Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Blood Adv ; 5(5): 1388-1402, 2021 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-33661300

RESUMEN

Sickle cell disease (SCD) is associated with hemolysis, vascular inflammation, and organ damage. Affected patients experience chronic painful vaso-occlusive events requiring hospitalization. Hypoxia-induced polymerization of sickle hemoglobin S (HbS) contributes to sickling of red blood cells (RBCs) and disease pathophysiology. Dilution of HbS with nonsickling hemoglobin or hemoglobin with increased oxygen affinity, such as fetal hemoglobin or HbS bound to aromatic aldehydes, is clinically beneficial in decreasing polymerization. We investigated a novel alternate approach to modify HbS and decrease polymerization by inhibiting methionine aminopeptidase 2 (MetAP2), which cleaves the initiator methionine (iMet) from Val1 of α-globin and ßS-globin. Kinetic studies with MetAP2 show that ßS-globin is a fivefold better substrate than α-globin. Knockdown of MetAP2 in human umbilical cord blood-derived erythroid progenitor 2 cells shows more extensive modification of α-globin than ß-globin, consistent with kinetic data. Treatment of human erythroid cells in vitro or Townes SCD mice in vivo with selective MetAP2 inhibitors extensively modifies both globins with N-terminal iMet and acetylated iMet. HbS modification by MetAP2 inhibition increases oxygen affinity, as measured by decreased oxygen tension at which hemoglobin is 50% saturated. Acetyl-iMet modification on ßS-globin delays HbS polymerization under hypoxia. MetAP2 inhibitor-treated Townes mice reach 50% total HbS modification, significantly increasing the affinity of RBCs for oxygen, increasing whole blood single-cell RBC oxygen saturation, and decreasing fractional flow velocity losses in blood rheology under decreased oxygen pressures. Crystal structures of modified HbS variants show stabilization of the nonpolymerizing high O2-affinity R2 state, explaining modified HbS antisickling activity. Further study of MetAP2 inhibition as a potential therapeutic target for SCD is warranted.


Asunto(s)
Anemia de Células Falciformes , Hemoglobina Falciforme , Aminopeptidasas , Anemia de Células Falciformes/tratamiento farmacológico , Animales , Antidrepanocíticos/farmacología , Humanos , Cinética , Metaloendopeptidasas , Metionil Aminopeptidasas , Ratones , Polimerizacion
4.
JCI Insight ; 2(20)2017 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-29046485

RESUMEN

Sickle cell disease (SCD) results from a point mutation in the ß-globin gene forming hemoglobin S (HbS), which polymerizes in deoxygenated erythrocytes, triggering recurrent painful vaso-occlusive crises and chronic hemolytic anemia. Reactivation of fetal Hb (HbF) expression ameliorates these symptoms of SCD. Nuclear factor (erythroid derived-2)-like 2 (Nrf2) is a transcription factor that triggers cytoprotective and antioxidant pathways to limit oxidative damage and inflammation and increases HbF synthesis in CD34+ stem cell-derived erythroid progenitors. We investigated the ability of dimethyl fumarate (DMF), a small-molecule Nrf2 agonist, to activate γ-globin transcription and enhance HbF in tissue culture and in murine and primate models. DMF recruited Nrf2 to the γ-globin promoters and the locus control region of the ß-globin locus in erythroleukemia cells, elevated HbF in SCD donor-derived erythroid progenitors, and reduced hypoxia-induced sickling. Chronic DMF administration in SCD mice induced HbF and increased Nrf2-dependent genes to detoxify heme and limit inflammation. This improved hematological parameters, reduced plasma-free Hb, and attenuated inflammatory markers. Chronic DMF administration to nonanemic primates increased γ-globin mRNA in BM and HbF protein in rbc. DMF represents a potential therapy for SCD to induce HbF and augment vasoprotection and heme detoxification.


Asunto(s)
Anemia de Células Falciformes/tratamiento farmacológico , Anemia/tratamiento farmacológico , Dimetilfumarato/metabolismo , Dimetilfumarato/farmacología , Hemoglobina Fetal/metabolismo , Hemo/metabolismo , Animales , Antioxidantes/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Humanos , Inflamación , Leucemia Eritroblástica Aguda/metabolismo , Ratones , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , ARN Mensajero/metabolismo , Bazo/metabolismo , gamma-Globinas/genética
5.
Br J Haematol ; 174(6): 970-82, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27291690

RESUMEN

Very Late Antigen-4 (VLA-4, α4ß1-integrin, ITGA4) orchestrates cell-cell and cell-endothelium adhesion. Given the proposed role of VLA-4 in sickle cell disease (SCD) pathophysiology, we evaluated the ability of the VLA-4 blocking antibody natalizumab to inhibit SCD blood cell adhesion. Natalizumab recognized surface VLA-4 on leucocytes and reticulocytes in whole blood from SCD subjects. SCD reticulocytes were positive for VLA-4, while VLA-4 staining of non-SCD reticulocytes was undetectable. Titrations with natalizumab revealed the presence of saturable levels of VLA-4 on both SCD reticulocytes and leucocytes similar to healthy subject leucocytes. Under physiological flow conditions, the adhesion of SCD whole blood cells and isolated SCD leucocytes to immobilized vascular cell adhesion molecule 1 (VCAM-1) was blocked by natalizumab in a dose-dependent manner, which correlated with cell surface receptor binding. Natalizumab also inhibited >50% of whole blood cell binding to TNF-α activated human umbilical vein endothelial cell monolayers under physiological flow at clinically relevant concentrations (10 to 100 µg/ml). This indicates that VLA-4 is the dominant receptor that drives SCD reticulocyte and mononuclear cell adhesion to VCAM-1 and that the VLA-4 adhesion to VCAM-1 is a significant contributor to SCD blood cell adhesion to endothelium. Thus, VLA-4 blockade may be beneficial in sickle cell disease.


Asunto(s)
Anemia de Células Falciformes/sangre , Adhesión Celular/efectos de los fármacos , Integrina alfa4beta1/antagonistas & inhibidores , Leucocitos/efectos de los fármacos , Leucocitos/metabolismo , Natalizumab/farmacología , Reticulocitos/efectos de los fármacos , Reticulocitos/patología , Adolescente , Adulto , Anemia de Células Falciformes/diagnóstico , Anemia de Células Falciformes/tratamiento farmacológico , Biomarcadores , Membrana Celular/metabolismo , Niño , Preescolar , Simulación por Computador , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Femenino , Citometría de Flujo , Hemodinámica , Humanos , Inmunoglobulina G/química , Inmunoglobulina G/metabolismo , Inmunoglobulina G/farmacología , Lactante , Masculino , Persona de Mediana Edad , Natalizumab/química , Natalizumab/metabolismo , Unión Proteica , Multimerización de Proteína , Reticulocitos/metabolismo , Molécula 1 de Adhesión Celular Vascular/metabolismo , Adulto Joven
6.
Thromb Res ; 136(6): 1266-72, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26514955

RESUMEN

INTRODUCTION: Recombinant factor VIII Fc fusion protein (rFVIIIFc) is a novel recombinant factor VIII with a prolonged half-life, developed for the treatment of hemophilia A. Studies that evaluated the toxicological effects of rFVIIIFc in 2 pharmacologically relevant species, cynomolgus monkeys and Sprague Dawley rats, are reported here. MATERIALS AND METHODS: In repeat-dose toxicology studies, rats and monkeys received 0, 50, 250, or 1000 IU/kg rFVIIIFc every other day for 4 weeks. In a high-dose tolerance study, monkeys received 1 rFVIIIFc dose of 3000, 10,000, or 20,000 IU/kg. Evaluations included in-life observations, laboratory and post-mortem evaluations, pharmacokinetics, and local tolerance. Allometric scaling, using data from 4 animal species and humans, was used to evaluate the relationship between animal and human pharmacokinetics. RESULTS: rFVIIIFc was well tolerated with no adverse toxicological findings directly attributable to rFVIIIFc. As expected, antibodies to this fully human protein developed in rats and monkeys in a time-dependent fashion following repeated dosing, leading to increased clearance in both species. There were no local reactions (infusion site) or evidence of thrombosis at high doses in rats and monkeys. Allometric scaling demonstrated more rapid clearance in small animals compared with humans and a volume of distribution (steady state) proportional to body weight across species, suggesting that animal pharmacokinetics are predictive of human pharmacokinetics. CONCLUSIONS: Repeated doses of rFVIIIFc in 2 relevant animal species and high doses of rFVIIIFc in monkeys were well tolerated. These results supported the clinical safety of rFVIIIFc observed in phase 1/2a and phase 3 clinical trials.


Asunto(s)
Factor VIII/farmacocinética , Fragmentos Fc de Inmunoglobulinas/química , Proteínas Recombinantes/farmacocinética , Animales , Área Bajo la Curva , Factor VIII/toxicidad , Femenino , Células HEK293 , Hemofilia A/inmunología , Humanos , Fragmentos Fc de Inmunoglobulinas/toxicidad , Macaca fascicularis , Masculino , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes/toxicidad , Especificidad de la Especie , Trombosis/inmunología
7.
PLoS One ; 10(4): e0124930, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25905473

RESUMEN

We recently developed a longer lasting recombinant factor VIII-Fc fusion protein, rFVIIIFc, to extend the half-life of replacement FVIII for the treatment of people with hemophilia A. In order to elucidate the biological mechanism for the elongated half-life of rFVIIIFc at a cellular level we delineated the roles of VWF and the tissue-specific expression of the neonatal Fc receptor (FcRn) in the biodistribution, clearance and cycling of rFVIIIFc. We find the tissue biodistribution is similar for rFVIIIFc and rFVIII and that liver is the major clearance organ for both molecules. VWF reduces the clearance and the initial liver uptake of rFVIIIFc. Pharmacokinetic studies in FcRn chimeric mice show that FcRn expressed in somatic cells (hepatocytes or liver sinusoidal endothelial cells) mediates the decreased clearance of rFVIIIFc, but FcRn in hematopoietic cells (Kupffer cells) does not affect clearance. Immunohistochemical studies show that when rFVIII or rFVIIIFc is in dynamic equilibrium binding with VWF, they mostly co localize with VWF in Kupffer cells and macrophages, confirming a major role for liver macrophages in the internalization and clearance of the VWF-FVIII complex. In the absence of VWF a clear difference in cellular localization of VWF-free rFVIII and rFVIIIFc is observed and neither molecule is detected in Kupffer cells. Instead, rFVIII is observed in hepatocytes, indicating that free rFVIII is cleared by hepatocytes, while rFVIIIFc is observed as a diffuse liver sinusoidal staining, suggesting recycling of free-rFVIIIFc out of hepatocytes. These studies reveal two parallel linked clearance pathways, with a dominant pathway in which both rFVIIIFc and rFVIII complexed with VWF are cleared mainly by Kupffer cells without FcRn cycling. In contrast, the free fraction of rFVIII or rFVIIIFc unbound by VWF enters hepatocytes, where FcRn reduces the degradation and clearance of rFVIIIFc relative to rFVIII by cycling rFVIIIFc back to the liver sinusoid and into circulation, enabling the elongated half-life of rFVIIIFc.


Asunto(s)
Factor VIII/metabolismo , Hepatocitos/metabolismo , Antígenos de Histocompatibilidad Clase I/fisiología , Receptores Fc/fisiología , Factor de von Willebrand/metabolismo , Animales , Ratones , Ratones Noqueados , Receptores Depuradores/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Distribución Tisular
8.
Thromb Res ; 136(2): 371-8, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25840744

RESUMEN

INTRODUCTION: Recombinant factor IX Fc fusion protein (rFIXFc) is a recombinant coagulation factor composed of a single molecule of recombinant factor IX (rFIX) covalently fused to the Fc domain of human immunoglobulin G1 (IgG1) with no intervening sequence. An extensive nonclinical program was performed to support the clinical development of rFIXFc for treatment of people with hemophilia B. MATERIALS AND METHODS: Repeat-dose toxicology studies of rFIXFc were performed in 2 relevant species: Sprague Dawley rats (4-week study) and cynomolgus monkeys (5- and 27-week studies). Assessments included in-life observations, electrocardiograms (monkeys only), laboratory evaluations (including hematology and blood chemistry), postmortem analyses, local tolerance, and pharmacokinetics (PK). Allometric scaling was performed with PK data from multiple species, including humans. Local tolerance (single-dose study) and thrombogenic potential (Wessler stasis model) of rFIXFc were tested in New Zealand White rabbits. RESULTS: There were no significant local or systemic toxicity findings in the repeat-dose studies. Allometric scaling data suggested that animal rFIXFc PK results are predictive of human PK parameters. There were no findings from the local tolerance study in rabbits; thrombogenic activity was less than that elicited by rFIX and a prothrombin complex concentrate, and similar to vehicle control. CONCLUSIONS: rFIXFc was well tolerated in toxicology studies and demonstrated a low thrombogenic potential. These results are consistent with phase 1/2a and phase 3 clinical studies of rFIXFc in people with hemophilia B.


Asunto(s)
Factor IX/farmacocinética , Factor IX/toxicidad , Fragmentos Fc de Inmunoglobulinas/toxicidad , Proteínas Recombinantes de Fusión/farmacocinética , Proteínas Recombinantes de Fusión/toxicidad , Trombosis/inducido químicamente , Animales , Antitrombinas/farmacocinética , Antitrombinas/toxicidad , Relación Dosis-Respuesta a Droga , Tolerancia a Medicamentos , Macaca fascicularis , Dosis Máxima Tolerada , Tasa de Depuración Metabólica , Conejos , Ratas , Ratas Sprague-Dawley , Especificidad de la Especie , Trombosis/prevención & control
9.
J Nucl Med ; 50(3): 435-43, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19223400

RESUMEN

UNLABELLED: To optimize in vivo tissue uptake kinetics and clearance of engineered monoclonal antibody (mAb) fragments for radiotherapeutic and radiodiagnostic applications, we compared the biodistribution and tumor localization of four (111)In- and (86)Y-labeled antibody formats, derived from a single antimindin/RG-1 mAb, in a prostate tumor model. The IgG, diabody, single-chain variable domain (scFv), and novel miniantibody formats, composed of the human IgE-C(H)4 and a modified IgG1 hinge linked to scFv domains, were compared. METHODS: Antibodies were first derivatized with the bifunctional chelator CHX-A''-diethylenetriamine pentaacetic acid and then bound to the radiometal to create radiolabeled immunoconjugates. Human LNCaP xenografts were grown in nude mice, and (111)In- or (86)Y-labeled antibodies were administered intravenously. Tissues were harvested at different times, and the level of antibody deposition was determined by measuring radioactivity. Whole-body small-animal PET of mice receiving (86)Y-labeled antibodies was performed at 6 time points and colocalized with simultaneous micro-CT imaging. RESULTS: The biodistributions of (111)In and (86)Y antibodies were quite similar. The blood, tumor, kidney, and liver tissues contained varying levels of radioactivity. The antibody accumulation in the tumor correlated with molecular size. The IgG steadily increased with time to 24.1 percentage injected dose per gram (%ID/g) at 48 h. The miniantibody accumulated at a similar rate to reach a lower level (14.2 %ID/g) at 48 h but with a higher tumor-to-blood ratio than the IgG. Tumor accumulation of the diabody peaked at 3 h, reaching a much lower level (3.7 %ID/g). A combination of rapid clearance and lower relative affinity of the scFv precluded deposition in the tumor. Small-animal PET results correlated well with the biodistribution results, with similar tumor localization patterns. CONCLUSION: The larger antibody formats (IgG and miniantibody) gave higher tumor uptake levels than did the smaller formats (diabody and scFv). These larger formats may be more suitable for radioimmunotherapy applications, evidenced by the preclinical efficacy previously shown by a report on the IgG format. The smaller formats were rapidly cleared from circulation, and the diabody, which accumulated in the tumor, may be more suitable for radiodiagnostic applications.


Asunto(s)
Anticuerpos Monoclonales/farmacocinética , Neoplasias de la Próstata/metabolismo , Radiofármacos/farmacocinética , Animales , Anticuerpos Monoclonales/química , Línea Celular Tumoral , Quelantes/química , Humanos , Radioisótopos de Indio , Isotiocianatos/química , Masculino , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Ácido Pentético/análogos & derivados , Ácido Pentético/química , Tomografía de Emisión de Positrones , Radiofármacos/química , Distribución Tisular , Tomografía Computarizada por Rayos X , Trasplante Heterólogo , Radioisótopos de Itrio
10.
J Biomol Screen ; 12(4): 536-45, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17384330

RESUMEN

Leukotriene A4 (LTA4) hydrolase catalyzes a rate-limiting final biosynthetic step of leukotriene B4 (LTB4), a potent lipid chemotactic agent and proinflammatory mediator. LTB4 has been implicated in the pathogenesis of various acute and chronic inflammatory diseases, and thus LTA4 hydrolase is regarded as an attractive therapeutic target for anti-inflammation. To facilitate identification and optimization of LTA4 hydrolase inhibitors, a specific and efficient assay to quantify LTB4 is essential. This article describes the development of a novel 384-well homogeneous time-resolved fluorescence assay for LTB4 (LTB4 HTRF assay) and its application to establish an HTRF-based LTA4 hydrolase assay for lead optimization. This LTB4 HTRF assay is based on competitive inhibition and was established by optimizing the reagent concentration, buffer composition, incubation time, and assay miniaturization. The optimized assay is sensitive, selective, and robust, with a Z' factor of 0.89 and a subnanomolar detection limit for LTB4. By coupling this LTB4 HTRF assay to the LTA4 hydrolase reaction, an HTRF-based LTA4 hydrolase assay was established and validated. Using a test set of 16 LTA4 hydrolase inhibitors, a good correlation was found between the IC50 values obtained using LTB4 HTRF with those determined using the LTB enzyme-linked immunoassay (R = 0.84). The HTRF-based LTA4 hydrolase assay was shown to be an efficient and suitable assay for determining compound potency and library screening to guide the development of potent inhibitors of LTA4 hydrolase.


Asunto(s)
Epóxido Hidrolasas/metabolismo , Leucotrieno B4/metabolismo , Animales , Anticuerpos Monoclonales/metabolismo , Unión Competitiva , Activación Enzimática , Epóxido Hidrolasas/química , Fluoroinmunoensayo , Humanos , Leucotrieno B4/química , Leucotrieno B4/inmunología , Ratones , Unión Proteica
11.
Thromb Haemost ; 97(1): 45-53, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17200770

RESUMEN

We have discovered a novel small-molecule (3-phosphinoylpropionic acid) inhibitor of activated thrombin activatable fibrinolysis inhibitor (TAFIa), BX 528, which had an IC (50) of 2 nM in an enzymatic assay and 50 nM in an in-vitro clot lysis assay, with 3,500- to 35,000-fold selectivity against other carboxypeptidases, such as CPN, CPZ and CPD, and 5- and 12-fold selectivity against CPE (CPH) and CPB, respectively. At 10 micro M, BX 528 had no significant activity (<50% inhibition or antagonism) in a panel of 137 enzymes and receptors. It had no effects on blood coagulation and platelet aggregation up to 300 and 10 micro M, respectively. The plasma half-life following intravenous administration was 0.85 hours in rats and 4.5 hours in dogs. No significant metabolism was detected in human, dog or rabbit hepatic microsomes, and no significant inhibition of cytochrome P450 3A4 and 2D6 up to 30 micro M. No cytotoxic or cell proliferative effects were found in three hepatic and renal cell lines up to 300 micro M and no mutagenic activity was seen in the Ames II screen. There were no significant hemodynamic effects in rats and dogs up to 100 and 30 mg/kg with peak plasma drug concentrations of approximately 1,000 and 300 micro M, respectively. In an in-vivo complement activation model in guinea pigs, BX 528 showed minimal inhibition of plasma CPN activity up to 60 mg/kg with peak plasma concentrations up to 250 micro M. Thus, these data demonstrate that BX 528 is a novel, potent, selective and safe TAFIa inhibitor.


Asunto(s)
Carboxipeptidasa B2/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacocinética , Animales , Coagulación Sanguínea/efectos de los fármacos , Carboxipeptidasas/antagonistas & inhibidores , Proliferación Celular/efectos de los fármacos , Perros , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Cobayas , Semivida , Humanos , Concentración 50 Inhibidora , Microsomas Hepáticos/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Propionatos/farmacocinética , Propionatos/farmacología , Conejos , Ratas , Especificidad por Sustrato
12.
J Biomol Screen ; 11(8): 985-95, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17092910

RESUMEN

A therapeutic antibody candidate (AT-19) isolated using multivalent phage display binds native tomoregulin (TR) as a mul-timer not as a monomer. This report raises the importance of screening and selecting phage antibodies on native antigen and reemphasizes the possibility that potentially valuable antibodies are discarded when a monomeric phage display system is used for screening. A detailed live cell panning selection and screening method to isolate multivalently active antibodies is described. AT-19 is a fully human antibody recognizing the cell surface protein TR, a proposed prostate cancer target for therapeutic antibody internalization. AT-19 was isolated from a multivalent single-chain variable fragment (scFv) antibody library rescued with hyperphage. The required multivalency for isolation of AT-19 is supported by fluorescence activated cell sorting data demonstrating binding of the multivalent AT-19 phage particles at high phage concentrations and failure of monovalent particles to bind. Pure monomeric scFv AT-19 does not bind native receptor on cells, whereas dimeric scFv or immunoglobulin G binds with nanomolar affinity. The isolation of AT-19 antibody with obligate bivalent binding activity to native TR is attributed to the use of a multivalent display of scFv on phage and the method for selecting and screening by alternate use of 2 recombinant cell lines.


Asunto(s)
Antígenos de Superficie/inmunología , Bacteriófagos/inmunología , Región Variable de Inmunoglobulina/inmunología , Proteínas de la Membrana/farmacología , Proteínas de Neoplasias/farmacología , Biblioteca de Péptidos , Animales , Anticuerpos , Bacteriófagos/genética , Células CHO , Línea Celular , Cricetinae , Cricetulus , Ensayo de Inmunoadsorción Enzimática/métodos , Humanos , Inmunoglobulina G/metabolismo , Análisis de Secuencia de Proteína , Transfección , Carga Viral
13.
Thromb Haemost ; 96(3): 317-24, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16953273

RESUMEN

Tissue factor (TF) exposure is a potent pro-thrombotic trigger that initiates activation of the coagulation cascade, while thrombomodulin (TM) is a potent anticoagulant protein that limits the extent of activation. Both TF neutralizing antibodies and soluble TM (sTM) are effective anticoagulants. We have developed a novel anticoagulant fusion protein, Ab(TF)-TM, by fusing a TFneutralizing single-chain antibody, Ab(TF), to an active fragment of TM. Ab(TF)-TM is a novel anticoagulant targeting to sites of TF exposure with a dual mechanism of action. The Ab(TF) portion of the molecule inhibitsTF/factorVIIa mediated activation of FIX and FX, and the TM portion of the molecule acts as a cofactor for activation of protein C. In-vitro coagulation assays show that Ab(TF)-TM more potently inhibits TF-initiated coagulation (prothrombin time) than can its individual components, Ab(TF) (20-fold) and sTM (80-fold) alone, or in combination (10-fold). In contrast, the potency of Ab(TF)-TM in the activated partial thromboplastin and thrombin clotting time assays was similar to sTM alone. In a rat model of disseminated intravascular coagulation (DIC), intravenous injection of a human TF-containing thromboplastin reagent (0.5 ml/kg) resulted in an immediate death in approximately 60% of the animals and a clinical score of approximately 2.5. Pre-injection of Ab(TF)-TM or Ab(TF) and sTM, given alone or in combination, showed dose-dependent efficacy. At a dose of 0.7 nmol/kg, Ab(TF)-TM completely prevented death and reduced clinical scores by 79%, while neitherAb(TF) nor sTM, given alone or in combination, showed significant therapeutic effects. Calculated effective doses that reduced mortality by 50% relative to that in the control group (ED(50), nmol/kg) were 0.21 for Ab(TF)-TM, 3.2 for an equimolar mixture of Ab(TF) and sTM, 4.3 for sTM and 20 for Ab(TF). Thus, Ab(TF)-TM presented 10- to 100-fold enhancement of the anticoagulant potency, relative to the ED(50) in Ab(TF) and sTM given either alone or in combination, in a rat DIC model.


Asunto(s)
Anticoagulantes/farmacología , Trombomodulina/sangre , Tromboplastina/metabolismo , Animales , Anticoagulantes/química , Coagulación Sanguínea , Dimerización , Relación Dosis-Respuesta a Droga , Humanos , Masculino , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Tromboembolia , Trombomodulina/química , Tromboplastina/inmunología , Factores de Tiempo
14.
Neoplasia ; 8(1): 18-30, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16533422

RESUMEN

The EphA2 receptor tyrosine kinase has been shown to be over-expressed in cancer and a monoclonal antibody (mAb) that activates and down-modulates EphA2 was reported to inhibit the growth of human breast and lung tumor xenografts in nude mice. Reduction of EphA2 levels by treatment with anti-EphA2 siRNA also inhibited tumor growth, suggesting that the anti-tumor effects of these agents are mediated by decreasing the levels of EphA2. As these studies employed human tumor xenograft models in nude mice with reagents whose cross reactivity with murine EphA2 is unknown, we generated a mAb (Ab20) that preferentially binds, activates, and induces the degradation of murine EphA2. Treatment of established murine CT26 colorectal tumors with Ab20 reduced EphA2 protein levels to approximately 12% of control tumor levels, yet had no effect on tumor growth. CT26 tumor cell colonization of the lung was also not affected by Ab20 administration despite having barely detectable levels of EphA2. We also generated and tested a potent agonistic mAb against human EphA2 (1G9-H7). No inhibition of humanMDA-231 breast tumor xenograft growth was observed despite evidence for >85% reduction of EphA2 protein levels in the tumors. These results suggest that molecular characteristics of the tumors in addition to EphA2 over-expression may be important for predicting responsiveness to EphA2-directed therapies.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias Colorrectales/metabolismo , Regulación Neoplásica de la Expresión Génica , Inmunoterapia/métodos , Neoplasias Mamarias Animales/metabolismo , Receptor EphA2/química , Animales , Anticuerpos Monoclonales/química , Neoplasias de la Mama/terapia , Línea Celular Tumoral , Neoplasias Colorrectales/terapia , Femenino , Humanos , Neoplasias Mamarias Animales/terapia , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Trasplante de Neoplasias , Receptor EphA2/inmunología
15.
Bioorg Med Chem Lett ; 15(21): 4752-6, 2005 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-16125385

RESUMEN

The activated Factor VII/tissue factor complex (FVIIa/TF) plays a key role in the formation of blood clots. Inhibition of this complex may lead to new antithrombotic drugs. An X-ray crystal structure of a fluoropyridine-based FVIIa/TF inhibitor bound in the active site of the enzyme complex suggested that incorporation of substitution at the 5-position of the hydroxybenzoic acid side chain could lead to the formation of more potent inhibitors through interactions with the S1'/S2' pocket.


Asunto(s)
Inhibidores Enzimáticos/síntesis química , Factor VIIa/química , Fibrinolíticos/síntesis química , Piridinas/síntesis química , Tromboplastina/química , Sitios de Unión , Cristalografía por Rayos X , Inhibidores Enzimáticos/química , Factor VIIa/antagonistas & inhibidores , Inhibidores del Factor Xa , Fibrinolíticos/química , Fibrinolíticos/farmacología , Humanos , Concentración 50 Inhibidora , Unión Proteica , Tiempo de Protrombina , Piridinas/química , Relación Estructura-Actividad , Tromboplastina/antagonistas & inhibidores
16.
Protein Expr Purif ; 42(2): 295-304, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15925519

RESUMEN

Prolyl-4-hydroxylase domain-containing enzymes (PHDs) mediate the oxygen-dependent regulation of the heterodimeric transcription factor hypoxia-inducible factor-1 (HIF-1). Under normoxic conditions, one of the subunits of HIF-1, HIF-1alpha, is hydroxylated on specific proline residues to target HIF-1alpha for degradation by the ubiquitin-proteasome pathway. Under hypoxic conditions, the hydroxylation by the PHDs is attenuated by lack of the oxygen substrate, allowing HIF-1 to accumulate, translocate to the nucleus, and mediate HIF-mediated gene transcription. In several mammalian species including humans, three PHDs have been identified. We report here the cloning of a full-length rat cDNA that is highly homologous to the human and murine PHD-1 enzymes and encodes a protein that is 416 amino acids long. Both cDNA and protein are widely expressed in rat tissues and cell types. We demonstrate that purified and crude baculovirus-expressed rat PHD-1 exhibits HIF-1alpha specific prolyl hydroxylase activity with similar substrate affinities and is comparable to human PHD-1 protein.


Asunto(s)
Clonación Molecular , Procolágeno-Prolina Dioxigenasa/química , Procolágeno-Prolina Dioxigenasa/genética , Secuencia de Aminoácidos , Animales , Humanos , Cinética , Ratones , Datos de Secuencia Molecular , Especificidad de Órganos/genética , Procolágeno-Prolina Dioxigenasa/biosíntesis , ARN Mensajero/metabolismo , Ratas , Spodoptera
17.
Eur J Pharmacol ; 512(2-3): 215-22, 2005 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-15840407

RESUMEN

Recent evidence indicates that the GTPase activated Rho/Rho-kinase pathway contributes angiotensin II-induced cardiac hypertrophy and vascular remodeling. We tested this hypothesis in vivo by determining the effects of fasudil, a Rho-kinase inhibitor, on angiotensin II-induced cardiac hypertrophy, coronary vascular remodeling, and ventricular dysfunction. Six-month-old apolipoprotein E deficient (apoE-KO) mice were subcutaneously infused with angiotensin II (1.44 mg/kg/day) using an osmotic mini-pump. Mice were randomly assigned to either vehicle or fasudil (136 or 213 mg/kg/day in drinking water) group. Infusion of angiotensin II for 4 weeks resulted in cardiac enlargement, myocyte hypertrophy, and myocardial interstitial and coronary artery perivascular fibrosis. These changes were accompanied by reduced aortic flow velocity and acceleration rate. Cardiac gene expression levels of atrial natriuretic peptide (ANP) and collagen type III detected by real-time reverse transcriptase polymerase chain reaction were significantly increased in angiotensin II-infused mice. Treatment with fasudil dose-dependently attenuated angiotensin II-induced cardiac hypertrophy, prevented perivascular fibrosis, blunted the increase in ANP and collagen type III expression, and improved cardiac function, without changing blood pressure. These data are consistent with a role for Rho-kinase activation in angiotensin II-induced cardiac remodeling and vascular wall fibrosis.


Asunto(s)
1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/análogos & derivados , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/farmacología , Angiotensina II/farmacología , Apolipoproteínas E/genética , Cardiomegalia/prevención & control , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Animales , Apolipoproteínas E/metabolismo , Factor Natriurético Atrial/genética , Presión Sanguínea/efectos de los fármacos , Cardiomegalia/inducido químicamente , Cardiomegalia/patología , Colágeno Tipo III/genética , Vasos Coronarios/efectos de los fármacos , Vasos Coronarios/patología , Relación Dosis-Respuesta a Droga , Fibrosis/prevención & control , Expresión Génica/efectos de los fármacos , Frecuencia Cardíaca/efectos de los fármacos , Péptidos y Proteínas de Señalización Intracelular , Masculino , Ratones , Ratones Noqueados , Miocardio/metabolismo , Miocardio/patología , Inhibidores de Proteínas Quinasas/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética , Quinasas Asociadas a rho
18.
Circulation ; 111(17): 2219-26, 2005 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-15851596

RESUMEN

BACKGROUND: Angiotensin II (Ang II) accelerates atherosclerosis and induces abdominal aortic aneurysm (AAA) in an experimental mouse model. Agonism of a G protein-coupled receptor by Ang II activates Rho-kinase and other signaling pathways and results in activation of proteolysis and apoptosis. Enhanced proteolysis and smooth muscle cell apoptosis are important mechanisms associated with AAA. In this study, we tested the hypothesis that fasudil, a Rho-kinase inhibitor, could attenuate Ang II-induced AAA formation by inhibiting vascular wall apoptosis and extracellular matrix proteolysis. METHODS AND RESULTS: Six-month-old apolipoprotein E-deficient mice were infused with Ang II (1.44 mg x kg(-1) x d(-1)) for 1 month. Animals were randomly assigned to treatment with fasudil (136 or 213 mg x kg(-1) x d(-1) in drinking water) or tap water. Ang II infusion induced AAA formation in 75% of the mice, which was accompanied by an increase in proteolysis detected by zymographic analysis and quantified by active matrix metalloproteinase-2 activity, as well as apoptosis detected by terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling and quantified by both caspase-3 activity and histone-associated DNA fragmentation. The level of DNA fragmentation in the suprarenal aorta correlated with AAA diameter. Ang II also increased atherosclerotic lesion area and blood pressure. Fasudil treatment resulted in a dose-dependent reduction in both the incidence and severity of AAA. At the higher dose, fasudil decreased AAA by 45% while significantly inhibiting both apoptosis and proteolysis, without affecting atherosclerosis or blood pressure. CONCLUSIONS: These data demonstrate that inhibition of Rho-kinase by fasudil attenuated Ang II-induced AAA through inhibition of both apoptosis and proteolysis pathways.


Asunto(s)
1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/análogos & derivados , Angiotensina II/farmacología , Aneurisma de la Aorta Abdominal/tratamiento farmacológico , Apolipoproteínas E/deficiencia , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/farmacología , Animales , Apoptosis , Endotelio Vascular/citología , Matriz Extracelular/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Ratones , Ratones Noqueados , Inhibidores de Proteasas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Quinasas Asociadas a rho
19.
Bioorg Med Chem Lett ; 15(9): 2249-52, 2005 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-15837303

RESUMEN

Reductive amination followed by acylation of polymer-linked formyl aryl amidines generate combinatorial libraries of aryl amidines 8-13. Potent small molecule naphthylamidine inhibitors 12 (Ki<100 nM) of FVIIa/TF have been discovered and their activity against other serine proteases in the coagulation cascade is reported.


Asunto(s)
Amidinas/síntesis química , Factor VIIa/antagonistas & inhibidores , Tromboplastina/antagonistas & inhibidores , Amidinas/química , Amidinas/farmacología , Sitios de Unión , Humanos , Cinética , Modelos Moleculares , Conformación Molecular , Naftoles/síntesis química , Naftoles/química , Naftoles/farmacología , Relación Estructura-Actividad
20.
Biochemistry ; 42(26): 7942-9, 2003 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-12834346

RESUMEN

13C[(15)N] and (13)C[(19)F] rotational-echo double-resonance NMR have been used to characterize the enzyme-bound structure of ZK-816042, an amidine-imidazoline inhibitor of human factor Xa (FXa). The NMR experiments were performed on a lyophilized FXa-inhibitor complex. The complex was formed in solution in the presence of stabilizing excipients and frozen after gradual supercooling prior to lyophilization. The results indicate that the inhibitor binds with a distribution of orientations of the imidazoline ring.


Asunto(s)
Amidinas/química , Factor Xa/química , Imidazoles/química , Piridinas/química , Sitios de Unión , Inhibidores del Factor Xa , Humanos , Espectroscopía de Resonancia Magnética/métodos , Modelos Moleculares , Conformación Molecular , Tripsina/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...