Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Toxicol Lett ; 342: 50-57, 2021 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-33581289

RESUMEN

Carbonyl reduction biotransformation pathway of anthracyclines (doxorubicin, daunorubicin) is a significant process, associated with drug metabolism and elimination. However, it also plays a pivotal role in anthracyclines-induced cardiotoxicity and cancer resistance. Herein, carbonyl reduction of eight anthracyclines, at in vivo relevant concentrations (20 µM), was studied in human liver cytosol, to describe the relationship between their structure and metabolism. Significant differences of intrinsic clearance between anthracyclines, ranging from 0,62-74,9 µL/min/mg were found and associated with data from in silico analyses, considering their binding in active sites of the main anthracyclines-reducing enzymes: carbonyl reductase 1 (CBR1) and aldo-keto reductase 1C3 (AKR1C3). Partial atomic charges of carbonyl oxygen atom were also determined and considered as a factor associated with reaction rate. Structural features, including presence or absence of side-chain hydroxy group, a configuration of sugar chain hydroxy group, and tetracyclic rings substitution, affecting anthracyclines susceptibility for carbonyl reduction were identified.


Asunto(s)
Aclarubicina/metabolismo , Citosol/metabolismo , Doxorrubicina/análogos & derivados , Hepatocitos/metabolismo , Oxidorreductasas/metabolismo , Aclarubicina/química , Oxidorreductasas de Alcohol/genética , Oxidorreductasas de Alcohol/metabolismo , Miembro C3 de la Familia 1 de las Aldo-Ceto Reductasas/genética , Miembro C3 de la Familia 1 de las Aldo-Ceto Reductasas/metabolismo , Antineoplásicos/química , Antineoplásicos/metabolismo , Sitios de Unión , Biotransformación , Doxorrubicina/química , Doxorrubicina/metabolismo , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Humanos , Modelos Moleculares , Simulación del Acoplamiento Molecular , Estructura Molecular , Conformación Proteica
2.
Biochem Pharmacol ; 163: 391-403, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30862505

RESUMEN

E-cadherin transcriptional activator EP300 is down-regulated in metaplastic breast carcinoma, a rare form of triple negative and E-cadherin-negative aggressive breast cancer with a poor clinical outcome. In order to shed light on the regulation of E-cadherin by EP300 in breast cancer we analyzed by immunohistochemistry 41 cases of invasive breast cancer with both E-cadherinhigh and E-cadherinlow expression levels, together with 20 non-malignant breast tissues. EP300 and E-cadherin showed a positive correlation in both non-malignant and cancer cases and both markers together were better predictors of lymph node metastasis than E-cadherin alone. These data support a metastasis suppressor role for EP300 in breast cancer. However, some reports suggest an oncogenic role for EP300. We generated a breast cancer cell model to study E-cadherin-independent effects of EP300 by over-expression of EP300 in HS578T cells which have E-cadherin promoter hypermethylated. In this cell system, EP300 led to up-regulation of mesenchymal (vimentin, Snail, Slug, Zeb1) and stemness (ALDH+ and CD44high/CD24low) markers, increases in migration, invasion, anchorage-independent growth and drug resistance. Genome-wide expression profiling identified aldo-keto reductases AKR1C1-3 as effectors of stemness and drug resistance, since their pharmacological inhibition with flufenamic acid restored both doxorubicin and paclitaxel sensitivity and diminished mammosphere formation. Thus, in cells with a permissive E-cadherin promoter, EP300 acts as a tumour/metastasis supressor by up-regulating E-cadherin expression, maintenance of the epithelial phenotype and avoidance of an epithelial-to-mesenchymal transition. In cells in which the E-cadherin promoter is hypermethylated, EP300 functions as an oncogene via up-regulation of aldo-keto reductases. This offers the rationale of using current aldo-keto reductase inhibitors in breast cancer treatment.


Asunto(s)
Aldo-Ceto Reductasas/antagonistas & inhibidores , Neoplasias de la Mama/enzimología , Proteína p300 Asociada a E1A/antagonistas & inhibidores , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Antineoplásicos/farmacología , Biomarcadores de Tumor , Cadherinas , Línea Celular Tumoral , Movimiento Celular , Doxorrubicina/farmacología , Resistencia a Antineoplásicos , Proteína p300 Asociada a E1A/genética , Proteína p300 Asociada a E1A/metabolismo , Transición Epitelial-Mesenquimal/efectos de los fármacos , Femenino , Humanos , Inmunohistoquímica , Invasividad Neoplásica , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Paclitaxel/farmacología
3.
Oncol Rep ; 37(4): 2025-2032, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28259989

RESUMEN

Resistance to anticancer medications often leads to poor outcomes. The present study explored an effective approach for enhancing chemotherapy targeted against human cancer cells. Real-time quantitative real-time polymerase chain reaction (qRT-PCR) analysis revealed overexpression of members of aldo-keto reductase (AKR) 1C family, AKR1C1, AKR1C2, AKR1C3, and AKR1C4, in cisplatin, cis-diamminedichloroplatinum (II) (CDDP)-resistant human cancer cell lines, HeLa (cervical cancer cells) and Sa3 (oral squamous cell carcinoma cells). The genes were downregulated using small-interfering RNA (siRNA) transfection, and the sensitivity to CDDP or 5-fluorouracil (5-FU) was investigated. When the genes were knocked down, sensitivity to CDDP and 5-FU was restored. Furthermore, we found that administration of mefenamic acid, a widely used non-steroidal anti-inflammatory drug (NSAID) and a known inhibitor of AKR1Cs, enhanced sensitivity to CDDP and 5-FU. The present study suggests that AKR1C family is closely associated with drug resistance to CDDP and 5-FU, and mefenamic acid enhances their sensitivity through its inhibitory activity in drug-resistant human cancer cells. Thus, the use of mefenamic acid to control biological function of AKR1C may lead to effective clinical outcomes by overcoming anticancer drug resistance.


Asunto(s)
20-Hidroxiesteroide Deshidrogenasas/biosíntesis , 3-Hidroxiesteroide Deshidrogenasas/biosíntesis , Hidroxiprostaglandina Deshidrogenasas/biosíntesis , Hidroxiesteroide Deshidrogenasas/biosíntesis , Ácido Mefenámico/administración & dosificación , Neoplasias/tratamiento farmacológico , 20-Hidroxiesteroide Deshidrogenasas/antagonistas & inhibidores , 20-Hidroxiesteroide Deshidrogenasas/genética , 3-Hidroxiesteroide Deshidrogenasas/antagonistas & inhibidores , 3-Hidroxiesteroide Deshidrogenasas/genética , Miembro C3 de la Familia 1 de las Aldo-Ceto Reductasas , Cisplatino/administración & dosificación , Resistencia a Antineoplásicos/efectos de los fármacos , Fluorouracilo/administración & dosificación , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HeLa , Humanos , Hidroxiprostaglandina Deshidrogenasas/antagonistas & inhibidores , Hidroxiprostaglandina Deshidrogenasas/genética , Hidroxiesteroide Deshidrogenasas/antagonistas & inhibidores , Hidroxiesteroide Deshidrogenasas/genética , Neoplasias/genética , Neoplasias/patología , Oxidorreductasas
4.
PLoS One ; 12(2): e0172244, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28196134

RESUMEN

Intrinsic or acquired drug resistance is a major impediment to the successful treatment of women with breast cancer using chemotherapy. We have observed that MCF-7 breast tumor cells selected for resistance to doxorubicin or epirubicin (MCF-7DOX2 and MCF-7EPI cells, respectively) exhibited increased expression of several members of the aldo-keto reductase (AKR) gene family (in particular AKR1C3 and AKR1B10) relative to control MCF-7CC cells selected by propagation in the absence of drug. Normal cellular roles for the AKRs include the promotion of estrogen (E2) synthesis from estrone (E1) and the hydroxylation and detoxification of exogenous xenobiotics such as anthracycline chemotherapy drugs. While hydroxylation of anthracyclines strongly attenuates their cytotoxicity, it is unclear whether the enhanced AKR expression in the above anthracycline-resistant cells promotes E2 synthesis and/or alterations in E2 signalling pathways and whether such changes contribute to enhanced survival and anthracycline resistance. To determine the role of AKRs and E2 pathways in doxorubicin resistance, we examined changes in the expression of E2-related genes and proteins upon acquisition of doxorubicin resistance. We also assessed the effects of AKR overexpression or downregulation or the effects of activators or inhibitors of E2-dependent pathways on previously acquired resistance to doxorubicin. In this study we observed that the enhanced AKR expression upon acquisition of anthracycline resistance was, in fact, associated with enhanced E2 production. However, the expression of estrogen receptor α (ERα) was reduced by 2- to 5-fold at the gene transcript level and 2- to 20-fold at the protein level upon acquisition of anthracycline resistance. This was accompanied by an even stronger reduction in ERα phosphorylation and activity, including highly suppressed expression of two proteins under E2-dependent control (Bcl-2 and cyclin D1). The diminished Bcl-2 and cyclin D1 expression would be expected to reduce the growth rate of the cells, a hypothesis which was confirmed in subsequent cell proliferation experiments. AKR1C3 or AKR1B10 overexpression alone had no effect on doxorubicin sensitivity in MCF-7CC cells, while siRNA-mediated knockdown of AKR1C3 and/or AKR1B10 expression had no significant effect on sensitivity to doxorubicin in MCF-7DOX2 or MCF-7EPI cells. This suggested that enhanced or reduced AKR expression/activity is insufficient to confer anthracycline resistance or sensitivity to breast tumor cells, respectively. Rather, it would appear that AKR overexpression acts in concert with other proteins to confer anthracycline resistance, including reduced E2-dependent expression of both an important apoptosis inhibitor (Bcl-2) and a key protein associated with activation of cell cycle-dependent kinases (cyclin D1).


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Doxorrubicina/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Estrógenos/metabolismo , Transducción de Señal/efectos de los fármacos , 3-Hidroxiesteroide Deshidrogenasas/biosíntesis , 3-Hidroxiesteroide Deshidrogenasas/genética , Aldehído Reductasa/biosíntesis , Aldehído Reductasa/genética , Miembro C3 de la Familia 1 de las Aldo-Ceto Reductasas , Aldo-Ceto Reductasas , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Ciclina D1/genética , Ciclina D1/metabolismo , Resistencia a Antineoplásicos/genética , Estrógenos/genética , Femenino , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Hidroxiprostaglandina Deshidrogenasas/biosíntesis , Hidroxiprostaglandina Deshidrogenasas/genética , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Transducción de Señal/genética
5.
BMC Cancer ; 12: 381, 2012 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-22938713

RESUMEN

BACKGROUND: Since proteins involved in chemotherapy drug pharmacokinetics and pharmacodynamics have a strong impact on the uptake, metabolism, and efflux of such drugs, they likely play critical roles in resistance to chemotherapy drugs in cancer patients. METHODS: To investigate this hypothesis, we conducted a whole genome microarray study to identify difference in the expression of genes between isogenic doxorubicin-sensitive and doxorubicin-resistant MCF-7 breast tumour cells. We then assessed the degree of over-representation of doxorubicin pharmacokinetic and pharmacodynamic genes in the dataset of doxorubicin resistance genes. RESULTS: Of 27,958 Entrez genes on the array, 7.4 per cent or 2,063 genes were differentially expressed by ≥ 2-fold between wildtype and doxorubicin-resistant cells. The false discovery rate was set at 0.01 and the minimum p value for significance for any gene within the "hit list" was 0.01. Seventeen and 43 per cent of doxorubicin pharmacokinetic genes were over-represented in the hit list, depending upon whether the gene name was identical or within the same gene family, respectively. The most over-represented genes were within the 1C and 1B families of aldo-keto reductases (AKRs), which convert doxorubicin to doxorubicinol. Other genes convert doxorubicin to other metabolites or affect the influx, efflux, or cytotoxicity of the drug. In further support of the role of AKRs in doxorubicin resistance, we observed that, in comparison to doxorubicin, doxorubincol exhibited dramatically reduced cytotoxicity, reduced DNA-binding activity, and strong localization to extra nuclear lysosomes. Pharmacologic inhibition of the above AKRs in doxorubicin-resistant cells increased cellular doxorubicin levels, restored doxorubicin cytotoxicity and re-established doxorubicin localization to the nucleus. The properties of doxorubicinol were unaffected. CONCLUSIONS: These findings demonstrate the utility of using curated pharmacokinetic and pharmacodynamic knowledge bases to identify highly relevant genes associated with doxorubicin resistance. The induction of one or more of these genes was found to be correlated with changes in the drug's properties, while inhibiting one specific class of these genes (the AKRs) increased cellular doxorubicin content and restored drug DNA binding, cytotoxicity, and subcellular localization.


Asunto(s)
Aldehído Reductasa/genética , Aldehído Reductasa/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Doxorrubicina/análogos & derivados , Doxorrubicina/farmacocinética , 20-Hidroxiesteroide Deshidrogenasas/biosíntesis , 20-Hidroxiesteroide Deshidrogenasas/genética , 20-Hidroxiesteroide Deshidrogenasas/metabolismo , 3-Hidroxiesteroide Deshidrogenasas/biosíntesis , 3-Hidroxiesteroide Deshidrogenasas/genética , 3-Hidroxiesteroide Deshidrogenasas/metabolismo , Aldehído Reductasa/biosíntesis , Miembro C3 de la Familia 1 de las Aldo-Ceto Reductasas , Aldo-Ceto Reductasas , Neoplasias de la Mama/metabolismo , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Ácidos Cólicos/farmacología , Ciclosporina/farmacología , ADN de Neoplasias/metabolismo , Doxorrubicina/farmacología , Resistencia a Antineoplásicos , Perfilación de la Expresión Génica , Humanos , Hidroxiprostaglandina Deshidrogenasas/biosíntesis , Hidroxiprostaglandina Deshidrogenasas/genética , Hidroxiprostaglandina Deshidrogenasas/metabolismo , Espacio Intracelular/efectos de los fármacos , Espacio Intracelular/metabolismo , Células MCF-7 , Análisis de Secuencia por Matrices de Oligonucleótidos
6.
J Pharmacol Exp Ther ; 335(3): 533-45, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20837989

RESUMEN

Doxorubicin (DOX) and daunorubicin (DAUN) are effective anticancer drugs; however, considerable interpatient variability exists in their pharmacokinetics. This may be caused by altered metabolism by nonsynonymous single-nucleotide polymorphisms (ns-SNPs) in genes encoding aldo-keto reductases (AKRs) and carbonyl reductases. This study examined the effect of 27 ns-SNPs, in eight human genes, on the in vitro metabolism of both drugs to their major metabolites, doxorubicinol and daunorubicinol. Kinetic assays measured metabolite levels by high-performance liquid chromatography separation with fluorescence detection using purified, histidine-tagged, human wild-type, and variant enzymes. Maximal rate of activity (V(max)), substrate affinity (K(m)), turnover rate (k(cat)), and catalytic efficiency (k(cat)/K(m)) were determined. With DAUN as substrate, variants for three genes exhibited significant differences in these parameters compared with their wild-type counterparts: the A106T, R170C, and P180S variants significantly reduced metabolism compared with the AKR1C3 wild-type (V(max), 23-47% decrease; k(cat), 22-47%; k(cat)/K(m), 38-44%); the L311V variant of AKR1C4 significantly decreased V(max) (47% lower) and k(cat) and k(cat)/K(m) (both 43% lower); and the A142T variant of AKR7A2 significantly affected all kinetic parameters (V(max) and k(cat), 61% decrease; K(m), 156% increase; k(cat)/K(m), 85% decrease). With DOX, the R170C and P180S variants of AKR1C3 showed significantly reduced V(max) (41-44% decrease), k(cat) (39-45%), and k(cat)/K(m) (52-69%), whereas the A142T variant significantly altered all kinetic parameters for AKR7A2 (V(max), 41% decrease; k(cat), 44% decrease; K(m), 47% increase; k(cat)/K(m), 60% decrease). These findings suggest that ns-SNPs in human AKR1C3, AKR1C4, and AKR7A2 significantly decrease the in vitro metabolism of DOX and DAUN.


Asunto(s)
Oxidorreductasas de Alcohol/genética , Oxidorreductasas de Alcohol/metabolismo , Daunorrubicina/metabolismo , Doxorrubicina/metabolismo , Polimorfismo de Nucleótido Simple/fisiología , 20-Hidroxiesteroide Deshidrogenasas/genética , 20-Hidroxiesteroide Deshidrogenasas/metabolismo , 3-Hidroxiesteroide Deshidrogenasas/genética , 3-Hidroxiesteroide Deshidrogenasas/metabolismo , Aldehído Reductasa/genética , Aldehído Reductasa/metabolismo , Miembro C3 de la Familia 1 de las Aldo-Ceto Reductasas , Aldo-Ceto Reductasas , Biocatálisis , Frecuencia de los Genes , Gliceraldehído/metabolismo , Humanos , Hidroxiprostaglandina Deshidrogenasas/genética , Hidroxiprostaglandina Deshidrogenasas/metabolismo , Hidroxiesteroide Deshidrogenasas/genética , Hidroxiesteroide Deshidrogenasas/metabolismo , Indanos/metabolismo , Cinética , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Modelos Moleculares , NAD(P)H Deshidrogenasa (Quinona)/genética , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Oxidorreductasas/genética , Oxidorreductasas/metabolismo , Fenantrenos/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Vitamina K 3/metabolismo
7.
Int J Cancer ; 120(9): 2019-27, 2007 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-17266043

RESUMEN

Dihydrodiol dehydrogenase (DDH) is a member of the aldo-keto reductases superfamily (AKR1C1-AKR1C4), which plays central roles in the metabolism of steroid hormone, prostaglandin and xenobiotics. We have previously detected overexpression of DDH as an indicator of poor prognosis and chemoresistance in human non-small lung cancer (NSCLC). We also found DDH expression to be closely related to chronic inflammatory conditions. The aim of this study was to investigate the links between inflammation, DDH expression and drug resistance in NSCLC cells. We showed that pro-inflammatory mediators including interleukin-6 (IL-6) could induce AKR1C1/1C2 expression in NSCLC cells and increase cellular resistance to cisplatin and adriamycin. This effect was nullified by Safingol, a protein kinase C inhibitor. Moreover, the expression of AKR1C1/1C2 was inversely correlated to NBS1 and apoptosis-inducing factor (AIF). We also showed that IL-6-induced AKR1C1/1C2 expression and drug resistance were inhibited by wogonin and chrysin, which are major flavonoids in Scutellaria baicalensis, a widely used traditional Chinese and Japanese medicine. In conclusion, this study demonstrated novel links of pro-inflammatory signals, AKR1C1/1C2 expression and drug resistance in NSCLC. The protein kinase C pathway may play an important role in this process. Overexpression of AKR1C1/1C2 may serve as a marker of chemoresistance. Further studies are warranted to evaluate wogonin and chrysin as a potential adjuvant therapy for drug-resistant NSCLC, especially for those with AKR1C1/1C2 overexpression.


Asunto(s)
20-Hidroxiesteroide Deshidrogenasas/antagonistas & inhibidores , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Flavanonas/farmacología , Flavonoides/farmacología , Hidroxiesteroide Deshidrogenasas/antagonistas & inhibidores , Neoplasias Pulmonares/tratamiento farmacológico , 20-Hidroxiesteroide Deshidrogenasas/fisiología , Apoptosis , Ciclo Celular , Línea Celular Tumoral , Reparación del ADN , Relación Dosis-Respuesta a Droga , Doxorrubicina/farmacología , Resistencia a Antineoplásicos , Humanos , Hidroxiesteroide Deshidrogenasas/fisiología , Interleucina-6/farmacología
8.
Curr Med Res Opin ; 21(3): 363-8, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15811204

RESUMEN

OBJECTIVE: Doxorubicin, irinotecan and etoposide are all associated with the debilitating side-effects of nausea and vomiting, thereby necessitating concomitant antiemetic therapy. However, this may increase the potential for drug-drug interactions by inhibition or induction of the cytochrome P450 enzymes. A study was undertaken to investigate the effects of the 5-HT(3) -receptor antagonist granisetron on the metabolism of doxorubicin, irinotecan and etoposide in vitro in human liver microsomal preparations. RESEARCH DESIGN AND METHODS: Doxorubicin, 20 microM, irinotecan, 10 microM, and etoposide, 50 microM, were incubated in the presence of granisetron, 0 nM, 20 nM, 200 nM and 2000 nM, in human liver microsomal preparations (250 microg). The levels of unchanged doxorubicin, irinotecan and etoposide in the incubation mixture were determined by high-performance liquid chromatography. Positive controls were ketoconazole, 20 microM, a potent inhibitor of CYP3A metabolism, for irinotecan and etoposide and quercitrin, 2 mM, a potent inhibitor of aldo-keto reductase, for doxorubicin. RESULTS: In the absence of granisetron, unchanged doxorubicin, irinotecan and etoposide levels decreased by 34.2 +/- 5.5%, 21.3 +/- 2.9% and 13.4 +/- 1.6% of control, respectively. Ketoconazole prevented the breakdown of both irinotecan and etoposide, while quercitrin inhibited the breakdown of doxorubicin. Granisetron had no effect on the rate of reduction of doxorubicin, irinotecan or etoposide. CONCLUSIONS: The results from this study suggest that granisetron neither inhibits nor induces the enzymes involved in the metabolism of doxorubicin, irinotecan or etoposide. Thus, granisetron can be used safely to treat nausea and vomiting induced by these agents with minimal risk of drug-drug interactions.


Asunto(s)
Antibióticos Antineoplásicos/metabolismo , Antieméticos/farmacología , Antineoplásicos Fitogénicos/metabolismo , Camptotecina/análogos & derivados , Camptotecina/metabolismo , Doxorrubicina/metabolismo , Etopósido/metabolismo , Granisetrón/farmacología , Técnicas de Cultivo de Célula , Sistema Enzimático del Citocromo P-450/farmacología , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Humanos , Irinotecán , Microsomas Hepáticos
9.
Drug Metab Dispos ; 25(1): 66-74, 1997 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-9010632

RESUMEN

Pharmacokinetic parameters-including tissue distribution, biliary excretion, and urinary excretion of M1-M4-were compared after an intravenous administration of DA-125 (a new anthracycline derivative; 20 mg/kg body weight) to male spontaneously hypersensitive rats (SHRs) at 16 weeks (an animal model for human primary hypertension) and at 6 weeks (corresponding to the early phase of the development of hypertension, at which time blood pressure remains within the normotensive range) of age and their age-matched control Kyoto-Wistar rats, and male deoxycorticosterone acetate-salt-induced Sprague-Dawley rats (DOCA-salt rats, an animal model for human secondary hypertension) at 16 weeks of age and their age-matched control Sprague-Dawley rats. Mean plasma concentrations of both M2 and M4, and the resultant area under the plasma concentration-time curve from time 0 to last measured time [AUCT; M2 (68.9 vs. 29.3 micrograms-min/ml) and M4 (53.4 vs. 33.4 micrograms-min/ml)], increased significantly in SHRs at 16 weeks of age, compared with their control rats. Similar results were also obtained from DOCA-salt rats at 16 weeks of age, compared with their control rats. However, values were not significantly different between SHRs at 6 weeks of age and their control rats. Previous data indicated that the significant increase in plasma concentrations and the resultant AUCT values of both M2 and M4 in SHRs at 16 weeks of age were due to the hypertension state itself, and not to any hereditary characteristics of the SHRs. The significantly increased plasma concentrations and the resultant AUCT values of M2 in both SHRs and DOCA-salt rats at 16 weeks of age were due to the significantly decreased biliary excretion of M2 and possibly to the increased amount of aldo-keto reductase in the liver. However, the increase in the two aforementioned pharmacokinetic parameters in the case of M4 were possibly due solely to the increased amount of aldo-keto reductase in the liver.


Asunto(s)
Antibióticos Antineoplásicos/farmacocinética , Doxorrubicina/análogos & derivados , Hipertensión/metabolismo , Factores de Edad , Oxidorreductasas de Alcohol/metabolismo , Aldehído Reductasa , Aldo-Ceto Reductasas , Animales , Antibióticos Antineoplásicos/metabolismo , Bilis/química , Desoxicorticosterona , Doxorrubicina/metabolismo , Doxorrubicina/farmacocinética , Hipertensión/sangre , Hipertensión/inducido químicamente , Hipertensión/orina , Hígado/enzimología , Masculino , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Ratas Sprague-Dawley , Cloruro de Sodio
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...