Skip to main content
American Journal of Physiology - Cell Physiology logoLink to American Journal of Physiology - Cell Physiology
. 2021 Aug 18;321(4):C721–C734. doi: 10.1152/ajpcell.00269.2021

Plasminogen activator receptor assemblies in cell signaling, innate immunity, and inflammation

Steven L Gonias 1,
PMCID: PMC8560384  PMID: 34406905

Abstract

Tissue-type plasminogen activator (tPA) and urokinase-type plasminogen activator (uPA) are serine proteases and major activators of fibrinolysis in mammalian systems. Because fibrinolysis is an essential component of the response to tissue injury, diverse cells, including cells that participate in the response to injury, have evolved receptor systems to detect tPA and uPA and initiate appropriate cell-signaling responses. Formation of functional receptor systems for the plasminogen activators requires assembly of diverse plasma membrane proteins, including but not limited to: the urokinase receptor (uPAR); integrins; N-formyl peptide receptor-2 (FPR2), receptor tyrosine kinases (RTKs), the N-methyl-d-aspartate receptor (NMDA-R), and low-density lipoprotein receptor-related protein-1 (LRP1). The cell-signaling responses elicited by tPA and uPA impact diverse aspects of cell physiology. This review describes rapidly evolving knowledge regarding the structure and function of plasminogen activator receptor assemblies. How these receptor assemblies regulate innate immunity and inflammation is then considered.

Keywords: inflammation, innate immunity, tissue-type plasminogen activator, uPAR, urokinase-type plasminogen activator

INTRODUCTION

The response to tissue injury is an important process in evolution, impacting the ability of organisms to survive and reproduce. To understand wound repair at the cellular and molecular levels, this process is frequently divided into four temporally overlapping phases, including: 1) hemostasis; 2) inflammation; 3) the proliferative phase; and 4) tissue remodeling (13). The purpose of hemostasis is to terminate blood loss by the eventual formation of a provisional fibrin-based matrix. Hemostasis begins immediately after wound formation and is followed by the inflammatory phase, during which microorganisms are cleared from the injury site together with foreign materials and damaged tissue. This process is initially accomplished by neutrophils and subsequently, by macrophages and other inflammatory cells. The proliferative phase encompasses a variety of processes that lead to tissue regeneration and/or scarring. Fibroblasts play a central role in the proliferative phase, depositing extracellular matrix (ECM) proteins that form granulation tissue and eventually, fibrillar collagen. Other cellular elements also are important, including endothelial cells, which initiate angiogenesis and revascularization, and epithelial cells, which grow into injured tissue from wound edges. Finally, during the remodeling phase, collagen matrices mature through reorganization and cross-linking, accompanied by apoptosis of various cellular elements to form mature regenerated tissue and/or scar tissue.

Like wound healing, hemostasis may be divided into three-to-four overlapping phases, which have been discussed in depth in excellent reviews (49). The earliest event that occurs in hemostasis, when the injury affects blood vessels with smooth muscle cells, is vasoconstriction, which results in blood vessel narrowing. Vasoconstriction limits but does not stop blood loss. Primary hemostasis is achieved by platelets, small cellular fragments that adhere to injured tissue surfaces, aggregate with one another, and then release factors that support subsequent steps in hemostasis and the response to tissue injury. The next major system activated is coagulation, in which cascades of protease activation events result in thrombin generation. Thrombin cleaves fibrinopeptides A and B from the N-termini of the fibrinogen α and β chains to drive fibrin polymerization. Although primary hemostasis terminates blood loss, at least temporarily, coagulation is necessary to stabilize the platelet plug. In the final stage of hemostasis, fibrin clots are dissolved and removed by the fibrinolysis system to allow optimal tissue regeneration during the proliferative phase.

Cell surfaces regulate various steps of hemostasis and wound repair. This is particularly relevant to endothelial cells in hemostasis (10, 11). To assure that blood clots do not propagate unnecessarily at a distance from a blood vessel injury, uninjured endothelial cells display cell-surface macromolecules with anticoagulant activity. For example, the thrombomodulin/endothelial protein C receptor system facilitates thrombin-mediated activation of protein C, which proteolytically inactivates factors Va and VIIIa (12, 13). Uninjured endothelial cells also express heparan sulfate proteoglycans, which bind and activate the anticoagulant Serpin, antithrombin III (11, 14).

Approximate relationships between the four phases of wound repair and three phases of hemostasis are shown in Fig. 1. Overlap between the inflammatory phase of wound repair and the time when fibrinolysis is active is evident. It is thus not surprising that pathways have evolved whereby factors in the fibrinolysis system regulate innate immunity and inflammation. This review begins by briefly describing the fibrinolysis system as it functions independently of cells. Cell-surface receptors that bind plasminogen and amplify fibrinolysis are then described. Finally, two multicomponent receptor systems that recognize and bind the major mammalian plasminogen activators, urokinase-type plasminogen activator (uPA) and tissue-type plasminogen activator (tPA), are discussed. Plasminogen activator receptor assemblies are robust regulators of cell signaling. Rapidly evolving knowledge regarding the activity of these receptor assemblies in innate immunity and inflammation is considered.

Figure 1.

Figure 1.

Overlap between the four phases of the response to injury (wounding) (color coded in green) and the three major phases of hemostasis (color coded in blue).

THE FIBRINOLYSIS SYSTEM

The serine protease that is principally responsible for fibrin degradation in mammalian systems is plasmin, which circulates as a 92-kDa zymogen called plasminogen (7, 15, 16). Cleavage of a single Arg-Val peptide bond in the structure of plasminogen converts it into its active, two chain form (plasmin). In mammalian systems, plasminogen activation is catalyzed by uPA and tPA. tPA is expressed by endothelial cells and smooth muscle cells in normal blood vessels whereas uPA is expressed only by macrophages in blood vessels in which atherosclerotic lesions have formed (17). It is, thus, not surprising that fibrinolysis within the vascular system is typically attributed mainly to tPA. However, at least in mice, when the gene encoding tPA (Plat) is deleted, uPA prevents massive, multiorgan fibrin deposition (18). Thus, both plasminogen activators may contribute to intravascular fibrinolysis.

Once plasmin is generated, it degrades fibrin and fibrinogen in a predictable manner (19, 20), generating soluble products of defined size and sequence. The products of fibrinolysis and fibrinogenolysis differ mainly due to covalent cross-linking of fibrin by Factor XIIIa/transglutaminase. Fibrinolysis in vivo is detected clinically by measuring the degradation product, D-dimer, which forms only when fibrin polymerizes and is cross-linked by Factor XIIIa, before becoming a substrate for plasmin (21).

uPA and tPA are similar to plasmin(ogen) in that these proteins exist in single-chain and two-chain forms (16, 2224). The single-chain form of uPA demonstrates only minimal activity as a serine protease until it is converted into the two-chain form (24). By contrast, both single-chain and two-chain tPA are active as serine proteases and plasminogen activators (22, 23). Although a number of proteases may convert uPA and tPA into the two-chain forms, plasmin is paramount among these enzymes (23, 24). Thus, plasminogen activation may be viewed as a bi-directional reaction in which plasmin(ogen) and its activators function as enzymes and substrates.

When proteases in the fibrinolysis system circulate freely in the plasma, they are targeted by protease inhibitors in the Serpin gene family, including α2-antiplasmin (α2AP), which inhibits free plasmin with an initial association rate constant >1.0 × 107 M−1·s−1 (25), and plasminogen activator inhibitor-1 (PAI-1), which inhibits both tPA and uPA, also with initial association rate constants exceeding 1.0 × 107 M−1·s−1 (26, 27). The activity of α2AP as a plasmin inhibitor is supported by α2-macroglobulin (α2M), which reacts slower with plasmin but is highly abundant in the plasma and thus, relevant as a regulator of fibrinolysis (2830).

A number of mechanisms exist to limit fibrinolysis to mature clots and protect circulating fibrinogen so that the ability of the organism to respond to future injuries is not compromised. First, activation of plasminogen by tPA is greatly accelerated by fibrin, compared with the solution phase reaction (31, 32). Furthermore, once plasmin is associated with fibrin, it is relatively protected from its major inhibitors, α2AP and α2M (33, 34). These interactions are facilitated by multiple domains within the structure of plasmin and the plasminogen activators (Fig. 2). Binding of plasminogen to fibrin requires its tandem kringle domains, which contain binding sites for lysine and lysine-like compounds, including, most importantly, C-terminal lysine residues in fibrin and other proteins (16, 35, 36). tPA also binds to fibrin via its kringle-2 and fibronectin type-1/finger domains (37, 38). Thus, fibrin serves as a template, assembling enzyme (tPA), and substrate (plasminogen) so that the catalytic efficiency of plasminogen activation is increased. Importantly, plasmin is a lysine-specific serine protease (16). Thus, as fibrin is degraded, additional C-terminal lysine residues are exposed, allowing for additional plasminogen binding and acceleration of fibrinolysis (39).

Figure 2.

Figure 2.

The multidomain structure of the fibrinolysis proteases, plasminogen (Plg), tissue-type plasminogen activator (tPA), and urokinase-type plasminogen activator (uPA) is shown. The structure of Plg is remarkable for five tandem kringle (K) domains. Plg is activated to form plasmin (Pm) by tPA and uPA (cleavage site shown). The 77 amino acid pre-activation peptide (PAP) is removed from Plg by plasmin cleavage, converting Glu-Plg into Lys-Plg. This reaction does not activate Plg but accelerates the kinetics of Plg activation by the plasminogen activators under certain circumstances. In addition to two kringle domains, the structure of tPA includes a fibronectin type-1/finger domain (FD) and an epidermal growth factor receptor (EGF)-like/growth factor domain (GFD). In uPA, the EGF-like/growth factor domain (GFD) and kringle domain are collectively referred to as the amino-terminal fragment, which is responsible for binding to urokinase receptor (uPAR). Both uPA and tPA may be cleaved by plasmin. These reactions convert the plasminogen activators into their two-chain forms.

Fibrin is less effective in promoting the activity of uPA as a plasminogen activator, compared with tPA (40, 41). In uPA-mediated plasminogen activation, the most important effect of fibrin may be to bind the full-length form of plasminogen (referred to as Glu-plasminogen) and convert it into a more open and readily activated conformation. Glu-plasminogen is cleaved by plasmin between Lys77 and Lys78, eliminating what was originally referred to as the “pre-activation peptide” and generating a form of the zymogen (Lys78-plasminogen) with a more open conformation (16, 42, 43). Lys78-plasminogen is a preferred substrate for the plasminogen activators in the absence of fibrin.

An exosite interaction involving the lysine binding sites in plasmin is very important for the rapid and irreversible inhibition of free plasmin by α2AP (25, 33). The fact that these lysine binding sites are occupied when plasmin associates with fibrin explains the substantial decrease in the rate of plasmin inhibition by α2AP. A similar principle also applies when plasmin binds other proteins, including specific ECM proteins and cell surface receptors. For example, when plasmin associates with receptors on cell surfaces, the response is mediated by its kringle domains and the bound plasmin is protected from inhibition by α2AP and α2M (44, 45).

PAI-2 has been identified as a rapid inhibitor of tPA and uPA (46). Because of an inefficient signal sequence, PAI-2 is usually retained within cells, where it expresses unique biological activities (47). In specific conditions, such as pregnancy, PAI-2 becomes available in extracellular spaces and plays a role in controlling fibrinolysis (46). Other protease inhibitors that may regulate fibrinolysis under specific conditions and in specific tissue microenvironments include: PAI-3/protein C inhibitor (48, 49); protease nexin-1 (50); and neuroserpin, which functions mainly in the central nervous system (CNS) (51). Thrombin-activatable fibrinolysis inhibitor (TAFI) is the precursor for plasma carboxypeptidase B, which inhibits fibrinolysis by cleaving and removing C-terminal Lys residues from fibrin and other substrates (52).

PLASMINOGEN ACTIVATION AT THE CELL SURFACE

The macromolecules that maintain an anticoagulant state on the surfaces of uninjured cells (1014) are supported by cell-surface proteins with plasminogen-binding activity (35, 53, 54). Plasminogen receptors that have been identified using discovery-based methodologies are listed in Table 1, together with representative references (5564). Plasminogen receptors are structurally diverse and include both integral plasma membrane proteins and extrinsic membrane-associated proteins. In many cells, the total plasminogen binding capacity exceeds 106 sites/cell. The dissociation constant (KD) for plasminogen binding to most receptors is ∼1.0 µM (53, 54). Because the concentration of plasminogen in plasma is ∼1.7 µM, for cells such as endothelium and inflammatory cells that are in direct contact with plasma, ∼60% of the plasminogen receptors should be occupied, as determined by the formula: B/Bmax = [plasminogen, Plg]/([Plg] + KD). Some plasminogen receptors have been described as moonlighting proteins because they have, in addition to their role as plasminogen-binding proteins, alternative activities.

Table 1.

Examples of known cell-surface receptors for plasminogen

Receptor References
α-Enolase Miles et al. (55)
Annexin-A2/S100 complex Hajjar et al. (56)
Miller et al. (57)
Cytokeratin-8 Hembrough et al. (58)
Histone H2B Das et al. (59)
Actin Dudani et al. (60)
Plg-RKT Andronicos et al. (61)
Integrin αMβ2/Mac-1 Pluskota et al. (62)
Amphoterin Parkkinen et al. (63)
Megalin (LRP-2/gp330) Kanalas et al. (64)

Plasminogen receptors share common properties. First, plasminogen receptors typically are synthesized with C-terminal lysines or posttranslationally modified so that they have C-terminal lysines; this is critical for plasmin(ogen) binding (35, 53, 54). Second, many identified plasminogen receptors increase the catalytic efficiency (kcat/KM) of plasminogen activation, either in isolation or in concert with other cellular proteins that bind plasminogen activators. Finally, once plasmin is generated in association with receptors, it is protected from inhibitors (44, 45). These properties allow healthy cells that are directly exposed to blood plasma to coat themselves with sufficient quantities of plasmin so that any nidus of inappropriate fibrin formation is reversed.

Plasmin that is generated on the surfaces of cells targets important substrates beyond fibrin and fibrinogen. Thus, many biological activities, which have been attributed to plasminogen activators, reflect the activity of plasmin as a protease. For example, plasmin cleaves the amino-terminal component of latent transforming growth factor-β (TGF-β), resulting in release of active TGF-β (65). Plasmin also targets the chondroitin sulfate/heparan sulfate proteoglycan and TGF-β receptor, betaglycan, allowing for redistribution of active TGF-β between its diverse receptors and between neighboring cells (66).

Plasmin activates the “pro-form” of matrix metalloprotease-2 (MMP2) by a mechanism that requires the plasma membrane-anchored protein, membrane-type 1 matrix metalloprotease/MT1-MMP (67). Plasmin also activates the pro-form of MMP-3/stromelysin-1 in a reaction that leads to activation of MMP9 (68). Multiple glycoproteins in the ECM are plasmin substrates (6971). Cleavage of these proteins may result in remodeling of the ECM, altered interaction with cellular integrins, or release of growth factors from the ECM. Many complex interactions involving plasminogen activators, plasmin(ogen), MMPs, ECM proteins, growth factors, and cell surface protease receptors were elegantly reviewed a number of years ago by Mignatti and Rifkin (72). How these interactions may regulate innate immunity, inflammation, and the response to pathogens was more recently reviewed by Medcalf and Keragala (73).

THE uPA RECEPTOR ASSEMBLY: STRUCTURE AND CELL-SIGNALING ACTIVITY

The principal high-affinity receptor for uPA is urokinase receptor (uPAR), a three-domain glycosylphosphatidylinositol (GPI)-anchored membrane protein (74). The crystal structure of uPAR revealed a central cavity formed by its three domains (D1–D3) into which the amino terminal fragment of uPA, which includes the growth factor and kringle domains, inserts (75). When early studies were published indicating that uPA binding to uPAR activates cell signaling, these results were unanticipated because uPAR lacks transmembrane and intracellular domains (7681). The solution to this problem emerged as uPAR coreceptors, with known cell-signaling activity, were discovered.

One way to conceptualize this system is to consider uPAR a membrane-anchored ligand as opposed to a receptor. The function of uPAR as a ligand is activated by uPA binding, which triggers cell signaling through uPAR coreceptors. In support of this hypothesis, soluble forms of uPAR activate cell signaling similarly to uPA, apparently by direct binding to uPAR coreceptors (8286). This interaction is facilitated by cleavage of soluble uPAR between its N-terminal and central domains, which yields a D2–D3 fragment that is active in cell signaling (82, 84, 85, 87, 88). When uPAR is membrane-anchored, the principal protease responsible for uPAR cleavage between D1 and D2 is uPA (88). Full-length soluble uPAR is less readily cleaved by uPA but may be converted into D1 and the bioactive D2D3 fragment by chymotrypsin (85, 8789). Full-length soluble uPAR, the D1 fragment, and the D2D3 fragment have been identified as useful biomarkers for neoplastic and inflammatory diseases (88, 89).

Diverse integrins have been identified as uPAR coreceptors, which directly associate with uPAR and mediate cell signaling in response to uPA-uPAR complex and cleaved soluble uPAR, including but not limited to: α5β1, α4β1, αvβ5, and αMβ2/Mac-1 (83, 9094). The pertussis toxin-sensitive G protein-coupled receptor, N-formyl peptide receptor-2 (FPR2; formerly FPRL1/LXA4R), also was identified as an essential uPAR coreceptor (84). FPR2 responds to membrane-anchored uPA-uPAR complex and to cleaved soluble uPAR, suggesting collaboration between FPR2 with integrins within a single cell-signaling receptor assembly. Finally, receptor tyrosine kinases (RTKs) and specifically the epidermal growth factor receptor (EGF-R) have been implicated in uPA-initiated cell signaling (95, 96). The EGF-R may be transactivated by Src family kinases (SFKs), downstream of uPA-uPAR-integrin complexes. Ligand-independent EGF-R transactivation does not require physical association of the EGF-R with the uPA cell-signaling receptor assembly; however, physical association has been demonstrated (95). The uPA cell-signaling receptor assembly is shown in Fig. 3. Activation of cell signaling by uPA has diverse effects on cell physiology. Prominent among these is promotion of cell migration and cell survival (74, 78, 79, 92, 97, 98).

Figure 3.

Figure 3.

The urokinase-type plasminogen activator (uPA) cell-signaling receptor assembly is shown, including urokinase receptor (uPAR), N-formyl peptide receptor-2 (FPR2), integrins, and the epidermal growth factor receptor (EGF-R). Other plasma membrane proteins also may participate in this receptor assembly. The uPA cell-signaling receptor assembly is activated to trigger cell-signaling when uPA binds to glycosylphosphatidyl inositol (GPI)-anchored uPAR. Soluble uPAR also may trigger cell-signaling through the uPA cell-signaling receptor assembly, especially when soluble uPAR is cleaved between its first and second domains.

The collection of uPAR coreceptors shown in Fig. 3 may not be comprehensive. For example, at least in some cells, glycoprotein 130 (gp130) may associate with uPAR to mediate uPA-initiated cell signaling (99). gp130 is best known as a transmembrane protein that associates with cytokine receptors to mediate signal transduction in response to cytokines (100). Interestingly, gp130 also may directly bind uPA (101). These gp130 interactions appear to be essential for activation of JAK/STAT signaling by uPA (99).

Vitronectin is a second, distinct ligand for uPAR, which activates uPAR-dependent signal transduction (96, 102104). Vitronectin-triggered uPAR signaling does not require uPA and does not require the EGF-R as a coreceptor (96, 103). A major downstream mediator of vitronectin-initiated uPAR signaling is Rac1, as opposed to ERK1/2, which plays an important role in uPA-initiated cell signaling. Rac1 and ERK1/2 are complementary promoters of cell migration by distinct mechanisms (105).

THE tPA RECEPTOR ASSEMBLY: STRUCTURE AND CELL-SIGNALING ACTIVITY

For over twenty years, it has been apparent that tPA expresses biological activities unrelated to fibrinolysis, including, for example, regulation of endothelial cell proliferation (106), smooth muscle cell proliferation (107), and activation of microglia (108). These activities were described as either dependent on the protease activity of tPA or not. tPA activities that required its protease activity probably reflected plasminogen activation and modification of one or more plasmin substrates, including growth factors, ECM proteins, and MMP pro-forms. Plasmin that is generated on cell surfaces also may activate cell-signaling receptors in the protease-activated receptor (PAR) family, affecting diverse cell physiologic processes (109112).

Low-density lipoprotein receptor-related protein-1 (LRP1) is a 600-kDa transmembrane receptor in the LDL receptor gene family that undergoes endocytosis in clathrin-coated pits and rapid recycling to the cell surface (113, 114). LRP1-associated ligands are typically delivered to lysosomes. LRP1 was first identified as a tPA receptor in studies demonstrating the ability of hepatic LRP1 to clear tPA from the circulation (115, 116). As the LRP1 field advanced, it became apparent that LRP1 is capable of coupling ligand endocytosis with activation of cell signaling (113, 114). This occurs by at least two mechanisms. First, the cytoplasmic tail of LRP1 serves as a scaffold for assembling cell-signaling enzymes and adaptor proteins, facilitating activation of cell-signaling pathways (113, 117119). Second, based on its broad ligand-binding activity, LRP1 may sequester ligands and deliver these ligands to cell-signaling coreceptors.

Activation of cell-signaling by binding of tPA to LRP1 induces expression of genes encoding MMPs (120, 121); opens the blood-brain barrier (122); promotes hippocampal long-term potentiation (123); promotes cell survival (124126); increases cell migration (127); and promotes neurite outgrowth (128). To confirm that activities attributed to tPA reflect receptor-binding and not plasminogen activation, studies have been performed with enzymatically inactive tPA (EI-tPA), in which the active-site serine is mutated. To implicate LRP1, Lrp1 gene-silencing approaches have been applied. LRP receptor-associated protein (RAP) is a useful reagent because it blocks the interaction of LRP1 with other LRP1 ligands (129). In many cell culture model systems, RAP does not independently regulate cell signaling and thus, serves as a competitive LRP1 cell-signaling inhibitor; however, this is not uniformly true (130). A pitfall that should be recognized when studying RAP is its ability to bind to receptors in addition to LRP1 in the LDL receptor gene family (113). A variety of Lrp1 conditional gene knockout mice have been generated and have been highly useful in studying the diverse activities of this receptor (131).

A second receptor that has been implicated in tPA-initiated cell signaling is the N-methyl-d-aspartate receptor (NMDA-R) (132). Although the NMDA-R is best characterized as a neuronal ionotropic glutamate receptor (133), the NMDA-R has been identified in many cell types, including macrophages and Schwann cells in which tPA-initiated cell signaling is dependent on the NMDA-R (134, 135). Direct binding of tPA to the N-terminal region of the essential GluN1 NMDA-R subunit has been demonstrated (136, 137). This event may result in GluN1 cleavage (132, 138), although, at least in some model systems, NMDA-R subunit cleavage does not appear to be required for NMDA-R-mediated tPA-signaling because EI-tPA is active (127, 134, 135). Other NMDA-R subunits also may be cleaved when enzymatically active tPA interacts with the NMDA-R (139).

In the nervous system, the tPA-NMDA-R interaction has been reported to support neuronal survival and paradoxically, to be neurotoxic (126, 140). Experimental conditions may determine which result is observed. The effects of tPA-activated cell signaling on neuronal survival and other CNS responses also may depend on which member of the NMDA-R family mediates the tPA response. For example, blocking the activity of GluN2D-containing NMDA-Rs specifically inhibits tPA-mediated neurotoxicity (141). Interaction of tPA with GluN2B-containing NMDA-Rs in the hippocampus regulates contextual fear responses (142).

The possibility that LRP1 and the NMDA-R function together, as members of a single tPA receptor assembly, was first proposed over ten years ago (143, 144) and supported by more recent studies (127, 134, 135). A deeper dive into this synergy has shown that in PC12 cells, N2a cells, and Schwann cells, the requirement for the NMDA-R to mediate tPA-activated cell signaling is absolute (127, 135). By contrast, neutralizing LRP1 does not completely block tPA-activated cell signaling but instead, increases the tPA concentration required to elicit cell-signaling events. These results are consistent with a “sequester and deliver” model for the function of LRP1 in tPA-activated cell signaling. According to this model, most tPA signaling events are triggered by tPA, which associates first with LRP1 and is then delivered to the NMDA-R. In the absence of LRP1, signaling is limited to events in which tPA binds directly to the GluN1 NMDA-R subunit without first being sequestered at the cell surface. In support of this model, the NMDA-R and LRP1 have been reported to form a multiprotein complex, in tPA-treated neurons and neuron-like cells, bridged by the bifunctional intracellular adapter protein, postsynaptic density protein-95 (PSD-95) (127, 143).

The tPA cell-signaling receptor assembly is shown in Fig. 4. Compared with uPA, less is known about the various cell-signaling cascades activated by tPA. When tPA engages the NMDA-R/LRP1 complex, calcium influx occurs, which results in activation of SFKs (127, 128). SFKs are capable of activating diverse downstream substrates and controlling diverse cellular processes (145). RTKs are transactivated by SFKs, which expands the continuum of cell-signaling proteins that may be regulated (146). The receptor assembly shown in Fig. 4 may not be complete. For example, it has been shown that the nonpathogenic form of cellular prion protein/PrPC associates with LRP1 to mediate cell signaling in response to tPA (147). Furthermore, it has been reported that Annexin A2 mediates tPA-activated cell signaling (148). Whether Annexin A2 functions in concert with the NMDA-R/LRP1 receptor system or independently is an important question.

Figure 4.

Figure 4.

The tissue-type plasminogen activator (tPA) cell-signaling receptor assembly is shown. In the “sequester and deliver” model, LRP1 functions to initially bind tPA and then transfer tPA to the N-methyl-d-aspartate receptor (NMDA-R). In the absence of LRP1, tPA may bind directly to the NMDA-R; however, under these conditions, higher tPA concentrations are required to elicit cell-signaling responses. At least in some cell types, tPA binding to the tPA cell-signaling receptor assembly causes postsynaptic density protein-95 (PSD-95) to associate with both LRP1 and the NMDA-R, providing an intracytoplasmic bridge between these two receptors.

uPA RECEPTOR ASSEMBLIES IN INNATE IMMUNITY AND INFLAMMATION

In innate immunity, pattern recognition receptors (PRRs) recognize conserved products released by infectious agents and rapidly reprogram cells, generating proinflammatory mediators that support organism defense (149, 150). In mammalian systems, innate immunity provides an early response to infection. Unfortunately, dysregulated innate immunity contributes to many illnesses that involve chronic inflammation (151). Elucidating novel pathways that regulate innate immunity is very important in the anti-inflammatory therapeutics development field.

To understand the function of uPAR in innate immunity and inflammation, first, its expression pattern should be considered. In healthy adult humans, uPAR expression is fairly restricted, which has contributed to interest in developing uPAR-targeting drugs to treat cancer (152, 153). CD34-positive hematopoietic precursor cells in the bone marrow are uPAR-negative (154156). Monocytes and granulocytes acquire uPAR as they mature and differentiate. Resting lymphocytes also are uPAR negative; however, activated lymphocytes and NK cells may express uPAR (155, 157). The effects of cellular differentiation and/or activation on uPAR expression are probably related to the ability of uPAR to promote cell migration and cell survival. The effects of uPAR on migration of neutrophils and monocytes/macrophages have been convincingly demonstrated using both in vitro and in vivo model systems (158, 159). When the gene encoding uPAR (Plaur) is deleted in mice, neutrophil migration into lungs infected with Pseudomonas aeruginosa is impaired and, as a result, clearance of the pathogen also is impaired (159).

Agents that activate PRRs in the Toll-like receptor (TLR) family upregulate uPAR expression (160, 161). Moreover, uPA and the uPA cell-signaling receptor assembly regulate TLR activity. Kiyan et al. (162) demonstrated that uPAR is a member of the TLR4 interactome. uPAR-deficient leukocytes demonstrate attenuated cell-signaling responses and decreased proinflammatory cytokine expression when treated with the TLR4 agonist, lipopolysaccharide (LPS). The toxicity of systemically administered LPS is attenuated in uPAR-deficient mice (162). uPAR deficiency also protects mice from the effects of LPS on kidney barrier function (163). uPA potentiates activation of neutrophils by LPS, by a mechanism that depends on the integrin, αvβ3 (164). Similarly, uPAR may be necessary for optimal neutrophil activation in response to TLR2 agonists (165). Collectively, these studies point to a role for the uPA cell-signaling receptor assembly in supporting TLR activity. However, in some complex models of human disease, the role of uPAR and its coreceptors may be less straightforward. For example, in mouse models of colitis, uPAR deficiency is associated with increased inflammation, an effect that was explained by the ability of uPAR to support integrin αMβ2/Mac-1-dependent phagocytosis and bacterial product tolerance (166).

tPA RECEPTOR ASSEMBLIES IN INNATE IMMUNITY AND INFLAMMATION

Our current understanding of the activity of tPA in innate immunity and inflammation may be traced in part to earlier studies that focused on LRP1. Overton et al. (167) deleted Lrp1 in monocytes, macrophages, and neutrophils, under the control of the LysM promoter, in mice. When bone marrow from these mice was transplanted into irradiated LDL receptor-deficient mice, atherosclerosis in response to a Western diet was significantly increased. This result was explained mechanistically by studies demonstrating that LRP1 deficiency increases expression of proinflammatory mediators, such as monocyte chemoattractant protein-1/MCP1 and MMP9. LRP1-deficient macrophages demonstrated increased migration in vitro and LRP1-deficient monocytes migrated in greater numbers into atherosclerotic lesions in vivo (167). Similarly, we demonstrated that LRP1-deficient monocytes migrate in increased numbers into pancreatic tumor isografts in mice (168). Mechanistically, we attributed this result to increased expression of the promigratory chemokine, macrophage inflammatory protein-1α/CCL3, and its receptor CCR5.

May et al. (169) demonstrated that in macrophages, LRP1 deficiency is associated with increased expression of genes that define the proinflammatory M1 phenotype and decreased expression of gene products that define the M2 phenotype (169). The ability of LRP1 to regulate the expression of proinflammatory mediators is not restricted to macrophages. In a number of cell lines, deletion or silencing of the gene encoding LRP1 (Lrp1) enhanced the basal level of activity of the IκBα kinase-NFκB pathway, derepressing expression of proinflammatory mediators, including MCP-1, iNOS, and complement proteases (170).

The activities of LRP1 and tPA in inflammation were linked conceptually when we demonstrated that the effects of LRP1 on proinflammatory mediator expression are evident not only when Lrp1 is deleted but also in LRP1-expressing macrophages, when these cells are treated with LRP1 ligands (130). Importantly, the effects of LRP1 ligands on gene expression in macrophages are ligand-specific (130). In the absence of TLR agonists, the LRP1 ligands, RAP and lactoferrin, independently activate NFκB in macrophages, inducing expression of proinflammatory cytokines, promoting macrophage migration, and increasing the expression of the microRNA, miR-155. These changes mimic those observed when Lrp1 is deleted. By contrast, the LRP1 ligands, EI-tPA and activated α2M, block NFκB activation and cytokine expression in LPS-treated macrophages (130, 134).

LRP1 ligands number over 100, including proteins released by injured and dying cells (113, 171, 172). Although the number of ligands we have examined in macrophage gene expression studies is still limited, it is intriguing to consider a model in which LRP1 ligands are divided into “cell-signaling agonists,” which oppose production of proinflammatory mediators, and “cell-signaling antagonists,” which stimulate production of proinflammatory mediators. Under basal conditions, macrophages in cell culture probably produce sufficient levels of LRP1 cell-signaling agonists endogenously to suppress activation of the NFκB pathway. Addition of exogenous LRP1 agonists, such as EI-tPA, amplifies anti-inflammatory LRP1-signaling sufficiently so that even robust LPS responses are attenuated (130, 134). Similarly, therapeutically administered EI-tPA blocks the toxicity of LPS in mice (134).

When Lrp1 is deleted, autocrine LRP1 signaling is eliminated and with it, the effects of this system on NFκB activation. In LRP1-expressing cells, antagonists such as RAP competitively block binding of endogenously produced ligands to LRP1, creating a situation similar to that observed with Lrp1 deletion. Our model, which reconciles the effects of LRP1 ligands and Lrp1 deletion, is presented in Fig. 5. Further studies will be required to determine whether this model is correct. One interesting observation is that the intensity of the proinflammatory effect observed when Lrp1 is deleted depends on the approach applied to delete the gene (130, 168). This raises the question of whether other members of the LDL receptor gene family may compensate for LRP1 when Lrp1 is deleted during development.

Figure 5.

Figure 5.

A model showing how tissue-type plasminogen activator (tPA) may regulate NFκB activation in macrophages. It is assumed that LRP1 ligands include proteins secreted by macrophages, a fraction of which activates cell signaling via the tPA cell-signaling receptor assembly, similar to tPA. This autocrine pathway, involving endogenously produced LRP1 ligands, suppresses NFκB activation resulting from low levels of activation of pattern recognition receptors (PRRs) and other proinflammatory receptors under basal conditions (top left). When an LRP1 antagonist such as LRP receptor-associated protein (RAP) is added, or when Lrp1 is silenced or deleted, cell-signaling via the tPA receptor assembly is aborted, eliminating its activity in maintaining immune quiescence and increasing NFκB activity (top right). In the presence of LRP1, cell-signaling via the tPA receptor assembly, triggered by endogenously produced ligands, is insufficient to neutralize cell-signaling activated by high levels of PRR agonists such as lipopolysaccharide (LPS) (bottom left). Under these conditions, NFκB activation is observed. However, if tPA is administered exogenously, to supplement endogenously produced ligands, even responses triggered by high levels of PRR agonists may be neutralized (bottom right).

If the model shown in Fig. 5 is proven correct, it will be important to determine how LRP1 ligands that are cell-signaling agonists and antagonists differ. One possible explanation focuses on the NMDA-R, which is expressed by macrophages and essential for the anti-inflammatory activity of tPA (134). LRP1 agonists may be ligands that bind to LRP1 and also engage the NMDA-R. Expression of the NMDA-R in macrophages is differentiation state-dependent (173). Macrophages that are harvested from the mouse peritoneum without eliciting or activating agents express very low levels of the NMDA-R and are not responsive to tPA. However, when these cells are treated with colony-stimulating factor-1, the abundance of cell-surface NMDA-R increases and responsiveness to tPA is acquired. The importance of the NMDA-R as an essential LRP1 coreceptor in cell signaling has been demonstrated for activated α2M as well, in early studies with cultured neurons (174).

In addition to TLR4, tPA counteracts proinflammatory responses mediated by ligands that activate other TLRs, including TLR2 and TLR9 (173). In unpublished studies, we have shown that tPA also is effective at inhibiting TLR7; however, not all PRRs are antagonized by tPA. Responses to the PRRs, nucleotide binding oligomerization domain-containing 1 (NOD1) and NOD2, are not antagonized by tPA and instead, may be amplified (173). Furthermore, plasmin that is activated on macrophage cell surfaces stimulates proinflammatory responses by activating PARs, which also cannot be blocked by tPA (112). Thus, it may be most correct to refer to the tPA receptor assembly as an “anti-TLR system” as opposed to a generalized anti-inflammatory system.

Given the PRR-selective nature of tPA, we undertook studies to assess the efficacy of EI-tPA as a candidate anti-inflammatory agent in the dextran sulfate sodium (DSS) colitis model. DSS colitis is a complex model of a human inflammatory bowel disease (IBD) in which TLRs and diverse other PRRs contribute to the symptoms and pathology observed (175178). DSS is administered in the drinking water and causes a chemical colitis focused in the large intestines. Although this IBD model has little in common with the development of IBD in humans, once pathology develops in DSS colitis, it is characterized by epithelial denudation, crypt disruption, and extensive inflammatory infiltrates in the mucosa and submucosa.

We treated C57BL/6 mice with DSS for 5 or 7 days. A single dose of EI-tPA (2.5 mg⋅kg−1) was administered by intravenous injection immediately after DSS treatment was terminated. The effects of the EI-tPA on the rate of recovery were monitored. In studies examining mouse weight, abdominal tenderness, stool character and blood content, and in posteuthanasia studies examining colon length (an index of irreversible tissue damage and scarring) and colon histology, EI-tPA significantly and robustly accelerated recovery (179).

Our DSS colitis study justifies additional studies to examine EI-tPA and other ligands that engage the tPA cell-signaling receptor assembly as candidate therapeutics for complex human diseases in which inflammation plays a pivotal role. However, many important questions still remain. For example, although most of our research has focused on macrophages, the cell type or types that are targeted by EI-tPA in the DSS colitis model remains to be determined. Given the broad range of effects on cell biology attributed to the NMDA-R/LRP1 receptor complex, we cannot be certain whether the beneficial effects of EI-tPA in the DSS model system reflects its anti-TLR activity or, for example, effects on cell survival and proliferation. Considerable opportunities for future work exist.

Another area of importance is the cell-signaling cascade that couples activation of the tPA receptor assembly to regulation of NFκB activation. This cascade remains incompletely defined. When macrophages are treated with a TLR agonist and EI-tPA simultaneously, EI-tPA does not prevent IκBα phosphorylation, which is required for activation of NFκB as a transcription factor (134, 173). Instead, EI-tPA rapidly reverses IκBα phosphorylation. This process is sufficient to prevent proinflammatory cytokine expression. An important mediator may be phosphatidylinositol 3-kinase-γ, an inhibitor of TLR-induced NFκB activation, known to be activated downstream of the NMDA-R (180, 181). In response to TLR stimulation, the cytoplasmic tail of LRP1 is phosphorylated and recruits Rab8a, which leads to activation of phosphatidylinositol 3-kinase-γ (119). The activity of this pathway in cells treated with tPA will constitute an important area of investigation.

Finally, the effects of the principal tPA inhibitor, PAI-1, on tPA cell signaling deserves consideration. PAI-1 is present in the plasma at increased levels in conditions associated with inflammation, including metabolic syndrome, cardiovascular disease, and diabetes (182184). Whether PAI-1 is simply a biomarker of these serious illnesses or contributes mechanistically is not clear. We have shown that complex formation between catalytically active tPA and PAI-1 completely blocks the ability of tPA to inhibit NFκB activation and cytokine expression in macrophages treated with LPS (185). Similarly, PAI-1 blocks the cell-signaling activity of tPA in experiments with PC12 cells and Schwann cells. The effects of PAI-1 on tPA-activated cell signaling were unanticipated because the affinity of tPA-PAI-1 complex for LRP1 is actually higher than the affinity of tPA (186). Inhibiting the anti-TLR activity of tPA represents a plausible mechanism by which PAI-1 may be proinflammatory in diverse diseases.

CONCLUDING REMARKS/FUTURE DIRECTIONS

Although major components of the hemostasis system are, for the most part, well described, hemostasis factors have important activities in cell biology that remain only partially understood. The effect of fibrinolysis factors on innate immunity and inflammation is an important example. Considerably more work will be required to fully understand how fibrinolysis proteases and their inhibitors regulate these processes. It also is important to better understand how inflammatory mediators regulate hemostasis. This topic is not covered in this review.

Mining the hemostasis system to develop novel therapeutics to treat chronic inflammation constitutes an important opportunity. In this review, we focused on EI-tPA as an example of a candidate therapeutic derived from the fibrinolysis system. The activity of EI-tPA in a mouse model of IBD was discussed (179). In a second study, using a mouse model of spinal cord injury, EI-tPA again demonstrated efficacy, improving the ability of induced pluripotent stem cells to restore motor function (187). Although the activity of EI-tPA in this study was attributed to direct effects of EI-tPA on the stem cells, EI-tPA was administered into the spinal cord injury site with the stem cells. Inflammation is a prominent contributor to the pathology observed in spinal cord injury (188) and may have been modified by the EI-tPA.

The activity of EI-tPA, observed in the LPS challenge and DSS colitis models (134, 179), was somewhat surprising because the circulating half-life of inactive tPA in mice is less than 5 min (189). We assume that in these model systems, EI-tPA rapidly activated cell-signaling pathways in target cells, which induced sustained effects on the physiology of those cells. Because LRP1 may have opposing effects on the activity of EI-tPA in vivo, decreasing the EI-tPA concentration required to elicit NMDA-R-dependent cell signaling (114, 127, 134, 135), while, at the same time, clearing EI-tPA from the circulation (115, 116), there may be an opportunity to genetically engineer tPA and/or other ligands that engage the tPA cell-signaling receptor assembly to optimize the activity of candidate therapeutics.

Our studies in the DSS model system justify additional studies of EI-tPA in other models of diseases with chronic inflammation, including but not limited to rheumatoid arthritis, multiple sclerosis, and psoriasis. Finally, TLRs have been identified as important therapeutic targets in SARS-CoV-2 (190). Given the anti-TLR activity of the tPA cell-signaling receptor assembly, it may be legitimate to ask whether EI-tPA should be studied as a candidate therapeutic for this illness.

GRANTS

Dr. S. L. Gonias was partially supported by NIH Grants R01 HL136395 and R01 NS097590.

DISCLOSURES

No conflicts of interest, financial or otherwise, are declared by the author.

AUTHOR CONTRIBUTIONS

S.L.G. prepared figures; drafted manuscript; edited and revised manuscript; and approved final version of manuscript.

ACKNOWLEDGMENTS

The author thanks colleagues, Dr. Elisabetta Mantuano (UCSD) and Dr. Wendy M. Mars (University of Pittsburgh), for critical reading of this manuscript. The author also thanks Pardis Azmoon for assistance in preparing figures. Figures were created with BioRender.com.

REFERENCES

  • 1.Diegelmann RF, Evans MC. Wound healing: an overview of acute, fibrotic and delayed healing. Front Biosci 9: 283–289, 2004. doi: 10.2741/1184. [DOI] [PubMed] [Google Scholar]
  • 2.Reinke JM, Sorg H. Wound repair and regeneration. Eur Surg Res 49: 35–43, 2012. doi: 10.1159/000339613. [DOI] [PubMed] [Google Scholar]
  • 3.Rodrigues M, Kosaric N, Bonham CA, Gurtner GC. Wound healing: a cellular perspective. Physiol Rev 99: 665–706, 2019. doi: 10.1152/physrev.00067.2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Monroe DM, Hoffman M. What does it take to make the perfect clot? Arterioscler Thromb Vasc Biol 26: 41–48, 2006. doi: 10.1161/01.ATV.0000193624.28251.83. [DOI] [PubMed] [Google Scholar]
  • 5.Wolberg AS, Campbell RA. Thrombin generation, fibrin clot formation and hemostasis. Transfus Apher Sci 38: 15–23, 2008. doi: 10.1016/j.transci.2007.12.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Versteeg HH, Heemskerk JW, Levi M, Reitsma PH. New fundamentals in hemostasis. Physiol Rev 93: 327–358, 2013. doi: 10.1152/physrev.00016.2011. [DOI] [PubMed] [Google Scholar]
  • 7.Chapin JC, Hajjar KA. Fibrinolysis and the control of blood coagulation. Blood Rev 29: 17–24, 2015. doi: 10.1016/j.blre.2014.09.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Menter DG, Kopetz S, Hawk E, Sood AK, Loree JM, Gresele P, Honn KV. Platelet “first responders” in wound response, cancer, and metastasis. Cancer Metastasis Rev 36: 199–213, 2017. doi: 10.1007/s10555-017-9682-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Weisel JW. Fibrinogen and fibrin. Adv Protein Chem 70: 247–299, 2005. doi: 10.1016/S0065-3233(05)70008-5. [DOI] [PubMed] [Google Scholar]
  • 10.Wu KK, Thiagarajan P. Role of endothelium in thrombosis and hemostasis. Annu Rev Med 47: 315–331, 1996. doi: 10.1146/annurev.med.47.1.315. [DOI] [PubMed] [Google Scholar]
  • 11.Yau JW, Teoh H, Verma S. Endothelial cell control of thrombosis. BMC Cardiovasc Disord 15: 130, 2015. doi: 10.1186/s12872-015-0124-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Esmon CT. The roles of protein C and thrombomodulin in the regulation of blood coagulation. J Biol Chem 264: 4743–4746, 1989. doi: 10.1016/S0021-9258(18)83649-3. [DOI] [PubMed] [Google Scholar]
  • 13.Mohan Rao LV, Esmon CT, Pendurthi UR. Endothelial cell protein C receptor: a multiliganded and multifunctional receptor. Blood 124: 1553–1562, 2014. doi: 10.1182/blood-2014-05-578328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Pike RN, Buckle AM, Le Bonniec BF, Church FC. Control of the coagulation system by serpins. Getting by with a little help from glycosaminoglycans. FEBS J 272: 4842–4851, 2005. doi: 10.1111/j.1742-4658.2005.04880.x. [DOI] [PubMed] [Google Scholar]
  • 15.Vassalli JD, Sappino AP, Belin D. The plasminogen activator/plasmin system. J Clin Invest 88: 1067–1072, 1991. doi: 10.1172/JCI115405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Castellino FJ, Ploplis VA. Structure and function of the plasminogen/plasmin system. Thromb Haemost 93: 647–654, 2005. doi: 10.1160/TH04-12-0842. [DOI] [PubMed] [Google Scholar]
  • 17.Lupu F, Heim DA, Bachmann F, Hurni M, Kakkar VV, Kruithof EK. Plasminogen activator expression in human atherosclerotic lesions. Arterioscler Thromb Vasc Biol 15: 1444–1455, 1995. doi: 10.1161/01.atv.15.9.1444. [DOI] [PubMed] [Google Scholar]
  • 18.Bugge TH, Flick MJ, Danton MJ, Daugherty CC, Romer J, Dano K, Carmeliet P, Collen D, Degen JL. Urokinase-type plasminogen activator is effective in fibrin clearance in the absence of its receptor or tissue-type plasminogen activator. Proc Natl Acad Sci USA 93: 5899–5904, 1996. doi: 10.1073/pnas.93.12.5899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Pizzo SV, Schwartz ML, Hill RL, McKee PA. The effect of plasmin on the subunit structure of human fibrinogen. J Biol Chem 247: 636–645, 1972. doi: 10.1016/S0021-9258(19)45656-1. [DOI] [PubMed] [Google Scholar]
  • 20.Pizzo SV, Schwartz ML, Hill RL, McKee PA. The effect of plasmin on the subunit structure of human fibrin. J Biol Chem 248: 4574–4583, 1973. doi: 10.1016/s0021-9258(19)43703-4. [DOI] [PubMed] [Google Scholar]
  • 21.Adam SS, Key NS, Greenberg CS. D-dimer antigen: current concepts and future prospects. Blood 113: 2878–2887, 2009. doi: 10.1182/blood-2008-06-165845. [DOI] [PubMed] [Google Scholar]
  • 22.Urano T, Takada Y, Takada A. Stimulation of the amidolytic activity of single chain tissue-type plasminogen activator by fibrinogen degradation products: possible fibrin binding sites on single chain tissue-type plasminogen activator molecule. Biochim Biophys Acta 1077: 245–252, 1991. doi: 10.1016/0167-4838(91)90536-9. [DOI] [PubMed] [Google Scholar]
  • 23.Renatus M, Engh RA, Stubbs MT, Huber R, Fischer S, Kohnert U, Bode W. Lysine 156 promotes the anomalous proenzyme activity of tPA: X-ray crystal structure of single-chain human tPA. EMBO J 16: 4797–4805, 1997. doi: 10.1093/emboj/16.16.4797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Lijnen HR, Van Hoef B, Nelles L, Collen D. Plasminogen activation with single-chain urokinase-type plasminogen activator (scu-PA). Studies with active site mutagenized plasminogen (Ser740––Ala) and plasmin-resistant scu-PA (Lys158––Glu). J Biol Chem 265: 5232–5236, 1990. doi: 10.1016/S0021-9258(19)34111-0. [DOI] [PubMed] [Google Scholar]
  • 25.Wiman B, Collen D. On the kinetics of the reaction between human antiplasmin and plasmin. Eur J Biochem 84: 573–578, 1978. doi: 10.1111/j.1432-1033.1978.tb12200.x. [DOI] [PubMed] [Google Scholar]
  • 26.Alessi MC, Declerck PJ, De Mol M, Nelles L, Collen D. Purification and characterization of natural and recombinant human plasminogen activator inhibitor-1 (PAI-1). Eur J Biochem 175: 531–540, 1988. doi: 10.1111/j.1432-1033.1988.tb14225.x. [DOI] [PubMed] [Google Scholar]
  • 27.Lijnen HR, Van Hoef B, Collen D. On the reversible interaction of plasminogen activator inhibitor-1 with tissue-type plasminogen activator and with urokinase-type plasminogen activator. J Biol Chem 266: 4041–4044, 1991. doi: 10.1016/S0021-9258(20)64281-8. [DOI] [PubMed] [Google Scholar]
  • 28.Collen D. Identification and some properties of a new fast-reacting plasmin inhibitor in human plasma. Eur J Biochem 69: 209–216, 1976. doi: 10.1111/j.1432-1033.1976.tb10875.x. [DOI] [PubMed] [Google Scholar]
  • 29.Aoki N, Moroi M, Matsuda M, Tachiya K. The behavior of alpha2-plasmin inhibitor in fibrinolytic states. J Clin Invest 60: 361–369, 1977. doi: 10.1172/JCI108784. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Harpel PC. Plasmin inhibitor interactions. The effectiveness of alpha2-plasmin inhibitor in the presence of alpha2-macroglobulin. J Exp Med 146: 1033–1040, 1977. doi: 10.1084/jem.146.4.1033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Hoylaerts M, Rijken DC, Lijnen HR, Collen D. Kinetics of the activation of plasminogen by human tissue plasminogen activator. Role of fibrin. J Biol Chem 257: 2912–2919, 1982. [PubMed] [Google Scholar]
  • 32.Medved L, Nieuwenhuizen W. Molecular mechanisms of initiation of fibrinolysis by fibrin. Thromb Haemost 89: 409–419, 2003. doi: 10.1055/s-0037-1613368. [DOI] [PubMed] [Google Scholar]
  • 33.Wiman B, Lijnen HR, Collen D. On the specific interaction between the lysine-binding sites in plasmin and complementary sites in alpha2-antiplasmin and in fibrinogen. Biochim Biophys Acta 579: 142–154, 1979. doi: 10.1016/0005-2795(79)90094-1. [DOI] [PubMed] [Google Scholar]
  • 34.Anonick PK, Gonias SL. Soluble fibrin preparations inhibit the reaction of plasmin with alpha 2-macroglobulin. Comparison with alpha 2-antiplasmin and leupeptin. Biochem J 275: 53–59, 1991. doi: 10.1042/bj2750053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Plow EF, Herren T, Redlitz A, Miles LA, Hoover-Plow JL. The cell biology of the plasminogen system. FASEB J 9: 939–945, 1995. doi: 10.1096/fasebj.9.10.7615163. [DOI] [PubMed] [Google Scholar]
  • 36.Vali Z, Patthy L. The fibrin-binding site of human plasminogen. Arginines 32 and 34 are essential for fibrin affinity of the kringle 1 domain. J Biol Chem 259: 13690–13694, 1984. doi: 10.1016/S0021-9258(18)89800-3. [DOI] [PubMed] [Google Scholar]
  • 37.van Zonneveld AJ, Veerman H, Pannekoek H. On the interaction of the finger and the kringle-2 domain of tissue-type plasminogen activator with fibrin. Inhibition of kringle-2 binding to fibrin by epsilon-amino caproic acid. J Biol Chem 261: 14214–14218, 1986. doi: 10.1016/S0021-9258(18)67006-1. [DOI] [PubMed] [Google Scholar]
  • 38.Verheijen JH, Caspers MP, Chang GT, de Munk GA, Pouwels PH, Enger-Valk BE. Involvement of finger domain and kringle 2 domain of tissue-type plasminogen activator in fibrin binding and stimulation of activity by fibrin. EMBO J 5: 3525–3530, 1986. doi: 10.1002/j.1460-2075.1986.tb04678.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Fleury V, Loyau S, Lijnen HR, Nieuwenhuizen W, Angles-Cano E. Molecular assembly of plasminogen and tissue-type plasminogen activator on an evolving fibrin surface. Eur J Biochem 216: 549–556, 1993. doi: 10.1111/j.1432-1033.1993.tb18173.x. [DOI] [PubMed] [Google Scholar]
  • 40.Lijnen HR, Van Hoef B, De Cock F, Collen D. The mechanism of plasminogen activation and fibrin dissolution by single chain urokinase-type plasminogen activator in a plasma milieu in vitro. Blood 73: 1864–1872, 1989. doi: 10.1182/blood.V73.7.1864.bloodjournal7371864. [DOI] [PubMed] [Google Scholar]
  • 41.Takada A, Takada Y, Sugawara Y. Effects of fibrinogen and fibrin on the activation of Glu- and Lys-plasminogen by urokinase. Thromb Res 33: 561–569, 1984. doi: 10.1016/0049-3848(84)90111-7. [DOI] [PubMed] [Google Scholar]
  • 42.Markus G. Conformational changes in plasminogen, their effect on activation, and the agents that modulate activation rates—a review. Fibrinolysis 10: 75–85, 1996. doi: 10.1016/S0268-9499(96)80082-8. [DOI] [Google Scholar]
  • 43.Xue Y, Bodin C, Olsson K. Crystal structure of the native plasminogen reveals an activation-resistant compact conformation. J Thromb Haemost 10: 1385–1396, 2012. doi: 10.1111/j.1538-7836.2012.04765.x. [DOI] [PubMed] [Google Scholar]
  • 44.Hall SW, Humphries JE, Gonias SL. Inhibition of cell surface receptor-bound plasmin by alpha 2-antiplasmin and alpha 2-macroglobulin. J Biol Chem 266: 12329–12336, 1991. doi: 10.1016/S0021-9258(18)98900-3. [DOI] [PubMed] [Google Scholar]
  • 45.Plow EF, Freaney DE, Plescia J, Miles LA. The plasminogen system and cell surfaces: evidence for plasminogen and urokinase receptors on the same cell type. J Cell Biol 103: 2411–2420, 1986. doi: 10.1083/jcb.103.6.2411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Wyatt AR, Cater JH, Ranson M. PZP and PAI-2: structurally-diverse, functionally similar pregnancy proteins? Int J Biochem Cell Biol 79: 113–117, 2016. doi: 10.1016/j.biocel.2016.08.018. [DOI] [PubMed] [Google Scholar]
  • 47.Dickinson JL, Bates EJ, Ferrante A, Antalis TM. Plasminogen activator inhibitor type 2 inhibits tumor necrosis factor alpha-induced apoptosis. Evidence for an alternate biological function. J Biol Chem 270: 27894–27904, 1995. doi: 10.1074/jbc.270.46.27894. [DOI] [PubMed] [Google Scholar]
  • 48.Suzuki K. Protein C inhibitor (PAI-3): structure and multi-function. Fibrinolysis Proteol 14: 133–145, 2000. doi: 10.1054/fipr.2000.0063. [DOI] [Google Scholar]
  • 49.Schwartz BS, Espana F. Two distinct urokinase-serpin interactions regulate the initiation of cell surface-associated plasminogen activation. J Biol Chem 274: 15278–15283, 1999. doi: 10.1074/jbc.274.21.15278. [DOI] [PubMed] [Google Scholar]
  • 50.Madjene C, Boutigny A, Bouton MC, Arocas V, Richard B. Protease nexin-1 in the cardiovascular system: wherefore art thou? Front Cardiovasc Med 8: 652852, 2021. doi: 10.3389/fcvm.2021.652852. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Hastings GA, Coleman TA, Haudenschild CC, Stefansson S, Smith EP, Barthlow R, Cherry S, Sandkvist M, Lawrence DA. Neuroserpin, a brain-associated inhibitor of tissue plasminogen activator is localized primarily in neurons. Implications for the regulation of motor learning and neuronal survival. J Biol Chem 272: 33062–33067, 1997. doi: 10.1074/jbc.272.52.33062. [DOI] [PubMed] [Google Scholar]
  • 52.Bajzar L, Manuel R, Nesheim ME. Purification and characterization of TAFI, a thrombin-activable fibrinolysis inhibitor. J Biol Chem 270: 14477–14484, 1995. doi: 10.1074/jbc.270.24.14477. [DOI] [PubMed] [Google Scholar]
  • 53.Miles LA, Hawley SB, Baik N, Andronicos NM, Castellino FJ, Parmer RJ. Plasminogen receptors: the sine qua non of cell surface plasminogen activation. Front Biosci 10: 1754–1762, 2005. doi: 10.2741/1658. [DOI] [PubMed] [Google Scholar]
  • 54.Cesarman-Maus G, Hajjar KA. Molecular mechanisms of fibrinolysis. Br J Haematol 129: 307–321, 2005. doi: 10.1111/j.1365-2141.2005.05444.x. [DOI] [PubMed] [Google Scholar]
  • 55.Miles LA, Dahlberg CM, Plescia J, Felez J, Kato K, Plow EF. Role of cell-surface lysines in plasminogen binding to cells: identification of alpha-enolase as a candidate plasminogen receptor. Biochemistry 30: 1682–1691, 1991. doi: 10.1021/bi00220a034. [DOI] [PubMed] [Google Scholar]
  • 56.Hajjar KA, Jacovina AT, Chacko J. An endothelial cell receptor for plasminogen/tissue plasminogen activator. I. Identity with annexin II. J Biol Chem 269: 21191–21197, 1994. doi: 10.1016/S0021-9258(17)31947-6. [DOI] [PubMed] [Google Scholar]
  • 57.Miller VA, Madureira PA, Kamaludin AA, Komar J, Sharma V, Sahni G, Thelwell C, Longstaff C, Waisman DM. Mechanism of plasmin generation by S100A10. Thromb Haemost 117: 1058–1071, 2017. doi: 10.1160/TH16-12-0936. [DOI] [PubMed] [Google Scholar]
  • 58.Hembrough TA, Vasudevan J, Allietta MM, Glass WF II, Gonias SL. A cytokeratin 8-like protein with plasminogen-binding activity is present on the external surfaces of hepatocytes, HepG2 cells and breast carcinoma cell lines. J Cell Sci 108: 1071–1082, 1995. doi: 10.1242/jcs.108.3.1071. [DOI] [PubMed] [Google Scholar]
  • 59.Das R, Burke T, Plow EF. Histone H2B as a functionally important plasminogen receptor on macrophages. Blood 110: 3763–3772, 2007. doi: 10.1182/blood-2007-03-079392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Dudani AK, Ganz PR. Endothelial cell surface actin serves as a binding site for plasminogen, tissue plasminogen activator and lipoprotein(a). Br J Haematol 95: 168–178, 1996. doi: 10.1046/j.1365-2141.1996.7482367.x. [DOI] [PubMed] [Google Scholar]
  • 61.Andronicos NM, Chen EI, Baik N, Bai H, Parmer CM, Kiosses WB, Kamps MP, Yates JR 3rd, Parmer RJ, Miles LA. Proteomics-based discovery of a novel, structurally unique, and developmentally regulated plasminogen receptor, Plg-RKT, a major regulator of cell surface plasminogen activation. Blood 115: 1319–1330, 2010. doi: 10.1182/blood-2008-11-188938. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Pluskota E, Soloviev DA, Bdeir K, Cines DB, Plow EF. Integrin alphaMbeta2 orchestrates and accelerates plasminogen activation and fibrinolysis by neutrophils. J Biol Chem 279: 18063–18072, 2004. doi: 10.1074/jbc.M310462200. [DOI] [PubMed] [Google Scholar]
  • 63.Parkkinen J, Rauvala H. Interactions of plasminogen and tissue plasminogen activator (t-PA) with amphoterin. Enhancement of t-PA-catalyzed plasminogen activation by amphoterin. J Biol Chem 266: 16730–16735, 1991. doi: 10.1016/S0021-9258(18)55362-X. [DOI] [PubMed] [Google Scholar]
  • 64.Kanalas JJ, Makker SP. Identification of the rat Heymann nephritis autoantigen (GP330) as a receptor site for plasminogen. J Biol Chem 266: 10825–10829, 1991. doi: 10.1016/S0021-9258(18)99093-9. [DOI] [PubMed] [Google Scholar]
  • 65.Lyons RM, Gentry LE, Purchio AF, Moses HL. Mechanism of activation of latent recombinant transforming growth factor beta 1 by plasmin. J Cell Biol 110: 1361–1367, 1990. doi: 10.1083/jcb.110.4.1361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Lamarre J, Vasudevan J, Gonias SL. Plasmin cleaves betaglycan and releases a 60 kDa transforming growth factor-beta complex from the cell surface. Biochem J 302: 199–205, 1994. doi: 10.1042/bj3020199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Monea S, Lehti K, Keski-Oja J, Mignatti P. Plasmin activates pro-matrix metalloproteinase-2 with a membrane-type 1 matrix metalloproteinase-dependent mechanism. J Cell Physiol 192: 160–170, 2002. doi: 10.1002/jcp.10126. [DOI] [PubMed] [Google Scholar]
  • 68.Ramos-DeSimone N, Hahn-Dantona E, Sipley J, Nagase H, French DL, Quigley JP. Activation of matrix metalloproteinase-9 (MMP-9) via a converging plasmin/stromelysin-1 cascade enhances tumor cell invasion. J Biol Chem 274: 13066–13076, 1999. doi: 10.1074/jbc.274.19.13066. [DOI] [PubMed] [Google Scholar]
  • 69.Vlodavsky I, Fuks Z, Ishai-Michaeli R, Bashkin P, Levi E, Korner G, Bar-Shavit R, Klagsbrun M. Extracellular matrix-resident basic fibroblast growth factor: implication for the control of angiogenesis. J Cell Biochem 45: 167–176, 1991. doi: 10.1002/jcb.240450208. [DOI] [PubMed] [Google Scholar]
  • 70.Bonnefoy A, Legrand C. Proteolysis of subendothelial adhesive glycoproteins (fibronectin, thrombospondin, and von Willebrand factor) by plasmin, leukocyte cathepsin G, and elastase. Thromb Res 98: 323–332, 2000. doi: 10.1016/s0049-3848(99)00242-x. [DOI] [PubMed] [Google Scholar]
  • 71.Goldfinger LE, Stack MS, Jones JC. Processing of laminin-5 and its functional consequences: role of plasmin and tissue-type plasminogen activator. J Cell Biol 141: 255–265, 1998. doi: 10.1083/jcb.141.1.255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Mignatti P, Rifkin DB. Nonenzymatic interactions between proteinases and the cell surface: novel roles in normal and malignant cell physiology. Adv Cancer Res 78: 103–157, 2000. doi: 10.1016/s0065-230x(08)61024-6. [DOI] [PubMed] [Google Scholar]
  • 73.Medcalf RL, Keragala CB. Fibrinolysis: a primordial system linked to the immune response. Int J Mol Sci 22: 3406, 2021. doi: 10.3390/ijms22073406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Blasi F, Carmeliet P. uPAR: a versatile signalling orchestrator. Nat Rev Mol Cell Biol 3: 932–943, 2002. doi: 10.1038/nrm977. [DOI] [PubMed] [Google Scholar]
  • 75.Huai Q, Mazar AP, Kuo A, Parry GC, Shaw DE, Callahan J, Li Y, Yuan C, Bian C, Chen L, Furie B, Furie BC, Cines DB, Huang M. Structure of human urokinase plasminogen activator in complex with its receptor. Science 311: 656–659, 2006. doi: 10.1126/science.1121143. [DOI] [PubMed] [Google Scholar]
  • 76.Del Rosso M, Anichini E, Pedersen N, Blasi F, Fibbi G, Pucci M, Ruggiero M. Urokinase-urokinase receptor interaction: non-mitogenic signal transduction in human epidermal cells. Biochem Biophys Res Commun 190: 347–352, 1993. doi: 10.1006/bbrc.1993.1054. [DOI] [PubMed] [Google Scholar]
  • 77.Dumler I, Petri T, Schleuning WD. Interaction of urokinase-type plasminogenactivator (u-PA) with its cellular receptor (u-PAR) induces phosphorylation on tyrosine of a 38 kDa protein. FEBS Lett 322: 37–40, 1993. doi: 10.1016/0014-5793(93)81106-a. [DOI] [PubMed] [Google Scholar]
  • 78.Busso N, Masur SK, Lazega D, Waxman S, Ossowski L. Induction of cell migration by pro-urokinase binding to its receptor: possible mechanism for signal transduction in human epithelial cells. J Cell Biol 126: 259–270, 1994. doi: 10.1083/jcb.126.1.259. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Nguyen DH, Hussaini IM, Gonias SL. Binding of urokinase-type plasminogen activator to its receptor in MCF-7 cells activates extracellular signal-regulated kinase 1 and 2 which is required for increased cellular motility. J Biol Chem 273: 8502–8507, 1998. doi: 10.1074/jbc.273.14.8502. [DOI] [PubMed] [Google Scholar]
  • 80.Konakova M, Hucho F, Schleuning WD. Downstream targets of urokinase-type plasminogen-activator-mediated signal transduction. Eur J Biochem 253: 421–429, 1998. doi: 10.1046/j.1432-1327.1998.2530421.x. [DOI] [PubMed] [Google Scholar]
  • 81.Tang H, Kerins DM, Hao Q, Inagami T, Vaughan DE. The urokinase-type plasminogen activator receptor mediates tyrosine phosphorylation of focal adhesion proteins and activation of mitogen-activated protein kinase in cultured endothelial cells. J Biol Chem 273: 18268–18272, 1998. doi: 10.1074/jbc.273.29.18268. [DOI] [PubMed] [Google Scholar]
  • 82.Resnati M, Guttinger M, Valcamonica S, Sidenius N, Blasi F, Fazioli F. Proteolytic cleavage of the urokinase receptor substitutes for the agonist-induced chemotactic effect. EMBO J 15: 1572–1582, 1996. doi: 10.1002/j.1460-2075.1996.tb00502.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Wei Y, Lukashev M, Simon DI, Bodary SC, Rosenberg S, Doyle MV, Chapman HA. Regulation of integrin function by the urokinase receptor. Science 273: 1551–1555, 1996. doi: 10.1126/science.273.5281.1551. [DOI] [PubMed] [Google Scholar]
  • 84.Resnati M, Pallavicini I, Wang JM, Oppenheim J, Serhan CN, Romano M, Blasi F. The fibrinolytic receptor for urokinase activates the G protein-coupled chemotactic receptor FPRL1/LXA4R. Proc Natl Acad Sci USA 99: 1359–1364, 2002. doi: 10.1073/pnas.022652999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Jo M, Thomas KS, Wu L, Gonias SL. Soluble urokinase-type plasminogen activator receptor inhibits cancer cell growth and invasion by direct urokinase-independent effects on cell signaling. J Biol Chem 278: 46692–46698, 2003. doi: 10.1074/jbc.M308808200. [DOI] [PubMed] [Google Scholar]
  • 86.Gilder AS, Jones KA, Hu J, Wang L, Chen CC, Carter BS, Gonias SL. Soluble urokinase receptor is released selectively by glioblastoma cells that express epidermal growth factor receptor variant III and promotes tumor cell migration and invasion. J Biol Chem 290: 14798–14809, 2015. doi: 10.1074/jbc.M115.637488. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Fazioli F, Resnati M, Sidenius N, Higashimoto Y, Appella E, Blasi F. A urokinase-sensitive region of the human urokinase receptor is responsible for its chemotactic activity. EMBO J 16: 7279–7286, 1997. doi: 10.1093/emboj/16.24.7279. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Rasch MG, Lund IK, Almasi CE, Hoyer-Hansen G. Intact and cleaved uPAR forms: diagnostic and prognostic value in cancer. Front Biosci 13: 6752–6762, 2008. doi: 10.2741/3186. [DOI] [PubMed] [Google Scholar]
  • 89.Thuno M, Macho B, Eugen-Olsen J. suPAR: the molecular crystal ball. Dis Markers 27: 157–172, 2009. doi: 10.3233/DMA-2009-0657. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Ossowski L, Aguirre-Ghiso JA. Urokinase receptor and integrin partnership: coordination of signaling for cell adhesion, migration and growth. Curr Opin Cell Biol 12: 613–620, 2000. doi: 10.1016/s0955-0674(00)00140-x. [DOI] [PubMed] [Google Scholar]
  • 91.Simon DI, Wei Y, Zhang L, Rao NK, Xu H, Chen Z, Liu Q, Rosenberg S, Chapman HA. Identification of a urokinase receptor-integrin interaction site. Promiscuous regulator of integrin function. J Biol Chem 275: 10228–10234, 2000. doi: 10.1074/jbc.275.14.10228. [DOI] [PubMed] [Google Scholar]
  • 92.Aguirre Ghiso JA, Kovalski K, Ossowski L. Tumor dormancy induced by downregulation of urokinase receptor in human carcinoma involves integrin and MAPK signaling. J Cell Biol 147: 89–104, 1999. doi: 10.1083/jcb.147.1.89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Yebra M, Parry GC, Stromblad S, Mackman N, Rosenberg S, Mueller BM, Cheresh DA. Requirement of receptor-bound urokinase-type plasminogen activator for integrin alphavbeta5-directed cell migration. J Biol Chem 271: 29393–29399, 1996. doi: 10.1074/jbc.271.46.29393. [DOI] [PubMed] [Google Scholar]
  • 94.Tarui T, Mazar AP, Cines DB, Takada Y. Urokinase-type plasminogen activator receptor (CD87) is a ligand for integrins and mediates cell-cell interaction. J Biol Chem 276: 3983–3990, 2001. doi: 10.1074/jbc.M008220200. [DOI] [PubMed] [Google Scholar]
  • 95.Liu D, Aguirre Ghiso J, Estrada Y, Ossowski L. EGFR is a transducer of the urokinase receptor initiated signal that is required for in vivo growth of a human carcinoma. Cancer Cell 1: 445–457, 2002. doi: 10.1016/S1535-6108(02)00072-7. [DOI] [PubMed] [Google Scholar]
  • 96.Jo M, Thomas KS, O'Donnell DM, Gonias SL. Epidermal growth factor receptor-dependent and -independent cell-signaling pathways originating from the urokinase receptor. J Biol Chem 278: 1642–1646, 2003. doi: 10.1074/jbc.M210877200. [DOI] [PubMed] [Google Scholar]
  • 97.Alfano D, Franco P, Vocca I, Gambi N, Pisa V, Mancini A, Caputi M, Carriero MV, Iaccarino I, Stoppelli MP. The urokinase plasminogen activator and its receptor: role in cell growth and apoptosis. Thromb Haemost 93: 205–211, 2005. doi: 10.1160/TH04-09-0592. [DOI] [PubMed] [Google Scholar]
  • 98.Ma Z, Webb DJ, Jo M, Gonias SL. Endogenously produced urokinase-type plasminogen activator is a major determinant of the basal level of activated ERK/MAP kinase and prevents apoptosis in MDA-MB-231 breast cancer cells. J Cell Sci 114: 3387–3396, 2001. doi: 10.1242/jcs.114.18.3387. [DOI] [PubMed] [Google Scholar]
  • 99.Koshelnick Y, Ehart M, Hufnagl P, Heinrich PC, Binder BR. Urokinase receptor is associated with the components of the JAK1/STAT1 signaling pathway and leads to activation of this pathway upon receptor clustering in the human kidney epithelial tumor cell line TCL-598. J Biol Chem 272: 28563–28567, 1997. doi: 10.1074/jbc.272.45.28563. [DOI] [PubMed] [Google Scholar]
  • 100.Taga T, Kishimoto T. Gp130 and the interleukin-6 family of cytokines. Annu Rev Immunol 15: 797–819, 1997. doi: 10.1146/annurev.immunol.15.1.797. [DOI] [PubMed] [Google Scholar]
  • 101.Liang OD, Chavakis T, Linder M, Bdeir K, Kuo A, Preissner KT. Binding of urokinase plasminogen activator to gp130 via a putative urokinase-binding consensus sequence. Biol Chem 384: 229–236, 2003. doi: 10.1515/BC.2003.025. [DOI] [PubMed] [Google Scholar]
  • 102.Wei Y, Waltz DA, Rao N, Drummond RJ, Rosenberg S, Chapman HA. Identification of the urokinase receptor as an adhesion receptor for vitronectin. J Biol Chem 269: 32380–32388, 1994. doi: 10.1016/S0021-9258(18)31646-6. [DOI] [PubMed] [Google Scholar]
  • 103.Jo M, Takimoto S, Montel V, Gonias SL. The urokinase receptor promotes cancer metastasis independently of urokinase-type plasminogen activator in mice. Am J Pathol 175: 190–200, 2009. doi: 10.2353/ajpath.2009.081053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Kjoller L, Hall A. Rac mediates cytoskeletal rearrangements and increased cell motility induced by urokinase-type plasminogen activator receptor binding to vitronectin. J Cell Biol 152: 1145–1157, 2001. doi: 10.1083/jcb.152.6.1145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Cheresh DA, Leng J, Klemke RL. Regulation of cell contraction and membrane ruffling by distinct signals in migratory cells. J Cell Biol 146: 1107–1116, 1999. doi: 10.1083/jcb.146.5.1107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Welling TH, Huber TS, Messina LM, Stanley JC. Tissue plasminogen activator increases canine endothelial cell proliferation rate through a plasmin-independent, receptor-mediated mechanism. J Surg Res 66: 36–42, 1996. doi: 10.1006/jsre.1996.0369. [DOI] [PubMed] [Google Scholar]
  • 107.Herbert JM, Lamarche I, Prabonnaud V, Dol F, Gauthier T. Tissue-type plasminogen activator is a potent mitogen for human aortic smooth muscle cells. J Biol Chem 269: 3076–3080, 1994. doi: 10.1016/S0021-9258(17)42049-7. [DOI] [PubMed] [Google Scholar]
  • 108.Siao CJ, Tsirka SE. Tissue plasminogen activator mediates microglial activation via its finger domain through annexin II. J Neurosci 22: 3352–3358, 2002. doi: 10.1523/JNEUROSCI.22-09-03352.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Bock A, Tucker N, Kelher MR, Khan SY, Gonzalez E, Wohlauer M, Hansen K, Dzieciatkowska M, Sauaia A, Banerjee A, Moore EE, Silliman CC. Alpha-enolase causes proinflammatory activation of pulmonary microvascular endothelial cells and primes neutrophils through plasmin activation of protease-activated receptor 2. Shock 44: 137–142, 2015. doi: 10.1097/SHK.0000000000000394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Carmo AA, Costa BR, Vago JP, de Oliveira LC, Tavares LP, Nogueira CR, Ribeiro AL, Garcia CC, Barbosa AS, Brasil BS, Dusse LM, Barcelos LS, Bonjardim CA, Teixeira MM, Sousa LP. Plasmin induces in vivo monocyte recruitment through protease-activated receptor-1-, MEK/ERK-, and CCR2-mediated signaling. J Immunol 193: 3654–3663, 2014. doi: 10.4049/jimmunol.1400334. [DOI] [PubMed] [Google Scholar]
  • 111.Nagai T, Ito M, Nakamichi N, Mizoguchi H, Kamei H, Fukakusa A, Nabeshima T, Takuma K, Yamada K. The rewards of nicotine: regulation by tissue plasminogen activator-plasmin system through protease activated receptor-1. J Neurosci 26: 12374–12383, 2006. doi: 10.1523/JNEUROSCI.3139-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Zalfa C, Azmoon P, Mantuano E, Gonias SL. Tissue-type plasminogen activator neutralizes LPS but not protease-activated receptor-mediated inflammatory responses to plasmin. J Leukoc Biol 105: 729–740, 2019. doi: 10.1002/JLB.3A0818-329RRR. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Herz J, Strickland DK. LRP: a multifunctional scavenger and signaling receptor. J Clin Invest 108: 779–784, 2001. doi: 10.1172/JCI13992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Gonias SL, Campana WM. LDL receptor-related protein-1: a regulator of inflammation in atherosclerosis, cancer, and injury to the nervous system. Am J Pathol 184: 18–27, 2014. doi: 10.1016/j.ajpath.2013.08.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Bu G, Williams S, Strickland DK, Schwartz AL. Low density lipoprotein receptor-related protein/alpha 2-macroglobulin receptor is an hepatic receptor for tissue-type plasminogen activator. Proc Natl Acad Sci USA 89: 7427–7431, 1992. doi: 10.1073/pnas.89.16.7427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Warshawsky I, Bu G, Schwartz AL. 39-kD protein inhibits tissue-type plasminogen activator clearance in vivo. J Clin Invest 92: 937–944, 1993. doi: 10.1172/JCI116669. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Trommsdorff M, Borg JP, Margolis B, Herz J. Interaction of cytosolic adaptor proteins with neuronal apolipoprotein E receptors and the amyloid precursor protein. J Biol Chem 273: 33556–33560, 1998. doi: 10.1074/jbc.273.50.33556. [DOI] [PubMed] [Google Scholar]
  • 118.Ranganathan S, Liu CX, Migliorini MM, Von Arnim CA, Peltan ID, Mikhailenko I, Hyman BT, Strickland DK. Serine and threonine phosphorylation of the low density lipoprotein receptor-related protein by protein kinase Calpha regulates endocytosis and association with adaptor molecules. J Biol Chem 279: 40536–40544, 2004. doi: 10.1074/jbc.M407592200. [DOI] [PubMed] [Google Scholar]
  • 119.Luo L, Wall AA, Tong SJ, Hung Y, Xiao Z, Tarique AA, Sly PD, Fantino E, Marzolo MP, Stow JL. TLR crosstalk activates LRP1 to recruit Rab8a and PI3Kgamma for suppression of inflammatory responses. Cell Rep 24: 3033–3044, 2018. doi: 10.1016/j.celrep.2018.08.028. [DOI] [PubMed] [Google Scholar]
  • 120.Wang X, Lee SR, Arai K, Lee SR, Tsuji K, Rebeck GW, Lo EH. Lipoprotein receptor-mediated induction of matrix metalloproteinase by tissue plasminogen activator. Nat Med 9: 1313–1317, 2003. doi: 10.1038/nm926. [DOI] [PubMed] [Google Scholar]
  • 121.Hu K, Yang J, Tanaka S, Gonias SL, Mars WM, Liu Y. Tissue-type plasminogen activator acts as a cytokine that triggers intracellular signal transduction and induces matrix metalloproteinase-9 gene expression. J Biol Chem 281: 2120–2127, 2006. doi: 10.1074/jbc.M504988200. [DOI] [PubMed] [Google Scholar]
  • 122.Yepes M, Sandkvist M, Moore EG, Bugge TH, Strickland DK, Lawrence DA. Tissue-type plasminogen activator induces opening of the blood-brain barrier via the LDL receptor-related protein. J Clin Invest 112: 1533–1540, 2003. doi: 10.1172/JCI19212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Zhuo M, Holtzman DM, Li Y, Osaka H, DeMaro J, Jacquin M, Bu G. Role of tissue plasminogen activator receptor LRP in hippocampal long-term potentiation. J Neurosci 20: 542–549, 2000. doi: 10.1523/JNEUROSCI.20-02-00542.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Hu K, Lin L, Tan X, Yang J, Bu G, Mars WM, Liu Y. tPA protects renal interstitial fibroblasts and myofibroblasts from apoptosis. J Am Soc Nephrol 19: 503–514, 2008. doi: 10.1681/ASN.2007030300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Lin L, Jin Y, Hu K. Tissue-type plasminogen activator (tPA) promotes M1 macrophage survival through p90 ribosomal S6 kinase (RSK) and p38 mitogen-activated protein kinase (MAPK) pathway. J Biol Chem 290: 7910–7917, 2015. doi: 10.1074/jbc.M114.599688. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Yepes M. Tissue-type plasminogen activator is a neuroprotectant in the central nervous system. Front Cell Neurosci 9: 304, 2015. doi: 10.3389/fncel.2015.00304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Mantuano E, Lam MS, Gonias SL. LRP1 assembles unique co-receptor systems to initiate cell signaling in response to tissue-type plasminogen activator and myelin-associated glycoprotein. J Biol Chem 288: 34009–34018, 2013. doi: 10.1074/jbc.M113.509133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Shi Y, Mantuano E, Inoue G, Campana WM, Gonias SL. Ligand binding to LRP1 transactivates Trk receptors by a Src family kinase-dependent pathway. Sci Signal 2: ra18, 2009. doi: 10.1126/scisignal.2000188. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Williams SE, Ashcom JD, Argraves WS, Strickland DK. A novel mechanism for controlling the activity of alpha 2-macroglobulin receptor/low density lipoprotein receptor-related protein. Multiple regulatory sites for 39-kDa receptor-associated protein. J Biol Chem 267: 9035–9040, 1992. doi: 10.1016/S0021-9258(19)50384-2. [DOI] [PubMed] [Google Scholar]
  • 130.Mantuano E, Brifault C, Lam MS, Azmoon P, Gilder AS, Gonias SL. LDL receptor-related protein-1 regulates NFkappaB and microRNA-155 in macrophages to control the inflammatory response. Proc Natl Acad Sci USA 113: 1369–1374, 2016. doi: 10.1073/pnas.1515480113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Lillis AP, Van Duyn LB, Murphy-Ullrich JE, Strickland DK. LDL receptor-related protein 1: unique tissue-specific functions revealed by selective gene knockout studies. Physiol Rev 88: 887–918, 2008. doi: 10.1152/physrev.00033.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Nicole O, Docagne F, Ali C, Margaill I, Carmeliet P, MacKenzie ET, Vivien D, Buisson A. The proteolytic activity of tissue-plasminogen activator enhances NMDA receptor-mediated signaling. Nat Med 7: 59–64, 2001. doi: 10.1038/83358. [DOI] [PubMed] [Google Scholar]
  • 133.Bliss TV, Collingridge GL. A synaptic model of memory: long-term potentiation in the hippocampus. Nature 361: 31–39, 1993. doi: 10.1038/361031a0. [DOI] [PubMed] [Google Scholar]
  • 134.Mantuano E, Azmoon P, Brifault C, Banki MA, Gilder AS, Campana WM, Gonias SL. Tissue-type plasminogen activator regulates macrophage activation and innate immunity. Blood 130: 1364–1374, 2017. doi: 10.1182/blood-2017-04-780205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Mantuano E, Lam MS, Shibayama M, Campana WM, Gonias SL. The NMDA receptor functions independently and as an LRP1 co-receptor to promote Schwann cell survival and migration. J Cell Sci 128: 3478–3488, 2015. doi: 10.1242/jcs.173765. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Fernandez-Monreal M, Lopez-Atalaya JP, Benchenane K, Cacquevel M, Dulin F, Le Caer JP, Rossier J, Jarrige AC, Mackenzie ET, Colloc'h N, Ali C, Vivien D. Arginine 260 of the amino-terminal domain of NR1 subunit is critical for tissue-type plasminogen activator-mediated enhancement of N-methyl-d-aspartate receptor signaling. J Biol Chem 279: 50850–50856, 2004. doi: 10.1074/jbc.M407069200. [DOI] [PubMed] [Google Scholar]
  • 137.Lesept F, Chevilley A, Jezequel J, Ladepeche L, Macrez R, Aimable M, Lenoir S, Bertrand T, Rubrecht L, Galea P, Lebouvier L, Petersen KU, Hommet Y, Maubert E, Ali C, Groc L, Vivien D. Tissue-type plasminogen activator controls neuronal death by raising surface dynamics of extrasynaptic NMDA receptors. Cell Death Dis 7: e2466, 2016. doi: 10.1038/cddis.2016.279. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Macrez R, Bezin L, Le Mauff B, Ali C, Vivien D. Functional occurrence of the interaction of tissue plasminogen activator with the NR1 Subunit of N-methyl-d-aspartate receptors during stroke. Stroke 41: 2950–2955, 2010. doi: 10.1161/STROKEAHA.110.592360. [DOI] [PubMed] [Google Scholar]
  • 139.Ng KS, Leung HW, Wong PT, Low CM. Cleavage of the NR2B subunit amino terminus of N-methyl-d-aspartate (NMDA) receptor by tissue plasminogen activator: identification of the cleavage site and characterization of ifenprodil and glycine affinities on truncated NMDA receptor. J Biol Chem 287: 25520–25529, 2012. doi: 10.1074/jbc.M112.374397. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Chevilley A, Lesept F, Lenoir S, Ali C, Parcq J, Vivien D. Impacts of tissue-type plasminogen activator (tPA) on neuronal survival. Front Cell Neurosci 9: 415, 2015. doi: 10.3389/fncel.2015.00415. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Jullienne A, Montagne A, Orset C, Lesept F, Jane DE, Monaghan DT, Maubert E, Vivien D, Ali C. Selective inhibition of GluN2D-containing N-methyl-d-aspartate receptors prevents tissue plasminogen activator-promoted neurotoxicity both in vitro and in vivo. Mol Neurodegener 6: 68, 2011. doi: 10.1186/1750-1326-6-68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Norris EH, Strickland S. Modulation of NR2B-regulated contextual fear in the hippocampus by the tissue plasminogen activator system. Proc Natl Acad Sci USA 104: 13473–13478, 2007. doi: 10.1073/pnas.0705848104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Martin AM, Kuhlmann C, Trossbach S, Jaeger S, Waldron E, Roebroek A, Luhmann HJ, Laatsch A, Weggen S, Lessmann V, Pietrzik CU. The functional role of the second NPXY motif of the LRP1 beta-chain in tissue-type plasminogen activator-mediated activation of N-methyl-d-aspartate receptors. J Biol Chem 283: 12004–12013, 2008. doi: 10.1074/jbc.M707607200. [DOI] [PubMed] [Google Scholar]
  • 144.Samson AL, Nevin ST, Croucher D, Niego B, Daniel PB, Weiss TW, Moreno E, Monard D, Lawrence DA, Medcalf RL. Tissue-type plasminogen activator requires a co-receptor to enhance NMDA receptor function. J Neurochem 107: 1091–1101, 2008. doi: 10.1111/j.1471-4159.2008.05687.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Thomas SM, Brugge JS. Cellular functions regulated by Src family kinases. Annu Rev Cell Dev Biol 13: 513–609, 1997. doi: 10.1146/annurev.cellbio.13.1.513. [DOI] [PubMed] [Google Scholar]
  • 146.Wetzker R, Bohmer FD. Transactivation joins multiple tracks to the ERK/MAPK cascade. Nat Rev Mol Cell Biol 4: 651–657, 2003. doi: 10.1038/nrm1173. [DOI] [PubMed] [Google Scholar]
  • 147.Mattei V, Manganelli V, Martellucci S, Capozzi A, Mantuano E, Longo A, Ferri A, Garofalo T, Sorice M, Misasi R. A multimolecular signaling complex including PrP(C) and LRP1 is strictly dependent on lipid rafts and is essential for the function of tissue plasminogen activator. J Neurochem 152: 468–481, 2020. doi: 10.1111/jnc.14891. [DOI] [PubMed] [Google Scholar]
  • 148.Ortiz-Zapater E, Peiro S, Roda O, Corominas JM, Aguilar S, Ampurdanes C, Real FX, Navarro P. Tissue plasminogen activator induces pancreatic cancer cell proliferation by a non-catalytic mechanism that requires extracellular signal-regulated kinase 1/2 activation through epidermal growth factor receptor and annexin A2. Am J Pathol 170: 1573–1584, 2007. doi: 10.2353/ajpath.2007.060850. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Akira S, Uematsu S, Takeuchi O. Pathogen recognition and innate immunity. Cell 124: 783–801, 2006. doi: 10.1016/j.cell.2006.02.015. [DOI] [PubMed] [Google Scholar]
  • 150.Medzhitov R. Toll-like receptors and innate immunity. Nat Rev Immunol 1: 135–145, 2001. doi: 10.1038/35100529. [DOI] [PubMed] [Google Scholar]
  • 151.Shaw AC, Goldstein DR, Montgomery RR. Age-dependent dysregulation of innate immunity. Nat Rev Immunol 13: 875–887, 2013. doi: 10.1038/nri3547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Mazar AP. The urokinase plasminogen activator receptor (uPAR) as a target for the diagnosis and therapy of cancer. Anticancer Drugs 12: 387–400, 2001. doi: 10.1097/00001813-200106000-00001. [DOI] [PubMed] [Google Scholar]
  • 153.Gonias SL, Hu J. Urokinase receptor and resistance to targeted anticancer agents. Front Pharmacol 6: 154, 2015. doi: 10.3389/fphar.2015.00154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Plesner T, Behrendt N, Ploug M. Structure, function and expression on blood and bone marrow cells of the urokinase-type plasminogen activator receptor, uPAR. Stem Cells 15: 398–408, 1997. doi: 10.1002/stem.150398. [DOI] [PubMed] [Google Scholar]
  • 155.Mondino A, Blasi F. uPA and uPAR in fibrinolysis, immunity and pathology. Trends Immunol 25: 450–455, 2004. doi: 10.1016/j.it.2004.06.004. [DOI] [PubMed] [Google Scholar]
  • 156.Plesner T, Ralfkiaer E, Wittrup M, Johnsen H, Pyke C, Pedersen TL, Hansen NE, Dano K. Expression of the receptor for urokinase-type plasminogen activator in normal and neoplastic blood cells and hematopoietic tissue. Am J Clin Pathol 102: 835–841, 1994. doi: 10.1093/ajcp/102.6.835. [DOI] [PubMed] [Google Scholar]
  • 157.Nykjaer A, Moller B, Todd RF 3rd, Christensen T, Andreasen PA, Gliemann J, Petersen CM. Urokinase receptor. An activation antigen in human T lymphocytes. J Immunol 152: 505–516, 1994. [PubMed] [Google Scholar]
  • 158.Gyetko MR, Todd RF 3rd, Wilkinson CC, Sitrin RG. The urokinase receptor is required for human monocyte chemotaxis in vitro. J Clin Invest 93: 1380–1387, 1994. doi: 10.1172/JCI117114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Gyetko MR, Sud S, Kendall T, Fuller JA, Newstead MW, Standiford TJ. Urokinase receptor-deficient mice have impaired neutrophil recruitment in response to pulmonary Pseudomonas aeruginosa infection. J Immunol 165: 1513–1519, 2000. doi: 10.4049/jimmunol.165.3.1513. [DOI] [PubMed] [Google Scholar]
  • 160.Dekkers PE, ten Hove T, Te Velde AA, van Deventer SJ, van Der Poll T. Upregulation of monocyte urokinase plasminogen activator receptor during human endotoxemia. Infect Immun 68: 2156–2160, 2000. doi: 10.1128/IAI.68.4.2156-2160.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Coleman JL, Benach JL. The urokinase receptor can be induced by Borrelia burgdorferi through receptors of the innate immune system. Infect Immun 71: 5556–5564, 2003. doi: 10.1128/IAI.71.10.5556-5564.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Kiyan Y, Tkachuk S, Rong S, Gorrasi A, Ragno P, Dumler I, Haller H, Shushakova N. TLR4 response to LPS is reinforced by urokinase receptor. Front Immunol 11: 573550, 2020. doi: 10.3389/fimmu.2020.573550. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Wei C, Moller CC, Altintas MM, Li J, Schwarz K, Zacchigna S, Xie L, Henger A, Schmid H, Rastaldi MP, Cowan P, Kretzler M, Parrilla R, Bendayan M, Gupta V, Nikolic B, Kalluri R, Carmeliet P, Mundel P, Reiser J. Modification of kidney barrier function by the urokinase receptor. Nat Med 14: 55–63, 2008. doi: 10.1038/nm1696. [DOI] [PubMed] [Google Scholar]
  • 164.Kwak SH, Mitra S, Bdeir K, Strassheim D, Park JS, Kim JY, Idell S, Cines D, Abraham E. The kringle domain of urokinase-type plasminogen activator potentiates LPS-induced neutrophil activation through interaction with {alpha}V{beta}3 integrins. J Leukoc Biol 78: 937–945, 2005. doi: 10.1189/jlb.0305158. [DOI] [PubMed] [Google Scholar]
  • 165.Liu G, Yang Y, Yang S, Banerjee S, De Freitas A, Friggeri A, Davis KI, Abraham E. The receptor for urokinase regulates TLR2 mediated inflammatory responses in neutrophils. PLoS One 6: e25843, 2011. doi: 10.1371/journal.pone.0025843. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Genua M, D'Alessio S, Cibella J, Gandelli A, Sala E, Correale C, Spinelli A, Arena V, Malesci A, Rutella S, Ploplis VA, Vetrano S, Danese S. The urokinase plasminogen activator receptor (uPAR) controls macrophage phagocytosis in intestinal inflammation. Gut 64: 589–600, 2015. [Erratum in Gut 64: 1465, 2015]. doi: 10.1136/gutjnl-2013-305933. [DOI] [PubMed] [Google Scholar]
  • 167.Overton CD, Yancey PG, Major AS, Linton MF, Fazio S. Deletion of macrophage LDL receptor-related protein increases atherogenesis in the mouse. Circ Res 100: 670–677, 2007. doi: 10.1161/01.RES.0000260204.40510.aa. [DOI] [PubMed] [Google Scholar]
  • 168.Staudt ND, Jo M, Hu J, Bristow JM, Pizzo DP, Gaultier A, VandenBerg SR, Gonias SL. Myeloid cell receptor LRP1/CD91 regulates monocyte recruitment and angiogenesis in tumors. Cancer Res 73: 3902–3912, 2013. doi: 10.1158/0008-5472.CAN-12-4233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.May P, Bock HH, Nofer JR. Low density receptor-related protein 1 (LRP1) promotes anti-inflammatory phenotype in murine macrophages. Cell Tissue Res 354: 887–889, 2013. doi: 10.1007/s00441-013-1699-2. [DOI] [PubMed] [Google Scholar]
  • 170.Gaultier A, Arandjelovic S, Niessen S, Overton CD, Linton MF, Fazio S, Campana WM, Cravatt BF III, Gonias SL. Regulation of tumor necrosis factor receptor-1 and the IKK-NF-kappaB pathway by LDL receptor-related protein explains the antiinflammatory activity of this receptor. Blood 111: 5316–5325, 2008. doi: 10.1182/blood-2007-12-127613. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Fernandez-Castaneda A, Arandjelovic S, Stiles TL, Schlobach RK, Mowen KA, Gonias SL, Gaultier A. Identification of the low density lipoprotein (LDL) receptor-related protein-1 interactome in central nervous system myelin suggests a role in the clearance of necrotic cell debris. J Biol Chem 288: 4538–4548, 2013. doi: 10.1074/jbc.M112.384693. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Strickland DK, Gonias SL, Argraves WS. Diverse roles for the LDL receptor family. Trends Endocrinol Metab 13: 66–74, 2002. doi: 10.1016/s1043-2760(01)00526-4. [DOI] [PubMed] [Google Scholar]
  • 173.Das L, Azmoon P, Banki MA, Mantuano E, Gonias SL. Tissue-type plasminogen activator selectively inhibits multiple toll-like receptors in CSF-1-differentiated macrophages. PLoS One 14: e0224738, 2019. doi: 10.1371/journal.pone.0224738. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Bacskai BJ, Xia MQ, Strickland DK, Rebeck GW, Hyman BT. The endocytic receptor protein LRP also mediates neuronal calcium signaling via N-methyl-d-aspartate receptors. Proc Natl Acad Sci USA 97: 11551–11556, 2000. doi: 10.1073/pnas.200238297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Perse M, Cerar A. Dextran sodium sulphate colitis mouse model: traps and tricks. J Biomed Biotechnol 2012: 718617, 2012. doi: 10.1155/2012/718617. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Cooper HS, Murthy SN, Shah RS, Sedergran DJ. Clinicopathologic study of dextran sulfate sodium experimental murine colitis. Lab Invest 69: 238–249, 1993. doi: 10.1016/S0021-5198(19)41298-5. [DOI] [PubMed] [Google Scholar]
  • 177.Fukata M, Michelsen KS, Eri R, Thomas LS, Hu B, Lukasek K, Nast CC, Lechago J, Xu R, Naiki Y, Soliman A, Arditi M, Abreu MT. Toll-like receptor-4 is required for intestinal response to epithelial injury and limiting bacterial translocation in a murine model of acute colitis. Am J Physiol Gastrointest Liver Physiol 288: G1055–G1065, 2005. doi: 10.1152/ajpgi.00328.2004. [DOI] [PubMed] [Google Scholar]
  • 178.Kiesler P, Fuss IJ, Strober W. Experimental models of inflammatory bowel diseases. Cell Mol Gastroenterol Hepatol 1: 154–170, 2015. doi: 10.1016/j.jcmgh.2015.01.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Das L, Banki MA, Azmoon P, Pizzo D, Gonias SL. Enzymatically inactive tissue-type plasminogen activator reverses disease progression in the dextran sulfate sodium mouse model of inflammatory bowel disease. Am J Pathol 191: 590–601, 2021. doi: 10.1016/j.ajpath.2021.01.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Kaneda MM, Messer KS, Ralainirina N, Li H, Leem CJ, Gorjestani S, Woo G, Nguyen AV, Figueiredo CC, Foubert P, Schmid MC, Pink M, Winkler DG, Rausch M, Palombella VJ, Kutok J, McGovern K, Frazer KA, Wu X, Karin M, Sasik R, Cohen EE, Varner JA. PI3Kgamma is a molecular switch that controls immune suppression. Nature 539: 437–442, 2016. doi: 10.1038/nature19834. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Perkinton MS, Ip JK, Wood GL, Crossthwaite AJ, Williams RJ. Phosphatidylinositol 3-kinase is a central mediator of NMDA receptor signalling to MAP kinase (Erk1/2), Akt/PKB and CREB in striatal neurones. J Neurochem 80: 239–254, 2002. doi: 10.1046/j.0022-3042.2001.00699.x. [DOI] [PubMed] [Google Scholar]
  • 182.De Taeye B, Smith LH, Vaughan DE. Plasminogen activator inhibitor-1: a common denominator in obesity, diabetes and cardiovascular disease. Curr Opin Pharmacol 5: 149–154, 2005. doi: 10.1016/j.coph.2005.01.007. [DOI] [PubMed] [Google Scholar]
  • 183.Devaraj S, Xu DY, Jialal I. C-reactive protein increases plasminogen activator inhibitor-1 expression and activity in human aortic endothelial cells: implications for the metabolic syndrome and atherothrombosis. Circulation 107: 398–404, 2003. doi: 10.1161/01.cir.0000052617.91920.fd. [DOI] [PubMed] [Google Scholar]
  • 184.Alessi MC, Juhan-Vague I. PAI-1 and the metabolic syndrome: links, causes, and consequences. Arterioscler Thromb Vasc Biol 26: 2200–2207, 2006. doi: 10.1161/01.ATV.0000242905.41404.68. [DOI] [PubMed] [Google Scholar]
  • 185.Gonias SL, Banki MA, Gilder AS, Azmoon P, Campana WM, Mantuano E. PAI1 blocks NMDA receptor-mediated effects of tissue-type plasminogen activator on cell signaling and physiology. J Cell Sci 131: jcs217083, 2018. doi: 10.1242/jcs.217083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Stefansson S, Muhammad S, Cheng XF, Battey FD, Strickland DK, Lawrence DA. Plasminogen activator inhibitor-1 contains a cryptic high affinity binding site for the low density lipoprotein receptor-related protein. J Biol Chem 273: 6358–6366, 1998. doi: 10.1074/jbc.273.11.6358. [DOI] [PubMed] [Google Scholar]
  • 187.Shiga Y, Shiga A, Mesci P, Kwon H, Brifault C, Kim JH, Jeziorski JJ, Nasamran C, Ohtori S, Muotri AR, Gonias SL, Campana WM. Tissue-type plasminogen activator-primed human iPSC-derived neural progenitor cells promote motor recovery after severe spinal cord injury. Sci Rep 9: 19291, 2019. doi: 10.1038/s41598-019-55132-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Beck KD, Nguyen HX, Galvan MD, Salazar DL, Woodruff TM, Anderson AJ. Quantitative analysis of cellular inflammation after traumatic spinal cord injury: evidence for a multiphasic inflammatory response in the acute to chronic environment. Brain 133: 433–447, 2010. doi: 10.1093/brain/awp322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Fuchs HE, Berger H Jr, Pizzo SV. Catabolism of human tissue plasminogen activator in mice. Blood 65: 539–544, 1985. doi: 10.1182/blood.V65.3.539.bloodjournal653539. [DOI] [PubMed] [Google Scholar]
  • 190.Sallenave JM, Guillot L. Innate immune signaling and proteolytic pathways in the resolution or exacerbation of SARS-CoV-2 in Covid-19: key therapeutic targets? Front Immunol 11: 1229, 2020. doi: 10.3389/fimmu.2020.01229. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from American Journal of Physiology - Cell Physiology are provided here courtesy of American Physiological Society

RESOURCES