Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Int J Hematol ; 111(1): 51-56, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31667683

ABSTRACT

Congenital factor X (FX) deficiency is a rare bleeding disorder with an incidence of one in one million. The proband, a 2-year-old girl, exhibited easy bruising and a history of umbilical cord bleeding at birth. Prothrombin time (> 40 s) and activated partial thromboplastin time (65.0 s) were prolonged. Marked declines in FX activity (< 1%) and FX antigen levels (5%) were also observed. Genetic analysis of the proband identified two types of single-base substitutions, c.353G>A (p.Gly118Asp) and c.1303G>A (p.Gly435Ser), indicating compound heterozygous congenital FX deficiency. Genetic analysis of family members revealed that her father and older sister (5-year-old) were also heterozygous for p.Gly118Asp, and that her mother was heterozygous for p.Gly435Ser. To improve the bleeding tendency, the proband received regular replacement of 500 units of PPSB-HT, a prothrombin complex concentrate (PCC). Following continued regular replacement of 500 units of PPSB-HT once per week, the proband has exhibited no bleeding tendencies and no new bruises have been observed. There are no previous report of the use of PPSB-HT for regular FX replacement. Regular replacement therapy with PPSB-HT may be an effective method for preventative control of bleeding tendencies in FX deficiency.


Subject(s)
Blood Coagulation Factors/therapeutic use , Factor X Deficiency/drug therapy , Factor X Deficiency/genetics , Factor X/genetics , Adult , Child, Preschool , Factor X/metabolism , Factor X Deficiency/enzymology , Factor X Deficiency/pathology , Female , Genetic Testing , Genotype , Hemorrhage/genetics , Heterozygote , Humans , Male , Mutation, Missense , Partial Thromboplastin Time , Pedigree , Phenotype , Prothrombin Time
2.
Haemophilia ; 24(5): 774-785, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30152566

ABSTRACT

Congenital factor X (FX) deficiency is a rare bleeding disorder that is inherited as an autosomal recessive trait. In this study, a genetic analysis of the FX gene was performed in five families with this disorder. Four heterozygous mutations [p.Gly154Arg, p.Val236Met, p.Gly263Val and p.Arg387Cys] and one pair of compound heterozygous FX gene mutations consisting of p.Gly406Ser and p.Val424Phe were identified. Mutant FX proteins containing the identified amino acid substitutions were also expressed in cultured cells. These proteins were analysed by enzyme-linked immunosorbent assay and pulse-chase experiments. The results demonstrated normal intracellular synthesis and extracellular secretion of mutant FX proteins carrying the p.Val236Met, p.Arg387Cys and p.Gly406Ser amino acid substitutions. However, the results also showed that the p.Gly154Arg, p.Gly263Val and p.Val424Phe proteins were secreted less efficiently than the wild-type protein, although they were synthesized normally in the cell. Collectively, these observations suggest that the amino acid substitutions p.Gly154Arg, p.Gly263Val and p.Val424Phe induce protein misfolding, leading to the intracellular degradation of many FX proteins containing any of these mutations, and ultimately to the development of FX deficiency. On the other hand, for the p.Val236Met, p.Arg387Cys and p.Gly406Ser mutant proteins, we hypothesize that secreted FX proteins have impaired coagulant activities due to functional defects caused by these amino acid substitutions.


Subject(s)
Factor X Deficiency/genetics , Genetic Testing/methods , Mutant Proteins/genetics , Adult , Aged , Child, Preschool , Female , Humans , Male , Middle Aged , Mutation, Missense , Transfection , Young Adult
3.
Int J Hematol ; 107(4): 490-494, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29071478

ABSTRACT

Inherited antithrombin (AT) deficiency is one of the most clinically significant forms of congenital thrombophilia and follows an autosomal dominant mode of inheritance. We analyzed SERPINC1 in a patient who developed deep-vein thrombosis and low AT activity during pregnancy, and identified a novel missense mutation c.259A>G (p.Asn87Asp; N87D). Surprisingly, analysis of the parents' DNA showed that they did not possess this mutant, and thus, it may have been due to a de novo mutation. We also expressed this mutant AT protein in COS-1 cells and compared its intracellular localization and intracellular and extracellular antigen levels with that of wild-type AT. The expression experiment did not reveal a significant difference in the antigen levels of the mutant and wild-type AT in the cell lysate, but the mutant AT antigen level was markedly lower than that of its wild-type counterpart in the COS-1 cell supernatant. Immunofluorescence did not indicate any difference between the mutant and wild-type AT in terms of cytoplasmic localization of fluorescence signals. Our findings suggest that the patient's AT deficiency may have been caused by impaired extracellular secretion of mutant AT protein p.Asn87Asp.


Subject(s)
Antigens/metabolism , Antithrombin III Deficiency/genetics , Antithrombin III/genetics , Antithrombin Proteins/genetics , Antithrombin Proteins/physiology , Mutation, Missense , Pregnancy Complications/genetics , Adult , Animals , Antithrombin Proteins/immunology , Antithrombin Proteins/metabolism , COS Cells , Chlorocebus aethiops , Female , Humans , Pregnancy , Thrombophilia/genetics , Venous Thrombosis
4.
Clin Appl Thromb Hemost ; 23(7): 844-850, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28301899

ABSTRACT

INTRODUCTION: Careful monitoring of the hypercoagulable state is required during pregnancy. However, coagulation and fibrinolysis markers are not fully utilized because there are no reference values reflective of coagulation and fibrinolysis dynamics during pregnancy, which differ from the nonpregnant state. METHODS: Changes in antithrombin (AT), fibrinogen (Fbg), prothrombin fragment 1+2 (F1+2), thrombin-antithrombin complex (TAT), soluble fibrin (SF), D-dimer (DD), and protein S (PS) were investigated in healthy pregnant women, and reference ranges in the early, mid, late, and end stages of pregnancy were established. RESULTS: The AT was essentially constant throughout pregnancy. The Fbg, F1+2, TAT, and DD increased significantly as pregnancy progressed. In contrast, SF did not show a significant increase throughout the entire pregnancy period. Total PS antigen and total PS activity showed a corresponding decrease from early gestation. When test data in 3 cases in which deep vein thrombosis or intrauterine fetal death occurred during pregnancy were compared to the established reference ranges, all of the cases had multiple markers with values that exceeded the reference ranges. CONCLUSION: Establishing reference ranges for each week could potentially make it possible to evaluate abnormalities of the coagulation and fibrinolysis systems during pregnancy. Of note, SF might be a useful marker that reflects thrombus formation during pregnancy. Larger-scale studies will be required to establish reference ranges for every gestational week.


Subject(s)
Thrombosis/diagnosis , Adult , Biomarkers/analysis , Blood Coagulation , Female , Fibrin/analysis , Gestational Age , Humans , Predictive Value of Tests , Pregnancy , Pregnancy Complications, Cardiovascular/diagnosis , Reference Values , Young Adult
5.
Thromb Res ; 151: 8-16, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28088608

ABSTRACT

Congenital deficiency of protein S (PS), an anticoagulant factor, leads to venous thrombosis, with onset predominantly beginning in adolescence. In the present study, gene analysis of six unrelated Japanese families diagnosed with congenital PS deficiency identified five missense mutations in the PROS1 gene - c.757C>T (Ala139Val; A139V), c.1346 G>T (Cys449Phe; C449F), c.1352G>A (Arg451Gln; R451Q), c.1424G>T (Cys475Phe; C475F) and c.1574C>T (Ala525Val; A525V) - and one frameshift mutation, c.2135delA (Asp599ThrfsTer13; D599TfsTer13). C449F, R451Q, A525V and D599TfsTer13 are novel mutations. Results from ELISA to measure PS antigen levels in culture supernatant showed that the A139V variant was similar to wild-type, but other variants showed reductions when compared with wild-type. Results from pulse-chase analysis confirmed that the A139V variant exhibited secretion equivalent to wild-type, but for the other variants, there was no extracellular secretion, and it had nearly all been degraded inside the cell within six hours. Results from pulse-chase analysis using proteasome inhibitors also showed that intracellular degradation of mutant protein was inhibited. Activity of the A139V variant was decreased to 71% of wild-type, and the phospholipid binding capacity fell to as low as 45%. These results suggest that although the A139V variant has normal secretion, it has abnormal phospholipid binding capacity, and therefore causes type II PS deficiency, in which PS activity is decreased. It is also thought that with the other variants, misfolding due to amino acid mutations causes nearly all PS to be degraded intracellularly, therefore leading to type I PS deficiency.


Subject(s)
Blood Proteins/genetics , Frameshift Mutation , Mutation, Missense , Protein S Deficiency/genetics , Protein S/genetics , Adolescent , Adult , Asian People/genetics , Blood Proteins/metabolism , Female , Genotype , Humans , Japan/epidemiology , Male , Phospholipids/metabolism , Protein Binding , Protein S/metabolism , Protein S Deficiency/epidemiology , Protein S Deficiency/metabolism
6.
Int J Hematol ; 105(3): 287-294, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27858332

ABSTRACT

We summarize causative genetic mutations for antithrombin (AT) deficiency and their clinical background in Japanese patients. A total of 19 mutations, including seven novel mutations, were identified. We also summarize clinical symptoms of thrombosis, age at onset, family history, and contributing factors for thrombosis, and review the use of prophylactic anticoagulation in pregnant women with heterozygous type II heparin binding site defects (HBS) AT deficiency. The prevalence of thrombosis in probands with type I AT deficiency (88%) was double that observed in those with type II AT deficiency (50%). The prevalence of thrombotic episodes among family members was also higher for type I AT deficiency subjects (82%) than for those with type II AT deficiency (0%). The most common contributing factor for thrombosis among women with type I AT deficiency was pregnancy. Forty-five percent of women with type I AT deficiency developed thrombotic events before the 20th week of gestation. In contrast, women with type II (HBS) AT deficiency appear to be at a lower risk of thrombosis during pregnancy. In conclusion, thrombotic risk varies among different subtypes. Risk assessments based on genetic/clinical backgrounds may contribute to appropriate diagnosis, treatment, and prophylaxis for patients with AT deficiency.


Subject(s)
Antithrombin Proteins/deficiency , Mutation/physiology , Adolescent , Adult , Aged , Antithrombin Proteins/genetics , Asian People , Female , Humans , Male , Middle Aged , Pregnancy , Pregnancy Complications, Cardiovascular/diagnosis , Risk Factors , Thrombophilia/complications , Thrombophilia/diagnosis , Thrombosis/etiology
7.
PLoS One ; 10(7): e0133196, 2015.
Article in English | MEDLINE | ID: mdl-26186226

ABSTRACT

Protein S (PS) acts as a cofactor for activated protein C in the plasma anticoagulant system. PS Lys196-to-Glu (K196E) mutation is a genetic risk factor for venous thromboembolism in Japanese individuals. Because of the substantial overlap in PS anticoagulant activity between KK (wild-type) and KE (heterozygous) genotypes, it is difficult to identify PS K196E carriers by measuring PS activity. Here, we generated monoclonal antibodies specific to the PS K196E mutant and developed a simple and reliable method for the identification of PS K196E carriers. We immunized mice with a keyhole limpet hemocyanin-conjugated synthetic peptide with Glu196. The hybridoma cells were screened for the binding ability of the produced antibodies to recombinant mutant EGF-like domains of PS (Ile117-Glu283). We obtained three hybridoma cell lines producing PS K196E mutation-specific antibodies. We established a sandwich enzyme-linked immunosorbent assay (ELISA) system in which the PS K196E mutation-specific monoclonal antibody was used as a detection antibody. We measured human plasma samples by using this system and successfully discriminated 11 individuals with the KE genotype from 122 individuals with the KK genotype. The ELISA system using the PS K196E mutation-specific antibody is a useful tool for the rapid identification of PS K196E carriers, who are at a higher risk for venous thromboembolism.


Subject(s)
Amino Acid Substitution , Mutation , Protein S/genetics , Venous Thromboembolism/genetics , Animals , Antibodies, Monoclonal/immunology , Antibody Specificity , Enzyme-Linked Immunosorbent Assay , Genetic Predisposition to Disease , Genotype , Humans , Mice , Protein S/immunology , Risk Factors , Venous Thromboembolism/blood
9.
J Atheroscler Thromb ; 22(7): 660-8, 2015 Jul 23.
Article in English | MEDLINE | ID: mdl-25735397

ABSTRACT

AIM: 3-Hydroxy-3-methylglutaryl coenzyme A reductase inhibitors (statins) are cholesterol-lowering drugs with a variety of pleiotropic effects including antithrombotic properties. Tissue factor pathway inhibitor (TFPI), which is produced predominantly in endothelial cells and platelets, inhibits the initiating phase of clot formation. We investigated the effect of fluvastatin on TFPI expression in cultured endothelial cells. METHODS: Human umbilical vein endothelial cells (HUVECs) were treated with fluvastatin (0-10µM). The expression of TFPI mRNA and antigen were detected by RT-PCR and western blotting, respectively. The effects of mevalonate intermediates, small GTP-binding inhibitors, and signal transduction inhibitors were also evaluated to identify which pathway was involved. A luciferase reporter assay was performed to evaluate the effect of fluvastatin on TFPI transcription. The stability of TFPI mRNA was estimated by quantitating its levels after actinomycin D treatment. RESULTS: Fluvastatin increased TFPI mRNA expression and antigen in HUVECs. Fluvastatin-induced TFPI expression was reversed by co-treatment with mevalonate or geranylgeranylpyrophosphate (GGPP). NSC23766 and Y-27632 had no effect on TFPI expression. SB203580, GF109203, and LY294002 reduced fluvastatin-induced TFPI upregulation. Moreover, fluvastatin did not significantly affect TFPI promoter activity. TFPI mRNA degradation in the presence of actinomycin D was delayed by fluvastatin treatment. CONCLUSIONS: Fluvastatin increases endothelial TFPI expression through inhibition of mevalonate-, GGPP-, and Cdc42-dependent signaling pathways, and activation of the p38 MAPK, PI3K, and PKC pathways. This study revealed unknown mechanisms of the anticoagulant effect of statins and gave a new insight to its therapeutic potential for the prevention of thrombotic diseases.


Subject(s)
Fatty Acids, Monounsaturated/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Indoles/pharmacology , Lipoproteins/analysis , Anticoagulants/pharmacology , Antigens/analysis , Blotting, Western , Cells, Cultured , Chromones/pharmacology , Dactinomycin/pharmacology , Fluvastatin , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Imidazoles/pharmacology , Lipoproteins/genetics , Lipoproteins/immunology , Luciferases/analysis , Maleimides/pharmacology , Mevalonic Acid/pharmacology , Morpholines/pharmacology , Pyridines/pharmacology , RNA, Messenger/analysis , Real-Time Polymerase Chain Reaction , Up-Regulation/drug effects
10.
Thromb Res ; 132(2): e118-23, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23809926

ABSTRACT

INTRODUCTION: Inherited antithrombin (AT) deficiency is associated with a predisposition to familial venous thromboembolic disease. We analyzed the AT gene in three unrelated patients with an AT deficiency who developed thrombosis. MATERIALS AND METHODS: We analyzed the SERPINC1 gene in three patients. Additionally, we expressed the three mutants in the COS-1 cells and compared their secretion rates and levels of AT activity with those of the wild-type (WT). RESULTS: We identified three distinct heterozygous mutations of c.2534C>T: p.56Arginine → Cysteine (R56C), c.13398C>A: p.459Alanine → Aspartic acid (A459D) and c.2703C>G: p.112 Proline → Arginine (P112R). In the in vitro expression experiments, the AT antigen levels in the conditioned media (CM) of the R56C mutant were nearly equal to those of WT. In contrast, the AT antigen levels in the CM of the A459D and P112R mutants were significantly decreased. The AT activity of R56C was decreased in association with a shorter incubation time in a FXa inhibition assay and a thrombin inhibition-based activity test. However, the AT activity of R56C was comparable to that of WT when the incubation time was increased. CONCLUSIONS: We concluded that the R56C mutant is responsible for type II HBS deficiency. We considered that the A459D and P112R mutants can be classified as belonging to the type I AT deficiency.


Subject(s)
Antithrombin III Deficiency/genetics , Antithrombin III/genetics , Point Mutation , Adult , Aged , Animals , Antithrombin III/metabolism , Antithrombin III Deficiency/blood , Blood Coagulation Tests , COS Cells , Chlorocebus aethiops , Female , Humans , Japan , Young Adult
11.
Thromb Res ; 130(3): e188-93, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22819264

ABSTRACT

INTRODUCTION: Heme oxygenase-1 (HO-1) is the rate limiting enzyme that catalyzes the conversion of heme into biliverdin, free iron, and carbon monoxide (CO). The first human case of HO-1 deficiency showed abnormalities in blood coagulation and the fibrinolytic system. Thus, HO-1 or HO-1 products, such as CO, might regulate coagulation and the fibrinolytic system. This study examined whether tricarbonyldichlororuthenium (II) dimer (CORM-2), which liberates CO, modulates the expression of tissue factor (TF) and plasminogen activator inhibitor type 1 (PAI-1) in human umbilical vein endothelial cells (HUVECs), and TF expression in peripheral blood mononuclear cells (PBMCs). Additionally, we examined the mechanism by which CO exerts its effects. MATERIALS AND METHODS: HUVECs were pretreated with 50 µM CORM-2 for 3 hours, and stimulated with tumor necrosis factor-α (TNF-α, 10 ng/ml) for an additional 0-5 hours. PBMCs were pretreated with 50-100 µM CORM-2 for 1 hour followed by stimulating with lipopolysaccharid (LPS, 10 ng/ml) for additional 0-9 hours. The mRNA and protein levels were determined by RT-PCR and western blotting, respectively. RESULTS: Pretreatment with CORM-2 significantly inhibited TNF-α-induced TF and PAI-1 up-regulation in HUVECs, and LPS-induced TF expression in PBMCs. CORM-2 inhibited TNF-α-induced activation of p38 MAPK, ERK1/2, JNK, and NF-κB signaling pathways in HUVECs. CONCLUSIONS: CORM-2 suppresses TNF-α-induced TF and PAI-1 up-regulation, and MAPKs and NF-κB signaling pathways activation by TNF-α in HUVECs. CORM-2 suppresses LPS-induced TF up-regulation in PBMCs. Therefore, we envision that the antithrombotic activity of CORM-2 might be used as a pharmaceutical agent for the treatment of various inflammatory conditions.


Subject(s)
Carbon Dioxide/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Organometallic Compounds/pharmacology , Plasminogen Activator Inhibitor 1/metabolism , Thromboplastin/metabolism , Cells, Cultured , Humans
12.
Rinsho Ketsueki ; 53(3): 357-60, 2012 Mar.
Article in Japanese | MEDLINE | ID: mdl-22499055

ABSTRACT

Congenital factor VII (FVII) deficiency is a bleeding disorder that requires optimal hemostatic management for each case due to its wide variety of bleeding symptoms. We experienced a patient with inherited FVII deficiency who demonstrated different FVII activities depending on tissue thromboplastins used for assays. An 82-year-old woman without any episodes of abnormal bleeding was found to have different FVII activities of 1.4% and 32% when assayed using thromboplastins from rabbit brain and human placenta, respectively. DNA sequencing analysis revealed a homozygous missense mutation of G10828A (FVII Padua) that caused an amino acid substitution of Arg304 to Gln (R304Q). Carriers of 304Q alleles are usually clinically asymptomatic and do not require FVII replacement therapies even in cases of homozygotes. In case a prolonged prothrombin time or reduced FVII activity is detected, re-examination using thromboplastins of other sources can be helpful for preliminary diagnosis of R304Q, in order to prevent unnecessary FVII replacement therapies.


Subject(s)
Factor VII Deficiency/congenital , Factor VII Deficiency/diagnosis , Factor VII/genetics , Mutation, Missense , Thromboplastin , Aged, 80 and over , Amino Acid Substitution/genetics , Animals , Factor VII Deficiency/genetics , Female , Homozygote , Humans , Rabbits
13.
J Atheroscler Thromb ; 19(1): 98-104, 2012.
Article in English | MEDLINE | ID: mdl-22104176

ABSTRACT

AIM: Obstructive sleep apnea syndrome (OSAS) has been associated with high cardiovascular morbidity and mortality, and patients suffer from repeated episodes of hypoxia. Platelet-derived microparticles (PDMPs) are released via platelet activation by various agonists, including inflammatory cytokines or high shear stress. Plasminogen activator inhibitor -1 (PAI-1) is a fibrinolytic marker and soluble fibrin (SF) is a coagulation activation marker. We examined plasma levels of PDMPs, PAI-1 and SF in patients with OSAS. We also examined the effects of continuous positive airway pressure (CPAP) on plasma levels of PDMPs. METHODS: Full polysomnography (PSG) monitoring was performed on 27 patients. The apneahypopnea index (AHI) of 5 events/h or less than 30 events/h indicated mild to moderate OSAS, and an AHI of 30 events/h or more indicated severe OSAS. Plasma levels of PDMPs were measured using an ELISA kit, and PAI and SF were determined by a latex immunoassay. In addition, the effects of CPAP treatment were studied in 7 patients. RESULT: The plasma level of PDMPs was significantly higher in patients with severe OSAS (15.8±10.4 U/mL) than normal controls (10.8±7.1 U/mL, p < 0.05) and patients with mild to moderate OSAS (9.2±3.5 U/mL, p < 0.05). The plasma levels of PDMPs correlated with the AHI (r = 0.39, p < 0.05). In addition, CPAP treatment decreased the plasma level of PDMPs (11.9±5.6 U/mL to 6.7±3.2 U/mL, p < 0.05). CONCLUSIONS: Patients with OSAS might be at increased cardiovascular risk due to elevated PDMPs. Moreover a decrease in the plasma level of PDMPs by treatment with CPAP might reduce cardiovascular risk.


Subject(s)
Blood Platelets/pathology , Cell-Derived Microparticles/pathology , Sleep Apnea, Obstructive/diagnosis , Blood Platelets/metabolism , Case-Control Studies , Cell-Derived Microparticles/metabolism , Female , Fibrin/metabolism , Humans , Male , Middle Aged , Plasminogen Activator Inhibitor 1/blood , Platelet Activation , Polysomnography , Prognosis , Sleep Apnea, Obstructive/blood
14.
Int J Hematol ; 93(2): 216-219, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21240680

ABSTRACT

An inherited antithrombin deficiency is an autosomal dominant thrombotic disorder. We identified two pedigrees of inherited type I antithrombin deficiency and two responsible mutations in each. A novel 21-22delAA appeared to have caused a frameshift with a premature termination at amino acid +63 in one patient and a large deletion including all seven exons was identified by multiplex ligation-dependent probe amplification in the other. Some asymptomatic relatives of the second patient had the same mutation. The present findings support the value of using more than one method of gene analysis and of studying the families of probands with inherited thrombotic disorders.


Subject(s)
Antithrombin III Deficiency/genetics , Antithrombin III/genetics , Exons/genetics , Frameshift Mutation , Gene Deletion , Adult , Antithrombin III Deficiency/diagnosis , Female , Humans , Male , Middle Aged , Pedigree
15.
Nihon Ronen Igakkai Zasshi ; 46(4): 348-51, 2009 Jul.
Article in Japanese | MEDLINE | ID: mdl-19713668

ABSTRACT

The proband is a 69-year-old woman with purpura and subcutaneous hematoma.We investigated this patient with prekallikrein (PK) deficiency, using both standard coagulation study and molecular genetic analysis of the PK gene. In a coagulation study, the prothrombin time (PT) was normal but the activated partial thromboplastin time (APTT) was prolonged. Preincubation of normal plasma with APTT reagent caused shortening of abnormal clotting time. Plasma PK activity was <1%. Her parents were cousins. Molecular genetic analysis showed a homozygous Gly401Glu substitution in exon 11 in the PK gene. This mutation has already been reported in a Japanese patient as PK Tokushima. Gly401 is positioned in PK light chain, which encodes the serine protease domain. The disulfide binding is formed between Cys400 and Cys416, thus Gly401 is located next to His415, which is one of the activation peptides and is important in supporting the correct conformation of proteins. Therefore, we suggest that this mutation may prevent formation of disulfide binding and reduce enzyme activity. In conclusion, in the elderly case with prolonged APTT, we should consider the contact factor deficiency and determine PK activity apart from the abnormality of coagulation factor VIII, IX, XI and XII activities.


Subject(s)
Prekallikrein/deficiency , Aged , Female , Humans , Partial Thromboplastin Time , Prekallikrein/genetics
16.
Int J Hematol ; 88(3): 272-277, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18751872

ABSTRACT

Previous studies have found markedly elevated serum concentrations of proinflammatory cytokines in patients with Graves' disease (GD). We investigated the role of macrophage colony-stimulating factor (M-CSF) in GD. We assayed concentrations of M-CSF in sera from 32 patients with GD (25 untreated; 7 receiving thiamazole therapy). We also studied 32 age-matched healthy subjects as controls. Relationships between serum M-CSF and both thyroid state and serum lipids were examined. Moreover, to examine the effect of thyroid hormone alone on serum M-CSF, T3 was administered orally to normal subjects. Serum concentrations of M-CSF in GD patients who were hyperthyroid were significantly increased compared with GD patients who were euthyroid (P < 0.05) and control subjects (P < 0.0001). Serum M-CSF concentrations correlated closely with T3 levels in patients (r = 0.51, P < 0.005). Serial measurement of five individual patients revealed that serum concentrations of M-CSF were significantly decreased (P < 0.05), reaching normal control values upon attainment of euthyroidism. Furthermore, oral T3 administered to 15 volunteers for 7 days produced significant increases in serum levels of M-CSF (P < 0.05). The close correlation between serum M-CSF and serum thyroid hormone levels suggests that high circulating levels of thyroid hormones may directly or indirectly potentiate the production of M-CSF in patients with GD.


Subject(s)
Graves Disease/blood , Macrophage Colony-Stimulating Factor/blood , Triiodothyronine/administration & dosage , Triiodothyronine/pharmacokinetics , Administration, Oral , Adult , Female , Graves Disease/drug therapy , Humans , Inflammation Mediators/blood , Male , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL
...