Next Article in Journal
Treating BCG-Induced Cystitis with Combined Chondroitin and Hyaluronic Acid Instillations in Bladder Cancer
Previous Article in Journal
Pre-Exposure Prophylaxis and Treatment with Tixagevimab/Cilgavimab for COVID-19 among Immunocompromised Pediatric Patients
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Endothelial Protein C Receptor and Its Impact on Rheumatic Disease

by
Zachary Daniel O’Hehir
1,
Tom Lynch
2,
Sean O’Neill
3,
Lyn March
2,3 and
Meilang Xue
1,2,*
1
Sutton Arthritis Research Laboratory, Sydney Musculoskeletal Health, Kolling Institute, Faculty of Medicine and Health, The University of Sydney at Royal North Shore Hospital, Sydney, NSW 2065, Australia
2
The Australian Arthritis and Autoimmune Biobank Collaborative (A3BC), Institute of Bone and Joint Research, Kolling Institute, Faculty of Medicine and Health, University of Sydney at Royal North Shore Hospital, St Leonards, NSW 2065, Australia
3
Department of Rheumatology, Royal North Shore Hospital, Syndey, NSW 2065, Australia
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2024, 13(7), 2030; https://doi.org/10.3390/jcm13072030
Submission received: 12 March 2024 / Revised: 28 March 2024 / Accepted: 29 March 2024 / Published: 31 March 2024
(This article belongs to the Section Immunology)

Abstract

:
Endothelial Protein C Receptor (EPCR) is a key regulator of the activated protein C anti-coagulation pathway due to its role in the binding and activation of this protein. EPCR also binds to other ligands such as Factor VII and X, γδ T-cells, plasmodium falciparum erythrocyte membrane protein 1, and Secretory group V Phospholipases A2, facilitating ligand-specific functions. The functions of EPCR can also be regulated by soluble (s)EPCR that competes for the binding sites of membrane-bound (m)EPCR. sEPCR is created when mEPCR is shed from the cell surface. The propensity of shedding alters depending on the genetic haplotype of the EPCR gene that an individual may possess. EPCR plays an active role in normal homeostasis, anti-coagulation pathways, inflammation, and cell stemness. Due to these properties, EPCR is considered a potential effector/mediator of inflammatory diseases. Rheumatic diseases such as rheumatoid arthritis and systemic lupus erythematosus are autoimmune/inflammatory conditions that are associated with elevated EPCR levels and disease activity, potentially driven by EPCR. This review highlights the functions of EPCR and its contribution to rheumatic diseases.

1. Introduction

The Endothelial Protein C Receptor (EPCR, CD201) is a transmembrane protein that is expressed by a variety of cell types in the human body, including myeloid cells, tissue-resident cells, and even platelets. It plays a vital role in mediating the anti-clotting and anti-inflammatory functions of a natural anticoagulant, activated protein C (APC) [1]. EPCR can also interact with various other ligands, eliciting ligand-dependent functions. Additionally, EPCR is homologous to MHCI/CD1 family proteins [2], with the potential to regulate both innate and adaptive immunity. Moreover, EPCR has been identified as a promising stem cell marker for multiple cell types in both humans and mice. It is worth noting that EPCR’s functions can be affected by its soluble form (sEPCR), which competes for the binding sites of membrane-bound (m)EPCR. sEPCR is created when mEPCR is shed from the cell surface, and its circulating level depends mainly on the genetic haplotype of the EPCR gene that an individual may possess. Higher levels of EPCR have been found in many inflammatory/autoimmune diseases and may contribute to disease pathogenesis. This review summarises the functions of EPCR and its contribution to rheumatic diseases.

2. Endothelial Cell Protein C Receptor

The human EPCR gene is located at position 20q11.2 with a length of 8 kb and comprises four exons separated by three introns [3,4]. The 5′ untranslated region and a single peptide is coded by exon 1, the extracellular domain is coded by exon 2 and 3, and exon 4 is responsible for the 3′ untranslated region as well as the transmembrane domain and the cytoplasmic tail [4]. The encoded human EPCR protein is an N-glycosylated type 1 membrane protein. It has a similar 3D representation to the MHCI/CD1 proteins; however, a characterising deep groove that is usually required for antigen binding instead contains a lipid in EPCR [1,5]. In most cases, this lipid will be phosphatidylcholine; however, recent studies have found a variety of lipids that can be bound to EPCR with varying frequency [6]. The bound lipid contributes to the binding of EPCR to Protein C (PC), likely by maintaining the structure of EPCR rather than through direct contact [1,7] (Figure 1).
Human EPCR has also been found to bear a great homology of 62% to murine protein CCD41 as they come from the same gene and are altered through post-transcriptional or post-translational processes [9,10]. The proteins are distinguished by their unique cell position. CCD41 is predominantly located in the centrosome close to the nucleus and in the perinuclear vesicles during cell cycle progression and, as a result, can only indirectly interact with APC. EPCR has both surface cell receptor and soluble forms that can directly bind to APC, unlike CCD41 [11,12].

2.1. EPCR Expression and Localisation

EPCR expression was originally detected in the endothelium of large blood vessels [13,14] and the microvascular endothelium of the lungs, heart, and skin to a lesser extent [15]. Subsequently, other cell types found to express EPCR included neutrophils, natural killer cells, monocytes, keratinocytes, smooth muscle cells, cardiomyocytes, eosinophils neurons, placental trophoblasts, and dendritic cells [16,17,18,19], mucosal tissues such as the lung and gut, and joint synovial fibroblasts [20]. Additionally, hematopoietic, epithelial, neuronal, and multipotent progenitor cells; breast cancer stem cells; and skin epidermal stem cells express EPCR, and EPCR acts as a potential stem cell marker for these cells [21,22,23,24,25,26,27,28].
EPCR on the cell surface, also known as mEPCR, is predominantly localised in membrane microdomains such as caveolin-1. EPCR can also be detected in the endosomes and colocalised with lysosome protein marker 1 in smaller quantities [14]. Through endocytosis and recycling of ligand-bound surface EPCR, the cell can internalise certain ligands through a dynamin and caveolar-dependent pathway such that they can be translocated to extravascular tissues, thereby impacting ligand bioavailability [14,29]. This process is regulated in a temporospatial manner by specific Ras-like small GTPases [30]. Intracellular EPCR has been found in endothelial cells, mostly around the perinuclear region, with some dispersed throughout the cytoplasm [14].

2.2. EPCR Function

EPCR is crucial in maintaining normal homeostasis and regulating coagulation and inflammation. Most of the experimental evidence on the physiological and pathological importance of EPCR is derived from studies on gene-modified mice.

2.2.1. Regulation of Normal Homeostasis

Deletion of the EPCR gene causes early embryonic lethality in mice [31]. Further investigation revealed critical EPCR expression on trophoblast giant cells [31]. Mice with EPCR expression on placenta giant trophoblasts being carried to term develop normally and are healthy [32]. Mice with a variant of EPCR that cannot bind to PC/APC develop splenomegaly due to bone marrow (BM) failure [33], a phenotype that was not observed in the aforementioned EPCR-deficient mice [32]. BM transplant experiments suggest that EPCR modulates haematopoiesis [33].

2.2.2. Regulation of Coagulation

EPCR is known to promote the activation of PC to APC on the cell surface [34] and regulate the anti-coagulation pathways. In mice, disrupting the EPCR gene can lead to fibrin deposition and placental thrombosis [31]. As EPCR-deficient mice age, they generate more thrombin and activate less PC in response to procoagulant stimuli, resulting in increased spontaneous thrombin formation [32]. In a mouse model of haemophilia, this EPCR deficiency is responsible for the reduced intra-articular bleeding initially and the reduced severity of related arthropathy in the longer term [35]. Furthermore, mice with an EPCR variant that cannot bind to PC/APC have impaired activation of PC and increased generation of thrombin in response to thrombotic and inflammatory challenges compared to wild-type mice [33].

2.2.3. Regulation of Inflammation

EPCR plays a crucial role in controlling inflammation [1]. Overexpression of EPCR protects mice from endotoxin-induced injury [36], and EPCR-deficient mice displayed a higher mortality rate, more thrombin generation, platelet consumption, and immune cell sequestration in the lung tissue in response to lipopolysaccharide (LPS) [37]. These mice are more susceptible to dextran sulphate sodium -induced colitis, manifested by inflammation and mucosal barrier disruption [38]. These findings are consistent with previous studies on EPCR low-expression mice [39] and baboons treated with EPCR-blocking antibodies [40]. EPCR can inhibit Th17 cells, and T cell-specific EPCR deficiency exacerbates experimental autoimmune encephalomyelitis in mice [41]. Paradoxically, EPCR on circulating T cells positively correlates with disease activity in psoriasis [42]. Similarly, elevated levels of EPCR can predict poor outcomes of severe lung infection and inflammation [43], colorectal and lung cancers [44,45], and lupus nephritis patients [46]. Conversely, EPCR deficiency ameliorates murine inflammatory arthritis, protects against bacterial-induced lung injury [47], inhibits joint bleeding-induced inflammation [35], and deters the development of lupus and antiphospholipid syndrome [48] in mice. The underlying mechanisms of EPCR’s conflicting functions in different diseases are not clear but may be associated with its ligands present on specific cells/organs.

2.2.4. Regulation of Stemness

EPCR has been identified as a promising stem cell marker for several cell types. It is a dependable marker of long-term haematopoietic stem cells (HSCs) [49] and can be used to specifically identify HSCs in murine BM [50]. EPCR also marks human foetal liver HSCs [51]; human epidermal stem cells [28]; human cord blood HSCs [25]; and progenitor cells of the endothelium [52], neuron [53] and epithelium [54,55]. EPCR is also expressed by highly aggressive basal-like breast cancer subtypes. In aggressive triple-negative breast cancer cells, EPCR expression is a characteristic of cancer stem cell-like populations with tumour-initiating properties in vivo [56]. Interestingly, the functions of EPCR in coagulation and inflammation are mainly controlled by EPCR on non-hematopoietic cells [37,57]. Selective deletion of non-hematopoietic EPCR almost completely abolishes PC activation [37].

2.3. EPCR Function Mediators

2.3.1. Soluble EPCR

Cell surface EPCR can be shed through a process mediated by TNF-α converting enzyme/ADAM17 (TACE) [58] or via alternative mRNA splicing [59] to generate sEPCR. Additionally, pre-treatment of samples with phorbol-12-myristate 13-acetate (PMA), TNF-α, IL-1β, or LPS induces EPCR shedding (Figure 2) [60,61,62,63]. sEPCR does not have the transmembrane and cytoplasmic tail domain of mEPCR [64], but it can bind both PC and APC with an affinity similar to mEPCR. Binding PC to sEPCR rather than mEPCR blocks surface interactions with negatively charged phospholipids that are required for the efficient inactivation of factor (F)V and FVIIIa. This binding also inhibits the activation of PC to APC, thus interfering with the anti-inflammatory and anti-coagulative effects of EPCR [65,66,67]. High plasma sEPCR can result in a low mEPCR level and reduced APC activity, leading to a pro-thrombotic state in the body [68]. Plasma sEPCR, therefore, has the potential as a marker for hypercoagulable states [64,69].

2.3.2. Anti-EPCR Autoantibodies

High levels of anti-EPCR autoantibodies have been observed in patients with antiphospholipid syndrome (APS) [70], a condition that is linked with thrombosis and foetal death. These autoantibodies sometimes obstruct the binding of PC to EPCR, thus hampering the generation of APC [70]. Recently, anti-EPCR autoantibodies have also been detected in patients with Takayasu arteritis—a type of large vessel vasculitis. Interestingly, patients with these autoantibodies were more prone to strokes [71]. Additionally, more than 60% of patients with primary ulcerative colitis (UC) tested positive for anti-EPCR autoantibodies [71,72], which suggests that these antibodies may prove useful in diagnosing UC. In the general population, the presence of anti-EPCR autoantibodies is a moderate risk factor for deep vein thrombosis [73]. Among young women, anti-EPCR autoantibodies have been associated with acute myocardial infarction [74] and are independent risk factors for foetal death [70].

2.3.3. EPCR Genetic Variants

The EPCR gene has several functional genetic mutations that lead to reduced expression of EPCR or receptor dysfunction. Such mutations may be the result of a rare 23-bp insertion in exon 3 [75] or a variety of single-nucleotide polymorphisms (SNPs) in coding or non-coding regions of the EPCR gene. There are sixteen SNPs present in the EPCR gene, which can be divided into four haplotypes (H1–4) [67,76,77].
H1 contains the combination of 10 specific alleles that have been altered from the common alleles of the H2 haplotype. These are the 1451T (rs2069943), 1541A (rs2069944), 1880C (rs2069945), 2532C (rs2069948), 2897A (rs945960), 3424C (rs871480), 3997C (rs2069952), 4678C (rs9574), 5632G (rs1415773), and 5663A (rs1415774) alleles. This haplotype causes an increase in mEPCR that subsequently increases the levels of APC and provides anti-coagulative effects (Figure 2) [78,79]. The specific SNP responsible for this increased level of mEPCR is currently unknown.
The H3 haplotype is tagged by the g.4600A>G minor allele (Ser219Gly; rs867186), with the rs867186-GG genotype, in particular, being responsible for the increased production of plasma sEPCR [67,77,79,80,81,82,83,84], causing an increase in risk for pro-coagulative disease states such as venous and arterial thrombosis, deep vein thrombosis, and miscarriage [65,67,85,86,87]. This mutation predicts an amino acid change in Ser219Gly in the receptor’s transmembrane region, facilitating the removal of EPCR from the cytomembrane via metalloproteinases (MMPs). Furthermore, it is associated with a truncated form of EPCR mRNA that lacks the transmembrane and intracellular domains and is likely responsible for additional cleavage of mEPCR to sEPCR (Figure 2) [59]. SNP rs867186 explains ~85% of the phenotypic variance [82] and is responsible for elevated levels of plasma sEPCR during inflammatory disorders. The frequency of Ser219Gly polymorphism in the general population is about 12% [83]. EPCR H3 is also associated with an increase in the levels of plasma PC that is speculated to be caused by a decrease in mEPCR levels and EPCR-dependent activation of PC due to excess shedding [79,80,88,89]. Similarly, higher levels of plasma FVII are associated with the H3 haplotype [90].
Finally, the H2 haplotype is the set of common alleles for the gene [67,77], while H4 rarely occurs and is associated with a slight increase in the risk of venous thromboembolism [77].
Figure 2. Regulation of EPCR shedding. mEPCR is cleaved from the cell surface through a process mediated by TACE to create sEPCR. TACE cleavage is regulated by Polo-like kinase 2 (PLK2), mitogen-activated protein kinases (MAPKs), and protein kinase C (PKC) [91] and can be induced by pre-treatment of cells with PMA, TNF-α, or IL-1β [60,61,62,63]. Once cleaved, the mEPCR and sEPCR will compete for ligand binding, making fewer mEPCR-ligand functions realised [64]. H1 favours mEPCR compared to the common allele of H2 due to decreased TACE cleavage. In contrast, H3 favours sEPCR via increased TACE cleavage and increased production of truncated mRNA that creates a functionally similar isoform of sEPCR [59,67,77,78].
Figure 2. Regulation of EPCR shedding. mEPCR is cleaved from the cell surface through a process mediated by TACE to create sEPCR. TACE cleavage is regulated by Polo-like kinase 2 (PLK2), mitogen-activated protein kinases (MAPKs), and protein kinase C (PKC) [91] and can be induced by pre-treatment of cells with PMA, TNF-α, or IL-1β [60,61,62,63]. Once cleaved, the mEPCR and sEPCR will compete for ligand binding, making fewer mEPCR-ligand functions realised [64]. H1 favours mEPCR compared to the common allele of H2 due to decreased TACE cleavage. In contrast, H3 favours sEPCR via increased TACE cleavage and increased production of truncated mRNA that creates a functionally similar isoform of sEPCR [59,67,77,78].
Jcm 13 02030 g002

3. EPCR Ligands and Ligand Associated Functions

Many functions of EPCR depend on its interaction with a variety of ligands. A previous report showed that EPCR was constitutively translocated into the nucleus [92], indicating that EPCR may internalise its ligands and translocate them into the nucleus, which could directly influence gene expression [14]. Below are some of the ligands that EPCR binds to and their resulting outcomes upon binding.

3.1. Activated Protein C

The PC/APC pathway provides several cytoprotective properties, including anti-clotting, anti-inflammatory, anti-apoptotic, and barrier-protective functions [93]. The binding of thrombomodulin to thrombin activates PC to APC, which in turn allows for the inactivation of FV and FVIII via a complex made of APC, Protein S (PS), phospholipids, and calcium [94] (Figure 3). The binding of EPCR to PC enhances the activation of PC by 20-fold, thus providing an anti-coagulation effect [34,95,96]. Blocking EPCR binding to PC accelerates thrombus development in a murine model of thrombosis, confirming that EPCR plays a crucial role in regulating coagulation [64,97]. EPCR-deficient mice are significantly protected from acquired haemophilia, suggesting that the EPCR-mediated APC anticoagulant pathway plays a critical role in haemophilia [98].
After being activated, APC can either dissociate from EPCR and perform anticoagulant functions or remain bound to EPCR and exhibit cell-signalling cytoprotective activities (Figure 3) [1]. When bound to EPCR, APC cleaves PAR1 at Arg-46, which triggers β-arrestin-2 biased PAR1 signalling [99,100,101,102] and exerts its anti-apoptotic, anti-inflammatory, and barrier stabilising functions [103,104,105]. Additionally, when bound to EPCR, APC elicits biased cytoprotective signalling through the cleavage of PAR3 at the Arg41 noncanonical site [49,106]. Moreover, the interaction of APC with EPCR, PAR3, and neutrophil-expressed Mac-1 generates the signalling required to inhibit PMA-induced neutrophil extracellular trap (NET) formation [107]. The APC-EPCR signalling pathways are the main mechanisms for APC’s beneficial effects in various autoimmune and inflammatory diseases, including type 1 diabetes [108], inflammatory arthritis [109], systemic lupus erythematosus (SLE), and lupus nephritis (LN) [110].
Figure 3. EPCR-PC pathways. When PC is bound to EPCR the activation of PC by thrombin–thrombomodulin complex is increased by approximately 20-fold [95,96]. Once activated, APC will either dissociate from EPCR to directly inactivate coagulation factors FVa or FVIIa with the assistance of PS [94] or APC will remain bound to EPCR to illicit cytoprotective functions via cleavage and activation of PARs [93].
Figure 3. EPCR-PC pathways. When PC is bound to EPCR the activation of PC by thrombin–thrombomodulin complex is increased by approximately 20-fold [95,96]. Once activated, APC will either dissociate from EPCR to directly inactivate coagulation factors FVa or FVIIa with the assistance of PS [94] or APC will remain bound to EPCR to illicit cytoprotective functions via cleavage and activation of PARs [93].
Jcm 13 02030 g003

3.2. Factor VII

EPCR may exhibit APC/PC-independent anticoagulant activities via FVII interaction. Human recombinant FVIIa in pharmacological concentrations inhibits EPCR-mediated APC production on endothelial cells, suggesting competition between FVIIa and PC/APC for EPCR binding [98,111]. Further studies have confirmed that EPCR is a receptor for FVII, FVIIa, PC, and APC with similar affinity [14,64]. When bound to EPCR, FVIIa is internalised, which could indicate that EPCR assists FVIIa translocation and signalling pathways (Figure 4) [64]. Like APC, FVIIa bound to EPCR does not modulate thrombin-induced endothelial barrier permeability [112]; rather, it elicits a PAR1 and β-arrestin-1 dependent anti-inflammatory signalling pathway [113]. This binding attenuates LPS-induced vascular leakage in the mouse lung and kidney [114]. Furthermore, PAR1-mediated p44/42 mitogen-activated protein kinase (MAPK) activation is induced upon EPCR binding and activation of endogenous PAR1 [113]. Additionally, the binding of EPCR and FVIIa downregulates the production of APC and contributes to a haemostatic effect in haemophilia [98].

3.3. Factor X

FX is a zymogen that plays a significant role in the coagulation cascade. Once activated to FXa, it competes with APC for the binding site of EPCR in a calcium-dependent manner [115]. When bound to EPCR, FXa cleaves and activates PARs, which are crucial for the activation of coagulation pathways. Activation of PAR1 through FXa depended on the EPCR expressed in Chinese hamster ovary cells [116]. EPCR promotes efficient cleavage of PAR1 and PAR2 by the Tissue Factor (TF)-FVIIa-FXa complex (Figure 4); however, it does not change the efficacy of PAR2 cleavage by TF-FVIIa [49]. Furthermore, EPCR activates PAR2 via the ternary TF-FVIIa-FXa [117], acting as a necessary coreceptor in cell signalling. This is further supported by EPCR-dependent TF-FVIIa-FXa-mediated activation of the p44/42 MAPK signalling cascade [118]. Moreover, EPCR greatly improves the efficacy of noncanonical PAR3 cleavage by FXa (Figure 4). This FXa-EPCR-induced PAR3 cleavage results in prolonged activation of the barrier-protective tunica intima endothelial receptor tyrosine kinase 2 [119].

3.4. γδ T Cells

γδ T cells are important for immunity at barrier surfaces [120]. EPCR has been identified as a ligand for the subpopulation of T lymphocytes that bear the γδ T-cell antigen receptor [93,121]. A subpopulation of γδ T cells called Vδ2¯− γδ T cells can recognise cytomegalovirus-infected cells through binding to EPCR. The β sheet of EPCR is the binding site for this subpopulation of γδ T cells, and the binding is dependent on conformational integrity instead of lipid binding [122]. This binding of γδ T cells to EPCR may play a role in the surveillance of endothelium for viral infections or malignancies (Figure 4) [122,123].

3.5. Plasmodium Falciparum Erythrocyte Membrane Protein 1 (PfEMP1)

PfEMP1 is found on the surface of erythrocytes infected by Plasmodium falciparum. During malaria infection, PfEMP1 will compete with APC for the binding site of EPCR, resulting in the cytoprotective activation of PAR1 not being achieved (Figure 4) [124]. Furthermore, during malaria infection, EPCR is a major host receptor for the sequestration of Plasmodium falciparum-infected erythrocytes in the brain and other vital organs. The increased expression of EPCR-binding PfEMP1 is associated with progressively more severe disease [125].

3.6. Secretory Group V Phospholipases A2 (sPLA2V)

Phospholipases A2 are part of a superfamily of enzymes responsible for catalysing glycerophospholipids’ hydrolysis [126]. sPLA2V inhibited the generation and anti-apoptotic effects of APC by binding to EPCR on the endothelial cells (Figure 4) [127,128]. The binding of sPLA2V to EPCR also mediates the aggressive behaviour of RA synovial fibroblasts [129]. The binding occurs by exchanging phosphatidylcholine for lysophosphatidylcholine in the hydrophobic groove

3.7. Proteinase-3 (PR3)/Macrophage-1 Antigen (Mac-1)

PR3, also named myeloblastin, is mainly recognised for its role in breaking down enzymes and structural proteins. It is stored within the azurophilic granules of polymorphonuclear neutrophils alongside cathepsin G and neutrophil elastase [130,131]. When neutrophils are activated, PR3 is released, which binds activated neutrophils to sEPCR [132,133]. Additionally, PR3 plays a role in the degradation of EPCR as it cleaves EPCR at multiple sites. Further studies have revealed that β2 integrin Mac-1 forms a heteromeric complex with PR3, suggesting that Mac-1 contributes to the binding between sEPCR and PR3 [134]. However, studies have shown that EPCR can bind to Mac-1 directly on monocytes and share the same site as APC binding [134,135,136].

3.8. Autoantibodies to Phospholipids

aPLs are a category of antibodies that target proteins that bind to phospholipids and are associated with various inflammatory/autoimmune diseases, including antiphospholipid syndrome (APS), SLE, and RA. These antibodies can activate coagulation pro-inflammatory pathways. A recent study discovered that EPCR is a cell surface target for aPLs [48]. In APS and SLE, the pathogenic effects of aPLs appear to be mediated by their interaction with EPCR. The lipid-bound in the groove of EPCR is phosphatidylcholine in most cases. However, it can be exchanged with lysophosphatidic acid (LBPA), resulting in an EPCR-LBPA complex that facilitates the endosomal trafficking of aPLs [48,137]. EPCR has been shown to interact with aPLs to activate trophoblast cells, monocytes, and dendritic cells. Monocytes and trophoblasts produce more pro-inflammatory cytokines (TNF and type 1 IFN) when aPLs bind to EPCR, which leads to the internalisation of aPLs (Figure 4) [48,137].
These observations indicate that EPCR may play a broader role in influencing various pathophysiological processes by interacting with different ligands in different milieus.
Figure 4. Ligand function summary. EPCR binds to FVII/FVIIa with similar affinity to APC and will compete with APC for ligand binding. When bound to EPCR, FVIIa will be internalised with EPCR and activate endogenous PAR1. FXa can bind to EPCR in a TF-FVIIa-FXa complex, or it can bind to EPCR by itself. The TF-FVIIa-FXa will incite PAR2 activation, which is necessary for TLR4 and interferon regulation host defences [117]. By itself, FXa bound to EPCR will cleave and activate PAR3 to prolong activation of the barrier-protective tunica intima endothelial receptor tyrosine kinase 2 [119]. γδ T cells bind to EPCR at a different site to APC and will cause a potential increase in endothelial surveillance for viral infections or malignancies [122,123]. PfEMP1 on malaria-infected erythrocytes competes for the APC binding site on EPCR [124]. It is associated with the severity of malaria [125]. sPLA2-V competes with APC for EPCR binding and mediates the aggressive behaviour of synovial fibroblasts from rheumatoid arthritis (RA) [127,129]. aPLs undergo internalisation when bound to EPCR. The lipid bound in the groove of EPCR is phosphatidylcholine in most cases. However, it can be exchanged with LBPA, resulting in an EPCR-LBPA complex that facilitates the endosomal trafficking of aPLs [48,137].
Figure 4. Ligand function summary. EPCR binds to FVII/FVIIa with similar affinity to APC and will compete with APC for ligand binding. When bound to EPCR, FVIIa will be internalised with EPCR and activate endogenous PAR1. FXa can bind to EPCR in a TF-FVIIa-FXa complex, or it can bind to EPCR by itself. The TF-FVIIa-FXa will incite PAR2 activation, which is necessary for TLR4 and interferon regulation host defences [117]. By itself, FXa bound to EPCR will cleave and activate PAR3 to prolong activation of the barrier-protective tunica intima endothelial receptor tyrosine kinase 2 [119]. γδ T cells bind to EPCR at a different site to APC and will cause a potential increase in endothelial surveillance for viral infections or malignancies [122,123]. PfEMP1 on malaria-infected erythrocytes competes for the APC binding site on EPCR [124]. It is associated with the severity of malaria [125]. sPLA2-V competes with APC for EPCR binding and mediates the aggressive behaviour of synovial fibroblasts from rheumatoid arthritis (RA) [127,129]. aPLs undergo internalisation when bound to EPCR. The lipid bound in the groove of EPCR is phosphatidylcholine in most cases. However, it can be exchanged with LBPA, resulting in an EPCR-LBPA complex that facilitates the endosomal trafficking of aPLs [48,137].
Jcm 13 02030 g004

4. EPCR in Rheumatic Diseases

Rheumatic diseases refer to a group of autoimmune conditions that cause inflammation and pain in the joints, muscles, or fibrous tissues. EPCR plays a potential role in the development and ongoing activity of these debilitating conditions due to its role as a mediator of coagulation and inflammatory pathways. This review summarises current knowledge of EPCR functions in rheumatic diseases, focusing on RA and SLE.

4.1. EPCR and RA

RA is one of the most prevalent chronic inflammatory diseases, affecting ~0.5–1% of the population, predominantly women. It primarily affects synovial joints but also impacts other organ systems [138], resulting in disability and premature death. The inflammation and tissue damage in RA are mediated by a coordinated response from the innate and adaptive immune system, including inflammatory cells and their autoantibody and cytokine production [139,140]. Currently, 269 SNPs have been identified as risk genes for RA development [141]. The gene with the closest correlation to RA disease progression is the human leukocyte antigen DRB1*04 epitope, which is shared by 80% of RA patients [142,143,144].
EPCR is significantly elevated in the synovium of patients with RA, while it aids APC in suppressing RA monocyte activation and migration [20]. This points to the possibility of EPCR having a protective role in mediating inflammatory states in RA. Additionally, it has been found that higher concentrations of EPCR may be associated with anti-inflammatory and protective effects due to its essential role in creating APC [99,145]. In fact, mice models that were treated with APC showed a decrease in arthritis severity by approximately 50% and a reduction in disease incidence by 40% [109].
Despite the cytoprotective properties of EPCR through the APC pathway, EPCR is observed to promote inflammatory responses by interacting with other ligands or through sEPCR. In RA synovial fibroblasts, EPCR promotes an inflammatory response through interacting with sPLA2V. This ligand competes with APC for the same ligand binding site [129], which interferes with regular APC-EPCR interactions and stimulates RA synovial proliferation and destruction [109]. This may also contribute to the production of sEPCR since the sera of arthritic mice have significantly higher levels of sEPCR than healthy controls [109,146]. Short-cut mRNA is increased in RA patients compared to osteoarthritis patients, indicating that variable EPCR mRNA splicing plays a key role in the overproduction of sEPCR in RA patients [146].
EPCR is known to interact with T cells and neutrophils in patients with RA, which can have both protective and destructive effects. In RA patients, lower levels of CD4+ EPCR+ T cells are observed compared to patients with osteoarthritis. This may lead to an increase in pathogenic Th17 cell differentiation in RA patients [146]. Contrarily, mice with severe EPCR deficiency displayed more than 40% reduced arthritis incidence and 50% decreased disease severity when compared to normal mice [147].
A potential mechanism for how EPCR may regulate RA is through the γδ T cell ligand. The concentration of γδ T cells is higher in human RA patients [148,149], and the percentage of Vδ2 T cells was negatively correlated to RA disease activity [121]. Another mechanism could involve NETs; the expression of NETs appears to increase when treated with EPCR-neutralising antibodies. Therefore, EPCR may regulate the secretion of NETs in RA patients through the APC-EPCR signalling pathway, thereby affecting the progression of disease in RA patients [150].

4.2. EPCR and SLE

SLE is a potentially fatal, chronic, multisystem autoimmune disorder that typically affects women between puberty and menopause. The disease is characterised by an abnormal response of the immune system involving B cells, T cells, dendritic cells, and macrophages [151]. The exact pathogenesis of the disease is still unknown; however, research suggests that environmental, genetic, and epigenetic factors may play a role in the development and progression of SLE. Currently, there are 60 genes identified as risk factors for SLE [151].
Several studies have observed increased sEPCR levels in SLE patients [152,153,154]. The severity of the disease and the use of corticosteroid treatment can impact the levels of sEPCR in SLE patients [152]. An individual’s EPCR haplotype may also play a role in the development of SLE, as the H3 haplotype is more commonly observed in SLE patients than in healthy controls. However, some patients with elevated sEPCR levels still have the H1, H2, and H4 haplotypes [153]. Moreover, complications of SLE, such as lupus nephritis (LN), can also affect the levels of EPCR. While SLE causes elevated sEPCR levels, patients with LN have significantly higher levels of sEPCR than patients with SLE who do not develop LN [155,156,157]. The increase in sEPCR levels in LN patients may be explained by the conversion of elevated mEPCR levels, which are found in the cortical peritubular capillaries of these patients, to sEPCR due to the shedding by the inflammatory cytokines IFN-γ and IL-1 [46,155].
APS is an autoimmune disease that often occurs in patients with SLE. It is characterised by repeated blood clots and is associated with the presence of aPLs. aPLs can be specifically recognised by the EPCR-LBPA complex on cell surfaces. This recognition can activate trophoblast cells, monocytes, and dendritic cells, leading to the development of APS autoimmunity [48]. In mice, blocking the EPCR-LBPA signalling pathway can prevent the development of APS, SLE-like syndrome, and kidney pathology associated with SLE [48]. Therefore, EPCR-LPBA signalling is a potential central mechanism for the development of SLE-like APS-related autoimmune diseases.

4.3. EPCR and Other Rheumatic Diseases

Scleroderma describes a group of rare autoimmune diseases that cause the hardening and tightening of skin. Arthritis and tendinopathy are common developments of scleroderma. In scleroderma, there is a downregulation of endothelial EPCR by Fli1 that could induce a hypercoagulable state, leading to tissue fibrosis and disruption of peripheral circulation [158].
Multiple Sclerosis (MS) is an autoimmune disease that affects the central nervous system and is associated with an increase in the subsequent diagnosis of RA [159]. Like RA, there are elevated levels of sEPCR in MS patients, which exceed that of SLE patients [160].
Haemophilic arthropathy (HA) is a debilitating joint condition. A murine study showed that mice with EPCR deficiency were afflicted with less severe HA due to reduced joint bleeding from lack of EPCR. The therapeutic, recombinant FVIIa was also more effective at preventing HA in the EPCR-deficient mice. EPCR deficiency is thought to have reduced the severity of HA by reducing the recurrence of spontaneous joint bleeding. It is also possible that the lack of APC generation results in enough thrombin generation to prevent further joint bleeding in HA [35].

5. Conclusions

EPCR is a transmembrane protein with various functions in several disease states, its most prominent contribution being its involvement in the PC/APC pathway. It is regulated by competition for its binding site between its membrane-bound and soluble form. The propensity for EPCR to come in either of these forms can vary depending on the haplotype of the EPCR gene, with H3 producing more sEPCR and H1 maintaining more mEPCR. In this review, we identified PC/APC, FVIIa, FXa, γδ T cells, PfEMP1, sPLA2V, Mac-1, PR3, and aPLs as ligands that can bind to EPCR, each of them providing unique cytoprotective or destructive effects. Many of these ligands have cytoprotective properties when bound to EPCR, such as through the cleavage of PARs; however, ligands such as sPLA2V and PfEMP1 have destructive properties when bound to EPCR. EPCR can regulate immune function by inhibiting Th17 cells and is useful as a marker for stem cells such as human epidermal stem cells or HSCs.

6. Clinical Implications and Future Perspectives

EPCR is a highly flexible protein that is expressed by different cell types and plays a vital role in coagulation, inflammation, and haemostasis. These key properties of EPCR make it a potential biomarker and target for treating various diseases. Among the rheumatic diseases, RA and SLE stand out as having significant associations with EPCR. In both SLE and APS, the binding of aPLs with the EPCR-LBPA complex induces pathogenic effects. Disrupting the formation of the EPCR-LBPA complex on the cell surface or preventing aPLs from binding to the EPCR-LBPA complex could be an effective approach for EPCR-targeted therapy for SLE and APS. The same mechanism may apply to RA, although there is no confirmation yet. However, studies have shown that mice with severe EPCR deficiency had a much lower disease incidence and milder arthritis [147], indicating that EPCR could also be a potential therapeutic target for RA. Therefore, it is essential to validate these findings in large clinical cohorts to assess EPCR function and its potential as a therapeutic target for rheumatoid diseases.
To create therapeutics that target EPCR for rheumatoid diseases, it is important to design them in a way that preserves EPCR’s ability to bind with other ligands, especially with PC/APC. This is necessary to maintain the anti-coagulation and anti-inflammatory properties of APC, as APC has been shown to have beneficial effects on inflammatory arthritis [109], SLE, and LN [110]. However, targeting EPCR may have both positive and negative effects, given its well-established functions in homeostasis, coagulation, inflammation, and stemness.
It is still uncertain how exactly EPCR contributes to pathogenesis in rheumatic diseases, and the role of other ligands such as sPLA2V, PR3, Mac-1, γδ T cells, FVIIa, and FXa in these diseases has not been fully explored. Therefore, future research should aim to expand our knowledge of how these ligands interact with EPCR in order to develop targeted adjunctive therapies for rheumatic diseases.

Author Contributions

Conceptualisation, M.X., L.M. and S.O.; Software, Z.D.O., M.X. and T.L.; Resources, M.X. and L.M.; Writing—Original Draft Preparation, Z.D.O. and M.X.; Writing—Review and Editing, M.X., Z.D.O., T.L., L.M. and S.O.; Supervision, M.X.; Funding Acquisition, M.X. and L.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

This project was partly supported by the Woodend Foundation, the Ulysses Club, The Lincoln Centre, Arthritis Australia, and CLEARbridge Foundation. Figures were created with BioRender.com.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Esmon, C.T. Structure and functions of the endothelial cell protein C receptor. Crit. Care Med. 2004, 32, S298–S301. [Google Scholar] [CrossRef] [PubMed]
  2. Fukudome, K.; Esmon, C.T. Identification, cloning, and regulation of a novel endothelial cell protein c activated protein c receptor. J. Biol. Chem. 1994, 269, 26486–26491. [Google Scholar] [CrossRef] [PubMed]
  3. Liang, Z.; Rosen, E.D.; Castellino, F.J. Nucleotide structure and characterization of the murine gene encoding the endothelial cell protein C receptor. Thromb. Haemost. 1999, 81, 585–588. [Google Scholar] [CrossRef] [PubMed]
  4. Simmonds, R.E.; Lane, D.A. Structural and functional implications of the intron/exon organization of the human endothelial cell protein C/activated protein C receptor (EPCR) gene: Comparison with the structure of CD1/major histocompatibility complex alpha1 and alpha2 domains. Blood 1999, 94, 632–641. [Google Scholar] [CrossRef]
  5. Liaw, P.C.; Mather, T.; Oganesyan, N.; Ferrell, G.L.; Esmon, C.T. Identification of the protein C/activated protein C binding sites on the endothelial cell protein C receptor. Implications for a novel mode of ligand recognition by a major histocompatibility complex class 1-type receptor. J. Biol. Chem. 2001, 276, 8364–8370. [Google Scholar] [CrossRef] [PubMed]
  6. Erausquin, E.; Moran-Garrido, M.; Saiz, J.; Barbas, C.; Dichiara-Rodriguez, G.; Urdiciain, A.; Lopez-Sagaseta, J. Identification of a broad lipid repertoire associated to the endothelial cell protein C receptor (EPCR). Sci. Rep. 2022, 12, 15127. [Google Scholar] [CrossRef] [PubMed]
  7. Oganesyan, V.; Oganesyan, N.; Terzyan, S.; Qu, D.; Dauter, Z.; Esmon, N.L.; Esmon, C.T. The crystal structure of the endothelial protein C receptor and a bound phospholipid. J. Biol. Chem. 2002, 277, 24851–24854. [Google Scholar] [CrossRef]
  8. Ly, D.; Moody, D.B. The CD1 size problem: Lipid antigens, ligands, and scaffolds. Cell. Mol. Life Sci. 2014, 71, 3069–3079. [Google Scholar] [CrossRef] [PubMed]
  9. Fukudome, K.; Esmon, C.T. Molecular cloning and expression of murine and bovine endothelial cell protein C/activated protein C receptor (EPCR). The structural and functional conservation in human, bovine, and murine EPCR. J. Biol. Chem. 1995, 270, 5571–5577. [Google Scholar] [CrossRef]
  10. Rothbarth, K.; Dabaghian, A.R.; Stammer, H.; Werner, D. One single mRNA encodes the centrosomal protein CCD41 and the endothelial cell protein C receptor (EPCR). FEBS Lett. 1999, 458, 77–80. [Google Scholar] [CrossRef]
  11. Rothbarth, K.; Petzelt, C.; Lu, X.; Todorov, I.T.; Joswig, G.; Pepperkok, R.; Ansorge, W.; Werner, D. cDNA-derived molecular characteristics and antibodies to a new centrosome-associated and G2/M phase-prevalent protein. J. Cell Sci. 1993, 104 Pt 1, 19–30. [Google Scholar] [CrossRef] [PubMed]
  12. Fukudome, K.; Kurosawa, S.; Stearns-Kurosawa, D.J.; He, X.; Rezaie, A.R.; Esmon, C.T. The endothelial cell protein C receptor. Cell surface expression and direct ligand binding by the soluble receptor. J. Biol. Chem. 1996, 271, 17491–17498. [Google Scholar] [CrossRef] [PubMed]
  13. Laszik, Z.; Mitro, A.; Taylor, F.B., Jr.; Ferrell, G.; Esmon, C.T. Human protein C receptor is present primarily on endothelium of large blood vessels: Implications for the control of the protein C pathway. Circulation 1997, 96, 3633–3640. [Google Scholar] [CrossRef] [PubMed]
  14. Nayak, R.C.; Sen, P.; Ghosh, S.; Gopalakrishnan, R.; Esmon, C.T.; Pendurthi, U.R.; Rao, L.V. Endothelial cell protein C receptor cellular localization and trafficking: Potential functional implications. Blood 2009, 114, 1974–1986. [Google Scholar] [CrossRef] [PubMed]
  15. Ye, X.; Fukudome, K.; Tsuneyoshi, N.; Satoh, T.; Tokunaga, O.; Sugawara, K.; Mizokami, H.; Kimoto, M. The endothelial cell protein C receptor (EPCR) functions as a primary receptor for protein C activation on endothelial cells in arteries, veins, and capillaries. Biochem. Biophys. Res. Commun. 1999, 259, 671–677. [Google Scholar] [CrossRef] [PubMed]
  16. Kerschen, E.; Hernandez, I.; Zogg, M.; Jia, S.; Hessner, M.J.; Fernandez, J.A.; Griffin, J.H.; Huettner, C.S.; Castellino, F.J.; Weiler, H. Activated protein C targets CD8+ dendritic cells to reduce the mortality of endotoxemia in mice. J. Clin. Investig. 2010, 120, 3167–3178. [Google Scholar] [CrossRef] [PubMed]
  17. Gleeson, E.M.; O’Donnell, J.S.; Preston, R.J. The endothelial cell protein C receptor: Cell surface conductor of cytoprotective coagulation factor signaling. Cell. Mol. Life Sci. 2012, 69, 717–726. [Google Scholar] [CrossRef] [PubMed]
  18. Xue, M.; Campbell, D.; Sambrook, P.N.; Fukudome, K.; Jackson, C.J. Endothelial protein C receptor and protease-activated receptor-1 mediate induction of a wound-healing phenotype in human keratinocytes by activated protein C. J. Investig. Dermatol. 2005, 125, 1279–1285. [Google Scholar] [CrossRef] [PubMed]
  19. Feistritzer, C.; Sturn, D.H.; Kaneider, N.C.; Djanani, A.; Wiedermann, C.J. Endothelial protein C receptor-dependent inhibition of human eosinophil chemotaxis by protein C. J. Allergy Clin. Immunol. 2003, 112, 375–381. [Google Scholar] [CrossRef]
  20. Xue, M.; March, L.; Sambrook, P.N.; Fukudome, K.; Jackson, C.J. Endothelial protein C receptor is overexpressed in rheumatoid arthritic (RA) synovium and mediates the anti-inflammatory effects of activated protein C in RA monocytes. Ann. Rheum. Dis. 2007, 66, 1574–1580. [Google Scholar] [CrossRef]
  21. Shipitsin, M.; Campbell, L.L.; Argani, P.; Weremowicz, S.; Bloushtain-Qimron, N.; Yao, J.; Nikolskaya, T.; Serebryiskaya, T.; Beroukhim, R.; Hu, M.; et al. Molecular definition of breast tumor heterogeneity. Cancer Cell 2007, 11, 259–273. [Google Scholar] [CrossRef] [PubMed]
  22. Iwasaki, H.; Arai, F.; Kubota, Y.; Dahl, M.; Suda, T. Endothelial protein C receptor-expressing hematopoietic stem cells reside in the perisinusoidal niche in fetal liver. Blood 2010, 116, 544–553. [Google Scholar] [CrossRef] [PubMed]
  23. Park, S.Y.; Lee, H.E.; Li, H.; Shipitsin, M.; Gelman, R.; Polyak, K. Heterogeneity for Stem Cell–Related Markers According to Tumor Subtype and Histologic Stage in Breast Cancer. Clin. Cancer Res. 2010, 16, 876–887. [Google Scholar] [CrossRef] [PubMed]
  24. Balazs, A.B.; Fabian, A.J.; Esmon, C.T.; Mulligan, R.C. Endothelial protein C receptor (CD201) explicitly identifies hematopoietic stem cells in murine bone marrow. Blood 2006, 107, 2317–2321. [Google Scholar] [CrossRef] [PubMed]
  25. Fares, I.; Chagraoui, J.; Lehnertz, B.; MacRae, T.; Mayotte, N.; Tomellini, E.; Aubert, L.; Roux, P.P.; Sauvageau, G. EPCR expression marks UM171-expanded CD34(+) cord blood stem cells. Blood 2017, 129, 3344–3351. [Google Scholar] [CrossRef] [PubMed]
  26. Che, J.L.C.; Bode, D.; Kucinski, I.; Cull, A.H.; Bain, F.; Becker, H.J.; Jassinskaja, M.; Barile, M.; Boyd, G.; Belmonte, M.; et al. Identification and characterization of in vitro expanded hematopoietic stem cells. EMBO Rep. 2022, 23, e55502. [Google Scholar] [CrossRef] [PubMed]
  27. Lin, D.S.; Trumpp, A. Differential expression of endothelial protein C receptor (EPCR) in hematopoietic stem and multipotent progenitor cells in young and old mice. Cells Dev. 2023, 174, 203843. [Google Scholar] [CrossRef]
  28. Xue, M.; Dervish, S.; Chan, B.; Jackson, C.J. The endothelial protein C receptor is a potential stem cell marker for epidermal keratinocytes. Stem Cells 2017, 35, 1786–1798. [Google Scholar] [CrossRef]
  29. Clark, C.A.; Vatsyayan, R.; Hedner, U.; Esmon, C.T.; Pendurthi, U.R.; Rao, L.V. Endothelial cell protein C receptor-mediated redistribution and tissue-level accumulation of factor VIIa. J. Thromb. Haemost. 2012, 10, 2383–2391. [Google Scholar] [CrossRef]
  30. Nayak, R.C.; Keshava, S.; Esmon, C.T.; Pendurthi, U.R.; Rao, L.V. Rab GTPases regulate endothelial cell protein C receptor-mediated endocytosis and trafficking of factor VIIa. PLoS ONE 2013, 8, e59304. [Google Scholar] [CrossRef]
  31. Gu, J.M.; Crawley, J.T.; Ferrell, G.; Zhang, F.; Li, W.; Esmon, N.L.; Esmon, C.T. Disruption of the endothelial cell protein C receptor gene in mice causes placental thrombosis and early embryonic lethality. J. Biol. Chem. 2002, 277, 43335–43343. [Google Scholar] [CrossRef] [PubMed]
  32. Li, W.; Zheng, X.; Gu, J.M.; Ferrell, G.L.; Brady, M.; Esmon, N.L.; Esmon, C.T. Extraembryonic expression of EPCR is essential for embryonic viability. Blood 2005, 106, 2716–2722. [Google Scholar] [CrossRef] [PubMed]
  33. Pepler, L.; Yu, P.; Dwivedi, D.J.; Trigatti, B.L.; Liaw, P.C. Characterization of mice harboring a variant of EPCR with impaired ability to bind protein C: Novel role of EPCR in hematopoiesis. Blood 2015, 126, 673–682. [Google Scholar] [CrossRef] [PubMed]
  34. Feldmann, M.; Maini, R.N.; Bondeson, J.; Taylor, P.; Foxwell, B.M.; Brennan, F.M. Cytokine blockade in rheumatoid arthritis. Adv. Exp. Med. Biol. 2001, 490, 119–127. [Google Scholar] [PubMed]
  35. Magisetty, J.; Pendurthi, U.R.; Esmon, C.T.; Rao, L.V.M. EPCR deficiency or function-blocking antibody protects against joint bleeding-induced pathology in hemophilia mice. Blood 2020, 135, 2211–2223. [Google Scholar] [CrossRef] [PubMed]
  36. Li, W.; Zheng, X.; Gu, J.; Hunter, J.; Ferrell, G.L.; Lupu, F.; Esmon, N.L.; Esmon, C.T. Overexpressing endothelial cell protein C receptor alters the hemostatic balance and protects mice from endotoxin. J. Thromb. Haemost. 2005, 3, 1351–1359. [Google Scholar] [CrossRef] [PubMed]
  37. Zheng, X.; Li, W.; Song, Y.; Hu, Y.; Ferrell, G.L.; Esmon, N.L.; Esmon, C.T. Non-hematopoietic EPCR regulates the coagulation and inflammatory responses during endotoxemia. J. Thromb. Haemost. 2007, 5, 1394–1400. [Google Scholar] [CrossRef] [PubMed]
  38. Kondreddy, V.; Keshava, S.; Esmon, C.T.; Pendurthi, U.R.; Rao, L.V.M. A critical role of endothelial cell protein C receptor in the intestinal homeostasis in experimental colitis. Sci. Rep. 2020, 10, 20569. [Google Scholar] [CrossRef]
  39. Iwaki, T.; Cruz, D.T.; Martin, J.A.; Castellino, F.J. A cardioprotective role for the endothelial protein C receptor in lipopolysaccharide-induced endotoxemia in the mouse. Blood 2005, 105, 2364–2371. [Google Scholar] [CrossRef]
  40. Taylor, F.B., Jr.; Stearns-Kurosawa, D.J.; Kurosawa, S.; Ferrell, G.; Chang, A.C.; Laszik, Z.; Kosanke, S.; Peer, G.; Esmon, C.T. The endothelial cell protein C receptor aids in host defense against Escherichia coli sepsis. Blood 2000, 95, 1680–1686. [Google Scholar] [CrossRef]
  41. Kishi, Y.; Kondo, T.; Xiao, S.; Yosef, N.; Gaublomme, J.; Wu, C.; Wang, C.; Chihara, N.; Regev, A.; Joller, N.; et al. Protein C receptor (PROCR) is a negative regulator of Th17 pathogenicity. J. Exp. Med. 2016, 213, 2489–2501. [Google Scholar] [CrossRef] [PubMed]
  42. Yu, Y.; Chen, Z.; Wang, Y.; Li, Y.; Lu, J.; Cui, L.; Yu, Z.; Ding, Y.; Guo, C.; Zhang, X.; et al. Infliximab modifies regulatory T cells and co-inhibitory receptor expression on circulating T cells in psoriasis. Int. Immunopharmacol. 2021, 96, 107722. [Google Scholar] [CrossRef] [PubMed]
  43. Kager, L.M.; Schouten, M.; Wiersinga, W.J.; de Boer, J.D.; Lattenist, L.C.; Roelofs, J.J.; Meijers, J.C.; Levi, M.; Dondorp, A.M.; Esmon, C.T.; et al. Overexpression of the endothelial protein C receptor is detrimental during pneumonia-derived gram-negative sepsis (Melioidosis). PLoS Negl. Trop. Dis. 2013, 7, e2306. [Google Scholar] [CrossRef] [PubMed]
  44. Lal, N.; Willcox, C.R.; Beggs, A.; Taniere, P.; Shikotra, A.; Bradding, P.; Adams, R.; Fisher, D.; Middleton, G.; Tselepis, C.; et al. Endothelial protein C receptor is overexpressed in colorectal cancer as a result of amplification and hypomethylation of chromosome 20q. J. Pathol. Clin. Res. 2017, 3, 155–170. [Google Scholar] [CrossRef] [PubMed]
  45. Anton, I.; Molina, E.; Luis-Ravelo, D.; Zandueta, C.; Valencia, K.; Ormazabal, C.; Martinez-Canarias, S.; Perurena, N.; Pajares, M.J.; Agorreta, J.; et al. Receptor of Activated Protein C Promotes Metastasis and Correlates with Clinical Outcome in Lung Adenocarcinoma. Am. J. Respir. Crit. Care Med. 2012, 186, 6. [Google Scholar] [CrossRef] [PubMed]
  46. Izmirly, P.M.; Barisoni, L.; Buyon, J.P.; Kim, M.Y.; Rivera, T.L.; Schwartzman, J.S.; Weisstuch, J.M.; Liu, D.T.; Bernstein, S.; Tseng, C.E.; et al. Expression of endothelial protein C receptor in cortical peritubular capillaries associates with a poor clinical response in lupus nephritis. Rheumatology 2009, 48, 513–519. [Google Scholar] [CrossRef]
  47. Keshava, S.; Magisetty, J.; Tucker, T.A.; Kujur, W.; Mulik, S.; Esmon, C.T.; Idell, S.; Rao, L.V.M.; Pendurthi, U.R. Endothelial Cell Protein C Receptor Deficiency Attenuates Streptococcus Pneumoniae-induced Pleural Fibrosis. Am. J. Respir. Cell Mol. Biol. 2021, 64, 14. [Google Scholar] [CrossRef]
  48. Muller-Calleja, N.; Hollerbach, A.; Royce, J.; Ritter, S.; Pedrosa, D.; Madhusudhan, T.; Teifel, S.; Meineck, M.; Hauser, F.; Canisius, A.; et al. Lipid presentation by the protein C receptor links coagulation with autoimmunity. Science 2021, 371, eabc0956. [Google Scholar] [CrossRef]
  49. Pendurthi, U.R.; Rao, L.V.M. Endothelial cell protein C receptor-dependent signaling. Curr. Opin. Hematol. 2018, 25, 219–226. [Google Scholar] [CrossRef]
  50. Kent, D.G.; Copley, M.R.; Benz, C.; Wohrer, S.; Dykstra, B.J.; Ma, E.; Cheyne, J.; Zhao, Y.; Bowie, M.B.; Zhao, Y.; et al. Prospective isolation and molecular characterization of hematopoietic stem cells with durable self-renewal potential. Blood 2009, 113, 6342–6350. [Google Scholar] [CrossRef]
  51. Subramaniam, A.; Talkhoncheh, M.S.; Magnusson, M.; Larsson, J. Endothelial protein C receptor (EPCR) expression marks human fetal liver hematopoietic stem cells. Haematologica 2019, 104, e47–e50. [Google Scholar] [CrossRef] [PubMed]
  52. Do, M.H.; Shi, W.; Ji, L.; Ladewig, E.; Zhang, X.; Srivastava, R.M.; Capistrano, K.J.; Edwards, C.; Malik, I.; Nixon, B.G.; et al. Reprogramming tumor-associated macrophages to outcompete endovascular endothelial progenitor cells and suppress tumor neoangiogenesis. Immunity 2023, 56, 2555–2569.e5. [Google Scholar] [CrossRef] [PubMed]
  53. Ivanova, N.B.; Dimos, J.T.; Schaniel, C.; Hackney, J.A.; Moore, K.A.; Lemischka, I.R. A Stem Cell Molecular Signature. Science 2002, 298, 601–604. [Google Scholar] [CrossRef] [PubMed]
  54. Blanpain, C.; Lowry, W.E.; Geoghegan, A.; Polak, L.; Fuchs, E. Self-renewal, multipotency, and the existence of two cell populations within an epithelial stem cell niche. Cell 2004, 118, 635–648. [Google Scholar] [CrossRef] [PubMed]
  55. Ramalho-Santos, M.; Yoon, S.; Matsuzaki, Y.; Mulligan, R.C.; Melton, D.A. “Stemness”: Transcriptional profiling of embryonic and adult stem cells. Science 2002, 298, 597–600. [Google Scholar] [CrossRef] [PubMed]
  56. Hwang-Verslues, W.W.; Kuo, W.H.; Chang, P.H.; Pan, C.C.; Wang, H.H.; Tsai, S.T.; Jeng, Y.M.; Shew, J.Y.; Kung, J.T.; Chen, C.H.; et al. Multiple lineages of human breast cancer stem/progenitor cells identified by profiling with stem cell markers. PLoS ONE 2009, 4, e8377. [Google Scholar] [CrossRef] [PubMed]
  57. Kirino, Y.; Remmers, E.F. Genetic architectures of seropositive and seronegative rheumatic diseases. Nat. Rev. Rheumatol. 2015, 11, 401–414. [Google Scholar] [CrossRef]
  58. Qu, D.; Wang, Y.; Esmon, N.L.; Esmon, C.T. Regulated endothelial protein C receptor shedding is mediated by tumor necrosis factor-alpha converting enzyme/ADAM17. J. Thromb. Haemost. 2007, 5, 395–402. [Google Scholar] [CrossRef] [PubMed]
  59. Saposnik, B.; Lesteven, E.; Lokajczyk, A.; Esmon, C.T.; Aiach, M.; Gandrille, S. Alternative mRNA is favored by the A3 haplotype of the EPCR gene PROCR and generates a novel soluble form of EPCR in plasma. Blood 2008, 111, 3442–3451. [Google Scholar] [CrossRef]
  60. Kang, H.; Lee, T.; Bae, J.S. Suppressive Effects of Pelargonidin on Endothelial Protein C Receptor Shedding via the Inhibition of TACE Activity and MAP Kinases. Am. J. Chin. Med. 2016, 44, 771–784. [Google Scholar] [CrossRef]
  61. Ku, S.K.; Han, M.S.; Bae, J.S. Sulforaphane inhibits endothelial protein C receptor shedding in vitro and in vivo. Vascul. Pharmacol. 2014, 63, 13–18. [Google Scholar] [CrossRef] [PubMed]
  62. Ku, S.K.; Yang, E.J.; Song, K.S.; Bae, J.S. Rosmarinic acid down-regulates endothelial protein C receptor shedding in vitro and in vivo. Food Chem. Toxicol. 2013, 59, 311–315. [Google Scholar] [CrossRef] [PubMed]
  63. Ku, S.K.; Han, M.S.; Park, E.J.; Na, D.H.; Bae, J.S. Exendin-4 inhibits endothelial protein C receptor shedding in vitro and in vivo. Pharmacol. Res. 2014, 84, 18–25. [Google Scholar] [CrossRef] [PubMed]
  64. Navarro, S.; Bonet, E.; Estellés, A.; Montes, R.; Hermida, J.; Martos, L.; España, F.; Medina, P. The endothelial cell protein C receptor: Its role in thrombosis. Thromb. Res. 2011, 128, 410–416. [Google Scholar] [CrossRef] [PubMed]
  65. Liaw, P.C.; Neuenschwander, P.F.; Smirnov, M.D.; Esmon, C.T. Mechanisms by which soluble endothelial cell protein C receptor modulates protein C and activated protein C function. J. Biol. Chem. 2000, 275, 5447–5452. [Google Scholar] [CrossRef] [PubMed]
  66. Van de Wouwer, M.; Collen, D.; Conway, E.M. Thrombomodulin-protein C-EPCR system: Integrated to regulate coagulation and inflammation. Arterioscler. Thromb. Vasc. Biol. 2004, 24, 1374–1383. [Google Scholar] [CrossRef] [PubMed]
  67. Saposnik, B.; Reny, J.L.; Gaussem, P.; Emmerich, J.; Aiach, M.; Gandrille, S. A haplotype of the EPCR gene is associated with increased plasma levels of sEPCR and is a candidate risk factor for thrombosis. Blood 2004, 103, 1311–1318. [Google Scholar] [CrossRef]
  68. Chen, X.D.; Tian, L.; Li, M.; Jin, W.; Zhang, H.K.; Zheng, C.F. Relationship between endothelial cell protein C receptor gene 6936A/G polymorphisms and deep venous thrombosis. Chin. Med. J. 2011, 124, 72–75. [Google Scholar] [PubMed]
  69. Stearns-Kurosawa, D.J.; Swindle, K.; D’Angelo, A.; Della Valle, P.; Fattorini, A.; Caron, N.; Grimaux, M.; Woodhams, B.; Kurosawa, S. Plasma levels of endothelial protein C receptor respond to anticoagulant treatment. Blood 2002, 99, 526–530. [Google Scholar] [CrossRef]
  70. Hurtado, V.; Montes, R.; Gris, J.C.; Bertolaccini, M.L.; Alonso, A.; Martinez-Gonzalez, M.A.; Khamashta, M.A.; Fukudome, K.; Lane, D.A.; Hermida, J. Autoantibodies against EPCR are found in antiphospholipid syndrome and are a risk factor for fetal death. Blood 2004, 104, 1369–1374. [Google Scholar] [CrossRef]
  71. Mutoh, T.; Shirai, T.; Ishii, T.; Shirota, Y.; Fujishima, F.; Takahashi, F.; Kakuta, Y.; Kanazawa, Y.; Masamune, A.; Saiki, Y.; et al. Identification of two major autoantigens negatively regulating endothelial activation in Takayasu arteritis. Nat. Commun. 2020, 11, 1253. [Google Scholar] [CrossRef]
  72. Shirai, T.; Kakuta, Y.; Fujii, H. Distinct Autoantibodies Against Endothelial Protein C Receptor in Ulcerative Colitis. Gastroenterology 2021, 161, 1724–1725. [Google Scholar] [CrossRef]
  73. van Hylckama Vlieg, A.; Montes, R.; Rosendaal, F.R.; Hermida, J. Autoantibodies against endothelial protein C receptor and the risk of a first deep vein thrombosis. J. Thromb. Haemost. 2007, 5, 1449–1454. [Google Scholar] [CrossRef] [PubMed]
  74. Montes, R.; Hurtado, V.; Alonso, A.; Foco, L.; Zonzin, P.; Mannucci, P.M.; Hermida, J. Autoantibodies against the endothelial receptor of protein C are associated with acute myocardial infarction in young women. J. Thromb. Haemost. 2005, 3, 1454–1458. [Google Scholar] [CrossRef]
  75. Biguzzi, E.; Merati, G.; Liaw, P.C.; Bucciarelli, P.; Oganesyan, N.; Qu, D.; Gu, J.M.; Fetiveau, R.; Esmon, C.T.; Mannucci, P.M.; et al. A 23bp insertion in the endothelial protein C receptor (EPCR) gene impairs EPCR function. Thromb. Haemost. 2001, 86, 945–948. [Google Scholar] [PubMed]
  76. Medina, P.; Navarro, S.; Estelles, A.; Vaya, A.; Woodhams, B.; Mira, Y.; Villa, P.; Migaud-Fressart, M.; Ferrando, F.; Aznar, J.; et al. Contribution of polymorphisms in the endothelial protein C receptor gene to soluble endothelial protein C receptor and circulating activated protein C levels, and thrombotic risk. Thromb. Haemost. 2004, 91, 905–911. [Google Scholar] [CrossRef] [PubMed]
  77. Uitte de Willige, S.; Van Marion, V.; Rosendaal, F.R.; Vos, H.L.; de Visser, M.C.; Bertina, R.M. Haplotypes of the EPCR gene, plasma sEPCR levels and the risk of deep venous thrombosis. J. Thromb. Haemost. 2004, 2, 1305–1310. [Google Scholar] [CrossRef]
  78. Medina, P.; Navarro, S.; Bonet, E.; Martos, L.; Estellés, A.; Bertina, R.M.; Vos, H.L.; España, F. Functional analysis of two haplotypes of the human endothelial protein C receptor gene. Arterioscler. Thromb. Vasc. Biol. 2014, 34, 684–690. [Google Scholar] [CrossRef]
  79. Martos, L.; Oto, J.; Fernandez-Pardo, A.; Plana, E.; Solmoirago, M.J.; Cana, F.; Hervas, D.; Bonanad, S.; Ferrando, F.; Espana, F.; et al. Increase of Neutrophil Activation Markers in Venous Thrombosis-Contribution of Circulating Activated Protein C. Int. J. Mol. Sci. 2020, 21, 5651. [Google Scholar] [CrossRef]
  80. Qu, D.; Wang, Y.; Song, Y.; Esmon, N.L.; Esmon, C.T. The Ser219-->Gly dimorphism of the endothelial protein C receptor contributes to the higher soluble protein levels observed in individuals with the A3 haplotype. J. Thromb. Haemost. 2006, 4, 229–235. [Google Scholar] [CrossRef]
  81. Cespedes, J.C.; Hibbert, J.; Krishna, S.; Yan, F.; Bharti, P.K.; Stiles, J.K.; Liu, M. Association of EPCR Polymorphism rs867186-GG With Severity of Human Malaria. Front. Genet. 2020, 11, 56. [Google Scholar] [CrossRef] [PubMed]
  82. Yamagishi, K.; Cushman, M.; Heckbert, S.R.; Tsai, M.Y.; Folsom, A.R. Lack of association of soluble endothelial protein C receptor and PROCR 6936A/G polymorphism with the risk of venous thromboembolism in a prospective study. Br. J. Haematol. 2009, 145, 221–226. [Google Scholar] [CrossRef] [PubMed]
  83. Li, Y.Y.; Wu, J.J.; Yang, X.X.; Geng, H.Y.; Gong, G.; Kim, H.J. EPCR Gene Ser219Gly Polymorphism and Venous Thromboembolism: A Meta-Analysis of 9,494 Subjects. Front. Physiol. 2017, 8, 339. [Google Scholar] [CrossRef] [PubMed]
  84. Zoheir, N.; Eldanasouri, N.; Abdel-Aal, A.A.; Hosny, K.A.; Abdel-Ghany, W.M. Endothelial cell protein C receptor gene 6936A/G and 4678G/C polymorphisms as risk factors for deep venous thrombosis. Blood Coagul. Fibrinolysis 2016, 27, 259–265. [Google Scholar] [CrossRef] [PubMed]
  85. Navarro, S.; Medina, P.; Mira, Y.; Estelles, A.; Villa, P.; Ferrando, F.; Vaya, A.; Bertina, R.M.; Espana, F. Haplotypes of the EPCR gene, prothrombin levels, and the risk of venous thrombosis in carriers of the prothrombin G20210A mutation. Haematologica 2008, 93, 885–891. [Google Scholar] [CrossRef]
  86. Cochery-Nouvellon, E.; Chauleur, C.; Demattei, C.; Mercier, E.; Fabbro-Peray, P.; Mares, P.; Mismetti, P.; Lissalde-Lavigne, G.; Gris, J.C. The A6936G polymorphism of the endothelial protein C receptor gene is associated with the risk of unexplained foetal loss in Mediterranean European couples. Thromb. Haemost. 2009, 102, 656–667. [Google Scholar] [PubMed]
  87. Lavigne-Lissalde, G.; Cochery-Nouvellon, E.; Mercier, E.; Marès, P.; Gris, J.C. High plasma levels of endothelial protein C receptor are associated with the risk of unexplained fetal death. J. Thromb. Haemost. 2005, 3, 393–395. [Google Scholar] [CrossRef] [PubMed]
  88. Tang, W.; Basu, S.; Kong, X.; Pankow, J.S.; Aleksic, N.; Tan, A.; Cushman, M.; Boerwinkle, E.; Folsom, A.R. Genome-wide association study identifies novel loci for plasma levels of protein C: The ARIC study. Blood 2010, 116, 5032–5036. [Google Scholar] [CrossRef] [PubMed]
  89. Reiner, A.P.; Carty, C.L.; Jenny, N.S.; Nievergelt, C.; Cushman, M.; Stearns-Kurosawa, D.J.; Kurosawa, S.; Kuller, L.H.; Lange, L.A. PROC, PROCR and PROS1 polymorphisms, plasma anticoagulant phenotypes, and risk of cardiovascular disease and mortality in older adults: The Cardiovascular Health Study. J. Thromb. Haemost. 2008, 6, 1625–1632. [Google Scholar] [CrossRef]
  90. Smith, N.L.; Chen, M.H.; Dehghan, A.; Strachan, D.P.; Basu, S.; Soranzo, N.; Hayward, C.; Rudan, I.; Sabater-Lleal, M.; Bis, J.C.; et al. Novel associations of multiple genetic loci with plasma levels of factor VII, factor VIII, and von Willebrand factor: The CHARGE (Cohorts for Heart and Aging Research in Genome Epidemiology) Consortium. Circulation 2010, 121, 1382–1392. [Google Scholar] [CrossRef]
  91. Zunke, F.; Rose-John, S. The shedding protease ADAM17: Physiology and pathophysiology. Biochim. Biophys. Acta Mol. Cell Res. 2017, 1864, 2059–2070. [Google Scholar] [CrossRef] [PubMed]
  92. Xu, J. Endothelial cell protein C receptor (EPCR) is constitutively translocated into the nucleus and also mediates activated protein C, but not protein C, nuclear translocation. Thromb. Haemost. 1999, 114, 652a. [Google Scholar]
  93. Gavlovsky, P.J.; Tonnerre, P.; Guitton, C.; Charreau, B. Expression of MHC class I-related molecules MICA, HLA-E and EPCR shape endothelial cells with unique functions in innate and adaptive immunity. Hum. Immunol. 2016, 77, 1084–1091. [Google Scholar] [CrossRef] [PubMed]
  94. Dahlback, B. Advances in understanding pathogenic mechanisms of thrombophilic disorders. Blood 2008, 112, 19–27. [Google Scholar] [CrossRef] [PubMed]
  95. Dennis, J.; Johnson, C.Y.; Adediran, A.S.; de Andrade, M.; Heit, J.A.; Morange, P.E.; Tregouet, D.A.; Gagnon, F. The endothelial protein C receptor (PROCR) Ser219Gly variant and risk of common thrombotic disorders: A HuGE review and meta-analysis of evidence from observational studies. Blood 2012, 119, 2392–2400. [Google Scholar] [CrossRef] [PubMed]
  96. Stearns-Kurosawa, D.J.; Kurosawa, S.; Mollica, J.S.; Ferrell, G.L.; Esmon, C.T. The endothelial cell protein C receptor augments protein C activation by the thrombin-thrombomodulin complex. PNAS 1996, 93, 10212–10216. [Google Scholar] [CrossRef] [PubMed]
  97. Gu, J.M.; Fukudome, K.; Esmon, C.T. Characterization and regulation of the 5’-flanking region of the murine endothelial protein C receptor gene. J. Biol. Chem. 2000, 275, 12481–12488. [Google Scholar] [CrossRef] [PubMed]
  98. Keshava, S.; Sundaram, J.; Rajulapati, A.; Esmon, C.; Pendurthi, U.; Rao, L.V.M. Factor VIIa interaction with EPCR modulates the hemostatic effect of rFVIIa in hemophilia therapy: Mode of its action. Blood Adv. 2017, 1, 1206–1214. [Google Scholar] [CrossRef]
  99. Bouwens, E.A.; Stavenuiter, F.; Mosnier, L.O. Mechanisms of anticoagulant and cytoprotective actions of the protein C pathway. J. Thromb. Haemost. 2013, 11 (Suppl. S1), 242–253. [Google Scholar] [CrossRef]
  100. Coughlin, S.R. Protease-activated receptors in hemostasis, thrombosis and vascular biology. J. Thromb. Haemost. 2005, 3, 1800–1814. [Google Scholar] [CrossRef]
  101. Riewald, M.; Petrovan, R.J.; Donner, A.; Mueller, B.M.; Ruf, W. Activation of endothelial cell protease activated receptor 1 by the protein C pathway. Science 2002, 296, 1880–1882. [Google Scholar] [CrossRef] [PubMed]
  102. Roy, R.V.; Ardeshirylajimi, A.; Dinarvand, P.; Yang, L.; Rezaie, A.R. Occupancy of human EPCR by protein C induces beta-arrestin-2 biased PAR1 signaling by both APC and thrombin. Blood 2016, 128, 1884–1893. [Google Scholar] [CrossRef]
  103. Dahlback, B.; Villoutreix, B.O. Regulation of blood coagulation by the protein C anticoagulant pathway: Novel insights into structure-function relationships and molecular recognition. Arterioscler. Thromb. Vasc. Biol. 2005, 25, 1311–1320. [Google Scholar] [CrossRef] [PubMed]
  104. Mosnier, L.O.; Zlokovic, B.V.; Griffin, J.H. The cytoprotective protein C pathway. Blood 2007, 109, 3161–3172. [Google Scholar] [CrossRef]
  105. Griffin, J.H.; Zlokovic, B.V.; Mosnier, L.O. Protein C anticoagulant and cytoprotective pathways. Int. J. Hematol. 2012, 95, 333–345. [Google Scholar] [CrossRef]
  106. Burnier, L.; Mosnier, L.O. Novel mechanisms for activated protein C cytoprotective activities involving noncanonical activation of protease-activated receptor 3. Blood 2013, 122, 807–816. [Google Scholar] [CrossRef] [PubMed]
  107. Healy, L.D.; Puy, C.; Fernandez, J.A.; Mitrugno, A.; Keshari, R.S.; Taku, N.A.; Chu, T.T.; Xu, X.; Gruber, A.; Lupu, F.; et al. Activated protein C inhibits neutrophil extracellular trap formation in vitro and activation in vivo. J. Biol. Chem. 2017, 292, 8616–8629. [Google Scholar] [CrossRef]
  108. Xue, M.; Dervish, S.; Harrison, L.C.; Fulcher, G.; Jackson, C.J. Activated protein C inhibits pancreatic islet inflammation, stimulates T regulatory cells and prevents diabetes in NOD mice. J. Biol. Chem. 2012, 287, 16356–16364. [Google Scholar] [CrossRef] [PubMed]
  109. Xue, M.; Dervish, S.; McKelvey, K.J.; March, L.; Wang, F.; Little, C.B.; Jackson, C.J. Activated protein C targets immune cells and rheumatoid synovial fibroblasts to prevent inflammatory arthritis in mice. Rheumatology 2019, 58, 1850–1860. [Google Scholar] [CrossRef]
  110. Lichtnekert, J.; Rupanagudi, K.V.; Kulkarni, O.P.; Darisipudi, M.N.; Allam, R.; Anders, H.J. Activated protein C attenuates systemic lupus erythematosus and lupus nephritis in MRL-Fas(lpr) mice. J. Immunol. 2011, 187, 3413–3421. [Google Scholar] [CrossRef]
  111. Lopez-Sagaseta, J.; Montes, R.; Puy, C.; Diez, N.; Fukudome, K.; Hermida, J. Binding of factor VIIa to the endothelial cell protein C receptor reduces its coagulant activity. J. Thromb. Haemost. 2007, 5, 1817–1824. [Google Scholar] [CrossRef]
  112. Bae, J.S.; Yang, L.; Manithody, C.; Rezaie, A.R. The ligand occupancy of endothelial protein C receptor switches the protease-activated receptor 1-dependent signaling specificity of thrombin from a permeability-enhancing to a barrier-protective response in endothelial cells. Blood 2007, 110, 3909–3916. [Google Scholar] [CrossRef] [PubMed]
  113. Kondreddy, V.; Wang, J.; Keshava, S.; Esmon, C.T.; Rao, L.V.M.; Pendurthi, U.R. Factor VIIa induces anti-inflammatory signaling via EPCR and PAR1. Blood 2018, 131, 2379–2392. [Google Scholar] [CrossRef]
  114. Sen, P.; Gopalakrishnan, R.; Kothari, H.; Keshava, S.; Clark, C.A.; Esmon, C.T.; Pendurthi, U.R.; Rao, L.V. Factor VIIa bound to endothelial cell protein C receptor activates protease activated receptor-1 and mediates cell signaling and barrier protection. Blood 2011, 117, 3199–3208. [Google Scholar] [CrossRef]
  115. Schuepbach, R.A.; Riewald, M. Coagulation factor Xa cleaves protease-activated receptor-1 and mediates signaling dependent on binding to the endothelial protein C receptor. J. Thromb. Haemost. 2010, 8, 379–388. [Google Scholar] [CrossRef]
  116. Disse, J.; Petersen, H.H.; Larsen, K.S.; Persson, E.; Esmon, N.; Esmon, C.T.; Teyton, L.; Petersen, L.C.; Ruf, W. The endothelial protein C receptor supports tissue factor ternary coagulation initiation complex signaling through protease-activated receptors. J. Biol. Chem. 2011, 286, 5756–5767. [Google Scholar] [CrossRef] [PubMed]
  117. Liang, H.P.; Kerschen, E.J.; Hernandez, I.; Basu, S.; Zogg, M.; Botros, F.; Jia, S.; Hessner, M.J.; Griffin, J.H.; Ruf, W.; et al. EPCR-dependent PAR2 activation by the blood coagulation initiation complex regulates LPS-triggered interferon responses in mice. Blood 2015, 125, 2845–2854. [Google Scholar] [CrossRef] [PubMed]
  118. Yuan, Q.; Zhang, D.; Wu, S.; Yu, J.; Yu, L.; Sun, Y.; Du, Z.; Li, Z.; Zhou, L.; Wu, X.; et al. FVIIa prevents the progressive hemorrhaging of a brain contusion by protecting microvessels via formation of the TF-FVIIa-FXa complex. Neuroscience 2017, 348, 114–125. [Google Scholar] [CrossRef]
  119. Stavenuiter, F.; Mosnier, L.O. Noncanonical PAR3 activation by factor Xa identifies a novel pathway for Tie2 activation and stabilization of vascular integrity. Blood 2014, 124, 3480–3489. [Google Scholar] [CrossRef]
  120. Chien, Y.H.; Meyer, C.; Bonneville, M. γδ T cells: First line of defense and beyond. Ann. Rev. Immunol. 2014, 32, 121–155. [Google Scholar] [CrossRef]
  121. Mo, W.X.; Yin, S.S.; Chen, H.; Zhou, C.; Zhou, J.X.; Zhao, L.D.; Fei, Y.Y.; Yang, H.X.; Guo, J.B.; Mao, Y.J.; et al. Chemotaxis of Vdelta2 T cells to the joints contributes to the pathogenesis of rheumatoid arthritis. Ann. Rheum. Dis. 2017, 76, 2075–2084. [Google Scholar] [CrossRef] [PubMed]
  122. Willcox, C.R.; Pitard, V.; Netzer, S.; Couzi, L.; Salim, M.; Silberzahn, T.; Moreau, J.F.; Hayday, A.C.; Willcox, B.E.; Dechanet-Merville, J. Cytomegalovirus and tumor stress surveillance by binding of a human gammadelta T cell antigen receptor to endothelial protein C receptor. Nat. Immunol. 2012, 13, 872–879. [Google Scholar] [CrossRef] [PubMed]
  123. Witherden, D.A.; Havran, W.L. EPCR: A stress trigger for gammadelta T cells. Nat. Immunol. 2012, 13, 812–814. [Google Scholar] [CrossRef] [PubMed]
  124. Turner, L.; Lavstsen, T.; Berger, S.S.; Wang, C.W.; Petersen, J.E.; Avril, M.; Brazier, A.J.; Freeth, J.; Jespersen, J.S.; Nielsen, M.A.; et al. Severe malaria is associated with parasite binding to endothelial protein C receptor. Nature 2013, 498, 502–505. [Google Scholar] [CrossRef] [PubMed]
  125. Shabani, E.; Hanisch, B.; Opoka, R.O.; Lavstsen, T.; John, C.C. Plasmodium falciparum EPCR-binding PfEMP1 expression increases with malaria disease severity and is elevated in retinopathy negative cerebral malaria. BMC Med. 2017, 15, 183. [Google Scholar] [CrossRef] [PubMed]
  126. Ait-Oufella, H.; Mallat, Z.; Tedgui, A. Lp-PLA2 and sPLA2: Cardiovascular biomarkers. Med. Sci. 2014, 30, 526–531. [Google Scholar]
  127. Lopez-Sagaseta, J.; Puy, C.; Tamayo, I.; Allende, M.; Cervero, J.; Velasco, S.E.; Esmon, C.T.; Montes, R.; Hermida, J. sPLA2-V inhibits EPCR anticoagulant and antiapoptotic properties by accommodating lysophosphatidylcholine or PAF in the hydrophobic groove. Blood 2012, 119, 2914–2921. [Google Scholar] [CrossRef]
  128. Tamayo, I.; Velasco, S.E.; Puy, C.; Esmon, C.T.; Dichiara, M.G.; Montes, R.; Hermida, J. Group V secretory phospholipase A2 impairs endothelial protein C receptor-dependent protein C activation and accelerates thrombosis in vivo. J. Thromb. Haemost. 2014, 12, 1921–1927. [Google Scholar] [CrossRef] [PubMed]
  129. Xue, M.; Shen, K.; McKelvey, K.; Li, J.; Chan, Y.K.; Hatzis, V.; March, L.; Little, C.B.; Tonkin, M.; Jackson, C.J. Endothelial protein C receptor associated invasiveness of rheumatoid synovial fibroblasts is likely driven by group V secretory phospholipase A2. Arthritis. Res. Ther. 2014, 16, R44. [Google Scholar] [CrossRef]
  130. Korkmaz, B.; Kuhl, A.; Bayat, B.; Santoso, S.; Jenne, D.E. A hydrophobic patch on proteinase 3, the target of autoantibodies in Wegener granulomatosis, mediates membrane binding via NB1 receptors. J. Biol. Chem. 2008, 283, 35976–35982. [Google Scholar] [CrossRef]
  131. Preston, G.A.; Zarella, C.S.; Pendergraft, W.F., 3rd; Rudolph, E.H.; Yang, J.J.; Sekura, S.B.; Jennette, J.C.; Falk, R.J. Novel effects of neutrophil-derived proteinase 3 and elastase on the vascular endothelium involve in vivo cleavage of NF-kappaB and proapoptotic changes in JNK, ERK, and p38 MAPK signaling pathways. J. Am. Soc. Nephrol. 2002, 13, 2840–2849. [Google Scholar] [CrossRef] [PubMed]
  132. Bruner, B.F.; Vista, E.S.; Wynn, D.M.; Harley, J.B.; James, J.A. Anti-neutrophil cytoplasmic antibodies target sequential functional proteinase 3 epitopes in the sera of patients with Wegener’s granulomatosis. Clin. Exp. Immunol. 2010, 162, 262–270. [Google Scholar] [CrossRef]
  133. Mohan Rao, L.V.; Esmon, C.T.; Pendurthi, U.R. Endothelial cell protein C receptor: A multiliganded and multifunctional receptor. Blood 2014, 124, 1553–1562. [Google Scholar] [CrossRef] [PubMed]
  134. Kurosawa, S.; Esmon, C.T.; Stearns-Kurosawa, D.J. The soluble endothelial protein C receptor binds to activated neutrophils: Involvement of proteinase-3 and CD11b/CD18. J. Immunol. 2000, 165, 4697–4703. [Google Scholar] [CrossRef] [PubMed]
  135. Villegas-Mendez, A.; Montes, R.; Ambrose, L.R.; Warrens, A.N.; Laffan, M.; Lane, D.A. Proteolysis of the endothelial cell protein C receptor by neutrophil proteinase 3. J. Thromb. Haemost. 2007, 5, 980–988. [Google Scholar] [CrossRef] [PubMed]
  136. Fink, K.; Busch, H.J.; Bourgeois, N.; Schwarz, M.; Wolf, D.; Zirlik, A.; Peter, K.; Bode, C.; von Zur Muhlen, C. Mac-1 directly binds to the endothelial protein C-receptor: A link between the protein C anticoagulant pathway and inflammation? PLoS ONE 2013, 8, e53103. [Google Scholar] [CrossRef] [PubMed]
  137. Chu, C.Q. The pivotal role of endothelial protein C receptor for antiphospholipid antibody-mediated pathologies. Rheumatology 2022, 61, 883–885. [Google Scholar] [CrossRef] [PubMed]
  138. Conforti, A.; Di Cola, I.; Pavlych, V.; Ruscitti, P.; Berardicurti, O.; Ursini, F.; Giacomelli, R.; Cipriani, P. Beyond the joints, the extra-articular manifestations in rheumatoid arthritis. Autoimmun. Rev. 2021, 20, 102735. [Google Scholar] [CrossRef]
  139. Bartok, B.; Firestein, G.S. Fibroblast-like synoviocytes: Key effector cells in rheumatoid arthritis. Immunol. Rev. 2010, 233, 233–255. [Google Scholar] [CrossRef]
  140. O’Neil, L.J.; Kaplan, M.J. Neutrophils in Rheumatoid Arthritis: Breaking Immune Tolerance and Fueling Disease. Trends. Mol. Med. 2019, 25, 215–227. [Google Scholar] [CrossRef]
  141. Rostami, S.; Hoff, M.; Brown, M.A.; Hveem, K.; Videm, V. Comparison of methods to construct a genetic risk score for prediction of rheumatoid arthritis in the population-based Nord-Trondelag Health Study, Norway. Rheumatology 2020, 59, 1743–1751. [Google Scholar] [CrossRef]
  142. Smolen, J.S.; Aletaha, D.; Koeller, M.; Weisman, M.H.; Emery, P. New therapies for treatment of rheumatoid arthritis. Lancet 2007, 370, 1861–1874. [Google Scholar] [CrossRef]
  143. Jang, S.; Kwon, E.J.; Lee, J.J. Rheumatoid Arthritis: Pathogenic Roles of Diverse Immune Cells. Int. J. Mol. Sci. 2022, 23, 905. [Google Scholar] [CrossRef]
  144. Wouters, F.; Maurits, M.P.; van Boheemen, L.; Verstappen, M.; Mankia, K.; Matthijssen, X.M.E.; Dorjee, A.L.; Emery, P.; Knevel, R.; van Schaardenburg, D.; et al. Determining in which pre-arthritis stage HLA-shared epitope alleles and smoking exert their effect on the development of rheumatoid arthritis. Ann. Rheum. Dis. 2022, 81, 48–55. [Google Scholar] [CrossRef] [PubMed]
  145. Xue, M.; March, L.; Sambrook, P.N.; Jackson, C.J. Differential regulation of matrix metalloproteinase 2 and matrix metalloproteinase 9 by activated protein C: Relevance to inflammation in rheumatoid arthritis. Arthritis. Rheum. 2007, 56, 2864–2874. [Google Scholar] [CrossRef] [PubMed]
  146. Bai, L.; Liu, W.; Guo, P.; Bai, J.; Liu, Y.; Hua, Y.; Pang, C.; Zhang, W.; Yin, F.; Wang, Y. Elevated levels of soluble Endothelial protein C receptor in rheumatoid arthritis and block the therapeutic effect of protein C in collagen-induced arthritis. Int. Immunopharmacol. 2020, 81, 106255. [Google Scholar] [CrossRef] [PubMed]
  147. Xue, M.; Lin, H.; Liang, H.; Bereza-Malcolm, L.; Lynch, T.; Sinnathurai, P.; Weiler, H.; Jackson, C.; March, L. EPCR deficiency ameliorates inflammatory arthritis in mice via suppressing the activation and migration of T cells and dendritic cells. Rheumatology 2023, 63, 571–580. [Google Scholar] [CrossRef]
  148. Liu, M.F.; Yang, C.Y.; Chao, S.C.; Li, J.S.; Weng, T.H.; Lei, H.Y. Distribution of double-negative (CD4- CD8-, DN) T subsets in blood and synovial fluid from patients with rheumatoid arthritis. Clin. Rheumatol. 1999, 18, 227–231. [Google Scholar] [CrossRef]
  149. Andreu, J.L.; Trujillo, A.; Alonso, J.M.; Mulero, J.; Martinez, C. Selective expansion of T cells bearing the gamma/delta receptor and expressing an unusual repertoire in the synovial membrane of patients with rheumatoid arthritis. Arthritis. Rheum. 1991, 34, 808–814. [Google Scholar] [CrossRef]
  150. Liu, X.; Huo, Y.; Zhao, J.; Wang, G.; Liu, H.; Yin, F.; Pang, C.; Wang, Y.; Bai, L. Endothelial cell protein C receptor regulates neutrophil extracellular trap-mediated rheumatoid arthritis disease progression. Int. Immunopharmacol. 2022, 112, 109249. [Google Scholar] [CrossRef]
  151. Wu, H.; Chang, C.; Lu, Q. The Epigenetics of Lupus Erythematosus. Adv. Exp. Med. Biol. 2020, 1253, 185–207. [Google Scholar] [PubMed]
  152. Koarada, S.; Tsuneyoshi, N.; Haruta, Y.; Tada, Y.; Mitamura, M.; Inoue, H.; Ohta, A.; Fukudome, K.; Nagasawa, K. Effect of disease activity and corticosteroids on serum levels of soluble endothelial cell protein C receptor in patients with systemic lupus erythematosus. Mod. Rheumatol. 2009, 19, 173–179. [Google Scholar] [CrossRef] [PubMed]
  153. Kurosawa, S.; Stearns-Kurosawa, D.J.; Carson, C.W.; D’Angelo, A.; Della Valle, P.; Esmon, C.T. Plasma levels of endothelial cell protein C receptor are elevated in patients with sepsis and systemic lupus erythematosus: Lack of correlation with thrombomodulin suggests involvement of different pathological processes. Blood 1998, 91, 725–727. [Google Scholar] [CrossRef] [PubMed]
  154. Wu, J.; Zhou, Z.; Ye, S.; Dai, H.; Ma, L.; Xu, X.; Li, X. Detection of soluble endothelial protein C receptor (sEPCR) in patients with CHD, DM and SLE. Zhonghua Xue Ye Xue Za Zhi 2000, 21, 472–474. [Google Scholar] [PubMed]
  155. Leibovich, S.J. Vasculopathy and renal injury in lupus erythematosus: Does shedding of the endothelial protein C receptor play a role? Kidney Int. 2005, 68, 407–408. [Google Scholar] [CrossRef] [PubMed]
  156. Sesin, C.A.; Yin, X.; Esmon, C.T.; Buyon, J.P.; Clancy, R.M. Shedding of endothelial protein C receptor contributes to vasculopathy and renal injury in lupus: In vivo and in vitro evidence. Kidney Int. 2005, 68, 110–120. [Google Scholar] [CrossRef] [PubMed]
  157. Asmaa, A.; Nadia, A.; Hebatallah, A.; Rana, A.; Nadia, G.; Nesrine, A. Soluble and membranous endothelial protein C receptor in systemic lupus erythematosus patients: Relation to nephritis. Egypt. Rheumatol. 2019, 41, 6. [Google Scholar]
  158. Saigusa, R.; Asano, Y.; Yamashita, T.; Taniguchi, T.; Takahashi, T.; Ichimura, Y.; Toyama, T.; Yoshizaki, A.; Miyagaki, T.; Sugaya, M.; et al. Fli1 deficiency contributes to the downregulation of endothelial protein C receptor in systemic sclerosis: A possible role in prothrombotic conditions. Br. J. Dermatol. 2016, 174, 338–347. [Google Scholar] [CrossRef]
  159. Tseng, C.C.; Chang, S.J.; Tsai, W.C.; Ou, T.T.; Wu, C.C.; Sung, W.Y.; Hsieh, M.C.; Yen, J.H. Increased incidence of rheumatoid arthritis in multiple sclerosis: A nationwide cohort study. Medicine 2016, 95, e3999. [Google Scholar] [CrossRef]
  160. Festoff, B.W.; Li, C.; Woodhams, B.; Lynch, S. Soluble thrombomodulin levels in plasma of multiple sclerosis patients and their implication. J. Neurol. Sci. 2012, 323, 61–65. [Google Scholar] [CrossRef]
Figure 1. The structures of EPCR vs. CD1. The EPCR molecule forms a deep hydrophobic groove between two anti-parallel α-helices, the α1 and α2 domains, sitting atop a β-pleated sheet that houses the phospholipid phosphatidylcholine. This lipid is bound to EPCR and assists in its binding to PC by maintaining the structure of EPCR. The ligands of EPCR do not bind inside the hydrophobic groove [7]. The group of CD1 proteins possesses a similar structure to EPCR; however, they possess an α3 domain that associates with β2-microglobulin. The deep grooves formed from these structures are used for ligand binding in the CD1 molecules and can vary between each of the CD1 molecules depending on the structure of the groove [8].
Figure 1. The structures of EPCR vs. CD1. The EPCR molecule forms a deep hydrophobic groove between two anti-parallel α-helices, the α1 and α2 domains, sitting atop a β-pleated sheet that houses the phospholipid phosphatidylcholine. This lipid is bound to EPCR and assists in its binding to PC by maintaining the structure of EPCR. The ligands of EPCR do not bind inside the hydrophobic groove [7]. The group of CD1 proteins possesses a similar structure to EPCR; however, they possess an α3 domain that associates with β2-microglobulin. The deep grooves formed from these structures are used for ligand binding in the CD1 molecules and can vary between each of the CD1 molecules depending on the structure of the groove [8].
Jcm 13 02030 g001
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

O’Hehir, Z.D.; Lynch, T.; O’Neill, S.; March, L.; Xue, M. Endothelial Protein C Receptor and Its Impact on Rheumatic Disease. J. Clin. Med. 2024, 13, 2030. https://doi.org/10.3390/jcm13072030

AMA Style

O’Hehir ZD, Lynch T, O’Neill S, March L, Xue M. Endothelial Protein C Receptor and Its Impact on Rheumatic Disease. Journal of Clinical Medicine. 2024; 13(7):2030. https://doi.org/10.3390/jcm13072030

Chicago/Turabian Style

O’Hehir, Zachary Daniel, Tom Lynch, Sean O’Neill, Lyn March, and Meilang Xue. 2024. "Endothelial Protein C Receptor and Its Impact on Rheumatic Disease" Journal of Clinical Medicine 13, no. 7: 2030. https://doi.org/10.3390/jcm13072030

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop