Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Mol Genet Metab ; 114(2): 129-37, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25467058

ABSTRACT

BACKGROUND: Antibody formation can interfere with effects of enzyme replacement therapy (ERT) in lysosomal storage diseases. Biomarkers are used as surrogate marker for disease burden in MPS I, but large systematic studies evaluating the response of biomarkers to ERT are lacking. We, for the first time, investigated the response of a large panel of biomarkers to long term ERT in MPS I patients and correlate these responses with antibody formation and antibody mediated cellular uptake inhibition. METHODS: A total of 428 blood and urine samples were collected during long-term ERT in 24 MPS I patients and an extensive set of biomarkers was analyzed, including heparan sulfate (HS) and dermatan sulfate (DS) derived disaccharides; total urinary GAGs (DMBu); urinary DS:CS ratio and serum heparin co-factor II thrombin levels (HCII-T). IgG antibody titers and the effect of antibodies on cellular uptake of the enzyme were determined for 23 patients. RESULTS: Median follow-up was 2.3 years. In blood, HS reached normal levels more frequently than DS (50% vs 12.5%, p=0.001), though normalization could take several years. DMBu normalized more rapidly than disaccharide levels in urine (p=0.02). Nineteen patients (83%) developed high antibody titers. Significant antibody-mediated inhibition of enzyme uptake was observed in 8 patients (35%), and this correlated strongly with a poorer biomarker response for HS and DS in blood and urine as well as for DMBu, DS:CS-ratio and HCII-T (all p<0.006). CONCLUSIONS: This study shows that, despite a response of all studied biomarkers to initiation of ERT, some biomarkers were less responsive than others, suggesting residual disease activity. In addition, the correlation of cellular uptake inhibitory antibodies with a decreased biomarker response demonstrates a functional role of these antibodies which may have important clinical consequences.


Subject(s)
Biomarkers/analysis , Enzyme Replacement Therapy , Iduronidase/immunology , Iduronidase/therapeutic use , Immunoglobulin G/blood , Mucopolysaccharidosis I/drug therapy , Mucopolysaccharidosis I/immunology , Adolescent , Adult , Child , Child, Preschool , Dermatan Sulfate/analysis , Disaccharides/analysis , Disaccharides/blood , Disaccharides/urine , Female , Follow-Up Studies , Heparin Cofactor II/analysis , Heparitin Sulfate/analysis , Heparitin Sulfate/blood , Heparitin Sulfate/urine , Humans , Infant , Infant, Newborn , Male , Mucopolysaccharidosis I/blood , Mucopolysaccharidosis I/urine , Recombinant Proteins/immunology , Recombinant Proteins/therapeutic use , Thrombin/analysis , Young Adult
2.
J Diabetes Investig ; 12(12): 2172-2182, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34043882

ABSTRACT

AIMS/INTRODUCTION: Thrombin exerts various pathophysiological functions by activating protease-activated receptors (PARs). Recent data have shown that PARs influence the development of glomerular diseases including diabetic kidney disease (DKD) by regulating inflammation. Heparin cofactor II (HCII) specifically inactivates thrombin; thus, we hypothesized that low plasma HCII activity correlates with DKD development, as represented by albuminuria. MATERIALS AND METHODS: Plasma HCII activity and spot urine biomarkers, including albumin and liver-type fatty acid-binding protein (L-FABP), were determined as the urine albumin-to-creatinine ratio (uACR) and the urine L-FABP-to-creatinine ratio (uL-FABPCR) in 310 Japanese patients with diabetes mellitus (176 males and 134 females). The relationships between plasma HCII activities and those DKD urine biomarkers were statistically evaluated. In addition, the relationship between plasma HCII activities and annual uACR changes was statistically evaluated for 201/310 patients (115 males and 86 females). RESULTS: The mean plasma HCII activity of all participants was 93.8 ± 17.7%. Multivariate-regression analysis including confounding factors showed that plasma HCII activity independently contributed to the suppression of the uACR and log-transformed uACR values (P = 0.036 and P = 0.006, respectively) but not uL-FABPCR (P = 0.541). In addition, plasma HCII activity significantly and inversely correlated with annual uACR and log-transformed uACR increments after adjusting for confounding factors (P = 0.001 and P = 0.014, respectively). CONCLUSIONS: The plasma HCII activity was inversely and specifically associated with glomerular injury in patients with diabetes. The results suggest that HCII can serve as a novel predictive factor for early-stage DKD development, as represented by albuminuria.


Subject(s)
Albuminuria/blood , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/urine , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/urine , Heparin Cofactor II/analysis , Adult , Aged , Albumins/metabolism , Albuminuria/urine , Biomarkers/blood , Biomarkers/urine , Creatinine/urine , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 2/complications , Diabetic Nephropathies/etiology , Fatty Acid-Binding Proteins/urine , Female , Humans , Male , Middle Aged , Receptors, Proteinase-Activated/blood , Regression Analysis , Thrombin/metabolism
3.
Mol Genet Metab ; 99(3): 269-74, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19926322

ABSTRACT

Mucopolysaccharide (MPS) diseases are lysosomal storage disorders caused by deficiencies of enzymes catabolising glycosaminoglycans (GAGs). Abnormal GAG accumulation leads to symptoms including severe progressive neurological decline, skeletal deformities, organomegally, respiratory compromise and premature death. Treatment is available for some MPS diseases; enzyme replacement therapy for MPS I, II and VI, and haematopoietic stem cell transplantation for MPS I, VI and VII. These treatments are reliant on early diagnosis of the disease and accurate monitoring of treatment outcomes. Blood enzyme levels and total urinary GAGs are commonly used biomarkers in diagnosis of MPS but are not good measures of treatment outcome. Serum heparin cofactor II-thrombin complex (HCII-T), which is a GAG regulated serpin-protease complex, has recently been identified as a promising biomarker for MPS diseases. Here we present an assessment of the HCII-T biomarker in mouse models of MPS I, IIIA and IIIB, which suggests that HCII-T is a reliable marker for MPS I when measured in serum or dried blood spots stored for over a year at 4 degrees C, but that murine MPS IIIA and IIIB cannot be reliably detected using this biomarker. We also show that HCII-T formation in vivo is dependent on the presence of excess intravenous dermatan sulphate (DS), whilst intravenous heparan sulphate (HS), does not promote complex formation effectively. This suggests that HCII-T will prove effective as a biomarker for MPS I, II, VI and VII diseases, storing dermatan sulphate but may not be as appropriate for MPS III, storing heparan sulphate. With careful sample preparation, HCII-T ELISA could prove to be a useful biomarker for both newborn screening and measurement of treatment outcomes in selected MPS diseases.


Subject(s)
Biomarkers/blood , Heparin Cofactor II/metabolism , Mucopolysaccharidosis III/diagnosis , Mucopolysaccharidosis I/diagnosis , Thrombin/metabolism , Animals , Biomarkers/metabolism , Blood Specimen Collection/methods , Dermatan Sulfate/blood , Dermatan Sulfate/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Heparin Cofactor II/analysis , Heparitin Sulfate/blood , Heparitin Sulfate/metabolism , Humans , Mice , Mice, Inbred C57BL , Mucopolysaccharidosis I/blood , Mucopolysaccharidosis I/metabolism , Mucopolysaccharidosis III/blood , Mucopolysaccharidosis III/metabolism , Thrombin/analysis
4.
Exp Mol Pathol ; 87(3): 178-83, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19747479

ABSTRACT

Heparin cofactor II (HCII) is a serine protease inhibitor (serpin) that has been shown to be a predictor of decreased atherosclerosis in the elderly and protective against atherosclerosis in mice. HCII inhibits thrombin in vitro and HCII-thrombin complexes have been detected in human plasma. Moreover, the mechanism of protection against atherosclerosis in mice was determined to be the inhibition of thrombin. Despite this evidence, the presence of HCII in human atherosclerotic tissue has not been reported. In this study, using samples of coronary arteries obtained from the Pathobiological Determinants of Atherosclerosis in Youth (PDAY) study, we explore the local relationship between HCII and (pro)thrombin in atherosclerosis. We found that HCII and (pro)thrombin are co-localized in the lipid-rich necrotic core of atheromas. A significant positive correlation between each protein and the severity of the atherosclerotic lesion was present. These results suggest that HCII is in a position to inhibit thrombin in atherosclerotic lesions where thrombin can exert a proatherogenic inflammatory response. However, these results should be tempered by the additional findings from this, and other studies, that indicate the presence of other plasma proteins (antithrombin, albumin, and alpha(1)-protease inhibitor) in the same localized region of the atheroma.


Subject(s)
Atherosclerosis/metabolism , Coronary Vessels/metabolism , Heparin Cofactor II/metabolism , Prothrombin/metabolism , Adolescent , Adult , Animals , Atherosclerosis/pathology , Coronary Vessels/pathology , Heparin Cofactor II/analysis , Humans , Immunohistochemistry , Mice , Necrosis , Prothrombin/analysis , Prothrombin/antagonists & inhibitors , Young Adult
5.
Mol Genet Metab ; 94(4): 456-461, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18511319

ABSTRACT

The mucopolysaccharidoses are a group of lysosomal storage disorders caused by defects in the degradation of glycosaminoglycans. Each disorder is characterized by progressive multi-system disease with considerable clinical heterogeneity. The clinical heterogeneity of these disorders is thought to be related to the degree of the metabolic block in glycosaminoglycan degradation which in turn is related to the underlying mutation at the respective locus. There are currently no objective means other than longitudinal clinical observation, or the detection of a recurrent genetic mutation to accurately predict the clinical course for an individual patient, particularly when diagnosed early. In addition, there are no specific disease biomarkers that reflect the total body burden of disease. The lack of specific biomarkers has made monitoring treatment responses and predicting disease course difficult in these disorders. The recent introduction of enzyme replacement therapy for MPS I, II, and VI highlights the need for objective measures of disease burden and disease responsiveness. We show that serum levels of heparin cofactor II-thrombin complex is a reliable biomarker of the mucopolysaccharidoses. Untreated patients have serum levels that range from 3- to 112-fold above control values. In a series of patients with varying severity of mucopolysaccharidosis I, the serum complex concentration was reflective of disease severity. In addition, serum heparin cofactor II-thrombin levels showed responsiveness to various treatment regimens. We propose that serum levels of heparin cofactor II-thrombin complex may provide an important assessment and monitoring tool for patients with mucopolysaccharidosis.


Subject(s)
Biomarkers/blood , Heparin Cofactor II/metabolism , Mucopolysaccharidoses/blood , Thrombin/metabolism , Animals , Blood Chemical Analysis , Enzyme-Linked Immunosorbent Assay , Female , Heparin Cofactor II/analysis , Humans , Longitudinal Studies , Male , Mice , Mucopolysaccharidoses/therapy , Thrombin/analysis
6.
Chin J Nat Med ; 16(4): 241-251, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29703324

ABSTRACT

Respiratory syncytial virus (RSV) is a leading cause of acute lower respiratory tract infections. Qingfei oral liquid (QFOL), a traditional Chinese medicine, is widely used in clinical treatment for RSV-induced pneumonia. The present study was designed to reveal the potential targets and mechanism of action for QFOL by exploring its influence on the host cellular network following RSV infection. We investigated the serum proteomic changes and potential biomarkers in an RSV-infected mouse pneumonia model treated with QFOL. Eighteen BALB/c mice were randomly divided into three groups: RSV pneumonia model group (M), QFOL-treated group (Q) and the control group (C). Serum proteomes were analyzed and compared using a label-free quantitative LC-MS/MS approach. A total of 172 protein groups, 1009 proteins, and 1073 unique peptides were successfully identified. 51 differentially expressed proteins (DEPs) were identified (15 DEPs when M/C and 43 DEPs when Q/M; 7 DEPs in common). Classification and interaction network showed that these proteins participated in various biological processes including immune response, blood coagulation, complement activation, and so forth. Particularly, fibrinopeptide B (FpB) and heparin cofactor II (HCII) were evaluated as important nodes in the interaction network, which was closely involved in coagulation and inflammation. Further, the FpB level was increased in Group M but decreased in Group Q, while the HCII level exhibited the opposite trend. These findings not only indicated FpB and HCII as potential biomarkers and targets of QFOL in the treatment of RSV pneumonia, but also suggested a regulatory role of QFOL in the RSV-induced disturbance of coagulation and inflammation-coagulation interactions.


Subject(s)
Biomarkers/blood , Drugs, Chinese Herbal/pharmacology , Fibrinopeptide B/analysis , Heparin Cofactor II/analysis , Proteome/drug effects , Proteomics , Respiratory Syncytial Virus Infections/blood , Respiratory Syncytial Viruses/drug effects , Animals , Chromatography, Liquid , Disease Models, Animal , Drugs, Chinese Herbal/therapeutic use , Fibrinopeptide B/genetics , Gene Expression Regulation/drug effects , Heparin Cofactor II/genetics , Lung/pathology , Mice, Inbred BALB C , Respiratory Syncytial Virus Infections/drug therapy , Tandem Mass Spectrometry
8.
Circulation ; 109(4): 481-6, 2004 Feb 03.
Article in English | MEDLINE | ID: mdl-14744972

ABSTRACT

BACKGROUND: Thrombin plays an important role in the development of atherosclerosis and restenosis after percutaneous coronary intervention. Because heparin cofactor II (HCII) inhibits thrombin action in the presence of dermatan sulfate, which is abundantly present in arterial wall, HCII may affect vascular remodeling by modulating thrombin action. We hypothesized that patients with high plasma HCII activity may show a reduced incidence of in-stent restenosis (ISR). METHODS AND RESULTS: Sequential coronary arteries (n=166) with NIR stent (Boston Scientific Corp) implantation in 134 patients were evaluated before, immediately after, and at 6 months after percutaneous coronary intervention. Patients were divided into the following groups: high HCII (> or =110%, 45 lesions in 36 patients), normal HCII (> or =80% and <110%, 81 lesions in 66 patients), and low HCII (<80%, 40 lesions in 32 patients). Percent diameter stenosis at follow-up in the high-HCII group (18.7%) was significantly lower (P=0.046) than that in the normal-HCII group (30.3%) or the low-HCII group (29.0%). The ISR rate in the high-HCII group (6.7%) was significantly lower than that in the low-HCII group (30.0%) (P=0.0039). Furthermore, multivariate analysis demonstrated that high plasma HCII activity is an independent factor in reducing the incidence of angiographic restenosis (odds ratio, 0.953/1% increase of HCII; 95% CI, 0.911 to 0.998). CONCLUSIONS: The results demonstrate that HCII may have a hitherto unrecognized effect in inhibiting ISR. The effect of HCII may be mediated by inactivating thrombin in injured arteries, thereby inhibiting vascular smooth muscle cell migration and proliferation.


Subject(s)
Angioplasty, Balloon, Coronary , Coronary Restenosis/epidemiology , Heparin Cofactor II/analysis , Stents/adverse effects , Aged , Coronary Angiography , Coronary Restenosis/diagnosis , Coronary Restenosis/etiology , Female , Humans , Incidence , Male , Middle Aged , Risk Factors
9.
Circulation ; 109(22): 2761-5, 2004 Jun 08.
Article in English | MEDLINE | ID: mdl-15148272

ABSTRACT

BACKGROUND: Thrombin plays a crucial role in atherothrombotic changes. Because heparin cofactor II (HCII) inhibits thrombin actions after binding to dermatan sulfate at injured arterial walls, HCII may negatively regulate thrombin actions in vascular walls. We hypothesized that plasma HCII activity is a preventive factor against atherosclerotic changes, especially in elderly individuals who already have atherosclerotic vascular injuries. METHODS AND RESULTS: Maximum plaque thickness (MPT) in the carotid artery was measured by ultrasonography in 306 Japanese elderly individuals (154 men and 152 women; age, 40 to 91 years; 68.9+/-11.1 years, mean+/-SD). The relevance of cardiovascular risk factors including plasma HCII activity to the severity of MPT was statistically evaluated. Plasma HCII activity decreased with age. Simple linear regression analysis after adjustments for age and sex showed that lipoprotein(a), glycosylated hemoglobin A1c, and presence of diabetes mellitus significantly contributed to an increase in MPT values (r=0.119, P<0.05; r=0.196, P<0.001; and r=0.227, P<0.0001, respectively). In contrast, high-density lipoprotein (HDL) cholesterol and HCII activity were negatively correlated with MPT values (r=-0.117, P<0.05, and r=-0.202, P<0.0005, respectively). Multiple regression analysis revealed that plasma HCII activity and HDL cholesterol independently contributed to the suppression of MPT values and that the antiatherogenic contribution of HCII activity was stronger than that of HDL cholesterol (P<0.001 and P<0.05, respectively). CONCLUSIONS: These results suggest that HCII can be a novel and independent antiatherogenic factor. Moreover, HCII is a stronger predictive factor than HDL cholesterol against carotid atherosclerosis in elderly individuals.


Subject(s)
Carotid Artery Diseases/blood , Heparin Cofactor II/physiology , Adult , Aged , Aged, 80 and over , Aging/blood , Antithrombins/analysis , Cardiovascular Diseases/blood , Carotid Artery Diseases/diagnostic imaging , Female , Heparin Cofactor II/analysis , Humans , Male , Middle Aged , Regression Analysis , Risk Factors , Severity of Illness Index , Ultrasonography
10.
Eur J Obstet Gynecol Reprod Biol ; 119(2): 185-8, 2005 Apr 01.
Article in English | MEDLINE | ID: mdl-15808377

ABSTRACT

OBJECTIVES: (1) To study the changes in the haemostatic variables during normal pregnancy, and (2) to compare them with the corresponding variables in a control group of nonpregnant women. SETTING: University Hospital. SUBJECTS: The study involves two groups of women: pregnant women (in 35th-40th gestational weeks (GW)) (N = 35) and a control group of nonpregnant women (N = 35). METHODS: We examined the global tests, the variables relating to coagulation and fibrinolysis and the group of natural inhibitors of coagulation, and compared them between the two groups. RESULTS: The pregnant women had statistically significantly higher values for: prothrombin time (PT) (P < 0.0001), thrombin time (TT) (P < 0.0001), fibrinogen (P < 0.0001), activity of factor VII (P < 0.0001), factor X (P < 0.0001) and alpha2-antiplasmin (P < 0.002), plasma concentration of D-dimer (plsDD) (P < 0.0001) and activity of heparin cofactor II (HCII) (P < 0.002). They had statistically significantly lower activity of protein C (PrC) (P < 0.0001) and of total protein S (TPrS) (P < 0.0001). CONCLUSION: During normal pregnancy the activation of coagulation is counterbalanced by the activation of fibrinolysis, which maintains the haemostatic balance.


Subject(s)
Hemostasis , Pregnancy/blood , Factor VII/analysis , Factor X/analysis , Female , Fibrin Fibrinogen Degradation Products/analysis , Fibrinogen/analysis , Gestational Age , Heparin Cofactor II/analysis , History, 16th Century , Humans , Protein C/analysis , Protein S/analysis , Prothrombin Time , Reference Values , Thrombin Time , alpha-2-Antiplasmin/analysis
11.
Thromb Haemost ; 92(5): 1108-13, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15543340

ABSTRACT

High heparin cofactor II (HCII) activity has recently been described to protect from coronary instent restenosis, presumably by inactivating thrombin in injured arteries. In this study, we investigated the association of HCII activity and restenosis after femoropopliteal stenting. We studied 63 consecutive patients with peripheral artery disease who underwent femoropopliteal stent implantation after initial failure of plain balloon angioplasty due to a significant residual stenosis (>30% lumen diameter reduction) or a flow limiting dissection. HCII activity was measured before stenting and patients were followed for median 10 months (interquartile range 6 to 17) for the occurrence of a first instent restenosis, defined as a >50% lumen diameter reduction by color coded duplex sonography and confirmed by angiography. Cumulative freedom from restenosis at 6 and 12 months in patients with lower HCII activity (100%, lower tertile, n=20) was 84% and 35% as compared to 93% and 72% in patients with high HCII activity (>100%, middle and upper tertile, n=43; p=0.024 by Log Rank test). Adjusting for the material of the implanted stents (nitinol vs. Wallstents), patients with a high HCII activity had a 0.39-fold reduced risk for instent restenosis (95% CI 0.17 to 0.90, p=0.028), additional adjustment for diabetes mellitus, poor run-off, critical limb ischemia and cumulative length of the stented segment did not alter the observed effect. Higher activity of heparin cofactor II may exert a protective effect against instent restenosis also in the femoropopliteal vessel area, confirming a prior observation after coronary stenting.


Subject(s)
Arterial Occlusive Diseases/surgery , Coronary Restenosis/etiology , Heparin Cofactor II/physiology , Popliteal Artery/surgery , Stents/adverse effects , Aged , Arterial Occlusive Diseases/complications , Cohort Studies , Female , Follow-Up Studies , Heparin Cofactor II/analysis , Humans , Male , Middle Aged , Popliteal Artery/pathology , Prospective Studies , Risk Factors
12.
Thromb Haemost ; 69(3): 231-5, 1993 Mar 01.
Article in English | MEDLINE | ID: mdl-8470046

ABSTRACT

OBJECTIVES: To determine whether functional antithrombin III (AT-III) levels measured by a factor Xa inhibition (AT-III-Xa) assay identifies AT-III deficient individuals more reliably than functional AT-III levels measured by a thrombin inhibition (AT-III-IIa) assay. STUDY DESIGN: Cross-sectional study. PATIENT POPULATION: Sixty-seven members of a large family with type 2 AT-III deficiency. INTERVENTION: DNA analysis was used as the reference diagnostic standard for AT-III status and subjects were classified as AT-III deficient or non deficient according to these results. Functional AT-III levels were measured in all subjects using: 1) a chromogenic substrate for thrombin and added human thrombin (AT-III-IIa), and 2) a chromogenic substrate for factor Xa and added bovine factor Xa (AT-III-Xa). Functional heparin cofactor II (HC-II) levels were measured using a commercially available kit. The proportions of 125I-alpha-thrombin complexed to AT-III and HC-II were measured by polyacrylamide gel electrophoresis and autoradiography. RESULTS: Thirty-one (46%) individuals were classified as AT-III deficient and 36 (54%) as AT-III non deficient. AT-III-Xa assay measured a significantly lower mean AT-III value and a narrower range for individuals classified as AT-III deficient than the AT-III-IIa assay. Using the AT-III-IIa assay, six subjects had borderline AT-III levels compared to none with the AT-III-Xa assay.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Antithrombin III/analysis , Blood Coagulation Disorders/diagnosis , Factor Xa Inhibitors , Adolescent , Adult , Aged , Antithrombin III Deficiency , Blood Coagulation Disorders/genetics , Child , Child, Preschool , Consanguinity , Evaluation Studies as Topic , Female , Heparin Cofactor II/analysis , Humans , Male , Middle Aged , Prothrombin/antagonists & inhibitors , Sensitivity and Specificity
13.
Thromb Haemost ; 81(4): 571-5, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10235441

ABSTRACT

Recent observations describe an increase in platelet aggregability and a decrease in fibrinolytic activity in the early morning hours. To determine whether anticoagulant proteins also show such a circadian variation we measured protein C (PC), protein S (PS), antithrombin (AT) and heparin cofactor-II (HC-II) levels on venous plasma samples taken from 10 healthy men at three-hour intervals throughout a 24-hour period. To investigate the possible temporal mapping of circadian periodicity, we also measured plasma levels of beta-thromboglobulin (beta-TG) as an indicator of platelet activation, and interleukin-6 (IL-6) as one of the possible regulatory factors that drive this rhythm. Blood samples were drawn at 6 a.m., 9 a.m., noon, 3 p.m., 6 p.m., 9 p.m. and midnight. PC, IL-6 and beta-TG were measured by ELISA, PS and AT by latex immune assay and HC-II by chromogenic substrate method. A significant circadian variation was found in PC, PS, AT, beta-TG and IL-6, but not in HC-II levels. PC, PS, IL-6 and beta-TG were at their peaks at 6 a.m., and nadirs at a time from noon to midnight. AT peak was at 6 p.m. and nadir at noon. The regression of PS on IL-6 was significant. Although the fluctuations of PS and AT were within the normal ranges during the day, some PC levels of two subjects were below the lower normal limit (0.70). These data indicate that PC, PS, and AT show a marked circadian periodicity as the other components of the blood coagulation and fibrinolytic system do. The similar trends in plasma concentrations of PC, PS, beta-TG and IL-6 may be coincidental, but could reflect a common regulatory mechanism or an effect on each other. The clinical implications of these physiological changes in coagulation inhibitors and the role of IL-6 in the anticoagulant response deserve further studies.


Subject(s)
Antithrombins/analysis , Blood Coagulation/drug effects , Circadian Rhythm , Interleukin-6/pharmacology , Protein C/analysis , Protein S/analysis , Adult , Anticoagulants/blood , Heparin Cofactor II/analysis , Humans , Male , Serine Proteinase Inhibitors/blood , Statistics as Topic , beta-Thromboglobulin/analysis
14.
Thromb Haemost ; 62(3): 856-60, 1989 Nov 24.
Article in English | MEDLINE | ID: mdl-2595660

ABSTRACT

Extrinsic coagulation pathway inhibitor may be an important regulator of haemostasis to prevent thrombosis after tissue damage. The functional activity of this inhibitor was determined using a chromogenic substrate assay, and compared to the activities of antithrombin, heparin cofactor II and protein C during the perioperative period of elective hip replacement (n = 28), cholecystectomy (n = 11), and vascular surgery (n = 5). Peroperatively, all the inhibitors decreased rather similarly and to the same degree as the decrease in albumin concentration. The decreases during hip surgery were about 2-fold the decreases observed during cholecystectomy. A significant peroperative increase in extrinsic pathway inhibitor activity was observed in vascular surgery, probably due to a bolus injection of heparin. Antithrombin, heparin cofactor II and protein C levels normalized on days 3-5 postoperatively in all three patient groups. Sustained low levels of extrinsic pathway inhibitor were observed on postoperative days 1 to 7 in hip surgery patients. Apparently, extrinsic pathway inhibitor is not an acute phase reactant. In uncomplicated surgery, the decreases of the coagulation inhibitor levels are mainly due to hemodilution.


Subject(s)
Factor VII/analysis , Lipoproteins/analysis , Protease Inhibitors/analysis , Thromboplastin/analysis , Adult , Aged , Factor VII/antagonists & inhibitors , Female , Hematocrit , Heparin Cofactor II/analysis , Hip/surgery , Humans , Male , Middle Aged , Postoperative Complications/etiology , Protein C/analysis , Serum Albumin/analysis , Thromboplastin/antagonists & inhibitors , Thrombosis/etiology
15.
Leuk Lymphoma ; 33(3-4): 361-4, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10221516

ABSTRACT

In this study, protein C (PC), protein S (PS), heparin cofactor II (HCFII), prothrombin fragment 1+2 (PF 1,2), thrombin-antithrombin III complex (TAT), von Willebrand factor (vWF) and thrombomodulin (TM) were investigated in 19 patients with acute lymphoblastic leukemia, (ALL) receiving combined chemotherapy including L-asparaginase (L-ASP) and high dose methylprednisolone (HDMP). HDMP was administered in doses of 30 mg/kg/day for 7 days, and 20 mg/kg/day for another 7 days. In order to evaluate the effect of HDMP on the hemostatic system, the 8 patients studied here received HDMP (30 mg/kg/day) therapy for 4 days before the combined chemotherapy. These parameters were also studied in 12 healthy children as a control group. PC levels were normal in the patients while PS levels were decreased both before and after combined chemotherapies. Patients with ALL have laboratory signs of coagulation activation such as PF 1,2, TAT prior to initiation of chemotherapy. With combined chemotherapy, TAT levels were found to be normal while PF1,2 were not. TM levels were found to be increased both before and after therapies whereas HCFII and vWF levels were not different from those of the control group. The short course of HDMP therapy did not prominently influence these hemostatic parameters. These results indicate that both the malignant process and the drugs used in combined chemotherapy cause a decrease in natural inhibitors and an increase in procoagulant activity and endothelial injury. These hemostatic changes may contribute to a thrombotic tendency in the patients with ALL.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Blood Proteins/analysis , Precursor Cell Lymphoblastic Leukemia-Lymphoma/blood , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Adolescent , Antithrombin III/analysis , Asparaginase/administration & dosage , Child , Child, Preschool , Female , Heparin Cofactor II/analysis , Humans , Male , Methylprednisolone/administration & dosage , Peptide Fragments/analysis , Peptide Hydrolases/analysis , Protein C/analysis , Protein S/analysis , Prothrombin/analysis , Thrombomodulin/blood , von Willebrand Factor/analysis
16.
Thromb Res ; 65(2): 281-7, 1992 Jan 15.
Article in English | MEDLINE | ID: mdl-1579900

ABSTRACT

HC II functional assays are generally preferred with respect to immunochemical assays. Nevertheless functional assays can be biased by the antithrombin III (AT III)-heparin complex activity; in fact trace amounts of heparin generally contaminate dermatan sulfate (DS) commercial preparations used as HC II reaction activators. We have employed a purified DS preparation showing these features: DS concentration 94.4%, chondroitin sulfate 5.6%, M.W. 21.4 kDa. The absence of any interference due to AT III-heparin complexes was verified in a kinetic HC II assay of some human plasma pools. The immunological inhibition of AT III by anti-AT III caused a minimal decrease (6-8%) of the reaction slope, attributable to AT III activity. Progressive increase of heparin concentration in the assay was effective only starting from 30 U/ml (the assay was carried out in the presence of polybrene to prevent any AT III activation). The reference interval (mean +/- SD) obtained from 157 normal subjects was 100.8 +/- 20.2%; there was a good correlation with immunoreactive HC II. The purified DS we have used seems suitable for routinary assays of HC II where a minimal interference due to AT III-heparin is required.


Subject(s)
Dermatan Sulfate/isolation & purification , Heparin Cofactor II/analysis , Amino Acid Sequence , Hexadimethrine Bromide , Humans , Kinetics , Molecular Sequence Data
17.
Thromb Res ; 61(3): 235-41, 1991 Feb 01.
Article in English | MEDLINE | ID: mdl-2028443

ABSTRACT

By a series of experiments it could be shown that an activity test of heparin cofactor II (HC II) is only specific after a complete depletion of antithrombin III. Also when dermatan sulfate which does not enhance the action of AT III is used for the activation of HC II there is a considerable influence on the remaining thrombin activity which alters the test results. Furthermore, in a system which contains plasma as well as thrombin the formation of a clot is likely to occur which by its opacity influences photometric results. A chromogenic substrate assay is described which excludes the influence of these variables. This assay was used to examine the activity of HC II in healthy persons as well as in patients. Three members of a family were found who had a heterozygous deficiency of HC II without any history of thrombosis. On the other hand, a total of 16 patients with heterozygous deficiency of HC II suffered from recurrent thromboembolic episodes. For this reason it is assumed that a deficiency of HC II has a certain importance in the occurrence of thrombophilia though it is apparently less thrombogenic than a deficiency of other inhibitors.


Subject(s)
Heparin Cofactor II/analysis , Photometry/methods , Antibodies/immunology , Antithrombin III/immunology , Chromogenic Compounds , Dermatan Sulfate , Female , Heparin Cofactor II/deficiency , Humans , Male , Thrombin , Thromboembolism/etiology
18.
Thromb Res ; 66(5): 489-98, 1992 Jun 01.
Article in English | MEDLINE | ID: mdl-1381849

ABSTRACT

We investigated heparin cofactor II (HC II) levels and their relationship to other haemostatic factors in the elderly in comparison with antithrombin III (AT III). We measured plasma HC II activity levels in 166 subjects aged from 61 to 99 years using a chromogenic method. HC II levels (94.4 +/- 18.5%) in the healthy elderly subjects were significantly (p less than 0.001) lower than in 40 healthy adult controls under 60 years of age (mean age: 51.5 years; 111.6 +/- 21.2%). HC II levels in the elderly subjects decreased further with age (r = 0.308, p less than 0.001) and the extent of the decrease was more marked than that for AT III (r = 0.179, p less than 0.05). There was no significant sex difference in HC II levels in the elderly. HC II levels correlated significantly with AT III levels and with acute phase reactants including sialic acid, fibrinogen, and PAI-1. HC II levels also correlated with factor VII, plasminogen, alpha 2-plasmin inhibitor, serum lipid, pseudocholinesterase, and albumin levels. These correlations were also found for AT III except active PAI-1 and tPA-PAI-1 complexes, but the correlations with acute phase reactants were stronger for HC II than AT III. We divided 154 elderly subjects into 4 groups by their pseudocholinesterase and albumin levels to estimate the effect of nutritional status on antithrombin activity in the elderly. HC II levels were normal in the elderly subjects with a good nutritional state (103 +/- 18%), but were significantly decreased in those with malnutrition (85 +/- 15%, p less than 0.001). AT III levels also showed the same tendency. These results indicate a decrease in the reserve capacity to inhibit thrombin generation at sites of atherosclerosis in response to trigger events. The deficiency of two major antithrombin factors in the elderly may indicate a tendency to thrombosis, especially in individuals with malnutrition. When considering the clinical significance of HC II, several other parameters, including age, nutritional status, hepatic synthetic ability, and the presence or absence of acute phase reaction should also be assessed.


Subject(s)
Aging/blood , Antithrombin III/analysis , Heparin Cofactor II/deficiency , Acute-Phase Proteins/analysis , Aged , Aged, 80 and over , Blood Coagulation Factors/analysis , Butyrylcholinesterase/blood , Female , Heparin Cofactor II/analysis , Humans , Lipids/blood , Male , Middle Aged , N-Acetylneuraminic Acid , Nutrition Disorders/blood , Sialic Acids/analysis
19.
Thromb Res ; 66(5): 591-8, 1992 Jun 01.
Article in English | MEDLINE | ID: mdl-1523613

ABSTRACT

An ELISA assay for quantitation of the thrombin-heparin cofactor II complex (T-HC II) in plasma was developed. Plasma was incubated with immobilized, specific antibodies to human thrombin. The second, biotinylated antibody was directed against human HC II. The assay was insensitive to thrombin-antithrombin complex (TAT) and to uncomplexed HC II. In plasma samples from 31 normal individuals (aged 21-68, mean 43.3 years), the T-HC II ranged 0.3-6.1 ng/ml; median 1.5, mean 2.0, and SD 1.6 ng/ml. In plasma samples from 13 patients with disseminated intravascular coagulation (DIC), T-HC II ranged 0.4-30.0 (median 13.5) ng/ml. In plasma samples from 6 patients in which the clinical suspicion of DIC was not verified, T-HC II complex ranged 1.4-14.3 (median 3.6) ng/ml. In plasma samples with elevated T-HC II levels, TAT was usually elevated, and on the average more than was T-HC II. These results indicate that HC II contributes significantly to the inactivation of in vivo generated thrombin.


Subject(s)
Disseminated Intravascular Coagulation/blood , Enzyme-Linked Immunosorbent Assay , Heparin Cofactor II/analysis , Thrombin/analysis , Adolescent , Adult , Aged , Antithrombin III/analysis , Child , Child, Preschool , Disseminated Intravascular Coagulation/etiology , Female , Humans , Infant , Infections/blood , Infections/complications , Male , Middle Aged , Peptide Hydrolases/analysis , Pre-Eclampsia/blood , Pregnancy
20.
Thromb Res ; 66(4): 435-44, 1992 May 15.
Article in English | MEDLINE | ID: mdl-1412208

ABSTRACT

We measured plasma heparin cofactor II (HC II) activity in patients with disseminated intravascular coagulation (DIC) due to various underlying diseases together with the levels of antithrombin III (AT III), pseudocholinesterase (a marker of hepatic synthesis), and various haemostatic molecular markers. Both HC II and AT III were decreased in DIC secondary to all the underlying diseases studied, except acute promyelocytic leukemia (APL), when compared with healthy subjects. The lowest HC II and AT III levels was observed in coagulopathy secondary to liver disease, the HC II level in sepsis was the second lowest. In DIC due to APL, the decrease in HC II was not accompanied by a decrease in AT III. Thus, we divided all 124 samples tested into APL and non-APL groups. The HC II level correlated positively with fibrinogen and plasminogen in both the APL and non-APL groups. In the APL group, the HC II level had a significant negative correlation with the thrombin-AT III complex (TAT), fibrinogen/fibrin degradation products, and D-dimer levels as well as the prothrombin time, while AT III showed no correlations with any of the haemostatic parameters. These results suggest that HC II may be consumed preferentially by thrombin in APL patients with DIC, and thus may spare the consumption of AT III. Accordingly, HC II seems to be a superior indicator of DIC than AT III in APL patients. Moreover, replacement therapy with HC II instead of AT III may be useful to treat DIC associated with APL. In the non-APL group, the HC II levels were positively correlated with the levels of AT III and pseudocholinesterase activity. This indicates that plasma HC II levels are closely related not only to consumption coagulopathy but also to hepatic synthetic activity, as is the case for plasma AT III.


Subject(s)
Disseminated Intravascular Coagulation/blood , Heparin Cofactor II/analysis , Leukemia, Promyelocytic, Acute/complications , Adult , Antithrombin III/analysis , Biomarkers/blood , Butyrylcholinesterase/blood , Disseminated Intravascular Coagulation/etiology , Hemostasis , Humans , Leukemia, Promyelocytic, Acute/blood , Liver Diseases/blood , Liver Diseases/complications , Middle Aged , Neoplasms/blood , Neoplasms/complications , Sepsis/blood , Sepsis/complications , Thrombin/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL