Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Clin Pharmacol Drug Dev ; 11(5): 640-653, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35172043

RESUMEN

This single 60-mg dose, 4-period crossover study assessed the effect of food and formulation change on navtemadlin (KRT-232) pharmacokinetics (PK) and macrophage inhibitory cytokine-1 (MIC-1) pharmacodynamics. Healthy subjects (N = 30) were randomized to 3 treatment sequences, A: new tablet, fasted (reference, dosed twice); B: new tablet, 30 minutes after a high-fat meal (test 1); C: old tablet, fasted (test 2). PK/pharmacodynamic parameters were measured over 0 to 96 hours. Adverse events were mild without any discontinuations. No serious adverse events or deaths occurred. In treatment A, navtemadlin mean (coefficient of variation) maximum concentration (Cmax ) was 525 (66) ng/mL, at median time to maximum concentration (tmax ) of 2 hours. Mean (coefficient of variation) area under the plasma concentration-time curve from time 0 to time t (AUC0-t ) was 3392 (63.3) ng • h/mL, and arithmetic mean terminal half-life was 18.6 hours. Acyl glucuronide metabolite (M1)/navtemadlin AUC0-t ratio was 0.2, and urine excretion of navtemadlin was negligible. After a meal (B vs A), navtemadlin tmax was delayed by 1 hour. Geometric least squares means ratios (90%CI) for navtemadlin Cmax and AUC0-t were 102.7% (87.4-120.6) and 81.4% (76.2-86.9), respectively. Old vs new tablet fasted formulations (C vs A) had geometric least squares means ratios (90%CI) of 78.4% (72.0-85.3) for Cmax and 85.9% (80.5-91.7) for AUC0-t . MIC-1 Cmax and AUC were comparable across groups; tmax was delayed relative to navtemadlin tmax by ≈8 hours. Navtemadlin AUC0-t and MIC-1 AUC0-t correlated significantly. In conclusion, navtemadlin can be administered safely with or without food; the new formulation does not affect navtemadlin PK. The 60-mg navtemadlin dose elicited a reproducible and robust MIC-1 response that correlated well with navtemadlin exposure, indicating that murine double minute 2 target engagement leads to p53 activation.


Asunto(s)
Interacciones Alimento-Droga , Proteínas Proto-Oncogénicas c-mdm2 , Humanos , Administración Oral , Estudios Cruzados , Citocinas , Voluntarios Sanos , Macrófagos , Comprimidos
2.
J Immunother ; 43(4): 107-120, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31899702

RESUMEN

Chimeric antigen receptor (CAR) T-cell therapy is a promising treatment for patients with CD19 B-cell malignancies. Combination strategies that improve CAR T-cell potency, limit tumor environment-mediated immune dysfunction, and directly reduce tumor burden may increase the potential for durable clinical benefit of CAR T-cell therapy. Lisocabtagene maraleucel (liso-cel) is a product therapy candidate being tested in patients with relapsed/refractory non-Hodgkin lymphoma or chronic lymphocytic leukemia. This study assessed the in vitro and in vivo functionality of CAR T cells transduced to express the anti-CD19 CAR of liso-cel in combination with ibrutinib or acalabrutinib. In prolonged stimulation assays, the presence of ibrutinib or acalabrutinib improved the CAR T-cell effector function. RNA-Seq analysis and surface marker profiling of these CAR T cells treated with ibrutinib but not acalabrutinib revealed gene expression changes consistent with skewing toward a memory-like, type 1 T-helper, Bruton tyrosine kinase phenotype. Ibrutinib or acalabrutinib improved CD19 tumor clearance and prolonged survival of tumor-bearing mice when used in combination with CAR T cells. A combination of the defined cell product therapy candidate, liso-cel, with ibrutinib or acalabrutinib is an attractive approach that may potentiate the promising clinical responses already achieved in CD19 B-cell malignancies with each of these single agents.


Asunto(s)
Antígenos CD19/inmunología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Inmunoterapia Adoptiva , Neoplasias/terapia , Receptores de Antígenos de Linfocitos T/inmunología , Receptores Quiméricos de Antígenos/inmunología , Adenina/administración & dosificación , Adenina/análogos & derivados , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Benzamidas/administración & dosificación , Biomarcadores , Terapia Combinada , Citocinas/metabolismo , Citotoxicidad Inmunológica , Modelos Animales de Enfermedad , Humanos , Inmunoterapia Adoptiva/métodos , Activación de Linfocitos/inmunología , Ratones , Neoplasias/etiología , Neoplasias/metabolismo , Piperidinas/administración & dosificación , Pirazinas/administración & dosificación , Receptores Quiméricos de Antígenos/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Eur J Pharm Biopharm ; 142: 435-448, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31306750

RESUMEN

Acalabrutinib (Calquence®) 100 mg (bid) has received accelerated approval by FDA for the treatment of adult patients with mantle cell lymphoma (MCL) who have received at least one prior therapy. Acalabrutinib is a substrate of PgP and CYP3A4, with a significant fraction of drug metabolized by first pass gut extraction and 25% absolute bioavailability. The absorption of acalabrutinib is affected by stomach pH, with lower pharmacokinetic exposure observed following co-administration with proton pump inhibitors. During dissolution at pH values below its highest basic pKa, the two basic moieties of acalabrutinib react with protons from the aqueous solution, leading to a higher pH at the drug surface than in the bulk solution. A batch-specific product particle size distribution (P-PSD), was derived from dissolution data using a mechanistic model that was based on the understanding of surface pH and the contribution of micelles to the dissolution rate. P-PSD values obtained for various batches of acalabrutinib products in simple buffers, or in complex fluids such as fruit juices, were successfully integrated into a physiologically based pharmacokinetic (PBPK) model developed using GastroPlus v9.0™. The integrated model allowed the prediction of clinical pharmacokinetics under normal physiological stomach pH conditions as well as following treatment with proton pump inhibitors. The model also accounted for lower pharmacokinetic exposure that was observed when acalabrutinib was co-administered with the acidic beverages, grapefruit juice, (which contains CYP3A inhibitors), and orange drink (which does not contain CYP3A inhibitors), relative to administration with water. The integration of dissolution data in the PBPK model enables mechanistic understanding and the establishment of more robust in vitro-in vivo correlations (IVIVC) under a variety of conditions. The model can then distinguish the interplay between dissolution and first pass extraction and how in vivo stomach pH, saturation of gut PgP, and saturation or inhibition of gut CYP3A4, will impact the pharmacokinetics of acalabrutinib.


Asunto(s)
Benzamidas/química , Benzamidas/farmacocinética , Interacciones Farmacológicas/fisiología , Jugos de Frutas y Vegetales/efectos adversos , Inhibidores de la Bomba de Protones/química , Inhibidores de la Bomba de Protones/farmacocinética , Pirazinas/química , Pirazinas/farmacocinética , Solubilidad/efectos de los fármacos , Disponibilidad Biológica , Química Farmacéutica/métodos , Humanos , Modelos Biológicos
4.
PLoS One ; 11(7): e0159607, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27434128

RESUMEN

Acalabrutinib (ACP-196) is a second-generation inhibitor of Bruton agammaglobulinemia tyrosine kinase (BTK) with increased target selectivity and potency compared to ibrutinib. In this study, we evaluated acalabrutinib in spontaneously occurring canine lymphoma, a model of B-cell malignancy similar to human diffuse large B-cell lymphoma (DLBCL). First, we demonstrated that acalabrutinib potently inhibited BTK activity and downstream effectors in CLBL1, a canine B-cell lymphoma cell line, and primary canine lymphoma cells. Acalabrutinib also inhibited proliferation in CLBL1 cells. Twenty dogs were enrolled in the clinical trial and treated with acalabrutinib at dosages of 2.5 to 20mg/kg every 12 or 24 hours. Acalabrutinib was generally well tolerated, with adverse events consisting primarily of grade 1 or 2 anorexia, weight loss, vomiting, diarrhea and lethargy. Overall response rate (ORR) was 25% (5/20) with a median progression free survival (PFS) of 22.5 days. Clinical benefit was observed in 30% (6/20) of dogs. These findings suggest that acalabrutinib is safe and exhibits activity in canine B-cell lymphoma patients and support the use of canine lymphoma as a relevant model for human non-Hodgkin lymphoma (NHL).


Asunto(s)
Antineoplásicos/administración & dosificación , Benzamidas/administración & dosificación , Linfoma de Células B Grandes Difuso/tratamiento farmacológico , Linfoma de Células B Grandes Difuso/veterinaria , Inhibidores de Proteínas Quinasas/administración & dosificación , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Pirazinas/administración & dosificación , Agammaglobulinemia Tirosina Quinasa , Animales , Anorexia/inducido químicamente , Anorexia/fisiopatología , Antineoplásicos/efectos adversos , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Linfocitos B/patología , Benzamidas/efectos adversos , Línea Celular Tumoral , Diarrea/inducido químicamente , Diarrea/fisiopatología , Modelos Animales de Enfermedad , Supervivencia sin Enfermedad , Perros , Esquema de Medicación , Evaluación Preclínica de Medicamentos , Femenino , Humanos , Letargia/inducido químicamente , Letargia/fisiopatología , Linfoma de Células B Grandes Difuso/enzimología , Linfoma de Células B Grandes Difuso/mortalidad , Masculino , Inhibidores de Proteínas Quinasas/efectos adversos , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , Pirazinas/efectos adversos , Vómitos/inducido químicamente , Vómitos/fisiopatología , Pérdida de Peso/efectos de los fármacos
5.
PLoS One ; 8(6): e67256, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23825648

RESUMEN

Rituximab, a monoclonal antibody targeting CD20 on B cells, is currently used to treat many subtypes of B cell lymphomas. However, treatment is not curative and response rates are variable. Recombinant interleukin-21 (rIL-21) is a cytokine that enhances immune effector function and affects both primary and transformed B cell differentiation. We hypothesized that the combination of rIL-21 plus rituximab would be a more efficacious treatment for B cell malignancies than rituximab alone. We cultured human and cynomolgus monkey NK cells with rIL-21 and found that their activity was increased and proteins associated with antibody dependent cytotoxicity were up-regulated. Studies in cynomolgus monkeys modeled the effects of rIL-21 on rituximab activity against CD20 B cells. In these studies, rIL-21 activated innate immune effectors, increased ADCC and mobilized B cells into peripheral blood. When rIL-21 was combined with rituximab, deeper and more durable B cell depletion was observed. In another series of experiments, IL-21 was shown to have direct antiproliferative activity against a subset of human lymphoma cell lines, and combination of murine IL-21 with rituximab yielded significant survival benefits over either agent alone in xenogeneic mouse tumor models of disseminated lymphoma. Therefore, our results do suggest that the therapeutic efficacy of rituximab may be improved when used in combination with rIL-21.


Asunto(s)
Anticuerpos Monoclonales de Origen Murino/farmacología , Antineoplásicos/farmacología , Linfocitos B/citología , Linfocitos B/efectos de los fármacos , Interleucinas/farmacología , Linfoma de Células B/tratamiento farmacológico , Linfoma de Células B/inmunología , Animales , Anticuerpos Monoclonales de Origen Murino/uso terapéutico , Citotoxicidad Celular Dependiente de Anticuerpos/efectos de los fármacos , Antineoplásicos/uso terapéutico , Linfocitos B/inmunología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Femenino , Humanos , Inmunidad Innata/efectos de los fármacos , Linfoma de Células B/patología , Macaca fascicularis , Masculino , Ratones , Rituximab , Análisis de Supervivencia
6.
J Immunotoxicol ; 10(4): 361-72, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23384295

RESUMEN

Non-human primates may be the only relevant species for pharmacology or toxicology studies of certain biologics, due to lack of activity in other species. Flow cytometry immunophenotyping is often included as a minimally invasive adjunct to standard toxicity testing. A retrospective inter-laboratory analysis was conducted to assess counts and variability of the main cell types monitored in toxicity studies, and to provide guidance for conduct and interpretation of immunophenotyping assessments in cynomolgus monkeys. Univariate and multivariate models were developed. Study design factors influencing cell counts and variability were identified and a power analysis was performed. Pre-study and on-study counts were generally similar; longitudinal analysis showed little drift in mean counts or within-animal variability over time. Within-animal variability was lower than inter-animal variability. Gender was associated with small but significant differences in mean counts and variability. Age was associated with significant differences in variability. Immunophenotype definitions were associated with significant differences in mean counts and within-animal variability for most cell types. Power analysis for groups of 6-8 animals showed that differences of ≈50% in counts of T-cells, T-cell subsets, and B-cells compared to pre-treatment values may be detected; for NK cells and monocytes, differences of ≈60-90% may be detected. This review yields some general points to consider for immunophenotyping studies, i.e. (a) analysis of log-transformed cell count data and comparisons using each animal as its own reference will improve ability to detect changes, (b) the magnitude of change detectable given study group size should be considered, (c) multiplication of sampling timepoints during a study seems unnecessary, (d) consideration should be given to using only one gender, when applicable, to increase power while minimizing animal usage, and (e) the choice of immunophenotype has impacts on cell counts and variability.


Asunto(s)
Linfocitos B/inmunología , Células Asesinas Naturales/inmunología , Subgrupos Linfocitarios/inmunología , Monocitos/inmunología , Linfocitos T/inmunología , Factores de Edad , Animales , Recuento de Células , Separación Celular , Citometría de Flujo , Humanos , Inmunofenotipificación/métodos , Macaca fascicularis , Modelos Animales , Variaciones Dependientes del Observador , Guías de Práctica Clínica como Asunto , Estudios Retrospectivos , Factores Sexuales
7.
J Immunotoxicol ; 8(3): 238-50, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21692639

RESUMEN

The Immunotoxicology Technical Committee of HESI sponsored a retrospective analysis of T-cell-dependent antibody responses in non-human primates (NHP). Antibody responses to keyhole limpet hemocyanin (KLH), tetanus toxoid (TT), and/or sheep red blood cells (SRBC) in 178 NHP (from 8 sponsors, 13 testing sites, 30 studies) were statistically analyzed. Rates of positive or negative anti-KLH, -TT, and -SRBC primary and secondary IgM and IgG responses were compared. The influence of gender, country of origin, and previous immunization with a different antigen on response rate and kinetics of anti-KLH and anti-TT responses were analyzed. In addition, the magnitude of the antibody responses and the impact of the above-mentioned factors were analyzed. In addition, based upon the inter-individual variability of the peak response values, power calculations were conducted. The analysis demonstrated that the rates of positive responses were similar between the two genders, were high for KLH, SRBC, and TT challenges by 21 days following immunization (87, 100, and 84%, respectively, for IgGs) and did not include statistically significant differences based on NHP country of origin. Mean peak secondary responses were greater than peak primary responses; the magnitude of the response to KLH was increased by incomplete Freund's adjuvant (IFA). Gender had little effect on the magnitude and variability of these responses. KLH and TT were associated with similar inter-animal variability, whereas in some situations KLH responses were less variable than responses to SRBC. The data suggested that inter-animal variability with KLH was similar with or without IFA. Power analysis illustrated that animal group sizes of typical standard toxicology studies (generally ≤ 4/sex) are likely to detect only fairly large treatment effects. However, combining males and females, when appropriate, will improve the power: an N of 8 to 12 could detect ≤ 3.1-fold differences in anti-KLH IgG responses.


Asunto(s)
Formación de Anticuerpos/inmunología , Proyectos de Investigación , Linfocitos T/inmunología , Animales , Interpretación Estadística de Datos , Determinación de Punto Final , Eritrocitos/inmunología , Femenino , Hemocianinas/inmunología , Inmunoglobulina G/sangre , Inmunoglobulina M/sangre , Técnicas Inmunológicas , Macaca fascicularis , Masculino , Ovinos , Especificidad de la Especie , Toxoide Tetánico/inmunología , Toxicología/métodos
8.
Arthritis Res Ther ; 12(2): R48, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20302641

RESUMEN

INTRODUCTION: B-lymphocyte stimulator (BLyS) and a proliferation-inducing ligand (APRIL) are members of the tumor necrosis factor (TNF) family that regulate B-cell maturation, survival, and function. They are overexpressed in a variety of autoimmune diseases and reportedly exist in vivo not only as homotrimers, but also as BLyS/APRIL heterotrimers. METHODS: A proprietary N-terminal trimerization domain was used to produce recombinant BLyS/APRIL heterotrimers. Heterotrimer biologic activity was compared with that of BLyS and APRIL in a 4-hour signaling assay by using transmembrane activator and CAML interactor (TACI)-transfected Jurkat cells and in a 4-day primary human B-cell proliferation assay. A bead-based immunoassay was developed to quantify native heterotrimers in human sera from healthy donors (n = 89) and patients with systemic lupus erythematosus (SLE; n = 89) or rheumatoid arthritis (RA; n = 30). Heterotrimer levels were compared with BLyS and APRIL homotrimer levels in a subset of these samples. RESULTS: The recombinant heterotrimers consisted mostly of one BLyS and two APRIL molecules. Heterotrimer signaling did not show any significant difference compared with APRIL in the TACI-Jurkat assay. Heterotrimers were less-potent inducers of B-cell proliferation than were homotrimeric BLyS or APRIL (EC(50), nMol/L: BLyS, 0.02; APRIL, 0.17; heterotrimers, 4.06). The soluble receptor fusion proteins atacicept and B-cell maturation antigen (BCMA)-immunoglobulin (Ig) neutralized the activity of BLyS, APRIL, and heterotrimers in both cellular assays, whereas B-cell activating factor belonging to the TNF family receptor (BAFF-R)-Ig neutralized only the activity of BLyS. In human sera, significantly more patients with SLE had detectable BLyS (67% versus 18%; P < 0.0001), APRIL (38% versus 3%; P < 0.0002), and heterotrimer (27% versus 8%; P = 0.0013) levels compared with healthy donors. Significantly more patients with RA had detectable APRIL, but not BLyS or heterotrimer, levels compared with healthy donors (83% versus 3%; P < 0.0001). Heterotrimer levels weakly correlated with BLyS, but not APRIL, levels. CONCLUSIONS: Recombinant BLyS/APRIL heterotrimers have biologic activity and are inhibited by atacicept and BCMA-Ig, but not by BAFF-R-Ig. A novel immunoassay demonstrated that native BLyS/APRIL heterotrimers, as well as BLyS and APRIL homotrimers, are elevated in patients with autoimmune diseases.


Asunto(s)
Enfermedades Autoinmunes/sangre , Factor Activador de Células B/sangre , Antígeno de Maduración de Linfocitos B/farmacología , Proliferación Celular/efectos de los fármacos , Proteínas Recombinantes de Fusión/farmacología , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/sangre , Linfocitos B/citología , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Humanos , Interleucina-4/farmacología , Células Jurkat , Activación de Linfocitos/efectos de los fármacos , Multimerización de Proteína , Proteínas Recombinantes
9.
J Biol Chem ; 285(21): 16116-24, 2010 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-20332089

RESUMEN

Glutamate cysteine ligase (GCL) catalyzes the rate-limiting step in the formation of the cellular antioxidant glutathione (GSH). The GCL holoenzyme consists of two separately coded proteins, a catalytic subunit (GCLC) and a modifier subunit (GCLM). Both GCLC and GLCM are controlled transcriptionally by a variety of cellular stimuli, including oxidative stress. This study addresses post-translational control of GCL activity, which increased rapidly in human lymphocytes following oxidative stress. Activation of GCL occurred within minutes of treatment and without any change in GCL protein levels and coincided with an increase in the proportion of GCLC in the holoenzyme form. Likewise, GCLM shifted from the monomeric form to holoenzyme and higher molecular weight species. Normal rat tissues also showed a distribution of monomeric and higher molecular weight forms. Neither GCL activation, nor the formation of holoenzyme, required a covalent intermolecular disulfide bridge between GCLC and GCLM. However, in immunoprecipitation studies, a neutralizing epitope associated with enzymatic activity was protected following cellular oxidative stress. Thus, the N-terminal portion of GCLC may undergo a change that stabilizes the GCL holoenzyme. Our results suggest that a dynamic equilibrium exists between low and high activity forms of GCL and is altered by transient oxidative stress. This provides a mechanism for the rapid post-translational activation of GCL and maintenance of cellular GSH homeostasis.


Asunto(s)
Glutamato-Cisteína Ligasa/metabolismo , Glutatión/metabolismo , Homeostasis/fisiología , Estrés Oxidativo/fisiología , Transcripción Genética/fisiología , Animales , Disulfuros/metabolismo , Activación Enzimática/fisiología , Holoenzimas/metabolismo , Humanos , Células Jurkat , Linfocitos/enzimología , Ratones , Especificidad de Órganos/fisiología , Estructura Terciaria de Proteína , Ratas
10.
Nat Rev Drug Discov ; 5(6): 507-21, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16763661

RESUMEN

Pharmacogenetic studies have traditionally focused on genes involved in processes that affect the pharmacokinetics of small-molecule drugs, such as drug metabolism. However, attention is shifting to the effects of genetic variations in drug targets and associated pathway components on drug responses. We describe how these variations are important for understanding differences in responses to the growing number of protein therapeutics that are entering clinical practice. Pharmacogenetic studies of these drugs are surveyed, and issues important to the success of such endeavours are discussed. As novel protein therapeutics are introduced, we anticipate that the use of pharmacogenetics will assume a key role in their development and clinical application.


Asunto(s)
Farmacogenética , Proteínas/uso terapéutico , Ensayos Clínicos como Asunto , Diseño de Fármacos , Variación Genética , Genotipo , Humanos , Proteínas/farmacocinética
11.
Reprod Toxicol ; 19(1): 117-29, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15336720

RESUMEN

The placenta and the yolk sac play critical roles in fetal development, including protection from oxidative stress through the presence of detoxifying enzymes. Glutathione (GSH; gamma-glutamylcysteinylglycine), a crucial molecule in the maintenance of cellular redox status, plays a critical role in development, and it is also protective against methylmercury toxicity. Glutamate-cysteine ligase (GCL), the enzyme that catalyzes the rate-limiting step in GSH synthesis, is widely expressed in the mouse embryo and extraembryonic membranes throughout development. The aim of this study was to investigate the effect of low-level subchronic methylmercury exposure on GCL expression in the mouse placenta and yolk sac, after describing the basal developmental expression of the enzyme in these tissues. We found that basal mRNA expression levels increased dramatically in the placenta and the yolk sac at gd 18, whereas protein levels did not increase in parallel with the mRNA. We also found that methylmercury induced GCLc mRNA expression in the placenta at gd 18 in a dose-dependent manner, suggesting an important role for this enzyme in the response of the placenta to toxicants. These changes in expression may be useful as a biomarker of MeHg exposure during development.


Asunto(s)
Contaminantes Ambientales/toxicidad , Glutamato-Cisteína Ligasa/biosíntesis , Compuestos de Metilmercurio/toxicidad , Placenta/efectos de los fármacos , Saco Vitelino/efectos de los fármacos , Administración Oral , Animales , Relación Dosis-Respuesta a Droga , Contaminantes Ambientales/administración & dosificación , Femenino , Técnica del Anticuerpo Fluorescente Indirecta , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Glutamato-Cisteína Ligasa/genética , Hibridación in Situ , Masculino , Compuestos de Metilmercurio/administración & dosificación , Ratones , Placenta/enzimología , Embarazo , ARN Mensajero/metabolismo , Abastecimiento de Agua , Saco Vitelino/enzimología
12.
Free Radic Biol Med ; 37(5): 632-42, 2004 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-15288121

RESUMEN

Glutathione (GSH) is important in free radical scavenging, maintaining cellular redox status, and regulating cell survival in response to a wide variety of toxicants. The rate-limiting enzyme in GSH synthesis is glutamate-cysteine ligase (GCL), which is composed of catalytic (GCLC) and modifier (GCLM) subunits. To determine whether increased GSH biosynthetic capacity enhances cellular resistance to tumor necrosis factor-alpha- (TNF-alpha-) induced apoptotic cell death, we have established several mouse liver hepatoma (Hepa-1) cell lines overexpressing GCLC and/or GCLM. Cells overexpressing GCLC alone exhibit modest increases in GCL activity, while cells overexpressing both subunits have large increases in GCL activity. Importantly, cells overexpressing both GCL subunits exhibit increased resistance to TNF-induced apoptosis as judged by a loss of redox potential; mitochondrial membrane potential; translocation of cytochrome c to the cytoplasm; and activation of caspase-3, caspase-8, and caspase-9. Analysis of the effects of TNF on these parameters indicates that maintaining mitochondrial integrity mediates this protective effect in GCL-overexpressing cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Glutamato-Cisteína Ligasa/metabolismo , Mitocondrias/patología , Factor de Necrosis Tumoral alfa/toxicidad , Animales , Carcinoma Hepatocelular , Línea Celular Tumoral , Glutatión/metabolismo , Humanos , Neoplasias Hepáticas , Ratones , Mitocondrias/efectos de los fármacos , Proteínas Recombinantes/metabolismo , Transfección
13.
Curr Opin Drug Discov Devel ; 6(4): 470-80, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12951810

RESUMEN

Computational methods are increasingly used to streamline and enhance the lead discovery and optimization process. However, accurate prediction of absorption, distribution, metabolism and excretion (ADME) and adverse drug reactions (ADR) is often difficult, due to the complexity of underlying physiological mechanisms. Modeling approaches have been hampered by the lack of large, robust and standardized training datasets. In an extensive effort to build such a dataset, the BioPrint database was constructed by systematic profiling of nearly all drugs available on the market, as well as numerous reference compounds. The database is composed of several large datasets: compound structures and molecular descriptors, in vitro ADME and pharmacology profiles, and complementary clinical data including therapeutic use information, pharmacokinetics profiles and ADR profiles. These data have allowed the development of computational tools designed to integrate a program of computational chemistry into library design and lead development. Models based on chemical structure are strengthened by in vitro results that can be used as additional compound descriptors to predict complex in vivo endpoints. The BioPrint pharmacoinformatics platform represents a systematic effort to accelerate the process of drug discovery, improve quantitative structure-activity relationships and develop in vitro/in vivo associations. In this review, we will discuss the importance of training set size and diversity in model development, the implementation of linear and neighborhood modeling approaches, and the use of in silico methods to predict potential clinical liabilities.


Asunto(s)
Biología Computacional/métodos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Preparaciones Farmacéuticas/metabolismo , Farmacocinética , Animales , Inteligencia Artificial , Inhibidores del Citocromo P-450 CYP2D6 , Sinergismo Farmacológico , Inhibidores Enzimáticos/farmacología , Humanos , Modelos Moleculares , Valor Predictivo de las Pruebas , Relación Estructura-Actividad Cuantitativa
14.
Anal Biochem ; 318(2): 175-80, 2003 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-12814619

RESUMEN

Glutamate-cysteine ligase (GCL; also known as gamma-glutamylcysteine synthetase) is the rate-limiting enzyme in glutathione (GSH) synthesis. Traditional assays for the activity of this enzyme are based either on coupled reactions with other enzymes or on high-performance liquid chromatography (HPLC) assessment of gamma-glutamylcysteine (gamma-GC) product formation. We took advantage of the reaction of naphthalene dicarboxaldehyde (NDA) with GSH or gamma-GC to form cyclized products that are highly fluorescent. Hepa-1 cells which were designed to overexpress mouse GCL and mouse liver homogenates were used to evaluate and compare the utility of the NDA method with an assay based on monobromobimane derivatization and HPLC analysis with fluorescence detection. Excellent agreement was found between GCL activities measured by HPLC and NDA-microtiter plate analyses. This assay should be useful for high-throughput GCL activity analyses.


Asunto(s)
Glutamato-Cisteína Ligasa/análisis , Glutamato-Cisteína Ligasa/metabolismo , Animales , Células Cultivadas , Fluorescencia , Concentración de Iones de Hidrógeno , Hígado/citología , Hígado/enzimología , Extractos Hepáticos , Ratones , Microquímica/métodos , Factores de Tiempo
15.
Am J Pathol ; 160(5): 1887-94, 2002 May.
Artículo en Inglés | MEDLINE | ID: mdl-12000740

RESUMEN

Apoptotic cell death is usually accompanied by activation of a family of cysteine proteases termed caspases. Caspases mediate the selective proteolysis of multiple cellular targets often resulting in the disruption of survival pathways. Intracellular levels of the antioxidant glutathione (GSH) are an important determinant of cellular susceptibility to apoptosis. The rate-limiting step in GSH biosynthesis is mediated by glutamate-L-cysteine ligase (GCL), a heterodimeric enzyme consisting of a catalytic (GCLC) and a modifier (GCLM) subunit. In this report we demonstrate that GCLC is a direct target for caspase-mediated cleavage in multiple models of apoptotic cell death. Mutational analysis revealed that caspase-mediated cleavage of GCLC occurs at Asp(499) within the sequence AVVD(499)G. GCLC cleavage occurs upstream of Cys(553), which is thought to be important for association with GCLM. GCLC cleavage is accompanied by a rapid loss of intracellular GSH due to caspase-mediated extrusion of GSH from the cell. However, while GCLC cleavage is dependent on caspase-3, GSH extrusion occurs by a caspase-3-independent mechanism. Our identification of GCLC as a target for caspase-3-dependent cleavage during apoptotic cell death suggests that this post-translational modification may represent a novel mechanism for regulating GSH biosynthesis during apoptosis.


Asunto(s)
Apoptosis/fisiología , Caspasas/metabolismo , Glutamato-Cisteína Ligasa/metabolismo , Animales , Apoptosis/efectos de los fármacos , Secuencia de Bases , Sitios de Unión/genética , Caspasa 3 , Dominio Catalítico , Relación Dosis-Respuesta a Droga , Activación Enzimática , Glutamato-Cisteína Ligasa/genética , Glutatión/metabolismo , Células HeLa , Humanos , Células Jurkat , Ratones , Receptores de Superficie Celular/fisiología , Factores de Tiempo , Células Tumorales Cultivadas , Factor de Necrosis Tumoral alfa/farmacología
16.
Mol Reprod Dev ; 62(1): 83-91, 2002 May.
Artículo en Inglés | MEDLINE | ID: mdl-11933164

RESUMEN

The tripeptide glutathione (GSH), which plays a crucial role in protecting cells against oxidative stress, is synthesized in a two-step process. The rate-limiting step is the binding of glutamate and cysteine, which is catalyzed by the enzyme glutamate-cysteine ligase (GCL). This enzyme is composed of two subunits: a large catalytic subunit (GCLc) and a smaller modifying subunit (GCLm), originating from different genes. Control of cellular GSH levels is essential for normal development. In the current study, we investigated the tissue distribution of Gclc and Gclm transcripts, as well as GCLc protein, in the developing mouse embryo. We found that both mRNAs were highly expressed in the liver and CNS at gestational day 10 (gd 10) and gd 12, with Gclm being more abundant than Gclc in the liver relative to other tissues. Also, the expression of the two subunit mRNAs was not always parallel in the embryo, in that some tissues expressed one of the subunits preferentially, suggesting that the two genes are differentially expressed during mouse development. The GCLc protein was also widely expressed throughout the embryo, and, in general, it co-localized with the Gclc mRNA.


Asunto(s)
Expresión Génica , Glutamato-Cisteína Ligasa/genética , Tejido Adiposo Pardo/enzimología , Animales , Dominio Catalítico , Sistema Nervioso Central/embriología , Sistema Nervioso Central/enzimología , Desarrollo Embrionario y Fetal , Femenino , Hígado/embriología , Hígado/enzimología , Ratones , Ratones Endogámicos C57BL , Embarazo , ARN Mensajero , Glándulas Salivales/embriología , Glándulas Salivales/enzimología , Distribución Tisular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...