Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 750
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Gastroenterology ; 160(3): 771-780.e4, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33098885

RESUMO

BACKGROUND AND AIMS: The largest cause of mortality in patients with inflammatory bowel disease (IBD) remains thromboembolic disease (TED). Recent reports have demonstrated that both monogenic and polygenic factors contribute to TED and 10% of healthy subjects are genetically at high risk for TED. Our aim was to utilize whole-exome sequencing and genome-wide genotyping to determine the proportion of IBD patients genetically at risk for TED and investigate the effect of genetic risk of TED in IBD. METHODS: The TED polygenic risk score was calculated from genome-wide genotyping. Thrombophilia pathogenic variants were extracted from whole-exome sequencing. In total, 792 IBD patients had both whole-exome sequencing and genotyping data. We defined patients at genetically high risk for TED if they had a high TED polygenic risk score or carried at least 1 thrombophilia pathogenic variant. RESULTS: We identified 122 of 792 IBD patients (15.4%) as genetically high risk for TED. Among 715 of 792 subjects whose documented TED status were available, 63 of the 715 patients (8.8%) had TED events. Genetic TED risk was significantly associated with increased TED event (odds ratio, 2.5; P = .0036). In addition, we confirmed an additive effect of monogenic and polygenic risk on TED (P = .0048). Patients with high TED genetic risk more frequently had thrombosis at multiple sites (78% vs 42%, odds ratio, 3.96; P = .048). CONCLUSIONS: Genetic risk (both poly- and monogenic) was significantly associated with TED history. Our results suggest that genetic traits identify approximately 1 in 7 patients with IBD who will experience 2.5-fold or greater risk for TED.


Assuntos
Predisposição Genética para Doença , Doenças Inflamatórias Intestinais/complicações , Tromboembolia/epidemiologia , Adulto , Idoso , Estudos de Casos e Controles , Feminino , Testes Genéticos/métodos , Testes Genéticos/estatística & dados numéricos , Voluntários Saudáveis , Humanos , Doenças Inflamatórias Intestinais/genética , Masculino , Pessoa de Meia-Idade , Herança Multifatorial , Prevalência , Medição de Risco/métodos , Medição de Risco/estatística & dados numéricos , Fatores de Risco , Tromboembolia/genética , Sequenciamento do Exoma
2.
J Pediatr ; 238: 259-267.e2, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34245770

RESUMO

OBJECTIVE: To clarify the incidence and genetic risk of neonatal-thromboembolism, we conducted a nationwide study exploring the impact of thrombophilia on neonatal-thromboembolism in Japan. STUDY DESIGN: A questionnaire survey was conducted for perinatal centers in Japan, focusing on the clinical expression, genotype, treatment, and outcome of patients who developed thromboembolism within 28 days of birth from 2014 to 2018. RESULTS: The estimated incidence of neonatal-thromboembolism was 0.39 cases per 10 000 live births. Intracranial lesions and purpura fulminans occurred in 66 and 5 of 77 patients, respectively. Fifty-eight (75.3%) infants presented within 3 days after birth. Four (5.2%) died, and 14 (18.2%) survived with disability. At the diagnosis, <20% plasma activity of protein C was noted in 16 infants, protein S (in 2), and antithrombin (in 1). Thirteen genetic tests identified 4 biallelic and 5 monoallelic protein C-variants but no protein S- or antithrombin-variants. Protein C-variants had purpura fulminans (P < .01), ocular bleeding (P < .01), positive-family history (P = .01), and death or disability (P = .03) more frequently than others. Protein C-variants were independently associated with disability (OR 5.74, 95% CI 1.16-28.4, P = .03) but not death. Four biallelic variants had serious thrombotic complications of neurologic disability, blindness, and/or amputation. Three monoallelic variants survived without complications. The only protein C-variant death was an extremely preterm heterozygote infant. CONCLUSIONS: Monoallelic protein C-variants had a higher incidence of neonatal-thromboembolism than biallelic variants. Thrombophilia genetic testing should be performed in the setting of neonatal-thromboembolism and low protein C to identify the underlying genetic defect.


Assuntos
Deficiência de Proteína C/complicações , Tromboembolia/epidemiologia , Feminino , Predisposição Genética para Doença , Humanos , Recém-Nascido , Japão , Masculino , Deficiência de Proteína C/genética , Estudos Retrospectivos , Fatores de Risco , Inquéritos e Questionários , Tromboembolia/genética
3.
Blood ; 143(17): 1686-1687, 2024 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-38662387
4.
Blood ; 131(17): 1903-1909, 2018 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-29483100

RESUMO

Combinations of proinflammatory and procoagulant reactions are the unifying principle for a variety of disorders affecting the cardiovascular system. The factor XII-driven contact system starts coagulation and inflammatory mechanisms via the intrinsic pathway of coagulation and the bradykinin-producing kallikrein-kinin system, respectively. The biochemistry of the contact system in vitro is well understood; however, its in vivo functions are just beginning to emerge. Challenging the concept of the coagulation balance, targeting factor XII or its activator polyphosphate, provides protection from thromboembolic diseases without interfering with hemostasis. This suggests that the polyphosphate/factor XII axis contributes to thrombus formation while being dispensable for hemostatic processes. In contrast to deficiency in factor XII providing safe thromboprotection, excessive FXII activity is associated with the life-threatening inflammatory disorder hereditary angioedema. The current review summarizes recent findings of the polyphosphate/factor XII-driven contact system at the intersection of procoagulant and proinflammatory disease states. Elucidating the contact system offers the exciting opportunity to develop strategies for safe interference with both thrombotic and inflammatory disorders.


Assuntos
Angioedemas Hereditários/metabolismo , Fator XII/metabolismo , Polifosfatos/metabolismo , Tromboembolia/metabolismo , Trombose/metabolismo , Angioedemas Hereditários/genética , Angioedemas Hereditários/patologia , Fator XII/genética , Humanos , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Tromboembolia/genética , Tromboembolia/patologia , Trombose/genética , Trombose/patologia
5.
Clin Sci (Lond) ; 134(2): 169-192, 2020 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-31971230

RESUMO

Stroke is the leading cause of serious disability in the world and a large number of ischemic strokes are due to thromboembolism from unstable carotid artery atherosclerotic plaque. As it is difficult to predict plaque rupture and surgical treatment of asymptomatic disease carries a risk of stroke, carotid disease continues to present major challenges with regard to clinical decision-making and revascularization. There is therefore an imminent need to better understand the molecular mechanisms governing plaque instability and rupture, as this would allow for the development of biomarkers to identify at-risk asymptomatic carotid plaque prior to disease progression and stroke. Further, it would aid in creation of therapeutics to stabilize carotid plaque. MicroRNAs (miRNAs) have been implicated as key protagonists in various stages of atherosclerotic plaque initiation, development and rupture. Notably, they appear to play a crucial role in carotid artery thromboembolism. As the molecular pathways governing the role of miRNAs are being uncovered, we are learning that their involvement is complex, tissue- and stage-specific, and highly selective. Notably, miRNAs can be packaged and secreted in extracellular vesicles (EVs), where they participate in cell-cell communication. The measurement of EV-encapsulated miRNAs in the circulation may inform disease mechanisms occurring in the plaque itself, and therefore may serve as sentinels of unstable plaque as well as therapeutic targets.


Assuntos
Artérias Carótidas/patologia , MicroRNAs/metabolismo , Tromboembolia/genética , Animais , Biomarcadores/metabolismo , Vesículas Extracelulares/metabolismo , Regulação da Expressão Gênica , Humanos , MicroRNAs/genética
6.
J Clin Pharm Ther ; 45(3): 547-560, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32168383

RESUMO

WHAT IS KNOWN AND OBJECTIVE: Despite an apparently sound pharmacological basis, clinical studies of genotype-guided warfarin dosing have yielded mixed and conflicting results, leading to reluctance in its clinical implementation. The objective of this critique is to re-evaluate key warfarin pharmacogenetic studies with a view to explaining why this may be so. METHODS: Major widely-cited warfarin pharmacogenetic studies as well as recent meta-analyses were identified and a critical analysis of these was undertaken to identify factors that may account for poor clinical implementation of pre-treatment genotyping. RESULTS AND DISCUSSION: Critical examination of major warfarin pharmacogenetic studies identified a number of methodological concerns such as marked variations in study designs with different variously-defined measures of outcome. Genotype testing involved only a limited number of CYP2C9/VKORC1 alleles. Claims of benefits of genotyping are based almost exclusively on INR-related parameters which are known to be highly time-labile and of limited value in predicting clinical risk or benefit. This is evidenced by lack of any significant effect of genotyping on rates of bleeding or thromboembolic events. Neither have the effects of potential phenoconversion or medication non-adherence in study populations been adequately investigated. Although the effect of ethnicity/race is now better characterised, studies lack the power to determine whether any benefits claimed are indication-sensitive. WHAT IS NEW AND CONCLUSION: Since 60% of inter-individual variability in warfarin dose/response is due to other factors (many of which are non-genetic), expectations of eliminating this variability simply by CYP2C9/VKORC1 genotyping are over-optimistic and efforts cost-ineffective. Real-world studies have not always corroborated trials-based claims of clinical benefit. It is time to consider redirecting scarce resources away from the study of warfarin pharmacogenetics to pharmacogenetic research of potentially greater clinical relevance.


Assuntos
Anticoagulantes/uso terapêutico , Tromboembolia/tratamento farmacológico , Varfarina/uso terapêutico , Genótipo , Humanos , Coeficiente Internacional Normatizado , Farmacogenética , Tromboembolia/genética
7.
Int J Mol Sci ; 20(3)2019 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-30759794

RESUMO

Chronic Thromboembolic Pulmonary Hypertension (CTEPH) is a debilitating disease, for which the underlying pathophysiological mechanisms have yet to be fully elucidated. Occurrence of a pulmonary embolism (PE) is a major risk factor for the development of CTEPH, with non-resolution of the thrombus being considered the main cause of CTEPH. Polymorphisms in the α-chain of fibrinogen have been linked to resistance to fibrinolysis in CTEPH patients, and could be responsible for development and disease progression. However, it is likely that additional genetic predisposition, as well as genetic and molecular alterations occurring as a consequence of tissue remodeling in the pulmonary arteries following a persistent PE, also play an important role in CTEPH. This review summarises the current knowledge regarding genetic differences between CTEPH patients and controls (with or without pulmonary hypertension). Mutations in BMPR2, differential gene and microRNA expression, and the transcription factor FoxO1 have been suggested to be involved in the processes underlying the development of CTEPH. While these studies provide the first indications regarding important dysregulated pathways in CTEPH (e.g., TGF-ß and PI3K signaling), additional in-depth investigations are required to fully understand the complex processes leading to CTEPH.


Assuntos
Hipertensão Pulmonar/genética , Tromboembolia/genética , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/genética , Doença Crônica , Progressão da Doença , Humanos , Embolia Pulmonar/genética , Transdução de Sinais/genética
8.
Angiol Sosud Khir ; 25(4): 28-33, 2019.
Artigo em Russo | MEDLINE | ID: mdl-31855198

RESUMO

BACKGROUND: Diabetes mellitus a commonly encountered pathology in the whole world and has a tendency towards steady growth of morbidity and development of vascular complications. The presence of haemostatic disorders and genetic susceptibility to thrombosis in patients with diabetes mellitus increases the risk for the development of thrombotic complications. AIM: The purpose of the study was to determine the incidence rate of thrombophilic conditions (TC) and thrombosis-associated gene carrier status (TAGCS) in patients suffering from neuroischaemic form of diabetic foot. PATIENTS AND METHODS: The study enrolled a total of 38 patients undergoing treatment at the Department of Vascular Surgery for critical ischaemia of lower limbs combined with diabetes mellitus during the period from 2016 to 2018. There were 29 (76.3%) men and 9 (23.7%) women. The mean age amounted to 58.9±7.3 years. The diagnosis of TC and TAGCS was made based on the study for the presence of the markers of antiphospholipid syndrome, level of homocysteine and antithrombin III, proteins C and S, as well as comprehensive genetic study in order to reveal gene polymorphisms (prothrombin, Leiden mutation, MTHFR gene, MTR gene and others). Based on the obtained findings we calculated the incidence rate of TC and TAGCS, as well as their combinations in the examined patients. RESULTS: In the studied group of patients we revealed various incidence of TC and TAGCS, and, most importantly, that of a combination of these conditions. All cases of thrombophilias were combined with TAGCS. Hyperhomocysteinemia was most commonly combined with the MTRR 66 A>G gene mutation, the presence of lupus anticoagulant - with PAI-1 675 5G>4G, whereas thrombophilic conditions - with MTRR 66 A>G and PAI-I - 675 5G>4G. Two patients were found to be carriers of the factor V Leiden mutation. CONCLUSION: The examined patients were diagnosed as having TC or TAGCS, as well as their combinations in one form or another, thus increasing the risk for the development of thromboses and embolisms in such patients.


Assuntos
Complicações do Diabetes/complicações , Pé Diabético/etiologia , Tromboembolia/etiologia , Trombofilia/complicações , Idoso , Complicações do Diabetes/genética , Pé Diabético/genética , Feminino , Heterozigoto , Humanos , Masculino , Pessoa de Meia-Idade , Prevalência , Fatores de Risco , Tromboembolia/genética , Trombofilia/genética
9.
Arterioscler Thromb Vasc Biol ; 37(1): 13-20, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27834692

RESUMO

Coagulation factor XII (FXII, Hageman factor) is a plasma protease that in its active form (FXIIa) initiates the procoagulant and proinflammatory contact system. This name arises from FXII's unique mechanism of activation that is induced by binding (contact) to negatively charged surfaces. Various substances have the capacity to trigger FXII contact-activation in vivo including mast cell-derived heparin, misfolded protein aggregates, collagen, nucleic acids, and polyphosphate. FXII deficiency is not associated with bleeding, and for decades, the factor was considered to be dispensable for coagulation in vivo. However, despite the fact that humans and animals with deficiency in FXII have a normal hemostatic capacity, animal models revealed a critical role of FXIIa-driven coagulation in thromboembolic diseases. In addition to its role in thrombosis, FXIIa contributes to inflammation through the activation of the inflammatory bradykinin-producing kallikrein-kinin system. Pharmacological inhibition of FXII/FXIIa interferes with thrombosis and inflammation in animal models. Thus, targeting the FXIIa-driven contact system seems to be a promising and safe therapeutic anticoagulation treatment strategy, with additional anti-inflammatory effects. Here, we discuss novel functions of FXIIa in cardiovascular thrombotic and inflammatory disorders.


Assuntos
Anti-Inflamatórios/farmacologia , Coagulação Sanguínea/efeitos dos fármacos , Fator XII/antagonistas & inibidores , Fibrinolíticos/uso terapêutico , Mediadores da Inflamação/antagonistas & inibidores , Inflamação/tratamento farmacológico , Sistema Calicreína-Cinina/efeitos dos fármacos , Tromboembolia/tratamento farmacológico , Animais , Modelos Animais de Doenças , Fator XII/genética , Fator XII/metabolismo , Fator XIIa/antagonistas & inibidores , Fator XIIa/metabolismo , Predisposição Genética para Doença , Humanos , Inflamação/sangue , Inflamação/genética , Inflamação/imunologia , Mediadores da Inflamação/sangue , Camundongos Knockout , Terapia de Alvo Molecular , Tromboembolia/sangue , Tromboembolia/genética , Tromboembolia/imunologia
10.
Klin Monbl Augenheilkd ; 235(1): 81-86, 2018 Jan.
Artigo em Alemão | MEDLINE | ID: mdl-27643601

RESUMO

BACKGROUND: The potential impact of elevated Lipoprotein (a) [Lp(a)] levels on retinal venous occlusive (RVO) diseases with regard to age and various risk factors has not been studied extensively. PATIENTS AND METHODS: In a retrospective case-control study, thrombophilia data of 106 young patients (< 60 years at the time of the RVO or a previous thromboembolic event) with RVO and 76 healthy subjects were evaluated. RESULTS: Elevated Lp(a) plasma levels were significantly more prevalent among RVO patients (29.2 %) than among controls (9.2 %; p = 0.0009). Lp(a) levels were found to be significantly (p = 0.012) different between patients and controls. Moreover, we found that an unusual personal or family history of thromboembolism was a strong predictor of elevated Lp(a) (p = 0.03). We observed a significant correlation between elevated Lp(a) and other coagulation disorders (p = 0.005). Multivariate analysis showed that elevated lipoprotein(a) levels (OR: 3.5; p = 0.003) were an independent risk factor for the development of RVO. CONCLUSIONS: Elevated plasma levels of Lp(a) are associated with the development of RVO. Selective screening of young patients and subjects with a personal or family history of thromboembolism may be helpful in identifying RVO patients with elevated Lp(a).


Assuntos
Lipoproteína(a)/sangue , Oclusão da Veia Retiniana/sangue , Adulto , Estudos de Casos e Controles , Estudos Transversais , Feminino , Predisposição Genética para Doença/genética , Humanos , Masculino , Pessoa de Meia-Idade , Valores de Referência , Oclusão da Veia Retiniana/genética , Estudos Retrospectivos , Fatores de Risco , Tromboembolia/sangue , Tromboembolia/genética , Trombofilia/sangue , Trombofilia/genética
12.
Circulation ; 133(15): 1449-57; discussion 1457, 2016 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-26939588

RESUMO

BACKGROUND: ABO blood groups have been shown to be associated with increased risks of venous thromboembolic and arterial disease. However, the reported magnitude of this association is inconsistent and is based on evidence from small-scale studies. METHODS AND RESULTS: We used the SCANDAT2 (Scandinavian Donations and Transfusions) database of blood donors linked with other nationwide health data registers to investigate the association between ABO blood groups and the incidence of first and recurrent venous thromboembolic and arterial events. Blood donors in Denmark and Sweden between 1987 and 2012 were followed up for diagnosis of thromboembolism and arterial events. Poisson regression models were used to estimate incidence rate ratios as measures of relative risk. A total of 9170 venous and 24 653 arterial events occurred in 1 112 072 individuals during 13.6 million person-years of follow-up. Compared with blood group O, non-O blood groups were associated with higher incidence of both venous and arterial thromboembolic events. The highest rate ratios were observed for pregnancy-related venous thromboembolism (incidence rate ratio, 2.22; 95% confidence interval, 1.77-2.79), deep vein thrombosis (incidence rate ratio, 1.92; 95% confidence interval, 1.80-2.05), and pulmonary embolism (incidence rate ratio, 1.80; 95% confidence interval, 1.71-1.88). CONCLUSIONS: In this healthy population of blood donors, non-O blood groups explain >30% of venous thromboembolic events. Although ABO blood groups may potentially be used with available prediction systems for identifying at-risk individuals, its clinical utility requires further comparison with other risk markers.


Assuntos
Sistema ABO de Grupos Sanguíneos/análise , Arteriopatias Oclusivas/epidemiologia , Doadores de Sangue/estatística & dados numéricos , Tromboembolia/epidemiologia , Sistema ABO de Grupos Sanguíneos/genética , Adulto , Arteriopatias Oclusivas/genética , Dinamarca/epidemiologia , Feminino , Seguimentos , Humanos , Incidência , Masculino , Pessoa de Meia-Idade , Gravidez , Complicações Cardiovasculares na Gravidez/epidemiologia , Complicações Cardiovasculares na Gravidez/genética , Embolia Pulmonar/epidemiologia , Embolia Pulmonar/genética , Recidiva , Análise de Regressão , Risco , Suécia/epidemiologia , Tromboembolia/genética , Trombofilia/genética , Trombose Venosa/epidemiologia , Trombose Venosa/genética , Adulto Jovem
13.
Blood Cells Mol Dis ; 67: 18-22, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28010922

RESUMO

Venous thrombosis (VTE) in children is increasingly diagnosed, as advanced medical care has increased treatment intensity of hospitalized pediatric patients. The aim of this review was to summarize the data available and to discuss the controversial issue of thrombophilia screening in the light of the pediatric data available. Follow-up data for VTE recurrence in children suggest a recurrence rate between 3% (neonates) and 21% in individuals with unprovoked VTE. Apart from underlying medical conditions, recently reported systematic reviews on pediatric VTE (70% provoked) have shown significant associations between thrombosis and presence of protein C-, protein S- and antithrombin deficiency, factor 5 (F5: rs6025), factor 2 (F2: rs1799963), even more pronounced when combined inherited thrombophilias [IT] were involved. The F2 mutation, protein C-, protein S-, and antithrombin deficiency did also play a significant role at VTE recurrence. Although we have learned more about the pathophysiology of VTE with the increased discovery of IT evidence is still lacking as to whether IT influence the clinical outcome in pediatric VTE. It still remains controversial as to whether children with VTE or offspring from thrombosis-prone families benefit from IT screening. Thus, IT testing in children should be individualized.


Assuntos
Tromboembolia/epidemiologia , Tromboembolia Venosa/epidemiologia , Adolescente , Criança , Estudo de Associação Genômica Ampla , Hemostasia , Humanos , Recidiva , Fatores de Risco , Tromboembolia/sangue , Tromboembolia/genética , Tromboembolia/patologia , Trombofilia/sangue , Trombofilia/complicações , Trombofilia/genética , Trombofilia/patologia , Trombose/sangue , Trombose/epidemiologia , Trombose/genética , Trombose/patologia , Tromboembolia Venosa/sangue , Tromboembolia Venosa/genética , Tromboembolia Venosa/patologia
14.
Pharmacogenomics J ; 17(4): 331-336, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-27001121

RESUMO

Single-nucleotide polymorphisms (SNPs) related to hereditary thrombophilia were investigated as risk factors for thromboembolism in cancer patients. Their effect in metastatic colorectal cancer (mCRC) has never been explored so far. Our aim was to analyse the effect of coagulation factor V (FVL G1691A), prothrombin (PT G20210A), methylenetetrahydrofolate reductase (MTHFR C677T and A1298C) and plasminogen activator inhibitor type 1 (PAI-1 5G/4G) allelic variants in this setting. Fifty-two patients treated with first-line chemotherapy plus bevacizumab who developed a thromboembolic event in their lifetime were initially genotyped. A contemporary cohort of 127 patients who did not experience any thromboembolic event was also analysed. DNA was extracted from peripheral blood and genotypes were determined by real-time PCR, using LightSNiP (TIB MOLBIOL) on LightCcler 480 (Roche). The association between thromboembolism and SNPs was investigated by univariable and multivariable analyses. All SNPs were in Hardy-Weinberg equilibrium (χ2 test P>0.20). FVL G1691A and PT G20210A were present only in heterozygosis in 4 (2.2%) and 7 (3.9%) patients, respectively; MTHFR C677T in homozygosis in 29 (16.2%), MTHFR A1298C in homozygosis in 13 (7.3%); PAI-1 5G/4G in 98 (54.7%) and 4G/4G in 41 (23%) patients. At univariable analysis, treatment duration was significantly associated with thromboembolism (P<0.001), whereas gender, age, obesity, platelets count and chemotherapy backbone were not. Similarly, FVL G1691A and PT G20210A as well as MTHFR C677T and PAI-1 4G allele were significantly associated, whereas MTHFR A1298C was not. At multivariable model including PT G20210A, MTHFR C677T and PAI-1 4G (age, obesity, treatment duration and chemotherapy backbone were included as adjustment factors), the three SNPs were significantlty associated with higher risk of thromboembolism (P=0.025, <0.0001 and P=0.033, respectively). Further validation studies are warranted in order to design a prospective trial of thromboprophylaxis in mCRC patients with high-risk genotypes.


Assuntos
Neoplasias Colorretais/genética , Fator V/genética , Metilenotetra-Hidrofolato Redutase (NADPH2)/genética , Inibidor 1 de Ativador de Plasminogênio/genética , Polimorfismo de Nucleotídeo Único/genética , Protrombina/genética , Tromboembolia/genética , Adulto , Idoso , Alelos , Bevacizumab/uso terapêutico , Neoplasias Colorretais/tratamento farmacológico , Feminino , Predisposição Genética para Doença/genética , Genótipo , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Risco
15.
Clin Sci (Lond) ; 131(9): 823-831, 2017 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-28424376

RESUMO

The gene SerpinC1 encodes a serine protease inhibitor named antithrombin III (ATIII). This protease demonstrates both anticoagulant and anti-inflammatory action. ATIII is the most important coagulation factor inhibitor, and even minor changes in ATIII can significantly alter the risk of thromboembolism. ATIII can also suppress inflammation via a coagulation-dependent or -independent effect. Moreover, apart from ATIII deficiency, ATIII and its gene SerpinC1 may also be related to many diseases (e.g. hypertension, kidney diseases). The present review summarizes how ATIII affects the progress of kidney disease and its mechanism. Further studies are required to investigate how ATIII affects renal function and the treatment.


Assuntos
Deficiência de Antitrombina III/metabolismo , Antitrombina III/metabolismo , Nefropatias/metabolismo , Tromboembolia/metabolismo , Antitrombina III/genética , Antitrombina III/uso terapêutico , Deficiência de Antitrombina III/genética , Coagulação Sanguínea/efeitos dos fármacos , Humanos , Inflamação/genética , Inflamação/metabolismo , Inflamação/prevenção & controle , Nefropatias/genética , Modelos Biológicos , Fatores de Risco , Transdução de Sinais , Tromboembolia/genética
16.
Pediatr Blood Cancer ; 64(5)2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-27748013

RESUMO

The prevalence of protein S (PS) deficiency in Asian patients with venous thromboembolism is around 8-30%, higher than that in Caucasian populations. The present study reports the genotypes (including one novel mutation) and phenotypes of children with PS deficiency at a tertiary care institute. A total of six patients were included, three with arterial ischemic stroke, two with cerebral venous sinus thrombosis, and one with deep vein thrombosis. PS mutations were identified in four patients: p.R355C, p.G336D, p.E67A, and p.N188KfsX9. p.N188KfsX9 is a novel mutation with less than 20% PS activity noted in heterozygotes.


Assuntos
Deficiência de Proteína S/epidemiologia , Deficiência de Proteína S/genética , Trombose dos Seios Intracranianos/genética , Acidente Vascular Cerebral/genética , Tromboembolia/epidemiologia , Tromboembolia/genética , Trombose Venosa/genética , Adolescente , Criança , Pré-Escolar , Feminino , Genótipo , Humanos , Masculino , Fenótipo , Reação em Cadeia da Polimerase , Deficiência de Proteína S/patologia , Estudos Retrospectivos , Tailândia/epidemiologia
17.
J Stroke Cerebrovasc Dis ; 26(6): 1383-1390, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28412319

RESUMO

BACKGROUND: Warfarin has a narrow therapeutic window. We hypothesized that genetic factors related to warfarin metabolism (CYP2C9) and activity (VKORC1) would show stronger associations than modifiable factors with the quality of anticoagulation control and risks for thromboembolism and hemorrhage. METHODS: In this retrospective cohort analysis, clinical and genetic data were collected from 380 patients with atrial fibrillation (AF) who were followed for an average observation period of 4 years. We evaluated the factors associated with time in therapeutic range (TTR, international normalized ratio [INR]: 2-3) and vascular events (either thromboembolic or hemorrhagic), including both genetic (CYP2C9 and VKORC1 genotype) and modifiable factors (anticoagulation service and warfarin dose assessment interval). RESULTS: The genotypic frequency of CYP2C9*3 (rs1057910) was 9.5% and that of VKORC1 1173C>T (rs9934438) was 16.3%. TTR showed dependence on VKORC1 polymorphism: TTR was higher in carriers of the VKORC1 1173C>T than of the VKORC1 TT genotype (61.7 ± 16.0% versus 56.7 ± 17.4%, P = .031). Multivariate testing showed that the VKORC1 genotype and anticoagulation service were independently related to labile INRs (TTR <65%). Vascular events were observed in 66 patients (18.4%) during the study period. A Cox proportional hazard model showed that the use of anticoagulation service and patients' characteristics, such as AF-thromboembolic risk (CHA2DS2-VASc score: Congestive heart failure, Hypertension, Age 75 years or older, Diabetes mellitus, previous Stroke or transient ischemic attack, Vascular disease, Age 65 to 74 years, female) and consequence (neurologic disability), but not genetic factors, were independently associated with vascular events. CONCLUSIONS: Both genetic factor (VKORC1 genotype) and clinical efforts (anticoagulation service) influenced the quality of anticoagulation control. However, clinical events were more strongly associated with patient characteristics and clinical efforts than with genetic factors.


Assuntos
Anticoagulantes/uso terapêutico , Fibrilação Atrial/tratamento farmacológico , Coagulação Sanguínea/efeitos dos fármacos , Citocromo P-450 CYP2C9/genética , Variantes Farmacogenômicos , Acidente Vascular Cerebral/prevenção & controle , Tromboembolia/prevenção & controle , Vitamina K Epóxido Redutases/genética , Idoso , Idoso de 80 Anos ou mais , Anticoagulantes/efeitos adversos , Fibrilação Atrial/sangue , Fibrilação Atrial/diagnóstico , Fibrilação Atrial/genética , Coagulação Sanguínea/genética , Distribuição de Qui-Quadrado , Citocromo P-450 CYP2C9/metabolismo , Monitoramento de Medicamentos/métodos , Feminino , Frequência do Gene , Hemorragia/induzido quimicamente , Hemorragia/genética , Humanos , Coeficiente Internacional Normatizado , Modelos Lineares , Modelos Logísticos , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Razão de Chances , Farmacogenética , Testes Farmacogenômicos , Modelos de Riscos Proporcionais , Estudos Retrospectivos , Medição de Risco , Fatores de Risco , Acidente Vascular Cerebral/sangue , Acidente Vascular Cerebral/diagnóstico , Acidente Vascular Cerebral/genética , Tromboembolia/sangue , Tromboembolia/diagnóstico , Tromboembolia/genética , Fatores de Tempo , Resultado do Tratamento
18.
Med Sci Monit ; 22: 4345-4353, 2016 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-27840402

RESUMO

BACKGROUND Ischemic stroke and myocardial infarction are fatal diseases and are among the top 10 causes of death in Korea, including arterial thromboembolic events. Many previous studies have described the function of interleukin-6 (IL-6) in arterial thromboembolic events and the association between promoter single-nucleotide polymorphism (SNP) (rs1800795; -174, G/C) of the IL-6 gene. However, these results were controversial. Therefore, we performed a meta-analysis to more precisely assess the association between the SNP of the IL-6 gene and susceptibility to arterial thromboembolic events. MATERIAL AND METHODS We used PubMed, Embase, Google Scholar, and Korean Studies Information Service System (KISS) electronic databases. Comprehensive Meta-analysis software (Corporation, NJ) was used to evaluate the relationship between rs1800795 SNP of IL-6 gene and risk of arterial thromboembolic events. Odds ratio (OR), 95% confidence interval (CI), and P value were also calculated. The 13 eligible studies were analyzed in the meta-analysis. RESULTS The present meta-analysis found that rs1800795 SNP of IL-6 gene is not significantly associated with susceptibility to arterial thromboembolic events (C allele vs. G allele, OR=1.04, 95% CI=0.91-1.19, P=0.619; CC vs. CG+GG, OR=1.09, 95% CI=0.91-1.31, P=0.364; CC+CG vs. GG, OR=0.97, 95% CI=0.78-1.21, P=0.763, respectively), and the SNP of IL-6 gene also did not show any significant association with ischemic stroke or myocardial infarction (P>0.05 in each model). CONCLUSIONS We found that rs1800795 SNP of IL-6 gene was not related to arterial thromboembolic events. However, further study will be needed to confirm these results.


Assuntos
Interleucina-6/genética , Infarto do Miocárdio/genética , Tromboembolia/genética , Alelos , Predisposição Genética para Doença , Humanos , Interleucina-6/metabolismo , Infarto do Miocárdio/metabolismo , Polimorfismo de Nucleotídeo Único , Regiões Promotoras Genéticas/genética , Acidente Vascular Cerebral/genética , Acidente Vascular Cerebral/metabolismo , Tromboembolia/metabolismo
19.
Pharmacogenet Genomics ; 25(2): 93-5, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25461249

RESUMO

Phenprocoumon is an anticoagulant used for thromboembolic disorder prophylaxis metabolized mainly by CYP3A4. However, polymorphisms in this gene did not explain the observed variability. PPARA (peroxisome proliferator-activated receptor-α) is a nuclear receptor that, among others, influences CYP3A4 gene expression. The aim of this study was to determine whether PPARA gene polymorphisms and the CYP3A4*22 allele are associated with phenprocoumon dose variability. A total of 198 patients on a stable dose of phenprocoumon were included in the study. Genotyping was performed by allele discrimination using standardized TaqMan assays. Differences between the average phenprocoumon dose and genotypes/haplotypes were assessed by analysis of variance and multiple linear regression analyses. Patients with the PPARA rs4253728A allele needed higher phenprocoumon doses. However, the effect size (3%) of this association was small. The CYP3A4*22 allele was not associated with the dose of phenprocoumon. As this is the first report of an association between PPARA gene polymorphisms and phenprocoumon dose, future studies are warranted to confirm these results.


Assuntos
Anticoagulantes/uso terapêutico , Biomarcadores Farmacológicos , PPAR alfa/genética , Femprocumona/uso terapêutico , Polimorfismo de Nucleotídeo Único , Adulto , Idoso , Idoso de 80 Anos ou mais , Anticoagulantes/farmacocinética , Relação Dose-Resposta a Droga , Feminino , Frequência do Gene , Genótipo , Humanos , Masculino , Pessoa de Meia-Idade , Femprocumona/farmacocinética , Tromboembolia/tratamento farmacológico , Tromboembolia/genética
20.
Am J Hum Genet ; 91(1): 152-62, 2012 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-22703881

RESUMO

Activated partial thromboplastin time (aPTT) and prothrombin time (PT) are clinical tests commonly used to screen for coagulation-factor deficiencies. One genome-wide association study (GWAS) has been reported previously for aPTT, but no GWAS has been reported for PT. We conducted a GWAS and meta-analysis to identify genetic loci for aPTT and PT. The GWAS for aPTT was conducted in 9,240 individuals of European ancestry from the Atherosclerosis Risk in Communities (ARIC) study, and the GWAS for PT was conducted in 2,583 participants from the Genetic Study of Three Population Microisolates in South Tyrol (MICROS) and the Lothian Birth Cohorts (LBC) of 1921 and 1936. Replication was assessed in 1,041 to 3,467 individuals. For aPTT, previously reported associations with KNG1, HRG, F11, F12, and ABO were confirmed. A second independent association in ABO was identified and replicated (rs8176704, p = 4.26 × 10(-24)). Pooling the ARIC and replication data yielded two additional loci in F5 (rs6028, p = 3.22 × 10(-9)) and AGBL1 (rs2469184, p = 3.61 × 10(-8)). For PT, significant associations were identified and confirmed in F7 (rs561241, p = 3.71 × 10(-56)) and PROCR/EDEM2 (rs2295888, p = 5.25 × 10(-13)). Assessment of existing gene expression and coronary artery disease (CAD) databases identified associations of five of the GWAS loci with altered gene expression and two with CAD. In summary, eight genetic loci that account for ∼29% of the variance in aPTT and two loci that account for ∼14% of the variance in PT were detected and supported by functional data.


Assuntos
Predisposição Genética para Doença , Tempo de Tromboplastina Parcial , Tempo de Protrombina , Tromboembolia/genética , Trombose/genética , Estudos de Coortes , Feminino , Perfilação da Expressão Gênica , Estudo de Associação Genômica Ampla , Humanos , Masculino , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único , Risco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA