Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
1.
J Thromb Haemost ; 15(8): 1601-1606, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28581691

RESUMO

Essentials Prophylaxis is the standard of care for congenital factor XIII-A (FXIII-A) deficiency. Six children with FXIII-A deficiency received once-monthly prophylaxis with recombinant FXIII-A. Prophylaxis was well tolerated and no anti-FXIII antibodies were detected. Prophylaxis was effective with an annualized bleeding rate of zero. SUMMARY: Background Factor XIII deficiency is a rare, severe congenital bleeding disorder. Monthly prophylaxis with recombinant FXIII A-Subunit (rFXIII) has demonstrated favorable safety and efficacy in patients aged ≥ 6 years, and may similarly benefit younger children. Objective To evaluate the long-term safety and efficacy of rFXIII in children aged < 6 years with congenital FXIII A-subunit deficiency. Patients/methods Six children, who had previously completed a single-dose pharmacokinetic trial of rFXIII, received 35 IU kg-1 rFXIII every 28 days (± 2 days) for a minimum of 52 weeks, and were evaluated for bleeding and adverse events. The Berichrom FXIII activity assay was used to monitor FXIII activity. Results The children, three girls and three boys, had an average age of 3.0 years (range: 1-4 years) at enrollment. The total treatment duration was 1.8-3.5 years, giving a total of 16.6 patient-years. No antibody development, thromboembolic events or allergic reactions occurred. There were 93 mild and seven moderate adverse events. Two adverse events (lymphopenia and gastroenteritis) were reported as probably or possibly related to rFXIII in two children. Two serious adverse events, unrelated to rFXIII, were reported in a single child, each related to head injury, and neither resulting in intracranial hemorrhage. The geometric mean FXIII activity trough was 0.19 IU mL-1 . No bleeding episodes requiring treatment with an FXIII-containing hemostatic agent occurred during the trial; thus, the annualized bleeding rate was 0. Conclusions Consistent with data from older age groups, prophylaxis with rFXIII appears to be safe and effective in young children with congenital FXIII A-subunit deficiency.


Assuntos
Coagulantes/administração & dosagem , Deficiência do Fator XIII/tratamento farmacológico , Fator XIII/administração & dosagem , Fatores Etários , Pré-Escolar , Coagulantes/efeitos adversos , Esquema de Medicação , Fator XIII/efeitos adversos , Deficiência do Fator XIII/sangue , Deficiência do Fator XIII/diagnóstico , Deficiência do Fator XIII/genética , Fator XIIIa/genética , Feminino , Humanos , Lactente , Masculino , Proteínas Recombinantes/administração & dosagem , Fatores de Tempo , Resultado do Tratamento
3.
J Thromb Haemost ; 12(12): 2038-43, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25263390

RESUMO

BACKGROUND: The use of monthly recombinant factor XIII (rFXIII) recently demonstrated favorable safety and efficacy for congenital FXIII A-subunit deficiency patients aged ≥ 6 years (mentor(™) 1 trial), although the pharmacokinetics (PK) were not fully evaluated. OBJECTIVES: To comprehensively evaluate the steady-state PK of rFXIII in patients aged ≥ 6 years with congenital FXIII A-subunit deficiency. PATIENTS/METHODS: mentor(™) 2 is an ongoing, multinational safety and efficacy trial in which patients are receiving monthly rFXIII (35 IU kg(-1) ) for ≥ 52 weeks. For this 28-day PK analysis, blood samples were collected immediately predosing, and 1 h, 2 h, 3, 7, 14, 21, and 28 days postdosing. FXIII activity was measured and PK parameters were calculated using non-compartmental analysis, without prior baseline adjustment. Information regarding adverse events and bleeding was collected at each visit. Antibody assessments were performed predosing and at day 28. RESULTS: PK analysis in 23 patients revealed first-order elimination of rFXIII with a geometric mean half-life of 13.6 days. Mean FXIII activity was > 0.1 IU mL(-1) throughout the 28-day period, with a geometric mean peak activity of 0.87 IU mL(-1) and trough of 0.16 IU mL(-1) . The geometric mean clearance was 0.15 mL h(-1) kg(-1) . No bleeding episodes occurred during the PK session, and no anti-rFXIII antibodies were detected. Peak and trough FXIII activities were constant over time, compared with previous activities (≥ 10 rFXIII doses) in the same patients. CONCLUSIONS: Clearance of rFXIII is unaffected over time, and monthly prophylaxis with 35 IU kg(-1) rFXIII provides FXIII activity > 0.1 IU mL(-1) throughout the dosing interval in patients with congenital FXIII A-subunit deficiency.


Assuntos
Deficiência do Fator XIII/tratamento farmacológico , Fator XIIIa/farmacocinética , Proteínas Recombinantes/farmacocinética , Adolescente , Adulto , Idoso , Anticorpos/análise , Área Sob a Curva , Criança , Estudos de Coortes , Esquema de Medicação , Deficiência do Fator XIII/sangue , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Segurança do Paciente , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/química , Fatores de Tempo , Adulto Jovem
4.
J Wound Care ; 22(3): 144-6, 148, 150-2 passim, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23665733

RESUMO

OBJECTIVE: To compare the efficacy and safety of negatively-charged polystyrene microspheres (NCM)with controls (saline soaks) in the treatment of hard-to-heal wounds of various aetiologies. METHOD: Patients with one or more hard-to-heal wounds, defined as refractory to healing for at least 4 weeks, or those with exposed bone, tendon or ligament, were eligible for inclusion and were randomised to either NCM (PolyHeal; MediWound Ltd.) or controls, both applied twice daily for 4 weeks. Patients were monitored bi-weekly for an additional 8 weeks, while treated by standard wound care, at the investigators' discretion, and were re-evaluated 2 years after inclusion. The primary endpoint was defined as coverage of> 75% of the wound area by light-red granulation tissue after 4 weeks of treatment. RESULTS: Fifty-eight patients completed the study, 32 in the NCM group and 26 in the control group. The two most common wound types were those with primary etiologies of venous insufficiency and postoperative/post trauma. In the NCM group 47% of patients achieved > 75% light red granulation tissue after 4 weeks compared with 15% of patients in the control group (p=O.O I). The mean wound surface area in the NCM group was reduced by 39.0% after 4 weeks compared with 14.9% in the control group (p=0.02).The achievement of> 75% light red granulation tissue and reduction of mean wound surface area was also observed in the two main sub-groups (venous insufficiency and postoperative/post trauma), although it was not statistically significant, possibly due to the small sample size in each sub-group. CONCLUSION: This study demonstrates that compared to control treatment, NCM treatment of hard to-heal and chronic wounds improves formation of healthy granulation tissue and reduces wound size thus in fact 'kick-starting' the healing process and 'dechronifying' chronic wounds.


Assuntos
Ânions/uso terapêutico , Tecido de Granulação/crescimento & desenvolvimento , Microesferas , Úlcera Cutânea/terapia , Cicatrização , Adulto , Idoso , Doença Crônica , Método Duplo-Cego , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Poliestirenos , Estudos Prospectivos , Solução Salina Hipertônica , Eletricidade Estática , Resultado do Tratamento
5.
Haemophilia ; 14 Suppl 3: 202-10, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18510543

RESUMO

During the haemostatic response, the formation of a primary platelet plug limits bleeding and provides a surface for clotting factors to assemble and become activated. The initial platelet plug is stabilized by fibrin monomers, covalently cross-linked by FXIII, forming a platelets-fibrin thrombus. Defects in platelets as well as inherited deficiencies of coagulation factors including fibrinogen, FII, FV, FV + FVIII, FVII, FX, FXI and FXIII deficiencies, generally lead to lifelong bleeding disorders, whose severity of bleeding symptoms is heterogeneous in platelets abnormalities but generally inversely proportional to the degree of the factor deficiency in rare bleeding disorders (RBDs). The prevalence of platelet defects among the general population has not been established, whereas for RBDs it ranges from approximately 1 in 2 million to 1 in 500,000, being higher in countries where consanguineous marriages are diffused. As a consequence of the rarity of these deficiencies, the type and severity of bleeding symptoms, the underlying molecular defects, and the actual management of bleeding episodes are not well established. In this review the main features, diagnosis, available treatment options and treatment complications of the platelet disorders, caused by abnormalities in platelet receptors for adhesive proteins, platelet receptors for soluble agonists, platelet granules, signal transduction pathways, or procoagulant phospholipids will be discussed by Dr Cattaneo, whereas fibrinogen deficiency and FXIII deficiency will be described by Dr Inbal and Dr de Moerloose, respectively. Finally, the update of the Rare Bleeding Disorders Database will be presented by Dr Spreafico.


Assuntos
Transtornos Herdados da Coagulação Sanguínea/diagnóstico , Transtornos Plaquetários/diagnóstico , Transtornos Hemorrágicos/diagnóstico , Transtornos Herdados da Coagulação Sanguínea/genética , Transtornos Herdados da Coagulação Sanguínea/terapia , Inibidores dos Fatores de Coagulação Sanguínea/uso terapêutico , Transtornos Plaquetários/genética , Transtornos Plaquetários/terapia , Bases de Dados Genéticas , Esquema de Medicação , Genótipo , Transtornos Hemorrágicos/genética , Transtornos Hemorrágicos/terapia , Humanos , Sistemas Computadorizados de Registros Médicos , Fenótipo , Guias de Prática Clínica como Assunto , Reino Unido
7.
Br J Haematol ; 135(3): 348-51, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16995886

RESUMO

Hereditary thrombocythaemia (HT) is an inherited autosomal dominant disorder. Recent studies reported six different mutations, four within the thrombopoietin (TPO) gene and two within c-Mpl (TPO receptor) gene in six unrelated families with HT. This study investigated the molecular basis of hereditary thrombocythaemia in an Israeli-Jewish family. We screened the genes for TPO and c-Mpl by amplification and sequencing of all the corresponding exons including exon/intron boundaries and promoters. In addition, plasma levels of TPO and erythropoietin (EPO) were measured. No abnormality in the TPO/c-Mpl genes has been identified in affected HT family members. Plasma TPO and EPO levels were found to be normal/low or normal respectively in the individuals affected. In conclusion, lack of a molecular lesion within either TPO or cMpl genes indicate that HT may be caused by factors other than TPO-cMpl axis in this family.


Assuntos
Saúde da Família , Receptores de Trombopoetina/genética , Trombocitose/genética , Trombopoetina/genética , Adolescente , Adulto , Pré-Escolar , DNA/genética , Eritropoetina/sangue , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Linhagem , Análise de Sequência de DNA/métodos , Trombocitose/sangue , Trombopoetina/sangue
8.
J Thromb Haemost ; 4(1): 19-25, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16129024

RESUMO

Factor XIII is a plasma transglutaminase that participates in the final stage of the coagulation cascade. Thrombin-activated FXIII (FXIIIa) catalyzes the formation of covalent cross-links between gamma-glutamyl and epsilon-lysyl residues on adjacent fibrin chains in polymerized fibrin to yield the mature clot. In addition to its role in hemostasis, FXIII is known to participate in wound healing and embryo implantation, which are processes involving angiogenesis. In this review, we discuss the role of FXIII in angiogenesis and the molecular mechanisms underlying its proangiogenic effects. The FXIII role in tissue repair and remodeling may at least in part be attributed to its pro-angiogenic activity.


Assuntos
Fator XIII/fisiologia , Neovascularização Fisiológica/fisiologia , Animais , Implantação do Embrião , Homeostase , Humanos , Cicatrização
9.
J Thromb Haemost ; 2(10): 1790-7, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15456491

RESUMO

BACKGROUND: Hereditary factor (F)XIII deficiency is a rare bleeding disorder mostly due to mutations in FXIII A subunit. OBJECTIVES: We studied the molecular basis of FXIII deficiency in patients from 10 unrelated families originating from Israel, India and Tunisia. METHODS: Exons 2-15 of genomic DNA consisting of coding regions and intron/exon boundaries were amplified and sequenced. Structural analysis of the mutations was undertaken by computer modeling. RESULTS: Seven novel mutations were identified in the FXIIIA gene. The propositus from the Ethiopian-Jewish family was found to be a compound heterozygote for two novel mutations: a 10-bp deletion in exon 12 at nucleotides 1652-1661 (followed by 22 altered amino acids and termination codon) and Ala318Val mutation. The propositus of the Tunisian family was homozygous for C insertion after nucleotide 863 within a stretch of six cytosines of exon 7. This insertion results in generation of eight altered amino acids followed by a termination codon downstream. The propositus from Indian-Jewish origin was found to be homozygous for G to T substitution at IVS 11 [+1] resulting in skipping of exons 10 and 11. In addition to the Ala318Val mutation, three of the novel mutations identified are missense mutations: Arg260Leu, Thr398Asn and Gly210Arg each occurring in a homozygous state in an Israeli-Arab and two Indian families, respectively. CONCLUSIONS: Structure-function correlation analysis by computer modeling of the new missense mutations predicted that Gly210Arg will cause protein misfolding, Ala318Val and Thr398Asn will interfere with the catalytic process or protein stability, and Arg260Leu will impair dimerization.


Assuntos
Deficiência do Fator XIII/genética , Fator XIII/genética , Mutação , Catálise , Códon sem Sentido , Análise Mutacional de DNA , Dimerização , Éxons , Fator XIII/química , Saúde da Família , Humanos , Modelos Moleculares , Mutação de Sentido Incorreto , Dobramento de Proteína , Subunidades Proteicas/genética , Deleção de Sequência
10.
Bone Marrow Transplant ; 34(5): 459-63, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15247931

RESUMO

Antithymocyte globulin (ATG) is increasingly used in pre-allogeneic stem cell transplantation (allo-SCT) conditioning regimens to prevent graft rejection and graft-versus-host disease. However, ATG was also found to be associated with increased incidence of thrombosis during organ transplantation. In the present study, we tested the coagulation status of 21 patients with hematologic malignancies undergoing allo-SCT who received ATG-based (11 patients) or non-ATG-based (10) conditioning treatment. We assessed several thrombophilia markers as well as circulating total and endothelial microparticles (TMP/EMP) and soluble CD40 ligand (CD40L). No significant difference in the mean values of prothrombin time, partial thromboplastin time, fibrinogen, antithrombin, protein C, protein S, thrombin-antithrombin III complex, homocysteine levels, prevalence of genetic thrombophilia markers and levels of EMP, TMP or CD40L was observed between the ATG-treated and ATG-untreated patients, as well as before and after conditioning in each group separately. Platelet counts decreased significantly in ATG-treated patients; however, this decrease was not associated with clinical or laboratory evidence of disseminated intravascular coagulation. No patient developed thromboembolic event or veno-occlusive liver disease. Our results suggest that allo-SCT is not associated with increased hypercoagulability and addition of ATG to conditioning regimen has no significant procoagulant effect.


Assuntos
Soro Antilinfocitário/administração & dosagem , Coagulação Sanguínea , Neoplasias Hematológicas/terapia , Transplante de Células-Tronco Hematopoéticas , Imunossupressores/administração & dosagem , Condicionamento Pré-Transplante , Adulto , Idoso , Ligante de CD40/sangue , Feminino , Neoplasias Hematológicas/epidemiologia , Neoplasias Hematológicas/genética , Humanos , Incidência , Masculino , Metilenotetra-Hidrofolato Redutase (NADPH2)/genética , Pessoa de Meia-Idade , Polimorfismo Genético , Prevalência , Fatores de Risco , Trombose/sangue , Trombose/epidemiologia , Trombose/genética , Transplante Homólogo
11.
J Thromb Haemost ; 1(3): 433-8, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12871446

RESUMO

The effect of thromboprophylaxis with low molecular weight heparin (LMWH), on the subsequent live birth rate, in thrombophilic women with recurrent miscarriage has not been sufficiently assessed. The present study is a cohort study undertaken to assess the effect of enoxaparin on the subsequent live birth rate in women with hereditary thrombophila. Eighty-five patients with three or more consecutive pregnancy losses and a hereditary thrombophilia subsequently conceived. Thirty-seven were treated with daily subcutaneous injections of enoxaparin 40 mg and 48 were not treated. The outcome of the subsequent pregnancy was assessed in both groups of patients in terms of live births or repeat miscarriage. Forty-seven of the 85 patients were subsequently delivered, 38 have miscarried. Twenty-six of the 37 pregnancies in treated patients (70.2%) resulted in live births, compared with 21 of 48 (43.8%) in untreated patients (P < 0.02, OR 3.03, 95% CI 1.12-8.36). The beneficial effect was seen mainly in primary aborters, i.e. women with no previous live births (P < 0.008, OR 9.75, 95% CI 1.59-52.48). This benefit was also found in patients with a poor prognosis for a live birth (five or more miscarriages), where the live birth rate was increased from 18.2% to 61.6%. However, the benefit was not statistically significant, probably due to the small number of patients. If the beneficial effects of enoxaparin are confirmed by additional studies, thromboprophylaxis can be recommended for patients with hereditary thrombophilia and recurrent pregnancy loss.


Assuntos
Coeficiente de Natalidade , Trombofilia/complicações , Trombose/prevenção & controle , Aborto Habitual/prevenção & controle , Adulto , Enoxaparina/uso terapêutico , Saúde da Família , Feminino , Fibrinolíticos/uso terapêutico , Humanos , Cariotipagem , Gravidez , Complicações Hematológicas na Gravidez , Resultado da Gravidez , Trombofilia/tratamento farmacológico , Trombofilia/genética , Trombose/complicações
12.
Haemophilia ; 8(5): 622-8, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12199669

RESUMO

The present study was undertaken to evaluate stability, pharmacokinetic profile and efficacy of continuous infusion of 8Y in patients with different types of von Willebrand disease (vWD). Following reconstitution, 8Y levels of von Willebrand factor ristocetin cofactor (vWF:Rco), vWF antigen and factor VIII coagulant activity (FVIII:C) decreased to about 80% of the baseline levels; addition of low molecular weight heparin decreased the level of FVIII:C even further. Reconstituted 8Y was found to be sterile for up to 6 days postreconstitution. Ten vWD patients (four with type 2A, three with type 3, two with type 1 and one with 2N) underwent pharmacokinetic analysis. The recovery of vWF: RCo was significantly lower in patients with type 3 vWD (1.4 +/- 0.05% U(-1) kg(-1)) compared with that of the patients with types 1 (2.3 +/- 0.52% U(-1) kg(-1)) or 2A (2.0 +/- 0.06% U(-1) kg(-1)) vWD (P = 0.015). Type 3 vWD patients exhibited significantly higher vWF:RCo clearance (5.1 +/- 1.1 mL kg(-1) h(-1)) compared with that of patients with type 2A (2.8 +/- 0.7 mL kg(-1) h(-1)) and type 1 (2.6 +/- 1.0 mL kg(-1) h(-1)) vWD (P = 0.028). Accordingly, terminal half-life was lower in patients with type 3 vWD (8.0 +/- 0.6 h(-1)) compared with type 2A (12.7 +/- 5.9 h(-1)) or type 1 (14 +/- 1.2 h(-1)) vWD patients. Multimeric pattern of vWF from patients' plasma was similar to that of 8Y. In two patients treated with 8Y by continuous infusion for prevention or treatment of bleeding haemostasis was achieved. Thus, 8Y is suitable and haemostatically effective for continuous infusion treatment in patients with vWD.


Assuntos
Avaliação de Medicamentos/métodos , Fator VIII/administração & dosagem , Doenças de von Willebrand/terapia , Fator de von Willebrand/administração & dosagem , Adulto , Idoso , Perda Sanguínea Cirúrgica/prevenção & controle , Fator VIII/análise , Fator VIII/farmacocinética , Feminino , Humanos , Infusões Intravenosas , Masculino , Pessoa de Meia-Idade , Hemorragia Pós-Operatória/terapia , Hemorragia Pós-Parto/terapia , Gravidez , Complicações Hematológicas na Gravidez/sangue , Complicações Hematológicas na Gravidez/classificação , Complicações Hematológicas na Gravidez/terapia , Doenças de von Willebrand/sangue , Doenças de von Willebrand/classificação , Fator de von Willebrand/análise , Fator de von Willebrand/farmacocinética
13.
Thromb Res ; 105(4): 317-23, 2002 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-12031826

RESUMO

Coagulation factor XIII (FXIII) is a transglutaminase that catalyzes crosslink formation in fibrin clots. Endothelial cells (EC) were demonstrated to bind FXIII via their alpha(v)beta3 integrin receptor. FXIII was also shown to bind platelet glycoprotein IIb/IIIa receptor. In the present study, we analyzed if FXIII can mediate platelet-EC interaction. Both FXIII and activated FXIII (FXIIIa) bound to EC monolayers; this binding was enhanced by the addition of Mn2+ and was inhibited by the monoclonal antibody L609 against alpha(v)beta3 integrin. Normal washed platelets also bound surface-immobilized or soluble FXIII and FXIIIa, and the binding was GPIIb/IIIa dependent. The effect of FXIII concentrate (Fibrogammin-P) treatment on the interaction of ECs with platelets from six FXIII-deficient patients was studied. Patients' platelets were radiolabeled with 3H-Adenine, washed, resuspended in autologous plasma and allowed to adhere to immortalized EC line EAhy926. Adhesion of platelets from FXIII-deficient patients to ECs increased 1.7+/-0.4-fold (P=.01) following intravenous infusion of FXIII concentrate. Similarly, addition of 1 U/ml of FXIII concentrate to the patients' PRP in vitro increased the adhesion 1.8+/-0.5-fold (P=.008). Preincubation of the EC monolayers with increasing concentrations of either FXIII or FXIIIa augmented the adhesion of normal washed platelets to ECs in a dose-dependent manner. At 10 U/ml of EC-bound FXIII or FXIIIa, platelet adhesion enhanced 1.7+/-0.25-fold (P=.03) and 2.5+/-0.5-fold (P=.02), respectively. The increase in platelet adhesion was completely abolished by pretreatment of ECs with the anti-alpha(v)beta3 antibody L609 or by preincubation of the platelets with the GPIIb/IIIa inhibitor Abciximab. Taken together, our data indicate that FXIII mediates the interaction of platelets with ECs by bridging between endothelial alpha(v)beta3 and platelet GPIIb/IIIa integrins. This interaction may be relevant for tissue remodeling and wound repair after vascular injury in FXIII-deficient patients.


Assuntos
Deficiência do Fator XIII/sangue , Fator XIII/fisiologia , Integrina alfaVbeta3/sangue , Adesividade Plaquetária/fisiologia , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/fisiologia , Linhagem Celular , Endotélio Vascular/fisiologia , Fator XIII/farmacologia , Fator XIIIa/fisiologia , Humanos , Técnicas In Vitro , Adesividade Plaquetária/efeitos dos fármacos , Solubilidade
14.
Stroke ; 32(12): 2753-8, 2001 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-11739968

RESUMO

BACKGROUND AND PURPOSE: Although risk factors for carotid artery stenosis caused by atherosclerosis are known, it is unclear what triggers "activation" of the atherosclerotic plaques and the ensuing thromboembolic cerebral events. The aim of this study was to evaluate whether thrombophilic factors, platelet glycoprotein (GP) polymorphisms, and homocysteine are associated with a risk of ischemic events in patients with significant carotid stenosis. METHODS: Consecutive patients with >/=50% carotid stenosis, whether symptomatic (with ipsilateral ischemic events) or asymptomatic, who were evaluated and followed in a neurovascular clinic were tested for plasma levels of homocysteine, C677T mutation in methylenetetrahydrofolate reductase, G20210A mutation of factor II, factor V Leiden, antiphospholipid antibodies, and polymorphisms of platelet membrane GP: human platelet antigen (HPA)-1, GP Ia (C807T), and GP Ib (variable number of tandem repeats, Kozak, and HPA-2). RESULTS: Eighty-six asymptomatic and 67 symptomatic patients were evaluated. The former group was older (73.7+/-6.9 versus 69.5+/-9.1 years, P=0.02). Major risk factors for stroke were similar in both groups. In symptomatic patients versus asymptomatic patients, hyperhomocysteinemia was 3-fold more frequent (34.3% versus 12.8%, respectively; P=0.002) and HPA-1a/b was almost 2-fold more common (38.8% versus 20.9%, respectively; P=0.01). All other thrombophilic factors and platelet polymorphisms studied did not differ significantly between the 2 groups. Multivariate analysis revealed that hyperhomocysteinemia and the HPA-1a/b genotype conferred a significant risk of cerebral ischemic events, with odds ratios (95% CI) of 4.07 (1.7 to 9.7) and 3.4 (1.5 to 7.8), respectively. CONCLUSIONS: Hyperhomocysteinemia and HPA-1a/b are independent risk factors for ischemic events in patients with significant carotid stenosis.


Assuntos
Antígenos de Plaquetas Humanas/genética , Estenose das Carótidas/sangue , Hiper-Homocisteinemia/sangue , Polimorfismo Genético/genética , Acidente Vascular Cerebral/sangue , Idoso , Substituição de Aminoácidos/genética , Anticorpos Antifosfolipídeos/sangue , Estenose das Carótidas/diagnóstico , Estenose das Carótidas/epidemiologia , Comorbidade , Fator V/genética , Feminino , Homocisteína/sangue , Humanos , Hiper-Homocisteinemia/diagnóstico , Hiper-Homocisteinemia/epidemiologia , Integrina beta3 , Masculino , Metilenotetra-Hidrofolato Redutase (NADPH2) , Análise Multivariada , Razão de Chances , Oxirredutases atuantes sobre Doadores de Grupo CH-NH/genética , Glicoproteínas da Membrana de Plaquetas/genética , Protrombina/genética , Medição de Risco , Fatores de Risco , Acidente Vascular Cerebral/diagnóstico , Acidente Vascular Cerebral/epidemiologia
15.
Int J Oncol ; 19(5): 897-902, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11604985

RESUMO

BAT is a monoclonal antibody produced against membranes of Daudi cells that induces anti-tumor activity in mice against a variety of solid murine and human tumors, mediated by its immune stimulatory properties on murine and human lymphocytes. The present study analyzes the effect of BAT on leukemia/lymphoma using the BCL1 model of leukemia/lymphoma in BALB/C mice. BAT antibody binds to BCL1 leukemia cells and recognizes a 48 kDa protein similar to the antigen on Daudi cells. Mice inoculated with leukemia cells were treated either by direct BAT injections or by adoptive transfer of lymphocytes from BAT-injected mice. Administration of BAT monoclonal antibody was either once, on day 14, or daily on days 10-13 post tumor inoculation. A single injection of BAT resulted in reduction of peripheral blood tumor cells, however additional injections further decreased the tumor cell number reaching a 95-fold reduction on day 20 post tumor inoculation. Anti-tumor effect was also obtained when animals were injected with splenocytes from BAT-treated donor mice. A significant prolongation of survival of BAT-treated mice was observed although with no cure. The results of this study indicate that BAT might be used for reducing the tumor burden in leukemia for immunotherapy and in combination with other treatment modalities.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Linfoma de Burkitt/imunologia , Leucemia Experimental/tratamento farmacológico , Linfoma/tratamento farmacológico , Transferência Adotiva , Animais , Western Blotting , Relação Dose-Resposta a Droga , Feminino , Citometria de Fluxo , Humanos , Leucemia Experimental/mortalidade , Leucemia Experimental/patologia , Linfoma/mortalidade , Linfoma/patologia , Camundongos , Camundongos Endogâmicos BALB C , Peso Molecular , Proteínas de Neoplasias/imunologia , Baço/citologia , Baço/imunologia , Taxa de Sobrevida , Células Tumorais Cultivadas
16.
Development ; 128(18): 3405-13, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11566847

RESUMO

Homothorax (HTH) is a homeobox-containing protein, which plays multiple roles in the development of the embryo and the adult fly. HTH binds to the homeotic cofactor Extradenticle (EXD) and translocates it to the nucleus. Its function within the nucleus is less clear. It was shown, mainly by in vitro studies, that HTH can bind DNA as a part of ternary HTH/EXD/HOX complexes, but little is known about the transcription regulating function of HTH-containing complexes in the context of the developing fly. Here we present genetic evidence, from in vivo studies, for the transcriptional-activating function of HTH. The HTH protein was forced to act as a transcriptional repressor by fusing it to the Engrailed (EN) repression domain, or as a transcriptional activator, by fusing it to the VP16 activation domain, without perturbing its ability to translocate EXD to the nucleus. Expression of the repressing form of HTH in otherwise wild-type imaginal discs phenocopied hth loss of function. Thus, the repressing form was working as an antimorph, suggesting that normally HTH is required to activate the transcription of downstream target genes. This conclusion was further supported by the observation that the activating form of HTH caused typical hth gain-of-function phenotypes and could rescue hth loss-of-function phenotypes. Similar results were obtained with XMeis3, the Xenopus homologue of HTH, extending the known functional similarity between the two proteins. Competition experiments demonstrated that the repressing forms of HTH or XMeis3 worked as true antimorphs competing with the transcriptional activity of the native form of HTH. We also describe the phenotypic consequences of HTH antimorph activity in derivatives of the wing, labial and genital discs. Some of the described phenotypes, for example, a proboscis-to-leg transformation, were not previously associated with alterations in HTH activity. Observing the ability of HTH antimorphs to interfere with different developmental pathways may direct us to new targets of HTH. The HTH antimorph described in this work presents a new means by which the transcriptional activity of the endogenous HTH protein can be blocked in an inducible fashion in any desired cells or tissues without interfering with nuclear localization of EXD.


Assuntos
Drosophila melanogaster/genética , Proteínas de Homeodomínio/genética , Fatores de Transcrição/genética , Ativação Transcricional , Proteínas de Xenopus , Sequência de Aminoácidos , Animais , Animais Geneticamente Modificados , Núcleo Celular/metabolismo , Sequência Conservada , Proteínas de Drosophila , Drosophila melanogaster/embriologia , Evolução Molecular , Extremidades/embriologia , Proteína Vmw65 do Vírus do Herpes Simples/genética , Proteína Vmw65 do Vírus do Herpes Simples/metabolismo , Proteínas de Homeodomínio/metabolismo , Fenótipo , Transporte Proteico , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Repressoras , Fatores de Transcrição/metabolismo
17.
Thromb Haemost ; 85(5): 806-9, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11372672

RESUMO

Acquired von Willebrand syndrome (AVWS) has been associated mainly with monoclonal gammopathy of uncertain significance (MGUS), clonal lymphoproliferative or myeloproliferative disorders and autoimmunity. In the present work we studied 6 patients with AVWS: four with MGUS IgG (lambda or kappa), one with small lymphocytic lymphoma and one with agnogenic myeloid metaplasia (AMM). All the patients underwent a pharmacokinetic analysis at presentation in order to study potential differences in recovery, clearance (CL) or terminal half-life (THL) following administration of von Willebrand factor (VWF) concentrate. In all the patients with AVWS an increase in clearance and a decrease in THL was observed as compared to these parameters in patients with hereditary type 3 von Willebrand disease (VWD). No difference in recovery was observed among the groups. The increase in clearance and the decrease in THL were significantly more pronounced in the group of MGUS patients (57.93 +/- 25.6 ml/h/kg, and 1.39 +/- 0.5 h, respectively) as compared to these parameters in the AMM (8.06 ml/h/kg, and 6.96 h, respectively) or the lymphoma (4.76 ml/h/kg, and 6.76 h. respectively) patients (p = 0.03 for clearance and 0.001 for THL). These data indicate that the pharmacokinetic analysis can be a useful tool to distinguish between MGUS-related and other causes of AVWS, and to plan an appropriate treatment accordingly.


Assuntos
Fator VIII/farmacocinética , Doenças de von Willebrand/diagnóstico , Fator de von Willebrand/farmacocinética , Idoso , Tempo de Sangramento , Fator VIII/administração & dosagem , Feminino , Meia-Vida , Humanos , Leucemia Linfocítica Crônica de Células B/complicações , Masculino , Taxa de Depuração Metabólica , Pessoa de Meia-Idade , Gamopatia Monoclonal de Significância Indeterminada/complicações , Mielofibrose Primária/complicações , Ristocetina , Doenças de von Willebrand/tratamento farmacológico , Doenças de von Willebrand/etiologia , Fator de von Willebrand/administração & dosagem
18.
Hematol J ; 2(1): 38-41, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11920232

RESUMO

Hyperhomocysteinemia is a defined risk factor for venous thromboembolism (VTE). Several polymorphisms of genes encoding for enzymes acting in the remethylation pathway of homocysteine metabolism, ie, methionine synthase (MS) A2756G, methylenetetrahydrofolate reductase (MTHFR) C677T and MTHFR A1298C, can cause increased homocysteine levels particularly in patients with deficiencies of folic acid, vitamin B6, or B12 and hence be potential risk factors for VTE. Indeed, homozygous MTHFR C677T was shown to be a mild risk factor for VTE by some, but not by all, investigators. In this study, we assessed the risk exerted by MS A2756G and MTHFR A1298C in a cohort of patients with idiopathic venous thromboembolism. Homozygosities for MS A2756G and MTHFR A1298C were not found to be statistically significant risk factors for VTE. In addition, no interactions were observed among MS A2756G, MTHFR A1298C and MTHFR C677T in conferring a risk of VTE.


Assuntos
5-Metiltetra-Hidrofolato-Homocisteína S-Metiltransferase/genética , Oxirredutases atuantes sobre Doadores de Grupo CH-NH/genética , Polimorfismo Genético , Tromboembolia/genética , Trombose Venosa/genética , Estudos de Casos e Controles , Fator V/genética , Testes Genéticos , Homozigoto , Humanos , Metilenotetra-Hidrofolato Redutase (NADPH2) , Mutação Puntual , Protrombina/genética , Fatores de Risco , Tromboembolia/etiologia , Trombose Venosa/etiologia
19.
Thromb Haemost ; 84(5): 912-7, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11127876

RESUMO

Antiplatelet drugs are the mainstays of therapy for acute and chronic cardiovascular diseases. S-nitroso-AR545C - an S-nitrosoderivative of a recombinant von Willebrand factor fragment AR545C spanning Ala 444 to Asp 730 and containing an Arg 545 Cys mutation, was previously found to inhibit ristocetin- and ADP-induced platelet aggregation and the interaction of platelets with extracellular matrix (ECM). In the current study we tested the antithrombotic properties of S-nitroso-AR545C on guinea pig platelets and in a platelet-rich thrombosis model in the guinea pig. Preincubation of guinea pig platelets with 0.1 microM of S-nitroso-AR545C decreased ristocetin-induced agglutination by 40% (p = 0.009) and completely abolished ADP-induced aggregation (p <0.0001). At concentration of 1.0 microM, S-nitroso-AR545C completely inhibited platelet adhesion (represented by surface coverage - SC) and decreased aggregate formation (represented by average aggregate size - AS) by more than 50%. Treatment of guinea pigs with 1.0 mg/kg S-nitroso-AR545C resulted in a significantly delayed time to arterial occlusion (31.7+/-6.0 min vs. 13.9+/-3.2 min, p <0.02). Similarly, total patency time was longer in the group injected with S-nitroso-AR545C compared to the control group. However, the difference was not statistically significant (33.8+/-6.3 min vs. 20.2+/-3.3 min, p = 0.07). No change in platelet count, hematocrit and bleeding time was observed 60 min after injection compared to baseline. In contrast, a significant decrease in SC (p <0.0001) and AS (p <0.01) were observed 60 min after the injection of S-nitroso-AR545C, whereas no change in these parameters was observed in the control group. These observations indicate that S-nitroso-AR545C exhibits significant antiadhesive and antiaggregating effects in-vitro and inhibits clot formation in-vivo suggesting that this compound may have potential therapeutic advantages.


Assuntos
Trombose das Artérias Carótidas/tratamento farmacológico , Fragmentos de Peptídeos/farmacologia , Inibidores da Agregação Plaquetária/farmacologia , Fator de von Willebrand/farmacologia , Animais , Modelos Animais de Doenças , Cobaias , Fragmentos de Peptídeos/uso terapêutico , Inibidores da Agregação Plaquetária/uso terapêutico , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Fator de von Willebrand/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...