Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Front Microbiol ; 12: 678012, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34194413

RESUMEN

This study focused on the microbial and bacteriophages identification and characterization in cheese-production facilities that use natural whey starter (NWS) cultures for Trentingrana production. Bacterial and phage screening was carried out on cooked not acidified whey and NWS samples isolated from six dairy factories, for 4 consecutive days in four different months. By means of a combined approach, using plate counts, bacterial isolation, and metataxonomic analysis Lactobacillus helveticus was found occurring as the dominant species in NWS cultures and Levilactobacillus brevis as codominant in the cheese factories where the temperature of NWS production was mainly lower than 40°C, suggesting that the variability in the parameters of the NWS culture preparation could differently modulate the bacterial species in NWS cultures. Using turbidity test approach on 303 bacterial isolates from the NWS cultures, 120 distinct phages were identified. L. helveticus phage contamination of NWS cultures was revealed in most of the analyzed samples, but despite the great recovery of bacteriophage contamination cases, the microbial quality of NWS cultures was high. Our results support the presence of natural bacteriophage resistance mechanisms in L. helveticus. The use of NWS cultures probably creates an ideal environment for the proliferation of different L. helveticus strains balanced with their phages without a clear dominance. It is evident, from this study, that the presence of a high biodiversity of NWS bacterial strains is relevant to avoid phages dominance in NWS cultures and consequently to keep a good acidification ability.

2.
J Lipid Res ; 55(6): 1052-65, 2014 06.
Artículo en Inglés | MEDLINE | ID: mdl-24771867

RESUMEN

Thyroid hormone responsive protein Spot 14 has been consistently associated with de novo fatty acid synthesis activity in multiple tissues, including the lactating mammary gland, which synthesizes large quantities of medium chain fatty acids (MCFAs) exclusively via FASN. However, the molecular function of Spot14 remains undefined during lactation. Spot14-null mice produce milk deficient in total triglyceride and de novo MCFA that does not sustain optimal neonatal growth. The lactation defect was rescued by provision of a high fat diet to the lactating dam. Transgenic mice overexpressing Spot14 in mammary epithelium produced total milk fat equivalent to controls, but with significantly greater MCFA. Spot14-null dams have no diminution of metabolic gene expression, enzyme protein levels, or intermediate metabolites that accounts for impaired de novo MCFA. When [(13)C] fatty acid products were quantified in vitro using crude cytosolic lysates, native FASN activity was 1.6-fold greater in control relative to Spot14-null lysates, and add back of Spot14 partially restored activity. Recombinant FASN catalysis increased 1.4-fold and C = 14:0 yield was enhanced 4-fold in vitro following addition of Spot14. These findings implicate Spot14 as a direct protein enhancer of FASN catalysis in the mammary gland during lactation when maximal MCFA production is needed.


Asunto(s)
Acido Graso Sintasa Tipo I/metabolismo , Lactancia/fisiología , Glándulas Mamarias Animales/metabolismo , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , Animales , Catálisis , Acido Graso Sintasa Tipo I/genética , Femenino , Ratones , Ratones Mutantes , Proteínas Nucleares/genética , Factores de Transcripción/genética
3.
PLoS One ; 8(8): e73267, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24039668

RESUMEN

The unfolded protein response (UPR) is an endoplasmic reticulum (ER)-based cytoprotective mechanism acting to prevent pathologies accompanying protein aggregation. It is frequently active in tumors, but relatively unstudied in gliomas. We hypothesized that UPR stress effects on glioma cells might protect tumors from additional exogenous stress (ie, chemotherapeutics), postulating that protection was concurrent with altered tumor cell metabolism. Using human brain tumor cell lines, xenograft tumors, human samples and gene expression databases, we determined molecular features of glioma cell UPR induction/activation, and here report a detailed analysis of UPR transcriptional/translational/metabolic responses. Immunohistochemistry, Western and Northern blots identified elevated levels of UPR transcription factors and downstream ER chaperone targets in gliomas. Microarray profiling revealed distinct regulation of stress responses between xenograft tumors and parent cell lines, with gene ontology and network analyses linking gene expression to cell survival and metabolic processes. Human glioma samples were examined for levels of the ER chaperone GRP94 by immunohistochemistry and for other UPR components by Western blotting. Gene and protein expression data from patient gliomas correlated poor patient prognoses with increased expression of ER chaperones, UPR target genes, and metabolic enzymes (glycolysis and lipogenesis). NMR-based metabolomic studies revealed increased metabolic outputs in glucose uptake with elevated glycolytic activity as well as increased phospholipid turnover. Elevated levels of amino acids, antioxidants, and cholesterol were also evident upon UPR stress; in particular, recurrent tumors had overall higher lipid outputs and elevated specific UPR arms. Clonogenicity studies following temozolomide treatment of stressed or unstressed cells demonstrated UPR-induced chemoresistance. Our data characterize the UPR in glioma cells and human tumors, and link the UPR to chemoresistance possibly via enhanced metabolism. Given the role of the UPR in the balance between cell survival and apoptosis, targeting the UPR and/or controlling metabolic activity may prove beneficial for malignant glioma therapeutics.


Asunto(s)
Resistencia a Antineoplásicos , Glioma/metabolismo , Respuesta de Proteína Desplegada , Animales , Antineoplásicos Alquilantes/farmacología , Línea Celular Tumoral , Dacarbazina/análogos & derivados , Dacarbazina/farmacología , Modelos Animales de Enfermedad , Resistencia a Antineoplásicos/genética , Estrés del Retículo Endoplásmico/genética , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Glioma/genética , Glioma/mortalidad , Glioma/patología , Humanos , Lipogénesis , Ratones , Chaperonas Moleculares/metabolismo , Clasificación del Tumor , Pronóstico , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal , Temozolomida , Factores de Transcripción/metabolismo , Transcripción Genética , Respuesta de Proteína Desplegada/genética , Ensayos Antitumor por Modelo de Xenoinjerto
4.
PLoS One ; 8(3): e58089, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23520486

RESUMEN

PURPOSE: The PI3K/AKT/mTOR pathway is frequently dysregulated in cancers and inhibition of mTOR has demonstrated the ability to modulate pro-survival pathways. As such, we sought to determine the ability of the mTOR inhibitor everolimus to potentiate the antitumor effects of irinotecan in colorectal cancer (CRC). EXPERIMENTAL DESIGN: The combinatorial effects of everolimus and irinotecan were evaluated in vitro and in vivo in CRC cell lines harboring commonly found mutations in PIK3CA, KRAS and/or BRAF. Pharmacokinetically-directed dosing protocols of everolimus and irinotecan were established and used to assess the in vivo antitumor effects of the agents. At the end of treatment, 3-6 tumors per treatment arm were harvested for biomarker analysis by NMR metabolomics. RESULTS: Everolimus and irinotecan/SN38 demonstrated synergistic anti-proliferative effects in multiple CRC cell lines in vitro. Combination effects of everolimus and irinotecan were determined in CRC xenograft models using clinically-relevant dosing protocols. Everolimus demonstrated significant tumor growth inhibition alone and when combined with irinotecan in HT29 and HCT116 tumor xenografts. Metabolomic analysis showed that HT29 tumors were more metabolically responsive than HCT116 tumors. Everolimus caused a decrease in glycolysis in both tumor types whilst irinotecan treatment resulted in a profound accumulation of lipids in HT29 tumors indicating a cytotoxic effect. CONCLUSIONS: Quantitative analysis of tumor growth and metabolomic data showed that the combination of everolimus and irinotecan was more beneficial in the BRAF/PIK3CA mutant HT29 tumor xenografts, which had an additive effect, than the KRAS/PIK3CA mutant HCT116 tumor xenografts, which had a less than additive effect.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Camptotecina/análogos & derivados , Neoplasias del Colon/tratamiento farmacológico , Inmunosupresores/farmacología , Sirolimus/análogos & derivados , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Antineoplásicos Fitogénicos/agonistas , Camptotecina/agonistas , Camptotecina/farmacología , Línea Celular Tumoral , Fosfatidilinositol 3-Quinasa Clase I , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Sinergismo Farmacológico , Everolimus , Femenino , Humanos , Inmunosupresores/agonistas , Irinotecán , Metaboloma/efectos de los fármacos , Metaboloma/genética , Ratones , Ratones Desnudos , Mutación , Trasplante de Neoplasias , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , Proteínas Proto-Oncogénicas p21(ras) , Sirolimus/agonistas , Sirolimus/farmacología , Trasplante Heterólogo , Proteínas ras/genética , Proteínas ras/metabolismo
5.
Biomark Med ; 3(3): 289-306, 2009 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-20160999

RESUMEN

Cancer cells possess a highly unique metabolic phenotype, which is characterized by high glucose uptake, increased glycolytic activity, decreased mitochondrial activity, low bioenergetic and increased phospholipid turnover. These metabolic hallmarks can be readily assessed by metabolic technologies - either in vitro or in vivo - to monitor responsiveness and resistance to novel targeted drugs, where specific inhibition of cell proliferation (cytostatic effect) occurs rather than direct induction of cell death (cytotoxicity). Using modern analytical technologies in combination with statistical approaches, 'metabolomics', a global metabolic profile on patient samples can be established and validated for responders and nonresponders, providing additional metabolic end points. Discovered metabolic end points should be translated into noninvasive metabolic imaging protocols.

6.
Cancer Chemother Pharmacol ; 61(2): 179-88, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17393165

RESUMEN

PURPOSE: Vandetanib (ZD6474, Zactima) is a novel, orally available inhibitor of vascular endothelial growth factor receptor-2 (VEGFR2) tyrosine kinase activity with additional activity against epidermal growth factor receptor (EGFR) tyrosine kinase. Vandetanib has demonstrated enhanced efficacy in combination with radiation therapy (RT) in human tumor models. This study aimed to evaluate the schedule-dependent interaction of clinically relevant dosing of vandetanib with RT in human head and neck cancer models that had been characterized as EGFR positive (EGFR+) or negative (EGFR-) in order to begin differentiating vandetanib and RT interactions at the level of antitumor (EGFR) or antivascular (VEGFR2) activities. METHODS: The human head and neck squamous cell carcinoma (HNSCC) cell lines UMSCC2 (EGFR+) and UMSCC10 (EGFR(-)) are sensitive and resistant to EGFR inhibitors, respectively, while having similar sensitivity to ionizing radiation. Nude mice with UMSCC2 or UMSCC10 tumor xenografts were treated with vandetanib or RT alone, or with combinations of concomitant and sequential therapy. Vandetanib was dosed at 30 mg kg(-1) day(-1) based on pharmacokinetic studies in nude mice showing that this dose results in drug exposure similar to that seen in humans at clinical doses. RT was dosed at 3 Gy twice a week for two consecutive weeks for a total dose of 12 Gy. RESULTS: Vandetanib alone caused regression in EGFR+ but not EGFR- tumors and RT therapy alone was similar in both tumor types. Combinations of vandetanib and RT showed concomitant use of vandetanib and RT was superior to RT followed by vandetanib or visa versa in EGFR- tumors. Therapeutic response of EGFR+ tumors was similar regardless of treatment sequencing. CONCLUSIONS: The combination of vandetanib and RT is active in both EGFR+ and EGFR- HNSCC tumor xenografts, however, vandetanib alone is only active in EGFR+ xenografts. EGFR+ tumor response to vandetanib and RT was independent of treatment sequencing, but concomitant treatment was superior to sequencing in EGFR- tumors. These results suggest that the anti-VEGFR2 antitumor activity of vandetanib is enhanced by RT as presumably the activity seen in EGFR- tumors is due to antiangiogenic activity, whereas the anti-EGFR antitumor activity dominates in EGFR+ tumors such that RT enhancement is not observed.


Asunto(s)
Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Neoplasias de Cabeza y Cuello/terapia , Piperidinas/administración & dosificación , Piperidinas/uso terapéutico , Quinazolinas/administración & dosificación , Quinazolinas/uso terapéutico , Receptores de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Animales , Western Blotting , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Terapia Combinada , Gefitinib , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Neoplasias de Cabeza y Cuello/radioterapia , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Trasplante de Neoplasias , Piperidinas/efectos adversos , Quinazolinas/efectos adversos , Transducción de Señal/efectos de los fármacos , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Clin Cancer Res ; 13(21): 6450-8, 2007 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-17975157

RESUMEN

PURPOSE: This in vivo study was designed to determine the optimal doses and schedules of vandetanib, a dual epidermal growth factor receptor (EGFR)-vascular endothelial growth factor receptor tyrosine kinase inhibitor, in combination with irinotecan in a murine xenograft model of human colon cancer. EXPERIMENTAL DESIGN: HT-29 tumor-bearing nude mice were treated with two doses of vandetanib (12.5 and 25 mg/kg/d) with or without irinotecan (100 mg/kg) using either sequential or concurrent schedules for 30 days. Tumor size was measured using standard variables, whereas the antiangiogenic response was evaluated using dynamic contrast-enhanced magnetic resonance imaging. Additionally, effects on EGFR-dependent signal transduction pathways and proliferation were assessed using immunohistochemistry. These pharmacodynamic end points were then evaluated for associations with antitumor efficacy and/or to plasma/tumor concentrations of vandetanib. RESULTS: The greatest antitumor efficacy was observed in the groups receiving the highest dose of vandetanib given continuously (concurrent schedule), alone or in combination with irinotecan. These dosing schedules resulted in significant effects on tumor vasculature, with decreased volume transfer constants, area under the curve, and permeability surface factor as well as increased gadolinium clearance after 30 days of treatment. In addition, these groups showed the greatest inhibition of EGFR signaling. Interestingly, tumor concentrations of vandetanib were increased by irinotecan in the concurrent schedule, possibly due to decreased tumor perfusion in this group. CONCLUSIONS: These data suggest that higher, sustained concentrations of vandetanib (versus intermittent), alone and in combination with irinotecan, result in optimal antitumor efficacy in this model and may have implications for the design of future clinical studies with this drug.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Camptotecina/análogos & derivados , Receptores ErbB/antagonistas & inhibidores , Piperidinas/administración & dosificación , Quinazolinas/administración & dosificación , Receptores de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Animales , Camptotecina/administración & dosificación , Línea Celular Tumoral , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/patología , Esquema de Medicación , Femenino , Humanos , Irinotecán , Imagen por Resonancia Magnética/métodos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Trasplante de Neoplasias , Resultado del Tratamiento
8.
J Pharmacol Exp Ther ; 318(2): 872-80, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16644900

RESUMEN

ZD6474 [N-(4-bromo-2-fluorophenyl)-6-methoxy-7-[(1-methylpiperidin-4-yl)methoxy]-quinazolin-4-amine; Zactima] is a tyrosine kinase inhibitor with antiangiogenic and antitumor activity currently undergoing human trials for cancer treatment. Pharmacokinetic studies in animal models are an important component in the clinical development of this agent to relate preclinical studies to patient treatment. In the studies presented here, the pharmacokinetics of ZD6474 was determined in plasma and tissues of MCF-7 tumor-bearing nude mice following single p.o. doses at 10, 25, and 50 mg/kg. Plasma area under the curve and Cmax were linear, increasing proportionally with dose. Tissue analysis showed that ZD6474 is extensively distributed to tissues, with liver and lung accumulating concentrations of 212 microg/g (approximately 450 microM) and 161 microg/g (approximately 340 microM), respectively. Tumor levels ranged from 27 to 71 microg/g at Cmax levels across the three dose ranges, and ZD6474 was distributed to all of the tissues in a dose-dependent manner. Analysis of putative ZD6474 metabolites in feces found four, with the N-demethyl-piperidinyl-ZD6474 metabolite being the most prominent but still accounting for less than 2% of the total amount of ZD6474 present. The lack of significant metabolism of ZD6474 is consistent with the relatively long half-life in mice (approximately 30 h), as well as that seen in humans (approximately 120 h), and the primary method of drug elimination appears to be unchanged in the feces (approximately 25%). The incorporation of an empirical approach to dosing in mouse models of cancer in preclinical studies may allow for better prediction of clinical efficacy for ZD6474 alone and in combination with other therapeutic modalities based on equivalent drug exposure.


Asunto(s)
Inhibidores de la Angiogénesis/farmacocinética , Antineoplásicos/farmacocinética , Piperidinas/farmacocinética , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Quinazolinas/farmacocinética , Animales , Área Bajo la Curva , Cromatografía Liquida , Relación Dosis-Respuesta a Droga , Sistemas de Liberación de Medicamentos , Estradiol/administración & dosificación , Estradiol/farmacología , Heces/química , Femenino , Hígado/efectos de los fármacos , Hígado/metabolismo , Espectrometría de Masas , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Reproducibilidad de los Resultados , Distribución Tisular , Trasplante Heterólogo
9.
Cancer Chemother Pharmacol ; 56(3): 248-54, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15856231

RESUMEN

PURPOSE: Paclitaxel (Taxol) is an effective agent against a broad range of human cancers. Studies on the metabolism and disposition of paclitaxel have shown that it is primarily eliminated via hepatic metabolism by P450 enzymes (2C8 and 3A4) to essentially inactive metabolites, and that biliary and gut transport by P-glycoprotein (PGP) as well as urinary elimination of the parent compound play relatively minor roles. Recent studies in vitro have shown that paclitaxel treatment increases the level of CYP2C8 and CYP3A4 in human hepatocytes as well as PGP in colon tumor cells. The data suggest that previous paclitaxel exposure may influence metabolism and elimination of subsequent doses. Further, since weekly paclitaxel dose schedules are becoming more common as opposed to the original every 21-day dosing, the likelihood of enzyme induction from previous doses impacting that from subsequent doses is increased. METHODS: To study the potential for such sequence-dependent alterations in paclitaxel pharmacokinetics, we carried out pharmacokinetic studies in mouse plasma and tissues following day 1 and days 1 and 5 dosing at 20 mg/kg. Paclitaxel concentrations were determined by a sensitive LC/MS/MS assay out to 16 h post-dosing in plasma, liver, kidney, gut and heart. The effect of paclitaxel treatment on hepatic expression of PGP and P450 isoforms (CYP2C and CYP3A) was determined to elucidate the mechanism by which paclitaxel disposition is altered by previous drug exposure. RESULTS: Pharmacokinetic analysis of the data showed that plasma and tissue AUC values after treatment on day 5 following a dose on day 1 were between 50% and 74% of those determined following a single dose on day 1. The terminal elimination half-life was not different. Activity and protein levels for CYP2C in liver were elevated at 24 and 96 h after paclitaxel dosing. Cremophor EL, a carrier solvent for paclitaxel, also caused elevated CYP2C activity. Neither CYP3A nor PGP levels in liver were altered by paclitaxel or Cremophor EL treatment at the 24-h and 96-h time points. The levels of 6alpha-OH-paclitaxel in feces were increased on day 5 as opposed to day 1 while paclitaxel levels in feces were unchanged. CONCLUSIONS: The results of our studies showed that paclitaxel pharmacokinetics are altered by previous paclitaxel exposure up to 96 h earlier.


Asunto(s)
Antineoplásicos Fitogénicos/farmacocinética , Sistema Enzimático del Citocromo P-450/biosíntesis , Paclitaxel/farmacocinética , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Animales , Área Bajo la Curva , Citocromo P-450 CYP3A , Inducción Enzimática , Femenino , Semivida , Inyecciones Intravenosas , Hígado/efectos de los fármacos , Hígado/enzimología , Ratones , Ratones Endogámicos BALB C , Distribución Tisular
10.
Cancer Lett ; 220(2): 161-9, 2005 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-15766591

RESUMEN

Doxorubicin (DOX) has excellent antitumor activity when combined with paclitaxel (PTX) and this combination is used as first-line treatment for metastatic breast cancer. Results from clinical studies on pharmacokinetic interaction of these agents are not conclusive and pre-clinical studies are still needed. Pharmacokinetic studies were carried out in female Balb/c mice with combined DOX (6 mg/kg) and PTX (10 mg/kg) treatment. Combined treatment with PTX and DOX leads to alterations in the pharmacokinetics of both agents, with the predominant effect being elevated drug levels in liver and gut tissues. DOX levels in kidney and heart tissues were unaffected by concurrent PTX treatment. Further, plasma levels of DOX are not changed by concurrent PTX treatment suggesting that monitoring of plasma levels of DOX, when used in combination with another drug that is a P-glycoprotein (PGP) substrate, will not reflect actual pharmacokinetic changes occurring in other tissues.


Asunto(s)
Antibióticos Antineoplásicos/farmacocinética , Antineoplásicos Fitogénicos/farmacocinética , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Animales , Neoplasias de la Mama/tratamiento farmacológico , Interacciones Farmacológicas , Femenino , Hígado/química , Ratones , Ratones Endogámicos BALB C , Distribución Tisular
11.
J Pharmacol Exp Ther ; 305(3): 1079-86, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12649308

RESUMEN

The bioreductive activation of the antitumor quinone mitomycin C (MMC) by NAD(P)H: quinone oxidoreductase 1 (NQO1) is complicated by the ability of MMC to also act as a mechanism-based inhibitor of NQO1 in a pH dependent manner. Inhibition of NQO1 by MMC has been studied in purified enzyme preparations and in cultured cells but has not determined in vivo. In the studies presented here, NQO1 activity was measured in mouse tissues following treatment with MMC or the potent mechanism-based human NQO1 inhibitor 5-methoxy-1,2-dimethyl-[(4-nitrophenoxy)methyl]indole-4,7-dione (ES936). NQO1 activity was significantly decreased at 1, 2, and 4 h following MMC (10 or 20 mg/kg) treatment in kidney and lung but was unchanged in brain, heart, liver, and bladder. ES936 (1 mg/kg) treatment led to a significant and much more potent inhibition of NQO1 in all murine tissues analyzed except for bladder. To extrapolate these in vivo results from mice to humans, the species-specific kinetics of NQO1 inactivation by MMC was determined in vitro using mouse, rat, and human recombinant NQO1 proteins. Results showed the inactivation kinetics of mouse and human proteins by MMC were similar. Treatment of human and murine endothelial cells with MMC or ES936 showed similar inhibition of NQO1 activity. The aforementioned results clearly demonstrate that MMC can serve as a substrate for NQO1 in vivo; however, the metabolism resulting in enzyme inactivation is possibly tissue-specific. Furthermore, the kinetic similarities for inactivation between murine and human forms of NQO1 show these results are apropos to clinical use of MMC.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Indolquinonas , Mitomicina/farmacología , NAD(P)H Deshidrogenasa (Quinona)/antagonistas & inhibidores , Animales , Interacciones Farmacológicas , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Femenino , Humanos , Indoles/farmacología , Cinética , Ratones , Ratones Endogámicos BALB C , Ratas , Especificidad de la Especie , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA