scholarly journals Biochemical, molecular and clinical aspects of coagulation factor VII and its role in hemostasis and thrombosis

Haematologica ◽  
2021 ◽  
Author(s):  
Francesco Bernardi ◽  
Guglielmo Mariani

Activated factor VII (FVIIa), the first protease of clotting, expresses its physiological procoagulant potential only after complexing with tissue factor (TF) exposed to blood. Deep knowledge of the FVIIa-TF complex and F7 gene helps to understand the Janus-faced clinical findings associated to low or elevated FVII activity (FVIIc). Congenital FVII deficiency, the most frequent among the recessively inherited bleeding disorders, is caused by heterogeneous mutations in the F7 gene. Complete FVII deficiency causes perinatal lethality. A wide range of bleeding symptoms, from life-threatening intracranial hemorrhage to mild mucosal bleeding, is observed in patients with apparently modest differences in FVIIc levels. Though clinically relevant FVIIc threshold levels are still uncertain, effective management, including prophylaxis, has been devised, substantially improving the quality of life of patients. The exposure of TF in diseased arteries fostered investigation on the role of FVII in cardiovascular disease. FVIIc levels were found to be predictors of cardiovascular death and to be markedly associated to F7 gene variation. These genotype-phenotype relationships are among the most extensively investigated in humans. Genome-wide analyses extended association to numerous loci that, together with F7, explain >50% of FVII level plasma variance. However, the ability of F7 variation to predict thrombosis was not consistently evidenced in the numerous population studies. Main aims of this review are to highlight i) the biological and clinical information that distinguishes FVII deficiency from the other clotting disorders and ii) the impact exerted by genetically predicted FVII level variation on bleeding as well as on the thrombotic states.

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 2412-2412
Author(s):  
Gennadii M. Galstian ◽  
Olesya A. Polevodova ◽  
Elena Yakovleva ◽  
Antonina Shchekina ◽  
Igor Davydkin ◽  
...  

Introduction and Objectives: Inherited FVII deficiency is a rare hemorrhagic disorder.Clinical bleeding does not correlate with the level of FVII plasma activity. FVII involves in the extrinsic coagulation pathway. ROTEM is point of care method. EXTEM is one of ROTEM screening tests. EXTEM analyses the extrinsic pathway of coagulation. Aim of the study was to evaluate hemostatic effect and safety of rFVIIa treatment in pts with inherited FVII deficiency. Materials and Methods: The results of rFVIIa treatment were investigated in 4 pts (1 male, 3 females) with inherited FVII deficiency. Pts were treated with rFVIIa before gynecological (2), abdominal (1) and orthopedic surgery (1). Pts received 30 mcg/kg of rFVIIa (Koagil VII, Generium, Russia) before surgery. Plasma FVII activity, fibrinogen, INR, APTT, ROTEM and thromboelastography (TEG) parameters were investigated before rFVIIa administration, then in 15 minutes, in 2 hours, in 6 hours and in 12 hours. Results: Before rFVIIa administration median plasma FVII activity was low, CT EXTEM was long and INR was greater than 3. TEG, APTT and fibrinogen were in normal ranges (table 1, fig. 1). Fifteen minutes after rFVIIa administration FVII:C activity increased, INR and CT EXTEM decreased. Hemostatic effect of rFVIIa remained during 12 hours. During this period CT EXTEM correlated with INR (r = 0.97, p<0.001). Other parameters did not change significantly. None patients had hemorrhagic complications. Conclusion: CT EXTEM allowed to evaluate hemostatic effect of rFVIIa in operating room and good correlated with INR in pts with inherited FVII deficiency. Disclosures No relevant conflicts of interest to declare.


2017 ◽  
Vol 20 (2) ◽  
pp. 19-25
Author(s):  
S Shahbazi ◽  
R Mahdian ◽  
K Karimi ◽  
A Mashayekhi

AbstractCoagulation factor VII (FVII) is a key enzyme of the extrinsic coagulation cascade that is predominantly produced by hepatocytes. TheF7gene mutations cause FVII deficiency with considerable molecular and phenotypic heterogeneity. We characterized the molecular alterations of theF7gene and their corresponding mRNA transcripts in Iranian patients from eight unrelated families. The mutations were detected by polymerase chain reaction (PCR)-sequencing of allF7gene exons, their flanking intronic sequences, as well as their corresponding cDNA fragments. Homozygous P303T, C91S and R304Q mutations were detected in patient 2, patient 5, and patient 6, respectively. Patient 7 was a compound heterozygote for S282R and H348R and patient 8 was a compound heterozygote for R304Q and IVS7+7A>G mutations. Furthermore, our investigation revealed three heterozygous individuals, patient 1 and patient 3 with the A244V mutation who were symptomatic and patient 4 with V(–39)I mutation who was also asymptomatic. TheF7mRNA expression analysis revealed that, except the transcript of V(–39)I, other mutation-harboring transcripts were expressed at detectable levels. In conclusion, this report reinforces the genetic and phenotypic heterogeneity of FVII deficiency. The findings of the mRNA study implied that decreased FVII protein activity subsequent to missense mutations does not completely reflect the degradation of mutation-harboring mRNA.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 3978-3978 ◽  
Author(s):  
Brett E. Skolnick ◽  
Lisa Payne Rojkjaer ◽  
Stephanie V. Seremetis

Abstract Background and Purpose: The safety of recombinant activated coagulation factor VII (rFVIIa) has been studied in a variety of clinical settings including von Willebrand disease, thrombocytopenia, trauma, liver surgery, and vitamin-K antagonist therapy, as well as upper gastrointestional and central nervous system bleeding. Although multiple interventions have been developed to treat bleeding through revised surgical management and blood product transfusions, current treatment often remains inadequate. Seventeen clinical trials have been conducted in the seven therapeutic areas described above to evaluate the efficacy and safety of rFVIIa for the reduction or elimination of excessive bleeding. The safety data are presented in this summary. Methods: A comprehensive review of rFVIIa safety data was performed, which included 1630 patients (rFVIIa: 1086; placebo: 544) from international studies in 7 therapeutic areas where patients received single or repeated doses ranging from 5–200 μg/kg, with a maximum of 8 doses. Results: Serious adverse events (SAEs) were evenly distributed among treatment groups: 350/1086 (32%) rFVIIa-treated patients experienced 491 SAEs, and 213/544 (39%) placebo-treated patients experienced 333 SAEs. Fifty-one thromboembolic SAEs occurred in 48 patients (4.4%) in the rFVIIa treatment group (frequency per patient: 5%), and 20 thromboembolic SAEs occurred in 20 patients (3.7%) in the placebo group (frequency: 4%). Of the 51 thromboembolic SAEs in the rFVIIa group, 28 were arterial (frequency: 2.6%) and 23 were venous (frequency: 2.1%). Of the 20 thromboembolic SAEs that occurred in the placebo group, 8 were arterial (frequency: 1.5%) and 12 were venous (frequency: 2.2%). Thromboembolic SAEs considered by the investigator to be related to treatment represented thirty-four thromboembolic SAEs in the rFVIIa group (frequency of events per patient: 3.1%) and 13 thromboembolic SAEs in the placebo group (frequency: 2.4%). A total of 231 deaths occurred among both treatment groups: 144 (13%) from the rFVIIa group (11 thromboembolic-related deaths), and 87 (16%) from the placebo group (3 thromboembolic-related deaths). Of the 11 (1%) thromboembolic deaths that occurred in the rFVIIa group, 6 were considered by the investigator to be possibly/probably related to treatment. Of the 3 (0.6%) thromboembolic deaths that occurred in the placebo group, 2 were considered by the investigator to be possibly/probably related to treatment. Conclusions: Recombinant FVIIa appears to be safely administered at total doses ranging from 5–1120 μg/kg for the treatment of a wide range of bleeding disorders. Continued safety and efficacy evaluations are underway to further elaborate on these observations. rFVIIa patients; n=1086 Placebo patients; n=544 Events Number of SAEs Frequency * Number of SAEs Frequency * * = number of SAEs/total patients x 100, TE = Thromboembolic, SAE = Serious Adverse Events TE SAEs 51 5% 20 4% Arterial TE SAEs 28 2.6% 8 1.5% Venous TE SAES 23 2.1% 12 2.2% TE SAEs related to treatment 34 3.1% 13 2.4% TE-related Deaths 11 (1%) - 3 (0.6%) -


Blood ◽  
1998 ◽  
Vol 92 (3) ◽  
pp. 920-926
Author(s):  
John H. McVey ◽  
Emma J. Boswell ◽  
Osamu Takamiya ◽  
Gabriel Tamagnini ◽  
Victor Valente ◽  
...  

We have studied a family with homozygous lethal, blood coagulation factor VII (FVII) deficiency. To identify the mutation responsible for the deficiency, exons 2 to 8 and the intron-exon junctions of their FVII genes were amplified from peripheral white blood cell DNA by polymerase chain reaction and screened by single-strand conformational polymorphism analysis. The fragment showing aberrant mobility was cloned and sequenced. We detected a single point mutation, a homozygous G to A substitution at nucleotide position 6070, in the invariant GT dinucleotide at the 5′ splice site of intron 4. Homozygosity was confirmed by loss of a site for the restriction endonuclease Mlu I. Analysis of the splicing pattern of ectopic transcripts in lymphocytes in the parents revealed that this mutation is associated with skipping of exon 4, which produces an mRNA encoding FVII with an in-frame deletion of the first epidermal growth factor–like domain (EGF 1). Transient transfection of COS-7 cells with an expression vector containing the ▵EGF 1 FVII cDNA shows that this mutant protein is not expressed. The identification of the molecular basis of the FVII deficiency in this family allowed mutation-specific prenatal diagnosis to be performed in a subsequent pregnancy. In this family complete FVII deficiency is associated with a severe bleeding diathesis but no developmental abnormalities, lending weight to the hypothesis that fetal FVII is not required for the putative angiogenic functions of tissue factor in humans. © 1998 by The American Society of Hematology.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 30-30 ◽  
Author(s):  
Matteo ND Di Minno ◽  
Angelika Batorova ◽  
Mariasanta Napolitano ◽  
Alberto Dolce ◽  
Giovanni Di Minno ◽  
...  

Abstract Abstract 30 Background: Inherited Factor VII (FVII) deficiency is the most common of the “rare” autosomal recessive bleeding disorders. Affected individuals display a wide range of clinical phenotypes and treatment demand may vary from prophylaxis to the need of rare or no replacement at all. As a matter of fact, it would be important to individualize the optimal and safest management with reference to the risk of bleeding. Objective: In a large number of subjects with FVII deficiency, we evaluated whether the type of symptom at disease presentation could help to predict further symptoms during the “Observation Period” (OP). Methods: Appropriate information in subjects with FVII deficiency, collected in the multicentre STER and IF7SG Registries, were employed. OP= time elapsed between the date of disease-presentation symptom and that of enrolment into the registry. Data for this analysis were complete for 687 individuals. Results: At diagnosis, among the 687 subjects (356 [51.8%] females, 331 [48.2%] males, mean age 29.6±19.63 yrs), 272 (39.6%) were asymptomatic; of the symptomatic individuals, 338 (49.2%) displayed a mild “Platelet- Like” (PL) bleeding defect (i.e. muco-cutaneous bleedings) and 77 (11.2%) had a severe, “Hemophilia-Like” (HL), phenotype. Mean age at diagnosis for the asymptomatic individuals was 23.8±18.8; mean ages at disease presentation were 10.54 ±11.8 for the PL, and 5.5 ±12.3 for the HL individuals ( p for trend= 0.000). FVII activity (FVIIc) was: females 17.53±19.68%, males 18.58±17.50% (p=0.460), overall: 18.04 ±18,66%. Of the 338 individuals with a PL-disorder, 268 (79.3%) had new muco-cutaneous bleedings; 51 (15.1%) developed severe bleedings, and 12 (4%) did not develop any subsequent bleed. Among the patients (n=77) with a HL-disorder, 38 (49.4%) experienced new severe bleeds, 34 (44.2%) developed mucosal bleedings, and only 5 (6.5%) had no further bleeds. Among those asymptomatic at diagnosis (n=272), 237 (87.1%) remained asymptomatic, while 35 (12.9%) developed only muco-cutaneous bleedings. Thus, 87.1% of the asymptomatic individual at diagnosis remained so, 79.3% of those with a platelet-like disorder at diagnosis displayed the same type of bleeding and, finally, 50% of those with severe bleedings at diagnosis had, again, severe bleeds. FVIIc helped to refine clinical information. Asymptomatic individuals at diagnosis that subsequently developed muco-cutaneous bleedings, had FVIIc similar to that of individuals with the same mild bleeds at onset (∼15–20%). FVIIc of individuals with PL-disorder at diagnosis who subsequently experienced a life- and limb-threatening bleeding episode, were as low (<5%) as those of individuals that had a severe phenotype at disease presentation. Regardless whether documented at diagnosis or during the OP, severe bleeding was invariably associated with FVIIc <5%. At diagnosis, the latter levels were found in 88.3% of individuals with severe symptoms, in 57.8% of those with muco-cutaneous bleedings and in 14.7% of asymptomatic individuals. Conclusions: In Factor VII deficiency, three clinical phenotypes can be identified: 1. the most prevalent cohort experience muco-cutaneous bleeding (PL-disorder); 2. Ten–15% of patients exhibit potentially life- or limb-threatening hemorrhages, such as hemartroses, central nervous system (CNS) or gastrointestinal (GI), a phenotype that may be as severe or more severe than that in hemophilia, and 3. about 1/3 of cases are asymptomatic and tend to remain so. Bleeding symptom at disease presentation predicts the subsequently developed bleeding phenotype. FVIIc levels refine such clinical prediction and help to identify individuals that call for early prophylaxis or on demand treatment. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
1998 ◽  
Vol 92 (3) ◽  
pp. 920-926 ◽  
Author(s):  
John H. McVey ◽  
Emma J. Boswell ◽  
Osamu Takamiya ◽  
Gabriel Tamagnini ◽  
Victor Valente ◽  
...  

Abstract We have studied a family with homozygous lethal, blood coagulation factor VII (FVII) deficiency. To identify the mutation responsible for the deficiency, exons 2 to 8 and the intron-exon junctions of their FVII genes were amplified from peripheral white blood cell DNA by polymerase chain reaction and screened by single-strand conformational polymorphism analysis. The fragment showing aberrant mobility was cloned and sequenced. We detected a single point mutation, a homozygous G to A substitution at nucleotide position 6070, in the invariant GT dinucleotide at the 5′ splice site of intron 4. Homozygosity was confirmed by loss of a site for the restriction endonuclease Mlu I. Analysis of the splicing pattern of ectopic transcripts in lymphocytes in the parents revealed that this mutation is associated with skipping of exon 4, which produces an mRNA encoding FVII with an in-frame deletion of the first epidermal growth factor–like domain (EGF 1). Transient transfection of COS-7 cells with an expression vector containing the ▵EGF 1 FVII cDNA shows that this mutant protein is not expressed. The identification of the molecular basis of the FVII deficiency in this family allowed mutation-specific prenatal diagnosis to be performed in a subsequent pregnancy. In this family complete FVII deficiency is associated with a severe bleeding diathesis but no developmental abnormalities, lending weight to the hypothesis that fetal FVII is not required for the putative angiogenic functions of tissue factor in humans. © 1998 by The American Society of Hematology.


Blood ◽  
2008 ◽  
Vol 111 (5) ◽  
pp. 2681-2684 ◽  
Author(s):  
Mirko Pinotti ◽  
Lara Rizzotto ◽  
Dario Balestra ◽  
Marzena Anna Lewandowska ◽  
Nicola Cavallari ◽  
...  

Small nuclear U1-RNAs (snRNAs), the spliceosome components selectively recognizing donor splice sites (5′ss), were engineered to restore correct mRNA processing in a cellular model of severe coagulation factor VII (FVII) deficiency, caused by the IVS7 9726 + 5g/a change. Three U1-snRNAs, complementary to the mutated 5′ss (U1 + 5a) or to neighboring sequences were expressed with FVII minigenes in a hepatoma cell line. The U1-snRNAs reduced from 80% to 40% the exon 7 skipping, thus increasing exon definition. The U1 + 5a construct also dramatically increased recognition of the correct 5′ss over the 37-bp downstream cryptic site preferentially activated by the mutation, thus inducing appreciable synthesis of normal transcripts (from barely detectable to 50%). This effect, which was dose-dependent, clearly demonstrated that impaired recognition by the U1-snRNA was the mechanism responsible for FVII deficiency. These findings suggest compensatory U1-snRNAs as therapeutic tools in coagulation factor deficiencies caused by mutations at 5′ss, a frequent cause of severe defects.


1996 ◽  
Vol 76 (04) ◽  
pp. 492-499 ◽  
Author(s):  
L I Mennen ◽  
E G Schouten ◽  
D E Grobbee ◽  
C Kluft

Sign in / Sign up

Export Citation Format

Share Document