Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Haemophilia ; 20(4): 593-600, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24471985

RESUMEN

Patients with haemophilia (PWH) are usually monitored by the one-stage activated partial thromboplastin time (aPTT) factor VIII (FVIII) assay. Different aPTT activators may affect clotting time (CT) and FVIII:C levels in patients treated with PEGylated FVIII. To evaluate the characteristics of PEGylated FVIII (BAY 94-9027) in various aPTT clotting assays, and to identify suitable aPTT reagents for monitoring BAY 94-9027 during the treatment of PWH, BAY 94-9027 and World Health Organization (WHO) 8th FVIII standards (WHO-8) were spiked into pooled and individual severe haemophilia A plasma at 1.0, 0.25 and 0.05 IU mL(-1) . Five commercial aPTT reagents widely used in clinical laboratories were compared and evaluated for BAY 94-9027 activity in plasma from PWH. BAY 94-9027 and WHO-8 bestowed similar CT and excellent precision when ellagic acid (SynthAFax, Dade Actin, and Cephascreen) aPTT reagents were used. In contrast, BAY 94-9027 showed significantly prolonged CT and poor precision compared with WHO-8 using silica aPTT reagents (APTT-SP and STA PTT 5). Furthermore, free 60-kDa polyethylene glycol (PEG), used for the conjugation of FVIII, showed a dose-dependent prolongation of CT in the APTT-SP assay. There was no effect on the SynthAFax-APTT, prothrombin time, or FXIa-initiated thrombin generation assay, demonstrating that the PEG moiety on FVIII has no general effect on the coagulation cascade. In summary, ellagic aPTT reagents (SynthAFax, Dade Actin, and Cephascreen) are most suitable for evaluating potency of BAY 94-9027 and should be the preferred aPTT reagents used in clinical laboratories for monitoring FVIII activity after infusion of BAY 94-9027 to PWH.


Asunto(s)
Factor VIII/química , Factor VIII/uso terapéutico , Hemofilia A/tratamiento farmacológico , Tiempo de Tromboplastina Parcial/métodos , Polietilenglicoles/química , Coagulación Sanguínea/efectos de los fármacos , Factor VIII/farmacología , Humanos , Tiempo de Tromboplastina Parcial/instrumentación , Polietilenglicoles/farmacología , Polietilenglicoles/uso terapéutico , Dióxido de Silicio/química , Resultado del Tratamiento
2.
Br J Pharmacol ; 152(7): 1070-80, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17934515

RESUMEN

BACKGROUND AND PURPOSE: Rho-kinase (ROCK) has been implicated in the pathophysiology of altered vasoregulation leading to hypertension. Here we describe the pharmacological characterization of a potent, highly selective and orally active ROCK inhibitor, the derivative of a class of azaindoles, azaindole 1 (6-chloro-N4-{3,5-difluoro-4-[(3-methyl-1H-pyrrolo[2,3-b]pyridin-4-yl)oxy]-phenyl}pyrimidine-2,4-diamine). EXPERIMENTAL APPROACH: Pharmacological characterization of azaindole 1 was performed with human recombinant ROCK in vitro. Vasodilator activity was determined using isolated vessels in vitro and different animal models in vivo. KEY RESULTS: This compound inhibited the ROCK-1 and ROCK-2 isoenzymes with IC50 s of 0.6 and 1.1 nM in an ATP-competitive manner. Although ATP-competitive, azaindole 1 was inactive against 89 kinases (IC50>10 microM) and showed only weak activity against an additional 21 different kinases (IC50=1-10 microM). Only the kinases TRK und FLT3 were inhibited by azaindole 1 in the sub-micromolar range, albeit with IC50 values of 252 and 303 nM, respectively. In vivo, azaindole 1 lowered blood pressure dose-dependently after i.v. administration in anaesthetized normotensive rats. In conscious normotensive and spontaneously hypertensive rats azaindole 1 induced a dose-dependent decrease in blood pressure after oral administration without inducing a significant reflex increase in heart rate. In anaesthetized dogs, azaindole 1 induced vasodilatation with a moderately elevated heart rate. CONCLUSIONS AND IMPLICATIONS: Azaindole 1 is representative of a new class of selective and potent ROCK inhibitors and is a valuable tool for the elucidation of the role of ROCK in the cardiovascular system.


Asunto(s)
Sistema Cardiovascular/efectos de los fármacos , Diaminas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacología , Quinasas Asociadas a rho/antagonistas & inhibidores , Administración Oral , Animales , Proteínas Reguladoras de la Apoptosis/antagonistas & inhibidores , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Sitios de Unión/efectos de los fármacos , Presión Sanguínea/efectos de los fármacos , Proteínas Quinasas Dependientes de Calcio-Calmodulina/antagonistas & inhibidores , Proteínas Quinasas Dependientes de Calcio-Calmodulina/genética , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Células Cultivadas , Simulación por Computador , Proteínas Quinasas Asociadas a Muerte Celular , Perros , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Inyecciones Intravenosas , Masculino , Ratones , Modelos Animales , Modelos Moleculares , Técnicas de Cultivo de Órganos , Fosforilación , Reacción en Cadena de la Polimerasa/métodos , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/química , Conejos , Ratas , Ratas Endogámicas SHR , Ratas Wistar , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Factores de Tiempo , Vasodilatadores/administración & dosificación , Vasodilatadores/química , Vasodilatadores/farmacología , Quinasas Asociadas a rho/genética , Quinasas Asociadas a rho/metabolismo
3.
J Mol Med (Berl) ; 77(1): 14-23, 1999 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-9930922

RESUMEN

Soluble guanylyl cyclase (sGC) is the main receptor for nitric oxide, a messenger molecule with multiple clinical implications. Understanding the activation of sGC is an important step for establishing new therapeutic principles. We have now overexpressed sGC in a baculovirus/Sf9 system optimized for high protein yields to facilitate spectral and kinetic studies of the activation mechanisms of this enzyme. It was expressed in a batch fermenter using a defined mixture of viruses encoding the alpha and beta1 subunits of the rat lung enzyme. The expressed enzyme was purified from the cytosolic fraction by anion exchange chromatography, hydroxyapatite chromatography, and size exclusion chromatography. By use of this new method 2.5 l culture yielded about 1 mg of apparently homogeneous sGC with a content of about one heme per heterodimer without the need of a heme reconstitution step. The enzyme did not contain stoichiometric amounts of copper. The basal activities of the purified enzyme were 153 and 1259 nmol min(-1) mg(-1) in the presence of Mg2+ and Mn2+, respectively. The nitric oxide releasing agent 2-(N,N-diethylamino)-diazenolate-2-oxide (DEA/NO) stimulated the enzyme 160-fold with Mg2+, whereas the NO-independent activator 3-(5'-hydroxymethyl-2'-furyl)-1-benzylindazole (YC-1) induced an increase in the activity of 101-fold at a concentration of 300 microM. The combination of DEA/NO (10 microM) and YC-1 (100 microM) elicited a dose-dependent synergistic stimulation with a maximum of a 792-fold increase over the basal activity in the presence of Mg2+, resulting in a specific activity of 121 micromol min(-1) mg(-1). The synergistic stimulation of DEA/NO and YC-1 was attenuated by the sGC inhibitor 1H-(1,2,4)oxadiazole(4,3-a)quinoxalin-1-one (ODQ) (10 microM) by 94%. In a different experimental setup a saturated carbon monoxide solution in the absence of ambient oxygen or NO stimulated the enzyme 15-fold in the absence and 1260-fold in the presence of YC-1 compared to an argon control. The heme spectra of the enzyme showed a shift of the Soret peak from 432 to 399 and 424 nm in the presence of DEA/NO or carbon monoxide, respectively. The heme spectra were not affected by YC-1 in the absence or in the presence of DEA/NO or of carbon monoxide, which reflects the fact that YC-1 does not interact directly with the heme group of the enzyme. In summary, this study shows that our expression/purification procedure is suitable for producing large amounts of highly pure sGC which contains one heme per heterodimer without a reconstitution step. The activator experiments show that in a synergistic stimulation with YC-1 sGC can be activated maximally both by nitric oxide and by carbon monoxide and that YC-1 does not directly act via heme. The described method should help to facilitate the investigation of the new therapeutic principle of NO-independent guanylyl cyclase activators.


Asunto(s)
Monóxido de Carbono/farmacología , Guanilato Ciclasa/metabolismo , Indazoles/farmacología , Óxido Nítrico/farmacología , Animales , Baculoviridae/enzimología , Baculoviridae/genética , Línea Celular , Cobre/análisis , Activación Enzimática , Guanilato Ciclasa/química , Guanilato Ciclasa/genética , Guanilato Ciclasa/aislamiento & purificación , Hemo/análisis , Hidrazinas/farmacología , Espectrometría de Masas , Óxidos de Nitrógeno , Proteínas Recombinantes/metabolismo , Spodoptera/virología
4.
J Biotechnol ; 84(3): 217-30, 2000 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-11164263

RESUMEN

Interleukin-4 (IL-4) is a multifunctional cytokine that plays an important role in the regulation of various immune responses. However, the development of IL-4 or IL-4 variants into potential therapeutic drugs is hindered by the low efficiency of the in vitro refolding process of this protein. In this work, we have investigated the improvement of the refolding yield of IL-4 using two different rational design approaches. The first one is based on the so-called inverse hydrophobic effect and involved the replacement of a solvent exposed, non-conserved, hydrophobic residue (W91) by serine. This led to an increase in stability of 1.4 kcal mol(-1) and shifted the midpoint transition temperature (Tm) from 62 to 70 degrees C. The second approach is based on the stabilization of alpha-helices through the introduction of favorable local interactions. This strategy resulted in the following helix sequence for helix C of IL-4, 68ASAAEANRHKQLIRFLKRLDRNLWGLAG95. The mutant protein was stabilized by 0.5 kcal mol(-1), the Tm shifted to 68 degrees C, and a two-fold increase in the refolding yield was consistently observed. Our results make the large-scale production of IL-4 derivatives economically more viable, suggest that a similar approach can be applied to other related proteins, and may represent a general strategy to improve in vitro refolding yields through the selective optimization of the stability of alpha-helices.


Asunto(s)
Interleucina-4/química , Interleucina-4/metabolismo , Pliegue de Proteína , Secuencia de Aminoácidos , Cromatografía Líquida de Alta Presión , Dicroismo Circular , Guanidina/metabolismo , Humanos , Interleucina-4/genética , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida/genética , Resonancia Magnética Nuclear Biomolecular , Péptidos/química , Péptidos/genética , Péptidos/metabolismo , Desnaturalización Proteica/genética , Estructura Secundaria de Proteína/genética , Temperatura , Termodinámica
5.
BMC Pharmacol ; 1: 13, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11801189

RESUMEN

BACKGROUND: The most important receptor for nitric oxide is the soluble guanylate cyclase (sGC), a heme containing heterodimer. Recently, a pyrazolopyridine derivative BAY 41-2272, structurally related to YC-1, was identified stimulating soluble guanylate cyclase in an NO-independent manner, which results in vasodilatation and antiplatelet activity. The study described here addresses the identification of the NO-independent site on soluble guanylate cyclase. RESULTS: We developed a photoaffinity label (3H-meta-PAL) for the direct and NO-independent soluble guanylate cyclase (sGC) stimulator BAY 41-2272 by introducing an azido-group into the tritium labeled compound. The synthesized photoaffinitylabel directly stimulates the purified sGC and shows in combination with NO a synergistic effect on sGC activity. Irradiation with UV light of 3H-meta-PAL together with the highly purified sGC leads to a covalent binding to the alpha1-subunit of the enzyme. This binding is blocked by unlabeled meta-PAL, YC-1 and BAY 41-2272. For further identification of the NO-independent regulatory site the 3H-meta-PAL labeled sGC was fragmented by CNBr digest. The 3H-meta-PAL binds to a CNBr fragment, consisting of the amino acids 236-290 of the alpha1-subunit. Determination of radioactivity of the single PTH-cycles from the sequencing of this CNBr fragment detected the cysteines 238 and 243 as binding residues of the 3H-meta-PAL. CONCLUSIONS: Our data demonstrate that the region surrounding the cysteines 238 and 243 in the alpha1-subunit of the sGC could play an important role in regulation of sGC activity and could be the target of this new type of sGC stimulators.


Asunto(s)
Activadores de Enzimas/farmacología , Guanilato Ciclasa/metabolismo , Indazoles/farmacología , Óxido Nítrico/metabolismo , Pirazoles/farmacología , Piridinas/farmacología , Animales , Células Cultivadas , Activación Enzimática , Guanilato Ciclasa/genética , Insectos/citología , Etiquetas de Fotoafinidad , Proteínas Recombinantes/efectos de los fármacos , Proteínas Recombinantes/metabolismo
6.
J Thromb Haemost ; 11(9): 1699-706, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23692404

RESUMEN

BACKGROUND: The rapid clearance of factor IX (FIX) necessitates frequent intravenous administration to achieve effective prophylaxis for patients with hemophilia B. Subcutaneous administration would be a preferred route of administration but is limited by bioavailability. OBJECTIVES: To improve the pharmacokinetics (PK) and bioavailability of FIX, a screen was performed to identify positions for the introduction of novel glycosylation sites with maximal effect on PK and maintenance of coagulation activity. METHODS: Two hundred fifty-one variants, each containing one additional N-linked glycosylation site, were screened in vitro, and the PK profiles of selected variants mapping to spatially distinct regions of FIX were evaluated in mice. Optimal variants were combined, and their PK and efficacy were determined in mice with hemophilia B. RESULTS: Variants that mapped to spatially distinct regions of the FIX structure exhibited different degrees of improved PK and enabled selection of optimized sites while minimizing the loss of FIX activity. Combining the most effective N-glycan sites in the same FIX molecule resulted in further improvements in PK. An optimized variant containing three novel N-glycan sites (at amino acids 103, 151, and 228), and the activity enhancing 338A variant had double the specific activity of wild-type FIX, exhibited 4.5-fold reduced clearance and 2.4-fold increased subcutaneous bioavailability, and was efficacious at a fivefold lower mass dose than wild-type FIX after subcutaneous injection in a bleeding model in mice with hemophilia B. CONCLUSIONS: Glycoengineering was used to significantly improve the subcutaneous PK and efficacy of FIX and may have advantages for subcutaneous dosing.


Asunto(s)
Factor IX/farmacocinética , Ingeniería de Proteínas , Animales , Disponibilidad Biológica , Modelos Animales de Enfermedad , Factor IX/administración & dosificación , Factor IX/genética , Factor IX/uso terapéutico , Glicosilación , Células HEK293 , Hemofilia B/tratamiento farmacológico , Humanos , Inyecciones Subcutáneas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutagénesis Sitio-Dirigida , Conformación Proteica
7.
Vaccine ; 26(12): 1552-65, 2008 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-18295380

RESUMEN

For the production of a chemically inactivated Parapoxvirus ovis (PPVO), an adherent bovine kidney cell line was cultivated on Cytodex-3 microcarriers in suspension culture. The inactivated and purified virus particles have shown immune modulatory activity in several animal models. PPVO was produced by a biphasic batch process at the 3.5 and 10 L scale. Aeration was realised by bubble-free membrane oxygenation via a tube stator with a central two-blade anchor impeller. In order to increase efficiency, process robustness and safety, the established process was optimised. The cell line was adapted to a protein-free medium (except recombinant insulin) in order to increase biosafety. A scale up to a 50 L pilot plant with direct cell expansion was performed successfully. In parallel, the biphasic batch process was optimised with special emphasis on different operating conditions (cell number, Multiplicity of Infection (MOI), etc.) and process management (fed-batch, dialysis, etc.). The quality and concentration of the purified virus particles was assessed by quantitative electron microscopy, residual host cell protein and DNA-content and, finally, biologic activity in a transgenic mouse model. This integrated approach led to a new, safe, robust and highly productive large-scale production process, called "Volume-Expanded-Fed" Batch with cell densities up to 6-7e06 cells/mL. By subsequent dilution of infected cells into the next process scale, an increase in total productivity by a factor of 40 (related to an established biphasic batch process) was achieved.


Asunto(s)
Parapoxvirus/crecimiento & desarrollo , Parapoxvirus/inmunología , Vacunas de Productos Inactivados/biosíntesis , Animales , Bovinos , Recuento de Células , Supervivencia Celular , Células Cultivadas , Medios de Cultivo , ADN Viral/biosíntesis , ADN Viral/inmunología , Hepatitis B/inmunología , Riñón/citología , Riñón/metabolismo , Ratones , Ratones Transgénicos , Microscopía Electrónica , Tripsina/química , Vacunas de Productos Inactivados/inmunología
8.
J Recept Signal Transduct Res ; 15(1-4): 117-30, 1995.
Artículo en Inglés | MEDLINE | ID: mdl-8903936

RESUMEN

The rat vascular smooth muscle cell (VSMC) line A10 (ATCC CRL 1476) was stably transfected with a human c-fos promoter-driven luciferase reporter gene to monitor thrombin receptor activation and subsequent induction of c-fos expression. Selective activation of the endogeneous thrombin receptor by the thrombin receptor activating peptide (TRAP1-6), SFLLRN, is shown here to result in a significant transient increase of intracellular [Ca2+], dose-dependent induction of c-fos promoter-mediated luciferase activity, and stimulation of DNA synthesis. These data demonstrate that A10 cells and reporter line derivatives thereof possess a functional thrombin receptor very similar or identical to that previously described. Results obtained with various signal transduction modulating or inhibiting agents support previous notions showing that thrombin receptor activation by SFLLRN is coupled to events involving p21ras activation, protein tyrosine kinase, and activation of PKC. The A10 reporter line described here proved to be a helpful and reliable tool to study alpha-thrombin and TRAP1-6-mediated intracellular events, since it retained most of the spectrum of biological responses found in primary VSMC cultures.


Asunto(s)
Genes fos , Músculo Liso Vascular/metabolismo , Receptores de Trombina/metabolismo , Animales , Línea Celular , ADN/biosíntesis , Regulación de la Expresión Génica , Genes Reporteros , Humanos , Luciferasas/genética , Fragmentos de Péptidos/farmacología , Ratas , Receptores de Trombina/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Trombina/farmacología , Transfección
9.
Eur J Biochem ; 247(3): 890-5, 1997 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-9288912

RESUMEN

An attractive target for anti-herpes chemotherapy is the herpes simplex virus 1 (HSV-1) protease encoded by the UL26 gene. Studies with HSV-1 strains that harbour mutations in the protease gene have demonstrated that the protease is essential for DNA packaging and virus maturation. The UL26 translation product is 635 amino acids long and undergoes autoproteolytic processing between residues Ala247/Ser248 and Ala610/Ser611. The N-terminal processing product (amino acids 1-247) contains the protease domain. To perform crystallization studies and high throughput screening for potent inhibitors, large amounts of the HSV-1 protease are required. However, expression of the natural HSV-1 protease gene in Escherichia coli using a T7-promoter-regulated system is low and does not allow for the efficient production of larger amounts of highly purified enzyme. In this report, we describe the use of a synthetic protease gene with optimized E. coli codon usage. The level of protease expression was at least 20 times higher with the synthetic gene as compared to the natural UL26 gene. The HSV-1 protease was purified to homogeneity in three steps using mixed-bed ion-exchange chromatography, affinity chromatography, and hydroxyapatite chromatography.


Asunto(s)
Cápside/genética , Genes Sintéticos , Herpesvirus Humano 1/enzimología , Serina Endopeptidasas/genética , Proteínas Virales , Secuencia de Aminoácidos , Secuencia de Bases , Cápside/aislamiento & purificación , Cromatografía Líquida de Alta Presión , Cromatografía por Intercambio Iónico , Clonación Molecular , ADN Recombinante , Electroforesis en Gel de Poliacrilamida , Escherichia coli/genética , Datos de Secuencia Molecular , Serina Endopeptidasas/aislamiento & purificación
10.
Nature ; 410(6825): 212-5, 2001 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-11242081

RESUMEN

Nitric oxide (NO) is a widespread, potent, biological mediator that has many physiological and pathophysiological roles. Research in the field of NO appears to have followed a straightforward path, and the findings have been progressive: NO and cyclic GMP are involved in vasodilatation; glycerol trinitrate relaxes vascular smooth muscles by bioconversion to NO; mammalian cells synthesize NO; and last, NO mediates vasodilatation by stimulating the soluble guanylate cyclase (sGC), a heterodimeric (alpha/beta) haem protein that converts GTP to cGMP2-4. Here we report the discovery of a regulatory site on sGC. Using photoaffinity labelling, we have identified the cysteine 238 and cysteine 243 region in the alpha1-subunit of sGC as the target for a new type of sGC stimulator. Moreover, we present a pyrazolopyridine, BAY 41-2272, that potently stimulates sGC through this site by a mechanism that is independent of NO. This results in antiplatelet activity, a strong decrease in blood pressure and an increase in survival in a low-NO rat model of hypertension, and as such may offer an approach for treating cardiovascular diseases.


Asunto(s)
Guanilato Ciclasa/química , Óxido Nítrico/química , Secuencia de Aminoácidos , Animales , Antihipertensivos/uso terapéutico , Sitios de Unión , Presión Sanguínea/efectos de los fármacos , Óxidos N-Cíclicos/farmacología , Cisteína/química , Modelos Animales de Enfermedad , Activación Enzimática , Femenino , Guanilato Ciclasa/metabolismo , Hemo/química , Humanos , Imidazoles/farmacología , Técnicas In Vitro , Indazoles/farmacología , Datos de Secuencia Molecular , Etiquetas de Fotoafinidad , Inhibidores de Agregación Plaquetaria/farmacología , Pirazoles/farmacología , Piridinas/farmacología , Ratas , Solubilidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA