Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 888
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Blood Cells Mol Dis ; 104: 102794, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37639740

RESUMO

BACKGROUND: Factor V (FV) is an essential cofactor in the coagulation cascade. The characterization of novel mutations is advantageous for the clinical management of FV-deficient patients. METHODS: Coagulation screening and thrombin generation assay were performed with the plate-poor plasma. All 25 exons of the F5 gene were amplified and sequenced. The ClustalX-2.1 software was applied to the multiple sequence alignment. The possible adverse effects of mutations were investigated with online bioinformatics software and protein modeling. RESULTS: Two unrelated families with FV deficiency were under investigation. Proband A was an 18-year-old youth with recurrent epistaxis. Proband B was a 29-year-old woman who did not present with any bleeding symptoms. Three heterozygous mutations (p.Gln1532*, p.Phe218Ser, and p.Asp2222Gly) were detected. Interestingly, they were compound heterozygotes and both contained the p.Asp2222Gly, a polymorphism. The thrombin generation assay showed that both patients had impaired ability of thrombin generation, and in particular, proband A was more severe. Conservation, pathogenicity and protein modeling studies all indicated that these three mutations could cause deleterious effects on the function and structure of FV. CONCLUSION: These three mutations are responsible for the FV-deficient in two pedigrees. Moreover, the nonsense variant p.Gln1532* is first reported in the world.


Assuntos
Deficiência do Fator V , Feminino , Adolescente , Humanos , Adulto , Deficiência do Fator V/genética , Trombina , Fator V/genética , Mutação , Heterozigoto
2.
Int J Mol Sci ; 24(7)2023 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-37047215

RESUMO

Proteostasis, i.e., the homeostasis of proteins, responsible for ensuring protein turnover, is regulated by proteases, which also participate in the etiopathogenesis of multiple conditions. The magic of proteases is such that, in blood coagulation, one same molecule, such as coagulation factor V, for example, can perform both a procoagulant and an anticoagulant function as a result of the activity of proteases. However, this magic has an insidious side to it, as it may also prevent the completion of the clinical value chain of factor V deficiency. This value chain encompasses the discovery of knowledge, the transfer of this knowledge, and its translation to clinical practice. In the case of rare and ultra-rare diseases like factor V deficiency, this value chain has not been completed as the knowledge acquisition phase has dragged out over time, holding up the transfer of knowledge to clinical practice. The reason for this is related to the small number of patients afflicted with these conditions. As a result, new indications must be found to make the therapies cost-effective. In the case of factor V, significant research efforts have been directed at developing a recombinant factor V capable of resisting the action of the proteases capable of inactivating this factor. This is where bioethics and health equity considerations come into the equation.


Assuntos
Deficiência do Fator V , Fator V , Humanos , Fator V/genética , Fator V/metabolismo , Anticoagulantes/farmacologia , Anticoagulantes/uso terapêutico , Peptídeo Hidrolases/farmacologia , Coagulação Sanguínea , Endopeptidases/farmacologia
3.
Zhonghua Yi Xue Za Zhi ; 103(17): 1323-1327, 2023 May 09.
Artigo em Zh | MEDLINE | ID: mdl-37150682

RESUMO

Objective: To analyze the gene variation of a genetic coagulation factor Ⅴ (FⅤ) deficiency pedigree and explore the molecular pathogenesis. Methods: The proband was a 32 years old female. The patient was prone to nose bleeding since childhood which was usually self-healed. On March 10, 2021, the proband went to the First Affiliated Hospital of Air Force Medical University for treatment of knee hematoma caused by a fall. None of the family members reported any history of bleeding. The prothrombin time (PT), activated partial thromboplastin time (APTT) and FⅤ activity (FⅤ: C) were detected by clotting method and the FⅤ antigen (FⅤ: Ag) was tested with enzyme-linked immunosorbent assay (ELISA). All exons and flanks of F5 gene were determined by Sanger sequencing. Clustalx-2.1-win, PolyPhen-2 and Swiss-PDBViewer software were used to analyze the conservatism of missense variation sites, whether the variations were harmful and their influences on protein structure and function. MutationTaster and NetGene2 software were used to analyze whether the splice site variation was harmful and its effect on the splice site. Results: The PT and APTT of the proband prolonged to 24.0 s and 69.8 s, respectively. The FⅤ: C and FⅤ: Ag decreased to 6% and 9%, respectively. There were compound heterozygous variations in F5 gene, which included c.911G>A heterozygous missense variation in exon 6 leading to p.Gly276Glu variation and c.5208+1G>A heterozygous missense variation in intron 15. The father and daughter had the p.Gly276Glu heterozygous variation. Her mother and son had the c.5208+1G>A heterozygous variation. Software analysis results of p.Gly276Glu heterozygous variation showed that Gly276 was conserved among homologous species, the variation was harmful, and it could affect the local structure and function of the protein. The c.5208+1G>A heterozygous variation was deleterious and resulted in the disappearance of the splice site, thereby affecting the protein function. Conclusion: The p.Gly276Glu and c.5208+1G>A compound heterozygous variants are deleterious variants associated with the patient's disease and may be the molecular pathogenesis of inherited FⅤ deficiency in this family.


Assuntos
Deficiência do Fator V , Fator V , Humanos , Feminino , Criança , Adulto , Linhagem , Fator V/genética , Mutação , Heterozigoto , Tempo de Tromboplastina Parcial , Deficiência do Fator V/genética
4.
Rinsho Ketsueki ; 64(2): 113-118, 2023.
Artigo em Japonês | MEDLINE | ID: mdl-36990730

RESUMO

Aortic regurgitation, a thoracoabdominal aortic aneurysm, chronic myeloid leukemia, and chronic kidney disease were all being treated at two hospitals for an 83-year-old man. He was admitted to the Department of Orthopedics at our hospital with a lumbar compression fracture. Later, he experienced melena, for which the Department of Internal Medicine was consulted. Due to the aberrant results of PT-INR (7.1) and a PTT > 200 seconds on a coagulation test, we suspected the presence of an autoimmune coagulation factor deficiency, and prednisolone immunosuppressive therapy medication was started right away. Due to a sharp decline in FV/5 activity, the presence of FV/5 inhibitors, and the presence of anti-FV/5 autoantibodies, a final diagnosis of autoimmune coagulation factor V (FV/5) deficiency was made. After the start of immunosuppressive therapy, the FV/5 inhibitor and anti-FV/5 autoantibodies disappeared, and the FV/5 activity progressively returned to normal. Disseminated intravascular coagulation-which may have been caused by a known aortic aneurysm-worsened while tapering off prednisolone. Due to the patient's advanced age and other problems, the aneurysm was extensive and inappropriate for surgical repair. The coagulation test findings improved gradually upon initiation of warfarin therapy. Herein, the patient had autoimmune FV/5 deficiency, a rare disorder that made diagnosis and therapy difficult because of the patient's several coexisting conditions.


Assuntos
Coagulação Intravascular Disseminada , Deficiência do Fator V , Masculino , Humanos , Idoso de 80 Anos ou mais , Fator V , Coagulação Intravascular Disseminada/etiologia , Coagulação Intravascular Disseminada/complicações , Prednisolona/uso terapêutico , Autoanticorpos
5.
Semin Thromb Hemost ; 48(2): 206-218, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-34942668

RESUMO

Coagulation factor V (or FV for the purpose of medical safety) is an essential cofactor of coagulation factor X in the common pathway of coagulation; severe FV deficiency leads to a bleeding tendency. Although both congenital and acquired FV deficiencies are widely recognized, FV deficiency also presents as an autoimmune disorder. A nationwide survey on autoimmune coagulation factor deficiencies (AiCFDs) conducted in Japan by our Japanese Collaborative Research Group identified 24 new patients with autoimmune FV deficiency (AiFVD) in the past 5 years. Furthermore, our extensive literature search confirmed that 177 AiFVD cases have been reported in previous articles published from Japan. Patients with AiFVD in Japan were predominantly men, with age similar to those with other AiCFDs. AiFVD was confirmed as a relatively mild type of bleeding diathesis, associated with lower mortality rate than that for AiFVD and other AiCFDs reported in previous studies. Patients with AiFVD had variable FV inhibitor titers and both neutralizing anti-FV autoantibodies and nonneutralizing counterparts. Although spontaneous resolution occurs in some patients, timely initiation of hemostatic and immunosuppressive therapies helps arrest the bleeding and eliminate anti-FV antibodies, resulting in a high cumulative recovery rate. Immunological anti-FV antibody detection is recommended to avoid missing AiFVD cases for the presence of nonneutralizing anti-FV autoantibodies. Further investigation is necessary to clarify the long-term prognosis and optimal management of AiFVD.


Assuntos
Transtornos da Coagulação Sanguínea , Deficiência do Fator V , Coagulação Sanguínea , Fator V , Humanos , Japão , Masculino
6.
Blood ; 136(23): 2679-2690, 2020 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-32663239

RESUMO

Quebec platelet disorder (QPD) is an autosomal dominant bleeding disorder with a unique, platelet-dependent, gain-of-function defect in fibrinolysis, without systemic fibrinolysis. The hallmark feature of QPD is a >100-fold overexpression of PLAU, specifically in megakaryocytes. This overexpression leads to a >100-fold increase in platelet stores of urokinase plasminogen activator (PLAU/uPA); subsequent plasmin-mediated degradation of diverse α-granule proteins; and platelet-dependent, accelerated fibrinolysis. The causative mutation is a 78-kb tandem duplication of PLAU. How this duplication causes megakaryocyte-specific PLAU overexpression is unknown. To investigate the mechanism that causes QPD, we used epigenomic profiling, comparative genomics, and chromatin conformation capture approaches to study PLAU regulation in cultured megakaryocytes from participants with QPD and unaffected controls. QPD duplication led to ectopic interactions between PLAU and a conserved megakaryocyte enhancer found within the same topologically associating domain (TAD). Our results support a unique disease mechanism whereby the reorganization of sub-TAD genome architecture results in a dramatic, cell-type-specific blood disorder phenotype.


Assuntos
Elementos Facilitadores Genéticos , Deficiência do Fator V , Duplicação Gênica , Regulação da Expressão Gênica , Megacariócitos/metabolismo , Proteínas de Membrana , Animais , Deficiência do Fator V/genética , Deficiência do Fator V/metabolismo , Deficiência do Fator V/patologia , Feminino , Humanos , Megacariócitos/patologia , Proteínas de Membrana/biossíntese , Proteínas de Membrana/genética , Peixe-Zebra
7.
J Pediatr Hematol Oncol ; 44(2): e482-e486, 2022 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-34387626

RESUMO

OBJECTIVE: The aim was to investigate the clinical characteristics and molecular pathogenic mechanism of twins with congenital factor V (FV) deficiency. METHODS: We comprehensively analyzed the clinical manifestations and laboratory test results of a set of twins and their parents and performed point mutation analysis with direct high-throughput exon sequencing. RESULTS: The prothrombin time and activated partial thromboplastin time were prolonged for both probands, and the FV activity levels were 13.0% and 9.8%. Next-generation sequencing showed that the affected individuals harbored a paternal c.5113A>C (p.S1705R) and a maternal c.4949C>T (p.A1650V) heterozygous variants in the FV gene, which conformed to an autosomal recessive inheritance pattern. This is the first report of these point mutations. The older boy also had a congenital patent foramen ovale. CONCLUSION: In this set of twins, missense mutations of the FV gene were related to congenital FV deficiency but unrelated to the patent foramen ovale observed in the older boy.


Assuntos
Proteínas de Ligação a Calmodulina/genética , Deficiência do Fator V , Forame Oval Patente , Proteínas dos Microfilamentos/genética , Resistência à Proteína C Ativada , Fator V/genética , Deficiência do Fator V/congênito , Deficiência do Fator V/genética , Heterozigoto , Humanos , Mutação , Linhagem , Fenótipo
8.
BMC Pregnancy Childbirth ; 22(1): 915, 2022 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-36482349

RESUMO

BACKGROUND: Factor V deficiency is a rare bleeding disorder that can be either congenital or acquired. Factor V deficiency mostly present with mucosal bleeding. Coagulation factor V does not increase considerably during normal gestation. Since pregnancy can be threatened by blood clotting disorders, abnormal changes in coagulation factors level can pose challenges to pregnant women. CASE PRESENTATION: We report a 40-year-old pregnant woman with prolonged gingival bleeding and epistaxis at 28 weeks of pregnancy. Her past medical history included two unexplained abortions. Physical examination was unremarkable, but the blood test showed elevated PT and PTT with a considerable decrease in factor V activity, while other factors were within normal range. Subsequently, the patient was diagnosed with congenital factor V deficiency. After treatment with fresh frozen plasma, she underwent vaginal delivery and a baby with factor V deficiency was born. CONCLUSIONS: This is the second report of recurrent miscarriage in congenital factor V deficiency patients. Clinicians should consider the possibility of factor V deficiency in women with a history of idiopathic miscarriage even in patients without any symptoms.


Assuntos
Aborto Habitual , Deficiência do Fator V , Feminino , Gravidez , Humanos , Adulto , Deficiência do Fator V/complicações , Deficiência do Fator V/diagnóstico , Aborto Habitual/etiologia
9.
J Clin Lab Anal ; 36(11): e24705, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36125894

RESUMO

BACKGROUND: Congenital coagulation factor V (FV) deficiency is a very rare hemorrhagic disease with an incidence of approximately one in a million. The common clinical manifestations of FV deficiency include ecchymosis and mucosal bleeding. Life-threatening intracranial bleeding is rare. It has been reported in several cases. However, the molecular basis has been established in only a few cases. METHODS: We reported a 2-month-old girl with congenital FV deficiency and intracranial hemorrhage. Coagulation screening combined with clinical manifestations was performed to diagnose congenital FV deficiency. Genetic testing was performed to identify the pathogenic genes. A literature review was included to emphasize the clinical manifestation, diagnosis, and treatment for congenital FV deficiency with intracranial bleeding. RESULTS: The coagulation tests revealed a significantly prolonged prothrombin time (PT) of 51 s and an activated partial thromboplastin time (APTT) of 73.7 s. The patient had a plasma FV activity of 0.9%. Genetic testing showed compound heterozygous mutations of the patient's FV gene. A literature review showed that patients with homozygous or compound heterozygous variants of the FV gene were often associated with a severe bleeding phenotype. CONCLUSION: Our study provides a direction for the rapid and accurate diagnosis and treatment for FV deficiency to avoid life-threatening bleeding. Infants with spontaneous cranial hematoma and intracranial hemorrhage should be investigated for underlying hemostatic defects. Congenital coagulation factor deficiency should be considered. Once congenital FV deficiency is diagnosed, fresh frozen plasma (FFP) should be given on a regular basis. Liver transplantation may be performed in severe cases.


Assuntos
Resistência à Proteína C Ativada , Deficiência do Fator V , Humanos , Fator V/genética , Deficiência do Fator V/complicações , Deficiência do Fator V/genética , Deficiência do Fator V/congênito , Hemorragias Intracranianas/genética
10.
J Cardiothorac Vasc Anesth ; 36(1): 242-245, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-33744111

RESUMO

Congenital factor V deficiency is an extremely rare abnormality and may be associated with significant bleeding as a result of trauma or surgery. Perioperative management primarily includes the administration of fresh frozen plasma to replace the deficient clotting factor. Acute administration of multiple blood products is a risk factor for transfusion-associated circulatory overload. Herein, the case of a 71-year-old man with an ejection fraction of 13% and a history of congenital factor V deficiency who was at risk for significant bleeding and transfusion-associated circulatory overload who underwent successful complex extraction of a right atrial pacing lead is reported.


Assuntos
Deficiência do Fator V , Reação Transfusional , Idoso , Fatores de Coagulação Sanguínea , Hemorragia , Humanos , Lasers , Masculino
11.
J Card Surg ; 37(12): 5493-5495, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36183387

RESUMO

BACKGROUND: Factor V deficiency is a rare disease, with an incidence of one in a million. Symptoms are mostly scant, and it is often diagnosed by the presence of an abnormality on PT-INR or APTT. In addition, no established therapy exists and platelet dysfunction is seldom found to be concomitant with this disease CASE PRESENTATION: A 64-year-old man who had both factor V deficiency and platelet dysfunction had angina in the past year. Coronary surgery was required, and we successfully performed coronary artery bypass grafting under strategic planned platelet transfusion with additional adequate cryoprecipitates transfusion. No perioperative problems nor any postoperative major bleeding issues were observed. The postoperative course was also uneventful. CONCLUSION: Strategic planned platelet transfusion with the additional transfusion of an adequate amount of cryoprecipitates is thus considered to be feasible for cases presenting with factor V deficiency and platelet dysfunction.


Assuntos
Procedimentos Cirúrgicos Cardíacos , Deficiência do Fator V , Masculino , Humanos , Pessoa de Meia-Idade , Deficiência do Fator V/terapia , Ponte de Artéria Coronária , Transfusão de Sangue , Hemorragia Pós-Operatória , Transfusão de Plaquetas
12.
Int J Mol Sci ; 23(10)2022 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-35628611

RESUMO

Factor V deficiency, an ultra-rare congenital coagulopathy, is characterized by bleeding episodes that may be more or less intense as a function of the levels of coagulation factor activity present in plasma. Fresh-frozen plasma, often used to treat patients with factor V deficiency, is a scarcely effective palliative therapy with no specificity to the disease. CRISPR/Cas9-mediated gene editing, following precise deletion by non-homologous end-joining, has proven to be highly effective for modeling on a HepG2 cell line a mutation similar to the one detected in the factor V-deficient patient analyzed in this study, thus simulating the pathological phenotype. Additional CRISPR/Cas9-driven non-homologous end-joining precision deletion steps allowed correction of 41% of the factor V gene mutated cells, giving rise to a newly developed functional protein. Taking into account the plasma concentrations corresponding to the different levels of severity of factor V deficiency, it may be argued that the correction achieved in this study could, in ideal conditions, be sufficient to turn a severe phenotype into a mild or asymptomatic one.


Assuntos
Deficiência do Fator V , Fator V , Sistemas CRISPR-Cas/genética , Fator V/genética , Deficiência do Fator V/genética , Edição de Genes , Humanos , Mutação
13.
Rinsho Ketsueki ; 63(7): 733-739, 2022.
Artigo em Japonês | MEDLINE | ID: mdl-35922940

RESUMO

Acquired factor V deficiency is a rare disease that presents with various bleeding symptoms because of the acquired production of factor V inhibitors and decrease in factor V activity. We have experienced five cases of acquired factor V deficiency diagnosed on the basis of abnormalities in coagulation tests in the last 10 years. All five patients were older men, of whom one had no bleeding symptoms, and three had a history of renal failure and malignant tumors. In the cross-mixing test, two of three cases demonstrated an inhibitor pattern, but one case showed a deficient pattern. In all cases, steroid treatment improved factor V activity as well as prothrombin time and activated partial thromboplastin time. However, patients with intracranial hemorrhage had a poor prognosis. Although this disease is rare, careful management is necessary, especially in the absence of bleeding symptoms and where cross-mixing test does not show an inhibitor pattern.


Assuntos
Deficiência do Fator V , Idoso , Testes de Coagulação Sanguínea/efeitos adversos , Fator V/genética , Deficiência do Fator V/complicações , Deficiência do Fator V/diagnóstico , Hemorragia/etiologia , Humanos , Masculino , Tempo de Tromboplastina Parcial , Tempo de Protrombina
14.
Blood ; 134(20): 1745-1754, 2019 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-31558466

RESUMO

Combined factor V (FV) and FVIII deficiency (F5F8D) is a rare autosomal-recessive bleeding disorder caused by mutations in lectin mannose binding-1 (LMAN1) and multiple coagulation factor deficiency-2 (MCFD2). Six causative homozygous mutations (5 in LMAN1 and 1 in MCFD2) were identified in 6 patients with F5F8D. A thrombin-generation assay, triggered with tissue factor (1 pM) in F5F8D plasma, paradoxically exhibited enhanced thrombin generation compared with normal plasma. Significantly lower free tissue factor pathway inhibitor (fTFPI) was found in F5F8D patients compared with healthy controls (P < .01). Normalizing tissue factor pathway inhibitor α (TFPIα) in F5F8D plasma greatly delayed and reduced thrombin generation. Increasing FV concentrations by adding plasma FV to F5F8D plasma only caused a gradual decrease in thrombin generation, suggesting that low levels of TFPIα and FV cocontributed to the elevated thrombin generation by reducing anticoagulant effects. On the contrary, thrombin generation in F5F8D platelet-rich plasma (PRP) was significantly lower than in normal controls (P < .05); however, it was fully corrected by normalizing FVIII or after 1-deamino-8-d-arginine vasopressin (DDAVP) infusion, indicating that the hypocoagulable state of F5F8D patients is associated with low FVIII levels. In addition, plasma and platelet FV in F5F8D PRP were sufficient to support normal thrombin generation, and low TFPIα may have no effect on thrombin generation. DDAVP infusion induced a complete response in 5 F5F8D patients and a partial response in the remaining patient. Based on our findings, we suggest that DDAVP may be considered a potential substitute for FVIII concentrates, and fresh-frozen plasma (FFP) infusion may not be necessary for F5F8D patients with minor bleeding challenges.


Assuntos
Deficiência do Fator V/sangue , Fator V/análise , Hemofilia A/sangue , Hemorragia/sangue , Adulto , Deficiência do Fator V/complicações , Feminino , Hemofilia A/complicações , Hemorragia/etiologia , Hemostasia , Humanos , Masculino , Pessoa de Meia-Idade , Trombina/análise , Adulto Jovem
15.
Transfusion ; 61(2): 405-409, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33166428

RESUMO

BACKGROUND: Allogeneic platelet (PLT) infusion is a strategy to raise Factor V (FV) levels in patients with congenital FV deficiency. However, since FV is labile in vitro, we hypothesized that FV activity could be low in PLT units. STUDY DESIGN AND METHODS: FV activity was tested using a prothrombin time-based platform in the supernatant and platelet lysate (PL) of apheresis PLT units (16 units stored in PLT additive solution with acetate and phosphate [PAS-C] and 10 units stored in plasma only), on post-collection days 3-6. Statistical analysis was performed using Student's t test (P < .05). RESULTS: FV activity was severely diminished in PAS-C PLTs (N = 16) supernatant (3.70% ± 1.02%) and PL (3.26% ± 1.02%). FV activity in plasma-only PLTs (N = 10) was lower in both supernatant (44.55% ± 6.46%) and lysate (39.67% ± 6.33%) relative to normal plasma levels, but both were significantly higher (P < .0001) compared to PAS-C PLTs. In a separate set of experiments, FV activity in PAS-C PLTs examined serially over storage time (N = 3 for these experiments) showed that FV levels were reduced by day 3 and not significantly different by day 5 of storage (Day 3 supernatant 5.03% ± 1.41%; Day 5 supernatant: 3.10% ± 0.57%; P = .2; Day 3 lysate: 3.89% ± 1.03%; Day 5 lysate: 2.61% ± 0.41%; P = .4). CONCLUSION: Plasma should be considered over PLTs as first-line therapy for non-complex FV deficiency-associated hemorrhage. If PLTs are considered for transfusion, plasma-only PLT units should be preferentially utilized, as PAS-C PLT have near-absent FV activity.


Assuntos
Plaquetas/química , Deficiência do Fator V/terapia , Fator V/análise , Transfusão de Plaquetas , Plaquetoferese , Transfusão de Componentes Sanguíneos , Meios de Cultivo Condicionados/química , Grânulos Citoplasmáticos/química , Deficiência do Fator V/sangue , Deficiência do Fator V/complicações , Hemorragia/etiologia , Hemorragia/prevenção & controle , Humanos , Plasma , Tempo de Protrombina
16.
Acta Haematol ; 144(1): 82-87, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32784304

RESUMO

Acquired factor V deficiency (AFVD) is a rare autoimmune bleeding disorder. Unlike acquired hemophilia, bypass therapies with recombinant activated factor VII and activated prothrombin complex concentrates are ineffective for severe bleeding due to AFVD. Although several treatment strategies have been attempted, a standard of care for severe hemorrhage induced by AFVD is lacking. Herein, we report a case of AFVD with severe bleeding that responded to plasma exchange (PE) combined with immunosuppression. We also reviewed previously reported AFVD cases with severe hemorrhage and suggest that PE may be an effective initial treatment for AFVD-induced severe hemorrhage.


Assuntos
Deficiência do Fator V/complicações , Hemorragia/etiologia , Hemorragia/terapia , Troca Plasmática , Autoimunidade , Biomarcadores , Testes de Coagulação Sanguínea , Deficiência do Fator V/diagnóstico , Deficiência do Fator V/etiologia , Hemorragia/sangue , Hemorragia/diagnóstico , Humanos , Masculino , Pessoa de Meia-Idade , Troca Plasmática/efeitos adversos , Troca Plasmática/métodos , Índice de Gravidade de Doença , Avaliação de Sintomas , Tomografia Computadorizada por Raios X , Resultado do Tratamento
17.
Acta Haematol ; 144(6): 712-716, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34280927

RESUMO

Hereditary factor V (FV) deficiency is a rare autosomal recessive bleeding disorder caused by F5 gene mutations. The objective of this study was to investigate the p.Phe218Ser and p.Gly304Glu variants found in 2 families with hereditary FV deficiency. The FV activity (FV:C) and FV antigen (FV:Ag) were measured by clotting and ELISA, respectively. The F5 gene and sequence conservation were analyzed by direct sequencing and ClustalX-2.1-win, respectively. One proband carried a homozygous p.Phe218Ser (c.653T>C) mutation, with FV:C and FV:Ag decreased to 11 and 14%, respectively. The other proband carried a heterozygous p.Gly304Glu (c.911G>A) mutation, with FV:C and FV:Ag reduced to 55 and 62%, respectively. Phe218 and Gly304 were highly conserved in the homologous gene in 9 other species. We hypothesized that the p.Phe218Ser and p.Gly304Glu variants are deleterious and responsible for the reduction in FV:C and FV:Ag.


Assuntos
Deficiência do Fator V/diagnóstico , Fator V/genética , Adulto , Sequência de Aminoácidos , Testes de Coagulação Sanguínea , Ensaio de Imunoadsorção Enzimática , Fator V/análise , Feminino , Heterozigoto , Homozigoto , Humanos , Masculino , Mutação de Sentido Incorreto , Linhagem , Análise de Sequência de DNA
18.
Int J Mol Sci ; 22(18)2021 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-34575869

RESUMO

Factor V is an essential clotting factor that plays a key role in the blood coagulation cascade on account of its procoagulant and anticoagulant activity. Eighty percent of circulating factor V is produced in the liver and the remaining 20% originates in the α-granules of platelets. In humans, the factor V gene is about 80 kb in size; it is located on chromosome 1q24.2, and its cDNA is 6914 bp in length. Furthermore, nearly 190 mutations have been reported in the gene. Factor V deficiency is an autosomal recessive coagulation disorder associated with mutations in the factor V gene. This hereditary coagulation disorder is clinically characterized by a heterogeneous spectrum of hemorrhagic manifestations ranging from mucosal or soft-tissue bleeds to potentially fatal hemorrhages. Current treatment of this condition consists in the administration of fresh frozen plasma and platelet concentrates. This article describes the cases of two patients with severe factor V deficiency, and of their parents. A high level of mutational heterogeneity of factor V gene was identified, nonsense mutations, frameshift mutations, missense changes, synonymous sequence variants and intronic changes. These findings prompted the identification of a new mutation in the human factor V gene, designated as Jaén-1, which is capable of altering the procoagulant function of factor V. In addition, an update is provided on the prospects for the treatment of factor V deficiency on the basis of yet-to-be-developed recombinant products or advanced gene and cell therapies that could potentially correct this hereditary disorder.


Assuntos
Análise Mutacional de DNA , Deficiência do Fator V/genética , Deficiência do Fator V/terapia , Fator V/genética , Adolescente , Coagulação Sanguínea , Transtornos Herdados da Coagulação Sanguínea/genética , Testes de Coagulação Sanguínea , Plaquetas/metabolismo , Pré-Escolar , Códon sem Sentido , DNA Complementar/metabolismo , Saúde da Família , Feminino , Mutação da Fase de Leitura , Humanos , Masculino , Paquistão , Proteínas Recombinantes/química , Análise de Sequência de DNA , Espanha
19.
Haemophilia ; 26(5): 826-833, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32700411

RESUMO

BACKGROUND: Factor V (FV) deficiency is a monogenic inherited coagulation disorder considered to be an ideal indication for gene therapy. To investigate the possibility of therapeutic application of genome editing, we generated induced pluripotent stem cells (iPSCs) from a FV-deficient patient and repaired the mutation of factor V gene (F5) using a clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated 9 (Cas9). METHODS: The patient's peripheral blood mononuclear cells were reprogrammed for iPSCs. The targeting vector was designed with homology arms against F5 containing the corrected sequence. Cas9 ribonucleoprotein (RNP) complex and targeting vector were electroporated into iPSCs. Gene-edited iPSCs were differentiated into hepatocyte-like cells (HLCs). RESULTS: The mutation of F5 in patient-derived iPSCs was repaired by CRISPR/Cas9. In concentrated culture supernatants of patient-derived iPS-HLCs, neither FV antigen nor activity was detected, while in those of gene-corrected iPS-HLCs, FV antigen and specific activity were 67.0 ± 13.1 ng/mL and 173.2 ± 41.1 U/mg, respectively. CONCLUSIONS: We successfully repaired the mutation of F5 using the CRISPR/Cas9 and confirmed the recovery of FV activity with gene-corrected iPS-HLCs. Gene-edited iPSCs are promising for elucidating the pathophysiology as well as for a modality of gene therapy.


Assuntos
Deficiência do Fator V/genética , Edição de Genes/métodos , Células-Tronco Pluripotentes Induzidas/metabolismo , Diferenciação Celular , Células Cultivadas , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Feminino , Humanos , Pessoa de Meia-Idade
20.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 37(5): 505-508, 2020 May 10.
Artigo em Zh | MEDLINE | ID: mdl-32335873

RESUMO

OBJECTIVE: To explore the genetic basis for a consanguineous pedigree affected with inherited coagulation factor V deficiency. METHODS: Genomic DNA was extracted from peripheral blood samples from the pedigree and subjected to next generation sequencing for screening variants of the F5 gene. Suspected pathogenic variant was verified by using Sanger sequencing. Pathogenicity of the variant was evaluated according to ACMG guidelines. RESULTS: A homozygous frameshifting variant, c.4096delC (p.Leu1366Phefs*3), was identified in the F5 gene in the proband, which was confirmed to be derived from her consanguineous parents. This variant was absent in all databases including 10 000 in-house Chinese exome sequences. Based on the ACMG guidelines, the c.4096delC was predicted to be a pathogenic variant. CONCLUSION: A novel pathogenic variant has been identified in the F5 gene in a consanguineous pedigree with inherited coagulation factor V deficiency, which has enriched the spectrum of F5 gene variants.


Assuntos
Deficiência do Fator V , Fator V , Variação Genética , Linhagem , Consanguinidade , Fator V/genética , Deficiência do Fator V/genética , Feminino , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA