scholarly journals Factor VIIa Induces Biased Cytoprotective Signaling through the Cleavage of Protease Activated Receptor 1 at Canonical Arg41 Site

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 481-481
Author(s):  
Vijay Kumar Reddy Kondreddy ◽  
Usha R. Pendurthi ◽  
Xiao Xu ◽  
John H. Griffin ◽  
L. Vijaya Mohan Rao

Endothelial cell protein C receptor (EPCR) interacts with diverse ligands, in addition to its known ligands protein C and activated protein C (APC). We reported earlier that procoagulant clotting factor VIIa (FVIIa) binds EPCR with the same affinity as APC. FVIIa binding to EPCR leads to the down regulation of the EPCR-mediated anticoagulation pathway. Our recent studies showed that FVIIa, like APC, induces EPCR-dependent cytoprotective signaling through activation of protease activated receptor 1 (PAR1). Recent studies of Griffin, Mosnier and their colleagues revealed that APC noncanonical cleavage of PAR1 at Arg46 site that generates a novel tethered ligand is responsible for APC-induced β-arrestin2-dependent PAR1 biased signaling. It is unknown at present whether FVIIa follows a similar mechanism as APC in inducing PAR1 biased signaling. PAR1 reporter constructs - wild-type and cleavage site-specific mutants - were routinely used to investigate PAR1 cleavage by thrombin, APC, or other proteases, and to determine protease-specific cleavage sites in PAR1. Unfortunately, this approach was not useful in determining any FVIIa cleavage site in PAR1. In contrast to thrombin or APC, FVIIa treatment failed to show a detectable cleavage (over the background) of transfected wild-type PAR1 reporter constructs expressed in cultured endothelial cells. However, in other studies, FVIIa was shown to cleave endogenous PAR1 in endothelial cells as assessed by the loss of cleavage-specific PAR1 mAb binding. The recent generation of transgenic mice strains carrying R41Q or R46Q homozygous point mutations in PAR1 has allowed us in the present study to investigate in vivo mechanisms for PAR1-dependent cytoprotective signaling of FVIIa. We employed two murine injury models, LPS-induced inflammation and VEGF-induced barrier disruption. Murine brain endothelial cells isolated from the PAR1 mutated strains and primary human endothelial cells were used to validate in vivo findings and extend the mechanistic studies. Our studies show that administration of rFVIIa (250 µg/kg body weight) reduced LPS-induced cytokine elaboration and neutrophil infiltration in the lung tissues of wild-type (WT) PAR1 and QQ46-PAR1 mice but not in QQ41-PAR1 mice. Similarly, FVIIa suppression of the VEGF-induced barrier disruption was abolished in the QQ41-PAR1 mice but not in WT and the QQ46-PAR1 mice. Parallel experiments conducted with APC showed, as expected, that it protected WT and QQ41-PAR1 mice but not QQ46-PAR1 mice against LPS-induced inflammation and VEGF-mediated barrier destabilization. In vitro signaling studies performed with brain endothelial cells isolated from WT, QQ41-PAR1 and QQ46-PAR1 mice showed that FVIIa activation of Akt in endothelial cells required Arg41 in PAR1. Additional studies showed that FVIIa-cleaved endogenous PAR1 was readily internalized, whereas APC-cleaved PAR1 remained on the cell surface. Very low concentrations of thrombin (< 1 nM) mimicked FVIIa in inducing PAR1-dependent cytoprotective signaling. However, very low concentration thrombin-induced cytoprotective signaling differed from EPCR-FVIIa-induced cytoprotective signaling in the isoform of ß-arrestin required for the protective effect. EPCR-FVIIa-induced PAR1-mediated cytoprotective signaling was soley mediated via the β-arrestin1-dependent pathway whereas very low dose thrombin-induced cytoprotective effects appear to be mediated by either β-arrestin1 or β-arrestin2. Silencing of ß-arrestin1 or ß-arrestin2 alone did not affect the protective effects of very low doses of thrombin, but the silencing both ß-arrestin1 and ß-arrestin2 together completely prevented a low dose of thrombin-induced protective effect. In summary, our data strongly support the hypothesis that the in vivo mechanism of action for FVIIa's pharmacologic benefits in curbing inflammation and endothelial barrier disruption involves biased signaling of PAR1 due to cleavage at Arg41. Moreover, our studies demonstrate that very low concentrations of thrombin are also capable of inducing PAR1 biased beneficial -cytoprotective signaling by coupling to either ß-arrestin1 or 2. These results emphasize the striking diversity of PAR1's conformational states and interactomes that initiate canonical and biased signaling. Disclosures No relevant conflicts of interest to declare.

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.


2020 ◽  
Vol 117 (9) ◽  
pp. 5039-5048 ◽  
Author(s):  
Ying Lin ◽  
Jacob M. Wozniak ◽  
Neil J. Grimsey ◽  
Sravan Girada ◽  
Anand Patwardhan ◽  
...  

Thrombin, a procoagulant protease, cleaves and activates protease-activated receptor-1 (PAR1) to promote inflammatory responses and endothelial dysfunction. In contrast, activated protein C (APC), an anticoagulant protease, activates PAR1 through a distinct cleavage site and promotes anti-inflammatory responses, prosurvival, and endothelial barrier stabilization. The distinct tethered ligands formed through cleavage of PAR1 by thrombin versus APC result in unique active receptor conformations that bias PAR1 signaling. Despite progress in understanding PAR1 biased signaling, the proteins and pathways utilized by thrombin versus APC signaling to induce opposing cellular functions are largely unknown. Here, we report the global phosphoproteome induced by thrombin and APC signaling in endothelial cells with the quantification of 11,266 unique phosphopeptides using multiplexed quantitative mass spectrometry. Our results reveal unique dynamic phosphoproteome profiles of thrombin and APC signaling, an enrichment of associated biological functions, including key modulators of endothelial barrier function, regulators of gene transcription, and specific kinases predicted to mediate PAR1 biased signaling. Using small interfering RNA to deplete a subset of phosphorylated proteins not previously linked to thrombin or APC signaling, a function for afadin and adducin-1 actin binding proteins in thrombin-induced endothelial barrier disruption is unveiled. Afadin depletion resulted in enhanced thrombin-promoted barrier permeability, whereas adducin-1 depletion completely ablated thrombin-induced barrier disruption without compromising p38 signaling. However, loss of adducin-1 blocked APC-induced Akt signaling. These studies define distinct thrombin and APC dynamic signaling profiles and a rich array of proteins and biological pathways that engender PAR1 biased signaling in endothelial cells.


Blood ◽  
2011 ◽  
Vol 117 (11) ◽  
pp. 3199-3208 ◽  
Author(s):  
Prosenjit Sen ◽  
Ramakrishnan Gopalakrishnan ◽  
Hema Kothari ◽  
Shiva Keshava ◽  
Curtis A. Clark ◽  
...  

Abstract Recent studies have shown that factor VIIa (FVIIa) binds to the endothelial cell protein C receptor (EPCR), a cellular receptor for protein C and activated protein C, but the physiologic significance of this interaction is unclear. In the present study, we show that FVIIa, upon binding to EPCR on endothelial cells, activates endogenous protease activated receptor-1 (PAR1) and induces PAR1-mediated p44/42 mitogen-activated protein kinase (MAPK) activation. Pretreatment of endothelial cells with FVIIa protected against thrombin-induced barrier disruption. This FVIIa-induced, barrier-protective effect was EPCR dependent and did not involve PAR2. Pretreatment of confluent endothelial monolayers with FVIIa before thrombin reduced the development of thrombin-induced transcellular actin stress fibers, cellular contractions, and paracellular gap formation. FVIIa-induced p44/42 MAPK activation and the barrier-protective effect are mediated via Rac1 activation. Consistent with in vitro findings, in vivo studies using mice showed that administration of FVIIa before lipopolysaccharide (LPS) treatment attenuated LPS-induced vascular leakage in the lung and kidney. Overall, our present data provide evidence that FVIIa bound to EPCR on endothelial cells activates PAR1-mediated cell signaling and provides a barrier-protective effect. These findings are novel and of great clinical significance, because FVIIa is used clinically for the prevention of bleeding in hemophilia and other bleeding disorders.


Blood ◽  
2012 ◽  
Vol 120 (26) ◽  
pp. 5237-5246 ◽  
Author(s):  
Laurent O. Mosnier ◽  
Ranjeet K. Sinha ◽  
Laurent Burnier ◽  
Eveline A. Bouwens ◽  
John H. Griffin

Abstract Activated protein C (APC) exerts endothelial cytoprotective actions that require protease-activated receptor 1 (PAR1), whereas thrombin acting via PAR1 causes endothelial disruptive, proinflammatory actions. APC's activities, but not thrombin's, require PAR1 located in caveolae. PAR1 is a biased 7-transmembrane receptor because G proteins mediate thrombin's signaling, whereas β-arrestin 2 mediates APC's signaling. Here we elucidate novel mechanisms for APC's initiation of signaling. Biochemical studies of APC's protease specificity showed that APC cleaved PAR1 sequences at both Arg41 and Arg46. That PAR1 cleavage at Arg46 can occur on cells was supported by APC's cleavage of N-terminal-SEAP-tagged R41Q-PAR1 but not R41Q/R46Q-PAR1 mutants transfected into cells and by anti-PAR1 epitope mapping of APC-treated endothelial cells. A synthetic peptide composing PAR1 residues 47-66, TR47, stimulated protective signaling in endothelial cells as reflected in Akt and glycogen synthase kinase 3β phosphorylation, Ras-related C3 botulinum toxin substrate 1 activation, and barrier stabilization effects. In mice, the TR47 peptide reduced VEGF-induced vascular leakage. These in vitro and in vivo data imply that the novel PAR1 N-terminus beginning at residue Asn47, which is generated by APC cleavage at Arg46, mediates APC's cytoprotective signaling and that this unique APC-generated N-terminal peptide tail is a novel biased agonist for PAR1.


Blood ◽  
2003 ◽  
Vol 102 (9) ◽  
pp. 3224-3231 ◽  
Author(s):  
Hiroshi Kataoka ◽  
Justin R. Hamilton ◽  
David D. McKemy ◽  
Eric Camerer ◽  
Yao-Wu Zheng ◽  
...  

AbstractDefining the relative importance of protease-activated receptors (PARs) for thrombin signaling in mouse endothelial cells is critical for a basic understanding of thrombin signaling in these cells and for the rational use of knockout mice to probe the roles of thrombin's actions on endothelial cells in vivo. We examined thrombin- and PAR agonist–induced increases in cytoplasmic calcium, phosphoinositide hydrolysis, extracellular signal-regulated kinase (ERK) phosphorylation, and gene expression in endothelial cells from wild-type and PAR-deficient mice. PAR1 and PAR4 agonists triggered responses in wild-type but not in Par1–/– and Par4–/– endothelial cells, respectively. Calcium imaging confirmed that a substantial fraction of individual endothelial cells responded to both agonists. Compared with wild-type cells, Par1–/– endothelial cells showed markedly decreased responses to low concentrations of thrombin, and cells that lacked both PAR1 and PAR4 showed no responses to even high concentrations of thrombin. Similar results were obtained when endothelial-dependent vasorelaxation of freshly isolated mouse aorta was used as an index of signaling in native endothelial cells. Thus PAR1 is the major thrombin receptor in mouse endothelial cells, but PAR4 also contributes. These receptors serve at least partially redundant roles in endothelial cells in vitro and in vivo and together are necessary for the thrombin responses measured.


2015 ◽  
Vol 114 (12) ◽  
pp. 1144-1155 ◽  
Author(s):  
Eveline A. M. Bouwens ◽  
Fabian Stavenuiter ◽  
Laurent O. Mosnier

SummaryThe protein C (PC) system conveys beneficial anticoagulant and cytoprotective effects in numerous in vivo disease models. The endothelial protein C receptor (EPCR) plays a central role in these pathways as cofactor for PC activation and by enhancing activated protein C (APC)-mediated protease-activated receptor (PAR) activation. During inflammatory disease, expression of EPCR on cell membranes is often diminished thereby limiting PC activation and APC’s effects on cells. Here a caveolae-targeting glycosylphosphatidylinositol (GPI)-anchored EPCR (EPCR-GPI) was engineered to restore EPCR’s bioavailability via “cell painting.” The painting efficiency of EPCR-GPI on EPCR-depleted endothelial cells was time- and dose-dependent. The EPCR-GPI bioavailability after painting was long lasting since EPCR surface levels reached 400 % of wild-type cells after 2 hours and remained > 200 % for 24 hours. EPCR-GPI painting conveyed APC binding to EPCR-depleted endothelial cells where EPCR was lost due to shedding or shRNA. EPCR painting normalised PC activation on EPCR-depleted cells indicating that EPCR-GPI is functional active on painted cells. Caveolin-1 lipid rafts were enriched in EPCR after painting due to the GPI-anchor targeting caveolae. Accordingly, EPCR painting supported PAR1 and PAR3 cleavage by APC and augmented PAR1-dependent Akt phosphorylation by APC. Thus, EPCR-GPI painting achieved physiological relevant surface levels on endothelial cells, restored APC binding to EPCR-depleted cells, supported PC activation, and enhanced APC-mediated PAR cleavage and cytoprotective signalling. Therefore, EPCRGPI provides a novel tool to restore the bioavailability and functionality of EPCR on EPCR- depleted and -deficient cells.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 137-137
Author(s):  
Madhusudhan Thati ◽  
Sanchita Ghosh ◽  
Hongjie Wang ◽  
Wolfram Ruf ◽  
Berend H. Isermann

Abstract Introduction Coagulation protease signaling via protease-activated receptors (PARs) is essential for maintenance of cellular homeostasis. Perturbed or aberrant activation of protease-dependent signaling via PARs propagates inflammation and pathological responses in disease models such as sepsis, neurological diseases and metabolic diseases including atherosclerosis, obesity and diabetic nephropathy (dNP). Disruption of protease-activated protein C (aPC) signaling in renal epithelial cells, i.e. podocytes, compromises adaptive endoplasmic reticulum (ER) signaling, promoting maladaptive ER-stress and ultimately dysfunction of the glomerular filtration barrier and dNP. While these results demonstrate that aPC-dependent ER-reprogramming is an essential component of its cytoprotective effect, currently the co-receptors that are involved in mediating aPC's cytoprotective effects in podocytes remain unclear. In endothelial cells, endothelial protein C receptor (EPCR) facilitates the binding and alternative proteolytic cleavage of PAR1 propagating biased anti-inflammatory and cytoprotective effects. Intriguingly, unlike in endothelial cells, podocytes do not express EPCR and the cytoprotective effects of aPC requires PAR3/PAR2 heterodimer (human podocytes) and PAR3/PAR1 heterodimer (mouse podocytes). Podocytes express integrins β1 and β3, which could potentially mediate biased protease signaling. Methods To characterize the receptor-dependent mechanism through which aPC-mediates its cytoprotective effects in podocytes, we generated transgenic mice that expresses constitutively higher levels of wild type-human aPC (APChigh mice) and integrin binding deficient mutant (RGE-APChigh mice). Additionally, the receptor-dependent mechanisms in dNP were evaluated in mouse models with podocyte specific genetic deletion of the integrin β3. After 26 weeks of persistent hyperglycaemia (streptozotocin-induced diabetes), markers of dNP were determined. In wild type diabetic mice, integrin β3 antagonist Cyclo-RGDfv and PAR1 antagonist (SCH79797) was administered to pharmacologically inhibit integrin and PAR1 signaling, respectively. Results In vitro data show that integrin αvβ3 in podocytes controls protease-dependent spatial and temporal regulation of PAR signaling by aPC. This protease binds to integrin αvβ3 and temporally regulates PAR signaling. Abolishing aPC-αvβ3 interaction or knockdown of αvβ3 integrin perturbs endosomal trafficking of PARs resulting in aberrant cell-disruptive signaling. Likewise, in vivo overexpression of integrin binding deficient aPC-mutant or podocyte-specific genetic deletion of integrin β3 abrogates aPC-dependent cytoprotective effects in a mouse model of dNP. Akin to wild type aPC, pharmacological inhibition of aberrant PAR1 signaling protects against dNP. Corroborating our results in murine dNP, analysis of human dNP samples revealed increased expression and activation of αvβ3 associated with altered PAR receptor localization specifically within the podocytes. Conclusion Our results show that aPC-αvβ3 interaction on podocytes modulates endosomal trafficking of PAR receptors and that loss of this interaction drives aberrant protease-dependent signaling and pathological podocyte migration. Complementary in vivo studies in newly developed integrin binding deficient aPC mutant mice demonstrate that integrin αvβ3 ligation is essential for nephroprotective aPC signaling. These results identify integrin αvβ3 as a rheostat that provides temporal and spatial regulation of PAR signaling in diabetic disease. Disclosures No relevant conflicts of interest to declare.


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.


2003 ◽  
Vol 373 (1) ◽  
pp. 65-70 ◽  
Author(s):  
Laurent O. MOSNIER ◽  
John H. GRIFFIN

In a model of staurosporine-induced apoptosis using EAhy926 endothelial cells, inhibition of apoptosis by activated protein C was dose-dependent and required the enzyme's active site, implicating activated protein C-mediated proteolysis. Consistent with this implication, both protease-activated receptor-1 (PAR-1) and endothelial cell protein C receptor (EPCR) were required for the anti-apoptotic effects of activated protein C.


Sign in / Sign up

Export Citation Format

Share Document