PMID- 8808830 OWN - NLM STAT- MEDLINE DCOM- 19961203 LR - 20161123 IS - 0300-9165 (Print) IS - 0300-9165 (Linking) VI - 48 IP - 8 DP - 1996 Aug TI - [Mechanism of tumor cell-induced extracellular matrix degradation--inhibition of cell-surface proteolytic activity might have a therapeutic effect on tumor cell invasion and metastasis]. PG - 623-32 AB - Tumor cells produce urokinase-type plasminogen activator (uPA) in an enzymatically inactive proenzyme form (pro-uPA). Secreted pro-uPA can immediately bind to the specific uPA receptors (uPAR) on tumor cell surface with high affinity. The uPAR specifically recognizes enzymatically inactive pro-uPA and active high molecular weight-uPA (HMW-uPA) by their growth factor-like terminal domain. uPAR is a glycoprotein of approximately 55 kDa; the affinity for uPA is high (0.2 nM) and the rate of dissociation is low. Receptor-bound uPA catalizes the formation of plasmin on the cell surface to generate the proteolytic cascade that contributes to the breakdown of basement membrane and extracellular matrix. The plasma membrane uPAR has attracted considerable attention because of its role in migration and tissue invasion by mononuclear phagocytes and malignant cells. In some cell types uPAR localizes uPA to cell-cell and cell-substratum contact sites, providing the possibility of a directional proteolysis that may be involved in cell migration and invasion. Recently it has been reported that competitive displacement of uPA from uPAR resulted in decreased proteolysis, suggesting that the cell surface is the preferred site for uPA-mediated protein degradation. Various very different approaches to interfere with the expression or reactivity of uPA or uPAR at the gene or protein level were successfully tested including antisense oligonucleotides, antibodies, inhibitors and recombinant or synthetic uPA and uPAR analogues. Recently we have reported that a highly purified urinary trypsin inhibitor (UTI) efficiently inhibits soluble and tumor cell-surface receptor-bound plasmin. UTI inhibits not only tumor cell invasion in an in vitro assay but also production of experimental and spontaneous lung metastasis in an in vivo mouse model. The anti-invasive effect is dependent on the anti-plasmin activity of UTI. UTI peptide, which inhibits plasmin activity, synthesized by an automated peptide synthesizer showed mouse 3LL cell invasion inhibitory activity. UTI and the effective peptide inhibited tumor cell invasion through Matrigel. UTI did not inhibit tumor cell proliferation or the binding of the cells to Matrigel. Also, UTI did not inhibit chemotactic migration of tumor cells to fibronectin. It is likely that UTI acts as a protease inhibitor. We attempted to synthesize conjugates between ATF and UTI. Thus, conjugating a physiological plasmin inhibitor to ATF might target it to reduce cell-associated proteolytic activity to the close environment of the uPAR-expressing tumor cell surface and subsequently may effectively inhibit tumor cell invasion and metastasis, because the cell surface uPAR might be a critical component of the metastatic machinery. A method of conjugation of the UTI domain II (HI-8), to the receptor-binding amino-terminal fragment (ATF) of uPA has been developed utilizing the heterobifunctional cross-linking reagent, N-succinimidyl-3-(2-pyridyldithio) propionate (SPDP). The conjugate retained its protease inhibiting activity and showed a binding reactivity to uPAR on the surface of tumor cells. We have shown that the conjugate exhibits plasmin inhibition to the close environment of the cell surface and subsequently inhibits the tumor cell invasion through Matrigel in an in vitro invasion assay. In order to extend our idea, we attempt to produce a novel hybrid molecule consisting of the ATF of uPA placed at the N-terminus of UTI domain II (HI-8) by protein engineering techniques. Exogenously applied ATFHI hybrid protein can immediately bind to the specific uPAR on cell surfaces with high affinity. The receptor-bound hybrid protein focuses the protease-inhibiting activity to the tumor cell surface. This is effectively a bifunctional molecule which, in addition to inhibiting trypsin and plasmin activities directly, is able toblock unoccupied uPAR, thereby preventing localization of uPA activity. FAU - Kobayashi, H AU - Kobayashi H AD - Department of Obstetrics and Gynecology, Hamamatsu University School of Medicine. LA - jpn PT - Journal Article PT - Review PL - Japan TA - Nihon Sanka Fujinka Gakkai Zasshi JT - Nihon Sanka Fujinka Gakkai zasshi JID - 7505749 RN - 0 (Glycoproteins) RN - 0 (Plasminogen Activator Inhibitor 1) RN - EC 3.4.21.73 (Urokinase-Type Plasminogen Activator) RN - OR3S9IF86U (urinastatin) SB - IM MH - Animals MH - Extracellular Matrix/*metabolism MH - Female MH - Glycoproteins/pharmacology/physiology/therapeutic use MH - Humans MH - Mice MH - Neoplasm Invasiveness/prevention & control MH - Neoplasm Metastasis/prevention & control MH - Plasminogen Activator Inhibitor 1/metabolism MH - Urokinase-Type Plasminogen Activator/metabolism MH - Uterine Cervical Neoplasms/drug therapy/*metabolism/*pathology RF - 14 EDAT- 1996/08/01 00:00 MHDA- 1996/08/01 00:01 CRDT- 1996/08/01 00:00 PHST- 1996/08/01 00:00 [pubmed] PHST- 1996/08/01 00:01 [medline] PHST- 1996/08/01 00:00 [entrez] PST - ppublish SO - Nihon Sanka Fujinka Gakkai Zasshi. 1996 Aug;48(8):623-32.