCA results in significant derangement of the coagulation system and a high incidence of postoperative bleeding. This review examines the impact of DHCA on bleeding and transfusion requirements in children and the pathophysiology of DHCA-induced platelet dysfunction. It also focuses on possible pharmacologic interventions to decrease bleeding following DHCA in children.
Collapse
Affiliation(s)
- Emad B Mossad
- Department of Cardiothoracic Anesthesia-G3, Cleveland Clinic, Cleveland, OH 44195, USA.
| | | | | |
Collapse
|
7
|
Abstract
Cardiopulmonary bypass (CPB) is a nonphysiologic state that has many detrimental effects on a patient's hemostatic integrity. Exposure to the extracorporeal circuit and subsequent activation of the coagulation and fibrinolytic systems are factors that contribute to morbidity and mortality in cardiac surgical patients. These effects can be prevented in part or appropriately treated if practitioners understand the basic mecha nisms. This article reviews the effects of CPB on platelet function, the relationship of platelet function to post operative bleeding, the monitors available to measure platelet function, and the impact of antiplatelet therapy on bleeding in cardiac surgery.
Collapse
Affiliation(s)
- Shari Samson
- Department of Anesthesiology, Mount Sinai Medical Center, New York, NY
| | | |
Collapse
|
8
|
Alacacioglu I, Ozcan MA, Piskin O, Yuksel F, Alacacioglu A, Demirkan F, Ozsan HG, Polat M, Ozgenc Y, Undar B. Increased Concentration of Soluble CD40 Ligand in Preeclampsia. Clin Appl Thromb Hemost 2016; 13:201-5. [PMID: 17456631 DOI: 10.1177/1076029606299076] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Preeclampsia has been associated with increased platelet activation detected before disease onset. Platelets are involved in hemostasis and also directly initiate an inflammatory response of the vessel wall. Inappropriate activation of platelets may be involved in pathogenesis in preeclampsia by promoting coagulation and thrombosis, and also as a mediator of inflammation. Platelets may release inflammatory mediators such as soluble CD40 ligand. The plasma level of soluble CD40 ligand was investigated during preeclamptic (n =20) and normal pregnancies (n = 20) to emphasize inflammatory response in preeclampsia. The mean soluble CD40 ligand levels were 1.08 ± 0.43 ng/mL in patients with preeclampsia and 0.76 ± 0.24 ng/mL in healthy pregnant women, which was statistically significant ( P = .01). To clarify whether inflammation may cause inappropriate endothelial cell activation or inappropriate endothelial cell activation may start this inflammatory response, future studies are needed in a larger study population.
Collapse
Affiliation(s)
- Inci Alacacioglu
- Department of Haematology, Dokuz Eylul University Faculty of Medicine, Izmir, Turkey.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Differential contributions of platelets and fibrinogen to early coagulopathy in a rat model of hemorrhagic shock. Thromb Res 2016; 141:58-65. [DOI: 10.1016/j.thromres.2016.03.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 03/02/2016] [Accepted: 03/05/2016] [Indexed: 11/20/2022]
|
10
|
Salem MAEK, Adly AAM, Ismail EAR, Darwish YW, Kamel HA. Platelets microparticles as a link between micro- and macro-angiopathy in young patients with type 1 diabetes. Platelets 2015; 26:682-8. [DOI: 10.3109/09537104.2015.1018880] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
11
|
Xin Y, Liu L, Chen B, Zhang L, Tong Y. Affinity purification of aprotinin from bovine lung. J Sep Sci 2015; 38:1441-8. [PMID: 25677462 DOI: 10.1002/jssc.201401188] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Revised: 01/27/2015] [Accepted: 01/28/2015] [Indexed: 11/06/2022]
Abstract
An affinity protocol for the purification of aprotinin from bovine lung was developed. To simulate the structure of sucrose octasulfate, a natural specific probe for aprotinin, the affinity ligand was composed of an acidic head and a hydrophobic stick, and was then linked with Sepharose. The sorbent was then subjected to adsorption analysis with pure aprotinin. The purification process consisted of one step of affinity chromatography and another step of ultrafiltration. Then purified aprotinin was subjected to sodium dodecyl sulfate polyacrylamide gel electrophoresis, trypsin inhibitor activity, gel-filtration, and thin-layer chromatography analysis. As calculated, the theoretical maximum adsorption (Qmax ) of the affinity sorbent was 25,476.0 ± 184.8 kallikrein inactivator unit/g wet gel; the dissociation constant of the complex "immobilized ligand-aprotinin" (Kd ) was 4.6 ± 0.1 kallikrein inactivator unit/mL. After the affinity separation of bovine lung aprotinin, reducing sodium dodecyl sulfate polyacrylamide gel electrophoresis analysis and gel-filtration chromatography revealed that the protein was a single polypeptide, and the purities were ∼ 97 and 100%, respectively; the purified peptide was also confirmed with aprotinin standard by gel-filtration chromatography and thin-layer chromatography. After the whole purification process, protein, and bioactivity recoveries were 2.2 and 92.6%, respectively; and the specific activity was up to 15,907.1 ± 10.2 kallikrein inactivator unit/mg.
Collapse
Affiliation(s)
- Yu Xin
- School of Biotechnology, Jiangnan University, Key Laboratory of Industry Biotechnology, Ministry of Education, Jiangsu, P. R. China
| | - Lanhua Liu
- Nanchang Wanhua Biochem Products Co., Ltd, Nanchang, Jiangxi, P. R. China
| | - Beizhan Chen
- Nanchang Wanhua Biochem Products Co., Ltd, Nanchang, Jiangxi, P. R. China
| | - Ling Zhang
- School of Biotechnology, Jiangnan University, Key Laboratory of Industry Biotechnology, Ministry of Education, Jiangsu, P. R. China
| | - Yanjun Tong
- School of Biotechnology, Jiangnan University, Key Laboratory of Industry Biotechnology, Ministry of Education, Jiangsu, P. R. China
| |
Collapse
|
12
|
Tykhomyrov AA, Zhernosekov DD, Roka-Moya YM, Diordieva SI, Grinenko TV. [THE EFFECTS OF LYS-PLASMINOGEN ON HUMAN PLATELET SECRETION]. ACTA ACUST UNITED AC 2015; 61:26-34. [PMID: 27025042 DOI: 10.15407/fz61.06.026] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The effects of Lys-plasminoge on platelet α-granule secretion were studied. The level of P-selectin exposed on the surface of plasma membranes of washed human platelets was measured by flow cytometry as a market of α-granule secretion. It was shown that Lys-plasminogen facilitates partial release of α-granules, but impedes thrombin-induced platelet exocytosis. It is suggested that Lys-plasminogen may affect platelet secretion rather through interaction of its non-catalytic (kringle) domains with membrane receptors than due to contaminating plasmin activity. In contrast to Lys-form, native proenzyme (Glu-plasminogen) had no effects on α-granule releasing. Here, we provide the first experimental demonstration that Lys-form of plasminogen is able to modulate platelet α-granule secretion, and this effect can be considered as one of the plausible mechanisms of its anti-aggregating activity.
Collapse
|
13
|
Tantawy AAG, Adly AAM, Ismail EAR, Habeeb NM. Flow cytometric assessment of circulating platelet and erythrocytes microparticles in young thalassemia major patients: relation to pulmonary hypertension and aortic wall stiffness. Eur J Haematol 2013; 90:508-18. [PMID: 23506251 DOI: 10.1111/ejh.12108] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/14/2013] [Indexed: 01/19/2023]
Abstract
Heart disease is the leading cause of mortality and morbidity in β-thalassemia major (β-TM). Aggregability of abnormal red cells and membrane-derived microparticles (MPs) stemming from activated platelets and erythrocytes are responsible for thrombotic risk. We measured platelet and erythrocyte MPs (PMPs and ErMPs) in 60 young β-TM patients compared with 40 age- and sex-matched healthy controls and assessed their relation to clinicopathological characteristics and aortic elastic properties. Patients were studied stressing on transfusion history, splenectomy, thrombotic events, chelation therapy, hematological and coagulation profiles, flow cytometric measurement of PMPs (CD41b(+) ) and ErMPs (glycophorin A(+) ) as well as echocardiographic assessment of aortic elastic properties. Aortic stiffness index and pulmonary artery pressure were significantly higher, whereas aortic strain and distensibility were lower in TM patients than controls (P < 0.001). Both PMPs and ErMPs were significantly elevated in TM patients compared with controls, particularly patients with risk of pulmonary hypertension, history of thrombosis, splenectomy or serum ferritin >2500 μg/L (P < 0.001). Compliant patients on chelation therapy had lower MPs levels than non-compliant patients (P < 0.001). PMPs and ErMPs were positively correlated to markers of hemolysis, serum ferritin, D-dimer, vWF Ag, and aortic stiffness, whereas negatively correlated to hemoglobin level and aortic distensibility (P < 0.05). We suggest that increased MPs may be implicated in vascular dysfunction, pulmonary hypertension risk, and aortic wall stiffness observed in thalassemia patients. Their quantification could provide utility for early detection of cardiovascular abnormalities and monitoring the biological efficacy of chelation therapy.
Collapse
Affiliation(s)
- Azza A G Tantawy
- Pediatrics Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt.
| | | | | | | |
Collapse
|
14
|
Tantawy AAG, Adly AAM, Ismail EAR, Habeeb NM, Farouk A. Circulating platelet and erythrocyte microparticles in young children and adolescents with sickle cell disease: Relation to cardiovascular complications. Platelets 2012; 24:605-14. [DOI: 10.3109/09537104.2012.749397] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
15
|
Ismail EAR, Youssef OI. Platelet-Derived Microparticles and Platelet Function Profile in Children With Congenital Heart Disease. Clin Appl Thromb Hemost 2012; 19:424-32. [DOI: 10.1177/1076029612456733] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Platelet microparticles (PMPs) and function profile in children with congenital heart disease (CHD) have not been widely explored. We investigated platelet aggregation, flow cytometric platelet surface receptors (P-selectin and glycoprotein (GP) IIb/IIIa) and PMPs in 23 children with cyanotic CHD (CCHD), 30 children with acyanotic CHD (ACHD) and 30 healthy controls correlating these variables to hematological and coagulation parameters including von Willebrand factor antigen (vWF Ag) as a marker of endothelial dysfunction. Hemoglobin, hematocrit (HCT), d-dimer, and vWF Ag were significantly higher in CCHD than ACHD group. Platelet MPs and P-selectin expression were increased in patients than controls, particularly in CCHD and positively correlated to HCT, d-dimer, and vWF Ag while platelet count, aggregation, and GP IIb/IIIa expression were decreased in CCHD compared with ACHD group and negatively correlated to HCT. The overproduction of PMPs and platelet activation with suppressed aggregation may be implicated in the pathogenesis of coagulation/hemostatic abnormalities in children with CCHD.
Collapse
|
16
|
Rozental T, Shore-Lesserson L. Pharmacologic Management of Coagulopathy in Cardiac Surgery: An Update. J Cardiothorac Vasc Anesth 2012; 26:669-79. [DOI: 10.1053/j.jvca.2012.03.030] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Indexed: 11/11/2022]
|
17
|
Sniecinski RM, Chandler WL. Activation of the Hemostatic System During Cardiopulmonary Bypass. Anesth Analg 2011; 113:1319-33. [DOI: 10.1213/ane.0b013e3182354b7e] [Citation(s) in RCA: 149] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
18
|
Landis RC, Murkin JM, Stump DA, Baker RA, Arrowsmith JE, De Somer F, Dain SL, Dobkowski WB, Ellis JE, Falter F, Fischer G, Hammon JW, Jonas RA, Kramer RS, Likosky DS, Milsom FP, Poullis M, Verrier ED, Walley K, Westaby S. Consensus Statement: Minimal Criteria for Reporting the Systemic Inflammatory Response to Cardiopulmonary Bypass. Heart Surg Forum 2011. [DOI: 10.1532/hsf98.20101182] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
19
|
Clive Landis R, Murkin JM, Stump DA, Baker RA, Arrowsmith JE, De Somer F, Dain SL, Dobkowski WB, Ellis JE, Falter F, Fischer G, Hammon JW, Jonas RA, Kramer RS, Likosky DS, Paget Milsom F, Poullis M, Verrier ED, Walley K, Westaby S. Consensus statement: minimal criteria for reporting the systemic inflammatory response to cardiopulmonary bypass. Heart Surg Forum 2010; 13:E116-23. [PMID: 20444674 DOI: 10.1532/hsf98.20101022] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The lack of established cause and effect between putative mediators of inflammation and adverse clinical outcomes has been responsible for many failed anti-inflammatory interventions in cardiopulmonary bypass (CPB). Candidate interventions that impress in preclinical trials by suppressing a given inflammation marker might fail at the clinical trial stage because the marker of interest is not linked causally to an adverse outcome. Alternatively, there exist examples in which pharmaceutical agents or other interventions improve clinical outcomes but for which we are uncertain of any antiinflammatory mechanism. The Outcomes consensus panel made 3 recommendations in 2009 for the conduct of clinical trials focused on the systemic inflammatory response. This panel was tasked with updating, as well as simplifying, a previous consensus statement. The present recommendations for investigators are the following: (1) Measure at least 1 inflammation marker, defined in broad terms; (2) measure at least 1clinical end point, drawn from a list of practical yet clinically meaningful end points suggested by the consensus panel; and(3) report a core set of CPB and perfusion criteria that maybe linked to outcomes. Our collective belief is that adhering to these simple consensus recommendations will help define the influence of CPB practice on the systemic inflammatory response, advance our understanding of causal inflammatory mechanisms, and standardize the reporting of research findings in the peer-reviewed literature.
Collapse
|
20
|
Yang IS, Kim TG, Park BS, Cho KJ, Lee JH, Park Y, Kim KH. Crystal structures of aprotinin and its complex with sucrose octasulfate reveal multiple modes of interactions with implications for heparin binding. Biochem Biophys Res Commun 2010; 397:429-35. [PMID: 20529698 DOI: 10.1016/j.bbrc.2010.05.113] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2010] [Accepted: 05/24/2010] [Indexed: 11/17/2022]
Abstract
The crystal structures of aprotinin and its complex with sucrose octasulfate (SOS), a polysulfated heparin analog, were determined at 1.7-2.6A resolutions. Aprotinin is monomeric in solution, which associates into a decamer at high salt concentrations. Sulfate ions serve to neutralize the basic amino acid residues of aprotinin to stabilize the decameric aprotinin. Whereas SOS interacts with heparin binding proteins at 1:1 molar ratio, SOS was surprisingly found to induce strong agglutination of aprotinins. Five molecules of aprotinin interact with one molecule of the sulfated sugar, which is stabilized by electrostatic interactions between the positively charged residues of aprotinin and sulfate groups of SOS. The multiple binding modes of SOS with five individual aprotinin molecules may represent the diverse patterns of potential heparin binding to aprotinin, reflecting the interactions of densely packed protein molecules along the heparin polymer.
Collapse
Affiliation(s)
- In Seok Yang
- Department of Biotechnology & Bioinformatics, Korea University, Chungnam 339-700, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
21
|
Sun HY, Szlam F, Levy JH, Csete ME, Tanaka KA. Antifibrinolytic agents reduce tissue plasminogen activator-mediated neuronal toxicity in vitro. Acta Anaesthesiol Scand 2009; 53:325-31. [PMID: 19243318 DOI: 10.1111/j.1399-6576.2008.01858.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
INTRODUCTION Serine proteases and their inhibitors play an important role in physiological homeostasis including neuronal activity, hemostasis, and wound healing. Tissue plasminogen activator (tPA) is involved in normal neuronal plasticity and memory formation but can also be neurotoxic. We hypothesized that the serine protease inhibitor aprotinin confers neuronal protection by inhibiting tPA activity. METHODS Using cultured rat dopaminergic neuroblasts (N27 line), tPA-induced cytotoxicity was quantitated by an MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] assay and flow cytometry using propidium iodide DNA staining. The anti-apoptotic effects of aprotinin and other protease inhibitors were also evaluated using these systems. RESULTS Treatment of cultured neuroblasts with tPA (10-20 microg/ml) caused a dose-dependent decrease in cell viability (71.3+/-2.4 at 10 microg/ml down to 52.7+/-2.5% at 20 microg/m tPA, 24-h treatment), which was potentiated in the absence of serum in the culture medium (59.5+/-6.3% at 10 microg/ml down to 47.9+/-4.7% at 20 microg/ml). Aprotinin was effective in ameliorating cell death when administered 30 min before tPA exposure as shown by increased cell viability (91.8+/-0.6% at tPA at 20 microg/ml), but this protection was significantly reduced when aprotinin was administered after tPA. The efficacy of aprotinin as a neuroprotectant was equivalent or superior to other direct tPA antagonist peptides Glu-Gly-Arg-chlormethylketone (EGRck) and Phe-Pro-Arg-chlormethylketone (FPRck) in this setting. CONCLUSION These data suggest that one of the mechanisms of neuroprotection afforded by aprotinin may be inhibition of tPA-mediated neurotoxicity.
Collapse
Affiliation(s)
- H-Y Sun
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | | | | | | |
Collapse
|
22
|
Ngaage DL, Cale AR, Cowen ME, Griffin S, Guvendik L. Aprotinin in primary cardiac surgery: operative outcome of propensity score-matched study. Ann Thorac Surg 2008; 86:1195-202. [PMID: 18805159 DOI: 10.1016/j.athoracsur.2008.06.048] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2008] [Revised: 06/06/2008] [Accepted: 06/09/2008] [Indexed: 10/21/2022]
Abstract
BACKGROUND Some recent multicenter series have questioned the safety of aprotinin in primary cardiac operations. We report a large, single-center experience with aprotinin therapy in primary cardiac operations and discuss the limitations and potential confounders of current treatment strategies. METHODS We compared myocardial infarction, neurologic events, renal insufficiency, and operative death after first-time coronary or valve procedures, or both, in 3334 patients treated with full-dose aprotinin with 3417 patients not treated with aprotinin who underwent operation between March 1998 and January 2007. Further analysis was performed for 341 propensity score-matched pairs. RESULTS There were substantial differences between the groups. Aprotinin patients were higher risk on account of older age, unstable symptoms, poor ejection fraction, preoperative hemodynamic support, emergency/urgent operations, and combined coronary/valve operations. Postoperative bleeding and blood product transfusion were considerably reduced in aprotinin patients, as was median duration of mechanical ventilation. Aprotinin was neither a predictor of postoperative myocardial infarction, renal insufficiency, neurologic dysfunction, or operative death. Achieving parity between the groups by propensity score matching eliminated the elevated rates of postoperative renal insufficiency, neurologic dysfunction, and operative death observed in aprotinin patients in the unmatched comparison. These adverse outcomes were evenly distributed between matched groups. Conversely, blood transfusion had univariate associations with all adverse outcome measures. CONCLUSIONS Full-dose aprotinin use was not associated with myocardial infarction, neurologic dysfunction, renal insufficiency, or death after coronary or valve operations. We observed less postoperative bleeding and blood product transfusion, and early extubation with the use of aprotinin.
Collapse
Affiliation(s)
- Dumbor L Ngaage
- Castle Hill Hospital, Kingston-Upon-Hull, East Yorkshire, United Kingdom.
| | | | | | | | | |
Collapse
|
23
|
Yamada T, Katori N, Tanaka KA, Takeda J. Impact of Sonoclot hemostasis analysis after cardiopulmonary bypass on postoperative hemorrhage in cardiac surgery. J Anesth 2007; 21:148-52. [PMID: 17458642 DOI: 10.1007/s00540-006-0477-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2006] [Accepted: 11/01/2006] [Indexed: 10/23/2022]
Abstract
PURPOSE The Sonoclot Analyzer provides a functional test of whole blood coagulation by measuring the viscous property of the blood sample. In this study, we used a modified Sonoclot assay, using cuvettes with a glass bead activator containing heparinase, and compared the Sonoclot data before and after cardiopulmonary bypass (CPB) to assess the usefulness in predicting postoperative hemorrhage. METHODS In 41 cardiac surgery patients, Sonoclot data were obtained immediately after heparin administration (pre-bypass) and just before protamine administration (post-bypass). Excessive bleeding was defined as chest tube drainage greater than 2 ml.kg(-1).h(-1) in 1 h during the first 4 h after surgery. RESULTS There were no significant differences in Sonoclot values before and after CPB in patients with acceptable bleeding (n = 29). In patients with excessive bleeding (n = 12), Sonoclot variables reflecting fibrin formation (activated clotting time [ACT], rate of fibrin formation [clot rate], and peak clot signal) were preserved after CPB; however, the variables reflecting platelet-fibrin interaction (time to peak, peak angle, and clot retraction rate) were significantly different from their respective pre-bypass values. Sonoclot analysis showed impairment of clot maturation after CPB in patients with excessive postoperative bleeding. CONCLUSION Our results suggest that abnormal postoperative hemorrhage can be predicted by Sonoclot analysis with a new glass bead-activated heparinase test performed after CPB.
Collapse
Affiliation(s)
- Tatsuya Yamada
- Department of Anesthesiology, School of Medicine, Keio University, 35 Shinanomachi, Tokyo 160-8582, Japan
| | | | | | | |
Collapse
|
24
|
Lösche W, Heptinstall S. Value of Platelet Activation Markers as Prothrombotic Risk Indicators. Transfus Med Hemother 2007. [DOI: 10.1159/000097497] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
25
|
Weant KA, Flynn JF, Akers WS. Management of antiplatelet therapy for minimization of bleeding risk before cardiac surgery. Pharmacotherapy 2007; 26:1616-25. [PMID: 17064207 DOI: 10.1592/phco.26.11.1616] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Antiplatelet therapy is commonly administered for primary and secondary prevention of stroke, recurrent angina, myocardial infarction, and death in patients with cardiovascular disorders. It also is associated with an increased risk of bleeding. We describe the management of antiplatelet therapy in patients undergoing coronary artery bypass graft surgery. In addition, we provide basic information about the mechanisms of action by which the most common antiplatelet agents inhibit platelet function. This information is integrated with results from pharmacologic studies and clinical trials. Determining the net effect in patients undergoing coronary artery bypass graft surgery requires knowledge about the pharmacokinetics, pharmacodynamics, and clinical efficacy of each drug, and an estimation of the absolute thrombotic versus hemorrhagic risk for each patient.
Collapse
Affiliation(s)
- Kyle A Weant
- University of North Carolina Hospitals and the School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, USA
| | | | | |
Collapse
|
26
|
Cicekcioglu F, Cagli K, Emir M, Topbas M, Catav Z, Sener E, Tasdemir O. Effects of minimal dose aprotinin on blood loss and fibrinolytic system-complement activation in coronary artery bypass grafting surgery. J Card Surg 2006; 21:336-41. [PMID: 16846409 DOI: 10.1111/j.1540-8191.2006.00267.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND To determine whether 500,000 KIU aprotinin is effective to reduce blood loss in coronary artery bypass grafting (CABG) and to evaluate the effects of this regimen on hematologic parameters. METHODS Forty-four patients scheduled for primary CABG were randomly assigned to the aprotinin (n = 24) or control group (n = 20). In aprotinin group, aprotinin was administered in two equal doses (before skin incision and added to the pump prime). Ventilation time, intensive care unit stay, mediastinal tube drainage, hospitalization, transfusion requirements, and postoperative morbidities and mortality were noted. Hematologic markers of fibrinolytic activity and complement activation were also measured pre- and postoperatively. RESULTS Although less mediastinal drainage occurred in aprotinin group, the difference was not statistically significant. Other postoperative variables like transfusion requirements, morbidities, and mortality were also found to be similar between groups. Among hematologic parameters, only postoperative levels of alpha2-antiplasmin and plasminogen activator inhibitor-1 were significantly higher in aprotinin group. CONCLUSIONS Although plasmin inhibitors begin to rise at this very low aprotinin dosage, it is not advisable to use this aprotinin regimen in CABG patients.
Collapse
Affiliation(s)
- Ferit Cicekcioglu
- Cardiovascular Surgery Clinic, Turkiye Yuksek Ihtisas Hospital, Ankara, Turkey
| | | | | | | | | | | | | |
Collapse
|
27
|
Day JRS, Landis RC, Taylor KM. Aprotinin and the protease-activated receptor 1 thrombin receptor: antithrombosis, inflammation, and stroke reduction. Semin Cardiothorac Vasc Anesth 2006; 10:132-42. [PMID: 16959740 DOI: 10.1177/1089253206288997] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Cardiopulmonary bypass, although remaining an indispensable asset in cardiac surgery, especially in more complex and repeat operations, is associated with significant thrombin generation in the bypass circuit, leading to the activation of platelets, the coagulation system, an inflammatory response, and perioperative stroke. Recent clinical studies and meta-analyses of clinical trials in coronary artery bypass grafting surgery have confirmed that aprotinin not only reduces transfusion requirements in cardiac surgery but also confers significant protection against platelet dysfunction, activation of the systemic inflammatory response, and perioperative stroke when administered at the full (or "Hammersmith") dose. This article reviews research from several independent groups to propose a novel mechanism through which the antithrombotic, anti-inflammatory, and neuroprotective mechanism might be mediated, via protection of the high-affinity thrombin receptor protease-activated receptor 1 (PAR1).
Collapse
Affiliation(s)
- J R S Day
- British Heart Foundation Cardiac Surgery Unit, Imperial College, London, UK.
| | | | | |
Collapse
|
28
|
Abstract
Skilful surgery combined with blood-saving methods and careful management of blood coagulation will all help reduce unnecessary blood loss and transfusion requirements. Excessive surgical bleeding causes hypovolaemia, haemodynamic instability, anaemia and reduced oxygen delivery to tissues, with a subsequent increase in postoperative morbidity and mortality. The role of anaesthetists in managing surgical blood loss has increased greatly in the last decade. Position of the patient during surgery and the provision of a hypotensive anaesthetic regimen were once considered the most important contributions of the anaesthetist to decreasing blood loss. Now, several pharmacological haemostatic agents are being used by anaesthetists as blood-saving agents. After a brief discussion of the physiology of haemostasis, this article will review the evidence for the role of such agents in reducing perioperative blood loss and transfusion requirements.
Collapse
Affiliation(s)
- A M Mahdy
- Academic Unit of Anaesthesia and Intensive Care, University of Aberdeen, Aberdeen, UK
| | | |
Collapse
|
29
|
Paparella D, Brister SJ, Buchanan MR. Coagulation disorders of cardiopulmonary bypass: a review. Intensive Care Med 2004; 30:1873-81. [PMID: 15278267 DOI: 10.1007/s00134-004-2388-0] [Citation(s) in RCA: 247] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2003] [Accepted: 06/24/2004] [Indexed: 01/17/2023]
Abstract
BACKGROUND Postoperative bleeding is one of the most common complications of cardiac surgery. DISCUSSION Extensive surgical trauma, prolonged blood contact with the artificial surface of the cardiopulmonary bypass (CPB) circuit, high doses of heparin, and hypothermia are all possible triggers of a coagulopathy leading to excessive bleeding. Platelet activation and dysfunction also occur and are caused mainly by heparin, hypothermia, and inadequate protamine administration. Heparin and protamine administration based on heparin concentrations as opposed to fixed doses may reduce coagulopathy and postoperative blood loss. CONCLUSIONS A better comprehension of the multifactorial mechanisms of activation of coagulation, inflammation, and fibrinolytic pathways during CPB may enable a more effective use of the technical and pharmaceutical options which are currently available.
Collapse
Affiliation(s)
- Domenico Paparella
- Division of Cardiac Surgery, Dipartimento di Emergenza e Trapianti di Organo, University of Bari, Piazza Giulio Cesare 11, 70100 Bari, Italy.
| | | | | |
Collapse
|
30
|
Kucharewicz I, Kowal K, Buczko W, Bodzenta-Łukaszyk A. The plasmin system in airway remodeling. Thromb Res 2004; 112:1-7. [PMID: 15013265 DOI: 10.1016/j.thromres.2003.10.011] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2003] [Revised: 10/14/2003] [Accepted: 10/15/2003] [Indexed: 11/22/2022]
Abstract
Recent studies suggest that the plasmin system plays an active role in tissue remodeling. Plasmin degrades the extracellular matrix (ECM), either directly removing glycoproteins from ECM or by activating matrix metalloproteinases (MMPs). PAI-1 blocking MMPs may prevent ECM degradation, but inhibiting fibrinolysis leads to fibrin accumulation and fibrosis. Components of the plasmin system including tissue plasminogen activator (t-PA), urokinase plasminogen activator (u-PA), and plasminogen activator inhibitors PAI-1 and PAI-2 are synthesised by airway cells, and inflammatory mediators affect their expression. The plasmin system, in turn, actively influences the production of inflammatory mediators and growth factors, extending pathological structural changes in the airway. Modulation of the plasmin system might be a new pharmacological strategy that could inhibit the development of airway remodeling.
Collapse
Affiliation(s)
- Iwona Kucharewicz
- Department of Allergology and Internal Diseases, Medical University of Bialystok, M. Sklodowska-Curie Street 24a, 15-276 Bialystok, Poland.
| | | | | | | |
Collapse
|
31
|
Maslow A, Schwartz C. Cardiopulmonary Bypass-Associated Coagulopathies and Prophylactic Therapy. Int Anesthesiol Clin 2004; 42:103-33. [PMID: 15205643 DOI: 10.1097/00004311-200404230-00010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Andrew Maslow
- Department of Anesthesiology, Rhode Island Hospital, Providence, 02903, USA
| | | |
Collapse
|
32
|
Chong AJ, Hampton CR, Verrier ED. Microvascular Inflammatory Response in Cardiac Surgery. Semin Cardiothorac Vasc Anesth 2003. [DOI: 10.1177/108925320300700308] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cardiac surgical procedures, with or without cardiopulmonary bypass, elicit a systemic inflammatory response in patients that induces the elaboration of multiple cytokines, chemokines, adhesion molecules, and destructive enzymes. This inflammatory reaction involves multiple interdependent and redundant cell types and humoral cascades, which allows for amplification and positive feedback at numerous steps. This systemic inflammatory response ultimately results in a broad spectrum of clinical manifestations, with multiple organ failure being the most severe form. Investigative efforts have focused on understanding the mechanism of this systemic inflammatory response syndrome in order to develop potential therapeutic targets to inhibit it, thereby possibly decreasing postoperative morbidity and mortality. Multiple therapeutic methods have been investigated, including pharmacologic inhibitors and modifications of surgical technique and the cardiopulmonary bypass circuit. Although studies have demonstrated that the use of these therapies in experimental and clinical settings has attenuated the systemic inflammatory response, they have failed to conclusively show clinical benefit from these therapies. These therapies may be too specific to minimize the deleterious effects of a systemic inflammatory response that results from the activation of multiple, interdependent, and redundant inflammatory cascades and cell types. Hence, further studies that investigate the molecular and cellular events underlying the systemic inflammatory response syndrome and the resultant effects of anti-inflammatory therapies are warranted to ultimately achieve improvements in clinical outcome after cardiac surgical procedures.
Collapse
Affiliation(s)
| | | | - Edward D. Verrier
- Division of Cardiothoracic Surgery, The University of Washington, Seattle, Washington
| |
Collapse
|
33
|
Landis RC. Aprotinin: Antithrombotic and Vasoactive Mechanisms of Action. Semin Cardiothorac Vasc Anesth 2002. [DOI: 10.1177/108925320200600405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Aprotinin is a serine protease inhibitor that has been in clinical use since the late 1980s to reduce blood loss in patients undergoing cardiopulmonary bypass surgery. Its hemostatic mechanism of action is mediated predominantly through inhibition of plasmin, thus exerting a net antifibrinolytic effect. Compared to other antifibrinolytics, however, aprotinin provides an additional patient benefit at the level of improved platelet function and diminished inflammatory response to bypass. Recent work on platelets has identified a cell-associated target for aprotinin: the thrombin-receptor, protease-activated receptor 1. Selective blockade of the protease-activated receptor 1 limits thrombin-induced activation and consequent “exhaustion” of platelets in the bypass circuit, while maintaining the hemostatic activity of platelets in the pericardial cavity in response to nonproteolytic agonists, such as collagen, adenosine diphosphate and epinephrine. While no specific cellular receptors have as yet been identified to explain the antiinflammatory and vasoactive properties of aprotinin, awareness is growing that serine protease-sensitive receptors belonging to the protease-activated receptor family (1-4) may represent important aprotinin targets, since these receptors are expressed by all major cells of the vasculature and act as sensors of the coagulation, inflammatory and vasoactive pathways activated by major surgery or trauma. The possibility is discussed that endothelial protease-activated receptor 2, whose natural ligands are trypsin, tryptase and the ternary tissue factor-Vlla-Xa complex, may be targeted by aprotinin.
Collapse
Affiliation(s)
- R. Clive Landis
- Cardiovascular Medicine Unit, Faculty of Medicine, Imperial College, Hammersmith Hospital, London W12 ONN, UK
| |
Collapse
|
34
|
Carr ME, Carr SL, Roa V, McCardell KA, Greilich PE. Aprotinin counteracts heparin-induced inhibition of platelet contractile force. Thromb Res 2002; 108:161-8. [PMID: 12590953 DOI: 10.1016/s0049-3848(02)00403-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Aprotinin interferes with heparin binding to platelets and decreases blood loss during cardiopulmonary bypass (CPB). Heparin abolishes platelet force during CPB, and the extent of platelet force recovery after protamine administration appears to correlate with blood loss. This study assessed the effect of aprotinin on heparin suppression of platelet force. METHODS Platelet force was measured using the Hemodyne Hemostasis Analyzer. Clots were formed from platelet-rich plasma (PRP) by the addition of batroxobin and 10 mM CaCl(2). Clotting conditions included pH 7.4, ionic strength 0.15 M, fibrinogen level 1 mg/ml and 75,000 platelets/microl. RESULTS After 1200 s of clotting, force was reduced from 7110+/-1190 to 450+/-450 dyn by 0.2 U/ml of heparin. Platelet force in aprotinin [20 microg/ml (140 KIU/ml)] containing PRP was not suppressed by heparin addition (7480+/-2410 dyn). Aprotinin [40 microg/ml (280 KIU/ml)] addition to previously heparinized plasma counteracted heparin force suppression. Aprotinin (40 microg/ml) increased platelet force from 5630 to 11,138+/-562 in PRP devoid of heparin. Aprotinin did not affect thrombin activity, fibrin structure, platelet aggregation or secretion. CONCLUSIONS Aprotinin counteracts heparin suppression of platelet force and enhances platelet force in the absence of heparin. Aprotinin-heparin-platelet interactions may help explain aprotinin's ability to reduce blood loss during CPB.
Collapse
Affiliation(s)
- Marcus E Carr
- Department of Internal Medicine, Medical College of Virginia, Virginia Commonwealth University, Richmond, 23298, USA.
| | | | | | | | | |
Collapse
|
35
|
Burysek L, Syrovets T, Simmet T. The serine protease plasmin triggers expression of MCP-1 and CD40 in human primary monocytes via activation of p38 MAPK and janus kinase (JAK)/STAT signaling pathways. J Biol Chem 2002; 277:33509-17. [PMID: 12093796 DOI: 10.1074/jbc.m201941200] [Citation(s) in RCA: 137] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The mechanism of proinflammatory activation of human monocytes by plasmin is unknown. Here we demonstrate that in human primary monocytes, plasmin stimulates mitogen-activated protein kinase (MAPK) signaling via phosphorylation of MAPK kinase 3/6 (MKK3/6) and p38 MAPK that triggers subsequent DNA binding of transcription factor activator protein-1 (AP-1). The AP-1 complex contained phosphorylated c-Jun and ATF2, and its DNA binding activity was blocked by the p38 MAPK inhibitor SB203580. In addition, plasmin elicits Janus kinase (JAK)/signal transducer and activator of transcription (STAT) signaling, as detected by phosphorylation of JAK1 tyrosine kinase and STAT1 and STAT3 proteins. Plasmin-induced DNA binding of STAT1 and STAT3 was blocked by SB203580 and AG490, inhibitors of p38 MAPK and JAK, respectively, but not by U0126, an inhibitor of MKK1/2. DNA binding of NF-kappaB remained unaffected by any of these inhibitors. The plasmin-induced signaling led to expression of monocyte chemoattractant protein-1 (MCP-1) and CD40, which required activation of both p38 MAPK and JAK/STAT signaling pathways. Additionally, signaling through both p38 MAPK and JAK is involved in the plasmin-mediated monocyte migration, whereas the formylmethionylleucylphenylalanine-induced chemotaxis remained unaffected. Taken together, our data demonstrate a novel function of the serine protease plasmin in a proinflammatory signaling network.
Collapse
Affiliation(s)
- Ladislav Burysek
- Department of Pharmacology of Natural Products and Clinical Pharmacology, University of Ulm, D-89081 Ulm, Germany
| | | | | |
Collapse
|
36
|
Horigome H, Hiramatsu Y, Shigeta O, Nagasawa T, Matsui A. Overproduction of platelet microparticles in cyanotic congenital heart disease with polycythemia. J Am Coll Cardiol 2002; 39:1072-7. [PMID: 11897453 DOI: 10.1016/s0735-1097(02)01718-7] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVES We sought to clarify the role of platelets in the pathogenesis of abnormal coagulation in patients with cyanotic congenital heart disease (CCHD) with polycythemia; we evaluated the production of platelet microparticles (MPs), platelet degranulation and aggregation response, as well as the correlations of these variables with polycythemia. BACKGROUND A shortened life span and suppressed aggregability of platelets are well known in patients with CCHD. Although platelet MPs are overproduced and play an important role in the coagulation process in various hematologic and cardiovascular disorders, the production of MPs remains to be elucidated in CCHD. We studied 19 patients who had CCHD with polycythemia and 21 age-matched subjects with acyanotic congenital heart disease (ACHD). Flow cytometry, using monoclonal antibodies, showed the presence of MPs as particles positive for the surface antigen (glycoprotein IIb/IIIa) specific to platelets, and platelet alpha-degranulation was recognized as platelets positive for the surface antigen of P-selectin. Platelet aggregation was assessed as the response to adenosine diphosphate (ADP). Relationships between these indexes and hematocrit (Hct) values were also evaluated. RESULTS Production of MPs correlated positively with Hct and markedly increased at Hct values above 60% in patients with CCHD. Surface P-selectin and the mean platelet volume in patients with CCHD were comparable with those in patients with ACHD. The platelet aggregation response to ADP significantly and negatively correlated with Hct. In two subjects who showed hemoptysis and underwent phlebotomy, MPs were reduced 6 h after the procedure. CONCLUSIONS Platelet MPs are overproduced in patients who have CCHD with polycythemia, probably due to a high shear stress derived from blood hyperviscosity. Circulating incompetent platelets, which have already been activated, as well as MPs, might play an important role in the coagulation abnormalities identified in such patients.
Collapse
Affiliation(s)
- Hitoshi Horigome
- Department of Pediatrics, Institute of Clinical Medicine, University of Tsukuba, Tsukuba, Japan.
| | | | | | | | | |
Collapse
|
37
|
Ekbäck G, Edlund B, Smolowicz A, Axelsson K, Kjellberg J, Carlsson O, Schött U. The effects of platelet apheresis in total hip replacement surgery on platelet activation. Acta Anaesthesiol Scand 2002; 46:68-73. [PMID: 11903075 DOI: 10.1046/j.0001-5172.2001.00367.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND Autologous platelet rich plasma (PRP) harvest with autotransfusion devices has been used for 10 years in cardiac surgery and recently in orthopedics as a blood saving method. The quality of the harvested platelets has not been adequately examined, in part because of methodological difficulties in studying platelet function during surgery. METHODS Twenty patients undergoing primary total hip replacement (THR) were studied. Ten patients underwent an immediate preoperative platelet apheresis to obtain concentrated platelet rich plasma (c-PRP). The other 10 patients not undergoing apheresis were allocated to a control group. Platelet activation was evaluated as the population expressing P-selectin on the surface of platelets in the c-PRP and in blood samples collected pre-, per- and postoperatively. The method used was flow cytometry. RESULTS AND CONCLUSIONS A minor population of activated platelets was found to be circulating in the patients' blood, with a highly significant difference between patients (P = 0.005), and with a range of 1-23% in peroperative activation. PRP harvest did not significantly alter platelet activity. The platelet apheresis procedure did not inhibit platelet function in the c-PRP, as judged by a high proportion of platelets that could be activated in ADP stimulation experiments (mean value +/- SD 86% +/- 7.5%).
Collapse
Affiliation(s)
- G Ekbäck
- Department of Anesthesiology, Orebro Medical Center Hospital, Orebro, Sweden.
| | | | | | | | | | | | | |
Collapse
|
38
|
Landis RC, Asimakopoulos G, Poullis M, Haskard DO, Taylor KM. The antithrombotic and antiinflammatory mechanisms of action of aprotinin. Ann Thorac Surg 2001; 72:2169-75. [PMID: 11789829 DOI: 10.1016/s0003-4975(01)02821-1] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Aprotinin (Trasylol) is generally regarded to be an effective hemostatic agent that prevents blood loss and preserves platelet function during cardiac surgery procedures requiring cardiopulmonary bypass (CBP). However, its clinical use has been limited by the concern that such a potent hemostatic agent might be prothrombotic, particularly in relation to coronary vein graft occlusion. In this review we present a mechanism of action that challenges such a viewpoint and explains how aprotinin can be simultaneously hemostatic and antithrombotic. Aprotinin achieves these two apparently disparate properties by selectively blocking the proteolytically activated thrombin receptor on platelets, the protease-activated receptor 1 (PAR1), while leaving other mechanisms of platelet aggregation unaffected. We also review recent research leading to the discovery of novel antiinflammatory targets for aprotinin. A better understanding of its mechanisms of action has led to the conclusion that aprotinin is a remarkable drug with the capacity to correct many of the imbalances that develop in the coagulation system and the inflammatory system after CPB. Nonetheless, it has been clinically underused for fear of causing thrombotic complications, a fear that in light of recent evidence may be unfounded.
Collapse
Affiliation(s)
- R C Landis
- The British Heart Foundation Unit of Cardiovascular Medicine, Hammersmith Hospital, National Heart and Lung Institute, Imperial College School of Medicine, London, England.
| | | | | | | | | |
Collapse
|
39
|
Abstract
The clinical benefit of aprotinin with respect to improved hemostasis, platelet function, and inflammatory response to cardiopulmonary bypass (CPB) surgery has been well documented, but these benefits have been overshadowed by the concern that such a potently hemostatic agent might also be prothrombotic. In this article, we discuss recent advances in the understanding of the basic mechanism of aprotinin that have led to the identification of new antiinflammatory targets and the discovery that aprotinin is, in fact, antithrombotic with respect to platelets. Its antithrombotic action is mediated by the selective blocking of the major thrombin receptor, the protease-activated receptor 1 (PAR1), but not other receptors of platelet activation (ie, collagen, adenosine diphosphate [ADP], or epinephrine receptors). The selective targeting of PAR1 enables aprotinin to protect platelets from unwanted activation by thrombin generated during CPB surgery (consistent with a role in platelet-preservation), while permitting the participation of platelets in the formation of hemostatic plugs at wound and suture sites, where collagen, ADP, and epinephrine are most likely to be expressed. Aprotinin therefore exerts a subtle hemostatic yet antithrombotic mechanism of action, which, when allied with its multitiered antiinflammatory effect, makes this drug a valuable companion to cardiac surgery.
Collapse
Affiliation(s)
- R C Landis
- National Heart and Lung Institute, Faculty of Medicine, Imperial College of Science, Technology and Medicine, London, England.
| | | | | |
Collapse
|
40
|
Hennein HA. Inflammation After Cardiopulmonary Bypass: Therapy for the Postpump Syndrome. Semin Cardiothorac Vasc Anesth 2001. [DOI: 10.1053/scva.2001.26129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Cardiopulmonary bypass (CPB) is used in most, but not all, complex heart operations. CPB is associated with a systemic inflammatory response in adults and children. Many materials-dependent (exposure of blood to non- physiologic surfaces and conditions) and materials-in dependent (surgical trauma, ischemia-perfusion to the organs, changes in body temperature, and release of endotoxin) factors during CPB have been implicated in the etiology of this complex response. The mechanisms involved may include complement activation, release of cytokines, leukocyte activation with expression of ad hesion molecules, and production of various vasoactive and immunoactive substances. Postpump inflamma tion may lead to postoperative complications and may result in respiratory failure, renal dysfunction, bleeding disorders, neurologic dysfunction, altered liver func tion, and ultimately multiple organ failure. Significant efforts are being made to decrease the generation and effects of postpump inflammation. Interventions to this end have included avoiding CPB when possible, im proving the biocompatibility of the involved mechani cal devices, and administering medications that main tain cellular integrity. This article provides an overview of the etiology, pathophysiology, and treatment of postpump inflammation. Perhaps with additional in sight into this syndrome, CPB can be made a safer and more efficacious modality of cardiorespiratory support. Copyright© 2001 by W.B. Saunders Company.
Collapse
Affiliation(s)
- Hani A. Hennein
- Department of Pediatric Cardiothoracic Surgery, Loyola University Medical Center, 2160 South First Ave, Maywood, IL 60153
| |
Collapse
|
41
|
Syrovets T, Jendrach M, Rohwedder A, Schüle A, Simmet T. Plasmin-induced expression of cytokines and tissue factor in human monocytes involves AP-1 and IKKbeta-mediated NF-kappaB activation. Blood 2001; 97:3941-50. [PMID: 11389038 DOI: 10.1182/blood.v97.12.3941] [Citation(s) in RCA: 127] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
It was previously shown that plasmin activates human peripheral monocytes in terms of lipid mediator release and chemotactic migration. Here it is demonstrated that plasmin induces proinflammatory cytokine release and tissue factor (TF) expression by monocytes. Plasmin 0.043 to 1.43 CTA U/mL, but not active site-blocked plasmin, triggered concentration-dependent expression of mRNA for interleukin-1alpha (IL-1alpha), IL-1beta, tumor necrosis factor-alpha (TNF-alpha), and TF with maximum responses after 4 hours. Plasmin-mediated mRNA expression was inhibited in a concentration-dependent manner by the lysine analogue trans-4-(aminomethyl)cyclohexane-1-carboxylic acid (t-AMCA). Increases in mRNA levels were followed by concentration- and time-dependent release of IL-1alpha, IL-1beta and TNF-alpha and by TF expression on monocyte surfaces. Neither cytokines nor TF could be detected when monocytes were preincubated with actinomycin D or cycloheximide. Electrophoretic mobility shift assays indicated plasmin-induced activation of NF-kappaB; DNA-binding complexes were composed of p50, p65, and c-Rel, as shown by supershift experiments. Nuclear translocation of NF-kappaB/Rel proteins coincided with IkappaBalpha degradation. At variance with endotoxic lipopolysaccharide, plasmin elicited the rapid degradation of another cytoplasmic NF-kappaB inhibitor, p105. Proteolysis of NF-kappaB inhibitors was apparently due to transient activation of IkappaB kinase (IKK) beta that reached maximum activity at 1 hour after plasmin stimulation. In addition, AP-1 binding was increased in plasmin-treated monocytes, with most complexes composed of JunD, c-Fos, and FosB. These findings further substantiate the role of plasmin as a proinflammatory activator of human monocytes and reveal an important new link between the plasminogen-plasmin system and inflammation. (Blood. 2001;97:3941-3950)
Collapse
Affiliation(s)
- T Syrovets
- Department of Pharmacology of Natural Products and Clinical Pharmacology, University of Ulm, Helmholtzstrasse 20, D-89081 Ulm, Germany
| | | | | | | | | |
Collapse
|
42
|
Poullis M, Manning R, Laffan M, Haskard DO, Taylor KM, Landis RC. The antithrombotic effect of aprotinin: actions mediated via the proteaseactivated receptor 1. J Thorac Cardiovasc Surg 2000; 120:370-8. [PMID: 10917956 DOI: 10.1067/mtc.2000.108531] [Citation(s) in RCA: 94] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
BACKGROUND Despite aprotinin being in widespread clinical use to prevent bleeding during cardiac surgery, there remains concern that such a powerful hemostatic agent may also be prothrombotic, particularly in relation to coronary vein graft occlusion. The major thrombin receptor on platelets, protease-activated receptor 1 (PAR1) requires proteolytic cleavage to transmit activating signals. Here we have investigated the effect of aprotinin on thrombin-induced PAR1 activation of platelets. METHODS AND RESULTS Proteolysis-dependent and -independent responses of washed platelets were studied in vitro. Platelet aggregation induced by trypsin was dependent on PAR1 (inhibited by the PAR1-specific antagonist peptide, FLLRN) and was completely blocked by aprotinin at doses more than 100 KIU/mL. Aggregation in response to thrombin, 1 nmol/L, was predominantly mediated through PAR1 and was inhibited 42.6% to 86.6% (P <.05-.001) by pharmacologic doses of aprotinin (50-160 KIU/mL). Aprotinin did not inhibit the nonproteolytic agonists collagen, epinephrine, adenosine diphosphate, or phorbol 12-myristate 13-acetate. Furthermore, blockade of the thrombin response by aprotinin did not prevent subsequent platelet aggregation through collagen or epinephrine. Experiments with intraplatelet Ca(2+) fluxes, which provided an earlier measure of platelet activation, placed the effect of aprotinin proximal to the PAR1 activation event. Since aprotinin did not inhibit platelet responses to the nonproteolytic PAR1 agonist peptide, SFLLRN, this implied that aprotinin acted by preventing PAR1 receptor cleavage by thrombin. CONCLUSIONS Aprotinin inhibits thrombin-induced platelet activation by preventing proteolysis of the PAR1 receptor. These findings argue against aprotinin being prothrombotic and suggest instead that aprotinin may have significant antithrombotic effects.
Collapse
Affiliation(s)
- M Poullis
- BHF Units of Cardiothoracic Surgery and Cardiovascular Medicine, National Heart and Lung Institute, and the Department of Haematology, Hammersmith Hospital, Imperial College School of Medicine, London, United Kingdom
| | | | | | | | | | | |
Collapse
|
43
|
Biswas AK, Lewis L, Sommerauer JF. Aprotinin in the management of life-threatening bleeding during extracorporeal life support. Perfusion 2000; 15:211-6. [PMID: 10866422 DOI: 10.1177/026765910001500305] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Contact with the synthetic surfaces of an extracorporeal circuit induces alterations in vascular components, derangements of the coagulation cascade and a systemic inflammatory response. Aprotinin reduces intraoperative and postoperative bleeding in adults undergoing cardiopulmonary bypass; however, trials in children have not had similar favorable results. While there have been some anecdotal reports, there have been no prospective clinical trials exploring the utility of aprotinin in the prevention of or as a therapy for bleeding while on extracorporeal life support (ECLS). We present a case series on our experience utilizing aprotinin for the treatment of life-threatening bleeding during ECLS. The combination of a loading dose followed by a continuous infusion resulted in significant reduction in blood loss and blood product utilization. This suggests that aprotinin may have clinical efficacy in the management of massive blood loss while on ECLS; however, larger controlled trials will be essential to determine the efficacy and appropriate dosing regimens before widespread use in ECLS can be advocated.
Collapse
Affiliation(s)
- A K Biswas
- Department of Pediatrics, Children's Medical Center of Dallas and the University of Texas Southwestern Medical Center, USA.
| | | | | |
Collapse
|
44
|
The Effects of Heparin, Protamine, and Heparinase 1 on Platelets in vitro Using Whole Blood Flow Cytometry. Anesth Analg 2000. [DOI: 10.1213/00000539-200004000-00007] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
45
|
Kozek-Langenecker SA, Mohammad SF, Masaki T, Kamerath C, Cheung AK. The effects of heparin, protamine, and heparinase 1 on platelets in vitro using whole blood flow cytometry. Anesth Analg 2000; 90:808-12. [PMID: 10735780 DOI: 10.1097/00000539-200004000-00007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
UNLABELLED The effects of heparinization and the reversal of heparin activity on platelet function after cardiopulmonary bypass have not been well defined. Flow cytometry has become a convenient and powerful technique for characterizing platelets. We examined the expression of a secretion marker (P-selectin) and an aggregation marker (activated fibrinogen receptor GP IIb-IIIa) on normal platelets in response to heparin, heparinase 1, and protamine in vitro using whole blood flow cytometry. Unfractionated heparin increased adenosine diphosphate-induced expression of P-selectin and GP IIb-IIIa in a dose-dependent manner. Heparinase 1 alone decreased both markers of platelet activation. Protamine alone increased P-selectin expression but had no effect on GP IIb-IIIa expression. Heparinase 1 antagonized the stimulatory effect of heparin on both markers. In contrast, protamine antagonized the effect of heparin on GP IIb-IIIa expression but potentiated the effect of heparin on P-selectin expression. These in vitro observations suggest that 1) both heparin and its reversal agents affect platelet secretion and aggregation, and 2) heparinase 1 reverses heparin-induced platelet preactivation more effectively than protamine. IMPLICATIONS This experimental in vitro study demonstrates that heparin and its reversal agents affect platelet secretion and aggregation.
Collapse
Affiliation(s)
- S A Kozek-Langenecker
- Department of General Anesthesiology and Intensive Care B, University of Vienna, School of Medicine, Vienna, Austria.
| | | | | | | | | |
Collapse
|
46
|
|
47
|
O'Connor CJ, Brown DV, Avramov M, Barnes S, O'Connor HN, Tuman KJ. The Impact of Renal Dysfunction on Aprotinin Pharmacokinetics During Cardiopulmonary Bypass. Anesth Analg 1999. [DOI: 10.1213/00000539-199911000-00006] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
48
|
|
49
|
Affiliation(s)
- D H Bevan
- Department of Haematology, St George's Hospital, London
| |
Collapse
|
50
|
Abstract
Platelets are the smallest of the blood cells and are known to be activated during cardiopulmonary bypass. They play a role in many associated complications. Both quantitative and qualitative platelet defects have been demonstrated, resulting in microvascular hemorrhage and thromboembolism. As their interactions with endothelium and other blood cells are unraveled, the important contribution they make toward the systemic inflammatory response to operation seen in cardiopulmonary bypass is increasingly evident. In this review, we consider platelet activation during cardiopulmonary bypass, the resultant clinical effects, and potential approaches to therapy and prevention.
Collapse
Affiliation(s)
- A Weerasinghe
- Department of Cardiothoracic Surgery, Imperial College of Science, Technology and Medicine, University of London, Hammersmith Hospital, England
| | | |
Collapse
|