CANCER METASTASIS: BIOLOGICAL AND CLINICAL ASPECTS
THE ROLE OF THE PLASMINOGEN ACTIVATION SYSTEM IN ANGIOGENESIS AND METASTASIS Shafaat A. Rabbani, MD, and Andrew P. Mazar, PhD
Tumors grow and disseminate by recruiting and forming new blood supplies through the process of tumor-mediated angiogenesi~.~~ Although recent evidence suggests that tumor cells also form blood-carrying structures that resemble vessels and may be able to compensate for a lack of angiogene~is;~there is still a significantbody of data that supports the requirement for true neovessel formation for tumor progression. Numerous preclinical animal models have demonstrated the use of antiangiogenic approaches in suppressing the growth and metastasis of tumors30 and some antiangiogenic compounds with antiangiogenic activity in preclinical models have begun to demonstrate similar activity in human clinical trials.137 Angiogenesis is a prerequisite for the hematogenous dissemination of tumor cells. Although primary metastasis can occur through the lymphatic system, the most predominant mode of tumor cell egress is intra~asation.~~ Thus, agents that inhibit angiogenesis may also inhibit metastasis. Further, metastasis and angiogenesis share many common phenotypic features that lead to the motility of tumor cells (in the case of metastasis) and endothelial cells (in the case of angiogenesis). These include the upregulation of protease and integrin expression, loss of cell-cell and cell-matrix contacts, an increase in responsiveness to growth and differentiation factors and the remodeling of extracellular matrix (ECM) and basement membrane This work was supported by Grant MT-16209 to SAR from the Medical Research Council of Canada.
From the Departments of Oncology and Physiology, McGill University Health Centre, Montreal, Quebec, Canada (SAR); and Attenuon, LLC, San Diego, California (APM)
SURGICAL ONCOLOGY CLINICS OF NORTH AMERICA -
-
-
-
VOLUME 10. NUMBER 2 a APRIL 2001
-
-
-
-
393
394
RABBANI & MAZAR
( ~ ~ ) . 784,114 2 . jyumerous molecules have been implicated in these processes and many of these molecules have provided the basis for targeted therapeutic and diagnostic approaches for the treatment and detection of ~ a n c e r . ' ~ , ~ ~ The authors' research over the past 11 years has focused on the role of the urokinase plasminogen activator (uPA) system in metastasis and angiogenesis, a system that the authors believe to be central to many of the cascades that have been demonstrated to be important in tumor progression (Fig. l).The uPA system, apart from its ability to promote extracellular fibrinolysis, plays an important role in initiating cascades that result in the activation of plasminogen,l12several matrix metalloproteases (MMPs),4O,73 the release and processing of latent growth factors,69, 92, lo6and the remodeling
Plasminogen
Piasmin
Extravascuiar Growth Factor Matrix proMMP fibrinolysis activation remodeling activation
Angiogenesis Metastasis Mitogenesis Figure 1. Model of the role of urokinase (uPA) system in tumor progression. uPA is localized within the tumor cell environment by binding to uPA receptor (uPAR) to promote activation of plasmin, matrix metalloproteases (MMPs), and growth factors resulting in increased angiogenesis, tumor metastases, and mitogenesis.
THE ROLE OF THE PLASMlNOGEN ACTIVATION SYSTEM IN ANGIOGENESIS
395
of BM and ECM.62,91 Further, the uPA system can also initiate localized fibrin turnover and this may contribute to the role of the uPA system in angiogenesis. Although the proteolytic activity of uPA has long been accepted as being important in the degradation of ECM, it is only recently that the nonproteolytic activities of uPA and its receptor, uPAR, have become recognized for their role in cell motility, including the migration of endothelial cells and the invasion of tumor cells. In this article, the authors summarize recent data on the integration of the various activities of the uPA system and how these contribute to tumor metastasis and angiogenesis. uPA-DEPENDENT PROTEOLYSIS
uPA is a serine protease that is highly specific for the activation of plasminogen. It is secreted in a proenzyme or single chain uPA (scuPA). ScuPA can be activated proteolytically by plasmin (to form catalyticallyactive uPA) or by several other protease such as cathepsins B and L, plasma kallikrein, and mast cell t r y p t a ~ e . ~ ~Physiologically, ,~~, plasmin is the most efficient activator of scuPA although other enzymes could potentially activate scuPA physiologically in the absence of plasmin. ScuPA also forms a catalytically active moiety in the absence of proteolytic activation by plasmin when it is bound to uPAR either on the monocyte cell surface70 or in solution.44This process may require the presence of certain c o f a ~ t o r and s~~ the physiologic relevance of this nonproteolytic activation is not presently understood. ScuPA bound to cell-surface uPAR is activated by plasmin much more efficientlythan scuPA in solution and thus, one role for suPAR in mediating cell-surface proteolysis is to potentiate the activation of SCUPA.~~ Plasminogen and plasmin can also bind to the cell surfaces of numerous cell types including tumor cells and thus scuPA/uPA and plasminogen/plasmin can exert their proteolytic effects while anchored to the cell surface. Because uPAR is often polarized and localized to the migrating or invading edge of a cell, uPA-dependent cascades are focally directed in the direction of cell movement rather than being uniformly activated all over the cell surface. Once plasminogen is activated to plasmin, several divergent pathways are initiated. Plasmin itself can cleave matrix componentssuch as fibrin and fibronectin.'j7, In addition, plasmin can activate several proMMPs (collagenases) such as proMMP2 and proMMP9 either alone or in combination with the activity of membrane-type (MT) MMPs such as MT-MMP1.40, 73, 82 This extends the activity of the pathway activated by uPA to the remodeling of type IV collagen and other matrix components through activated collagenases (MMPs).Type IV collagen is the major constituent of the basement membrane and is remodeled during angiogenesis.'05Plasmin can also process angiogenic growth factors such as vascular endothelial growth factor (VEGF)56or release latent forms of other growth factors (such as fibroblast growth factor 2 (FGF-2) and transforming growth factor B (TGFB))from
396
RABBANI & MAZAR
their binding sites in the matrix.92,'06 These growth factors can also function in a paracrine manner to regulate the expression of the uPA system, an aspect that is discussed in the section on the activation and release of growth factors. THE uPA SYSTEM IN MIGRATION, INVASION, AND ANGIOGENESIS
The exvression of uPA and uPAR has been demonstrated in numerous tumor types including prostate, breast, colon, glioblastoma, hepatocellular, The ~ ,levels ~ ~ , ~of~uPA and uPAR are typically inand renal cell c a r c i n ~ m a . ~ creased in more aggressive forms of disease and are often associated with ~ , ~ ~ ~recent studies have demonthe invasive front of the t ~ m o r . ' Several strated strong immunohistochemical staining for uPAR in blood vessels associated with the invasive front of breast (Fig. 2), colon, and renal cell c a r ~ i n o m a . ~Because , ~ ~ , ~angiogenesis ~~ most likely occurs at the tumorhost interface, these results implicate uPAR in the angiogenic process. High levels of uPA in breast cancer patients correlates with poor prognosis and this has also been observed in other cancer types such as head and neck and colon cancer.18, The expression of uPA and uPAR has also been observed on tumor-associated macrophages in several tumor types.23,55 uPA is chemotactic for monocytes and mediates adhesion and migration of these cells.31Adhesion and migration require only uPAR occupancy in the absence of uPA catalytic activity. uPAR occupancy, however, stimulates differential effects depending on the differentiation state of the monocytic Thus, less differentiated, more monocyte-like cells are stimulated to migrate in response to uPA binding (as might occur if a histiocyte was exposed to a uPA gradient in the region of a tumor) whereas a more differentiated macrophageslike cell might be induced to adhere in the presence of uPA (as might occur when the cell has reached the tumor). The role of macrophages in tumor progression has been controversial although recent evidence from patient biopsies demonstrates that macrophages are associated with areas of neovascularization in the tumor.46Thus, the uPA system may stimulate tumor angiogenesis by recruiting the primary inflammatory response to the tumor. Hildenbrand et a1 have demonstrated that only tumor-associated macrophages cultured from invasive breast carcinomas or ductal carcinoma in situ (DCIS) expressed high levels of uPAR whereas resident macrophages from normal breast tissue did not.47 Macrophages are known to secrete angiogenic factors such as VEGF, FGF-2 and interleukin-8 (IL-8) and regions of inflammation have been demonstrated to preferentially support tumor growth in model systems.47It is not clear why the inflammatory response does not lead to an immune response against the tumor but one hypothesis may be that tumors have evolved in such a way as to uncouple inflammation, which can promote tumor progression, from the immune response. Thus, the uPA system may initiate tumor angiogenesis by regulating the inflammatory response in tumors.
THE ROLE OF THE PLASMINOGEN ACTNATION SYSTEM IN ANGIOGENESIS
397
Figure 2. lmmunolocalization of uPAR in breast cancer. Normal (A) and malignant human tissue sections were stained with rabbit antihuman uPAR IgG to localize breast cancer (8) uPAR (original magnification x 400).
In addition to monocyte and macrophage chemotaxis, the uPA system plays a role in the migration and invasion of endothelial cells during angiogenesis. One of the major results of localized inflammatory response is the stimulation of vessel leaking by VEGF, which is secreted by macrophages and platelets that adhere in regions of inflammati~n.~~? This allows fibrinogen (and other plasma proteins) to diffuse into the extravascular space where it is converted to fibrin through the action of tissue factor (TF)-mediated clotting pathways.75Fibrin forms the transitional matrix on which endothelial cells migrate during angiogene~is.~~ uPA and uPAR have been localized to the invading, tube-forming front of microvessel endothelial cells in a fibrin matrix in ~ i t r o . ~ ~
398
RABBANI & MAZAR
The role of uPA in the angiogenic process was observed initially in models of corneal vasc~larization~~ and recent studies have extended this early work to implicate uPAR and uPA in the activities associated with neovascularization. Endothelial cells engineered to over-express uPA are more invasive than parental cells in ~ i t r oEndothelial .~~ cells can be induced to migrate in vitro by various proangiogenic factors such as basic FGF (bFGF) and VEGF. These factors upregulate uPA and uPAR in these cells.21,71 Hypoxia, which may stimulate tumor neovascularizationin vivo, also regulates uPAR expression in human umbilical vein endothelial cells (HUVEC) in vitro and this process appears to be mediated by a heme protein-dependent pathway.36An antagonist of uPA binding to uPAR has been demonstrated to inhibit endothelial cell branching morphogenesis in vitron and inhibitors of uPA expression in endothelial cells (such as testosterone and dexamethasone) also inhibit tube f ~ r m a t i o nThis . ~ ~ inhibition by steroid hormones could be reversed by the addition of exogenous uPA to the model system. In vivo evidence also supports a role for the hormonal regulation of angiogenesis. This has important implications for the growth of hormone-dependent tumors, many of which eventually become hormone independent after hormone ablation therapy. uPA and uPAR expression and tumor aggressiveness also increase as a tumor becomes hormone independent.loO Shionogicarcinoma cells (grownin a dorsal skin-fold chamber in SCID mice) depend on androgen for survival. Castration of the SCID hosts leads to tumor cell apoptosis, which is preceded by rarefaction of tumor-associated blood vessels.53After several weeks, however, a second wave of angiogenesis and tumor growth is observed in the castrated animals. An increase in uPA and uPAR levels in hormone-indevendent tumors may be one of the mechanisms responsible for relapse af
THE ROLE OF THE PLASMINOGEN ACTIVATION SYSTEM IN ANGIOGENESIS
399
Figure 3. Effect of uPA expression on skeletal metastases. Rat prostate cancer cells Mat Ly Lu overexpressing uPA (uPA-S) were inoculated into the left ventricle of syngeneic male Copenhagen rats. Animals were monitored for the development of hind limb paralysis. Comparison was made with control group of animals inoculated with wild type vector transfected Mat Ly Lu cells (top). Histologic analysis of these skeletal metastases from uPA-S inoculated cells showed an osteoblastic response at these affected lumbar vertebra (bottom).
r e p ~ r t e d . l ~The l , ' ~mitogenic ~ effects of the ATF have been reported in ovarian tumor cells and in bone cell^.^^^'^' ATF was originally identified from prostatic cancer cell conditioned media as a growth factor for bone cells10' and this observation was extended in vivo through gain of function experiments, which demonstrated that the over-expression of uPA in prostatic cancer cells led to the early onset of skeletal metastases to cause hind limb paralysis of experimental animals. Furthermore, because of the mitogenic effects of ATF (of uPA), these skeletal metastases found in the lumbar vertebra were of the osteoblastic variety2 (Fig. 3). These effects required the expression of uPAR on the target cells and the stimulation of bone cells with ATF resulted in the induction of early response genes such as c-fos, c-jun, and ~ - m y c . The ' ~ ~ maximum induction was seen in c-fos mRNA expression, effects which could be abrogated by coincubation of ATF with soluble uPAR to indicate that this response was mediated via the uPAR on osteoblasts (Fig.4). Similarly, uPA-mediated invasion through Matrigel was also dependent on the ability of tumor cells to bind to uPAR and the simple displacement of uPA from its receptor inhibited invasion in several
Cydophilin
600 Z
a f
500
..C
- h
rap
+5 3
400 300
0
200
0
'S
.-0 d
Cyclophilin
i@
2
6 00
E C ..-
500
-
a
- h
2E
$20
-
53 B0 E0
$2
300
2Y :e
200
U
100
.c.
400
'S
.-0 2
100
CI
0
0
I B 45 min Figure 4. Effect of ATF on oncogene expression. A, Treatment of human osteoblast-derived osteosarcoma cells SaOS, with ATF (2.5 pg/mL) for 45 minutes resulted in a significant induction of c-myc, c-jun, and c-fos mRNA expression. B, Coincubationof SaOS, cells with ATF and soluble uPAR (S-uPAR) (10 pg/mL) resulted in neutralizing the ability of ATF to induce c-fos mRNA expression.
THE ROLE OF THE PLASMINOGEN ACTIVATION SYSTEM IN ANGIOGENESIS
401
in vitro system^.^^,^^^ Several studies in vivo have also demonstrated that inhibiting the catalytic activity of uPA directly135or displacing uPA from its receptor19lead to a significant decrease in tumor growth and metastasis. Delivery of uPAR antisense using an adenovirus construct into established sc U87MG glioma tumors not only inhibited tumor progression but also induced regre~sion.~' These results were consistentwith data from Aguirre Ghiso et a1 that demonstrated the down-regulation of uPAR in Hep4 cells resulted in the dormancy of these cells when grown in a CAM model.3A polyclonal antibody raised against the N-terminus of rat uPAR inhibited tumor growth and resulted in tumor regression in a syngeneic rat breast cancer model.lo4 Although the therapeutic potential of inhibiting the uPA system in treating cancer has been evident for several years, the role of the uPA-uPAR interaction in tumor progression has been difficult to address in vivo. A species barrier prevents rodent uPA from binding to human uPAR and vice versa and this has proved problematic for testing antagonists developed to inhibit human uPAR in mouse tumor models. Human uPAR-specific antagonists would not be expected to bind to mouse uPAR expressed on macrophages and endothelial cells and could therefore not probe the contribution of the host stromal cells to tumor progression. This issue was addressed recently in an elegant proof-of-principle study that demonstrated the full therapeutic potential of inhibiting the binding of uPA to uPAR in a syngeneic system. The delivery of an adenovirus-encoded murine ATF directly into tumors resulted in the suppression of neovascularization and tumor growth arrest.64In addition, tumor growth arrest was observed in syngeneic and xenograft models, indicating that the inhibition was mediated through the suppression of the host angiogenic response (a murine ATF would not be expected to bind to human tumor cell uPAR but only to host endothelial cells and leukocytes). Although most studies have focused on either the uPAR-binding region of uPA or its catalytic activity, the authors have recently identified a novel epitope from within the connecting peptide region of uPA (amino acids 136-158 in the mature uPA sequence) which mediates cell motility and ~ o n t r a c t i l i t y .The ~~,~ authors ~ have also demonstrated that a peptide derived from this region of uPA (A6, comprised of amino acids 136-143) inhibits tumor cell invasion38and smooth muscle contraction in ~ i t r oEx.~~ perimental animals inoculated with rat (Mat B-111) or human (MDA-231) breast cancer cells receiving A6 showed a marked reduction in their tumor volume throughout the course of this study because of the antiangiogenic effects of A6 in vivo (Fig. 5). uPA that is phosphorylated in this region (Ser 138) is no longer chemotactic for m o n ~ c y t e s Proteolytic .~~ cleavage within this region has been demonstrated also to abrogate the stimulatory activity of uPA on vascular smooth muscle cell (SMC) proliferation3' and the ability of uPA to promote the binding of soluble uPAR to hematopoietic cells.79Mukhina et a1 have also recently demonstrated that the recombinant kringle of uPA containing the connecting peptide was still able to bind to SMC (as observed in competition studies) and stimulate their
402
RABBANI & MAZAR
Mat B-Ill
-
*
0 1 0
6
11
12
13 14 Time (days)
15
16
*
17
Time (weeks)
Figure 5. Effect of a6 on tumor volume. A, Syngeneic female Fischer rats were inoculated with rat breast cancer cells (mat B-Ill) and received either vehicle control (CTL) or 75 mglkgld of A6 by intraperitoneal injection (B). MDA-231 tumor bearing BALBIc (nulnu) female mice were injected with vehicle alone (CTL) or A6 (75 mg/kg/d) (B) and tumor volume was measured at timed intervals. Results represent the mean & SEM of 6 starting animals in each group in 4 different experiments. Significant difference in tumor-volume from control tumorbearing animals after ~6 treatment is shown by asterisks (P < .05).
migration with an EC50 6 nM, suggesting either a novel receptor for this domain or a secondary interaction of uPA with ul'AR.'l Although it is not known whether the connecting peptide directly modulates tumor progression when it is part of uPA or whether the uPA/uPAR interac@onsin a tumor are the same as those observed on SMC, the effect of the A6 peptide, isolated from this region, on tumor growth and vascular contractility suggest that it is its important in tumor progression as well.
THE ROLE OF THE PLASMINOGEN ACTIVATION SYSTEM IN ANGIOGENESIS
403
PLASMINOGEN ACTIVATOR INHIBITORS
Modulation of uPA proteolytic activity in tumors can also be exerted through specific plasminogen activator inhibitors (PAIs).Of this family of serpin inhibitors PAI-1 is probably the most relevant to tumor progression. Counter-intuitively, however, high PAI-1 levels are also associated with a poor prognosis and advanced disease in many human tumors including: breast ~ancer.'~ The activation of vroteolvtic cascades must be temvoraEy controlled in order for tumor ptogressi~nand angiogenesis to ockr. Uncontrolled and unfocused uPA-dependent proteolysis is not productive from the standpoint of tumor progression. Excessive proteolytic activity in angiogenic endothelial cells leads to hemangioma rather than tube formation, indicating a requirement for a balance between protease and inhibitor." Fibrin is the major component of transitional matrix and promotes angiogenesis when it is deposited in the extravascular space iA inflammation and in tumors.48PAI-1 may serve to protect the fibrin matrix in a tumor such that angiogenesis can occur. The preservation of fibrin in the angiogenic process may be especially critical to angiogenesis because fibrin is the major ligand for the integrin a,/?3, which is present on angiogenic but not on quiescent endothelial cells.33In addition, fibrin induces the expression of IL-8 and intercellular adhesioin molecule 1 (ICAM-1) in endothelial cells.99,104 IL-8 is a potent angiogenic factor and chemokine and mediates the recruitment of neutrophils (PMN)to tumor^.^ Neutrophils also secrete VEGF, thereby amplifying tumor angi~genesis.~~ In addition, many leukocytes (e.g., monocytes, macrophages, neutrophils) secrete chemokines, many of which have recently been demonstrated to be associated with tumors and to induce angiogenesis as ell.^,''^ The recent demonstration of CXCR4 (a major chemokine receptor) expression on endothelial cells further strengthens the link between inflammation and tumor angi~genesis.~~ Finally, fibrin itself is a major mediator of inflammatory cell recruitment through ligation of /?, integrins such as CD11/18 (Mac-1) on the surfaces of neutrophils and macrophages.lo9The presence of numerous inflammatory cells in many human tumors has been well documented and these cells may be fueling the angiogenic response rather than seeking out and destroying tumor cells. Thus, the role of the uPA system (and specifically PAI-1) in angiogenesis involves at least partially the initiation and preservation of the inflammatory response in tumors. PAI-1 is expressed by endothelial cells in the Matrigel system and is probably required for proper tube formation.l13 Pepper et a1 recently demonstrated that a new member of the VEGF family, VEGF-C, up-regulates PAI-1 expression in several endothelial cell lines.94PAI-1 expression has been demonstrated in fibroblasts contacting endothelial cells, which can occur when the basement membrane has been degraded as part of the angiogenic process and the endothelial cells are in the process of migrating5 Tumor progression and angiogenesis is also inhibited in PAI-1-deficient mice in ~ i v oPAI-1-deficient .~ mice, however, develop without any overt vascular abnormalities, and endothelial cell migration after denudation is not inhibited in these animals.95TNP-470, an antiangiogenic drug, inhibits
404
RABBANI & MAZAR
the expression of uPA and PAI-1 in human lung cancer cell lines51although it is not known if this down-regulation contributes to the antiangiogenic mechanism of this drug. Mice deficient in the various components of the uPA system (plasminogen, uPA, uPAR, PAI-1) have also been used to assess the role of these of components in angiogenesis and tumor growth and dissemination. Wound healing is impaired in uPA-deficient mice, at least in part because of reduced migration by leukocytes, fibroblasts, and smooth muscle cells. Re-endothelialization of denuded vessels, however, is not affected in mice deficient in uPA, uPAR PAI-1 or ~ l a s m i n o g e nIn . ~these ~ systems, the lack of fibrinolytic components results in an increased deposition of fibrin, which provides a strong stimulus for angi~genesis.~~ In tumors, it may be necessary to have profibrinolytic and procoagulant activities operating simultaneously but in different regions of the tumor or cell. Because uPA-dependentproteolysis is focalized, one can imagine an endothelial cell that is actively degrading matrix at one end while interacting with fibrin at the other end. Alternatively,tumor-associated fibrinolysis and coagulation may cycle. Increased levels of PAI-1 would promote fibrin deposition by inhibiting plasminogen activation. This would potentiate the procoagulant activity of TF, which is expressed by many tumor cells and endothelial cells and may initiate fibrin deposition in the tumor milieu.75TF also directly regulates VEGF expressionby tumor cells.' PAI-1 expression in tumor cells is stimulated by TGFB, which is itself released from binding sites within the matrix through the activity of the uPA cascade. TGFB directly inhibits endothelial cell proliferation in .~ vitro by inducing apoptosis but stimulates angiogenesis in v i v ~ TGFB also stimulates VEGF expression in fibroblasts, especially in conjunction with hypoxia, which is often present in a tumor.12The combination of VEGF and fibrin (formedbecause of the inhibition of uPA-dependent fibrinolysis) may stimulate endothelial cell migration, proliferation, and angiogenesis. The binding of factor VIIa to TF has also recently been demonstrated to upregulate uPAR expression in pancreatic cancer cells1" suggesting an emerging paradigm in which cross-talk between procoagulant and profibrinolytic pathways fuels tumor invasion, metastasis, and angiogenesis. One hypothesis of how both systems might function simultaneously is that an invasive or migrating cell may be considered polarized because the leading edge and the trailing edge are in contact with different milieus. Migrating cells usually display calcium gradients that range from high (Ca+') coincident with regions of actin depolymerization (at the trailing edge) to low (Ca+2)coincident with regions of actin polymerization (leading edge), indicating different environments at each end of the cell. Similarly, haptotactic contacts, which promote migration, may also be different within a procoagulant environment (fibrin, high PAI-1) at one end of the cell and a profibrinolytic (high uPA, uPAR) at the other end of the cell. Plasminogen (Plg) also has been implicated in the negative control of angiogenesis, primarily through the activity of its degradation product, angiostatin.1° Angiostatin is produced through the proteolytic cleavage
THE ROLE OF THE PLASMINOGEN ACTIVATION SYSTEM IN ANGIOGENESIS
405
of Plg and numerous enzymes have been implicated in this process.93, 118 O'Mahony et a1 recently demonstrated that PAI-1 inhibited the production of angiostatin in the presence of the pancreatic cell line, ASPC-1,88 which may provide an explanation hypothesis for the pro-angiogenic role of PAI-1. The induction of apoptosis and the inhibition of migration in endothelial cells by angiostatin have been d e ~ c r i b e d . ~Recently, ',~~ a second fragment of plasminogen, kringle 5, has also been demonstrated to inhibit endothelial cell and rnigrati~n.~' This fragment of plasminogen is generated through the activity of macrophage elastase, which could lead to its production in the tumor milieu as well. Thus, various components of the uPA system may alternately elicit positive and negative regulatory effects on angiogenesis although the control of these effectsat the molecular level is not understood. THE uPA SYSTEM AND GROWTH FACTOR ACTIVITY
uPA (directly, or through Plm formation) leads to the release or activation of many angiogenic growth factors such as bFGF, VEGF, HGF, IGF, EGF, and TGFB.56,83,92,106,142~he release of FGF-2 through the activity of the uPA system has also recently been demonstrated in vivo in the CAM model.lo7uPA itself is able to directly cleave VEGF to one of its active forms, VEGF189, and directly activates HGFS3in a stoichiometric complex which then reacts with the HGF receptor, c-met.14' Tumor promoting/ proangiogenic agents such as HGF, EGF, and IGF upregulate expression of the uPA-uPAR system in other tumor-associated cell population^.'^^^'^^^^^^ Agents that down-regulate the expression of the HGF receptor, c-met (e.g., the geldanamycins) also inhibited the expression of uPA and uPAR in SKLMS-1cells.129The HGF-stimulated motility of these cells was also suppressed using these compounds,presumably through the down-regulation of the uPA pathway. In addition to recruiting, focusing, and enhancing the activation of plasminogen by uPA locally on cell surfaces, uPAR also participates in the regulation of this activity through the internalization of uPA-PA1 complexes. While scuPA and uPA remain stably bound to cell surface uPAR, ternary complexes of uPAR-uPA-PA1 are rapidly endocytosed through recognition and uptake by the lipoprotein receptor-related protein (LRP, a,-macroglobulin receptor). The uPA and PA1 components are degraded while uPAR is recycled to the cell surface.87Cells deficient in LRP migrated to a much greater extent than cells that express LRP, indicating that the presence of a constitutive uPA-uPAR complex on the cell surface promotes migration and invasion.130In that study, however, no PAI-1 was added to quench the system and in the presence of PAI-1, LRP-mediated internalization of the uPA-uPAR complex would probably allow the system to be refreshed through uPAR re~yc1ing.l~~ Also implicit in this cycling is an oscillating interaction of uPAR with its adhesion partners although it is not known whether uPAR that is bound to both uPA and an integrin or matrix component will internalize in the presence of PAI-1.
406
RABBANI & MAZAR
SIGNALING THROUGH uPAR
uPA bound to uPAR also mediates intracellular signal transduction in endothelial cells. ATF alone is able to mediate certain aspects of capillary tube formation, as described above. Tang et a1 have demonstrated that uPAR occupancy on endothelial cells results in the phosphorylation of focal adhesion proteins and the activation of MAP kinase.lZ3Antisense oligonucleotides which target uPAR expression inhibited endothelial cell proliferation, migration, and invasion and diacylglycerol was identified as a second messenger, leading to activation of the protein kinase C (PKC).26 The down-regulation of uPAR expression in this study would be expected to inhibit signaling pathways that were dependent on uPA catalytic activity and uPAR occupancy. It is possible that uPA catalytic activity can lead to the activation of PKC (through the remodeling of matrix and the disruption of integrin ligation) whereas uPAR occupancy can signal through the MAP kinase pathway. These two uPA-dependent pathways could operate simultaneously or be regulated in a temporal fashion in a migrating or invading cell. Studies with cells other than endothelial cells have also implicated the MAP kinase (MAPK) pathway in uPAR-mediated cell migration in vitro. In MCF-7 breast cancer cells, uPAR occupancy resulted in cell migration, which occurred through the activation of ERKl/ERK2. An inhibitor targeting MAPK redundant k-kinase, a member of the JAK family of kinases, suppressed ligand-induced uPAR-dependent activation of ERKl / E m 2 in these cells.85This pathway has also been implicated in the migration and invasion of HT1080 fibrosarcoma cells.60The JAK/STAT pathway may also be involved in the uPAR-dependent migration of vascular smooth muscle cells.24A second, uPAR-dependent signaling pathway involving Src-like protein tyrosine kinases (TKs) has also been described in these cells, although the functional relevance of this second pathway is not yet understood in smooth muscle cells. In U937 myelomonocytic cells, however, activation of Src-family TKs in undifferentiated U937 cells led to enhanced migration whereas down-regulation of the expression of these same TKs in differentiating cells led to increased adhesion.17The JAK/STAT pathway was also activated by clustering the uPA-uPAR complex using a monoclonal antibody in the human kidney epithelial cell line TCL-598, resulting in the migration of this cell line.61The MAPK/JAK/STAT pathway is activated in cytokine-mediated signaling and has been implicated as a major signal-transducing pathway (stimulated by VEGF and bFGF) in angiogene~is.~~, lZ2 INTEGRINS AND uPAR SIGNALING
Recent evidence suggests that uPAR-dependent signal transduction and migration are mediated through integrins (Fig. 6). In addition to PIand &-integrin involvement in the uPAR-dependent adhesion and migra, ~interaction ' ~ , ~ ~ ~ of uPAR with integrins has also tion of l e u k o ~ y t e s , l ~ ~the
THE ROLE OF THE PLASMINOGEN ACTIVATION SYSTEM IN ANGIOGENESIS
407
Single chain uPA uPA receptor
lntegrin
@b
Vitronectin
0
Plasminogen activator inhibitor-1
Figure 6. uPA-uPAR interaction with integrins. Single chain uPA (scuPA), by binding to its cell surface receptor, can interactwith integrins through domain 2 and 3 of uPAR. Similarly through domain 1 and 2 scuPA-uPAR can complex with vitronectin to collectively induce intracellular signals to promote adhesion, migration, invasion, and differentiation.
been demonstrated on tumor cells. a,& binds to fibrin and vitronectin Xue et a1 (VN), and may regulate the expression of uPAR and PAI-1.57,86 have demonstrated the interaction of uPAR on HT1080 cells with various a and @ integrins including B1 and B3 and a,a3, a5, and a6.13$Migration, but not adhesion, of FG cells, which express a,B,, on VN was uPA-uPAR dependent.141Migration of several melanoma cell lines, however, that express only a,B3 occurred independently of uPAR.'~~ Transfection of HT1080 cells with VN resulted in localization of uPA to a,B5-containing focal adhesions, resulting in increased levels of cell adhesion concomitant with decreased levels of plasminogen a~tivati0n.l~~ Adhesion of myogenic cells to PAI-1 coated plates depended on both a,B3 and uPAR.~~ Yebra et a1have
408
RABBANI & MAZAR
demonstrated that ligation of uPAR (with ATF or uPA) enhances the migration of uPAR-transfected LnCAP cells on fibronectin (FN) (which was a5B1dependent) and leads to the phosphorylation of two proteins (FAK that are involved in integrin signaling.140 uPAR and coand p13OCaS) irnmunoprecipitate only after uPAR ligation. The mechanism of forming this signaling complex is unclear, however. uPAR ligation may lead to conformational changes in uPAR that allow it to interact with the integrin or it could allow for the reorganization of adhesive interactions that allow the uPAR-integin complex to form. In Hep3 cells, the uPAR-a5B1interaction was required for the adhesion of these cells to FN. The interaction with FN led to a persistent stimulation of ERK and this activation could be enhanced by the ligation of uPAR with scuPA. Inhibition of the uPAR-a5B1 interaction led to tumor cell dormancy in the CAM ~ y s t e mThe . ~ ligation of uPAR leading to enhanced adhesion and migration does not require the presence of the catalytic domain and has now been demonstrated to mediate migration in several systems including tumor T lymphocytes13and most recently in neutrophil migration in response to a neutrophil chemotactic agent in vivo.lZ8Similarly, in breast cancer cells that migrate on vitronectin, uPAR associated with the vitronectin-bindingintegrin, a,B5 .16Thus, the nature of the uPAR-integrin association may depend on the nature of the cellmatrix component involved in the interaction. Physiologically, the same cell may interact with different matrix components temporally during cell migration, invasion, and growth and the nature of the uPAR-integrin interaction may change to support these various cellular activities. Very little is known about how the uPAR-integrin complex initiates signal transduction. A recent study by Wei et a1 using smooth muscle cells suggests that uPAR helps stabilize a caveolin dependent complex between B1 integrins and Src family kinases.131 Disruption of this complex either through down-regulation of caveolin expression or through the disruption of the B1-uPAR interaction reduces the ability of these cells to migrate and blocks the B1-Src kinase association. Unfortunately, there are few data available for tumor and endothelial cells and for non-& integrindependent associations of uPAR on cell surfaces. Thus, it is difficult to speculate on whether uPAR-dependent signal transduction is generally dependent on caveolin. SUMMARY
A substantial body of data now supports the central role of the uPA system in tumor angiogenesis and metastasis. The components of this system have both negative and positive regulatory roles in these processes, which may depend on time, place, and location of the cell and the expression of the components of this system. A major challenge in the uPA field is how to translate the wealth of biological information now available to obtain therapeutically useful agents. As with other important biological cascades (e.g., MMPs) that have been implicated in tumor progression and angiogenesis, the relevance of the uPA system to human disease will
THE ROLE OF THE PLASMINOGEN ACTNATION SYSTEM IN ANGIOGENESIS
409
not be fully understood until relevant clinical approaches that specifically modulate the activity of this system are developed and moved into human testing. References 1. Abe K, ShojiM, Chen J, et al: Regulation of vascular endothelial growth factor production and angiogenesis by the cytoplasmic tail of tissue factor. Proc Natl Acad SciUSA 96:86638668,1999 2. ~ c h b a r o uA, Kaiser S, Tremblay G, et al: Urokinase overproduction results in increased skeletal metastasis by prostate cancer cells in vivo. Cancer Res 542372-2377,1994 3. 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 Biol14789-104,1999 4. Arras M, Ito WD, Scholz D, et al: Monocyte activation in angiogenesis and collateral growth in the rabbit hindlimb. J Clin Invest 101:40-50,1998 5. Bacharach E, Itin A, Keshet E: Apposition-dependent induction of plasminogen activator inhibitor type 1expression: A mechanism for balancing pericellular proteolysis during angiogenesis. Blood 92:939-945,1998 6. Bajou K, Noel A, Gerard RD, et al: Absence of host plasminogen activator inhibitor 1 prevents cancer invasion and vascularization. Nat Med 4923-928,1998 7. Bastholm L, et al: Increased vascular endothelial growth factor. Applied Immunohist Molec Morphol7:139,1999 8. Behzadian MA, Wang XL, Shabrawey M, et al: Effects of hypoxia on glial cell expression of angiogenesis-regulating factors VEGF and TGF-beta. Glia 24:216-225,1998 9. Bellocq A, Antoine M, Flahault A, et al: Neutrophil alveolitis in bronchioloalveolar carcinoma: Induction by tumor-derived interleukin-8 and relation to clinical outcome. Am J Pathol 15283-92,1998 10. Bergers G, Javaherian K, Lo K, et al: Effects of angiogenesis inhibitors on multistage carcinogenesis in mice. Science. 2849308-812,1999 11. Berman M, Winthrop S, Ausprunk D, et al: Plasminogen activator (urokinase) causes vascularization of the cornea. Invest Ophthalmol Vis Sci 22:191-199,1982 12. Berse B, Hunt JA, Diegel RJ, et al: Hypoxia augments cytokine (transforming growth factor-beta (TGF-beta) and interleukin 1(IL-1)-inducedvascular endothelial growth factor secretion by human synovial fibroblasts. Clin Exp Immunol115:176-182,1999 13. Bianchi E, Ferrero E, Fazioli F, et al: Integrin-dependent induction of functional urokinase receptors in primary T lymphocytes. J Clin Invest 98:1133-1141,1996 14. Buo L, Meling GI, Karlsrud TS, et al: Antigen levels of urokinase plasminogen activator and its receptor at the tumor-host interface of colorectal adenocarcinomas are related to tumor aggressiveness. Hum Pathol 26:1133-1138,1995 15. Burgle M, Koppitz M, Riemer C, et al: Inhibition of the interaction of urokinase-type plasminogen activator (uPA) with its receptor (uPAR)by synthetic peptides. Biol Chem 378:231-237,1997 16. Carriero MV, Del Vecchio S, Capozzoli M, et al: Urokinase receptor interacts with alpha(v)beta5 vitronectin receptor, promoting urokinase-dependent cell migration in breast cancer. Cancer Res 59:5307-5314,1999 17. Chiaradonna F, Fontana L, Iavarone C, et al: Urokinase receptor-dependent and -independent p56/59(hck) activation state is a molecular switch between myelomonocytic cell motility and adherence. EMBO J 18:3013-3023,1999 18. Cho JY, Chung HC, Noh SH, et al: High level of urokinase-type plasminogen activator is a new prognostic marker in patients with gastric carcinoma.Cancer 792378-883,1997 19. Crowley CW, Cohen RL, Lucas BK, et al: Prevention of metastasis by inhibition of the urokinase receptor. Proc Natl Acad Sci USA 90:5021-5025,1990 20. de Witte JH, Sweep CG, Klijn JG, et al: Prognostic value of tissue-type plasminogen activator (PA) and its complex with the type-1 inhibitor (PAI-1) in breast cancer. Br J Cancer 80:286-294,1999
410
RABBANI & MAZAR
21. Dell'Era P, Mohammadi M, Presta M: Different tyrosine autophosphorylation requirements in fibroblast growth factor receptor-1 mediate urokinase-type plasminogen activator induction and rnitogenesis. Mol Biol Cell 10:23-33,1999 22. Dubois-StringfellowN, Jonczyk A, Bautch VL: Perturbations in the fibrinolytic pathway abolish cyst formationbut not capillary-like organization of cultured murine endothelial cells. Blood 83:3206-327,1994 23. Dubuisson L, Monvoisin A, Nielsen BS, et al: Expression and cellular localization of the urokinase-type plasminogen activator and its receptor in human hepatocellular carcinoma. J Pathol 190:190-195,2000 24. Dumler I, Weis A, Mayboroda OA, et al: The Jak/Stat pathway and urokinase receptor signaling in human aortic vascular smooth muscle cells. J Biol Chem 273:315-321, 1998 25. Ellerby HM, Arap W, Ellerby LM, et al: Anti-cancer activity of targeted pro-apoptotic peptides. Nat Med 5:1032-1038,1999 26. Fibbi G, Caldini R, Chevanne M, et al: Urokinase-dependent angiogenesis invitro and diacylglycerolproduction are blocked by antisense oligonucleotides against the urokinase receptor. Lab Invest 78:1109-1119,1998 27. Fischer K, Lutz V, Wilhelm 0, et al: Urokinase induces proliferation of human ovarian cancer cells: Characterization of structural elements required for growth factor function. FEBS Lett 438:lOl-105,1998 28. Foekens JA, Peters HA, Look MP, et al: The urokinase system of plasminogen activation and prognosis in 2780 breast cancer patients. Cancer Res 60:636-643,2000 29. Folberg R, Hendrix MJ, Maniotis AJ: Vasculogenic mimicry and tumor angiogenesis. Am J Pathol 156:361-381,2000 30. Folkman J: Angiogenesis research: From laboratory to clinic. Forum (Genova) 9:59-62, 1999 31. Franco P, Iaccarino C, Chiaradonna F, et al: Phosphorylation of human pro-urokinase on Ser138/303 impairs its receptor-dependent ability to promote myelomonocytic adherence and motility. J Cell Biol137779-791,1997 32. Franco P, Iaccarino C, Chiaradonna F, et al: Phosphorylation of human pro-urokinase on Ser138/303 impairs its receptor-dependent ability to promote myelomonocytic adherence and motility. J Cell Biol137:779-791,1997 33. Gamble JR, Matthias LJ, Meyer G, et al: Regulation of in vitro capillary tube formation by anti-integrin antibodies. J Cell Biol121:931-943,1993 34. Goldfarb RH, Kitson RP, Brunson KW: Biology of cancer metastases. Hematogenous dissemination. In Lotze MT, Rubin JT (eds): Regional Therapy of Advanced Cancer. Philadelphia, Lipincott-Raven, 1997, pp 1-9 35. Goretzki L, Schmitt M, Mann K, et al: Effective activation of the proenzyme form of the urokinase-type plasminogen activator (pro-uPA) by the cysteine protease cathepsin L. FEBS Lett 297:112-118,1992 36. Graham CH, Fitzpatrick TE, McCrae KR:Hypoxia stimulates urokinase receptor expression through a heme protein-dependent pathway. Blood 91:3300-3307,1998 37. Gualandris A, Lopez Conejo T, Giunciuglio D, et al: Urokinase-type plasminogen activator overexpression enhances the invasive capacity of endothelial cells. Microvasc Res 53:254-260,1997 38. Guo Y-J, Higazi AA-R, Arakelian A, et al: A peptide derived from the non-receptor binding region of urokinase plasminogen activator (uPA) inhibits tumor progression and angiogenesis and induces tumor cell death in vivo. FASEB J 14:1400-1410,2000 39. Gupta SK, Lysko PG, Pillarisetti K, et al: Chemokine receptors in human endothelial cells. Functional expression of CXCR4 and its transcriptional regulation by inflammatory cytokines. J Biol Chem 273:42824287,1998 40. Hahn-Dantona E, Ramos-DeSimone N, Sipley J, et al: Activation of proMMP-9 by a plasmin/MMP-3 cascade in a tumor cell model. Regulation by tissue inhibitors of metalloproteinases. Ann N Y Acad Sci 878:372-387,1999 41. Haj-Yehia A, Nassar T, Sachais BS, et al: Urokinase-derived peptides regulate vascular smooth muscle contraction in vitro and in vivo. FASEB J 14:1411-1422,2000 42. Hanahan D, Folkrnan J: Patterns and emerging mechanisms of the angiogenic switch during tumorigenesis. Cell 86:353-364,1996
THE ROLE OF THE PLASMINOGEN ACTNATION SYSTEM IN ANGIOGENESIS
411
43. Higazi A, Cohen RL, Henkin J, et al: Enhancement of the enzymatic activity of singlechain urokinase plasminogen activator by soluble urokinase receptor. J Biol Chem 270:17375-17380,1995 44. Higazi AA, Mazar A, Wang J, et al: Single-chain urokinase-type plasminogen activator bound to its receptor is relatively resistant to plasminogen activator inhibitor type 1. Blood 823545-3549,1996 45. Higazi AA, Bdeir K, Hiss E, et al: Lysis of plasma clots by urokinase-soluble urokinase receptor complexes. Blood 92:2075-2083,1998 46. Hildenbrand R, Dilger I, Horlin A, et al: Urokinase and macrophages in tumour angiogenesis. Br J Cancer 72:818-823,1995 47. Hildenbrand R, Wolf G, Bohme B, et al: Urokinaseplasminogen activator receptor (CD87) expression of tumor-associated macrophages in ductal carcinoma in situ, breast cancer, and resident macrophages of normal breast tissue. J Leukoc Biol66:4049,1999 48. Holvoet P, Collen D: Thrombosis and atherosclerosis.Curr Opin Lipidol 8:320-328,1997 49. Hsu DW, Efird JT, Hedley-Whyte ET: Prognostic role of urokinase-type plasminogen activator in human gliomas. Am J Path01 142114-123,1995 50. Ilan N, Mahooti S, Madri JA: Distinct signal transduction pathways are utilized during the tube formation and survival phases of in vitro angiogenesis. J Cell Sci 111:3621-3631, 1998 51. Ishikawa H, Satoh H, Kamma H, et al: The effect of TNP-470 on cell proliferation and urokinase-type plasminogen activator and its inhibitor in human lung cancer cell lines. J Exp Ther Oncol1:390-396,1996 52. Ito H, Rovira 11, Bloom ML, et al: Endothelial progenitor cells as putative targets for angiostatin. Cancer Res 59:5875-5877,1999 53. Jain RK, Safabakhsh N, Sckell A, et al: Endothelial cell death, angiogenesis, and microvascular function after castration in an androgen-dependent tumor: Role of vascular endothelial growth factor. Proc Natl Acad Sci USA 95:10820-10825,1998 54. Jardi M, Ingles-Esteve J, Burgal M, et al: Distinct patterns of urokinase receptor (uPAR) expression by leukemic cells and peripheral blood cells. Thromb Haemost 76:1009-1019, 1996 55. Kennedy S, Duffy MJ, Duggan C, et al: Semi-quantitation of urokinase plasminogen activator and its receptor in breast carcinomas by immunocytochemistry. Br J Cancer 721638-1641,1998 56. Keyt BA, Berleau LT, Nguyen HV, et al: The carboxyl-terminal domainn'-"j5 of vascular endothelial growth factor is critical for its mitogenic potency. J Biol Chem 271:7788-7795, 1996 57. Khatib A-M, Nip J, Fallavollitta L, et al: Regulation of urokinase plasminogen activator/plasmin-mediated invasion of melanoma cells by the integrin vitronectin receptor a,B3 Int J Cancer 91:300-308,2001 58. Kobayashi H, Schmitt M, Goretzki L, et al: Cathepsin B efficiently activates the soluble and the tumor cell receptor-bound form of the proenzyme urokinase-type plasminogen activator (Pro-uPA).J Biol Chem 266:5147-5152,1991 59. Koivunen E, Arap W, Valtanen H, et al: Tumor targeting with a selective gelatinase inhibitor. Nat Biotechnol17:768-774,1999 60. Konakova M, Hucho F, Schleuning WD. Downstream targets of urokinase-type plasminogen-activator-mediated signal transduction. Eur J Biochem 253:421429, 1998 61. Koshelnick Y, Ehart M, Hufnagl P, et al: Urokinase receptor is associated with the components of the JAKl/STATl 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 62. Lang IM, Moser KM, Schleef RR: Elevated expression of urokinase-like plasminogen activator and plasminogen activator inhibitor type 1 during the vascular remodeling associated with pulmonary thromboembolism. Arterioscler Thromb Vasc Biol 18:808815,1998 63. Lansink M, Koolwijk P, van Hinsbergh V, et al: Effect of steroid hormones and retinoids on the formation of capillary-like tubular structures of human microvascular endothelial cells in fibrin matrices is related to urokinase expression. Blood 92927-938,1998
412
RABBANI & MAZAR
64. Li H, Lu H, Griscelli F, et al: Adenovirus-mediated delivery of a uPA/uPAR antagonist suppresses angiogenesis-dependent tumor growth and dissemination in mice. Gene Ther 5:1105-1113,1998 65. Lu H, Dhanabal M, Volk R, et al: Kringle 5 causes cell cycle arrest and apoptosis of endothelial cells. Biochem Biophys Res Commun 258:668-673,1999 66. Lu H, Mabilat C, Yeh P, et al: Blockage of urokinase receptor reduces in vitro the motility and the deformability of endothelial cells. FEBS Lett 380:21-24,1996 67. Lucas MA, Straight DL, Fretto LJ, et al: The effects of fibrinogen and its cleavage products on the kinetics of plasminogen activation by urokinase and subsequent plasrnin activity. J Biol Chem 258:12171-12177,1983 68. Marchina E, Barlati S: Degradation of human plasma and extracellular matrix fibronectin by tissue type plasminogen activator and urokinase. Int J Biochem Cell Biol28:1141-1150, 1996 69. Lyons RM, Gentry LE, Purchio AF, et al: Mechanism of activation of latent recombinant transforming growth factor beta 1by plasmin. J Cell Biol110:1361-1367,1990 70. Manchanda N, Schwartz BS: Single chainurokinase. Augmentation of enzymatic activity upon binding to monocytes. J Biol Chem 266:14580-14584,1991 71. Mandriota SJ, Pepper MS: Vascular endothelial growth factor-induced in vitro angiogenesis and plasminogen activator expression are dependent on endogenous basic fibroblast growth factor. J Cell Sci 110:2293-2302,1997 72. Manes S, Mira E, Gomez-Mouton C, et al: Cells on the move: A dialogue between polarization and motility. IUBNB Life 49894996,2000 73. Mazzieri R, Masiero L, Zanetta L, et al: Control of type IV collagenase activity by components of the urokinase-plasmin system: A regulatory mechanism with cell-bound reactants. EMBO J 16:2319-2332,1997 74. McCourt M, Wang JH, Sookhai S, et al: Prointlarnrnatory mediators stimulate neutrophildirected angiogenesis. Arch Surg 1341325-1331; discussion 1331-1332,1999 75. Mechtcheriakova D, Wlachos A, Holzmuller H, et al: Vascular endothelial cell growth factor-induced tissue factor expression in endothelial cells is mediated by EGR-1. Blood 93:3811-3823,1999 76. Mignatti P, Rifkin DB: Plasminogen activators and matrix metalloproteinases in angiogenesis. Enzyme Prot 49:117-137,1996 77. Min HY, Doyle LV, Vitt CR, et al: Urokinase receptor antagonists inhibit angiogenesis and primary tumor growth in syngeneic mice. Cancer Res 56:2428-2433,1996 78. Mizukami IF, Gami-Wagner BA, DeAngelo LM, et al: 3rd Immunologic detection of the cellular receptor for urokinase plasminogen activator. Clin Immunol Immunopathol 71:96-104,1994 79. Mizukami IF, Todd RF 3rd: A soluble form of the urokinase plasminogen activator receptor (suPAR) can bind to hematopoietic cells. J Leukoc Biol64:203-213,1998 80. Mohan PM, Chintala SK, Mohanam S, et al: Adenovirus-mediated delivery of antisense gene to urokinase-type plasminogen activator receptor suppresses glioma invasion and tumor growth. Cancer Res 59:3369-3373,1999 81. Mukhina S, Stepanova V, Traktouev D, et al: The chemotactic action of urokinase on smooth muscle cells is dependent on its kringle domain; Characterization of interactions and contribution to chemotaxis. J Biol Chem 275:16450-16458,2000 82. Murphy G, Stanton H, Cowell S, et al: Mechanisms for pro matrix metalloproteinase activation. Apmis 107:3844,1999 83. Naldini L, Vigna E, Bardelli A, et al: Biological activation of pro-HGF (hepatocyte growth factor) by urokinase is controlled by a stoichiometric reaction. J Biol Chem 270:603-611, 1995 84. Nelson AR, Fingleton B, Rothenberg ML, et al: Matrix metalloproteinases: Biologic activity and clinical implications. J Clin Oncol18:1135-1149,2000 85. Nguyen DH, Hussaini IM, Gonias SL, et al: 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 27323502-8507,1998 86. Nip J, Rabbani SA, Shibata HR, et al: Coordinated expression of the vitronectin receptor and the urokinase-type plasminogen activator receptor in metastatic melanoma cells. J Clin Invest 95:2096-20103,1995
THE ROLE OF THE PLASMINOGEN ACTIVATION SYSTEM IN ANGIOGENESIS
413
87. Nykjaer A, Conese M, Christensen EI, et al: Recycling of the urokinase receptor upon internalization of the uPA: Serpin complexes. EMBO J 16:2610-2620,1997 88. O'Mahony CA, Albo D, Tuszynski GP, et al: Transforming growth factor-beta 1 inhibits generation of angiostatin by human pancreatic cancer cells. Surgery 124:388-393, 1998 89. Odekon LE, Sato Y, Rifkin DB: Urokinase-type plasminogen activator mediates basic fibroblast growth factor-induced bovine endothelial cell migration independent of its proteolytic activity. J Cell Physiol150:258-263,1992 90. Ohtani H, Pyke C, Dano K, et al: Expression of urokinase receptor in various stromalcell populations in human colon cancer: Immunoelectron microscopical analysis. Int J Cancer 62:691-696,1995 91. Ohtani H: Stromal reaction in cancer tissue: Pathophysiologic significance of the expression of matrix-degrading enzymes in relation to matrix turnover and immune/inflammatory reactions. Pathol Int 48:l-9,1998 92. Park JE, Keller GA, Ferrara N: The vascular endothelial growth factor (VEGF) isoforms: Differential deposition into the subepithelial extracellular matrix and bioactivity of extracellular matrix-bound VEGF. Mol Biol Cell 4:1317-1326,1993 93. Patterson BC, Sang QA: Angiostatin-converting enzyme activities of human matrilysin (MMP-7) and gelatinase B/type IV collagenase (MMP-9).J Biol Chem 272:28823-28825, 1997 94. Pepper MS, Mandriota SJ, Jeltsch M, et al: Vascular endothelial growth factor (VEGF)-C synergizes with basic fibroblast growth factor and VEGF in the induction of angiogenesis invitro and alters endothelial cell extracellular proteolyticactivity.J Cell Physiol177:439452,1998 95. Pepper MS, Montesano R, Mandriota SJ, et al: Angiogenesis: A paradigm for balanced extracellular proteolysis during cell migration and morphogenesis. Enzyme Protein 49:138-162,1996 96. Pinsky DJ, Liao H, Lawson CA, et al: Coordinated induction of plasminogen activator inhibitor-1 (PAI-1) and inhibition of plasminogen activator gene expression by hypoxia promotes pulmonary vascular fibrin deposition. J Clin Invest 102:919-928,1998 97. Planus E, Barlovatz-Meimon G, Rogers RA, et al: Binding of urokinase to plasminogen activator inhibitor type-1 mediates cell adhesion and spreading. J Cell Sci 110:1091-1098, 1997 98. Proescholdt MA, Heiss JD, Walbridge S, et al: Vascular endothelial growth factor (VEGF) modulates vascular permeability and inflammation in rat brain. J Neuropathol Exp Neurol58:613-627,1999 99. Qi J, Kreutzer DL, Piela-Smith TH: Fibrin induction of ICAM-1 expression in human vascular endothelial cells. J Immunol158:1880-1886,1997 100. Rabbani SA, Xing RH: Role of urokinase (uPA) and its receptor (uPAR) in invasion and metastasis of hormone-dependent malignancies. Int J Oncol12:911-920,1998 101. Rabbani SA, Desjardins J, Bell AW, et al: An amino-terminal fragment of urokinase isolated from a prostate cancer cell line (PC-3) is mitogenic for osteoblast-like cells. Biochem Biophys Res Commun 173:1058-1064,1990 102. Rabbani SA, Gladu J, Mazar AP, et al: Induction in human osteoblastic cells (SaOS2) of the early response genes fos, jun, and myc by the amino terminal fragment (ATF) of urokinase. J Cell Physiol172:137-145,1997 103. Rabbani SA: Urokinase receptor directed immunodiagnostic and immunotherapeutic strategies for prostate and breast cancer. Proc Am Assoc Cancer Res 38:A1673,1997 104. Ramsby ML, Kreutzer DL: Fibrin induction of interleukin-8 expression in corneal endothelial cells in vitro. Invest Ophthalmol Vis Sci 35:3980-3990,1994 105. Ray JM, Stetler-Stevenson WG: The role of matrix metalloproteases and their inhibitors in tumour invasion, metastasis and angiogenesis. Eur Respir J 7:2062-2072,1994 106. Remacle-Bonnet MM, Garrouste FL, Pommier GJ: Surface-bound plasmin induces selective proteolysis of insulin-like-growth-factor (1GF)-bindingprotein-4 (IGFBP-4) and promotes autocrine IGF-I1 bio-availability in human colon-carcinoma cells. Int J Cancer 722335-843,1997 107. Ribatti D, Leali D, Vacca A, et al: In vivo angiogenic activity of urokinase: Role of endogenous fibroblast growth factor-2. J Cell Sci 112:42134221,1999
414
RABBANI & MAZAR
108. Ried S, Jager C, Jeffers M, et al: Activation mechanisms of the urokinase-type plasminogen activator promoter by hepatocyte growth factor/scatter factor. J Biol Chem 274:16377-16386,1999 109. Rogers C, Edelman ER, Simon DI: A mAb to the beta2-leukocyte integrin Mac-1 (CDllb/CD18) reduces intimal thickening after angioplasty or stent implantation in rabbits. Proc Natl Acad Sci USA 95:10134-10139,1998 110. Rosenthal EL, Johnson TM, Allen ED, et al: Role of the plasminogen activator and matrix metalloproteinase systems in epidermal growth factor- and scatter factor-stimulated invasion of carcinoma cells. Cancer Res 58:5221-5230,1998 111. Sakaguchi I, Ikeda N, Nakayama M, et al: 6,6'-dimycolate (Cord factor) enhances neovascularization through vascular endothelial growth factor production by neutrophils and macrophages. Infect Immun 68:2043-2052,2000 112. Saksela 0 : Plasminogen activation and regulation of pericellular proteolysis. Biochim Biophys Acta 823:35-65,1985 113. Schnaper HW, Barnathan ES, Mazar A, et al: Plasminogenactivators augment endothelial cell organization in vitro by two distinct pathways. J Cell Physiol165:107-118,1995 114. Schneller M, Vuori K, Ruoslahti E: Alphavbeta3 integrin associates with activated insulin and PDGFbeta receptors and potentiates the biological activity of PDGF. EMBO J 16:5600- 5607,1997 115. Sendo F, Suzuki K, Watanabe T, et al: Modulation of leukocyte transendothelialmigration by integrin-associated glycosyl phosphatidyl inositol (GP1)-anchoredproteins. Inflamm Res 47:133-136,1998 116. Simon DI, Wei Y, Zhang L, et al: Identification of a urokinase receptor-integrin interaction site. Promiscuous regulator of integrin function [In Process Citation]. J Biol Chem 275:10228-10234,2000 117. Stack MS, Johnson DA: Human mast cell tryptase activates single-chain urinary-type plasminogen activator (pro-urokinase). J Biol Chem 269:9416-9419,1994 118. Stathakis P, Lay AJ, Fitzgerald M, et al: Angiostatin formation involves disulfide bond reduction and proteolysis in kringle 5 of plasrnin. J Biol Chem 274:8910-8916,1999 119. Stephens RW, Brunner N, Janicke F, et al: The urokinase plasminogen activator system as a target for prognostic studies in breast cancer. Breast Cancer Res Treat 52:99-111, 1998 120. Strojan P, Budihna M, Smid L, et al: Urokinase-type plasminogen activator (uPA) and plasminogen activator inhibitor type 1 (PAI-1) in tissue and serum of head and neck squamous cell carcinoma patients. Eur J Cancer 34:1193-1197,1998 121. Stonelake PS, Jones CE, Neoptolemos JP, et al: Proteinase inhibitors reduce basement membrane degradation by human breast cancer cell lines. Br J Cancer 75:9515-9519, 1997 122. Tanaka K, Abe M, Sato Y Roles of extracellular signal-regulated kinase 1/2 and p38 mitogen-activated protein kinase in the signal transduction of basic fibroblast growth factor in endothelial cells during angiogenesis. Jpn J Cancer Res 90:647-654,1999 123. Tang H, Kerins DM, Hao Q, et al: The urokinase-type plasminogen activator receptor mediates tyrosine phosphorylation of focal adhesion proteins and activation of mitogenactivated protein kinase in cultured endothelial cells. J Biol Chem 273:8268-8272,1998 124. TaniguchiT, Kakkar AK, Tuddenham EG, et al: Enhanced expression of urokinase receptor induced through the tissue factor-factor VIIa pathway in human pancreatic cancer. Cancer Res 58:44614467,1998 125. Torisu H, Ono M, Kiryu H, et al: Macrophage infiltration correlates with tumor stage and angiogenesis in human malignant melanoma: Possible involvement of TNFalpha and IL-lalpha. Int J Cancer 85:182-188,2000 126. van ~ e e u w e nRL, Yoshinaga IG, Akasaka T, et al: Attachment, spreading and migration of melanoma cells on vitronectin. The role of aloha V beta 3 and aloha V beta 5 mtegrins. Exp Dermato15:308-315,1996 127. van HinsberghVW, Koolwijk P, Hanemaaijer R: Role of fibrin and plasminogen activators in repair-associated angiogenesis: In vitro studies with human endothelial cells. Exs 79:391411,1997 128. Waltz DA, Fujita RM, Yang X, et al: Nonproteolytic role for the urokinase receptor in cellular migration in vivo. Am J Respir Cell Mol Biol22:316-322,2000
.,
THE ROLE O F THE PLASMINOGEN ACTIVATION SYSTEM IN ANGIOGENESIS
415
129. Webb CP, Hose CD, Koochekpour S, et al: The geldanamycins are potent inhibitors of the hepatocyte growth factor/scatter factor-met-urokinaseplasminogen activator-plasmin proteolytic network. Cancer Res 60:342-349,2000 130. Webb DJ, Nguyen DH, Gonias SL: Extracellular signal-regulated kinase functions in the urokinase receptor-dependent pathway by which neutralization of low density lipoprotein receptor-related protein promotes fibrosarcoma cell migration and matrigel invasion. J Cell Sci 113:123-134,2000 131. Wei Y, Yang X, Liu Q, et al: A role for caveolin and the urokinase receptor in integrinmediated adhesion and signaling. J Cell Biol144:1285-1294,1999 132. Wilcox SA, Reho T, ~ i ~ PJ, ~ etal: i &Localization of urokinase to focal adhesions by human fibrosarcoma cells svnthesizinn " recombinant vitronectin. Biochem Cell Biol74:899810,1996 133. Wong WS, Simon DI, Rosoff PM, et al: Mechanisms of pertussis toxin-induced myelomonocytic cell adhesion: Role of Mac-l(CDllb/CD18) and urokinase receptor (CD87).Immunol88:90-97,1996 134. Wood JM, Bold G, Buchdunger E, et al: PTK787/ZK 222584, a novel and potent inhibitor of vascular endothelial growth factor receptor tyrosine kinases, impairs vascular endothelial growth factor-induced responses and tumor growth after oral administration. [In Process Citation] Cancer Res 60:2178-2189,2000 135. Xing RH, Mazar A, Henkin J, et al: Prevention of breast cancer growth, invasion, and metastasis by antiestrogen tamoxifen alone or in combination with urokinase inhibitor 8-428. Cancer Res 57:3585-3593,1997 136. Xing RH, Rabbani SA: Regulation of urokinase production by androgens in human prostate cancer cells: Effect on tumor growth and metastases in vivo. Endocrinology 140:40564064,1999 137. Xu Y, Hagege J, Doublet JD, et al: Endothelial and macrophage upregulation of urokinase receptor expression in human renal cell carcinoma. Hum Pathol 28:206-213,1997 138. Xue W, Mizukami I, Todd RF 3rd, et al: Urokinase-type plasminogen activator receptors associate with beta1 and beta3 integrins of fibrosarcoma cells: Dependence on extracellular matrix components. Cancer Res 57:1682-1689,1997 139. Yamamoto M, Sawaya R, Mohanam S, et al: Expression and localization of urokinasetype plasminogen activator receptor in human gliomas. Cancer Res 54:5016-5020,1994 140. Yebra M, Goretzki L, Pfeifer M, et al: Urokinase-type plasminogen activator binding to its receptor stimulates tumor cell migration by ehhancing integrin-mediated signal transduction. Exp Cell Res 250:231-240,1999 141. Yebra M, Parry GCN, Stromblad S, et al: Requirement of receptor-bound urokinase-type plasminogen activator for integrin alphavbeta5-directed cell migration. J Biol Chem 271:29393-29399,1996 142. Zanetti A, Stoppacciaro A, Marzullo A, et al: Expression of Met protein and urokinasetype plasminogen activator receptor (uPA-R) in papillary carcinoma of the thyroid. J Pathol 186:287-291,1998 143. Zhang JC, Sakthivel R, Kniss D, et al: The low density lipoprotein receptor-related protein/alpha2-macroglobulin receptor regulates cell surface plasminogen activator activity on human trophoblast cells. J Biol Chem 273:32273-32280,1998
Address reprint requests to Shafaat A. Rabbani, MD McGill University Health Centre 687 Pine Avenue West, Room H4.67 Montreal, Quebec H3A 1Al Canada