Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
2.
Clin Sci (Lond) ; 131(9): 823-831, 2017 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-28424376

RESUMEN

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.


Asunto(s)
Deficiencia de Antitrombina III/metabolismo , Antitrombina III/metabolismo , Enfermedades Renales/metabolismo , Tromboembolia/metabolismo , Antitrombina III/genética , Antitrombina III/uso terapéutico , Deficiencia de Antitrombina III/genética , Coagulación Sanguínea/efectos de los fármacos , Humanos , Inflamación/genética , Inflamación/metabolismo , Inflamación/prevención & control , Enfermedades Renales/genética , Modelos Biológicos , Factores de Riesgo , Transducción de Señal , Tromboembolia/genética
3.
Kidney Int ; 88(4): 796-803, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26108065

RESUMEN

Antithrombin III, encoded by SerpinC1, is a major anti-coagulation molecule in vivo and has anti-inflammatory effects. We found that patients with low antithrombin III activities presented a higher risk of developing acute kidney injury after cardiac surgery. To study this further, we generated SerpinC1 heterozygous knockout rats and followed the development of acute kidney injury in a model of modest renal ischemia/reperfusion injury. Renal injury, assessed by serum creatinine and renal tubular injury scores after 24 h of reperfusion, was significantly exacerbated in SerpinC1(+/-) rats compared to wild-type littermates. Concomitantly, renal oxidative stress, tubular apoptosis, and macrophage infiltration following this injury were significantly aggravated in SerpinC1(+/-) rats. However, significant thrombosis was not found in the kidneys of any group of rats. Antithrombin III is reported to stimulate the production of prostaglandin I2, a known regulator of renal cortical blood flow, in addition to having anti-inflammatory effects and to protect against renal failure. Prostaglandin F1α, an assayable metabolite of prostaglandin I2, was increased in the kidneys of the wild-type rats at 3 h after reperfusion. The increase of prostaglandin F1α was significantly blunted in SerpinC1(+/-) rats, which preceded increased tubular injury and oxidative stress. Thus, our study found a novel role of SerpinC1 insufficiency in increasing the severity of renal ischemia/reperfusion injury.


Asunto(s)
Lesión Renal Aguda/etiología , Deficiencia de Antitrombina III/complicaciones , Antitrombina III/metabolismo , Riñón/metabolismo , Daño por Reperfusión/etiología , Lesión Renal Aguda/genética , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Anciano , Animales , Antitrombina III/análisis , Antitrombina III/genética , Deficiencia de Antitrombina III/genética , Deficiencia de Antitrombina III/metabolismo , Apoptosis , Biomarcadores/sangre , Procedimientos Quirúrgicos Cardíacos/efectos adversos , Creatinina/sangre , Modelos Animales de Enfermedad , Femenino , Técnicas de Silenciamiento del Gen , Predisposición Genética a la Enfermedad , Heterocigoto , Humanos , Riñón/patología , Macrófagos/metabolismo , Masculino , Persona de Mediana Edad , Estrés Oxidativo , Fenotipo , Prostaglandinas F/metabolismo , Ratas Transgénicas , Daño por Reperfusión/genética , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Factores de Riesgo , Índice de Severidad de la Enfermedad , Transducción de Señal , Factores de Tiempo
4.
J Thromb Haemost ; 12(7): 1131-40, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24824609

RESUMEN

BACKGROUND: The characterization of natural mutants identified in patients with antithrombin deficiency has helped to identify functional domains or regions of this key anticoagulant and the mechanisms involved in the deficiency, as well as to define the clinical prognosis. Recently, we described an abnormal glycosylation in a pleiotropic mutant (K241E) that explained the impaired heparin affinity and the mild risk of thrombosis in carriers. OBJECTIVES: To evaluate the effects of different natural pleiotropic mutations on the glycosylation of antithrombin and their functional effects. METHODS: Five pleiotropic mutations identified in patients with antithrombin deficiency and located at each one of the strands of the C-sheet were selected (K241E, M251I, M315K, F402L, and P429L). Recombinant mutants were generated and purified. Glycoform heterogeneity and conformational sensitivity were studied with electrophoresis, proteomic analysis, and glycomic analysis. Heparin affinity was evaluated from intrinsic fluorescence. Reactivity assays with factor Xa, thrombin and neutrophil elastase in the presence or absence of heparin were also performed. RESULTS AND CONCLUSIONS: Pleiotropic mutants, except for that with the M315K mutation, which affects a non-exposed residue, showed two glycoforms. Variant 1, with abnormal glycosylation, had reduced heparin affinity and severely affected reactivity with the target proteases. In contrast, variant 2, with similar electrophoretic mobility and heparin affinity to wild-type antithrombin, had impaired inhibitory activity that was partially compensated for by activation with heparin. Our results suggest the C-sheet of antithrombin as a new region that is relevant for proper maturation of the N-glycans. Therefore, pleiotropic mutations lead to glycosylation defects that are responsible for the reduced heparin affinity.


Asunto(s)
Deficiencia de Antitrombina III/genética , Antitrombinas/química , Mutación , Polisacáridos/química , Adolescente , Adulto , Deficiencia de Antitrombina III/metabolismo , Electroforesis , Glicosilación , Heparina/química , Heterocigoto , Humanos , Elastasa de Leucocito/metabolismo , Persona de Mediana Edad , Mutagénesis Sitio-Dirigida , Pronóstico , Estructura Terciaria de Proteína , Proteómica , Proteínas Recombinantes/química , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Trombina/química , Trombosis/genética
5.
Clin Appl Thromb Hemost ; 20(4): 355-64, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24335249

RESUMEN

Population pharmacokinetic (PK) analyses were conducted to refine dosing recommendations for recombinant human anti-thrombin therapy in surgery and delivery patients with hereditary antithrombin deficiency (HD). Single-dose PK data from patients with HD and nonlinear mixed-effects modeling were used to devise a dosing regimen to target antithrombin (AT) activity levels between 80% and 120% of normal. External validation with data from a phase 3 trial confirmed the correctness of a covariate-free model for surgery patients, but dosing adjustment was necessary for delivery patients. After different covariates were tested, the model was updated to incorporate the influential covariate, delivery. Simulations were used to develop a therapeutic drug-monitoring scenario that results in steady state AT activity levels within the target range as quickly as practically feasible. Data from a second clinical trial provided additional external validation and confirmed the accuracy of the dosing model for both groups of patients.


Asunto(s)
Deficiencia de Antitrombina III/metabolismo , Antitrombinas/farmacocinética , Deficiencia de Antitrombina III/tratamiento farmacológico , Deficiencia de Antitrombina III/cirugía , Monitoreo de Drogas , Femenino , Humanos , Masculino , Modelos Biológicos , Proteínas Recombinantes/farmacocinética
6.
Thromb Res ; 132(1): 132-7, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23768451

RESUMEN

INTRODUCTION: Congenital antithrombin (AT) deficiency is an inherited thrombophilia with high thrombosis prevalence. It has been reported that functional laboratory tests have varying potential in recognizing type II defects, and that there is discrepancy between thrombin inhibition based and factor Xa inhibition based methods. MATERIALS AND METHODS: Patients with known AT deficiency (n=374) were interviewed and their current AT status was tested in a new blood sample (n=214). The samples were analyzed using five different commercial methods (either thrombin or FXa based and one thrombin based method using two different incubation times). Antigen assay was used for typing the deficiency. RESULTS: In 101 of 214 (47.2%) samples the results obtained by different methods were congruent: 91 low and 10 normal by all assays. All other 113 (52.8%) samples showed discrepant values between the assays: most of them had abnormal results by two methods and normal by other methods. The discrepancies were observed mainly in type II deficiency. The best correlation of results was observed between one thrombin based and one FXa based assay. CONCLUSIONS: There was great inter-assay variability especially in type II deficient patients, but also in patients with type I deficiency. However, most of the patients defined as having normal AT activity by some methods had thrombotic symptoms. Most tested assays find type I AT deficient patients accurately. In our study population only methods A1 and C could find most patients with type II AT deficiency, whereas methods A2, B and D misdiagnosed the majority of patients as non-deficient.


Asunto(s)
Deficiencia de Antitrombina III/diagnóstico , Deficiencia de Antitrombina III/metabolismo , Antitrombina III/metabolismo , Pruebas de Coagulación Sanguínea , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Deficiencia de Antitrombina III/sangre , Pruebas de Coagulación Sanguínea/métodos , Factor Xa/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Trombina/metabolismo , Adulto Joven
7.
Best Pract Res Clin Haematol ; 25(3): 333-44, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22959549

RESUMEN

Thrombosis in children is gaining increased awareness, as advanced medical care has increased treatment intensity of hospitalized pediatric patients. Guidelines for diagnosis and treatment of children and neonates with venous thromboembolism (VTE) are mostly extrapolated from adult data, despite the uniqueness of their hemostatic system. Whereas inherited thrombophilia (IT) have been established as risk factors for VTE in adults, in children with idiopathic VTE and in pediatric populations in which thromboses were associated with medical diseases, IT have been described as additional risk factors. Follow-up data for VTE recurrence in children suggest a recurrence rate between 3% (neonates) and 21% (idiopathic VTE). Apart from underlying medical conditions, recently reported systematic reviews on pediatric VTE and stroke have shown significant associations between thrombosis and presence of factor V G1691A, factor II G20210A, protein C-, protein S- and antithrombin deficiency, even more pronounced when combined IT were involved. The pooled odds ratios (OR: single IT) for VTE onset ranged from 2.4 for the factor II G20210A mutation (cerebrovascular occlusion) to 9.4 in children with antithrombin deficiency (venous VTE). In addition, the pooled OR for persistent antiphospholipid antibodies/lupus anticoagulants was 6.6 for children with cerebrovascular occlusion and 4.9 for pediatric cases with venous VTE. The factor II G20210A mutation (OR: 2.1), protein C- (OR: 2.4), S- (OR: 3.1), and antithrombin deficiency (OR: 3.0) did also play a significant role at recurrence. Among primarily asymptomatic family members of pediatric VTE index cases annual VTE incidences were 2.82% (95% confidence interval [95% CI], 1.63-4.80%) in carriers of antithrombin, protein C, or protein S-deficiency, 0.42% (0.12-0.53%) for factor II G202010A, 0.25% (0.12-0.53%) for factor V G1691A, and 0.10% (0.06-0.17%) in relatives with no IT. Based on these data diagnosis, screening and treatment issues will be discussed.


Asunto(s)
Trombofilia/diagnóstico , Tromboembolia Venosa/diagnóstico , Anticuerpos Antifosfolípidos/sangre , Deficiencia de Antitrombina III/metabolismo , Niño , Factor V/genética , Humanos , Recién Nacido , Inhibidor de Coagulación del Lupus/sangre , Mutación , Oportunidad Relativa , Proteína C/genética , Proteína S/genética , Protrombina/genética , Factores de Riesgo , Trombofilia/complicaciones , Trombofilia/congénito , Trombofilia/patología , Tromboembolia Venosa/complicaciones , Tromboembolia Venosa/patología
8.
Mol Med ; 18: 762-70, 2012 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-22481271

RESUMEN

Mutations affecting mobile domains of antithrombin induce conformational instability resulting in protein polymerization that associates with a severe clinical phenotype, probably by an unknown gain of function. By homology with other conformational diseases, we speculated that these variants might infect wild-type (WT) monomers reducing the anticoagulant capacity. Infective polymerization of WT polymers and different P1 mutants (p.R425del, p.R425C and p.R425H) were evaluated by using native gels and radiolabeled WT monomers and functional assays. Human embryonic kidney cells expressing the Epstein-Barr nuclear antigen 1 (HEK-EBNA) cells expressing inducible (p.R425del) or two novel constitutive (p.F271S and p.M370T) conformational variants were used to evaluate intracellular and secreted antithrombin under mild stress (pH 6.5 and 39°C for 5 h). We demonstrated the conformational sensitivity of antithrombin London (p.R425del) to form polymers under mild heating. Under these conditions purified antithrombin London recruited WT monomers into growing polymers, reducing the anticoagulant activity. This process was also observed in the plasma of patients with p.R425del, p.R425C and p.R425H mutations. Under moderate stress, coexpression of WT and conformational variants in HEK-EBNA cells increased the intracellular retention of antithrombin and the formation of disulfide-linked polymers, which correlated with impaired secretion and reduction of anticoagulant activity in the medium. Therefore, mutations inducing conformational instability in antithrombin allow its polymerization with the subsequent loss of function, which under stress could sequestrate WT monomers, resulting in a new prothrombotic gain of function, particularly relevant for intracellular antithrombin. The in vitro results suggest a temporal and severe plasma antithrombin deficiency that may contribute to the development of the thrombotic event and to the clinical severity of these mutations.


Asunto(s)
Deficiencia de Antitrombina III/metabolismo , Proteínas Antitrombina/química , Proteínas Antitrombina/metabolismo , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Proteínas Antitrombina/genética , Línea Celular , Humanos , Conformación Proteica , Multimerización de Proteína , Estabilidad Proteica , Estrés Fisiológico
10.
Int J Lab Hematol ; 33(3): 227-37, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21401902

RESUMEN

INTRODUCTION: Antithrombin (AT) deficiency is associated with an increased risk of deep vein thrombosis and pulmonary embolism which are major causes of morbidity and death. The incidence of deficiency in healthy populations has been reported to vary from 1/600 to 1/5000, with the variation being due to the different populations studied and detection methods used. When reduced activity levels are identified it is important to measure the AT antigen levels to differentiate type I from type II disorders, as type II defects have varying thrombotic risk. METHODS: Functional AT assays detect the ability of AT to inactivate thrombin or factor Xa, and AT antigen assays detect the quantity of AT in plasma. In functional assays, reducing the incubation time of sample with enzyme/heparin reagent may increase sensitivity to type II defects. An excess of antigen over activity level suggests the presence of functionally defective AT, which can be characterized further by assaying AT in the absence of heparin, electrophoresis to investigate the ability of heparin to bind to AT, and gene sequencing. RESULTS: Many patients with AT deficiency have a type II defect and these defects may not be detected by all routine diagnostic assays. Assays using human thrombin may lack specificity and assays that use factor Xa may fail to detect the common variant, AT Cambridge II, which can be detected by assays using bovine thrombin, especially if activity is compared to antigen by ratio. Factor Xa based assays may be particularly sensitive to certain heparin binding defects, and sensitivity of assays to both heparin binding and reactive site defects can be improved by shortening the incubation time with enzyme. CONCLUSION: uAT activity assays are essential for the detection of AT deficiency because type II defects are relatively common in patients with heritable deficiency. No one functional assay can be assumed to detect all forms of AT deficiency, and assays can sometimes be improved by reducing reaction time of AT with thrombin or factor Xa.


Asunto(s)
Deficiencia de Antitrombina III/genética , Deficiencia de Antitrombina III/metabolismo , Fenotipo , Antitrombina III/genética , Antitrombina III/metabolismo , Deficiencia de Antitrombina III/diagnóstico , Antitrombinas/sangre , Bioensayo , Humanos
11.
J Theor Biol ; 253(4): 725-38, 2008 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-18539301

RESUMEN

A mathematical model comprised of 23 reaction-diffusion equations is used to simulate the biochemical changes and transport of various reactants involved in coagulation and fibrinolysis in quiescent plasma. The growth and lysis of a thrombus, as portrayed by the model equations, is governed by boundary conditions that include the surface concentration of TF-VIIa, the generation of XIa by contact activation (in vitro), and the secretion of tPA due to endothelial activation. We apply the model to two clinically relevant hypercoagulable states, caused by deficiency of either antithrombin III or protein C. These predictions are compared with published experimental data which validate the utility of the developed model under the special case of static conditions. The incorporation of varying hemodynamic conditions in to the current fluid static model remains to be performed.


Asunto(s)
Deficiencia de Antitrombina III/metabolismo , Coagulación Sanguínea/fisiología , Plasma/metabolismo , Deficiencia de Proteína C/metabolismo , Factores de Coagulación Sanguínea/metabolismo , Fibrinólisis/fisiología , Humanos , Modelos Biológicos
12.
Thromb Res ; 119(5): 631-41, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-16844203

RESUMEN

Plasma-derived human antithrombin (pAT) is used for the treatments of disseminated intravascular coagulation (DIC) and hereditary antithrombin deficiencies. We expressed recombinant human antithrombin (rAT) in Chinese hamster ovary (CHO) cells. The purified rAT is composed of 55% alpha-isoform and 45% beta-isoform. The structure of the N-linked oligosaccharides of rAT is the same biantennary complex type as previously found in pAT with less sialylated on the non-reducing ends. Most of the oligosaccharides of rAT are fucosylated at the reducing ends of N-acetylglucosamine, while those of pAT are not fucosylated. Despite of the difference in sialylation and fucosylation of the oligosaccharide units, rAT and pAT showed indistinguishable heparin cofactor and progressive activities, and they bound to thrombin in a one-to-one stoichiometric manner. In lipopolysaccharide (LPS)-induced and thromboplastin-induced DIC rat models, rAT reduced fibrinogen and platelet consumption to a similar extent with pAT. In LPS-induced DIC model, both ATs similarly restrained the increase of alanine aminotransferase and aspartate aminotransferase activities. Finally, pharmacokinetic analysis showed that both ATs had similar half-lives in the circulation of normal rats. Together, the present study demonstrated that rAT prepared in CHO cells has potential for a substitute of pAT in therapeutic use.


Asunto(s)
Antitrombinas/uso terapéutico , Coagulación Intravascular Diseminada/tratamiento farmacológico , Proteínas Recombinantes/uso terapéutico , Alanina Transaminasa/efectos de los fármacos , Alanina Transaminasa/metabolismo , Animales , Deficiencia de Antitrombina III/tratamiento farmacológico , Deficiencia de Antitrombina III/metabolismo , Antitrombinas/biosíntesis , Antitrombinas/química , Aspartato Aminotransferasas/efectos de los fármacos , Aspartato Aminotransferasas/metabolismo , Células CHO , Cricetinae , Cricetulus , Modelos Animales de Enfermedad , Coagulación Intravascular Diseminada/metabolismo , Fibrinógeno/efectos de los fármacos , Glicosilación , Humanos , Lipopolisacáridos/farmacología , Masculino , Oligosacáridos/química , Oligosacáridos/metabolismo , Isoformas de Proteínas/biosíntesis , Isoformas de Proteínas/química , Isoformas de Proteínas/uso terapéutico , Ratas , Ratas Wistar , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Estereoisomerismo , Tromboplastina/farmacología , Factores de Tiempo
13.
Blood Coagul Fibrinolysis ; 17(3): 217-20, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16575261

RESUMEN

Quantitative antithrombin deficiency constitutes an important risk factor for venous thromboembolism, stillbirth, and other complications of pregnancy. Studies suggest, however, that individuals heterozygous for missense mutations involving the heparin-binding site of antithrombin do not have a significantly increased thrombotic risk. Owing to the rarity of such mutations, it remains unclear whether any specific heparin-binding site defects might be associated with thrombotic potential. We report here the case of a pregnant woman with an exceptionally rare Type II heparin-binding site antithrombin variant. This case highlights the difficult issues that are associated with the management of Type II antithrombin deficiency during pregnancy.


Asunto(s)
Antitrombina III/genética , Heparina/metabolismo , Mutación Missense/genética , Complicaciones Cardiovasculares del Embarazo/genética , Complicaciones Cardiovasculares del Embarazo/metabolismo , Tromboembolia/genética , Tromboembolia/metabolismo , Adulto , Antitrombina III/metabolismo , Deficiencia de Antitrombina III/genética , Deficiencia de Antitrombina III/metabolismo , Sitios de Unión/genética , Femenino , Heparina/administración & dosificación , Heparina/uso terapéutico , Heterocigoto , Humanos , Embarazo , Complicaciones Cardiovasculares del Embarazo/tratamiento farmacológico , Análisis de Secuencia de ADN , Tromboembolia/tratamiento farmacológico
14.
Blood Coagul Fibrinolysis ; 16(2): 149-55, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15741804

RESUMEN

As a major physiological inhibitor of thrombin and other coagulation proteases, antithrombin (AT) plays an important role in the maintenance of normal hemostasis and its deficiency is associated with a predisposition for familial venous thromboembolic disease. Recently, we found a novel mutation (13387-9delG) in the antithrombin gene that is associated with type I AT deficiency. To examine the molecular pathologic mechanism of this mutation causing type I AT deficiency, the wild-type and the mutant AT constructs were expressed in COS-7 cells or Chinese Hamster Ovary cells. No AT antigen could be detected by enzyme-linked immunosorbent assay in the conditioned media of cells expressing the mutant protein, and the AT antigen level was reduced in cell lysates. The mutant AT-expressing cells did not have less intracellular mRNA levels than the wild-type transfectants as estimated by quantitative reverse transcriptase-polymerase chain reaction. Metabolic and pulse-chase experiments showed the newly synthesized wild-type AT protein was gradually secreted into the media, whereas no labeled mutant AT protein was detected in the media and the total amount of radioactivity was significantly reduced in the cells during the chase periods. By immunofluorescence analysis, the staining of the mutant AT was weaker than that of the wild type, and was predominantly diffuse without perinuclear enhancement. These results indicate that the 13387-9delG mutation, which disrupts the disulfide bridge Cys247-Cys430, impairs the secretion and stability of the truncated AT protein associated with intracellular degradation.


Asunto(s)
Deficiencia de Antitrombina III/metabolismo , Antitrombina III/biosíntesis , Mutación , Adolescente , Animales , Antitrombina III/genética , Deficiencia de Antitrombina III/genética , Células CHO , Células COS , Chlorocebus aethiops , Cricetinae , Expresión Génica , Humanos , Masculino
15.
Int J Biochem Cell Biol ; 36(3): 386-9, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-14687916

RESUMEN

Antithrombin is a serine proteinase inhibitor (serpin) which controls the process of coagulation. It has a well defined structure, consisting of three beta-sheets, nine alpha-helices and a reactive centre loop (RCL). The RCL contains the reactive centre which harbours a bait sequence for target proteases; cleavage results in inhibition by a unique mechanism. The inhibitory activity of antithrombin is controlled by its interaction with the co-factor, heparin, which accelerates its interaction with target proteases. This ensures that heparin and its newer derivatives, such as heparin pentasaccharide, are the mainstay therapeutics for control of thrombosis or inappropriate clotting. The clinical importance of antithrombin is manifested by its clear association with thrombosis when deficiency states occur.


Asunto(s)
Antitrombina III/metabolismo , Coagulación Sanguínea , Inhibidores de Serina Proteinasa/metabolismo , Animales , Antitrombina III/química , Deficiencia de Antitrombina III/genética , Deficiencia de Antitrombina III/metabolismo , Heparina/metabolismo , Humanos , Conformación Proteica , Trombosis/etiología
16.
Wiad Lek ; 56(7-8): 341-7, 2003.
Artículo en Polaco | MEDLINE | ID: mdl-14969162

RESUMEN

We have analyzed the prothrombotic risk factors in 124 patients with ulcerative colitis (UC) as compared with control subjects with other gastrointestinal disorders. The patients were hospitalized from 1991 to 2000 in the Department of Internal Medicine. Platelets level was significantly higher (p < 0.001) in UC patients as compared with control group, and aPTT was significantly (p < 0.05) prolonged respectively in UC patients as compared with control group. In the prospective pilot study we observed the decrease of plasma antithrombin level and/or decrease of protein S in approximately 22% of UC patients. The plasma protein C activity was normal in all UC patients, whereas the mean level of protein S was significantly lower (p < 0.02) in UC patients as compared with controls. Thus our data indicate that coagulation abnormalities are potential risk factors of thromboembolic complications in UC. Plasma cholesterol and triglyceride levels were above upper limit of normal values in 38% and 18% UC patients respectively, but mean values of both parameters were not significantly different between UC and control group. Our results suggest that plasma lipid changes are not independent risk factor of vascular complication in UC. In UC patients smokers were observed 4 times less than no-smokers. However, positive correlation between smoking and rise of hematocrit (F = 4.48; p < 0.05) in UC suggests that smoking may be a risk factor of vascular complications in the disease. Thromboembolic events were found in 1.6% of UC patients. In addition, chronic coronary heart disease was accompanied approximately 6% of UC patients. Evaluation of prothrombotic risk factors and associated ischaemic heart diseases may have prognostic value in the management of UC.


Asunto(s)
Colitis Ulcerosa/complicaciones , Tromboembolia/etiología , Trombofilia/etiología , Reacción de Fase Aguda/complicaciones , Adulto , Anciano , Anciano de 80 o más Años , Deficiencia de Antitrombina III/metabolismo , Colitis Ulcerosa/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Proyectos Piloto , Polonia , Estudios Prospectivos , Deficiencia de Proteína S/metabolismo , Estudios Retrospectivos , Factores de Riesgo , Fumar/efectos adversos , Tromboembolia/metabolismo , Trombofilia/metabolismo
17.
Hum Mutat ; 20(2): 98-109, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12124990

RESUMEN

The inclusion of a mutation in a pathology-based database such as the Human Gene Mutation Database (HGMD) is a two-stage process: first, the mutation must occur at the DNA level, then it must cause a clinically detectable disease state. The likelihood of the latter step, termed the relative clinical observation likelihood (RCOL), can be regarded as a function of the structural/functional consequences of a mutation at the protein level. Following this paradigm, we modeled in silico all amino acid replacements that could potentially have arisen from an inherited single base pair substitution in five human genes encoding arylsulphatase A (ARSA), antithrombin III (SERPINC1), protein C (PROC), phenylalanine hydroxylase (PAH), and transthyretin (TTR). These proteins were chosen on the basis of 1) the availability of a crystallographic structure, and 2) a sufficiently large number of amino acid replacements being logged in HGMD. A total of 9,795 possible mutant structures were modeled and 20 different biophysical parameters assessed. Together with the HGMD-derived spectra of clinically detected mutations, these data allowed maximum likelihood estimation of RCOL profiles for the 20 parameters studied. Nine parameters (including energy difference between wild-type and mutant structures, accessibility of the mutated residue, and distance from the binding/active site) exhibited statistically significant variability in their RCOL profiles, indicating that mutation-associated changes affected protein function. As yet, however, a biological meaning could only be attributed to the RCOL profiles of solvent accessibility and, for three proteins, local energy change, disturbed geometry, and distance from the active center. The limited ability of the biophysical properties of mutations to explain clinical consequences is probably due to our current lack of understanding as to which amino acid residues are critical for protein folding. However, since the proteins examined here were unrelated, and our findings consistent, it may nevertheless prove possible to extrapolate to other proteins whose dysfunction underlies inherited disease.


Asunto(s)
Sustitución de Aminoácidos/genética , Biofisica/métodos , Enfermedades Genéticas Congénitas/genética , Neuropatías Amiloides Familiares/genética , Neuropatías Amiloides Familiares/metabolismo , Antitrombina III/química , Antitrombina III/genética , Antitrombina III/fisiología , Deficiencia de Antitrombina III/genética , Deficiencia de Antitrombina III/metabolismo , Arilsulfatasas/química , Arilsulfatasas/deficiencia , Arilsulfatasas/genética , Arilsulfatasas/fisiología , Bases de Datos Genéticas , Bases de Datos de Proteínas , Enfermedades Genéticas Congénitas/enzimología , Enfermedades Genéticas Congénitas/metabolismo , Genotipo , Humanos , Leucodistrofia Metacromática/enzimología , Leucodistrofia Metacromática/genética , Modelos Genéticos , Mutación Missense/genética , Fenotipo , Fenilalanina Hidroxilasa/química , Fenilalanina Hidroxilasa/deficiencia , Fenilalanina Hidroxilasa/genética , Fenilalanina Hidroxilasa/fisiología , Fenilcetonurias/enzimología , Fenilcetonurias/genética , Prealbúmina/química , Prealbúmina/deficiencia , Prealbúmina/genética , Prealbúmina/fisiología , Proteína C/química , Proteína C/genética , Proteína C/fisiología , Deficiencia de Proteína C/genética , Deficiencia de Proteína C/metabolismo , Estructura Secundaria de Proteína/genética
18.
J Extra Corpor Technol ; 33(4): 245-8, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11806438

RESUMEN

We report a case of heparin resistance and its management during cardiopulmonary bypass (CPB). A 63-year-old, 96 Kg female with a posterior myocardial infarction (MI) with previous deep venous thrombosis was treated with intravenous (IV) heparin infusion for 7 days before myocardial revascularization surgery. The patient required 1200 IU/Kg of beef lung heparin to extend the activated clotting time (ACT) in order to initiate CPB. A total of 1562 IU/Kg of heparin was administered throughout the procedure. This acquired heparin resistance was attributed to an antithrombin (AT III) deficiency, and was treated with fresh frozen plasma (FFP) to restore adequate anticoagulation. The patient's heparinized ACTs ranged between 368 seconds and 387 seconds before FFP administration as opposed to 626 seconds to 1329 seconds after treatment with FFP and additional heparin once on CBP. The patient experienced an uneventful postoperative course. Future treatment with AT III concentrate rather than FFP may reduce heparin requirements that will, in turn, reduce protamine reversal dose, postoperative bleeding attributable to heparin rebound, and its associated complications.


Asunto(s)
Anticoagulantes/administración & dosificación , Deficiencia de Antitrombina III/metabolismo , Trastornos de la Coagulación Sanguínea/tratamiento farmacológico , Monitoreo de Drogas/métodos , Heparina/administración & dosificación , Anticoagulantes/sangre , Transfusión de Componentes Sanguíneos , Puente Cardiopulmonar , Resistencia a Medicamentos , Femenino , Heparina/sangre , Humanos , Persona de Mediana Edad , Revascularización Miocárdica , Tiempo de Coagulación de la Sangre Total
19.
J Biol Chem ; 274(49): 34556-65, 1999 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-10574918

RESUMEN

Heparin cofactor II (HCII) is a serpin whose thrombin inhibition activity is accelerated by glycosaminoglycans. We describe the novel properties of a carboxyl-terminal histidine-tagged recombinant HCII (rHCII-CHis(6)). Thrombin inhibition by rHCII-CHis(6) was increased >2-fold at approximately 5 microgram/ml heparin compared with wild-type recombinant HCII (wt-rHCII) at 50-100 microgram/ml heparin. Enhanced activity of rHCII-CHis(6) was reversed by treatment with carboxypeptidase A. We assessed the role of the HCII acidic domain by constructing amino-terminal deletion mutants (Delta1-52, Delta1-68, and Delta1-75) in wt-rHCII and rHCII-CHis(6). Without glycosaminoglycan, unlike wt-rHCII deletion mutants, the rHCII-CHis(6) deletion mutants were less active compared with full-length rHCII-CHis(6). With glycosaminoglycans, Delta1-68 and Delta1-75 rHCIIs were all less active. We assessed the character of the tag by comparing rHCII-CHis(6), rHCII-CAla(6), and rHCII-CLys(6) to wt-rHCII. Only rHCII-CHis(6) had increased activity with heparin, whereas all three mutants have increased heparin binding. We generated a carboxyl-terminal histidine-tagged recombinant antithrombin III to study the tag on another serpin. Interestingly, this mutant antithrombin III had reduced heparin cofactor activity compared with wild-type protein. In a plasma-based assay, the glycosaminoglycan-dependent inhibition of thrombin by rHCII-CHis(6) was significantly greater compared with wt-rHCII. Thus, HCII variants with increased function, such as rHCII-CHis(6), may offer novel reagents for clinical application.


Asunto(s)
Cofactor II de Heparina/metabolismo , Inhibidores de Serina Proteinasa/metabolismo , Alanina/metabolismo , Anticoagulantes/farmacología , Antitrombina III/genética , Antitrombina III/metabolismo , Deficiencia de Antitrombina III/metabolismo , Antitrombinas/farmacología , Carboxipeptidasas/farmacología , Carboxipeptidasas A , Dermatán Sulfato/farmacología , Inhibidores del Factor Xa , Glicosaminoglicanos/metabolismo , Glicosaminoglicanos/farmacología , Heparina/farmacología , Cofactor II de Heparina/genética , Hirudinas/análogos & derivados , Hirudinas/farmacología , Humanos , Cinética , Lisina/metabolismo , Mutagénesis Sitio-Dirigida , Fragmentos de Péptidos/farmacología , Unión Proteica , Proteínas Recombinantes/metabolismo , Sefarosa/metabolismo , Inhibidores de Serina Proteinasa/genética , Trombina/antagonistas & inhibidores
20.
Fertil Steril ; 71(5): 860-2, 1999 May.
Artículo en Inglés | MEDLINE | ID: mdl-10231046

RESUMEN

OBJECTIVE: To present a case of ovarian hyperstimulation syndrome in which antithrombin III activity in plasma was decreased and in ascites was increased. DESIGN: Case report. SETTING: Hospital-based clinic for reproductive medicine. PATIENT(S): A 27-year-old woman who was transferred to our hospital because of ovarian hyperstimulation syndrome. INTERVENTION(S): Induced abortion. MAIN OUTCOME MEASURE(S): Antithrombin III activity in plasma and ascites. RESULT(S): Antithrombin III activity in ascites was slightly lower than that in plasma. CONCLUSION(S): The loss of antithrombin III into ascites probably caused its deficiency in this case.


Asunto(s)
Deficiencia de Antitrombina III/metabolismo , Antitrombina III/metabolismo , Líquido Ascítico/metabolismo , Síndrome de Hiperestimulación Ovárica/metabolismo , Adulto , Femenino , Humanos , Síndrome de Hiperestimulación Ovárica/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA