Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Haemophilia ; 21(1): 58-63, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25459309

RESUMO

Prophylaxis prevents joint and other bleeding episodes in patients with haemophilia A. Development of new factor concentrates with longer circulating half-lives may encourage patients to start, continue or resume prophylaxis. The aim of this study was to compare the pharmacodynamic effect of a PEGylated full-length recombinant factor VIII (rFVIII) concentrate with that of an unmodified rFVIII concentrate with respect to the duration of prophylactic efficacy in a murine model of haemophilic joint bleeding. Mice were pretreated with BAX 855 or unmodified rFVIII at specified times before right knee puncture to induce haemarthrosis; left knee joints served as controls. Joint bleeding was evaluated using a combination of visual and histological assessments. Administration of a single dose of unmodified rFVIII before joint puncture prevented haemarthrosis in mice up to 24 h, whereas pretreatment with BAX 855 protected the joint from bleeding up to 48 h. This pharmacodynamic study showed prolonged efficacy of BAX 855 compared to ADVATE in a haemophilia A mouse joint bleeding model. This finding supports the possibility of using BAX 855 to increase FVIII trough levels and/or extend the dosing interval in patients with haemophilia A on prophylaxis, which may potentially improve prophylactic efficacy and long-term adherence.


Assuntos
Fator VIII/administração & dosagem , Hemofilia A/tratamento farmacológico , Animais , Modelos Animais de Doenças , Hemorragia/prevenção & controle , Humanos , Camundongos , Proteínas Recombinantes/administração & dosagem
2.
Nat Med ; 2(11): 1250-4, 1996 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-8898755

RESUMO

Measles virus (MV) still incites one of the most contagious infections of humankind. Despite the development and use of an excellent live attenuated virus vaccine, over one million infants and children continue to die each year from measles. The main cause of morbidity and mortality is virus-induced immunosuppression of lymphocyte function, which allows secondary infections. Here we report an in vivo model for the study of MV-induced immunosuppression. Human peripheral blood leukocytes (PBLs) grafted onto mice with severe combined immunodeficiency disease (SCID mice) to create hu-PBLS-SCID mice produce human IgG that is suppressed by MV infection. Immunosuppression is dependent on the involvement of live virus and is dramatically more severe for PBLs obtained from newborns than PBLs from adults. Suppression of IgG synthesis by PBLs from newborns occurs as early as ten days after administration of MV to hu-PBLS-SCID mice compared with 44 days required for PBLs from adults. Further, MV infection of SCID mice reconstituted with PBLs from newborns.


Assuntos
Tolerância Imunológica/imunologia , Imunoglobulina G/imunologia , Leucócitos Mononucleares/imunologia , Vírus do Sarampo/imunologia , Adulto , Animais , Antígenos CD/imunologia , Sequência de Bases , Transplante de Células , Primers do DNA , Humanos , Recém-Nascido , Leucócitos Mononucleares/citologia , Vírus do Sarampo/genética , Proteína Cofatora de Membrana , Glicoproteínas de Membrana/imunologia , Camundongos , Camundongos SCID , Dados de Sequência Molecular , RNA Viral/análise
3.
J Thromb Haemost ; 2018 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-29888855

RESUMO

Essentials Patients with hemophilia A and inhibitors receiving emicizumab experience breakthrough bleeding. Safety concerns may exist when combining emicizumab with bypassing agents. Combined bypassing agent and bispecific antibody increased thrombin generation up to 17-fold. Thrombotic effects should be considered when combining emicizumab with plasma bypassing agent. SUMMARY: Background Investigational non-factor products such as emicizumab offer a treatment option for patients with hemophilia and inhibitors. However, their mechanism of action raises questions regarding safety when they are combined with treatments for breakthrough bleeding. Objectives To evaluate in vitro thrombin generation (TG) and clot formation for combinations of activated prothrombin complex concentrate (aPCC), recombinant activated factor VII (rFVIIa), and a sequence-identical analog of emicizumab (SIA). Methods Therapeutic concentrations of SIA (20-600 nm) alone or with aPCC (0.05-1 U mL-1 ), isolated aPCC components or rFVIIa (0.88-5.25 µg mL-1 ) were tested for TG and compared with reference ranges for healthy donor plasma. Coagulation of FVIII-inhibited blood was determined with a widely established method, i.e. rotational thromboelastometry (ROTEM), and confirmed with the Total Thrombus-formation Analysis System. Results and conclusions SIA (600 nm) or aPCC (0.5 U mL-1 ) alone resulted in peak thrombin levels of 21.4 nm and 38.6 nm, respectively, both of which are lower than normal (83.7 ± 29.8 nm). SIA plus aPCC (0.5 U mL-1 ) increased the peak thrombin level 17-fold over SIA alone, exceeding the reference plasma value by 4.2-fold. This hypercoagulable effect occurred with 600 nmSIA combined with as little as 0.25 U mL-1 aPCC, confirmed by ROTEM. FIX was the main driver for enhanced TG. SIA plus rFVIIa (1.75 µg mL-1 ) induced a 1.8-fold increase in the peak thrombin level in platelet-rich plasma, but it did not reach the normal range. These in vitro experiments demonstrate excessive TG after administration of a combination of aPCC and SIA at clinically relevant doses. Careful judgement may be required when breakthrough bleeding is treated in patients receiving emicizumab.

4.
J Thromb Haemost ; 16(6): 1176-1181, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29665242

RESUMO

Essentials Aggregation is a critical quality attribute of protein therapeutics influencing immunogenicity. Aggregates and subvisible particles in 9 recombinant factor VIII (rFVIII) products were analyzed. Major differences in aggregate and particle concentrations were detected after reconstitution. rFVIII product quality determined aggregation propensity under use-relevant stress. SUMMARY: Background Recombinant protein technologies have facilitated the development of novel factor VIII (FVIII) therapeutics with improved production efficiency, potency and half-live, and a low risk of viral transmission. The increasing number of recombinant FVIII (rFVIII) products and information on their efficacy, safety and cost allow patients and healthcare professionals to adjust treatment to individual needs. Nonetheless, 20-32% of previously untreated patients with severe hemophilia A develop inhibitory antibodies to rFVIII following treatment. The root cause of the immunogenicity of rFVIII products is not well understood. Data for human interferon and human insulin products suggest that critical quality parameters such as soluble protein aggregates (SPAs) and subvisible particles (SVPs) influence the immunogenicity of protein therapeutics. Therefore, we analyzed SPA and SVP concentrations in commercially available rFVIII products and determined how these parameters change upon exposure of rFVIII products to relevant stress conditions. Objectives Compare critical quality parameters such as SPA and SVP concentrations in rFVIII products under intended use and use-relevant stress conditions. Methods Nine rFVIII products (≥ 3 lots each) were analyzed by high-performance liquid chromatography-size exclusion chromatography (HPLC-SEC) and flow cytometry-based particle analysis. Results/conclusions SPAs and SVPs were present at different concentrations in all freshly reconstituted rFVIII products: SPA concentrations ranged from 0.2% to 11.6%; SVPs were 0.7 × 106 to 114.0 × 106 / 1000 IU. Under use-relevant stress conditions (agitation and shear stress) the products formed additional SPAs and SVPs to different degrees. The collected data indicate that product quality determines its propensity to form SVPs and SPAs, and highlights differences between marketed rFVIII products.


Assuntos
Fator VIII/química , Hemostáticos/química , Agregados Proteicos , Cromatografia em Gel , Cromatografia Líquida de Alta Pressão , Citometria de Fluxo , Tamanho da Partícula , Proteínas Recombinantes/química , Solubilidade , Estresse Mecânico
5.
J Thromb Haemost ; 2018 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-29888865

RESUMO

Essentials Glycosylation heterogeneity of recombinant proteins affects pharmacokinetics and immunogenicity. N-glycomics/glycoproteomics of plasma-derived Factor VIII and 6 recombinant FVIIIs were compared. Depending on cell line, significant differences to plasma-derived FVIII were observed. Recombinant FVIIIs expressed distinct and immunologically relevant epitopes. SUMMARY: Background/Objective Human factor VIII (FVIII) is a plasma glycoprotein, defects of which result in hemophilia A. Current substitution therapy uses FVIII products purified from human plasma or from various cell lines (recombinant FVIII) with different levels of B-domain deletion. Glycosylation is a post-translational protein modification in FVIII that has a substantial influence on its physical, functional and antigenic properties. Variation in glycosylation is likely to be the reason that FVIII products differ in their pharmacokinetics, pharmacodynamics and immunogenicity. However, the literature on FVIII glycosylation is inconsistent, preventing assembly into a coherent model. Seeking to better understand the glycosylation mechanisms underlying FVIII biology, we studied the N-glycosylation of human plasma-derived (pd)FVIII and six rFVIII products expressed in CHO, BHK or HEK cell lines. Methods FVIII samples were subjected to head-to-head detailed glycomic and glycoproteomic characterization using a combination of MALDI-MS and MS/MS, GC-MS and UPLC-UV-MSE technologies. Results/Conclusion The results of our study detail the N-glycan repertoire of pdFVIII to an unprecedented level, and for the first time, provide evidence of N-glycolylneuraminic acid (NeuGc) found on pdFVIII. Although site-specific glycosylation of rFVIII proved consistent with pdFVIII regardless of the expression system, the entire N-glycan content of each sample appeared significantly different. Although the proportion of biologically important epitopes common to all samples (i.e. sialylation and high-mannose) varied between samples, some recombinant products expressed distinct and immunologically relevant epitopes, such as LacdiNAc (LDN), fucosylated LacdiNAc (FucLDN), NeuGc, LewisX/Y and Galα1,3 Gal epitopes. rFVIII expressed in HEK cells showed the greatest glycomic differences to human pdFVIII.

6.
J Thromb Haemost ; 4(5): 1063-70, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16689760

RESUMO

INTRODUCTION: ADAMTS-13 is a member of A Disintegrin And Metalloprotease with ThromboSpondin type 1 repeats (ADAMTS) family, primarily synthesized in hepatic stellate cells (HSCs), one of the major cell types transdifferentiating into myofibroblasts during liver fibrosis. However, the association between ADAMTS-13 expression and HSC activation or liver fibrosis is not known. METHODS: In this study, we determined the ADAMTS-13 mRNA, protein, and activity in isolated primary HSCs upon activation on a plastic dish and in liver after administration of carbon tetrachloride (CCl(4)) in rats. RESULTS: We showed that ADAMTS-13 antigen and proteolytic activity in the activated rat HSCs were dramatically increased, whereas ADAMTS-13 mRNA in these cells was only minimally altered. Similarly, the ADAMTS-13 antigen and proteolytic activity in rat liver after CCl(4) injury were also significantly increased, whereas the ADAMTS-13 mRNAs in these liver tissues were only slightly increased compared with normal. Surprisingly, despite the dramatic up-regulation of ADAMTS-13 protein synthesis in the activated HSCs after CCl(4) administration, the plasma levels of ADAMTS-13 protease in rats did not increase concordantly. CONCLUSION: We conclude that the up-regulation of ADAMTS-13 protein expression in rat HSCs during activation in vitro and in vivo suggests the possibility of ADAMTS-13 proteolysis, an important part of function of the activated HSCs, perhaps through modulation of liver regeneration or formation of liver fibrosis after various injuries. The data also suggest the minimal contribution of the activated HSCs in regulation of plasma levels of ADAMTS-13 protease.


Assuntos
Proteínas ADAM/metabolismo , Fígado/metabolismo , Proteína ADAMTS13 , Animais , Western Blotting , Células Cultivadas , Hidrólise , Imuno-Histoquímica , Fígado/citologia , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Regulação para Cima
7.
J Thromb Haemost ; 14(7): 1410-9, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27371116

RESUMO

UNLABELLED: Essentials ADAMTS-13-deficiency is a cause of thrombotic thrombocytopenic purpura (TTP). Preclinical safety of recombinant human ADAMTS-13 (BAX930) was shown in animal models. Preclinical efficacy of BAX930 was shown in a mouse model of TTP. BAX930 showed advantageous efficacy over fresh frozen plasma, the current standard of care. Click to hear Dr Cataland and Prof. Lämmle present a seminar on Thrombotic Thrombocytopenic Purpura (TTP): new Insights in Pathogenesis and Treatment Modalities. SUMMARY: Background Thrombotic thrombocytopenic purpura (TTP) is a rare blood disorder characterized by microthrombosis in small blood vessels of the body, resulting in a low platelet count. Baxalta has developed a new recombinant ADAMTS-13 (rADAMTS-13) product (BAX930) for on-demand and prophylactic treatment of patients with hereditary TTP (hTTP). Objectives To evaluate the pharmacokinetics, efficacy and safety of BAX930 in different species, by use of an extensive preclinical program. Methods The prophylactic and therapeutic efficacies of BAX930 were tested in a previously established TTP mouse model. Pharmacokinetics were evaluated after single intravenous bolus injection in mice and rats, and after repeated dosing in cynomolgus monkeys. Toxicity was assessed in rats and monkeys, safety pharmacology in monkeys, and local tolerance in rabbits. Results BAX930 was shown to be efficacious, as demonstrated by a stabilized platelet count in ADAMTS-13 knockout mice that were thrombocytopenic when treated. Prophylactic efficacy was dose-dependent and comparable with that achieved by treatment with fresh frozen plasma, the mainstay of hTTP treatment. Therapeutic efficacy was treatment interval-dependent. Safety pharmacology evaluation did not show any deleterious effects of BAX930 on cardiovascular and respiratory functions in monkeys. The compound's pharmacokinetics were similar and dose-proportional in mice, rats, and monkeys. BAX930 was well tolerated in rats, monkeys, and rabbits, even at the highest doses tested. Conclusions These results demonstrate that BAX930 has a favorable preclinical profile, and support the clinical development of rADAMTS-13 for the treatment of hTTP.


Assuntos
Proteína ADAMTS13/farmacologia , Púrpura Trombocitopênica Trombótica/tratamento farmacológico , Proteína ADAMTS13/genética , Animais , Área Sob a Curva , Plaquetas/efeitos dos fármacos , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Feminino , Humanos , Macaca fascicularis , Masculino , Camundongos , Plasma/metabolismo , Contagem de Plaquetas , Púrpura Trombocitopênica Trombótica/sangue , Coelhos , Ratos , Proteínas Recombinantes/farmacologia , Especificidade da Espécie , Trombose/sangue , Resultado do Tratamento
8.
J Thromb Haemost ; 3(5): 1064-73, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15869605

RESUMO

Functional deficiency or absence of the human von Willebrand factor (VWF)-cleaving protease (VWF-cp), recently termed ADAMTS13, has been shown to cause acquired and congenital thrombotic thrombocytopenic purpura (TTP), respectively. As a first step towards developing a small animal model of TTP, we have cloned the complete (non-truncated) murine Adamts13 gene from BALB/c mice liver poly A+ mRNA. Murine ADAMTS13 is a 1426-amino-acid protein with a high homology and similar structural organization to the human ortholog. Transient expression of the murine Adamts13 cDNA in HEK 293 cells yielded a protein with a molecular weight of approximately 180 kDa which degraded recombinant murine VWF (rVWF) in a dose-dependent manner. The cleavage products of murine rVWF had the expected size of 140 and 170 kDa. Murine ADAMTS13 was inhibited by EDTA and the plasma from a TTP patient.


Assuntos
Metaloendopeptidases/genética , Metaloendopeptidases/metabolismo , Proteínas ADAM , Proteína ADAMTS13 , Sequência de Aminoácidos , Aminoácidos/química , Animais , Northern Blotting , Western Blotting , Linhagem Celular , Clonagem Molecular , Primers do DNA/química , DNA Complementar/metabolismo , Bases de Dados Genéticas , Relação Dose-Resposta a Droga , Ácido Edético/farmacologia , Humanos , Metaloendopeptidases/química , Camundongos , Camundongos Endogâmicos BALB C , Modelos Genéticos , Dados de Sequência Molecular , Filogenia , Poli A/metabolismo , Reação em Cadeia da Polimerase , Ligação Proteica , Estrutura Terciária de Proteína , Púrpura Trombocitopênica Trombótica/sangue , RNA/metabolismo , RNA Mensageiro/metabolismo , Proteínas Recombinantes/química , Homologia de Sequência de Aminoácidos , Distribuição Tecidual , Transfecção , Fator de von Willebrand/metabolismo
9.
J Thromb Haemost ; 13(11): 2053-62, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26340698

RESUMO

BACKGROUND: Acquired thrombotic thrombocytopenic purpura (TTP) is caused by an autoantibody-mediated deficiency of the von Willebrand factor-cleaving protease ADAMTS-13. Acute episodes of the disease are treated with a combination of immunosuppression and repeated cycles of plasma exchange to remove anti-ADAMTS-13 autoantibodies and, at the same time, replenish functional ADAMTS-13. Although this is often effective, the mortality rate has remained between 10% and 20%, highlighting the need for safer treatment options. OBJECTIVES: We previously showed that, in vitro, human recombinant ADAMTS-13 (rADAMTS-13) is able to override neutralizing antibodies and restore ADAMTS-13 activity in plasma from patients with acquired TTP. In the present study, we assessed the in vivo feasibility of this strategy by using a rat model. METHODS: Wild-type rats were adjusted to an ADAMTS-13 inhibitor (inhibitor) titer of ~ 10 BU mL(-1) with goat anti-ADAMTS-13 IgG, and treated with increasing doses of rADAMTS-13. Blood samples were drawn and analyzed for ADAMTS-13-specific parameters, including FRETS-VWF73 activity, inhibitor, and ADAMTS-13-specific immune complexes (ICs). The pharmacokinetics of ADAMTS-13 activity and inhibitors were evaluated. RESULTS: Administration of inhibitor titer-adjusted doses of rADAMTS-13 to inhibitor-treated rats predictably restored activity. Inhibitors were readily neutralized through formation of ADAMTS-13-specific ICs, which were cleared at a higher rate than the free inhibitor. Surplus protease was enzymatically active in plasma, and showed similar pharmacokinetics to ADAMTS-13 in not inhibitor-treated rats. CONCLUSIONS: Defined doses of rADAMTS-13 neutralized circulating anti-ADAMTS-13 antibodies and enabled reconstitution of ADAMTS-13 activity in plasma in our model, indicating that the protease may be a promising candidate for further exploration in treating acute episodes of acquired TTP.


Assuntos
Proteínas ADAM/uso terapêutico , Anticorpos Neutralizantes/sangue , Autoanticorpos/sangue , Púrpura Trombocitopênica Trombótica/imunologia , Proteínas ADAM/sangue , Proteínas ADAM/deficiência , Proteínas ADAM/imunologia , Proteína ADAMTS13 , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/toxicidade , Complexo Antígeno-Anticorpo/sangue , Autoanticorpos/imunologia , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Cabras/imunologia , Humanos , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Imunoglobulina G/toxicidade , Masculino , Processamento de Proteína Pós-Traducional , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/uso terapêutico , Fator de von Willebrand/metabolismo
10.
J Thromb Haemost ; 13(1): 92-100, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25348176

RESUMO

BACKGROUND: Tissue factor pathway inhibitor-α (TFPIα) inhibits factor Xa by forming a binary TFPI-FXa complex in a reaction that is stimulated by protein S. TF-FVIIa forms a quaternary complex with TFPIα and FXa, which shuts off the initiation of coagulation via the extrinsic pathway. AIM: To investigate whether direct inhibition of FXa by TFPIα independently of TF plays a role in downregulating coagulation. METHODS: Inhibition of FXa by TFPIα in plasma was determined by measuring thrombin generation triggered with FXa, the FX activator from Russell's viper venom (RVV-X), FXIa, or FIXa. TF-independent anticoagulant activities of TFPIα and its cofactor, protein S, were quantified: (i) after neutralization of TFPIα and protein S with anti-TFPI or anti-protein S antibodies; and (ii) in TFPI-depleted or protein S-depleted plasmas supplemented with varying amounts of TFPIα or protein S. RESULTS: Both anti-TFPI and anti-protein S antibodies enhanced thrombin generation in plasma triggered with RVV-X, FXa, FIXa, or FXIa. Anti-TFPI and anti-protein S antibodies decreased the lag time and increased the peak height of thrombin generation to the same extent, indicating that inhibition of FXa by TFPIα requires the presence of protein S. TFPIα and protein S titrations in TFPI-depleted or protein S-depleted plasma in which thrombin formation was initiated with triggers other than TF also revealed TF-independent anticoagulant activity of TFPIα, which was completely dependent on the presence of protein S. CONCLUSION: Direct inhibition of FXa by TFPIα contributes to the downregulation of coagulation.


Assuntos
Coagulação Sanguínea , Lipoproteínas/metabolismo , Trombina/metabolismo , Tromboplastina/metabolismo , Testes de Coagulação Sanguínea , Regulação para Baixo , Fator IXa/metabolismo , Fator Xa/metabolismo , Humanos , Cinética , Proteína S/metabolismo
11.
Gene ; 109(2): 307-8, 1991 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-1765277

RESUMO

A new plasmid pSC-O for generation of recombinant vaccinia virus was constructed. It offers additional advantages when compared with other widely used insertion vectors. This plasmid allows the expression of coding sequences lacking codons for the initiation as well as for termination of translation. Additional sequences modulating translation, but also transcription or affecting intracellular processing can be introduced. Sequences flanking the transcription unit of the gene of interest are complementary to SP6/T7 sequencing primers and thus may allow rapid sequencing (amplification) of the inserted DNA.


Assuntos
DNA Recombinante/genética , Vetores Genéticos/genética , Plasmídeos/genética , Vaccinia virus/genética , Sequência de Bases , Clonagem Molecular , Códon/genética , Regulação Viral da Expressão Gênica/genética , Dados de Sequência Molecular , Recombinação Genética/genética
12.
Gene ; 177(1-2): 43-6, 1996 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-8921843

RESUMO

The 'Modified Vaccinia Ankara' (MVA) strain is a potential live vaccine vector. The use of the hemagglutinin (ha) gene of the MVA strain as an insertion site for foreign genes was evaluated. To identify the molecular basis of the hemagglutinin-negative (HA-) phenotype of MVA, the ha gene and the region around this gene were sequenced. Amino acid (aa) sequence comparisons with functional hemagglutinins of other vaccinia strains predicted a functional polypeptide. The late part of the promoter region of the ha gene, however, was deleted, causing the apparent loss of the ha gene function. Nevertheless, insertion of foreign DNA into the ha gene allowed generation of functional recombinant viruses, indicating that the ha-gene region is a suitable insertion site.


Assuntos
Vetores Genéticos , Hemaglutininas Virais/genética , Vaccinia virus/genética , Sequência de Bases , DNA Viral , Estudos de Avaliação como Assunto , Genes Virais , Dados de Sequência Molecular , Deleção de Sequência
13.
Thromb Haemost ; 68(2): 119-24, 1992 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-1412155

RESUMO

We have worked out an efficient and time saving procedure for the expression of recombinant human prothrombin. The glycoprotein was expressed in the vaccinia virus expression system in several mammalian cell lines. The kidney cell lines Vero and BHK and the human cell line Hela were found to efficiently secrete prothrombin. Expression level of 3-4 micrograms of factor II per 10(6) cells per day corresponding to 18-23 mU per 10(6) cells per day were achieved. Since the expression levels obtained with the vaccinia virus/Vero cell system were comparable to those obtained in amplified transformed CHO cells it provides an alternative system for the efficient expression of human prothrombin and may allow to further elucidate structure-function relationships of (pro)thrombin and its various effectors.


Assuntos
Protrombina/genética , Vaccinia virus/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Linhagem Celular , Clonagem Molecular , DNA/genética , Expressão Gênica , Humanos , Dados de Sequência Molecular , Plasmídeos , Protrombina/biossíntese , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Vaccinia virus/metabolismo
14.
Wien Klin Wochenschr ; 111(5): 172-80, 1999 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-10226347

RESUMO

Hemophilic care was revolutionized in the late 50's, when factor VIII (FVIII) substitution therapy became accessible. Since then, plasma-derived FVIII preparations with varying degrees of purity have been widely used. The availability of such products has resulted in a great improvement of life quality and life expectancy of affected people. However, FVIII concentrates have been involved in the transmission of blood borne viruses like HIV and HCV. The tremendous development of molecular biology techniques, in association with modern biotechnology has allowed the large-scale production of first generation biopharmaceuticals such as recombinant FVIII, for clinical purposes. In parallel, intense research has revealed the genetic basis of hemophilia A, many aspects of FVIII biosynthesis and the biological function of FVIII. Synergistic efforts between genetic engineering and biotechnology have the potential to develop second generation products and therapeutic strategies based solely on "protein engineering". Integration of basic research results in new treatment concepts should allow the rational design of FVIII molecules endowed with improved functional properties. Such molecules may be highly beneficial in the development of future strategies in FVIII inhibitor therapy and in FVIII somatic gene therapy.


Assuntos
Hemofilia A/genética , Hemofilia A/metabolismo , Terapêutica/tendências , Animais , Fator VIII/biossíntese , Fator VIII/genética , Hemofilia A/sangue , Hemofilia A/terapia , Humanos
16.
Thromb Haemost ; 111(3): 429-37, 2014 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-24285223

RESUMO

Fucoidan is a highly complex sulfated polysaccharide commonly extracted from brown seaweed. In addition to their many biological activities, fucoidans have recently been demonstrated to inhibit or increase coagulation at different concentration ranges. Their structural features, i.e. molecular weight (Mw), Mw distribution, degree of sulfation, monosaccharide composition, and different linkages, are known to affect these activities. Therefore, structure-activity relationship (SAR) analysis of fucoidan is crucial for its potential use as a procoagulant. In this study, Fucus vesiculosus (F.v.) fucoidan was fractionated by charge and size as well as over- and desulfated to different degrees to yield preparations with various structural properties. The fractions' pro- and anticoagulant activities were assessed by calibrated automated thrombography (CAT) and activated partial thromboplastin time(aPTT) assays. Binding to and inhibition of the anticoagulant protein tissue factor pathway inhibitor (TFPI) and the ability to activate coagulation via the contact pathway were also investigated. This paper discusses the impact of charge density, size, and sugar composition on fucoidan's pro- and anticoagulant activities. Fucoidan requires a minimal charge density of 0.5 sulfates per sugar unit and a size of 70 sugar units to demonstrate desired procoagulant activities for improvement of haemostasis in factor VIII/factor IX-deficient plasma.


Assuntos
Anticoagulantes/uso terapêutico , Coagulantes/uso terapêutico , Hemofilia A/terapia , Hemofilia B/terapia , Polissacarídeos/uso terapêutico , Fracionamento Químico , Fator IX/genética , Fator VIII/genética , Fucus , Hemofilia A/genética , Hemofilia B/genética , Hemostasia , Humanos , Lipoproteínas/metabolismo , Estrutura Molecular , Peso Molecular , Tempo de Tromboplastina Parcial , Extratos Vegetais/química , Polissacarídeos/química , Ligação Proteica , Relação Estrutura-Atividade , Ésteres do Ácido Sulfúrico/química
17.
J Thromb Haemost ; 12(9): 1523-32, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25040659

RESUMO

BACKGROUND: Several static Bethesda-type assays are routinely used to determine ADAMTS-13-neutralizing autoantibodies in acquired thrombotic thrombocytopenic purpura (TTP), but the inhibitory activity of these antibodies has not been thoroughly evaluated under the more physiologic condition of flow. OBJECTIVES: We investigated whether ADAMTS-13 inhibitor assessment with the FRETS-VWF73 assay is predictive for evaluation under flow. METHODS: Anti-ADAMTS-13 autoantibodies were purified from patients with acquired TTP by chromatography involving an ADAMTS-13 affinity matrix and/or protein G. ADAMTS-13 activity was measured with the FRETS-VWF73 assay and a novel flow assay determining the ADAMTS-13-mediated decrease in platelet aggregate surface coverage, caused by perfusion of a suspension containing platelets, erythrocytes and von Willebrand factor (VWF) over a surface coated with extracellular matrix components. The neutralizing activities of ADAMTS-13 inhibitors were compared under static conditions and under flow by use of the two assays. RESULTS: The suitability of the flow-based ADAMTS-13 activity assay for quantification of ADAMTS-13 inhibitors could be demonstrated by reversibility of the ADAMTS-13-dependent decrease in surface coverage upon addition of goat ADAMTS-13 antiserum. Testing the neutralizing activity of purified autoantibodies from six patients in the flow assay according to their FRETS-VWF73-based inhibitor titers gave rise to vastly different inhibitory effects, indicating a discrepancy in inhibitor assessment between static and flow conditions. CONCLUSIONS: Anti-ADAMTS-13 autoantibodies may show inhibitory properties in vivo that are not consistent with the ADAMTS-13 inhibitor levels determined in routine static assays, possibly because certain epitopes are selectively exposed under shear. Consequently, the course of disease and treatment efficacy may vary among TTP patients, despite common inhibitor titers.


Assuntos
Proteínas ADAM/química , Proteínas ADAM/imunologia , Autoanticorpos/química , Testes de Coagulação Sanguínea/instrumentação , Testes Hematológicos/métodos , Púrpura Trombocitopênica Trombótica/sangue , Proteína ADAMTS13 , Testes de Coagulação Sanguínea/métodos , Proteínas do Citoesqueleto/química , Transferência Ressonante de Energia de Fluorescência , Humanos , Imunoglobulina G/química , Proteínas com Domínio LIM/química , Agregação Plaquetária , Ligação Proteica , Púrpura Trombocitopênica Trombótica/diagnóstico , Proteínas de Ligação a RNA , Resistência ao Cisalhamento , Estresse Mecânico , Fator de von Willebrand/química
18.
J Thromb Haemost ; 12(11): 1826-37, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25163770

RESUMO

BACKGROUND: TFPI is a Kunitz-type protease inhibitor that downregulates the extrinsic coagulation pathway by inhibiting factor Xa (FXa) and FVIIa. All three Kunitz domains (KD1, KD2, and KD3) and protein S are required for optimal inhibition of FXa and FVIIa. There is limited information on Kunitz domain requirements of the inhibition of TF:FVIIa-catalyzed FIX and FX activation by TFPI. AIM: To investigate the role of the Kunitz domains of TFPI and protein S in the inhibition of FX and FIX activation. METHODS: Inhibition of TF:FVIIa-catalyzed FX and FIX activation by full-length TFPI (TFPIFL ) and TFPI constructs was quantified from progress curves of FXa and FIXa generation measured with chromogenic substrates. RESULTS AND CONCLUSIONS: TFPIFL inhibited TF:FVIIa-catalyzed FIX activation with a Ki of 16.7 nmol L(-1) . Protein S reduced the Ki to 1.0 nmol L(-1) . TFPI1-150 and KD1-KD2 had 10-fold higher Ki values and were not stimulated by protein S. Single Kunitz domains were poor inhibitors of TF:FVIIa-catalyzed FIX activation (Ki >800 nm). FX activation was measured at limiting FVIIa and excess TF or vice versa. At both conditions, TFPIFL , TFPI1-150 , and KD1-KD2 showed similar inhibition of FX activation. However, at low phospholipid concentrations, TFPIFL was ~ 15-fold more active than TFPI1-150 or KD1-KD2. Apparently, excess phospholipids act as a kind of sink for TFPIFL , limiting its availability for TF:FVIIa inhibition. Preformed FXa:TFPIFL/1-150 complexes rapidly and stoichiometrically inhibited FIX and FX activation by TF:FVIIa, indicating that binary TFPI:FXa complex formation is the limiting step in TF:FVIIa inhibition. Protein S also enhanced inhibition of TF:FVIIa-catalyzed FX activation by TFPI.


Assuntos
Coagulação Sanguínea , Fator IXa/metabolismo , Fator VIIa/metabolismo , Inibidores do Fator Xa/metabolismo , Fator Xa/metabolismo , Lipoproteínas/metabolismo , Tromboplastina/metabolismo , Coagulação Sanguínea/efeitos dos fármacos , Catálise , Relação Dose-Resposta a Droga , Fator VIIa/antagonistas & inibidores , Inibidores do Fator Xa/farmacologia , Humanos , Cinética , Lipoproteínas/farmacologia , Fosfolipídeos/metabolismo , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Proteína S/metabolismo , Proteínas Recombinantes/metabolismo
19.
J Thromb Haemost ; 11(6): 1137-45, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23528042

RESUMO

BACKGROUND: Tissue factor pathway inhibitor (TFPI) is a multidomain protein that negatively regulates the coagulation cascade. TFPI inhibits the tissue factor (TF)-activated factor VII-activated FX (FXa) complex during TF-mediated coagulation initiation. The aptamer BAX 499 binds specifically to TFPI and inhibits its function, mediating a procoagulant effect in both in vitro and in vivo models of hemophilia. OBJECTIVES: This study sought to identify the regions of TFPI that are critical for BAX 499 binding, and to determine how binding mediates aptamer inhibition of TFPI. METHODS AND RESULTS: In vitro biochemical methods were used to evaluate the BAX 499 interaction with and inhibition of TFPI. Binding experiments indicated that the full-length TFPI protein is required for tight aptamer binding. Binding-competition experiments implicated the Kunitz 1, Kunitz 3 and C-terminal domains of TFPI in aptamer binding, a finding that is supported by hydrogen-deuterium exchange experiments, and indicated that aptamer and FXa can bind simultaneously to TFPI. In enzymatic assays, BAX 499 inhibited TFPI in a manner that is distinct from domain-specific antibodies, and aptamer inhibitory activity is reduced in the presence of the TFPI cofactor protein S. CONCLUSIONS: These studies demonstrate that BAX 499 binds to TFPI via multiple domains of the protein in a manner that is distinct from other TFPI inhibitors, mediating a mechanism of inhibition that does not involve direct competition with FXa. With this unique inhibitory mechanism, BAX 499 provides a useful tool for studying TFPI biology in health and disease.


Assuntos
Aptâmeros de Nucleotídeos/química , Lipoproteínas/antagonistas & inibidores , Lipoproteínas/química , Tromboplastina/química , Anticorpos/química , Coagulação Sanguínea/efeitos dos fármacos , Coagulantes/química , Medição da Troca de Deutério , Ensaio de Imunoadsorção Enzimática , Fator Xa/química , Hemofilia A/tratamento farmacológico , Humanos , Hidrogênio/química , Concentração Inibidora 50 , Peptídeos/química , Ligação Proteica , Proteína S/química , Estrutura Terciária de Proteína , Tromboplastina/antagonistas & inibidores
20.
J Thromb Haemost ; 11(4): 704-14, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23347185

RESUMO

BACKGROUND: Tissue factor pathway inhibitor (TFPI) is a multi-Kunitz domain protease inhibitor that down-regulates the extrinsic coagulation pathway by inhibiting FXa and FVIIa. OBJECTIVES: To investigate the role of the three Kunitz domains (KDs) of TFPI in FVIIa inhibition using full-length TFPI (TFPIfl ) and truncated TFPI constructs. METHODS: Inhibition of FVIIa with/without relipidated tissue factor (TF) or soluble TF (sTF) by TFPIfl /TFPI constructs was quantified with a FVIIa-specific chromogenic substrate. RESULTS AND CONCLUSIONS: TFPIfl inhibited TF-FVIIa via a monophasic reaction, which is rather slow at low TFPIfl concentrations (t½  ≈ 5 min at 2 nm TFPI) and has a Ki of 4.6 nm. In the presence of sTF and without TF, TFPIfl was a poor FVIIa inhibitor, with Ki values of 122 nm and 1118 nm, respectively. This indicates that phospholipids and TF significantly contribute to FVIIa inhibition by TFPIfl . TFPI constructs without the KD3-c-terminus (TFPI1-150 and KD1-KD2) were 7-10-fold less effective than TFPIfl in inhibiting TF-FVIIa and sTF-FVIIa, indicating that the KD3-C-terminus significantly contributes to direct inhibition of FVIIa by TFPI. Compared with KD1-KD2, KD1 was a poor TF-FVIIa inhibitor (Ki =434 nm), which shows that the KD2 domain of TFPI also contributes to FVIIa inhibition. Protein S stimulated TF-FVIIa inhibition by TFPIfl (Ki =0.7 nm). In the presence of FXa, a tight quaternary TF-FVIIa-TFPI-FXa complex is formed with TFPIfl , TFPI1-150 and KD1-KD2, with Ki values of < 0.15 nm, 0.5 nm and 0.8 nm, respectively, indicating the KD3-C-terminus is not a prerequisite for quaternary complex formation. Phospholipids and the Gla-domain of FXa are required for quaternary complex formation.


Assuntos
Fator VIIa/antagonistas & inibidores , Lipoproteínas/farmacologia , Sequência de Aminoácidos , Humanos , Lipoproteínas/química , Dados de Sequência Molecular , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA