Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Proc Natl Acad Sci U S A ; 110(27): 11199-204, 2013 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-23776246

RESUMEN

Oxaliplatin is an integral component of colorectal cancer therapy, but its clinical use is associated with a dose-limiting peripheral neurotoxicity. We found that the organic cation transporter 2 (OCT2) is expressed on dorsal root ganglia cells within the nervous system where oxaliplatin is known to accumulate. Cellular uptake of oxaliplatin was increased by 16- to 35-fold in cells overexpressing mouse Oct2 or human OCT2, and this process was associated with increased DNA platination and oxaliplatin-induced cytotoxicity. Furthermore, genetic or pharmacologic knockout of Oct2 protected mice from hypersensitivity to cold or mechanical-induced allodynia, which are established tests to assess acute oxaliplatin-induced neurotoxicity. These findings provide a rationale for the development of targeted approaches to mitigate this debilitating toxicity.


Asunto(s)
Síndromes de Neurotoxicidad/etiología , Síndromes de Neurotoxicidad/metabolismo , Proteínas de Transporte de Catión Orgánico/fisiología , Compuestos Organoplatinos/toxicidad , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/toxicidad , Femenino , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/metabolismo , Humanos , Masculino , Ratones , Ratones Noqueados , Síndromes de Neurotoxicidad/genética , Factor 1 de Transcripción de Unión a Octámeros/deficiencia , Factor 1 de Transcripción de Unión a Octámeros/genética , Factor 1 de Transcripción de Unión a Octámeros/fisiología , Proteínas de Transporte de Catión Orgánico/deficiencia , Proteínas de Transporte de Catión Orgánico/genética , Transportador 2 de Cátion Orgánico , Compuestos Organoplatinos/farmacocinética , Oxaliplatino
2.
Am J Physiol Cell Physiol ; 305(12): C1223-9, 2013 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-23986199

RESUMEN

Hydroxyurea is currently the only FDA-approved drug that ameliorates the pathophysiology of sickle cell anemia. Unfortunately, substantial interpatient variability in the pharmacokinetics (PK) of hydroxyurea may result in variation of the drug's efficacy. However, little is known about mechanisms that modulate hydroxyurea PK. Recent in vitro studies identifying hydroxyurea as a substrate for organic anion transporting polypeptide (OATP1B) transporters prompted the current investigation assessing the role of OATP1B transporters in modulating hydroxyurea PK. Using wild-type and Oatp1b knockout (Oatp1b(-/-)) mice, hydroxyurea PK was analyzed in vivo by measuring [(14)C]hydroxyurea distribution in plasma, kidney, liver, urine, or the exhaled (14)CO2 metabolite. Plasma levels were significantly reduced by 20% in Oatp1b(-/-) mice compared with wild-type (area under the curve of 38.64 or 48.45 µg·h(-1)·ml(-1), respectively) after oral administration, whereas no difference was observed between groups following intravenous administration. Accumulation in the kidney was significantly decreased by twofold in Oatp1b(-/-) mice (356.9 vs. 748.1 pmol/g), which correlated with a significant decrease in urinary excretion. Hydroxyurea accumulation in the liver was also decreased (136.6 vs. 107.3 pmol/g in wild-type or Oatp1b(-/-) mice, respectively) correlating with a decrease in exhaled (14)CO2. These findings illustrate that deficiency of Oatp1b transporters alters the absorption, distribution, and elimination of hydroxyurea thus providing the first in vivo evidence that cell membrane transporters may play a significant role in modulating hydroxyurea PK. Future studies to investigate other transporters and their role in hydroxyurea disposition are warranted for understanding the sources of variation in hydroxyurea's PK.


Asunto(s)
Antidrepanocíticos/farmacocinética , Hidroxiurea/farmacocinética , Transportadores de Anión Orgánico Sodio-Independiente/metabolismo , Animales , Área Bajo la Curva , Femenino , Regulación de la Expresión Génica/fisiología , Transportador 1 de Anión Orgánico Específico del Hígado , Ratones , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Transportadores de Anión Orgánico Sodio-Independiente/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo
3.
Mol Cell Biol ; 27(20): 7007-17, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17698581

RESUMEN

The conserved TOR (target of rapamycin) kinase is part of a TORC1 complex that regulates cellular responses to environmental stress, such as amino acid starvation and hypoxia. Dysregulation of Akt-TOR signaling has also been linked to the genesis of cancer, and thus, this pathway presents potential targets for cancer chemotherapeutics. Here we report that rapamycin-sensitive TORC1 signaling is required for the S-phase progression and viability of yeast cells in response to genotoxic stress. In the presence of the DNA-damaging agent methyl methanesulfonate (MMS), TOR-dependent cell survival required a functional S-phase checkpoint. Rapamycin inhibition of TORC1 signaling suppressed the Rad53 checkpoint-mediated induction of ribonucleotide reductase subunits Rnr1 and Rnr3, thereby abrogating MMS-induced mutagenesis and enhancing cell lethality. Moreover, cells deleted for RNR3 were hypersensitive to rapamycin plus MMS, providing the first demonstration that Rnr3 contributes to the survival of cells exposed to DNA damage. Our findings support a model whereby TORC1 acts as a survival pathway in response to genotoxic stress by maintaining the deoxynucleoside triphosphate pools necessary for error-prone translesion DNA polymerases. Thus, TOR-dependent cell survival in response to DNA-damaging agents coincides with increased mutation rates, which may contribute to the acquisition of chemotherapeutic drug resistance.


Asunto(s)
Supervivencia Celular , Daño del ADN , Proteínas de Saccharomyces cerevisiae/metabolismo , Transducción de Señal/fisiología , Adulto , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Quinasa de Punto de Control 2 , Cicloheximida/metabolismo , Humanos , Metilmetanosulfonato/metabolismo , Complejos Multiproteicos , Mutágenos/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Ribonucleósido Difosfato Reductasa/genética , Ribonucleósido Difosfato Reductasa/metabolismo , Fase S/fisiología , Saccharomyces cerevisiae/citología , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Sirolimus/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
4.
Mol Cell Biol ; 26(13): 4958-69, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16782883

RESUMEN

Covalent modification with SUMO alters protein function, intracellular localization, or protein-protein interactions. Target recognition is determined, in part, by the SUMO E2 enzyme, Ubc9, while Siz/Pias E3 ligases may facilitate select interactions by acting as substrate adaptors. A yeast conditional Ubc9P(123)L mutant was viable at 36 degrees C yet exhibited enhanced sensitivity to DNA damage. To define functional domains in Ubc9 that dictate cellular responses to genotoxic stress versus those necessary for cell viability, a 1.75-A structure of yeast Ubc9 that demonstrated considerable conservation of backbone architecture with human Ubc9 was solved. Nevertheless, differences in side chain geometry/charge guided the design of human/yeast chimeras, where swapping domains implicated in (i) binding residues within substrates that flank canonical SUMOylation sites, (ii) interactions with the RanBP2 E3 ligase, and (iii) binding of the heterodimeric E1 and SUMO had distinct effects on cell growth and resistance to DNA-damaging agents. Our findings establish a functional interaction between N-terminal and substrate-binding domains of Ubc9 and distinguish the activities of E3 ligases Siz1 and Siz2 in regulating cellular responses to genotoxic stress.


Asunto(s)
Daño del ADN , Proteínas de Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/enzimología , Enzimas Ubiquitina-Conjugadoras/química , Secuencia de Aminoácidos , Daño del ADN/genética , Humanos , Datos de Secuencia Molecular , Estructura Terciaria de Proteína/genética , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Saccharomyces cerevisiae/citología , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Especificidad por Sustrato , Ubiquitina/metabolismo , Enzimas Ubiquitina-Conjugadoras/genética , Enzimas Ubiquitina-Conjugadoras/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Difracción de Rayos X
5.
Clin Cancer Res ; 20(15): 4026-35, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24916697

RESUMEN

PURPOSE: Tubular secretion of cisplatin is abolished in mice deficient for the organic cation transporters Oct1 and Oct2 (Oct1/2(-/-)mice), and these animals are protected from severe cisplatin-induced kidney damage. Since tubular necrosis is not completely absent in Oct1/2(-/-)mice, we hypothesized that alternate pathways are involved in the observed injury. EXPERIMENTAL DESIGN: Studies were done in wild-type, Oct1/2(-/-), or p53-deficient animals, all on an FVB background, receiving cisplatin intraperitoneally at 15 mg/kg. Cisplatin metabolites were analyzed using mass spectrometry, and gene expression was assessed using Affymetrix microarrays and RT-PCR arrays. RESULTS: KEGG pathway analyses on kidneys from mice exposed to cisplatin revealed that the most significantly altered genes were associated with the p53 signaling network, including Cdnk1a and Mdm2, in both wild-type (P = 2.40 × 10(-11)) and Oct1/2(-/-)mice (P = 1.92 × 10(-8)). This was confirmed by demonstrating that homozygosity for a p53-null allele partially reduced renal tubular damage, whereas loss of p53 in Oct1/2(-/-)mice (p53(-/-)/Oct1/2(-/-)) completely abolished nephrotoxicity. We found that pifithrin-α, an inhibitor of p53-dependent transcriptional activation, inhibits Oct2 and can mimic the lack of nephrotoxicity observed in p53(-/-)/Oct1/2(-/-)mice. CONCLUSIONS: These findings indicate that (i) the p53 pathway plays a crucial role in the kidney in response to cisplatin treatment and (ii) clinical exploration of OCT2 inhibitors may not lead to complete nephroprotection unless the p53 pathway is simultaneously antagonized.


Asunto(s)
Antineoplásicos/toxicidad , Cisplatino/toxicidad , Enfermedades Renales/inducido químicamente , Enfermedades Renales/prevención & control , Factor 1 de Transcripción de Unión a Octámeros/fisiología , Proteínas de Transporte de Catión Orgánico/fisiología , Proteína p53 Supresora de Tumor/fisiología , Animales , Antineoplásicos/metabolismo , Benzotiazoles/farmacología , Biomarcadores/metabolismo , Cisplatino/metabolismo , Femenino , Perfilación de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Enfermedades Renales/patología , Masculino , Ratones , Ratones Noqueados , Factor 1 de Transcripción de Unión a Octámeros/antagonistas & inhibidores , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas de Transporte de Catión Orgánico/antagonistas & inhibidores , Transportador 2 de Cátion Orgánico , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tolueno/análogos & derivados , Tolueno/farmacología , Proteína p53 Supresora de Tumor/antagonistas & inhibidores
6.
Mol Cancer Ther ; 12(8): 1537-44, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23757163

RESUMEN

Expression of the human organic anion transporting polypeptides OATP1B1 and OATP1B3 has been previously believed to be restricted to hepatocytes. Here we show that the gene encoding OATP1B3, but not OATP1B1, is abundantly expressed in multiple human solid tumors that include hepatocellular, lung, and ovarian carcinomas. Surprisingly, OATP1B3 gene expression in a panel of 60 human tumor cell lines was linked with sensitivity to multiple cytotoxic agents, including the platinum anticancer drugs cisplatin, carboplatin, and oxaliplatin. In addition, overexpression of OATP1B3 in mammalian cells increased cellular accumulation of platinum agents and decreased cell survival. In mice with a targeted disruption of the ortholog transporter Oatp1b2, the liver-to-plasma ratio of cisplatin was significantly reduced compared with wild-type mice, without concurrent changes in expression profiles of other transporter genes. Our findings indicate an unexpected role for tumoral and host OATP1B-type carriers in the toxicity and disposition of platinum anticancer drugs, and may provide a foundation for understanding the extensive interindividual pharmacodynamic variability seen with these drugs in patients.


Asunto(s)
Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Transportadores de Anión Orgánico/metabolismo , Platino (Metal)/metabolismo , Platino (Metal)/farmacología , Animales , Antineoplásicos/farmacocinética , Transporte Biológico , Línea Celular Tumoral , Resistencia a Antineoplásicos/genética , Expresión Génica , Humanos , Transportador 1 de Anión Orgánico Específico del Hígado , Masculino , Ratones , Neoplasias/genética , Neoplasias/metabolismo , Transportadores de Anión Orgánico/deficiencia , Transportadores de Anión Orgánico/genética , Transportadores de Anión Orgánico Sodio-Independiente/genética , Transportadores de Anión Orgánico Sodio-Independiente/metabolismo , Platino (Metal)/farmacocinética , Miembro 1B3 de la Familia de los Transportadores de Solutos de Aniones Orgánicos
7.
Mol Cancer Ther ; 11(4): 921-9, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22389472

RESUMEN

OCTN2 is a bifunctional transporter that reabsorbs filtered carnitine in a sodium-dependent manner and secretes organic cations into urine as a proton antiport mechanism. We hypothesized that inhibition of OCTN2 by anticancer drugs can influence carnitine resorption. OCTN2-mediated transport inhibition by anticancer drugs was assessed using cells transfected with human OCTN2 (hOCTN2) or mouse Octn2 (mOctn2). Excretion of carnitine and acetylcarnitine was measured in urine collected from mice and pediatric patients with cancer before and after administration of etoposide. Five of 27 tested drugs (50-100 µmol/L) inhibited hOCTN2-mediated carnitine uptake by 42% to 85% (P < 0.001). Of these inhibitors, etoposide was itself a transported substrate of hOCTN2 and mOctn2. Etoposide uptake by hOCTN2 was reversed in the presence of excess carnitine. This competitive inhibitory mechanism was confirmed in an in silico molecular docking analysis. In addition, etoposide inhibited the transcellular apical-to-basolateral flux of carnitine in kidney cells. Etoposide was also associated with a significant urinary loss of carnitine in mice (~1.5-fold) and in patients with cancer (~2.4-fold). Collectively, these findings indicate that etoposide can inhibit hOCTN2 function, potentially disturb carnitine homeostasis, and that this phenomenon can contribute to treatment-related toxicities.


Asunto(s)
Carnitina/metabolismo , Etopósido/farmacología , Proteínas de Transporte de Catión Orgánico/antagonistas & inhibidores , Acetilcarnitina/orina , Adolescente , Animales , Antineoplásicos Fitogénicos/farmacocinética , Antineoplásicos Fitogénicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Transporte Biológico , Carnitina/farmacocinética , Carnitina/orina , Técnicas de Cultivo de Célula , Línea Celular , Niño , Etopósido/administración & dosificación , Etopósido/farmacocinética , Células HEK293 , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/orina , Masculino , Ratones , Proteínas de Transporte de Catión Orgánico/genética , Proteínas de Transporte de Catión Orgánico/metabolismo , Miembro 5 de la Familia 22 de Transportadores de Solutos , Porcinos , Transfección
8.
Clin Cancer Res ; 18(4): 1101-8, 2012 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-22223530

RESUMEN

PURPOSE: Knowledge of transporters responsible for the renal secretion of creatinine is key to a proper interpretation of serum creatinine and/or creatinine clearance as markers of renal function in cancer patients receiving chemotherapeutic agents. EXPERIMENTAL DESIGN: Creatinine transport was studied in transfected HEK293 cells in vitro and in wild-type mice and age-matched organic cation transporter 1 and 2-deficient [Oct1/2(-/-)] mice ex vivo and in vivo. Clinical pharmacogenetic and transport inhibition studies were done in two separate cohorts of cancer patients. RESULTS: Compared with wild-type mice, creatinine clearance was significantly impaired in Oct1/2(-/-) mice. Furthermore, creatinine inhibited organic cation transport in freshly isolated proximal tubules from wild-type mice and humans, but not in those from Oct1/2(-/-) mice. In a genetic association analysis (n = 590), several polymorphisms around the OCT2/SLC22A2 gene locus, including rs2504954 (P = 0.000873), were significantly associated with age-adjusted creatinine levels. Furthermore, in cancer patients (n = 68), the OCT2 substrate cisplatin caused an acute elevation of serum creatinine (P = 0.0083), consistent with inhibition of an elimination pathway. CONCLUSIONS: Collectively, this study shows that OCT2 plays a decisive role in the renal secretion of creatinine. This process can be inhibited by OCT2 substrates, which impair the usefulness of creatinine as a marker of renal function.


Asunto(s)
Creatinina/metabolismo , Túbulos Renales Proximales/metabolismo , Proteínas de Transporte de Catión Orgánico/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/uso terapéutico , Transporte Biológico , Línea Celular , Cisplatino/administración & dosificación , Cisplatino/uso terapéutico , Creatinina/sangre , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Proteínas de Transporte de Catión Orgánico/genética , Transportador 2 de Cátion Orgánico , Polimorfismo de Nucleótido Simple , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética
9.
Clin Cancer Res ; 18(16): 4433-40, 2012 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-22711709

RESUMEN

PURPOSE: Docetaxel is extensively metabolized by CYP3A4 in the liver but mechanisms by which the drug is taken up into hepatocytes remain poorly understood. We hypothesized that (i) liver uptake of docetaxel is mediated by the polymorphic solute carriers OATP1B1 and OATP1B3 and (ii) inherited genetic defects in this process may impair systemic drug elimination. EXPERIMENTAL DESIGN: Transport of docetaxel was studied in vitro using various cell lines stably transfected with OATP1B1*1A (wild-type), OATP1B1*5 [c.521T>C (V174A); rs4149056], OATP1B3, or the mouse transporter Oatp1b2. Docetaxel clearance was evaluated in wild-type and Oatp1b2-knockout mice as well as in two cohorts of patients with multiple variant transporter genotypes (n = 213). RESULTS: Docetaxel was found to be a substrate for OATP1B1, OATP1B3, and Oatp1b2 but was not transported by OATP1B1*5. Deficiency of Oatp1b2 in mice was associated with an 18-fold decrease in docetaxel clearance (P = 0.0099), which was unrelated to changes in intrinsic metabolic capacity in mouse liver microsomes. In patients, however, none of the studied common reduced function variants in OATP1B1 or OATP1B3 were associated with docetaxel clearance (P > 0.05). CONCLUSIONS: The existence of at least two potentially redundant uptake transporters in the human liver with similar affinity for docetaxel supports the possibility that functional defects in both of these proteins may be required to confer substantially altered disposition phenotypes. In view of the established exposure-toxicity relationships for docetaxel, we suggest that caution is warranted if docetaxel has to be administered together with agents that potently inhibit both OATP1B1 and OATP1B3.


Asunto(s)
Antineoplásicos/metabolismo , Transportadores de Anión Orgánico/genética , Polimorfismo Genético , Taxoides/metabolismo , Adulto , Anciano , Animales , Antineoplásicos/farmacocinética , Línea Celular , Cricetinae , Docetaxel , Femenino , Genotipo , Hepatocitos/metabolismo , Humanos , Inactivación Metabólica , Hígado/metabolismo , Transportador 1 de Anión Orgánico Específico del Hígado , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/metabolismo , Transportadores de Anión Orgánico/deficiencia , Transportadores de Anión Orgánico Sodio-Independiente/genética , Miembro 1B3 de la Familia de los Transportadores de Solutos de Aniones Orgánicos , Taxoides/farmacocinética
10.
Clin Cancer Res ; 16(19): 4789-99, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20858838

RESUMEN

PURPOSE: Carnitine is an essential cofactor for mitochondrial fatty acid oxidation that is actively reabsorbed by the luminal transporter Octn2 (Slc22a5). Because the nephrotoxic agent cisplatin causes urinary loss of carnitine in humans, we hypothesized that cisplatin may affect Octn2 function. EXPERIMENTAL DESIGN: Excretion of carnitine and acetylcarnitine was measured in urine collected from mice with or without cisplatin administration. The transport of carnitine was assessed in cells that were transfected with OCT1 or OCT2. The effect of cisplatin treatment on gene expression was analyzed using a mouse GeneChip array and validated using quantitative reverse transcriptase-PCR. RESULTS: In wild-type mice, urinary carnitine excretion at baseline was ∼3-fold higher than in mice lacking the basolateral cisplatin transporters Oct1 and Oct2 [Oct1/2(-/-) mice], indicating that carnitine itself undergoes basolateral uptake into the kidney. Transport of carnitine by OCT2, but not OCT1, was confirmed in transfected cells. We also found that cisplatin caused an increase in the urinary excretion of carnitine and acetylcarnitine in wild-type mice but not in Oct1/2(-/-) mice, suggesting that tubular transport of cisplatin is a prerequisite for this phenomenon. Cisplatin did not directly inhibit the transport of carnitine by Octn2 but downregulated multiple target genes of the transcription factor peroxisome proliferator activated receptor α, including Slc22a5, in the kidney of wild-type mice that were absent in Oct1/2(-/-) mice. CONCLUSION: Our study shows a pivotal role of Oct1 and Oct2 in cisplatin-related disturbances in carnitine homeostasis. We postulate that this phenomenon is triggered by deactivation of peroxisome proliferator activated receptor α and leads to deregulation of carnitine-shuttle genes.


Asunto(s)
Carnitina/orina , Cisplatino/farmacología , Regulación hacia Abajo/efectos de los fármacos , Proteínas de Transporte de Catión Orgánico/antagonistas & inhibidores , Acetilcarnitina/orina , Animales , Proteínas Portadoras/antagonistas & inhibidores , Proteínas Portadoras/metabolismo , Línea Celular , Perfilación de la Expresión Génica , Humanos , Masculino , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos , Ratones Noqueados , Proteínas de Transporte de Catión Orgánico/deficiencia , Proteínas de Transporte de Catión Orgánico/metabolismo , PPAR alfa/antagonistas & inhibidores , PPAR alfa/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Miembro 5 de la Familia 22 de Transportadores de Solutos , Simportadores
11.
Clin Cancer Res ; 16(16): 4198-206, 2010 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-20601443

RESUMEN

PURPOSE: This study aimed to test the influence of functional renal organic cation transporters (OCT2 in humans, Oct1 and Oct2 in mice) on biomarkers of cisplatin nephrotoxicity, such as urinary activity of N-acetyl-beta-D-glucosaminidase (NAG). EXPERIMENTAL DESIGN: Temporal cisplatin-induced nephrotoxicity was assessed by histopathology and biomarkers. Cisplatin-mediated NAG changes and survival were determined in wild-type and Oct1/2(-/-) mice. Identification of OCT2 inhibitors was done in transfected 293Flp-In cells, and the NCI(60) cell line panel was used to assess contribution of OCT2 to cisplatin uptake in cancer cells. RESULTS: Classical biomarkers such as blood urea nitrogen and serum creatinine were not elevated until 72 hours after cisplatin administration and substantial kidney damage had occurred. Oct1/2(-/-) mice had 2.9-fold lower NAG by 4 hours (P < 0.0001) and 2.3-fold increased survival (P = 0.0097). Among 16 agents, cimetidine strongly inhibited uptake of tetraethylammonium bromide (P = 0.0006) and cisplatin (P < 0.0001), but did not have an influence on cisplatin uptake in SK-OV-3 cells, the cancer line with the highest OCT2 mRNA levels. In wild-type mice, cimetidine inhibited cisplatin-induced NAG changes (P = 0.016 versus cisplatin alone) to a degree similar to that seen in Oct1/2(-/-) mice receiving cisplatin (P = 0.91). Cumulative NAG activity of >0.4 absorbance units (AU) was associated with 21-fold increased odds for severe nephrotoxicity (P = 0.0017), which was linked with overall survival (hazard ratio, 8.1; 95% confidence interval, 2.1-31; P = 0.0078). CONCLUSIONS: Cimetidine is able to inhibit OCT2-mediated uptake of cisplatin in the kidney, and subsequently ameliorate nephrotoxicity likely with minimal effect on uptake in tumor cells.


Asunto(s)
Acetilglucosaminidasa/orina , Antineoplásicos/toxicidad , Cisplatino/toxicidad , Factor 1 de Transcripción de Unión a Octámeros/deficiencia , Proteínas de Transporte de Catión Orgánico/deficiencia , Animales , Antineoplásicos/metabolismo , Biomarcadores/análisis , Línea Celular Tumoral , Cimetidina/farmacología , Cisplatino/metabolismo , Inhibidores Enzimáticos/farmacología , Humanos , Riñón/efectos de los fármacos , Riñón/metabolismo , Masculino , Ratones , Ratones Noqueados , Transportador 2 de Cátion Orgánico , Reacción en Cadena de la Polimerasa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA