151
|
Weiler H. Multiple receptor-mediated functions of activated protein C. Hamostaseologie 2012; 31:185-95. [PMID: 21826371 DOI: 10.5482/ha-1166] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2011] [Accepted: 06/17/2011] [Indexed: 01/08/2023] Open
Abstract
The central effector protease of the protein C pathway, activated protein C (APC), interacts with the endothelial cell protein C receptor, with protease activated receptors (PAR), the apolipoprotein E2 receptor, and integrins to exert multiple effects on haemostasis and immune cell function. Such receptor interactions modify the activation of PC and determine the biological response to endogenous and therapeutically administered APC. This review summarizes the current knowledge about interactions of APC with cell surface-associated receptors, novel substrates such as histones and tissue factor pathway inhibitor, and their implications for the biologic function of APC in the control of coagulation and inflammation.
Collapse
Affiliation(s)
- H Weiler
- Blood Research Institute, BloodCenter of Wisconsin, Milwaukee WI 53226, USA.
| |
Collapse
|
152
|
Conway EM. Thrombomodulin and its role in inflammation. Semin Immunopathol 2012; 34:107-25. [PMID: 21805323 DOI: 10.1007/s00281-011-0282-8] [Citation(s) in RCA: 220] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2011] [Accepted: 07/20/2011] [Indexed: 12/30/2022]
Abstract
The goal is to provide an extensive review of the physiologic role of thrombomodulin (TM) in maintaining vascular homeostasis, with a focus on its anti-inflammatory properties. Data were collected from published research. TM is a transmembrane glycoprotein expressed on the surface of all vascular endothelial cells. Expression of TM is tightly regulated to maintain homeostasis and to ensure a rapid and localized hemostatic and inflammatory response to injury. By virtue of its strategic location, its multidomain structure and complex interactions with thrombin, protein C (PC), thrombin activatable fibrinolysis inhibitor (TAFI), complement components, the Lewis Y antigen, and the cytokine HMGB1, TM exhibits a range of physiologically important anti-inflammatory, anti-coagulant, and anti-fibrinolytic properties. TM is an essential cofactor that impacts on multiple biologic processes. Alterations in expression of TM and its partner proteins may be manifest by inflammatory and thrombotic disorders. Administration of soluble forms of TM holds promise as effective therapies for inflammatory diseases, and infections and malignancies that are complicated by disseminated intravascular coagulation.
Collapse
Affiliation(s)
- Edward M Conway
- Division of Hematology-Oncology, Department of Medicine, Centre for Blood Research (CBR), University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
153
|
Genetic defects in Portuguese families with inherited protein C deficiency. Thromb Res 2011; 128:299-302. [DOI: 10.1016/j.thromres.2011.05.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2010] [Revised: 04/28/2011] [Accepted: 05/02/2011] [Indexed: 11/18/2022]
|
154
|
Yajima A, Kouno S, Dairi T, Mogi M, Katsuta R, Seto H, Nukada T. Synthesis of (±)-cyclic dehypoxanthine futalosine, the biosynthetic intermediate in an alternative biosynthetic pathway for menaquinones. Tetrahedron Lett 2011. [DOI: 10.1016/j.tetlet.2011.07.061] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
155
|
Ma L, Dorling A. The roles of thrombin and protease-activated receptors in inflammation. Semin Immunopathol 2011; 34:63-72. [PMID: 21809138 DOI: 10.1007/s00281-011-0281-9] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2011] [Accepted: 07/20/2011] [Indexed: 12/11/2022]
Abstract
Inflammation and coagulation constitute two host defence systems with complementary physiological roles in limiting tissue damage, restoring homeostasis and eliminating invading pathogens, functions reliant on effective regulation of both processes at a variety of levels. Dysfunctional activation or regulation of either pathway may lead to pathology and contribute to human diseases as diverse as myocardial infarction and septic shock. The serine protease thrombin, a key protein in the coagulation pathway, can activate cellular signalling directly via proteolytic cleavage of the N-terminal domain of a family of G protein-coupled receptors or indirectly through the generation of molecules such as activated protein C. These events transmit signals to many cell types and can elicit the production of various pro-inflammatory mediators such as cytokines, chemokines and growth factors, thereby influencing cell activation, differentiation, survival and migration. This review discusses recent progress in understanding how thrombin and protease-activated receptors influence biological processes, highlighting the detrimental and protective cellular effects of thrombin and its signalling pathways.
Collapse
Affiliation(s)
- Liang Ma
- Medical Research Council (MRC) Centre for Transplantation, King's College London, King's Health Partners, Guy's Hospital, London, UK
| | | |
Collapse
|
156
|
Kador K, Mamedov T, Schneider M, Subramanian A. Sequential co-immobilization of thrombomodulin and endothelial protein C receptor on polyurethane: activation of protein C. Acta Biomater 2011; 7:2508-17. [PMID: 21414427 DOI: 10.1016/j.actbio.2011.03.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2010] [Revised: 03/03/2011] [Accepted: 03/11/2011] [Indexed: 10/18/2022]
Abstract
In an effort to control the surface-mediated activation of thrombin and clot formation, proteins and molecules which mimic the anticoagulant properties of the vascular endothelial lining were immobilized on material surfaces. When immobilized on biomaterial surfaces, thrombomodulin (TM), an endothelial glycoprotein that binds thrombin and activates protein C (PC), was shown to generate activated PC (APC) and delay clot formation. However, TM-mediated activation of PC on biomaterial surfaces was shown to be limited by the transport of PC to the surface, with maximum activation obtained at a surface density of ∼40 fmole TM cm(-2). This work investigates surface immobilized with TM and endothelial protein C receptor (EPCR), a natural cofactor to TM which increases the rate of activation of PC on the native endothelium. A sequential and ordered immobilization of TM and EPCR on polyurethane at an enzymatically relevant distance (<10 nm) resulted in higher amounts of APC compared with surfaces with immobilized TM or with TM and EPCR immobilized randomly and at TM surface densities (1400 fmole cm(-2)) which were previously shown to be transport limited. Ordered TM and EPCR samples also showed increased time to clot formation in experiments with platelet-poor plasma, as measured by thromboelastography. Surfaces immobilized with TM and its natural cofactor EPCR at an enzymatically relevant distance are able to overcome transport limitations, increasing anticoagulant activation and time to clot formation.
Collapse
|
157
|
Bergrem A, Dahm AEA, Jacobsen AF, Mowinckel MC, Sandvik L, Sandset PM. Resistance to activated protein C is a risk factor for pregnancy-related venous thrombosis in the absence of the F5 rs6025 (factor V Leiden) polymorphism. Br J Haematol 2011; 154:241-7. [DOI: 10.1111/j.1365-2141.2011.08712.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
158
|
Zhu T, Ding Q, Bai X, Wang X, Kaguelidou F, Alberti C, Wei X, Hua B, Yang R, Wang X, Wang Z, Ruan C, Schlegel N, Zhao Y. Normal ranges and genetic variants of antithrombin, protein C and protein S in the general Chinese population. Results of the Chinese Hemostasis Investigation on Natural Anticoagulants Study I Group. Haematologica 2011; 96:1033-40. [PMID: 21486865 DOI: 10.3324/haematol.2010.037515] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Inherited deficiency of antithrombin, protein C and protein S, three important, naturally occurring coagulation inhibitors, might play a major role in the occurrence of venous thromboembolism in Chinese. The establishment of age- and gender-related normal ranges of these inhibitors is crucial for an accurate diagnosis of these deficiencies. DESIGN AND METHODS We designed a prospective cross-sectional study recruiting healthy adults from four university-affiliated hospitals in China. Antithrombin, protein C and protein S were studied by measuring their activity. Gene analysis was performed when natural anticoagulant deficiency was suspected. Polymorphisms of the factor V gene were searched for among subjects who were positive for activated protein C resistance. RESULTS In 3493 healthy Chinese adults (1734 men, 1759 women; age 17-83 years), we found higher age-adjusted activities for protein C and protein S in men than in women but no sex difference for antithrombin. In women, mean protein C and protein S activities increased with age. In men, mean protein C levels increased with age up to the age of 49 but decreased after 50 years old; mean protein S levels decreased after 50 years of age. Antithrombin levels remained stable over time in women but decreased significantly after 50 years of age in men. Reference values according to age and sex allowed the identification of 15 genetic variants (protein C:10, antithrombin:3, protein S:2) in subjects with protein activity below the 1(st) percentile. CONCLUSIONS This is the largest survey ever conducted in the healthy general Chinese population. These normal ranges provide the essential basis for the diagnosis and treatment of thrombosis in Chinese.
Collapse
Affiliation(s)
- Tienan Zhu
- Department of Hematology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, 100730 Beijing, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
159
|
Activated protein C up-regulates procoagulant tissue factor activity on endothelial cells by shedding the TFPI Kunitz 1 domain. Blood 2011; 117:6338-46. [PMID: 21474669 DOI: 10.1182/blood-2010-10-316257] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Thrombin and activated protein C (APC) signaling can mediate opposite biologic responses in endothelial cells. Given that thrombin induces procoagulant tissue factor (TF), we examined how TF activity is affected by APC. Exogenous or endogenously generated APC led to increased TF-dependent factor Xa activity. Induction required APC's proteolytic activity and binding to endothelial cell protein C receptor but not protease activated receptors. APC did not affect total TF antigen expression or the availability of anionic phospholipids on the apical cell membrane. Western blotting and cell surface immunoassays demonstrated that APC sheds the Kunitz 1 domain from tissue factor pathway inhibitor (TFPI). A TFPI Lys86Ala mutation between the Kunitz 1 and 2 domains eliminated both cleavage and the enhanced TF activity in response to APC in overexpression studies, indicating that APC up-regulates TF activity by endothelial cell protein C receptor-dependent shedding of the Kunitz 1 domain from membrane-associated TFPI. Our results demonstrate an unexpected procoagulant role of the protein C pathway that may have important implications for the regulation of TF- and TFPI-dependent biologic responses and for fine tuning of the hemostatic balance in the vascular system.
Collapse
|
160
|
Effects of long-term administration of recombinant human protein C in xenografted primates. Transplantation 2011; 91:161-8. [PMID: 21088649 DOI: 10.1097/tp.0b013e318200ba0e] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND The role potential of recombinant human activated protein C (rhaPC), a recently developed molecule with anticoagulant and antiinflammatory properties, in prolonging survival in immunosuppressed primate recipients of porcine renal xenografts has been evaluated. METHODS rhaPC was administered daily for 5 days (24 μg/kg/hr; group A; n = 3) or throughout the postoperative period (8-24 μg/kg/hr; group B; n = 2; or 24-48 μg/kg/hr; group C; n = 4). Animals in group D (n = 2) received rhaPC daily (24 μg/kg/hr) combined with recombinant human antithrombin (84 U/kg every 8 hr). Two animals served as control (group E). RESULTS The results indicate that rhaPC is protective against fibrin deposition early after transplantation but does not prevent fibrin deposition and the occurrence of acute humoral xenograft rejection (AHXR) later on. Animals in the study survived between 8 and 55 days. At the dose used, rhaPC is able to prevent fibrin deposition in the graft in the first 2 weeks after xenotransplantation, except when it is administered in conjunction with antithrombin. However, rhaPC did not prevent the eventual occurrence of AHXR in primate recipients of porcine xenografts. CONCLUSIONS In this pig to primate model, rhaPC confers a short advantage in the prevention of early perioperative xenograft damage but does not represent an effective strategy for preventing AHXR.
Collapse
|
161
|
Scaldaferri F, Lancellotti S, Pizzoferrato M, Cristofaro RD. Haemostatic system in inflammatory bowel diseases: New players in gut inflammation. World J Gastroenterol 2011; 17:594-608. [PMID: 21350708 PMCID: PMC3040331 DOI: 10.3748/wjg.v17.i5.594] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2010] [Revised: 03/25/2010] [Accepted: 04/01/2010] [Indexed: 02/06/2023] Open
Abstract
Inflammation and coagulation constantly influence each other and are constantly in balance. Emerging evidence supports this statement in acute inflammatory diseases, such as sepsis, but it also seems to be very important in chronic inflammatory settings, such as inflammatory bowel disease (IBD). Patients with Crohn’s disease and ulcerative colitis have an increased risk of thromboembolic events, and several abnormalities concerning coagulation components occur in the endothelial cells of intestinal vessels, where most severe inflammatory abnormalities occur. The aims of this review are to update and classify the type of coagulation system abnormalities in IBD, and analyze the strict and delicate balance between coagulation and inflammation at the mucosal level. Recent studies on possible therapeutic applications arising from investigations on coagulation abnormalities associated with IBD pathogenesis will also be briefly presented and critically reviewed.
Collapse
|
162
|
Raivio PM, Lassila R, Kuitunen AH, Eriksson H, Suojaranta-Ylinen RT, Petäjä J. Increased preoperative thrombin generation and low protein S level associated with unfavorable postoperative hemodynamics after coronary artery bypass grafting. Perfusion 2010; 26:99-106. [DOI: 10.1177/0267659110392442] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
In a previous study, preoperative levels of activated protein C (APC) were associated with unfavorable postoperative hemodynamics after coronary artery bypass grafting (CABG). Protein C is activated by thrombin. Protein S, the cofactor of activated protein C, has activated protein C-independent anticoagulant activity and cytoprotective effects. Therefore, the objective of this study was to test whether preoperative, baseline levels of either thrombin or protein S were associated with hemodynamic performance or markers of myocardial damage after CABG. One hundred patients undergoing elective on-pump CABG were prospectively studied. Prothrombin fragment F1+2 (a marker of thrombin generation) and free protein S were measured preoperatively and cardiac index, systemic vascular resistance index (SVRI), and pulmonary vascular resistance index (PVRI) were measured serially thereafter at fixed time points. Cardiac biomarkers CK-MBm and TnT were measured postoperatively. There was an inverse correlation between preoperative F1+2 and free protein S levels (r= —0.30, p=0.003). High preoperative F1+2 and low preoperative protein S levels were associated with a less favorable hemodynamic profile postoperatively. Patients with F1+2 in the highest decile (≥0.85 nmol/l) and patients with preoperative protein S in the lowest decile (≤63%) had lower CI values, and higher pulmonary and systemic vascular resistance index values postoperatively than comparison patients. Preoperative F1+2 or protein S did not correlate with postoperative cardiac biomarker levels. Baseline activation of coagulation and the balance between pro-coagulant and anti-coagulant factors preoperatively might have implications for postoperative hemodynamic recovery after CABG.
Collapse
Affiliation(s)
- Peter M Raivio
- Department of Cardiothoracic Surgery, Helsinki University Central Hospital, Helsinki, Finland,
| | - Riitta Lassila
- Department of Coagulation Disorders, Helsinki University Central Hospital, Helsinki, Finland, Laboratory Division (HUSLAB), Helsinki University Central Hospital, Helsinki, Finland
| | - Anne H Kuitunen
- Department of Anesthesiology and Intensive Care Medicine, Helsinki University Central Hospital, Helsinki, Finland
| | - Heidi Eriksson
- Department of Anesthesiology and Intensive Care Medicine, Helsinki University Central Hospital, Helsinki, Finland
| | - Raili T Suojaranta-Ylinen
- Department of Anesthesiology and Intensive Care Medicine, Helsinki University Central Hospital, Helsinki, Finland
| | - Jari Petäjä
- Department of Pediatrics, Helsinki University Central Hospital, Helsinki, Finland
| |
Collapse
|
163
|
The envelope glycoprotein domain III of Dengue virus type 2 induced the expression of anticoagulant molecules in endothelial cells. Mol Cell Biochem 2010; 342:215-21. [PMID: 20526654 DOI: 10.1007/s11010-010-0486-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2010] [Accepted: 05/04/2010] [Indexed: 12/25/2022]
Abstract
Dengue virus (DV) causes a non-specific febrile illness known as Dengue fever (DF), and a severe life-threatening illness, Dengue hemorrhagic fever/Dengue shock syndrome (DHF/DSS). Hemostatic changes induced by this virus involve three main factors: thrombocytopenia, endothelial cell damage, and significant abnormalities of the coagulation and fibrinolysis systems. The pathogenesis of bleeding in DV infections remains unknown. In this article, we focused on the DV activating endothelial cells and altering the parameters of hemostasis system. The expression of hemostasis-related factors, Thrombomodulin, TF, TFPI, t-PA, and PAI-1, in DV-infected cells were determined by RT-PCR. Flow cytometry analysis and immunofluorescence staining confirmed that the expression levels of TM in the DV-infected HMEC-1 and THP-1 cells were increased. In addition, the purified recombinant domain III of the envelope glycoprotein of DV (EIII) could induce the expression of TM in the HMEC-1 cells and THP-1 cells. The TM expression induced by DV or EIII in the endothelial cells and monocytic cells suggests that the EIII of DV plays an important role in the pathogenesis of DHF/DSS.
Collapse
|
164
|
Kahn NN, Feldman SP, Bauman WA. Lower-extremity functional electrical stimulation decreases platelet aggregation and blood coagulation in persons with chronic spinal cord injury: a pilot study. J Spinal Cord Med 2010; 33:150-8. [PMID: 20486534 PMCID: PMC2869270 DOI: 10.1080/10790268.2010.11689690] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Individuals with spinal cord injury (SCI) develop premature cardiovascular disease. Regular exercise reduces the incidence and symptoms of cardiovascular disease in able-bodied individuals; these salutary effects of exercise have not been documented in persons with SCI. OBJECTIVE To evaluate the effects of functional electrical stimulation leg cycle ergometry (FES-LCE) exercise training on platelet aggregation and blood coagulation in persons with SCI. PARTICIPANTS Subjects (n=14) with stable chronic (>1 year) paraplegia (T1-T10) or tetraplegia (C4-C8). METHODS Blood samples were collected before and after the first and eighth sessions (2 sessions per week for 4 weeks) of FES exercise. RESULTS Platelet aggregation was inhibited by 20% after the first session and by 40% (P < 0.001) after the eighth session. Thrombin activity was unchanged after the first session (10.7 +/- 0.85 s to 10.43 +/- 0.56 s) and decreased after the eighth session (12.5 +/- 1.98 s to 11.1 +/- 1.7 s; P < 0.0003). Antithrombin III activity increased after the first (103.8% +/- 8.9% to 110% +/- 6.9%; P < 0.0008) and eighth sessions (107.8% +/- 12.1% to 120.4% +/- 13.1%; P < 0.0001). Cyclic adenosine monophosphate increased after the first (9.9% + 2.5% to 15.8% +/- 3%; P < 0.001) and eighth sessions (17.8% +/- 4.2% to 36.5% +/- 7.6%; P < 0.0001). After the eighth session, factors V and X increased significantly (88% +/- 27% to 103% +/- 23%, P < 0.0001; 100% +/- 40% to 105% +/- 7%, P < 0.01, respectively); factors VII and VIII and fibrinogen did not change significantly. A significant reduction in platelet activation/aggregation was demonstrated in response to FES-LCE. The decrease in thrombin level was caused by the simultaneous increase in antithrombin activity. CONCLUSION These findings provide new insight into the potential protective effects of FES-LCE against the risk of cardiovascular disease.
Collapse
Affiliation(s)
- Nighat N Kahn
- Center of Excellence, James J. Peters VA Medical Center, 130 W. Kingsbridge Road, Bronx, NY 10468, USA.
| | | | - William A Bauman
- James J. Peters VA Medical Center, Bronx, New York
,Mount Sinai School of Medicine, New York, New York
| |
Collapse
|
165
|
Abstract
Activated protein C (APC) is a natural anticoagulant that plays an important role in coagulation homeostasis by inactivating the procoagulation factor Va and VIIIa. In addition to its anticoagulation functions, APC also has cytoprotective effects such as anti-inflammatory, anti-apoptotic, and endothelial barrier protection. Recently, a recombinant form of human APC (rhAPC or drotrecogin alfa activated; known commercially as 'Xigris') was approved by the US Federal Drug Administration for treatment of severe sepsis associated with a high risk of mortality. Sepsis, also known as systemic inflammatory response syndrome (SIRS) resulting from infection, is a serious medical condition in critical care patients. In sepsis, hyperactive and dysregulated inflammatory responses lead to secretion of pro- and anti-inflammatory cytokines, activation and migration of leucocytes, activation of coagulation, inhibition of fibrinolysis, and increased apoptosis. Although initial hypotheses focused on antithrombotic and profibrinolytic functions of APC in sepsis, other agents with more potent anticoagulation functions were not effective in treating severe sepsis. Furthermore, APC therapy is also associated with the risk of severe bleeding in treated patients. Therefore, the cytoprotective effects, rather than the anticoagulant effect of APC are postulated to be responsible for the therapeutic benefit of APC in the treatment of severe sepsis.
Collapse
Affiliation(s)
- Pranita P Sarangi
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | | | | |
Collapse
|
166
|
Abstract
OBJECTIVE To review new findings about the function of the protein C system during inflammation and coagulation. MAIN FINDINGS Coagulation proteases and their cofactors modify the outcome of severe inflammation by engaging signaling-competent cell surface receptors. The central effector protease of the protein C pathway, activated protein C, interacts with the endothelial cell protein C receptor, protease-activated receptors, and other receptors to exert multiple effects on hemostasis and immune cell function. Thrombomodulin controls the complement arm of the innate immune system in a thrombin-dependent manner through activation of the thrombin activatable inhibitor of fibrinolysis, and in a thrombin-independent, constitutive manner via its lectin-like extracellular domain; and inhibits the inflammatory effects of high-mobility box group 1 protein. Protein S not only suppresses coagulation as an enhancing cofactor for the coagulation inhibitors activated protein C and tissue factor pathway inhibitor but also is also a physiologic ligand for the Tyro/axl/Mer-family of receptor tyrosine kinases that mediate an anti-inflammatory regulatory loop of dendritic cell and monocyte inflammatory function. CONCLUSIONS The immune-regulatory capacity of the protein C pathway and its individual components emerge as the dominant action of this pathway in the setting of severe inflammation.
Collapse
|
167
|
Horai Y, Miyamura T, Hirata A, Nakamura M, Takahama S, Ando H, Minami R, Yamamoto M, Suematsu E. Idiopathic portal hypertension in a patient with mixed connective tissue disease and protein C deficiency. Intern Med 2010; 49:2013-6. [PMID: 20847509 DOI: 10.2169/internalmedicine.49.3834] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We report a 29-year-old woman with a 2.5 year history of mixed connective tissue disease (MCTD) who developed idiopathic portal hypertension (IPH) and thrombocytopenia as a result of hypersplenism. She had recurrent esophagogastric variceal rupture. Hematological examination also revealed low levels of protein C activity. The liver biopsy specimen showed non-specific mild inflammation and no thrombi. However, portal vein thrombosis developed after splenectomy. This was a rare case of severe complications of IPH accompanying MCTD and protein C deficiency.
Collapse
Affiliation(s)
- Yoshiro Horai
- Department of Internal Medicine and Rheumatology, Clinical Research Center, National Hospital Organization Kyushu Medical Center, Fukuoka.
| | | | | | | | | | | | | | | | | |
Collapse
|
168
|
Vitamin-K-abhängige Gerinnungsfaktoren. Hamostaseologie 2010. [DOI: 10.1007/978-3-642-01544-1_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
169
|
Activated Protein C in Ischemia-Reperfusion Injury After Experimental Lung Transplantation. J Heart Lung Transplant 2009; 28:1180-4. [DOI: 10.1016/j.healun.2009.06.026] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2009] [Revised: 06/19/2009] [Accepted: 06/26/2009] [Indexed: 11/21/2022] Open
|
170
|
Panwar R, Venkatesh B, Kruger P, Bird R, Gill D, Nunnink L, Dimeski G. Plasma protein C levels in immunocompromised septic patients are significantly lower than immunocompetent septic patients: a prospective cohort study. J Hematol Oncol 2009; 2:43. [PMID: 19840396 PMCID: PMC2772189 DOI: 10.1186/1756-8722-2-43] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2009] [Accepted: 10/19/2009] [Indexed: 12/30/2022] Open
Abstract
Introduction Activated Protein C [APC] improves outcome in immunocompetent patients with severe sepsis particularly in those who are perceived to have high mortality risk. Before embarking on a trial of APC administration in immunocompromised septic patients, a preliminary study on plasma levels of protein C in this cohort is essential. Objective To assess serum Protein C concentrations in immunocompromised patients as compared to immunocompetent patients during sepsis, severe sepsis, septic shock and recovery. Methods Prospective cohort study in a tertiary hospital. Patients satisfying inclusion criteria were enrolled after informed consent. Clinical variables were noted with sample collection when patients met criteria for sepsis, severe sepsis, septic shock and recovery. Protein C levels were measured using monoclonal antibody based fluorescence immunoassay. Results Thirty one patients participated in this study (22 immunocompromised, 9 immunocompetent). Protein C levels were found to be significantly lower in the immunocompromised group compared to the immunocompetent group, particularly observed in severe sepsis [2.27 (95% CI: 1.63-2.9) vs 4.19 (95% CI: 2.87-5.52) mcg/ml] (p = 0.01) and sepsis [2.59 (95% CI: 1.98-3.21) vs 3.64 (95% CI: 2.83-4.45) mcg/ml] (p = 0.03). SOFA scores were similar in both the groups across sepsis, severe sepsis and septic shock categories. Protein C levels improved significantly in recovery (p = 0.001) irrespective of immune status. Conclusion Protein C levels were significantly lower in immunocompromised patients when compared to immunocompetent patients in severe sepsis and sepsis categories. Our study suggests a plausible role for APC in severely septic immunocompromised patients which need further elucidation.
Collapse
Affiliation(s)
- Rakshit Panwar
- Intensive Care, Monash Medical Centre, Clayton, VIC 3168, Australia.
| | | | | | | | | | | | | |
Collapse
|
171
|
Krisinger MJ, Guo LJ, Salvagno GL, Guidi GC, Lippi G, Dahlbäck B. Mouse recombinant protein C variants with enhanced membrane affinity and hyper-anticoagulant activity in mouse plasma. FEBS J 2009; 276:6586-602. [PMID: 19817854 DOI: 10.1111/j.1742-4658.2009.07371.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Mouse anticoagulant protein C (461 residues) shares 69% sequence identity with its human ortholog. Interspecies experiments suggest that there is an incompatibility between mouse and human protein C, such that human protein C does not function efficiently in mouse plasma, nor does mouse protein C function efficiently in human plasma. Previously, we described a series of human activated protein C (APC) Gla domain mutants (e.g. QGNSEDY-APC), with enhanced membrane affinity that also served as superior anticoagulants. To characterize these Gla mutants further in mouse models of diseases, the analogous mutations were now made in mouse protein C. In total, seven mutants (mutated at one or more of positions P(10)S(12)D(23)Q(32)N(33)) and wild-type protein C were expressed and purified to homogeneity. In a surface plasmon resonance-based membrane-binding assay, several high affinity protein C mutants were identified. In Ca(2+) titration experiments, the high affinity variants had a significantly reduced (four-fold) Ca(2+) requirement for half-maximum binding. In a tissue factor-initiated thrombin generation assay using mouse plasma, all mouse APC variants, including wild-type, could completely inhibit thrombin generation; however, one of the variants denoted mutant III (P10Q/S12N/D23S/Q32E/N33D) was found to be a 30- to 50-fold better anticoagulant compared to the wild-type protein. This mouse APC variant will be attractive to use in mouse models aiming to elucidate the in vivo effects of APC variants with enhanced anticoagulant activity.
Collapse
Affiliation(s)
- Michael J Krisinger
- Department of Laboratory Medicine, Division of Clinical Chemistry, Lund University, University Hospital, Malmö, Sweden
| | | | | | | | | | | |
Collapse
|
172
|
Guermazi S, Conard J. [Congenital protein S deficiencies; diagnostic difficulties]. PATHOLOGIE-BIOLOGIE 2009; 57:483-487. [PMID: 18583066 DOI: 10.1016/j.patbio.2008.04.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2008] [Accepted: 04/16/2008] [Indexed: 05/26/2023]
Abstract
Protein S is a physiologic inhibitor of coagulation acting as a cofactor of activated protein C (APC) that inhibits factor Va and VIII. Approximately 60% of PS is bound to C4bBP, a protein of the complement system and only the free PS has a cofactor PCa role. Congenital PS deficiencies are diagnosed by immunologic dosage of free and total PS and functional assay evaluating APC cofactor activity. However, it has been demonstrated a direct anticoagulant activity of free PS, non-dependant of APC on the cascade coagulation and even PS bound to C4bBP seems to have anticoagulant properties. So, it appears that functional assays available estimate only a part of PS anticoagulant activities and, in addition, many interferences are reported with these tests (lupus anticoagulant, factor V Leiden, factor VIII excess...). Immunologic dosages are more reliable in spite of rare qualitative PS deficiencies that could be non-diagnosed. PS deficiencies are often difficult to diagnose because of an overlapping between normal and pathological values. Familial studies are necessary to prove the hereditary origin because there are several causes of acquired and sometimes persistent PS deficiencies (liver insufficiency, vitamin K absence, hormonal therapy in women, PS auto immune deficiency). About 200 different mutations were retrieved and, therefore, molecular studies are not of current practice. It is recommended currently to measure in first intention the free PS, if possible in association with PCa cofactor activity.
Collapse
Affiliation(s)
- S Guermazi
- Laboratoire d'hématologie, hôpital Charles-Nicolle, 01006 Tunis, Tunisie.
| | | |
Collapse
|
173
|
Abstract
Severe protein C deficiency (i.e. protein C activity <1 IU dL(-1)) is a rare autosomal recessive disorder that usually presents in the neonatal period with purpura fulminans (PF) and severe disseminated intravascular coagulation (DIC), often with concomitant venous thromboembolism (VTE). Recurrent thrombotic episodes (PF, DIC, or VTE) are common. Homozygotes and compound heterozygotes often possess a similar phenotype of severe protein C deficiency. Mild (i.e. simple heterozygous) protein C deficiency, by contrast, is often asymptomatic but may involve recurrent VTE episodes, most often triggered by clinical risk factors. The coagulopathy in protein C deficiency is caused by impaired inactivation of factors Va and VIIIa by activated protein C after the propagation phase of coagulation activation. Mutational analysis of symptomatic patients shows a wide range of genetic mutations. Management of acute thrombotic events in severe protein C deficiency typically requires replacement with protein C concentrate while maintaining therapeutic anticoagulation; protein C replacement is also used for prevention of these complications around surgery. Long-term management in severe protein C deficiency involves anticoagulation with or without a protein C replacement regimen. Although many patients with severe protein C deficiency are born with evidence of in utero thrombosis and experience multiple further events, intensive treatment and monitoring can enable these individuals to thrive. Further research is needed to better delineate optimal preventive and therapeutic strategies.
Collapse
Affiliation(s)
- N A Goldenberg
- Hemophilia & Thrombosis Center, Department of Pediatrics, University of Colorado Denver and The Children's Hospital, Aurora, CO, USA
| | | |
Collapse
|
174
|
Shao WH, Zhen Y, Eisenberg RA, Cohen PL. The Mer receptor tyrosine kinase is expressed on discrete macrophage subpopulations and mainly uses Gas6 as its ligand for uptake of apoptotic cells. Clin Immunol 2009; 133:138-44. [PMID: 19631584 DOI: 10.1016/j.clim.2009.06.002] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2009] [Revised: 05/21/2009] [Accepted: 06/04/2009] [Indexed: 12/22/2022]
Abstract
The Mer receptor tyrosine kinase is both an important mediator of apoptotic cell phagocytosis and a regulator of macrophage and DC cytokine production. Since phenotypically distinguishable macrophages are known to have different functions, we have examined Mer expression of murine splenic macrophages. We also used serum deficient in the Mer ligand, growth arrest-specific protein 6 (Gas6) to define better the role of this Mer ligand in macrophage function. By immunofluorescence staining, we found Mer to be strongly expressed in splenic red pulp, largely on platelets. We also found Mer expression on marginal zone macrophages. Strikingly, all tingible body macrophages bore Mer. In functional phagocytosis assays of apoptotic cells, Gas6 appeared to be the sole ligand for Mer, and this system accounted for about 30% of splenic macrophage phagocytosis of apoptotic cells. Taken together, the expression pattern of Mer on macrophage subpopulations in the spleen and its Gas6-dependent role in macrophage phagocytosis suggest an important role for Mer in the modulation of immune responses.
Collapse
Affiliation(s)
- Wen-Hai Shao
- Department of Medicine, Division of Rheumatology, Temple University, Philadelphia, PA 19140, USA
| | | | | | | |
Collapse
|
175
|
An alternative menaquinone biosynthetic pathway operating in microorganisms: an attractive target for drug discovery to pathogenic Helicobacter and Chlamydia strains. J Antibiot (Tokyo) 2009; 62:347-52. [PMID: 19557031 DOI: 10.1038/ja.2009.46] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Menaquinone is an essential vitamin as an obligatory component of the electron transfer pathway in microorganisms. Menaquinone has been shown to be derived from chorismate by eight enzymes, designated MenA to -H in Escherichia coli. However, bioinformatic analyses of whole-genome sequences have suggested that some microorganisms, such as Helicobacter pylori and Campylobacter jejuni, which are known to cause gastric carcinoma and diarrhea, respectively, do not have orthologs of most of the men genes, although they synthesize menaquinone. The (13)C-labeling pattern of menaquinone purified from Streptomyces coelicolor A3(2) grown on [U-(13)C]glucose was quite different from that of E. coli, suggesting that an alternative pathway was operating in the strain. We searched for candidate genes participating in the alternative pathway by in silico screening, and the involvement of these genes in the pathway was confirmed by gene-disruption experiments. We also used mutagenesis to isolate mutants that required menaquinone for their growth and used these mutants as hosts for shotgun cloning experiments. Metabolites that accumulated in the culture broth of mutants were isolated and their structures were determined. Taking these results together, we deduced the outline of the alternative pathway, which branched at chorismate in a similar manner to the known pathway but then followed a completely different pathway. As humans and some useful intestinal bacteria, such as lactobacilli, lack the alternative pathway, it would be an attractive target for the development of chemotherapeutics.
Collapse
|
176
|
An exosite-specific ssDNA aptamer inhibits the anticoagulant functions of activated protein C and enhances inhibition by protein C inhibitor. ACTA ACUST UNITED AC 2009; 16:442-51. [PMID: 19389630 DOI: 10.1016/j.chembiol.2009.03.007] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2008] [Revised: 02/25/2009] [Accepted: 03/05/2009] [Indexed: 11/22/2022]
Abstract
Activated protein C (APC) is a serine protease with anticoagulant, anti-inflammatory, and cytoprotective properties. Using recombinant APC, we identified a class of single-stranded DNA aptamers (HS02) that selectively bind to APC with high affinity. Interaction of HS02 with APC modulates the protease activity in a way such that the anticoagulant functions of APC are inhibited and its reactivity toward the protein C inhibitor is augmented in a glysoaminoglycan-like fashion, whereas APC's antiapoptotic and cytoprotective functions remain unaffected. Based on these data, the binding site of HS02 was localized to the basic exosite of APC. These characteristics render the exosite-specific aptamers a promising tool for the development of APC assays and a potential therapeutic agent applicable for the selective control of APC's anticoagulant activity.
Collapse
|
177
|
Danese S. Nonimmune cells in inflammatory bowel disease: from victim to villain. Trends Immunol 2009; 29:555-64. [PMID: 18838297 DOI: 10.1016/j.it.2008.07.009] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2008] [Revised: 07/21/2008] [Accepted: 07/23/2008] [Indexed: 12/13/2022]
Abstract
Nonimmune cells have traditionally been viewed as target cells of the aberrant inflammatory process present in chronic immune-mediated conditions such as inflammatory bowel disease (IBD). However, the discovery that many of the functions traditionally attributed to immune cells are also performed by nonimmune cells has caused a shift to a multidirectional hypothesis in which nonimmune cells and acellular elements play active roles. Many types of interactions occur within this multidirectional system, and the difficulties associated with modeling these complex interactions currently limit our understanding of the cellular network that occurs in IBD. I describe the current knowledge of the roles played by nonimmune cells in the pathogenesis of IBD, as they emerge as crucial alternative targets for therapeutic intervention.
Collapse
Affiliation(s)
- Silvio Danese
- Division of Gastroenterology, Istituto Clinico Humanitas, IRCCS in Gastroenterology, Rozzano, Milan 20089, Italy.
| |
Collapse
|
178
|
Mosnier LO, Zampolli A, Kerschen EJ, Schuepbach RA, Banerjee Y, Fernández JA, Yang XV, Riewald M, Weiler H, Ruggeri ZM, Griffin JH. Hyperantithrombotic, noncytoprotective Glu149Ala-activated protein C mutant. Blood 2009; 113:5970-8. [PMID: 19244160 PMCID: PMC2700330 DOI: 10.1182/blood-2008-10-183327] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Activated protein C (APC) reduces mortality in severe sepsis patients. APC exerts anticoagulant activities via inactivation of factors Va and VIIIa and cytoprotective activities via endothelial protein C receptor and protease-activated receptor-1. APC mutants with selectively altered and opposite activity profiles, that is, greatly reduced anticoagulant activity or greatly reduced cytoprotective activities, are compared here. Glu149Ala-APC exhibited enhanced in vitro anticoagulant and in vivo antithrombotic activity, but greatly diminished in vitro cytoprotective effects and in vivo reduction of endotoxin-induced murine mortality. Thus, residue Glu149 and the C-terminal region of APC's light chain are identified as functionally important for expression of multiple APC activities. In contrast to Glu149Ala-APC, 5A-APC (Lys191-193Ala + Arg229/230Ala) with protease domain mutations lacked in vivo antithrombotic activity, although it was potent in reducing endotoxin-induced mortality, as previously shown. These data imply that APC molecular species with potent antithrombotic activity, but without robust cytoprotective activity, are not sufficient to reduce mortality in endotoxemia, emphasizing the need for APC's cytoprotective actions, but not anticoagulant actions, to reduce endotoxin-induced mortality. Protein engineering can provide APC mutants that permit definitive mechanism of action studies for APC's multiple activities, and may also provide safer and more effective second-generation APC mutants with reduced bleeding risk.
Collapse
Affiliation(s)
- Laurent O Mosnier
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
179
|
Chaireti R, Jennersjö C, Lindahl TL. Thrombin generation and D-dimer concentrations in a patient cohort investigated for venous thromboembolism. Relations to venous thrombosis, factor V Leiden and prothrombin G20210A. The LIST study. Thromb Res 2009; 124:178-84. [DOI: 10.1016/j.thromres.2008.12.033] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2008] [Revised: 10/22/2008] [Accepted: 12/09/2008] [Indexed: 11/24/2022]
|
180
|
Neyrinck AP, Liu KD, Howard JP, Matthay MA. Protective mechanisms of activated protein C in severe inflammatory disorders. Br J Pharmacol 2009; 158:1034-47. [PMID: 19466992 DOI: 10.1111/j.1476-5381.2009.00251.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The protein C system is an important natural anticoagulant mechanism mediated by activated protein C (APC) that regulates the activity of factors VIIIa and Va. Besides well-defined anticoagulant properties, APC also demonstrates anti-inflammatory, anti-apoptotic and endothelial barrier-stabilizing effects that are collectively referred to as the cytoprotective effects of APC. Many of these beneficial effects are mediated through its co-receptor endothelial protein C receptor, and the protease-activated receptor 1, although exact mechanisms remain unclear and are likely pleiotropic in nature. Increased insight into the structure-function relationships of APC facilitated design of APC variants that conserve cytoprotective effects and reduce anticoagulant features, thereby attenuating the risk of severe bleeding with APC therapy. Impairment of the protein C system plays an important role in acute lung injury/acute respiratory distress syndrome and severe sepsis. The pathophysiology of both diseases states involves uncontrolled inflammation, enhanced coagulation and compromised fibrinolysis. This leads to microvascular thrombosis and organ injury. Administration of recombinant human APC to correct the dysregulated protein C system reduced mortality in severe sepsis patients (PROWESS trial), which stimulated further research into its mechanisms of action. Several other clinical trials evaluating recombinant human APC have been completed, including studies in children and less severely ill adults with sepsis as well as a study in acute lung injury. On the whole, these studies have not supported the use of APC in these populations and challenge the field of APC research to search for additional answers.
Collapse
Affiliation(s)
- Arne P Neyrinck
- University of California San Francisco, Cardiovascular Research Institute, San Francisco, CA, USA
| | | | | | | |
Collapse
|
181
|
Schuepbach RA, Feistritzer C, Fernández JA, Griffin JH, Riewald M. Protection of vascular barrier integrity by activated protein C in murine models depends on protease-activated receptor-1. Thromb Haemost 2009; 101:724-33. [PMID: 19350118 PMCID: PMC2680237 DOI: 10.1160/th08-10-0632] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Protease activated receptor-1 (PAR1) mediates barrier protective signalling of activated protein C (APC) in human endothelial cells in vitro and may contribute to APC's beneficial effects in patients with severe sepsis. Mouse models are of key importance for translational research but species differences may limit conclusions for the human system. We analysed whether mouse APC can cleave, activate and induce signalling through murine PAR1 and tested in newly established mouse models if long-term infusion of APC prevents from vascular leakage. Cell surface immunoassays demonstrated efficient cleavage of endogenous murine endothelial PAR1 by either murine or human APC. Pharmacological concentrations of APC of either species had powerful barrier protective effects on cultured murine endothelial cells that required PAR1 cleavage. Vascular endothelial growth factor-mediated hyperpermeability in the skin was reduced by either endogenously generated as well as directly infused recombinant mouse APC in wild-type mice. However APC did not significantly alter the vascular barrier function in PAR1-deficient mice. In endotoxin-challenged mice, infused APC significantly prevented from pulmonary fluid accumulation in the wild-type mice but not in mice lacking PAR1. Our results directly show that murine APC cleaves and signals through PAR1 in mouse endothelial cells. APC reduces vascular permeability in mouse models and PAR1 plays a major role in mediating these effects. Our data in vitro and in vivo support the paradigm that PAR1 contributes to protective effects of APC on vascular barrier integrity in sepsis.
Collapse
Affiliation(s)
- Reto A. Schuepbach
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, USA
| | - Clemens Feistritzer
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, USA
| | - José A. Fernández
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, USA
| | - John H. Griffin
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, USA
| | - Matthias Riewald
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, USA
| |
Collapse
|
182
|
Inherited protein C deficiency with acute ischemic stroke in a young adult: a case report. Blood Coagul Fibrinolysis 2008; 19:601-4. [PMID: 18685446 DOI: 10.1097/mbc.0b013e3283009c7c] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Hereditary protein C deficiency is inherited primarily as an autosomal dominant trait with incomplete penetrance. Arterial thrombosis, especially of the intracranial arteries, due to this deficiency is relatively rare. A 31-year-old man was admitted to our department because of sudden onset of neurological symptoms. Magnetic resonance imaging of the brain disclosed an acute ischemic infarction of the area supplied by the left middle cerebral artery. Protein C antigen was 40.7% (77-129%) and protein C activity was 46.3% (70-140%). No other possible associated causes of stroke were present. A survey of his relatives for protein C deficiency showed this deficiency in his mother, brother, sister and nephews. Protein C concentrations should be determined in cases of ischemic stroke in all young patients with no other major risk factors. Once protein C deficiency is detected, a search for protein C deficiency in the patient's relatives should be performed to prevent the occurrence of ischemic strokes.
Collapse
|
183
|
Abstract
PURPOSE OF REVIEW To discuss the potential use of recombinant activated protein C (aPC) variants with altered bioactivity in sepsis therapy. RECENT FINDINGS Since the initial Protein C Worldwide Evaluation in Severe Sepsis trial demonstrating efficacy of aPC therapy to reduce mortality of severe sepsis, follow-up studies have failed to resolve concerns about the low overall risk-to-benefit ratio of this therapy and suggest that it might only be effective in severely ill patients with the most aggravated forms of coagulopathy. New studies begin to shed light on the potential mechanisms of how aPC therapy may alter sepsis outcome, and how recombinant aPC variants with altered bioactivities may improve the efficacy and safety of this therapy. SUMMARY aPC variants with selectively diminished antithrombotic activity, but normal cytoprotective potential, may allow more efficient dosing without increasing adverse bleeding effects and therefore provide a safer and possibly more efficient alternative to normal aPC. Critical questions about the precise mechanisms by which aPC therapy reduces mortality remain to be resolved in order to identify patients most likely to benefit from it and to reevaluate potential efficacy of aPC therapy in children and patients with less than severe sepsis.
Collapse
|
184
|
Rinaldi L, Marietta M, Mignini MA, Donno L, Busani S, Codeluppi M, Masetti M, Girardis M. Use of activated protein C in liver transplantation patients with septic shock. Liver Transpl 2008; 14:1598-602. [PMID: 18972572 DOI: 10.1002/lt.21589] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Recombinant human activated protein C (rhAPC) has been approved for use in patients with severe sepsis at high risk of death. Because of the high risk of bleeding, liver transplantation (LT) patients have been excluded from the randomized control trials that evaluated efficacy and safety of rhAPC and, thus, few data are available on the use of this drug in LT patients with severe sepsis. We describe our experience with 5 LT recipients treated for septic shock with the best conventional therapy and rhAPC. Before rhAPC therapy, all the patients showed septic shock, with > or =3 organ dysfunctions and thrombocytopenia with impairment of coagulation. rhAPC therapy started within 30 hours after septic shock onset in all the patients who recovered from sepsis-induced circulatory failure, improved organ dysfunction, and completed the 96 hours of rhAPC therapy. During rhAPC infusion, 4 patients received fresh frozen plasma and/or platelet concentrates because of thrombocytopenia and severe hemostasis dysfunction. No major bleeding occurred and only 1 patient presented with minor bleeding events.
Collapse
Affiliation(s)
- Laura Rinaldi
- Department of Anaesthesiology and Intensive Care Unit, University of Modena and Reggio Emilia, University Hospital of Modena [corrected] Eli-Lilly Italia, Sesto Fiorentino, Italy
| | | | | | | | | | | | | | | |
Collapse
|
185
|
Malm K, Arnljots B, Dahlbäck B. Human activated protein C variants in a rat model of arterial thrombosis. Thromb J 2008; 6:16. [PMID: 18957140 PMCID: PMC2584025 DOI: 10.1186/1477-9560-6-16] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2008] [Accepted: 10/29/2008] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Activated protein C (APC) inhibits coagulation by degrading activated factor V (FVa) and factor VIII (FVIIIa), protein S (PS) functioning as a cofactor to APC. METHODS By mutagenesis of the vitamin K-dependent Gla domain of APC, we have recently created an APC variant having enhanced anticoagulant activity due to increased affinity for negatively charged phospholipid membranes. In the present study, the potential antithrombotic effects of this APC variant, and of a variant APC that is additionally mutated in the serine protease domain, have been evaluated in a blind randomized study in a rat model of arterial thrombosis. In this model, we have previously found the combination of bovine APC and PS to be highly antithrombotic. Four treatment groups each containing 10 rats were, in a blind random fashion, given intravenous bolus injections of wild-type or mutant variants of APC (0.8 mg/kg) together with human PS (0.6 mg/kg) or human PS (0.6 mg/kg) alone. A control group with 20 animals where given vehicle only. RESULTS A trend to increased patency rates was noted in a group receiving one of the APC variants, but it did not reach statistical significance. CONCLUSION In conclusion, administration of human APC variants having enhanced anticoagulant efficacy together with human PS in a rat model of arterial thrombosis did not give an efficient antithrombotic effect. The lack of effect may be due to species-specific differences between the human protein C system and the rat hemostatic system.
Collapse
Affiliation(s)
- Karl Malm
- Department of Laboratory Medicine, Division of Clinical Chemistry, Lund University, University Hospital, SE-20502 Malmö, Sweden.
| | | | | |
Collapse
|
186
|
Abstract
Sepsis, a systemic inflammatory response to infection, is a leading cause of death in intensive care units. Recent investigations into the pathogenesis of sepsis reveal a biphasic inflammatory process. An early phase is characterized by pro-inflammatory cytokines (e.g. tumour necrosis factor-alpha), whereas a late phase is mediated by an inflammatory high-mobility group box 1 and an anti-inflammatory interleukin-10. Inflammation aberrantly activates coagulation cascades as sepsis progresses. This dual inflammatory response concomitant with dysregulated coagulation partially accounts for unsuccessful anti-cytokine therapies that have solely targeted early pro-inflammatory mediators (e.g. tumour necrosis factor-alpha). In contrast, activated protein C, which modifies both inflammatory and coagulatory pathways, has improved survival in patients in severe sepsis. Inhibition of the late mediator high-mobility group box 1 improves survival in established sepsis in pre-clinical studies. In addition, recent advances in molecular medicine have shed light on two novel experimental interventions against sepsis. Accelerated apoptosis of lymphocytes has been shown to play an important role in organ dysfunction in sepsis and techniques to suppress apoptosis have improved survival rate in sepsis models. The vagus nerve system has also been shown to suppress innate immune response through endogenous release and exogenous administration of cholinergic agonists, ameliorating inflammation and lethality in sepsis models.
Collapse
|
187
|
Linger RMA, Keating AK, Earp HS, Graham DK. TAM receptor tyrosine kinases: biologic functions, signaling, and potential therapeutic targeting in human cancer. Adv Cancer Res 2008; 100:35-83. [PMID: 18620092 DOI: 10.1016/s0065-230x(08)00002-x] [Citation(s) in RCA: 567] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Tyro-3, Axl, and Mer constitute the TAM family of receptor tyrosine kinases (RTKs) characterized by a conserved sequence within the kinase domain and adhesion molecule-like extracellular domains. This small family of RTKs regulates an intriguing mix of processes, including cell proliferation/survival, cell adhesion and migration, blood clot stabilization, and regulation of inflammatory cytokine release. Genetic or experimental alteration of TAM receptor function can contribute to a number of disease states, including coagulopathy, autoimmune disease, retinitis pigmentosa, and cancer. In this chapter, we first provide a comprehensive review of the structure, regulation, biologic functions, and downstream signaling pathways of these receptors. In addition, we discuss recent evidence which suggests a role for TAM receptors in oncogenic mechanisms as family members are overexpressed in a spectrum of human cancers and have prognostic significance in some. Possible strategies for targeted inhibition of the TAM family in the treatment of human cancer are described. Further research will be necessary to evaluate the full clinical implications of TAM family expression and activation in cancer.
Collapse
Affiliation(s)
- Rachel M A Linger
- Department of Pediatrics, University of Colorado at Denver and Health Sciences Center, Aurora, CO, USA
| | | | | | | |
Collapse
|
188
|
Association of endothelial protein C receptor haplotypes, factor V Leiden and recurrent first trimester pregnancy loss. Clin Biochem 2008; 41:1022-4. [DOI: 10.1016/j.clinbiochem.2008.05.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2008] [Revised: 05/03/2008] [Accepted: 05/13/2008] [Indexed: 11/21/2022]
|
189
|
Abstract
Venous thromboembolism is a major medical problem, annually affecting 1 in 1000 individuals. It is a typical multifactorial disease, involving both genetic and circumstantial risk factors that affect a delicate balance between procoagulant and anticoagulant forces. In the last 50 years, the molecular basis of blood coagulation and the anticoagulant systems that control it have been elucidated. This has laid the foundation for discoveries of both common and rare genetic traits that tip the natural balance in favor of coagulation, with a resulting lifelong increased risk of venous thrombosis. Multiple mutations in the genes for anticoagulant proteins such as antithrombin, protein C, and protein S have been identified and constitute important risk factors. Two single mutations in the genes for coagulation factor V (FV Leiden) and prothrombin (20210G>A), resulting from approximately 20,000-year-old mutations with subsequent founder effects, are common in the general population and constitute major genetic risk factors for thrombosis. In celebration of the 50-year anniversary of the American Society of Hematology, this invited review highlights discoveries that have contributed to our present understanding of the systems that control blood coagulation and the genetic factors that are involved in the pathogenesis of venous thrombosis.
Collapse
|
190
|
Steen M, Tran S, Autin L, Villoutreix BO, Tholander AL, Dahlbäck B. Mapping of the factor Xa binding site on factor Va by site-directed mutagenesis. J Biol Chem 2008; 283:20805-12. [PMID: 18502757 DOI: 10.1074/jbc.m802703200] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Activated coagulation factor V functions as a cofactor to factor Xa in the conversion of prothrombin to thrombin. Based on the introduction of extra carbohydrate side chains in recombinant factor V, we recently proposed several regions in factor Va to be important for factor Xa binding. To further define which residues are important for factor Xa binding, we prepared fifteen recombinant factor V variants in which clusters of charged amino acid residues were mutated, mainly to alanines. The factor V variants were expressed in COS-1 cells, and their functional properties evaluated in a prothrombinase-based assay, as well as in a direct binding test. Four of the factor V variants, 501A/510A/511D, 501A/510A/511D/513A, 513A/577A/578A, and 501A/510A/511D/513A/577A/578A exhibited markedly reduced factor Xa-cofactor activity tested in the prothrombinase assay, and reduced binding affinity as judged by the direct binding assay. These factor Va variants were normally cleaved at Arg-506 by activated protein C, and the interaction between the factor Xa-factor Va complex and prothrombin was unaffected by the introduced mutations. Based on the integration of all available data, we propose a key factor Xa binding surface to be centered on Arg-501, Arg-510, Ala-511, Asp-513, Asp-577, and Asp-578 in the factor Va A2 domain. These residues form an elongated charged factor Xa binding cluster on the factor Va surface.
Collapse
Affiliation(s)
- Mårten Steen
- Department of Laboratory Medicine, Division of Clinical Chemistry, Lund University, The Wallenberg Laboratory, MAS, SE-205 02 Malmö, Sweden
| | | | | | | | | | | |
Collapse
|
191
|
Maternal Par4 and platelets contribute to defective placenta formation in mouse embryos lacking thrombomodulin. Blood 2008; 112:585-91. [PMID: 18490515 DOI: 10.1182/blood-2007-09-111302] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Absence of the blood coagulation inhibitor thrombomodulin (Thbd) from trophoblast cells of the mouse placenta causes a fatal arrest of placental morphogenesis. The pathogenesis of placental failure requires tissue factor, yet is not associated with increased thrombosis and persists in the absence of fibrinogen. Here, we examine the role of alternative targets of coagulation that might contribute to the placental failure and death of Thbd(-/-) embryos. We demonstrate that genetic deficiency of the protease-activated receptors, Par1 or Par2, in the embryo and trophoblast cells does not prevent the death of Thbd(-/-) embryos. Similarly, genetic ablation of the complement pathway or of maternal immune cell function does not decrease fetal loss. In contrast, Par4 deficiency of the mother, or the absence of maternal platelets, restores normal development in one-third of Thbd-null embryos. This finding generates new evidence implicating increased procoagulant activity and thrombin generation in the demise of thrombomodulin-null embryos, and suggests that platelets play a more prominent role in placental malfunction associated with the absence of thrombomodulin than fibrin formation. Our findings demonstrate that fetal prothrombotic mutations can cause localized activation of maternal platelets at the feto-maternal interface in a mother with normal hemostatic function.
Collapse
|
192
|
Bellido-Martín L, de Frutos PG. Vitamin K-dependent actions of Gas6. VITAMINS AND HORMONES 2008; 78:185-209. [PMID: 18374195 DOI: 10.1016/s0083-6729(07)00009-x] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Gas6 (growth arrest-specific gene 6) is the last addition to the family of plasma vitamin K-dependent proteins. Gas6 was cloned and characterized in 1993 and found to be similar to the plasma anticoagulant protein S. Soon after it was recognized as a growth factor-like molecule, as it interacted with receptor tyrosine kinases (RTKs) of the TAM family; Tyro3, Axl, and MerTK. Since then, the role of Gas6, protein S, and the TAM receptors has been found to be important in inflammation, hemostasis, and cancer, making this system an interesting target in biomedicine. Gas6 employs a unique mechanism of action, interacting through its vitamin K-dependent Gla module with phosphatidylserine-containing membranes and through its carboxy-terminal LG domains with the TAM membrane receptors. The fact that these proteins are affected by anti-vitamin K therapy is discussed in detail.
Collapse
Affiliation(s)
- Lola Bellido-Martín
- Department of Cell Death and Proliferation, Institute for Biomedical Research of Barcelona, IIBB-CSIC-IDIBAPS, Barcelona 08036, Spain
| | | |
Collapse
|
193
|
Sperandio O, Miteva MA, Segers K, Nicolaes GAF, Villoutreix BO. Screening Outside the Catalytic Site: Inhibition of Macromolecular Inter-actions Through Structure-Based Virtual Ligand Screening Experiments. Open Biochem J 2008; 2:29-37. [PMID: 18949072 PMCID: PMC2570552 DOI: 10.2174/1874091x00802010029] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2008] [Revised: 02/08/2008] [Accepted: 02/23/2008] [Indexed: 12/11/2022] Open
Abstract
During these last 15 years, drug discovery strategies have essentially focused on identifying small molecules able to inhibit catalytic sites. However, other mechanisms could be targeted. Protein-protein interactions play crucial roles in a number of biological processes, and, as such, their disruption or stabilization is becoming an area of intense activity. Along the same line, inhibition of protein-membrane could be of major importance in several disease indications. Despite the many challenges associated with the development of such classes of interaction modulators, there has been considerable success in the recent years. Importantly, through the existence of protein hot-spots and the presence of druggable pockets at the macromolecular interfaces or in their vicinities, it has been possible to find small molecule effectors using a variety of screening techniques, including combined virtual ligand-in vitro screening strategy. Indeed such in silico-in vitro protocols emerge as the method of choice to facilitate our quest of novel drug-like compounds or of mechanistic probes aiming at facilitating the understanding of molecular reactions involved in the Health and Disease process. In this review, we comment recent successes of combined in silico-in vitro screening methods applied to modulating macromolecular interactions with a special emphasis on protein-membrane interactions.
Collapse
Affiliation(s)
- Olivier Sperandio
- Inserm U648, University of Paris 5, 45 rue des Sts Peres, 75006 Paris, France
| | - Maria A Miteva
- Inserm U648, University of Paris 5, 45 rue des Sts Peres, 75006 Paris, France
| | - Kenneth Segers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands
| | - Gerry A. F Nicolaes
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands
| | - Bruno O Villoutreix
- Inserm U648, University of Paris 5, 45 rue des Sts Peres, 75006 Paris, France
| |
Collapse
|
194
|
Berny MA, White TC, Tucker EI, Bush-Pelc LA, Di Cera E, Gruber A, McCarty OJ. Thrombin Mutant W215A/E217A Acts as a Platelet GPIb Antagonist. Arterioscler Thromb Vasc Biol 2008; 28:329-34. [DOI: 10.1161/atvbaha.107.156273] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Michelle A. Berny
- From the Departments of Biomedical Engineering (M.A.B., T.C.W., E.I.T., A.G., O.J.T.M.) and Cell and Developmental Biology (O.J.T.M.), Oregon Health and Science University, Portland; and Biochemistry and Molecular Biophysics (L.A.B.-P., E.D.C.), Washington University, St. Louis, Mo
| | - Tara C. White
- From the Departments of Biomedical Engineering (M.A.B., T.C.W., E.I.T., A.G., O.J.T.M.) and Cell and Developmental Biology (O.J.T.M.), Oregon Health and Science University, Portland; and Biochemistry and Molecular Biophysics (L.A.B.-P., E.D.C.), Washington University, St. Louis, Mo
| | - Erik I. Tucker
- From the Departments of Biomedical Engineering (M.A.B., T.C.W., E.I.T., A.G., O.J.T.M.) and Cell and Developmental Biology (O.J.T.M.), Oregon Health and Science University, Portland; and Biochemistry and Molecular Biophysics (L.A.B.-P., E.D.C.), Washington University, St. Louis, Mo
| | - Leslie A. Bush-Pelc
- From the Departments of Biomedical Engineering (M.A.B., T.C.W., E.I.T., A.G., O.J.T.M.) and Cell and Developmental Biology (O.J.T.M.), Oregon Health and Science University, Portland; and Biochemistry and Molecular Biophysics (L.A.B.-P., E.D.C.), Washington University, St. Louis, Mo
| | - Enrico Di Cera
- From the Departments of Biomedical Engineering (M.A.B., T.C.W., E.I.T., A.G., O.J.T.M.) and Cell and Developmental Biology (O.J.T.M.), Oregon Health and Science University, Portland; and Biochemistry and Molecular Biophysics (L.A.B.-P., E.D.C.), Washington University, St. Louis, Mo
| | - András Gruber
- From the Departments of Biomedical Engineering (M.A.B., T.C.W., E.I.T., A.G., O.J.T.M.) and Cell and Developmental Biology (O.J.T.M.), Oregon Health and Science University, Portland; and Biochemistry and Molecular Biophysics (L.A.B.-P., E.D.C.), Washington University, St. Louis, Mo
| | - Owen J.T. McCarty
- From the Departments of Biomedical Engineering (M.A.B., T.C.W., E.I.T., A.G., O.J.T.M.) and Cell and Developmental Biology (O.J.T.M.), Oregon Health and Science University, Portland; and Biochemistry and Molecular Biophysics (L.A.B.-P., E.D.C.), Washington University, St. Louis, Mo
| |
Collapse
|
195
|
Bathum L, Münster AM, Nybo M, Madsen JS, Larsen TB, Kristensen SR. Denaturing high-performance liquid chromatography mutation analysis in patients with reduced Protein S levels. Clin Chim Acta 2008; 390:76-81. [PMID: 18242167 DOI: 10.1016/j.cca.2007.12.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2007] [Revised: 12/28/2007] [Accepted: 12/28/2007] [Indexed: 11/15/2022]
Abstract
BACKGROUND Patients with congenital Protein S deficiency have increased risk of venous thromboembolism. However, Protein S levels show large intra-individual variation and the biochemical assays have low accuracy and a high interlaboratory variability. Genetic analysis might aid in a more precise diagnosis and risk estimation. The aim was to design a high-throughput genetic analysis based on denaturing high-performance liquid chromatography to identify sequence variations in the gene coding for Protein S. PATIENTS In total, 55 patients referred to the Section of Thrombosis and Haemostasis, Odense University Hospital, in the period 1998-2004 were included in the study. RESULTS Mutations were found in ten of the 55 patients: Six different variants were identified, of which four were not previously reported: One were a nonsense mutation substituting a glutamine with a stopcodon (c.790C>T) and the rest were missense mutations (c.932T>G; c.1367A>G; c.1378T>C). Furthermore, four patients carried the same mutation (c.1045G>A), while two carried the Heerlen mutation (c.1378T>C). CONCLUSIONS The reported method will be useful for rapidly detecting sequence variations in the gene coding for Protein S, giving a precise diagnosis and subsequently a better risk estimation.
Collapse
Affiliation(s)
- Lise Bathum
- Department of Biochemistry, Pharmacology and Genetics, Odense University Hospital, Denmark.
| | | | | | | | | | | |
Collapse
|
196
|
Eisert WG, Schlachetzki F. Vascular endothelium and the blood-brain barrier. HANDBOOK OF CLINICAL NEUROLOGY 2008; 92:197-214. [PMID: 18790276 DOI: 10.1016/s0072-9752(08)01910-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Wolfgang G Eisert
- Center for Thrombosis and Atherosclerosis Research, University of Hanover, Hanover, Germany.
| | | |
Collapse
|
197
|
Mosnier LO, Yang XV, Griffin JH. Activated protein C mutant with minimal anticoagulant activity, normal cytoprotective activity, and preservation of thrombin activable fibrinolysis inhibitor-dependent cytoprotective functions. J Biol Chem 2007; 282:33022-33. [PMID: 17872949 DOI: 10.1074/jbc.m705824200] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Activated protein C (APC) reduces mortality in severe sepsis patients and exhibits beneficial effects in multiple animal injury models. APC anticoagulant activity involves inactivation of factors Va and VIIIa, whereas APC cytoprotective activities involve the endothelial protein C receptor and protease-activated receptor-1 (PAR-1). The relative importance of the anticoagulant activity of APC versus the direct cytoprotective effects of APC on cells for the in vivo benefits is unclear. To distinguish cytoprotective from the anticoagulant activities of APC, a protease domain mutant, 5A-APC (RR229/230AA and KKK191-193AAA), was made and compared with recombinant wild-type (rwt)-APC. This mutant had minimal anticoagulant activity but normal cytoprotective activities that were dependent on endothelial protein C receptor and protease-activated receptor-1. Whereas anticoagulantly active rwt-APC inhibited secondary-extended thrombin generation and concomitant thrombin-dependent activation of thrombin activable fibrinolysis inhibitor (TAFI) in plasma, secondary-extended thrombin generation and the activation of TAFI were essentially unopposed by 5A-APC due to its low anticoagulant activity. Compared with rwt-APC, 5A-APC had minimal profibrinolytic activity and preserved TAFI-mediated anti-inflammatory carboxypeptidase activities toward bradykinin and presumably toward the anaphlatoxins, C3a and C5a, which are well known pathological mediators in sepsis. Thus, genetic engineering can selectively alter the multiple activities of APC and provide APC mutants that retain the beneficial cytoprotective effects of APC while diminishing bleeding risk due to reduction in APC's anticoagulant and APC-dependent profibrinolytic activities.
Collapse
Affiliation(s)
- Laurent O Mosnier
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | | | | |
Collapse
|
198
|
Chen P, Zhang Y, Qiao M, Yuan Y. Activated protein C, an anticoagulant polypeptide, ameliorates severe acute pancreatitis via regulation of mitogen-activated protein kinases. J Gastroenterol 2007; 42:887-96. [PMID: 18008033 DOI: 10.1007/s00535-007-2104-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2006] [Accepted: 08/13/2007] [Indexed: 02/07/2023]
Abstract
BACKGROUND Our aim was to investigate the changes of mitogen-activated protein kinases (MAPKs) by activated protein C (APC) treatment in rats with severe acute pancreatitis (SAP), and relate them to changes in SAP severity, thus providing evidence for developing clinical therapies. METHODS Sprague-Dawley rats were given an intravenous injection of saline (SAP group), APC (50 microg/kg or 10 microg/kg), or CNI1493 just before SAP induction. One group of rats underwent a sham operation (control group). Experimental samples were harvested 16 h after SAP induction. The gene expression of pancreatic MAPKs was evaluated by cDNA microarrays. The mRNA and protein/phosphorylated protein levels of p38 MAPK, extracellular signal-regulated protein kinase (ERK) 1/2, and c-Jun N-terminal kinase (JNK) and the protein levels of tumor necrosis factor (TNF)-alpha and interleukin (IL)-1beta were determined in pancreatic tissue. The severity of disease was evaluated by pancreatic histology, the pancreatic wet/dry weight ratio, and the serum amylase level. RESULTS In rats treated with APC (50 microg/kg) or CNI1493, the severity of pancreatitis and expression of pancreatic TNF-alpha and IL-1beta proteins were attenuated by the decreased expression and activity of p38 MAPK and JNK (vs. the SAP group, P < 0.01). The expression and activity of ERK1/2 were increased in APC-treated rats, especially in the group treated with APC 50 microg/kg (vs. the SAP or CNI1493-treated group, P < 0.01, respectively). CONCLUSIONS Inhibition of expression of pancreatic p38 MAPK and JNK and upregulation of ERK1/2 expression by APC treatment may protect against pancreatic injury, thus ameliorating severity of the disease.
Collapse
Affiliation(s)
- Ping Chen
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | | | | | | |
Collapse
|
199
|
The cost-benefit ratio of screening pregnant women for thrombophilia. BLOOD TRANSFUSION = TRASFUSIONE DEL SANGUE 2007; 5:189-203. [PMID: 19204775 DOI: 10.2450/2007.0022-07] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Subscribe] [Scholar Register] [Received: 05/16/2007] [Accepted: 06/07/2007] [Indexed: 11/21/2022]
|
200
|
Borgel D, Bornstain C, Reitsma PH, Lerolle N, Gandrille S, Dali-Ali F, Esmon CT, Fagon JY, Aiach M, Diehl JL. A comparative study of the protein C pathway in septic and nonseptic patients with organ failure. Am J Respir Crit Care Med 2007; 176:878-85. [PMID: 17673691 DOI: 10.1164/rccm.200611-1692oc] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Severe sepsis is associated with an exacerbated procoagulant state with protein C (PC) system impairment. In contrast, the inflammatory and coagulation status of nonseptic patients with organ failure (OF) is less documented. OBJECTIVES To compare coagulation activation, focusing on the PC system, and inflammatory status in septic and nonseptic patients with OF. METHODS Thirty patients with severe sepsis and 30 nonseptic patients were recruited at the onset of OF and compared with 30 matched healthy subjects. We performed an extensive analysis of the PC pathway, including plasma protein measurements and quantification of leukocyte expression of PC system receptors. In addition, we analyzed the inflammatory status, based on inflammation-related gene leukocyte expression. MEASUREMENTS AND MAIN RESULTS We observed coagulation activation, reflected by a similar increase in tissue factor mRNA expression, in the two patient groups when compared with the healthy subjects. Soluble thrombomodulin levels were higher in septic patients than in healthy control subjects, whereas PC, protein S, and soluble endothelial cell PC receptor levels were lower. Similar results were obtained in nonseptic patients with OF. Monocyte thrombomodulin overexpression, together with increased circulating levels of activated PC, suggests that the capacity for PC activation is at least partly preserved in both settings. No difference in the inflammatory profile was found between septic and nonseptic patients. CONCLUSIONS The pathogenesis of OF in critical care patients is characterized by an overwhelming systemic inflammatory response and by exacerbated coagulation activation, independently of whether or not infection is the triggering event. Clinical trial registered with www.clinicaltrials.gov (NCT 00361725).
Collapse
|