Thromb Haemost 2000; 84(05): 871-875
DOI: 10.1055/s-0037-1614130
Review Article
Schattauer GmbH

Effect of Stabilizing versus Destabilizing Interactions on Plasminogen Activator Inhibitor-1

Nele Vleugels
1   From the Laboratory for Pharmaceutical Biology and Phytopharmacology, Faculty of Pharmaceutical Sciences, Katholieke Universiteit Leuven, Belgium
,
John Leys
1   From the Laboratory for Pharmaceutical Biology and Phytopharmacology, Faculty of Pharmaceutical Sciences, Katholieke Universiteit Leuven, Belgium
,
Isabelle Knockaert
1   From the Laboratory for Pharmaceutical Biology and Phytopharmacology, Faculty of Pharmaceutical Sciences, Katholieke Universiteit Leuven, Belgium
,
Paul J. Declerck
1   From the Laboratory for Pharmaceutical Biology and Phytopharmacology, Faculty of Pharmaceutical Sciences, Katholieke Universiteit Leuven, Belgium
› Author Affiliations
This work was supported in part by a grant from the Fund for Scientific Research (F.W.O.-Vlaanderen, project G.0266.97) and by a grant from the Research Fund K.U. Leuven (OT/98/37). N.V. is a research assistant of the Fund for Scientific Research (F.W.O.-Vlaanderen).
We are grateful to Dr. A. Rabijns (Laboratory of Analytical Chemistry and Medical Physicochemistry, KULeuven, Belgium) for assistance in the preparation of Fig. 1. and to Mrs I. Christ and Dr. J. Stassen (Cardiovascular Department, Boehringer Ingelheim Pharma KG, Biberach, Germany) for the measurement of the affinity constants.
Further Information

Publication History

Received 24 November 1999

Accepted after resubmission 26 June 2000

Publication Date:
13 December 2017 (online)

Summary

Plasminogen activator inhibitor-1 (PAI-1) is a unique member of the serpin family, as it spontaneously converts into a latent conformation. However, the exact mechanism of this conversion is not known. Previous studies reported that neutralizing monoclonal antibodies as well as reversal or removal of charges on the s3C-s4C turn results in a destabilization of PAI-1 leading to an accelerated conversion to its latent form.

In this study the effect of the reversal or removal of charges in this “gate region” (R186E/R187E, H190E/K191E, H190L/K191L and R356E) on a stable PAI-1-variant (PAI-1-stab) was investigated. Whereas PAI-1-stab has a half-life of 150 ± 66 h, PAI-1-stab-R186ER187E, PAI-1-stab-H190E-K191E, PAI-1-stab-H190L-K191L and PAI-1-stab-R356E have a strongly decreased half-life (p< 0.005 versus PAI-1-stab) of 175 ± 48 min, 75 ± 34 min, 68 ± 38 min and 79 ± 16 min, respectively. Wild-type PAI-1 (wtPAI-1) had a half-life of 55 ± 19 min. These data indicate that the stabilization induced by the mutated residues in PAI-1-stab is counteracted by the additional mutations, resulting in half-lives similar to that of wtPAI-1, thereby suggesting that the stabilizing and destabilizing forces act mainly independently in these mutants. Extrapolation of these data to other (stable) serpins leads to the hypothesis that the s3C-s4C turn and the distal hinge region of the reactive site loop plays a role for the stability of serpins in general.

 
  • References

  • 1 Pannekoek H, Veerman H, Lambers H, Diergaarde P, Verweij CL, van Zonneveld AJ, van Mourik JA. Endothelial plasminogen activator inhibitor (PAI): a new member of the Serpin gene family. EMBO J 1986; 05: 2539-44.
  • 2 Ny T, Sawdey M, Lawrence D, Millan JL, Loskutoff DJ. Cloning and sequence of a cDNA coding for the human beta-migrating endothelial-celltype plasminogen activator inhibitor. Proc Natl Acad Sci USA 1986; 83: 6776-80.
  • 3 Declerck PJ, De Mol M, Vaughan DE, Collen D. Identification of a conformationally distinct form of plasminogen activator inhibitor-1, acting as a noninhibitory substrate for tissue-type plasminogen activator. J Biol Chem 1992; 267: 11693-6.
  • 4 Mottonen J, Strand A, Symersky J, Sweet RM, Danley DE, Geoghegan KF, Gerard RD, Goldsmith EJ. Structural basis of latency in plasminogen activator inhibitor-1. Nature 1992; 355: 270-3.
  • 5 Hekman CM, Loskutoff DJ. Endothelial cells produce a latent inhibitor of plasminogen activators that can be activated by denaturants. J Biol Chem 1985; 260: 11581-7.
  • 6 Berkenpas MB, Lawrence DA, Ginsburg D. Molecular evolution of plasminogen activator inhibitor-1 functional stability. EMBO J 1995; 14: 2969-77.
  • 7 Gils A, Lu J, Aertgeerts K, Knockaert I, Declerck PJ. Identification of positively charged residues contributing to the stability of plasminogen activator inhibitor 1. FEBS Lett 1997; 415: 192-5.
  • 8 Aertgeerts K, De Bondt HL, De Ranter CJ, Declerck PJ. Mechanisms contributing to the conformational and functional flexibility of plasminogen activator inhibitor-1. Nat Struct Biol 1995; 02: 891-7.
  • 9 Stanssens P, Opsomer C, McKeown YM, Kramer W, Zabeau M, Fritz HJ. Efficient oligonucleotide-directed construction of mutations in expression vectors by the gapped duplex DNA method using alternating selectable markers. Nucleic Acids Res 1989; 17: 4441-54.
  • 10 Audenaert AM, Knockaert I, Collen D, Declerck PJ. Conversion of plasminogen activator inhibitor-1 from inhibitor to substrate by point mutations in the reactive-site loop. J Biol Chem 1994; 269: 19559-64.
  • 11 Vleugels N, Gils A, Mannaerts S, Knockaert I, Declerck PJ. Evaluation of the mechanism of inactivation of plasminogen activator inhibitor-1 by monoclonal antibodies using a stable variant. Fibrinolysis & Proteolysis 1998; 12: 277-82.
  • 12 Gils A, Knockaert I, Declerck PJ. Substrate behavior of plasminogen activator inhibitor-1 is not associated with a lack of insertion of the reactive site loop. Biochemistry 1996; 35: 7474-81.
  • 13 Ngo TH, Declerck PJ. Immunological quantitation of rabbit plasminogen activator inhibitor-1 in biological samples. Evidence that rabbit platelets do not contain PAI-1. Thromb Haemost 1999; 82: 1510-5.
  • 14 Debrock S, Declerck PJ. Neutralization of plasminogen activator inhibitor-1 inhibitory properties: identification of two different mechanisms. Biochim Biophys Acta 1997; 1337: 257-66.
  • 15 Verheijen JH, Chang GT, Kluft C. Evidence for the occurrence of a fastacting inhibitor for tissue-type plasminogen activator in human plasma. Thromb Haemost 1984; 51: 392-5.
  • 16 Jönsson U, Malmqvist M. Real time biospecific interaction analysis. The integration of surface plasmon resonance detection, general biospecific interface chemistry and microfluidics into one analytical system. In: Advances in biosensors. JAI Press; 1992: 291-336.
  • 17 Egelund R, Schousboe SL, Sottrupjensen L, Rodenburg KW, Andreasen PA. Type 1 plasminogen activator inhibitor: conformational differences between latent, active, reactive centre cleaved and plasminogen activator complexed forms, as probed by proteolytic susceptibility. Eur J Biochem 1997; 248: 775-85.
  • 18 Urano T, Strandberg L, Johansson LB, Ny T. A substrate-like form of plasminogen-activator-inhibitor type 1. Conversions between different forms by sodium dodecyl sulphate. Eur J Biochem 1992; 209: 985-92.
  • 19 Munch M, Heegaard CW, Andreasen PA. Interconversions between active, inert and substrate forms of denatured/refolded type-1 plasminogen activator inhibitor. Biochim Biophys Acta 1993; 1202: 29-37.
  • 20 Gils A, Declerck PJ. Proteinase specificity and functional diversity in point mutants of plasminogen activator inhibitor 1. J Biol Chem 1997; 272: 12662-6.
  • 21 Gils A, Knockaert I, Declerck PJ. Construction and characterization of plasminogen activator inhibitor-1 mutants in which part of the active site loop is deleted. Fibrinolysis & Proteolysis 1997; 11 (5/6): 265-71.
  • 22 Madison EL, Goldsmith EJ, Gething MJ, Sambrook JF, Gerard RD. Restoration of serine protease-inhibitor interaction by protein engineering. J Biol Chem 1990; 265: 21423-6.
  • 23 Lawrence DA, Olson ST, Palaniappan S, Ginsburg D. Serpin reactive center loop mobility is required for inhibitor function but not for enzyme recognition. J Biol Chem 1994; 269: 27657-62.
  • 24 Stein PE, Carrell RW. What do dysfunctional serpins tell us about molecular mobility and disease?. Nat Struct Biol 1995; 02: 96-113.
  • 25 Chang WS, Whisstock J, Hopkins PC, Lesk AM, Carrell RW, Wardell MR. Importance of the release of strand 1C to the polymerization mechanism of inhibitory serpins. Protein Sci 1997; 06: 89-98.
  • 26 Nar H, Bauer M, Stassen JM, Lang D, Gils A, Declerck PJ. Plasminogen activator inhibitor-1:structure of the native serpin, comparison to its other conformers and implications for serpin inactivation. J Mol Biol 2000; 297: 683-95.
  • 27 Sharp AM, Stein PE, Pannu NS, Carrell RW, Berkenpas MB, Ginsburg D, Lawrence DA, Read RJ. The active conformation of plasminogen activator inhibitor 1, a target for drugs to control fibrinolysis and cell adhesion. Structure 1999; 07: 111-8.
  • 28 Schechter NM, Jordan LM, James AM, Cooperman BS, Wang ZM, Rubin H. Reaction of human chymase with reactive site variants of alpha 1-antichymotrypsin. Modulation of inhibitor versus substrate properties. J Biol Chem 1993; 268: 23626-33.
  • 29 Gils A, Declerck PJ. Structure-function relationship in serpins: current concepts and controversies. Thromb Haemost 1998; 80: 531-41.