Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Thromb Haemost ; 118(3): 451-460, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29448295

RESUMO

Recombinant factor XIII-A2 (rFXIII-A2) was developed for prophylaxis and treatment of bleeds in patients with congenital FXIII A-subunit deficiency. mentor™2 (NCT00978380), a multinational, open-label, single-arm, multiple-dosing extension to the pivotal mentor™1 trial, assessed long-term safety and efficacy of rFXIII-A2 prophylaxis in eligible patients (patients with severe [<0.05 IU/mL] congenital FXIII subunit A deficiency) aged ≥6 years. Patients received 35 IU/kg rFXIII-A2 (exact dosing) every 28 ± 2 days for ≥52 weeks. Primary endpoint was safety (adverse events including immunogenicity); secondary endpoints were rate of bleeds requiring FXIII treatment, haemostatic response after one 35 IU/kg rFXIII-A2 dose for breakthrough bleeds and withdrawals due to lack of rFXIII-A2 efficacy. Steady-state pharmacokinetic variables were also summarized. Elective surgery was permitted during the treatment period. Sixty patients were exposed to rFXIII-A2; their median age was 26.0 years (range: 7.0-77.0). rFXIII-A2 was well tolerated without any safety concerns. No non-neutralizing or neutralizing antibodies (inhibitors) against FXIII were detected. Mean annualized bleeding rate (ABR) was 0.043/patient-year. Mean spontaneous ABR was 0.011/patient-year. No patients withdrew due to lack of efficacy. Geometric mean FXIII trough level was 0.17 IU/mL. Geometric terminal half-life was 13.7 days. rFXIII-A2 prophylaxis provided sufficient haemostatic coverage for 12 minor surgeries without the need for additional FXIII therapy; eight procedures were performed within 7 days of the patient's last scheduled rFXIII-A2 dose, and four were performed 10 to 21 days after the last dose.


Assuntos
Fator VIIIa/uso terapêutico , Deficiência do Fator XIII/terapia , Hemorragia/prevenção & controle , Procedimentos Cirúrgicos Operatórios , Adolescente , Adulto , Idoso , Criança , Deficiência do Fator XIII/congênito , Deficiência do Fator XIII/cirurgia , Feminino , Hemostasia , Humanos , Masculino , Pessoa de Meia-Idade , Segurança do Paciente , Proteínas Recombinantes/uso terapêutico , Adulto Jovem
2.
Acta Haematol ; 137(4): 183-190, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28419992

RESUMO

BACKGROUND: Refractoriness to platelet transfusion, prevalent among 15-20% of hemato-oncological patients, is associated with multitransfusions and inferior outcomes. We evaluated the effectiveness of extended slow-dose transfusion (ESDT) in increasing platelet increments in multitransfused patients. METHODS: Patients treated after the implementation of ESDT were compared with historical controls treated with standard single-donor platelet (SDP) transfusions. Cohorts of early and late recipients were assembled for comparison, i.e. the 8th or 9th and 11th platelet unit per patient, respectively. Patients in the ESDT group received transfusions equal to half an SDP unit, administered over 4 h. Effectiveness was defined as a higher corrected count increment (CCI) at 1, 12, and 24 h after transfusion. RESULTS: In the early-recipients cohort, 24-h-posttransfusion increments were available for 29 ESDT patients and 6 standard patients, and did not differ significantly between the groups (p = 0.078). The 24-h-posttransfusion increment was available for 20 ESDT patients and 7 standard patients in the late-recipients cohort. The CCI was significantly higher in the ESDT group (p = 0.042). ABO compatibility improved the CCI (p = 0.01). CONCLUSIONS: ESDT demonstrated slightly higher increments at 24 h after transfusion in late recipients, suggesting this could be a cost-effective approach for the treatment of thrombocytopenic multitransfused hemato-oncological patients.


Assuntos
Neoplasias Hematológicas/terapia , Transfusão de Plaquetas/métodos , Adulto , Idoso , Estudos de Coortes , Feminino , Neoplasias Hematológicas/sangue , Neoplasias Hematológicas/complicações , Humanos , Leucemia/sangue , Leucemia/complicações , Leucemia/terapia , Linfoma/sangue , Linfoma/complicações , Linfoma/terapia , Masculino , Pessoa de Meia-Idade , Contagem de Plaquetas , Transfusão de Plaquetas/efeitos adversos , Fatores de Risco , Trombocitopenia/sangue , Trombocitopenia/etiologia , Trombocitopenia/terapia , Fatores de Tempo , Resultado do Tratamento , Adulto Jovem
3.
Leuk Lymphoma ; 58(6): 1455-1467, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-27733071

RESUMO

Tyrosine kinase inhibitors (TKIs) have revolutionized the prognosis of chronic myeloid leukemia. With the advent of highly efficacious therapy, the focus has shifted toward managing TKI adverse effects, such as vascular adverse events (VAEs). We used an in vitro angiogenesis model to investigate the TKI-associated VAEs. Our data show that imatinib, nilotinib, and ponatinib reduce human umbilical vein endothelial cells (HUVECs) viability. Pharmacological concentrations of ponatinib induced apoptosis, reduced migration, inhibited tube formation of HUVECs, and had a negative effect on endothelial progenitor cell (EPC) function. Furthermore, in HUVECs transfected with VEGF receptor 2 (VEGFR2), the effect of ponatinib on tube formation and on all parameters representing normal endothelial cell function was less prominent than in control cells. This is the first report regarding the pathogenesis of ponatinib-associated VAEs. The antiangiogenic effect of ponatinib, possibly mediated by VEGFR2 inhibition, as shown in our study, is another piece in the intricate puzzle of TKI-associated VAEs.


Assuntos
Antineoplásicos/farmacologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Imidazóis/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Piridazinas/farmacologia , Antineoplásicos/efeitos adversos , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Biomarcadores , Movimento Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Ensaio de Unidades Formadoras de Colônias , Expressão Gênica , Células Endoteliais da Veia Umbilical Humana , Humanos , Imidazóis/efeitos adversos , Imidazóis/uso terapêutico , Imunofenotipagem , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Fenótipo , Inibidores de Proteínas Quinases/efeitos adversos , Inibidores de Proteínas Quinases/uso terapêutico , Piridazinas/efeitos adversos , Piridazinas/uso terapêutico , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
5.
Semin Thromb Hemost ; 43(1): 59-68, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27556350

RESUMO

Congenital factor XIII (FXIII) deficiency is a rare, autosomal recessive bleeding disorder with potentially life-threatening consequences. FXIII is composed of two subunits (A and B), and a deficiency or dysfunction of either can result in FXIII deficiency. Traditionally, FXIII deficiency has been managed by infusing plasma-derived products containing FXIII (fresh frozen plasma, cryoprecipitate, and plasma-derived FXIII concentrates), all of which contain both subunits. Despite the increased safety of plasma-derived products, concern remains regarding potential viral safety issues. This review describes the development, from concept to clinical use, of a recombinant FXIII molecule (containing subunit A only; rFXIII-A2) for congenital FXIII-A subunit deficiency. Unmet needs and ongoing challenges in congenital FXIII deficiency are also discussed. Despite the challenges in developing a product for a very rare bleeding disorder, the information gathered on efficacy, safety, and pharmacokinetics of FXIII replacement therapy represents the largest dataset on congenital FXIII-A subunit deficiency in the world. It also provides evidence for the safety and efficacy of monthly prophylaxis with 35 IU/kg of rFXIII-A2 in patients with FXIII-A subunit deficiency. The issues encountered and overcome, along with lessons learned, may be applied to and encourage the development of new recombinant products for other rare bleeding disorders.


Assuntos
Deficiência do Fator XIII/tratamento farmacológico , Fator XIII/uso terapêutico , Proteínas Recombinantes/uso terapêutico , Humanos
6.
Thromb Res ; 136(1): 118-24, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25496996

RESUMO

The aim of the study was to further investigate the role of fibrinogen-like protein 2 (FGL-2), a transmembrane prothrombinase that directly cleaves prothrombin to thrombin, in angiogenesis and tumor development and the mechanism(s) underlying these processes. To study angiogenesis HUVEC clones with decreased fgl-2 mRNA were generated by specific siRNA. To study tumorigenesis SCID mice were implanted with intact (wild type) and fgl-2-silenced PC-3 clones. IFN-γ treated HUVEC expressing increased fgl-2 mRNA exhibited significant capillary sprouting that was not inhibited by hirudin, whereas fgl-2 silencing completely inhibited blood-vessel formation. Tumors (poorly differentiated carcinoma) developed in all 12 mice injected with wild type PC-3 compared with 8/12 mice injected with the fgl-2-silenced PC-3 clone. The tumors developed by fgl-2-silenced PC-3 clones were smaller and less aggressive and contained significantly fewer blood vessels (p<0.05). All tumors' sections were negative for thrombin staining, indicating that FGL-2-induced tumorigenesis was not mediated by thrombin. In fgl-2-silenced tumors there was a decrease in fgl-2 mRNA (p=0.02) and ERK1/2 phosphorylation (p<0.05) by 80% and a 20%, respectively. The mechanism underlying these processes, studied in PC-3 clones, revealed that fgl-2 silencing was associated with a 65% decrease in FGF-2 mRNA (p<0.01) and a 30% down regulation of ERK1/2 phosphorylation (p<0.05). Together, these results suggest that FGL-2 mediates angiogenesis and tumorigenesis not by thrombin-mediated mechanism but rather through FGF-2/ERK signaling pathway. FGL-2 may serve as a valuable therapeutic target in the future.


Assuntos
Carcinogênese/metabolismo , Fibrinogênio/metabolismo , Sistema de Sinalização das MAP Quinases , Neovascularização Patológica/metabolismo , Neoplasias da Próstata/metabolismo , Tromboplastina/metabolismo , Animais , Carcinogênese/genética , Carcinogênese/patologia , Linhagem Celular Tumoral , Proliferação de Células , Fibrinogênio/genética , Células Endoteliais da Veia Umbilical Humana , Humanos , Masculino , Camundongos SCID , Neovascularização Patológica/genética , Neovascularização Patológica/patologia , Próstata/metabolismo , Próstata/patologia , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Interferência de RNA , RNA Interferente Pequeno/genética , Trombina/metabolismo , Tromboplastina/genética
7.
PLoS One ; 9(10): e109648, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25303152

RESUMO

Fibrinogen-like protein 2, FGL-2, was reported to be overexpressed in various cancer tissues, where it acts as a transmembrane prothrombinase. This study aims to determine the prothrombinase activity of FGL-2 in peripheral blood mononuclear cells (PBMC) of patients with B-cell lymphoma. FGL-2 activity was determined in patients with B-cell lymphoma (n = 53), and healthy controls (n = 145). FGL-2 activity in patients at diagnosis increased 3 ± 0.3 fold (p < 0.001). Sensitivity and specificity of the test was established at 73.6% and 80.7%, respectively, using a cutoff of 150% activity over control. Moreover, FGL-2 activity in 10 of 11 patients in remission decreased by 76%. In contrast, no significant difference was observed in expression levels of fgl-2 gene in patients and controls. Taken together, our study indicates that FGL-2 prothrombinase activity in PBMC of lymphoma patients is increased in active disease and normalizes during remission, thus being a potential marker for follow up of lymphoma patients.


Assuntos
Fibrinogênio/metabolismo , Leucócitos Mononucleares/metabolismo , Linfoma de Células B/metabolismo , Tromboplastina/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores/metabolismo , Membrana Celular/metabolismo , Feminino , Humanos , Linfoma de Células B/patologia , Masculino , Pessoa de Meia-Idade
8.
Isr Med Assoc J ; 16(5): 307-10, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24979837

RESUMO

BACKGROUND: A 75 year old patient presenting with mucocutaneous bleeding was diagnosed with acquired thrombasthenia. The diagnosis was based on lack of platelet aggregation with adenosine diphosphate (ADP), arachidonic acid and collagen, and normal aggregation induced by ristocetin. OBJECTIVE: To study the mechanism of platelet function inhibition in a patient with acquired thrombasthenia. METHODS: Aggregation assays of platelets from the patient and healthy controls were performed. In addition, anti-glycoprotein (GP) IIbIIIa antibodies bindingto normal in the presence or absence of the patient's serum was by flow cytometry. RESULTS: Aggregation of normal platelets in the presence of patient's plasma was inhibited four- and 2.5-fold in the presence of ADP and arachidonic acid respectively, while collagen-induced aggregation was completely abolished. Ristocetin-induced aggregation was normal. The patient's serum inhibited binding of commercial anti-glycoprotein IIbIIIa antibodies to normal platelets twofold by flow cytometry. Treatment with anti-CD20 monoclonal antibody (rituximab) normalized the patient's platelet aggregation. CONCLUSIONS: These results suggest that the patient developed inhibitory anti-GPIIbIIIa autoantibodies that caused acquired thrombasthenia.


Assuntos
Autoanticorpos/análise , Transtornos Plaquetários , Agregação Plaquetária , Testes de Função Plaquetária/métodos , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/imunologia , Difosfato de Adenosina , Idoso , Anticorpos Monoclonais Murinos/administração & dosagem , Ácido Araquidônico , Transtornos Plaquetários/diagnóstico , Transtornos Plaquetários/etiologia , Transtornos Plaquetários/imunologia , Colágeno , Monitoramento de Medicamentos/métodos , Feminino , Humanos , Fatores Imunológicos/administração & dosagem , Lúpus Eritematoso Sistêmico/complicações , Lúpus Eritematoso Sistêmico/imunologia , Agregação Plaquetária/efeitos dos fármacos , Agregação Plaquetária/imunologia , Indução de Remissão , Ristocetina , Rituximab , Resultado do Tratamento
10.
Thromb Res ; 131(4): 338-41, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23276530

RESUMO

BACKGROUND: Factor XIII (FXIII), a plasma pro-transglutaminase, consists of two A subunits and two B subunits (FXIIIA2B2). Following activation by thrombin, it cross-links fibrin chains at the final step of coagulation. We previously reported that FXIII subunit A (FXIIIA) serves as a protein disulfide isomerase (PDI), and that PDI promotes platelet adhesion and aggregation. OBJECTIVE: This study sought to examine possible mechanistic effect of FXIII on platelet adhesion to fibrinogen; specifically, the role of its PDI activity. METHODS: Ex vivo experiments: Blood platelets derived from five patients with hereditary FXIIIA deficiency before and after treatment with Fibrogammin-P (FXIIIA2B2 concentrate) were washed and incubated on immobilized fibrinogen. Bound platelets were stained and counted by microscopy. In vitro experiments: Platelets derived from patients before treatment and five healthy controls were washed and analyzed for adhesion in the presence or absence of Fibrogammin-P or recombinant FXIII (FXIIIA2 concentrate). RESULTS: In ex vivo experiments, one hour after Fibrogammin-P treatment, mean (±SEM) platelet adhesion to fibrinogen increased by 27±2.32% (p<0.001). In in vitro experiments, treatment with Fibrogammin-P or recombinant FXIII (10IU/mL each) enhanced platelet adhesion to fibrinogen (in patients, by 29.95±6.7% and 29.05±5.3%, respectively; in controls, by 26.06±3.24% and 26.91±4.72, respectively; p<0.04 for all). Iodoacetamide-treated FXIII (I-FXIII), where transglutaminase activity is blocked, showed similar enhanced adhesion as untreated FXIII. By contrast, addition of an antibody that specifically blocks FXIIIA-PDI activity inhibited FXIII-mediated platelet adhesion to fibrinogen by 65%. CONCLUSION: These findings indicate that FXIII-induced enhancement of platelet adhesion is mediated by FXIII-PDI activity.


Assuntos
Plaquetas/efeitos dos fármacos , Fator XIIIa/farmacologia , Fibrinogênio/metabolismo , Adesividade Plaquetária/efeitos dos fármacos , Isomerases de Dissulfetos de Proteínas/metabolismo , Plaquetas/enzimologia , Plaquetas/metabolismo , Estudos de Casos e Controles , Deficiência do Fator XIII/sangue , Deficiência do Fator XIII/tratamento farmacológico , Fibrinogênio/química , Humanos , Proteínas Imobilizadas/química , Proteínas Imobilizadas/metabolismo , Proteínas Recombinantes/farmacologia
11.
PLoS One ; 7(2): e31393, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22384017

RESUMO

The histone deacetylase inhibitor (HDACI) butyroyloxymethyl diethylphosphate (AN-7) synergizes the cytotoxic effect of doxorubicin (Dox) and anti-HER2 on mammary carcinoma cells while protecting normal cells against their insults. This study investigated the concomitant changes occurring in heart tissue and tumors of mice bearing a subcutaneous 4T1 mammary tumor following treatment with AN-7, Dox, or their combination. Dox or AN-7 alone led to inhibition of both tumor growth and lung metastases, whereas their combination significantly increased their anticancer efficacy and attenuated Dox- toxicity. Molecular analysis revealed that treatment with Dox, AN-7, and to a greater degree, AN-7 together with Dox increased tumor levels of γH2AX, the marker for DNA double-strand breaks and decreased the expression of Rad51, a protein needed for DNA repair. These events culminated in increased apoptosis, manifested by the appearance of cytochrome-c in the cytosol. In the myocardium, Dox-induced cardiomyopathy was associated with an increase in γH2AX expression and a reduction in Rad51 and MRE11 expression and increased apoptosis. The addition of AN-7 to the Dox treatment protected the heart from Dox insults as was manifested by a decrease in γH2AX levels, an increase in Rad51 and MRE11 expression, and a diminution of cytochrome-c release. Tumor fibrosis was high in untreated mice but diminished in Dox- and AN-7-treated mice and was almost abrogated in AN-7+Dox-treated mice. By contrast, in the myocardium, Dox alone induced a dramatic increase in fibrosis, and AN7+Dox attenuated it. The high expression levels of c-Kit, Ki-67, c-Myc, lo-FGF, and VEGF in 4T1 tumors were significantly reduced by Dox or AN-7 and further attenuated by AN-7+Dox. In the myocardium, Dox suppressed these markers, whereas AN-7+Dox restored their expression. In conclusion, the combination of AN-7 and Dox results in two beneficial effects, improved anticancer efficacy and cardioprotection.


Assuntos
Butiratos/farmacologia , Inibidores de Histona Desacetilases/farmacologia , Neoplasias Mamárias Animais/metabolismo , Compostos Organofosforados/farmacologia , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Quebras de DNA de Cadeia Dupla , Doxorrubicina/farmacologia , Feminino , Marcadores Genéticos/genética , Histonas/metabolismo , Humanos , Recém-Nascido , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/secundário , Neoplasias Mamárias Animais/tratamento farmacológico , Camundongos , Camundongos Endogâmicos BALB C , Modelos Genéticos , Metástase Neoplásica , Ratos , Espécies Reativas de Oxigênio
12.
Blood ; 119(22): 5111-7, 2012 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-22451421

RESUMO

Congenital factor XIII (FXIII) deficiency is a rare, autosomal-recessive disorder, with most patients having an A-subunit (FXIII-A) deficiency. Patients experience life-threatening bleeds, impaired wound healing, and spontaneous abortions. In many countries, only plasma or cryoprecipitate treatments are available, but these carry a risk for allergic reactions and infection with blood-borne pathogens. The present study was a multinational, open-label, single-arm, phase 3 prophylaxis trial evaluating the efficacy and safety of a novel recombinant FXIII (rFXIII) in congenital FXIII-A subunit deficiency. Forty-one patients ≥ 6 years of age (mean, 26.4; range, 7-60) with congenital FXIII-A subunit deficiency were enrolled. Throughout the rFXIII prophylaxis, only 5 bleeding episodes (all trauma induced) in 4 patients were treated with FXIII-containing products. The crude mean bleeding rate was significantly lower than the historic bleeding rate (0.138 vs 2.91 bleeds/patient/year, respectively) for on-demand treatment. Transient, non-neutralizing, low-titer anti-rFXIII Abs developed in 4 patients, none of whom experienced allergic reactions, any bleeds requiring treatment, or changes in FXIII pharmacokinetics during the trial or follow-up. These non-neutralizing Abs declined below detection limits in all 4 patients despite further exposure to rFXIII or other FXIII-containing products. We conclude that rFXIII is safe and effective in preventing bleeding episodes in patients with congenital FXIII-A subunit deficiency. This study is registered at http://www..clinicaltrials.gov as number NCT00713648.


Assuntos
Fator VIII/administração & dosagem , Hemofilia A/tratamento farmacológico , Hemorragia/tratamento farmacológico , Adolescente , Adulto , Anticorpos/sangue , Criança , Hipersensibilidade a Drogas/etiologia , Fator VIII/efeitos adversos , Feminino , Hemofilia A/sangue , Hemorragia/sangue , Humanos , Masculino , Pessoa de Meia-Idade , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/efeitos adversos , Fatores de Risco , Segurança
13.
Invest New Drugs ; 30(1): 130-43, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20862515

RESUMO

The histone deacetylase inhibitor (HDACI) butyroyloxymethyl diethylphosphate (AN-7) has been shown to synergize doxorubicin (Dox) anticancer activity while attenuating its cardiotoxicity. In this study we further explored the selectivity of AN-7's action in several cancer and normal cells treated with anticancer agents. The cells studied were murine mammary 4T1, human breast T47D and glioblastoma U251 cancer cell lines, neonatal rat cardiomyocytes, cardiofibroblasts and astrocytes, and immortalized cardiomyocyte H9C2 cells. Cell death, ROS production and changes in protein expression were measured and in vivo effects were evaluated in Balb-c mice. AN-7 synergized Dox and anti-HER2 cytotoxicity against mammary carcinoma cells with combination indices of 0.74 and 0.79, respectively, while it protected cardiomyocytes against their toxicity. Additionally AN-7 protected astrocytes from Dox-cytoxicity. Cell-type specific changes in the expression of proteins controlling survival, angiogenesis and inflammation by AN-7 or AN-7+Dox were observed. In mice, the protective effect of AN-7 against Dox cardiotoxicity was associated with a reduction in inflammatory factors. In summary, AN-7 augmented the anticancer activity of Dox and anti-HER2 and attenuated their toxicity against normal cells. AN-7 modulation of c-Myc, thrombospondin-1, lo-FGF-2 and other proteins were cell type specific. The effects of AN-7, Dox and their combination were preserved in vivo indicating the potential benefit of combining AN-7 and Dox for clinical use.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Astrócitos/efeitos dos fármacos , Neoplasias Encefálicas/patologia , Neoplasias da Mama/patologia , Fibroblastos/efeitos dos fármacos , Glioblastoma/patologia , Miócitos Cardíacos/efeitos dos fármacos , Proteínas Angiogênicas/metabolismo , Animais , Anticorpos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/toxicidade , Astrócitos/patologia , Neoplasias Encefálicas/enzimologia , Neoplasias da Mama/enzimologia , Neoplasias da Mama/imunologia , Butiratos/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Citoproteção , Relação Dose-Resposta a Droga , Doxorrubicina/farmacologia , Sinergismo Farmacológico , Feminino , Fibroblastos/patologia , Glioblastoma/enzimologia , Inibidores de Histona Desacetilases/farmacologia , Humanos , Mediadores da Inflamação/metabolismo , Concentração Inibidora 50 , Camundongos , Camundongos Endogâmicos BALB C , Miócitos Cardíacos/patologia , Compostos Organofosforados/farmacologia , Ratos , Espécies Reativas de Oxigênio/metabolismo , Receptor ErbB-2/imunologia , Fatores de Tempo
14.
Isr Med Assoc J ; 13(7): 398-401, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21838180

RESUMO

Acquired thrombotic thrombocytopenic purpura (TTP) is an uncommon disease in adults, characterized by fever, neurological manifestations, microangiopathic hemolytic anemia, thrombocytopenia, renal dysfunction, and the presence of antibodies against the enzyme ADAMTS13. Treatment with plasmapheresis has increased the survival from 10% to more than 90%. Still, there is a subset of patients with resistant TTP who fail to respond to plasmapheresis or remain dependent on this procedure. There is mounting evidence that rituximab may play an important role in remission induction of resistant/relapsing TTP, but the extent of the remission is unknown. We present here four patients with chronic-relapsing TTP who responded favorably to rituximab. All four patients achieved prolonged remission of 23 to 82 months after the treatment. One patient relapsed 6 years afterthe initial treatment with rituximab and re-entered remission following retreatment.


Assuntos
Anticorpos Monoclonais Murinos/uso terapêutico , Fatores Imunológicos/uso terapêutico , Adulto , Antígenos CD20 , Feminino , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Púrpura Trombocitopênica Trombótica/tratamento farmacológico , Recidiva , Indução de Remissão , Estudos Retrospectivos , Rituximab , Fatores de Tempo , Adulto Jovem
15.
Harefuah ; 149(5): 283-7, 336, 2010 May.
Artigo em Hebraico | MEDLINE | ID: mdl-20929067

RESUMO

Von Willebrand disease (VWD) is the most common hereditary bleeding disorder, characterized by skin and mucosaL bleeding in most of the cases. In the Western world, the approximate number of bleeding VWD patients is 6.9 to 17 million. The disease is represented by several types with specific treatment for each type. Diagnosis can be troublesome and, therefore, n order to prevent bleeding complications, a professional clinical/laboratory team is required.


Assuntos
Doenças de von Willebrand/diagnóstico , Doenças de von Willebrand/terapia , Hemorragia/etiologia , Hemorragia/prevenção & controle , Humanos , Incidência , Equipe de Assistência ao Paciente , Doenças de von Willebrand/classificação , Doenças de von Willebrand/epidemiologia
16.
Thromb Haemost ; 104(4): 709-17, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20664907

RESUMO

Factor XIII subunit A (FXIII-A) is one of the most overrepresented genes that is expressed during the alternative activation of macrophages. Based on its substrate profile and its cellular localisation, FXIII-A is thought to function as an intracellular/intranuclear transglutaminase. Our aim was to find role for the intracellular FXIII-A by comparing the microarray profiles of alternatively activated monocyte-derived macrophages. Microarray analyses of FXIII-A-deficient patients and healthy controls were evaluated, followed by functional clustering of the differentially expressed genes. After a 48-hour differentiation in the presence of interleukin 4 (IL4), 1,017 probes out of the 24,398 expressed in macrophages from FXIII-A- deficient samples were IL4 sensitive, while only 596 probes were IL4 sensitive in wild-type samples. Of these genes, 307 were induced in both the deficient and the wild-type macrophages. Our results revealed that FXIII-A has important role(s) in mediating gene expression changes in macrophages during alternative activation. Functional clustering of the target genes carried out using Cytoscape/BiNGO and Ingenuity Pathways Analysis programs showed that, in the absence of FXIII-A, the most prominent differences are related to immune functions and to wound response. Our findings suggest that functional impairment of macrophages at the level of gene expression regulation plays a role in the wound healing defects of FXIII-A-deficient patients.


Assuntos
Deficiência do Fator XIII/genética , Deficiência do Fator XIII/imunologia , Fator XIIIa/metabolismo , Ativação de Macrófagos , Macrófagos/metabolismo , Diferenciação Celular , Linhagem da Célula , Células Cultivadas , Simulação por Computador , Deficiência do Fator XIII/metabolismo , Fator XIIIa/genética , Fator XIIIa/imunologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/imunologia , Redes Reguladoras de Genes/imunologia , Humanos , Interleucina-4/imunologia , Interleucina-4/metabolismo , Ativação de Macrófagos/genética , Macrófagos/imunologia , Macrófagos/patologia , Análise em Microsséries , Cicatrização/genética
18.
Thromb Haemost ; 101(5): 840-4, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19404536

RESUMO

Tissue transglutaminase was reported to act as protein disulfide isomerase (PDI). We studied whether plasma transglutaminase - coagulation factor XIII (FXIII) - has PDI activity as well. PDI activity was measured by determining the ability to renature reduced-denatured RNase (rdRNase). We found that FXIII can renature rdRNase, with efficiency comparable to commercial PDI. This PDI activity was inhibited by bacitracin. Like tissue transglutaminase, FXIII-mediated PDI activity is independent of its transglutaminase activity and is located on the A subunit. Surface-associated PDI has been previously shown to catalyse two distinct functions: transnitrosation with subsequent release of intracellular nitric oxide and disulfide bond rearrangement during platelet integrin ligation. Our results imply that FXIII-PDI activity may have a role in platelet function.


Assuntos
Fator XIII/metabolismo , Fator XIIIa/metabolismo , Isomerases de Dissulfetos de Proteínas/sangue , Ribonuclease Pancreático/metabolismo , Animais , Anticorpos , Bacitracina/farmacologia , Bovinos , Inibidores Enzimáticos/farmacologia , Fator XIII/antagonistas & inibidores , Fator XIII/química , Fator XIII/imunologia , Fator XIIIa/antagonistas & inibidores , Fator XIIIa/química , Fator XIIIa/imunologia , Humanos , Isomerases de Dissulfetos de Proteínas/antagonistas & inibidores , Isomerases de Dissulfetos de Proteínas/química , Isomerases de Dissulfetos de Proteínas/imunologia , Renaturação Proteica , Subunidades Proteicas
19.
Clin Exp Metastasis ; 25(7): 703-16, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18506586

RESUMO

Histone deacetylase inhibitory prodrugs that are metabolized to butyric acid and formaldehyde possess antineoplastic properties and low toxicity. We sought to characterize the antiangiogenic and antimetastatic activities of two lead prodrugs, pivaloyloxymethyl butyrate (AN-9) and butyroyloxymethyl-diethyl phosphate (AN-7) in murine cancer models. In the sc implanted human colon carcinoma HT-29 xenograft model AN-7, exhibited superior anticancer activity compared to AN-9, as was evident by the significantly greater inhibition of tumor growth and reduction of serum CEA. AN-7 was also more effective in reducing mean vessel density (MVD) by 7-fold, bFGF, Ki-67 (7-fold) and HIF-1alpha in immunohistochemically stained tumor sections. Semi-quantitative evaluation of the levels of bFGF, HDAC1 and HIF-1alpha by Western blot analysis showed a decrease in expression only in the tumors of mice treated with AN-7. The level of bFGF was reduced 3-fold in the tumor and that of TIMP1 was elevated (by 3-fold) in the serum of AN-7 treated mice. In a 4T1 metastatic breast carcinoma model, AN-7 inhibited the formation of lung lesions by 76% and AN-9 by 47%, further demonstrating the greater efficacy of AN-7 compared to AN-9 (P<0.02). Both AN-7 and AN-9 exhibited antimetastatic and antiangiogenic activities by reducing vascularization, bFGF expression and HIF-1alpha. Yet, AN-7 was more potent than AN-9.


Assuntos
Inibidores da Angiogênese/farmacologia , Antineoplásicos/farmacologia , Butiratos/farmacologia , Inibidores Enzimáticos/farmacologia , Inibidores de Histona Desacetilases , Metástase Neoplásica/prevenção & controle , Compostos Organofosforados/farmacologia , Animais , Antígenos CD34/análise , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/antagonistas & inibidores , Camundongos , PTEN Fosfo-Hidrolase/análise , Inibidor Tecidual de Metaloproteinase-1/análise
20.
Cell Physiol Biochem ; 19(1-4): 113-20, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17310105

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 crosslinks between gamma-glutamyl and epsilon-lysyl residues on fibrin molecules to yield the mature clot. In addition to its role in hemostasis, FXIIIa was previously shown by us to stimulate endothelial cells to exhibit pro-angiogenic activity. In this work, we studied the effect of FXIIIa on other cells that participate in angiogenesis and tissue repair, such as monocytes and fibroblasts. FXIIIa significantly enhanced migration and proliferation, and inhibited apoptosis of monocytes and fibroblasts. Similar to our previous observations with endothelial cells, the stimulating effect of FXIIIa on monocytes and fibroblasts was elicited via its binding to alpha (v)beta (3) integrin leading to cJun upregulation and TSP-1 downregulation. Since monocytes and fibroblasts are essential components of the tissue repair process, the results of this study, together with the proangiogenic activity of FXIIIa, further substantiate a significant role of FXIII in tissue repair.


Assuntos
Fator XIII/farmacologia , Fibroblastos/efeitos dos fármacos , Monócitos/efeitos dos fármacos , Apoptose , Movimento Celular , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Fibroblastos/fisiologia , Humanos , Monócitos/fisiologia , Proteínas Proto-Oncogênicas c-jun/metabolismo , Trombospondina 1/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...