scholarly journals Mutations causing hemophilia B in Algeria: Identification of two novel mutations of the factor 9 gene

2018 ◽  
Vol 19 (1) ◽  
pp. 52-58 ◽  
Author(s):  
ZIDANI ABLA ◽  
YAHIA MOULOUD ◽  
EL MAHMOUDI HEJER ◽  
GOUIDER EMNA ◽  
ABDI MERIEM ◽  
...  

Abla Z, Mouloud Y, Hejer El, Emna G, Abdi Meriem A, Ouarhlent Yamina O, Naouel S. 2018. Mutations causing hemophilia B in Algeria: Identification of two novel mutations of the factor 9 gene. Biodiversitas 19: 52-58. Hemophilia B (HB) (also known as Christmas disease; Christmas is the family name of the first patient.) is an X linked recessive hemorrhagic disorder caused by mutations in factor 9 (F9: is used for the gene) gene that leads to deficient or defective coagulation factor IX (FIX: is used for the protein). The variable phenotype of HB results from wide range of mutations affecting the F9 gene. Our study was aimed at molecular analysis of HB to identify the causative mutation in known patients with HB in a part of Algeria. For genotyping, polymerase chain reaction (PCR) and direct sequencing have been applied to all the essential regions of the F9 gene from 39 Algerian HB patients belonging to 13 unrelated families. We identified 10 different mutations. The identified mutations included 1 duplication and 9 substitutions. In total 9 point mutations were identified, of which 5 are located in exon 8, the hotspot region in the F9 gene. Among the 10 mutations, 2 are novel and not deposited in database sites nor described in recently published articles. The results of this study emphasize the heterogeneity of HB. In summary, our preliminary results will be used to build an Algerian mutation database which would facilitate genetic counseling.

1994 ◽  
Vol 72 (01) ◽  
pp. 074-077 ◽  
Author(s):  
J Walter ◽  
I Pabinger-Fasching ◽  
H H Watzke

SummaryIn this report we describe the molecular basis of the factor IX (FIX) deficiency in nine patients with severe (n = 6), moderate (n = 1) or mild (n = 2) hemophilia B. The following genetic defects were identified by enzymatic amplification with the polymerase chain reaction (PCR) and subsequent direct sequencing of all exons and exon-intron-junctions: patient B.B. (FIX “Vienna I”): deletion of nucleotides 6343 to 6362; patient M.H. and W. J. (FIX “Vienna II”): nucleotide 17704 (C to G), Gin 97 to Glu; patient L. K. (FIX “Vienna III”): nucleotide 17761 (C to T), Arg 116 to stop; patient U. A. (FIX “Vienna IV”): nucleotide 10415 (C to G), Pro 55 to Ala; patient H.G. (FIX “Vienna V”): nucleotide 6488 (C to T), Thr 38 to lie; patient H. M. (FIX “Vienna VI”): nucleotide 31276 (G to C), Trp 385 to Cys; patient L. C. (FIX “Vienna VII”): deletion of nucleotide 6700; patient S.F. (FIX “Vienna VIII”): nucleotide 10392 (A to T), Asp 47 to Val. The causative mutation was detected in the FIX gene in each of the nine patients with hemophilia B. There was one small deletion, one point deletion and seven point mutations. The latter include six missense mutations and one nonsense mutation. The mutations in Vienna III, IV and V have already been described in previous studies. The two deletions, Vienna I and Vienna VII have not been reported previously. The genetic defects observed in Vienna II, VI and VIII are novel missense mutations which result in amino acid changes at residues 97,47 and 385, respectively.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 4790-4790
Author(s):  
Rong-Fu Zhou ◽  
Bo Gao ◽  
Jian Ou-yang

FTo investigate the molecular mutation leading to Hemophilia B in one patient. Methods FOne-stage method was used to detect APTT, PT, TT, Fibrinogen and FVIII:C, FIX:C. The genomic DNA was extracted from the peripheral blood of proband. All exons and their flanks of Factor IX gene were amplified by polymerase chain reaction (PCR). The PCR products were sequenced directly. Results FThe proband was a 20-month boy presenting with scalp hematoma after trauma. Regular coagulation tests showed that his APTT was 135.1s, PT 11.9s, TT 15.6s and Fibrinogen 2g/l. Normal mixed plasma could correct the prolonged APTT to 35s. His FIX:C was 6.4% and FVIII:C was normal. Direct sequencing of PCR products suggested that there was a 5085T>C mutation (NG_007994.1) in Exon1, and a 36060G>C mutation in Exon8 of F9 gene. The former mutation caused the substitution of Leu19 by Pro, which lies in -28th in signal peptide. The later mutation lead to the substituion of Gln370 by His. Conclusion FMutations of 5085T>C in Exon1 and 36060G>C in Exon8 might be the causes of coagulation factor IX defiency for the patient. These mutations are de novo ones according to the database presenting in http://www.factorix.org/. Disclosures: No relevant conflicts of interest to declare.


1993 ◽  
Vol 69 (03) ◽  
pp. 247-252 ◽  
Author(s):  
Shu-Wha Lin ◽  
Ming-Ching Shen

SummaryWe have characterized the genetic defects of 17 hemophilia B patients of Chinese origin by means of the polymerase chain reaction (PCR) and direct sequencing. The single-strand conformation polymorphism (SSCP) was used as an initial screening method to analyze the entire coding region and the flanking introns of each individual’s factor IX gene. The abnormal exons were subsequently amplified and the nucleotide sequence determined. Of the 17 patients studied, 16 had single point mutations and one had a gross gene deletion of exons VII and VIII of factor IX. Among these 16 factor IX variants with point mutations 13 were missense and two were nonsense mutations. The remaining one had a nucleotide deleted, resulting in frame shifting at amino acid residue 97. A total of ten novel mutations, including the one with gross gene deletion, are reported in this study which have not been described previously. Five of the remaining seven variants were missense mutations with novel amino acids substituted for residues 127, 132, 180, 207, and 215, respectively. Mutations containing different amino acid residues at those positions have been reported. The last two are variants that have already been described to contain mutations at amino acid residues 333 and 365, respectively. To evaluate the efficiency of SSCP analysis in assessing the mutated exons and to further confirm our results we sequenced the entire exons of all 17 factor IX genes. The mutations detected by SSCP method were indeed the only mutation identified in each factor IX variant. The SSCP analysis and direct sequencing have also allowed us to circumvent the difficulties of carrier determination for Chinese by direct detection of the abnormal factor IX alleles inherited by the females.


1995 ◽  
Vol 73 (05) ◽  
pp. 774-778 ◽  
Author(s):  
Marianne Schwartz ◽  
Jørgen Ingerslev ◽  
Elma Scheibel ◽  
Lise Rud Nielsen

SummaryHemophilia B is caused by a wide range of mutations. In order to characterize the mutations among patients in Denmark, we have systematically screened the entire coding region, the promoter region and exon flanking sequences of the gene encoding factor IX using single strand conformation and heteroduplex analyses. Patients from 32 different families were examined, and point mutations (23 different) were found in all of them. Ten of the mutations have not been reported by others; they include a splice site mutation, a single base pair deletion, and missense mutations. Notably, the study contains a female patient and a previously described Leyden mutation. In ten families with sporadic cases of hemophilia B, all 10 mothers were found to be carriers. The origin of two of these mutations was established.


Blood ◽  
1989 ◽  
Vol 73 (3) ◽  
pp. 743-746 ◽  
Author(s):  
PH Reitsma ◽  
T Mandalaki ◽  
CK Kasper ◽  
RM Bertina ◽  
E Briet

Abstract Hemophilia B Leyden is characterized by low levels of factor IX antigen and activity before the age of 15 years, whereas after puberty factor IX levels rise at a rate of about 5% per year. Two distinct point mutations (deletion of A, A----G) were identified at position +13 of the factor IX gene of a Greek and an American patient with hemophilia B Leyden. The nucleotide changes have occurred 32 basepairs downstream of a previously reported point mutation in a Dutch kindred with the same hemophilic phenotype. The results point to the importance of sequences surrounding the putative start site for the constitutive expression of the factor IX gene and to the possible significance of an imperfect direct repeat of DNA.


2013 ◽  
Vol 109 (01) ◽  
pp. 24-33 ◽  
Author(s):  
Liliana Carmen Rossetti ◽  
Miguel Martín Abelleyro ◽  
Miguel Candela ◽  
Raúl Pérez Bianco ◽  
Miguel de Tezanos Pinto ◽  
...  

SummaryIn haemophilia B (HB) (factor IX [FIX] deficiency), F9 genotype largely determines clinical phenotype. Aimed to characterise Argentinian families with HB, this study presents F9 genotype frequencies and their specific FIX inhibitor risk and 10 novel F9 mutations. Ninety-one DNA samples from HB patients and relatives were subjected to a new scheme: a primary screen for large deletions, a secondary screen for point mutations using conformation sensitive gel electrophoresis, DNA-sequencing and bioinformatic analysis. Our unbiased HB population (N=52) (77% with severe, 11.5% moderate and 11.5% mild HB) showed 32 missense (61.5%), including three novel mutations predicting specific structural/functional defects in silico, seven nonsense (13.5%) (one novel), five large deletions, four splice including three novel mutations affecting predicted splicing scores, three indels (two novel) and one Leiden mutation. Our comprehensive HB population included five patients with long-lasting FIX inhibitors: three nonsense (p.E35* (novel), p.R75*, p.W240*) and two entire-F9 deletions. Another patient with an indel (p.A26Rfs*14) developed transient inhibitors. A case-control analysis, based on our global prevalence of 3.05% for developing inhibitors in HB revealed that missense mutations were associated with a low risk odds ratio (OR) of 0.05 and a prevalence of 0.39%, whereas nonsense and entire-F9 deletions had significantly higher risks (OR 11.0 and 32.7) and prevalence (14.3% and 44.5%, respectively). Our cost-effective practical approach enabled identification of the causative mutation in all 55 Argentine families with HB, analysis of the molecular pathology of novel F9 defects and determination of mutation-associated FIX inhibitor risks.


2013 ◽  
Vol 11 (7) ◽  
pp. 1329-1340 ◽  
Author(s):  
P. M. Rallapalli ◽  
G. Kemball-Cook ◽  
E. G. Tuddenham ◽  
K. Gomez ◽  
S. J. Perkins

1999 ◽  
Vol 46 (3) ◽  
pp. 721-726 ◽  
Author(s):  
K Wulff ◽  
K Bykowska ◽  
S Lopaciuk ◽  
F H Herrmann

We examined the molecular basis of factor IX deficiency in 53 unrelated Polish patients with hemophilia B. Heteroduplex analysis and direct sequencing of polymerase chain reaction (PCR) products were applied to identify the gene defect. Forty-three different point mutations were detected in the factor IX gene of 47 patients. There were 29 missense mutations, 9 nonsense mutations, 4 splice site mutations and 1 point mutation in the promoter region. Twelve mutations were novel. The results of this study emphasize a very high degree of heterogeneity of hemophilia B.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2202-2202
Author(s):  
David Markusic ◽  
Sushrusha Nayak ◽  
Roland W Herzog

Abstract Abstract 2202 Hemophilia B is the X-linked bleeding disorder caused by loss of coagulation factor IX (F.IX) function. Current treatment relies on infusion of plasma derived or recombinant F.IX protein. Approximately 2–4% of hemophilia B patients develop inhibitory antibodies to F.IX protein, and those with F.IX gene deletions are at risk for anaphylaxis. We recently described a murine mode for this pathogenic antibody response, namely C3H/HeJ hemophilia B (HB) mice with a F9 gene deletion (PNAS 107:7101). Anaphylactic reactions were the result of a strong Th2-driven antibody response, comprised of IgG1 and IgE. In new experiments, we again found that repeated exposure to F.IX protein from weekly intravenous injections of recombinant human F.IX (1 IU/mouse) resulted in inhibitors, with 53% of the treated mice experiencing fatal anaphylaxis following the third administration. Surviving animals were given three additional intravenous F.IX protein injections (the fifth and sixth injections were given with antihistamine) and resulted in an average inhibitor titer of 6.2 BU/ml corresponding to an IgG1 titer of 5 mg/ml. Previously, we generated C3H/HeJ HB mice expressing non-functional crm- hF.IX variants due a missense mutation analogous to the UNC-Chapel Hill strain of hemophilia B dog (HB-CH) or due to a stop codon at amino acid residue 338 (HB-338). These mice, in contrast to the gene deletion mutation, failed to develop CD8+ T cell responses to hF.IX, but still formed inhibitors (albeit at reduced titers) upon muscle-directed gene transfer with AAV vector (Mol. Ther 17:1733). Furthermore, HB mice transgenic for a crim+ hF.IX missense mutation (HB-180) were entirely unresponsive to functional hF.IX. When challenged by a total of 6 weekly injections of hF.IX protein, all three strains (HB-338, HB-CH, and HB-180) tolerated the hF.IX antigen without any indication of inhibitors/IgG formation or anaphylaxis, which was in sharp contrast to the gene deletion mice. These results indicate that anaphylaxis in the murine model is F.IX genotype dependent, as it is in humans. In other experiments, naïve gene deletion mice were successfully tolerized to hF.IX by hepatic gene transfer. AAV8 or AAV2 (Y444/500/730F) capsid vectors containing a livers-specific expression cassette were injected into the tail vein. AAV8 vector induced tolerance over a wide range of vector doses. For optimized AAV2, hF.IX levels of 8–14% of normal were achieved with a dose of 2×10^11 vg/mouse, which protected from inhibitor formation and anaphylaxis upon subsequent challenge with F.IX protein. Next, we attempted to reverse the inhibitor response that had formed after protein therapy in gene deletion mice. These were repeatedly treated with hF.IX protein resulting in an average inhibitor of 6 BU/ml. Shortly afterwards, AAV8 vector expressing hF.IX (1×10^11 vg/mouse, n=4) was injected via the tail vein for hepatic gene transfer. Strikingly, this resulted in a complete reversal of antibody titers and systemic hF.IX levels of >40% of normal and aPTTs in the normal range. There was no evidence for an amnestic response, and the animals could be challenged with intravenous hF.IX protein without anaphylaxis. These results demonstrate that inhibitor formation resulting from F.IX replacement therapy can be reversed by liver gene transfer. Moreover, gene therapy in pre-immune mice corrected coagulation and protected from subsequent anaphylactic reactions after gene transfer. We are currently testing whether the protocol is safe for even higher pre-existing inhibitor titers. Disclosures: Herzog: Genzyme Corp: Patents & Royalties.


1990 ◽  
Vol 64 (02) ◽  
pp. 302-306 ◽  
Author(s):  
N S Wang ◽  
S H Chen ◽  
A R Thompson

SummaryPoint mutations in factor IX genes of four unrelated Chinese patients with hemophilia B have been identified by direct sequencing of amplified genomic DNA fragments. These four mutations occur in exon 8 of the factor IX gene. A C to T transition at nucleotide 30,863 changes codon 248 from Arg (CGA) to a new Stop codon (TGA), described in a previous family as factor IXMalmo3 (Green P M et al., EMBO J 1989; 8: 1067). A G to A transition, at nucleotide 31,051 changes codon 310 from Trp (TGG) to a nonsense or Stop codon (TGA; factor IXchongqing2)- A G to A transition at nucleotide 31,119 changes codon 333 which is for Arg (CGA) in normal factor IX, to one for Gin (CAA) in the variant previously described as factor IXLondon2 (Tsang T C et al., EMBO J 1988; 7: 3009) in a patient with moderately severe hemophilia B. The fourth patient has a novel C to A transversion at nucleotide 31,290, which corresponds to replacement of codon 390 which is for Ala (GCA) in normal factor IX, to one for Glu (GAA) in a patient with moderately severe hemophilia B (factor IXChongqing3)- DNA sequences of amplified fragments from mothers of three showed both their son’s variant and a normal nucleotide at the appropriate position, indicating that they are carriers. The fourth patient’s (factor IXMalmo3) mother, whose DNA was not evaluable, was most probably a carrier because of her low plasma factor IX levels.


Sign in / Sign up

Export Citation Format

Share Document