Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Blood ; 124(11): 1799-807, 2014 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-25051961

RESUMO

Rare missense mutations in the von Willebrand factor (VWF) A3 domain that disrupt collagen binding have been found in patients with a mild bleeding phenotype. However, the analysis of these aberrant VWF-collagen interactions has been limited. Here, we have developed mouse models of collagen-binding mutants and analyzed the function of the A3 domain using comprehensive in vitro and in vivo approaches. Five loss-of-function (p.S1731T, p.W1745C, p.S1783A, p.H1786D, A3 deletion) and 1 gain-of-function (p.L1757A) variants were generated in the mouse VWF complementary DNA. The results of these various assays were consistent, although the magnitude of the effects were different: the gain-of-function (p.L1757A) variant showed consistent enhanced collagen binding whereas the loss-of-function mutants showed variable degrees of functional deficit. We further analyzed the impact of direct platelet-collagen binding by blocking glycoprotein VI (GPVI) and integrin α2ß1 in our ferric chloride murine thrombosis model. The inhibition of GPVI demonstrated a comparable functional defect in thrombosis formation to the VWF(-/-) mice whereas α2ß1 inhibition demonstrated a milder bleeding phenotype. Furthermore, a delayed and markedly reduced thrombogenic response was still evident in VWF(-/-), GPVI, and α2ß1 blocked animals, suggesting that alternative primary hemostatic mechanisms can partially rescue the bleeding phenotype associated with these defects.


Assuntos
Colágeno/metabolismo , Integrina alfa2beta1/metabolismo , Glicoproteínas da Membrana de Plaquetas/metabolismo , Trombose/metabolismo , Fator de von Willebrand/metabolismo , Substituição de Aminoácidos , Animais , Cloretos/efeitos adversos , Cloretos/farmacologia , Colágeno/genética , Modelos Animais de Doenças , Compostos Férricos/efeitos adversos , Compostos Férricos/farmacologia , Células HEK293 , Humanos , Integrina alfa2beta1/genética , Camundongos , Camundongos Knockout , Mutação de Sentido Incorreto , Noxas/efeitos adversos , Noxas/farmacologia , Glicoproteínas da Membrana de Plaquetas/genética , Estrutura Terciária de Proteína , Trombose/induzido quimicamente , Trombose/genética , Trombose/patologia , Fator de von Willebrand/genética
2.
Blood ; 123(26): 4045-53, 2014 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-24829206

RESUMO

Ex vivo gene therapy strategies avoid systemic delivery of viruses thereby mitigating the risk of vector-associated immunogenicity. Previously, we delivered autologous factor VIII (FVIII)-expressing blood outgrowth endothelial cells (BOECs) to hemophilia A mice and showed that these cells remained sequestered within the implanted matrix and provided therapeutic levels of FVIII. Prior to translating this strategy into the canine (c) model of hemophilia A, we increased cFVIII transgene expression by at least 100-fold with the use of the elongation factor 1 alpha (EF1α) promoter and a strong endothelial enhancer element. BOECs isolated from hemophilia A dogs transduced with this lentiviral vector express levels of cFVIII ranging between 1.0 and 1.5 U/mL per 10(6) cells over 24 hours. Autologous BOECs have been implanted into the omentum of 2 normal and 3 hemophilia A dogs. These implanted cells formed new vessels in the omentum. All 3 hemophilia A dogs treated with FVIII-expressing autologous BOECs developed anti-FVIII immunoglobulin G2 antibodies, but in only 2 of the dogs were these antibodies inhibitory. FVIII antigen levels >40% in the absence of FVIII coagulant function were detected in the circulation for up to a year after a single gene therapy treatment, indicating prolonged cellular viability and synthesis of FVIII.


Assuntos
Autoanticorpos , Inibidores dos Fatores de Coagulação Sanguínea , Células Endoteliais , Fator VIII , Expressão Gênica , Terapia Genética/métodos , Hemofilia A , Animais , Autoanticorpos/sangue , Autoanticorpos/imunologia , Autoenxertos , Inibidores dos Fatores de Coagulação Sanguínea/sangue , Inibidores dos Fatores de Coagulação Sanguínea/imunologia , Modelos Animais de Doenças , Cães , Células Endoteliais/metabolismo , Células Endoteliais/transplante , Fator VIII/biossíntese , Fator VIII/genética , Fator VIII/imunologia , Vetores Genéticos , Hemofilia A/sangue , Hemofilia A/genética , Hemofilia A/imunologia , Hemofilia A/terapia , Humanos , Lentivirus , Camundongos , Fator 1 de Elongação de Peptídeos/genética , Regiões Promotoras Genéticas , Transdução Genética , Transgenes
3.
Blood ; 117(16): 4358-66, 2011 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-21346256

RESUMO

Type 1 VWD is the mild to moderate reduction of VWF levels. This study examined the mechanisms underlying 2 common type 1 VWD mutations, the severe R1205H and more moderate Y1584C. In vitro biosynthesis was reduced for both mutations in human and mouse VWF, with the effect being more severe in R1205H. VWF knockout mice received hydrodynamic injections of mouse Vwf cDNA. Lower VWF antigen levels were demonstrated in both homozygous and heterozygous forms for both type 1 mutations from days 14-42. Recombinant protein infusions and hydrodynamic-expressed VWF propeptide to antigen ratios demonstrate that R1205H mouse VWF has an increased clearance rate, while Y1584C is normal. Recombinant ADAMTS13 digestions of Y1584C demonstrated enhanced cleavage of both human and mouse VWF115 substrates. Hydrodynamic-expressed VWF shows a loss of high molecular weight multimers for Y1584C compared with wild-type and R1205H. At normal physiologic levels of VWF, Y1584C showed reduced thrombus formation in a ferric chloride injury model while R1205H demonstrated similar thrombogenic activity to wild-type VWF. This study has elucidated several novel mechanisms for these mutations and highlights that the type 1 VWD phenotype can be recapitulated in the VWF knockout hydrodynamic injection model.


Assuntos
Mutação , Doenças de von Willebrand/genética , Fator de von Willebrand/genética , Proteínas ADAM/metabolismo , Proteína ADAMTS13 , Animais , Linhagem Celular , Modelos Animais de Doenças , Humanos , Metaloendopeptidases/metabolismo , Camundongos , Camundongos Knockout , Fenótipo , Multimerização Proteica , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Trombose/genética , Fator de von Willebrand/administração & dosagem , Fator de von Willebrand/metabolismo
4.
Blood ; 115(23): 4862-9, 2010 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-20371742

RESUMO

Type 2B von Willebrand disease (2B VWD) results from von Willebrand factor (VWF) A1 mutations that enhance VWF-GPIbalpha binding. These "gain of function" mutations lead to an increased affinity of the mutant VWF for platelets and the binding of mutant high-molecular-weight VWF multimers to platelets in vivo, resulting in an increase in clearance of both platelets and VWF. Three common 2B VWD mutations (R1306W, V1316M, and R1341Q) were independently introduced into the mouse Vwf cDNA sequence and the expression vectors delivered to 8- to 10-week-old C57Bl6 VWF(-/-) mice, using hydrodynamic injection. The resultant phenotype was examined, and a ferric chloride-induced injury model was used to examine the thrombogenic effect of the 2B VWD variants in mice. Reconstitution of only the plasma component of VWF resulted in the generation of the 2B VWD phenotype in mice. Variable thrombocytopenia was observed in mice expressing 2B VWF, mimicking the severity seen in 2B VWD patients: mice expressing the V1316M mutation showed the most severe thrombocytopenia. Ferric chloride-induced injury to cremaster arterioles showed a marked reduction in thrombus development and platelet adhesion in the presence of circulating 2B VWF. These defects were only partially rescued by normal platelet transfusions, thus emphasizing the key role of the abnormal plasma VWF environment in 2B VWD.


Assuntos
Plaquetas/metabolismo , Mutação de Sentido Incorreto , Adesividade Plaquetária , Doença de von Willebrand Tipo 2/metabolismo , Fator de von Willebrand/metabolismo , Substituição de Aminoácidos , Animais , Cloretos/toxicidade , Modelos Animais de Doenças , Compostos Férricos/toxicidade , Humanos , Integrina alfa2/genética , Integrina alfa2/metabolismo , Camundongos , Camundongos Knockout , Noxas/toxicidade , Trombocitopenia/genética , Trombocitopenia/metabolismo , Trombose/induzido quimicamente , Trombose/genética , Trombose/metabolismo , Trombose/terapia , Doença de von Willebrand Tipo 2/genética , Doença de von Willebrand Tipo 2/terapia , Fator de von Willebrand/genética
5.
Mol Ther ; 19(4): 723-30, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21285959

RESUMO

The objective to use gene therapy to provide sustained, therapeutic levels of factor VIII (FVIII) for hemophilia A is compromised by the emergence of inhibitory antibodies that prevent FVIII from performing its essential function as a cofactor for factor IX (FIX). FVIII appears to be more immunogenic than FIX and an immune response is associated more frequently with FVIII than FIX gene therapy strategies. We have evaluated a modified lentiviral delivery strategy that facilitates liver-restricted transgene expression and prevents off-target expression in hematopoietic cells by incorporating microRNA (miRNA) target sequences. In contrast to outcomes using this strategy to deliver FIX, this modified delivery strategy was in and of itself insufficient to prevent an anti-FVIII immune response in treated hemophilia A mice. However, pseudotyping the lentivirus with the GP64 envelope glycoprotein, in conjunction with a liver-restricted promoter and a miRNA-regulated FVIII transgene resulted in sustained, therapeutic levels of FVIII. These modifications to the lentiviral delivery system effectively restricted FVIII transgene expression to the liver. Plasma levels of FVIII could be increased to around 9% that of normal levels when macrophages were depleted prior to treating the hemophilia A mice with the modified lentiviral FVIII delivery system.


Assuntos
Fator VIII/metabolismo , Hemofilia A/terapia , Lentivirus/genética , MicroRNAs/genética , Animais , Modelos Animais de Doenças , Fator VIII/genética , Terapia Genética/métodos , Camundongos , Camundongos Endogâmicos C57BL
6.
Blood ; 114(3): 677-85, 2009 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-19458355

RESUMO

Under certain instances, factor VIII (FVIII) stimulates an immune response, and the resulting neutralizing antibodies present a significant clinical challenge. Immunotherapies to re-establish or induce long-term tolerance would be beneficial, and an in-depth knowledge of mechanisms involved in tolerance induction is essential to develop immune-modulating strategies. We have developed a murine model system for studying mechanisms involved in induction of immunologic tolerance to FVIII in hemophilia A mice. We used lentiviral vectors to deliver the canine FVIII transgene to neonatal hemophilic mice and demonstrated that induction of long-term FVIII tolerance could be achieved. Hemophilia A mice are capable of mounting a robust immune response to FVIII after neonatal gene transfer, and tolerance induction is dependent on the route of delivery and type of promoter used. High-level expression of FVIII was not required for tolerance induction and, indeed, tolerance developed in some animals without evidence of detectable plasma FVIII. Tolerance to FVIII could be adoptively transferred to naive hemophilia recipient mice, and FVIII-stimulated splenocytes isolated from tolerized mice expressed increased levels of interleukin-10 and decreased levels of interleukin-6 and interferon-gamma. Finally, induction of FVIII tolerance mediated by this protocol is associated with a FVIII-expandable population of CD4(+)CD25(+)Foxp3(+) regulatory T cells.


Assuntos
Transferência Adotiva/métodos , Fator VIII/imunologia , Hemofilia A/imunologia , Tolerância Imunológica , Linfócitos T Reguladores/imunologia , Animais , Animais Recém-Nascidos , Citocinas/biossíntese , Citocinas/efeitos dos fármacos , Fator VIII/administração & dosagem , Técnicas de Transferência de Genes , Vetores Genéticos , Hemofilia A/terapia , Camundongos , Modelos Animais , Baço/citologia , Linfócitos T/transplante
7.
Br J Haematol ; 143(4): 552-8, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18986390

RESUMO

The multimeric plasma protein von Willebrand factor (VWF) is regulated in size by its protease, ADAMTS13 (a disintegrin and metalloproteinase with thrombospondin type 1 motif, member 13). Y1605-M1606 cleavage site mutations and single nucleotide polymorphisms (SNPs) in the VWF A1 and A2 domains were examined for alteration in ADAMTS13-mediated cleavage of VWF. Recombinant human full-length VWF (rVWF) was digested with recombinant human ADAMTS13 (rADAMTS13) using a dialysis membrane method with 1.5 mol/l urea, and analyzed via multimer migration distance. The glutathione-S-transferase (GST) and histidine-tagged construct, E1554-R1668 of VWF (VWF115) was assayed via enzyme-linked immunosorbent assay: VWF115 was bound to anti-GST coated plates, digested with rADAMTS13, and intact VWF115 detected via horseradish peroxidase-labelled anti-histidine tag antibody. All alterations examined in the Y1605-M1606 cleavage site greatly reduced the cleavability of VWF by ADAMTS13 in the rVWF assay. Greatest cleavage resistance in both assays was observed in Y1605A/M1606A. In contrast, Y1605H and M1606L show a loss of cleavability only in the rVWF assay, suggesting that an aromatic ring at 1605 is critical for ADAMTS13 recognition. Additionally, under our rVWF assay conditions, the G1643S polymorphism showed increased cleavage, suggesting a Type 2A VWD phenotype, while D1472H, Q1571H and P1601T showed slightly decreased ADAMTS13 cleavage. Our two complementary assay conditions show that A-domain changes in VWF alter ADAMTS13-mediated proteolysis.


Assuntos
Proteínas ADAM/genética , Fator de von Willebrand/genética , Proteínas ADAM/sangue , Proteína ADAMTS13 , Clivagem do DNA , Humanos , Mutagênese , Polimorfismo de Nucleotídeo Único , Estrutura Terciária de Proteína/genética , Proteínas Recombinantes/metabolismo , Fator de von Willebrand/metabolismo
8.
Stem Cells ; 25(10): 2660-9, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17615271

RESUMO

Novel therapeutic strategies for hemophilia must be at least as effective as current treatments and demonstrate long-term safety. To date, several small clinical trials of hemophilia gene transfer have failed to show the promise of preclinical evaluations. Therefore, we wanted to develop and evaluate the feasibility of a novel ex vivo gene transfer strategy whereby cells derived from progenitor cells are engineered to express factor VIII (FVIII) and then implanted subcutaneously to act as a depot for FVIII expression. Circulating blood outgrowth endothelial cells (BOECs) were isolated from canine and murine blood and transduced with a lentiviral vector encoding the canine FVIII transgene. To enhance safety, these cells were implanted subcutaneously in a Matrigel scaffold, and the efficacy of this strategy was compared with i.v. delivery of engineered BOECs in nonhemophilic nonobese diabetic/severe combined immunodeficiency mice. Therapeutic levels of FVIII persisted for 15 weeks, and these levels of stable expression were extended to 20 weeks when the cytomegalovirus promoter was replaced with the thrombomodulin regulatory element. Subsequent studies in immunocompetent hemophilic mice, pretreated with tolerizing doses of FVIII or with transient immunosuppression, showed therapeutic FVIII expression for 27 weeks before the eventual return to baseline levels. This loss of transgene expression appears to be due to the disappearance of the implanted cells. The animals treated with either of the two tolerizing regimens did not develop anti-FVIII antibodies. Biodistribution analysis demonstrated that BOECs were retained inside the subcutaneous implants. These results indicate, for the first time, that genetically modified endothelial progenitor cells implanted in a subcutaneous scaffold can provide sustained therapeutic levels of FVIII and are a promising and safe treatment modality for hemophilia A. Disclosure of potential conflicts of interest is found at the end of this article.


Assuntos
Células Endoteliais/transplante , Fator VIII/genética , Terapia Genética/métodos , Vetores Genéticos/uso terapêutico , Hemofilia A/terapia , Lentivirus/genética , Animais , Células Cultivadas/metabolismo , Células Cultivadas/transplante , Dessensibilização Imunológica , Cães , Células Endoteliais/metabolismo , Fator VIII/administração & dosagem , Fator VIII/biossíntese , Fator VIII/imunologia , Estudos de Viabilidade , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Sobrevivência de Enxerto , Hemofilia A/sangue , Hemofilia A/genética , Injeções Subcutâneas , Camundongos , Camundongos Endogâmicos NOD , Camundongos Mutantes , Camundongos SCID , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/fisiologia , Alicerces Teciduais , Transdução Genética
9.
Mol Ther Methods Clin Dev ; 2: 15033, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26636112

RESUMO

Recombinant FVIII manufacturing is characterized by poor product stability and low yields. Codon-optimization of transgenes accelerates translation by exploiting the synonymous codon usage bias of a species. However, this can alter the performance of the final product. Additionally, the effects of transgene design across diverse cell types are not well understood and are of interest for next-generation protein and gene therapies. To investigate the effects of transgene design across different host cells, B-domain-deleted (BDD) and modified codon-optimized (CO-N6) transgenes were inserted via lentiviral delivery into cBOECs, HEK293T, and MDCK cells. The CO-N6 cFVIII transgene produced threefold more protein per transgene in HEK293T cells, and sixfold more protein in the two canine cell lines. However, pharmacokinetic analysis in hemophilia A dogs demonstrated that cFVIII produced from cBOECs transduced with the CO-N6 transgene had significantly reduced in vivo recovery. Furthermore, this product showed reduced in vitro stability and activity on thrombin activation versus the BDD product. This trend was reversed in HEK293T lines. Overall, our results demonstrate the need for an integrated approach that not only assesses protein expression levels but also considers the influence that host-cells have on preserving the molecular and biochemical properties of the naturally occurring FVIII.

10.
Blood Coagul Fibrinolysis ; 24(7): 719-26, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23846000

RESUMO

von Willebrand factor (vWF) antigen levels are elevated in patients with end-stage kidney disease (ESKD). We determined the quantitative and qualitative abnormalities of vWF and factors influencing vWF proteolysis in participants with ESKD compared with age-matched controls and determined the association between abnormalities in vWF and mortality over 4 years of follow-up. vWF : Ag and von Willebrand factor propeptide (vWFpp) levels, vWF functional activity (vWF :RCo), vWF multimer profiles, ADAMTS-13, thrombospondin 1 (TSP-1), and interleukin 6 (IL-6) were evaluated before and after a single hemodialysis treatment in 55 individuals with vascular disease and an age-matched group of controls (n = 21). vWF : Ag and vWF activity were significantly higher in the ESKD patients and levels increased further following the dialysis procedure. The percentage of high molecular weight multimers (%HMWMs) was significantly elevated in the ESKD patients compared with controls. TSP-1 was lower and IL-6 was higher providing possible explanation for the increase in %HMWM in ESKD. The %HMWM dropped significantly in the postdialysis sample. Mortality at 4 years was significantly associated with vWF : Ag. There are higher plasma vWF : Ag levels and a small increase in HMWMs in the ESKD milieu. The acute drop in the %HMWM of vWF postdialysis appears to be due to shear forces encountered during the dialysis procedure. The contribution of these abnormalities to either a pro-thrombotic and/or pro-bleeding phenotype in this population requires further study.


Assuntos
Falência Renal Crônica/sangue , Fator de von Willebrand/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Humanos , Masculino , Pessoa de Meia-Idade
11.
Blood ; 109(1): 145-54, 2007 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-17190853

RESUMO

In order to evaluate the changes within the VWF gene that might contribute to the pathogenesis of type 1 von Willebrand disease (VWD), a large multicenter Canadian study was undertaken. We present data from the sequence analysis of the VWF gene in 123 type 1 VWD index cases and their families. We have identified putative mutations within the VWF gene in 63% (n = 78) of index cases, leaving 37% (n = 45) with no identified changes. These changes comprise 50 different putative mutations: 31 (62%) missense mutations, 8 (16%) changes involving the VWF transcriptional regulatory region, 5 (10%) small deletions/insertions, 5 (10%) splicing consensus sequence mutations, and 1 nonsense mutation. Twenty-one of the index cases had more than one putative VWF mutation identified. We were somewhat more likely to identify putative mutations in cases with lower VWF levels, and the contribution of other factors, such as ABO blood group, seems more important in milder cases. Taken as a whole, our data support a complex spectrum of molecular pathology resulting in type 1 VWD. In more severe cases, genetic changes are common within the VWF gene and are highly penetrant. In milder cases, the genetic determinants are more complex and involve factors outside of the VWF gene.


Assuntos
Mutação , Doenças de von Willebrand/genética , Fator de von Willebrand/genética , Sistema ABO de Grupos Sanguíneos/genética , Adolescente , Adulto , Idoso , Substituição de Aminoácidos , Canadá/epidemiologia , Criança , Pré-Escolar , Estudos de Coortes , Análise Mutacional de DNA , Saúde da Família , Feminino , Frequência do Gene , Genótipo , Humanos , Lactente , Masculino , Pessoa de Meia-Idade , Mutação de Sentido Incorreto , Fenótipo , Mutação Puntual , Doenças de von Willebrand/sangue , Doenças de von Willebrand/classificação , Doenças de von Willebrand/epidemiologia , Fator de von Willebrand/análise
12.
Blood ; 105(4): 1531-9, 2005 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-15498853

RESUMO

Mechanisms of tissue-restricted patterns of von Willebrand factor (VWF) expression involve activators and repressors that limit expression to endothelial cells and megakaryocytes. The relative transcriptional activity of the proximal VWF promoter was assessed in VWF-producing and -nonproducing cells, and promoter activity was highest in endothelial cells followed by megakaryocytes. Only basal VWF promoter activity was seen in nonendothelial cells. Here we identify a negative response element located at nucleotides (nts) +96/+105 and demonstrate, using chromatin immunoprecipitation (ChIP) analysis, that in vivo this sequence interacts with the E4BP4 transcriptional repressor. Differences in size and relative abundance of nuclear E4BP4 were observed. In HepG2 cells, low levels of larger forms of E4BP4 are present that directly interact with the negative response element. In VWF-expressing cells, high levels of smaller forms predominate with no evidence of direct DNA binding. However, in endothelial cells, mutation of the VWF E4BP4 binding motif not only restores but also further elevates VWF promoter activity, suggesting that E4BP4 may be part of a coordinated binding complex. These observations implicate this binding motif in repressing both activated and basal levels of VWF transcription by different cell type-specific mechanisms, and support the hypothesis that E4BP4 sequesters negative regulators of transcription, thereby enhancing activated gene expression.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Proteínas Repressoras/fisiologia , Fatores de Transcrição/fisiologia , Fator de von Willebrand/biossíntese , Fator de von Willebrand/metabolismo , Animais , Sequência de Bases , Fatores de Transcrição de Zíper de Leucina Básica , Western Blotting , Bovinos , Linhagem Celular , Linhagem Celular Tumoral , Cricetinae , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Éxons/genética , Fatores de Ligação G-Box , Células HeLa , Humanos , Camundongos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Proteínas Nucleares/análise , Proteínas Nucleares/metabolismo , Regiões Promotoras Genéticas/genética , Ligação Proteica/genética , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transfecção , Fator de von Willebrand/antagonistas & inibidores , Fator de von Willebrand/genética
13.
Blood ; 104(9): 2739-45, 2004 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-15226181

RESUMO

In this manuscript, we describe a case of type 2A von Willebrand disease (VWD) caused by the novel heterozygous G>A transition at nucleotide 3538, which should result in the putative, nonconservative substitution of G1180R. This mutation was reproduced by site-directed mutagenesis; however, the recombinant mutant protein was efficiently secreted from cells and assembled correctly into multimers. Because the substitution is located at the last nucleotide of exon 26, the patient's platelet von Willebrand factor (VWF) mRNA was analyzed and 3 transcripts were observed: the normal transcript without the 3538G>A transition, a transcript with the in-frame deletion of exon 26, and a transcript with the in-frame deletions of exons 23 and 26. These deletion VWF cDNA constructs were created and the resulting recombinant proteins were analyzed following transfection into COS-7 cells. Cotransfection results demonstrate that the exon-skipped transcripts led to intracellular retention, and the levels of VWF antigen (VWF:Ag) produced by these constructs were as follows: del23/26A transition at nucleotide (nt) 3538 does not result in the expression of the G1180R missense mutation, but rather leads to exon skipping, which is the pathogenic basis of the patient's phenotype. This is the first report of a coding region mutation resulting in the skipping of 2 nonadjacent exons.


Assuntos
Éxons , Mutação Puntual/fisiologia , Deleção de Sequência , Fator de von Willebrand/genética , Adolescente , Sequência de Aminoácidos , Saúde da Família , Feminino , Humanos , RNA/genética , Doenças de von Willebrand/etiologia , Doenças de von Willebrand/genética , Fator de von Willebrand/metabolismo
14.
Blood ; 102(2): 549-57, 2003 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-12649144

RESUMO

To date, no dominant mutation has been identified in a significant proportion of patients with type 1 von Willebrand disease (VWD). In this study, we examined 70 families as part of the Canadian Type 1 VWD Study. The entire VWF gene was sequenced for 1 index case, revealing 2 sequence variations: intron 30 (5312-19A>C) and exon 28 at Tyr1584Cys (4751A>G). The Tyr1584Cys variation was identified in 14.3% (10 of 70) of the families and was in phase with the 5312-19A>C variation in 7 (10.0%) families. Both variants were observed in 2 of 10 UK families with type 1 VWD, but neither variant was found in 200 and 100 healthy, unrelated persons, respectively. Mean von Willebrand factor antigen (VWF:Ag), VWF ristocetin cofactor (VWF:RCo), and factor VIII coagulant activity (FVIII:C) for the index cases in these families are 0.4 U/mL, 0.36 U/mL, and 0.54 U/mL, respectively, and VWF multimer patterns show no qualitative abnormalities. Aberrant VWF splicing was not observed in these patients, and both alleles of the VWF gene are expressed as RNA. Molecular dynamic simulation was performed on a homology model of the VWF-A2 domain containing the Tyr1584Cys mutation. This showed that no significant structural changes occur as a result of the substitution but that a new solvent-exposed reactive thiol group is apparent. Expression studies revealed that the Tyr1584Cys mutation results in increased intracellular retention of the VWF protein. We demonstrate that all the families with the Tyr1584Cys mutation share a common, evolved VWF haplotype, suggesting that this mutation is ancient. This is the first report of a mutation that segregates in a significant proportion of patients with type 1 VWD.


Assuntos
Substituição de Aminoácidos , Efeito Fundador , Íntrons/genética , Mutação de Sentido Incorreto , Doenças de von Willebrand/genética , Fator de von Willebrand/genética , Alelos , Canadá/epidemiologia , Códon/genética , Simulação por Computador , Evolução Molecular , França/epidemiologia , França/etnologia , Genótipo , Haplótipos , Humanos , Modelos Moleculares , Polimorfismo de Nucleotídeo Único , Regiões Promotoras Genéticas/genética , Conformação Proteica , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Compostos de Sulfidrila/análise , Reino Unido/epidemiologia , Reino Unido/etnologia , Doenças de von Willebrand/epidemiologia , Fator de von Willebrand/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA