Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
J Pediatr Hematol Oncol ; 42(6): e527-e530, 2020 08.
Article in English | MEDLINE | ID: mdl-31343480

ABSTRACT

INTRODUCTION: Inherited factor VII (FVII) deficiency is the most common of the rare bleeding disorders and shows a heterogenous distribution of bleeding phenotypes independent of factor activity level. The bleeding score (BS) evaluates the phenotype of patients with rare bleeding disorders. Thromboelastography (TEG) and thrombin generation assays (TGAs) are 2 methods to evaluate global hemostasis, and controversially both tests are useful for identifying different bleeding tendency phenotypes. The purpose of this study was to investigate the use of the BS and global assays (TEG and TGAs) to predict the bleeding phenotype of inherited FVII deficiency. MATERIALS AND METHODS: A total of 27 patients with FVII deficiency were evaluated with the BS and global hemostasis assays. RESULTS: The BS was compatible with disease severity according to the FVII activity level (P<0.05) but the BS and bleeding grade of patients did not show a statistically significant correlation with factor activity level (P>0.05). No significant correlation was observed between the factor activity level and any TEG parameter (P>0.05). The factor activity level was negatively correlated with the lag time of the TGA on the contrary positively correlated with the peak thrombin time of the TGA (P<0.05). CONCLUSIONS: The global assays do not successfully predict the bleeding phenotype. The BS is a more suitable tool than conventional and global assays for predicting the bleeding phenotype.


Subject(s)
Blood Coagulation Disorders, Inherited/diagnosis , Blood Coagulation Tests/methods , Factor VII Deficiency/diagnosis , Severity of Illness Index , Thrombelastography/methods , Thrombin/analysis , Adolescent , Blood Coagulation Disorders, Inherited/blood , Blood Coagulation Disorders, Inherited/metabolism , Case-Control Studies , Child , Child, Preschool , Factor VII Deficiency/blood , Factor VII Deficiency/metabolism , Female , Follow-Up Studies , Hemostasis , Humans , Male , Phenotype , Predictive Value of Tests
2.
Transfus Apher Sci ; 58(5): 569-571, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31447397

ABSTRACT

Factor VII (FVII) deficiency is the most common of the Rare Inherited Coagulation Disorders. The inheritance is autosomal recessive but there is variable penetrance. Overall there is poor correlation between the FVII level and the bleeding phenotype. Heterozygotes may have significant bleeding and severe homozygotes, or compound heterozygotes can be asymptomatic. Typically, homozygotes have FVII levels <10% and heterozygotes have levels above that. In most cases bleeding is uncommon with FVII levels>10-20%. A personal and family history is essential to determine the bleeding risk and to plan for surgical and obstetrical prophylaxis. Severe bleeding complications including central nervous system bleeding, gastrointestinal system bleeding and bleeding into the joints occurs in 10-15% of FVII deficient patients. Mucocutaneous bleeding is a common symptom but 30% of patients are asymptomatic. Fifty to 69% of women have heavy menstrual bleeding. Due to the limited number of publications regarding this rare disorder there are no consensus guidelines. There is registry data which has led to the best recommendations for treatment of bleeding episodes, initiation of long-term prophylaxis in addition to surgical plus ante and peripartum prophylaxis. Recombinant FVII concentrate is the best replacement therapy and a review of treatment and prophylaxis dosing is discussed.


Subject(s)
Factor VII Deficiency/drug therapy , Factor VII/therapeutic use , Hemorrhage/drug therapy , Factor VII Deficiency/metabolism , Factor VII Deficiency/pathology , Female , Hemorrhage/metabolism , Hemorrhage/pathology , Humans , Male
3.
Int J Biochem Cell Biol ; 113: 67-74, 2019 08.
Article in English | MEDLINE | ID: mdl-31185295

ABSTRACT

Factor VII (FVII) is a key serine protease in blood coagulation. N-glycosylation in FVII has been shown to be critical for protein secretion. To date, however, the underlying biochemical mechanism remains unclear. Recently, we found that N-glycans in the transmembrane serine protease corin are critical for calnexin-assisted protein folding and extracellular expression. In this study, we tested the hypothesis that N-glycans in the FVII protease domain mediate calnexin-assisted protein folding and that naturally occurring F7 mutations abolishing N-glycosylation impair FVII secretion. We expressed human FVII wild-type (WT) and mutant proteins lacking one or both N-glycosylation sites in HEK293 and HepG2 cells in the presence or absence of a glucosidase inhibitor. FVII expression, secretion and binding to endoplasmic reticulum chaperones were examined by immune staining, co-immunoprecipitation, Western blotting, and ELISA. We found that N-glycosylation at N360 in the protease domain, but not N183 in the pro-peptide domain, of human FVII is required for protein secretion. Elimination of N-glycosylation at N360 impaired calnexin-assisted FVII folding and secretion. Similar results were observed in WT FVII when N-glycan-calnexin interaction was blocked by glucosidase inhibition. Naturally occurring F7 mutations abolishing N-glycosylation at N360 reduced FVII secretion in HEK293 and HepG2 cells. These results indicate that N-glycans in the FVII protease domain mediate calnexin-assisted protein folding and subsequent extracellular expression. Naturally occurring F7 mutations abolishing N-glycosylation in FVII may impair this mechanism, thereby reducing FVII levels in patients.


Subject(s)
Calnexin/metabolism , Endoplasmic Reticulum/metabolism , Factor VII Deficiency/metabolism , Factor VII/metabolism , Polysaccharides/metabolism , Calnexin/genetics , Factor VII/genetics , Factor VII Deficiency/genetics , Genetic Variation , Glycosylation , HEK293 Cells , Hep G2 Cells , Humans , Mutant Proteins/genetics , Mutant Proteins/metabolism , Mutation , Protein Folding
4.
Mol Med Rep ; 17(2): 2738-2742, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29207137

ABSTRACT

Factor VII (FVII) serves an essential role in the initiation of blood coagulation. Mutations in conserved residues within its serine protease domain may lead to dysregulated coagulation activity. The objective of the present study was to elucidate the impact of altering two conserved residues, H348R and S282R, on the functional properties of the FVII protein. The mutation­harboring fragments were derived from genomic DNA of a FVII deficient patient. The fragments were integrated into a pcDNA vector containing FVII cDNA of HepG2 cells. The wild-type and mutated FVII constructs were transfected into CHO­K1 cells as a mammalian cell model. The coagulation activity, antigen levels and intracellular localization of the recombinant proteins were studied in association with their pathological importance. Results indicated that FVII activity was not detectable in conditioned media of the cells transfected with the mutated constructs. The H348R mutation reduced the expression of intracellular and secreted forms of the FVII protein. Following S282R transfection, intracellular FVII expression showed no significant variation; however, extracellular protein was reduced. The pattern of intracellular localization of mutated FVII remained unaltered in comparison to the wild-type protein. In conclusion, the present study suggested that missense mutations within the serine protease domain of FVII affect extracellular levels in addition to the coagulation activity of FVII. These results may contribute to further understanding of the molecular pathogenesis of FVII deficiency and the development of pharmaceutical candidates with improved therapeutic properties.


Subject(s)
Factor VII Deficiency/genetics , Factor VII/genetics , Gene Expression , Mutation , Amino Acid Substitution , Animals , CHO Cells , Cell Line , Cricetulus , Factor VII/metabolism , Factor VII Deficiency/blood , Factor VII Deficiency/metabolism , Genetic Association Studies , Humans , Immunohistochemistry , Transfection
5.
Blood Cells Mol Dis ; 67: 86-90, 2017 09.
Article in English | MEDLINE | ID: mdl-28038846

ABSTRACT

Severe congenital factor VII (FVII) deficiency is a rare bleeding disorder. Prophylaxis with replacement therapy has been suggested to patients, yet the most beneficial dosing regimens and therapy intervals are still to be defined. Due to the lack of evidence-based data, we hereby present our experience with long-term administration and monitoring primary prophylaxis in children with severe FVII deficiency and an extremely high bleeding risk. Four children with familial FVII deficiency, treated by prophylactic recombinant activated factor VII (rFVIIa), 15-30µg/kg/dose, given 2-3 times weekly since infancy, are discussed. Clinical follow up and monitoring laboratory assays, including thrombin generation, measured at various time points after prophylactic rFVIIa administration are presented. Among our treated patients neither FVII activity nor thrombin generation parameters (both already declined 24h post rFVIIa administration) were able to predict the impact of prophylaxis, and could not be used as surrogate markers in order to assess the most beneficial treatment frequency. However, the long clinical follow-up and comprehensive laboratory assessment performed, have shown that early primary prophylaxis as administered in our cohort was safe and effective.


Subject(s)
Factor VII Deficiency/prevention & control , Factor VIIa/therapeutic use , Child , Child, Preschool , Factor VII Deficiency/blood , Factor VII Deficiency/complications , Factor VII Deficiency/metabolism , Factor VIIa/administration & dosage , Female , Hemorrhage/blood , Hemorrhage/etiology , Hemorrhage/metabolism , Hemorrhage/prevention & control , Humans , Male , Recombinant Proteins/administration & dosage , Recombinant Proteins/therapeutic use , Thrombin/metabolism
6.
Sci Rep ; 6: 28304, 2016 Jun 24.
Article in English | MEDLINE | ID: mdl-27341548

ABSTRACT

Tailored approaches to restore defective transcription responsible for severe diseases have been poorly explored. We tested transcription activator-like effectors fused to an activation domain (TALE-TFs) in a coagulation factor VII (FVII) deficiency model. In this model, the deficiency is caused by the -94C > G or -61T > G mutation, which abrogate the binding of Sp1 or HNF-4 transcription factors. Reporter assays in hepatoma HepG2 cells naturally expressing FVII identified a single TALE-TF (TF4) that, by targeting the region between mutations, specifically trans-activated both the variant (>100-fold) and wild-type (20-40-fold) F7 promoters. Importantly, in the genomic context of transfected HepG2 and transduced primary hepatocytes, TF4 increased F7 mRNA and protein levels (2- to 3-fold) without detectable off-target effects, even for the homologous F10 gene. The ectopic F7 expression in renal HEK293 cells was modestly affected by TF4 or by TALE-TF combinations. These results provide experimental evidence for TALE-TFs as gene-specific tools useful to counteract disease-causing promoter mutations.


Subject(s)
Factor VII Deficiency/therapy , Factor VII/genetics , Promoter Regions, Genetic , Transcription Factors/genetics , Transcription, Genetic , Base Sequence , Binding Sites , Factor VII/metabolism , Factor VII Deficiency/genetics , Factor VII Deficiency/metabolism , Genes, Reporter , Genetic Therapy , HEK293 Cells , Hep G2 Cells , Hepatocytes/metabolism , Humans , Luciferases, Firefly/biosynthesis , Luciferases, Firefly/genetics , Mutation , Protein Engineering , Recombinant Fusion Proteins , Transcriptional Activation
7.
Semin Thromb Hemost ; 41(4): 366-73, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25973586

ABSTRACT

Factor VII (FVII) deficiency is one of the two congenital coagulation disorders that was not discovered by the description of a new bleeding patient whose clotting pattern did not fit the blood coagulation knowledge of the time (the other is factor XIII deficiency). The existence of an additional factor capable of accelerating the conversion of prothrombin into thrombin was suspected before 1951, the year in which the first family with FVII deficiency was discovered. As several investigators were involved in the discovery of FVII deficiency from both sides of the Atlantic, several different names were tentatively suggested to define this entity, namely stable factor (in contrast with labile factor or FV), cothromboplastin, proconvertin, serum prothrombin conversion accelerator, prothrombin acceleration, and autoprothrombin I. The last term was proposed by those who denied the existence of this new entity, which was instead considered to be a derivate of prothrombin activation, namely autoprothrombin. The description of several families, from all over the world, of the same defect, however clearly demonstrated the singularity of the condition. Factor VII was then proposed to define this protein. In subsequent years, several variants were described with peculiar reactivity toward tissue thromboplastins of different origin. Molecular biology techniques demonstrated several gene mutations, usually missense mutations, often involving exon 8 of the FVII gene. Later studies dealt with the relation of FVII with tissue factor and activated FVII (FVIIa). The evaluation of circulating FVIIa was made possible by the use of a truncated form of tissue factor, which is only sensitive to FVIIa present in the circulation. The development of FVII concentrates, both plasma derived and recombinant, has facilitated therapeutic management of FVII-deficient patients. The use of FVIIa concentrates was noted to be associated with the occasional occurrence of thrombotic events, mainly venous. Total or partial liver transplants have been performed with success in these patients and have "cured" their deficiencies. Prenatal diagnosis has also been performed and recent research involves the development of inhibitors of FVII + tissue factor complex or of FVIIa. This approach, if successful, could provide another antithrombotic therapeutics tool. The story of FVII well summarizes the efforts of both theoretical and clinical approaches in the characterization of a coagulation disorder, that is, among the rare bleeding conditions, most frequently encountered in clinical practice.


Subject(s)
Factor VII Deficiency , Factor VII , Factor VII/genetics , Factor VII/history , Factor VII/metabolism , Factor VII/therapeutic use , Factor VII Deficiency/drug therapy , Factor VII Deficiency/genetics , Factor VII Deficiency/history , Factor VII Deficiency/metabolism , History, 20th Century , History, 21st Century , Humans
8.
Thromb Res ; 132(1): 116-22, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23731565

ABSTRACT

Recombinant activated factor VII (rFVIIa) and plasma-derived factor VII (pdFVII) are used to prevent bleedings in severe FVII deficient patients, despite their short half-lifes. It is suggested that FVII levels of 15-20 IU/dL are sufficient to maintain hemostasis. We analyzed the pharmacodynamic effects of FVII substitution therapy in the Nijmegen Hemostasis Assay (NHA) that simultaneously measures thrombin and plasmin generation. Ten severe FVII deficient patients were treated with 20 µg/kg rFVIIa or 25 IU/kg pdFVII in a cross-over design. Thrombin generation lag-time (TG-LT) was identified as an effect-response parameter. Pharmacodynamic analysis using a maximum effect model showed 50% reduction of the TG-LT effect at ~2 IU/dL FVII activity for both rFVIIa and pdFVII. The FVII activity to obtain TG-LT comparable to the upper limit of normal range in healthy controls (4 min) was given by the effective concentration (ECnormal), showing sufficient hemostasis at 3-4 IU/dL FVII activity. No association was seen between FVII activity and other thrombin or plasmin generation parameters as measured by NHA. In conclusion, 3-4 IU/dL FVII activity seems sufficient to maintain hemostasis in patients with severe FVII deficiency during prophylaxis. These data may suggest a potential value for measurement of TG-LT in the monitoring of FVII(a) therapy.


Subject(s)
Factor VII Deficiency/blood , Factor VII Deficiency/drug therapy , Factor VII/therapeutic use , Factor VIIa/therapeutic use , Adult , Cohort Studies , Factor VII/pharmacology , Factor VII Deficiency/metabolism , Factor VIIa/pharmacology , Female , Fibrinolysin/metabolism , Hemostasis/drug effects , Humans , Male , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Thrombin/metabolism , Thrombin Time , Young Adult
9.
Thromb Res ; 131(2): 166-72, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23141848

ABSTRACT

INTRODUCTION: Congenital coagulation factor VII (FVII) deficiency is a rare coagulation disease. We investigated the molecular mechanisms of this FVII deficiency in a patient with compound heterozygous mutations. METHODS: A 22-year-old Japanese female was diagnosed with asymptomatic FVII deficiency. The FVII activity and antigen were greatly reduced (activity, 13.0%; antigen, 10.8%). We analyzed the F7 gene of this patient and characterized mutant FVII proteins using in vitro expression studies. RESULTS: Sequence analysis revealed that the patient was compound heterozygous with a point mutation (p.Leu13Pro) in the central hydrophobic core of the signal peptides and a novel non-sense mutation (p.Tyr294*) in the catalytic domain. Expression studies revealed that mutant FVII with p.Leu13Pro (FVII13P) showed less accumulation in the cells (17.5%) and less secretion into the medium (64.8%) than wild type showed. Truncated FVII resulting from p.Tyr294* (FVII294X) was also decreased in the cells (32.0%), but was not secreted into the medium. Pulse-chase experiments revealed that both mutants were extensively degraded intracellularly compared to wild type. The majority of FVII13P cannot translocate into endoplasmic reticulum (ER). However, a small amount of FVII13P was processed normally with post-translational modifications and was secreted into the medium. The fact that FVII294X was observed only in ER suggests that it is retained in ER. Proteasome apparently plays a central role in these degradations. CONCLUSIONS: These findings demonstrate that both mutant FVIIs impaired secretion through ineffective translocation to and retention in ER with extensive intracellular degradation, resulting in an insufficient phenotype.


Subject(s)
Factor VII Deficiency/blood , Factor VII Deficiency/genetics , Factor VII/metabolism , Point Mutation , Protein Processing, Post-Translational , Animals , COS Cells , Chlorocebus aethiops , Factor VII Deficiency/metabolism , Female , Humans , Transfection , Young Adult
10.
Hematology ; 17 Suppl 1: S156-8, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22507808

ABSTRACT

Inherited deficiencies of blood coagulation factors are usually associated with lifelong bleeding tendency. In addition to Haemophilias A and B and von Willebrand disease, congenital deficiencies of such factors as fibrinogen, prothrombin (FII)), FV, FVII, FX, FXI, FXIII, and combined deficiencies occur and can lead to a diversity of clinical conditions. Paradoxically, for some of these disorders associated with significant bleeding tendency there are reports of thrombotic events, both arterial and venous. Thrombosis in hemophilia patients has a multifactorial pathogenesis and the main conditions associated with this complication are the use of long-term central venous catheters, intensive replacement therapy usually in the setting of surgical procedures, the use of bypassing agents or the coexistence of acquired or inherited prothrombotic risk factors. Regarding other rare bleeding disorders, thrombotic phenomena has been described particularly in patients with afibrinogenemia, FXI and FVII deficiency and the events can occur even in young patients, in the presence of concomitant risk factors or spontaneously. Replacement therapy must be individualized and should take into account past history of haemostatic challenges, family history of bleeding and thrombosis, just like the level of factor. For mild deficiencies when patients are asymptomatic the use of antithrombotic prophylaxis must be considered with or without concomitant use of replacement therapy. In patients with history of thrombosis it may be helpful to perform a thrombophilia screening to exclude coexisting prothrombotic defects and for all patients it is recommended to control known cardiovascular disease risk factors.


Subject(s)
Hemorrhage/complications , Hemorrhagic Disorders/complications , Rare Diseases/complications , Thrombosis/complications , Blood Coagulation Disorders, Inherited/complications , Blood Coagulation Disorders, Inherited/metabolism , Factor VII Deficiency/complications , Factor VII Deficiency/metabolism , Factor XI Deficiency/complications , Factor XI Deficiency/metabolism , Fibrinogen/metabolism , Hemorrhage/metabolism , Hemorrhagic Disorders/metabolism , Humans , Rare Diseases/metabolism , Thrombosis/metabolism
11.
Haematologica ; 97(5): 705-9, 2012 May.
Article in English | MEDLINE | ID: mdl-22180436

ABSTRACT

We report 2 asymptomatic homozygotes for the nonsense p.R462X mutation affecting the carboxy-terminus of coagulation factor VII (FVII, 466 aminoacids). FVII levels of 3-5% and 2.7 ± 0.4% were found in prothrombin time-based and activated factor X (FXa) generation assays with human thromboplastins. Noticeably, FVII antigen levels were barely detectable (0.7 ± 0.2%) which suggested a gain-of-function effect. This effect was more pronounced with bovine thromboplastin (4.8 ± 0.9%) and disappeared with rabbit thromboplastin (0.7 ± 0.2%). This suggests that the mutation influences tissue factor/FVII interactions. Whereas the recombinant rFVII-462X variant confirmed an increase in specific activity (~400%), a panel of nonsense (p.P466X, p.F465X, p.P464X, p.A463X) and missense (p.R462A, p.R462Q, p.R462W) mutations of the FVII carboxy-terminus resulted in reduced secretion but normal specific activity. These data provide evidence for counteracting pleiotropic effects of the p.R462X mutation, which explains the asymptomatic FVII deficiency, and contributes to our understanding of the role of the highly variable carboxy-terminus of coagulation serine proteases.


Subject(s)
Blood Coagulation/genetics , Codon, Nonsense/genetics , Factor VII Deficiency/genetics , Factor VII Deficiency/metabolism , Factor VII/genetics , Factor VII/metabolism , Animals , Cattle , Child , Enzyme-Linked Immunosorbent Assay , Female , Heterozygote , Homozygote , Humans , Male , Middle Aged , Mutagenesis, Site-Directed , Prothrombin Time , Rabbits , Thromboplastin/metabolism
12.
Hematology ; 16(5): 308-12, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21902896

ABSTRACT

Congenital FVII deficiency is usually subdivided into two forms: type I and type II. Type I is characterized by a concomitant deficiency of FVII activity and FVII antigen (true deficiency). Type II is characterized by a discrepancy between FVII activity which is always low and FVII antigen which may be normal, near normal, or reduced. Thromboplastins of different origins may show a discrepant behaviour towards type II FVII deficiencies. The abnormal factor VII present in these forms may, in fact show, different levels of activity, according to the thromboplastin used in the assay system. Typical of these variants is the Arg304Gln mutation (know as FVII Padua). In this variant, FVII level is low when rabbit brain thromboplastin is used, whereas the level is perfectly normal when ox-brain thromboplastin is employed. Intermediate levels are obtained if human placenta or human recombinant is used. Since ox-brain thromboplastin is very sensitive to activated FVII, the normal FVII levels obtained in FVII Padua could be due to abnormally high circulating levels of activated FVII. The purpose of the present paper was to investigate the level of activated FVII present in homozygotes and heterozygotes with FVII Padua. For comparison, a group of patients with type I or 'true' deficiency was also investigated. A group of 21 normal patients served as controls. The activated FVII level found in FVII Padua was 8·4 and 41·0 mU/ml for homozygotes and heterozygotes, respectively. The level found in homozygous true deficiency was unassayable, whereas that found in heterozygotes was 36·2 mU/ml. The level found in the control population was 64·9 mU/ml in agreement with other reports. The low levels of activated FVIIa found in homozygotes with FVII Padua indicate that the normal FVII activity found with ox-brain thromboplastin cannot be attributed to higher than normal circulating levels of FVIIa.


Subject(s)
Factor VII Deficiency/genetics , Factor VII Deficiency/metabolism , Factor VII/metabolism , Factor VIIa/metabolism , Heterozygote , Homozygote , Adult , Blood Coagulation Tests , Female , Humans , Male , Middle Aged , Young Adult
13.
Clin Appl Thromb Hemost ; 17(6): E205-10, 2011.
Article in English | MEDLINE | ID: mdl-21441234

ABSTRACT

We investigated the mechanisms responsible for factor VII (FVII) deficiency in a compound heterozygous Japanese patient with mutations both in the signal peptide and in the catalytic domain. FVII activity (FVII:C) and antigen (FVII:Ag) levels of the patient were 14.5% and 12.5% of those of the normal controls, respectively. In all, 2 heterozygous point mutations were identified in the patient: one was the mutation substituting Pro for Leu-48 in the prepeptide domain of FVII; the other one was a novel mutation substituting Leu for Pro260 in the catalytic domain. FVII activity and FVII:Ag levels in the condition medium that transiently coexpressed the 2 different FVII mutants in baby hamster kidney (BHK) cells were 4.81% and 5.18% of the wild-type FVII. Factor VII defect of the patient may be combined with both impairing endoplasmic reticulum (ER) targeting and altering FVII folding/biosynthesis, but cotransfection of 2 different FVII mutants may interfere with their expression in BHK cells.


Subject(s)
Factor VII Deficiency/genetics , Leucine/genetics , Mutation , Animals , Cell Line , Child , Cricetinae , Factor VII Deficiency/metabolism , Heterozygote , Humans , Immunohistochemistry , Leucine/metabolism , Male , Transfection
14.
Pediatr Blood Cancer ; 52(3): 394-5, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19137513

ABSTRACT

We present the case of a 2-year-old female with Wilms tumor whose initial evaluation revealed a prolonged prothrombin time (PT) and normal activated partial thromboplastin time. Mixing studies demonstrated correction of the PT and the Factor VII activity was 17% in the absence of a Factor VII inhibitor. She underwent successful resection of the tumor with fresh frozen plasma support and no excessive bleeding. Post-operative testing demonstrated normal PT at 3 days and 1-month. Although acquired von Willebrand factor deficiency has a known association with Wilms tumor, paraneoplastic factor VII deficiency associated with Wilms tumor is previously unreported.


Subject(s)
Factor VII Deficiency/complications , Wilms Tumor/complications , Child, Preschool , Factor VII/metabolism , Factor VII Deficiency/metabolism , Factor VII Deficiency/therapy , Female , Humans , Wilms Tumor/metabolism , Wilms Tumor/surgery
16.
Br J Haematol ; 127(5): 576-84, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15566361

ABSTRACT

We report the results of in vitro expression and biochemical characterization of the naturally occurring type II mutation Pro303Thr (P303T) in the factor VII (FVII) gene. Recombinant activated mutated FVII (FVIIa303T), compared with the activated wild-type FVII (FVIIaWT), showed reduced amidase activity toward synthetic substrates, especially when the observed reduced binding affinity for human soluble tissue factor (TF) (K(d) from 4.4 nmol/l for FVIIaWT to 17.3 nmol/l for FVIIa303T) was overcome by a fully saturating TF concentration. Likewise, factor X (FX) hydrolysis by FVIIa303T showed a reduced activity in the absence (and more severely in the presence) of TF (k(cat)/K(m) from 2.3 x 10(7)/mol/l s for FVIIaWT to 8.7 x 10(5)/mol/l s for FVIIa303T). These results showed that the mutant FVIIa is more shifted toward a zymogen-like form compared to FVIIaWT, suggesting that P303 facilitates the conformational transitions that stabilize the active form of FVIIa. The alteration of these allosteric equilibria is especially evident in the presence of TF, which was unable to shift the equilibrium toward a fully active FVIIa form. Additional experiments showed that both TF-catalysed FVII303T autoactivation and FVII303T activation by activated FX in the presence of TF were severely impaired, mainly because of an increase of the K(m) value. Altogether, these defects may explain the severe bleeding symptoms in a patient carrying the FVIIP303T mutation.


Subject(s)
Factor VII Deficiency/genetics , Factor VII/genetics , Point Mutation , Adult , Blood Coagulation , Cell Line , Factor VII/metabolism , Factor VII Deficiency/metabolism , Factor VIIa/metabolism , Factor Xa/metabolism , Humans , Male , Protein Conformation , Recombinant Proteins/metabolism , Thromboplastin/metabolism , Transfection/methods
17.
Clin Lab ; 50(9-10): 529-38, 2004.
Article in English | MEDLINE | ID: mdl-15481628

ABSTRACT

Recombinant Factor VIIa (rVIIa) is a potent hemostatic agent for the management of refractory bleeding in patients with Factor VII deficiency or Factor VIII inhibitors. While the current recommended dose is usually effective, the most appropriate dose remains a subject of debate. Since factor VII levels and shortening of the pro-thrombin time do not appear to correlate with response, an appropriate laboratory marker of clinical response has not been identified. In this article we report changes noted in thrombin generation, platelet function and clot structure in blood from patients treated with rVIIa. Thrombin generation was assessed via a thrombin generation time (TGT) assay using a Hemodyne HAS instrument. Changes in clot structure were assessed as changes in clot elastic modulus in the HAS, changes in maximum amplitude in the TEG and changes in maximum clot firmness in the ROTEG. The cases presented confirmed improvement in thrombin generation with administration of rVIIa. The cases also illustrate that: a) in the factor VII deficient patient, 25% of the 90 microg/kg dose is sufficient to totally correct the defect, b) patients with high level factor VIII inhibitors may require significantly more than the recommended dose of 90 microg/kg, c) thrombin generation may not be completely corrected despite dramatic shortening of the prothrombin time, and d) increasing rVIIa doses does not by itself ensure improved thrombin generation.


Subject(s)
Blood Coagulation Disorders/drug therapy , Drug Monitoring , Factor VII/therapeutic use , Hemostasis , Hemostatics/therapeutic use , Recombinant Proteins/therapeutic use , Aged , Biomarkers , Blood Coagulation/drug effects , Blood Coagulation/physiology , Blood Coagulation Disorders/metabolism , Blood Platelets/drug effects , Blood Platelets/physiology , Child , Elasticity/drug effects , Factor VII Deficiency/drug therapy , Factor VII Deficiency/metabolism , Factor VIII/antagonists & inhibitors , Factor VIIa , Female , Hemostasis/drug effects , Humans , Male , Middle Aged , Thrombin/drug effects , Thrombin/metabolism
20.
Transplantation ; 39(4): 374-7, 1985 Apr.
Article in English | MEDLINE | ID: mdl-3920795

ABSTRACT

Procoagulant activity (PCA) of leukocytes of renal transplant recipients was studied. This material, which activates coagulation, has previously been shown to be released from macrophages after they interact with mitogen-stimulated or antigen-stimulated T lymphocytes. Under endotoxin-free conditions, PCA of peripheral blood leukocytes, incubated for 90 min in tissue culture, was elevated in postoperative transplant recipients and in many transplant patients tested around the time of a rejection episode. The response to lipopolysaccharide added during culture was also increased in these populations. The PCA response was factor-VII-dependent when tested with washed peripheral blood mononuclear cells (PBMC), but was factor-VII-independent when tested with unwashed PBMC in their original culture medium. The results indicate a possible link between immunologic events and coagulation in transplant recipients.


Subject(s)
Blood Coagulation Factors/metabolism , Kidney Transplantation , Culture Techniques , Factor VII Deficiency/metabolism , Factor VIII/physiology , Graft Rejection , Humans , Leukocytes/metabolism , Lipopolysaccharides/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...