Two cross-reacting material–positive (CRM+) factor VII (FVII) mutations, associated with similar reductions in coagulant activity (2.5%) but with mild to asymptomatic (Gly331Ser, c184 [in chymotrypsin numbering]) or severe (Gly283Ser, c140) hemorrhagic phenotypes, were investigated. The affected glycines belong to structurally conserved regions in the c184 through c193 and c140s activation domain loops, respectively. The natural mutants 331Ser-FVII and 283Ser-FVII were expressed, and in addition 331Ala-FVII and 283Ala-FVII were expressed because 3 functional serine-proteases bear alanine at these positions. The 331Ser-FVII, present in several asymptomatic subjects, showed detectable factor Xa generation activity in patient plasma (0.7% ± 0.2%) and in reconstituted system with the recombinant molecules (2.7% ± 1.1%). The reduced activity of recombinant 283Ala-FVII (7.2% ± 2.2%) indicates that the full function of FVII requires glycine at this position, and the undetectable activity of 283Ser-FVII suggests that the oxydrile group of Ser283 participates in causing severe CRM+ deficiency. Furthermore, in a plasma system with limiting thromboplastin concentration, 283Ser-FVII inhibited wild-type FVIIa activity in a dose-dependent manner.

Factor VII (FVII) deficiency is associated with a wide spectrum of coagulation and hemorrhagic phenotypes.1,2 Although a large number of FVII mutations have been described,3 the relationship of molecular and clotting defects to the clinical picture has been poorly defined,3-6 particularly for cross-reacting material–positive (CRM+) deficiencies.7-10 

Functional studies were conducted in plasma and with recombinant variants to provide molecular elements useful to define this relationship in 2 homozygous CRM+ FVII deficiencies associated with markedly different hemorrhagic phenotypes.

Patients

Patients with normal FVII antigen and low FVII coagulant activity (FVIIc) (2.5%), not previously characterized, were studied.

Patient FVII33 (male, age 83, FVIIc 2%); patient FVII34 (male, age 26, FVIIc 0.7%); and patient FVII35 (female, age 30, FVIIc 0.7%) were clinically asymptomatic, while patient FVII36 (male, age 39, FVIIc 0.6%) experienced epistaxis and gum hemorrhage. Patient FVII37 (female, age 71, FVIIc 1.1%) was referred for anemia following chronic bleeding (rectorrhagia, hemorrhoids).

Patient FVII38 (female, age 30, FVIIc 2.5%) experienced recurring hemorrhages (menometrorrhagia, melena) and had hemarthroses. Two episodes of hemoperitoneum occurred following rupture of ovarian cysts. Fresh plasma, FVII concentrates, or FVII (Provertin-UM, Immuno, Pisa, Italy) were administered on several occasions.

Genetic analysis

Mutation search and detection of FVII polymorphisms were performed as previously described.11,12 Mutagenesis and transfection expression vectors were created by site-directed mutagenesis of plasmid pCDNA3-FVII.13Oligonucleotides for mutagenesis spanned the following complementary DNA nucleotides: (1) 1193 through 1220 containing the 1206G > A transition (Gly331Ser) or the 1207G > C transversion (Gly331Ala); (2) 1040 through 1073 containing the 1062G > A transition (Gly283Ser) or the 1063 G > C transversion (Gly283Ala).

BHK cells were transiently transfected13 with the use of serum-free medium. FVII antigen and coagulant activity, FVII coagulant activity, and FVII antigen in plasma or conditioned medium were determined as described.13 

Factor Xa generation assay

Plasma.

The reaction was initiated by adding an excess (30 μL) of thromboplastin (Thromborel S) (Behring, Marburg, Germany) to 50 μL diluted plasma (1:20, 1:40, 1:80, and 1:140 in 20 mM HEPES, 150 mM NaCl, 0.1% PEG 8000, 5 mM CaCl2, pH 7.4.

Recombinant FVII.

FVII in 2 nM conditioned medium was incubated 5 minutes at 37°C with 1 nM human factor Xa (hFXa) (Sigma, St Louis, MO); 300 μM 75% phosphatidylcholine/25% phosphatidylserine14; and 5 mM CaCl2 in 50 μL final volume. After addition of 30 μL thromboplastin, FXa generation was started with 40 nM hFX (Sigma).

Quenching reactions.

Reactions were quenched with 30 μL 20 mM HEPES, 150 mM NaCl, 0.1% PECT 8000, 50 mM EDTA, pH 7.4 after 5 minutes' incubation at ambient temperature. FXa fluorogenic substrate (MeSO2-D-CHA-Gly-Arg-AMCAcOH) (American Diagnostica, Greenwich, CT) was added (200 μM), and fluorescence (360 nm excitation, 465 nm emission) was measured on SpectraFluorPlus microplate reader (TECAN, Salzburg, Austria).

Standardizing assays.

Assays were standardized with dilutions of pooled normal plasma (PNP) or of recombinant wild-type (rWt) FVII. Molecular modeling of FVII was performed by means of Swiss Model (GLAXO, Geneva, Switzerland) through the ProMod program,15 and CHARMs software. The crystal coordinates of the tissue factor–FVIIa complex (1DAN), FVIIa (1QFK, 1CVW, 1DVA) and FVII (1JBU),16-20 were from the protein data bank.

FVII gene sequencing indicated that 5 patients (patient FVII33 through FVII37), all from southern Italy, were homozygous for the 10908G > A transition, resulting in the Gly331Ser substitution. In contrast, patient FVII38 was homozygous for the 10764G > A transition, resulting in the Gly283Ser substitution. In spite of a similar reduction in FVIIc levels (Table1), the Gly331Ser homozygotes showed asymptomatic to mild phenotypes, whereas the Gly283Ser homozygote was characterized by severe bleeding tendency.

Table 1.

Activity and antigen of factor VII variants in patient's plasma or in conditioned medium

Wt331Ser283Ser331Ala283Ala
Activity, %*      
 Plasma, FVIIc 100 0.6-1.9 2.5 NA NA 
 Plasma 100 0.7 ± 0.2 ND NA NA 
 rFVII 100 2.7 ± 1.1 ND 18.1 ± 1.1 7.2 ± 2.2  
Antigen      
 Plasma, % 100 85-100 100 NA NA  
 rFVII, ng/mL 119 ± 70 105 ± 71 101 ± 80 145 ± 84 85 ± 58 
Wt331Ser283Ser331Ala283Ala
Activity, %*      
 Plasma, FVIIc 100 0.6-1.9 2.5 NA NA 
 Plasma 100 0.7 ± 0.2 ND NA NA 
 rFVII 100 2.7 ± 1.1 ND 18.1 ± 1.1 7.2 ± 2.2  
Antigen      
 Plasma, % 100 85-100 100 NA NA  
 rFVII, ng/mL 119 ± 70 105 ± 71 101 ± 80 145 ± 84 85 ± 58 

Results obtained in 5 independent sets of experiments are reported as mean ± SD.

Wt indicates wild type; FVIIc, factor VIIc; rFVII, recombinant FVII; ND, not detectable, and NA, not applicable.

*

Percentage of pooled normal plasma (PNP) or of rWtFVII.

Factor Xa generation activity.

Range of FVII levels in plasma of 5 homozygous patients.

Several genetic and methodological factors could contribute to loosening the relationship among causative mutations, FVII activity, and clinical severity of these CRM+ FVII deficiencies. Among the genetic factors, we have investigated the influence of FVII polymorphisms, previously found to be associated with FVII levels in plasma.12 All patients were homozygous for a common genotype characterized by the most frequent allelic forms A1, b, and M1,12 thus excluding their differential contribution to FVII levels.

Since the low sensitivity of the routine laboratory assays hampers the comparative evaluation of FVIIc values,5,21 the residual FVII activity in patient plasma was further evaluated by measuring the FXa generation with the use of an FXa fluorogenic substrate (Table1). Activity in plasma from a Gly331Ser homozygote (0.7% ± 0.2%) was significantly higher (P < .001, analysis of variance) than in plasma from the Gly283Ser homozygote, which, investigated at different dilutions, was indistinguishable (P = .97) from FVII-depleted plasma. This indicates very low, if any, activity of the stable Gly283Ser-FVII. FXa generation curves from 2 Gly331Ser homozygotes overlapped (not shown).

To validate the causative role of mutations, the recombinant FVII molecules (r331Ser-FVII, r283Ser-FVII) were transiently expressed in BHK cells. Their antigen levels (Table 1) did not differ significantly (P = .93) from those of rWt-FVII. Functional assays in a reconstituted system showed a clearly detectable FXa generation activity (Table1) for the r331Ser-FVII. In contrast, the activity of the r283Ser-FVII was indistinguishable from that of the mock medium, and of medium containing the active site (rSer344Ala-FVII) or the cleavage site (rArg152Gln-FVII) mutants,9,22 used as 2 inactive reference CRM+ FVII variants.

Findings in plasma and reconstituted system help to interpret the markedly different hemorrhagic phenotypes associated to the mutations. The 331Ser-FVII residual activity would trigger coagulation, thus preventing severe bleeding symptoms in the several Gly331Ser homozygotes. On the other hand, the undetectable activity of the 283Ser-FVII is in accordance with the severe phenotype observed in PFVII38.

Additional insights into the causative role of these mutations were sought by inspection of FVII crystallographic structures,16-20 molecular modeling,15 and comparative analysis,23 coupled with further mutagenesis, expression, and functional studies.

Gly331 (c184 in chymotrypsin numbering) belongs to the activation loop c184 through c193, makes a hydrogen bond with Pro303 of the shifting β-strand-B220 (c153 through c162) and is in close contact with Asp338 at the bottom of the specificity pocket of FVIIa.

Gly283 (c140) is at the hinge of the c140s loop19 and, both in the zymogen and activated form, is in close contact with Asp343, which forms the salt bridge with Ile153.

In FVII models, the Gly283Ser and Gly331Ser substitutions are tolerated without major structural changes, which could explain the presence of normal amounts of circulating proteins resulting in CRM+deficiencies.

Gly283 and Gly331 belong to structurally conserved regions in serine-proteases23 (Table 2). Among functional members, only urokinase and complement factors B and 2 present alanine at the topologically equivalent positions (Table 2, italics), which led us to investigate the r331Ala-FVII and r283Ala-FVII (Table 1). Although clearly lower than that of rWt-FVII, FXa generation by r331Ala-FVII and r283Ala-FVII was remarkably higher than that of the corresponding natural variants. These findings suggest that the full activity of FVII is not compatible with the presence of side chains at positions 331 and 283. The 331Ala-FVII, lacking the serine oxydrile group, showed a 7-fold higher activity than the 331Ser-FVII. The appreciable activity of 283Ala-FVII, compared with the impaired function of the 283Ser-FVII, suggests that the oxydrile group of Ser283, potentially affecting proper salt bridge formation and c140s loop conformational changes, participates in producing a clinically severe form of CRM+FVII deficiency.

Table 2.

Structurally conserved regions containing the affected residues in factor VII

SCRAmino acid residues
(c139-c142)(c180-c184)
FVII Ser-Gly-Trp-Gly Met-Phe-Cys-Ala-Gly 
FX Ser-Gly-Phe-Gly Met-Phe-Cys-Ala-Gly 
FIX Ser-Gly-Trp-Gly Met-Phe-Cys-Ala-Gly 
PC Thr-Gly-Trp-Gly Met-Leu-Cys-Ala-Gly 
UPA Thr-Gly-Phe-Gly Met-Leu-Cys-Ala-Ala 
CFB Lys-Ala-Leu-Phe Phe-Leu-Cys-Thr-Gly 
CF2 Pro-Ala-His-Phe Phe-Leu-Cys-Ser-Gly 
CHT Thr-Gly-Trp-Gly Met-Ile-Cys-Ser-Gly 
SCRAmino acid residues
(c139-c142)(c180-c184)
FVII Ser-Gly-Trp-Gly Met-Phe-Cys-Ala-Gly 
FX Ser-Gly-Phe-Gly Met-Phe-Cys-Ala-Gly 
FIX Ser-Gly-Trp-Gly Met-Phe-Cys-Ala-Gly 
PC Thr-Gly-Trp-Gly Met-Leu-Cys-Ala-Gly 
UPA Thr-Gly-Phe-Gly Met-Leu-Cys-Ala-Ala 
CFB Lys-Ala-Leu-Phe Phe-Leu-Cys-Thr-Gly 
CF2 Pro-Ala-His-Phe Phe-Leu-Cys-Ser-Gly 
CHT Thr-Gly-Trp-Gly Met-Ile-Cys-Ser-Gly 

Residues c139 through c142 and c180 through c184 in chymotrypsin numbering corresponds to residues 282 through 285 and 327 through 331 in FVII.

SCR indicates structurally conserved region; FVII, factor VII; FX, factor X; FIX, factor IX; PC, protein C; UPA, urokinase plasminogen activator; CFB, complement factor B; CF2, complement factor 2; CHT, chymotrypsin.

Previous studies of FVIIa inhibition at tissue factor–limiting concentrations24 prompted us to investigate the activity of the recombinant FVII (rFVIIa) in Gly283Ser homozygote plasma (Figure1A). In 3 independent experiments, a reduction of 54.6% ± 8.2% in rFVIIa activity was observed at a 1:100 ratio between rFVIIa and plasma 283Ser-FVII. The rFVIIa was not inhibited in plasma from a severe CRM-FVII–deficient patient (FVIIc, FVIIag less than 1%) homozygous for the double mutation Ala294Val/404delC.25 Furthermore, FXa generation rate by rFVIIa in Ala294Val/404delC homozygote plasma was inhibited in a dose-dependent manner by the addition of medium containing r283Ser-FVII (Figure 1B). A 500-fold excess of r283Ser-FVII (1 nM) reduced rFVIIa (2 pM) activity of 68.8% ± 14.7%. A dose-dependent inhibition was confirmed in plasma-free reconstituted system (not shown).

Fig. 1.

Inhibition of the rFVIIa activity in plasma by 283Ser-FVII.

(A) FXa generation curves produced by 2 pM rFVIIa (NovoNordisk, Bagsvaed, Denmark) in diluted (1:20) patient FVII38 plasma (FVIIa/283Ser-FVII molar ratio of 1:100) (▪), Ala294ValdelC homozygote plasma (●), or FVII-depleted plasma (○). (B) FXa generation curves produced by 2 pM rFVIIa in Ala294ValdelC homozygote diluted plasma (1:20) with the addition of conditioned medium containing 0.2 nM (■), 1 nM (▪) r283Ser-FVII, or mock medium (●). Reactions were initiated by adding limiting amount of thromboplastin, 20 nM hFX, and 100 μM FXa fluorogenic substrate, and fluorescence was measured over time. The limiting amount was determined before each assay by evaluating the FXa generation obtained with 2 pM rFVIIa concentration and decreasing the amounts of thromboplastin. The decrease in activity was estimated by comparison of FXa generation rates (relative fluorescence units per minute) between 4 and 8 minutes. Rfu indicates relative fluorescence units.

Fig. 1.

Inhibition of the rFVIIa activity in plasma by 283Ser-FVII.

(A) FXa generation curves produced by 2 pM rFVIIa (NovoNordisk, Bagsvaed, Denmark) in diluted (1:20) patient FVII38 plasma (FVIIa/283Ser-FVII molar ratio of 1:100) (▪), Ala294ValdelC homozygote plasma (●), or FVII-depleted plasma (○). (B) FXa generation curves produced by 2 pM rFVIIa in Ala294ValdelC homozygote diluted plasma (1:20) with the addition of conditioned medium containing 0.2 nM (■), 1 nM (▪) r283Ser-FVII, or mock medium (●). Reactions were initiated by adding limiting amount of thromboplastin, 20 nM hFX, and 100 μM FXa fluorogenic substrate, and fluorescence was measured over time. The limiting amount was determined before each assay by evaluating the FXa generation obtained with 2 pM rFVIIa concentration and decreasing the amounts of thromboplastin. The decrease in activity was estimated by comparison of FXa generation rates (relative fluorescence units per minute) between 4 and 8 minutes. Rfu indicates relative fluorescence units.

Close modal

Because of the decrease over time of infused FVII and the constant amount of endogenous dysfunctional variants, further studies are needed to establish whether the inhibition observed in vitro might have implications for substitution therapy of severe CRM+deficiency.

We thank Dr F. H. Herrmann, University of Greifswald, Germany, for providing us with plasma from the A294V/404delC homozygote.

Support by the Consiglio Nazionale Della Ricerche Biotech Target Project and Ministero Dell 'Universitá, Della Ricerca Scientifica E Tecnologica.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 U.S.C. section 1734.

1
Hedner
U
Davie
EW
Introduction of haemostasis and the vitamin K-dependent coagulation factors.
The Metabolic Basis of Inherited Disease. Vol 2.
Scriver
CR
Beaudet
AL
Sly
WS
et al
84
1989
2107
2134
McGraw-Hill
New York, NY
2
Triplett
DA
Brandt
JT
Batard
MA
Dixon
JL
Fair
DS
Hereditary factor VII deficiency: heterogeneity defined by combined functional and immunochemical analysis.
Blood.
66
1985
1284
1287
3
McVey
JH
Boswell
E
Mumford
AD
Kemball-Cook
G
Tuddenham
EGD
Factor VII deficiency and the FVII mutation database.
Hum Mutat.
17
2001
3
17
4
Cooper
DN
Millar
DS
Wacey
A
Banner
DW
Tuddenham
GD
Inherited factor VII deficiency: molecular genetics and pathophysiology.
Thromb Haemost.
78
1997
151
156
5
Mariani
G
Lo Coco
L
Bernardi
F
Pinotti
M
Molecular and clinical aspects of factor VII deficiency.
Blood Coagul Fibrinolysis.
9(suppl 1)
1998
S83
S88
6
Millar
DS
Kemball-Cook
G
McVey
JH
et al
Molecular analysis of the genotype-phenotype relationship in factor VII deficiency.
Hum Genet.
107
2000
327
342
7
O'Brien
DP
Gale
KM
Anderson
JS
et al
Purification and characterization of FVII-304 Gln: a variant molecule with reduced activity isolated from a clinically unaffected male.
Blood.
78
1991
132
140
8
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.
89
1992
497
502
9
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.
83
1994
3524
3535
10
Zheng
DQ
Shurafa
M
James
HL
Factor VII G331D: a variant molecule involving replacement of a residue in the substrate-binding region of the catalytic domain.
Blood Coagul Fibrinolysis.
7
1996
93
96
11
Bernardi
F
Patracchini
P
Gemmati
D
et al
Molecular analysis of factor VII deficiency in Italy: a frequent mutation (FVII Lazio) in a repeated intronic region.
Hum Genet.
92
1993
446
450
12
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 population.
Arterioscler Thromb Vasc Biol.
17
1997
2548
2553
13
Pinotti
M
Toso
R
Redaelli
R
Berrettini
M
Marchetti
G
Bernardi
F
Molecular mechanisms of FVII deficiency: expression of mutations clustered in the IVS7 donor splice site of factor VII gene.
Blood.
92
1998
1646
1651
14
Hoekema
L
Castoldi
E
Tans
G
et al
Functional properties of factor V and factor Va encoded by the R2-gene.
Thromb Haemost.
85
2001
75
81
15
Peitsch
MC
PROMOD and Swiss-model: Internet-based tools for automated comparative protein modelling.
Biochem Soc Trans.
24
1996
274
279
16
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.
380
1996
41
44
17
Pike
ACW
Brzozowski
AM
Roberts
SM
Olsen
OH
Persson
E
Structure of human factor VIIa and its implications for the triggering of blood coagulation.
Proc Nat Acad Sci U S A.
96
1999
8925
8930
18
Kemball-Cook
G
Johnson
DJD
Tuddenham
EGD
Harlos
K
Crystal structure of active site-inhibited human coagulation factor VIIa (des-Gla).
J Struct Biol.
127
1999
213
223
19
Dennis
MS
Eigenbrot
C
Skelton
NJ
et al
Peptide exosite inhibitors of factor VIIa as anticoagulants.
Nature.
404
2000
465
470
20
Eigenbrot
C
Kirchhofer
D
Dennis
MS
Santell
L
et al
The factor VII zymogen structure reveals reregistration of beta strands during activation.
Structure.
9
2001
627
636
21
Massignon
D
Moulsma
M
Bondon
P
et al
Prothrombin time sensitivity and specificity to mild clotting factor deficiencies of the extrinsic pathway: evaluation of eight commercial thromboplastins.
Thromb Haemost.
75
1996
590
594
22
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.
93
1996
14379
14384
23
Greer
J
Comparative modelling methods: applications to the family of the mammalian serine proteases.
Proteins.
7
1990
317
334
24
Van 't Veer
C
Golden
NJ
Mann
KG
Inhibition of thrombin generation by the zymogen factor VII: implications for the treatment of hemophilia A by factor VIIa.
Blood.
95
2000
1330
1335
25
Herrmann
FH
Wulff
K
Auberger
K
Aumann
V
et al
Molecular biology and clinical manifestation of hereditary factor VII deficiency.
Semin Thromb Hemost.
26
2000
393
400

Author notes

Francesco Bernardi, Dipartimento di Biochimica e Biologia Molecolare, Università di Ferrara, Via L Borsari 46, 44100 Ferrara, Italy; e-mail: ber@unife.it.

Sign in via your Institution