Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Blood Cells Mol Dis ; 48(4): 254-9, 2012 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-22424603

RESUMO

Inherited antithrombin (AT) deficiency carries a 50% risk of venous thromboembolism (VTE) during pregnancy. Here, we investigated the molecular basis of type I AT deficiency in two women with recurrent VTE in the first trimester of pregnancy. Phenotype analysis showed both probands had almost 50% of normal AT levels. Two novel heterozygous AT mutations were identified: g.7920C>T resulting in a Trp225Cys mutation in case 1 and g.13863C>A causing an Ala404Asp mutation in case 2. Transient expression of either wild-type (WT) or mutant AT expression vectors in HEK293T and CHO cells showed impaired secretion of both AT mutant proteins. Immunofluorescence analysis revealed that the staining of AT-Trp225Cys in both endoplasmic reticulum (ER) and Golgi apparatus was similar to that of AT-WT, and the staining of AT-Ala404Asp was mainly present in ER but was weaker than that of AT-WT. These results revealed that the type I AT deficiency in two patients was caused by impaired secretion of the AT-Trp225Cys and AT-Ala404Asp mutant proteins, respectively. The two mutations are associated with a high risk of thrombotic onset and women with these AT mutations are prone to VTE in early pregnancy.


Assuntos
Deficiência de Antitrombina III/complicações , Deficiência de Antitrombina III/genética , Antitrombina III/genética , Complicações Hematológicas na Gravidez/genética , Tromboembolia Venosa/etiologia , Adulto , Deficiência de Antitrombina III/diagnóstico , Linhagem Celular , Feminino , Genótipo , Humanos , Mutação , Linhagem , Gravidez , Primeiro Trimestre da Gravidez , Fatores de Risco
2.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 29(5): 524-8, 2012 Oct.
Artigo em Zh | MEDLINE | ID: mdl-23042386

RESUMO

OBJECTIVE: To analyze the phenotype and genotype of two Chinese pedigrees with von Willebrand diseases, and to investigate the molecular pathogenesis. METHODS: Bleeding time (BT), activated partial thromboplastin time (APTT), ristocetin-induced platelet aggregation (RIPA), von Willebrand factor-ristocetin cofactor (vWF:Rco), von Willebrand factor antigen (vWF:Ag), von Willebrand factor activity (vWF:A), von Willebrand factor collagen binding assay (vWF:CB) and multimer analysis were used for phenotype diagnosis. DNA was extracted. All of the 52 exons and exon-intron bounda ries of the VWF gene were amplified with polymerase chain reaction(PCR) and analyzed by direct sequencing. RESULTS: APTT and BT were prolonged. Plasma RIPA, vWF:Rco, vWF:Ag, vWF:A and vWF:CB was significantly decreased. No VWF multimer can be found by plasma VWF multimer analysis. Homozygous insertional mutation g.82888_82889insCATG in exon 17 was found in proband A. Compound heterozygous mutations g.94865 G to A (Trp856stop) in exon 20 and g.110698_110699delinsG in exon 28 were found in proband B. CONCLUSION: Homozygous insertional mutation g.82888_82889insCATG and compound heterozygous mutations g.94865G to A(Trp856X) and g.110698_110699delinsG probably have respectively induced type 3 von Willebrand diseases in the two probands.


Assuntos
Doença de von Willebrand Tipo 3/genética , Adolescente , Feminino , Genótipo , Humanos , Masculino , Mutação , Linhagem , Fenótipo , Fator de von Willebrand/genética
3.
Zhonghua Nei Ke Za Zhi ; 51(10): 788-92, 2012 Oct.
Artigo em Zh | MEDLINE | ID: mdl-23290978

RESUMO

OBJECTIVE: To analyze the phenotype and genotype of three patients with von Willebrand disease (vWD), and to explore its molecular pathogenesis. METHODS: Bleeding time (BT), APTT, ristocetin induced platelet aggregation (RIPA), von Willebrand factor (vWF):ristocetin cofactor (Rco) (vWF:Rco), vWF antigen (vWF:Ag), vWF activity (vWF:A) test, vWF collagen binding assay (vWF:CB) and multimer analysis were detected for phenotype diagnosis. The dynamic process of blood coagulation was evaluated by using the thrombelastography. Genomic DNA was extracted from the peripheral blood. The vWF gene mutation was detected by sequencing. RESULTS: APTT, BT were prolonged in the three probands. Plasma vWF:Rco, vWF:Ag, vWF:A and vWF:CB were decreased in different degrees. RIPA was reduced in probands B and C. vWF multimer analysis found the lost of the large molecular weight multimers in proband B, while basically normal in probands A and C. The dynamic process of blood coagulation of proband C presented obvious hypocoagulability by using the thrombelastography. Heterozygous missense mutation g.106782G > T resulting in Cys1130Phe in exon 26, g.110988G > A resulting in Gly1579Arg in exon 28 and g.110373C > T resulting in Arg1374Cys in exon 28 were found in the probands A, B and C, respectively. CONCLUSION: Three probands were diagnosed as type 1, type 2A or type 2M vWD by phenotype detection. Heterozygous missense mutation Cys1130Phe, Gly1579Arg and Arg1374Cys induced vWD of three probands, respectively.


Assuntos
Mutação , Doenças de von Willebrand/diagnóstico , Doenças de von Willebrand/genética , Adulto , Feminino , Genótipo , Humanos , Masculino , Linhagem , Fenótipo , Adulto Jovem , Fator de von Willebrand
4.
Blood Cells Mol Dis ; 42(1): 44-50, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-18976939

RESUMO

BACKGROUND: Glanzmann thrombasthenia (GT) is an autosomal recessive bleeding disorder characterized by lack of platelet aggregation in response to most physiological agonists and caused by either a lack or dysfunction of the platelet integrin alphaIIbbeta3 (glycoprotein IIb/IIIa). PATIENTS: Mucocutaneous bleeding manifestations and platelet dysfunction consistent with GT were observed in a 20-year-old proband of a Chinese family. OBJECTIVES: To determine the molecular basis of GT and characterize the mutation by in vitro expression studies. RESULTS: Analysis of the patient's platelets by fluorescence-activated cell sorting demonstrated the presence of trace amounts of beta3, exposed on her platelet surface, but a complete absence of alphaIIbbeta3. Sequence analysis revealed a novel C470A transversion in exon 4 of the alphaIIb gene predicting a Pro126His alteration in the blade 2 of the alphaIIb beta propeller domain. The proband was homozygous for the mutation, the mother and the father were heterozygous, whereas 100 healthy subjects lacked this transversion. Chinese hamster ovary cells cotransfected with cDNAs of mutated alphaIIb and wild-type beta3 failed to express alphaIIbbeta3 on the cell surface as shown by FACS. Western blot analysis of the cell lysates showed no detectable mature alphaIIb. Immunoprecipitation with antibody against beta3 demonstrated pro-alphaIIb in the cells expressing the mutant alphaIIbbeta3, indicating pro-alphaIIbbeta3 complex formation. Intracellular immunofluorescence studies demonstrated the pro-alphaIIbbeta3 complex that co-localized with an ER marker, but showed minimal co-localization with a Golgi marker. CONCLUSIONS: A novel Pro126His mutation in alphaIIb compromised transport of the pro-alphaIIbbeta3 complex from the endoplasmic reticulum to the Golgi, leading to intracellular retention. The impaired alphaIIbbeta3 transport is responsible for the thrombasthenia in this patient.


Assuntos
Plaquetas/metabolismo , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Glicoproteína IIb da Membrana de Plaquetas/genética , Trombastenia/genética , Trombastenia/metabolismo , Animais , Células CHO , Cricetinae , Cricetulus , Retículo Endoplasmático/genética , Retículo Endoplasmático/metabolismo , Éxons/genética , Família , Feminino , Complexo de Golgi/genética , Complexo de Golgi/metabolismo , Heterozigoto , Homozigoto , Humanos , Masculino , Mutação de Sentido Incorreto/genética , Glicoproteína IIb da Membrana de Plaquetas/metabolismo , Transporte Proteico/genética , Transporte Proteico/fisiologia , Trombastenia/etiologia
5.
Zhonghua Yi Xue Za Zhi ; 88(26): 1832-6, 2008 Jul 08.
Artigo em Zh | MEDLINE | ID: mdl-19040019

RESUMO

OBJECTIVE: To explore the molecular mechanisms of Glanzmann thrombasthenia caused by alpha IIb P126H mutation. METHODS: Eukaryotic vector of alpha IIb P126H was constructed by PCR site-directed mutagenesis and then co-transfected with eukaryotic vector PCDM8 II a expressing the subunit beta3 into human renal epithelial cells of the line 293& and Chinese hamster ovarian cancer cells of the line CHO after sequencing. The membrane expression of alpha IIb P126H mutant was analyzed by flow cytometry and the whole expression was confirmed by Western blotting. The alpha IIb P126H mutant subcellular localization was determined by laser confocal scanning microscopy. RESULTS: The 293T cells cotransfected with cDNAs of mutated alpha IIb and wild-type beta3 failed to express alpha II bbeta3 on the cell surface as shown by FACS. Western blotting of the cell lysate showed no detectable mature alpha IIb. Immunofluorescence studies demonstrated proa II bbeta3 complex colocalized with an endoplasmic reticulum (ER) marker, but showed minimal colocalization with an Golgi marker. CONCLUSION: The P126H mutation in alpha IIb prevents transport of the pro-alpha II bbeta3 complex from ER to the Golgi body, thus hindering its maturation and surface expression. The impaired alpha II bp33 transport is responsible for the thrombasthenia.


Assuntos
Mutação , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/genética , Trombastenia/genética , Animais , Sequência de Bases , Western Blotting , Células CHO , Linhagem Celular , Cricetinae , Cricetulus , Retículo Endoplasmático/metabolismo , Citometria de Fluxo , Humanos , Microscopia Confocal , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Transporte Proteico , Trombastenia/metabolismo , Transfecção
6.
Chin Med J (Engl) ; 131(15): 1780-1785, 2018 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-29848837

RESUMO

BACKGROUND: Although much attention has been paid to the pharmacokinetics (PKs) of different factor VIII (FVIII) concentrates in persons with hemophilia A (HA), limited information is available in young boys with severe HA. In this study, we aimed to assess the PK parameters of FVIII products in boys with severe HA in China. METHODS: A total of 36 boys (plasma-derived [pd]-FVIII, n = 15; recombinant [r] FVIII, n = 21) were enrolled between January 2015 and May 2016 in Beijing Children's Hospital. PK characteristics of FVIII products were studied according to a reduced 4-sampling time point design (1 h, 9 h, 24 h, and 48 h postinfusion). RESULTS: The mean FVIII half-life (t1/2) was 10.99 ± 3.45 h (range 5.52-20.02 h), the mean in vivo recovery (IVR) was 2.01 ± 0.42 IU/dl per IU/kg (range 1.24-3.02 IU/dl per IU/kg) and mean clearance (CL) of FVIII is 4.34 ± 1.58 ml·kg-1·h-1 (range 2.29-7.90 ml·kg-1·h-1). We also analyzed the influence of several parameters that potentially modulate FVIII PK. The age was closely associated with FVIII half-life (R2 = 0.32, P < 0.01). The t1/2of FVIII increased by 0.59 h per year. Besides age, von Willebrand factor antigen (VWF:Ag) also was associated with FVIII half-life (R2 = 0.52, P < 0.01). Patients with blood Group O had a shorter FVIII half-life than patients with non-O blood group (9.40 ± 0.68 h vs. 12.3 ± 0.79 h, t = 2.70, P = 0.01). The FVIII IVR correlated with age (R2 = 0.21, P < 0.01) and VWF:Ag level (R2 = 0.28, P < 0.01). CL rates were faster in young patients and in those with low-VWF:Ag levels. CL rates of FVIII are higher in blood Group O versus non-blood Group O persons (5.02 ± 0.38 vs. 4.00 ± 0.32 ml·kg-1·h-1, t = 2.53, P = 0.02). CONCLUSIONS: Chinese boys with severe HA have similar PK values to other ethnic groups and large differences in FVIII PK between individual patients. Age, blood group, and VWF:Ag levels are important determining factors for FVIII CL.


Assuntos
Fator VIII/farmacocinética , Hemofilia A/tratamento farmacológico , Adolescente , Testes de Coagulação Sanguínea , Criança , Pré-Escolar , China , Humanos , Masculino , Fator de von Willebrand
7.
Zhonghua Yi Xue Za Zhi ; 87(14): 987-90, 2007 Apr 10.
Artigo em Zh | MEDLINE | ID: mdl-17650426

RESUMO

OBJECTIVE: To study the molecular mechanisms of antithrombin (AT) deficiency caused by AT gene mutations T98I and A404T. METHODS: Wild-type and mutant AT cDNA expression plasmids (ATwt, AT T98I and AT A404T) were constructed and transfected into the monkey fibroblast of the line COS-7 or Chinese hamster ovary (CHO) cells. NH4Cl. ALLN and brefeldin A were added. ELISA was used to detect the AT: Ag. Pulse-chase experiment and immunofluorescence assay were used to detect the radioactivity of the 35S-labeled AT. Fluorescence real-time PCR was used to detect the expression of AT mRNA, protein degradation inhibition was used to elucidate the mutant T98I degradation pathway inside the cells. RESULTS: AT T98I was not secreted from the cells and was gradually degraded inside the cells. There was partial secretion of AT A404T, but most of the molecule was not secreted but was degraded intracellularly. Fluorescence real-time PCR indicated that the mutant AT mRNA was transcribed at a similar or even higher level as that of wild-type (wt). Pulse-chase labeling studies suggested both AT variants did not accumulate, but degraded intracellularly. Protein degradation inhibition experiment showed that mutant AT T98I was degraded intracellularly through the proteasome pathway. Immunohistochemical staining of the transfected cells revealed that CHO cells expressing the AT T98I mutant were stained diffusely without perinuclear enhancement and cells expressing AT A404T mutant mainly in the whole cytoplasm with weaker perinuclear enhancement. CONCLUSION: Impaired secretion of the mutant AT molecules, due to intracellular degradation, is the molecular mechanism of AT deficiency caused by T98I and A404T mutation.


Assuntos
Deficiência de Antitrombina III/genética , Antitrombinas/genética , Mutação , Animais , Antitrombinas/metabolismo , Células CHO , Células COS , Chlorocebus aethiops , Cricetinae , Cricetulus , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção
8.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 24(6): 1850-1855, 2016 Dec.
Artigo em Zh | MEDLINE | ID: mdl-28024507

RESUMO

OBJECTIVE: To investigate the correlation of patients with thrombosis or prothrombotic status with hyperhomocysteinemia (HHcy), activated protein C-resistance(APCR) and gene polymorphism of coagulation factor V. METHODS: Three hundred healthy voluteers were selected as controls, 223 cases of thrombosis (80 cases of cerebral infarction of CT, the MI of 82 cases of myocardial infarction, venous thrombosis of VTE 61 cases), 270 cases of patients with prothrombotic state (76 cases of pregnancy disease of PIH, 62 cases of chronic obstructive pulmonary disease (COPD), 60 cases of diabetes(DM) and 72 cases of cancer) were enrolled in this study. The plasma APCR and hyperhomocysteinemia were detected by APTT coagulation method and cycling enzyme method respectively, and restriction fragment length polymorphism(RFLP) were was used to detect the gene polymorphism of FV G1691-A, G1091-C and A1090-G in the patient and control groups. RESULTS: APCR positive rate was 62.29% and 7.33%, and the positive hyperhomocysteinemia accounted for 68.42% and 10.00% respectively in the group of the patients with venous thrombosis and the normal control group. 3 cases of heterozygous FV gene mutations were found in the APCR-positive patients with venous thrombosis. CONCLUSION: HHcy possitive rate of patients with venous thrombosis is signiticantly higher than that in control, the HHcy is one of the important causes resulting in thrombosis, the patients with venous thrombosis have proved to be with APCR, and the possitive APCR may be related with the coagulation factor V gene polymorphism.


Assuntos
Polimorfismo Genético , Resistência à Proteína C Ativada , Fator V , Feminino , Humanos , Mutação , Gravidez , Trombose , Trombose Venosa
9.
Thromb Haemost ; 94(6): 1172-6, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16411389

RESUMO

We investigated the molecular mechanisms responsible for type I congenital antithrombin (AT) deficiency in two unrelated Chinese pedigrees manifesting multiple site venous thrombosis. Phenotype analysis showed both probands had almost 50% of normal AT levels. Direct sequencing of amplified DNA revealed 2757C > T in proband 1 and 13328G > A in proband 2, predicting a heterozygous Thr98Ile (T981) and Ala404Thr (A404T), respectively. No proband had 20210A allele or factor V Leiden mutation. Transient expression of complementary DNA coding for the mutations in COS-7 cells showed impaired secretion of the mutant molecules. Real-time quantitative PCR indicated that the mutant AT mRNA was transcribed at a similar or even higher level as that of wild-type (wt). Pulse-chase labeling studies suggested both AT variants did not accumulate, but degraded intracellularly. Immunohistochemical staining of the transfected cells revealed that CHO cells expressing the AT-198 mutant were stained diffusely without perinuclear enhancement and cells expressing AT-T404 mutant mainly in the whole cytoplasm with weaker perinuclear enhancement. We conclude that the impaired secretion of the mutant AT molecules, due to intracellular degradation, is the molecular pathogenesis of AT deficiency caused by T981 and A404T mutation for the two families, respectively.


Assuntos
Antitrombinas/genética , Transtornos Herdados da Coagulação Sanguínea/genética , RNA Mensageiro/metabolismo , Trombose Venosa/genética , Animais , Antitrombinas/metabolismo , Transtornos Herdados da Coagulação Sanguínea/sangue , Transtornos Herdados da Coagulação Sanguínea/complicações , Células CHO/metabolismo , Células COS/metabolismo , China , Chlorocebus aethiops , Cricetinae , Cricetulus , Genótipo , Heterozigoto , Humanos , Linhagem , Fenótipo , Mutação Puntual , Transfecção , Trombose Venosa/sangue , Trombose Venosa/etiologia
10.
Haematologica ; 90(12): 1659-64, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16330440

RESUMO

BACKGROUND AND OBJECTIVES: Most secreted proteins, including coagulation factor X (FX), are synthesized with a signal peptide, which is necessary for targeting the nascent polypeptide into the endoplasmic reticulum. Characterization of naturally occurring mutations may provide insights into the functional roles of the amino acids in the signal peptide. DESIGN AND METHODS: A 52-year old male patient with type I FX deficiency was studied. Mutations were searched for by FX gene (F10) sequencing. The wild-type and the mutant FX proteins were expressed in transfected cells and then immunological assays were performed. Pulse-chase experiments and cell-free expression studies were conducted to determine the cellular fate of the mutant FX molecules. RESULTS: The patient we studied was homozygous for a substitution of arginine for serine at codon -30 in the signal sequence of F10. Immunoassays detected low FX antigen levels in both the conditioned media and lysates of the cells expressing the mutant protein. Pulse-chase analysis showed that only trace amounts of the mutant FX protein were detectable in the conditioned media, and that the mutant molecules did not accumulate inside the cells either. The results of cell-free expression studies showed that although the transcription and translation of the mutant construct were normal, no post-translational processing, such as N-linked glycosylation, occurred in the presence of microsomes. INTERPRETATION AND CONCLUSIONS: These findings suggest that substitution of a neutral polar amino acid, serine by arginine, in the hydrophobic core of FX signal peptide severely impairs the ability of the protein to enter the endoplasmic reticulum and results in FX deficiency.


Assuntos
Retículo Endoplasmático/metabolismo , Deficiência do Fator X/genética , Fator X/genética , Mutação de Sentido Incorreto , Transporte Proteico , Substituição de Aminoácidos , Sistema Livre de Células , Células Cultivadas , Consanguinidade , Fator X/química , Fator X/metabolismo , Deficiência do Fator X/metabolismo , Hemorragia Gastrointestinal/etiologia , Humanos , Interações Hidrofóbicas e Hidrofílicas , Masculino , Pessoa de Meia-Idade , Sinais Direcionadores de Proteínas , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/biossíntese , Relação Estrutura-Atividade , Transfecção
11.
Blood Coagul Fibrinolysis ; 16(2): 149-55, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15741804

RESUMO

As a major physiological inhibitor of thrombin and other coagulation proteases, antithrombin (AT) plays an important role in the maintenance of normal hemostasis and its deficiency is associated with a predisposition for familial venous thromboembolic disease. Recently, we found a novel mutation (13387-9delG) in the antithrombin gene that is associated with type I AT deficiency. To examine the molecular pathologic mechanism of this mutation causing type I AT deficiency, the wild-type and the mutant AT constructs were expressed in COS-7 cells or Chinese Hamster Ovary cells. No AT antigen could be detected by enzyme-linked immunosorbent assay in the conditioned media of cells expressing the mutant protein, and the AT antigen level was reduced in cell lysates. The mutant AT-expressing cells did not have less intracellular mRNA levels than the wild-type transfectants as estimated by quantitative reverse transcriptase-polymerase chain reaction. Metabolic and pulse-chase experiments showed the newly synthesized wild-type AT protein was gradually secreted into the media, whereas no labeled mutant AT protein was detected in the media and the total amount of radioactivity was significantly reduced in the cells during the chase periods. By immunofluorescence analysis, the staining of the mutant AT was weaker than that of the wild type, and was predominantly diffuse without perinuclear enhancement. These results indicate that the 13387-9delG mutation, which disrupts the disulfide bridge Cys247-Cys430, impairs the secretion and stability of the truncated AT protein associated with intracellular degradation.


Assuntos
Deficiência de Antitrombina III/metabolismo , Antitrombina III/biossíntese , Mutação , Adolescente , Animais , Antitrombina III/genética , Deficiência de Antitrombina III/genética , Células CHO , Células COS , Chlorocebus aethiops , Cricetinae , Expressão Gênica , Humanos , Masculino
12.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 20(4): 279-83, 2003 Aug.
Artigo em Zh | MEDLINE | ID: mdl-12903033

RESUMO

OBJECTIVE: To investigate the genotypes of mutations of an inherited coagulation factor VII(F VII) deficiency pedigree. METHODS: The diagnosis was validated by coagulant parameters. F VII gene mutations were analysed in the proband and her family members by DNA direct sequencing. The PCR fragments were cleaved by the Msp I restriction enzyme to confirm the mutations detected by sequencing was performed in this study. RESULTS: Double heterozygous mutations at the same coding site of amino acid were detected in propositus of the pedigree: a C to T mutation at position 11348 resulting in Arg304Trp substitution combined with a G to A mutation at position 11349 resulting in Arg304Gln substitution. Her farther had a G to A mutation at position 11349 and her mother had a C to T mutation at position 11348, respectively. Both were heterozygous mutations. One of her brothers had normal genotype, the other brother and all her three offsprings had heterozygous mutations. CONCLUSION: Double heterozygous mutations coding the same amino acid were found in a pedigree with hereditary coagulation factor VII deficiency.


Assuntos
Deficiência do Fator VII/genética , Fator VII/genética , Mutação , Análise Mutacional de DNA , Feminino , Heterozigoto , Humanos , Masculino , Linhagem
13.
Zhonghua Nei Ke Za Zhi ; 42(10): 692-6, 2003 Oct.
Artigo em Zh | MEDLINE | ID: mdl-14633462

RESUMO

OBJECTIVE: To identify the genetic mutations of a severe inherited coagulation factor VII (FVII) deficiency pedigree. METHODS: The diagnosis was validated by coagulant and haemostatic parameters. FVII gene mutations were screened in the propositus and his family members by DNA direct sequencing and confirmed by digestions of the restriction enzymes of the PCR production. RESULTS: Two heterozygous missense mutations were found in the propositus of the pedigree: a G to T transversion at position 9482 in exon 6 and a C to T mutation at position 11348 in exon 8 resulting in the amino acid substitution of Arg152 with Leu and Arg304 with Trp, respectively. A heterozygous single nucleotide deletion (C) at position 11487-11489(CCC) within exon 8 was identified, which predicted the frameshift mutation at position His351 followed by the changes of six corresponding amino acids and appearance of a premature protein caused by stop codon. The heterozygous mutations identified in the proband were derived from his father (Arg152 to Leu) and his mother (Arg304 to Trp mutation) and a heterozygous deletion (C) at position 11487-9(CCC). By tracing the other pedigree members, it was found that his grandmother had a heterozygous mutation of Arg304Trp and a heterozygous polymorphism of Arg353Gln and his grandfather had a heterozygous Arg152Leu mutation. CONCLUSION: Three heterozygous mutations were found in a pedigree with hereditary coagulation factor VII deficiency. Arg152Leu and deletion C at position 11487-9(CCC) were novel mutations.


Assuntos
Transtornos da Coagulação Sanguínea/genética , Deficiência do Fator VII/genética , Fator VII/genética , Adulto , Idoso , Testes de Coagulação Sanguínea , Feminino , Mutação da Fase de Leitura , Deleção de Genes , Heterozigoto , Humanos , Masculino , Pessoa de Meia-Idade , Linhagem , Mutação Puntual
14.
Zhonghua Yi Xue Za Zhi ; 83(4): 312-5, 2003 Feb 25.
Artigo em Zh | MEDLINE | ID: mdl-12812650

RESUMO

OBJECTIVE: To discover the gene mutations of a pedigree with inherited factor V (FV) deficiency. METHODS: The activated partial thromboplastin time (APTT), prothrombin time (PT), FV activity (FV:C) and FV antigen test were adopted for phenotype diagnosis. The genomic DNA was extracted from the peripheral blood of the 16-year-old propositus, female. All the 25 exons and their flanks in the FV gene of the propositus were amplified by polymerase chain reaction (PCR). The PCR products were screened by direct sequencing and the mutations were further confirmed by restricted enzyme digestion. Six persons in the pedigree (grandfather, grandmother, father, mother, uncle, and aunt) were examined too. 108 healthy blood donors were used as controls. RESULTS: The APTT, PT, FV:C, and FV:Ag of the propositus were 126.6s, 42.8s, 0.3% and 1.3% respectively. The Fbg and FII, FVII, FVIII, FIX, FX activities were in normal range. FV:C of the members of the pedigree was 36% - 70%, and the FV:Ag of the pedigree members was 26.4% - 45.3% that of the mixture of 30 normal plasma samples. Taking the GeneBank Z99572 sequence as the reference, totally five variations in the FV gene were found in the propositus. The mutations, A1348G and 4887 approximately 8delG, were traced to her father and her mother respectively. No 1348G-->T mutation was found in the 108 controls. CONCLUSION: The FV deficiency of the propositus is caused by missense mutation of G1348T and frameshift mutation of 4887 approximately 8delG, which haven't been identified previously.


Assuntos
Deficiência do Fator V/congênito , Deficiência do Fator V/genética , Fator V/genética , Mutação , Adolescente , Sequência de Bases , Feminino , Humanos , Masculino , Dados de Sequência Molecular , Linhagem
15.
Zhonghua Yi Xue Za Zhi ; 83(23): 2054-7, 2003 Dec 10.
Artigo em Zh | MEDLINE | ID: mdl-14703415

RESUMO

OBJECTIVE: To identify the mutations of fibrinogen genes in a Chinese family with inherited afibrinogenemia. METHODS: Samples of peripheral blood were collected from 17 members of 3 generations in a Chinese family with inherited afibrinogenemia, including the proband, female, aged 8. All the exons and exon-intron boundaries of the three fibrinogen genes were analyzed by direct sequencing. RESULTS: The sequencing results of the proband revealed compound 2 heterozygous mutations in fibrinogen FGA gene, one being a splice mutation (g.1892-1899delAGTAorGTAA) in the boundary between exon3 and intron3 of the FGA gene and traced back to her patriline and the other being a 1,238 bp large deletion (g.1978-3215) in the same gene and originating from her matriline. CONCLUSION: Inherited afibrinogenemia is caused by the compound heterozygous deletion in the fibrinogen FGA gene.


Assuntos
Fibrinogênio/genética , Criança , Feminino , Fibrinogênio/análise , Deleção de Genes , Humanos , Mutação
16.
Zhonghua Yi Xue Za Zhi ; 83(19): 1694-7, 2003 Oct 10.
Artigo em Zh | MEDLINE | ID: mdl-14642106

RESUMO

OBJECTIVE: To identify the gene mutation in a Chinese pedigree of type I hereditary protein C deficiency. METHODS: The plasma levels of protein C activity (PC:A), protein C antigen (PC:Ag), protein S activity, and anti-thrombin activity (AT:A) of the propositus, male, aged 7, and 11 members of the pedigree were detected using ELISA and chromogenic assay respectively. All of the nine exons and intron-exon boundaries of protein C gene of the propositus were analyzed by direct sequencing of the corresponding amplified PCR products in DNA from the propositus. Restriction enzyme site analysis was used to confirm the mutation. RESULTS: The plasma concentrations of protein C activity and antigen of the propositus were 26% and 1.43 g/L respectively. The PC:Ag and PC:A of his father were normal. Decreased PC:A level was seen in his mother and 4 of his maternal pedigree. PS:A and AT:A were all normal in all of the pedigree members. A C5498T heterozygous mutation in exon 3, resulting in the substitution of Arg for Trp at the 15th amino acid, was identified in the propositus and 8 of his relatives. This mutation was confirmed by restriction enzyme site analysis. Mutations C/T at position 2405, A/G at position 2418, and A/T at position 2583 in the protein C promoter region were confirmed in the propositus and all members of the pedigree. CONCLUSION: C5498T heterozygous mutation in exon 3 of protein C gene, first reported in China, leads to type I hereditary.


Assuntos
Mutação , Deficiência de Proteína C/genética , Proteína C/genética , Criança , Feminino , Humanos , Masculino , Linhagem , Proteína C/análise
17.
Zhonghua Yi Xue Za Zhi ; 83(24): 2158-61, 2003 Dec 25.
Artigo em Zh | MEDLINE | ID: mdl-14720426

RESUMO

OBJECTIVE: To investigate the mechanisms of two novel missense mutations of factor XIIIA subunit gene (Arg77-->Cys,Ser413-->Trp) in the pathogenesis of hereditary factor XIII deficiency. METHODS: Site-directed mutagenesis was conducted to obtain 2 mutant human XIII A recombinant plasmids, mut-PCI/FXIIIA. Normal wild type factor XIII A recombinant plasmid, wt-PCI/FXIIIA, and mut-PCI/FXIIIA, were transfected into cultured COS7 cells line, renal fibroid cell of African green monkey using Superfect reagent respectively, The expression levels of DNA, RNA and protein of human factor XIII, both wild type and mutant, were detected by PCR, RT-PCR and Western blotting. Pulse-chase experiment was used to look into the changing of factor XIII A in the cytoplasm. Factor XIIIA activity was assayed by Biotin-pentylamine incorporation technique. RESULTS: The mRNA levels of the two mutants in transfected cells were similar to that of the wild type factor XIIIA. But the amount of mutant factor XIIIA protein and its activity in cells decreased markedly, even disappeared. Pulse-chase experiment revealed that at the two mutants existed chase time 0.5 h and 1 h considerable amounts in cells and then disappeared rapidly later. CONCLUSION: The 2 mutations of the factor XIIIA cause the instability, degradation, and rapid disappearance of FXIIIA in cytoplasm, thus resulting in hereditary factor XIII deficiency.


Assuntos
Deficiência do Fator XIII/genética , Fator XIII/genética , Mutação , Animais , Células COS , Fator XIII/química , Humanos , RNA Mensageiro/análise
18.
Pathology ; 46(7): 630-5, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25393254

RESUMO

We report two compound heterozygous mutants that caused severe type I protein C (PC) deficiency in two independent Chinese families.PC antigen was determined by enzyme-linked immunosorbent assay (ELISA), and PC activity was measured by chromogenic assay. Genetic mutations were screened with polymerase chain reaction (PCR) followed by direct sequencing. PC mutants were transiently expressed in COS-7 cells for the evaluation of PC secretory activity and function. The subcellular location was visualised by immunofluorescence assay. The structural analysis of mutation was performed as well.Compound heterozygous mutations of Arg178Trp and Asp255His with reduced PC activity and antigen levels were identified in Proband 1, a 28-year-old male with deep vein thrombosis (DVT) and pulmonary embolism. The other mutations of Leu-34Pro and Thr295Ile with reduced PC activity and antigen levels were identified in Proband 2, a 19-year-old male with DVT. The PC activities with Arg178Trp, Asp255His, Leu-34Pro and Thr295Ile mutations decreased significantly. Immunofluorescence assay demonstrated that only trace amount of PC with novel Thr295Ile mutation was transported to the Golgi apparatus. Subsequent structural analysis indicated severe impairments of intracellular folding and secretion.The two rare compound heterozygous mutations could cause type I PC deficiency via impairment of secretory activity of PC.


Assuntos
Povo Asiático/genética , Heterozigoto , Mutação/genética , Deficiência de Proteína C/genética , Adulto , Animais , Células COS , Linhagem Celular , Ensaio de Imunoadsorção Enzimática/métodos , Humanos , Masculino , Linhagem , Embolia Pulmonar/genética , Trombose Venosa/genética , Adulto Jovem
19.
Zhonghua Xue Ye Xue Za Zhi ; 34(8): 691-5, 2013 Aug.
Artigo em Zh | MEDLINE | ID: mdl-23978021

RESUMO

OBJECTIVE: To investigate the binding mechanisms of FVIII Trp1707Ser mutation-associated inhibitor. METHODS: The APPT, PT, TT, Fg and FVIII:C were detected to make phenotypic diagnosis of haemophilia A. Inhibitors titer were measured by Bethesda method. Long distance-PCR (LD-PCR) and sequence-specific PCR were adopted for screening the intron 22 and intron 1 inversions respectively. FVIII coding and boundary sequences were analyzed by direct DNA sequencing. Inhibitor was reacted with different segments of FVIII, including heavy chain and its components A1 and A2, light chain and its components A3, C1 and C2. Corrected test was used to measure the remaining F VIII:C (% ) by adding pooled normal plasmas. After labeling purified inhibitors with biotin, western blot was performed to further confirm the binding reactions between inhibitors and segments. RESULTS: The haemophilia A patient had mild deficiency of FVIII:C (1.1%) and had high FVIII inhibitor titer of 18.4 BU. A mutation c.97223C>G in exon 14 of F8 gene resulted to p.Trp1707Ser was identified by DNA sequencing. Corrected test showed that the remaining F VIII:C was increased when inhibitors reacted with heavy chain and light chain, especially with heavy chain. The remaining FVIII:C was also increased in the A2 and C2 domain reactions. No significant differences were seen in the A1, A3 and C1 domain reactions. Antigen-antibody reaction bands were confirmed by western blots when degenerated B-domain deleted recombinant FVIII, A2 and C2 were used as antigens. CONCLUSION: The binding sites of FVIIITrp1707Ser mutation inhibitor were the A2 domain of heavy chain and C2 domain of light chain. The binding reaction with heavy chain was more intense.


Assuntos
Fator VIII/genética , Hemofilia A/genética , Mutação , Sítios de Ligação/genética , Éxons , Fator VIII/antagonistas & inibidores , Humanos , Masculino , Adulto Jovem
20.
Zhonghua Xue Ye Xue Za Zhi ; 34(3): 190-4, 2013 Mar.
Artigo em Zh | MEDLINE | ID: mdl-23683413

RESUMO

OBJECTIVE: To investigate the function of abnormal fibrinogen in two inherited dysfibrinogenemia pedigrees. METHODS: Routine coagulation tests were conducted in the probands and related family members. The antigen and activity levels of fibrinogen were detected by immunoturbidimetry assay and clauss assay, respectively. All the exons and exon-intron boundaries of the three fibrinogen genes and antithrombin gene(AT3)were analyzed by PCR amplification and direct sequencing. Routine thrombelastography (TEG) test and functional fibrinogen TEG test were both used to make a comprehensive evaluation of coagulation status and functional fibrinogen level in patients. The molecular weights of the three peptides from fibrinogen were measured by Western blot. The function of abnormal fibrinogen was assessed by fibrinogen dynamic polymerization and fibrinolysis velocity. RESULTS: The coagulation routine tests were normal in two probands except for prolonged thrombin time (TT) and reptilase time (RT), as well as reduced activity levels of 0.5 g/L and 0.6 g/L fibrinogen, respectively. The antigen levels of fibrinogen were 2.32 g/L and 2.66 g/L in two probands, which were in the normal reference range. The genotype analysis showed that Arg275His in fibrinogen γ chain (γ Arg275His) existed in both probands and patients in these two pedigrees. Meanwhile, proband B's grandfather and aunt also carried heterozygote g.5876T>C (Ser116Pro) mutation in AT3. The results of routine TEG test demonstrated that the α values of proband B and his father were close to and lower than the lower limit of reference range, respectively, while the MA values were normal in both of them. However, functional fibrinogen TEG test revealed obviously reduced MA value. All the probands and patients demonstrated prolonged lag-off time and reduced peak value in fibrinogen dynamic polymerization tests. Meanwhile, most of fibrin formed from the patients' plasma could not be dissolved completely by plasminogen (PLG) and urokinase-typeplasminogenactivator (u-PA) at a certain time. CONCLUSION: We first reported cases of inherited dysgibrinogenemia associated with inherited AT deficiency. γArg275His mutation caused the abnormal fibrinogen in terms of fibrin mono polymerization and possibly in fibrinolysis. Combined use of routine TEG test and functional fibrinogen TEG test with comprehensive analyses of the parameters in both tests could better evaluate the level of functional fibrinogen and predict the risk of hemorrhage and thrombosis in patients with inherited dysfibrinogenemia.


Assuntos
Afibrinogenemia/genética , Fibrinogênio/genética , Fibrinogênios Anormais/fisiologia , Mutação , Adulto , Criança , Feminino , Fibrinogênios Anormais/genética , Genótipo , Humanos , Masculino , Linhagem , Fenótipo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA