Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Biotechnol Prog ; 34(2): 515-520, 2018 03.
Article in English | MEDLINE | ID: mdl-29086495

ABSTRACT

The production of recombinant vitamin K dependent (VKD) proteins for therapeutic purposes is an important challenge in the pharmaceutical industry. These proteins are primarily synthesized as precursor molecules and contain pre-propeptide sequences. The propeptide is connected to γ-carboxylase enzyme through the γ-carboxylase recognition site for the direct γ-carboxylation of VKD proteins that has a significant impact on their biological activity. Propeptides have different attitudes toward γ-carboxylase and certain amino acids in propeptide sequences are responsible for the differences in γ-carboxylase affinity. By aiming to replace amino acids in hFIX propeptide domain based on the prothrombin propeptide, pMT-hFIX-M14 expression cassette, containing cDNA of hFIX with substituted -14 residues (Asp to Ala) was made. After transfection of Drosophila S2 cells, expression of the active hFIX was analyzed by performing ELISA and coagulation test. A 1.4-fold increase in the mutant recombinant hFIX expression level was observed in comparison with that of a native recombinant hFIX. The enhanced hFIX activity and specific activity of the hFIXD-14A (2.2 and 1.6 times, respectively) were further confirmed by comparing coagulation activity levels of substituted and native hFIX. Enrichment for functional, fully γ-carboxylated hFIX species via barium citrate adsorption demonstrated 2-fold enhanced recovery in the S2-expressing hFIXD-14A relative to that expressed native hFIX. These results show that changing -14 residues leads to a decrease in the binding affinity to substrate, increase in γ-carboxylation and activity of recombinant hFIX. © 2017 American Institute of Chemical Engineers Biotechnol. Prog., 34:515-520, 2018.


Subject(s)
Carbon-Carbon Ligases/chemistry , Peptides/chemistry , Protein S/biosynthesis , Recombinant Proteins/biosynthesis , Amino Acid Sequence/genetics , Amino Acid Substitution/genetics , Animals , CHO Cells/chemistry , Cricetulus , Factor IX/chemistry , Factor IX/genetics , Humans , Protein S/chemistry , Prothrombin/chemistry , Recombinant Proteins/chemistry , Transfection , Vitamin K/chemistry , Vitamin K/genetics
2.
J Immunol ; 196(8): 3375-84, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26962228

ABSTRACT

Multifocal inflammatory lesions featuring destruction of lipid-rich myelin are pathologic hallmarks of multiple sclerosis. Lesion activity is assessed by the extent and composition of myelin uptake by myeloid cells present in such lesions. In the inflamed CNS, myeloid cells are comprised of brain-resident microglia, an endogenous cell population, and monocyte-derived macrophages, which infiltrate from the systemic compartment. Using microglia isolated from the adult human brain, we demonstrate that myelin phagocytosis is dependent on the polarization state of the cells. Myelin ingestion is significantly enhanced in cells exposed to TGF-ß compared with resting basal conditions and markedly reduced in classically activated polarized cells. Transcriptional analysis indicated that TGF-ß-treated microglia closely resembled M0 cells. The tyrosine kinase phagocytic receptor MerTK was one of the most upregulated among a select number of differentially expressed genes in TGF-ß-treated microglia. In contrast, MerTK and its known ligands, growth arrest-specific 6 and Protein S, were downregulated in classically activated cells. MerTK expression and myelin phagocytosis were higher in CNS-derived microglia than observed in monocyte-derived macrophages, both basally and under all tested polarization conditions. Specific MerTK inhibitors reduced myelin phagocytosis and the resultant anti-inflammatory biased cytokine responses for both cell types. Defining and modulating the mechanisms that regulate myelin phagocytosis has the potential to impact lesion and disease evolution in multiple sclerosis. Relevant effects would include enhancing myelin clearance, increasing anti-inflammatory molecule production by myeloid cells, and thereby permitting subsequent tissue repair.


Subject(s)
Multiple Sclerosis/immunology , Myelin Sheath/immunology , Myeloid Cells/immunology , Phagocytosis/immunology , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Adult , Brain/cytology , Brain/immunology , Cell Polarity/physiology , Cells, Cultured , Down-Regulation , Humans , Inflammation/immunology , Inflammation/pathology , Intercellular Signaling Peptides and Proteins/biosynthesis , Macrophages/immunology , Microglia/cytology , Microglia/immunology , Multiple Sclerosis/pathology , Protein S/biosynthesis , Proto-Oncogene Proteins/biosynthesis , Receptor Protein-Tyrosine Kinases/biosynthesis , Transforming Growth Factor beta/pharmacology , Up-Regulation , c-Mer Tyrosine Kinase
3.
Immunology ; 135(1): 40-50, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22043818

ABSTRACT

Activation of Toll-like receptors (TLRs) triggers rapid inflammatory cytokine production in various cell types. The exogenous product of growth-arrest-specific gene 6 (Gas6) and Protein S (ProS) inhibit the TLR-triggered inflammatory responses through the activation of Tyro3, Axl and Mer (TAM) receptors. However, regulation of the Gas6/ProS-TAM system remains largely unknown. In the current study, mouse macrophages are shown to constitutively express Gas6 and ProS, which synergistically suppress the basal and TLR-triggered production of inflammatory cytokines, including those of tumour necrosis factor-α, interleukin-6 and interleukin-1ß, by the macrophages in an autocrine manner. Notably, TLR signalling markedly decreases Gas6 and ProS expression in macrophages through the activation of the nuclear factor-κB. Further, the down-regulation of Gas6 and ProS by TLR signalling facilitates the TLR-mediated inflammatory cytokine production in mouse macrophages. These results describe a self-regulatory mechanism of TLR signalling through the suppression of Gas6 and ProS expression.


Subject(s)
Cytokines/immunology , Inflammation Mediators/immunology , Intercellular Signaling Peptides and Proteins/immunology , Macrophages, Peritoneal/immunology , Protein S/immunology , Toll-Like Receptors/immunology , Animals , Cells, Cultured , Cytokines/biosynthesis , Cytokines/genetics , Gene Expression Regulation/immunology , Inflammation Mediators/metabolism , Intercellular Signaling Peptides and Proteins/biosynthesis , Intercellular Signaling Peptides and Proteins/genetics , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred C57BL , NF-kappa B/immunology , NF-kappa B/metabolism , Protein S/biosynthesis , Protein S/genetics , Toll-Like Receptors/metabolism
5.
J Infect ; 58(5): 368-74, 2009 May.
Article in English | MEDLINE | ID: mdl-19307023

ABSTRACT

OBJECTIVES: Dengue virus (DV) infections can cause severe life-threatening dengue hemorrhagic fever/dengue shock syndrome (DHF/DSS). However, the mechanism to cause hemorrhage in DV infections remains poorly understood. Thrombomodulin (TM), expressed on the surface of endothelial cells and monocytes, is very important in regulation of coagulation and inflammation. Therefore, the effect of DV on the TM expression was studied in vitro using both endothelial cells and monocytes. METHODS AND RESULTS: The expression of TM in human endothelial cell line, HMEC-1, monocytic cell line THP-1 and peripheral blood mononuclear cells derived from human blood was increased after DV infection, UV-inactivated DV or recombinant DV envelop protein domain III stimulation as demonstrated by flow cytometry and immunofluorescent staining. Western blot analysis further confirmed only DV but not enterovirus 71 infection of HMEC-1 cells increased TM protein expression. In addition, RT-PCR analysis showed the increase of TM mRNA as well as other protein C activation-related molecules in DV stimulated HMEC-1 in a dose-dependent manner. CONCLUSION: These results suggest that DV stimulation of human endothelial cells and monocytes can increase the expression of TM, which may contribute to the anticoagulant properties of cells during DV infection.


Subject(s)
Dengue Virus/physiology , Endothelial Cells/metabolism , Monocytes/metabolism , Severe Dengue/metabolism , Thrombomodulin/biosynthesis , Transcriptional Activation , Antigens, CD/biosynthesis , Antigens, CD/genetics , Cell Line , Endothelial Cells/virology , Endothelial Protein C Receptor , Enterovirus A, Human/physiology , Enterovirus Infections/genetics , Enterovirus Infections/metabolism , Enterovirus Infections/virology , Humans , Monocytes/virology , Protein S/biosynthesis , Protein S/genetics , Protein Structure, Tertiary , Receptors, Cell Surface/biosynthesis , Receptors, Cell Surface/genetics , Recombinant Proteins/biosynthesis , Severe Dengue/genetics , Severe Dengue/virology , Thrombomodulin/genetics , Tissue Plasminogen Activator/biosynthesis , Tissue Plasminogen Activator/genetics , Viral Envelope Proteins/biosynthesis , Viral Envelope Proteins/chemistry
7.
J Gastroenterol Hepatol ; 23(8 Pt 1): 1169-74, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18505413

ABSTRACT

The discovery of "ground glass" hepatocytes (GGH) that contain hepatitis B virus (HBV) surface antigens by Hadziyannis and Popper in 1973 represents a historical landmark in the pathology of chronic HBV infection. Different types of GGH have been correlated to the expression patterns of surface/core antigens and the stages of virus replication. The original two types (designated types I & II) of GGH were found to contain specific pre-S mutants with deletions over either pre-S1 or pre-S2 regions, respectively. Type II GGH consistently harbor pre-S2 deletion mutants, which can escape from immune attack and grow preferentially to form clusters. Both types of pre-S mutants can induce endoplasmic reticulum (ER) stress and oxidative DNA damage. The pre-S2 mutants, albeit inducing a weaker level of ER stress signals, could additionally initiate ER stress-independent retinoblastoma/adenovirus E2 promoter binding factor/cyclin A signaling through their interaction with c-Jun activation domain binding protein 1 to degrade p27, illustrating the growth advantage of type II GGH. The combined effects of genomic instability and the proliferation of hepatocytes harboring pre-S mutants could potentially lead to hepatocarcinogenesis over the decades of chronic HBV infection. The presence of pre-S mutants in sera was reported to carry a high risk of developing hepatocellular carcinoma (HCC). Furthermore, transgenic mice harboring pre-S2 mutant plasmids have been shown to develop a dysplastic change of hepatocytes and HCC. Therefore, in addition to being a histological marker of chronic HBV infection, GGH, particularly type II GGH, may represent the preneoplastic lesions of HBV-related HCC.


Subject(s)
Hepatitis B, Chronic/genetics , Hepatocytes/pathology , Liver Neoplasms/genetics , Protein Precursors/genetics , Protein S/genetics , Animals , Hepatitis B, Chronic/pathology , Hepatocytes/virology , Humans , Liver Neoplasms/pathology , Mice , Mutation , Protein Precursors/biosynthesis , Protein S/biosynthesis
8.
Haematologica ; 93(4): 574-80, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18322254

ABSTRACT

BACKGROUND: The molecular mechanisms by which PROS1 mutations result in protein S deficiency are still unknown for many of the mutations, particularly for those that result in a premature termination codon. The aim of this study was to analyze the functional relevance on mRNA and protein expression of 12 natural PROS1 mutations associated with protein S deficiency. DESIGN AND METHODS: Five mutations were nonsense, three were small frameshift deletions, one was c.258,259AG>GT at the 3' end of exon 3, one was p.M640T and the last two were c.-7C>G and p.L15H, found in double heterozygosis as [c.-7C>G;44T>A]. The apparently neutral variant p.R233K was also analyzed. PROS1 cDNA was assessed by reverse transcriptase polymerase chain reaction of platelet mRNA. Expression of mutant proteins was determined by site-directed mutagenesis and analyses of transiently transfected PROS1 mutants in COS-7 cells. RESULTS: Only cDNA from the normal allele was observed from the five nonsense mutations, the frameshift deletion c.1731delT and from c.258,259AG>GT. Both the normal and the mutated alleles were observed from [c.-7C>G;44T>A], c.187,188delTG and p.M640T. Transient expression analyses of PROS1 mutants whose mRNA was normally expressed revealed greatly reduced secretion of p.L15H and c.1272delA, mild secretion values of p.M640T and normal secretion levels of c.-7C>G and, as expected, p.R233K. CONCLUSIONS: Whereas the main cause of quantitative protein S deficiency associated with missense mutations is defective synthesis, stability or secretion of the mutated protein, the main mechanism for the deficiency associated with mutations that generate a premature termination codon is not the synthesis of a truncated protein, but the exclusion of the mutated allele, probably by nonsense-mediated mRNA decay.


Subject(s)
Mutation , Protein S Deficiency/genetics , Protein S/genetics , Animals , COS Cells , Chlorocebus aethiops , Codon, Nonsense , Frameshift Mutation , Genotype , Humans , Mutagenesis, Site-Directed , Mutation, Missense , Protein S/biosynthesis , RNA Stability/genetics , RNA, Messenger/metabolism , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Sequence Deletion , Transfection
9.
Haematologica ; 92(2): 279-80, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17296591

ABSTRACT

Prothrombotic coagulation abnormalities were analyzed in patients with untreated multiple myeloma. Increases in factor VIII, in von Willebrand factor (vWF) and a decrease in protein S were observed and these changes were strongly associated with disease stage. No difference in baseline coagulation parameters was found between patients with and without subsequent venous thromboembolism.


Subject(s)
Blood Coagulation Disorders/complications , Blood Coagulation Disorders/diagnosis , Blood Coagulation , Multiple Myeloma/blood , Multiple Myeloma/diagnosis , Venous Thrombosis/complications , Venous Thrombosis/diagnosis , Aged , Antibodies, Antiphospholipid/chemistry , Case-Control Studies , Factor VIII/biosynthesis , Female , Humans , Male , Middle Aged , Protein S/biosynthesis , von Willebrand Factor/biosynthesis
10.
Thromb Res ; 120(5): 639-45, 2007.
Article in English | MEDLINE | ID: mdl-17184825

ABSTRACT

BACKGROUND: Estrogen and the estrogen receptors alpha (ESR1) and beta (ESR2) play a role in regulating genes, including coagulation and fibrinolysis genes. OBJECTIVE: We investigated the association between ESR1 c.454-397T>C and c.454-351A>G and ESR2 1082A>G and 1730A>G polymorphisms and the risk of deep vein thrombosis (DVT), in 134 patients and 134 controls with acquired risk factors for thrombosis associated with estrogen alterations, such as pregnancy, puerperium, oral contraceptives (OC), and hormone replacement therapy (HRT). We also analysed 134 men with DVT. We investigated the relationship of these polymorphisms and the levels of fibrinogen, protein C (PC), protein S (PS), and antithrombin (AT) activity. METHODS: Gene polymorphisms were identified by using PCR and RFLP. Coagulation methods were used to measure PC, PS, and fibrinogen. Chromogenic methods were used to quantify AT. RESULTS AND CONCLUSIONS: The presence of the AA genotype of the 1730G>A polymorphism (OR=0.18; 95%CI=0.05-0.62) suggests a protective effect for DVT in women using OC. As the GG genotype of the 1730G>A polymorphism is associated with increased PS activity in all control women and women using OC, this suggested that a protective effect must occur by another pathway not related to PS. The AA and AG genotypes of the c.454-351A>G and GG genotype of the 1082G>A polymorphisms are associated with increased fibrinogen concentration in pregnant women. The GG haplotype in the ESR2 gene (P<0.001) was related to factor V Leiden or G20210A mutation in the prothrombin gene, or both, as predictive factors of DVT.


Subject(s)
Estrogen Receptor alpha/genetics , Estrogen Receptor beta/genetics , Polymorphism, Genetic , Venous Thrombosis/diagnosis , Venous Thrombosis/genetics , Adolescent , Adult , Aged , Antithrombins/biosynthesis , Blood Coagulation , Case-Control Studies , Female , Fibrinogen/metabolism , Genotype , Hormone Replacement Therapy/adverse effects , Humans , Male , Middle Aged , Protein C/biosynthesis , Protein S/biosynthesis
11.
Br J Haematol ; 135(4): 538-46, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17061980

ABSTRACT

Protein S is expressed in a number of tissue types, one of the most physiologically relevant being the liver. However, transcriptional control of protein S gene expression is poorly understood. We have characterised a 638 bp area in the 5' flanking region of the human protein S gene, spanning all 10 previously reported transcription initiation sites, which demonstrates promoter activity in the human liver-derived cell line HepG2. More refined reporter gene analysis of this region enabled the identification of three transcription initiation sites whose absence is associated with significantly reduced promoter activity, together with a number of positively and negatively acting transcriptional regulatory elements. Consistent with these findings, DNaseI footprinting analysis identified eleven sites (I-XI) from within this 638 bp region that show evidence of binding nuclear proteins. We present evidence to show that the liver-specific factors hepatocyte nuclear factor 1 (HNF1) and HNF4 bind regions of the protein S promoter, which lie within the identified protein binding sites V and VIII, respectively, and that HNF4 activates the protein S promoter. Reporter gene analysis suggests that members of the CCAAT/enhancer binding protein (C/EBP) family of transcription factors are potent activators of protein S gene transcription in HepG2 cells.


Subject(s)
Gene Expression Regulation , Liver/metabolism , Promoter Regions, Genetic , Protein S/genetics , Base Sequence , DNA Footprinting , Genes, Reporter , Hepatocyte Nuclear Factor 1/metabolism , Hepatocyte Nuclear Factor 4/genetics , Humans , Molecular Sequence Data , Plasmids , Protein Binding , Protein S/biosynthesis , Transcription Factors/genetics , Transcription, Genetic
12.
J Thromb Haemost ; 4(8): 1763-73, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16879219

ABSTRACT

BACKGROUND: The vitamin K-dependent protein S (PS), mainly synthesized in hepatocytes and endothelial cells, plays a critical role in the anticoagulant activity of plasma. The decreased plasma level of PS in sepsis is associated with thrombotic tendency, but the mechanism is unclear. OBJECTIVES: In the present study, we examined the effect of lipopolysaccharide (LPS) on PS expression in vivo in rat liver, and in vitro in isolated hepatocytes and sinusoidal endothelial cells (SECs) from normal rats. RESULTS: LPS induced a progressive decrease of plasma PS antigen level up to 12 h with a slight recovery at 24 h, and a transient decrease of liver PS mRNA level at 4-8 h with a complete recovery at 24 h. In the in vitro studies, LPS decreased PS antigen and mRNA levels in both hepatocytes and SECs. After LPS treatment, tumor necrosis factor-alpha (TNF-alpha), interleukin-6 (IL-6) and interferon-gamma (IFN-gamma) transiently increased in plasma. IL-6 increased the protein expression of PS from hepatocytes, while TNF-alpha decreased it from SECs. LPS increased CD14 in hepatocytes and decreased it in SECs, but did not affect toll-like receptor-4 (TLR-4) expression in both cells. Antirat CD14 and antirat TLR-4 antibodies inhibited LPS-induced NFkappaB activation, and a NFkappaB inhibitor suppressed LPS-induced decreased PS expression in both cells. Furthermore, MEK inhibitor blocked LPS-induced decreased PS expression in both cells. CONCLUSIONS: These findings suggest that LPS-induced decreased PS expression in hepatocytes and SECs is mediated by MEK/ERK signaling and NFkappaB activation and that membrane-bound CD14 and TLR-4 are involved in this mechanism. These findings may explain in part the decreased level of plasma PS and thrombotic tendency in sepsis.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Lipopolysaccharide Receptors/physiology , Lipopolysaccharides/metabolism , Liver/metabolism , MAP Kinase Kinase Kinases/metabolism , NF-kappa B/metabolism , Protein S/biosynthesis , Toll-Like Receptor 4/physiology , Animals , Anticoagulants/pharmacology , Humans , Lipopolysaccharide Receptors/biosynthesis , Lipopolysaccharides/chemistry , Male , RNA, Messenger/metabolism , Rats , Rats, Wistar , Signal Transduction , Toll-Like Receptor 4/biosynthesis
13.
Arterioscler Thromb Vasc Biol ; 26(9): 2168-74, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16840717

ABSTRACT

OBJECTIVE: The protein C anticoagulant pathway is an essential process for attenuating thrombin generation by the membrane-bound procoagulant complexes tenase and prothrombinase. In this pathway, protein S (PS) serves as a cofactor for activated protein C. PS circulates in plasma both in a free form and in complex with complement component 4b-binding protein (C4BP). C4BP is a known acute phase reactant, thereby suggesting a relation between PS and the acute phase response. Interleukin (IL)-6 has been shown to increase both PS and C4BP gene expression. Our objective was to study the regulation of PS gene expression by IL-6 in detail. METHODS AND RESULTS: IL-6 upregulates both PS mRNA and protein levels in liver-derived HepG2 cells. The promoter of the PS gene (PROS1) was cloned upstream from a luciferase reporter gene. After transfection in HepG2 cells, the luciferase activity was shown to be stimulated by the addition of IL-6. IL-6 exerts its effect through Signal Transducer and Activator of Transcription 3 (STAT3) that interacts with the PROS1 promoter at a binding site in between nucleotides 229 to 207 upstream from the translational start. CONCLUSIONS: IL-6 induces PS expression via STAT3. A possible function for IL-6-induced PS expression in cell survival is discussed.


Subject(s)
Interleukin-6/physiology , Protein S/biosynthesis , STAT3 Transcription Factor/physiology , Cell Line, Tumor , Gene Expression Regulation , Humans , Interleukin-6/pharmacology , Phosphorylation , Promoter Regions, Genetic , Protein S/genetics , Protein S/metabolism , RNA, Messenger/biosynthesis , Response Elements , STAT3 Transcription Factor/metabolism
14.
J Thromb Haemost ; 4(10): 2215-22, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16824189

ABSTRACT

BACKGROUND: Protein S (PS) has activated protein C-independent, direct anticoagulant activity (PS-direct). We reported that both multimers and monomers of affinity-purified PS have PS-direct similar to that in plasma, in contrast to another report. OBJECTIVE: We extended our studies to establish the molecular forms and activity of plasma PS. METHODS: Novel ELISAs were developed that could detect only multimeric, not monomeric, PS because they employed the same monoclonal antibody for capture and detection. PS forms were also examined on native PAGE immunoblots. A new activity assay for PS-direct was applied to plasma and gel-filtered plasma fractions. RESULTS: Plasma PS multimers were clearly demonstrated using the ELISAs; 30-60% of free plasma PS appeared to be multimeric, a proportion similar to that of affinity-purified PS. On immunoblots, plasma PS multimers were more easily detected after gel filtration; plasma PS monomers and several apparent multimers comigrated with respective forms of affinity-purified PS. Antigen elution profiles after gel filtration of plasma revealed at least one major peak of apparent PS multimers (40-55% of free PS appeared multimeric). Biotin-factor Xa could bind to both plasma PS monomers and multimers. Strong plasma PS-direct was demonstrated, and plasma PS monomers, multimers, and PS-C4b-binding protein complexes each reconstituted PS-depleted plasma to similar levels of PS-direct. CONCLUSION: Our data are in disagreement with a report that monomeric purified PS has little PS-direct and that only monomeric PS exists in plasma. We find that both affinity-purified and plasma PS exist as monomers and multimers with similar PS-direct.


Subject(s)
Anticoagulants/pharmacology , Blood Coagulation Tests/methods , Protein S/biosynthesis , Protein S/chemistry , Antibodies, Monoclonal/chemistry , Antigens/chemistry , Chemistry, Clinical/methods , Chromatography , Chromatography, Gel , Enzyme-Linked Immunosorbent Assay , Humans , Ligands , Protein Binding
15.
Thromb Haemost ; 94(1): 101-6, 2005 Jul.
Article in English | MEDLINE | ID: mdl-16113792

ABSTRACT

Retinal vein occlusion (RVO) is a multifactorial disease involving vessel damage, stasis, viscosity and thrombosis. Conflicting findings on hereditary thrombophilic risk factors have been reported and their impact on RVO features remains to be defined. The aim of the present study was to evaluate the prevalence of hereditary thrombophilic risk factors (HTRF) and characteristics of RVO in patients with or without HTRF. The design of the study was a prospective, observational case series. Two hundred and thirty-four patients with RVO were included consecutively. A French healthy population of the same region was studied as control group. The HTRF studied were protein C (PC), protein S (PS) and antithrombin (AT) deficiencies, factor V Leiden (FVL) and factor II 20210A polymorphisms. Chi-Square was used for comparison with the healthy subjects and between RVO patient with and without HTRF according to localisation (branch vs. central), type of RVO (ischemic or non-ischemic), recurrence, age at first event and classical vascular risk factors. Twenty-two patients had HTRF (12 FV Leiden heterozygotes, 9 FII 20210A heterozygotes and 1 PS deficiency). No AT or PC deficiency was detected. Frequencies of PS deficiency, FVL and FII 20210A allele were similar to the reference population as well as to published data in the general caucasian population. Eighty-six patients experienced their first episode before the age of 60 years. Systemic hypertension, glaucoma and angina were significantly less frequent in patients with RVO before 60 years. Fourteen of the 22 patients with one HTRF (64%) experienced their first episode of RVO before the age of 60 years compared to 72 of 212 without HTRF (34%) (p = 0.006). Heterozygote status for FV Leiden was significantly more frequent in patients who had experienced their first episode of RVO before 60 years (p = 0.027). In conclusion, this study suggests a role of FV Leiden in the occurrence of RVO in patients younger than 60 years who exhibit fewer acquired vascular risk factors than in older patients.


Subject(s)
Factor V/genetics , Retinal Vein Occlusion/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Alleles , Antithrombins/biosynthesis , Factor V/biosynthesis , Female , Heterozygote , Humans , Male , Middle Aged , Prevalence , Prospective Studies , Protein C/biosynthesis , Protein S/biosynthesis , Prothrombin/biosynthesis , Retinal Vein Occlusion/epidemiology , Risk Factors , Thrombophilia/epidemiology , Thrombophilia/genetics , Thrombosis , Time Factors
16.
Thromb Haemost ; 93(5): 853-9, 2005 May.
Article in English | MEDLINE | ID: mdl-15886799

ABSTRACT

We determined anticoagulant parameters that depend on protein S function in plasma, i.e. the APC-independent anticoagulant activity of protein S (expressed as pSR) and APC resistance determined with thrombin generation-based tests (expressed as APCsr) as well as plasma levels of total and free protein S and prothrombin in men, women not using oral contraceptives (OC), and in women using second or third generation OC. Thrombin generation in the APC resistance assays was initiated either with factor Xa (Xa-APCsr) or tissue factor (TF-APCsr). The APC-independent anticoagulant activity of protein S was highest in men (pSR=1.69) and gradually decreased from women not using OC (pSR=1.49) via women using second generation (pSR=1.35) to women using third generation OC (pSR=1.27). The pSR correlated inversely with nAPCsr determined with the tissue factor-based APC resistance test (TF-APCsr) but not with nAPCsr determined with the factor Xa-based assay (Xa-APCsr). Multiple linear regression analysis in which sex, OC use, and protein S and prothrombin levels were included as independent variables and the pSR, TF-APCsr or Xa-APCsr as dependent variables indicated that plasma protein S levels poorly predict the pSR and the TF-APCsr, but are the main determinant of the Xa-APCsr. This indicates that OC use alters the expression of protein S activity. This phenomenon can be caused by differences in modulation of the activity of protein S by other plasma proteins that change during OC use or by OC-induced changes in the protein S molecule that impair its anticoagulant activity. Functional impairment of protein S as a result of hormonal influence may, at least in part, contribute to the thrombotic risk of OC users.


Subject(s)
Anticoagulants/pharmacology , Contraceptives, Oral/pharmacology , Protein S/biosynthesis , Thrombosis/chemically induced , Activated Protein C Resistance/blood , Adult , Anticoagulants/chemistry , Anticoagulants/metabolism , Blood Coagulation Tests , Dose-Response Relationship, Drug , Factor Xa/biosynthesis , Female , Humans , Linear Models , Male , Phospholipids/metabolism , Protein C/biosynthesis , Protein Structure, Tertiary , Prothrombin/biosynthesis , Prothrombin/chemistry , Risk , Thrombin/biosynthesis , Thrombin/chemistry , Thromboplastin/biosynthesis , Thrombosis/blood
17.
Thromb Haemost ; 93(5): 867-71, 2005 May.
Article in English | MEDLINE | ID: mdl-15886801

ABSTRACT

From 1998 to 2003, 133 Caucasian women aged 17-40 years (median 29 years) suffering from unexplained recurrent miscarriage (uRM) were consecutively enrolled. In patients and 133 age-matched healthy controls prothrombotic risk factors (factor V (FV) G1691A, factor II (FII) G20210A, MTHFR T677T, 4G/5G plasminogen activator inhibitor (PAI)-1, lipoprotein (Lp) (a), protein C (PC), protein S (PS), antithrombin (AT), antiphospholipid/anticardiolipin (APA/ACA) antibodies) as well as associated environmental conditions (smoking and obesity) were investigated. 70 (52.6%) of the patients had at least one prothrombotic risk factor compared with 26 control women (19.5%; p<0.0001). Body mass index (BMI; p=0.78) and smoking habits (p=0.44) did not differ significantly between the groups investigated. Upon univariate analysis the heterozygous FV mutation, Lp(a) > 30 mg/dL, increased APA/ACA and BMI > 25 kg/m(2) in combination with a prothrombotic risk factor were found to be significantly associated with uRM. In multivariate analysis, increased Lp(a) (odds ratio (OR): 4.7/95% confidence interval (CI): 2.0-10.7), the FV mutation (OR:3.8/CI:1.4-10.7), and increased APA/ACA (OR: 4.5/CI: 1.1-17.7) had independent associations with uRM.


Subject(s)
Abortion, Habitual/blood , Lipoprotein(a)/chemistry , Thrombosis/blood , Abortion, Habitual/diagnosis , Adolescent , Adult , Antibodies, Anticardiolipin/biosynthesis , Antibodies, Antiphospholipid/blood , Anticoagulants/pharmacology , Antithrombins/biosynthesis , Body Mass Index , Case-Control Studies , Factor V/biosynthesis , Female , Follow-Up Studies , Heterozygote , Humans , Lipoprotein(a)/biosynthesis , Logistic Models , Methylenetetrahydrofolate Dehydrogenase (NAD+)/biosynthesis , Multivariate Analysis , Mutation , Odds Ratio , Plasminogen Activator Inhibitor 1/biosynthesis , Protein C/biosynthesis , Protein S/biosynthesis , Prothrombin/biosynthesis , Risk Factors , Thrombosis/diagnosis
18.
J Thromb Haemost ; 3(5): 929-34, 2005 May.
Article in English | MEDLINE | ID: mdl-15869586

ABSTRACT

BACKGROUND: In recent years there has been a significant increase in the diagnosis of sudden sensorineural hearing loss (SSHL) in western, countries with an incidence of 20 of 100,000 people affected every year. No clear causes for this disease have been found thus far, but cochlear ischemia has been hypothesized in patients in whom an infectious episode or acoustic neurinoma have been excluded. OBJECTIVES: The aim of this case-control study was to investigate a number of acquired and inherited thrombophilic risk factors [antithrombin, protein C and S; factor V (FV) Leiden, FII polymorphism; lupus anticoagulant (LA); anticardiolipin (aCL) antibodies; fasting homocysteine (Hcy); lipoprotein(a) (Lp(a)); plasminogen activator inhibitor-1 (PAI-1)] in addition to cardiovascular risk factors in patients with idiopathic SSHL (ISSHL). PATIENTS AND METHODS: We investigated 155 patients (67 male/88 female; age: 55 (range 19-79 years) with a diagnosis of ISSHL within 30 days from the onset of symptoms, and 155 controls (67 male/88 female; age 54 (range 19-78 years). Fasting Hcy levels were significantly higher in patients than in controls [11.6 (6.7-60) micromol/L vs. 8.7 (5.0-24) micromol/L] as well as PAI-1 levels [19 (2-95) mg/dL vs. 14.5 (4.0-87) mg/dL]. Lupus anticoagulant was present in 13 of 155 (8.4%) patients; 20 patients (12.9%) had positivity of aCL (four IgM and 16 IgG). In no patient was a deficiency of physiological clotting inhibitors antithrombin, protein C and protein S found. No significant differences between patients and controls were observed for Lp(a) plasma levels [111 (1-1146) mg/L vs. 103 (11-695) mg/L] and for the presence of FV Leiden (4.5% vs. 4.5%) and FII variant G20210A (3.8% vs. 3.2%). RESULTS AND CONCLUSIONS: Independent risk factors for ISSHL at the multivariate analysis (adjusted for age, sex and the traditional cardiovascular risk factors) were the positivity of aCL: OR 5.6 (95% CI 2.0-15.3); cholesterol levels within the second and third tertiles (with respect to the first tertile): T2 = OR 4.8 (95% CI 1.9-12.6)/T3 = OR 19 (95% CI 7-50.1); PAI-1 and Hcy levels within the third tertile (with respect to the first tertile): OR 20 (95% CI 7.8-78) and OR 4.0 (95% CI 2.0-8.1), respectively. These preliminary data suggest that hypercholesterolemia, hyperhomocysteinemia, elevated PAI-1 levels and anticardiolipin antibodies are associated with ISSHL, so indirectly supporting the hypothesis of a vascular occlusion in the pathogenesis of the disease.


Subject(s)
Hearing Loss, Sensorineural/diagnosis , Thrombophilia/diagnosis , Adult , Aged , Antibodies, Anticardiolipin/biosynthesis , Antithrombins/biosynthesis , Case-Control Studies , Factor V/genetics , Female , Hearing Loss, Sensorineural/complications , Homocysteine/biosynthesis , Humans , Hypercholesterolemia/complications , Hyperhomocysteinemia/complications , Lipoprotein(a)/biosynthesis , Lupus Coagulation Inhibitor/biosynthesis , Male , Middle Aged , Multivariate Analysis , Odds Ratio , Plasminogen Activator Inhibitor 1/biosynthesis , Protein C/biosynthesis , Protein S/biosynthesis , Risk Factors , Time Factors
19.
J Thromb Haemost ; 3(5): 935-8, 2005 May.
Article in English | MEDLINE | ID: mdl-15869587

ABSTRACT

Oral contraceptive (OC) use increases venous thrombosis (VTE) risk and causes activated protein C (APC) resistance. Plasma glucosylceramide (GlcCer) deficiency is associated with VTE and GlcCer functions as an APC anticoagulant cofactor. Because estradiol decreases GlcCer in cultured cells, we hypothesized OC use would decrease plasma GlcCer and contribute to APC resistance. In a pilot study, seven female adults alternatively took second and third generation OCs and plasma samples were analyzed for GlcCer using high performance liquid chromatography and for APC sensitivity using modified prothrombin time assays. Second and third generation OC usage decreased the APC sensitivity ratio by 8.1% +/- 4.7% (P = 0.004) and 11.7% +/- 8.2% (P = 0.013) and plasma GlcCer levels by 10.1% +/- 6.8% (P = 0.008) and 11.0% +/- 5.1% (P = 0.002), respectively. The plasma GlcCer level correlated with the sensitivity of plasma to APC (P = 0.017, r = 0.51, n = 21 plasma samples). Thus, both second and third generation OC usage decreased plasma GlcCer which could cause a reduction in the plasma sensitivity to APC/protein S, thereby potentially increasing VTE risk.


Subject(s)
Activated Protein C Resistance/chemically induced , Contraceptives, Oral/pharmacology , Glucosylceramides/blood , Protein C/metabolism , Activated Protein C Resistance/blood , Chromatography, High Pressure Liquid , Contraceptives, Oral, Combined/administration & dosage , Desogestrel/pharmacology , Estradiol/metabolism , Ethinyl Estradiol/pharmacology , Female , Glucosylceramides/deficiency , Humans , Levonorgestrel/pharmacology , Pilot Projects , Protein S/biosynthesis , Prothrombin Time , Risk , Sensitivity and Specificity , Venous Thrombosis/blood , Venous Thrombosis/chemically induced
20.
Semin Thromb Hemost ; 31(1): 25-32, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15706472

ABSTRACT

Laboratory tests to detect congenital thrombophilia are frequently requested by clinicians. This review attempts to define (1) whether and to what extent laboratory testing may help clinicians make decisions on patient management, (2) the types of conditions to be investigated, and (3) the types of testing to be performed for each condition.


Subject(s)
Chemistry, Clinical/methods , Hematology/methods , Thrombophilia/congenital , Thrombophilia/diagnosis , Activated Protein C Resistance/diagnosis , Antithrombins/biosynthesis , Factor V/genetics , Fibrinogens, Abnormal/genetics , Genotype , Homocysteine/metabolism , Humans , Mutation , Protein C/biosynthesis , Protein S/biosynthesis , Prothrombin/genetics , Reproducibility of Results , Thrombophilia/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...