Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Pediatr Res ; 83(2): 527-535, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28922349

RESUMEN

BackgroundAge-dependent differences in pharmacokinetics exist for metabolically cleared medications. Differential contributions in the cytochrome P450 3A (CYP3A), CYP2C, and flavin-containing monooxygenases (FMOs) families have an important role in the metabolic clearance of a large number of drugs administered to children.MethodsUnlike previous semiquantitative characterization of age-dependent changes in the expression of genes and proteins (western blot analysis), this study quantifies both gene and absolute protein expression in the same fetal, pediatric, and adult hepatic tissue. Expression was then correlated with the corresponding functional activities in the same samples.ResultsCYP3A and FMO families showed a distinct switch from fetal (CYP3A7 and FMO1) to adult isoforms (CYP3A4 and FMO3) at birth, whereas CYP2C9 showed a linear maturation from birth into adulthood. In contrast, analysis of CYP2C19 revealed higher expression and catalytic efficiency in pediatric samples compared with that in fetal and adult samples. Further, CYP3A and FMO enzymes exhibited an unexpectedly higher functional activity in fetal samples not entirely explained by protein expression.ConclusionThese surprising findings suggest that CYP and FMO enzymes may encounter development-related differences in their microenvironments that can influence the enzyme activity in addition to protein expression levels.


Asunto(s)
Factores de Edad , Sistema Enzimático del Citocromo P-450/metabolismo , Dinitrocresoles/metabolismo , Adulto , Niño , Preescolar , Citocromo P-450 CYP2C9/metabolismo , Citocromo P-450 CYP3A/metabolismo , Perfilación de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Humanos , Lactante , Recién Nacido , Hígado/embriología , Hígado/metabolismo , Microsomas Hepáticos/metabolismo , Persona de Mediana Edad , Oxidación-Reducción , Oxigenasas/metabolismo , Proteínas Recombinantes/metabolismo
2.
J Extra Corpor Technol ; 49(3): 150-159, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28979038

RESUMEN

Invasive candidiasis is common and often fatal in patients supported with extracorporeal membrane oxygenation (ECMO), and treatment relies on optimal antifungal dosing. The ECMO circuit can extract drug and decrease drug exposure, placing the patient at risk of therapeutic failure. This ex vivo study determined the extraction of antifungal drugs by the ECMO circuit. Fluconazole and micafungin were studied separately in three closed-loop circuit configurations to isolate the impact of the oxygenator, hemofilter, and tubing on circuit extraction. Each circuit was primed with human blood, and flow was set to 1 L/min. Drug was dosed to achieve therapeutic concentrations. Each antifungal was added to a separate tube of blood to serve as a control. Serial blood samples were collected over 24 hours and concentrations were quantified with a validated assay. Drug recovery was calculated at each time point: (C t /C i )*100, with C t and C i the concentrations at time = t and 1 minute, respectively. After 24 hours of recirculation, mean recovery of fluconazole in the ECMO circuit (95-98%) and controls (101%) was high. In contrast, mean recovery of micafungin was dependent on the time and circuit configuration. Recovery at 4 hours was only 46% when a hemofilter was in-line but was much higher when the hemofilter was removed (91%). By 24 hours, however, micafungin recovery was low in all circuit configurations (26-43%), regardless of the presence of a hemofilter, as well as in the controls (57%). In conclusion, these results suggest that micafungin is extracted by the ECMO circuit, which may result in decreased drug exposure in vivo.


Asunto(s)
Candidiasis/tratamiento farmacológico , Equinocandinas/administración & dosificación , Oxigenación por Membrana Extracorpórea/métodos , Fluconazol/administración & dosificación , Lipopéptidos/administración & dosificación , Tiempo de Circulación Sanguínea , Candidiasis/sangre , Relación Dosis-Respuesta a Droga , Equinocandinas/farmacocinética , Oxigenación por Membrana Extracorpórea/instrumentación , Fluconazol/farmacocinética , Hemofiltración/instrumentación , Hemofiltración/métodos , Humanos , Lipopéptidos/farmacocinética , Micafungina , Unión Proteica , Albúmina Sérica/metabolismo
3.
Int J Cancer ; 138(9): 2281-92, 2016 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-26669511

RESUMEN

The antidiabetic drug metformin exerts antineoplastic effects against breast cancer and other cancers. One mechanism by which metformin is believed to exert its anticancer effect involves activation of its intracellular target, adenosine monophosphate-activated protein kinase (AMPK), which is also implicated in the antidiabetic effect of metformin. It is proposed that in cancer cells, AMPK activation leads to inhibition of the mammalian target of rapamycin (mTOR) and the downstream pS6K that regulates cell proliferation. Due to its hydrophilic and cationic nature, metformin requires cation-selective transporters to enter cells and activate AMPK. This study demonstrates that expression levels of cation-selective transporters correlate with the antiproliferative and antitumor efficacy of metformin in breast cancer. Metformin uptake and antiproliferative activity were compared between a cation-selective transporter-deficient human breast cancer cell line, BT-20, and a BT-20 cell line that was engineered to overexpress organic cation transporter 3 (OCT3), a representative of cation-selective transporters and a predominant transporter in human breast tumors. Metformin uptake was minimal in BT-20 cells, but increased by >13-fold in OCT3-BT20 cells, and its antiproliferative potency was >4-fold in OCT3-BT20 versus BT-20 cells. This increase in antiproliferative activity was associated with greater AMPK phosphorylation and decreased pS6K phosphorylation in OCT3-BT20 cells. In vitro data were corroborated by in vivo observations of significantly greater antitumor efficacy of metformin in xenograft mice bearing OCT3-overexpressing tumors versus low transporter-expressing wildtype tumors. Collectively, these findings establish a clear relationship between cation-selective transporter expression, the AMPK-mTOR-pS6K signaling cascade, and the antiproliferative activity of metformin in breast cancer.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Antineoplásicos/farmacología , Neoplasias de la Mama/patología , Metformina/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Femenino , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Desnudos , Proteínas de Transporte de Catión Orgánico , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Drug Metab Dispos ; 44(7): 975-83, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26839369

RESUMEN

Flavin-containing monooxygenases (FMOs) have a significant role in the metabolism of small molecule pharmaceuticals. Among the five human FMOs, FMO1, FMO3, and FMO5 are the most relevant to hepatic drug metabolism. Although age-dependent hepatic protein expression, based on immunoquantification, has been reported previously for FMO1 and FMO3, there is very little information on hepatic FMO5 protein expression. To overcome the limitations of immunoquantification, an ultra-performance liquid chromatography (UPLC)-multiple reaction monitoring (MRM)-based targeted quantitative proteomic method was developed and optimized for the quantification of FMO1, FMO3, and FMO5 in human liver microsomes (HLM). A post-in silico product ion screening process was incorporated to verify LC-MRM detection of potential signature peptides before their synthesis. The developed method was validated by correlating marker substrate activity and protein expression in a panel of adult individual donor HLM (age 39-67 years). The mean (range) protein expression of FMO3 and FMO5 was 46 (26-65) pmol/mg HLM protein and 27 (11.5-49) pmol/mg HLM protein, respectively. To demonstrate quantification of FMO1, a panel of fetal individual donor HLM (gestational age 14-20 weeks) was analyzed. The mean (range) FMO1 protein expression was 7.0 (4.9-9.7) pmol/mg HLM protein. Furthermore, the ontogenetic protein expression of FMO5 was evaluated in fetal, pediatric, and adult HLM. The quantification of FMO proteins also was compared using two different calibration standards, recombinant proteins versus synthetic signature peptides, to assess the ratio between holoprotein versus total protein. In conclusion, a UPLC-MRM-based targeted quantitative proteomic method has been developed for the quantification of FMO enzymes in HLM.


Asunto(s)
Cromatografía Liquida , Hígado/enzimología , Microsomas Hepáticos/enzimología , Oxigenasas/metabolismo , Proteómica/métodos , Adulto , Factores de Edad , Anciano , Calibración , Niño , Cromatografía Liquida/normas , Edad Gestacional , Humanos , Hígado/embriología , Persona de Mediana Edad , Proteómica/normas , Estándares de Referencia , Reproducibilidad de los Resultados , Espectrometría de Masas en Tándem
5.
J Pharmacol Exp Ther ; 352(3): 519-28, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25563903

RESUMEN

Metformin is the frontline therapy for type II diabetes mellitus. The oral bioavailability of metformin is unexpectedly high, between 40 and 60%, given its hydrophilicity and positive charge at all physiologic pH values. Previous studies in Caco-2 cell monolayers, a cellular model of the human intestinal epithelium, showed that during absorptive transport metformin is taken up into the cells via transporters in the apical (AP) membrane; however, predominant transport to the basolateral (BL) side occurs via the paracellular route because intracellular metformin cannot egress across the BL membrane. Furthermore, these studies have suggested that the AP transporters can contribute to intestinal accumulation and absorption of metformin. Transporter-specific inhibitors as well as a novel approach involving a cocktail of transporter inhibitors with overlapping selectivity were used to identify the AP transporters that mediate metformin uptake in Caco-2 cell monolayers; furthermore, the relative contributions of these transporters in metformin AP uptake were also determined. The organic cation transporter 1, plasma membrane monoamine transporter (PMAT), serotonin reuptake transporter, and choline high-affinity transporter contributed to approximately 25%, 20%, 20%, and 15%, respectively, of the AP uptake of metformin. PMAT-knockdown Caco-2 cells were constructed to confirm the contribution of PMAT in metformin AP uptake because a PMAT-selective inhibitor is not available. The identification of four intestinal transporters that contribute to AP uptake and potentially intestinal absorption of metformin is a significant novel finding that can influence our understanding of metformin pharmacology and intestinal drug-drug interactions involving this highly prescribed drug.


Asunto(s)
Sistemas de Transporte de Aminoácidos Básicos/metabolismo , Hipoglucemiantes/metabolismo , Metformina/metabolismo , Transporte Biológico/fisiología , Células CACO-2 , Proteínas de Transporte de Nucleósido Equilibrativas/metabolismo , Células HEK293 , Humanos , Proteínas de Transporte de Membrana/metabolismo , Transportador 1 de Catión Orgánico/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo
6.
J Am Pharm Assoc (2003) ; 54(5): 552-5, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25216886

RESUMEN

OBJECTIVE: The objective of this commentary is to explore the potential role of the pharmacist in the field of global health by understanding the definition of global health and how it can be applied to the profession of pharmacy. SUMMARY: While the role of the pharmacist in public health has been defined, the literature is limited with respect to the role of the pharmacist in global health. It is suggested that the "global" in global health should refer to the scope of a problem and not necessarily the geographic location. Through this lens, pharmacists have the opportunity to play an important role in both public health and global health. In particular, pharmacists can look at the varied global health careers established in medicine and use this as a framework to understand the potential role of the pharmacist within global health practice and program delivery, research, and policy. CONCLUSIONS: To further define the role of the pharmacist in global health, pharmacists may have to change their perception of what services they can provide and where these services can be applied.


Asunto(s)
Salud Global , Servicios Farmacéuticos/organización & administración , Farmacéuticos/organización & administración , Actitud del Personal de Salud , Atención a la Salud/organización & administración , Política de Salud , Humanos , Rol Profesional , Salud Pública
7.
Mol Pharmacol ; 84(2): 182-9, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23680637

RESUMEN

Organic cation transporters (OCTs) are members of the solute carrier 22 family of transporter proteins that are involved in absorption, distribution, and excretion of organic cations. OCT3 is localized in the apical (AP) membrane of enterocytes, but the literature is ambiguous about OCT1 (mOct1) localization, with some evidence suggesting a basolateral (BL) localization in human and mouse enterocytes. This is contrary to our preliminary findings showing AP localization of OCT1 in Caco-2 cell monolayers, an established model of human intestinal epithelium. Therefore, this study aims at determining the localization of OCT1 (mOct1) in Caco-2 cells, and human and mouse enterocytes. Functional studies using OCT1-specific substrate pentamidine showed transporter-mediated AP but not BL uptake in Caco-2 cells and human and mouse intestinal tissues. OCT1 inhibition decreased AP uptake of pentamidine by ∼50% in all three systems with no effect on BL uptake. A short hairpin RNA-mediated OCT1 knockdown in Caco-2 cells decreased AP uptake of pentamidine by ∼50% but did not alter BL uptake. Immunostaining and confocal microscopy in all three systems confirmed AP localization of OCT1 (mOct1). Our studies unequivocally show AP membrane localization of OCT1 (mOct1) in Caco-2 cells and human and mouse intestine. These results are highly significant as they will require reinterpretation of previous drug disposition and drug-drug interaction studies where conclusions were drawn assuming BL localization of OCT1 in enterocytes. Most importantly, these results will require revision of the regulatory guidance for industry in the United States and elsewhere because it has stated that OCT1 is basolaterally localized in enterocytes.


Asunto(s)
Enterocitos/metabolismo , Mucosa Intestinal/metabolismo , Transportador 1 de Catión Orgánico/metabolismo , Animales , Células CACO-2 , Línea Celular Tumoral , Membrana Celular/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Transporte de Proteínas
8.
Drug Metab Dispos ; 41(9): 1695-702, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23821186

RESUMEN

Recent studies showed that P-glycoprotein (P-gp) increases the portal bioavailability (FG) of loperamide by sparing its intestinal first-pass metabolism. Loperamide is a drug whose oral absorption is strongly attenuated by intestinal P-gp-mediated efflux and first-pass metabolism by cytochrome P450 3A (CYP3A). Here the effect of the interplay of P-gp and Cyp3a in modulating intestinal first-pass metabolism and absorption was investigated for another Cyp3a/P-gp dual substrate amprenavir, which is less efficiently effluxed by P-gp than loperamide. After oral administration of amprenavir, the portal concentrations and FG of amprenavir were approximately equal in P-gp competent and P-gp deficient mice. Mechanistic studies on the effect of P-gp on Cyp3a-mediated metabolism of amprenavir using intestinal tissue from P-gp competent and P-gp deficient mice (Ussing-type diffusion chamber) revealed that P-gp-mediated efflux caused only a slight reduction of oxidative metabolism of amprenavir. Studies in which portal concentrations and FG were measured in P-gp competent and P-gp deficient mice whose cytochrome P450 (P450) enzymes were either intact or inactivated showed that intestinal first-pass metabolism attenuates the oral absorption of amprenavir by approximately 10-fold, whereas P-gp efflux has a relatively small effect (approximately 2-fold) in attenuating the intestinal absorption. Cumulatively, these studies demonstrate that P-gp has little influence on the intestinal first-pass metabolism and FG of amprenavir and that intestinal P450-mediated metabolism plays the dominant role in attenuating the oral absorption of this drug.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Carbamatos/metabolismo , Citocromo P-450 CYP3A/metabolismo , Absorción Intestinal/fisiología , Mucosa Intestinal/metabolismo , Intestinos/enzimología , Sulfonamidas/metabolismo , Animales , Disponibilidad Biológica , Transporte Biológico , Furanos , Masculino , Ratones , Permeabilidad
9.
Drug Metab Dispos ; 41(3): 642-50, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23288866

RESUMEN

P-glycoprotein (P-gp) and CYP3A (cytochrome P450 3A, generally; Cyp3a, rodent enzyme) in the intestine can attenuate absorption of orally administered drugs. While some suggest that P-gp enhances intestinal metabolism by CYP3A/Cyp3a during absorption of a dual substrate, others suggest that P-gp reduces the metabolism in the intestine when substrates are at subsaturating concentrations. Hence, to elucidate the cellular mechanisms that can address these divergent reports, we studied intestinal absorption of the dual substrate loperamide in portal vein-cannulated P-gp-competent and P-gp-deficient mice. These studies showed that at low doses of loperamide, which produced intestinal concentrations near the apparent K(m) for oxidative metabolism, the bioavailability across the intestine (F(G)) was 6-fold greater in the P-gp-competent mice than in P-gp-deficient mice. The higher F(G) of loperamide in the presence of P-gp was attributed to lower loperamide intestinal metabolism. However, at high doses of loperamide, the sparing of first-pass metabolism by P-gp was balanced against the attenuation of absorption by apical efflux, resulting in no net effect on F(G). In vitro studies with intestinal tissue from P-gp-competent and -deficient mice confirmed that P-gp reduced the metabolic rate of loperamide during absorptive flux at concentrations near K(m) but had little effect on metabolism at higher (saturating) concentrations. Further, studies in which Cyp3a was chemically inactivated by aminobenzotriazole in P-gp-competent and -deficient mice, showed that P-gp and Cyp3a individually attenuated F(G) by 8-fold and 70-fold, respectively. These results confirmed that P-gp effectively protects loperamide at low doses from intestinal first-pass metabolism during intestinal absorption.


Asunto(s)
Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Absorción Intestinal , Mucosa Intestinal/metabolismo , Loperamida/farmacocinética , Subfamilia B de Transportador de Casetes de Unión a ATP/deficiencia , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Administración Oral , Animales , Disponibilidad Biológica , Citocromo P-450 CYP3A/metabolismo , Inhibidores del Citocromo P-450 CYP3A , Inhibidores Enzimáticos/farmacología , Absorción Intestinal/efectos de los fármacos , Intestinos/efectos de los fármacos , Loperamida/administración & dosificación , Loperamida/sangre , Masculino , Ratones , Ratones Noqueados , Modelos Biológicos , Modelos Estadísticos , Permeabilidad , Vena Porta , Triazoles/farmacología
10.
Drug Metab Dispos ; 41(2): 518-28, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23223498

RESUMEN

DB868 [2,5-bis [5-(N-methoxyamidino)-2-pyridyl] furan], a prodrug of the diamidine DB829 [2,5-bis(5-amidino-2-pyridyl) furan], has demonstrated efficacy in murine models of human African trypanosomiasis. A cross-species evaluation of prodrug bioconversion to the active drug is required to predict the disposition of prodrug, metabolites, and active drug in humans. The phase I biotransformation of DB868 was elucidated using liver microsomes and sandwich-cultured hepatocytes from humans and rats. All systems produced four NADPH-dependent metabolites via O-demethylation (M1, M2) and N-dehydroxylation (M3, M4). Compartmental kinetic modeling of the DB868 metabolic pathway suggested an unusual N-demethoxylation reaction that was supported experimentally. A unienzyme Michaelis-Menten model described the kinetics of M1 formation by human liver microsomes (HLMs) (K(m), 11 µM; V(max), 340 pmol/min/mg), whereas a two-enzyme model described the kinetics of M1 formation by rat liver microsomes (RLMs) (K(m1), 0.5 µM; V(max1), 12 pmol/min/mg; K(m2), 27 µM; V(max2), 70 pmol/min/mg). Human recombinant CYP1A2, CYP3A4, and CYP4F2, rat recombinant Cyp1a2 and Cyp2d2, and rat purified Cyp4f1 catalyzed M1 formation. M2 formation by HLMs exhibited allosteric kinetics (S(50), 18 µM; V(max), 180 pmol/mg), whereas M2 formation by RLMs was negligible. Recombinant CYP1A2/Cyp1a2 catalyzed M2 formation. DB829 was detected in trace amounts in HLMs at the end of the 180-min incubation and was detected readily in sandwich-cultured hepatocytes from both species throughout the 24-h incubation. These studies demonstrated that DB868 biotransformation to DB829 is conserved between humans and rats. An improved understanding of species differences in the kinetics of DB829 formation would facilitate preclinical development of a promising antitrypanosomal prodrug.


Asunto(s)
Sistema Nervioso Central/efectos de los fármacos , Sistema Enzimático del Citocromo P-450/metabolismo , Hígado/enzimología , Modelos Biológicos , Profármacos/metabolismo , Profármacos/farmacología , Tripanocidas/metabolismo , Tripanocidas/farmacología , Animales , Biotransformación , Células Cultivadas , Remoción de Radical Alquila , Femenino , Hepatocitos/enzimología , Humanos , Hidroxilación , Isoenzimas , Cinética , Masculino , Metilación , Microsomas Hepáticos/enzimología , Estructura Molecular , Oxidación-Reducción , Profármacos/química , Ratas , Proteínas Recombinantes/metabolismo , Especificidad de la Especie , Tripanocidas/química
11.
Drug Metab Dispos ; 41(6): 1179-86, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23482500

RESUMEN

Sorafenib is an orally active tyrosine kinase inhibitor used in the treatment of renal and hepatocellular carcinoma. This study was designed to establish whether transport proteins are involved in the hepatic uptake of sorafenib and to determine the extent of biliary excretion of sorafenib and its metabolites in human hepatocytes. Initial uptake was assessed in freshly isolated, suspended human hepatocytes in the presence of inhibitors and modulators. [(14)C]Sorafenib (1 µM) uptake at 4°C was reduced by about 61-63% of the uptake at 37°C, suggesting a high degree of passive diffusion. Hepatocyte uptake of [(14)C]sorafenib was not Na(+) dependent or influenced by the organic anion transporter 2 inhibitor ketoprofen. However, initial [(14)C]sorafenib hepatocyte uptake was reduced by 46 and 30% compared with control values in the presence of the organic anion transporting polypeptide inhibitor rifamycin SV and the organic cation transporter (OCT) inhibitor decynium 22, respectively. [(14)C]Sorafenib (0.5-5 µM) uptake was significantly higher in hOCT1-transfected Chinese hamster ovary cells compared with mock cells, and inhibited by the general OCT inhibitor, 1-methyl-4-phenylpryidinium. OCT1-mediated uptake was saturable with a Michaelis-Menten constant of 3.80 ± 2.53 µM and a V(max) of 116 ± 42 pmol/mg/min. The biliary excretion index and in vitro biliary clearance of sorafenib (1 µM) in sandwich-cultured human hepatocytes were low (∼11% and 11 ml/min/kg, respectively). Results suggest that sorafenib uptake in human hepatocytes occurs via passive diffusion, by OCT1, and by organic anion transporting polypeptide(s). Sorafenib undergoes modest biliary excretion, predominantly as a glucuronide conjugate(s).


Asunto(s)
Hepatocitos/metabolismo , Niacinamida/análogos & derivados , Compuestos de Fenilurea/metabolismo , Inhibidores de Proteínas Quinasas/metabolismo , Adulto , Animales , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Femenino , Hepatocitos/efectos de los fármacos , Humanos , Masculino , Persona de Mediana Edad , Niacinamida/metabolismo , Niacinamida/farmacología , Compuestos de Fenilurea/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Sorafenib
12.
J Biol Chem ; 286(14): 12407-16, 2011 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-21266572

RESUMEN

Phospholipase C (PLC) enzymes are an important family of regulatory proteins involved in numerous cellular functions, primarily through hydrolysis of the polar head group from inositol-containing membrane phospholipids. U73122 (1-(6-((17ß-3-methoxyestra-1,3,5(10)-trien-17-yl)amino)hexyl)-1H-pyrrole-2,5-dione), one of only a few small molecules reported to inhibit the activity of these enzymes, has been broadly applied as a pharmacological tool to implicate PLCs in diverse experimental phenotypes. The purpose of this study was to develop a better understanding of molecular interactions between U73122 and PLCs. Hence, the effects of U73122 on human PLCß3 (hPLCß3) were evaluated in a cell-free micellar system. Surprisingly, U73122 increased the activity of hPLCß3 in a concentration- and time-dependent manner; up to an 8-fold increase in enzyme activity was observed with an EC50=13.6±5 µm. Activation of hPLCß3 by U73122 required covalent modification of cysteines as evidenced by the observation that enzyme activation was attenuated by thiol-containing nucleophiles, l-cysteine and glutathione. Mass spectrometric analysis confirmed covalent reaction with U73122 at eight cysteines, although maximum activation was achieved without complete alkylation; the modified residues were identified by LC/MS/MS peptide sequencing. Interestingly, U73122 (10 µm) also activated hPLCγ1 (>10-fold) and hPLCß2 (∼2-fold); PLCδ1 was neither activated nor inhibited. Therefore, in contrast to its reported inhibitory potential, U73122 failed to inhibit several purified PLCs. Most of these PLCs were directly activated by U73122, and a simple mechanism for the activation is proposed. These results strongly suggest a need to re-evaluate the use of U73122 as a general inhibitor of PLC isozymes.


Asunto(s)
Estrenos/farmacología , Inhibidores de Fosfodiesterasa/farmacología , Pirrolidinonas/farmacología , Fosfolipasas de Tipo C/metabolismo , Secuencia de Aminoácidos , Activación Enzimática/efectos de los fármacos , Estrenos/química , Humanos , Datos de Secuencia Molecular , Inhibidores de Fosfodiesterasa/química , Pirrolidinonas/química , Fosfolipasas de Tipo C/química
13.
Mol Pharm ; 8(5): 1677-86, 2011 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-21780830

RESUMEN

Fexofenadine is a nonsedative antihistamine that exhibits good oral bioavailability despite its zwitterionic chemical structure and efflux by P-gp. Evidence exists that multiple uptake and efflux transporters play a role in hepatic disposition of fexofenadine. However, the roles of specific transporters and their interrelationship in intestinal absorption of this drug are unclear. This study was designed to elucidate vectorial absorptive transport of fexofenadine across Caco-2 cells involving specific apical uptake and efflux transporters as well as basolateral efflux transporters. Studies with cellular models expressing single transporters showed that OATP2B1 expression stimulated uptake of fexofenadine at pH 6.0. Apical uptake of fexofenadine into Caco-2 cells was decreased by 45% by pretreatment with estrone 3-sulfate, an OATP inhibitor, at pH 6.0 but not at pH 7.4, indicating that OATP2B1 mediates apical uptake of fexofenadine into these cells. Examination of fexofenadine efflux from preloaded Caco-2 cells in the presence or absence of (i) the MRP inhibitor MK-571 and (ii) the P-gp inhibitor GW918 showed that apical efflux is predominantly mediated by P-gp, with a small contribution by MRP2, whereas basolateral efflux is predominantly mediated by MRP3. These results also showed that while OSTαß is functionally active in the basolateral membrane of Caco-2 cells, it does not play a role in the export of fexofenadine. MK-571 decreased the absorptive transport of fexofenadine by 17%. However, the decrease in absorptive transport by MK-571 was 42% when P-gp was inhibited by GW918. The results provide a novel insight into a vectorial transport system mainly consisting of apical OATP2B1 and basolateral MRP3 that may play an important role in delivering hydrophilic anionic and zwitterionic drugs such as pravastatin and fexofenadine into systemic circulation upon oral administration.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Enterocitos/metabolismo , Antagonistas de los Receptores Histamínicos H1 no Sedantes/metabolismo , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Transportadores de Anión Orgánico/metabolismo , Terfenadina/análogos & derivados , Subfamilia B de Transportador de Casetes de Unión a ATP , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/antagonistas & inhibidores , Animales , Células CACO-2 , Línea Celular , Polaridad Celular , Perros , Enterocitos/citología , Enterocitos/efectos de los fármacos , Humanos , Absorción Intestinal/efectos de los fármacos , Túbulos Renales Distales/efectos de los fármacos , Túbulos Renales Distales/metabolismo , Moduladores del Transporte de Membrana/farmacología , Modelos Biológicos , Proteína 2 Asociada a Resistencia a Múltiples Medicamentos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/antagonistas & inhibidores , Transportadores de Anión Orgánico/antagonistas & inhibidores , Transportadores de Anión Orgánico/genética , Profármacos/metabolismo , Transporte de Proteínas , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Terfenadina/metabolismo
14.
Biopharm Drug Dispos ; 32(4): 222-32, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21449041

RESUMEN

Micafungin, a new echinocandin antifungal agent, has been used widely for the treatment of various fungal infections in human populations. Micafungin is predominantly cleared by biliary excretion and it binds extensively to plasma proteins. Micafungin body weight-adjusted clearance is higher in neonates than in adults, but the mechanisms underlying this difference are not understood. Previous work had revealed the roles of sinusoidal uptake (Na(+) -taurocholate co-transporting peptide, NTCP; organic anion transporting polypeptide, OATP) as well as canalicular efflux (bile salt export pump, BSEP; breast cancer resistance protein, BCRP) transporters in micafungin hepatobiliary elimination. In the present study, the relative protein expression of hepatic transporters was compared between liver homogenates from neonates and adults. Also, the extent of micafungin binding to serum from neonates and adults was measured in vitro. The results indicate that relative expression levels of NTCP, OATP1B1/3, BSEP, BCRP and MRP3 were similar in neonates and in adults. However, the micafungin fraction unbound (f(u) ) in neonatal serum was about 8-fold higher than in the adult serum (0.033±0.012 versus 0.004±0.001, respectively). While there was no evidence for different intrinsic hepatobiliary clearance of micafungin between neonates and adults, our data suggest that age-dependent serum protein binding of micafungin is responsible for its higher clearance in neonates compared with adults.


Asunto(s)
Antifúngicos/farmacocinética , Equinocandinas/farmacocinética , Lipopéptidos/farmacocinética , Adulto , Factores de Edad , Antifúngicos/sangre , Antifúngicos/química , Antifúngicos/uso terapéutico , Sistema Biliar/fisiología , Proteínas Sanguíneas/metabolismo , Equinocandinas/sangre , Equinocandinas/química , Equinocandinas/uso terapéutico , Humanos , Recién Nacido , Lipopéptidos/sangre , Lipopéptidos/química , Lipopéptidos/uso terapéutico , Hígado/metabolismo , Micafungina , Transportadores de Anión Orgánico/metabolismo , Unión Proteica , Simportadores/efectos de los fármacos , Simportadores/metabolismo
15.
Drug Metab Dispos ; 38(1): 25-31, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19841059

RESUMEN

Voriconazole is a broad spectrum antifungal agent for treating life-threatening fungal infections. Its clearance is approximately 3-fold higher in children compared with adults. Voriconazole is cleared predominantly via hepatic metabolism in adults, mainly by CYP3A4, CYP2C19, and flavin-containing monooxygenase 3 (FMO3). In vitro metabolism of voriconazole by liver microsomes prepared from pediatric and adult tissues (n = 6/group) mirrored the in vivo clearance differences in children versus adults, and it showed that the oxidative metabolism was significantly faster in children compared with adults as indicated by the in vitro half-life (T(1/2)) of 33.8 + or - 15.3 versus 72.6 + or - 23.7 min, respectively. The K(m) for voriconazole metabolism to N-oxide, the major metabolite formed in humans, by liver microsomes from children and adults was similar (11 + or - 5.2 versus 9.3 + or - 3.6 microM, respectively). In contrast, apparent V(max) was approximately 3-fold higher in children compared with adults (120.5 + or - 99.9 versus 40 + or - 13.9 pmol/min/mg). The calculated in vivo clearance from in vitro data was found to be approximately 80% of the observed plasma clearance values in both populations. Metabolism studies in which CYP3A4, CYP2C19, or FMO was selectively inhibited provided evidence that contribution of CYP2C19 and FMO toward voriconazole N-oxidation was much greater in children than in adults, whereas CYP3A4 played a larger role in adults. Although expression of CYP2C19 and FMO3 is not significantly different in children versus adults, these enzymes seem to contribute to higher metabolic clearance of voriconazole in children versus adults.


Asunto(s)
Envejecimiento/metabolismo , Hidrocarburo de Aril Hidroxilasas/metabolismo , Hígado/metabolismo , Oxigenasas/metabolismo , Pirimidinas/metabolismo , Triazoles/metabolismo , Adulto , Antifúngicos/metabolismo , Antifúngicos/farmacocinética , Hidrocarburo de Aril Hidroxilasas/antagonistas & inhibidores , Biocatálisis/efectos de los fármacos , Niño , Preescolar , Citocromo P-450 CYP2C19 , Citocromo P-450 CYP3A/metabolismo , Inhibidores del Citocromo P-450 CYP3A , Inhibidores Enzimáticos del Citocromo P-450 , Sistema Enzimático del Citocromo P-450/metabolismo , Inhibidores Enzimáticos/farmacología , Humanos , Cinética , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/enzimología , Persona de Mediana Edad , Modelos Biológicos , Pirimidinas/farmacocinética , Triazoles/farmacocinética , Voriconazol , Adulto Joven
16.
Drug Metab Dispos ; 38(10): 1848-56, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20606004

RESUMEN

The purpose of the present study was to elucidate the transport mechanisms responsible for elimination of micafungin, a new semisynthetic echinocandin antifungal agent, which is predominantly cleared by biliary excretion in humans and rats. In vitro studies using sandwich-cultured rat and human hepatocytes were conducted. Micafungin uptake occurred primarily (∼75%) by transporter-mediated mechanisms in rat and human. Micafungin uptake into hepatocytes was inhibited by taurocholate (K(i) = 61 µM), Na(+) depletion (45-55% reduced), and 10 µM rifampin (20-25% reduced); these observations support the involvement of Na(+)-taurocholate-cotransporting polypeptide (NTCP/Ntcp) and, to a lesser extent, organic anion-transporting polypeptides in the hepatic uptake of micafungin. The in vitro biliary clearance of micafungin, as measured by the B-CLEAR technique, amounted to 14 and 19 µl/(min · mg protein) in human and rat, respectively. In vitro biliary excretion of micafungin was reduced by 80 and 75% in the presence of the bile salt export pump (BSEP) inhibitors taurocholate (100 µM) and nefazodone (25 µM), respectively. Biliary excretion of micafungin also was reduced in the presence of breast cancer resistance protein inhibitors [N-(4-[2-(1,2,3,4-tetrahydro-6,7-dimethoxy-2-isoquinolinyl)ethyl]-phenyl)-9,10-dihydro-5-methoxy-9-oxo-4-acridine carboxamide (GF120918) (10 µM) and fumitremorgin C (10 µM)]. In vitro biliary excretion of micafungin was not significantly altered by coincubation with P-glycoprotein or multidrug resistance-associated protein 2 inhibitors. These results suggest that NTCP/Ntcp and BSEP/Bsep are primarily responsible for hepatobiliary disposition of micafungin in human and rat. Interference with hepatic bile acid disposition could be one mechanism underlying hepatotoxicity associated with micafungin in some patients.


Asunto(s)
Antifúngicos/farmacocinética , Sistema Biliar/metabolismo , Equinocandinas/farmacocinética , Lipopéptidos/farmacocinética , Hígado/metabolismo , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Antifúngicos/metabolismo , Canalículos Biliares/metabolismo , Técnicas de Cultivo de Célula , Células Cultivadas , Cromatografía Líquida de Alta Presión , Relación Dosis-Respuesta a Droga , Equinocandinas/metabolismo , Hepatocitos/metabolismo , Humanos , Inactivación Metabólica , Lipopéptidos/metabolismo , Masculino , Micafungina , Transportadores de Anión Orgánico Sodio-Dependiente/metabolismo , Ratas , Ratas Wistar , Simportadores/metabolismo , Distribución Tisular
17.
Drug Metab Dispos ; 37(2): 424-30, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18971316

RESUMEN

The antiparasitic activity of aromatic diamidine drugs, pentamidine and furamidine, depends on their entry into the pathogenic protozoa via membrane transporters. However, no such diamidine transporter has been identified in mammalian cells. The goal of this study is to investigate whether these dicationic drugs are substrates for human organic cation transporters (hOCTs, solute carrier family 22A1-3) and whether hOCTs play a role in their tissue distribution, elimination, and toxicity. Inhibitory and substrate activities of pentamidine and furamidine were studied in stably transfected Chinese hamster ovary (CHO) cells. The results of [(3)H]1-methyl-4-phenylpyridinium uptake study showed that pentamidine is a potent inhibitor for all three OCT isoforms (IC50 < 20 microM), whereas furamidine is a potent inhibitor for hOCT1 and hOCT3 (IC50 < 21 microM) but a less potent inhibitor for hOCT2 (IC50 = 189.2 microM). Both diamidines are good substrates for hOCT1 (Km = 36.4 and 6.1 microM, respectively), but neither is a substrate for hOCT2 or hOCT3. The cytotoxicity of pentamidine and furamidine was 4.4- and 9.3-fold greater, respectively, in CHO-hOCT1 cells compared with the mock cells. Ranitidine, an hOCT1 inhibitor, reversed this hOCT1-mediated potentiation of cytotoxicity. This is the first finding that dicationic drugs, such as pentamidine and furamidine, are substrates for hOCT1. In humans, aromatic diamidines are primarily eliminated in the bile but are distributed and cause toxicity in both liver and kidney. These transporters may play important roles in the disposition of aromatic diamidines in humans, as well as resultant drug-drug interactions and toxicity involving diamidine drugs.


Asunto(s)
Antifúngicos/farmacología , Benzamidinas/farmacología , Transporte Biológico/fisiología , Cationes/metabolismo , Proteínas de Transporte de Catión Orgánico/metabolismo , Pentamidina/farmacología , Femenino , Glucurónidos/química , Humanos , Proteínas de Transporte de Catión Orgánico/antagonistas & inhibidores , Transportador 1 de Catión Orgánico/antagonistas & inhibidores , Transportador 1 de Catión Orgánico/metabolismo
18.
Br J Pharmacol ; 176(15): 2724-2735, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31032880

RESUMEN

BACKGROUND AND PURPOSE: It has been extensively reported that the leading anti-diabetic drug, metformin, exerts significant anticancer effects. This hydrophilic, cationic drug requires cation transporters for cellular entry where it activates its intracellular target, the AMPK signalling pathway. However, clinical results on metformin therapy (used at antidiabetic doses) for breast cancer are ambiguous. It is likely that the antidiabetic dose is inadequate in patients that have breast tumours with low cation transporter expression, resulting in non-responsiveness to the drug. We postulate that cation transporter expression and metformin dose are key determinants in its antitumour efficacy in breast cancer. EXPERIMENTAL APPROACH: Antitumour efficacy of metformin was compared between low cation transporter-expressing MCF-7 breast tumours and MCF-7 tumours overexpressing organic cation transporter 3 (OCT3-MCF7). A dose-response relationship of metformin in combination with standard-of-care paclitaxel (for oestrogen receptor-positive MCF-7 breast tumours) or carboplatin (for triple-negative MDA-MB-468 breast tumours) was investigated in xenograft mice. KEY RESULTS: Metformin had greater efficacy against tumours with higher cation transporter expression, as observed in OCT3-MCF7 versus MCF-7 tumours and MDA-MB-468 versus MCF-7 tumours. In MCF-7 tumours, a threefold higher metformin dose was required to achieve intratumoural exposure that was comparable to exposure in MDA-MB-468 tumours and enhance antitumour efficacy of standard-of-care in MCF-7 tumours versus MDA-MB-468 tumours. Antitumour efficacy correlated with intratumoural AMPK activation and metformin concentration. CONCLUSIONS AND IMPLICATIONS: An efficacious metformin dose for breast cancer varies among tumour subtypes based on cation transporter expression, which provides a useful guide for dose selection.


Asunto(s)
Antineoplásicos/uso terapéutico , Hipoglucemiantes/uso terapéutico , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Metformina/uso terapéutico , Proteínas de Transporte de Catión Orgánico/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Antineoplásicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Carboplatino/uso terapéutico , Línea Celular Tumoral , Sinergismo Farmacológico , Femenino , Humanos , Hipoglucemiantes/farmacología , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Metformina/farmacología , Ratones Desnudos , Paclitaxel/uso terapéutico , Carga Tumoral/efectos de los fármacos
19.
CPT Pharmacometrics Syst Pharmacol ; 8(10): 759-768, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31420944

RESUMEN

Vincristine is a cytotoxic chemotherapeutic agent used as first-line therapy for pediatric acute lymphocytic leukemia. It is cleared by hepatic oxidative metabolism by CYP3A4 and CYP3A5 and via hepatic (biliary) efflux mediated by P-glycoprotein (P-gp) transporter. Bottom-up physiologically based pharmacokinetic (PBPK) models were developed to predict vincristine disposition in pediatric and adult populations. The models incorporated physicochemical properties, metabolism by CYP3A4/5, efflux by P-gp, and intracellular binding to ß-tubulin. The adult and pediatric PBPK models predicted pharmacokinetics (PK) within twofold of the observed PK parameters (area under the curve, terminal half-life, volume of distribution, and clearance). Simulating a higher hypothetical (4.9-fold) pediatric expression of ß-tubulin relative to adult improved predictions of vincristine PKs. To our knowledge, this is the first time that intracellular binding has been incorporated into a pediatric PBPK model. Utilizing this PBPK modeling approach, safe and effective doses of vincristine could be predicted.


Asunto(s)
Antineoplásicos Fitogénicos/farmacocinética , Citocromo P-450 CYP3A/metabolismo , Tubulina (Proteína)/metabolismo , Vincristina/farmacocinética , Administración Intravenosa , Adulto , Animales , Antineoplásicos Fitogénicos/administración & dosificación , Área Bajo la Curva , Línea Celular , Niño , Preescolar , Perros , Humanos , Lactante , Recién Nacido , Persona de Mediana Edad , Modelos Biológicos , Vincristina/administración & dosificación , Adulto Joven
20.
Drug Metab Dispos ; 36(8): 1650-8, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18458049

RESUMEN

The purpose of the study was to elucidate mechanisms of metformin absorptive transport to explain the dose-dependent absorption observed in humans. Apical (AP) and basolateral (BL) uptake and efflux as well as AP to BL (absorptive) transport across Caco-2 cell monolayers were evaluated over a range of concentrations. Transport was concentration-dependent and consisted of saturable and nonsaturable components (K(m) approximately 0.05 mM, J(max) approximately 1.0 pmol min(-1) cm(-2), and K(d, transport) approximately 10 nl min(-1) cm(-2)). AP uptake data also revealed the presence of saturable and nonsaturable components (K(m) approximately 0.9 mM, V(max) approximately 330 pmol min(-1) mg of protein(-1), and K(d, uptake) approximately 0.04 microl min(-1) mg of protein(-1)). BL efflux was rate-limiting to transcellular transport of metformin; AP efflux was 7-fold greater than BL efflux and was not inhibited by N-(4-[2-(1,2,3,4-tetrahydro-6,7-dimethoxy-2-isoquinolinyl)ethyl]-phenyl)-9,10-dihydro-5-methoxy-9-oxo-4-acridine carboxamide (GW918), a P-glycoprotein inhibitor. AP efflux was trans-stimulated by metformin and prototypical substrates of organic cation transporters, suggesting that a cation-specific bidirectional transport mechanism mediated the AP efflux of metformin. BL efflux of intracellular metformin was much less efficient in comparison with the overall transport, with BL efflux clearance accounting for approximately 7 and approximately 13% of the overall transport clearance at 0.05 and 10 mM metformin concentrations, respectively. Kinetic modeling of cellular accumulation and transport processes supports the finding that transport occurs almost exclusively via the paracellular route (approximately 90%) and that the paracellular transport is saturable. This report provides strong evidence for a saturable mechanism in the paracellular space and provides insight into possible mechanisms for the dose dependence of metformin absorption in vivo.


Asunto(s)
Sistema Enzimático del Citocromo P-450/metabolismo , Hipoglucemiantes/farmacocinética , Absorción Intestinal , Metformina/farmacocinética , Células CACO-2 , Cromatografía Líquida de Alta Presión , ADN Complementario , Humanos , Espectrometría de Masas , Microsomas Hepáticos/enzimología , Microsomas Hepáticos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA