Skip to main content
Log in

Multiple pathways of cell invasion are regulated by multiple families of serine proteases

  • Published:
Clinical & Experimental Metastasis Aims and scope Submit manuscript

Abstract

The complex process of tumor invasion requires the coordinated expression and activity of cell-substratum adhesive interactions and of cell-associated protease systems, which destroy the extracellular matrix (ECM), in order to enable the invading cells to simultaneously grip and destroy the anatomical barriers that control cell spreading. A number of data indicate that such a `grip and go' process may be performed by an enlarging series of cell membrane-associated serine proteases and serine protease receptors, which provide the invasive cells with a functional unit (the protease and its receptor), able to mediate cell-substratum adhesion through specific receptor domains, to proteolytically degrade ECM and to deliver into the cell signals that up-regulate the expression either of the protease/receptor complex, or of other adhesion molecules, such as integrins. There is evidence that some proteases and protease receptor expression are under the control of tumor hypoxia, which is the result of an imbalance in oxygen supply and demand. The urokinase-type plasminogen activator (u-PA) receptor (u-PAR) is under hypoxic control and cooperates with other serine proteases of the blood coagulation pathways that may extravasate in the tumor milieu as a result of hypoxia-simulated increase of vessel permeability. Other serine proteases and their receptors cooperate with the cell-associated fibrinolytic system to promote cell invasion. Among these, tissue factor and its ligand coagulation factor VII, thrombin and its protease-activated receptors, and type II trans-membrane serine proteases seem to play a crucial role. This Review takes into consideration the complex scenario of the single serine proteases and related receptors that are involved in cell invasion, as well as the protease receptor/adhesion molecule interplay which is necessary to focus the cell surface-driven proteolysis where adhesion provides a grip to the invading cell.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Institutional subscriptions

References

  1. Dano K, Andreasen PA, Grondahl-Hansen J et al. Plasminogen activators, tissue degradation, and cancer. Adv Cancer Res 1985; 44: 139–266.

    PubMed  CAS  Google Scholar 

  2. Goldhaber P, Cornman I, Ormsbee RA. Experimental alteration of the ability of tumor cells to lyse plasma clots in vitro. Proc Soc Exp Biol Med 1947; 66: 590–5.

    CAS  Google Scholar 

  3. Guillemin K, Krasnow MA. The hypoxic response: Huffing and HIFing. Cell 1997; 89: 9–12.

    PubMed  CAS  Google Scholar 

  4. Blancher C, Harris AL. The molcular basis of the hypoxia response pathway: Tumor hypoxia as a therapy target. Cancer Metastasis 1998; 17: 187–94.

    CAS  Google Scholar 

  5. Srinivas V, Zhu X, Salceda S et al. Hypoxia inducible factor alpha (HIF-alpha) is a non-heme protein. Implications for oxygen sensing. J Biol Chem 1998; 273: 18019–22.

    PubMed  CAS  Google Scholar 

  6. Dachs GU, Tozer GM. Hypoxia modulated gene expression: Angiogenesis, metastasis and therapeutic exploitation. Eur J Cancer 2000; 36: 1649–60.

    PubMed  CAS  Google Scholar 

  7. Graham CH, Forsdike J, Fitzgerald CJ et al. Hypoxia-mediated stimulation of carcinoma cell invasiveness via upregulation of urokinase receptor expression. Int J Cancer 1999; 80: 617–23.

    PubMed  CAS  Google Scholar 

  8. Folkman J. Angiogenesis in cancer, vascular, rheumatoid and other diseases. Nat Med 1995; 1: 27–31.

    PubMed  CAS  Google Scholar 

  9. Kook YH, Adamski J, Zelent A et al. The effect of antisense inhibition of urokinase receptor in human squamous cell carcinoma on malignancy. EMBO J 1994; 13: 3983–91.

    PubMed  CAS  Google Scholar 

  10. Yu W, Kim J, Ossowski L. Reduction in surface urokinase receptor forces malignant cells into a protracted state of dormancy. J Cell Biol 1997; 137: 767–77.

    PubMed  CAS  Google Scholar 

  11. Ott I, Fisher EG, Miyagi Y et al. A role for tissue factor in cell adhesion and migration mediated by interaction with Actin-binding Protein 280. J Cell Biol 1998; 140: 1241–53.

    PubMed  CAS  Google Scholar 

  12. O'Meara RAQ, Jackson RD. Cytological observations on carcinoma. Ir J Med Sci 1958; 6: 327–8.

    Google Scholar 

  13. Dvorak HF, Senger DR, Dvorak AM. Fibrin as a component of the tumor stroma: origins and biological significance. Cancer Metastasis Rev 1983; 2: 41–73.

    PubMed  CAS  Google Scholar 

  14. Gerner C, Steinkellner W, Holzmann K et al. Elevated plasma levels of crosslinked fibrinogen gamma-chain dimer indicate cancer-related fibrin deposition and fibrinolysis. Thromb Haemost 2001; 85: 494–501.

    PubMed  CAS  Google Scholar 

  15. Paleeck SP, Loftus JC, Ginsberg MH et al. Integrin-ligand binding properties govern cell migration speed through cell-substratum adhesiveness. Nature 1996; 385: 537–40.

    Google Scholar 

  16. Andreasen PA, Egelund R, Petersen HH. The plasminogen activation system in tumor growth, invasion, and metastasis. Cell Mol Life Sci 2000; 57: 25–40.

    PubMed  CAS  Google Scholar 

  17. Miles LA, Dahlberg CM, Plescia J et al. Role of cell-surface lysines in plasminogen binding to cells: Identification of alpha-enolase as a candidate plasminogen receptor.

  18. Harder T, Simons K, Caveolae, DIGS and the dynamics of sphingolipid-cholesterol mierodomains. Curr Opin Cell Biol 1997; 9: 543–2. Biochemistry 1991; 30: 1682–91.

    Google Scholar 

  19. Horejsi V, Drbal K, Cebekauer M et al. GPI-microdomains: A role in signalling via immunoreceptors. Immunol Today 1999; 20: 356–61.

    PubMed  CAS  Google Scholar 

  20. Estreicher A, Muhlhauser J, Carpentier JL, et al. The receptor for urokinase type plasminogen activator polarizes expression of the protease to the leading edge of migrating monocytes and promotes degradation of enzyme inhibitor complexes. J Cell Biol 1990 111(2): 783–92.

    PubMed  CAS  Google Scholar 

  21. Mignatti P, Rifkin DB. Nonenzymatic interactions between proteinases and the cell surface: Novel roles in normal and malignant cell physiology. Adv Cancer Res 2000; 78: 103–157.

    PubMed  CAS  Google Scholar 

  22. Hoyer-Hansen G, Ploug M, Behrendt N et al. Cell-surface acceleration of urokinase-catalyzed receptor cleavage. Eur J Biochem 1997; 243: 21–6.

    PubMed  CAS  Google Scholar 

  23. Pedersen N, Schmitt M, Ronne E et al. A ligand-free, soluble urokinase receptor is present in the ascitic fluid from patients with ovarian cancer. J Clin Invest 1993; 92: 2160–7.

    PubMed  CAS  Google Scholar 

  24. Møller LB, Pöllänen J, Rønne E et al. N-linked glycosylation of the ligand-binding domain of the human urokinase receptor contributes to the affinity for its ligand. J Biol Chem 1993; 268: 11152–9.

    PubMed  Google Scholar 

  25. Pucei M, Fibbi G, Magnelli L et al. Regulation of urokinase/urokinase receptor interaction by heparin-like glycosaminoglycans. J Biol Chem 2001; 276: 4756–65.

    Google Scholar 

  26. Andreasen PA, Kjoller L, Christensen L et al. The urokinase-type plasminogen activator system in cancer metastasis: A review. Int J Cancer 1997; 72: 1–22.

    PubMed  CAS  Google Scholar 

  27. Reijerkerk A, Voest EE, Gebbink MF. No grip, no growth: The conceptual basis of excessive proteolysis in the treatment of cancer. Eur J Cancer 2000; 36(13 Spec No): 1695–705.

    PubMed  CAS  Google Scholar 

  28. 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 Physiol 1992; 150: 258–63.

    PubMed  CAS  Google Scholar 

  29. Wei Y, Waltz DA, Rao N et al. Identification of the urokinase receptor as an adhesion receptor for vitronectin. J Biol Chem 1994; 269: 32380–8.

    PubMed  CAS  Google Scholar 

  30. Kanse SM, Kost C, Wilhelm OG et al. The urokinase receptor is a major vitroncetin-binding protein on endothelial cells. Exp Cell Res 1996; 224: 344–53.

    PubMed  CAS  Google Scholar 

  31. Deng G, Curriden SA, Wang S et al. Is plasminogen activator inhibitor-1 the molecular switch that governs urokinase receptor-mediated cell adhesion and release? J Cell Biol 1996; 134: 1563–71.

    PubMed  CAS  Google Scholar 

  32. Colman RW, Pixley RA, Najamunnisa S et al. Binding of high molecular weight kininogen to human endothelial cells is mediated via a site within domains 2 and 3 of the urokinase receptor. J Clin Invest 1997; 100: 1481–7.

    PubMed  CAS  Google Scholar 

  33. Chavakis T, Kanse SM, Lupu F et al. Different mechanisms define the antiadhesive function of high molecular weight kininogen in integrin-and urokinase receptor-dependent interactions.

  34. Godar S, Horejsi V, Weidle UH et al. M6P/IGFII-receptor complexes urokinase and plasminogen for activation of transforming growth factor betal. Eur J Immunol 1999; 29: 1004–13.

    PubMed  CAS  Google Scholar 

  35. Koshelnick Y, Ehart M, Hufnagl P et al. Urokinase receptor is associated with the components of the JAK1/STAT1 signaling pathway and leads to activation of this pathway upon clustering in the human kidney epithelial tumor cell line TLC-598. J Biol Chem 1997; 272: 28563–7.

    PubMed  CAS  Google Scholar 

  36. Behrendt N, Jensen ON, Engelholm LH et al. A urokinase receptor-associated protein with specific collagen binding properties. J Biol Chem 2000; 275: 1993–2002.

    PubMed  CAS  Google Scholar 

  37. Chapman HA. Plasminogen activators, integrins and the coordinated ragulation of cell adhesion and migration. Curr Opin Cell Biol 1997; 9: 714–24.

    PubMed  CAS  Google Scholar 

  38. Chapman HA, Wei Y, Simon DI et al. Role of urokinase receptor and caveolin in regulation of integrin signaling. Thromb Haemostasis 1999; 82: 291–7.

    CAS  Google Scholar 

  39. Stefansson S, Lawrence DA. The serpin PAI-1 inhibits cell migration by blocking integrin αvβ3 binding to vitronectin. Nature 1996; 383: 441–3.

    PubMed  CAS  Google Scholar 

  40. Kjoller L, Kanse SM, Kirkegaard T et al. Plasminogen activator inhibitor-1 represses integrin-and vitroncetin-mediated cell migration independently of its function as an inhibitor of plasminogen activation. Exp Cell Res 1997; 232: 420–9.

    PubMed  CAS  Google Scholar 

  41. Janicke F, Schmitt M, Pache L et al. Urokinase (uPA) and ist inhibitor PAI-1 are strong and independent prognostic factors in node-negative breast cancer. Brest Cancer Res Treat 1993; 24: 195–208.

    CAS  Google Scholar 

  42. Grondahl-Hansen J, Christensen U, Rosenquist C et al. High levels of urokinase-type plasmiongen activator and its inhibitor PAI-1 in cytosolic extracts of breast carcinomas are associated with poor prognosis. Cancer Res 1993; 53: 2513–21.

    PubMed  CAS  Google Scholar 

  43. Hofman R, Lehmer A, Buresh M et al. Clinical relevance of urokinase plasminogen activator, its receptor, and its inhibitor in patients with renal carcinoma. Cancer 1996; 78: 487-92.

    Google Scholar 

  44. Allgayer H, Babic R, Grutzner KU et al. Tumor-associated proteases and inhibitors in gastric cancer: Analysis of prognostic impact and individual risk protease patterns. Clin Exp Metastasis 1998; 16: 62–73.

    PubMed  CAS  Google Scholar 

  45. Carmeliet P, Schoojans L, Kieckens L et al. Physiological consequences of loss of plasminogen activator gene function in mice. Nature 1994; 368: 419–24.

    PubMed  CAS  Google Scholar 

  46. Dewerchin M, Nuffelen AV, Wallays G et al. Generation and characterization of urokinase receptor-deficient mice. J Clin Invest 1996; 97: 870–8.

    PubMed  CAS  Google Scholar 

  47. Pedersen TL, Yong K, Pedersen JO et al. Impaired migration in vitro of neutrophils from patients with paroxysmal nocturnal haemoglobinuria. Br J Haematol 1996; 95: 45–51.

    PubMed  CAS  Google Scholar 

  48. Fibbi G, Ziche M, Morbidelli L et al. Interaction of urokinase with specific receptors stimulates mobilization of bovine adrenal capillary endothelial cells. Exp Cell Res 1988; 179: 385–95.

    PubMed  CAS  Google Scholar 

  49. Ossowski L, Aguirre-Ghiso JA. Urokinase receptor and integrin partnership: Coordination of signaling for cell adhesion, migration and growth. Curr Opin Cell Biol 2000; 12: 613–20.

    PubMed  CAS  Google Scholar 

  50. Fazioli F, Resnati M, Sidenius N et al. A urokinase-sensitive region of the human urokinase receptor is responsible for its chemotactic activity. EMBO J 1997; 16: 7279–86.

    PubMed  CAS  Google Scholar 

  51. 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 2000; 275: 16450–8.

    PubMed  CAS  Google Scholar 

  52. Del Rosso M, Fibbi G, Dini G et al. Role of specific membrane receptors in urokinase-dependent migration of human keratinocytes. J Invest Dermatol 1990; 94: 310–6.

    PubMed  CAS  Google Scholar 

  53. Grondahl-Hansen J, Lund LR, Ralfkiaer et al. Urokinase and tissue-type plasminogen activators in keratinocytes during wound reepithelization in vivo. J Invest Dermatol 1988; 90: 790–5.

    PubMed  CAS  Google Scholar 

  54. Hajjar KA, Acharya SS. Annexin II and regulation of cell surface fibrinolysis. Ann NY Acad Sci 2000; 902: 265–71.

    PubMed  CAS  Google Scholar 

  55. Mai J, Wasman DM, Sloane BF. Cell surface complex of cathepsin B/annexin II tetramer in malignant progression. Biochim Biophys Acta 2000; 1477: 215–30.

    PubMed  CAS  Google Scholar 

  56. Kurschat P, Mauch C. Mechanisms of metastasis. Clin Dermatol 2000; 25: 482–9.

    CAS  Google Scholar 

  57. Martin DMA, Boys CWG, Ruf W. Tissue factor: Molecular recognition and cofactor function. FASEB J 1995; 9: 852–9.

    PubMed  CAS  Google Scholar 

  58. Drake TA, Morrissey JH, Edgington TS. Selective cellular expression of tissue factor in human tissues. Am J Pathol 1989; 134: 1087–97.

    PubMed  CAS  Google Scholar 

  59. Andoh K et al. Observations on the cell biology of tissue factor in endothelial cells. Thromb Haemost 1990; 63; 298–302.

    PubMed  CAS  Google Scholar 

  60. Edwards RL, Rickles FR. The role of leukocytes in the activation of blood coagulation. Semin Hematol 1992; 29: 202–12.

    PubMed  CAS  Google Scholar 

  61. Hillen HF. Thrombosis in cancer patients, Ann Oncol 2000; 11(3): 273–6.

    PubMed  Google Scholar 

  62. Ott I, Fischer EG, Miyagi Y et al. A role for tissue factor in cell adhesion and migration mediated by interaction with actin-binding protein 280. J Cell Biol 1998; 140: 1241–53.

    PubMed  CAS  Google Scholar 

  63. Bromberg ME, Konigsberg WH, Madison JF et al. Tissue factor promotes melanoma metastasis by a pathway independent of blood coagulation. Proc Natl Acad Sci USA 1995; 92: 8205–9.

    PubMed  CAS  Google Scholar 

  64. Carson SD, Kuszynski CA, Pirruccello SJ. Fibroblasts restrict tissue factor from vescicles which form in response to low concentrations of detergent. Blood Coagul Fibrinolysis 1996; 7: 314–24.

    PubMed  CAS  Google Scholar 

  65. Sprecher CA, Kisiel W, Mathews S et al. Molecular cloning, expression, and partial characterization of a second human tissue-factor pathway inhibitor. Proc Natl Acad Sci USA 1994; 91: 3353–57.

    PubMed  CAS  Google Scholar 

  66. Rao CN, Réddy P, Liu YY et al. Extrecellular matrix-associated serine protease inhibitors (Mr 33000, 31000 and 27000) are single-gene products with differential glycosylation: cDNA cloning of the 33-kDa inhibitor reveals its identity to tissue factor pathway inhibitor-2. Arch Biochem Biophys 1996; 335: 82–92.

    PubMed  CAS  Google Scholar 

  67. Broze GJ. Tissue factor pathway inhibitor and the revised theory of coagulation. Annu Rev Med 1995; 46: 103–12.

    PubMed  CAS  Google Scholar 

  68. Konduri SD, Tasiou A, Chandrasekar N et al. Overexpression of tissue factor pathway inhibitor-2 (TFPI-2) decreases the invasiveness of prostate cancer cells in vitro. Int J Oncol 2001; 18: 127-31.

    PubMed  CAS  Google Scholar 

  69. Taniguchi T, 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 1998; 58: 4461–7.

    PubMed  CAS  Google Scholar 

  70. Kung C, Hayes E, Mann KG. A membrane-mediated catalytic event in prothrombin activation. J Biol Chem 1994; 269: 25838–48.

    PubMed  CAS  Google Scholar 

  71. Mosesson MW. The roles of fibrinogen and fibrin in hemostasis and thrombosis. Semin Hematol 1992; 29: 177–88.

    PubMed  CAS  Google Scholar 

  72. Herbert JM, Dupuy E, Laplace MC et al. Thrombin induces endothelial cell growth via both proteolytic and non-proteolytic pathway. Biochem J 1994; 303: 227–31.

    PubMed  CAS  Google Scholar 

  73. McNamara CA, Sarembock IJ, Gimple LW et al. Thrombin stimulates proliferation of eultured rat aortie smooth muscle cells by a proteolytically activated receptor. J Clin Invest 1993; 91: 94–8.

    PubMed  CAS  Google Scholar 

  74. Macfarlane SR, Seatter MJ, Kanke T et al. Proteinase-activated receptors. Pharmacol Rev 2001; 53: 245–82.

    PubMed  CAS  Google Scholar 

  75. Scarborough RM, Naughton MA, Teng W et al. Tethered ligand agonist peptides. Structural requirements for thrombin receptor activation reveal mechanism of proteolytic unmasking of agonist function. J Biol Chem 1992; 267: 13146–9.

    PubMed  CAS  Google Scholar 

  76. Dery O, Corvera CU, Steinhoff M, et al. Proteinase-activated receptors: Novel mechanisms of signaling by serine proteases. Am J Physiol 1998; 274(Cell Physiol 43): C1429–52.

    PubMed  CAS  Google Scholar 

  77. Tsopanoglou NE, Pipilisynetos E, Maragoudakis ME. Thrombin promotes angiogenesis by a mechanism independent of fibrin formation. Am J Physiol 1993; 264: C1302–7.

    PubMed  CAS  Google Scholar 

  78. Haralabopoulos GC, Grant DS, Kleinman HK et al. Thrombin promotes endothelial cell alignement in matrigel in vitro and angiogenesis in vivo. Am J Cell Physiol 1997; 42: C239–45.

    Google Scholar 

  79. Maragoudakis ME, Tsopanoglou NE, Andriopolou P et al. Effects of thrombin/thrombosis in angiogenesis and tumor progression. Matrix Biol 2000; 19: 345–51.

    PubMed  CAS  Google Scholar 

  80. Bachhuber BG, Sarembock IJ, Gimple LW et al. Alpha-thrombin induces transforming growth factor-beta(1) mRNA and protein in cultured vascular smooth muscle cells via a proteolytically activated receptor. Endocrinology 1990; 34: 41–8.

    Google Scholar 

  81. Zucker S, Conner C, Di Massimo BI et al. Thrombin induces the activation of progelatinase A in vascular endothelial cells. Physiologic regulation of angiogenesis. J Biol Chem 1995; 270: 23730–8.

    PubMed  CAS  Google Scholar 

  82. Nguyen M, Arkell J, Jackson CJ. Thrombin rapidly and efficiently activates gelatinase A in human microvascular endothelial cells via a mechanism independent of active MT1 matrix metalloproteinase. Lab Invest 1999; 79: 467–75.

    PubMed  CAS  Google Scholar 

  83. Chiarugi VP, Magnelli L, Dello Sbarba P et al. Tumor angiogenesis: Thrombin and metalloproteinases in focus. Exp Mol Pathol 2000; 69: 63–6.

    PubMed  CAS  Google Scholar 

  84. Wojtukiewicz MZ, Tang DG, Benjosef E et al. Solid tumor cells express functional tethered ligand threombin receptor. Cancer Res 1995; 55: 698–704.

    PubMed  CAS  Google Scholar 

  85. Zacharski LR, Memoli VA, Morain WD et al. Cellular localization of enzymatically active thrombin in intact human tissues by hirudin binding. Thromb Haemostasis 1995; 73: 793–7.

    CAS  Google Scholar 

  86. Even-Ram S, Uziely B, Cohen P et al. Thrombin receptor overexpression in malignant and physiological invasion processes. Nature Med 1998; 4: 909–14.

    PubMed  CAS  Google Scholar 

  87. Henrikson KP, Salazar SL, Fenton JW et al. Role of thrombin receptor in breast cancer invasiveness, Br J Cancer 1999; 79: 401–6.

    PubMed  CAS  Google Scholar 

  88. Klepfish A, Greco MA, Karpatkin S. Thrombin stimulates melanoma tumor cell binding to endothelial cells and subendothelial matrix. Int J Cancer 1993; 53: 978–82.

    PubMed  CAS  Google Scholar 

  89. Wojtukiewicz MZ, Tang DG, Ciarelli JJ et al. Thrombin increases the metastatic potential of tumor cells. Int J Cancer 1993; 54: 793–806.

    PubMed  CAS  Google Scholar 

  90. Nierodzik ML, Klepfish A, Karpatkin S. Role of platelets, thrombin, integrin IIb-IIIa, fibronectin and Von Willenrand factor on tumor adhesion in vitro and metastasis in vivo. Thromb Haemostasis 1995; 74: 282–290.

    CAS  Google Scholar 

  91. Dardik R, Savion N, Kaufman Y et al. Thrombin promotes platelet-mediated melanoma cell adhesion to endothelial cells under flow conditions: Role of platelet glycoproteins P-selectin and GPIIb-IIIa. Br J Cancer 1998; 77: 2069–75.

    PubMed  CAS  Google Scholar 

  92. Even-Ram SC, Maoz M, Pokroy E et al. Tumor cell invasion is promoted by activation of protease activated receptor-1 in cooperation with the α y β 5 integrin. J Biol Chem 2001; 276: 10952–62.

    PubMed  CAS  Google Scholar 

  93. Miyata S, Kishikawa N, Yasumitsu H et al. Trypsin stimulates integrin alpha(5)beta(1)-dependent adhesion to fibronectin and proliferation of human gastric carcinoma cells through activation of proteinase-activated receptor-2. J Biol Chem 2000; 275: 4592–8.

    PubMed  CAS  Google Scholar 

  94. Noda-Heiny H, Sobel BE. Vascular smooth muscle cell migration mediated by thrombin and urokinase receptor. Am J Physiol 1995; 268: C1195–201.

    PubMed  CAS  Google Scholar 

  95. Yoshida E, Verrusio EN, Mihara H et al. Enhancement of the expression of urokinase-type plasminogen activator from PC-3 human prostate cancer cells by thrombin. Cancer Res 1994; 54: 3300–34.

    PubMed  CAS  Google Scholar 

  96. Esmon CT. The roles of protein C and thrombomodulin in the regulation of blood coagulation. J Biol Chem 1989; 264: 4743–6.

    PubMed  CAS  Google Scholar 

  97. Jackson DE, Teras TJ, Salem HH et al. Purification and characterization of two forms of soluble TM from human urine. Eur J Biochem 1994; 221: 1079–87.

    PubMed  CAS  Google Scholar 

  98. Suehiro T, Shimada M, Matsumata A et al. Thrombomodulin inhibits intrahepatic spread in human hepatocellular carcinoma. Hepatology 1995; 21: 1285–90.

    PubMed  CAS  Google Scholar 

  99. Tabata M, Sugihara K, Tonezawa S et al. An immunohistochemical study of thrombomodulin in oral squamous cell carcinoma and its association with invasive and metastatic potential. J Oral Pathol Med 1997; 26: 258–64.

    PubMed  CAS  Google Scholar 

  100. Imada S, Yamaguchi H, Nagumo M et al. Identification of fetomodulin, a surface marker protein of fetal development, as thrombomodulin by gene cloning and function assay. Dev Biol 1990; 140: 113–22.

    PubMed  CAS  Google Scholar 

  101. Hosaka Y, Higuchi T, Tsumagari M et al. Inhibition of invasion and experimental metastasis of murine melanoma cells by human soluble thrombomodulin. Cancer Lett 2000; 161: 231–40.

    PubMed  CAS  Google Scholar 

  102. Hooper JD, Clements JA, Quigley JP et al. Type II transmembrane serine proteases. Insights into an emerging class of cell surface proteolytic enzymes. J Biol Chem 2001; 276: 857–60.

    PubMed  CAS  Google Scholar 

  103. Takeuchi T, Harris J, Huang W et al. Cellular localization of membrane-type serine protease 1 and identification of protease activated receptor-2 and single chain urokinase-type plasminogen activator as substrates. J Biol Chem 2000; 275: 26333–42.

    PubMed  CAS  Google Scholar 

  104. Yan W, Sheng N, Seto M et al. Corin, a mosaic transmembrane serine protease encoded by a novel cDNA from human heart. J Biol Chem 1999; 274: 14926–35.

    PubMed  CAS  Google Scholar 

  105. Torres-Rosado A, O'Shea KS, Tsuji A et al. Hepsin, a putative cell-surface serine protease, is required for mammalian cell growth. Proc Natl Acad Sci USA 1993; 90: 7181–5.

    PubMed  CAS  Google Scholar 

  106. Tanimoto H, Yan Y, Clarke J et al. Hepsin, a cell surface serine protease identified in hepatoma cells, is overexpressed in ovarian cancer. Cancer res 1997; 57: 2884–7.

    PubMed  CAS  Google Scholar 

  107. Lin CY, Anders J, Johnson M et al. Purification and characterization of a complex containing matriptase and a Kunitz-type serine protease inhibitor from human milk. J Biol Chem 1999; 274: 18237–42.

    PubMed  CAS  Google Scholar 

  108. Ariga N, Sato E, Ohuchi N et al. Stromal expression of fibroblast activation protein/seprase, a cell membrane serine proteinase and gelatinase, is associated with longer survival in patients with invasive ductal carcinoma of breast. Int J Cancer 2001; 95: 67–72.

    PubMed  CAS  Google Scholar 

  109. Lin B, Ferguson C, White JT et al. Prostate-localized and androgen-regulated expression of the membrane-bound serine protease TM-PRSS2. Cancer Res 1999; 59: 4180–4.

    PubMed  CAS  Google Scholar 

  110. Wallrapp C, Hahnel S, Muller-Pillasch F et al. A novel transmembrane serine protease (TMPRSS3) overexpressed in pancreatic cancer. Cancer Res 2000; 60: 2602–6.

    PubMed  CAS  Google Scholar 

  111. Kong W, McConalogue K, Khitin LM et al. Luminal trypsin may regulate enterocytes through proteinase-activated receptor 2. Proc Natl Acad Sci USA 1997; 94: 8884–9.

    PubMed  CAS  Google Scholar 

  112. Kazama Y, Hamamoto T, Foster DC et al. Hepsin, a putative membrane-associated serine protease, activates human factor VII and initiates a pathway of blood coagulation on the cell surface leading to thrombin formation. J Biol Chem 1995; 270: 66–72.

    PubMed  CAS  Google Scholar 

  113. Lee SL, Dickson RB, Lin CY. Activation of hepatocyte growth factor and urokinase/plasminogen activator by matriptase, an epithelial membrane serine protease. J Biol Chem 2000; 275: 36720–5.

    PubMed  CAS  Google Scholar 

  114. Pineiro-Sanchez ML, Goldstein LA, Dodt J et al. Identification of the 170-kDa melanoma membrane-bound gelatinase (seprase) as a serine integral membrane protease. J Biol Chem 1997; 272: 7595–601.

    PubMed  CAS  Google Scholar 

  115. Park JE, Lenter MC, Rainer MC et al. Fibroblast activation protein, a dual specificity serine protease expressed in reactive human tumor stromal fibroblasts. J Biol Chem 1999; 274: 36505–12.

    PubMed  CAS  Google Scholar 

  116. Monski WL, Lin CY, Aoyama A et al. A potential marker protease of invasiveness, seprase, is localized on invadopodia of human malignant melanoma cells. Cancer Res 1994; 54: 5702–10.

    Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Mario Del Rosso.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Del Rosso, M., Fibbi, G., Pucci, M. et al. Multiple pathways of cell invasion are regulated by multiple families of serine proteases. Clin Exp Metastasis 19, 193–207 (2002). https://doi.org/10.1023/A:1015531321445

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1023/A:1015531321445

Navigation