Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Inherit Metab Dis ; 35(6): 1107-17, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22450713

RESUMEN

The success of enzymatic replacement in Gaucher disease has stimulated development of targeted protein replacement for other lysosomal disorders, including Anderson-Fabry disease, which causes fatal cardiac, cerebrovascular and renal injury: deficiency of lysosomal α-Galactosidase A induces accumulation of glycosphingolipids. Endothelial cell storage was the primary endpoint in a clinical trial that led to market authorization. Two α-Galactosidase A preparations are licensed worldwide, but fatal outcomes persist, with storage remaining in many tissues. We compare mechanisms of uptake of α -Galactosidase A into cells relevant to Fabry disease, in order to investigate if the enzyme is targeted to the lysosomes in a mannose-6-phosphate receptor dependent fashion, as generally believed. α -Galactosidase A uptake was examined in fibroblasts, four different endothelial cell models, and hepatic cells in vitro. Uptake of europium-labeled human α -Galactosidase A was measured by time-resolved fluorescence. Ligand-specific uptake was quantified in inhibitor studies. Targeting to the lysosome was determined by precipitation and by confocal microscopy. The quantity and location of cation-independent mannose-6-phosphate receptors in the different cell models were investigated using confocal microscopy. Uptake and delivery of α -Galactosidase A to lysosomes in fibroblasts is mediated by the canonical mannose-6-phosphate receptor pathway, but in endothelial cells in vitro this mechanism does not operate. Moreover, this observation is supported by a striking paucity of expression of cation independent mannose-6-phosphate receptors on the plasma membrane of the four endothelial cell models and by little delivery of enzyme to lysosomes, when compared with fibroblasts. If these observations are confirmed in vivo, alternative mechanisms will be needed to explain the ready clearance of storage from endothelial cells in patients undergoing enzyme replacement therapy.


Asunto(s)
Terapia de Reemplazo Enzimático/métodos , Enfermedad de Fabry/tratamiento farmacológico , Enfermedad de Fabry/enzimología , Lisosomas/enzimología , Línea Celular , Células Cultivadas , Sistemas de Liberación de Medicamentos/métodos , Células Endoteliales/enzimología , Fibroblastos/enzimología , Humanos , Isoenzimas/administración & dosificación , Isoenzimas/farmacocinética , Microscopía Confocal , Modelos Biológicos , Receptor IGF Tipo 2/metabolismo , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/farmacocinética , alfa-Galactosidasa/administración & dosificación , alfa-Galactosidasa/farmacocinética
2.
Clin Pharmacol Drug Dev ; 10(9): 1075-1088, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33876577

RESUMEN

Approved therapies for Fabry disease (FD) include migalastat, an oral pharmacological chaperone, and agalsidase beta and agalsidase alfa, 2 forms of enzyme replacement therapy. Broad tissue distribution may be beneficial for clinical efficacy in FD, which has severe manifestations in multiple organs. Here, migalastat and agalsidase beta biodistribution were assessed in mice and modeled using physiologically based pharmacokinetic (PBPK) analysis, and migalastat biodistribution was subsequently extrapolated to humans. In mice, migalastat concentration was highest in kidneys and the small intestine, 2 FD-relevant organs. Agalsidase beta was predominantly sequestered in the liver and spleen (organs unaffected in FD). PBPK modeling predicted that migalastat 123 mg every other day resulted in concentrations exceeding the in vitro half-maximal effective concentration in kidneys, small intestine, skin, heart, and liver in human subjects. However, extrapolation of mouse agalsidase beta concentrations to humans was unsuccessful. In conclusion, migalastat may distribute to tissues that are inaccessible to intravenous agalsidase beta in mice, and extrapolation of mouse migalastat concentrations to humans showed adequate tissue penetration, particularly in FD-relevant organs.


Asunto(s)
1-Desoxinojirimicina/análogos & derivados , Isoenzimas/farmacocinética , Modelos Biológicos , alfa-Galactosidasa/farmacocinética , 1-Desoxinojirimicina/farmacocinética , Adulto , Animales , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Persona de Mediana Edad , Especificidad de la Especie , Distribución Tisular , Adulto Joven , alfa-Galactosidasa/genética
3.
J Pharmacokinet Pharmacodyn ; 36(5): 443-59, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19789963

RESUMEN

Patients that are exposed to biotechnology-derived therapeutics often develop antibodies to the therapeutic, the magnitude of which is assessed by measuring antibody titers. A statistical approach for analyzing antibody titer data conditional on seroconversion is presented. The proposed method is to first transform the antibody titer data based on a geometric series using a common ratio of 2 and a scale factor of 50 and then analyze the exponent using a zero-inflated or hurdle model assuming a Poisson or negative binomial distribution with random effects to account for patient heterogeneity. Patient specific covariates can be used to model the probability of developing an antibody response, i.e., seroconversion, as well as the magnitude of the antibody titer itself. The method was illustrated using antibody titer data from 87 male seroconverted Fabry patients receiving Fabrazyme. Titers from five clinical trials were collected over 276 weeks of therapy with anti-Fabrazyme IgG titers ranging from 100 to 409,600 after exclusion of seronegative patients. The best model to explain seroconversion was a zero-inflated Poisson (ZIP) model where cumulative dose (under a constant dose regimen of dosing every 2 weeks) influenced the probability of seroconversion. There was an 80% chance of seroconversion when the cumulative dose reached 210 mg (90% confidence interval: 194-226 mg). No difference in antibody titers was noted between Japanese or Western patients. Once seroconverted, antibody titers did not remain constant but decreased in an exponential manner from an initial magnitude to a new lower steady-state value. The expected titer after the new steady-state titer had been achieved was 870 (90% CI: 630-1109). The half-life to the new steady-state value after seroconversion was 44 weeks (90% CI: 17-70 weeks). Time to seroconversion did not appear to be correlated with titer at the time of seroconversion. The method can be adequately used to model antibody titer data.


Asunto(s)
Anticuerpos/sangre , Isoenzimas/inmunología , Isoenzimas/farmacocinética , alfa-Galactosidasa/inmunología , alfa-Galactosidasa/farmacocinética , Adolescente , Algoritmos , Formación de Anticuerpos/fisiología , Pueblo Asiatico , Niño , Interpretación Estadística de Datos , Ensayo de Inmunoadsorción Enzimática , Enfermedad de Fabry/tratamiento farmacológico , Enfermedad de Fabry/metabolismo , Semivida , Humanos , Isoenzimas/uso terapéutico , Masculino , Modelos Estadísticos , Estudios Multicéntricos como Asunto , Distribución de Poisson , Proteínas/inmunología , Ensayos Clínicos Controlados Aleatorios como Asunto , Población Blanca , Adulto Joven , alfa-Galactosidasa/uso terapéutico
4.
Alcohol Alcohol ; 43(3): 281-6, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18304992

RESUMEN

AIMS: The goal of this paper was to determine Glutamate dehydrogenase's (GLDH) increased activity and rapid decrease in alcoholics according to last consumption of alcohol as well as to confirm that quick normalisation cannot be a sign of hepatocyte necrosis and that GLDH from rough endoplasmic reticulum exists in the serum of alcoholics. METHODS: GLDH activity was assessed in 238 alcoholics admitted to hospital. A blood sample was taken from every subject three times: on admission to hospital, after 24 hours and after 7 days. We established our own reference activities for GLDH in serum, i.e., up to 124.0 nkat/l in males and 64.5 nkat/l in females. RESULTS: Alcoholics were ascertained to have statistically significantly higher mean GLDH serum activities (men 351.53 nkat/L, women 296.08 nkat/L); the higher the level, the less elapsed time there was after the latest alcohol intake. There was an increased GLDH activity in 65.5% of alcoholics; furthermore, the percentage rose sharply to 72.2% with those who had last consumed alcohol within 48 hours. In the serum of alcoholics, it was found that, on average, it was 32.4% thermo-stable and 67.6% thermo-labile GLDH, which means that almost one third of GLDH serum originates from rough endoplasmic reticulum and rest from mitochondria. This is a completely new finding. CONCLUSIONS: A statistically significant fast decrease of GLDH serum activity after a break in alcohol consumption has been confirmed. It is estimated that increased GLDH activity in the sera of alcohol dependents and its fast decrease after total abstinence has been restored are specific for alcohol addiction.


Asunto(s)
Alcoholismo/sangre , Alcoholismo/enzimología , Glutamato Deshidrogenasa/sangre , Glutamato Deshidrogenasa/farmacocinética , Adolescente , Adulto , Anciano , Femenino , Humanos , Isoenzimas/sangre , Isoenzimas/farmacocinética , Cinética , Masculino , Persona de Mediana Edad , Templanza
5.
J Clin Invest ; 99(1): 55-61, 1997 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-9011576

RESUMEN

Cardiac myocytes coexpress multiple protein kinase C (PKC) isoforms which likely play distinct roles in signaling pathways leading to changes in contractility, hypertrophy, and ischemic preconditioning. Although PKC has been reported to be activated during myocardial ischemia, the effect of ischemia/hypoxia on individual PKC isoforms has not been determined. This study examines the effect of hypoxia on the subcellular distribution of individual PKC isoforms in cultured neonatal rat ventricular myocytes. Hypoxia induces the redistribution of PKC alpha and PKC epsilon from the soluble to the particulate compartment. This effect (which is presumed to represent activation of PKC alpha and PKC epsilon) is detectable by 1 h, sustained for up to 24 h, and reversible within 1 h of reoxygenation. Inhibition of phospholipase C with tricyclodecan-9-yl-xanthogenate (D609) prevents the hypoxia-induced redistribution of PKC alpha and PKC epsilon, whereas chelation of intracellular calcium with 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid (BAPTA) blocks the redistribution of PKC alpha, but not PKC epsilon; D609 and BAPTA do not influence the partitioning of PKC alpha and PKC epsilon in normoxic myocytes. Hypoxia, in contrast, decreases the membrane association of PKC delta via a mechanism that is distinct from the hypoxia-induced translocation/activation of PKC alpha/PKC epsilon, since the response is slower in onset, slowly reversible upon reoxygenation, and not blocked by D609 or BAPTA. The hypoxia-induced shift of PKC delta to the soluble compartment does not prevent subsequent 4-beta phorbol 12-myristate-13-acetate-dependent translocation/activation of PKC delta. Hypoxia does not alter the abundance of any PKC isoform nor does it alter the subcellular distribution of PKC lambda. The selective hypoxia-induced activation of PKC isoforms through a pathway involving phospholipase C (PKC alpha/PKC epsilon) and intracellular calcium (PKC alpha) may critically influence cardiac myocyte contractility, gene expression, and/or tolerance to ischemia.


Asunto(s)
Hipoxia/enzimología , Isoenzimas/metabolismo , Isoenzimas/farmacocinética , Proteína Quinasa C/metabolismo , Proteína Quinasa C/farmacocinética , Animales , Hidrocarburos Aromáticos con Puentes/farmacología , Calcio/metabolismo , Calcio/fisiología , Membrana Celular/enzimología , Células Cultivadas , Ácido Egtácico/análogos & derivados , Ácido Egtácico/farmacología , Immunoblotting , Miocardio/citología , Norbornanos , Inhibidores de Fosfodiesterasa/farmacología , Ratas , Ratas Wistar , Acetato de Tetradecanoilforbol/farmacología , Tiocarbamatos , Tionas/farmacología , Fosfolipasas de Tipo C/antagonistas & inhibidores , Fosfolipasas de Tipo C/metabolismo , Fosfolipasas de Tipo C/fisiología
6.
Genet Med ; 9(8): 504-9, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17700388

RESUMEN

PURPOSE: This 10-week study was conducted to determine the pharmacokinetics of varying doses of agalsidase alfa and evaluate the effect of dose and dosing frequency on plasma Gb3 levels. METHODS: Eighteen adult male Fabry patients, naive to enzyme replacement therapy, were randomized to one of five regimens: 0.1, 0.2, or 0.4 mg/kg weekly; 0.2 mg/kg every other week (the approved dose); or 0.4 mg/kg every other week. Intravenous infusion rate was 0.1 mg/kg per 20 minutes. Plasma Gb3 levels were assessed at baseline and periodically during the study. RESULTS: The mean half-life was 56-76 minutes, and the mean volume of distribution at steady state was 17%-18% of body weight, with no significant association between dose and half-life, clearance, or volume of distribution at steady state. The area under the curve was linearly proportional to the dose from 0.1 to 0.4 mg/kg. Baseline average plasma Gb3 was 9.12 +/- 2.61 nmol/mL and after 10 weeks of treatment was significantly reduced by about 50% in each group with no statistically significant differences between groups. CONCLUSIONS: Reduction of plasma Gb3 levels was independent of dose or dose frequency in the range tested. These observations, coupled with the clinical trial experience of both agalsidase alfa and agalsidase beta, indicate that the standard dose of agalsidase alfa is sufficient to maximally reduce plasma Gb3. However, because plasma Gb3 is not a validated surrogate of disease severity in Fabry disease, further clinical study will be required to determine the optimal dosing regimen for providing maximal clinical benefit.


Asunto(s)
Enfermedad de Fabry/terapia , alfa-Galactosidasa/farmacología , Adolescente , Adulto , Protocolos Clínicos , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Enfermedad de Fabry/enzimología , Humanos , Infusiones Intravenosas , Isoenzimas/administración & dosificación , Isoenzimas/efectos adversos , Isoenzimas/farmacocinética , Isoenzimas/farmacología , Masculino , Proteínas Recombinantes , Trihexosilceramidas/sangre , alfa-Galactosidasa/administración & dosificación , alfa-Galactosidasa/efectos adversos , alfa-Galactosidasa/farmacocinética
7.
Drugs ; 67(3): 435-55, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17335299

RESUMEN

Agalsidase beta (Fabrazyme) is a recombinant human alpha-galactosidase A enzyme approved for intravenous use in the treatment of Fabry disease. Fabry disease is a progressive, multisystemic, potentially life threatening disorder caused by a deficiency of alpha-galactosidase A. This deficiency results in accumulation of glycosphingolipids, particularly globotriaosylceramide (GL-3), in the lysosomes of various tissues. This accumulation is the underlying driver of disease progression. Agalsidase beta provides an exogenous source of alpha-galactosidase A.Intravenous agalsidase beta is effective and well tolerated in patients with Fabry disease. In a phase III trial, agalsidase beta was shown to clear GL-3 from various target cells and, in a subsequent extension of this trial, prevent GL-3 reaccumulation. In a post-approval trial, agalsidase beta was shown to provide significant clinical benefit by reducing the risk of a major clinical event. Thus, agalsidase beta represents an important advance in the treatment of Fabry disease, and agalsidase beta therapy should be strongly considered in patients with Fabry disease who are suitable candidates.


Asunto(s)
Enfermedad de Fabry/tratamiento farmacológico , Isoenzimas/uso terapéutico , Proteínas Recombinantes/uso terapéutico , alfa-Galactosidasa/uso terapéutico , Adulto , Pueblo Asiatico , Niño , Femenino , Humanos , Isoenzimas/efectos adversos , Isoenzimas/farmacocinética , Isoenzimas/farmacología , Japón , Masculino , Calidad de Vida , Proteínas Recombinantes/efectos adversos , Proteínas Recombinantes/farmacocinética , Proteínas Recombinantes/farmacología , Trihexosilceramidas , alfa-Galactosidasa/efectos adversos , alfa-Galactosidasa/farmacocinética , alfa-Galactosidasa/farmacología
8.
J Clin Pharmacol ; 47(10): 1222-30, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17698592

RESUMEN

This multicenter, open-label study evaluated pharmacokinetics, pharmacodynamics, and safety of agalsidase alpha in pediatric compared with adult patients with Fabry disease. The pharmacokinetic parameters of pediatric patients (19 boys, 5 girls, 6-18 years old; mean age, 11.8 years) were compared to those of adult male and female patients who participated in other clinical studies. All patients received agalsidase alpha at a dose of 0.2 mg/kg infused over 40 minutes every other week. Agalsidase alpha exhibited a biphasic serum elimination profile with a maximum serum concentration at the end of the 40-minute infusion; <1% of the maximum concentration was detected 8 hours after dosing. In children, serum clearance was 2.0 to 9.4 mL/min/kg and tended to decrease with increasing age. The average clearance in children, 3.7 +/- 1.5 mL/min/kg (mean +/- SD), was significantly greater than that measured in 33 adults (2.3 +/- 0.7 mL/min/kg, P < .0001). Mean terminal elimination half-life of agalsidase alpha was prolonged in week 25 compared with baseline (150 vs 66 minutes) in 8 of 19 male children. The magnitude of the reduction of plasma globotriaosylceremide was similar in all age groups and was independent of area under the curve and other pharmacokinetic parameters. Except for clearance in younger patients, agalsidase alpha appears to have comparable pharmacokinetic and pharmacodynamic profiles in pediatric and adult Fabry patients of both genders.


Asunto(s)
Enfermedad de Fabry/tratamiento farmacológico , alfa-Galactosidasa/farmacocinética , alfa-Galactosidasa/uso terapéutico , Adolescente , Adulto , Factores de Edad , Anciano , Área Bajo la Curva , Niño , Esquema de Medicación , Femenino , Semivida , Humanos , Inmunoglobulina G/sangre , Infusiones Intravenosas , Isoenzimas/administración & dosificación , Isoenzimas/inmunología , Isoenzimas/farmacocinética , Isoenzimas/uso terapéutico , Masculino , Persona de Mediana Edad , Factores Sexuales , Resultado del Tratamiento , Trihexosilceramidas/sangre , alfa-Galactosidasa/administración & dosificación , alfa-Galactosidasa/inmunología
9.
Oncogene ; 24(6): 992-1000, 2005 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-15592506

RESUMEN

Protein kinase (PK)C theta is a critical regulator of mature T-cell activation and proliferation, being implicated in TCR-triggered nuclear factor (NF)-kappa B activation and providing important survival signals to leukemic T cells. We previously showed that overexpression of pT alpha/pre-TCR and constitutive activation of NF-kappa B characterize the T-cell leukemia/lymphoma developing in Notch3-IC transgenic mice. We report here that PKC theta is a downstream target of Notch3 signaling and that its activation and membrane translocation require a functional pre-TCR in order to trigger NF-kappa B activation in thymocytes and lymphoma cells of transgenic mice. Furthermore, deletion of PKC theta in Notch3-IC transgenic mice reduces the incidence of leukemia, correlating with decreased NF-kappa B activation. This paper therefore suggests that PKC theta mediates the activation of NF-kappa B by pre-TCR in immature thymocytes and contributes to the development of Notch3-dependent T-cell lymphoma.


Asunto(s)
Isoenzimas/genética , Isoenzimas/farmacología , Leucemia de Células T/genética , Leucemia de Células T/fisiopatología , Proteína Quinasa C/genética , Proteína Quinasa C/farmacología , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/farmacología , Receptores de Superficie Celular/genética , Animales , Membrana Celular , Isoenzimas/farmacocinética , Linfoma de Células T , Glicoproteínas de Membrana , Ratones , Ratones Transgénicos , FN-kappa B/genética , FN-kappa B/farmacología , Proteína Quinasa C/farmacocinética , Proteína Quinasa C-theta , Receptor Notch3 , Receptor Notch4 , Receptores de Antígenos de Linfocitos T alfa-beta , Receptores Notch , Transducción de Señal , Timo/citología , Dedos de Zinc
10.
Biochim Biophys Acta ; 1380(2): 163-76, 1998 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-9565679

RESUMEN

Rabbit muscle enolase stimulates immunoglobulin production by a human hybridoma line, HB4C5 cells under serum-free condition. IgM productivity of HB4C5 cells was enhanced more than 20-fold by this enzyme at 220 micro/ml. Human peripheral blood lymphocytes were also facilitated their IgM and IgG productivity in the serum-free medium. However, baker's yeast enolase was ineffective to accelerate immunoglobulin production by HB4C5 cells, in spite of the same specific enzymatic activity as rabbit muscle enolase. There were differences in sensitivities against heat treatment and trypsin digestion between IPSF and enzymatic activities of enolase. These results imply that the immunoglobulin production stimulating effect of rabbit muscle enolase is irrelevant to its enzymatic function and reaction products. This fact also means that this enzyme has another function other than enzymatic one in glycolysis. Rabbit muscle enolase enhanced IgM production of transcription-suppressed HB4C5 cells treated with actinomycin D. Cycloheximide treatment of HB4C5 cells was useless to inhibit the expression of immunoglobulin production stimulating activity. However, inhibition of post-transcriptional process by monensin invalidated the activity of enolase. These findings suggest that enolase from rabbit muscle accelerates the steps between translation and post-translational processes to enhance immunoglobulin productivity. In addition, laser confocal microscopic analysis revealed that enolase from rabbit muscle was subsequently incorporated by HB4C5 cells.


Asunto(s)
Isoenzimas/fisiología , Músculos/enzimología , Fosfopiruvato Hidratasa/fisiología , Animales , Línea Celular , Cromatografía en Gel , Técnica del Anticuerpo Fluorescente , Humanos , Hibridomas , Inmunoglobulina M/biosíntesis , Inmunoglobulinas/biosíntesis , Isoenzimas/análisis , Isoenzimas/farmacocinética , Linfocitos/inmunología , Linfocitos/metabolismo , Microscopía Confocal , Fosfopiruvato Hidratasa/análisis , Fosfopiruvato Hidratasa/farmacocinética , Biosíntesis de Proteínas/efectos de los fármacos , Biosíntesis de Proteínas/genética , Biosíntesis de Proteínas/fisiología , Procesamiento Proteico-Postraduccional/genética , Procesamiento Proteico-Postraduccional/fisiología , Conejos , Transcripción Genética/genética , Transcripción Genética/fisiología
11.
J Leukoc Biol ; 51(1): 84-92, 1992 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-1311015

RESUMEN

Granule-poor human neutrophil cytoplasts, prepared without heat or cytochalasin B treatment so as to preserve both motile function and activatable respiratory burst oxidase, were investigated for their content of several isoforms of protein kinase C (PKC). Immunoblotting with isoform-specific rabbit antibodies (Abs) to PKC revealed that both the alpha-specific and the beta(I and II)- specific Abs recognized a protein band of 78 kd comigrating with PKC from rat brain cytosol. The gamma-specific antiserum did not detect any protein of this molecular mass. The cytoplast beta-PKC band was more readily detected than the cytoplast alpha-PKC band. Antibodies to beta I- or beta-II- specific PKC sequences showed the beta II subtype to be the predominant form of beta-PKC, although some beta I was also found. The identity of the 78-kd cytoplast bands as PKC was established by the fact that phorbol ester treatment of intact cytoplasts induced translocation of the bands from cytosol to membrane fractions. However, whereas PKC specific activity was similar in cytoplast lysates and brain cytosol, immunoreactivity of cytoplast alpha- and beta-PKC bands was considerably less than that of rat brain. Hydroxylapatite chromatography of partially purified cytoplast PKC revealed two major peaks of PKC activity precisely coeluting with brain alpha- and beta-PKC and displaying comparable enzymatic activities despite the relatively weak immunoreactivity of cytoplast alpha- and beta-PKC. To our knowledge, this is the first demonstration that human neutrophil-derived cytoplasts contain alpha, beta I, and beta II forms of PKC and that each isoform translocates from cytosol to membrane upon exposure to phorbol ester at concentrations that induce superoxide production. In addition, our evidence raises the possibility that cytoplasts may also possess other isoforms of PKC that we are unable to detect with our alpha, beta, and gamma antibodies. Finally, the granule-poor cytoplasts seem a particularly useful preparation in which to examine the role of individual PKC isoforms in neutrophil activation.


Asunto(s)
Isoenzimas/análisis , Neutrófilos/enzimología , Proteína Quinasa C/análisis , Secuencia de Aminoácidos , Animales , Transporte Biológico/efectos de los fármacos , Transporte Biológico/fisiología , Supervivencia Celular/fisiología , Cromatografía/métodos , Citocalasina B/farmacología , Humanos , Sueros Inmunes/inmunología , Immunoblotting , Isoenzimas/inmunología , Isoenzimas/farmacocinética , Masculino , Datos de Secuencia Molecular , Neutrófilos/citología , Neutrófilos/metabolismo , Ésteres del Forbol/farmacología , Proteína Quinasa C/inmunología , Proteína Quinasa C/farmacocinética , Ratas , Ratas Endogámicas , Homología de Secuencia de Ácido Nucleico , Superóxidos/metabolismo
12.
Clin Pharmacol Ther ; 98(2): 196-204, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25869840

RESUMEN

Preclinical development of new biological entities (NBEs), such as human protein therapeutics, requires considerable expenditure of time and costs. Poor prediction of pharmacokinetics in humans further reduces net efficiency. In this study, we show for the first time that pharmacokinetic data of NBEs in humans can be successfully obtained early in the drug development process by the use of microdosing in a small group of healthy subjects combined with ultrasensitive accelerator mass spectrometry (AMS). After only minimal preclinical testing, we performed a first-in-human phase 0/phase 1 trial with a human recombinant therapeutic protein (RESCuing Alkaline Phosphatase, human recombinant placental alkaline phosphatase [hRESCAP]) to assess its safety and kinetics. Pharmacokinetic analysis showed dose linearity from microdose (53 µg) [(14) C]-hRESCAP to therapeutic doses (up to 5.3 mg) of the protein in healthy volunteers. This study demonstrates the value of a microdosing approach in a very small cohort for accelerating the clinical development of NBEs.


Asunto(s)
Fosfatasa Alcalina/administración & dosificación , Fosfatasa Alcalina/farmacocinética , Radioisótopos de Carbono , Isoenzimas/administración & dosificación , Isoenzimas/farmacocinética , Administración Intravenosa , Adolescente , Adulto , Fosfatasa Alcalina/efectos adversos , Área Bajo la Curva , Método Doble Ciego , Cálculo de Dosificación de Drogas , Proteínas Ligadas a GPI/administración & dosificación , Proteínas Ligadas a GPI/efectos adversos , Proteínas Ligadas a GPI/farmacocinética , Semivida , Voluntarios Sanos , Humanos , Isoenzimas/efectos adversos , Modelos Lineales , Masculino , Espectrometría de Masas/métodos , Tasa de Depuración Metabólica , Modelos Biológicos , Países Bajos , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/farmacocinética , Adulto Joven
13.
J Clin Pharmacol ; 44(12): 1405-11, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15545312

RESUMEN

Inhibition of drug metabolism is generally avoided but can be useful in limited circumstances, such as reducing the formation of toxic metabolites. Acetylation is a major pathway for drug elimination that can also convert substrates into toxic species, including carcinogens. Sulfamethoxazole, a widely used antibiotic, is metabolized via arylamine N-acetyltransferase 1. p-Aminosalicylate, used for antitubercular treatment, is also metabolized by N-acetyltransferase 1 and could potentially inhibit sulfamethoxazole metabolism. Human hepatocytes from 4 donors were incubated in vitro with sulfamethoxazole and paminosalicylate at clinically achievable concentrations. p-Aminosalicylate competitively reduced the acetylation of sulfamethoxazole in vitro by 61% to 83% at 200 microM. Four healthy volunteers were studied following doses of 500 mg sulfamethoxazole either alone or during administration of paminosalicylate (4 g ter in die). Plasma concentrations of paminosalicylate exceeded 100 microM. With each subject as his or her own control, p-aminosalicylate reduced by 5-fold the ratio of plasma concentrations of acetylsulfamethoxazole relative to parent drug (P < .001). Metabolic drug-drug interaction studies in vitro successfully predicted inhibition of acetylation via N-acetyltransferase 1 in vivo. Although no specific toxic species was investigated in this work, the potential was demonstrated for improving the therapeutic index of drugs that have toxic metabolites.


Asunto(s)
Arilamina N-Acetiltransferasa/antagonistas & inhibidores , Arilamina N-Acetiltransferasa/farmacocinética , Sistemas de Liberación de Medicamentos/métodos , Sistemas de Liberación de Medicamentos/tendencias , Isoenzimas/antagonistas & inhibidores , Isoenzimas/farmacocinética , Sulfametoxazol/análogos & derivados , Adulto , Ácido Aminosalicílico/administración & dosificación , Ácido Aminosalicílico/sangre , Ácido Aminosalicílico/farmacocinética , Área Bajo la Curva , Arilamina N-Acetiltransferasa/efectos de los fármacos , Células Cultivadas , Técnicas de Cultivo/métodos , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Femenino , Hepatocitos/efectos de los fármacos , Hepatocitos/patología , Humanos , Isoenzimas/efectos de los fármacos , Masculino , Persona de Mediana Edad , Sulfametoxazol/administración & dosificación , Sulfametoxazol/antagonistas & inhibidores , Sulfametoxazol/metabolismo , Sulfametoxazol/farmacocinética
14.
Clin Chim Acta ; 258(1): 47-58, 1997 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-9049442

RESUMEN

gamma-Glutamyltransferase is eliminated from the circulation via the asialoglycoprotein receptor in liver. After purifying the enzyme from human liver, a subfractionation into differently sialylated forms was obtained using MonoQ ion exchange chromatography. The uptake of such forms from rat circulation was studied and the slowest rate was measured for the most sialylated form. To test if the uptake of the sialylated enzymes was dependent on prior desialylation in the circulation the enzyme was recovered from liver after uptake and from serum after inhibiting the uptake with asialofetuin. Analysis of these recovered forms showed no apparent alteration in charge. The enzyme is apparently eliminated without prior desialylation through available galactose units which bind with low affinity to the receptor.


Asunto(s)
Isoenzimas/sangre , Isoenzimas/farmacocinética , Ácido N-Acetilneuramínico/metabolismo , Receptores de Superficie Celular/fisiología , gamma-Glutamiltransferasa/sangre , gamma-Glutamiltransferasa/farmacocinética , Animales , Receptor de Asialoglicoproteína , Asialoglicoproteínas/metabolismo , Glicosilación , Humanos , Cinética , Masculino , Neuraminidasa/metabolismo , Ratas , Ratas Sprague-Dawley
15.
Oncol Res ; 11(6): 249-54, 1999.
Artículo en Inglés | MEDLINE | ID: mdl-10691026

RESUMEN

The action of the anticancer drug amsacrine appears to involve molecular interactions with both DNA and topoisomerase II. It has been shown previously that DNA intercalators can inhibit the action of amsacrine and several other topoisomerase II poisons, presumably as a result of interference with the DNA binding sites for the enzyme. We show here that drug molecules such as N-phenylmethanesulfonamide, which mimic the anilino side chain of amsacrine, inhibit the cytotoxicity against cultured Lewis lung murine carcinoma of amsacrine, amsacrine analogues including asulacrine and DACA (N-[2-(dimethylamino)-ethyl]acridine-4-carboxamide dihydrochloride), and etoposide. In contrast, the cytotoxicity of doxorubicin was slightly increased by co-incubation with N-phenylmethanesulfonamide. The cytotoxicity of amsacrine was also modulated in human Jurkat leukemia, HCT-8 colon, and HT-29 colon cell lines. Because o-AMSA, an amsacrine analogue containing a methoxy group in the ortho rather than in the meta position, is known to be inactive as an antitumor drug, the abilities of the ortho and meta methoxy-substituted derivatives of methyl-N-phenylcarbamate to reverse the cytotoxicity of amsacrine, asulacrine, and DACA were compared. The ortho substitution decreased activity while meta substitution slightly increased it, suggesting that the side chains were binding to a similar site to that occupied by amsacrine. To determine whether the side chain variants actively inhibited the formation of DNA-topoisomerase II covalent complexes, cultured cells were treated with amsacrine or asulacrine, harvested, and lysed directly on acrylamide gels before electrophoresis and Western blotting to identify non-DNA-bound topoisomerase II. Extractable topoisomerase II was depleted in cells incubated with amsacrine but partially restored by coculture with methyl-N-phenylcarbamate. The findings are consistent with the hypothesis that low molecular weight molecules can modulate the effects of topoisomerase II poisons by directly interacting with the enzyme.


Asunto(s)
Amsacrina/antagonistas & inhibidores , Compuestos de Anilina/farmacocinética , Antineoplásicos/antagonistas & inhibidores , ADN-Topoisomerasas de Tipo II , ADN-Topoisomerasas de Tipo II/farmacocinética , Inhibidores Enzimáticos/farmacología , Isoenzimas/farmacocinética , Amsacrina/análogos & derivados , Antígenos de Neoplasias , Supervivencia Celular/efectos de los fármacos , ADN-Topoisomerasas de Tipo II/química , Proteínas de Unión al ADN , Interacciones Farmacológicas , Células HT29 , Humanos , Isoenzimas/química , Células Jurkat , Relación Estructura-Actividad
16.
J Physiol Pharmacol ; 54(4): 643-52, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14726617

RESUMEN

Brain histamine participates in central regulation of the hypothalamic-pituitary-adrenal (HPA) axis. Endogenous prostaglandins modulate signal transduction of different neurotransmitters involved in activation of HPA axis. In the present experiment we investigated whether endogenous prostaglandins are involved in the stimulation of ACTH and corticosterone secretion by histaminergic systems in the rat brain. Histamine (50 microg), histamine-trifluoromethyl-toluidine derivative (HTMT, 75microg) a selective and potent H(1)-receptor agonist, and amthamine (50 microg) a H(2)-receptor agonist given intracerebroventricularly (i.c.v.) to non-anesthetized rats considerably increased ACTH and corticosterone secretion 1h after administration. A non-selective cyclooxygenase inhibitor indomethacin (2 mg/kg i.p. or 10 microg i.c.v.), piroxicam (0.02 and 0.2 microg i.c.v.) a more potent antagonist of constitutive cyclooxygenase (COX-1) and compound NS-398 (0.1 and 1.0 microg i.c.v.), a selective inhibitor of inducible cyclooxygenase (COX-2) were given 15 min before histamine and histamine receptor agonists. One hour after the last injection trunk blood from decapitated rats was collected for hormones determination. The histamine-induced ACTH and corticosterone secretion was significantly diminished by piroxicam and was not markedly altered by indomethacin and compound NS-398. The HTMT-elicited increase in ACTH and corticosterone secretion was significantly prevented by indomethacin and was not affected by piroxicam or compound NS-398. The amthamine-evoked increase in ACTH and corticosterone secretion was not markedly influenced by any cyclooxygenase blocker applied in the present experiment. These results indicate that the histamine H(1)-receptor transmitted central stimulation of the HPA axis is considerably mediated by prostaglandins generated by consititutive cyclooxygenase, whereas stimulation transmitted via H(2)-receptor does not significantly depend on endogenous prostaglandins mediation.


Asunto(s)
Inhibidores de la Ciclooxigenasa/farmacología , Histamina/análogos & derivados , Histamina/farmacocinética , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Sistema Hipófiso-Suprarrenal/efectos de los fármacos , Hormona Adrenocorticotrópica/antagonistas & inhibidores , Hormona Adrenocorticotrópica/sangre , Hormona Adrenocorticotrópica/metabolismo , Animales , Corticosterona/antagonistas & inhibidores , Corticosterona/sangre , Corticosterona/metabolismo , Ciclooxigenasa 1 , Ciclooxigenasa 2 , Inhibidores de la Ciclooxigenasa 2 , Inhibidores de la Ciclooxigenasa/metabolismo , Esquema de Medicación , Histamina/administración & dosificación , Histamina/fisiología , Agonistas de los Receptores Histamínicos/administración & dosificación , Agonistas de los Receptores Histamínicos/farmacocinética , Indometacina/administración & dosificación , Indometacina/farmacocinética , Inyecciones Intraperitoneales , Inyecciones Intraventriculares , Isoenzimas/administración & dosificación , Isoenzimas/antagonistas & inhibidores , Isoenzimas/farmacocinética , Masculino , Proteínas de la Membrana , Nitrobencenos/administración & dosificación , Piroxicam/administración & dosificación , Piroxicam/farmacocinética , Prostaglandina-Endoperóxido Sintasas/administración & dosificación , Prostaglandina-Endoperóxido Sintasas/farmacocinética , Ratas , Ratas Wistar , Sulfonamidas/administración & dosificación , Tiazoles/administración & dosificación , Tiazoles/farmacocinética
17.
Colloids Surf B Biointerfaces ; 35(2): 99-105, 2004 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-15261042

RESUMEN

The interfacial behavior differences of two glutathione peroxidase isoforms have been investigated. The first isoform is the phospholipid-hydroperoxide glutathione peroxidase (EC 1.11.1.12) (GPx-4) isolated from rat testes and the second one is the cytosolic glutathione peroxidase (EC 1.11.1.9) (GPx-1) from bovine erythrocytes. Injected in the subphase buffer of a Langmuir trough, GPx-4 was able to adsorb quickly at the air-water interface whereas the GPx-1 was not. Then, the protein interaction with phospholipid monolayers was explored. Indeed, a monolayer of phospholipids containing a different number of polyunsaturated fatty acyl chains was prepared at the air-water interface. Under each kind of monolayer, the protein solution was injected and its adsorption was visualized by the measurement of successive pressure-area isotherms. We have, then, determined the molecular area increase due to the protein adsorption. It was found that the GPx-4 is adsorbed in each kind of monolayer tested whereas no molecular area increase was detected with the GPx-1. This indicates that the GPx-4 has a higher affinity for the interface, recovered or not by lipids, than the GPx-1. Moreover, the GPx-4 presents a different affinity for the phospholipid monolayers depending on the number of polyunsaturated fatty acyl chains.


Asunto(s)
Aire , Glutatión Peroxidasa/química , Lípidos de la Membrana/química , Fosfolípidos/química , Agua/química , Adsorción , Animales , Bovinos , Fenómenos Químicos , Química Física , Glutatión Peroxidasa/farmacocinética , Isoenzimas/química , Isoenzimas/farmacocinética , Fosfolípido Hidroperóxido Glutatión Peroxidasa , Ratas , Propiedades de Superficie , Factores de Tiempo
18.
Ups J Med Sci ; 92(3): 293-300, 1987.
Artículo en Inglés | MEDLINE | ID: mdl-3448803

RESUMEN

Release characteristics of S-LD, S-LD1, S-ASAT, S-CK and S-CK-MB were studied in 47 consecutive AMI patients. In addition, previously obtained data for serum myoglobin (S-MYO) were compared. Serum was sampled at regular intervals after admission to the Coronary Care Unit (CCU). The release rate and half lives of the enzymes were calculated according to a one-compartment kinetic model. The time to peak values, the time of total release and the half lives were interrelated in the following order: MYO less than CK-MB less than CK less than ASAT less than LD1 less than LD which coincides with the wellknown appearance and disappearance rates in serum. The ratio between mean peak values and upper reference limits followed the reverse order. The finding that the release rate of enzymes and half-lives co-vary is hypothetically suggested to be attributed to differences in rate of membrane diffusion. There is indirect evidence that a slow indicator such as LD1 reflects infarct size better than fast indicators with rapid release and removal such as MYO or CK-MB. However, these fast markers have a better signal to noise ratio, whereby they probably reflect changes in the infarction process better.


Asunto(s)
Aspartato Aminotransferasas/sangre , Creatina Quinasa/sangre , Isoenzimas/sangre , L-Lactato Deshidrogenasa/sangre , Infarto del Miocardio/diagnóstico , Anciano , Aspartato Aminotransferasas/farmacocinética , Creatina Quinasa/farmacocinética , Femenino , Semivida , Humanos , Isoenzimas/farmacocinética , L-Lactato Deshidrogenasa/farmacocinética , Masculino , Persona de Mediana Edad , Infarto del Miocardio/enzimología
19.
Vopr Med Khim ; 40(3): 18-20, 1994.
Artículo en Ruso | MEDLINE | ID: mdl-8079432

RESUMEN

The king crab collagenase was shown to permeate through both aseptic and purulent wounds into rat circulation. The rate of enzyme permeation was higher in purulent wounds, while the enzymatic activity did not affect the rate of collagenase transport into blood. After intravenous administration the enzyme was mainly accumulated in liver and kidney, while protease was not found in the heart, spleen, lung and muscle tissues. Collagenase was rapidly eliminated from circulation and its isozymes were contained in blood not in free state, but they were bound and inactive.


Asunto(s)
Braquiuros/enzimología , Colagenasas/farmacocinética , Isoenzimas/farmacocinética , Animales , Colagenasas/uso terapéutico , Infecciones/tratamiento farmacológico , Masculino , Ratas , Distribución Tisular , Cicatrización de Heridas
20.
J Clin Pharmacol ; 54(6): 675-81, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24408305

RESUMEN

ISU303 is a new recombinant agalsidase beta (Agal) enzyme replacement therapy under investigation for Fabry disease, caused by a deficiency in α-galactosidase A activity that leads to fatty deposits in tissues. We evaluated the pharmacokinetic (PK) parameters, safety and tolerability of ISU303 in healthy adult volunteers. The study was a dose block-randomized, double-blinded, placebo-controlled, single-dosing, and dose escalation phase 1 clinical trial. A total of 18 healthy subjects were enrolled (0.3 mg/kg, n = 6; 1.0 mg/kg, n = 6; placebo, n = 6). Blood samples for PK analysis were collected according to planned time. The PK parameters in each 0.3 and 1.0 mg/kg Agal group were as follows: Cmax (mU/mL) 43.19 ± 5.9 and 195.86 ± 32.3; AUClast (h·mU/mL) 207.91 ± 25.1 and 939.96 ± 158.3; t1/2 (hours) 1.13 ± 0.3 and 1.46 ± 0.2; Cl (mL/min/kg) 1.79 ± 0.2 and 1.34 ± 0.2, respectively. There were seven adverse events (AE) overall. All AEs were resolved without any complications. None were related to the study drug. There were no immunogenicity or any significant infusion-related reactions. The new Agal product exhibited a dose-dependent PK and was well tolerated with no significant AEs in healthy adult volunteers.


Asunto(s)
Isoenzimas/farmacocinética , alfa-Galactosidasa/farmacocinética , Adulto , Método Doble Ciego , Voluntarios Sanos , Humanos , Inmunoglobulina G/sangre , Isoenzimas/efectos adversos , Isoenzimas/sangre , Isoenzimas/inmunología , Masculino , Proteínas Recombinantes/efectos adversos , Proteínas Recombinantes/sangre , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/farmacocinética , Adulto Joven , alfa-Galactosidasa/efectos adversos , alfa-Galactosidasa/sangre , alfa-Galactosidasa/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA