Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
Leukemia ; 34(10): 2648-2659, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32107471

RESUMO

Myelofibrosis (MF) occurs as part of the natural history of polycythemia vera (PV) and essential thrombocythemia (ET), and remarkably shortens survival. Although JAK2V617F and CALR allele burden are the main transformation risk factors, inflammation plays a critical role by driving clonal expansion toward end-stage disease. NF-κB is a key mediator of inflammation-induced carcinogenesis. Here, we explored the involvement of miR-146a, a brake in NF-κB signaling, in MPN susceptibility and progression. rs2910164 and rs2431697, that affect miR-146a expression, were analyzed in 967 MPN (320 PV/333 ET/314 MF) patients and 600 controls. We found that rs2431697 TT genotype was associated with MF, particularly with post-PV/ET MF (HR = 1.5; p < 0.05). Among 232 PV/ET patients (follow-up time=8.5 years), 18 (7.8%) progressed to MF, being MF-free-survival shorter for rs2431697 TT than CC + CT patients (p = 0.01). Multivariate analysis identified TT genotype as independent predictor of MF progression. In addition, TT (vs. CC + CT) patients showed increased plasma inflammatory cytokines. Finally, miR-146a-/- mice showed significantly higher Stat3 activity with aging, parallel to the development of the MF-like phenotype. In conclusion, we demonstrated that rs2431697 TT genotype is an early predictor of MF progression independent of the JAK2V617F allele burden. Low levels of miR-146a contribute to the MF phenotype by increasing Stat3 signaling.


Assuntos
MicroRNAs/genética , Transtornos Mieloproliferativos/genética , Mielofibrose Primária/genética , Idoso , Alelos , Animais , Citocinas/genética , Progressão da Doença , Feminino , Genótipo , Humanos , Inflamação/genética , Inflamação/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Mutação/genética , Transtornos Mieloproliferativos/patologia , NF-kappa B/genética , Policitemia Vera/genética , Policitemia Vera/patologia , Transdução de Sinais/genética , Trombocitemia Essencial/genética , Trombocitemia Essencial/patologia
2.
J Autoimmun ; 82: 31-40, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28465139

RESUMO

OBJECTIVES: 1) To assess the association of NETosis and NETosis-derived products with the activity of the disease and the development of cardiovascular disease in RA; 2) To evaluate the involvement of NETosis on the effects of biologic therapies such as anti-TNF alpha (Infliximab) and anti-IL6R drugs (Tocilizumab). METHODS: One hundred and six RA patients and 40 healthy donors were evaluated for the occurrence of NETosis. Carotid-intimae media thickness was analyzed as early atherosclerosis marker. Inflammatory and oxidative stress mediators were quantified in plasma and neutrophils. Two additional cohorts of 75 RA patients, treated either with Infliximab (n = 55) or Tocilizumab (n = 20) for six months, were evaluated. RESULTS: NETosis was found increased in RA patients, beside myeloperoxidase and neutrophil elastase protein levels. Cell-free nucleosomes plasma levels were elevated, and strongly correlated with the activity of the disease and the positivity for autoantibodies, alongside inflammatory and oxidative profiles in plasma and neutrophils. Moreover, ROC analyses showed that cell-free nucleosomes levels could identify RA patients showing early atherosclerosis with high specificity. RA patients treated either with IFX or TCZ for six months exhibited decreased generation of NETs. Concomitantly, clinical parameters and serum markers of inflammation were found reduced. Mechanistic in vitro analyses showed that inhibition of NETs extrusion by either DNase, IFX or TCZ, further abridged the endothelial dysfunction and the activation of immune cells, thus influencing the global activity of the vascular system. CONCLUSIONS: NETosis-derived products may have diagnostic potential for disease activity and atherosclerosis, as well as for the assessment of therapeutic effectiveness in RA.


Assuntos
Artrite Reumatoide/complicações , Aterosclerose/diagnóstico , Aterosclerose/etiologia , Armadilhas Extracelulares/metabolismo , Idoso , Antirreumáticos/uso terapêutico , Aterosclerose/terapia , Biomarcadores , Estudos de Casos e Controles , Comorbidade , Feminino , Humanos , Mediadores da Inflamação/metabolismo , Interleucina-6/antagonistas & inibidores , Interleucina-6/metabolismo , Masculino , Pessoa de Meia-Idade , Estresse Oxidativo , Peroxidase , Curva ROC , Fatores de Risco , Índice de Gravidade de Doença , Resultado do Tratamento , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/metabolismo
3.
J Thromb Haemost ; 14(12): 2410-2418, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27681307

RESUMO

Essentials Vitamin K-dependent coagulant factor deficiency (VKCFD) is a rare autosomal recessive disorder. We describe a case of inherited VKCFD due to uniparental disomy. The homozygous mutation caused the absence of GGCX isoform 1 and overexpression of Δ2GGCX. Hepatic and non-hepatic vitamin K-dependent proteins must be assayed to monitor VKCFD treatment. SUMMARY: Background Inherited deficiency of all vitamin K-dependent coagulant factors (VKCFD) is a rare autosomal recessive disorder caused by mutations in the γ-glutamyl carboxylase gene (GGCX) or the vitamin K epoxide reductase gene (VKORC1), with great heterogeneity in terms of both clinical presentation and response to treatment. Objective To characterize the molecular basis of VKCFD in a Spanish family. Methods and Results Sequencing of candidate genes, comparative genomic hybridization and massive sequencing identified a new mechanism causing VKCFD in the proband. Uniparental disomy (UPD) of chromosome 2 caused homozygosity of a mutation (c.44-1G>A) resulting in aberrant GGCX splicing. This change contributed to absent expression of the mRNA coding for the full-length protein, and to four-fold overexpression of the smaller mRNA isoform lacking exon 2 (Δ2GGCX). Δ2GGCX might be responsible for two unexpected clinical observations in the patient: (i) increased plasma osteocalcin levels following vitamin K1 supplementation; and (ii) a mild non-bleeding phenotype. Conclusions Our study identifies a new autosomal disease, VKCFD1, caused by UPD. These data suggest that the Δ2GGCX isoform may retain enzymatic activity, and strongly encourage the evaluation of both hepatic and non-hepatic vitamin K-dependent proteins to assess differing responses to vitamin K supplementation in VKCFD patients.


Assuntos
Coagulação Sanguínea , Dissomia Uniparental , Vitamina K Epóxido Redutases/deficiência , Vitamina K/metabolismo , Carbono-Carbono Ligases/genética , Hibridização Genômica Comparativa , Feminino , Hemostasia , Homozigoto , Humanos , Lactente , Perda de Heterozigosidade , Masculino , Mutação , Fenótipo , Regiões Promotoras Genéticas , RNA Mensageiro/metabolismo , Espanha , Vitamina K Epóxido Redutases/genética
4.
Sci Rep ; 6: 31375, 2016 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-27502756

RESUMO

MicroRNAs markedly affect the immune system, and have a relevant role in CVD and autoimmune diseases. Yet, no study has analyzed their involvement in atherothrombosis related to APS and SLE patients. This study intended to: 1) identify and characterize microRNAs linked to CVD in APS and SLE; 2) assess the effects of specific autoantibodies. Six microRNAs, involved in atherothrombosis development, were quantified in purified leukocytes from 23 APS and 64 SLE patients, and 56 healthy donors. Levels of microRNAs in neutrophils were lower in APS and SLE than in healthy donors. Gene and protein expression of miRNA biogenesis-related molecules were also reduced. Accordingly, more than 75% of identified miRNAs by miRNA profiling were underexpressed. In monocytes, miR124a and -125a were low, while miR-146a and miR-155 appeared elevated. Altered microRNAs' expression was linked to autoimmunity, thrombosis, early atherosclerosis, and oxidative stress in both pathologies. In vitro treatment of neutrophils, monocytes, and ECs with aPL-IgG or anti-dsDNA-IgG antibodies deregulated microRNAs expression, and decreased miRNA biogenesis-related proteins. Monocyte transfections with pre-miR-124a and/or -125a caused reduction in atherothrombosis-related target molecules. In conclusion, microRNA biogenesis, significantly altered in neutrophils of APS and SLE patients, is associated to their atherothrombotic status, further modulated by specific autoantibodies.


Assuntos
Síndrome Antifosfolipídica/sangue , Lúpus Eritematoso Sistêmico/sangue , MicroRNAs/sangue , Trombose/sangue , Adulto , Autoanticorpos/sangue , Biomarcadores/metabolismo , Espessura Intima-Media Carotídea , Estudos de Casos e Controles , Biologia Computacional , Epigênese Genética , Feminino , Humanos , Imunoglobulina G/sangue , Inflamação , Leucócitos/citologia , Masculino , Pessoa de Meia-Idade , Monócitos/citologia , Neutrófilos/metabolismo , Estresse Oxidativo , Transfecção
5.
J Thromb Haemost ; 14(6): 1226-37, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26999003

RESUMO

UNLABELLED: Essentials Estrogens are known to influence the expression of microRNAs in breast cancer cells. We looked at microRNAs in estrogenic regulation of tissue factor pathway inhibitor α (TFPIα). Estrogen upregulated microRNA-27a/b and microRNA-494 through the estrogen receptor α. MicroRNA-27a/b and microRNA-494 are partly involved in estrogenic downregulation of TFPIα. SUMMARY: Background Tissue factor pathway inhibitor (TFPI) has been linked to breast cancer pathogenesis. We have recently reported TFPI mRNA levels to be downregulated by estrogens in a breast cancer cell line (MCF7) through the estrogen receptor α (ERα). Accumulating evidence also indicates that activation of ERα signaling by estrogens may modulate the expression of target genes indirectly through microRNAs (miRNAs). Objectives To examine if miRNAs are involved in the estrogenic downregulation of TFPIα. Methods Computational analysis of the TFPI 3'-untranslated region (UTR) identified potential binding sites for miR-19a/b, miR-27a/b, miR-494, and miR-24. Transient overexpression or inhibition of the respective miRNAs was achieved by transfection of miRNA mimics or inhibitors. Direct targeting of TFPI 3'-UTR by miR-27a/b and miR-494 was determined by luciferase reporter assay in HEK293T cells. Effects of 17α-ethinylestradiol (EE2) and fulvestrant on relative miR-27a/b, miR-494, and TFPI mRNA levels in MCF7 cells were determined by qRT-PCR and secreted TFPIα protein by ELISA. Transient knockdown of ERα was achieved by siRNA transfection. Results EE2 treatment lead to a significant increase in miR-19a, miR-27a/b, miR-494, and miR-24 mRNA levels in MCF7 cells through ERα. miR-27a/b and miR-494 mimics lead to reduced TFPI mRNA and protein levels. Luciferase assay showed direct targeting of miR-27a/b and miR-494 on TFPI mRNA. Impaired estrogen-mediated downregulation of TFPI mRNA was detected in anti-miR-27a/b and anti-miR-494 transfected cells. Conclusions Our results provide evidence that miR-27a/b and miR-494 regulate TFPIα expression and suggest a possible role of these miRNAs in the estrogen-mediated downregulation of TFPIα.


Assuntos
Regulação para Baixo , Estrogênios/química , Lipoproteínas/metabolismo , MicroRNAs/metabolismo , Regiões 3' não Traduzidas , Sítios de Ligação , Linhagem Celular Tumoral , Receptor alfa de Estrogênio/metabolismo , Estrogênios/metabolismo , Fator Xa/química , Células HEK293 , Humanos , Células MCF-7 , Ligação Proteica , Transfecção
6.
J Thromb Haemost ; 8(5): 1012-7, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20149073

RESUMO

SUMMARY BACKGROUND: There is currently intense debate as to whether pharmacogenetic algorithms for estimating the initial dose of coumarins provide a more accurate dose than the fixed-dose approach. Recently, it has been suggested that the greatest benefit of pharmacogenetic algorithms is observed in patients with extreme dose requirements. OBJECTIVES: To identify clinical and genetic factors that better characterize patients who need extreme acenocoumarol doses for steady anticoagulation state. PATIENTS/METHODS: We reviewed 9538 patients with a steady acenocoumarol dose from three Spanish hospitals, selecting 83 who took or= 30.00 mg week(-1) (p95). We also selected patients matched by gender and age taking 13.50-14.00 mg week(-1) (p50). We genotyped VKORC1 (rs9923231), CALU (rs1043550), GGCX (rs699664), CYP2C9 (rs1799853; rs1057910), CYP4F2 (rs2108622) and F7 (rs5742910) single-nucleotide polymorphisms (SNPs). RESULTS: Comparison between p5 and p95 revealed five parameters with significant differences: body surface area (BSA) (P = 0.006), age, VKORC1, CYP2C9 and CYP4F2 genotypes (all P < 0.001). First VKORC1, and second, CYP2C9 SNPs played a strong effect by determining extreme doses, particularly in p95. Only one out of 203 p95 had the VKORC1 A-1639A genotype, but this subject was CYP2C9*1/*1. In contrast, nine out of 83 p5 carried the VKORC1 G-1639G genotype, although six of them were CYP2C9*3 homozygotes and another two were heterozygotes. Surprisingly, CYP4F2 V433M SNP displayed prevalences that suggest that its influence might only be evident when patients are treated with high doses. CONCLUSION: Two clinical data, age and BSA, and three SNPs in the VKORC1, CYP2C9 and CYP4F2 genes strongly predict outlier patients treated with acenocoumarol.


Assuntos
Acenocumarol/farmacologia , Anticoagulantes/farmacologia , Farmacogenética , Acenocumarol/administração & dosagem , Adulto , Idoso , Anticoagulantes/administração & dosagem , Hidrocarboneto de Aril Hidroxilases/genética , Sequência de Bases , Citocromo P-450 CYP2C9 , Sistema Enzimático do Citocromo P-450/genética , Família 4 do Citocromo P450 , Primers do DNA , Relação Dose-Resposta a Droga , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Oxigenases de Função Mista/genética , Polimorfismo de Nucleotídeo Único , Vitamina K Epóxido Redutases
7.
J Thromb Haemost ; 5(8): 1701-6, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17596133

RESUMO

BACKGROUND: The efficacy of oral anticoagulant therapy is largely conditioned by both environmental and genetic factors. OBJECTIVES: To attempt to define the genetic profile involved in the response to this treatment. PATIENTS AND METHODS: We selected 100 men younger than 75 years, with non-valvular atrial fibrillation, who started anticoagulation with acenocoumarol following the same protocol: 3 mg for three consecutive days. Then, doses were individually adjusted to achieve a steady International Normalized Ratio (INR). The basal plasma level and the level after 3 days were obtained, and the INR was determined. We studied five functional polymorphisms: FVII -323 Del/Ins, CYP2C*9, VKORC1 c1173t, calumenin (CALU) R4Q and CALU a29809g. The dose required for a steady INR was also recorded. RESULTS: Only the VKORC1 genotype had significant impact on the efficacy of therapy. Carriers of the 1173t allele were significantly more sensitive to therapy for 3 days [INR 2.07 (1.59-2.87) vs. 1.74 (1.30-2.09); P = 0.015] and they needed lower acenocoumarol doses to stabilize their INR (15.8 +/- 5.6 vs. 19.5 +/- 6.0 mg week(-1); P = 0.004). Its effect was exacerbated by combination with the CALU a29809g polymorphism. Carriers of both variants (27% of the sample) achieved the highest INR [2.26 (1.70-3.32)] and required the lowest dose (14.1 +/- 5.1 mg week(-1)). This genetic profile was particularly relevant in patients with INR >or= 3.5 at the start of therapy (P = 0.005; odds ratio = 6.67, 95% confidence interval = 1.32-37.43). CONCLUSIONS: Our results suggest that CALU a29809g might be a new genetic factor involved in the pharmacogenetics of anticoagulant therapy, and confirm that specific genetic profiles defined by different polymorphisms will determine the initial response and dose required to achieve a stable and safe INR.


Assuntos
Acenocumarol/farmacologia , Anticoagulantes/metabolismo , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Oxigenases de Função Mista/genética , Idoso , Proteínas Sanguíneas/metabolismo , Variação Genética , Humanos , Masculino , Pessoa de Meia-Idade , Modelos Biológicos , Modelos Genéticos , Farmacogenética , Polimorfismo Genético , Vitamina K/metabolismo , Vitamina K Epóxido Redutases
10.
Clín. investig. arterioscler. (Ed. impr.) ; 17(3): 122-129, mayo-jun. 2005. ilus, tab
Artigo em Es | IBECS | ID: ibc-037866

RESUMO

Introducción/objetivos. El factor XII desempeña un papel fundamental tanto en la coagulación como en la fibrinólisis; sin embargo, se desconoce la relevancia patológica de su deficiencia. El polimorfismo C46T, localizado en la región Kozak de este gen, se asocia con menores valores plasmáticos de factor XII al afectar la traducción del ARNm. Nuestro objetivo fue evaluar el papel de este polimorfismo en alteraciones trombóticas y hemorrágicas. Pacientes y métodos. Realizamos 3 estudios de casos y controles, que incluyeron a 55 pacientes con hemorragia intracraneal espontánea, 206 con trombosis venosa profunda, y 281 con infarto agudo de miocardio prematuro (infarto agudo de miocardio < 45). Los controles (n = 550) fueron sujetos sanos de la misma zona geográfica. La determinación del polimorfismo se realizó mediante reacción en cadena de la polimerasa y análisis de conformación de cadenas sencillas. El genotipo se confirmó mediante secuenciación. Resultados. El polimorfismo C46T no modifica significativamente el riesgo de presentar hemorragia intracraneal espontánea o trombosis venosa profunda. Sin embargo, el alelo 46T aparece como factor de riesgo débil para el desarrollo de infarto agudo de miocardio < 45 (odds ratio ajustada = 1,64; intervalo de confianza del 95%, 1,14-2,37; p = 0,008). Los portadores del alelo 46T mostraron valores de proteína C reactiva superiores (p = 0,002) y peor evolución que los controles. Además, encontramos un sinergismo entre la hipercolesterolemia y este polimorfismo. Finalmente, describimos una nueva variante genética en el gen que codifica el factor XII (C42T), infrecuente, cercana a la secuencia Kozak y posiblemente ligada a la variante 46C, pero que no parece afectar a los valores circulantes de factor XII. Conclusiones. El alelo 46T, asociado con menores valores plasmáticos de factor XII, parece predisponer al desarrollo de infarto agudo de miocardio prematuro, especialmente en pacientes hipercolesterolémicos


Introduction/Aim. Factor XII (FXII) plays a key role in both coagulation and fibrinolysis. However, the pathological importance of FXII deficiency remains unknown. The C46T polymorphism in the Kozak region of gene coding FXII is related to lower FXII plasma levels by mRNA translation alterations. Our aim was to assess the role of this polymorphism in thrombotic and hemorrhagic disease. Patients and methods. We performed 3 case-control studies including 55 patients with spontaneous intracranial hemorrhage (SIH), 206 with deep venous thrombosis (DVT) and 281 with premature myocardial infarction (AMI < 45). Controls (n = 550) were healthy subjects from the same geographic area. Polymorphism identification was performed by PCR and single-stranded conformational polymorphism (SSCP) analysis. Genotype was confirmed by sequencing. Results. The C46T polymorphism modifies neither the risk for SIH nor for DVT. Nevertheless, the 46T allele arises as a weak risk factor for the development of AMI < 45 (adjusted OR = 1.64; 95% CI, 1.14-2.37; p = 0.008). Allele 46T carriers showed higher CRP levels (p = 0.002) and worse outcome than controls. Moreover, we found a synergic interaction between hypercholesterolemia and this polymorphism. Finally, we described a new genetic variant in the gene coding FXII: C42T, found rarely, close to the Kozak sequence and possibly linked to the 46C variant, which, however, does not seem to affect FXII circulating levels


Assuntos
Adulto , Humanos , Fator XII/genética , Fator XII/fisiologia , Trombose/complicações , Trombose/fisiopatologia , Infarto do Miocárdio/epidemiologia , Infarto do Miocárdio/fisiopatologia , Hipercolesterolemia/complicações , Hipercolesterolemia/prevenção & controle , Polimorfismo Genético/genética , Polimorfismo Genético/fisiologia , Fator XII , Genótipo
12.
J Biol Regul Homeost Agents ; 18(2): 166-71, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15471222

RESUMO

Within the past decade our understanding of thromboembolic disorders has become even more sophisticated as recent discoveries have suggested the influence of gene variants on the development of atherosclerotic disease and arterial thrombosis. Candidate genes encode proteins involved in processes relevant to atherosclerosis, ranging from cholesterol metabolism to arterial thrombosis. Platelets are key elements in primary hemostasis, but also in arterial thrombosis. Moreover, a number of genetic polymorphisms of platelet proteins may also induce gain or loss of function, supporting a role predisposing some individuals to thrombotic events. However, after thousands of studies, much controversy remains whether individual platelet polymorphisms contribute to an increased likelihood of thromboembolic disorders. Although platelet polymorphisms are a promising addition to more established cardiovascular risk factors, identifying genetic variants as a single cause of cardiovascular disease would be an oversimplification; instead, the contribution of these polymorphisms should also be considered in the context of a multifactorial disease. Gene-gene and gene-environment studies would identify specific combinations associated with a high risk to suffer from these diseases. The platelet's genetic heterogeneity should also be considered in every aspect of clinical medicine, ranging from susceptibility to diseases, pathogenesis, and clinical outcome to diversity in responses to drug treatment (pharmacogenomics), and bleeding.


Assuntos
Plaquetas/fisiologia , Tromboembolia/genética , Anexina A5/genética , Antígenos CD/genética , Cromograninas , Subunidades alfa Gs de Proteínas de Ligação ao GTP/genética , Frequência do Gene , Glicoproteínas/genética , Humanos , Glicoproteínas de Membrana/genética , Modelos Biológicos , Selectina-P/genética , Polimorfismo Genético , Receptores de IgG/genética
13.
J Thromb Haemost ; 2(6): 931-9, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15140129

RESUMO

BACKGROUND: Missense mutations causing conformational alterations in serpins can be responsible for protein deficiency associated with human diseases. However, there are few data about conformational consequences of mutations affecting antithrombin, the main hemostatic serpin. OBJECTIVES: To investigate the conformational and clinical effect of mutations affecting the shutter region of antithrombin. PATIENTS AND METHODS: We identified two families with significant reduction of circulating antithrombin displaying early and severe venous thrombosis, frequently associated with pregnancy or infection. Mutations were determined by standard molecular methods. Biochemical studies were performed on plasma samples. One variant (P80S) was purified by heparin-affinity chromatography and gel filtration, and evaluated by proteomic analysis. Finally, we modelled the structure of the mutant dimer. RESULTS: We identified two missense mutations affecting the shutter region of antithrombin: P80S and G424R. Carriers of both mutations presented traces of a similar abnormal antithrombin, supporting inefficiently expressed rather than non-expressed variants. The abnormal antithrombin purified from P80S carriers is an inactive disulfide-linked dimer of mutant antithrombin whose properties are consistent with head-to-head insertion of the reactive loop. CONCLUSIONS: Our data support the conclusion that missense mutations affecting the shutter region of serpins have specific conformational effects resulting in the formation of mutant oligomers. The consequent inefficiency of secretion explains the accompanying deficiency and loss of function, but the severity of thrombosis associated with these mutations suggests that the oligomers also have new and undefined pathological properties that could be exacerbated by pregnancy or infection.


Assuntos
Deficiência de Antitrombina III/genética , Antitrombina III/genética , Dissulfetos , Mutação de Sentido Incorreto , Trombose Venosa/genética , Adulto , Idoso , Antitrombina III/química , Antitrombina III/isolamento & purificação , Deficiência de Antitrombina III/complicações , Análise Mutacional de DNA , Dimerização , Saúde da Família , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Linhagem , Conformação Proteica , Serpinas/química , Serpinas/genética , Espanha , Trombose Venosa/sangue
14.
Br J Haematol ; 125(5): 621-8, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15147378

RESUMO

A polymorphism of the gene encoding the extra-large stimulatory G-protein alpha-subunit (XLalphas), originally identified in three patients with a bleeding tendency, involved a 36-bp insertion and two missense changes. A paternally-inherited insertion displayed a moderate platelet Gsalpha over-expression, which lead to platelet hypo-reactivity. These data prompted us to investigate the genetic, functional and clinical relevance of this polymorphism in the Mediterranean population. We included 414 healthy subjects and three case/control studies: 263 consecutive patients with a first episode of primary intracerebral haemorrhage, 195 patients with deep venous thrombosis, and 104 patients with cerebrovascular disease. Controls were selected by approximating criteria to match selected risk factors to patients. Moreover, we performed studies of platelet function. We developed a simple method to determine the methylated allele, by digestion of genomic DNA with Sma I before polymerase chain reaction amplification. We identified two new rare variants, resulting from the loss of repeat units 7 and 5. The AB genotype was present in 3.6% of healthy population and the prevalence of the B allele was similar among cases and controls. Accordingly, the non-methylated B allele did not modify either the expression of platelet Gsalpha or the platelet response to Gs-agonists. Thus, our study suggests a minor functional role of XLalphas polymorphism in thrombotic or in haemorrhagic disorders.


Assuntos
Subunidades alfa Gs de Proteínas de Ligação ao GTP/genética , Hemorragia/genética , Polimorfismo Genético/genética , Trombose/genética , Idoso , Plaquetas/fisiologia , AMP Cíclico/metabolismo , Metilação de DNA , Feminino , Frequência do Gene , Genótipo , Humanos , Masculino , Região do Mediterrâneo , Pessoa de Meia-Idade , Linhagem , Fatores de Risco
15.
Transfusion ; 44(5): 771-6, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15104661

RESUMO

BACKGROUND: Alloimmune incompatibility in allo-geneic stem cell transplantation (ASCT), pregnancy, and blood transfusion might trigger an immune response with clinical consequences. Human PLT antigens (HPAs), which play a significant role in pregnancy or blood transfusion-associated alloimmune thrombocytopenia, are also expressed on the surface of tissues affected by GVHD. Thus, HPA mismatch in HLA-identical ASCT could play a potential role in PLT engraftment and GVHD. STUDY DESIGN AND METHODS: We studied the HPA-1, -2, and -5 genotypes in Caucasian donors and patients involved in 77 HLA-identical ASCTs. We evaluated the association of HPA compatibility with clinical outcome, analyzing the relevance of host-versus-donor HPA incompatibility in PLT engraftment and donor-versus-host HPA incompatibility in GVHD. RESULTS: PLT engraftment and transfusion require-ments were similar in HPA-compatible and HPA-incompatible ASCT. Cases with severe thrombocytopenia or significant delayed PLT engraftment did not display host-versus-donor HPA incompatibility. Moreover, the incidence of GVHD did not correlate with HPA compatibility. CONCLUSION: Our results support no role for these antigens in immune complications of ASCT: PLT engraftment, requirement of PLT transfusions, and GVHD.


Assuntos
Antígenos de Plaquetas Humanas/imunologia , Doença Enxerto-Hospedeiro/etiologia , Transplante de Células-Tronco de Sangue Periférico/efeitos adversos , Transfusão de Plaquetas , Adolescente , Adulto , Idoso , Antígenos de Plaquetas Humanas/análise , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Transplante Homólogo
16.
Blood Coagul Fibrinolysis ; 13(2): 95-103, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11914651

RESUMO

Activation of the prothrombinase complex, which catalyzes the formation of thrombin from prothrombin, is crucial for the (patho)physiological processes of hemostasis and thrombosis. We here report that washed platelets supplemented with prothrombin can be irreversibly aggregated with otherwise non-aggregant doses of adenosine diphosphate (10 micromol/l), thrombin (0.06 U/ml), or collagen (1 microg/ml). Prothrombinase-catalyzed prothrombin to thrombin conversion most probably supports this aggregation response, since inhibitors of thrombin (hirudin or heparin) and an inhibitor of activated factor X (DX-9065a) impair the response. A certain degree of agonist-induced platelet activation seems to be required for this prothrombin-supported aggregation response, since prothrombin alone does not induce aggregation, and blockade of glycoprotein Ia/IIa with a specific antibody inhibits the platelet aggregation response to collagen and prothrombin. These results may suggest that activation of the prothrombinase complex could be a common step of the platelet response to distinct agonists, which may be achieved at low levels of platelet stimulation.


Assuntos
Agregação Plaquetária/efeitos dos fármacos , Tromboplastina/metabolismo , Difosfato de Adenosina/farmacologia , Animais , Bovinos , Colágeno/farmacologia , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Ativação Enzimática/efeitos dos fármacos , Fator X/farmacologia , Fator Xa/farmacologia , Humanos , Integrina alfa2beta1 , Protrombina/metabolismo , Protrombina/farmacologia , Receptores de Colágeno/antagonistas & inibidores , Trombina/farmacologia , Tromboplastina/efeitos dos fármacos
17.
Cephalalgia ; 21(8): 837-41, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11737010

RESUMO

At present, it is contradictory to determine if the combination of certain prothrombotic polymorphisms and migraine increases the risk to develop ischaemic cerebrovascular disease. Recently, the common Val34Leu polymorphism of the A-chain factor XIII gene, associated with variations in factor XIII activity, has been suggested to play a significant role in the development of arterial and venous thrombotic disorders. We analysed the prevalence of this polymorphism in 17 patients with coexisting ischaemic cerebrovascular disease and migraine (5 with aura, and 12 without aura), 89 patients with migraine (43 with aura, and 46 without aura), 116 patients with ischaemic cerebrovascular disease, and 467 healthy Caucasian controls from the South of Spain. Genomic PCR amplification, using a mutated oligonucleotide, and allele-specific restriction assays were used for genotyping. The factor XIII Leu 34 variant was present in 47.1; 40.5; 34.9; and 35.1% of patients with coexisting ischaemic cerebrovascular disease and migraine, ischaemic cerebrovascular disease, migraine, and control subjects, respectively. These data suggest that the factor XIII Leu 34 allele does not play a protective role against these disorders in our population.


Assuntos
Transtornos Cerebrovasculares/genética , Fator XIII/genética , Transtornos de Enxaqueca/genética , Adulto , Idoso , Transtornos Cerebrovasculares/sangue , Transtornos Cerebrovasculares/complicações , Feminino , Genótipo , Humanos , Leucina , Masculino , Pessoa de Meia-Idade , Transtornos de Enxaqueca/sangue , Transtornos de Enxaqueca/complicações , Reação em Cadeia da Polimerase , Polimorfismo Genético , Valina
18.
Thromb Haemost ; 85(4): 686-93, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11341506

RESUMO

The interaction of lipoprotein(a) [Lp(a)] with platelets is not well defined, particularly with regards to the individual contribution of the protein components of Lp(a), the apo B-100 and the apolipoprotein apo(a). This study investigated the binding of different recombinant apo(a) [r-apo(a)] isoforms, to human platelets and its effect on platelet aggregation. Scatchard analysis of saturation binding experiments demonstrated that human platelets display a single class of high affinity r-apo(a) binding sites (71 +/- 46 molec./platelet, Kd = 5.6 +/- 2.0 nmol/L). Platelet activation with strong agonists (thrombin, arachidonic acid) increased 2- to 10-fold the r-apo(a) binding, without affecting the affinity. Competition assays showed that the binding sites are highly specific for r-apo(a) and Lp(a). At high concentration t-PA could also bind to the r-apo(a) binding sites. By contrast, neither fibrinogen nor plasminogen inhibited to the r-apo(a) binding. The lysine analogue EACA inhibits the binding of r-apo(a) to platelets, thus suggesting the involvement of lysine residues in that interaction. Moreover, the r-apo(a) binding to platelets is unlikely mediated by GPIIb/IIIa-attached fibrin since it is not affected by platelet treatment with either LJ-CP8, a monoclonal antibody that specifically blocks fibrinogen binding to GPIIb/IIIa, nor GPRP, an inhibitor of fibrin polymerisation. Finally, we show that the distinct recombinant apo(a) proteins, as well as native Lp(a), promote an aggregation response of platelets to otherwise subaggregant doses of arachidonic acid. This proaggregant effect of r-apo(a) is dependent on its binding to platelets since it requires a minimum incubation time, and it is prevented by EACA at concentration inhibiting the r-apo(a)-platelet interaction. These results suggest that the prothrombotic action of Lp(a) may be in part mediated by modulating the platelet function through the interaction of its apo(a) subunit with a specific receptor at the platelet surface.


Assuntos
Apolipoproteínas/metabolismo , Lipoproteína(a)/metabolismo , Agregação Plaquetária/efeitos dos fármacos , Difosfato de Adenosina/farmacologia , Sequência de Aminoácidos , Ácido Aminocaproico/farmacologia , Apolipoproteínas/genética , Apolipoproteínas/farmacologia , Apoproteína(a) , Ácido Araquidônico/farmacologia , Ligação Competitiva , Colágeno/farmacologia , Fibrinogênio/farmacologia , Humanos , Lipoproteína(a)/genética , Lipoproteína(a)/farmacologia , Dados de Sequência Molecular , Plasminogênio/farmacologia , Ligação Proteica , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/farmacologia , Proteínas/farmacologia , Receptores de Trombina , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Trombina/farmacologia , Ativador de Plasminogênio Tecidual/metabolismo , Ativador de Plasminogênio Tecidual/farmacologia
19.
Blood ; 97(10): 2979-82, 2001 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-11342420

RESUMO

Intracranial hemorrhage is the third most frequent cause of cerebrovascular disease, but few genetic risk factors have been associated with its development. Recently, it has been reported that some polymorphisms that affect clotting factors increase the risk for thrombosis. However, reports have analyzed the effect of polymorphisms influencing the hemostatic state in bleeding disorders insufficiently. A case-control study was conducted of 201 patients with spontaneous intracranial hemorrhage and 201 control subjects matched for age, race, sex, and selected risk factors (hypertension, smoking, and alcohol consumption). Genomic polymerase chain reaction was used to analyze the prevalence of 4 polymorphisms: factor V Leiden, prothrombin 20210A, factor VII-323 Del/Ins of a decanucleotide, and factor XIII V34L. Subjects with factor V Leiden had decreased risk for spontaneous intracranial hemorrhage (odds ratio, 0.19; 95% confidence interval, 0.03-0.95). The frequency of the prothrombin 20210A/G genotype was also lower among patients than controls (1.5% vs 3%, respectively). Moreover, carriers of the -323 Ins allele of factor VII had a 1.54-fold risk for intracranial hemorrhage (95% CI, 1.03-2.72). Finally, no significant differences were observed in the prevalence of factor XIII V34L polymorphism between patients and controls. Therefore, new genetic factors affecting the risk for spontaneous intracranial hemorrhage were identified. These data, together with the relevance of these polymorphisms in thrombotic diseases, support the idea that a polymorphism may play opposite roles in thrombosis and hemorrhage, suggesting an explanation for the high frequency of these polymorphisms in the general population.


Assuntos
Fatores de Coagulação Sanguínea/genética , Predisposição Genética para Doença , Hemorragias Intracranianas/genética , Mutação , Idoso , Estudos de Casos e Controles , Fator V/genética , Fator VII/genética , Fator XIII/genética , Feminino , Frequência do Gene , Genótipo , Humanos , Masculino , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase , Protrombina/genética , Fatores de Risco , Trombose/genética
20.
Br J Haematol ; 115(4): 969-76, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11843835

RESUMO

P-selectin glycoprotein ligand (PSGL-1) shares common features with platelet glycoprotein Ibalpha. A recently described polymorphism in this receptor that results in a variable number of tandem repeats (VNTR) sequence present either 16, 15 or 14 times (alleles A, B or C) could, similar to GPIbalpha, be functionally relevant. The allelic frequency of this polymorphism was investigated in 469 individuals from the south of Spain, and was similar to that previously described in other Caucasian populations: 85% A, 14% B and 1% C alleles. We identified two new polymorphisms genetically linked to the C isoform, resulting in the Ser273Phe and Met274Val changes. To assess the functional consequence of the polymorphisms in the receptor, we performed flow cytometric analysis of P-selectin dependent neutrophil-platelet interaction. Neutrophils carrying the shortest C allele and the amino acid variations in residues 273 and 274 exhibited a significantly lower capacity to bind activated platelets than A/B and A/A samples (mean fluorescence intensity of CD42b+ neutrophils 262 versus 303 and 319 respectively, P < 0.05). The distribution of the VNTR was analysed in three case-control studies including 104 cerebrovascular (CVD), 101 coronary heart disease (CHD) and 150 deep venous thrombosis (DVT) patients. The results showed that smaller (B and C) alleles seem to be associated with a lower risk of developing CVD (P = 0.008) but not to be related to CHD or DVT. In conclusion, polymorphisms of the PSGL-1 receptor may influence the neutrophil-platelet binding, and represent a risk factor for CVD.


Assuntos
Plaquetas/fisiologia , Glicoproteínas de Membrana/genética , Repetições Minissatélites , Neutrófilos/fisiologia , Trombose Venosa/genética , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Estudos de Casos e Controles , Adesão Celular , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Isquemia Miocárdica/genética , Adesividade Plaquetária , Prevalência , Estudos Retrospectivos , Análise de Sequência de DNA , Acidente Vascular Cerebral/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...