Endothelial barrier protection by activated protein C through PAR1-dependent sphingosine 1–phosphate receptor-1 crossactivation

Blood ◽  
2005 ◽  
Vol 105 (8) ◽  
pp. 3178-3184 ◽  
Author(s):  
Clemens Feistritzer ◽  
Matthias Riewald

AbstractEndothelial cells normally form a dynamically regulated barrier at the blood-tissue interface, and breakdown of this barrier is a key pathogenic factor in inflammatory disorders such as sepsis. Pro-inflammatory signaling by the blood coagulation protease thrombin through protease activated receptor-1 (PAR1) can disrupt endothelial barrier integrity, whereas the bioactive lipid sphingosine 1-phosphate (S1P) recently has been demonstrated to have potent barrier protective effects. Activated protein C (APC) inhibits thrombin generation and has potent anti-inflammatory effects. Here, we show that APC enhanced endothelial barrier integrity in a dual-chamber system dependent on binding to endothelial protein C receptor, activation of PAR1, and activity of cellular sphingosine kinase. Small interfering RNA that targets sphingosine kinase-1 or S1P receptor-1 blocked this protective signaling by APC. Incubation of cells with PAR1 agonist peptide or low concentrations of thrombin (∼ 40 pM) had a similar barrier-enhancing effect. These results demonstrate that PAR1 activation on endothelial cells can have opposite biologic effects, reveal a role for cross-communication between the prototypical barrier-protective S1P and barrier-disruptive PAR1 pathway, and suggest that S1P receptor-1 mediates protective effects of APC in systemic inflammation.

2009 ◽  
Vol 101 (04) ◽  
pp. 724-733 ◽  
Author(s):  
José Fernández ◽  
John Griffin ◽  
Reto Schuepbach ◽  
Clemens Feistritzer ◽  
Matthias Riewald

SummaryProtease 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.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 28-28
Author(s):  
Clemens Feistritzer ◽  
Laurent O. Mosnier ◽  
Enrico Di Cera ◽  
John H. Griffin ◽  
Matthias Riewald

Abstract Protein C (PC) is activated by thrombomodulin-bound thrombin on the endothelial cell surface and activated protein C (APC) inhibits blood coagulation in a negative feedback loop. Endothelial PC receptor (EPCR) can bind PC/APC and activation of EPCR-bound PC is enhanced. Exogenous APC has barrier protective effects on endothelial cells that depend on EPCR binding and protease activated receptor-1 (PAR1) cleavage and that may contribute to the anti-inflammatory effects of APC. Plasma APC concentrations in vivo are low compared to the substrate PC and in order to induce protective signaling exogenous APC has to compete with PC for EPCR binding. In this study we investigated whether the endogenous PC activation pathway may be linked to efficient protective responses analyzing endothelial barrier permeability in a dual chamber system. When endothelial EA.hy926 cells were incubated for 3 h in the presence of 80 nM purified PC and different concentrations of thrombin a dose-dependent linear increase of APC activity in the cell medium was observed over time. APC generation was detectable upon incubation with 20 pM thrombin or higher and a significant barrier protective response to 20 pM thrombin was found only in the presence of PC. 40 pM thrombin enhanced barrier integrity in the presence and absence of PC, consistent with our previous results. To exclude direct thrombin effects on endothelial permeability and to compare protective effects of exogenous and endogenously generated APC, we used the anticoagulant double mutant thrombin W215A/E217A (WE). WE was about 10 times less active than wildtype thrombin for PC activation in our system. However, PAR1-dependent induction of MAP kinase phosphorylation required more than 1000-fold higher concentrations of the thrombin mutant. Thus, 1–10 nM WE leads to APC generation without directly inducing PAR1-dependent signaling. When cells were incubated with various concentrations of exogenous APC or WE+80 nM PC, barrier protective effects of 5 nM exogenous APC and 2 nM WE+80 nM PC (1.3 nM APC generated after 3 h) were similar. Because APC is generated at a constant rate during the incubation period, the average concentration of generated APC in the cell medium was only about 0.65 nM, suggesting that signaling by endogenously generated APC was significantly more efficient. To conclusively demonstrate that protective effects in response to WE are mediated by APC generation, we used recombinant zymogen wildtype PC and a PC variant with a substitution of the active site serine with alanine (PC S360A). Cells were incubated with control or 80 nM wildtype PC and PC S360A, in the presence or absence of WE (4 nM) and exogenous APC (3.3 nM). WE induced protective signaling only in the presence of wildtype PC but not PC S360A. Barrier protective effects of exogenous APC were blocked by both wildtype PC and PC S360A, consistent with their expected role as competitive inhibitors for APC binding to EPCR. These data demonstrate that efficient barrier enhancement by APC is indeed mechanistically coupled to the PC activation pathway. Signaling by endogenously generated APC may play an important role in the regulation of inflammation.


Blood ◽  
2011 ◽  
Vol 118 (14) ◽  
pp. 3952-3959 ◽  
Author(s):  
Jong-Sup Bae ◽  
Alireza R. Rezaie

Abstract A pathogenic role for high-mobility group box 1 (HMGB1) protein has been postulated in severe sepsis. Activated protein C (APC) is the only drug approved by the Food and Drug Administration for severe sepsis; however, its effect on HMGB1 signaling has never been investigated. Here, we monitored the effect of APC on the lipopolysaccharide-mediated release of HMGB1 and the HMGB1-mediated modulation of proinflammatory responses in HUVECs. APC potently inhibited the release of HMGB1 and down-regulated the adhesion of the monocytic cell line, THP-1, to HMGB1-activated endothelial cells. HMGB1 up-regulated proinflammatory responses by interacting with 3 pathogen-related pattern recognition receptors: TLR2 and TLR4 and the receptor for advanced glycation end products. APC not only inhibited HMGB1 release but also down-regulated the cell surface expression of all 3 HMGB1 receptors in endothelial cells. The protective effects of APC were mediated through endothelial cell protein C receptor (EPCR) and protease-activated receptor 1 (PAR-1). Interestingly, a thrombin derivative containing the Gla-domain of APC recapitulated all protective effects of APC with a 20- to 50-fold higher efficacy. These results suggest that the EPCR- and PAR-1–dependent protective effects of APC in severe sepsis may partially be mediated through the inhibition of HMGB1 signaling and that the chimeric thrombin mutant has potential therapeutic utility for severe sepsis.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 1752-1752
Author(s):  
Jong-Sup Bae ◽  
Alireza R. Rezaie

Abstract It is known that residue 192 (chymotrypsin numbering) plays a key role in determining the P3 binding specificity of coagulation proteases. This residue is a Glu in both thrombin and activated protein C. Previous studies have indicated that the substrates containing an acidic residue at the P3 position are not recognized efficiently by either thrombin or APC in the absence of a cofactor. Protease activated receptor 1 (PAR-1), a common substrate for both thrombin and APC contains an acidic Asp at the P3 position. In this study we examined the ability of Glu-192 to Gln (E192Q) substitution mutants of both thrombin and APC to recognize and cleave PAR-1 in human umbilical vein endothelial cells (HUVEC) transfected with a PAR-1 cleavage reporter plasmid in which the exodomain of the receptor has been fused to a cDNA fragment encoding the soluble alkaline phosphatase. Thrombin E192Q cleaved PAR-1 with a catalytic efficiency that was approximately 5-fold higher than that of wild-type thrombin. On the other hand, the activity of APC E192Q toward PAR-1 was improved approximately 100-fold. Thus, unlike an approximately 1000-fold lower activity for APC in cleaving PAR-1 relative to thrombin, the activity of APC E192Q toward PAR-1 was only 10-fold lower than that of thrombin. These results suggest that the inhibitory interaction of Glu-192 of APC with P3-Asp of PAR-1 is responsible for its poor activity toward PAR-1. It is known that the cleavage of PAR-1 by thrombin in endothelial cells elicits a proinflammatory response. However, the cleavage of the same receptor by APC in complex with endothelial protein C receptor (EPCR) invokes a protective antiinflammatory response. The mechanism of the paradoxical effect of PAR-1 signaling by the two proteases is not known. We used these mutants to investigate the possibility that the level of PAR-1 activation by either thrombin or APC dictates the type of the response in endothelial cells. Furthermore, we used these mutants in TNF-a-stimulated endothelial cell permeability and apoptosis assays to understand the mechanism by which EPCR enables APC to activate PAR-1 in endothelial cells. We discovered that the dose of receptor activation is not responsible for the paradoxical effect of PAR-1 signaling by APC and thrombin in endothelial cells. Furthermore, the interaction of APC with EPCR is not associated with an improvement in the catalytic efficiency of the protease toward PAR-1. The possible EPCR and PAR-1 dependent recognition and signaling mechanism of APC and thrombin is discussed.


Blood ◽  
2009 ◽  
Vol 114 (9) ◽  
pp. 1968-1973 ◽  
Author(s):  
Geerte L. Van Sluis ◽  
Tatjana M.H. Niers ◽  
Charles T. Esmon ◽  
Wikky Tigchelaar ◽  
Dick J. Richel ◽  
...  

Activated protein C (APC) has both anticoagulant activity and direct cell-signaling properties. APC has been reported to promote cancer cell migration/invasion and to inhibit apoptosis and therefore may exacerbate metastasis. Opposing these activities, APC signaling protects the vascular endothelial barrier through sphingosine-1-phosphate receptor-1 (S1P1)activation, which may counteract cancer cell extravasation. Here, we provide evidence that endogenous APC limits cancer cell extravasation, with in vivo use of monoclonal antibodies against APC. The protective effect of endogenous APC depends on its signaling properties. The MAPC1591 antibody that only blocks anticoagulant activity of APC does not affect cancer cell extravasation as opposed to MPC1609 that blocks anticoagulant and signaling properties of APC. Combined administration of anti-APC antibodies and S1P1 agonist (SEW2871) resulted in a similar number of pulmonary foci in mice in presence and absence of APC, indicating that the protective effect of APC depends on the S1P1 pathway. Moreover, endogenous APC prevents cancer cell–induced vascular leakage as assessed by the Evans Blue Dye assay, and SEW2871 treatment reversed MPC1609-dependent vascular leakage. Finally, we show that cancer cells combined with MPC1609 treatment diminished endothelial VE-cadherin expression. In conclusion, endogenous APC limits cancer cell extravasation because of S1P1-mediated VE-cadherin–dependent vascular barrier enhancement.


2013 ◽  
Vol 305 (2) ◽  
pp. L185-L192 ◽  
Author(s):  
Boniface Obiako ◽  
Wendy Calchary ◽  
Ningyong Xu ◽  
Ryan Kunstadt ◽  
Bianca Richardson ◽  
...  

It is becoming increasingly apparent that cAMP signals within the pulmonary endothelium are highly compartmentalized, and this compartmentalization is critical to maintaining endothelial barrier integrity. Studies demonstrate that the exogenous soluble bacterial toxin, ExoY, and heterologous expression of the forskolin-stimulated soluble mammalian adenylyl cyclase (AC) chimera, sACI/II, elevate cytosolic cAMP and disrupt the pulmonary microvascular endothelial barrier. The barrier-disruptive effects of cytosolic cAMP generated by exogenous soluble ACs are in contrast to the barrier-protective effects of subplasma membrane cAMP generated by transmembrane AC, which strengthens endothelial barrier integrity. Endogenous soluble AC isoform 10 (AC10 or commonly known as sAC) lacks transmembrane domains and localizes within the cytosolic compartment. AC10 is uniquely activated by bicarbonate to generate cytosolic cAMP, yet its role in regulation of endothelial barrier integrity has not been addressed. Here we demonstrate that, within the pulmonary circulation, AC10 is expressed in pulmonary microvascular endothelial cells (PMVECs) and pulmonary artery endothelial cells (PAECs), yet expression in PAECs is lower. Furthermore, pulmonary endothelial cells selectively express bicarbonate cotransporters. While extracellular bicarbonate generates a phosphodiesterase 4-sensitive cAMP pool in PMVECs, no such cAMP response is detected in PAECs. Finally, addition of extracellular bicarbonate decreases resistance across the PMVEC monolayer and increases the filtration coefficient in the isolated perfused lung above osmolality controls. Collectively, these findings suggest that PMVECs have a bicarbonate-sensitive cytosolic cAMP pool that disrupts endothelial barrier integrity. These studies could provide an alternative mechanism for the controversial effects of bicarbonate correction of acidosis of acute respiratory distress syndrome patients.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 3274-3274
Author(s):  
Jong-Sup Bae ◽  
Alireza R. Rezaie

Abstract Abstract 3274 A pathogenic role for high mobility group box 1 (HMGB1) protein has been postulated in severe sepsis. Activated protein C (APC) is the only FDA-approved drug for severe sepsis, however, its effect on HMGB1 signaling has never been investigated. Here, we monitored the effect of APC on the LPS-mediated release of HMGB1 and the HMGB1-mediated modulation of pro-inflammatory signaling responses in human umbilical vein endothelial cells. APC potently inhibited the LPS-mediated release of HMGB1 and down-regulated the cell surface expression of VCAM-1, ICAM-1 and E-selectin as well as the adhesion of the monocytic cell line, THP-1, to HMGB1-activated endothelial cells. HMGB1 up-regulated pro-inflammatory responses by interacting with three pathogen-related pattern recognition receptors: toll-like receptors 2, 4 and the receptor for advanced glycation end products. APC not only inhibited the LPS-mediated HMGB1 release, but also down-regulated the cell surface expression of all three HMGB1 receptors on endothelial cells. The protective effects of APC were mediated through both EPCR and PAR-1, as evidenced by the function-blocking antibodies to either receptor abrogating the signaling function of APC. Interestingly, a thrombin derivative containing the Gla-domain of APC recapitulated all protective effects of APC with a 20–50-fold higher efficacy. These results suggest that the EPCR- and PAR-1-dependent protective effects of APC in severe sepsis may partially be mediated through the inhibition of HMGB1 signaling and that the chimeric thrombin mutant has potential therapeutic utility for severe sepsis. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 2802-2802
Author(s):  
Fabian Stavenuiter ◽  
Laurent O. Mosnier

Abstract Introduction: Endothelial barrier protective effects of activated protein C (APC) require the endothelial protein C receptor (EPCR), protease activated receptor 1 (PAR1), and PAR3. In contrast, PAR1 and PAR3 activation by thrombin results in barrier disruption. Non-canonical PAR1 and PAR3 activation by APC versus canonical activation by thrombin provide an explanation for the functional selectivity of these proteases. APC induces non-canonical PAR3 activation at Arg41 and synthetic peptides representing the tethered-ligand sequence of PAR3 after non-canonical cleavage (P3R) induce barrier protective effects in vitro and vascular integrity in vivo. However, signaling mechanisms employed by PAR3 remain undefined. To obtain better insights into the relation between coagulation proteases with endothelial barrier protective effects and canonical/non-canonical PAR1 and PAR3 activation, the PAR proteolysis analysis was extended to factor Xa (FXa). Similar to APC, FXa-mediates endothelial barrier protective effects that involve both PAR1 and EPCR. To date, however, no role for PAR3 in FXa-induced barrier integrity has been implicated. Results: In the presence of EPCR, FXa cleaved PAR1 at Arg41 similar to thrombin and not at Arg46 alike APC, whereas FXa cleaved PAR3 at the non-canonical Arg41 similar to APC but not at the canonical Lys38 corresponding to cleavage by thrombin. Surprisingly, changes in electric cell-substrate impedance sensing (ECIS) using the iCelligence system showed FXa induced an immediate drop in endothelial cell index (~60%) comparable to that induced by thrombin, indicating that FXa induced a loss of cell barrier function. Notwithstanding, after incubation of endothelial cells with FXa for 3 hours, FXa protected (~40%) against TRAP-induced loss of barrier function, similar to that induced by APC, confirming barrier protective effects of FXa. PAR1 blocking antibodies prevented the early FXa-mediated loss of barrier function, indicating that PAR1 cleavage at Arg41 was responsible for this.In contrast,a combinationofPAR1 and PAR2 blocking antibodies was needed to inhibit late (3h) FXa-mediated barrier protection. Blocking antibodies against PAR3 confirmed that canonical PAR3 activation enhanced PAR1-mediated barrier disruptive effects of thrombin (~15%). PAR3 blocking antibodies also significantly reduced the barrier protective effect of FXa (~15%), indicating a functional role for non-canonical PAR3 activation by FXa. Neither canonical (P3K) nor non-canonical (P3R) PAR3 tethered-ligand peptides directly induced significant phosphorylation of ERK1/2 or Akt in endothelial cells. The P3K however, but not the P3R peptide, enhanced TRAP induced ERK1/2 phosphorylation. No Akt phosphorylation was observed in endothelial cells treated with TRAP in the presence of either P3K or P3R. Interestingly, both APC and FXa but not thrombin induced prolonged activation of the endothelial cell specific Tie2 receptor, determined by phosphorylation of Y992 and S1119. Tie2 activation by FXa required PAR3 and EPCR with a partial contribution of PAR1 and PAR2. P3R induced potent activation of Tie2 achieving maximal activation at ~0.8 µM P3R, whereas P3K failed to do so. Additionally, neither (non-)canonical PAR1 nor PAR2 tethered-ligand peptides induced activation of Tie2. Activation of Tie2 by P3R was relatively fast and reached half-maximal activation in about 5 minutes. Blocking antibodies against Tie2 reduced FXa-mediated barrier protective effects by approximately 34%, whereas inhibition of Tie2 did not affect thrombin mediated barrier disruption. Immunohistochemistry indicated that Tie2 activation by FXa and P3R resulted in clustering of activated Tie2 at the cell borders. Accordingly, Tie2 activation by FXa and P3R resulted in changes in the cellular distribution of the tight-junction-associated protein zona occludens (ZO-1) in time. Conclusion: Here we identified a novel pathway for Tie2 activation by non-canonical PAR3 activation that promoted tight-junction formation and endothelial barrier protective effects. In contrast, canonical activation of PAR3 enhanced PAR1-mediated barrier disruptive effects by thrombin. These results exemplify the novel dimensions that non-canonical activation of PARs provides for the possible molecular mechanisms that are responsible for the functional selectivity of protease signaling. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2008 ◽  
Vol 111 (5) ◽  
pp. 2667-2673 ◽  
Author(s):  
Reto A. Schuepbach ◽  
Clemens Feistritzer ◽  
Lawrence F. Brass ◽  
Matthias Riewald

Activated protein C (APC) signals in endothelial cells ex vivo through protease activated receptor-1 (PAR1). However, it is controversial whether PAR1 can mediate APC's protective effects in sepsis because the inflammatory response results in thrombin generation and thrombin proteolytically activates PAR1 much more efficiently than APC. Here we show that APC can induce powerful barrier protective responses in an endothelial cell monolayer in the presence of thrombin. Using cell surface immunoassays with conformation sensitive monoclonal anti-PAR1 antibodies we analyzed cleavage of endogenous PAR1 on the endothelial cell surface by APC in the absence and presence of thrombin. Incubation with APC caused efficient PAR1 cleavage and upon coincubation with thrombin APC supported additional PAR1 cleavage. Thrombin-cleaved PAR1 rapidly disappeared from the cell surface whereas, unexpectedly, the APC-cleaved PAR1 remained and could be detected on the cell surface, even when thrombin at concentrations of up to 1 nM was also present. Our findings demonstrate for the first time directly that APC can generate a distinct PAR1 population on endothelial cells in the presence of thrombin. The data suggest that different trafficking of activated PAR1 might explain how PAR1 signaling by APC can be relevant when thrombin is present.


1986 ◽  
Vol 56 (02) ◽  
pp. 115-119 ◽  
Author(s):  
Eugene G Levin ◽  
David M Stern ◽  
Peter P Nawroth ◽  
Richard A Marlar ◽  
Daryl S Fair ◽  
...  

SummaryThe addition of thrombin (9 nM) to primary cultures of human endothelial cells induces a 6- to 7-fold increase in the rate of release of tissue plasminogen activator (tPA). Several other serine proteases which specifically interact with endothelial cells were also analyzed for their effect on tPA release. Gamma-thrombin, an autocatalytic product of α-thrombin, promoted tPA release but was less effective than α-thrombin. A maximum increase of 5.5-fold was observed, although a concentration of γ-thrombin 20 times greater than α-thrombin was required. The response to Factor Xa was similar to α-thrombin, although the stimulation was significantly reduced by the addition of hirudin or DAPA suggesting that prothrombin activation was occurring. The simultaneous addition of prothrombin with Factor Xa resulted in enhanced tPA release equal to that observed with an equimolar concentration of active α-thrombin. Thus, under these conditions, Factor Xa-cell surface mediated activation of prothrombin can lead to a secondary effect resulting from cell-thrombin interaction. Activated protein C, which has been implicated as a profibrinolytic agent, was also tested. No change in tPA release occurred after the addition of up to 325 nM activated protein C in the presence or absence of proteins. Factor IXa and plasmin were also ineffective. The effect of thrombin on the endothelial cell derived plasminogen activator specific inhibitor was also studied. Thrombin produced a small but variable release of the inhibitor with an increase of less than twice that of non-thrombin treated controls.


Sign in / Sign up

Export Citation Format

Share Document