scholarly journals Factor VIIa binding to endothelial cell protein C receptor: Differences between mouse and human systems

2012 ◽  
Vol 107 (05) ◽  
pp. 951-961 ◽  
Author(s):  
Prosenjit Sen ◽  
Curtis A. Clark ◽  
Ramakrishnan Gopalakrishnan ◽  
Ulla Hedner ◽  
Charles T. Esmon ◽  
...  

SummaryRecent in vitro studies have shown that the zymogen and activated form of factor (F)VII bind to endothelial cell protein C receptor (EPCR). At present, there is no evidence that FVIIa binds to EPCR on vascular endothelium in vivo in the presence of circulating protein C, a primary ligand for EPCR. The present study was carried out to investigate the interaction of murine and human ligands with murine EPCR both in vivo and in vitro. Measurement of endogenous plasma levels of FVII in wild-type, EPCR-deficient and EPCR-over expressing mice showed slightly lower levels of FVII in EPCR-over expressing mice. However, infusion of high concentrations of competing ligands, either human APCi or FVIIai, to EPCR-over expressing mice failed to increase plasma levels of mouse FVII whereas they increased the plasma levels of protein C by two- to three-fold. Examining the association of exogenously administered mouse FVIIa or human FVIIa by immunohistochemistry revealed that human, but not murine FVIIa, binds to the murine endothelium in an EPCR-dependent manner. In vitro binding studies performed using surface plasmon resonance and endothelial cells revealed that murine FVIIa binds murine EPCR negligibly. Human FVIIa binding to EPCR, particularly to mouse EPCR, is markedly enhanced by availability of Mg2+ ions. In summary, our data show that murine FVIIa binds poorly to murine EPCR, whereas human FVIIa binds efficiently to both murine and human EPCR. Our data suggest that one should consider the use of human FVIIa in mouse models to investigate the significance of FVIIa and EPCR interaction.

PLoS ONE ◽  
2014 ◽  
Vol 9 (8) ◽  
pp. e103505 ◽  
Author(s):  
Rit Vatsyayan ◽  
Hema Kothari ◽  
Nigel Mackman ◽  
Usha R. Pendurthi ◽  
L. Vijaya Mohan Rao

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 3435-3435
Author(s):  
Kazuhiro Abeyama ◽  
Yasushi Yoshimoto ◽  
Ikuro Maruyama

Abstract Thrombomodulin (TM) is an endothelial anticoagulant cofactor that promotes thrombin-mediated formation of activated protein C (APC), the latter an enzyme with potent anti-coagulant and anti-inflammatory properties. We have found that the N-terminal, lectin-like domain (D1) of thrombomodulin has unique anti-inflammatory properties. Thrombomodulin, via D1, binds high mobility group-B1 DNA binding protein (HMGB1), a factor closely associated with necrotic cell damage following its release from the nucleus, thereby preventing leukocyte activation in vitro, and ultraviolet radiation-induced cutaneous inflammation and lipopolysaccharide-induced lethality in vivo. Our data also demonstrate anti-inflammatory properties of a peptide spanning the D1 domain of TM and suggest its therapeutic potential. These findings highlight a novel mechanism through which an endothelial cofactor, TM, suppresses inflammation; i.e., sequestration of mediators thereby preventing their interaction with cell surface receptors on effector cells in the vasculature. Results: TM binds HMGB1 and prevents expression of pro-inflammatory activity. Our co-culture studies of leukocytes and HUVEC, and results in the cutaneous irritation model suggested that early release of a mediator, such as HMGB1, might contribute importantly to cellular activation in inflammation at later time points. In this context, TM might have the ability to decrease HMGB1-mediated inflammatory events. Binding studies using surface plasmon resonance (SPR), performed to directly assess the interaction of TM and immobilized HMGB1, demonstrated dose-dependent binding in the nanomolar range (Kd ~232 nM). Furthermore, addition of rhs-TM decreased, in a dose-dependent manner, the binding of HMGB1 to RAGE through the its N-terminal domain, but not anti-coagulant domain. TM and the N-terminal-derived TM peptide have anti-inflammatory effects in settings where HMGB1 is a likely key mediator. In HMGB1-mediated skin inflammation model, systemic administration of rhs-TM, its lectin-like domain and sRAGE resulted in a significant blunting of the inflammatory response. In contrast, the effect of anti-coagulant domain, although showing a trend toward decreased ear swelling, did not achieve statistical significance (anticoagulant domain has anti-inflammatory effects in vivo that probably reflect its ability to support thrombin-mediated activation of protein C; the latter does not occur in vitro after inactivation of the protein C zymogen by heat treatment). In view of recent data suggesting a link between HMGB1 released from injured tissue and endotoxin-induced lethality in mice, we also tested whether rhs-TM and its lectin-like domain might also have protective effects in this model. We employed a dose of intraperitoneal (IP) LPS (10 mg/kg) resulting in 100% lethality by 96 hrs. Systemic (IP) treatment of animals with anti-HMGB1 IgY had a protective effect with respect to lethality at 4 days, whereas the same regimen of nonimmune IgY was without effect. Similarly, IP administration of rhs-TM and its N-teminal lectin domain, but not anti-coagulant domain had complete protective effects compared with anti-HMGB1 IgY. Conclusion: Our findings have elucidated an unexpected anti-inflammatory property of TM residing in the D1 domain, namely binding of HMGB1.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 532-532
Author(s):  
Curtis A Clark ◽  
Rit Vatsyayan ◽  
Ramakrishnan Gopalakrishnan ◽  
Charles T Esmon ◽  
Ulla Hedner ◽  
...  

Abstract Abstract 532 Recombinant factor VIIa (FVIIa) has been demonstrated to be an effective hemostatic agent for treatment of severe hemophilia patients. Recent clinical studies suggest that the prophylactic use of FVIIa markedly reduces the number of bleeding episodes in hemophilic patients with inhibitors. Clinical evidence from hemophilic patients during prophylaxis and post-prophylaxis suggest that the hemostatic effect of FVIIa surpassed the short circulatory half-life of FVIIa. At present, mechanisms by which this phenomenon occurs are not precisely known. Our laboratory has formerly established that FVIIa is capable of binding to the endothelial protein C receptor (EPCR) on endothelial cells, yet the significance of this interaction in hemostasis, and especially in prophylaxis, is not fully understood. Recent studies from our laboratory using a murine model have demonstrated that pharmacologically administered FVIIa rapidly associates with the endothelium, enters into extravasculature where it can interact with TF, and remains functionally active in tissues for an extended period of time. The aim of the present study was to determine if interaction of FVIIa with EPCR on the endothelium and subsequent EPCR-dependent transcytosis serves as a mechanism by which pharmacologically administered FVIIa is transported to tissues and retained for greater periods of time compared to its circulatory half-life. We have employed transgenic murine lines, including EPCR-over expressing, EPCR-deficient, and wild-type (WT) mice, to investigate the function of EPCR in the biodistribution of FVIIa and its extended stay in tissues. Mice were injected with human FVIIa (120 μg/kg b.w.) via tail vein and at 30 min, 3 h, 8 h, and 24 h following FVIIa administration, blood was collected via cardiac puncture and various tissues were harvested for antigen and immunohistochemical analyses. Our studies reveal that, following intravenous injection, FVIIa rapidly disappears from plasma (plasma levels of FVIIa in WT mice at 30 min and 3 h post-injection were ∼ 15% and <1%, respectively, of that which was injected) and associates with the endothelium in an EPCR-dependent manner. At 30 min post-injection, plasma levels of FVIIa were significantly higher in EPCR-deficient mice as compared to WT mice whereas plasma FVIIa levels were significantly lower in EPCR-over expressing mice compared to WT mice, suggesting that EPCR facilitates the sequestration of FVIIa from the bloodstream. This EPCR-dependent decrease in plasma FVIIa coincides with an EPCR-dependent increase in antigen levels of FVIIa in tissue homogenates. FVIIa antigen in the lungs of EPCR-over expressing mice at 30 min following FVIIa administration was approximately 6-fold higher than that which was found in WT mice. At the corresponding time, FVIIa antigen in EPCR-deficient mice was approximately 35% lower than that which was measured in WT mice. FVIIa antigen in the lung tissue of EPCR-over expressing mice remained relatively stable or slightly increased at 3, 8 and 24 h post FVIIa administration. FVIIa levels in tissue homogenates of WT and EPCR-deficient mice remained significantly lower compared to EPCR-over expressing mice at all time points. Immunohistochemical analysis of lung, skin, and bone-joint sections revealed that association of FVIIa with the endothelium is more pronounced in EPCR-over expressing mice as compared to lesser, albeit appreciable, levels in WT mice; EPCR-deficient mice showed negligible association of FVIIa with the endothelium. Association of FVIIa with the endothelium in EPCR-over expressing mice was maximal at 30 min and was thereafter progressively reduced at each time point; FVIIa association with the endothelium was undetectable at 24 h post FVIIa administration. The observation that EPCR-dependent association of FVIIa with the endothelium is most pronounced soon after injection and subsequently declines time-wise, combined with the retention of FVIIa in tissue homogenates for extended periods, implies that FVIIa bound to the endothelium enters tissues in an EPCR-dependent manner. These findings put forward an EPCR-dependent mechanism by which pharmacologically administered FVIIa can relocate to and be retained within tissues where TF resides, which conceivably may prime coagulation thereby contributing to hemostasis. Disclosures: Hedner: Novo Nordisk: Consultancy. Rao:Novo Nordisk: Research Funding.


2014 ◽  
Vol 12 (5) ◽  
pp. 690-700 ◽  
Author(s):  
J. Sundaram ◽  
S. Keshava ◽  
R. Gopalakrishnan ◽  
C. T. Esmon ◽  
U. R. Pendurthi ◽  
...  

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1181-1181
Author(s):  
Prosenjit Sen ◽  
Curtis A Clark ◽  
Ramakrishnan Gopalakrishnan ◽  
Ulla Hedner ◽  
Charles T Esmon ◽  
...  

Abstract Abstract 1181 Recent studies from our laboratory and others have shown that both the zymogen and activated form of FVII bind to endothelial cell protein C receptor (EPCR), a cellular receptor for protein C and activated protein C (APC). Although at present the pathophysiological significance of this interaction is unclear, recent studies indicate that FVIIa binding to EPCR may facilitate FVIIa transport from blood to extravasculature, mediate FVIIa-induced cell signaling and provide endothelial barrier protection. However, at present, there is no direct evidence demonstrating FVIIa actually associates with EPCR in vivo. Further, the Gla region involved in the EPCR binding is not fully conserved in mouse FVII, which raises questions on the ability of mouse FVIIa to associate with EPCR and the validity of mouse model systems in investigating the importance of the interaction between FVIIa and EPCR. Thus, it is important to first characterize the interaction of mouse FVIIa with mouse EPCR before employing murine model systems to investigate the importance of FVIIa interaction with EPCR in hemostasis and inflammation. Measurement of plasma levels of FVII, using both clotting and antigen assays, in wild-type, EPCR-deficient and EPCR-over expressing mice showed a small but insignificant increase (∼10%) in the circulating levels of FVII in EPCR-deficient mice and a ∼15% reduction in EPCR-over expressing mice in comparison to plasma FVII levels in wild-type mice. In comparison, the plasma level of protein C was decreased by more than 60% in EPCR-over expressing mice. Infusion of high concentrations of either human APCi or human FVIIai (400 μg/mice) to EPCR-over expressing mice failed to displace and increase the plasma levels of endogenous mouse FVII, while they increased plasma levels of endogenous mouse protein C by 2–3-fold. In additional studies, either mouse FVIIa or human FVIIa (120 μg/kg, tagged with AF488 probe) was administered exogenously via tail-vein to the wild-type, EPCR-deficient and EPCR-over expressing mice and FVIIa association with EPCR was evaluated by immunohistochemistry using anti-AF488 antibodies. In the case of mice injected with human FVIIa, the staining of FVIIa was undetectable or negligible in EPCR-deficient mice, whereas visible staining for FVIIa was clearly observed on the endothelium lining of the vessel walls in the wild-type mice. Furthermore, very intense staining (more so than wild-type) of FVIIa was observed on the endothelium of EPCR-over expressing mice. Although exogenously infused mouse FVIIa also appears to associate with the endothelium, there are no discernable differences in the staining intensity among wild-type, EPCR-deficient and EPCR-over expressing mice, suggesting that mouse FVIIa association with the endothelium is unaffected by EPCR. Next, in vitro binding studies were performed using mouse and human ligands and EPCR to further evaluate differences between binding of human and mouse ligands to EPCR. Surface Plasmon Resonance binding studies showed negligible binding of mouse FVIIa to either soluble human EPCR or mouse EPCR immobilized on the sensor chip. Under identical experimental conditions, human FVIIa was shown to bind both human and mouse EPCR. Experiments conducted with mouse and human endothelial cells or CHO cells transfected to express mouse or human EPCR also showed that mouse FVIIa does not bind, in any appreciable amount, to mouse EPCR and binds very poorly to human EPCR. Conversely, human FVIIa was found to bind to both human and mouse EPCR with a similar efficiency, both of which were approximately equivalent to that of human APC binding to EPCR. Interestingly, the binding of human FVIIa and APC to mouse EPCR is more dependent on Mg2+ ions as compared to their binding to human EPCR, indicating that differences between human and murine EPCR may also contribute to the differential binding of human and mouse ligands to EPCR. In summary, our data indicate that significant differences exist between human and mouse FVIIa in their ability to interact with EPCR. Given that human but not mouse FVIIa binds to EPCR, it may be more appropriate to use human FVII/FVIIa in mouse model systems to properly investigate the importance of FVIIa interaction with EPCR in hemostasis and inflammation. Disclosures: Hedner: Novo Nordisk: Consultancy. Rao:Novo Nordisk: Research Funding.


Blood ◽  
2001 ◽  
Vol 97 (6) ◽  
pp. 1685-1688 ◽  
Author(s):  
Fletcher B. Taylor ◽  
Glenn T. Peer ◽  
Marion S. Lockhart ◽  
Gary Ferrell ◽  
Charles T. Esmon

Endothelial cell protein C receptor (EPCR) augments protein C activation by the thrombin-thrombomodulin complex about 5-fold in vitro. Augmentation is EPCR concentration dependent even when the EPCR concentration is in excess of the thrombomodulin. EPCR is expressed preferentially on large blood vessel endothelium, raising questions about the importance of protein C-EPCR interaction for augmenting systemic protein C activation. In these studies, this question was addressed directly by infusing thrombin into baboons in the presence or absence of a monoclonal antibody to EPCR that blocks protein C binding. Activated protein C levels were then measured directly by capturing the enzyme on a monoclonal antibody and assaying with chromogenic substrate. Blocking protein C-EPCR interaction resulted in about an 88% decrease in circulating activated protein C levels generated in response to thrombin infusion. Leukocyte changes, fibrinogen consumption, fibrin degradation products, and vital signs were similar between the animals infused with thrombin alone and those infused with thrombin and the anti-EPCR antibody. The results indicate that EPCR plays a major role in protein C activation and suggest that defects in the EPCR gene might contribute to increased risk of thrombosis.


2020 ◽  
Vol 40 (5) ◽  
pp. 1275-1288 ◽  
Author(s):  
Vijay Kondreddy ◽  
Usha R. Pendurthi ◽  
Xiao Xu ◽  
John H. Griffin ◽  
L. Vijaya Mohan Rao

Objective: Recent studies showed that FVIIa (factor VIIa), upon binding to EPCR (endothelial cell protein C receptor), elicits endothelial barrier stabilization and anti-inflammatory effects via activation of PAR (protease-activated receptor)-1–mediated signaling. It is unknown whether FVIIa induces PAR1-dependent cytoprotective signaling through cleavage of PAR1 at the canonical site or a noncanonical site, similar to that of APC (activated protein C). Approach and Results: Mouse strains carrying homozygous R41Q (canonical site) or R46Q (noncanonical site) point mutations in PAR1 (QQ41-PAR1 and QQ46-PAR1 mice) were used to investigate in vivo mechanism of PAR1-dependent pharmacological beneficial effects of FVIIa. Administration of FVIIa reduced lipopolysaccharide-induced inflammation, barrier permeability, and VEGF (vascular endothelial cell growth factor)-induced barrier disruption in wild-type (WT) and QQ46-PAR1 mice but not in QQ41-PAR1 mice. In vitro signaling studies performed with brain endothelial cells isolated from WT, QQ41-PAR1, and QQ46-PAR1 mice showed that FVIIa activation of Akt (protein kinase B) in endothelial cells required R41 cleavage site in PAR1. Our studies showed that FVIIa cleaved endogenous PAR1 in endothelial cells, and FVIIa-cleaved PAR1 was readily internalized, unlike APC-cleaved PAR1 that remained on the cell surface. Additional studies showed that pretreatment of endothelial cells with FVIIa reduced subsequent thrombin-induced signaling. This process was dependent on β-arrestin1. Conclusions: Our results indicate that in vivo pharmacological benefits of FVIIa in mice arise from PAR1-dependent biased signaling following the cleavage of PAR1 at the canonical R41 site. The mechanism of FVIIa-induced cytoprotective signaling is distinctly different from that of APC. Our data provide another layer of complexity of biased agonism of PAR1 and signaling diversity.


Blood ◽  
2021 ◽  
Author(s):  
Kaushik Das ◽  
Shiva Keshava ◽  
Shabbir A Ansari ◽  
Vijay Kumar Reddy Kondreddy ◽  
Charles Esmon ◽  
...  

Recombinant FVIIa (rFVIIa) is used as a hemostatic agent to treat bleeding disorders in hemophilia patients with inhibitors and other groups of patients. Our recent studies showed that FVIIa binds endothelial cell protein C receptor (EPCR) and induces protease-activated receptor 1 (PAR1)-mediated biased signaling. The importance of FVIIa-EPCR-PAR1-mediated signaling in hemostasis is unknown. In the present study, we show that FVIIa induces the release of extracellular vesicles (EVs) from endothelial cells both in vitro and in vivo. Silencing of EPCR or PAR1 in endothelial cells blocked the FVIIa-induced generation of EVs. Consistent with these data, FVIIa treatment enhanced the release of EVs from murine brain endothelial cells isolated from wild-type, EPCR overexpressors, and PAR1-R46Q mutant mice, but not EPCR-deficient or PAR1-R41Q mutant mice. In vivo studies revealed that administration of FVIIa to wild-type, EPCR overexpressors, and PAR1-R46Q mutant mice, but not EPCR-deficient or PAR1-R41Q mutant mice, increase the number of circulating EVs. EVs released in response to FVIIa treatment exhibit enhanced procoagulant activity. Infusion of FVIIa-generated EVs and not control EVs to platelet-depleted mice increased thrombin generation at the site of injury and reduced blood loss. Administration of FVIIa-generated EVs or generation of EVs endogenously by administering FVIIa augmented the hemostatic effect of FVIIa. Overall, our data reveal that FVIIa treatment, through FVIIa-EPCR-PAR1 signaling, releases EVs from the endothelium into the circulation, and these EVs contribute to the hemostatic effect of FVIIa.


Sign in / Sign up

Export Citation Format

Share Document