Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Haemophilia ; 20(1): e40-4, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24354485

RESUMO

The use of induced pluripotent stem cells (iPSCs) as an autologous cell source has shed new light on cell replacement therapy with respect to the treatment of numerous hereditary disorders. We focused on the use of iPSCs for cell-based therapy of haemophilia. We generated iPSCs from mesenchymal stem cells that had been isolated from C57BL/6 mice. The mouse iPSCs were generated through the induction of four transcription factor genes Oct3/4, Klf-4, Sox-2 and c-Myc. The derived iPSCs released functional coagulation factor VIII (FVIII) following transduction with a simian immunodeficiency virus vector. The subcutaneous transplantation of iPSCs expressing FVIII into nude mice resulted in teratoma formation, and significantly increased plasma levels of FVIII. The plasma concentration of FVIII was at levels appropriate for human therapy at 2-4 weeks post transplantation. Our data suggest that iPSCs could be an attractive and prospective autologous cell source for the production of coagulation factor, and that engineered iPSCs expressing coagulation factor might provide a cell-based therapeutic strategy appropriate for haemophilia.


Assuntos
Fator VIII/biossíntese , Fator VIII/genética , Vetores Genéticos/genética , Células-Tronco Pluripotentes Induzidas/metabolismo , Vírus da Imunodeficiência Símia/genética , Animais , Diferenciação Celular , Células Cultivadas , Expressão Gênica , Ordem dos Genes , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Mesenquimais/citologia , Camundongos , Fatores de Tempo , Transdução Genética
2.
J Laryngol Otol ; 126(11): 1114-20, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22989870

RESUMO

OBJECTIVE: This study aimed to investigate the function of tissue plasminogen activator in the olfactory epithelium of mice following neural injury. METHOD: Transmission electron microscopy was used to study the changes in the morphology of the olfactory epithelium 1-7 days after surgical ablation of the olfactory bulb (bulbectomy). RESULTS: Prior to bulbectomy, a uniformly fine material was observed within some regions of the olfactory epithelium of mice deficient in tissue plasminogen activator. At 2-3 days after bulbectomy, there were degenerative changes in the olfactory epithelium. At 5-7 days after bulbectomy, we noted drastic differences in olfactory epithelium morphology between mice deficient in tissue plasminogen activator and wild-type mice (comparisons were made using findings from a previous study). The microvilli seemed to be normal and olfactory vesicles and receptor neuron dendrites were largely intact in the olfactory epithelium of mice deficient in tissue plasminogen activator. CONCLUSION: The tissue plasminogen activator plasmin system may inhibit the regeneration of the olfactory epithelium in the early stages following neural injury.


Assuntos
Bulbo Olfatório/fisiologia , Bulbo Olfatório/cirurgia , Mucosa Olfatória/fisiologia , Regeneração/fisiologia , Ativador de Plasminogênio Tecidual/deficiência , Animais , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica de Transmissão , Mucosa Olfatória/citologia , Ativador de Plasminogênio Tecidual/fisiologia
3.
J Thromb Haemost ; 10(9): 1802-13, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22784361

RESUMO

BACKGROUND: Transplantation of cells overexpressing a target protein represents a viable gene therapeutic approach for treating hemophilia. Here, we focused on the use of autologous mesenchymal stem cells (MSCs) expressing coagulation factor for the treatment of coagulation factor VIII (FVIII) deficiency in mice. METHODS AND RESULTS: Analysis of luciferase gene constructs driven by different promoters revealed that the plasminogen activator inhibitor-1 (PAI-1) gene promoter coupled with the cytomegalovirus promoter enhancer region was one of the most effective promoters for producing the target protein. MSCs transduced with the simian immunodeficiency virus (SIV) vector containing the FVIII gene driven by the PAI-1 promoter expressed FVIII for several months, and this expression was maintained after multiple mesenchymal lineage differentiation. Although intravenous injection of cell supernatant derived from MSCs transduced with an SIV vector containing the FVIII gene driven by the PAI-1 promoter significantly increased plasma FVIII levels, subcutaneous implantation of the MSCs resulted in a transient and weak increase in plasma FVIII levels in FVIII-deficient mice. Interestingly, intra-articular injection of the transduced MSCs significantly ameliorated the hemarthrosis and hemophilic arthropathy induced by knee joint needle puncture in FVIII-deficient mice. The therapeutic effects of a single intra-articular injection of transduced MSCs to inhibit joint bleeding persisted for at least 8 weeks after administration. CONCLUSIONS: MSCs provide a promising autologous cell source for the production of coagulation factor. Intra-articular injection of MSCs expressing coagulation factor may offer an attractive treatment approach for hemophilic arthropathy.


Assuntos
Fatores de Coagulação Sanguínea/metabolismo , Transplante de Células , Fator VIII/genética , Hemofilia A/terapia , Artropatias/terapia , Células-Tronco Mesenquimais/citologia , Animais , Hemofilia A/complicações , Injeções Intra-Articulares , Artropatias/complicações , Células-Tronco Mesenquimais/metabolismo , Camundongos , Inibidor 1 de Ativador de Plasminogênio/genética , Regiões Promotoras Genéticas
4.
Haemophilia ; 18(3): e323-30, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22044430

RESUMO

Haemophilia A is a life long bleeding disorder caused by an inherited deficiency of factor VIII (FVIII). About 30% of haemophilia A patients develop neutralizing antibodies as a consequence of treatment with FVIII concentrates. Immune tolerance protocols for the eradication of inhibitors require daily delivery of intravenous FVIII. We evaluated the immune responses to serial intravenous administration of FVIII in preimmunized haemophilia A mice. We introduced an implantable venous-access device (iVAD) system into haemophilia A mice to facilitate sequential infusion of FVIII. After preimmunization with FVIII, the haemophilia A mice were subjected to serial intravenous administration of FVIII through the iVAD system. In all mice with serial infusion of FVIII, high titers of anti-FVIII inhibitory antibodies developed at 10 exposure days (EDs). However, the anti-FVIII IgG titers were decreased after 150 EDs of sequential low-dose infusion of FVIII [0.05 U g(-1) body weight (BW) five times per week]. Proliferative response to ex vivo FVIII stimulation was significantly suppressed in splenic CD4(+) T cells from mice with serial low-dose FVIII infusion compared with those from mice with high-dose FVIII infusion (0.5 U g(-1) BW five times per week) or preimmunized mice. Moreover, splenic CD4(+) T cells from mice with serial low-dose infusion of FVIII failed to produce interleukin-2 and interferon-γ. These data suggest that serial infusion of FVIII could induce T-cell anergy in haemophilia A mice with inhibitor antibodies.


Assuntos
Inibidores dos Fatores de Coagulação Sanguínea/imunologia , Coagulantes/imunologia , Fator VIII/imunologia , Hemofilia A/imunologia , Tolerância Imunológica/efeitos dos fármacos , Animais , Inibidores dos Fatores de Coagulação Sanguínea/sangue , Cateterismo Venoso Central , Cateteres de Demora , Proliferação de Células/efeitos dos fármacos , Coagulantes/administração & dosagem , Citocinas/metabolismo , Modelos Animais de Doenças , Fator VIII/administração & dosagem , Hemofilia A/tratamento farmacológico , Hemofilia A/metabolismo , Imunoglobulina G/sangue , Infusões Intravenosas , Isoanticorpos/sangue , Camundongos
5.
J Thromb Haemost ; 7(5): 811-24, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19220731

RESUMO

SUMMARY BACKGROUND: Hemophilia A is a congenital bleeding disorder caused by a deficiency of coagulation factor VIII. Approximately 30% of hemophilia A patients develop inhibitors against FVIII following replacement therapy. We have reported that neonatal exposure of FVIII antigen can induce antigen-specific immune tolerance by interferon-gamma (IFN-gamma)-dependent T-cell anergy in hemophilia A mice. OBJECTIVE: The thymus plays crucial roles in self-tolerance, with negative selection of self-reactive effector T cells and positive selection of self-reactive regulatory T cells. We investigated the possibility of the induction of antigen-specific immune tolerance by intrathymic injection of FVIII in hemophilia A mice. METHODS: Hemophilia A mice were injected with recombinant FVIII into the thymus under real-time high-resolution image guidance. RESULTS: Anti-FVIII inhibitory antibody titers in mice challenged with intravenous administration of FVIII were significantly lower in mice (n = 22) that had received thymic FVIII injection than in mice (n = 18) without thymic injection (9.4 +/- 2.3 vs. 122.5 +/- 27.6 BU mL(-1), respectively, P = 0.00078). The CD4(+) T cells from thymic-injected mice could not proliferate or produce interleukin (IL)-2, IL-12 and IFN-gamma in response to FVIII. The CD4(+)CD25(+) T cells generated from thymic-treated mice but not from naïve mice efficiently suppressed the in vitro proliferative response of CD4(+) T cells and blocked the in vivo development of anti-FVIII antibodies in the adoptive transfer. CONCLUSION: These data suggest that intrathymic administration of FVIII could result in immune tolerance by induction of FVIII-specific regulatory T cells.


Assuntos
Fator VIII/imunologia , Hemofilia A/imunologia , Timo/metabolismo , Animais , Autoanticorpos/biossíntese , Autoanticorpos/imunologia , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/imunologia , Proliferação de Células , Fator VIII/administração & dosagem , Citometria de Fluxo , Camundongos
6.
J Thromb Haemost ; 4(8): 1738-46, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16879216

RESUMO

OBJECTIVE: The main objective was to study the relationships of the molecular defects in 38 dysfibrinogens with their fibrin networks. METHODS AND RESULTS: Scanning electron microscopic analyses revealed that all the fibrins formed under the same conditions had networks composed of either normal thickness fibers or thin fibers, accompanied by a variety of alterations in the network structure and characteristics. We classified these fibrin networks into five classes, designated normal, less-ordered, porous A, porous B and lace-like networks. The dysfibrinogens with defects in fibrinopeptide A release or the E:D binding sites formed normal or less-ordered networks, while those with defects in the D:D association formed porous A networks composed of many tapered terminating fibers, despite having fibers of normal width, and containing many pores or spaces. The porous B and lace-like networks were composed of highly branched thin fibers because of defects in the lateral association among protofibrils, and the major difference between them was the porosity of the porous B networks. All the porous B networks were easily damaged by mechanical stress, whereas the lace-like networks retained high resistance to such stress, indicating that the network strength was not dependent on the fiber width, but on the porosity that led to fragility of the network. CONCLUSION: Impairment of the D:D association is the major disturbing factor that leads to the formation of porous fibrin networks. The porosity may be introduced by severe impairment of the D:D association, as well as the lateral association, as has often been observed by extra glycosylation or defects in Ca2+ binding.


Assuntos
Afibrinogenemia/sangue , Fibrina/química , Fibrina/ultraestrutura , Fibrinogênio/química , Fibrinogênios Anormais/genética , Coagulação Sanguínea , Fibrina/classificação , Fibrinogênios Anormais/classificação , Fibrinogênios Anormais/ultraestrutura , Heterozigoto , Homozigoto , Humanos , Microscopia Eletrônica de Varredura , Fatores de Tempo
7.
J Thromb Haemost ; 4(6): 1271-8, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16706971

RESUMO

OBJECTIVES: Although the concept of aspirin resistance is extensively reported in medical literature, its precise mechanisms and clinical outcomes are largely unknown. In this study, we examined individual thromboxane biosynthesis and platelet aggregation in aspirin-treated patients, and whether the results of a platelet aggregation test influenced clinical outcomes. RESULTS: Subjects taking 81 mg of aspirin (n = 50) and controls (n = 38) were evaluated for platelet aggregation and platelet cyclooxygenase-1 (COX-1) activity by measuring collagen-induced thromboxane B2 production. For aggregometry, both light transmission (LT) and laser-light scattering methods were employed to quantitatively evaluate aggregate sizes and numbers. Aspirin treatment resulted in the inhibition of collagen-induced platelet aggregation, particularly the transition from small to large platelet aggregates. Although platelet COX-1 activity seemed to be uniformly inhibited in all patients, platelet aggregation studies showed great inter-individual differences; variation in platelet COX-1 activity only accounted for 6-20% of the individual aggregations. Factor analysis revealed the existence of a common factor (other than platelet COX-1) that explained 48.4% of the variations in platelet aggregation induced by collagen, adenosine diphosphate (ADP), and collagen-related peptide. We then prospectively enrolled 136 aspirin-treated patients in our study, and we found that being in the upper quartile level of LT, or with large aggregate formation induced by collagen, was an independent risk factor for developing cardiovascular events within 12 months [hazard ratio (HR) = 7.98, P = 0.008 for LT; HR = 7.76, P = 0.007 for large aggregates]. On the other hand, the existence of diabetes mellitus was an independent risk factor for overall outcomes (HR 1.30-11.9, P = 0.015-0.033). CONCLUSIONS: Aspirin resistance expressed as unsuppressed platelet COX-1 activity is a rare condition in an out-patient population. Other factor(s) affecting collagen-induced platelet aggregation may influence early outcomes in aspirin-treated patients.


Assuntos
Aspirina/farmacologia , Plaquetas/efeitos dos fármacos , Doenças Cardiovasculares/sangue , Inibidores de Ciclo-Oxigenase/farmacologia , Resistência a Medicamentos , Inibidores da Agregação Plaquetária/farmacologia , Agregação Plaquetária/efeitos dos fármacos , Idoso , Aspirina/efeitos adversos , Plaquetas/enzimologia , Doenças Cardiovasculares/etiologia , Doenças Cardiovasculares/urina , Colágeno/farmacologia , Ciclo-Oxigenase 1/metabolismo , Inibidores de Ciclo-Oxigenase/efeitos adversos , Feminino , Humanos , Masculino , Inibidores da Agregação Plaquetária/efeitos adversos , Testes de Função Plaquetária/métodos , Estudos Prospectivos , Fatores de Risco , Transdução de Sinais , Tromboxano B2/análogos & derivados , Tromboxano B2/metabolismo , Tromboxano B2/urina , Resultado do Tratamento
8.
J Thromb Haemost ; 2(5): 754-62, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15099282

RESUMO

Inhibitory antibody formation is the most serious complication of factor (F)VIII replacement therapy in hemophilia A patients. FVIII-deficient mice were used to study new approaches for induction of immune tolerance. Neither antiFVIII inhibitory antibodies nor antiFVIII IgGs were observed in 13 of 14 adult mice that received 0.05 U g(-1) body weight of human FVIII intravenously within 24 h after birth and repeated injections as adults. In contrast, high FVIII antibody titers (>50 Bethesda Units mL(-1)) developed in seven of 13 mice injected on day 3 postpartum and in all adult mice not treated neonatally. One of nine mice and three of 17 mice developed high-titer antiFVIII inhibitory antibody when they were treated initially with 2-fold (0.1 U g(-1) body weight) and 10-fold higher doses (0.5 U g(-1) body weight) FVIII on day 0, respectively. A human FVIII-specific T-cell proliferative response was absent in splenocytes from neonatally treated mice. The tolerance was FVIII specific because antitoxoid antibodies developed after immunization with tetanus toxoid. Splenocytes failed to proliferate or produce interferon (IFN)-gamma in response to FVIII stimulation, yet still secreted interleukin-2. A proliferative response was restored with exogenous IFN-gamma or interleukin-12, suggesting that lack of inhibitor to FVIII was due to IFN-gamma-dependent anergy. Thus, exposure on day 0 to physiological levels of FVIII antigen might be important for induction of immune tolerance. This immune tolerance model may provide a basis for new approaches to prevention of FVIII inhibitors during replacement therapy.


Assuntos
Fator VIII/administração & dosagem , Hemofilia A/imunologia , Tolerância Imunológica/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Anticorpos Heterófilos/sangue , Coagulação Sanguínea/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Citocinas/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Fator VIII/imunologia , Fator VIII/farmacologia , Hemofilia A/tratamento farmacológico , Humanos , Injeções Intravenosas , Camundongos , Camundongos Knockout , Baço/citologia , Linfócitos T/efeitos dos fármacos , Fatores de Tempo
9.
J Thromb Haemost ; 2(2): 275-80, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14995990

RESUMO

After screening for species-specific antihuman factor (F)IX monoclonal antibodies, we found that antibody 3A6 did not bind to cynomolgus FIX. The 3A6 epitope was found to include Ala262 of human FIX. The 3A6 antibody was used as a catching antibody in an enzyme immunoassay (EIA) for specific detection of human FIX in cynomolgus macaque plasma. No significant increase of substrate hydrolysis was observed when EIA buffer containing cynomolgus macaque plasma was subjected to the 3A6-based EIA. Addition of up to 30% cynomolgus macaque plasma or canine plasma to the assay did not alter detection of human FIX. Three cynomolgus macaques were injected with human FIX (10 U kg-1; i.v.) and the circulating human FIX was quantified in the macaque plasma. The FIX level in the circulation increased to 470 +/- 37.6 ng mL-1 at 1 h after the injection and gradually decreased to 1.79 +/- 1.1 ng mL-1 by day 5, which is approximately 0.06% of the normal human plasma FIX concentration. These data suggest that the cynomolgus macaque can be used as a primate model for studying hemophilia B gene therapy by transduction of macaque organs with vectors to express human FIX in vivo and detection of human FIX using the 3A6 monoclonal antibody.


Assuntos
Modelos Animais de Doenças , Fator IX/farmacocinética , Animais , Anticorpos Monoclonais , Epitopos , Fator IX/administração & dosagem , Fator IX/análise , Hemofilia B , Humanos , Técnicas Imunoenzimáticas , Injeções , Fígado/química , Macaca fascicularis , Distribuição Tecidual
10.
Gene Ther ; 11(3): 253-9, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14737084

RESUMO

We demonstrate that transduction of adipocytes with a simian immunodeficiency virus agm TYO1 (SIVagm)-based lentiviral vector carrying the human coagulation factor VIII gene (SIVhFVIII) resulted in expression of the human FVIII transgene in vitro and in db/db mice in vivo. Cultured human adipocytes were transduced with the SIVagm vector carrying the GFP gene in a dose-dependent manner and transduction of adipocytes with SIVhFVIII resulted in efficient expression of human coagulation factor VIII (hFVIII; 320 +/- 39.8 ng/10(6) adipocytes/24 h) in vitro. Based upon successful transduction of adipocytes by SIV vectors carrying the lacZ gene in vivo in mice, the adipose tissue of db/db mice was transduced with SIVhFVIII. There was a transient appearance of human FVIII in mouse plasma (maximum 1.8 ng/ml) on day 11 after the injection. Transcripts of human FVIII transgene and human FVIII antigen also were detected in the adipose tissue by RT-PCR and immunofluorescence, respectively, on day 14. Emergence of anti-human FVIII antibodies 14 days after the injection of SIVhFVIII may explain the disappearance of human FVIII from the circulation. These results suggest that transduction of the adipocytes with vectors carrying the human FVIII gene may be potentially applicable for gene therapy of hemophilia A.


Assuntos
Adipócitos/metabolismo , Fator VIII/metabolismo , Terapia Genética/métodos , Hemofilia A/terapia , Transdução Genética/métodos , Animais , Células Cultivadas , Fator VIII/genética , Vetores Genéticos , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Vírus da Imunodeficiência Símia/genética , beta-Galactosidase/metabolismo
11.
J Thromb Haemost ; 1(11): 2356-9, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14629469

RESUMO

Congenital hypofibrinogenemia, fibrinogen Tottori II, caused by a nonsense mutation in the fibrinogen Bbeta chain gene, was found in a 68-year-old Japanese female. The plasma fibrinogen level was 99.2 mg dL(-1) as determined by the thrombin time method. No overt molecular abnormalities were observed in purified patient fibrinogen by SDS-PAGE analysis. After sequencing all exons and exon-intron boundaries of three fibrinogen genes, we found a heterozygous single point mutation of T-->G at position 3356 of the patient fibrinogen Bbeta chain gene. This nucleotide mutation results in a nonsense mutation (TAT sequence for Bbeta 41Tyr to TAG sequence for a translation termination signal). The mutation was confirmed by polymerase chain reaction-restriction fragment length polymorphism analysis, since this nucleotide mutation results in a new NheI recognition sequence at this position. These data indicated that the nonsense mutation of the fibrinogen Bbeta chain gene caused a truncated fibrinogen Bbeta chain, which may not be assembled in the fibrinogen molecule.


Assuntos
Transtornos de Proteínas de Coagulação/genética , Códon sem Sentido , Fibrinogênios Anormais/genética , Idoso , Transtornos de Proteínas de Coagulação/congênito , Análise Mutacional de DNA/métodos , Desoxirribonucleases de Sítio Específico do Tipo II , Feminino , Fibrinogênio/análise , Fibrinogênio/genética , Humanos , Mutação Puntual
12.
Blood ; 97(12): 3783-9, 2001 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-11389017

RESUMO

Acquired coagulation factor inhibitors include pathologic immunoglobulins that specifically bind to coagulation factors and either neutralize their procoagulant activity, accelerate their clearance from the circulation, or have proteolytic activity to degrade them into inactive polypeptides. Here, an autoantibody against prothrombin is described in a patient with serious hemorrhagic diatheses. The autoantibody exerts its influence by a previously unknown mechanism in which it inhibits coagulation through aberrant activation of the proenzyme in a catalytic manner. The antibody-bound prothrombin formed a stable stoichiometric complex with antithrombin III, consisting of intact prothrombin and an antithrombin III molecule cleaved at the (393)Arg-(394)Ser bond. The antibody dissociated from prothrombin after the complex formation with antithrombin III. Although the bound antibody elicited protease activity from prothrombin, the complex was not able to convert fibrinogen to fibrin or to activate protein C. Thus, this is the first description of an autoantibody that induces protease-like activity from a human proenzyme, permitting subsequent neutralization by its physiological inhibitor. (Blood. 2001;97:3783-3789)


Assuntos
Antitrombina III/metabolismo , Autoanticorpos/sangue , Transtornos da Coagulação Sanguínea/etiologia , Protrombina/imunologia , Sequência de Aminoácidos , Autoanticorpos/farmacologia , Transtornos da Coagulação Sanguínea/imunologia , Ativação Enzimática/efeitos dos fármacos , Epitopos/análise , Fibrinogênio/metabolismo , Hematoma/imunologia , Humanos , Cinética , Masculino , Pessoa de Meia-Idade , Ligação Proteica , Protrombina/metabolismo , Espaço Retroperitoneal/patologia , Serina Endopeptidases/metabolismo
13.
Gene Ther ; 8(22): 1690-7, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11892836

RESUMO

We were able to facilitate plasminogen activator inhibitor 1 (PAI-1) promoter activity approximately by 14-fold using an enhancer element. This enhanced PAI-1 promoter has a strong basal activity, comparable to CAG promoter activity, and has a response similar to the PAI-1 promoter with respect to TGFbeta 1 and TNFalpha stimulation. The characteristics of the enhanced PAI-1 promoter are thought to be suited to timely and tissue-specific expression of anticoagulant molecules in the vascular cells. Thus, we developed recombinant adeno-associated virus (rAAV) vectors using the enhanced PAI-1 promoter and were successful in transducing vascular endothelial cells to express the thrombomodulin transgene under the regulation of the enhanced PAI-1 promoter in vitro. Thromobomodulin transgene expression driven by the enhanced PAI-1 promoter in rAAV vector-transduced cultured endothelial cells was between 600- and 1000-fold higher than constitutive thrombomodulin gene expression in cultured human umbilical vein endothelial cells and was up-regulated by TGFbeta1 and TNFalpha stimulation which may down-regulate endogenous thrombomodulin gene expression in endothelial cells. The brain vascular endothelial cells of Mongolian gerbils could also be transduced by the same rAAV vector in vivo. Transduction of endothelial cells by rAAV vectors to express enhanced PAI-1 promoter-driven transgenes may be a useful gene therapy approach for vascular diseases.


Assuntos
Dependovirus/genética , Endotélio Vascular/metabolismo , Vetores Genéticos/administração & dosagem , Inibidor 1 de Ativador de Plasminogênio/genética , Regiões Promotoras Genéticas , Trombomodulina/genética , Animais , Artéria Carótida Primitiva , Bovinos , Células Cultivadas , Regulação da Expressão Gênica/efeitos dos fármacos , Gerbillinae , Humanos , Transdução Genética , Fator de Crescimento Transformador beta/farmacologia , Fator de Crescimento Transformador beta1 , Fator de Necrose Tumoral alfa/farmacologia
14.
Blood ; 96(12): 3779-85, 2000 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-11090060

RESUMO

The authors have identified a 12-residue carboxyl-terminal extension of Lys-Ser-Pro-Met-Arg-Arg-Phe-Leu-Leu-Phe-Cys-Met in a dysfibrinogen derived from a woman heterozygotic for this abnormality and associated with severe bleeding. This extension is due to a T-to-A mutation that creates AAG encoding Lys at the stop (TAG) codon, thus translating 36 base pairs in the noncoding region of the Bbeta gene. The extra Cys residues appear to be involved in 1 or 2 disulfide bonds between 2 adjacent abnormal fibrinogen molecules, forming a fibrinogen homodimer as indicated by sodium dodecyl sulfate-polyacrylamide gel electrophoresis. Indeed, about half of the fibrinogen molecules exist as end-linked dimers oriented in parallel or with an angle, as observed by transmission electron microscopy. These end-linked dimers may well alter the conformations of D and DD regions on fibrin assembly, leading to increased fiber branching at their sites in the growing protofibrils. By scanning electron microscopy, the Osaka VI fibrin network appears to have a lacelike structure composed of highly branched, thinner fibers than the normal fibrin architecture. Such fibrin networks may be easily damaged to form large pores when fluids are allowed to pass through the gels. The fragility of Osaka VI fibrin clots, further confirmed by permeation and compaction studies, may account for the massive bleeding observed in this patient. (Blood. 2000;96:3779-3785)


Assuntos
Fibrinogênios Anormais/química , Adulto , Coagulação Sanguínea/genética , Cromatografia Líquida de Alta Pressão , Cisteína/química , Dimerização , Dissulfetos/química , Endopeptidases/metabolismo , Feminino , Fibrinogênios Anormais/genética , Fibrinogênios Anormais/ultraestrutura , Humanos , Microscopia Eletrônica de Varredura , Fragmentos de Peptídeos/química , Permeabilidade , Análise de Sequência de Proteína
15.
Blood ; 94(11): 3806-13, 1999 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-10572095

RESUMO

A novel BbetaAsn-160 (TAA) to Ser (TGA) substitution has been identified in fibrinogen Niigata derived from a 64-year-old asymptomatic woman, who is heterozygotic for this abnormality. The mutation creates an Asn-X-Ser-type glycosylation sequence, and a partially sialylated biantennary oligosaccharide was linked to the BbetaAsn-158 residue. The functional abnormality was attributed to delayed lateral association of normally formed double-stranded protofibrils based on normal cross-linking of fibrin gamma-chains and tissue-type plasminogen activator-catalyzed plasmin generation by polymerizing fibrin monomers. Enzymatic removal of all the N-linked oligosaccharides from fibrinogen Niigata accelerated fibrin monomer polymerization that reached the level of untreated normal fibrin monomers, but the thrombin time was prolonged from 18.2 seconds to 113 seconds (normal: 11.2 seconds to 8.9 seconds). By scanning electron micrographic analysis, Niigata fibrin fibers were found to be more curvilinear than normal fibrin fibers. After deglycosylation, Niigata fibers became straight being similar to untreated normal fibrin fibers, whereas normal deglycosylated fibrin appeared to be less-branched than untreated normal or deglycosylated Niigata fibrin. Although normal and Niigata fibrins were similar to each other in permeation and compaction studies, deglycosylated normal and Niigata fibrins had much higher permeability and compaction values, indicating that deglycosylation had brought about the formation of more porous networks. The enzymatic deglycosylation necessitates an Asn to Asp change at position Bbeta-158 that is responsible for reducing the fiber thickness because of either local repulsive forces or steric hindrance in the coiled-coil region.


Assuntos
Fibrina/química , Fibrinogênio/química , Fibrinogênio/genética , Substituição de Aminoácidos , Asparagina , Feminino , Fibrina/metabolismo , Fibrinogênio/metabolismo , Glicosilação , Humanos , Pessoa de Meia-Idade , Mutação Puntual , Serina
16.
Nihon Rinsho ; 57 Suppl: 678-81, 1999 Sep.
Artigo em Japonês | MEDLINE | ID: mdl-10543211
17.
Thromb Haemost ; 81(6): 940-4, 1999 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10404772

RESUMO

A new type of substitution, Arg to Ser at gamma275, has been found in a heterozygous dysfibrinogen derived from a 23-year-old woman with no major bleeding or thrombosis. By sequence analyses of the affected gamma-chain and its gene. we found a single amino acid substitution of gamma Arg-275 to Ser in an aberrant gamma (274-302) residue peptide isolated from lysyl endopeptidase-digests of the patient's fibrinogen. In agreement with this amino acid substitution, we identified a single nucleotide exchange of A for C at position 5728 in the gamma-chain gene creating a codon (AGC) encoding Ser instead of the codon (CGC) encoding Arg at position gamma 275. Like two other known types of mutants with a His or Cys substitution at this position, the functional abnormality was characterized by delayed fibrin polymerization, most likely due to impaired abutting of two D domains of adjacent fibrin monomers in the same strand of fibrin protofibrils. The structural derangement that affects the D:D association may not be so severe as compared with those of Cys and His mutants, possessing an additional disulfide-linked Cys molecule and an imidazole ring at the mutation site, respectively.


Assuntos
Coagulação Sanguínea , Fibrina/metabolismo , Fibrinogênio/genética , Mutação Puntual , Adulto , Substituição de Aminoácidos , Arginina/genética , Feminino , Fibrinogênio/metabolismo , Humanos , Serina/genética
18.
Blood ; 94(2): 475-82, 1999 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-10397715

RESUMO

Plasminogen activator inhibitor-1 (PAI-1) is present in the platelet alpha-granule and is released on activation. However, there is some debate as to whether the megakaryocyte and platelet synthesize PAI-1, take it up from plasma, or both. We examined the expression of PAI-1 in differentiating megakaryocytic progenitor cells (UT-7) and in CD34(+)/CD41(-) cells from cord blood. UT-7 cells differentiated with thrombopoietin (TPO) resembled megakaryocytes (UT-7/TPO) with respect to morphology, ploidy, and the expression of glycoprotein IIb-IIIa. PAI-1 messenger RNA (mRNA) expression was upregulated and PAI-1 protein synthesized in the UT-7/TPO cells accumulated in the cytoplasm without being released spontaneously. In contrast, erythropoietin (EPO)-stimulated UT-7 cells (UT-7/EPO) did not express PAI-1 mRNA after stimulation with TPO because they do not have endogenous c-Mpl. After cotransfection with human wild-type c-mpl, the cells (UT-7/EPO-MPL) responded to phorbol 12-myristate 13-acetate (PMA), tumor necrosis factor-alpha (TNF-alpha), and interleukin-1beta (IL-1beta) with enhanced PAI-1 mRNA expression within 24 to 48 hours. However, induction of PAI-1 mRNA in UT-7/EPO-MPL cells by TPO required at least 14-days stimulation. UT-7/EPO cells expressing c-Mpl changed their morphology and the other characteristics similar to the UT-7/TPO cells. TPO also differentiated human cord blood CD34(+)/CD41(-) cells to CD34(-)/CD41(+) cells, generated morphologically mature megakaryocytes, and induced the expression of PAI-1 mRNA. These results suggest that both PAI-1 mRNA and de novo PAI-1 protein synthesis is induced after differentiation of immature progenitor cells into megakaryocytes by TPO.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Megacariócitos/metabolismo , Inibidor 1 de Ativador de Plasminogênio/biossíntese , Receptores de Citocinas , Trombopoetina/farmacologia , Biomarcadores , Diferenciação Celular/efeitos dos fármacos , Ensaio de Unidades Formadoras de Colônias , DNA Complementar/genética , Eritropoetina/farmacologia , Sangue Fetal/citologia , Humanos , Leucemia Megacarioblástica Aguda/patologia , Megacariócitos/efeitos dos fármacos , Proteínas de Neoplasias/metabolismo , Inibidor 1 de Ativador de Plasminogênio/genética , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptores de Trombopoetina , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Acetato de Tetradecanoilforbol/farmacologia , Transfecção , Células Tumorais Cultivadas
20.
Histochem Cell Biol ; 110(5): 449-55, 1998 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9826124

RESUMO

We evaluated a possible role for fibrinolytic components in nasal secretion by tissue localization with immunohistochemical techniques and by measuring their antigen concentrations in nasal discharge by means of ELISA and fibrin autography. Nasal mucosa was obtained surgically from the inferior turbinate. Urokinase-type plasminogen activator (u-PA) specific staining was observed in pseudostratified ciliated epithelium and was predominant in mucous cells of the seromucinous gland, while serous cells were almost devoid of stain. The pattern of staining of plasminogen activator inhibitor-2 was similar to that of u-PA. In contrast, plasminogen activator inhibitor-1 (PAI-1) immunoreactive material was localized exclusively in serous cells of seromucinous glands. Positive staining for tissue-type plasminogen activator (t-PA) was observed in endothelial cells and basal cells, which differentiate into either ciliated or goblet cells. Nasal secretions were partially fractionated by immunospecific antibody-immobilized Sepharose. Subsequent fibrin autography patterns indicated the presence of u-PA, PAI-1, and t-PA. After methacholine provocation, the level of t-PA increased transiently but decreased rapidly with subsequent challenges. These differential stainings of fibrinolytic components and the existence of PAs and PAI-1 in the nasal discharge suggest that the fibrinolytic system may play a role in the movement and fluidity of nasal secretion.


Assuntos
Mucosa Nasal/metabolismo , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Autorradiografia , Ensaio de Imunoadsorção Enzimática , Feminino , Fibrina/metabolismo , Fibrinólise , Humanos , Imuno-Histoquímica , Masculino , Mucosa Nasal/imunologia , Inibidor 1 de Ativador de Plasminogênio/imunologia , Ativador de Plasminogênio Tipo Uroquinase/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...