• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 14
  • 7
  • 4
  • 2
  • 1
  • 1
  • 1
  • Tagged with
  • 38
  • 38
  • 11
  • 8
  • 5
  • 5
  • 5
  • 5
  • 5
  • 4
  • 4
  • 4
  • 4
  • 4
  • 4
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
11

Usefulness of delE746-A750 and L858R Mutation-Specific Antibodies of EGFR for Predicting Treatment Outcome of Tyrosine Kinase Inhibitors

Tang, En-kuei 24 July 2012 (has links)
Efficacy of tyrosine kinase inhibitor (TKI) therapy depends on epidermal growth factor receptor (EGFR) mutation status in patients with non-small cell lung cancer (NSCLC). There has been an increasing interest in studying mutation-specific rabbit monoclonal antibodies of delE746-A750 mutation in exon 19 and L858R point mutation in exon 21 for detecting EGFR mutants. These two mutations account for approximately 90% of all EGFR mutations. We evaluated the two mutation-specific monoclonal antibodies for the detection of EGFR mutations by immunohistochemistry (IHC) and the relationship with treatment outcome and survival. Twenty-five patients (58.1%) harbored EGFR mutations. These mutations include delE746-A750 mutation for seven patients, L858R point mutation for in eighteen patients. IHC showed, for the delE746-A750 and L858R mutations, sensitivity (57.1% and 66.7%), specificity (97.3% and 100%), positive predictive value (80.0% and 100%), and negative predictive value (94.7% and 80.6%). Analysis for progression-free survival was not correlated to IHC staining, but the overall survival was correlated to IHC staining. These mutation-specific antibodies for delE746-A750 and L858R mutations have high positive predictive value and specificity for predefined EGFR mutations and may be suitable for screening for these predefined mutations. However, negative IHC results required further mutation analyses before excluding EGFR TKI therapy.
12

Farmakogenetika v revmatologii. / Pharmacogenetics in rheumatoid arthritis

Kobrlová, Martina January 2017 (has links)
Charles University in Prague Faculty of Pharmacy in Hradec Králové Department of Pharmacology & Toxicology Student: Martina Kobrlová Supervisor: prof. PharmDr. Petr Pávek, Ph.D. Title of diploma thesis: Pharmacogenetics in rheumatoid arthritis Based on scientific progress in the research of human genome and the discovery of polymorphisms, which are involved in the interindividual differences in human population, there is also a growing interest in pharmacogenetics. It is a field combining pharmacology and genetics with the aim of identifying specific features that could explain the different responses of patients to treatment by clinically used drugs. Applying this knowledge could contribute to a simpler choice of medication for a particular patient and it could reduce the risk of side effects or poor response. In this diploma thesis I dealt with the latest scientific knowledge on pharmacogenetics in rheumatology, in particular the rheumatoid arthritis. From available studies, reviews, and meta-analyzes that have been published, I summarized current data on the relationship between polymorphisms and disease modifying drugs (DMARDs) used for the treatment of this disease. The largest amount of data was found on the most commonly used methotrexate. Further, the work examines the leflunomide and other...
13

Trombose da veia porta em crianças e adolescentes : deficiência das proteínas C, S e Antitrombina e pesquisa das mutações fator V Leiden, G20210A da Protrombina e C677T da Metileno-tetraidrofolato redutase

Pinto, Raquel Borges January 2000 (has links)
Objetivo: A trombose da veia porta é uma causa importante de hiper-tensão porta em crianças e adolescentes, porém, em uma proporção importante dos casos, não apresenta fator etiológico definido. O objetivo desse estudo é determinar a freqüência de deficiência das proteínas inibidoras da coagulação – proteínas C, S e antitrombina − e das mutações fator V Leiden, G20210A no gene da protrombina e C677T da metileno-tetraidrofolato redutase em crianças e adolescentes com trom-bose da veia porta, definir o padrão hereditário de uma eventual deficiência das pro-teínas inibidoras da coagulação nesses pacientes e avaliar a freqüência da deficiên-cia dessas proteínas em crianças e adolescentes com cirrose. Casuística e Métodos: Foi realizado um estudo prospectivo com 14 crianças e adolescentes com trombose da veia porta, seus pais (n = 25) e dois gru-pos controles pareados por idade, constituídos por um grupo controle sem hepato-patia (n = 28) e um com cirrose (n = 24). A trombose da veia porta foi diagnosticada por ultra-sonografia abdominal com Doppler e/ou fase venosa do angiograma celíaco seletivo. A dosagem da atividade das proteínas C, S e antitrombina foi determinada em todos os indivíduos e a pesquisa das mutações fator V Leiden, G20210A da pro-trombina e C677T da metileno-tetraidrofolato redutase, nas crianças e adolescentes com trombose da veia porta, nos pais, quando identificada a mutação na criança, e nos controles sem hepatopatia. Resultados: Foram avaliados 14 pacientes caucasóides, com uma média e desvio padrão de idade de 8 anos e 8 meses ± 4 anos e 5 meses e do diagnóstico de 3 anos e 8 meses ± 3 anos e seis meses. Metade dos pacientes pertenciam ao gênero masculino. O motivo da investigação da trombose da veia porta foi hemorra-gia digestiva alta em 9/14 (64,3%) e achado de esplenomegalia ao exame físico em 5/14 (35,7%). Anomalias congênitas extra-hepáticas foram identificadas em 3/14 (21,4%) e fatores de risco adquiridos em 5/14 (35,7%) dos pacientes. Nenhum pa-ciente tinha história familiar de consangüinidade ou trombose venosa. A deficiência das proteínas C, S e antitrombina foi constatada em 6/14 (42,9%) (p < 0,05 vs con-troles sem hepatopatia), 3/14 (21,4%) (p > 0,05) e 1/14 (7,1%) (p > 0,05) pacientes com trombose da veia porta, respectivamente. A deficiência dessas proteínas não foi identificada em nenhum dos pais ou controles sem hepatopatia. A mutação G20210A no gene da protrombina foi identificada em um paciente com trombose da veia porta e em um controle sem hepatopatia (p = 0,999), mas em nenhum desses foi identificado a mutação fator V Leiden. A mutação C677T da metileno-tetraidrofo-lato redutase foi observada na forma homozigota, em 3/14 (21,4%) dos pacientes com trombose da veia porta e em 5/28 (17,9%) controles sem hepatopatia (p = 0,356). A freqüência da deficiência das proteínas C, S e antitrombina nos pacientes com cir-rose foi de 14/24 (58,3%), 7/24 (29,2%) e 11/24 (45,8%), respectivamente (p < 0,05 vs controles sem hepatopatia), sendo mais freqüente nos pacientes do subgrupo Child-Pugh B ou C, que foi de 11/12 (91,7%), 5/12 (41,7%) e 9/12 (75%), respectivamente (p < 0,05 vs controles sem hepatopatia). Conclusões: A deficiência de proteína C foi freqüente nas crianças e adolescentes com trombose da veia porta e não parece ser de origem genética. A deficiência de proteína S, antitrombina e as presenças das mutações G20210A da protrombina e C677T da metileno-tetraidrofolato redutase foram observadas mas não apresentaram diferença estatística significativa em relação ao grupo controle sem hepatopatia. O fator V Leiden não foi identificado. Os resultados deste estudo sugerem que a deficiência da proteína C pode ocorre como conseqüência da hiper-tensão porta. Os distúrbios pró-trombóticos hereditários não parecem apresentar um papel importante em relação à trombose nas crianças e adolescentes estudadas. / Objective: Portal vein thrombosis is a major cause of portal hypertension in children and adolescents; yet, its etiology is not clearly defined in a considerable number of cases. The present study aims at determining the prevalence of blood coagulation disorders – protein C, protein S and antithrombin – and factor V Leiden, G20210A prothrombin, and C677T methylenetetrahydrofolate reductase mutations in children and adolescents with portal vein thrombosis, as well as assessing the hereditary character of these disorders in these patients, and also evaluating the prevalence of blood coagulation disorders in children and adolescents with cirrhosis. Study design: A prospective study was carried out, including children and adolescents with portal vein thrombosis (n = 14), their parents (n = 25), two age-matched control groups, one without liver disease (n = 28), and another with cirrhosis (n = 24). Portal vein thrombosis was diagnosed through abdominal Doppler ultrasonography and/or venous phase of selective coeliac angiograms. The activity of protein C, protein S and antithrombin was evaluated for all individuals; the presence of factor V Leiden, G20210A prothrombin, and C677T methylenetetrahydrofolate reductase gene mutations was investigated in children and adolescents with portal vein thrombosis, in parents when their respective children presented any of these mutations, and in the control group without liver disease. Results: 14 Caucasian patients were assessed. The mean and standard deviation for age were 8 years and 8 months ± 4 years and 5 months while the mean and standard deviation for diagnosis were 3 years and 8 months ± 3 years and six months. Half of the patients were males. Initial clinical manifestations upon diagnosis were digestive hemorrhage in 9/14 (64.3%) and splenomegaly on physical examination in 5/14 individuals (35.7%). Patients presented extrahepatic anomalies in 3/14 (21.4%) and acquired risk factors in 5/14 (35.7%) of the cases. None of the patients had a family history of consanguinity or venous thrombosis. The frequency of protein C, protein S and antithrombin deficiency was observed in 6/14 (42.9%) (p < 0.05 vs. controls without liver disease), 3/14 (21.4%) (p > 0.05) and 1/14 (7.1%) (p > 0.05) of patients, respectively. None of the portal vein thombosis patients or controls presented protein C, S or antithrombin deficiency. One portal vein patient and one control (p = 0.999) presented G20210A prothrombin mutation. None of these patients presented the factor V Leiden. The homozygous form of C677T methylenetetrahydrofolate reductase mutation was observed in 3/14 patients with portal vein thrombosis (21.4%) and in 5/28 controls (17.9%) (p = 0.356). The frequency of coagulation inhibitor deficiency was high in cirrhotic patients (14/24 (58.3%) PC, 7/24 (29.2%) PS and 11/24 (45.8%) AT; p < 0.05 vs. controls), especially in Child-Pugh B and C patients (11/12 (91.7%) PC, 5/12 (41.7%) PS and 9/12 (75%) AT; p < 0.05 vs. controls). Conclusions: Protein C deficiency was frequent in children and adolescents with portal vein thrombosis and does not seem to be an inherited condition. Protein S and antithrombin deficiency, and G20210A prothrombin and C677T methylenetetrahydrofolate reductase mutations were observed but did not present statistically significant differences when compared to the controls without liver disease. Factor V Leiden was not observed. The results suggest the protein C deficiency may originates from portal hypertension. The hereditary prothrombotic disorders do not seem to play a vital role in thrombosis in children and adolescents with portal vein thrombosis.
14

Trombose da veia porta em crianças e adolescentes : deficiência das proteínas C, S e Antitrombina e pesquisa das mutações fator V Leiden, G20210A da Protrombina e C677T da Metileno-tetraidrofolato redutase

Pinto, Raquel Borges January 2000 (has links)
Objetivo: A trombose da veia porta é uma causa importante de hiper-tensão porta em crianças e adolescentes, porém, em uma proporção importante dos casos, não apresenta fator etiológico definido. O objetivo desse estudo é determinar a freqüência de deficiência das proteínas inibidoras da coagulação – proteínas C, S e antitrombina − e das mutações fator V Leiden, G20210A no gene da protrombina e C677T da metileno-tetraidrofolato redutase em crianças e adolescentes com trom-bose da veia porta, definir o padrão hereditário de uma eventual deficiência das pro-teínas inibidoras da coagulação nesses pacientes e avaliar a freqüência da deficiên-cia dessas proteínas em crianças e adolescentes com cirrose. Casuística e Métodos: Foi realizado um estudo prospectivo com 14 crianças e adolescentes com trombose da veia porta, seus pais (n = 25) e dois gru-pos controles pareados por idade, constituídos por um grupo controle sem hepato-patia (n = 28) e um com cirrose (n = 24). A trombose da veia porta foi diagnosticada por ultra-sonografia abdominal com Doppler e/ou fase venosa do angiograma celíaco seletivo. A dosagem da atividade das proteínas C, S e antitrombina foi determinada em todos os indivíduos e a pesquisa das mutações fator V Leiden, G20210A da pro-trombina e C677T da metileno-tetraidrofolato redutase, nas crianças e adolescentes com trombose da veia porta, nos pais, quando identificada a mutação na criança, e nos controles sem hepatopatia. Resultados: Foram avaliados 14 pacientes caucasóides, com uma média e desvio padrão de idade de 8 anos e 8 meses ± 4 anos e 5 meses e do diagnóstico de 3 anos e 8 meses ± 3 anos e seis meses. Metade dos pacientes pertenciam ao gênero masculino. O motivo da investigação da trombose da veia porta foi hemorra-gia digestiva alta em 9/14 (64,3%) e achado de esplenomegalia ao exame físico em 5/14 (35,7%). Anomalias congênitas extra-hepáticas foram identificadas em 3/14 (21,4%) e fatores de risco adquiridos em 5/14 (35,7%) dos pacientes. Nenhum pa-ciente tinha história familiar de consangüinidade ou trombose venosa. A deficiência das proteínas C, S e antitrombina foi constatada em 6/14 (42,9%) (p < 0,05 vs con-troles sem hepatopatia), 3/14 (21,4%) (p > 0,05) e 1/14 (7,1%) (p > 0,05) pacientes com trombose da veia porta, respectivamente. A deficiência dessas proteínas não foi identificada em nenhum dos pais ou controles sem hepatopatia. A mutação G20210A no gene da protrombina foi identificada em um paciente com trombose da veia porta e em um controle sem hepatopatia (p = 0,999), mas em nenhum desses foi identificado a mutação fator V Leiden. A mutação C677T da metileno-tetraidrofo-lato redutase foi observada na forma homozigota, em 3/14 (21,4%) dos pacientes com trombose da veia porta e em 5/28 (17,9%) controles sem hepatopatia (p = 0,356). A freqüência da deficiência das proteínas C, S e antitrombina nos pacientes com cir-rose foi de 14/24 (58,3%), 7/24 (29,2%) e 11/24 (45,8%), respectivamente (p < 0,05 vs controles sem hepatopatia), sendo mais freqüente nos pacientes do subgrupo Child-Pugh B ou C, que foi de 11/12 (91,7%), 5/12 (41,7%) e 9/12 (75%), respectivamente (p < 0,05 vs controles sem hepatopatia). Conclusões: A deficiência de proteína C foi freqüente nas crianças e adolescentes com trombose da veia porta e não parece ser de origem genética. A deficiência de proteína S, antitrombina e as presenças das mutações G20210A da protrombina e C677T da metileno-tetraidrofolato redutase foram observadas mas não apresentaram diferença estatística significativa em relação ao grupo controle sem hepatopatia. O fator V Leiden não foi identificado. Os resultados deste estudo sugerem que a deficiência da proteína C pode ocorre como conseqüência da hiper-tensão porta. Os distúrbios pró-trombóticos hereditários não parecem apresentar um papel importante em relação à trombose nas crianças e adolescentes estudadas. / Objective: Portal vein thrombosis is a major cause of portal hypertension in children and adolescents; yet, its etiology is not clearly defined in a considerable number of cases. The present study aims at determining the prevalence of blood coagulation disorders – protein C, protein S and antithrombin – and factor V Leiden, G20210A prothrombin, and C677T methylenetetrahydrofolate reductase mutations in children and adolescents with portal vein thrombosis, as well as assessing the hereditary character of these disorders in these patients, and also evaluating the prevalence of blood coagulation disorders in children and adolescents with cirrhosis. Study design: A prospective study was carried out, including children and adolescents with portal vein thrombosis (n = 14), their parents (n = 25), two age-matched control groups, one without liver disease (n = 28), and another with cirrhosis (n = 24). Portal vein thrombosis was diagnosed through abdominal Doppler ultrasonography and/or venous phase of selective coeliac angiograms. The activity of protein C, protein S and antithrombin was evaluated for all individuals; the presence of factor V Leiden, G20210A prothrombin, and C677T methylenetetrahydrofolate reductase gene mutations was investigated in children and adolescents with portal vein thrombosis, in parents when their respective children presented any of these mutations, and in the control group without liver disease. Results: 14 Caucasian patients were assessed. The mean and standard deviation for age were 8 years and 8 months ± 4 years and 5 months while the mean and standard deviation for diagnosis were 3 years and 8 months ± 3 years and six months. Half of the patients were males. Initial clinical manifestations upon diagnosis were digestive hemorrhage in 9/14 (64.3%) and splenomegaly on physical examination in 5/14 individuals (35.7%). Patients presented extrahepatic anomalies in 3/14 (21.4%) and acquired risk factors in 5/14 (35.7%) of the cases. None of the patients had a family history of consanguinity or venous thrombosis. The frequency of protein C, protein S and antithrombin deficiency was observed in 6/14 (42.9%) (p < 0.05 vs. controls without liver disease), 3/14 (21.4%) (p > 0.05) and 1/14 (7.1%) (p > 0.05) of patients, respectively. None of the portal vein thombosis patients or controls presented protein C, S or antithrombin deficiency. One portal vein patient and one control (p = 0.999) presented G20210A prothrombin mutation. None of these patients presented the factor V Leiden. The homozygous form of C677T methylenetetrahydrofolate reductase mutation was observed in 3/14 patients with portal vein thrombosis (21.4%) and in 5/28 controls (17.9%) (p = 0.356). The frequency of coagulation inhibitor deficiency was high in cirrhotic patients (14/24 (58.3%) PC, 7/24 (29.2%) PS and 11/24 (45.8%) AT; p < 0.05 vs. controls), especially in Child-Pugh B and C patients (11/12 (91.7%) PC, 5/12 (41.7%) PS and 9/12 (75%) AT; p < 0.05 vs. controls). Conclusions: Protein C deficiency was frequent in children and adolescents with portal vein thrombosis and does not seem to be an inherited condition. Protein S and antithrombin deficiency, and G20210A prothrombin and C677T methylenetetrahydrofolate reductase mutations were observed but did not present statistically significant differences when compared to the controls without liver disease. Factor V Leiden was not observed. The results suggest the protein C deficiency may originates from portal hypertension. The hereditary prothrombotic disorders do not seem to play a vital role in thrombosis in children and adolescents with portal vein thrombosis.
15

Trombose da veia porta em crianças e adolescentes : deficiência das proteínas C, S e Antitrombina e pesquisa das mutações fator V Leiden, G20210A da Protrombina e C677T da Metileno-tetraidrofolato redutase

Pinto, Raquel Borges January 2000 (has links)
Objetivo: A trombose da veia porta é uma causa importante de hiper-tensão porta em crianças e adolescentes, porém, em uma proporção importante dos casos, não apresenta fator etiológico definido. O objetivo desse estudo é determinar a freqüência de deficiência das proteínas inibidoras da coagulação – proteínas C, S e antitrombina − e das mutações fator V Leiden, G20210A no gene da protrombina e C677T da metileno-tetraidrofolato redutase em crianças e adolescentes com trom-bose da veia porta, definir o padrão hereditário de uma eventual deficiência das pro-teínas inibidoras da coagulação nesses pacientes e avaliar a freqüência da deficiên-cia dessas proteínas em crianças e adolescentes com cirrose. Casuística e Métodos: Foi realizado um estudo prospectivo com 14 crianças e adolescentes com trombose da veia porta, seus pais (n = 25) e dois gru-pos controles pareados por idade, constituídos por um grupo controle sem hepato-patia (n = 28) e um com cirrose (n = 24). A trombose da veia porta foi diagnosticada por ultra-sonografia abdominal com Doppler e/ou fase venosa do angiograma celíaco seletivo. A dosagem da atividade das proteínas C, S e antitrombina foi determinada em todos os indivíduos e a pesquisa das mutações fator V Leiden, G20210A da pro-trombina e C677T da metileno-tetraidrofolato redutase, nas crianças e adolescentes com trombose da veia porta, nos pais, quando identificada a mutação na criança, e nos controles sem hepatopatia. Resultados: Foram avaliados 14 pacientes caucasóides, com uma média e desvio padrão de idade de 8 anos e 8 meses ± 4 anos e 5 meses e do diagnóstico de 3 anos e 8 meses ± 3 anos e seis meses. Metade dos pacientes pertenciam ao gênero masculino. O motivo da investigação da trombose da veia porta foi hemorra-gia digestiva alta em 9/14 (64,3%) e achado de esplenomegalia ao exame físico em 5/14 (35,7%). Anomalias congênitas extra-hepáticas foram identificadas em 3/14 (21,4%) e fatores de risco adquiridos em 5/14 (35,7%) dos pacientes. Nenhum pa-ciente tinha história familiar de consangüinidade ou trombose venosa. A deficiência das proteínas C, S e antitrombina foi constatada em 6/14 (42,9%) (p < 0,05 vs con-troles sem hepatopatia), 3/14 (21,4%) (p > 0,05) e 1/14 (7,1%) (p > 0,05) pacientes com trombose da veia porta, respectivamente. A deficiência dessas proteínas não foi identificada em nenhum dos pais ou controles sem hepatopatia. A mutação G20210A no gene da protrombina foi identificada em um paciente com trombose da veia porta e em um controle sem hepatopatia (p = 0,999), mas em nenhum desses foi identificado a mutação fator V Leiden. A mutação C677T da metileno-tetraidrofo-lato redutase foi observada na forma homozigota, em 3/14 (21,4%) dos pacientes com trombose da veia porta e em 5/28 (17,9%) controles sem hepatopatia (p = 0,356). A freqüência da deficiência das proteínas C, S e antitrombina nos pacientes com cir-rose foi de 14/24 (58,3%), 7/24 (29,2%) e 11/24 (45,8%), respectivamente (p < 0,05 vs controles sem hepatopatia), sendo mais freqüente nos pacientes do subgrupo Child-Pugh B ou C, que foi de 11/12 (91,7%), 5/12 (41,7%) e 9/12 (75%), respectivamente (p < 0,05 vs controles sem hepatopatia). Conclusões: A deficiência de proteína C foi freqüente nas crianças e adolescentes com trombose da veia porta e não parece ser de origem genética. A deficiência de proteína S, antitrombina e as presenças das mutações G20210A da protrombina e C677T da metileno-tetraidrofolato redutase foram observadas mas não apresentaram diferença estatística significativa em relação ao grupo controle sem hepatopatia. O fator V Leiden não foi identificado. Os resultados deste estudo sugerem que a deficiência da proteína C pode ocorre como conseqüência da hiper-tensão porta. Os distúrbios pró-trombóticos hereditários não parecem apresentar um papel importante em relação à trombose nas crianças e adolescentes estudadas. / Objective: Portal vein thrombosis is a major cause of portal hypertension in children and adolescents; yet, its etiology is not clearly defined in a considerable number of cases. The present study aims at determining the prevalence of blood coagulation disorders – protein C, protein S and antithrombin – and factor V Leiden, G20210A prothrombin, and C677T methylenetetrahydrofolate reductase mutations in children and adolescents with portal vein thrombosis, as well as assessing the hereditary character of these disorders in these patients, and also evaluating the prevalence of blood coagulation disorders in children and adolescents with cirrhosis. Study design: A prospective study was carried out, including children and adolescents with portal vein thrombosis (n = 14), their parents (n = 25), two age-matched control groups, one without liver disease (n = 28), and another with cirrhosis (n = 24). Portal vein thrombosis was diagnosed through abdominal Doppler ultrasonography and/or venous phase of selective coeliac angiograms. The activity of protein C, protein S and antithrombin was evaluated for all individuals; the presence of factor V Leiden, G20210A prothrombin, and C677T methylenetetrahydrofolate reductase gene mutations was investigated in children and adolescents with portal vein thrombosis, in parents when their respective children presented any of these mutations, and in the control group without liver disease. Results: 14 Caucasian patients were assessed. The mean and standard deviation for age were 8 years and 8 months ± 4 years and 5 months while the mean and standard deviation for diagnosis were 3 years and 8 months ± 3 years and six months. Half of the patients were males. Initial clinical manifestations upon diagnosis were digestive hemorrhage in 9/14 (64.3%) and splenomegaly on physical examination in 5/14 individuals (35.7%). Patients presented extrahepatic anomalies in 3/14 (21.4%) and acquired risk factors in 5/14 (35.7%) of the cases. None of the patients had a family history of consanguinity or venous thrombosis. The frequency of protein C, protein S and antithrombin deficiency was observed in 6/14 (42.9%) (p < 0.05 vs. controls without liver disease), 3/14 (21.4%) (p > 0.05) and 1/14 (7.1%) (p > 0.05) of patients, respectively. None of the portal vein thombosis patients or controls presented protein C, S or antithrombin deficiency. One portal vein patient and one control (p = 0.999) presented G20210A prothrombin mutation. None of these patients presented the factor V Leiden. The homozygous form of C677T methylenetetrahydrofolate reductase mutation was observed in 3/14 patients with portal vein thrombosis (21.4%) and in 5/28 controls (17.9%) (p = 0.356). The frequency of coagulation inhibitor deficiency was high in cirrhotic patients (14/24 (58.3%) PC, 7/24 (29.2%) PS and 11/24 (45.8%) AT; p < 0.05 vs. controls), especially in Child-Pugh B and C patients (11/12 (91.7%) PC, 5/12 (41.7%) PS and 9/12 (75%) AT; p < 0.05 vs. controls). Conclusions: Protein C deficiency was frequent in children and adolescents with portal vein thrombosis and does not seem to be an inherited condition. Protein S and antithrombin deficiency, and G20210A prothrombin and C677T methylenetetrahydrofolate reductase mutations were observed but did not present statistically significant differences when compared to the controls without liver disease. Factor V Leiden was not observed. The results suggest the protein C deficiency may originates from portal hypertension. The hereditary prothrombotic disorders do not seem to play a vital role in thrombosis in children and adolescents with portal vein thrombosis.
16

Recherche de nouvelles cibles moléculaires dans les syndromes myélodysplasiques et leucémies aiguës myéloïdes / Identification of new molecular targets in myelodysplastic syndromes and acute myeloid leukemias

Rocquain, Julien 29 November 2010 (has links)
Au sein des hémopathies myéloïdes malignes, les syndromes myélodysplasiques(SMD) et les leucémies aiguës myéloïdes (LAM) représentent des pathologies complexes ethétérogènes résultant d’anomalies clonales des cellules souches médullaires. Elles sontcaractérisées par une hématopoïèse inefficace provoquant des cytopénies sanguines graves.Les connaissances sur les anomalies moléculaires des SMD et des LAM, notammentà caryotype normal, sont globalement pauvres et leur physiopathologie encore mal connue.Une meilleure définition moléculaire est nécessaire pour une évaluation pronostique plusprécise de ces hémopathies et pour optimiser secondairement les stratégies thérapeutiques.Cette thèse présente un panorama des classifications cytogénétiques et moléculairesactuelles des SMD et LAM ainsi que l’étude de certaines altérations moléculairesrencontrées dans ces maladies.Grâce à l’apport des techniques d’analyse génomique à grande échelle, notamment laCGH-array, notre laboratoire a identifié de nouvelles altérations génétiques, parmi lesquellesles mutations du gène ASXL1, ainsi que des altérations des gènes codant les protéines de laCohésine et des régulateurs de la protéine CBL. Nous avons analysé une combinaison demutations de gène et émis l’hypothèse d’un modèle de leucémogenèse à 4 classes demutations, afin d’apporter des pistes dans la compréhension de la physiopathologie des SMDet LAM. / Among myeloid malignancies, myelodysplastic syndromes (MDSs) represent a groupof complex diseases characterized by clonal abnormalities of bone marrow hematopoieticprecursor cells. They are defined by an ineffective hematopoiesis leading to peripheralcytopenias. About 40% of MDSs secondarily evolve to acute myeloid leukemia (AML).This risk of transformation is evaluated by several international prognostic scoringsystems like IPSS and WPSS. The WHO classification recognizes several classes of MDSsessentially based on morphology and cytogenetics features, some with a high progressionrisk, like refractory anemia with excess of blasts type 2, others with a low risk, likerefractory anemia with ringed sideroblasts. However, the classification of MDSs is stillunsatisfactory and relevant prognostic markers allowing earlier treatments for patients with ahigh risk of transformation are still lacking. The physiopathology of SMDs and AMLs withnormal karyotype remains unclear. Currently, the only potentially curative treatment isallogenic stem cell transplant, which is feasible for a restricted number of patients and candisplay side effects and failures.A better knowledge of the molecular biology of MDSs and AMLs is necessary for abetter understanding of these diseases and may provide new early prognosis indicators andbetter strategies of treatments.
17

A resposta SOS de Caulobacter crescentus e relações dos mecanismos de reparo com a progressão do ciclo celular. / The SOS response of Caulobacter crescentus and the relationship between DNA repair mechanisms and the cell cycle progression.

Rocha, Raquel Paes da 17 May 2011 (has links)
Caulobacter crescentus pertence ao grupo das proteobactérias e apresenta a característica distinta de diferenciação celular a cada divisão. Este trabalho visou desvendar os mecanismos de reparo de DNA em C. crescentus. Identificamos 44 genes pertecentes ao regulon SOS através da construção de um mutante para o repressor deste, LexA. Caracterizamos funcionalmente alguns dos genes do regulon, como CC_2272 (que codifica uma proteína da família das endonucleases III) e CC_2433. A cepa deficiente em LexA apresentou morfologia filamentosa, e por esse motivo, buscamos também desvendar quais seriam os fatores genéticos responsáveis por esta morfologia. Investigamos também os processos de controle do ciclo celular após a introdução de danos na molécula de DNA pela luz UVC, em mutantes deficientes para diferentes vias de reparo. Estes experimentos nos mostraram que as células procariontes possuem mecanismos para acoplar a progressão do ciclo celular a integridade do material genético. Este trabalho abre novas e excitantes possibilidades no campo da biologia bacteriana. / Caulobacter crescentus belongs to the proteobacteria group and exhibts the distinctive feature of cellular differentiation after each division. This work aimed to reveal the DNA repair mechanisms in C. crescentus. We have identified 44 genes belonging to the SOS regulon through the construction of a mutant strain to its repressor. We have functionally characterized some of its genes, like CC_2272 (that encodes an endonuclease III family protein) and CC_2433. The lexA strain showed filamentous morphology, e because of that, we have tried to discover which the genetic factors responsible for this morphology were. We have also investigated the cell cycle control processes after the introduction of damages in the DNA by the UVC light, in mutant strains deficient in different repair pathways. These experiments showed us that prokaryotic cells possess mechanisms to couple the cell cycle progression to the integrity of the genetic material. This work opens new and exciting possibilities in the field of bacterial biology.
18

Breast Cancer Susceptibility Gene 1 (BRCA1) And Breast Cancer

Lakhotia, Smita 02 1900 (has links)
Breast Cancer susceptibility gene 1 (BRCA1) & Breast Cancer Breast cancer is one of the most common malignancies affecting women worldwide. About 5-10% of all cases are estimated to be familial. Mutations in the BRCA1 (Breast Cancer susceptibility gene 1) gene account for about 15-20% of inherited breast cancer cases and 60-80% of families predisposed to both breast and ovarian cancer. BRCA1 mutations also result in susceptibility to early-onset breast and ovarian cancer. The human BRCA1 gene encodes a multi-domain 1,863 amino acid nuclear protein that is expressed in a wide variety of adult human tissues. The N-terminal end of BRCA1 contains a RING-finger domain. Exon 11 of BRCA1 contains two nuclear localization signals towards its N-terminal for targeting BRCA1 to the nucleus. The carboxyl terminus contains two BRCT (BRCA1 C-terminal) domains and a transcriptional activation domain. This study was carried out to functionally characterize BRCA1 and to find out the percentage in which BRCA1 gene is mutated in Indian familial breast and/or ovarian cancer families. The work has been divided into three sections: 1. Identification & characterization of a BRCA1 Associated Protein 2 (BAP2). 2. Germ-line BRCA1 mutation Analysis in Indian Breast and/or Ovarian Cancer Families. 3. Characterization of a novel missense mutation (E116K) in BRCA1. BRCA1 is known to interact with large number of proteins and is involved in various cellular functions like tumorigenesis, transcription, DNA damage repair, cell-cycle control, ubiquitinylation, genetic stability, cell growth and apoptosis. The interacting partners of BRCA1 have given a lot of clue about the functions of this complex protein. In the first project, we used the yeast two-hybrid system to identify novel interacting proteins of BRCA1. We used the 1-500 amino acid region of BRCA1 as bait in library screen and picked up a novel clone (clone 89) showing interaction with BRCA1. Clone 89 contains approximately 2.3 Kb long cDNA sequence. Using the nucleotide blast search, we obtained a full-length cDNA of approximately 5.4 Kb (KIAA0657) that is located on chromosome 2, 2q36.1 region. We have named this new protein BRCA1 Associated Protein 2 (BAP2). Translation of this coding sequence gave a protein that has homology to Titin protein. This protein, which has 1,236 amino acids, contains 9 Immunoglobulin like domains. The homologues of this protein exists in many other organisms but the function is not known. We have confirmed the interaction between BRCA1 and c89 using in vitro GST pull-down assay. We have studied the influence of BAP2 on various functions of BRCA1 like transcription, colony suppression and cell cycle. In the transcription assays, BAP2 activated p21 promoter activity perhaps by using endogenous BRCA1 as simultaneous ectopic expression of truncated BRCA1 (containing aa 1-500) abolished this activity. Further, BAP2 also increased the ability of BRCA1 to activate p21 promoter suggesting that BAP2 may act as a co-activator of BRCA1 functions. Surprisingly, we observed that BAP2 inhibited p53-mediated transcription both in the absence and presence of BRCA1. BAP2 failed to inhibit colony growth by itself as well as in combination with BRCA1. In the cell-cycle study, we found that BAP2 did not have any significant effect on cell cycle profile by itself. However, it drastically augmented the G2/M arrest mediated by BRCA1. Thus we conclude that we have identified a novel interacting protein of BRCA1 that regulates certain functions of BRCA1. Detection of mutations is of central importance in the study of genetic and malignant diseases. Mutation detection helps us in understanding the protein structure, function and expression. More than that, it is also important for pre-symptomatic/antenatal diagnosis, confirmation of the genetic cause of the disease and the mode of inheritance of a disease in a particular family, the prediction of clinical phenotype and the potentiation of diagnostic analysis in the case of families with incomplete pedigrees or with new mutations. Therefore, the importance of direct mutation analysis cannot be understated. The second project deals with screening of mutations in BRCA1 gene in 50 familial breast and/or ovarian cancer families using the technique of Conformation Sensitive Gel Electrophoresis (CSGE). CSGE can be used to detect mismatches in DNA heteroduplexes that contain one strand of wild type and one strand of mutated DNA. In a collaborative study with Kidwai Memorial Hospital for Oncology, Bangalore, we screened 50 families suffering from breast and/or ovarian cancer. We detected 13 mutations in this study out of which 3 are novel and 10 have already been reported earlier (Breast Information Core). All the mutations obtained in our study result in truncation of the BRCA1 protein either because of non-sense mutation or frame-shift mutation. Interestingly, 8 of the mutations detected are 185delAG mutations – the most commonly occurring mutation in Ashkenazi Jewish population. From this study, we conclude that BRCA1 is mutated in 26% of familial breast and/or ovarian cancer cases in India. Genetic testing in individuals with family history of breast, ovarian or both has become very common. It is difficult to interpret the result of genetic screen if a DNA change in the gene does not result in truncation of the protein. Rare missense changes of unknown functional and pathogenic significance are called unclassified variants. It is important to study the functional implications of these unclassified variants in order to determine the risk associated with the presence of such variations. The third project deals with characterization of one such missense variation. In an earlier mutation analysis study for BRCA1 gene in breast cancer samples, we found a novel missense variation resulting in Glu116Lys (E116K) change. In order to determine if this variant is a disease associated missense mutation or a benign sequence alteration; we introduced this variation into full length BRCA1 cDNA and studied its effect on the known functions of BRCA1, namely, transcription, colony suppression and cell cycle. We found that E116K is defective for activating transcription. However, it continued to inhibit growth in colony formation assay and arrest cells in G2/M phase of cell cycle. We conclude that E116K mutation results in loss of transactivation function of BRCA1 but has no effect on colony formation and cell cycle regulation; thus it can be categorized as a novel missense mutation.
19

A resposta SOS de Caulobacter crescentus e relações dos mecanismos de reparo com a progressão do ciclo celular. / The SOS response of Caulobacter crescentus and the relationship between DNA repair mechanisms and the cell cycle progression.

Raquel Paes da Rocha 17 May 2011 (has links)
Caulobacter crescentus pertence ao grupo das proteobactérias e apresenta a característica distinta de diferenciação celular a cada divisão. Este trabalho visou desvendar os mecanismos de reparo de DNA em C. crescentus. Identificamos 44 genes pertecentes ao regulon SOS através da construção de um mutante para o repressor deste, LexA. Caracterizamos funcionalmente alguns dos genes do regulon, como CC_2272 (que codifica uma proteína da família das endonucleases III) e CC_2433. A cepa deficiente em LexA apresentou morfologia filamentosa, e por esse motivo, buscamos também desvendar quais seriam os fatores genéticos responsáveis por esta morfologia. Investigamos também os processos de controle do ciclo celular após a introdução de danos na molécula de DNA pela luz UVC, em mutantes deficientes para diferentes vias de reparo. Estes experimentos nos mostraram que as células procariontes possuem mecanismos para acoplar a progressão do ciclo celular a integridade do material genético. Este trabalho abre novas e excitantes possibilidades no campo da biologia bacteriana. / Caulobacter crescentus belongs to the proteobacteria group and exhibts the distinctive feature of cellular differentiation after each division. This work aimed to reveal the DNA repair mechanisms in C. crescentus. We have identified 44 genes belonging to the SOS regulon through the construction of a mutant strain to its repressor. We have functionally characterized some of its genes, like CC_2272 (that encodes an endonuclease III family protein) and CC_2433. The lexA strain showed filamentous morphology, e because of that, we have tried to discover which the genetic factors responsible for this morphology were. We have also investigated the cell cycle control processes after the introduction of damages in the DNA by the UVC light, in mutant strains deficient in different repair pathways. These experiments showed us that prokaryotic cells possess mechanisms to couple the cell cycle progression to the integrity of the genetic material. This work opens new and exciting possibilities in the field of bacterial biology.
20

Preimplantační genetická haplotypizace v geneticky rizikových rodinách / Preimplantation genetic haplotyping in genetically risk families

Borgulová, Irena January 2018 (has links)
PREIMPLANTATION GENETIC HAPLOTYPING IN GENETICALLY RISK FAMILIES Abstract of Irena Borgulova's PhD study Page 1/1 ABSTRACT Preimplantation genetic diagnosis (PGD) is at the intersection of assisted reproduction and clinical genetics. PGD precedes prenatal diagnosis because consists in biopsy of a single embryonic cell and its examination excluding genetic risks before embryo transfer back to mother uterus. Methods within PGD can offer all spectrums of possible investigations of a single cell, whether focused on monogenic disorders, chromosomal aberration or abnormality of whole genome. Monogenic diseases in embryos can be detected by direct or indirect linkage analysis. Indirect linkage analysis has the advantage compared to direct analysis that it is able to indentify pertinent aberration of examined chromosome. Indirect linkage analysis is characterised by preimplantation genetic haplotyping (PGH) which is prime and important constituent of PGD cycle. PGH is based on family anamnesis for determination of pathologic/ high-risk (mutation-associated) haplotype and healthy/ low-risk (without mutation) haplotype by comparison with the haplotypes of other family members. PGD cycle requires in vitro fertilisation (IVF). IVF cycle includes hormonal stimulation, biopsy of oocytes and their fertilisation outside...

Page generated in 0.4279 seconds