Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
1.
J Pediatr Hematol Oncol ; 45(6): e781-e783, 2023 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-37494615

RESUMO

Coagulation factor VII (FVII) deficiency is a congenital disorder with heterogeneous clinical phenotypes ranging from asymptomatic to life-threatening bleeding and/or thrombotic events. We present the case of an adolescent male who developed acute deep and superficial venous thromboses of the upper extremities in the setting of multiple peripheral venous line insertions and shortly after receiving his second coronavirus disease of 2019 immunization dose. A hemostatic work-up revealed low FVII activity levels associated with 4 different FVII genetic variants. We highlight the need to better understand the pathophysiologic mechanisms behind FVII deficiency-associated prothrombotic risk and the role that specific FVII genetic variants may play in the clinical presentation of these patients.


Assuntos
Infecções por Coronavirus , Coronavirus , Deficiência do Fator VII , Trombose , Masculino , Humanos , Deficiência do Fator VII/complicações , Deficiência do Fator VII/genética , Fator VII/genética , Imunização
2.
J Clin Lab Anal ; 37(1): e24768, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36572978

RESUMO

BACKGROUND: Congenital coagulation factor VII (FVII) deficiency is a rare, autosomal-recessive haemorrhagic disorder with an estimated incidence of 1:500,000. This disorder is caused by mutations in the F7 gene. CASE DESCRIPTION: Here, we report a pedigree of congenital FVII deficiency. The proband was a 30-year-old female with severely low FVII activity and a history of menorrhagia and epistaxis since her childhood who was subsequently diagnosed with congenital compound heterozygous FVII deficiency. A genetic study revealed a novel combination of compound heterozygous mutations (c.64G 〉 A, p.Gly22Ser and c.1027G 〉 A, p.Gly343Ser). Her father and older son had the c.64G 〉 A, p.Gly22Ser (heterozygous) mutation. Her mother and younger son had the c.1027G 〉 A, p.Gly343Ser (heterozygous) mutation. The predicted results of PolyPhen-2 and MutationTaster indicated that these mutations were probably damaging and disease-causing, respectively. CONCLUSION: In this study, we identified a novel combination of genetic mutations that could expand the mutant library and help in elucidating the pathogenesis of hereditary human coagulation FVII deficiency. A novel combination of compound heterozygous mutations was reported for the first time in Chinese individuals.


Assuntos
Transtornos da Coagulação Sanguínea , Deficiência do Fator VII , Humanos , Feminino , Criança , Adulto , Fator VII/genética , Linhagem , População do Leste Asiático , Deficiência do Fator VII/genética , Heterozigoto , Mutação/genética
3.
Haemophilia ; 29(2): 591-599, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36571800

RESUMO

INTRODUCTION: Inherited Factor VII (FVII) deficiency is commonest among the rare bleeding disorders. A small number of patients present in infancy with severe bleeding, and many may remain asymptomatic but detected before surgery/invasive procedures. Genetic testing may be helpful in predictive testing/prenatal diagnosis in severe cases. AIM: Characterisation of clinical and genotypic spectrum of patients with inherited FVII deficiency. METHODS: Retro-prospectively, 35 cases with prolonged prothrombin time and FVII activity (FVII:C) <50 IU/dl were subjected to targeted resequencing. After in-silico analysis, variant/s were validated by Sanger sequencing in index cases and family members. Haplotype analysis was done for F7 polymorphisms. RESULTS: Severe FVII deficiency was found in 50% of patients (FVII:C ≤1 IU/dl), and 42.9% were asymptomatic. Clinical severity assessment revealed 17% severe, 17% moderate and 22.9% patients with mild bleeds. FVII levels ranged from .3 to 38 IU/dl. Molecular analysis revealed variants in 30/35 cases, of which 17 were homozygous, 10 were compound heterozygous and 3 were heterozygous. Twelve genetic variants were identified, one promoter variant c.-30A>C; seven missense (c.215C>G, c.244T>C, c.253G>C, c.904G>A, c.961C>T, c.1109G>T, c.1211G>A), two deletions (c.21delG, c.868_870delATC), and one each of nonsense c.634C>T and splice-site variant c.316+1G>A. Recurrent variants c.1109G>T and c.215C>G were found in 17 and 8 cases, 12 of the former cases were homozygous. They had the same haplotype, indicating the founder effect in North Indians. CONCLUSION: This is the largest cohort of FVII genotyping from India, confirming heterogeneity in terms of clinical manifestations, FVII activity and zygosity of the variants with a limited genotypic phenotypic correlation.


Assuntos
Transtornos da Coagulação Sanguínea , Deficiência do Fator VII , Humanos , Efeito Fundador , Mutação , Deficiência do Fator VII/diagnóstico , Deficiência do Fator VII/genética , Fator VII/genética , Hemorragia
4.
Blood Coagul Fibrinolysis ; 33(8): 468-472, 2022 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-35867939

RESUMO

The congenital factor VII deficiency (FVIID) is a rare autosomal recessive haemorrhagic disease caused by mutations in the F7 gene. The aim of this study was to identify the mutations causing FVII deficiency and explain the genotype-phenotype association in two unrelated Chinese patients. Mutation detection was conducted by sequencing the whole F7 gene coding exons, exon-intron boundaries and the untranslated regions of 3' and 5'. Then, the genetic information was analyzed to predict the structures of the mutated proteins. A total of four different mutations were detected, including three missense mutations (c.64G>A, c.286A>G, and c.722C>A, predicting p.Gly22Ser, p.Arg96Gly, p.Thr241Asn, respectively) and one insertion mutation (c.204_205insCGGC, predicting p. Leu68Argfs ∗ 37), among which two were reported for the first time (p.Arg96Gly, p.Leu68Argfs ∗ 37). Multiple sequence alignments of FVII protein revealed that the residues p.Arg96 and p.Thr241 were highly conserved. The novel missense mutation p.Arg96Gly was determined as damaging with online software Polyphen-2 and SIFT. We investigated two asymptomatic patients diagnosed with severe FVII deficiency and identified two novel mutations (the mutation p.Arg96Gly and p.Leu68Argfs ∗ 37). Identification of the F7 mutations was important for genetic counseling and accurate prediction of the inheritance pattern.


Assuntos
Deficiência do Fator VII , Humanos , Deficiência do Fator VII/genética , Fator VII/química , Fenótipo , Genótipo , Éxons , Mutação
5.
J Clin Lab Anal ; 36(5): e24349, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35349734

RESUMO

BACKGROUND: Congenital factor VII (FVII) deficiency is a rare inherited autosomal recessive disorder characterized by prolongation of prothrombin time and low FVII coagulation activity, which may increase the risk of bleeding. CASE PRESENTATION: A 66-year-old man with acute postoperative intracranial hemorrhage was transferred to our hospital owing to coagulation dysfunction. In coagulation tests, the FVII coagulation activity was less than 2%. Genetic analysis of the gene encoding FVII identified compound heterozygous mutations: c. 681+1 G>T and c. C1286T (p. Ala429Val). CONCLUSIONS: To our knowledge, this is the first report describing the c. C1286T (p. Ala429Val) mutation in the FVII-encoding gene. We suggest that these mutations resulted in the reduced FVII activity and abnormal clotting in our patient after brain surgery.


Assuntos
Deficiência do Fator VII , Idoso , Fator VII/genética , Deficiência do Fator VII/complicações , Deficiência do Fator VII/genética , Heterozigoto , Humanos , Masculino , Mutação/genética , Tempo de Protrombina
6.
BMJ Case Rep ; 14(2)2021 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-33542014

RESUMO

A 32-year-old multiparous obese woman was referred to our center at 37 weeks of twin gestation. She was referred for birth planning following an accidentally discovered high international normalised ratio (INR) in routine preoperative labs. Her history was significant for recurrent pregnancy-associated deep venous thrombosis as well as two early pregnancy losses. Further work-up revealed transaminitis, mild splenomegaly and high lupus anticoagulant titre. A multidisciplinary team of physicians from the high-risk pregnancy, anaesthesiology, haematology, gastroenterology and hepatology departments put a management plan; it culminated into uncomplicated delivery of the patient by repeated caesarian section. The team was also able to figure out the cause of the patient's high INR that is associated with thrombophilia rather than haemophilia.


Assuntos
Deficiência do Fator VII/complicações , Coeficiente Internacional Normatizado , Trombofilia/complicações , Trombose Venosa , Adulto , Antineoplásicos Fitogênicos/uso terapêutico , Cesárea , Deficiência do Fator VII/genética , Feminino , Humanos , Inibidor de Coagulação do Lúpus/sangue , Hepatopatia Gordurosa não Alcoólica/diagnóstico , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , Plasma , Gravidez , Complicações Hematológicas na Gravidez/sangue , Complicações Hematológicas na Gravidez/tratamento farmacológico , Silibina/uso terapêutico , Trombofilia/genética , Trombose Venosa/complicações , Trombose Venosa/genética
7.
J Pediatr Hematol Oncol ; 43(8): e1059-e1061, 2021 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-33480651

RESUMO

Factor VII (FVII) deficiency is a rare bleeding disorder normally caused by homozygous and compound heterozygous mutations in the F7 gene. Whole-exome sequencing was performed to identify F7 mutations in 3 individuals from 2 unrelated families who were diagnosed with FVII deficiency. Four compound heterozygous mutations were identified and validated in these 3 probands with FVII deficiency. Among the 4 identified mutations, NM_000131.4:c.572-1_581del, NM_000131.4:c.1250A>G (p.Tyr417Cys), and NM_000131.4:c.647G>T (p.Gly216Val) were novel. All 3 novel mutations were predicted to be likely pathogenic by the American College of Medical Genetics and Genomics/Association for Molecular Pathology guidelines.


Assuntos
Deficiência do Fator VII/patologia , Fator VII/genética , Heterozigoto , Mutação , Adolescente , Criança , Deficiência do Fator VII/congênito , Deficiência do Fator VII/genética , Família , Feminino , Humanos , Masculino , Prognóstico
8.
Ann Biol Clin (Paris) ; 78(2): 198-200, 2020 04 01.
Artigo em Francês | MEDLINE | ID: mdl-32319949

RESUMO

Factor VII deficiency is the most common of the rare coagulation deficiencies. A hemorrhagic syndrome may occur in patients with FVII deficiency below 20%, although no correlation exist between the plasma FVII activity level (FVII:C) and the bleeding risk. Therefore, the management of surgery in patients with FVII deficiency remains challenging. Laboratory monitoring of FVII:C level may be helpful but should be interpreted with caution, because the dosage of FVII:C level may vary depending on the origin of the thromboplastin used. Herein, we report the case of the management of a woman who had been fortuitously diagnosed during pregnancy with FVII deficiency due to FVII variant Padua, which have induced discrepant results between two different laboratories.


Assuntos
Deficiência do Fator VII/diagnóstico , Deficiência do Fator VII/genética , Fator VII/genética , Fator VII/metabolismo , Complicações Hematológicas na Gravidez/diagnóstico , Tromboplastina/metabolismo , Adulto , Substituição de Aminoácidos , Coagulação Sanguínea/genética , Testes de Coagulação Sanguínea , Fator VII/análise , Deficiência do Fator VII/sangue , Feminino , Humanos , Achados Incidentais , Recém-Nascido , Mutação de Sentido Incorreto , Gravidez , Complicações Hematológicas na Gravidez/genética , Tromboplastina/análise
9.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 36(10): 1006-1009, 2019 Oct 10.
Artigo em Chinês | MEDLINE | ID: mdl-31598947

RESUMO

OBJECTIVE: To detect potential mutations of the coagulation factor Ⅶ (F7) gene in a pedigree affected with hereditary FⅦ deficiency and explore its molecular pathogenesis. METHODS: The FⅦ antigen (FⅦ:Ag) was analyzed by an enzyme-linked immunosorbent assay (ELISA) method. Prothrombin time (PT), FⅦ activity (FⅦ:C) and other coagulant parameters were quantified with an one-stage clotting assay. The F7 gene was amplified by PCR and sequenced. Mutational sites were confirmed by reverse sequencing. Impact of amino acid substitution was assessed using SIFT and PolyPhen-2 software. Structure of the mutant protein was analyzed using Swiss-pdb Viewer software based on the three-dimensional structure in the Protein Data Bank. RESULTS: The propositus had prolonged PT (36.3 s), with FⅦ:C and FⅦ:Ag significantly reduced to 2% and 44%, respectively. Her father, mother, younger sister and daughter had slightly prolonged PT and reduced FⅦ:C (86%-120%). The FⅦ:Ag of her father and younger sister were also reduced. DNA sequencing revealed that the propositus has carried compound heterozygous mutations (Lys341Glu and IVS6-1G>A) of the F7 gene. Her father and younger sister were heterozygous for the IVS6-1G>A mutation, while her mother and daughter were heterozygous for the Lys341Glu mutation. Bioinformatics analysis indicated that Lys341Glu mutation may affect the stability and function of the FⅦ protein. CONCLUSION: The Lys341Glu and IVS6-1G>A mutations probably underlie the reduced activity of FⅦ in this pedigree.


Assuntos
Deficiência do Fator VII/genética , Fator VII/genética , Feminino , Testes Genéticos , Heterozigoto , Humanos , Masculino , Mutação , Linhagem
10.
J Investig Med High Impact Case Rep ; 7: 2324709619872657, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31496295

RESUMO

Acquired isolated factor VII deficiency is a rare bleeding disorder and has been reported in 31 cases. This is in contrast to congenital factor VII deficiency, which while also infrequent is the most common rare congenital bleeding disorder. Acquired isolated factor VII deficiency has been described primarily in patients with solid malignancies, sepsis, and in the presence of anti-factor VII autoantibodies. We report a case of acute myelogenous leukemia with an associated trisomy 8 cytogenetic abnormality presenting with factor VII deficiency. The factor VII deficiency cleared after induction chemotherapy and with the disappearance of the cytogenetic and molecular abnormalities. We discuss a possible link between trisomy 8 and vitamin K metabolism, which might result in acquired factor VII deficiency in acute myelogenous leukemia.


Assuntos
Deficiência do Fator VII/etiologia , Leucemia Mieloide Aguda/complicações , Trissomia/patologia , Adulto , Antineoplásicos/uso terapêutico , Cromossomos Humanos Par 8 , Deficiência do Fator VII/sangue , Deficiência do Fator VII/genética , Feminino , Humanos , Leucemia Mieloide Aguda/sangue , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/genética
11.
Int J Biochem Cell Biol ; 113: 67-74, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31185295

RESUMO

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.


Assuntos
Calnexina/metabolismo , Retículo Endoplasmático/metabolismo , Deficiência do Fator VII/metabolismo , Fator VII/metabolismo , Polissacarídeos/metabolismo , Calnexina/genética , Fator VII/genética , Deficiência do Fator VII/genética , Variação Genética , Glicosilação , Células HEK293 , Células Hep G2 , Humanos , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutação , Dobramento de Proteína
12.
J Clin Pediatr Dent ; 43(4): 288-291, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31094634

RESUMO

The 13q deletion syndrome is a rare genetic disorder caused by structural and functional monosomy of chromosome 13. On 13q34, which is the terminal of the long arm, causative genes of coagulation factors VII and X (FVII and FX) are mapped. Patients with a combination of FVII and FX deficiencies are extremely rare and there have been few articles about perioperative coagulation support for such patients. Herein, we report on a case of bilateral cleft lip and palate accompanied by 13q deletion syndrome with deficiencies of FVII and FX. The chromosomal investigation indicated 46, XX, del(13)(q33) by G-banding. Prothrombin time and activated partial thromboplastin time were found to be 21.0 seconds (sec) (prothrombin time-international normalized ratio 1.76) and 41.6 sec (normal range; 23.9 - 39.7 sec), respectively. The activities of coagulation FVII and FX were 22% and 36%, respectively. A two-stage cheiloplasty was performed at 4 and 7 months of age followed by a palatoplasty at 1 year and 6 months. Tranexamic acid was given intravenously three times a day for three days after each surgery. There were no adverse events such as bleeding from the oral or nasal cavities and healing of the surgical wound was good without dehiscence.


Assuntos
Deleção Cromossômica , Transtornos Cromossômicos , Cromossomos Humanos Par 13 , Fenda Labial , Fissura Palatina , Deficiência do Fator VII , Transtornos Cromossômicos/complicações , Transtornos Cromossômicos/genética , Cromossomos Humanos Par 13/genética , Fenda Labial/genética , Fissura Palatina/genética , Fator VII , Deficiência do Fator VII/genética , Humanos , Palato
13.
BMC Med Genet ; 19(1): 163, 2018 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-30208845

RESUMO

BACKGROUND: Hemophagocytic lymfohistiocytosis (HLH) is a rare, life-threatening hyperinflammation, characterized by immune system over-activation resulting in hemophagocytosis. HLH could appear as a primary disease caused by mutations of immune-regulatory genes, or develop as a result of viral or bacterial infections, or malignancy. Congenital factor VII (FVII) deficiency is a rare autosomal recessive disorder characterized by prolonged prothrombin time (PT) and low FVII, which may increase bleeding risk. CASE PRESENTATION: A 50-year-old woman was admitted for a fever persisted for 20 days, presenting with cytopenia, high hyperferritinemia, low activity of NK cells. Bone marrow aspiration showed hemophagocytosis. CT scanning found pulmonary infection. EBV and CMV were not detected. Genetic scanning did not find pathogenic mutation of a HLH NGS panel including 26 genes. This patient was treated as recommended by the HLH 2004 Guidelines. Coagulation tests identified FVII deficiency. Genetic analysis of F7 gene in the patient and her family members identified recurrent compound heterozygous F7 c.64 + 5G > A and c.1224 T > G (p.His408Gln) mutations in this patient and her brother who showed postoperative hemorrhage after surgical resection of renal cell carcinoma. Heterozygotes in this family were asymptomatic. CONCLUSIONS: To our knowledge, this is the first report of HLH in combination with congenital FVII deficiency in Chinese population.


Assuntos
Deficiência do Fator VII/genética , Fator VII/genética , Linfo-Histiocitose Hemofagocítica/genética , Mutação , Adulto , Povo Asiático , Sequência de Bases , Análise Mutacional de DNA , Deficiência do Fator VII/congênito , Deficiência do Fator VII/etnologia , Deficiência do Fator VII/patologia , Feminino , Expressão Gênica , Heterozigoto , Humanos , Linfo-Histiocitose Hemofagocítica/etnologia , Linfo-Histiocitose Hemofagocítica/patologia , Masculino , Pessoa de Meia-Idade , Linhagem
14.
BMC Med Genet ; 19(1): 15, 2018 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-29368589

RESUMO

BACKGROUND: Preimplantation genetic diagnosis (PGD) is a powerful tool for preventing the transmission of Mendelian disorders from generation to generation. However, PGD only can identify monogenically inherited diseases, but not other potential monogenic pathologies. We aimed to use PGD to deliver a healthy baby without congenital FVII deficiency or other common Mendelian diseases in a couple in which both individuals carried a deleterious mutation in the F7 gene. METHODS: After both members of the couple were confirmed to be carriers of the F7 gene mutation by Sanger sequencing, expanded carrier screening (ECS) for 623 recessive inheritance diseases was performed to detect pathological mutations in other genes. PGD and preimplantational genetic screening (PGS) were employed to exclude monogenic disorders and aneuploidy for their embryos. RESULTS: ECS using targeted capture sequencing technology revealed that the couple carried the heterozygous disease-causative mutations c.3659C > T (p.Thr1220Ile) and c.3209G > A (p.Arg1070Gln) in the CFTR gene. After PGD and PGS, one of their embryos that was free of congenital FVII deficiency, cystic fibrosis (CF) and aneuploidy was transferred, resulting in the birth of a healthy 3200 g male infant. CONCLUSION: We successfully implemented PGD for congenital FVII deficiency and PGD after ECS to exclude CF for the first time to the best of our knowledge. Our work significantly improved the reproductive outcome for the couple and provides a clear example of the use of ECS combined with PGD to avoid the delivery of offspring affected not only by identified monogenically inherited diseases but also by other potential monogenic pathologies and aneuploidy.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/genética , Deficiência do Fator VII/diagnóstico , Deficiência do Fator VII/genética , Testes Genéticos , Diagnóstico Pré-Implantação , Aneuploidia , Fibrose Cística/diagnóstico , Fibrose Cística/genética , Fibrose Cística/prevenção & controle , Fator VII/genética , Fator VII/metabolismo , Deficiência do Fator VII/prevenção & controle , Feminino , Fertilização in vitro , Deleção de Genes , Genes Recessivos , Doenças Genéticas Inatas/diagnóstico , Doenças Genéticas Inatas/genética , Doenças Genéticas Inatas/prevenção & controle , Predisposição Genética para Doença , Humanos , Lactente , Masculino , Linhagem , Gravidez , Resultado da Gravidez , Análise de Sequência de DNA , Sequenciamento Completo do Genoma
15.
Biochim Biophys Acta Mol Basis Dis ; 1864(3): 660-667, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29246447

RESUMO

Activated factor (F) VII is a vitamin K-dependent glycoprotein that initiates blood coagulation upon interaction with tissue factor. FVII deficiency is the most common of the rare congenital bleeding disorders. While the mutational pattern has been extensively characterized, the pathogenic molecular mechanisms of mutations, particularly at the intracellular level, have been poorly defined. Here, we aimed at elucidating the mechanisms underlying altered FVII biosynthesis in the presence of three mutation types in the catalytic domain: a missense change, a microdeletion and a frameshift/elongation, associated with severe or moderate to severe phenotypes. Using CHO-K1 cells transiently transfected with expression vectors containing the wild-type FVII cDNA (FVIIwt) or harboring the p.I289del, p.G420V or p.A354V-p.P464Hfs mutations, we found that the secretion of the FVII mutants was severely decreased compared to FVIIwt. The synthesis rate of the mutants was slower than the FVIIwt and delayed, and no degradation of the FVII mutants by proteasomes, lysosomes or cysteine proteases was observed. Confocal immunofluorescence microscopy studies showed that FVII variants were localized into the endoplasmic reticulum (ER) but were not detectable within the Golgi apparatus. These findings suggested that a common pathogenic mechanism, possibly a defective folding of the mutant proteins, was triggered by the FVII mutations. The misfolded state led to impaired trafficking of these proteins causing ER retention, which would explain the low to very low FVII plasma levels observed in patients carrying these mutations.


Assuntos
Domínio Catalítico/genética , Deficiência do Fator VII/genética , Fator VII/química , Fator VII/genética , Mutação de Sentido Incorreto , Substituição de Aminoácidos , Animais , Células CHO , Cricetinae , Cricetulus , Retículo Endoplasmático/metabolismo , Humanos , Modelos Moleculares , Simulação de Dinâmica Molecular , Proteínas Mutantes/química , Proteínas Mutantes/genética , Dobramento de Proteína , Transporte Proteico/genética , Transdução de Sinais/genética
16.
Transfus Apher Sci ; 55(3): 364-367, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27776919

RESUMO

Thrombosis is known to occur in patients with rare inherited bleeding disorders, usually in the presence of a thrombotic risk factor such as surgery and/or factor replacement therapy, but sometimes spontaneously. We present the case of a 72-year-old African American male diagnosed with congenital factor VII (FVII) deficiency after presenting with ischemic stroke, presumably embolic, in the setting of atherosclerotic carotid artery stenosis. The patient had an international normalized ratio (INR) of 2.0 at presentation, with FVII activity of 6% and normal Extem clotting time in rotational thromboelastometry. He was treated with aspirin (325 mg daily) and clopidogrel (75 mg daily) with no additional bleeding or thrombotic complications throughout his admission. This case provides further evidence that moderate to severe FVII deficiency does not protect against thrombosis.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Deficiência do Fator VII/complicações , Deficiência do Fator VII/genética , Padrões de Herança/genética , Acidente Vascular Cerebral/complicações , Idoso , Testes de Coagulação Sanguínea , Isquemia Encefálica/complicações , Deficiência do Fator VII/tratamento farmacológico , Humanos , Masculino , Acidente Vascular Cerebral/tratamento farmacológico
17.
Anaesthesist ; 65(10): 746-754, 2016 Oct.
Artigo em Alemão | MEDLINE | ID: mdl-27586406

RESUMO

BACKGROUND: A hereditary deficiency in coagulation factor VII (FVII) may affect the international normalized ratio (INR) value. However, FVII deficiency is occasionally associated with a tendency to bleed spontaneously. We hypothesized that perioperative substitution with coagulation factor concentrates might not be indicated in most patients. METHODS: In this retrospective data analysis, we included all patients with hereditary heterozygous FVII deficiency who underwent surgical procedures at the University Hospital Basel between December 2010 and November 2015. In addition, by searching the literature, we identified publications reporting patients with FVII deficiency undergoing surgical procedures without perioperative substitution. RESULTS: We identified 22 patients undergoing 46 surgical procedures, resulting in a prevalence of 1:1500-2000. Coagulation factor concentrates were administered during the perioperative period in 15 procedures (33 %), whereas in the other 31 procedures (66 %), FVII deficiency was not substituted. No postoperative bleeding or thromboembolic events were reported. In addition, we found no differences in pre- and postoperative hemoglobin and coagulation parameters, with the exception of an improved postoperative INR value in the substituted group. In the literature review, we identified five publications, including 125 patients with FVII deficiency, undergoing 213 surgical procedures with no perioperative substitution. DISCUSSION: Preoperative substitution using coagulation factor concentrates does not seem to be mandatory in patients with an FVII level ≥15 %. For decision-making on preoperative substitution, patient history of an increased tendency to bleed may be more important than the FVII level or increased INR value.


Assuntos
Deficiência do Fator VII/complicações , Adulto , Idoso , Fator VII/uso terapêutico , Deficiência do Fator VII/epidemiologia , Deficiência do Fator VII/genética , Feminino , Heterozigoto , Humanos , Coeficiente Internacional Normatizado , Masculino , Pessoa de Meia-Idade , Assistência Perioperatória , Complicações Pós-Operatórias/sangue , Complicações Pós-Operatórias/prevenção & controle , Hemorragia Pós-Operatória/sangue , Hemorragia Pós-Operatória/prevenção & controle , Prevalência , Estudos Retrospectivos , Tromboembolia/sangue , Tromboembolia/prevenção & controle , Vitamina K/uso terapêutico
18.
Sci Rep ; 6: 28304, 2016 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-27341548

RESUMO

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.


Assuntos
Deficiência do Fator VII/terapia , Fator VII/genética , Regiões Promotoras Genéticas , Fatores de Transcrição/genética , Transcrição Gênica , Sequência de Bases , Sítios de Ligação , Fator VII/metabolismo , Deficiência do Fator VII/genética , Deficiência do Fator VII/metabolismo , Genes Reporter , Terapia Genética , Células HEK293 , Células Hep G2 , Hepatócitos/metabolismo , Humanos , Luciferases de Vaga-Lume/biossíntese , Luciferases de Vaga-Lume/genética , Mutação , Engenharia de Proteínas , Proteínas Recombinantes de Fusão , Ativação Transcricional
19.
Hematology ; 21(8): 486-9, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26872273

RESUMO

OBJECTIVE: To investigate the prevalence and type of thrombotic events reported in patients with congenital factor XI (FXI) or factor VII (FVII) deficiency. PATIENTS AND METHODS: Data on all patients with congenital FXI or FVII deficiency and a thrombotic event were gathered by means of a time unlimited PubMed search carried out in June 2014 and in February 2015. Appropriate keywords including the medical subject headings were used in both instances. Side tables were also consulted and cross-checking of the references was carried out to avoid omissions. The thrombosis event had to be proven by objective methods. RESULTS: Forty-three patients with FXI deficiency had arterial thrombosis and only eight had venous thrombosis. On the contrary, only five patients with FVII deficiency had arterial thrombosis whereas 31 patients had venous thrombosis. The arterial/venous ratios were 5.37 and 0.17 for FXI or FVII, respectively. CONCLUSIONS: Arterial thrombosis is frequent in FXI deficiency whereas venous thrombosis is rare. The reverse is true for FVII deficiency. The significance of these findings is discussed especially in view of the recent use of synthetic anti-FXI compounds in the prophylaxis of post-orthopedic surgery of venous thrombosis complications.


Assuntos
Deficiência do Fator VII/genética , Fator XI/genética , Trombose Venosa/genética , Humanos
20.
Blood ; 127(5): 565-71, 2016 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-26702064

RESUMO

Factor VII (FVII) deficiency is a rare autosomal recessive bleeding disorder treated by infusion of fresh-frozen plasma, plasma-derived FVII concentrates and low-dose recombinant activated FVII. Clinical data suggest that a mild elevation of plasma FVII levels (>10% normal) results in improved hemostasis. Research dogs with a G96E missense FVII mutation (FVII-G96E) have <1% FVII activity. By western blot, we show that they have undetectable plasmatic antigen, thus representing the most prevalent type of human FVII deficiency (low antigen/activity). In these dogs, we determine the feasibility of a gene therapy approach using liver-directed, adeno-associated viral (AAV) serotype 8 vector delivery of a canine FVII (cFVII) zymogen transgene. FVII-G96E dogs received escalating AAV doses (2E11 to 4.95E13 vector genomes [vg] per kg). Clinically therapeutic expression (15% normal) was attained with as low as 6E11 vg/kg of AAV and has been stable for >1 year (ongoing) without antibody formation to the cFVII transgene. Sustained and supraphysiological expression of 770% normal was observed using 4.95E13 vg/kg of AAV (2.6 years, ongoing). No evidence of pathological activation of coagulation or detrimental animal physiology was observed as platelet counts, d-dimer, fibrinogen levels, and serum chemistries remained normal in all dogs (cumulative 6.4 years). We observed a transient and noninhibitory immunoglobulin G class 2 response against cFVII only in the dog receiving the highest AAV dose. In conclusion, in the only large-animal model representing the majority of FVII mutation types, our data are first to demonstrate the feasibility, safety, and long-term duration of AAV-mediated correction of FVII deficiency.


Assuntos
Deficiência do Fator VII/genética , Deficiência do Fator VII/terapia , Fator VII/genética , Terapia Genética , Vetores Genéticos/genética , Vetores Genéticos/uso terapêutico , Precursores de Proteínas/genética , Adenoviridae/genética , Animais , Cães , Deficiência do Fator VII/sangue , Expressão Gênica , Vetores Genéticos/administração & dosagem , Células HEK293 , Humanos , Mutação Puntual , Transgenes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA