Thrombomodulin mutant mice with a strongly reduced capacity to generate activated protein C have an unaltered pulmonary immune response to respiratory pathogens and lipopolysaccharide

Blood ◽  
2004 ◽  
Vol 103 (5) ◽  
pp. 1702-1709 ◽  
Author(s):  
Anita W. Rijneveld ◽  
Sebastiaan Weijer ◽  
Sandrine Florquin ◽  
Charles T. Esmon ◽  
Joost C. M. Meijers ◽  
...  

AbstractThe thrombomodulin–protein C–protein S (TM-PC-PS) pathway exerts anticoagulant and anti-inflammatory effects. We investigated the role of TM in the pulmonary immune response in vivo by the use of mice with a mutation in the TM gene (TMpro/pro) that was earlier found to result in a minimal capacity for activated PC (APC) generation in the circulation. We here demonstrate that TMpro/pro mice also display a strongly reduced capacity to produce APC in the alveolar compartment upon intrapulmonary delivery of PC and thrombin. We monitored procoagulant and inflammatory changes in the lung during Gram-positive (Streptococcus pneumoniae) and Gram-negative (Klebsiella pneumoniae) pneumonia and after local administration of lipopolysaccharide (LPS). Bacterial pneumonia was associated with fibrin(ogen) depositions in the lung that colocalized with inflammatory infiltrates. LPS also induced a rise in thrombin-antithrombin complexes in bronchoalveolar lavage fluid. These pulmonary procoagulant responses were unaltered in TMpro/pro mice, except for enhanced fibrin(ogen) deposition during pneumococcal pneumonia. In addition, TMpro/pro mice displayed unchanged antibacterial defense, neutrophil recruitment, and cytokine/chemokine levels. These data suggest that the capacity of TM to generate APC does not play a role of importance in the pulmonary response to respiratory pathogens or LPS.

2012 ◽  
Vol 107 (03) ◽  
pp. 468-476 ◽  
Author(s):  
Ilze Dienava-Verdoold ◽  
Marina R. Marchetti ◽  
Liane C. J. te Boome ◽  
Laura Russo ◽  
Anna Falanga ◽  
...  

SummaryThe natural anticoagulant protein S contains a so-called thrombin-sensitive region (TSR), which is susceptible to proteolytic cleavage. We have previously shown that a platelet-associated protease is able to cleave protein S under physiological plasma conditions in vitro. The aim of the present study was to investigate the relation between platelet-associated protein S cleaving activity and in vivo protein S cleavage, and to evaluate the impact of in vivo protein S cleavage on its anticoagulant activity. Protein S cleavage in healthy subjects and in thrombocytopenic and thrombocythaemic patients was evaluated by immunological techniques. Concentration of cleaved and intact protein S was correlated to levels of activated protein C (APC)-dependent and APC-independent protein S anticoagulant activity. In plasma from healthy volunteers 25% of protein S is cleaved in the TSR. While in plasma there was a clear positive correlation between levels of intact protein S and both APC-dependent and APC-independent protein S anticoagulant activities, these correlations were absent for cleaved protein S. Protein S cleavage was significantly increased in patients with essential thrombocythaemia (ET) and significantly reduced in patients with chemotherapy-induced thrombocytopenia. In ET patients on cytoreductive therapy, both platelet count and protein S cleavage returned to normal values. Accordingly, platelet transfusion restored cleavage of protein S to normal values in patients with chemotherapy-induced thrombocytopenia. In conclusion, proteases from platelets seem to contribute to the presence of cleaved protein S in the circulation and may enhance the coagulation response in vivo by down regulating the anticoagulant activity of protein S.


2003 ◽  
Vol 90 (08) ◽  
pp. 227-234 ◽  
Author(s):  
Björn Dahlbäck ◽  
Björn Arnljots ◽  
Karl Malm

SummaryThe antithrombotic effect of bovine activated protein C (bAPC) given with or without bovine protein S (bPS) was investigated in a rat model of deep arterial injury. A segment of the left common carotid artery was isolated between vascular clamps and opened longitudinally. An endarterectomy was performed and the arteriotomy was closed with a running suture, whereafter the vessel was reperfused by removing the clamps. The antithrombotic effect (vascular patency rates 31 minutes after reperfusion) and the arteriotomy bleeding were measured. Ten treatment groups each containing 10 rats and a control group of 20 animals were in a blind random fashion given intravenous bolus injections of increasing doses of activated protein C, with or without co-administration of protein S. The groups received either bAPC alone (0.8, 0.4, 0.2 or 0.1 mg/kg), bAPC (0.8, 0.4, 0.2, 0.1 or 0.05 mg/kg) combined with bPS (0.6 mg/kg), or bPS alone (0.6 mg/kg) whereas the control group received vehicle only. Administered alone, bAPC or bPS had no antithrombotic effect, regardless of dosage. In contrast, all groups that were treated with bAPC in combination with bPS demonstrated a significant antithrombotic effect, as compared to controls. Neither bAPC, bPS, nor the combination of bAPC and bPS increased the arteriotomy bleeding significantly compared to controls. In vitro clotting assays using bAPC or bPS alone yielded only minor prolongation of clotting time, whereas bAPC combined with bPS prolonged the clotting time considerably, demonstrating the dependence on the APC-cofactor activity of bPS for expression of anticoagulant activity by bAPC. In conclusion, our study shows the in vivo significance of protein S as a cofactor to activated protein C, and that potent anti-thrombotic effect can be achieved by low doses of bAPC combined with bPS, without producing hemorrhagic side effects.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 2704-2704
Author(s):  
Laurent O. Mosnier ◽  
Jose A. Fernandez ◽  
Antonella Zampolli ◽  
Xia V. Yang ◽  
Zaverio M. Ruggeri ◽  
...  

Abstract Activated protein C (APC) has both anticoagulant activity via inactivation of factors Va and VIIIa and cytoprotective activities on cells that include anti-apoptotic and anti-inflammatory activities, alterations of gene expression profiles and protection of endothelial barrier function. The relative importance of APC’s anticoagulant activity vs. APC’s direct cytoprotective effects on cells for reduction of mortality in severe sepsis patients and protective effects in animal injury models is not entirely clear. In this current study, genetically engineered APC variants with different activity spectra were tested for in vivo anti-thrombotic potency. Recently we made a non-anticoagulant APC variant, 5A-APC (RR229/230AA and KKK191-193AAA), that retains normal in vitro cytoprotective effects and an ability to reduce mortality in murine sepsis models (Kerschen et al, ASH2006, J Exper Med, 2007). In contrast to 5A-APC, mutation of E149 to A in APC increased anticoagulant activity in clotting assays while diminishing cytoprotective effects on cells. Murine APC variants, E149A-APC and 5A-APC (KKK192-194AAA + RR230/231AA) were used to determine in vivo anti-thrombotic potency in an acute carotid artery thrombosis model in mice, using FeCl3-induced injury. Under the conditions employed, first occlusion occurred within 3.5 min (mean: 171 sec; range 150-200 sec) in the absence of APC. Murine wild type (wt)-APC effectively delayed time to first occlusion in a dose-dependent manner (0 to 1.8 mg/kg wt-APC; mean: 561 sec; range 400-960 sec). The E149A-APC variant exhibited potent in vivo anti-thrombotic activity (1.8 mg/kg; mean: 1020 sec; range 540- >1600 sec) and was superior to wt-APC as evident by the absence of appreciable occlusion in 2/6 E149A-APC vs. 0/6 wt-APC treated animals. Thus E149A-APC was hyperactive in plasma clotting assays as well as hyperactive in an acute FeCl3-induced arterial thrombosis model. To test the hypothesis that an increased protein S cofactor activity contributed to its enhanced anticoagulant activity, E149A-APC anticoagulant activity was tested in normal and protein S deficient plasma. Compared to wt-APC, E149A-APC showed 3-fold increased anticoagulant activity in normal plasma but not in protein S deficient plasma. In studies with purified proteins, protein S concentrations required for half-maximal stimulation of factor Va inactivation by E149A-APC were 3-fold lower compared to wt-APC, whereas factor Va inactivation rates were indistinguishable in the absence of protein S. These data support our hypothesis that increased protein S cofactor activity is, at least partially, responsible for the observed hyper anticoagulant and anti-thrombotic potency in vitro and in vivo. In contrast to E149A-APC, 5A-APC was severely deficient in anti-thrombotic activity in vivo. Even at concentrations up to 8 mg/kg, 5A-APC (mean: 245 sec; range 172-300 sec) failed to delay significantly time to first occlusion compared to no APC. These data highlight important distinctions between structural requirements for APC’s anticoagulant, anti-thrombotic and cytoprotective functions. Engineered APC variants with differentially altered activities (e.g. cytoprotective vs. anticoagulant) may lead to safer or better therapeutic APC variants for a variety of indications including sepsis, ischemic stroke or other pathologies.


Blood ◽  
2009 ◽  
Vol 114 (11) ◽  
pp. 2307-2314 ◽  
Author(s):  
François Saller ◽  
Anne C. Brisset ◽  
Svetlana N. Tchaikovski ◽  
Monica Azevedo ◽  
Roman Chrast ◽  
...  

AbstractProtein S (PS) is an important natural anticoagulant with potentially multiple biologic functions. To investigate further the role of PS in vivo, we generated Pros+/− heterozygous mice. In the null (−) allele, the Pros exons 3 to 7 have been excised through conditional gene targeting. Pros+/− mice did not present any signs of spontaneous thrombosis and had reduced PS plasma levels and activated protein C cofactor activity in plasma coagulation and thrombin generation assays. Tissue factor pathway inhibitor cofactor activity of PS could not be demonstrated. Heterozygous Pros+/− mice exhibited a notable thrombotic phenotype in vivo when challenged in a tissue factor–induced thromboembolism model. No viable Pros−/− mice were obtained through mating of Pros+/− parents. Most E17.5 Pros−/− embryos were found dead with severe intracranial hemorrhages and most likely presented consumptive coagulopathy, as demonstrated by intravascular and interstitial fibrin deposition and an increased number of megakaryocytes in the liver, suggesting peripheral thrombocytopenia. A few E17.5 Pros−/− embryos had less severe phenotype, indicating that life-threatening manifestations might occur between E17.5 and the full term. Thus, similar to human phenotypes, mild heterozygous PS deficiency in mice was associated with a thrombotic phenotype, whereas total homozygous deficiency in PS was incompatible with life.


2006 ◽  
Vol 95 (02) ◽  
pp. 236-242 ◽  
Author(s):  
Jeroen Brugge ◽  
Guido Tans ◽  
Jan Rosing ◽  
Elisabetta Castoldi

SummaryElevated plasma prothrombin levels, due to the prothrombin 20210 G/A mutation or to acquired causes, area risk factor for venous thrombosis,partly because of prothrombin-mediated inhibition of the protein C anticoagulant pathway and consequent activated proteinC (APC) resistance. We determined the effect of plasma prothrombin concentration on the APC resistance phenotype and evaluated the role of protein S levels asa modulating variable. The effect of prothrombin and protein S levels on APC resistance was investigated in reconstituted plasma systems and in a population of healthy individuals using both the aPTT-based and the thrombin generation-based APC resistance tests. In reconstituted plasma, APC resistance increased at increasing prothrombin concentration in both assays. Enhanced APC resistance was caused by the effect of prothrombin on the clotting time in the absence of APC in the aPTT-based test, and on thrombin formation in the presence of APC in the thrombin generation-based test. In plasma from healthy individuals prothrombin levels were highly correlated to protein S levels. Since prothrombin and proteinS had opposite effects on the APC resistance phenotype, the prothrombin/protein S ratio was a better predictor of APC resistance than the levels of either protein alone. Prothrombin titrations in plasmas containing different amounts of proteinS confirmed that proteinS levels modulate the ability of prothrombin to induce APC resistance. These findings suggest that carriers of the prothrombin 20210 G/A mutation, who have a high prothrombin/protein S ratio, may experience a higher thrombosis risk than non-carriers with comparable prothrombin levels.


Blood ◽  
2002 ◽  
Vol 100 (2) ◽  
pp. 539-546 ◽  
Author(s):  
Julie A. Oliver ◽  
Dougald M. Monroe ◽  
Frank C. Church ◽  
Harold R. Roberts ◽  
Maureane Hoffman

Abstract The protein C/protein S system is known to regulate thrombin generation in vivo by cleaving factors Va and VIIIa. We have examined the activity of activated protein C in several tissue factor–initiated models of coagulation. We used 4 models: monocytes as the tissue factor source with platelets as the thrombin-generating surface; endothelial cells as the tissue factor source with platelets as the thrombin-generating surface; endothelial cells as both the tissue factor source and the thrombin-generating surface; and relipidated tissue factor with lipid vesicles providing the surface for thrombin generation. With the lipid surface, activated protein C dose-dependently reduced thrombin generation. Similarly, when endothelial cells provided the only surface for thrombin generation, activated protein C dose-dependently decreased thrombin generation significantly. By contrast, whenever platelets were present, activated protein C only minimally affected the amount of thrombin generated. When endothelial cells were the tissue factor source with platelets providing the surface for thrombin generation, activated protein C did increase the time until the burst of thrombin generation but had minimal effects on the total amount of thrombin generated. Activated protein C had essentially no effect on thrombin generation when monocytes were the tissue factor source with platelets providing the surface for thrombin generation. From the studies reported here, we conclude that in vivo, despite the important role of the protein C system in regulating thrombosis, activated protein C does not serve as a primary regulator of platelet-dependent thrombin generation.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 93-93
Author(s):  
Amelia R. Wilhelm ◽  
Nicole A. Parsons ◽  
Charles T. Esmon ◽  
Rodney M. Camire ◽  
Lindsey A. George

Activated factor VIII (FVIIIa) is an essential cofactor in the intrinsic tenase (Xase) enzyme complex that generates factor Xa and propagates clot formation. The FVIIIa heterotrimer is comprised of a metal ion linked dimer (A1/A3-C1-C2 domains) that is associated with the A2 domain by weak non-covalent interactions. Regulation of FXa formation by the intrinsic Xase enzyme complex occurs by FIXa inhibition and mechanisms contributing to FVIIIa inactivation, including: 1) rapid A2 domain dissociation and 2) activated protein C (APC) cleavage of FVIIIa. While FVIIIa inactivation by APC is considered important, there are surprisingly no in vivo studies documenting the hemostatic role of APC in FVIIIa regulation. Further, published data demonstrate APC cleavage of FVIIIa at physiologic protein concentrations occurs over hours while A2 dissociation occurs rapidly over minutes. Thus, it is thought that the predominant mechanism of FVIIIa inactivation is A2 dissociation and APC likely plays a marginal role in FVIIIa regulation. Additionally, unlike described A2 mutations that enhance dissociation and cause hemophilia A (HA), there is no known disease state attributed to altered FVIIIa cleavage by APC. This is in contrast to FVIII's homologous protein, FVa, whereby resistance to APC cleavage is the most common inherited thrombophilia (FV-Leiden [FVL]). Understanding the physiologically relevant mechanisms of FVIIIa inactivation has immediate clinical applicability for understanding safety considerations in HA therapeutics that bypass FVIIIa regulation (FVIII mimetic antibodies, e.g. emicizumab). Further, as evidenced by successful hemophilia B gene therapy trials using a gain of function FIX variant (FIX-Padua), altering FVIIIa inactivation could be exploited for therapeutic benefit in the setting of gene transfer. We aimed to determine the in vivo hemostatic role of APC in FVIIIa regulation and pair these studies with purified system analysis. We introduced Arg to Gln mutations at FVIII APC cleavage sites (R336Q and R562Q, herein called FVIII-QQ) on a B-domain deleted FVIII (FVIII-WT) backbone and produced recombinant FVIII-QQ and FVIII-WT. Unlike FVIII-WT, western blot analysis of FVIII-QQ incubated with APC and phospholipids had no evidence of cleavage. Enzyme kinetic studies using purified components demonstrated no appreciable difference in the Km or Vmax for FX within the intrinsic Xase enzyme complex or A2 dissociation of FVIII-QQ relative to FVIII-WT. These data confirmed no unexpected differences in FVIII-QQ relative to FVIII-WT. To begin to evaluate the role of APC in FVIIIa regulation, we measured thrombin generation in murine and human HA plasma reconstituted with FVIII-QQ or FVIII-WT in the presence of increasing APC concentrations. The IC50 of APC was 2-3-fold higher for FVIII-QQ than FVIII-WT. To evaluate the in vivo hemostatic effect of APC in FVIIIa regulation, HA mice were infused with FVIII-QQ or FVIII-WT and evaluated by tail clip injury and 7.5% FeCl3 carotid artery occlusion models. Required doses of FVIII-QQ to normalize blood loss from a tail clip assay and time to vessel occlusion in a FeCl3 assay were 4-5 fold lower than necessary FVIII-WT doses; the superior hemostatic effect of FVIII-QQ supported the physiologic significance of APC in FVIIIa inactivation. To isolate the role of APC in FVIIIa regulation from APC inactivation of FVa, we backcrossed HA mice with FVL mice to create homozygous HA/FVL mice. HA/FVL mice were infused with FVIII-QQ or FVIII-WT and underwent tail clip assay analysis. Doses of FVIII-QQ required to normalize blood loss were again less than FVIII-WT. To further isolate the enhanced hemostatic effect of FVIII-QQ to APC resistance, we performed the tail clip assay in HA/FVL mice infused with FVIII-QQ or FVIII-WT in the presence or absence of MPC1609, an antibody that blocks murine APC function (Xu et al. J Thromb Haemost 2008). In the presence of MPC1609, the same dose of FVIII-WT and FVIII-QQ was required to normalize blood loss (Figure 1). Collectively, our in vitro and in vivo data support the physiologic significance of APC in FVIIIa regulation. To our knowledge these data are the first to demonstrate the in vivo hemostatic effect of APC in FVIIIa inactivation. Our data may be translated to rationally exploit APC regulation of FVIIIa to develop novel HA therapeutics or further delineate safety considerations in therapies that bypass FVIIIa regulation. Figure 1 Disclosures Camire: Pfizer: Research Funding. George:University of Pennyslvania: Employment; Pfizer: Consultancy; Avrobio: Membership on an entity's Board of Directors or advisory committees.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3186-3186
Author(s):  
Rinku Majumder

Abstract 3186 Poster Board III-123 Thrombosis is a serious problem in the United States. The overall estimated incidence (annual occurrence) of deep venous thrombosis is 1 episode for every 1000 persons. Protein S, a vitamin K-dependent protein, is one of the natural anticoagulants found in the blood. Deficiency of protein S is most common protein deficiencies associated with familial venous thrombosis There are studies that suggest an association between arterial thrombosis (stroke, heart attack) in patients with protein S deficiency. At this time, the exact role of protein S deficiency and its relative importance in arterial disease is still being explored by physicians and scientists. Protein S is known as a non-enzymatic cofactor of activated Protein C in the inactivation of factors Va and VIIIa, as part of a negative feedback loop to regulate blood coagulation. Plasma coagulation assays in the absence of activated protein C suggest that Protein S may have other anticoagulant role(s). For example, it has been suggested that Protein S down-regulates thrombin generation by stimulating FXa inhibition by the tissue factor pathway inhibitor (Rosing, J., et al., Thromb Res, 2008. 122 Suppl 1: p. S60-3). It has also been proposed that protein S can directly inhibit the intrinsic Xase complex (Takeyama, M., et al.. Br J Haematol, 2008. 143(3): p. 409-20). But the exact mechanism of how Protein S exerts its anticoagulant effect on factor IXa/VIIIa complex is still unclear. In order to determine the role of Protein S as an anticoagulant in the intrinsic Xase Complex, we have used C6PS (a small six carbon chain synthetic Phosphatidylserine (PS) molecule) that does not occur in vivo, but has been used as a powerful tool in demonstrating the regulation of both factors Xa and Va by binding of molecular PS. Soluble lipid binding can offer invaluable insights into events that would be next to impossible to document on a membrane surface which is complicated as it has surface condensation effect and allosteric effects of different factors. We focus here on the conformation changes of the proteins by using C6PS as a tool. We have determined the binding of Protein S with C6PS by using tryptophan fluorescence and observed a stoichiometric Kd of ∼180 μM.We checked for micelles formation under each experimental condition. We have also determined the direct binding of factor IXa with Protein S by using DEGR-IXa ((5-(dimethylamino)1-naphthalenesulfonyl]-glutamylglycylarginyl chloromethyl ketone) in the presence and absence of C6PS. Our results show that the affinity of binding of DEGR-IXa to Protein S in the presence of C6PS is ∼22 fold tighter (Kd ∼15 nM compared to 324 nM) than without C6PS. We also measured the rate of factor X activation by factor IXa with the addition of increasing concentration of C6PS in the presence and absence of Protein S. We observed that Protein S decreased factor IXa mediated factor X activation by 14 fold. We had previously shown that apparent Kd of factor IXa binding to C6PS during factor X activation was ∼125 μM. But addition of Protein S had an effect on the apparent Kd as it increased to 700 μM indicating the affinity of factor IXa towards C6PS was decreased with the addition of Protein S during factor X activation. From these data we can speculate that Protein S induces a conformational change in factor IXa in the presence of C6PS which may affect the interaction of factor IXa with factor VIIIa, thus affecting the intrinsic Xase complex. Using this useful tool (C6PS), we will characterize the anticoagulant role of Protein S in the intrinsic Xase complex which in turn will give us some insights into this important protein which is a crucial target for therapeutic drugs for venous thrombosis. Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document