Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
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.
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
3.
J Cell Biol ; 110(1): 155-63, 1990 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-2104856

RESUMO

The interactions between exogenously added tissue-type plasminogen activator (t-PA) and the active form of type 1 plasminogen activator inhibitor (PAI-1) produced by and present in cultured human umbilical vein endothelial cells (HUVECs) were investigated. Immunoblotting analysis of the conditioned media obtained from monolayers of HUVECs treated with increasing concentrations of t-PA (less than or equal to 10 micrograms/ml) revealed a dose-dependent formation of both t-PA/PAI-1 complexes, and of a 42,000-Mr cleaved or modified form of the inhibitor. Immunoradiometric assays indicated that t-PA treatment resulted in a fourfold increase in PAI-1 antigen present in the conditioned media. This increase did not result from the release of PAI-1 from intracellular stores, but rather reflected a t-PA-dependent decrease in the PAI-1 content of the Triton X-100 insoluble extracellular matrix (ECM). Although the rate of t-PA-mediated release of PAI-1 was increased by the removal of the monolayer, similar quantities of PAI-1 were removed in the presence or absence of the cells. These results suggest that the cells only represent a semipermeable barrier between ECM-associated PAI-1 and exogenous t-PA. Treatment of HUVECs with t-PA (1 microgram/ml, 2 h) to deplete the ECM of PAI-1 did not affect the subsequent rate of PAI-1 production and deposition into the ECM. Immunogold electron microscopy of HUVECs not only confirmed the location of PAI-1 primarily in the region between the culture substratum and ventral cell surface but failed to demonstrate significant (less than 1%) PAI-1 on the cell surface. Thus, the majority of PAI-1 associated with cultured HUVEC monolayers is present under the cells in the ECM and is accessible to solution-phase t-PA.


Assuntos
Endotélio Vascular/metabolismo , Inativadores de Plasminogênio/análise , Ativador de Plasminogênio Tecidual/metabolismo , Divisão Celular , Células Cultivadas , Endotélio Vascular/citologia , Endotélio Vascular/ultraestrutura , Matriz Extracelular/metabolismo , Matriz Extracelular/ultraestrutura , Feminino , Fibrinólise , Humanos , Immunoblotting , Imuno-Histoquímica , Microscopia Eletrônica , Peso Molecular , Inativadores de Plasminogênio/metabolismo , Gravidez , Radioimunoensaio , Soluções , Veias Umbilicais
4.
J Clin Invest ; 77(3): 1006-13, 1986 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-2419360

RESUMO

When blood coagulation takes place in the presence of calcium ions, alpha 2-plasmin inhibitor (alpha 2PI) is cross-linked to fibrin by activated coagulation Factor XIII (XIIIa) and thereby contributes to the resistance of fibrin to fibrinolysis. It was previously shown that the cross-linking reaction is a reversible one, since the alpha 2PI-fibrinogen cross-linked complex could be dissociated. In the present study we have shown that the alpha 2PI-fibrin cross-linking reaction is also a reversible reaction and alpha 2PI which had been cross-linked to fibrin can be released from fibrin by disrupting the equilibrium, resulting in a decrease of its resistance to fibrinolysis. When the fibrin clot formed from normal plasma in the presence of calcium ions was suspended in alpha 2PI-deficient plasma of buffered saline, alpha 2PI was gradually released from fibrin on incubation. When alpha 2PI was present in the suspending milieu, the release was decreased inversely to the concentrations of alpha 2PI in the suspending milieu. The release was accelerated by supplementing XIIIa or the presence of a high concentration of the NH2-terminal 12-residue peptide of alpha 2PI (N-peptide) which is cross-linked to fibrin in exchange for the release of alpha 2PI. When the release of alpha 2PI from fibrin was accelerated by XIIIa or N-peptide, the fibrin became less resistant to the fibrinolytic process, resulting in an acceleration of fibrinolysis which was proportional to the degree of the release of alpha 2PI. These results suggest the possibility that alpha 2PI could be released from fibrin in vivo by disrupting the equilibrium of the alpha 2PI-fibrin cross-linking reaction, and that the release would result in accelerated thrombolysis.


Assuntos
Fator XIII/metabolismo , Fibrina/metabolismo , Fibrinólise , alfa 2-Antiplasmina/metabolismo , alfa-Macroglobulinas/metabolismo , Eletroforese em Gel de Poliacrilamida , Humanos , Cinética , Fragmentos de Peptídeos/farmacologia
5.
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
6.
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
7.
Biochim Biophys Acta ; 1160(3): 325-34, 1992 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-1477106

RESUMO

We have shown that the urokinase (UK) kringle domain contains a high-affinity plasminogen activator inhibitor-1 (PAI-1) binding site, responsible for the 10-fold faster complex formation between UK and PAI-1 than between PAI-1 and low-molecular-weight urokinase (LMWUK). Complex formation between UK and PAI-1, but not between LMWUK and PAI-1, was suppressed 10-fold in the presence of peptide U-107 derived from the UK kringle domain. Peptide U-373 derived from the UK catalytic domain slowed complex formation between UK and PAI-1 and also LMWUK and PAI-1. Inactivation of tissue-type plasminogen activator (tPA) by PAI-1 was slowed 10-fold in the presence of peptides derived from the tPA finger and kringle-2 domains. DFP-inactivated (DIP) UK and both forms of DIP-tPA inhibited PAI-1 binding to U-107 and to U-373 whereas single-chain urokinase-type PA (scuPA) was unable to compete with either peptide for PAI-1 binding. These data suggest that the reversible PAI-1 binding site in the UK A-chain plays a role in the rapid association with PAI-1 as important as those that reside in the tPA A-chain and that reversible PAI-1 binding sites are expressed on the surface of UK upon conversion from scuPA, in contrast to tPA.


Assuntos
Inibidor 1 de Ativador de Plasminogênio/química , Ativadores de Plasminogênio/química , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais , Sítios de Ligação , Linhagem Celular , Fibrinolisina , Humanos , Cinética , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Estrutura Molecular , Inibidor 2 de Ativador de Plasminogênio/química , Ativador de Plasminogênio Tipo Uroquinase/química , Ativador de Plasminogênio Tipo Uroquinase/imunologia
8.
Biochim Biophys Acta ; 1245(1): 69-75, 1995 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-7654768

RESUMO

Activation of Glu- and Lys-plasminogen by various concentrations of recombinant staphylokinase (SAK) were studied by the generation of amidolytic activity from the chromogenic substrate S-2251(H-D-Val-Leu-Lys-pNA) and by SDS-PAGE analysis. Surprisingly, excess SAK decreased and fixed the rate of S-2251 hydrolysis in a mixture of Lys-plasminogen and SAK. Since the effect of SAK on S-2251 hydrolysis by plasma was similar, the hydrolysis kinetics by free plasmin and plasmin-SAK complex were studied. Hydrolysis by either enzyme form followed Michaelis-Menten kinetics with a Km of 0.38 mM for plasma and 3.74 mM for SAK-plasmin complex. The catalytic rate constant was 22.7 s-1 for plasmin and 21.0 s-1 for the SAK-plasmin complex. With excess SAK and vigorous removal of plasmin activity from plasminogen, the pre-activation lag period differed greatly between Glu- and Lys-plasminogen. Based on the different substrate specificity of plasmin and plasmin-SAK complex, we analyzed the Glu-plasminogen activation with either catalytic or excess SAK. With excess SAK, almost no Lys-plasminogen was detectable and whole Glu-plasminogen was converted directly to Glu-plasmin, then gradually to Lys-plasmin. In contrast, Lys-plaminogen appeared rapidly with catalytic amount of SAK. These results suggest that inhibition of Glu-plasminogen to Lys-plasminogen to Lys-plasminogen conversion in the plasminogen-SAK complex in the presence of excess SAK prolonged the initial lag phase of activation.


Assuntos
Fibrinolisina/metabolismo , Metaloendopeptidases/farmacologia , Plasminogênio/metabolismo , Relação Dose-Resposta a Droga , Ativação Enzimática , Cinética , Metaloendopeptidases/metabolismo , Conformação Proteica , Proteínas Recombinantes/farmacologia , Especificidade por Substrato
9.
Biochim Biophys Acta ; 1226(3): 300-6, 1994 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-8054362

RESUMO

We studied the effect of the glucosidase I inhibitor, N-methyl-1-deoxynojirimycin (MdN) and the mannosidase inhibitor, 1-deoxymannojirimycin (dMM) on the biosynthesis and secretion of alpha 2-plasmin inhibitor (alpha 2-PI) and antithrombin III (ATIII) in cultures of human hepatoma (Hep-G2) cells. Incubation with 1 mM MdN decreased secreted alpha 2-PI activity and antigen levels by about 40%, whereas those of ATIII were not affected. Neither inhibitor affected the messenger RNA levels as determined by Northern blotting. Pulse-chase studies using [35S]-methionine showed that MdN decreased alpha 2-PI and ATIII secretion rates. By the 18 h chase, MdN had decreased secreted alpha 2-PI to 50-60%, with little effect on ATIII. Intracellular forms of alpha 2-PI or ATIII synthesized by cells treated with 1 mM MdN were sensitive to endoglycosidase H (Endo H), whereas almost all the secreted forms were resistant, suggesting the presence of complex-type oligosaccharides. In the presence of 1 mM dMM, cells synthesized Endo H-sensitive alpha 2-PI and ATIII with similar secretion rates. These results suggest that retention of glucose on N-linked oligosaccharides not only retards the exit of alpha 2-PI and ATIII, but also changes the catabolic rate of alpha 2-PI in the endoplasmic reticulum.


Assuntos
1-Desoxinojirimicina/análogos & derivados , 1-Desoxinojirimicina/farmacologia , Antitrombina III/biossíntese , Glicosídeo Hidrolases/antagonistas & inibidores , alfa 2-Antiplasmina/biossíntese , Animais , Cães , Glicosídeo Hidrolases/genética , Hexosaminidases , Humanos , Manosidases/antagonistas & inibidores , Manosidases/genética , Manosil-Glicoproteína Endo-beta-N-Acetilglucosaminidase , RNA Mensageiro/análise , Células Tumorais Cultivadas/metabolismo
10.
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
11.
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
12.
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
13.
Pediatrics ; 77(5): 670-6, 1986 May.
Artigo em Inglês | MEDLINE | ID: mdl-3754634

RESUMO

Severe and recurrent purpura fulminans developed in a Chinese boy at one day of age. Results of coagulation studies performed on the patient during attacks were compatible with the diagnosis of disseminated intravascular coagulation. Subsequent investigations have revealed that the patient is homozygous and that his parents are heterozygous for protein C deficiency. Cryoprecipitate and fresh frozen plasma induced a remission, and administration of warfarin has been successful in preventing recurrence of attacks for as long as 8 months without infusion of any plasma components. None of the family members who are heterozygous for protein C deficiency have had thrombotic episodes.


Assuntos
Glicoproteínas/deficiência , Deficiência de Proteína/genética , Púrpura/complicações , Terapia Combinada , Crioglobulinas/uso terapêutico , Heparina/uso terapêutico , Homozigoto , Humanos , Recém-Nascido , Masculino , Necrose/patologia , Plasma , Proteína C , Deficiência de Proteína/complicações , Púrpura/patologia , Púrpura/terapia , Pele/patologia , Varfarina/uso terapêutico
14.
Thromb Haemost ; 76(4): 569-76, 1996 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-8902998

RESUMO

We have studied the expression of plasminogen activator inhibitor 2 (PAI-2) in the adult mouse and during embryonic development using immunohistochemistry and the polymerase-chain reaction. Mouse PAI-2 mRNA was mainly expressed in the skin, bone-marrow, spleen, lung, thymus, and urinary bladder. Immunohistochemical studies suggested that PAI-2 was synthesized in macrophages and epithelial cells such as skin epithelial cells, transitional cells of the urinary bladder, and mesothelial cell of peritoneum and pleura. PAI-2 mRNA and antigen expression was observed only in the skin of 15 day-old mouse embryos. These data suggest that PAI-2 may play a role in protecting the mouse embryo from the protease attack in the amniotic fluid and also in maintaining and/or protecting the structure of various surfaces in the mouse.


Assuntos
Envelhecimento/metabolismo , Macrófagos/metabolismo , Inibidor 2 de Ativador de Plasminogênio/genética , Pele/metabolismo , Animais , Sequência de Bases , Desenvolvimento Embrionário e Fetal/fisiologia , Células Epiteliais , Epitélio/metabolismo , Escherichia coli , Feminino , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Inibidor 2 de Ativador de Plasminogênio/biossíntese , Reação em Cadeia da Polimerase , Gravidez , Proteínas Recombinantes/biossíntese , Pele/citologia , Pele/embriologia
15.
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
16.
J Biochem ; 111(2): 244-8, 1992 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-1314812

RESUMO

We have shown that plasminogen activator inhibitor-1 (PAI-1) inhibits the fibrin binding of both the single chain and two chain forms of tissue-type plasminogen activator (tPA) through two different mechanisms. PAI-1 inhibits the finger domain-dependent fibrin binding of diisopropylfluorophosphate-inactivated single chain tPA and the kringle-2 domain-dependent fibrin binding of diisopropylfluorophosphate-inactivated two chain tPA. In accordance with the data, preformed complexes of single chain tPA/PAI-1 and of two chain tPA/PAI-1 lost the fibrin binding abilities mediated by the finger and kringle-2 domains, respectively. These effects of PAI-1 appear to be mediated by steric hindrance of the fibrin binding sites after PAI-1 binding to adjacent regions in the functional domains of tPA. We thus propose a model in which a PAI-1 binding site resides in the finger domain of a single chain, and plays a role in the reversible association of single chain tPA and PAI-1. Conformational changes may take place during the conversion of single chain tPA to two chain tPA, resulting in burying of the original PAI-1 binding site and exposure of an alternate PAI-1 binding site on the surface of the kringle-2 domain.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Fibrina/metabolismo , Fragmentos de Peptídeos/metabolismo , Inativadores de Plasminogênio/farmacologia , Ativador de Plasminogênio Tecidual/metabolismo
17.
J Biochem ; 123(1): 71-7, 1998 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9504411

RESUMO

We have evaluated the effect of lysine binding sites in kringle structures on the activation of plasminogen with plasmin and staphylokinase (SAK) complex and on the binding of plasminogen to SAK. Activation of native plasminogen (Glu-plasminogen) by a catalytic amount of plasmin-SAK complex increased in the presence of epsilon-amino-n-caproic acid (EACA) and then decreased with higher concentrations of EACA. By contrast, activation of modified plasminogen (Lys-plasminogen) decreased in an EACA-concentration-dependent manner. This decrease was explained by a more than 10-fold higher Km for activation of Lys-plasminogen with a catalytic amount of plasmin-SAK complex in the presence of EACA. EACA was a competitive inhibitor with Ki 0.23 mM. In addition, the Km for activation of mini-plasminogen, which lacks first four kringle structures (K1+2+3+4), was at least 3.5-fold higher than that for the activation of Lys-plasminogen. Furthermore, EACA showed a negligible inhibitory effect on the activation of mini-plasminogen by the plasmin-SAK complex. We observed a similar biphasic effect of EACA on the binding of Glu-plasminogen to SAK and a dose-dependent effect on the Lys-plasminogen binding to SAK by gel filtration methods. Since EACA binds to plasminogen via lysine binding sites in the kringle structure, we propose that the lysine binding site in K1+2+3+4 domain plays a role in the activation of plasminogen by plasmin SAK complex, and in the binding of plasminogen to SAK.


Assuntos
Kringles/fisiologia , Metaloendopeptidases/metabolismo , Plasminogênio/metabolismo , Ácido Aminocaproico/metabolismo , Sítios de Ligação , Catálise , Fibrinolisina/metabolismo , Humanos , Lisina/metabolismo , Ligação Proteica , Proteínas Recombinantes/metabolismo
18.
J Biochem ; 121(2): 278-87, 1997 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9089401

RESUMO

Two groups of anti-plasminogen monoclonal antibodies, whose epitope was either in the kringle 1 + 2 + 3 domain (F3P2, F11P5, F11P6, and F12P18) or the kringle 5 domain (F1P6 and F12P16), were isolated and their effects on the conformation of plasminogen were explored. All antibodies except F1P6 had 3- to 10-fold higher affinity toward Lys-plasminogen than Glu-plasminogen. F1P6 exhibited a comparable affinity to Glu- and Lys-plasminogen. Among these, only F11P5 binding was inhibited by epsilon-amino-nu-caproic acid (EACA) in a concentration-dependent manner, with half maximal inhibition at 3 mM. From a competition assay, we concluded that the epitopes of F11P5, F11P6, and F12P18 should be very close, and located at or near the low affinity lysine binding site on the kringle 2 + 3. These three antibodies dramatically enhanced the binding of Glu-plasminogen to the other antibodies, except to F1P6. Interestingly, F3P2, whose non-overlapping epitope was in the kringle 2 + 3 domain, also augmented the binding of Glu-plasminogen to the other antibodies. In contrast, we did not observe enhanced binding of Lys-plasminogen to one antibody in the presence of the other antibodies, and the binding of Glu-plasminogen to these antibodies did not increase in the presence of 10 mM EACA. In the presence of these antibodies, including F1P6, Glu-plasminogen bound more efficiently to immobilized degraded fibrin, with a binding profile similar to Lys-plasminogen. All antibodies except F1P6 enhanced the conversion rate of plasminogen to plasmin remarkably. Taken together, we propose that these two groups of monoclonal antibodies can dissociate the intramolecular interactions of Glu-plasminogen and induce the conformational transition of Glu-plasminogen to Lys-plasminogen. In addition, the kringle 2 + 3 and kringle 5 structures of Glu-plasminogen liganded with EACA are distinct from the Lys-plasminogen structure.


Assuntos
Anticorpos Monoclonais/imunologia , Fragmentos de Peptídeos/imunologia , Plasminogênio/imunologia , Animais , Anticorpos Monoclonais/metabolismo , Ligação Competitiva , Western Blotting , Catálise , Fibrina/metabolismo , Fibrinolisina/metabolismo , Humanos , Cinética , Kringles/imunologia , Lisina , Camundongos , Fragmentos de Peptídeos/metabolismo , Plasminogênio/metabolismo , Ligação Proteica , Conformação Proteica , Relação Estrutura-Atividade , Propriedades de Superfície , Ativador de Plasminogênio Tipo Uroquinase/metabolismo
19.
Int J Hematol ; 57(1): 9-14, 1993 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-8477066

RESUMO

Protein C Yonago is a dysfunctional protein C characterized by defective anticoagulant activity determined by a coagulation assay and normal amidolytic activity measured on a synthetic substrate S-2366 (Iijima et al., Thromb Res 1991;63:249-257). We have identified a single point mutation of C to G in codon 57 (CGG-->GGG) of the gene for protein C Yonago by genetic analysis utilizing the polymerase chain reaction. The mutation should have resulted in an amino acid substitution of Gly for Arg at position 15 of the light chain of mature protein C. No mutations were found in nucleotides spanning the putative gamma-carboxylase recognition site or gamma-carboxyglutamic acid residues of protein C. Protein C Yonago was non-reactive to monoclonal antibodies JTC-1 and -3 that solely recognized the calcium-dependent conformation of the gamma-carboxyglutamic acid domain. This indicated that the mutation had critically perturbed the highly conserved structure of the gamma-carboxyglutamic acid domain. Thus, the calcium-dependent conformation required for the phospholipids binding to exert the physiological functions of protein C may not have been elicited normally in this abnormal protein C, resulting in defective generation of anticoagulant activities in plasma. Consequently, no anticoagulant activities may have been generated in vivo.


Assuntos
Arginina/genética , Códon , Glicina/genética , Mutação , Proteína C/genética , Ácido 1-Carboxiglutâmico/genética , Sequência de Aminoácidos , Sequência de Bases , Cálcio/fisiologia , Humanos , Dados de Sequência Molecular , Estrutura Terciária de Proteína , Trombose/genética
20.
Int J Hematol ; 56(2): 129-34, 1992 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-1421174

RESUMO

In four abnormal fibrinogens with a point mutation in the gamma chain, all characterized by impaired fibrin polymerization, we identified single base exchanges in the respective mutant gamma chain genes by polymerase chain reaction followed by DNA sequence analysis. These base exchanges accounted for the amino acid substitutions previously reported from our laboratory. They were exchanges of C to T (CGC for gamma Arg-275 to TGC for Cys) in fibrinogen Osaka II, T to G (AAT for gamma Asn-308 to AAG for Lys) in fibrinogen Kyoto I, T to C (ATG for gamma Met-310 to ACG for Thr) in fibrinogen Asahi, and G to T (GAT for gamma Asp-330 to TAT for Tyr) in fibrinogen Kyoto III. These base exchanges were found to reside in exon VIII of the gamma chain gene. Since many abnormal molecules are associated with polymerization defects, unless associated with the impaired release of fibrinopeptides A and/or B, exon VIII of the gamma chain gene may deserve careful study to define the structural alterations.


Assuntos
Fibrinogênio/genética , Fibrinogênios Anormais/genética , Mutação Puntual , Análise Mutacional de DNA , Humanos , Reação em Cadeia da Polimerase
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA