Thromb Haemost 2004; 91(01): 28-37
DOI: 10.1160/TH03-05-0258
Blood Coagulation, Fibrinolysis and Cellular Haemostasis
Schattauer GmbH

A novel gene mutation in the 60s loop of human coagulation factor VII – inhibition of interdomain crosstalk

Anita Kavlie
1   Biotechnology Centre of Oslo, University of Oslo, Oslo, Norway
,
Merete Thune Wiiger
1   Biotechnology Centre of Oslo, University of Oslo, Oslo, Norway
,
Mette Husbyn
1   Biotechnology Centre of Oslo, University of Oslo, Oslo, Norway
,
Helge Stormorken
1   Biotechnology Centre of Oslo, University of Oslo, Oslo, Norway
,
Hans Prydz
1   Biotechnology Centre of Oslo, University of Oslo, Oslo, Norway
› Author Affiliations
Grant support: This work was supported by grants to Hans Prydz from the Research Council of Norway (NFR), Hafslund Nycomed, The Norwegian Council on Cardiovascular Diseases and CA no. BMHI-CT94-1202 (Clotart). Merete Thune Wiiger is a Research Fellow of the Norwegian Foundation for Health and Rehabilitation.
Further Information

Publication History

Received 01 May 2003

Accepted after revision 17 September 2003

Publication Date:
30 November 2017 (online)

Summary

A novel mutation in the factor VII gene resulting in procoagulant activity of 7.5% and antigen levels of 23% is presented. Single-stranded conformational polymorphism and DNA sequencing analysis revealed heterozygous shifts, and mutations were detected in exons 5, 7 and 8. The mutant L204P in exon 7 was novel, while the common polymorphisms, H115H and R353Q, were located in exons 5 and 8, respectively.The molecular effect of the L204P mutation was characterized using recombinant mammalian expression in Chinese hamster ovary cells. Low levels (4 ng/ml) of secreted mutant protein were found in transiently transfected cells compared to wild-type factor VII (83 ng/ml). Metabolic labeling demonstrated that the rate of mutant protein synthesis was similar to that of wild-type FVII, and the mutant protein accumulated intracellularly with no signs of increased degradation during a four-hour chase. No interaction between secreted P204 protein and immobilized soluble tissue factor was detected using surface plasmon resonance. The activation rate of recombinant mutant FVII protein was strongly reduced compared to wild-type FVII. A 9-fold reduction in the rate of FX activation was detected whereas Km was nearly the same for wild-type and the mutant. This slow rate was caused by a correspondingly lowered rate of P204 activation. A synthetic peptide sequence comprising amino acids 177−206 blocked binding of FVIIa to the TF-chip, and the subsequent factor X activation with an IC50 value of 0.5 μM in a chromogenic factor Xa assay. Additionally, evaluation of the peptide by surface plasmon resonance analysis resulted in inhibition of complex formation with an apparent Ki of 7 μM.

 
  • References

  • 1 Alexander B, Goldstein R, Landwehr G. The prothrombin conversion accelerator of serum (SPCA): its partial purification and its properties compared with serum Ac-globulin. J Clin Invest 1950; 29: 881.
  • 2 Owren P. Proconvertin, the new clotting factor. Scand J Clin Lab Invest 1951; 03: 168.
  • 3 Koller F, Loeliger A, Duckert F. Experiments on a new clotting factor (factor VII). Acta Haematol 1951; 06: 1.
  • 4 Gladhaug A, Prydz H. Purification of the coagulation factors VII and X from human serum Some properties of factor VII. Biochim Biophys Acta 1970; 215: 105-11.
  • 5 Wilcox JN, Noguchi S, Casanova JR. et al. Extrahepatic synthesis of FVII in human ather-oma and smooth muscle cells in vitro . Ann N Y Acad Sci 2001; 947: 433-8.
  • 6 O’Hara PJ, Grant FJ, Haldeman BA. et al. Nucleotide sequence of the gene coding for human factor VII, a vitamin K-dependent protein participating in blood coagulation. Proc Natl Acad Sci U S A 1987; 84: 5158-62.
  • 7 Hagen FS, Gray CL, O’Hara P. et al. Characterization of a cDNA coding for human factor VII. Proc Natl Acad Sci U S A 1986; 83: 2412-6.
  • 8 Mackman N, Morrissey JH, Fowler B. et al. Complete sequence of the human tissue factor gene, a highly regulated cellular receptor that initiates the coagulation protease cascade. Biochemistry 1989; 28: 1755-62.
  • 9 Banner DW, D’Arcy A, Chene C. et al. The crystal structure of the complex of blood coagulation factor VIIa with soluble tissue factor. Nature 1996; 380: 41-6.
  • 10 Ashton AW, Kemball-Cook G, Johnson DJ, Martin DM, O’Brien DP, Tuddenham EG. et al. Factor VIIa and the extracellular domains of human tissue factor form a compact complex: a study by X-ray and neutron solution scattering. FEBS Lett 1995; 374: 141-6.
  • 11 Ashton AW, Boehm MK, Johnson DJ, KemballCook G, Perkins SJ. The solution structure of human coagulation factor VIIa in its complex with tissue factor is similar to free factor VIIa: a study of a heterodimeric receptor-ligand complex by X-ray and neutron scattering and computational modeling. Biochemistry 1998; 37: 8208-17.
  • 12 Tuddenham EG. The tissue factor-factor VII complex: recent advances towards elucidating the structure and function of the initiator of haemostasis. Haemostasis 1996; 26 (Suppl. 01) 20-4. 20-24.
  • 13 Perera L, Darden TA, Pedersen LG. Predicted solution structure of zymogen human coagulation FVII. J Comput Chem 2002; 35-47.
  • 14 Eigenbrot C, Kirchhofer D, Dennis MS. et al. The factor VII zymogen structure reveals reregistration of beta strands during activation. Structure 2001; 09: 627-36.
  • 15 Pike AC, Brzozowski AM, Roberts SM. et al. Structure of human factor VIIa and its implications for the triggering of blood coagulation. Proc Natl Acad Sci U S A 1999; 96: 8925-30.
  • 16 Persson E. Structure of human coagulation activated factor VII. Blood Coagul Fibrinolysis 2000; 11 (Suppl. 01) S15-S17.
  • 17 Edgington TS, Dickinson CD, Ruf W. The structural basis of function of the TF VIIa complex in the cellular initiation of coagulation. Thromb Haemost 1997; 78: 401-5.
  • 18 Kavlie A, Orning L, Grindflek A. et al. Characterization of a factor VII molecule carrying a mutation in the second epidermal growth factor-like domain. Thromb Haemost 1998; 79: 1136-43.
  • 19 Millar DS, Kemball-Cook G, McVey JH. et al. Molecular analysis of the genotype-phenotype relationship in factor VII deficiency. Hum Genet 2000; 107: 327-42.
  • 20 Wulff K, Herrmann FH. Twenty two novel mutations of the factor VII gene in factor VII deficiency. Hum Mutat 2000; 15: 489-96.
  • 21 Kavlie A, Stormorken H, Orning L. et al. Characterization of mutations in the human factor VII gene: modelling and expression. Thromb Haemost. Suppl 1997 Poster PS-1681.
  • 22 Orita M, Suzuki Y, Sekiya T. et al. Rapid and sensitive detection of point mutations and DNA polymorphisms using the polymerase chain reaction. Genomics 1989; 05: 874-9.
  • 23 Orita M, Iwahana H, Kanazawa H. et al. Detection of polymorphisms of human DNA by gel electrophoresis as single-strand conformation polymorphisms. Proc Natl Acad Sci U S A 1989; 86: 2766-70.
  • 24 Hvatum M, Prydz H. Studies on tissue thromboplastin-its splitting into two separable parts. Thromb Diath Haemorrh 1969; 02: 217-22.
  • 25 Hvatum M, Prydz H. Studies on Tissue Thromboplastin. Biochim Biophys Acta 1966; 130: 92-101.
  • 26 Kraulis P. MOLSCRIPT: A Program to Produce Both Detailed and Schematic Plots of Protein Structures. Journal of Applied Crystallography 1991; 24: 946-50.
  • 27 Sayle RA, Milner-White EJ. RASMOL: biomolecular graphics for all. Trends Biochem Sci 1995; 20: 374.
  • 28 Vriend G. WHAT IF: a molecular modeling and drug design program. J Mol Graph 1990; 08: 526 29.
  • 29 Howell S, Kenmore M, Kirkland M. et al. High-density immobilization of an antibody fragment to a carboxymethylated dextran-linked biosensor surface. J Mol Recognit 1998; 11: 200-3.
  • 30 Andersen A, Warren DJ, Slordal L. Quantitation of cell-associated doxorubicin by high-performance liquid chromatography after enzymatic desequestration. Cancer Chemother Pharmacol 1994; 34: 197-202.
  • 31 Gulowsen AC, Myrseth LE, Brandtzaeg P. et al. A monoclonal antibody to a megakaryoblastlike cell line detects a protein found in blood cells and in the epithelial cell lining of various rat tissues. Eur J Cell Biol 1990; 52: 142-6.
  • 32 Husbyn M, Cuthbertson A. A novel approach to the synthesis of EGF-like domains: a method for the one-pot regioselective formation of the three disulfide bonds of a human blood coagulation factor VII EGF-1 analogue. J Pept Res 2002; 60: 121-7.
  • 33 Husbyn M, Orning L, Sakariassen KS. et al. Peptides corresponding to the second epidermal growth factor-like domain of human blood coagulation factor VII: synthesis, folding and biological activity. J Pept Res 1997; 50: 475-82.
  • 34 Dennis MS, Eigenbrot C, Skelton NJ. et al. Peptide exosite inhibitors of factor VIIa as anticoagulants. Nature 2000; 404: 465-70.
  • 35 Cheng Y, Prusoff WH. Relationship between the inhibition constant (K1) and the concentration of inhibitor which causes 50 per cent inhibition (I50) of an enzymatic reaction. Biochem Pharmacol 1973; 22: 3099-108.
  • 36 Bohm G, Muhr R, Jaenicke R. Quantitative analysis of protein far UV circular dichroism spectra by neural networks. Protein Eng 1992; 05: 191-5.
  • 37 Marchetti G, Patracchini P, Papacchini M. et al. A polymorphism in the 5’ region of coagulation factor VII gene (F7) caused by an inserted decanucleotide. Hum Genet 1993; 90: 575-6.
  • 38 Marchetti G, Patracchini P, Gemmati D. et al. Detection of two missense mutations and characterization of a repeat polymorphism in the factor VII gene (F7). Hum Genet 1992; 89: 497502.
  • 39 Green F, Kelleher C, Wilkes H. et al. A common genetic polymorphism associated with lower coagulation factor VII levels in healthy individuals. Arterioscler Thromb 1991; 11: 540-6.
  • 40 Humphries SE, Lane A, Dawson S. et al. The study of gene-environment interactions that influence thrombosis and fibrinolysisGenetic variation at the loci for factor VII and plasminogen activator inhibitor-1. Arch Pathol Lab Med 1992; 116: 1322-9.
  • 41 Bernardi F, Arcieri P, Bertina RM. et al. Contribution of factor VII genotype to activated FVII levels Differences in genotype frequencies between northern and southern European populations. Arterioscler Thromb Vasc Biol 1997; 17: 2548-53.
  • 42 Triplett DA, Brandt JT, Batard MA. et al. Hereditary factor VII deficiency: heterogeneity defined by combined functional and immunochemical analysis. Blood 1985; 66: 1284-7.
  • 43 Wildgoose P, Kazim AL, Kisiel W. The importance of residues 195-206 of human blood clotting factor VII in the interaction of factor VII with tissue factor. Proc Natl Acad Sci U S A 1990; 87: 7290-4.
  • 44 Kumar A, Blumenthal DK, Fair DS. Identification of molecular sites on factor VII which mediate its assembly and function in the extrinsic pathway activation complex. J Biol Chem 1991; 266: 915-21.
  • 45 Kumar A, Fair DS. Specific molecular interaction sites on factor VII involved in factor X activation. Eur J Biochem 1993; 217: 509-18.
  • 46 Roberge M, Santell L, Dennis MS. et al. A novel exosite on coagulation factor VIIa and its molecular interactions with a new class of peptide inhibitors. Biochemistry 2001; 40: 9522-31.
  • 47 Chaing S, Clarke B, Sridhara S. et al. Severe factor VII deficiency caused by mutations abolishing the cleavage site for activation and altering binding to tissue factor. Blood 1994; 83: 3524-35.
  • 48 Dickinson CD, Kelly CR, Ruf W. Identification of surface residues mediating tissue factor binding and catalytic function of the serine protease factor VIIa. Proc Natl Acad Sci U S A 1996; 93: 14379-84.
  • 49 Iino M, Foster DC, Kisiel W. Functional consequences of mutations in Ser-52 and Ser-60 in human blood coagulation factor VII. Arch Biochem Biophys 1998; 352: 182-92.
  • 50 Peyvandi F, Jenkins PV, Mannucci PM. et al. Molecular characterisation and three-dimensional structural analysis of mutations in 21 unrelated families with inherited factor VII deficiency. Thromb Haemost 2000; 84: 250-7.
  • 51 Giansily-Blaizot M, Aguilar-Martinez P, BironAndreani C. et al. Analysis of the genotypes and phenotypes of 37 unrelated patients with inherited factor VII deficiency. Eur J Hum Genet 2001; 09: 105-12.
  • 52 Huber R, Bode W. Structural basis of activation and action of trypsin [ISI]. Acc Chem Res 1978; 11: 114-22.
  • 53 Jin J, Perera L, Stafford D. et al. Four loops of the catalytic domain of factor viia mediate the effect of the first EGF-like domain substitution on factor viia catalytic activity. J Mol Biol 2001; 307: 1503-17.
  • 54 Bode W, Mayr I, Baumann U. et al. The refined 1.9 A crystal structure of human alpha-thrombin: interaction with D-Phe-Pro-Arg chloromethylketone and significance of the Tyr-ProPro-Trp insertion segment. EMBO J 1989; 08: 3467-75.
  • 55 Sichler K, Banner DW, D’Arcy A. et al. Crystal structures of uninhibited factor VIIa link its cofactor and substrate-assisted activation to specific interactions. J Mol Biol 2002; 322: 591-603.
  • 56 Dickinson CD, Shobe J, Ruf W. Influence of cofactor binding and active site occupancy on the conformation of the macromolecular substrate exosite of factor VIIa. J Mol Biol 1998; 277: 959-71.
  • 57 Peyvandi F, Carew JA, Perry DJ. et al. Abnormal secretion and function of recombinant human factor VII as the result of modification to a calcium binding site caused by a 15-base pair insertion in the F7 gene. Blood 2001; 97: 960-5.
  • 58 Toso R, Pinotti M, High KA. et al. A frequent human coagulation Factor VII mutation (A294V, c152) in loop 140s affects the interaction with activators, tissue factor and substrates. Biochem J 2002; 363: 411-6.