Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 111
Filter
1.
Protein Expr Purif ; 137: 26-33, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28651975

ABSTRACT

Recombinant factor VII (rFVII) is the main therapeutic choice for hemophilia patients who have developed inhibitory antibodies against conventional treatments (FVIII and FIX). Because of the post-translational modifications, rFVII needs to be produced in mammalian cell lines. In this study, for the first time, we have shown efficient rFVII production in HepG2, Sk-Hep-1, and HKB-11 cell lines. Experiments in static conditions for a period of 96 h showed that HepG2-FVII produced the highest amounts of rhFVII, with an average of 1843 ng/mL. Sk-hep-1-FVII cells reached a maximum protein production of 1432 ng/mL and HKB-11-FVII cells reached 1468 ng/mL. Sk-Hep-1-rFVII and HKB-11-rFVII were selected for the first step of scale-up. Over 10 days of spinner flask culture, HKB-11 and SK-Hep-1 cells showed a cumulative production of rFVII of 152 µg and 202.6 µg in 50 mL, respectively. Thus, these human cell lines can be used for an efficient production of recombinant FVII. With more investment in basic research, human cell lines can be optimized for the commercial production of different bio therapeutic proteins.


Subject(s)
Factor VII , Gene Expression , Cell Line , Factor VII/biosynthesis , Factor VII/genetics , Factor VII/isolation & purification , Humans , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification
2.
Blood ; 119(4): 957-66, 2012 Jan 26.
Article in English | MEDLINE | ID: mdl-22134170

ABSTRACT

We explored adeno-associated viral vector (AAV)-mediated gene transfer in the perinatal period in animal models of severe congenital factor VII (FVII) deficiency, a disease associated with early postnatal life-threatening hemorrhage. In young adult mice with plasma FVII < 1% of normal, a single tail vein administration of AAV (1 × 10(13) vector genomes [vg]/kg) resulted in expression of murine FVII at 266% ± 34% of normal for ≥ 67 days, which mediated protection against fatal hemorrhage and significantly improved survival. Codon optimization of human FVII (hFVIIcoop) improved AAV transgene expression by 37-fold compared with the wild-type hFVII cDNA. In adult macaques, a single peripheral vein injection of 2 × 10(11) vg/kg of the hFVIIcoop AAV vector resulted in therapeutic levels of hFVII expression that were equivalent in males (10.7% ± 3.1%) and females (12.3% ± 0.8%). In utero delivery of this vector in the third trimester to fetal monkeys conferred expression of hFVII at birth of 20.4% ± 3.7%, with a gradual decline to > 1% by 7 weeks. Re-administration of an alternative serotype at 12 months postnatal age increased hFVII levels to 165% ± 6.2% of normal, which remained at therapeutic levels for a further 28 weeks without toxicity. Thus, perinatal AAV-mediated gene transfer shows promise for disorders with onset of pathology early after birth.


Subject(s)
Dependovirus , Factor VII Deficiency/therapy , Factor VII/therapeutic use , Genetic Therapy/methods , Genetic Vectors , Hemorrhage/prevention & control , Perinatal Care , Animals , Animals, Newborn , Codon , Dependovirus/genetics , Factor VII/analysis , Factor VII/biosynthesis , Factor VII/genetics , Factor VII Deficiency/blood , Factor VII Deficiency/genetics , Factor VII Deficiency/physiopathology , Female , Fetal Therapies/adverse effects , Gene Expression , Genetic Therapy/adverse effects , Genetic Vectors/administration & dosage , Genetic Vectors/adverse effects , Hemorrhage/etiology , Hep G2 Cells , Humans , Injections, Intravenous , Macaca mulatta , Male , Mice , Pregnancy , Sex Characteristics , Survival Analysis
3.
Biochem J ; 449(1): 231-9, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23050902

ABSTRACT

Coagulation FVII (Factor VII) is a vitamin K-dependent glycoprotein synthesized in hepatocytes. It was reported previously that FVII gene (F7) expression was up-regulated by ribavirin treatment in hepatitis C virus-infected haemophilia patients; however, its precise mechanism is still unknown. In the present study, we investigated the molecular mechanism of ribavirin-induced up-regulation of F7 expression in HepG2 (human hepatoma cell line). We found that intracellular GTP depletion by ribavirin as well as other IMPDH (inosine-5'-monophosphate dehydrogenase) inhibitors, such as mycophenolic acid and 6-mercaptopurine, up-regulated F7 expression. FVII mRNA transcription was mainly enhanced by accelerated transcription elongation, which was mediated by the P-TEFb (positive-transcription elongation factor b) complex, rather than by promoter activation. Ribavirin unregulated ELL (eleven-nineteen lysine-rich leukaemia) 3 mRNA expression before F7 up-regulation. We observed that ribavirin enhanced ELL3 recruitment to F7, whereas knockdown of ELL3 diminished ribavirin-induced FVII mRNA up-regulation. Ribavirin also enhanced recruitment of CDK9 (cyclin-dependent kinase 9) and AFF4 to F7. These data suggest that ribavirin-induced intracellular GTP depletion recruits a super elongation complex containing P-TEFb, AFF4 and ELL3, to F7, and modulates FVII mRNA transcription elongation. Collectively, we have elucidated a basal mechanism for ribavirin-induced FVII mRNA up-regulation by acceleration of transcription elongation, which may be crucial in understanding its pleiotropic functions in vivo.


Subject(s)
Antimetabolites/pharmacology , Factor VII/genetics , Gene Expression Regulation , Guanosine Triphosphate/deficiency , Intracellular Fluid/metabolism , Ribavirin/pharmacology , Transcription Elongation, Genetic/physiology , Factor VII/biosynthesis , Gene Expression Regulation/drug effects , Guanosine Triphosphate/antagonists & inhibitors , Hep G2 Cells , Humans , Transcription Elongation, Genetic/drug effects
4.
Genet Mol Res ; 12(4): 6813-24, 2013 Dec 16.
Article in English | MEDLINE | ID: mdl-24391029

ABSTRACT

Human coagulation factor VII (FVII) plays an important role in the blood coagulation process and exists in micro amounts in human plasma; therefore, any attempt at the large-scale production of FVII in significant quantities is challenging. The purpose of this study was to express and obtain biologically active recombinant FVII (rFVII) from Chinese hamster ovary K1 (CHO-K1) cells. The full-length FVII cDNA was isolated from a HepG2 cell line and then subcloned in pcDNA3.1 to construct an expression vector, pcDNA-FVII. CHO-K1 cells were transfected with 1 µg pcDNA-FVII. The cell line that stably expressed secretory FVII was screened using 900 µg/mL G418. The FVII copy number in CHO-K1 cells was detected by quantitative polymerase chain reaction (qPCR). The rFVII was purified in ligand affinity chromatography medium. The purified protein was detected by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and Western blot analysis. The biological activity of the purified FVII protein was determined by a prothrombin time assay. Three cell lines that permanently expressed rFVII were screened. The qPCR results demonstrated that each CHO-K1 cell harbored two FVII DNA copies. The SDS-PAGE and Western blot analysis showed that the purified protein was about 50 kDa. The purity of the target protein was 95%. The prothrombin time assay indicated that the FVII-specific activity of rFVII was 2573 ± 75 IU/mg. This method enabled the fast preparation of high-purity rFVII from CHO-K1 cells, and the purified protein had good biological activity.


Subject(s)
Cloning, Molecular , Factor VII/genetics , Recombinant Proteins/genetics , Animals , Base Sequence , Blood Coagulation/genetics , Blood Coagulation/physiology , CHO Cells , Cell Line , Cricetinae , Cricetulus , Factor VII/biosynthesis , Hep G2 Cells , Humans , Real-Time Polymerase Chain Reaction , Recombinant Proteins/biosynthesis , Sequence Analysis, DNA
5.
Blood ; 113(25): 6461-4, 2009 Jun 18.
Article in English | MEDLINE | ID: mdl-19387004

ABSTRACT

Our previous studies with genomic minigenes have demonstrated that an engineered small nuclear RNA-U1 (U1+5a) partially rescued coagulation factor VII (FVII) mRNA processing impaired by the 9726+5G>A mutation. Here, to evaluate the U1+5a effects on FVII function, we devised a full-length FVII splicing-competent construct (pSCFVII-wt). This construct drove in COS-1 cells the synthesis of properly processed FVII transcripts and of secreted functional FVII (23 +/- 4 ng/mL), which were virtually undetectable upon introduction of the 9726+5G>A mutation (pSCFVII-9726+5a). Cotransfection of pSCFVII-9726+5a with pU1+5a resulted in a partial rescue of FVII splicing and protein biosynthesis. The level increase in medium was dose dependent and, with a molar excess (1.5x) of pU1+5a, reached 9.5% plus or minus 3.2% (5.0 +/- 2.8 ng/mL) of FVII-wt coagulant activity. These data provide the first insights into the U1-snRNA-mediated rescue of donor splice sites at protein level, thus further highlighting its therapeutic implications in bleeding disorders, which would benefit even from tiny increase of functional levels.


Subject(s)
Factor VII/genetics , RNA Splice Sites/genetics , RNA, Small Nuclear/physiology , Animals , COS Cells/metabolism , Chlorocebus aethiops , Factor VII/biosynthesis , Factor VII/metabolism , Genes, Synthetic , Genetic Engineering , Humans , Point Mutation , RNA/genetics , RNA/physiology , RNA Splicing , RNA, Small Nuclear/chemistry , RNA, Small Nuclear/genetics , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Transfection
6.
J Biomed Biotechnol ; 2011: 873874, 2011.
Article in English | MEDLINE | ID: mdl-21912483

ABSTRACT

The variety of recombinant protein expression systems have been developed as a resource of FVII gene expression. In the current study, the authors used a novel protein expression system based on the Iranian Lizard Leishmania, a trypanosomatid protozoan as a host for expression of FVII. Plasmid containing cDNA encoding full-length human FVII was introduced into Lizard Leishmania and positive transfectants were analyzed by SDS-PAGE and Western blot analysis. Furthermore, biological activity of purified protein was detected by PT assay. The recombinant strain harboring a construct was analyzed for expression of FVII at the mRNA and protein level. Purified rFVII was obtained and in order to confirm the purified compound was in fact rFVII. Western blot analysis was carried out. Clotting time in PT assay was reduced about 30 seconds with the purified rFVII. In Conclusion, this study has demonstrated, for the first time, that Leishmania cells can be used as an expression system for producing recombinant FVII.


Subject(s)
Factor VII/biosynthesis , Leishmania/metabolism , Protein Engineering/methods , Recombinant Proteins/biosynthesis , Animals , Blood Coagulation Tests , Blotting, Western , Cloning, Molecular , Electrophoresis, Polyacrylamide Gel , Factor VII/chemistry , Factor VII/genetics , Hep G2 Cells , Humans , Leishmania/cytology , Leishmania/genetics , Lizards/parasitology , Models, Molecular , Plasmids/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics
7.
J Exp Med ; 159(4): 1042-57, 1984 Apr 01.
Article in English | MEDLINE | ID: mdl-6368733

ABSTRACT

In the present study we demonstrate that human monocytes can be induced by the model stimulus, lipopolysaccharide (LPS), to produce and assemble on their surface functional Factor VII/VIIa. This protease was not induced in relatively purified monocytes alone following exposure to LPS; but was induced in the presence of Leu-3a positive helper/inducer T cells. The Factor VII/VIIa protease activity represented 35-40% of the potential initiating activity for the extrinsic coagulation pathway and was demonstrated using functional coagulation assays, as well as in amidolytic assays for the activation of Factor X. This activity of cell-bound Factor VII/VIIa appeared to involve a tight adduct of calcium. The identity of the Factor X-activating protease as Factor VII/VIIa was confirmed by the capacity of antibody specific for Factor VII/VIIa to neutralize the cell-bound protease. Further propagation of the extrinsic pathway following generation of Factor Xa required addition of exogenous Factor Va. These results expand the repertoire of proteases that have been identified with appropriately triggered cells of the monocyte/macrophage series, and suggest that initiation and propagation of the extrinsic coagulation protease network on induced monocytes involves not only expression of the initiating cofactor molecule, tissue factor, but also production of Factor VII and its organization into the molecular assembly. Thus, in the absence of exogenous Factor VII/VIIa a directly proteolytic effector cell can be generated. Further molecular assembly of the extrinsic pathway on the monocyte surface sequentially expands the proteolytic capacity of this response. The synthesis and assembly of the extrinsic activation complex by the monocyte and its derived progeny, the macrophage, provides a mechanism by which coagulation is initiated under T cell instruction at sites of immunologic responses.


Subject(s)
Factor VII/biosynthesis , Monocytes/enzymology , Peptide Hydrolases/blood , T-Lymphocytes/immunology , Antibodies/physiology , Blood Coagulation Tests , Calcium/blood , Cell Communication , Factor VII/immunology , Factor VII/physiology , Factor VIIa , Humans , In Vitro Techniques , Lipopolysaccharides/pharmacology , Lymphocyte Activation , Monocytes/immunology , T-Lymphocytes/classification
8.
Biotechnol Lett ; 32(6): 803-9, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20213530

ABSTRACT

Recombinant coagulation factor VII (FVII) is used as a potential therapeutic intervention in hemophilia patients who produce antibodies against the coagulation factors. Mammalian cell lines provide low levels of expression, however, the Spodoptera frugiperda Sf9 cell line and baculovirus expression system are powerful systems for high-level expression of recombinant proteins, but due to the lack of endogenous vitamin K-dependent carboxylase, expression of functional FVII using this system is impossible. In the present study, we report a simple but versatile method to overcome the defect for high-level expression of the functional recombinant coagulation FVII in Sf9 cells. This method involves simultaneous expression of both human gamma-carboxylase (hGC) and human FVII genes in the host. It may be possible to express other vitamin K-dependent coagulation factors using this method in the future.


Subject(s)
Baculoviridae/genetics , Factor VII/biosynthesis , Gene Expression , Genetic Vectors , Animals , Carbon-Carbon Ligases/biosynthesis , Carbon-Carbon Ligases/genetics , Cell Line , Factor VII/genetics , Humans , Molecular Sequence Data , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Sequence Analysis, DNA , Spodoptera
9.
Vox Sang ; 96(4): 309-15, 2009 May.
Article in English | MEDLINE | ID: mdl-19175565

ABSTRACT

BACKGROUND: Factor VII (FVII) is a plasma glycoprotein that participates in the coagulation process leading to generation of fibrin. It is converted to factor VIIa that plays an important role in the coagulation cascade. The aim of this study was isolating and cloning the genes of human factor VII and hepsin and subsequent co-transfection of the constructs to Chinese hamster ovary (CHO) cell line to obtain rFVIIa. METHODS: Factor VII and hepsin cDNAs were isolated from HepG2 cell line and cloned into pcDNA3.1 (+) vector. The constructs were co-transfected to CHO cell line. A cell line that permanently expressed recombinant factor VII (rFVII) and hepsin was established. The expression of rFVII was confirmed by reverse transcriptase-polymerase chain reaction, enzyme-linked immunosorbent assay and Western blot analysis. Biological activity of rFVII was evaluated by prothrombin time assay. RESULTS: The results showed that the genes of FVII and hepsin were successfully cloned and expressed. Stable CHO cells co-transfected with pcNDA3.1-FVII and pcNDA3.1-hepsin expressed FVII and hepsin mRNA, but there was no expression in the CHO cells transfected with insert free pcDNA3.1. FVIIa protein was secreted to medium of CHO cells co-transfected with pcNDA3.1-FVII and pcNDA3.1-hepsin. The expected band of rFVII was detected in Western blot analysis. A three- to fourfold decrease in clotting time was observed when human FVII-depleted plasma was used in combination with human thromboplastin in the presence of rFVII, confirming the biological activity of rFVII. CONCLUSION: As we are aware, this is the first report of establishing a cell line expressing FVIIa using genetic engineering methods.


Subject(s)
Factor VII/genetics , Factor VIIa/metabolism , Genetic Engineering/methods , Animals , Blotting, Western , CHO Cells , Cell Line, Tumor , Cloning, Molecular , Cricetinae , Cricetulus , DNA, Complementary/genetics , Enzyme-Linked Immunosorbent Assay , Factor VII/biosynthesis , Factor VII/metabolism , Humans , Prothrombin Time , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Serine Endopeptidases/biosynthesis , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Transfection/methods , Tumor Cells, Cultured
10.
J Clin Invest ; 87(1): 50-7, 1991 Jan.
Article in English | MEDLINE | ID: mdl-1670636

ABSTRACT

Initiation and regulation of localized selective proteolysis is an important effector property of cells of macrophage (Mo) lineage. Among such effector responses is the induced expression of tissue factor (TF) by cells of Mo lineage. In characterizing the regulation of the Mo responses that may influence the magnitude of the effector phase of the cellular immune response, we have identified a role for the cell surface adhesive receptor CD11b/CD18 (Mac-1, CR3) to amplify the induced TF response. Occupancy of CD11b/CD18 by MAb as surrogate ligands does not directly initiate a TF response. In contrast, after either T cell-derived cytokine or LPS as initial signals, engagement of CD11b/CD18 by MAb induces a two- to eight-fold functional enhancement of the TF response in murine and human Mo. This pathway of CD11b/CD18 enhancement of this Mo effector response was also confirmed with recognized ligands for CD11b/CD18 by exposure of Mo to immobilized fibrinogen. A quantitative increase of Mo surface expression of TF was validated by flow cytometry. We suggest that engagement of CD11b/CD18 by complementary ligands including adherence to extracellular matrix, and possibly in antigen-driven TH:Mo collaborative responses, results in the transduction of cellular signals that quantitatively enhance the expression of TF per se and thereby enhance the inflammatory component of Mo mediated response.


Subject(s)
Antigens, CD/analysis , Blood Coagulation Factors/biosynthesis , Macrophage-1 Antigen/physiology , Macrophages/immunology , Receptors, Leukocyte-Adhesion/physiology , Thromboplastin/biosynthesis , Animals , Blood Coagulation Factors/analysis , CD18 Antigens , Calcium/physiology , Factor VII/biosynthesis , Factor X/metabolism , Humans , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred Strains
11.
J Clin Invest ; 75(6): 2030-7, 1985 Jun.
Article in English | MEDLINE | ID: mdl-4008651

ABSTRACT

Both fibrin and tissue macrophages are prominent in the histopathology of chronic inflammatory pulmonary disease. We therefore examined the procoagulant activity of freshly lavaged human alveolar macrophages in vitro. Intact macrophages (5 X 10(5) cells) from 13 healthy volunteers promoted clotting of whole plasma in a mean of 65 s. Macrophage procoagulant activity was at least partially independent of exogenous Factor VII as judged by a mean clotting time of 99 s in Factor VII-deficient plasma and by neutralization of procoagulant activity by an antibody to Factor VII. Immunoprecipitation of extracts of macrophages metabolically labeled with [35S]methionine by Factor VII antibody and analyzed by sodium dodecyl sulfate-polyacrylamide gel electrophoresis revealed a labeled protein consistent in size with the known molecular weight of blood Factor VII, 48,000. The addition of 50 micrograms of unlabeled, purified Factor VII blocked recovery of the 48,000-mol wt protein. In addition, supernatants of cultured macrophages from six normal volunteers had Factor X-activating activity that was suppressed an average of 71% after culture in the presence of 50 microM coumadin or entirely by the Factor VII antibody indicating that Factor VII synthesized by the cell was biologically active. Endotoxin in vitro induced increases in cellular tissue factor but had no consistent effect on macrophage Factor VII activity. We also examined the tissue factor and Factor VII activities of freshly lavaged alveolar cells from nine subjects with clinical and/or histologic evidence of sarcoidosis. Four of the nine subjects expressed increased tissue factor and seven of nine had increased Factor VII activity over the normal range (P less than 0.01). We estimate the mean Factor VII associated with the cells of sarcoid patients to be 4.7 ng/10(6) cells (range 0.4-20) as compared to a mean of 0.74 ng/10(6) cells (range 0.2-2) for that of normal subjects. Along with previous data showing synthesis of plasminogen activator, these findings indicate that human alveolar macrophages normally synthesize and express measurable amounts of the initial enzymes of proteolytic reactions regulating both fibrin deposition and fibrin resorption. Abnormalities in Factor VII activity in a small group of patients with sarcoidosis raise the possibility that modulation of fibrin turnover by macrophages may contribute to the pathology of this and perhaps other interstitial lung diseases.


Subject(s)
Factor VII/biosynthesis , Lung Diseases/metabolism , Macrophages/metabolism , Pulmonary Alveoli/cytology , Sarcoidosis/metabolism , Blood Coagulation/drug effects , Cells, Cultured , Endotoxins/pharmacology , Factor X/metabolism , Humans , Macrophages/drug effects , Warfarin/pharmacology
12.
J Am Coll Cardiol ; 46(4): 707-13, 2005 Aug 16.
Article in English | MEDLINE | ID: mdl-16098440

ABSTRACT

OBJECTIVES: The purpose of this study was to test the hypothesis that activated monocytes with soluble plasma tissue factor (pTF) activate factors VII and X to generate thrombin. BACKGROUND: Despite heparin, thrombin is progressively generated during cardiac surgery with cardiopulmonary bypass (CPB), produces intravascular fibrin and fibrinolysis, and causes serious thromboembolic and nonsurgical bleeding complications. Thrombin is primarily produced in the surgical wound, but mechanisms are unclear. METHODS: In 13 patients, interactions of mononuclear cells, platelets, pTF, and pTF fractions to activate factors VII and X were evaluated in pre-bypass, perfusate, and pericardial wound blood before and during CPB. RESULTS: Monocytes are activated in wound, but not in pre-bypass or perfusate plasma (monocyte chemotactic protein-1 = 29.5 +/- 2.1 pmoles/l vs. 2.8 +/- 1.2 pmoles/l and 3.3 +/-1.4 pmoles/l, respectively). Wound pTF is substantially elevated compared to other locations (3.64 +/- 0.45 pmoles/l vs. 0.71 +/- 0.65 pmoles/l and 1.31 +/- 1.4 pmoles/l). Supernatant wound pTF contains 81.7% of TF antigen; wound microparticle pTF contains 18.3%. Wound monocytes and all C5a-stimulated monocytes (but not activated platelets) completely convert factor VII to factor VIIa with wound pTF. Activated monocytes more efficiently activate factor X with wound supernatant TF/factor VII(VIIa) complex than with wound microparticle TF/factor VII(fVIIa). The correlation coefficient (r) between wound thrombin generation (F1.2) and wound pTF concentration is 0.944 (p = 0.0004). CONCLUSIONS: During cardiac surgery with CPB, wound monocytes plus wound pTF or wound microparticle-free supernatant pTF preferentially accelerate activation of factor VII and factor X. This system represents a novel mechanism for thrombin generation via the TF coagulation pathway.


Subject(s)
Blood Coagulation/physiology , Cardiopulmonary Bypass/adverse effects , Factor VII/biosynthesis , Factor X/biosynthesis , Monocytes/physiology , Plasma/chemistry , Postoperative Hemorrhage/physiopathology , Thrombin/biosynthesis , Thromboplastin/analysis , Aged , Aged, 80 and over , Blood Coagulation Factors/physiology , Blotting, Western , Factor VII/analysis , Factor X/analysis , Female , Humans , Male , Middle Aged , Postoperative Care , Wound Healing
13.
Bioengineered ; 7(3): 189-97, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27116572

ABSTRACT

Signal peptides play an important role in directing and efficiently transporting secretory proteins to their proper locations in the endoplasmic reticulum of mammalian cells. The aim of this study was to enhance the expression of recombinant coagulation factor VII (rFVII) in CHO cells by optimizing the signal peptides and type of fed-batch culture medium used. Five sub-clones (O2, I3, H3, G2 and M3) with different signal peptide were selected by western blot (WB) analysis and used for suspension culture. We compared rFVII expression levels of 5 sub-clones and found that the highest rFVII expression level was obtained with the IgK signal peptide instead of Ori, the native signal peptide of rFVII. The high protein expression of rFVII with signal peptide IgK was mirrored by a high transcription level during suspension culture. After analyzing culture and feed media, the combination of M4 and F4 media yielded the highest rFVII expression of 20 mg/L during a 10-day suspension culture. After analyzing cell density and cell cycle, CHO cells feeding by F4 had a similar percentage of cells in G0/G1 and a higher cell density compared to F2 and F3. This may be the reason for high rFVII expression in M4+F4. In summary, rFVII expression was successfully enhanced by optimizing the signal peptide and fed-batch medium used in CHO suspension culture. Our data may be used to improve the production of other therapeutic proteins in fed-batch culture.


Subject(s)
Cloning, Molecular/methods , Factor VII/genetics , Immunoglobulins/genetics , Protein Sorting Signals/genetics , Transcription, Genetic , Animals , Batch Cell Culture Techniques , CHO Cells , Cell Count , Cell Cycle/genetics , Cell Survival , Cricetulus , Culture Media/chemistry , Factor VII/biosynthesis , Gene Expression Regulation , Immunoglobulins/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Replication Origin
14.
Arterioscler Thromb Vasc Biol ; 23(1): 136-41, 2003 Jan 01.
Article in English | MEDLINE | ID: mdl-12524237

ABSTRACT

OBJECTIVE: Coagulation is initiated by the interaction of tissue factor (TF) with plasma coagulation factors VII (FVII) and X (FX). TF is highly expressed in atherosclerotic lesions, but little is known about the synthesis of FX or FVII outside of the liver. Previous studies suggested that macrophages synthesize FVII. We therefore hypothesized that macrophages within atherosclerotic lesions may produce FVII, leading to partial activation of the coagulation cascade. METHODS AND RESULTS: Immunohistochemistry was performed using antibodies against FVII, FX, and TF on normal and atherosclerotic vessels. In atherosclerotic lesions, FVII immunostaining was colocalized with TF in macrophages and spindle-shaped smooth muscle cells. FVII mRNA was also detected in these cells using in situ hybridization, suggesting the local synthesis of FVII in atherosclerosis. Reverse transcriptase-polymerase chain reaction confirmed the presence of FVII mRNA in normal and atherosclerotic vessels as well as smooth muscle cells, fibroblasts, and keratinocytes in vitro. CONCLUSIONS: The localization of FVII synthesis outside the liver may be indicative of other cellular functions for this coagulation protein. The observed coexpression of TF and FVII may contribute to autocrine signaling via thrombin-independent mechanisms and may represent a novel mechanism contributing to growth in the setting of vascular disease.


Subject(s)
Aorta/metabolism , Arteriosclerosis/blood , Factor VII/biosynthesis , Liver/blood supply , Liver/metabolism , Animals , Aorta/chemistry , Aorta/pathology , Aortic Diseases/blood , Brachial Artery/chemistry , Carotid Arteries/chemistry , Carotid Arteries/metabolism , Carotid Arteries/pathology , Carotid Artery Diseases/blood , Factor VII/immunology , Factor X/immunology , Factor X/metabolism , Fibroblasts/chemistry , Fibroblasts/pathology , Humans , Immunohistochemistry , In Situ Hybridization , Keratinocytes/chemistry , Keratinocytes/pathology , Liver/pathology , Macrophages/chemistry , Macrophages/pathology , Muscle, Smooth, Vascular/chemistry , Muscle, Smooth, Vascular/pathology , Papio , RNA, Messenger/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Thromboplastin/immunology , Thromboplastin/metabolism
15.
J Leukoc Biol ; 44(3): 172-9, 1988 Sep.
Article in English | MEDLINE | ID: mdl-3411233

ABSTRACT

Murine exudate macrophages elicited by different stimuli and bone marrow-derived macrophages were studied for their capacity to synthesize factor VII and tissue factor in a basal state and on stimulation with endotoxin (LPS). Cells elicited by different stimuli varied in their production of both factors. Thioglycollate-elicited cells generally made more, but not significantly more, tissue factor in response to endotoxin than cells elicited with periodate or streptococci. Cells elicited with proteose-peptone, fetal calf serum (FCS), or LPS produced less or very little tissue factor. Thioglycollate-elicited cells and cells elicited with streptococci or proteose-peptone consistently made more factor VII than cells elicited with periodate, FCS, and LPS. Bone marrow-derived macrophages were responsive to LPS by the production of tissue factor by the fifth day of culture, and this rose to a maximum by day 10. The maximal production of factor VII occurred on day 5 of culture and declined with longer cultivation. Factor VII production was not enhanced by LPS, and prolonged cultivation in the presence of LPS turned off the synthesis of both tissue factor and factor VII. We conclude that exudate cells are heterogeneous in the production of coagulant factors and that the production of these factors varies with the maturity of the cells. In addition, the production of the tissue factor and the factor VII were not necessarily expressed in a coordinate fashion.


Subject(s)
Blood Coagulation Factors/biosynthesis , Macrophages/metabolism , Animals , Bone Marrow/metabolism , Cells, Cultured , Exudates and Transudates/metabolism , Factor VII/biosynthesis , Female , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C3H
16.
Cell Transplant ; 24(12): 2541-55, 2015.
Article in English | MEDLINE | ID: mdl-25622096

ABSTRACT

Hepatocyte transplantation is a promising alternative therapy for the treatment of hepatic failure, hepatocellular deficiency, and genetic metabolic disorders. Hypothermic preservation of isolated human hepatocytes is potentially a simple and convenient strategy to provide on-demand hepatocytes in sufficient quantity and of the quality required for biotherapy. In this study, first we assessed how cold storage in three clinically safe preservative solutions (UW, HTS-FRS, and IGL-1) affects the viability and in vitro functionality of human hepatocytes. Then we evaluated whether such cold-preserved human hepatocytes could engraft and repopulate damaged livers in a mouse model of liver failure. Human hepatocytes showed comparable viabilities after cold preservation in the three solutions. The ability of fresh and cold-stored hepatocytes to attach to a collagen substratum and to synthesize and secrete albumin, coagulation factor VII, and urea in the medium after 3 days in culture was also equally preserved. Cold-stored hepatocytes were then transplanted in the spleen of immunodeficient mice previously infected with adenoviruses containing a thymidine kinase construct and treated with a single dose of ganciclovir to induce liver injury. Engraftment and liver repopulation were monitored over time by measuring the blood level of human albumin and by assessing the expression of specific human hepatic mRNAs and proteins in the recipient livers by RT-PCR and immunohistochemistry, respectively. Our findings show that cold-stored human hepatocytes in IGL-1 and HTS-FRS preservative solutions can survive, engraft, and proliferate in a damaged mouse liver. These results demonstrate the usefulness of human hepatocyte hypothermic preservation for cell transplantation.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Cryopreservation/methods , Cryoprotective Agents/pharmacology , Hepatocytes/transplantation , Liver Diseases/therapy , Organ Preservation Solutions/pharmacology , Adult , Aged , Albumins/biosynthesis , Animals , Cell Proliferation , Cell Survival , Factor VII/biosynthesis , Female , Ganciclovir/adverse effects , Hepatocytes/cytology , Humans , Liver/cytology , Male , Mice , Mice, Inbred BALB C , Middle Aged , Serum Albumin/analysis , Spleen/cytology , Transplantation, Heterologous , Urea/metabolism
17.
Gene ; 139(2): 275-9, 1994 Feb 25.
Article in English | MEDLINE | ID: mdl-8112618

ABSTRACT

Recombinant human proteins are generally recovered in low yields from mammalian tissue culture following transfection with commercially available vectors. We have constructed a novel vector containing both the neomycin-resistance-encoding gene (neo) as a dominant selectable marker, and the dihydrofolate reductase-encoding gene (DHFR) to enable amplification of transfected DNA followed by stable expression in mammalian cell lines. Levels of 5 micrograms/ml of the coagulation proteins, factor VII (FVII) and factor XI (FXI), have been achieved in serum-free media. N-terminal sequencing of the purified proteins, and of their separated chains after proteolytic activation, demonstrated correct processing of the recombinant products. In addition, the ratios of clotting activity to antigen for each are close to unity, and the recombinant and plasma-derived proteins had identical mobilities upon electrophoresis in the presence of SDS. The vector described will be of use for the synthesis of recombinant proteins, both wild-type and variants produced by site-directed mutagenesis, especially where complex post-translational modification of the protein makes it essential to use mammalian cells.


Subject(s)
Factor VII/biosynthesis , Factor XI/biosynthesis , Genetic Vectors/genetics , Animals , CHO Cells/drug effects , Cricetinae , Culture Media, Serum-Free , Drug Resistance/genetics , Factor VII/genetics , Factor XI/genetics , Humans , Methotrexate/pharmacology , Molecular Sequence Data , Neomycin , Plasmids/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Tetrahydrofolate Dehydrogenase/genetics , Transcription, Genetic/genetics , Transfection/genetics
18.
J Thromb Haemost ; 1(10): 2179-85, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14521602

ABSTRACT

AIMS: The aim of this study was to investigate associations between coronary heart disease risk and polymorphisms in the coagulation factor (F)VII gene in participants of a large prospective study. METHODS: One thousand nine hundred and fifty-seven men were genotyped for four FVII polymorphisms, -670A-->C, -402G-->A, a 10 base pair insertion at -323 (0 > 10) in the promoter, and R353Q in the structural gene. Associations among genotypes and estimated haplotypes, plasma FVII levels, and coronary heart disease risk were evaluated, and the function of the promoter polymorphisms was assessed in reporter gene assays. RESULTS: The -670A-->C and -402G-->A polymorphisms were in complete allelic association. The haplotype containing -670C and -402A (frequency =0.23) was associated with significantly increased plasma FVII coagulant activity and increased risk of an initial coronary event, particularly acute myocardial infarction, which remained after correction for conventional risk factors. In contrast, the -323 insertion and Q353 alleles (frequency =0.11 and 0.10, respectively) were associated with decreased plasma FVII levels, but hazard ratios for coronary events in carriers of these alleles were not significantly different from unity. In transiently transfected hepatoma cells, increased basal expression of the reporter gene was directed by a promoter fragment with rare haplotype -670C/-630G/-402A rather than by a promoter fragment with common haplotype -670A/-630A/-402G; -402A was not responsible for this effect. CONCLUSIONS: The promoter haplotype, -670C/-630G/402A, was associated with significantly increased plasma FVII coagulant activity, risk of an initial coronary event, particularly acute myocardial infarction, and reporter gene expression.


Subject(s)
Coronary Disease/genetics , Factor VII/genetics , Alleles , Carcinoma, Hepatocellular/metabolism , Cells, Cultured , Exons , Factor VII/biosynthesis , Genes, Reporter , Genotype , Haplotypes , Humans , Introns , Male , Middle Aged , Myocardial Infarction/genetics , Polymorphism, Genetic , Promoter Regions, Genetic , Proportional Hazards Models , Risk , Transfection
19.
J Thromb Haemost ; 2(12): 2107-14, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15613014

ABSTRACT

BACKGROUND: Thrombin promotes angiogenesis and cell proliferation in cancer. Whether thrombin turnover influences cancer incidence is unknown. OBJECTIVES: To explore the relation between the status of the coagulant pathway and cancer incidence by population survey. METHODS: Of 4,009 middle-aged men clinically free of malignancy, 3052 (76.1%) were recruited. Measurements of hemostatic status were made annually for 4 years, and follow-up for morbidity and mortality was maintained thereafter. Persistent activation of the coagulant pathway was diagnosed when prothrombin fragment 1+2 and fibrinopeptide A concentrations exceeded the upper quartiles of the population distribution in two consecutive annual examinations. Cancer incidence rates in men developing persistent activation (taking the time of onset of activation as baseline) were compared with those in men remaining free of this condition. RESULTS: Persistent activation of the hemostatic pathway was a distinct entity found in 111 men [43 expected by chance alone (P <0.001)], and associated with activation throughout the coagulation pathway. Total mortality (/1000 person-years) was higher in those with persistent activation than in others (17.1 and 9.7, respectively, P=0.015), owing to a higher mortality from all cancers (11.3 and 5.1, respectively, P=0.01), due in turn largely to a higher mortality from cancers of the digestive tract (6.3 and 1.9, respectively, P=0.004). Trends were similar for non-fatal cancers. CONCLUSIONS: Persistent activation of the coagulant pathway plays a role in the preclinical phase of cancer and is associated with an increased incidence of clinical malignancy, especially of the digestive tract.


Subject(s)
Coagulants/metabolism , Digestive System Neoplasms/complications , Digestive System Neoplasms/epidemiology , Enzyme-Linked Immunosorbent Assay , Factor IX/biosynthesis , Factor VII/biosynthesis , Fibrinopeptide A/biosynthesis , Follow-Up Studies , Hemostasis , Humans , Male , Middle Aged , Prevalence , Prothrombin/biosynthesis , Thrombin/metabolism , Time Factors
20.
Thromb Haemost ; 71(6): 773-7, 1994 Jun.
Article in English | MEDLINE | ID: mdl-7974346

ABSTRACT

We found hereditary factor VII deficiency in a clinically asymptomatic family, and characterized their factor VII gene and the abnormal molecule using recombinant DNA techniques. The propositus was a 45-year-old woman who was noted to have a prolonged prothrombin time. The level of factor VII antigen of the patient was 25.9% of that of normal individuals and the level of factor VII activity was 28% and 24%, when tested using rabbit brain tissue factor and human placental tissue factor in a one-stage clotting assay, respectively. Two of her sisters had almost the same reduced levels of factor VII antigen and activity, and her parents who are first cousins, a son, a daughter and a niece had moderately reduced leves of both factor VII activity and antigen. To identify the mutation site, all the coding exons and exon-intron boundaries of the factor VII gene of the propositus were amplified using the polymerase chain reaction (PCR), then subcloned and sequenced. One missense mutation (G to A) was identified in exon VII of the gene resulting in an amino acid substitution of His(CAC) for Arg(247)(CGC) in the gene product. PCR using a mutagenic primer to introduce a new ApaL I site into the mutant allele of the patient's factor VII gene revealed that this allele was inherited in the affected individuals in the pedigree. Transient expression assays using BHK cells transfected with an expression vector containing the mutant factor VII cDNA suggested that this mutation leads to factor VII deficiency by impairing secretion of the mutated factor VII.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Factor VII Deficiency/genetics , Factor VII/genetics , Base Sequence , Binding Sites , Catalysis , Cell Line , Exons , Factor VII/biosynthesis , Female , Homozygote , Humans , Male , Middle Aged , Molecular Sequence Data , Pedigree , Point Mutation , Polymerase Chain Reaction , Protein Structure, Tertiary , Recombinant Proteins/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL