Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Clin Invest ; 81(5): 1445-54, 1988 May.
Artículo en Inglés | MEDLINE | ID: mdl-3284913

RESUMEN

Protein S is a vitamin K-dependent plasma protein which serves as the cofactor for activated protein C. Protein S circulates in both an active, free form and in an inactive complex with C4b-binding protein. To elucidate the role of protein S in disease states and during oral anticoagulation, we developed a functional assay for protein S that permits evaluation of the distribution of protein S between free and bound forms and permits determination of the specific activity of the free protein S. In liver disease, free protein S antigen is moderately reduced and the free protein S has significantly reduced specific activity. In disseminated intravascular coagulation, reduced protein S activity occurs due to a redistribution of protein S to the inactive bound form. During warfarin anticoagulation, reduction of free protein S antigen and the appearance of forms with abnormal electrophoretic mobility significantly decrease protein S activity. After the initiation of warfarin, the apparent half-life of protein S is 42.5 h. In patients with thromboembolic disease, transient protein S deficiency occurs due to redistribution to the complexed form. Caution should be exercised in diagnosing protein S deficiency in such patients by use of functional assays.


Asunto(s)
Coagulación Intravascular Diseminada/metabolismo , Hepatopatías/metabolismo , Warfarina/uso terapéutico , Adulto , Anciano , Coagulación Sanguínea , Semivida , Humanos , Técnicas de Inmunoadsorción , Persona de Mediana Edad , Proteína C/metabolismo
2.
J Clin Invest ; 77(2): 416-25, 1986 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-3511097

RESUMEN

Protein C is a natural vitamin K-dependent plasma anticoagulant, deficiencies of which have been found in patients with recurrent thrombosis and warfarin-induced skin necrosis. To appreciate more fully the role of protein C in disease states and during oral anticoagulation, a new functional assay for protein C involving adsorption of plasma protein C on a Ca+2-dependent monoclonal antibody, elution, quantitative activation, and assessment of plasma anticoagulant activity, has been developed. When oral anticoagulation is initiated, the anticoagulant activity of protein C decreases to a greater extent than either the amidolytic or immunologic levels. During stabilized warfarin treatment, there is no correlation between either amidolytic or antigenic levels and the functional protein C activity, suggesting that measurement of protein C anticoagulant activity may be necessary to reflect adequately the anticoagulant protection afforded by this protein. In contrast, there was a strong correlation between anticoagulant and amidolytic and immunologic levels in liver failure and disseminated intravascular coagulation. Two patients with thromboembolic disease have been identified who exhibit a marked decrease in anticoagulant activity, but who have normal immunologic and amidolytic levels. Thus, this assay permits assessment of protein C in individuals who have received anticoagulant treatment and identification of a new class of protein C-deficient individuals.


Asunto(s)
Coagulación Sanguínea , Coagulación Intravascular Diseminada/sangre , Glicoproteínas/sangre , Hepatopatías/sangre , Tromboflebitis/sangre , Warfarina/uso terapéutico , Adulto , Anciano , Antígenos/análisis , Compuestos Cromogénicos , Enfermedad Crónica , Dipéptidos/metabolismo , Factor VII/metabolismo , Factor X/metabolismo , Glicoproteínas/inmunología , Humanos , Técnicas de Inmunoadsorción , Persona de Mediana Edad , Proteína C , Tromboflebitis/tratamiento farmacológico
3.
J Clin Invest ; 79(3): 918-25, 1987 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-3102560

RESUMEN

Gram-negative septicemia elicits multiple abnormalities of the coagulation system. Although products of coagulation can lead to clot formation, thereby potentiating organ damage, recent work has shown that low concentrations of thrombin can protect animals from the shock state. Because these amounts of thrombin also lead to formation in vivo of the anticoagulant enzyme, activated protein C, we examined the role of protein C in modulation of Escherichia coli shock in baboons. First, we infused activated protein C and lethal concentrations of E. coli organisms, which prevented the coagulopathic, hepatotoxic, and lethal effects of E. coli. Second, using an antibody to protein C we blocked protein C activation in vivo to determine if this influenced the response to lethal and sublethal concentrations of E. coli organisms. Under these conditions the response to lethal concentrations of E. coli organisms was made more severe and the response to sublethal concentrations of E. coli was made lethal. The coagulopathic, hepatotoxic, and lethal responses in this latter case were prevented by infusion of exogenous protein C.


Asunto(s)
Trastornos de la Coagulación Sanguínea/etiología , Escherichia coli , Proteína C/fisiología , Choque Séptico/complicaciones , Alanina Transaminasa/metabolismo , Animales , Trastornos de la Coagulación Sanguínea/metabolismo , Activación Enzimática , Factor V/metabolismo , Factor VIII/metabolismo , Fibrinógeno/metabolismo , Recuento de Leucocitos , Hepatopatías/etiología , Hepatopatías/metabolismo , Papio , Choque Séptico/prevención & control
4.
J Thromb Haemost ; 4(1): 186-91, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16409468

RESUMEN

OBJECTIVE: To characterize the first type II protein S (PS) deficiency affecting the epidermal growth factor (EGF)4 domain, a calcium-binding module with a poorly defined functional role. PATIENTS: The proband suffered from recurrent deep vein thrombosis and showed reduced PS anticoagulant activity (31%), and total, free PS antigen and C4bBP levels in the normal range. RESULTS: Reverse transcription-polymerase chain reaction analysis showed the presence of the IVSg-2A/T splicing mutation that, by activating a cryptic splice site, causes the deletion of codons Ile203 and Asp204. Free PS, immunopurified from proband's plasma, showed an altered electrophoretic pattern in native condition or in the presence of Ca2+. The recombinant PS (rPS) mutant showed reduced anticoagulant (<10%) and activated protein C-independent activities (24-38%) when compared with wild-type rPS (rPSwt). Binding of the rPS variant to phospholipid vesicles (Kd 235.7 +/- 30.8 nM, rPSwt; Kd 15.2 +/- 0.9 nM) as well as to Ca2+-dependent conformation-specific monoclonal antibodies for GLA domain was significantly reduced. CONCLUSIONS: These data aid in the characterization of the functional role of the EGF4 domain in the anticoagulant activities of PS and in defining the thrombophilic nature of type II PS deficiency.


Asunto(s)
Deficiencia de Proteína S/genética , Proteína S/química , Eliminación de Secuencia , Adulto , Calcio/farmacología , Proteínas de Unión al Calcio/genética , Proteína de Unión al Complemento C4b/análisis , Factor de Crecimiento Epidérmico/química , Humanos , Proteína S/análisis , Proteína S/genética , Deficiencia de Proteína S/complicaciones , Deficiencia de Proteína S/etiología , Estructura Terciaria de Proteína/genética , Sitios de Empalme de ARN/genética , Recurrencia , Trombosis de la Vena/etiología , Trombosis de la Vena/genética
5.
Thromb Haemost ; 79(6): 1092-5, 1998 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-9657429

RESUMEN

In a prospective longitudinal study, 130 primigravidae at risk for preeclampsia were examined and plasma sampling performed in 45 of them. Plasma thrombomodulin (pTM) was sequentially measured at weeks 12, 24 and 32 of gestation and after delivery in 20 primigravidae who developed either mild preeclampsia (n = 8) or gestational hypertension (n = 12) between weeks 32 and 39 of gestation and in 25 (age-matched) primigravidae who had uneventful pregnancies. pTM elevations were not observed until week 32 in uneventful pregnancies, but were present by week 24 (p = 0.002) in patients who later developed hypertensive complications. A net individual pTM increase > or = 4.2 ng/ml between weeks 12 and 24 (more than 8 times that of normotensive primigravidae) and/or pTM level > or = 47.5 ng/ml at week 32 predicted the development of hypertensive complications with 80% accuracy. Serial pTM determinations can be useful to select pregnancies who may benefit from early pharmacological intervention.


Asunto(s)
Preeclampsia/sangre , Trimestres del Embarazo/sangre , Trombomodulina/sangre , Adulto , Biomarcadores , Creatinina/sangre , Femenino , Humanos , Recién Nacido , Pruebas de Función Renal , Preeclampsia/epidemiología , Valor Predictivo de las Pruebas , Embarazo , Resultado del Embarazo , Estudios Prospectivos , Curva ROC , Factores de Riesgo
6.
Thromb Haemost ; 57(1): 44-8, 1987 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-3590079

RESUMEN

Five functional assays and two immunoassays for protein C (PC) were evaluated in parallel for the same plasma samples collected from healthy subjects, patients with congenital and acquired PC deficiencies or patients with conditions associated with high PC levels. For 7 patients starting warfarin therapy and for 15 patients during stabilized warfarin therapy, there were significant between-assay differences. For these groups immunoassays gave higher values than most functional assays and the latter also gave varied results, probably depending on their respective capacity for recognizing a carboxylated PC. On the other hand, there were no significant between-assay differences nor discrepancies between PC activity and antigen levels for healthy subjects (n = 39), patients with congenital PC deficiency (n = 10), myocardial infarction (n = 25), chronic liver disease (n = 19), disseminated intravascular coagulation (n = 35), in the post-operative period (n = 20) or in women taking oral contraceptives (n = 20). This comparison of PC assays indicates that PC levels measured by different functional or immunological assays are very close in the majority of clinical conditions, but not for patients on oral anticoagulants.


Asunto(s)
Proteína C/análisis , Pruebas de Coagulación Sanguínea/métodos , Femenino , Humanos , Inmunoensayo/métodos , Masculino , Proteína C/genética , Proteína C/inmunología , Deficiencia de Proteína C , Valores de Referencia , Warfarina/uso terapéutico
7.
Thromb Haemost ; 70(6): 946-50, 1993 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-7513091

RESUMEN

Four functional assays for protein S were evaluated by 4 different laboratories, each center using its own method. The aim of this study was to compare these different assays and to establish a relationship with results of immunological assays of total and free protein S antigen and C4bBP. The same plasma samples were distributed to each center and tested in blind. In 47 normal subjects, there was no significant difference between the 4 functional assays, with mean values ranging from 93 to 100%. These values were in good agreement with those of free and total protein S antigen. In 34 patients with a quantitative congenital deficiency of protein S the mean values of protein S activity were decreased with the 4 assays, ranging from 25 to 40%. Free protein S antigen was reduced to a similar extent, whereas total antigen was either normal or decreased. The correlation of protein S activity with free protein S antigen was satisfactory for 3 methods, with coefficients of correlation varying from 0.84 to 0.92 whereas it was only 0.70 in one lab. When total protein S antigen was reduced, protein S activity was decreased in all the patients with the 4 assays. In contrast when total protein S antigen was normal an important overlap of protein S activity between normals and patients was observed in one lab with 12 patients misclassified. In 8 patients with a functional defect, results of protein S activity differed substantially according to the assay used and about half of these patients were misclassified.(ABSTRACT TRUNCATED AT 250 WORDS)


Asunto(s)
Deficiencia de Proteína S , Proteína S/sangre , Administración Oral , Bioensayo , Dicumarol/administración & dosificación , Europa (Continente) , Femenino , Humanos , Recién Nacido , Inflamación/sangre , Masculino , Reproducibilidad de los Resultados
8.
Thromb Haemost ; 57(2): 183-6, 1987 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-3037717

RESUMEN

The partial characterization of a dysfunctional protein C (PC), provisionally named "PC Cádiz", in a 45-year-old male patient suffering from recurrent venous thrombosis is described. The only defect found in laboratory assays for haemostasis and hepatic function was a half normal level of both amidolytic and anticoagulant protein C activity, measured by different functional assays that use thrombin-thrombomodulin complex and a snake venom to activate protein C. Protein C antigen was always found to be within normal levels. Two young daughters of the propositus were found to have the same defect. Double-crossed immunoelectrophoresis, performed in the presence and absence of Ca2+ in the first dimension, showed no clear differences between patient and control PC. PC adsorption to barium salts was also found to be normal. Measurement of the PC activation peptide in the barium citrate eluates after PC activation showed no significant differences between patient and 10 normal controls, the concentration of this peptide being very similar to that of PC zymogen in the same eluates before PC activation. These results indicate that this abnormal PC is able to be normally activated by thrombin-thrombomodulin complex but does not exhibit serine protease activity, probably due to a defect in the PC molecule near the active site center.


Asunto(s)
Proteína C/fisiología , Tromboflebitis/sangre , Humanos , Inmunoelectroforesis , Masculino , Persona de Mediana Edad , Linaje , Proteína C/sangre , Receptores de Superficie Celular , Receptores de Trombina , Trombina , Tromboflebitis/genética
9.
Thromb Haemost ; 72(1): 39-43, 1994 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-7974373

RESUMEN

A series of coagulation parameters and lipoprotein(a) (Lp(a)) were explored in plasma from 40 patients with central retinal vein occlusion (CRVO, non-ischemic type n = 12; ischemic type n = 28) free of local and systemic predisposing factors, 1 to 12 months after the acute event. Forty age- and sex-matched patients with cataract served as controls. Prothrombin fragment 1.2 (F1.2), D-dimer, FVII:C--but not FVII:Ag--were higher and fibrinogen was lower in CRVO patients than in controls. Patients with non-ischemic CRVO had higher F1.2 and FVII:C and lower heparin cofactor II than patients with ischemic CRVO. Lp(a) levels greater than 300 mg/l were observed in 12 patients with CRVO and in 4 controls (30% vs 10%, p < 0.025). Patients with high Lp(a)--consistently associated with the S2 phenotype--had higher FVII:C, FVII:C/Ag ratio, and fibrinogen than the remaining CRVO patients. Plasma F1.2 and D-dimer correlated fairly in controls (r = 0.41) and patients with normal Lp(a) levels (r = 0.55), but they did not in the group of patients with high Lp(a) (r = 0.19), where the latter parameter was negatively related to D-dimer (r = -0.55). There was no dependence of the abnormalities observed on the time elapsed from vein occlusion. The findings of activated FVII and high F1.2, D-dimer, and Lp(a) are not uncommon in patients with CRVO. Increased thrombin formation with fibrin deposition and impaired fibrinolysis may play a role in the pathophysiology of CRVO and require specific treatment.


Asunto(s)
Trastornos de la Coagulación Sanguínea/sangre , Lipoproteína(a)/sangre , Oclusión de la Vena Retiniana/sangre , Adulto , Anciano , Femenino , Humanos , Incidencia , Análisis de los Mínimos Cuadrados , Masculino , Persona de Mediana Edad , Prevalencia , Oclusión de la Vena Retiniana/epidemiología
10.
Thromb Res ; 59(6): 985-94, 1990 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-2148229

RESUMEN

The changes in plasma levels of the vitamin K-dependent natural anticoagulants protein C (PC) and protein S (PS) and procoagulant factors II, IX and X were evaluated in 8 adult patients during treatment with L-asparaginase (L-ase i.v. 120,000 U/m2 over 10 days). PC anticoagulant activity and factor IX, X and II coagulant activity decreased proportionally to their half-lives to a nadir of 50-60% of pretreatment values after 2-5 L-ase infusions, suggesting that inhibition of protein synthesis rather than consumption is the main mechanism responsible for the observed changes. Free PS antigen levels declined at a rate similar to total PS antigen, reaching a nadir of 56% of pretreatment values after 3 L-ase infusions; however due to C4b-binding protein levels higher than total PS levels (p less than 0.05), they were constantly lower than the corresponding total PS antigen levels (0.05 less than p less than 0.001). This implicates that total PS antigen levels cannot be taken as an indicator of PS activity. No differences between the antigenic levels and the anticoagulant activities of PC and free PS could be observed suggesting that L-ase does not affect the mechanisms of vitamin K-dependent carboxylation of Gla-residues. The faster rate of decline of PC and PS activities relative to that of factor II may be responsible for the onset of an hypercoagulable state during the early phase of L-ase treatment.


Asunto(s)
Asparaginasa/uso terapéutico , Coagulación Sanguínea/efectos de los fármacos , Ligasas de Carbono-Carbono , Proteínas Inactivadoras de Complemento , Ligasas/efectos de los fármacos , Proteína C/antagonistas & inhibidores , Adulto , Amidas/metabolismo , Factores de Coagulación Sanguínea/efectos de los fármacos , Proteínas Sanguíneas/efectos de los fármacos , Proteínas Portadoras/efectos de los fármacos , Femenino , Glicoproteínas/efectos de los fármacos , Humanos , Ligasas/metabolismo , Masculino , Persona de Mediana Edad , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Proteína S
11.
Thromb Res ; 80(4): 333-7, 1995 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-8585046

RESUMEN

We describe a case of primary amyloidosis (AL) with severe factor X (FX) deficiency in an amateur cyclist presenting with muscular pain at rest and ecchymoses in his legs. No circulating inhibitor of FX was found by mixing studies and there was no deficiency of other vitamin K-dependent coagulation factors and inhibitors or of alpha 2-antiplasmin. Thrombin-time and reptilase time were abnormally prolonged and were not corrected by mixing with normal plasma. Administration of plasma or prothrombin complex concentrate (PCC) were unsuccessful in controlling bleeding: the apparent half-life of transfused FX was 6 minutes. Resting resulted in cessation of muscular pain and bleeding. Renal and cardiac deterioration led the patient to death 3 years after presentation. No further bleeding manifestations did occur during this period. FX levels remained consistently below 3%, but prothrombin fragment 1.2 and thrombin-antithrombin complex--measured at distance from PCC administration and prior to deterioration of renal and cardiac function--were markedly elevated. At autopsy, disseminated amyloidosis was found with sparing of the skeletal muscles and of the skin. This is the first report of increased in vivo prothrombin activation and activity in AL-associated FX deficiency.


Asunto(s)
Amiloidosis/complicaciones , Deficiencia del Factor X/complicaciones , Hemorragia/etiología , Trombina/metabolismo , Amiloidosis/sangre , Resultado Fatal , Hemorragia/sangre , Humanos , Masculino , Persona de Mediana Edad
12.
Adv Exp Med Biol ; 214: 47-54, 1987.
Artículo en Inglés | MEDLINE | ID: mdl-2959034

RESUMEN

Proteins C and S are two vitamin K-dependent plasma proteins that work in concert as a natural anticoagulant system. Activated protein C is the proteolytic component of the complex and protein S serves as an activated protein C binding protein that is essential for assembly of the anticoagulant complex on cell surfaces. The anticoagulant activity is expressed through the selective inactivation of Factors Va and VIIIa. Many patients deficient in proteins C and S have been described and have an associated thrombotic tendency, but not all heterozygous protein C and S deficient individuals experience thrombotic complications. Multiple mechanisms and/or drugs can lead to acquired deficiencies of these proteins: oral anticoagulation, liver disease, DIC and in the case of protein S, lupus erythematosus, nephrotic syndrome, pregnancy and certain hormones. The anticoagulant activity of protein C decreases rapidly after administration of warfarin (i.e., with a time course similar to Factor VII). This rapid decrease may lead to a transient imbalance and contribute to coumarin induced skin necrosis. Protein S antigen levels do not decrease as rapidly, but protein S functional levels are often low in patients with an acute thrombus. The discrepancy between antigen and function results from elevations in C4b-binding protein, which complexes reversibly with protein S. Unlike free protein S, the complex does not function in the anticoagulant pathway. The available information all suggest that deficiency of protein C and protein S should be considered a risk factor contributing to recurrent thrombotic disease and that the function of these proteins is altered by many common clinical conditions which have associated an increased risk of thrombosis.


Asunto(s)
Anticoagulantes , Coagulación Sanguínea , Glicoproteínas , Proteína C , Humanos , Proteína S , Trombosis/fisiopatología , Warfarina/efectos adversos
17.
Ann Intern Med ; 123(10): 747-53, 1995 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-7574192

RESUMEN

OBJECTIVE: To evaluate the prevalence of moderate hyperhomocysteinemia and inherited thrombophilia disorders (congenital defects of the natural anticoagulant or fibrinolytic mechanisms) in patients with early-onset venous or arterial thromboembolic disease. DESIGN: Cross-sectional 2-year evaluation of consecutive unrelated patients with a history of venous or arterial occlusive disease occurring before the age of 45 years or at unusual sites, in the absence of local predisposing factors. SETTING: Thrombosis research unit of a community hospital. PATIENTS: 107 patients with venous thromboembolism (mean age at event, 32.9 +/- 11.9 years) and 50 patients with arterial occlusive disease (mean age at event, 31.1 +/- 10 years) who did not have acquired coagulation defects, overt cancer, or acquired conditions affecting methionine metabolism. MEASUREMENTS: Total plasma homocysteine (fasting levels), antithrombin III, protein C, protein S, activated protein C resistance, plasminogen, and heparin cofactor II were measured at least 3 months after the event. In 87 patients, total plasma homocysteine levels were also measured 8 hours after an oral methionine load was administered (L-methionine, 0.1 g/kg body weight). Ninety-fifth percentiles of the distribution of these variables were established in 60 apparently healthy persons; sex-specific ranges were used for protein S and total plasma homocysteine. Relatives of patients with laboratory abnormalities were studied to confirm inheritance of the defects. RESULTS: Moderate hyperhomocysteinemia was detected in 13.1% (95% CI, 7.6% to 21.3%) and in 19.2% (CI, 9.0% to 31.9%) of patients with venous or arterial occlusive disease. The prevalence of hyperhomocysteinemia was almost twice as high when based on homocysteine measurements done after oral methionine load as when based on fasting levels. The remaining defects were detected only in patients with venous occlusive disease (activated protein C resistance in 11.2% of patients, protein S or C deficiency in 6.6%, and plasminogen deficiency in 0.9%), with an overall prevalence of 18.7% (CI, 12.1% to 27.6%). Inheritance of hyperhomocysteinemia and of the other defects was confirmed in 26 of the 30 families studied. Event-free survival analysis showed that the relative risk for occlusive disease in patients with moderate hyperhomocysteinemia and other defects was 1.70 times (CI, 1.19 to 2.42; P < 0.01) greater than in patients without defects. After adjustment for the presence of predisposing factors (for example, use of contraceptive drugs, pregnancy, surgery, prolonged bedrest, smoking, mild hypertension or dyslipidemia) and a family history of thrombosis, the age at first event of patients with moderate hyperhomocysteinemia was similar to that of patients with the other defects (26.4 +/- 11.2 years compared with 25.2 +/- 10.6 years), and the 43 patients with defects were significantly younger at first event than the 114 patients without defects (25.5 +/- 11.1 years compared with 31.0 +/- 12.3; P < 0.005). Patients with mild hyperhomocysteinemia had a higher rate of recurrence than those without defects (52% compared with 25%; P = 0.01); among the 56 patients who had their first event more than 1 year before observation, the recurrence rate was higher (80% [CI, 51% to 95%]) in patients with defects than in patients without defects (41% [CI, 26% to 57%] P = 0.01). CONCLUSIONS: Moderate hyperhomocysteinemia may have pathogenic significance in premature venous and arterial occlusive disease and should be included among the (inherited) disorders of venous and arterial thrombophilia.


Asunto(s)
Arteriopatías Oclusivas/sangre , Homocisteína/sangre , Tromboembolia/sangre , Adulto , Estudios Transversales , Femenino , Humanos , Masculino , Metionina , Factores de Riesgo , Factores Sexuales , Tromboembolia/genética
18.
Haematologica ; 82(2): 211-9, 1997.
Artículo en Inglés | MEDLINE | ID: mdl-9175329

RESUMEN

BACKGROUND AND OBJECTIVE: In spite of the large number of reports showing that hyperhomocysteinemia (HHcy) is an independent risk factor for atherosclerosis and arterial occlusive disease, this metabolite of the methionine pathway is measured in relatively few laboratories and its importance is not fully appreciated. Recent data strongly suggest that mild HHcy is also involved in the pathogenesis of venous thromboembolic disease. The aim of this paper is to analyze the most recent advances in this field. EVIDENCE AND INFORMATION SOURCES: The material examined in the present review includes articles and abstracts published in journals covered by the Science Citation Index and Medline. In addition the authors of the present article have been working in the field of mild HHcy as cause of venous thromboembolic disease. STATE OF ART AND PERSPECTIVES: The studies examined provide very strong evidence supporting the role of moderate HHcy in the development of premature and/or recurrent venous thromboembolic disease. High plasma homocysteine levels are also a risk factor for deep vein thrombosis in the general population. Folic acid fortification of food has been proposed as a major tool for reducing coronary artery disease mortality in the United States. Vitamin supplementation may also reduce recurrence of venous thromboembolic disease in patients with HHcy. At the present time, however, the clinical efficacy of this approach has not been tested. In addition, the bulk of evidence indicates that fasting total homocysteine determinations can identify up to 50% of the total population of hyperhomocysteinemic subjects. Patients with isolated methionine intolerance may benefit from vitamin B6 supplementation. Homocysteine-lowering vascular disease prevention trials are urgently needed. Such controlled studies, however, should not focus exclusively on fasting homocysteine determinations and folic acid monotherapy.


Asunto(s)
Homocisteína/sangre , Tromboflebitis/etiología , Humanos , Factores de Riesgo
19.
Minerva Anestesiol ; 70(5): 339-50, 2004 May.
Artículo en Italiano | MEDLINE | ID: mdl-15181414

RESUMEN

The last few years have clarified the tight link between inflammation and coagulation. In addition to the identification of new regulatory mechanisms of the coagulation system and of an explosive number of mediators of inflammation, it is now clear that the existence of a positive feed-back between inflammation and coagulation leads to reciprocal activation of both pathways. Plasma levels of acute phase proteins involved in coagulation and fibrinolysis are elevated during inflammation, while natural anticoagulant mechanisms are depressed. Pro-inflammatory cytokines "activate" cell membranes exposed to flowing blood (endothelium, platelets, monocytes, neutrophils) which from physiologically inert or anticoagulant become procoagulant. Increased tissue factor expression results in increased thrombin formation within the microcirculation. Thrombin is central to fibrin deposition but it also plays a key role in cell-mediated mechanisms involving inflammation, cell proliferation and activation of the natural anticoagulant protein C. Depression of natural anticoagulant mechanisms, occurring in severe sepsis, results in uncontrolled thrombin formation, with pro-inflammatory activity prevailing, and the feed-back loop of inflammation and coagulation ultimately leading to multi-organ failure. However, both in the clinical setting and in animal experiments, heparin or direct anticoagulants have shown no effect on survival even if blocking fibrin deposition. Organ failure is only partially due to the thrombotic occlusion of the microcirculation, while other mechanisms of endothelial damage are probably more relevant in the development of ischemia. The endothelium is central to the maintenance of the natural anticoagulant mechanisms (TFPI, antithrombin, protein C). The protein C system, in addition to dumping thrombin formation, specifically modulates inflammation by cell signaling. This system is markedly depressed in severe sepsis. The infusion of activated protein C, or restoring normal levels of protein C within the circulation - depending on the individual bleeding risk are powerful tools to treat the endothelitis responsible for the clinical sequelae of severe sepsis.


Asunto(s)
Coagulación Sanguínea , Proteína C/fisiología , Sepsis/sangre , Animales , Endotelio Vascular , Humanos , Inflamación/etiología , Sepsis/tratamiento farmacológico , Sepsis/inmunología , Índice de Severidad de la Enfermedad
20.
Int J Clin Lab Res ; 21(2): 171-5, 1991.
Artículo en Inglés | MEDLINE | ID: mdl-1815762

RESUMEN

C4b-binding protein (C4b-BP) is a high molecular weight plasma protein which inhibits the activity of the classical complement pathway C3 convertase. In addition to multiple binding sites for C4b, C4b-BP possesses a single binding site for vitamin K-dependent protein S, an inhibitor of blood coagulation. As protein S bound to C4b-BP has no anticoagulant activity, C4b-BP participates in the regulation of both the complement and the coagulation pathways. We have produced and immunochemically characterized a series of murine monoclonal antibodies to human C4b-BP. A mixture of four monoclonal antibodies precipitating C4b-BP both in agarose gel and in solution was used to develop a highly reproducible radial immunodiffusion method for the measurement of C4b-BP in human serum. C4b-BP levels were measured in sera from 284 patients referred to our central laboratory. Samples from subjects with an increased erythrocyte sedimentation rate (ESR), a1-acid glycoprotein (a1-AGP) or C-reactive protein (CRP) had significantly higher C4b-BP levels (307 mg/l, 292-322 mg/l, geometric mean and 95% confidence limits of the mean) than those from subjects without elevation of the aforementioned established acute phase reactants (231 mg/l, 226-237 mg/l, P less than 0.00001). C4b-BP was significantly (P less than 0.001) correlated with ESR (r = 0.715), a1-AGP (r = 0.692) and CRP (r = 0.567). There was no gender-related difference in C4b-BP levels. In subjects with no increased acute phase reactants there was a significant correlation between C4b-BP levels and age (r = 0.387, P less than 0.001). High C4b-BP might contribute to the increased thrombotic risk associated with inflammation and aging.


Asunto(s)
Proteínas Portadoras/análisis , Complemento C4b/metabolismo , Proteínas Inactivadoras de Complemento , Glicoproteínas , Inflamación/sangre , Receptores de Complemento/análisis , Adulto , Factores de Edad , Animales , Anticuerpos Monoclonales/inmunología , Sedimentación Sanguínea , Proteína C-Reactiva/análisis , Proteínas Portadoras/inmunología , Femenino , Humanos , Inmunodifusión , Inmunoglobulina G/inmunología , Cadenas kappa de Inmunoglobulina/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Persona de Mediana Edad , Orosomucoide/análisis , Receptores de Complemento/inmunología , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA