Thromb Haemost 2017; 117(07): 1338-1347
DOI: 10.1160/TH16-11-0891
Coagulation and Fibrinolysis
Schattauer GmbH

A long-acting PAI-1 inhibitor reduces thrombus formation

Shuangzhou Peng
1   State Key Laboratory of Structural Chemistry and Danish-Chinese Centre for Proteases and Cancer, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, Fujian, China
2   College of life sciences, Fujian Agriculture and Forestry University, Fuzhou, Fujian, China
,
Guangpu Xue
1   State Key Laboratory of Structural Chemistry and Danish-Chinese Centre for Proteases and Cancer, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, Fujian, China
5   College of Life science, Fujian Normal University, Fuzhou, Fujian, China
,
Lihu Gong
1   State Key Laboratory of Structural Chemistry and Danish-Chinese Centre for Proteases and Cancer, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, Fujian, China
,
Chao Fang
6   Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
7   School of Basic Medicine, Tongji Medical College, Huazhong University of Science & Technology, Huazhong, China
,
Jingfei Chen
4   Qingdao Institute of Biomass Energy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, China
,
Cai Yuan
3   Fuzhou University,Fuzhou, Fujian, China
,
Zhuo Chen
1   State Key Laboratory of Structural Chemistry and Danish-Chinese Centre for Proteases and Cancer, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, Fujian, China
,
Lishan Yao
4   Qingdao Institute of Biomass Energy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, China
,
Bruce Furie
6   Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
,
Mingdong Huang
1   State Key Laboratory of Structural Chemistry and Danish-Chinese Centre for Proteases and Cancer, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, Fujian, China
3   Fuzhou University,Fuzhou, Fujian, China
› Institutsangaben
Weitere Informationen

Publikationsverlauf

Received: 28. November 2016

Accepted after major revision: 25. März 2017

Publikationsdatum:
11. November 2017 (online)

Summary

Plasminogen activator inhibitor 1 (PAI-1) is the main inhibitor of tissue-type and urokinase-type plasminogen activators (t/uPA) and plays an important role in fibrinolysis. Inhibition of PAI-1 activity prevents thrombosis and accelerates fibrinolysis, indicating that PAI-1 inhibitors may be used as effective antithrombotic agents. We previously designed a PAI-1 inhibitor (PAItrap) which is a variant of inactivated urokinase protease domain. In the present study, we fused PAItrap with human serum albumin (HSA) to develop a long-acting PAI-1 inhibitor. Unfortunately, the fusion protein PAItrap-HSA lost some potency compared to PAItrap (33 nM vs 10 nM). Guided by computational method, we carried out further optimisation to enhance inhibitory potency for PAI-1. The new PAItrap, denominated PAItrap(H37R)-HSA, which was the H37R variant of PAItrap fused to HSA, gave a six-fold improvement of IC50 (5 nM) for human active PAI-1 compared to PAItrap-HSA, and showed much longer plasma half-life (200-fold) compared to PAItrap. We further demonstrated that the PAItrap(H37R)-HSA inhibited exogenous or endogenous PAI-1 to promote fibrinolysis in fibrin-clot lysis assay. PAItrap(H37R)-HSA inhibits murine PAI-1 with IC50 value of 12 nM, allowing the inhibitor to be evaluated in murine models. Using an intravital microscopy, we demonstrated that PAItrap(H37R)-HSA blocks thrombus formation and platelet accumulation in vivo in a laser-induced vascular injury mouse model. Additionally, mouse tail bleeding assay showed that PAItrap(H37R)-HSA did not affect the global haemostasis. These results suggest that PAItrap(H37R)-HSA have the potential benefit to prevent thrombosis and accelerates fibrinolysis.

 
  • References

  • 1 Ha H, Oh EY, Lee HB. The role of plasminogen activator inhibitor 1 in renal and cardiovascular diseases. Nat Rev Nephrol 2009; 5: 203-211.
  • 2 Schulman S, Wiman B. The significance of hypofibrinolysis for the risk of recurrence of venous thromboembolism. Duration of Anticoagulation (DURAC) Trial Study Group. Thromb Haemost 1996; 75: 607-611.
  • 3 Meade TW, Ruddock V, Stirling Y. et al. Fibrinolytic activity, clotting factors, and long-term incidence of ischaemic heart disease in the Northwick Park Heart Study. Lancet 1993; 342: 1076-1079.
  • 4 Hamsten A, de Faire U, Walldius G. et al. Plasminogen activator inhibitor in plasma: risk factor for recurrent myocardial infarction. Lancet 1987; 2: 3-9.
  • 5 Fernandez-Cadenas I, Alvarez-Sabin J, Ribo M. et al. Influence of thrombin-activatable fibrinolysis inhibitor and plasminogen activator inhibitor-1 gene polymorphisms on tissue-type plasminogen activator-induced recanalization in ischemic stroke patients. J Thromb Haemost 2007; 5: 1862-1868.
  • 6 Eren M, Painter CA, Atkinson JB. et al. Age-dependent spontaneous coronary arterial thrombosis in transgenic mice that express a stable form of human plasminogen activator inhibitor-1. Circulation 2002; 106: 491-496.
  • 7 Erickson LA, Fici GJ, Lund JE. et al. Development of Venous Occlusions in Mice Transgenic for the Plasminogen-Activator Inhibitor-1 Gene. Nature 1990; 346: 74-76.
  • 8 Kawasaki T, Dewerchin M, Lijnen HR. et al. Vascular release of plasminogen activator inhibitor-1 impairs fibrinolysis during acute arterial thrombosis in mice. Blood 2000; 96: 153-160.
  • 9 Eitzman DT, Westrick RJ, Nabel EG. et al. Plasminogen activator inhibitor-1 and vitronectin promote vascular thrombosis in mice. Blood 2000; 95: 577-580.
  • 10 Farrehi PM, Ozaki CK, Carmeliet P. et al. Regulation of arterial thrombolysis by plasminogen activator inhibitor-1 in mice. Circulation 1998; 97: 1002-1008.
  • 11 Carmeliet P, Stassen JM, Schoonjans L. et al. Plasminogen activator inhibitor-1 gene-deficient mice. II. Effects on hemostasis, thrombosis, and thrombolysis. J Clin Invest 1993; 92: 2756-2760.
  • 12 Eitzman DT, Westrick RJ, Xu ZJ. et al. Plasminogen activator inhibitor-1 deficiency protects against atherosclerosis progression in the mouse carotid artery. Blood 2000; 96: 4212-4215.
  • 13 Fay WP, Parker AC, Condrey LR. et al. Human plasminogen activator inhibitor-1 (PAI-1) deficiency: Characterization of a large kindred with a null mutation in the PAI-1 gene. Blood 1997; 90: 204-208.
  • 14 Fay WP, Shapiro AD, Shih JL. et al. Brief Report - Complete Deficiency of Plasminogen-Activator Inhibitor Type-1 Due to a Frame-Shift Mutation. N Engl J Med 1992; 327: 1729-1733.
  • 15 Wu Q, Zhao Z. Inhibition of PAI-1: a new anti-thrombotic approach. Curr Drug Targets Cardiovasc Haematol Dis 2002; 2: 27-42.
  • 16 Lin ZH, Jensen JK, Hong ZB. et al. Structural Insight into Inactivation of Plasminogen Activator Inhibitor-1 by a Small-Molecule Antagonist. Chem Biol 2013; 20: 253-261.
  • 17 Rouch A, Vanucci-Bacque C, Bedos-Belval F. et al. Small molecules inhibitors of plasminogen activator inhibitor-1-An overview. Eur J Med Chem 2015; 92: 619-636.
  • 18 Mathiasen L, Dupont DM, Christensen A. et al. A peptide accelerating the conversion of plasminogen activator inhibitor-1 to an inactive latent state. Mol Pharmacol 2008; 74: 641-653.
  • 19 Komissarov AA, Andreasen PA, Bodker JS. et al. Additivity in effects of vitronectin and monoclonal antibodies against alpha-helix F of plasminogen activator inhibitor-1 on its reactions with target proteinases. J Biol Chem 2005; 280: 1482-1489.
  • 20 Zhou XH, Hendrickx MLV, Hassanzadeh-Ghassabeh G. et al. Generation and in vitro characterisation of inhibitory nanobodies towards plasminogen activator inhibitor 1. Thromb Haemost 2016; 116: 1032-1040.
  • 21 Damare J, Brandal S, Fortenberry YM. Inhibition of PAI-1 Antiproteolytic Activity Against tPA by RNA Aptamers. Nucleic Acid Ther 2014; 24: 239-249.
  • 22 Thompson LC, Goswami S, Ginsberg DS. et al. Metals affect the structure and activity of human plasminogen activator inhibitor-1. I. Modulation of stability and protease inhibition. Protein Sci 2011; 20: 353-365.
  • 23 Bucci JC, Trelle MB, McClintock CS. et al. Copper(II) Ions Increase Plasminogen Activator Inhibitor Type 1 Dynamics in Key Structural Regions That Govern Stability. Biochemistry 2016; 55: 4386-4398.
  • 24 Levi M, Biemond BJ, Vanzonneveld AJ. et al. Inhibition of Plasminogen-Activator Inhibitor-1 Activity Results in Promotion of Endogenous Thrombolysis and Inhibition of Thrombus Extension in Models of Experimental Thrombosis. Circulation 1992; 85: 305-312.
  • 25 Izuhara Y, Yamaoka N, Kodama H. et al. A novel inhibitor of plasminogen activator inhibitor-1 provides antithrombotic benefits devoid of bleeding effect in nonhuman primates. J Cerebr Blood F Met 2010; 30: 904-912.
  • 26 Gong LH, Proulle V, Fang C. et al. A specific plasminogen activator inhibitor-1 antagonist derived from inactivated urokinase. J Cell Mol Med 2016; 20: 1851-1860.
  • 27 Berkenpas MB, Lawrence DA, Ginsburg D. Molecular Evolution of Plasminogen-Activator Inhibitor-1 Functional Stability. Embo J 1995; 14: 2969-2977.
  • 28 Cochran BJ, Gunawardhana LP, Vine KL. et al. The CD-loop of PAI-2 (SERPINB2) is redundant in the targeting, inhibition and clearance of cell surface uPA activity. Bmc Biotechnol 2009; 9: 43.
  • 29 Lin ZH, Jiang LG, Yuan C. et al. Structural Basis for Recognition of Urokinase-type Plasminogen Activator by Plasminogen Activator Inhibitor-1. J Biol Chem 2011; 286: 7027-7032.
  • 30 Guerois R, Nielsen JE, Serrano L. Predicting changes in the stability of proteins and protein complexes: A study of more than 1000 mutations. J Mol Biol 2002; 320: 369-387.
  • 31 Hess B, Kutzner C, van der Spoel D. et al. GROMACS 4: Algorithms for highly efficient, load-balanced, and scalable molecular simulation. J Chem Theory Comput 2008; 4: 435-447.
  • 32 Essmann U, Perera L, Berkowitz ML. et al. A Smooth Particle Mesh Ewald Method. J Chem Phys 1995; 103: 8577-8593.
  • 33 Hess B, Bekker H, Berendsen HJC. et al. LINCS: A linear constraint solver for molecular simulations. J Comput Chem 1997; 18: 1463-1472.
  • 34 Martins SA, Perez MAS, Moreira IS. et al. Computational Alanine Scanning Mutagenesis: MM-PBSA vs TI. J Chem Theory Comput 2013; 9: 1311-1319.
  • 35 Kollman PA, Massova I, Reyes C. et al. Calculating structures and free energies of complex molecules: Combining molecular mechanics and continuum models. Accounts Chem Res 2000; 33: 889-897.
  • 36 Li R, Zheng K, Hu P. et al. A Novel Tumor Targeting Drug Carrier for Optical Imaging and Therapy. Theranostics 2014; 4: 642-659.
  • 37 Torrbrown SR, Sobel BE. Attenuation of Thrombolysis by Release of Plasminogen-Activator Inhibitor Type-1 from Platelets. Thromb Res 1993; 72: 413-421.
  • 38 Falati S, Gross P, Merrill-Skoloff G. et al. Real-time in vivo imaging of platelets, tissue factor and fibrin during arterial thrombus formation in the mouse. Nature Med 2002; 8: 1175-1181.
  • 39 Jasuja R, Passam FH, Kennedy DR. et al. Protein disulfide isomerase inhibitors constitute a new class of antithrombotic agents. J Clin Invest 2012; 122: 2104-2113.
  • 40 Sambrano GR, Weiss EJ, Zheng YW. et al. Role of thrombin signalling in platelets in haemostasis and thrombosis. Nature 2001; 413: 74-78.
  • 41 Matsuo O, Lijnen HR, Ueshima S. et al. A guide to murine fibrinolytic factor structure, function, assays, and genetic alterations. J Thromb Haemost 2007; 5: 680-689.
  • 42 Zheng K, Li R, Zhou XL. et al. Dual actions of albumin packaging and tumor targeting enhance the antitumor efficacy and reduce the cardiotoxicity of doxorubicin in vivo. Int J Nanomed 2015; 10: 5327-5342.
  • 43 Gradishar WJ, Tjulandin S, Davidson N. et al. Phase III trial of nanoparticle albumin-bound paclitaxel compared with polyethylated castor oil-based paclitaxel in women with breast cancer. J Clin Oncol 2005; 23: 7794-7803.
  • 44 Yang F, Bian CB, Zhu LL. et al. Effect of human serum albumin on drug metabolism: Structural evidence of esterase activity of human serum albumin. J Struct Biol 2007; 157: 348-355.
  • 45 Hennan JK, Morgan GA, Swillo RE. et al. Effect of tiplaxtinin (PAI-039), an orally bioavailable PAI-1 antagonist, in a rat model of thrombosis. J Thromb Haemost 2008; 6: 1558-1564.
  • 46 Gorlatova NV, Tale JM, Elokdah H. et al. Mechanism of inactivation of plasminogen activator inhibitor-1 by a small molecule inhibitory. J Biol Chem 2007; 282: 9288-9296.
  • 47 Lucking AJ, Visvanathan A, Philippou H. et al. Effect of the small molecule plasminogen activator inhibitor-1 (PAI-1) inhibitor, PAI-749, in clinical models of fibrinolysis. J Thromb Haemost 2010; 8: 1333-1339.