Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Neuroepidemiology ; 37(2): 109-13, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21986109

RESUMEN

BACKGROUND: Several functional prothrombotic gene variants have been associated with cerebral ischemia and myocardial infarction. We hypothesized that such gene variants may also be associated with mortality after cerebral ischemia of arterial origin because of an increased risk of fatal vascular events. METHODS: We performed a case-control study in 316 long-term survivors and 887 patients with recent cerebral ischemia of arterial origin. False discovery rate q values were calculated to account for multiple testing. The mean duration between occurrence of cerebral ischemia and DNA collection was 16.8 years in long-term survivors and 3.2 months in recent patients. RESULTS: Two of 23 variants were associated with mortality: the 95Arg allele of the coagulation factor XIII subunit B (F13B) His95Arg variant (OR, 1.5 for Arg/Arg and His/Arg vs. His/His genotype; 95% CI, 1.1-2.2, q = 0.29) and the 4G allele of the plasminogen activator inhibitor-1 (PAI-1) 4G/5G variant (OR, 1.5 for 4G/4G and 5G/4G vs. 5G/5G genotype; 95% CI, 1.1-2.0, q = 0.29). Both associations disappeared after accounting for multiple testing. Data analysis restricted to recently deceased patients (n = 133) yielded similar results. CONCLUSIONS: In this hospital-based study none of 23 prothrombotic gene variants were associated with long-term mortality after cerebral ischemia of arterial origin. Prothrombotic gene variants do not appear to play an important role in long-term mortality after cerebral ischemia.


Asunto(s)
Isquemia Encefálica/genética , Isquemia Encefálica/mortalidad , Arterias Cerebrales , Variación Genética/genética , Anciano , Estudios de Casos y Controles , Arterias Cerebrales/patología , Factor XIII/genética , Femenino , Estudios de Seguimiento , Frecuencia de los Genes/genética , Humanos , Masculino , Persona de Mediana Edad
2.
Biochemistry ; 49(39): 8520-34, 2010 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-20722419

RESUMEN

Factor Va, the cofactor of prothrombinase, is composed of heavy and light chains associated noncovalently in the presence of divalent metal ions. The COOH-terminal region of the heavy chain contains acidic amino acid clusters that are important for cofactor activity. In this work, we have investigated the role of amino acid region 659-663, which contains five consecutive acidic amino acid residues, by site-directed mutagenesis. We have generated factor V molecules in which all residues were mutated to either lysine (factor V(5K)) or alanine (factor V(5A)). We have also constructed a mutant molecule with this region deleted (factor V(Δ659-663)). The recombinant molecules along with wild-type factor V (factor V(WT)) were transiently expressed in mammalian cells, purified, and assessed for cofactor activity. Two-stage clotting assays revealed that the mutant molecules had reduced clotting activities compared to that of factor Va(WT). Kinetic analyses of prothrombinase assembled with the mutant molecules demonstrated diminished k(cat) values, while the affinity of all mutant molecules for factor Xa was similar to that for factor Va(WT). Gel electrophoresis analyses of plasma-derived and recombinant mutant prothrombin activation demonstrated delayed cleavage of prothrombin at both Arg(320) and Arg(271) by prothrombinase assembled with the mutant molecules, resulting in meizothrombin lingering throughout the activation process. These results were confirmed after analysis of the cleavage of FPR-meizothrombin. Our findings provide new insights into the structural contribution of the acidic COOH-terminal region of factor Va heavy chain to factor Xa activity within prothrombinase and demonstrate that amino acid region 659-663 from the heavy chain of the cofactor contributes to the regulation of the rate of cleavage of prothrombin by prothrombinase.


Asunto(s)
Factor Va/química , Factor Va/metabolismo , Factor Xa/metabolismo , Tromboplastina/metabolismo , Secuencia de Aminoácidos , Animales , Precursores Enzimáticos/metabolismo , Factor V/genética , Factor V/metabolismo , Factor Va/genética , Expresión Génica , Humanos , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutación Puntual , Protrombina/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Trombina/metabolismo
3.
Thromb Haemost ; 101(1): 62-7, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19132190

RESUMEN

Protein C is an important inhibitor of blood coagulation. We investigated the effect of two polymorphisms within the promoter region of the protein C gene (C/T at position 2405 and A/G at position 2418) on risk of venous thrombosis and on plasma protein C levels. In addition the combined effect of the two polymorphisms with factor V Leiden and oral contraceptive use was investigated. Previous studies on these polymorphisms were small and were not able to investigate synergistic effects. In the Multiple Environmental and Genetic Assessment of risk factors for venous thrombosis (MEGA study), protein C levels were determined in 2,043 patients with venous thrombosis and 2,857 control subjects, and the two polymorphisms in 4,285 patients and 4,863 control subjects. The CC/GG genotype was associated with the lowest protein C levels. Compared to carriers of the TT/AA genotype - a genotype associated with higher protein C levels - the risk of venous thrombosis in CC/GG carriers was 1.3-fold increased (95% confidence interval 1.09-1.48). The combination of factor V Leiden with the CC/GG genotype led to a 4.7-fold increased risk, compared to non-carriers with the TT/AA genotype. Oral contraceptive use together with the CC/GG genotype resulted in a 5.2-fold increased risk. In conclusion, the CC/GG genotype is associated with lower levels of protein C and an elevated risk of venous thrombosis compared to the TT/AA genotype. There is no clear synergistic effect of the CC/GG genotype with factor V Leiden or oral contraceptive use on thrombotic risk.


Asunto(s)
Polimorfismo de Nucleótido Simple , Proteína C/genética , Trombosis de la Vena/genética , Adulto , Anciano , Estudios de Casos y Controles , Anticonceptivos Orales/efectos adversos , Factor V/genética , Femenino , Frecuencia de los Genes , Predisposición Genética a la Enfermedad , Haplotipos , Humanos , Masculino , Persona de Mediana Edad , Oportunidad Relativa , Fenotipo , Regiones Promotoras Genéticas , Proteína C/metabolismo , Medición de Riesgo , Factores de Riesgo , Trombosis de la Vena/sangre
4.
Thromb Haemost ; 101(6): 1078-84, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19492150

RESUMEN

Genetic determinants of venous thromboembolism (VTE) in the African-American population are poorly characterised. It was recently shown that fibrinogen gamma gene (FGG) polymorphisms 10034C>T and 9340T>C influence VTE risk in the Caucasian population. In the African-American population these polymorphisms are common, with allele frequencies above 25%. Here we evaluated whether these and other FGG 3'-end polymorphisms were associated with VTE risk in the African-American population and aimed to replicate the association in the Caucasian population. We examined 557 Caucasian patients and 678 Caucasian controls, and 537 African-American patients and 586 African-American controls from the ;Genetic Attributes and Thrombosis Epidemiology' (GATE) study. In the African-American population, 10034C>T and 9340T>C marginally influenced VTE-risk, with a 20% increase in risk for 10034TT carriers and a 20% reduction in risk for 9340CC carriers. In the Caucasian population, 10034TT was associated with a 1.7-fold increase in risk, which increased to 2.1-fold for idiopathic VTE patients. 9340CC significantly reduced VTE risk approximately two-fold. In conclusion, both FGG polymorphisms 10034C>T and 9340T>C influence VTE-risk, with the strongest effects observed in the Caucasian population, confirming previous data on these polymorphisms in this population.


Asunto(s)
Región de Flanqueo 3'/genética , Negro o Afroamericano , Fibrinógenos Anormales/genética , Predisposición Genética a la Enfermedad , Tromboembolia Venosa/genética , Población Blanca , Adolescente , Adulto , Anciano , Análisis Mutacional de ADN , Femenino , Fibrinógenos Anormales/metabolismo , Frecuencia de los Genes , Humanos , Masculino , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple , Factores de Riesgo , Tromboembolia Venosa/epidemiología , Tromboembolia Venosa/fisiopatología
5.
Stroke ; 39(3): 1033-5, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18239174

RESUMEN

BACKGROUND AND PURPOSE: To determine the contribution of fibrinogen gamma' levels and FGG haplotypes to ischemic stroke. METHODS: Associations between fibrinogen gamma' levels, fibrinogen gamma'/total fibrinogen ratio, and FGG haplotypes with the risk of ischemic stroke were determined in 124 cases and 125 controls. RESULTS: Fibrinogen gamma'/total fibrinogen ratio was higher in patients than in controls during the acute phase of the stroke and lower in the convalescent phase 3 months after the stroke. FGG haplotype 3 (H3) was associated with a reduced risk of ischemic stroke (odds ratio 0.60; 95% CI, 0.38 to 0.94), but not with the fibrinogen gamma'/total fibrinogen ratio. In contrast, FGG-H2 was associated with a decreased fibrinogen gamma'/total fibrinogen ratio, but not with risk of stroke. CONCLUSIONS: Fibrinogen gamma'/total fibrinogen ratio is associated with ischemic stroke, especially in the acute phase of the disease. In addition, FGG-H3 haplotype appears to be protective against ischemic stroke.


Asunto(s)
Isquemia Encefálica/complicaciones , Fibrinógenos Anormales/metabolismo , Accidente Cerebrovascular/sangre , Accidente Cerebrovascular/etiología , Adulto , Anciano , Alelos , Estudios de Casos y Controles , Femenino , Variación Genética , Haplotipos , Heterocigoto , Humanos , Ataque Isquémico Transitorio/sangre , Masculino , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple , Accidente Cerebrovascular/prevención & control
6.
Arterioscler Thromb Vasc Biol ; 27(6): 1486-91, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17413037

RESUMEN

OBJECTIVE: It has been suggested that the overall effect of the major proinflammatory cytokine interleukin-1 (IL-1) on coagulation and fibrinolysis is prothrombotic. The aim of this study was to investigate whether common variations in IL1B, IL1RN, IL1R1, and IL1R2 influence the risk of venous thrombosis. METHODS AND RESULTS: In a case-control study on the causes of deep venous thrombosis, the Leiden Thrombophilia Study (LETS), we genotyped 18 single nucleotide polymorphisms (SNPs) in IL1B, IL1RN, IL1R1, and IL1R2, enabling us to tag a total of 25 haplotype groups. Overall testing of the haplotype frequency distribution in patients and controls indicated that a recessive effect was present in IL1RN (P=0.031). Subsequently the risk of venous thrombosis was calculated for each haplotype of IL1RN. Increased thrombotic risk was found for homozygous carriers of haplotype 5 (H5, tagged by SNP 13888T/G, rs2232354) of IL1RN (Odds ratio=3.9; 95% confidence interval: 1.6 to 9.7; P=0.002). No risk was associated with haplotype 3 of IL1RN, which contains the frequently examined allele 2 variant of the intron 2 VNTR. CONCLUSIONS: We found that IL1RN-H5H5 carriership increases the risk of venous thrombosis.


Asunto(s)
Haplotipos , Proteína Antagonista del Receptor de Interleucina 1/genética , Interleucina-1beta/genética , Receptores Tipo II de Interleucina-1/genética , Receptores Tipo I de Interleucina-1/genética , Trombosis de la Vena/genética , Adolescente , Adulto , Anciano , Biomarcadores/sangre , Proteína C-Reactiva/metabolismo , Estudios de Casos y Controles , Femenino , Fibrinógeno/metabolismo , Frecuencia de los Genes , Predisposición Genética a la Enfermedad , Genotipo , Homocigoto , Humanos , Inflamación/sangre , Inflamación/genética , Intrones , Masculino , Persona de Mediana Edad , Oportunidad Relativa , Fenotipo , Polimorfismo de Nucleótido Simple , Medición de Riesgo , Factores de Riesgo , Trombosis de la Vena/sangre
7.
Arch Intern Med ; 167(5): 497-501, 2007 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-17353498

RESUMEN

BACKGROUND: Increased homocysteine levels are related to the occurrence of venous thrombosis, but whether this relation is causal is unclear. The T-variant of the common methylenetetrahydrofolate reductase (MTHFR) 677C-->T polymorphism mildly increases homocysteine levels. Meta-analyses have demonstrated a weak effect of the MTHFR 677TT genotype on risk but are sensitive to selective publication of positive results. The aim of the present study was to evaluate the effect of the MTHFR genotype on the risk of venous thrombosis, overall and in subgroups of known risk factors, in a single large study. METHODS: In the Multiple Environmental and Genetic Assessment of risk factors for venous thrombosis (MEGA Study), a population-based case-control study, we collected DNA from 4375 patients with a first deep vein thrombosis of the leg or pulmonary embolism and from 4856 control subjects. Information about risk factors for venous thrombosis was obtained from questionnaires. RESULTS: MTHFR 677C-->T was not associated with the risk of venous thrombosis (odds ratio [95% confidence interval], 0.99 [0.91-1.08] for the CT genotype and 0.94 [0.81-1.08] for the TT genotype). Stratification by known risk factors for venous thrombosis provided no evidence of an association in specific groups. CONCLUSIONS: In a single large study, MTHFR 677C-->T was not associated with the risk of venous thrombosis, and the narrow confidence interval excludes even a small effect. Therefore, mildly elevated homocysteine levels as a result of MTHFR 677TT do not seem to cause venous thrombosis. There is no rationale for measuring the MTHFR 677C-->T variant for clinical purposes.


Asunto(s)
ADN/genética , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Polimorfismo Genético , Trombosis de la Vena/genética , Adolescente , Adulto , Anciano , Alelos , Biomarcadores/sangre , Factor V/genética , Factor V/metabolismo , Femenino , Estudios de Seguimiento , Genotipo , Homocisteína/sangre , Humanos , Incidencia , Masculino , Persona de Mediana Edad , Países Bajos/epidemiología , Mutación Puntual , Reacción en Cadena de la Polimerasa , Pronóstico , Protrombina/genética , Protrombina/metabolismo , Embolia Pulmonar/sangre , Embolia Pulmonar/epidemiología , Embolia Pulmonar/genética , Estudios Retrospectivos , Factores de Riesgo , Encuestas y Cuestionarios , Trombosis de la Vena/sangre , Trombosis de la Vena/epidemiología
8.
Thromb Haemost ; 97(4): 534-41, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17393014

RESUMEN

ABO blood group is a genetic determinant of von Willebrand factor (VWF) levels. We investigated the effect of ABO genotypes on VWF and factor VIII (FVIII) levels and on the degree to which VWF is loaded with A- and B-antigens, expressed as normalized ratios, nA-ratio and nB-ratio, respectively, in the Leiden Thrombophilia Study, a large case-control study on venous thrombosis. We found that the ABO locus had an allele-specific, dosage dependent effect on VWF and FVIII levels and on the loading of VWF with A-antigen and B-antigen. The highest mean nA- and nB-ratios were found in A(1)A(1) and BB genotypes, respectively. Four A(1)O carriers had four 43-bp repeats in the minisatellite region of the ABO gene in stead of the expected one repeat. All had a reduced nA-ratio compared to A(1)O carriers with one repeat in their A(1) allele. The amount of A- and B-antigens expressed onVWF (nA-ratio and nB-ratio) explained about 18% (R(2)) of the variation in VWF levels.


Asunto(s)
Sistema del Grupo Sanguíneo ABO/genética , Sistema del Grupo Sanguíneo ABO/metabolismo , Factor VIII/metabolismo , Trombosis de la Vena/sangre , Factor de von Willebrand/metabolismo , Sistema del Grupo Sanguíneo ABO/sangre , Adolescente , Adulto , Anciano , Alelos , Antígenos de Grupos Sanguíneos/genética , Antígenos de Grupos Sanguíneos/metabolismo , Estudios de Casos y Controles , Ensayo de Inmunoadsorción Enzimática/métodos , Femenino , Dosificación de Gen , Genotipo , Humanos , Masculino , Persona de Mediana Edad , Repeticiones de Minisatélite , Vigilancia de la Población , Unión Proteica , Trombosis de la Vena/genética , Trombosis de la Vena/metabolismo
9.
Arterioscler Thromb Vasc Biol ; 26(9): 2168-74, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16840717

RESUMEN

OBJECTIVE: The protein C anticoagulant pathway is an essential process for attenuating thrombin generation by the membrane-bound procoagulant complexes tenase and prothrombinase. In this pathway, protein S (PS) serves as a cofactor for activated protein C. PS circulates in plasma both in a free form and in complex with complement component 4b-binding protein (C4BP). C4BP is a known acute phase reactant, thereby suggesting a relation between PS and the acute phase response. Interleukin (IL)-6 has been shown to increase both PS and C4BP gene expression. Our objective was to study the regulation of PS gene expression by IL-6 in detail. METHODS AND RESULTS: IL-6 upregulates both PS mRNA and protein levels in liver-derived HepG2 cells. The promoter of the PS gene (PROS1) was cloned upstream from a luciferase reporter gene. After transfection in HepG2 cells, the luciferase activity was shown to be stimulated by the addition of IL-6. IL-6 exerts its effect through Signal Transducer and Activator of Transcription 3 (STAT3) that interacts with the PROS1 promoter at a binding site in between nucleotides 229 to 207 upstream from the translational start. CONCLUSIONS: IL-6 induces PS expression via STAT3. A possible function for IL-6-induced PS expression in cell survival is discussed.


Asunto(s)
Interleucina-6/fisiología , Proteína S/biosíntesis , Factor de Transcripción STAT3/fisiología , Línea Celular Tumoral , Regulación de la Expresión Génica , Humanos , Interleucina-6/farmacología , Fosforilación , Regiones Promotoras Genéticas , Proteína S/genética , Proteína S/metabolismo , ARN Mensajero/biosíntesis , Elementos de Respuesta , Factor de Transcripción STAT3/metabolismo
10.
Eur J Intern Med ; 42: 54-60, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28579309

RESUMEN

BACKGROUND: Use of ethinylestradiol, one of the active ingredients in combined oral contraceptives, affects the incidence of venous thrombosis. To explain why some women develop thrombosis when using oral contraceptives and others do not, we hypothesized a role for the first-pass metabolism of ethinylestradiol in the liver. We set out to determine the association between genetic variation in the first-pass metabolism of ethinylestradiol, venous thrombosis risk and the effect on Sex-hormone-binding-globulin (SHBG) levels. METHODS: Premenopausal women were included from two case-control studies: LETS (103 cases; 159 controls) and MEGA (397 cases; 796 controls). Haplotype-tagging SNPs were selected in 11 candidate genes; COMT, CYP1A2, CYP2C9, CYP3A4, CYP3A5, SULT1A1, SULT1E1, UGT1A1, UGT1A3, UGT1A9, UGT2B7. Venous thrombosis risk was expressed as odds ratios (OR) with 95% confidence intervals (CI). For SHBG levels, mean differences with 95%CI were estimated in combined oral contraceptive-using control subjects from the MEGA study. RESULTS: Two copies of haplotype D in the UGT2B7 gene increased venous thrombosis risk (ORLETS: 3.78; ORMEGA: 2.61) as well as SHBG levels (mean difference 27.6nmol/L, 95%CI: -61.7 to 116.9 compared with no copies) in oral contraceptive users and not in non-users. In oral contraceptive users, haplotype A and B in the CYP3A4 gene were associated with venous thrombosis risk, but not in non-users; however, the effect on SHBG levels was not directional with the risk. None of the other haplotypes were associated with venous thrombosis. CONCLUSION: Genetic variation in the UGT2B7 gene may, in part, explain venous thrombosis risk in combined oral contraceptive users.


Asunto(s)
Anticonceptivos Orales Combinados/sangre , Etinilestradiol/sangre , Glucuronosiltransferasa/genética , Globulina de Unión a Hormona Sexual/análisis , Trombosis de la Vena/genética , Adolescente , Adulto , Estudios de Casos y Controles , Citocromo P-450 CYP3A/genética , Femenino , Variación Genética , Humanos , Modelos Lineales , Modelos Logísticos , Persona de Mediana Edad , Países Bajos , Factores de Riesgo , Trombosis de la Vena/sangre , Trombosis de la Vena/epidemiología , Adulto Joven
11.
Thromb Haemost ; 95(6): 942-8, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16732372

RESUMEN

Levels of factor VIII (FVIII) are associated with the risk of venous thrombosis. The FVIII variation D1241E has been reported to be associated with decreased levels of FVIII. Our objective was to study whether D1241E is associated with levels of FVIII and the risk of venous thrombosis and whether this association is caused by D1241E or another linked variation. We analyzed the association of three FVIII gene haplotypes encoding 1241E (further denoted as HT1, HT3, and HT5) with FVIII levels and thrombosis risk. This analysis was performed in the Leiden Thrombophilia Study (LETS). The control populations of two case-controls studies on arterial thrombosis in men and women, respectively, were used to confirm the effects observed on FVIII:C in the LETS. In men, HT1 was associated with a 6% reduction in FVIII:C and with a reduced risk of venous thrombosis [odds ratio 0.4 (CI95 0.2-0.8)]. Logistic regression showed that the risk reduction was only partially dependent of the reduction in FVIII levels. HT1 showed no effects in women on either FVIII:C or risk of thrombosis. The number of carriers of HT3 and HT5 was too low to make an accurate estimate of the risk of venous thrombosis. Neither HT3 nor HT5 showed effects on levels of FVIII:C. When we consider that all three haplotypes encoding 1241E show different effects on FVIII:C and thrombosis risk, it is possible that D1241E is not the functional variation. However, FVIII gene variations do contribute to both levels of FVIII and the risk of thrombosis.


Asunto(s)
Factor VIII/genética , Factor VIII/metabolismo , Trombosis de la Vena/genética , Anciano , Estudios de Casos y Controles , Femenino , Haplotipos , Humanos , Modelos Logísticos , Masculino , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple , Proteína C/metabolismo , Factores de Riesgo , Análisis de Secuencia de ADN , Factores Sexuales , Trombosis de la Vena/sangre
12.
Thromb Haemost ; 96(6): 717-24, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17139364

RESUMEN

Von Willebrand factor (VWF) contains a large number of cysteine residues, which all form disulfide bonds. Mutations of cysteines located in the cystine-knot (CK) domain of VWF have been identified in both qualitative type 2A (IID) and quantitative type 3 von Willebrand disease (VWD). Our objective was to test the hypothesis that the difference in phenotype is related to whether the mutated cysteine residue is involved in either interchain- or intrachain-disulfide-bond formation. The effects of three cysteine mutations which are all located in the CK-domain of VWF, C2773S (type 2A(IID)), C2739Y (type 3), and C2754W (type 3), were studied by transient expression in 293T cells. Cotransfection of wild-type (wt) and C2773S VWF constructs reproduced the plasma phenotype of heterozygous type 2A(IID) patients, with normal to high levels of VWF antigen (VWF:Ag), absence of high-molecular-weight multimers, and the presence of intervening bands between the normal multimers. In contrast, single transfections of C2739Y or C2754W resulted in a quantitative VWF defect with low VWF:Ag levels, and co-transfections of wt and mutant constructs resulted in a 50% reduction of VWF:Ag and only a minor effect on VWF multimerization. We demonstrated N-terminal dimerization of VWF-C2773S and both N- and C-terminal dimerization of VWF-C2754W. Our data suggest that loss of a single disulfide bond in the CK-domain of VWF leads to a recessive, quantitative VWF deficiency if an intrachain-disulfide bond is involved, and to a dominant-negative, qualitative defect of VWF if an interchain-disulfide bond is involved.


Asunto(s)
Coagulación Sanguínea , Motivos Nodales de Cisteina , Disulfuros/química , Procesamiento Proteico-Postraduccional , Enfermedades de von Willebrand/metabolismo , Factor de von Willebrand/metabolismo , Proteínas ADAM/metabolismo , Proteína ADAMTS13 , Línea Celular , Cisteína , Análisis Mutacional de ADN , Dimerización , Electroforesis en Gel de Poliacrilamida , Femenino , Humanos , Masculino , Mutación Missense , Linaje , Fenotipo , Conformación Proteica , Transfección , Enfermedades de von Willebrand/sangre , Enfermedades de von Willebrand/genética , Factor de von Willebrand/química , Factor de von Willebrand/genética
15.
Atherosclerosis ; 181(2): 275-84, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16039281

RESUMEN

BACKGROUND: Elevated plasminogen activator inhibitor-1 (PAI-1) concentrations are associated with cardiovascular diseases. PAI-1 antigen levels are influenced by environmental factors such as body mass index (BMI), and by genetic factors. The PAI-1 promoter of the PAI-1 gene contains two common polymorphisms (-844A/G and -675(4G/5G)) and the 4G allele of the -675(4G/5G) variation has been associated with elevated PAI-1 concentrations and on some occasions with an increased risk of cardiovascular disease. OBJECTIVES: The aim of our study was to investigate the effect of the PAI-1 promoter haplotype on PAI-1 concentrations and to determine the role of BMI. METHODS: The association between the PAI-1 promoter haplotype and PAI-1 antigen levels was investigated in two independent populations, each including 600 healthy Caucasians. Furthermore, to assess the effect of the PAI-1 promoter haplotype on PAI-1 promoter activity, in vitro reporter gene assays were performed in HepG2 and BAEC cells. RESULTS: We observed significantly higher PAI-1 concentrations in A-4G homozygotes than in G-5G carriers in lean subjects (BMI in the lowest quartile). In these lean subjects, the PAI-1 concentrations in A-4G/G-5G heterozygotes were reduced to 60-75%, and the concentrations in G-5G homozygotes to 45-55%, compared to the PAI-1 concentrations of A-4G homozygotes (p < 0.01). PAI-1 concentrations increased approximately four-fold from the lowest to the highest BMI quartile (p < 0.01). The reporter gene assays did not support a direct effect of the PAI-1 promoter haplotype on promoter activity in HepG2 or BEAC cells. CONCLUSIONS: Our study suggests that the PAI-1 promoter haplotype and BMI affect PAI-1 concentrations and that BMI is a stronger determinant than PAI-1 promoter variation.


Asunto(s)
Índice de Masa Corporal , Peso Corporal/genética , Inhibidor 1 de Activador Plasminogénico/sangre , Inhibidor 1 de Activador Plasminogénico/genética , Regiones Promotoras Genéticas/genética , Adulto , Anciano , Carcinoma Hepatocelular , Línea Celular Tumoral , Femenino , Genes Reporteros , Variación Genética , Haplotipos , Humanos , Neoplasias Hepáticas , Masculino , Persona de Mediana Edad
16.
Arterioscler Thromb Vasc Biol ; 24(5): 975-81, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15044210

RESUMEN

OBJECTIVE: It is generally assumed that C-reactive protein (CRP) induces synthesis of tissue factor (TF) in monocytic cells, thereby potentially initiating intravascular blood coagulation. We aimed to elucidate the mechanism of CRP-induced TF expression in monocytes and monocyte-derived macrophages (MDMs) in vitro. METHODS AND RESULTS: Monocytes were isolated from the blood of healthy donors and cultured with or without CRP or lipopolysaccharide (LPS) to study the time course of TF antigen and TF mRNA expression. Addition of 100 microg/mL CRP did not result in a significant increase in TF antigen (range: 9 to 163 pg/10(6) cells, n=11) and TF mRNA (relative number of TF transcripts; N(TF)=0.01 to 0.33), when compared with nonstimulated cells (TF antigen 7 to 46 pg/10(6) cells, N(TF)=0.01 to 0.13). Variation of CRP concentration and exposure time did not affect the TF response. Similar results were obtained in monocytes cultured in suspension and in MDMs. In contrast, TF was strongly induced by 10 microg/mL LPS (TF antigen 1125 to 6120 pg/10(6) cells, N(TF)=5.94 to 23.43). Cultured monocytes did express FcRgammaII, a putative CRP receptor, and addition of CRP induced a 7-fold increase in the production of monocyte chemoattractant protein-1 (MCP-1). Interestingly, CRP addition to peripheral blood mononuclear cells (PBMCs) did result in TF expression on monocytic cells. CONCLUSIONS: The absence of TF induction after incubation of purified monocytes with CRP indicates that CRP is unable to induce TF expression in monocytes and MDMs directly. The presence of CRP-induced TF expression in PBMCs suggests that CRP can induce TF indirectly, probably through cross-talk between cells.


Asunto(s)
Proteína C-Reactiva/farmacología , Monocitos/efectos de los fármacos , Tromboplastina/biosíntesis , Células Sanguíneas/fisiología , Proteína C-Reactiva/fisiología , Células Cultivadas/efectos de los fármacos , Células Cultivadas/metabolismo , Quimiocina CCL2/biosíntesis , Quimiocina CCL2/genética , Sistemas de Computación , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Lipopolisacáridos/farmacología , Monocitos/metabolismo , Reacción en Cadena de la Polimerasa , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Tromboplastina/genética
17.
Eur J Hum Genet ; 12(6): 478-82, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15054398

RESUMEN

A re-emerging strategy in the search for disease susceptibility genes is the evaluation of candidate genes, which are thought to play a role in disease pathogenesis. Candidate genes are screened for single nucleotide polymorphisms (SNPs) in a case-control study. The factor V Leiden (FVL) mutation (1691G --> A in the F5 gene) is an important risk factor for venous thrombosis. We asked ourselves whether the FVL mutation would have been found using the candidate gene approach in the absence of prior knowledge of the haplotype structure of the F5 gene. We typed four SNPs in the F5 gene in the Leiden Thrombophilia study, that is, promoter (99930G --> A), exon 13 (55907A --> G), exon 16 (42855A --> G), and intron 19 (37833T --> G). These SNPs were known to have different population frequencies, making their presence in distinct haplotypes likely. None of these SNPs has previously been associated with venous thrombotic risk. Subsequently we derived haplotypes. One haplotype was clearly more frequent in patients than controls (GAAT; 20 versus 9%), suggesting that a polymorphism in or near the F5 gene in this haplotype is associated with an increased thrombotic risk. If we had sequenced the F5 gene in patients homozygous for this haplotype, in order to locate the possible causal polymorphism, we would have found that 16 (76%) patients were homozygous or heterozygous for a missense mutation in exon 10 (1691G --> A), which predicts the replacement of Arg506 by Gln in one of the cleavage sites for activated protein C, a mutation that we now know as the FVL mutation.


Asunto(s)
Factor V/genética , Predisposición Genética a la Enfermedad/genética , Mutación/genética , Polimorfismo de Nucleótido Simple/genética , Trombofilia/genética , Trombosis de la Vena/genética , Estudios de Casos y Controles , Frecuencia de los Genes , Genética de Población , Humanos , Factores de Riesgo
18.
Thromb Haemost ; 89(3): 429-37, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12624624

RESUMEN

The R2 haplotype of the FV gene spans from exon 8 through 25 and comprises several strongly linked polymorphisms in the FV gene, including some missense mutations. Carriership of the R2-FV allele has been associated with reduced plasma FV levels, increased FV1/FV2 ratios and mild APC resistance. Some studies have reported that carriership of the R2-FV allele is associated with an increased risk of venous thombosis. At this moment, the individual contribution to the R2-associated phenotypes of the different mutations linked to the R2 haplotype of FV is unclear. The main objective of our study was to obtain insight in the influence of the R2-related Asp2194Gly mutation on FV expression, FV structure and FV function using Bdomainless rFV mutants. Replacing Asp at position 2194 by Gly resulted in a more than threefold reduction of rFV expression compared to rFV wild-type. Therefore, we propose that the R2-linked Asp2194Gly mutation is an important determinant of the association of the R2-FV allele with lower FV levels. Furthermore, the light chains from Asp2194Gly containing rFV mutants showed similar molecular weights as the light chains of the non-glycosylated rFVwt or the plasma FV2 isoform, indicating that glycosylation at Asn2181 is not stimulated by the presence of a glycine in position 2194. Finally, the apparent K(d) for dissociation of the FXaVa complex (K(1/2Xa)) was not higher in rFV mutants with the Asp2194Gly mutation than for rFVwt, suggesting that also the affinity for negatively charged phospholipids is not affected by substitution of Asp into Gly at position at 2194.


Asunto(s)
Factor V/química , Factor V/genética , Alelos , Sustitución de Aminoácidos , Secuencia de Bases , ADN Complementario/genética , Factor V/metabolismo , Factor Va/química , Factor Va/genética , Factor Va/metabolismo , Factor Xa/química , Factor Xa/genética , Factor Xa/metabolismo , Expresión Génica , Glicosilación , Haplotipos , Humanos , Técnicas In Vitro , Cinética , Mutagénesis Sitio-Dirigida , Polimorfismo Genético , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
19.
Thromb Haemost ; 90(3): 439-45, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12958613

RESUMEN

Deep vein thrombosis (DVT) and pulmonary embolism (PE) are considered to be two forms of the same disease, however it is not fully understood what determines their clinical presentation. Proteins encoded by the FXIIIA and TAFI genes are involved in stabilizing the fibrin clot and in making it more lysis resistant. The FXIIIA 34Leu and TAFI -438A alleles might protect against DVT. Information on such an association with PE is either contradictory or missing. We hypothesized that both polymorphisms might influence the formation and fate of emboli and accordingly the risk of PE. We determined the frequencies of both polymorphisms in patients with objectively demonstrated PE. The frequency of FXIIIA Leu34Leu in PE patients and non-PE patients was 4.5% and 8.8%, [OR 0.5 (95% CI: 0.1 to 1.9)], respectively. For -438 A/A TAFI genotype the frequency was 1.5% and 8.1% [OR 0.1 (95% CI: 0.02 to 1.1)], respectively.


Asunto(s)
Carboxipeptidasa B2/genética , Factor XIII/genética , Polimorfismo Genético/fisiología , Embolia Pulmonar/genética , Adulto , Anciano , Carboxipeptidasa B2/fisiología , Estudios de Casos y Controles , Análisis Mutacional de ADN , Factor XIII/fisiología , Femenino , Frecuencia de los Genes , Predisposición Genética a la Enfermedad/genética , Humanos , Masculino , Persona de Mediana Edad , Mutación Missense/fisiología , Oportunidad Relativa , Mutación Puntual/fisiología , Embolia Pulmonar/sangre , Embolia Pulmonar/etiología , Trombosis de la Vena
20.
Hypertens Pregnancy ; 23(2): 211-8, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15369653

RESUMEN

OBJECTIVE: Recently, a polymorphism of the gene encoding for the G protein beta3-subunit (GNB3) has been described. The T allele of this polymorphism (825T) is associated with endothelium dysfunction. Endothelium dysfunction has been described in women with preeclampsia. We, therefore, tested the hypothesis that in women who have had preeclampsia the T allele is more prevalent than in controls. STUDY DESIGN: We conducted a case-control study of 157 women with preeclampsia during 1991-1996. Cases and controls were tested for the presence of 825T by genotyping. Logistic regression methods were used for data analysis. RESULTS: The frequency of the T allele of the GNB3 gene was similar in cases (0.30) and controls (0.27) (odds ratio 1.24, 95% confidence interval 0.88-1.75). Compared to controls, we found a high frequency of the T allele in patients with the hemolysis, elevated liver enzymes, and low platelet count (HELLP) syndrome (odds ratio 4.25, 95% confidence interval 1.12-16.05). CONCLUSION: The frequency of the T allele of the GNB3 gene, which serves as a marker for endothelium dysfunction, was not different between women with preeclampsia and controls. Women with the HELLP syndrome had a high frequency of the T allele. This suggests that this polymorphism in the GNB3 gene does not contribute to endothelium dysfunction in women with preeclampsia while it does contribute in women with the HELLP syndrome.


Asunto(s)
Subunidades beta de la Proteína de Unión al GTP/genética , Polimorfismo Genético/genética , Preeclampsia/genética , Adulto , Estudios de Casos y Controles , Femenino , Frecuencia de los Genes/genética , Predisposición Genética a la Enfermedad/genética , Genotipo , Síndrome HELLP/genética , Humanos , Países Bajos , Embarazo , Índice de Severidad de la Enfermedad , Estadística como Asunto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA