Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
Add more filters










Publication year range
1.
J Thromb Haemost ; 15(4): 709-720, 2017 04.
Article in English | MEDLINE | ID: mdl-28109042

ABSTRACT

Essentials Recombinant factor VIII (FVIII) is known to be expressed at a low level in cell culture. To increase expression, we used codon-optimization of a B-domain deleted FVIII (BDD-FVIII). This resulted in 7-fold increase of the expression level in cell culture. The biochemical properties of codon-optimized BDD-FVIII were similar to the wild-type protein. SUMMARY: Background Production of recombinant factor VIII (FVIII) is challenging because of its low expression. It was previously shown that codon-optimization of a B-domain-deleted FVIII (BDD-FVIII) cDNA resulted in increased protein expression. However, it is well recognized that synonymous mutations may affect the protein structure and function. Objectives To compare biochemical properties of a BDD-FVIII variants expressed from codon-optimized and wild-type cDNAs (CO and WT, respectively). Methods Each variant of the BDD-FVIII was expressed in several independent Chinese hamster ovary (CHO) cell lines, generated using a lentiviral platform. The proteins were purified by two-step affinity chromatography and analyzed in parallel by PAGE-western blot, mass spectrometry, circular dichroism, surface plasmon resonance, and chromogenic, clotting and thrombin generation assays. Results and conclusion The average yield of the CO was 7-fold higher than WT, whereas both proteins were identical in the amino acid sequences (99% coverage) and very similar in patterns of the molecular fragments (before and after thrombin cleavage), glycosylation and tyrosine sulfation, secondary structures and binding to von Willebrand factor and to a fragment of the low-density lipoprotein receptor-related protein 1. The CO preparations had on average 1.5-fold higher FVIII specific activity (activity normalized to protein mass) than WT preparations, which was attributed to better preservation of the CO structure as a result of considerably higher protein concentrations during the production. We concluded that the codon-optimization of the BDD-FVIII resulted in significant increase of its expression and did not affect the structure-function properties.


Subject(s)
Codon , Factor VIII/genetics , Protein Engineering , Animals , CHO Cells , Cell Line , Cricetinae , Cricetulus , DNA, Complementary/metabolism , Factor VIII/metabolism , Genetic Vectors , Glycosylation , Humans , Lentivirus , Mutation , Peptide Fragments/genetics , Protein Structure, Secondary , Structure-Activity Relationship , Tyrosine/chemistry
2.
Am J Transplant ; 17(1): 272-280, 2017 01.
Article in English | MEDLINE | ID: mdl-27376583

ABSTRACT

Allograft transplantation into sensitized recipients with antidonor antibodies results in accelerated antibody-mediated rejection (AMR), complement activation, and graft thrombosis. We have developed a membrane-localizing technology of wide applicability that enables therapeutic agents, including anticoagulants, to bind to cell surfaces and protect the donor endothelium. We describe here how this technology has been applied to thrombin inhibitors to generate a novel class of drugs termed thrombalexins (TLNs). Using a rat model of hyperacute rejection, we investigated the potential of one such inhibitor (thrombalexin-1 [TLN-1]) to prevent acute antibody-mediated thrombosis in the donor organ. TLN-1 alone was able to reduce intragraft thrombosis and significantly delay rejection. The results confirm a pivotal role for thrombin in AMR in vivo. This approach targets donor organs rather than the recipient and is intended to be directly translatable to clinical use.


Subject(s)
Graft Rejection/prevention & control , Kidney Failure, Chronic/surgery , Kidney Transplantation/adverse effects , Peptides/pharmacology , Thrombin/antagonists & inhibitors , Thrombosis/prevention & control , Animals , Glomerular Filtration Rate , Graft Rejection/etiology , Graft Survival , Kidney Function Tests , Male , Prognosis , Rats , Rats, Inbred Lew , Risk Factors , Thrombosis/etiology
3.
Cell Death Discov ; 1: 15051, 2015.
Article in English | MEDLINE | ID: mdl-27551480

ABSTRACT

We previously demonstrated PAR2 starts upstreamed with tissue factor (TF) and factor VII (FVII), inhibited autophagy via mTOR signaling in HCC. However, the mechanism underlying for merging functions of PAR2 with the coagulation system in HCC progression remained unclear. The present study aimed to investigate the role of TF, FVII and PAR2 in tumor progression of HCC. The expressions of TF, FVII and PAR2 from HCC specimens were evaluated by immunohistochemical stains and western blotting. We found that the expression of FVII, but not TF and PAR2, directly related to the vascular invasion and the clinical staging. Importantly, a lower level of FVII expression was significantly associated with the longer disease-free survival. The addition of FVII but not TF induced the expression of PAR2 and phosphorylation of ERK1/2, whereas knockdown of FVII decreased PAR2 expression and ERK1/2 phosphorylation in HCC cell lines. Furthermore, levels of phosphor-TSC2 (Ser664) were increased after treatment with FVII and PAR2 agonist whereas these were significantly abolished in the presence of a potent and specific MEK/ERK inhibitor U0126. Moreover, mTOR knockdown highly reduced Hep3B migration, which could be reverted by FVII but not TF and PAR2. These results indicated that FVII/PAR2 signaling through MEK/ERK and TSC2 axis for mTOR activation has potent effects on the migration of HCC cells. In addition, FVII/PAR2 signaling elicits an mTOR-independent signaling, which promotes hepatoma cell migration in consistent with the clinical observations. Our study indicates that levels of FVII, but not TF, are associated with tumor migration and invasiveness in HCC, and provides clues that evaluation of FVII expression in HCC may be useful as a prognostic indicator in patients with HCC and may form an alternative target for further therapy.

4.
J Thromb Haemost ; 11(5): 963-74, 2013 May.
Article in English | MEDLINE | ID: mdl-23516969

ABSTRACT

BACKGROUND: CD34(+) α-smooth muscle actin (SMA)(+) cells mediate intimal hyperplasia (IH) after mechanical endoluminal injury. We previously found that IH is tissue factor (TF) dependent. The precise phenotype of the CD34(+) cells mediating IH is unknown and the mechanisms of TF are also unknown. OBJECTIVE: To define the phenotype of cells mediating IH and compare the effects of inhibiting TF on different subsets of CD34(+) cells. METHODS: Endoluminal injury was induced in C57BL/6 and two strains of mice expressing a human tissue factor pathway inhibitor (hTFPI) fusion protein on different subsets of CD34(+) cells. Confocal microscopy, immunocytofluorescence and real-time PCR were used to determine phenotype. RESULTS: Neointimal cells in C57BL/6 mice were defined as a subset of fibrocytes (CD34(+) CD45(+) collagen-1(+) ) expressing SMA, CD31, TIE-2, CXCR4 and CXCL12. Similar cells circulated post-injury and were also found in mice expressing hTFPI on CD34(+) CD31(+) cells, though in these mice, hTFPI inhibited CD31(+) fibrocyte hyperplasia, so no IH developed. Mice with hTFPI on all CD34(+) α-SMA(+) cells repaired arteries back to a pre-injured state. No CD31(+) fibrocytes were found in these mice unless an anti-hTFPI antibody was administered. Similar findings in protease activated receptor (PAR)-1-deficient mice suggested hTFPI prevented thrombin signaling through PAR-1. In vitro, thrombin increased the number of CD31(+) fibrocytes. CONCLUSIONS: Inhibition of TF on CD31(+) fibrocytes inhibits IH whereas inhibition on all CD34(+) α-SMA(+) cells (or PAR-1 deficiency) inhibits the appearance of CD31(+) fibrocytes and promotes repair. These data enhance our understanding of IH and suggest novel ways to promote regenerative repair.


Subject(s)
Fibroblasts/pathology , Hyperplasia/pathology , Thromboplastin/physiology , Tunica Intima/pathology , Animals , Fibroblasts/immunology , Immunophenotyping , Mice , Real-Time Polymerase Chain Reaction
6.
Gene Ther ; 19(9): 936-46, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22071970

ABSTRACT

The efficient delivery of genetic material to the developing fetal brain represents a powerful research tool and a means to supply therapy in a number of neonatal lethal neurological disorders. In this study, we have delivered vectors based upon adenovirus serotype 5 (Ad5) and adeno-associated virus (AAV) pseudotypes 2/5, 2/8 and 2/9 expressing green fluorescent protein to the E16 fetal mouse brain. One month post injection, widespread caudal to rostral transduction of neural cells was observed. In discrete areas of the brain these vectors produced differential transduction patterns. AAV2/8 and 2/9 produced the most extensive gene delivery and had similar transduction profiles. All AAV pseudotypes preferentially transduced neurons whereas Ad5 transduced both neurons and glial cells. None of the vectors elicited any significant microglia-mediated immune response when compared with control uninjected mice. Whole-body imaging and immunohistological evaluation of brains 9 months post injection revealed long-term expression using these non-integrating vectors. These data will be useful in targeting genetic material to discrete or widespread areas of the fetal brain with the purpose of devising therapies for early neonatal lethal neurodegenerative disease and for studying brain development.


Subject(s)
Adenoviridae/genetics , Brain/metabolism , Dependovirus/genetics , Gene Transfer Techniques , Genetic Vectors , Animals , Brain/embryology , Female , Green Fluorescent Proteins/genetics , Mice , Neuroglia/metabolism , Neurons/metabolism , Transduction, Genetic
8.
J Thromb Haemost ; 7(7): 1106-13, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19422457

ABSTRACT

BACKGROUND: Mouse tissue factor pathway inhibitor (TFPI) is produced in three alternatively spliced isoforms that differ in domain structure and mechanism for cell surface binding. Tissue expression of TFPI isoforms in mice was characterized as an initial step for identification of their physiological functions. METHODS AND RESULTS: Sequence homology demonstrates that TFPIalpha existed over 430 Ma while TFPIbeta and TFPIgamma evolved more recently. In situ hybridization studies of heart and lung did not reveal any cells exclusively expressing a single isoform. Although our previous studies have demonstrated that TFPIalpha mRNA is more prevalent than TFPIbeta or TFPIgamma mRNA in mouse tissues, western blot studies demonstrated that TFPIbeta is the primary protein isoform produced in adult tissues, while TFPIalpha is expressed during embryonic development and in placenta. Consistent with TFPIbeta as the primary isoform produced within adult vascular beds, the TFPI isoform in mouse plasma migrates like TFPIbeta in SDS-PAGE and mice have a much smaller heparin-releasable pool of plasma TFPIalpha than humans. CONCLUSIONS: The data demonstrate that alternatively spliced isoforms of TFPI are temporally expressed in mouse tissues at the level of protein production. TFPIalpha and TFPIbeta are produced in embryonic tissues and in placenta while adult tissues produce almost exclusively TFPIbeta.


Subject(s)
Alternative Splicing , Lipoproteins/genetics , Amino Acid Sequence , Animals , Blotting, Western , Conserved Sequence , Enzyme-Linked Immunosorbent Assay , Female , In Situ Hybridization , Lipoproteins/chemistry , Lung/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Molecular Sequence Data , Myocardium/metabolism , Placenta/metabolism , RNA, Messenger/genetics , Sequence Homology, Amino Acid
9.
Am J Transplant ; 7(3): 499-506, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17229071

ABSTRACT

Coagulation proteases are involved in generating fibrin after vascular injury (hemostasis) but they also have multiple other effects, many of which are mediated independently of fibrin generation, via interactions with specific cell membrane-expressed "protease activated receptors". In inflammation, this family of proteins has a complex influence, the facets of which are still incompletely understood, though a common feature in different models appears to be amplification of innate signals that are initially generated by pathogenic elements or, in the context of transplantation, ischemia or anti-graft antibodies, for instance. There is increasing evidence that these proteases may also have specific effects on cells involved in adaptive immunity and on cells that mediate chronic inflammation and fibrosis. Understanding whether these effects are relevant in the responses generated against transplanted organs is important, as it could lead ultimately to the development of novel ways to promote long-term graft survival.


Subject(s)
Blood Coagulation Factors/physiology , Graft Survival , Organ Transplantation , Peptide Hydrolases/physiology , Thrombin/physiology , Thrombosis/enzymology , Animals , Blood Coagulation , Humans , Immunity , Immunosuppressive Agents/adverse effects , Inflammation/enzymology , Inflammation/immunology , Mice , Receptors, Thrombin/agonists
10.
J Thromb Haemost ; 4(10): 2191-8, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16803463

ABSTRACT

BACKGROUND: Coagulation proteins promote neointimal hyperplasia and vascular remodelling after vessel injury, but the precise mechanisms by which they act in vivo remain undetermined. OBJECTIVES: This study, using an injury model in which the neointima is derived from bone marrow (BM)-derived cells, compared inhibition of tissue factor or thrombin on either BM-derived or existing vascular smooth muscle cells. METHODS: Two transgenic (Tg) mouse strains expressing membrane-tethered tissue factor pathway inhibitor (TFPI) or hirudin (Hir) fusion proteins driven by an alpha smooth muscle actin (SMA) promoter were generated (alpha-TFPI-Tg and alpha-Hir-Tg) and the phenotype after wire-induced endovascular injury was compared with that in wild-type (WT) controls. RESULTS: WT mice developed progressive neointimal expansion, whereas injury in either Tg was followed by repair back to a preinjured state. This was also seen when WT mice were reconstituted with BM from Tg mice but not when Tgs were reconstituted with WT BM, in which injury was followed by slowly progressive neointimal expansion. Injection of CD34+ cells from Tg mice into injured WT mice resulted in the accumulation of fusion protein-expressing cells from day 3 onwards and an absence of neointimal hyperplasia in those areas. CONCLUSIONS: Neointimal development after wire-induced endovascular injury in mice was completely inhibited when BM-derived cells infiltrating the damaged artery expressed membrane tethered anticoagulant fusion proteins under an alpha-SMA promoter. These findings enhance our understanding of the pathological role that coagulation proteins play in vascular inflammation.


Subject(s)
Anticoagulants/metabolism , Antigens, CD34/biosynthesis , Bone Marrow Cells/metabolism , Cell Membrane/metabolism , Gene Expression Regulation , Recombinant Fusion Proteins/metabolism , Stem Cells/metabolism , Animals , Aorta/metabolism , Arteriosclerosis/therapy , Blood Vessels/pathology , Carotid Arteries/pathology , Humans , Inflammation , Mice , Mice, Transgenic , Muscle, Smooth/metabolism , Phenotype
11.
Br J Haematol ; 123(3): 496-501, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14617013

ABSTRACT

We report a kindred in which two siblings suffered spontaneous venous thromboses in the second decade of life. Further investigation showed reduced coagulation factor V (FV) activity and activated protein C resistance (APCR) ratio but no other thrombophilic abnormalities. The reduction in APCR ratio persisted in a modified APCR assay in which FV activity was normalized between test and control plasmas. Analysis of the FV gene showed that the thrombotic individuals had a complex genotype that included two novel point mutations c.529G>T and c.1250T>C resulting in FV E119X and FV I359T substitutions inherited on different alleles. Individuals in the kindred with FV E119X or FV I359T substitutions alone were asymptomatic. We suggest that the FV I359T substitution confers pro-thrombotic risk and APCR, but that this is only clinically manifest when co-inherited with the FV E119X allele. The FV I359T substitution creates a new consensus sequence for N-linked glycosylation within the FV heavy chain and we speculate that this abnormal glycosylation may disrupt activated protein C-mediated proteolysis of the variant FV and FVa.


Subject(s)
Activated Protein C Resistance/genetics , Factor V/genetics , Point Mutation , Thrombosis/genetics , Activated Protein C Resistance/diagnosis , Adolescent , Genotype , Humans , Male , Pedigree , Sequence Analysis, DNA , Thrombosis/diagnosis
12.
J Thromb Haemost ; 1(9): 1935-44, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12941034

ABSTRACT

UNLABELLED: Coagulation factors (F)VIIa, FXa and thrombin are implicated in cellular responses in vascular, mesenchymal and inflammatory cells. Fibroblasts are the most abundant cells in connective tissue, and damage to blood vessels places coagulation factors in contact with these and other cell types. OBJECTIVES: To investigate cellular responses of primary dermal fibroblasts to FVIIa, FXa and thrombin by following changes in expression of candidate proteins: monocyte chemotactic protein-1 (MCP-1), interleukin-8 (IL-8), interleukin-6 (IL-6) and vascular endothelial growth factor (VEGF), and to determine the expression of receptors implicated in signaling by these coagulation factors. METHODS: Steady-state mRNA levels were quantified by RNase protection assay, and protein secretion by ELISA. PAR gene expression was assessed by ribonuclease protection assay and conventional and quantitative reverse-transcription-polymerase chain reaction. RESULTS: FVIIa did not induce the candidate genes. In contrast, FXa and thrombin induced MCP-1 mRNA and protein secretion strongly, IL-8 moderately, and IL-6 weakly. Neither FXa nor thrombin induced VEGF mRNA or protein secretion, although FXa induced VEGF protein secretion in lung fibroblasts. Comparison of the presence of candidate receptors in the two fibroblast subtypes demonstrated higher levels of PAR-1 and PAR-3 in lung fibroblasts relative to their dermal counterparts and the additional expression of PAR-2. CONCLUSIONS: FXa and thrombin induce expression of MCP-1, IL-8 and IL-6, and distribution and expression of PARs on dermal fibroblasts is reduced relative to their lung counterparts. Tissue origin may influence the cellular response of fibroblasts to coagulation proteases.


Subject(s)
Blood Coagulation Factors/pharmacology , Fibroblasts/drug effects , Gene Expression Regulation/drug effects , Skin/cytology , Cells, Cultured , Chemokine CCL2/genetics , Factor VIIa/pharmacology , Factor X/pharmacology , Fibroblasts/metabolism , Humans , Interleukin-6/genetics , Interleukin-8/genetics , Lung/cytology , RNA, Messenger/analysis , RNA, Messenger/drug effects , Receptor, PAR-1/genetics , Receptor, PAR-2/genetics , Receptors, Thrombin/genetics , Thrombin/pharmacology , Vascular Endothelial Growth Factor A/genetics
13.
J Thromb Haemost ; 1(1): 139-46, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12871551

ABSTRACT

Inhibitor antibody formation is a complication of factor VIII (FVIII) replacement therapy due to a failure to synthesize sufficient FVIII protein to induce immune tolerance. The incidence of nonsense mutations in inhibitor patients is high, however, this association is variable according to the position of the mutation. We have studied the effect of nonsense mutations on accumulation of FVIII mRNA, protein translation and secretion. Appropriately processed mRNA was detected in cells transfected with wild-type R1966X and R2116X expression constructs and no evidence of nonsense-mediated decay was observed. All constructs directed the translation of detectable intracellular FVIII antigen, however, secreted FVIII was detected only in conditioned media of cells transfected with wild-type cDNA. We have also analyzed ectopic FVIII mRNA transcripts in the lymphocytes of six hemophilia A patients with nonsense mutations (Q139X, R583X, R1941X, R1966X and two unrelated patients with R2116X). FVIII mRNA was detectable in every case. In R1941X and R1966X only normally spliced transcripts were present. In Q139X, R583X and R2116X aberrantly spliced transcripts were observed with two distinct patterns in two individuals with the R2116X mutation. No correlation between mutation, transcript pattern and incidence of inhibitor development was apparent.


Subject(s)
Codon, Nonsense/genetics , Codon, Nonsense/immunology , Factor VIII/genetics , Factor VIII/immunology , Alternative Splicing/genetics , Animals , Antibodies/blood , Antibodies/genetics , Antibodies/immunology , Antigens/genetics , Base Sequence , CHO Cells , Codon, Nonsense/metabolism , Cricetinae , DNA, Complementary/genetics , DNA, Complementary/metabolism , Factor VIII/metabolism , Hemophilia A/blood , Hemophilia A/genetics , Hemophilia A/immunology , Humans , Lymphocytes/immunology , Lymphocytes/metabolism , Point Mutation , Protein Biosynthesis , RNA, Messenger/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Transfection
14.
J Thromb Haemost ; 1(7): 1487-94, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12871284

ABSTRACT

In mammalian blood coagulation, five proteases (factor VII [FVII]; factor IX [FIX]; factor X [FX]; protein C [PC] and prothrombin [PT]) act with five cofactors (tissue factor [TF]; factor V [FV]; factor VIII [FVIII]; thrombomodulin and protein S) to control the generation of fibrin. Biochemical evidence, molecular cloning data and comparative sequence analysis support the existence of all components of this network in all jawed vertebrates, and strongly suggest that it evolved before the divergence of teleosts over 430 million years ago. Phylogenetic analysis of the amino acid sequences of the Gla-EGF1-EGF2-SP domain serine proteases (FVII, FIX, FX, PC) and the A domain-containing cofactors (FV and FVIII) strongly supports the evolution of the blood coagulation network through two rounds of gene duplication, and supports the hypothesis that vertebrate evolution benefited from two global genome duplications. The jawless vertebrates (hagfish and lamprey) that diverged over 450 million years ago have a blood coagulation network involving TF, PT and fibrinogen. Preliminary evidence indicates that they may have a smaller complement of Gla-EGF1-EGF2-SP domain proteins, suggesting the existence of a 'primitive' coagulation system in jawless vertebrates.


Subject(s)
Hemostasis/physiology , Animals , Biological Evolution , Factor IX/chemistry , Factor VII/chemistry , Factor X/chemistry , Fibrinogen/chemistry , Humans , Models, Biological , Models, Genetic , Phylogeny , Protein C/chemistry , Protein Structure, Tertiary , Prothrombin/chemistry
15.
Xenotransplantation ; 8(4): 258-65, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11737851

ABSTRACT

Aside from their critical role in thrombosis, activated coagulation factors also have inflammatory properties and these may be important during delayed xenograft rejection (DXR). This study assessed whether porcine EC could be activated by factor Xa (FXa) and thrombin (FIIa) and whether expression of tissue factor pathway inhibitor (TFPI)-CD4 and hirudin-CD4 fusion proteins could prevent such activation. Incubation of porcine EC with human FXa and FIIa induced cell surface expression of E-selectin, VCAM and tissue factor (TF) in a time-dependent and concentration-dependent manner. In contrast, porcine EC transfected with a human TFPI-CD4 fusion protein were selectively resistant to these pro-inflammatory effects of FXa but not FIIa. Likewise, the transfectants expressing the hirudin-CD4 fusion protein were selectively resistant to the pro-inflammatory effects of FIIa but not those of FXa. When combined, the FXa and FIIa had an additive effect on the activation of control EC. In contrast, coexpression of both hirudin-CD4 and TFPI-CD4 fusion proteins completely inhibited the upregulation of VCAM with the FXa/FIIa mix. These results indicate that expression of novel anticoagulant fusion proteins on the surface of porcine EC can protect against EC activation induced by human coagulation factors FXa and FIIa. In vivo, we anticipate that expression of these fusion proteins on the endothelium of transplanted xenografts, besides preventing intravascular thrombosis, will also protect against EC activation induced by trace amounts of FIIa and FXa, thereby further protecting the grafts from DXR.


Subject(s)
Endothelium, Vascular/cytology , Factor Xa/pharmacology , Graft Rejection/prevention & control , Hirudins/physiology , Lipoproteins/physiology , Thrombin/pharmacology , Thrombosis/prevention & control , Animals , Blood Coagulation , CD4 Antigens/genetics , CD4 Antigens/physiology , Cell Adhesion , Cells, Cultured/drug effects , E-Selectin/biosynthesis , E-Selectin/genetics , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Enzyme Activation , Gene Expression Regulation/drug effects , Hirudins/genetics , Humans , Lipoproteins/genetics , Postoperative Period , Prothrombin/pharmacology , Recombinant Fusion Proteins/physiology , Swine , Thromboplastin/biosynthesis , Thromboplastin/genetics , Thromboplastin/metabolism , Transfection , Vascular Cell Adhesion Molecule-1/biosynthesis , Vena Cava, Inferior/cytology
16.
Br J Haematol ; 113(3): 604-15, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11380445

ABSTRACT

In haemophilia A, the functional defect at the molecular level of most factor VIII (FVIII) missense mutations remains unknown. Site-directed mutagenesis of B domain-deleted FVIII cDNA (FVIIISQ) was used to introduce two mutations associated with severe cross-reacting material (CRM)-negative (FVIII-C329S) or mild/moderate CRM-reduced (FVIII-G1948D) haemophilia A. Wild-type (FVIIISQ-WT) and variant FVIIISQ proteins were successfully expressed after stable transfection in Chinese hamster ovary (CHO) cells, and partially characterized at the intracellular, molecular and functional levels. Reverse transcription polymerase chain reaction analysis confirmed that both transcription and mRNA processing appeared normal in CHO cells transfected with both the wild-type and two variant constructs. In contrast to FVIIISQ-WT, immunofluorescence analysis of both CRM(-) and CRM(r) variants showed intracellular FVIII accumulation within the rough endoplasmic reticulum, suggesting secretion defects in transfected CHO cells. Immunoblot analysis of the FVIIISQ variant proteins that were secreted showed that they were expressed as mixed populations of uncleaved 170 kDa polypeptides, processed 90 kDa heavy chains and 80 kDa light chains, similar to FVIIISQ-WT. Phenotypic analysis of the B domain-deleted FVIIISQ variants expressed in CHO cells correlated well with the patients' reduced FVIII activity and, in addition, surface plasmon resonance studies demonstrated that both missense mutations were associated with increased rates of A2 domain dissociation following thrombin activation. We conclude that the mutations found are responsible for the haemophilia A phenotype, through intracellular retention and decreased stability of the active cofactor FVIIIa.


Subject(s)
Factor VIII/genetics , Hemophilia A/genetics , Mutation, Missense , Animals , Biosensing Techniques , CHO Cells , Cricetinae , Factor VIII/analysis , Fluorescent Antibody Technique , Gene Expression , Humans , Immunoblotting , Mutagenesis, Site-Directed , Phenotype , Regression Analysis , Reverse Transcriptase Polymerase Chain Reaction , Thrombin/therapeutic use , Transgenes
17.
FEBS Lett ; 497(1): 31-8, 2001 May 18.
Article in English | MEDLINE | ID: mdl-11376658

ABSTRACT

Micronemes are specialised organelles, found in all apicomplexan parasites, which secrete molecules that are essential for parasite attachment to and invasion of host cells. Regions of several microneme proteins have sequence similarity to the Apple domains (A-domains) of blood coagulation factor XI (FXI) and plasma pre-kallikrein (PK). We have used mass spectrometry on a recombinant-expressed, putative A-domain from the microneme protein EtMIC5 from Eimeria tenella, to demonstrate that three intramolecular disulphide bridges are formed. These bridges are analogous to those that stabilise A-domains in FXI and PK. The data confirm that the apicomplexan domains are structural homologues of A-domains and are therefore novel members of the PAN module superfamily, which also includes the N-terminal domains of members of the plasminogen/hepatocyte growth factor family. The role of A-domains/PAN modules in apicomplexan parasites is not known, but their presence in the microneme suggests that they may be important for mediating protein-protein or protein-carbohydrate interactions during parasite attachment and host cell invasion.


Subject(s)
Apicomplexa/physiology , Factor XI/chemistry , Organelles/metabolism , Prekallikrein/chemistry , Protozoan Proteins/chemistry , Amino Acid Motifs/physiology , Animals , Conserved Sequence , Disulfides/chemistry , Eimeria tenella , Gas Chromatography-Mass Spectrometry , Glycoproteins/chemistry , Glycoproteins/genetics , Glycoproteins/metabolism , Host-Parasite Interactions/physiology , Multigene Family , Peptide Fragments/analysis , Protein Structure, Tertiary/physiology , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid
18.
Hum Mutat ; 17(1): 3-17, 2001.
Article in English | MEDLINE | ID: mdl-11139238

ABSTRACT

Factor VII (FVII) is a zymogen for a vitamin K-dependent serine protease essential for the initiation of blood coagulation. It is synthesized primarily in the liver and circulates in plasma at a concentration of approximately 0.5 microg/ml (10 nmol/L). The FVII gene (F7) is located on chromosome 13 (13q34), consists of 9 exons, and spans approximately 12kb. It encodes a mature protein of 406 amino acids, which has an N-terminal domain (Gla) post-translationally modified by gamma-carboxylation of glutamic acid residues, two domains with homology to epidermal growth factor (EGF1 and 2), and a C-terminal serine protease domain. The single chain zymogen is activated by proteolytic cleavage at Arg152-Ile153. There are 238 individuals described in the world literature with mutations in their F7 genes (FVII mutation database; europium.csc. mrc.ac.uk). Complete absence of FVII activity in plasma is usually incompatible with life, and individuals die shortly after birth due to severe hemorrhage. The majority of individuals with mutations in their F7 gene(s), however, are either asymptomatic or the clinical phenotype is unknown. In general, a severe bleeding phenotype is only observed in individuals homozygous for a mutation in their F7 genes with FVII activities (FVII:C) below 2% of normal, however, a considerable proportion of individuals with a mild-moderate bleeding phenotype have similar FVII:C by in vitro assay. The failure of in vitro tests to differentiate between these groups may be due to lack of sensitivity in the assays to the very low amounts of FVII:C, which are sufficient to initiate coagulation in vivo. A number of polymorphisms have been identified in the F7 gene and some have been shown to influence plasma FVII antigen levels.


Subject(s)
Databases, Factual , Factor VII Deficiency/genetics , Factor VII/genetics , Mutation/genetics , Animals , Genes, Lethal/genetics , Humans
19.
Thromb Haemost ; 84(5): 833-40, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11127865

ABSTRACT

Coagulation factor XI (FXI) deficiency is an inherited autosomal recessive mild bleeding disorder. In this study, we report the molecular genetic analysis of FXI deficiency in six unrelated families of Portuguese origin. The Jewish type II mutation was found in two families, of seemingly Portuguese origin. Haplotype analysis in these families demonstrated that this mutation is of Jewish origin. In the remaining families, five novel FXI mutations have been identified. Two of these mutations (FXI IVS K -10T-->A and FXI 1026G-->T, cd 324) affect the FXI pre-mRNA splicing. A further two (FXI 307 ins AAGCAAT, cd 85 and FXI 1072 del A, cd 340) introduce frameshifts leading to premature termination codons. The FXI splicing mutation, 1026G-->T cd 324, was found in compound heterozygosity with missense mutation FXI K518N. Analysis of the FXI mRNA from the latter genotype demonstrated new donor splice site usage. All reported mutations most likely result in functional null-alleles. In addition, three novel polymorphisms have been identified: at nt -138 in intron A, at codon D125 in exon 5 and at codon T249 in exon 8.


Subject(s)
Factor XI Deficiency/genetics , Factor XI/genetics , Alleles , Female , Humans , Male , Mutation , Pedigree , Portugal
20.
Hum Genet ; 107(4): 327-42, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11129332

ABSTRACT

Factor VII (FVII) deficiency is a rare haemorrhagic condition, normally inherited as an autosomal recessive trait, in which clinical presentation is highly variable and correlates poorly with laboratory phenotype. The FVII (F7) gene was sequenced in 48 unrelated individuals with FVII deficiency, yielding a total of 23 novel lesions including 15 missense mutations, 2 micro-deletions, 5 splice junction mutations and a single base-pair substitution in the 5' untranslated region. Family studies were performed in order to distinguish the contributions of individual mutant F7 alleles to the clinical and laboratory phenotypes. Specific missense mutations were evaluated by molecular modelling in the context of the FVIIa-tissue factor crystal structure. Single base-pair substitutions in splice sites and the 5' untranslated region were studied by in vitro splicing assay and luciferase reporter gene assay, respectively. All probands were also typed for four previously reported F7 polymorphisms. In the majority of cases of FVII deficiency studied here, consideration of both mutational and polymorphism data permitted the derivation of plausible explanations for the FVII activity and antigen levels measured in the laboratory. Inter-familial variation in FVII activity and the antigen levels of heterozygous relatives of probands was found to be significantly higher than intra-familial variation, consistent with the view that the nature of the F7 gene lesion(s) segregating in a given family is a prime determinant of laboratory phenotype. Although no relationship could be discerned between laboratory phenotype and polymorphism genotype, the frequencies of the A2 and M2 polymorphic alleles were significantly higher in the FVII-deficient individuals tested than in controls. This suggests that the presence of these alleles may have served to increase the likelihood of pathological F7 gene lesions coming to clinical attention.


Subject(s)
Factor VII Deficiency/genetics , Factor VII/genetics , Mutation , 5' Untranslated Regions , Amino Acid Substitution , Base Sequence , DNA Mutational Analysis , DNA Primers/genetics , Factor VII/chemistry , Genotype , Humans , In Vitro Techniques , Models, Molecular , Molecular Biology , Mutation, Missense , Phenotype , Point Mutation , Polymerase Chain Reaction , Polymorphism, Genetic , Protein Conformation , RNA Splicing/genetics , Sequence Deletion
SELECTION OF CITATIONS
SEARCH DETAIL
...