Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Thromb Res ; 237: 184-195, 2024 May.
Article in English | MEDLINE | ID: mdl-38631156

ABSTRACT

BACKGROUND AND AIMS: Blood disorders, such as sickle cell disease, and other clinical conditions are often accompanied by intravascular hemolytic events along with the development of severe coagulopathies. Hemolysis, in turn, leads to the accumulation of Fe(II/III)-protoporphyrin IX (heme) in the intravascular compartment, which can trigger a variety of proinflammatory and prothrombotic reactions. As such, heme binding to the blood coagulation proteins factor VIII (FVIII), fibrinogen, and activated protein C with functional consequences has been demonstrated earlier. METHODS: We herein present an in-depth characterization of the FVIII-heme interaction at the molecular level and its (patho-)physiological relevance through the application of biochemical, biophysical, structural biology, bioinformatic, and diagnostic tools. RESULTS: FVIII has a great heme-binding capacity with seven heme molecules associating with the protein. The respective binding sites were identified by investigating heme binding to FVIII-derived peptides in combination with molecular docking and dynamic simulation studies of the complex as well as cryo-electron microscopy, revealing three high-affinity and four moderate heme-binding motifs (HBMs). Furthermore, the relevance of the FVIII-heme complex formation was characterized in physiologically relevant assay systems, revealing a ~ 50 % inhibition of the FVIII cofactor activity even in the protein-rich environment of blood plasma. CONCLUSION: Our study provides not only novel molecular insights into the FVIII-heme interaction and its physiological relevance, but also strongly suggests the reduction of the intrinsic pathway and the accentuation of the final clotting step (by, for example, fibrinogen crosslinking) in hemolytic conditions as well as a future perspective in the context of FVIII substitution therapy of hemorrhagic events in hemophilia A patients.


Subject(s)
Factor VIII , Heme , Humans , Binding Sites , Blood Coagulation , Factor VIII/metabolism , Factor VIII/chemistry , Heme/metabolism , Molecular Docking Simulation , Protein Binding , Structure-Activity Relationship
2.
J Thromb Haemost ; 22(6): 1591-1604, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38453023

ABSTRACT

BACKGROUND: Antibodies against factor (F)VIII are a major complication in the treatment of patients with severe hemophilia A. The Nijmegen-Bethesda assay (NBA) is the gold standard for detection of neutralizing antibodies (inhibitors), whereas both inhibitors and nonneutralizing antibodies can be detected by immunoassays such as enzyme-linked immunosorbent assay (ELISA) and multiplex bead-based assays. OBJECTIVES: Evaluation of an in-house Luminex bead-based assay (LumiTope) compared with a commercially available ELISA and NBA. METHODS: The LumiTope method comprised full-length and B-domain-deleted FVIII as well as 9 purified FVIII single or multidomains. The respective proteins were coupled to magnetic beads to detect domain-specific immunoglobulin (IgG; IgG1-4) anti-FVIII antibodies in a large cohort of patients with hemophilia A with and without inhibitors. RESULTS: Overall, LumiTope assay had a high sensitivity (94.9%) and specificity (91.2%), particularly in patients with low-titer inhibitors compared with ELISA (sensitivity of 72.2% vs 27.7%). IgG4 was the most abundant IgG subclass in NBA-positive patients. NBA-positive and -negative patients showed different domain profiles. Patients with genetic variants in the heavy chain predominantly exhibited antibodies specific to this chain, while those with a light-chain variant showed a more diverse distribution of antibody specificities. Patients with an intron 22 inversion resembled those with a light-chain defect, with a majority of antibodies targeting the light chain. CONCLUSION: LumiTope assay provides a sensitive and specific method for not only detection but also domain specification of anti-FVIII-antibodies. Implementation of bead-based assays could improve antibody detection, profiling, and comparability of results and complement NBA.


Subject(s)
Enzyme-Linked Immunosorbent Assay , Factor VIII , Hemophilia A , Immunoglobulin G , Humans , Factor VIII/immunology , Hemophilia A/immunology , Hemophilia A/blood , Hemophilia A/diagnosis , Immunoglobulin G/blood , Immunoglobulin G/immunology , Enzyme-Linked Immunosorbent Assay/methods , Immunoassay/methods , Predictive Value of Tests , Reproducibility of Results , Male , Protein Domains , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Adolescent , Microspheres
3.
Int J Mol Sci ; 25(4)2024 Feb 17.
Article in English | MEDLINE | ID: mdl-38397060

ABSTRACT

Congenital factor VII (FVII) deficiency is a rare genetic bleeding disorder characterized by deficient or reduced activity of coagulation FVII. It is caused by genetic variants in the F7 gene. We aimed to evaluate the rate of detection of pathogenic variants in the F7 gene in a large group of patients with FVII deficiency and investigate the correlations between the F7 genotype and FVII activity (FVII:C). Moreover, the influence of the common genetic variant rs6046: c.1238G>A; p.(Arg413Gln), designated as the M2 allele, on FVII:C was investigated. Genetic analysis of the F7 gene was performed on 704 index patients (IPs) using either direct Sanger- or next-generation sequencing. Genetic variants were detected in 390 IPs, yielding a variant detection rate (VDR) of 55%. Notably, the VDR exhibited a linear decline with increasing FVII:C levels. We identified 124 genetic variants, of which 48 were not previously reported. Overall, the frequency of the M2 allele was considerably higher in patients with mild deficiency (FVII:C > 20 IU/dl). Furthermore, IPs lacking an identified pathogenic variant exhibited a significantly higher prevalence of the M2 allele (69%) compared to IPs with a disease-causing variant (47%). These results strongly support the association of the M2 allele with decreased FVII:C levels. This study shows the utility of FVII:C as a predictive marker for identifying pathogenic variants in patients with FVII deficiency. The M2 allele contributes to the reduction of FVII:C levels, particularly in cases of mild deficiency.


Subject(s)
Factor VII Deficiency , Humans , Factor VII Deficiency/genetics , Mutation , Phenotype , Factor VII/genetics , Genotype
4.
J Thromb Haemost ; 22(2): 379-393, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37832789

ABSTRACT

BACKGROUND: The characterization of inherited mild factor XIII deficiency is more imprecise than its rare, inherited severe forms. It is known that heterozygosity at FXIII genetic loci results in mild FXIII deficiency, characterized by circulating FXIII activity levels ranging from 20% to 60%. There exists a gap in information on 1) how genetic heterozygosity renders clinical bleeding manifestations among these individuals and 2) the reversal of unexplained bleeding upon FXIII administration in mild FXIII-deficient individuals. OBJECTIVES: To assess the prevalence and burden of mild FXIII deficiency among the apparently healthy German-Caucasian population and correlate it with genetic heterozygosity at FXIII and fibrinogen gene loci. METHODS: Peripheral blood was collected from 752 donors selected from the general population with essentially no bleeding complications to ensure asymptomatic predisposition. These were assessed for FXIII and fibrinogen activity, and FXIII and fibrinogen genes were resequenced using next-generation sequencing. For comparison, a retrospective analysis was performed on a cohort of mild inherited FXIII deficiency patients referred to us. RESULTS: The prevalence of mild FXIII deficiency was high (∼0.8%) among the screened German-Caucasian population compared with its rare-severe forms. Although no new heterozygous missense variants were found, certain combinations were relatively dominant/prevalent among the mild FXIII-deficient individuals. CONCLUSION: This extensive, population-based quasi-experimental approach revealed that the burden of heterozygosity in FXIII and fibrinogen gene loci causes the clinical manifestation of inherited mild FXIII deficiency, resulting in ''unexplained bleeding'' upon provocation.


Subject(s)
Factor XIII Deficiency , Factor XIII , Hemostatics , Humans , Factor XIII/genetics , Factor XIII Deficiency/diagnosis , Factor XIII Deficiency/genetics , Fibrinogen/genetics , Hemorrhage/diagnosis , Hemorrhage/genetics , Retrospective Studies
5.
J Thromb Haemost ; 21(6): 1503-1514, 2023 06.
Article in English | MEDLINE | ID: mdl-36934798

ABSTRACT

BACKGROUND: The standard therapy for patients with hemophilia A (HA) is the replacement with factor VIII (FVIII) therapeutics. To overcome the limitation of short half-life of wild-type FVIII protein, polyethylene glycol (PEG) can be coupled to therapeutic FVIII to improve pharmacokinetics. OBJECTIVES: We aimed to characterize antibodies developed against a FVIII therapeutic PEGylated with a 40-kDa PEG (40PEG-BDDFVIII) in 2 patients with mild HA. METHODS: An inhouse bead-based immunoassay was developed to characterize and confirm the specificity of the detected antibodies. The neutralizing nature of the antibodies toward PEGylated therapeutics was determined by a modified Nijmegen-Bethesda assay. RESULTS: Two out of 46 patients treated with 40PEG-BDDFVIII developed inhibitory antibodies toward the drug. Switching to a non-PEGylated FVIII successfully increased the FVIII activity in both patients. In patient 1, antibodies were raised against FVIII and PEG. Anti-FVIII antibodies were of the immunoglobulin (Ig)G isotype, whereas anti-PEG antibodies were of IgG, IgM, and IgA isotypes. In patient 2, antibodies of IgG and IgA isotypes were directed only against the PEG moiety. Competitive assays confirmed the specificity of the antibodies against PEG. The applied Nijmegen-Bethesda assay revealed that patients' anti-PEG antibodies and AGP3, an antibody against the backbone of PEG, can inhibit all currently available PEGylated therapeutics but to different degrees. No inhibitory FVIII antibodies were detected. CONCLUSION: Antibodies against the PEG moiety of 40PEG-BDDFVIII abolished the efficacy of the drug. This is the first report on real-world experiences with the development of neutralizing anti-PEG antibodies after treatment with PEGylated FVIII therapeutics in mild HA.


Subject(s)
Hemophilia A , Hemostatics , Humans , Factor VIII , Polyethylene Glycols/therapeutic use , Polyethylene/therapeutic use , Hemophilia A/drug therapy , Hemostatics/therapeutic use , Immunoglobulin G , Immunoglobulin A
6.
Hamostaseologie ; 42(6): 390-399, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36549291

ABSTRACT

Hemophilia A and hemophilia B are rare congenital, recessive X-linked disorders caused by lack or deficiency of clotting factor VIII (FVIII) or IX (FIX), respectively. The severity of the disease depends on the reduction of coagulation FVIII or FIX activity levels, which is determined by the type of the pathogenic variants in the genes encoding the two factors (F8 and F9, respectively). Molecular genetic analysis is widely applied in inherited bleeding disorders. The outcome of genetic analysis allows genetic counseling of affected families and helps find a link between the genotype and the phenotype. Genetic analysis in hemophilia has tremendously improved in the last decades. Many new techniques and modifications as well as analysis softwares became available, which made the genetic analysis and interpretation of the data faster and more accurate. Advances in genetic variant detection strategies facilitate identification of the causal variants in up to 97% of patients. In this review, we discuss the milestones in genetic analysis of hemophilia and highlight the importance of identification of the causative genetic variants for genetic counseling and particularly for the interpretation of the clinical presentation of hemophilia patients.


Subject(s)
Hemophilia A , Hemophilia B , Humans , Hemophilia A/diagnosis , Hemophilia A/genetics , Hemophilia B/diagnosis , Hemophilia B/genetics , Factor VIII/genetics , Genetic Testing/methods , Phenotype , Molecular Biology , Factor IX/genetics
8.
Int J Mol Sci ; 22(4)2021 Feb 20.
Article in English | MEDLINE | ID: mdl-33672736

ABSTRACT

Inactivation of thrombin by the endogenous inhibitor antithrombin (AT) is a central mechanism in the regulation of hemostasis. This makes hereditary AT deficiency, which is caused by SERPINC1 gene mutations, a major thrombophilic risk factor. Aim of this study was to assess to what extent AT mutations impair thrombin inhibition kinetics. The study population included 36 thrombophilic patients with 19 different mutations and mean AT levels of 65% in a thrombin-based functional assay, and 26 healthy controls. To assess thrombin inhibition kinetics, thrombin (3.94 mU/mL final concentration) was added to citrated plasma. Subsequently, endogenous thrombin inhibition was stopped by addition of the reversible thrombin inhibitor argatroban and the amount of argatroban-complexed thrombin quantified using an oligonucleotide-based enzyme capture assay. The plasma half-life of human thrombin was significantly longer in patients with AT mutations than in the controls (119.9 versus 55.9 s). Moreover, it was disproportionately prolonged when compared with preparations of wild type AT in plasma, in whom a comparable thrombin half-life of 120.8 s was reached at a distinctly lower AT level of 20%. These findings may help to better understand the increased thrombotic risk of SERPINC1 mutations with near normal AT plasma levels in functional assays.


Subject(s)
Antithrombins/metabolism , Mutation/genetics , Thrombin/antagonists & inhibitors , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Antithrombins/blood , Cattle , Child , Child, Preschool , Female , Half-Life , Humans , Kinetics , Middle Aged , Young Adult
9.
J Clin Med ; 10(2)2021 Jan 18.
Article in English | MEDLINE | ID: mdl-33477601

ABSTRACT

BACKGROUND: Familial multiple coagulation factor deficiencies (FMCFDs) are a group of inherited hemostatic disorders with the simultaneous reduction of plasma activity of at least two coagulation factors. As consequence, the type and severity of symptoms and the management of bleeding/thrombotic episodes vary among patients. The aim of this study was to identify the underlying genetic defect in patients with FMCFDs. METHODS: Activity levels were collected from the largest cohort of laboratory-diagnosed FMCFD patients described so far. Genetic analysis was performed using next-generation sequencing. RESULTS: In total, 52 FMCFDs resulted from coincidental co-inheritance of single-factor deficiencies. All coagulation factors (except factor XII (FXII)) were involved in different combinations. Factor VII (FVII) deficiency showed the highest prevalence. The second group summarized 21 patients with FMCFDs due to a single-gene defect resulting in combined FV/FVIII deficiency or vitamin K-dependent coagulation factor deficiency. In the third group, nine patients with a combined deficiency of FVII and FX caused by the partial deletion of chromosome 13 were identified. The majority of patients exhibited bleeding symptoms while thrombotic events were uncommon. CONCLUSIONS: FMCFDs are heritable abnormalities of hemostasis with a very low population frequency rendering them orphan diseases. A combination of comprehensive screening of residual activities and molecular genetic analysis could avoid under- and misdiagnosis.

10.
Front Cell Dev Biol ; 8: 669, 2020.
Article in English | MEDLINE | ID: mdl-32850803

ABSTRACT

Hemophilia A (HA) is a F8 gene mutational disorder resulting in deficiency or dysfunctional FVIII protein. However, surprisingly, in few cases, HA is manifested even without mutations in F8. To understand this anomaly, we recently sequenced microRNAs (miRNAs) of two patients with mild and moderate HA with no F8 gene mutations and selected two highly expressing miRNAs, miR-374b-5p and miR-30c-5p, from the pool to explain the FVIII deficiency that could be mediated by miRNA-based F8/FVIII suppression. In this report, an established orthogonal in vivo RNA-affinity purification approach was utilized to directly identify a group of F8-interacting miRNAs and we tested them for F8/FVIII suppression. From this pool, two miRNAs, miR-19b-3p and miR-186-5p, were found to be upregulated in a severe HA patient with a mutation in the F8 coding sequence and two HA patients without mutations in the F8 coding sequence were selected to demonstrate their role in F8 gene expression regulation in mammalian cells. Overall, these results provide further evidence for the hypothesis that by targeting the 3'UTR of F8, miRNAs can modulate FVIII protein levels. This mechanism could either be the primary cause of HA in patients who lack F8 mutations or control the severity of the disease in patients with F8 mutations.

11.
Nucleic Acid Ther ; 30(5): 276-288, 2020 10.
Article in English | MEDLINE | ID: mdl-32486960

ABSTRACT

Activated protein C (APC) is a serine protease with anticoagulant and cytoprotective activities. Nonanticoagulant APC mutants show beneficial effects as cytoprotective agents. To study, if such biased APC signaling can be achieved by APC-binding ligands, the aptamer technology has been used. A G-quadruplex-containing aptamer, G-NB3, has been selected that binds to the basic exosite of APC with a KD of 0.2 nM and shows no binding to APC-related serine proteases or the zymogen protein C. G-NB3 inhibits the inactivation of activated cofactors V and VIII with IC50 values of 11.6 and 13.1 nM, respectively, without inhibiting the cytoprotective and anti-inflammatory functions of APC as tested using a staurosporine-induced apoptosis assay and a vascular barrier protection assay. In addition, G-NB3 prolongs the plasma half-life of APC through inhibition of APC-serine protease inhibitor complex formation. These physicochemical and functional characteristics qualify G-NB3 as a promising therapeutic agent usable to enhance the cytoprotective functions of APC without increasing the risk of APC-related hemorrhage.


Subject(s)
Aptamers, Nucleotide/pharmacology , Hemorrhage/drug therapy , Protein C/pharmacology , Serine Proteases/pharmacology , Anticoagulants/pharmacology , Aptamers, Nucleotide/genetics , Blood Coagulation/drug effects , G-Quadruplexes , Hemorrhage/pathology , Humans , Ligands , Protein Binding/genetics , Protein C/genetics , Serine Proteases/genetics , Signal Transduction/drug effects , Thrombin/genetics
12.
Transfusion ; 60(2): 401-413, 2020 02.
Article in English | MEDLINE | ID: mdl-31785023

ABSTRACT

BACKGROUND: Hemophilia A (HA) is associated with mutations in the F8 gene that expresses factor VIII (FVIII). Unexpectedly, HA also manifests in a small subset of individuals with no mutations (exonic or intronic) in their F8 gene. MicroRNAs (miRNAs) cause translational interference, affecting protein quality and stoichiometry. Here, by analyzing miRNAs of two patients from this subset, we evaluated miRNA-based FVIII suppression as a testable hypothesis to explain FVIII deficiency in patients with HA with no F8 gene mutations. STUDY DESIGN AND METHODS: To test the hypothesis, miRNA sequencing from two patients with mild and moderate HA with no mutations in their F8 gene, followed by experimental verification, was used to identify a group of upregulated miRNAs in patients with HA compared to normal controls; with binding sites in the 3' untranslated region (UTR) of F8 messenger RNA (mRNA), a prerequisite for miRNA-based gene regulation. From this pool, miR-374b-5p and miR-30c-5p, known to be expressed in human liver, where FVIII is expressed, were subjected to extensive characterization. RESULTS: In two cell lines that constitutively express FVIII, we demonstrated that overexpression of miR-374b or miR-30c decreased FVIII expression, while an miR-30c inhibitor partially restored FVIII expression. CONCLUSION: These data support a role for microRNAs in fine-tuning F8 gene regulation. Based on our findings, our current model suggests that in HA cases where the F8 gene is normal and is predicted to express normal levels of FVIII, F8 mRNA 3' UTR targeting miRNAs may be responsible for a FVIII-deficiency phenotype clinically manifesting as HA.


Subject(s)
Factor VIII/genetics , Hemophilia A/genetics , Hemophilia A/pathology , Mutation/genetics , Blotting, Western , Cell Line , Fluorescent Antibody Technique , HEK293 Cells , HeLa Cells , Human Umbilical Vein Endothelial Cells , Humans , MicroRNAs/genetics , RNA, Messenger/genetics , Sequence Analysis, RNA
13.
Front Genet ; 10: 508, 2019.
Article in English | MEDLINE | ID: mdl-31191618

ABSTRACT

Diverse DNA structural variations (SVs) in human cancers and several other diseases are well documented. For genomic inversions in particular, the disease causing mechanism may not be clear, especially if the inversion border does not cross a coding sequence. Understanding about the molecular processes of these inverted genomic sequences, in a mainly epigenetic context, may provide additional information regarding sequence-specific regulation of gene expression in human diseases. Herein, we study one such inversion hotspot at Xq28, which leads to the disruption of F8 gene and results in hemophilia A phenotype. To determine the epigenetic consequence of this rearrangement, we evaluated DNA methylation levels of 12 CpG rich regions with the coverage of 550 kb by using bisulfite-pyrosequencing and next-generation sequencing (NGS)-based bisulfite re-sequencing enrichment assay. Our results show that this inversion prone area harbors widespread methylation changes at the studied regions. However, only 5/12 regions showed significant methylation changes, specifically in case of intron 1 inversion (two regions), intron 22 inversion (two regions) and one common region in both inversions. Interestingly, these aberrant methylated regions were found to be overlapping with the inversion proximities. In addition, two CpG sites reached 100% sensitivity and specificity to discriminate wild type from intron 22 and intron 1 inversion samples. While we found age to be an influencing factor on methylation levels at some regions, covariate analysis still confirms the differential methylation induced by inversion, regardless of age. The hemophilia A methylation inversion "HAMI" assay provides an advantage over conventional PCR-based methods, which may not detect novel rare genomic rearrangements. Taken together, we showed that genomic inversions in the F8 (Xq28) region are associated with detectable changes in methylation levels and can be used as an epigenetic diagnostic marker.

14.
PLoS One ; 14(6): e0218258, 2019.
Article in English | MEDLINE | ID: mdl-31194850

ABSTRACT

Replacement therapy in severe hemophilia A leads to factor VIII (FVIII) inhibitors in 30% of patients. Factor VIII gene (F8) mutation type, a family history of inhibitors, ethnicity and intensity of treatment are established risk factors, and were included in two published prediction tools based on regression models. Recently investigated immune regulatory genes could also play a part in immunogenicity. Our objective is to identify bio-clinical and genetic markers for FVIII inhibitor development, taking into account potential genetic high order interactions. The study population consisted of 593 and 79 patients with hemophilia A from centers in Bonn and Frankfurt respectively. Data was collected in the European ABIRISK tranSMART database. A subset of 125 severely affected patients from Bonn with reliable information on first treatment was selected as eligible for risk stratification using a hybrid tree-based regression model (GPLTR). In the eligible subset, 58 (46%) patients developed FVIII inhibitors. Among them, 49 (84%) were "high risk" F8 mutation type. 19 (33%) had a family history of inhibitors. The GPLTR model, taking into account F8 mutation risk, family history of inhibitors and product type, distinguishes two groups of patients: a high-risk group for immunogenicity, including patients with positive HLA-DRB1*15 and genotype G/A and A/A for IL-10 rs1800896, and a low-risk group of patients with negative HLA-DRB1*15 / HLA-DQB1*02 and T/T or G/T for CD86 rs2681401. We show associations between genetic factors and the occurrence of FVIII inhibitor development in severe hemophilia A patients taking into account for high-order interactions using a generalized partially linear tree-based approach.


Subject(s)
Factor VIII/genetics , Hemophilia A/genetics , Risk Assessment/methods , B7-2 Antigen/genetics , Biomarkers, Pharmacological , Blood Coagulation Tests , Factor VIII/antagonists & inhibitors , Factor VIII/metabolism , Genotype , Germany , HLA-DRB1 Chains/genetics , Hemophilia A/therapy , Humans , Interleukin-10/genetics , Multivariate Analysis , Mutation , Polymorphism, Single Nucleotide/genetics , Prospective Studies , Risk Factors
15.
Ann Hematol ; 98(8): 1855-1865, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30997536

ABSTRACT

Haemophilia A (HA) is caused by a lack or reduced amount of factor VIII protein (FVIII). About one-third of patients with non-severe HA carrying specific missense mutations show discrepant results between FVIII activity (FVIII:C), measured by one-stage or chromogenic two-stage assays. The aim of this study was to elucidate the mechanism underlying the assay discrepancy in vitro and in silico. Thirteen missense mutations in the Factor 8-gene associated with discrepant results in patients were transiently expressed. FVIII:C of the mutations was determined using two one-stage assays (FVIII:C1st, FVIII:CBonn) and a two-stage chromogenic assay (FVIII:Cchr). Furthermore, thrombin generation test (TGT) and in silico analysis were performed to investigate the haemostatic potential as well as the structural impact of the variants, respectively. For the majority (9/13) of the analysed mutations, the discrepancy was confirmed. Moreover, we established a modified TGT protocol for in vitro characterization of FVIII. Hence, TGT parameters were significantly impaired in the group of variants associated with higher chromogenic values. Additionally, in silico analysis revealed the impact of the mutations on FVIII protein structure leading to assay discrepancy. Moreover, the data shows that also among one-stage clotting assays, assay discrepancy is observed. Our results show that for the majority of mutations, application of a global assay like TGT method could help to improve diagnosis or correct assessment of the severity of HA.


Subject(s)
Biological Assay/standards , Factor VIII/chemistry , Hemophilia A/diagnosis , Hemophilia A/genetics , Mutation, Missense , Blood Coagulation Tests , Computer Simulation , Factor VIII/genetics , Factor VIII/metabolism , Gene Expression , Hemophilia A/blood , Hemophilia A/pathology , Humans , Male , Models, Molecular , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Severity of Illness Index , Thrombin/chemistry , Thrombin/metabolism
16.
Ann Hematol ; 97(6): 1061-1069, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29450643

ABSTRACT

One of the most common and unwanted side effects during oral anticoagulant therapy (OAT) is bleeding complications. In rare cases, vitamin K antagonist (VKA)-related bleeding events are associated with mutations affecting the F9 propeptide at amino acid position 37 due to a substitution of alanine to either valine or threonine. Based on our actual cohort of 18 patients, we update the knowledge on this rare phenotype and its origin. A founder mutation for both variants was reconfirmed by haplotype analysis of intronic and extragenic short tandem repeat (STR) polymorphisms with a higher prevalence in Switzerland than in other regions of Europe. Screening of healthy individuals for the presence of these F9 gene mutations did not identify any of these variants, thus proving the rare occurrence of this genotype. Furthermore, both variants were expressed in vitro and warfarin dose responses were studied. Our warfarin dose response analysis confirmed higher sensitivity of both variants to warfarin with the effect being more apparent for Ala37Thr. Thus, although F9 propeptide mutation-associated hypersensitivity to VKA is a rare phenomenon, awareness towards this bleeding phenotype is important to identify patients at risk.


Subject(s)
Anticoagulants/pharmacology , Factor IX/genetics , Mutation , Polymorphism, Genetic , Vitamin K/antagonists & inhibitors , Adult , Aged , Aged, 80 and over , Amino Acid Substitution , Anticoagulants/adverse effects , Cohort Studies , Factor IX/analysis , Factor IX/metabolism , Genetic Association Studies , Genetic Predisposition to Disease , HEK293 Cells , Hemorrhage/blood , Hemorrhage/chemically induced , Hemorrhage/genetics , Hemorrhage/metabolism , Humans , Male , Middle Aged , Protein Interaction Domains and Motifs , Recombinant Proteins/metabolism , Switzerland , Tandem Repeat Sequences , Warfarin/adverse effects , Warfarin/pharmacology
17.
PLoS One ; 12(7): e0180456, 2017.
Article in English | MEDLINE | ID: mdl-28683085

ABSTRACT

Coagulation Factor VIII is activated by an ordered limited thrombin proteolysis with different catalytic efficiency at three P1 Arginine residues: Arg759> Arg1708>Arg391, indicating the flanking residues of the latter to be less optimal. This study aimed to investigate, in silico and in vitro, the impact of possessing hypothetically optimized residues at these three catalytic cleavage sites. The structural impact of the residues flanking Arginine cleavage sites was studied by in silico analysis through comparing the cleavage cleft of the native site with a hypothetically optimized sequence at each site. Moreover, recombinant FVIII proteins were prepared by replacing the sequences flanking native thrombin cleavage sites with the proposed cleavage-optimized sequence. FVIII specific activity was determined by assessing the FVIII activity levels in relation to FVIII antigen levels. We further investigated whether thrombin generation could reflect the haemostatic potential of the variants. Our in silico results show the impact of the residues directly in the cleavage bond, and their neighboring residues on the insertion efficiency of the loop into the thrombin cleavage cleft. Moreover, the in vitro analysis shows that the sequences flanking the Arg1708 cleavage site seem to be the most close to optimal residues for achieving the maximal proteolytic activation and profactor activity of FVIII. The residues flanking the scissile bonds of FVIIII affect the cleavage rates and modulate the profactor activation. We were able to provide insights into the mechanisms of the specificity of thrombin for the P1 cleavage sites of FVIII. Thus, the P4-P2´ residues surrounding Arg1708 of FVIII have the highest impact on rates of thrombin proteolysis which contributes to thrombin activation of the profactor and eventually to the thrombin generation potential.


Subject(s)
Arginine/chemistry , Factor VIII/chemistry , Thrombin/chemistry , Amino Acid Motifs , Animals , Arginine/metabolism , Binding Sites , Biocatalysis , COS Cells , Chlorocebus aethiops , Cloning, Molecular , Factor VIII/genetics , Factor VIII/metabolism , Gene Expression , Humans , Kinetics , Models, Molecular , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Structure, Tertiary , Proteolysis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Substrate Specificity , Thermodynamics , Thrombin/metabolism
18.
Semin Thromb Hemost ; 40(8): 895-902, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25377322

ABSTRACT

Molecular genetic analysis is widely applied in inherited bleeding disorders. The outcome of genetic analysis allows genetic counselling in affected families and helps to find a link between the genotype and phenotype. Genetic analysis in hemophilia A (HA) has tremendously improved in the past decades. Many new techniques and modifications as well as analysis software have become available, which has enabled genetic analysis and interpretation of data to become faster and more accurate. The advances in mutation detection strategies facilitate the identification of the causal mutation in up to 97% of patients with HA. This review discusses the milestones in genetic analysis of HA and highlights the importance of identification of the causative mutations for genetic counseling and particularly for the interpretation of the clinical presentation of HA patients.


Subject(s)
Genetic Testing/history , Genetic Testing/methods , Hemophilia A/genetics , Hemophilia A/history , History, 20th Century , History, 21st Century , Humans
19.
Thromb Haemost ; 111(5): 851-61, 2014 May 05.
Article in English | MEDLINE | ID: mdl-24452774

ABSTRACT

About one-third of patients with non-severe haemophilia A (HA) show a discrepancy of factor (F)VIII activity (FVIII:C) measured by one-stage (FVIII:C1st), two-stage assays or the chromogenic method (FVIII:Cchr). The aim of the study was to characterise the mutation profile in patients with FVIII:C assay discrepancies. FVIII:C discrepancy was considered significant if the calculated ratio between FVIII:C1st and FVIII:Cchr was ≤ 0.6. In 16 patients FVIII:C1st was higher than FVIII:Cchr. The reversephenomenon was observed in 83 patients. Genetic analysis revealed 23 different missense mutations of which 17 were novel. Most mutations, exhibiting a higher FVIII:C1st were localised in the A1-A2-A3 interface. The majority of mutations associated with FVIII:Cchr>FVIII:C1st discrepancy were located close to or within the thrombin cleavage sites, FIX or vWF binding sites. Our data show a correlation between FVIII:C and thrombin generation testing with a clear differentiation between patients with haemophilia and normal controls. However, in the subgroup of FVIII:C1st>FVIII:Cchr discrepancy, the endogenous thrombin potential and peak thrombin parameters were similar to non-discrepant haemophilia patients, while in the inverse discrepancy FVIII:Cchr>FVIII:C1st, these variables showed values close to that of the normal control group. Certain hereditary F8 missense mutations cause discrepancy in FVIII:C as measured with different assays. This can lead to failure in diagnosing HA or incorrectly classifying the severity of the disease. Therefore, we recommend that initial diagnosis of non-severe HA phenotypes should be based on results of both FVIII:C1st and FVIII:Cchr assays.


Subject(s)
Blood Coagulation Tests/methods , Factor VIII/genetics , Hemophilia A/epidemiology , Hemophilia A/genetics , Mutation, Missense/genetics , Chromogenic Compounds/chemistry , Chromogenic Compounds/metabolism , Disease Progression , Early Diagnosis , Factor VIII/chemistry , Factor VIII/metabolism , Genetic Testing , Hemophilia A/diagnosis , Humans , Observer Variation , Phenotype , Protein Conformation
SELECTION OF CITATIONS
SEARCH DETAIL
...