Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Drug Metab Dispos ; 45(1): 76-85, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27803021

RESUMEN

Inhibition of thiamine transporters has been proposed as a putative mechanism for the observation of Wernicke's encephalopathy and subsequent termination of clinical development of fedratinib, a Janus kinase inhibitor (JAKi). This study aimed to determine the potential for other JAKi to inhibit thiamine transport using human epithelial colorectal adenocarcinoma (Caco-2) and thiamine transporter (THTR) overexpressing cells and to better elucidate the structural basis for interacting with THTR. Only JAKi containing a 2,4-diaminopyrimidine were observed to inhibit thiamine transporters. Fedratinib inhibited thiamine uptake into Caco-2 cells (IC50 = 0.940 µM) and THTR-2 (IC50 = 1.36 µM) and, to a lesser extent, THTR-1 (IC50 = 7.10 µM) overexpressing cells. Two other JAKi containing this moiety, AZD1480 and cerdulatinib, were weaker inhibitors of the thiamine transporters. Other JAKi-including monoaminopyrimidines, such as momelotinib, and nonaminopyrimidines, such as filgotinib-did not have any inhibitory effects on thiamine transport. A pharmacophore model derived from the minimized structure of thiamine suggests that 2,4-diaminopyrimidine-containing compounds can adopt a conformation matching several key features of thiamine. Further studies with drugs containing a 2,4-diaminopyrimidine resulted in the discovery that the antibiotic trimethoprim also potently inhibits thiamine uptake mediated by THTR-1 (IC50 = 6.84 µM) and THTR-2 (IC50 = 5.56 µM). Fedratinib and trimethoprim were also found to be substrates for THTR, a finding with important implications for their disposition in the body. In summary, our results show that not all JAKi have the potential to inhibit thiamine transport and further establish the interaction of these transporters with xenobiotics.


Asunto(s)
Inhibidores de las Cinasas Janus/farmacología , Proteínas de Transporte de Membrana/metabolismo , Pirimidinas/química , Pirrolidinas/farmacología , Sulfonamidas/farmacología , Trimetoprim/farmacología , Células CACO-2 , Interacciones Farmacológicas , Células HEK293 , Humanos , Inhibidores de las Cinasas Janus/química , Proteínas de Transporte de Membrana/genética , Estructura Molecular , Pirrolidinas/química , Especificidad por Sustrato , Sulfonamidas/química , Tiamina/metabolismo , Trimetoprim/química
2.
PLoS Genet ; 9(4): e1003464, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23637631

RESUMEN

Gene fusions, like BCR/ABL1 in chronic myelogenous leukemia, have long been recognized in hematologic and mesenchymal malignancies. The recent finding of gene fusions in prostate and lung cancers has motivated the search for pathogenic gene fusions in other malignancies. Here, we developed a "breakpoint analysis" pipeline to discover candidate gene fusions by tell-tale transcript level or genomic DNA copy number transitions occurring within genes. Mining data from 974 diverse cancer samples, we identified 198 candidate fusions involving annotated cancer genes. From these, we validated and further characterized novel gene fusions involving ROS1 tyrosine kinase in angiosarcoma (CEP85L/ROS1), SLC1A2 glutamate transporter in colon cancer (APIP/SLC1A2), RAF1 kinase in pancreatic cancer (ATG7/RAF1) and anaplastic astrocytoma (BCL6/RAF1), EWSR1 in melanoma (EWSR1/CREM), CDK6 kinase in T-cell acute lymphoblastic leukemia (FAM133B/CDK6), and CLTC in breast cancer (CLTC/VMP1). Notably, while these fusions involved known cancer genes, all occurred with novel fusion partners and in previously unreported cancer types. Moreover, several constituted druggable targets (including kinases), with therapeutic implications for their respective malignancies. Lastly, breakpoint analysis identified new cell line models for known rearrangements, including EGFRvIII and FIP1L1/PDGFRA. Taken together, we provide a robust approach for gene fusion discovery, and our results highlight a more widespread role of fusion genes in cancer pathogenesis.


Asunto(s)
Fusión Génica , Proteínas Tirosina Quinasas , Genómica , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Proteínas de Fusión Oncogénica/genética , Proteínas Tirosina Quinasas/genética , Proteínas Proto-Oncogénicas/genética
3.
Mol Pharm ; 12(12): 4301-10, 2015 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-26528626

RESUMEN

The biguanide metformin is widely used as first-line therapy for the treatment of type 2 diabetes. Predominately a cation at physiological pH's, metformin is transported by membrane transporters, which play major roles in its absorption and disposition. Recently, our laboratory demonstrated that organic cation transporter 1, OCT1, the major hepatic uptake transporter for metformin, was also the primary hepatic uptake transporter for thiamine, vitamin B1. In this study, we tested the reverse, i.e., that metformin is a substrate of thiamine transporters (THTR-1, SLC19A2, and THTR-2, SLC19A3). Our study demonstrated that human THTR-2 (hTHTR-2), SLC19A3, which is highly expressed in the small intestine, but not hTHTR-1, transports metformin (Km = 1.15 ± 0.2 mM) and other cationic compounds (MPP(+) and famotidine). The uptake mechanism for hTHTR-2 was pH and electrochemical gradient sensitive. Furthermore, metformin as well as other drugs including phenformin, chloroquine, verapamil, famotidine, and amprolium inhibited hTHTR-2 mediated uptake of both thiamine and metformin. Species differences in the substrate specificity of THTR-2 between human and mouse orthologues were observed. Taken together, our data suggest that hTHTR-2 may play a role in the intestinal absorption and tissue distribution of metformin and other organic cations and that the transporter may be a target for drug-drug and drug-nutrient interactions.


Asunto(s)
Interacciones Farmacológicas/fisiología , Proteínas de Transporte de Membrana/análisis , Metformina/metabolismo , Tiamina/metabolismo , Animales , Transporte Biológico/fisiología , Línea Celular , Células HEK293 , Humanos , Absorción Intestinal/efectos de los fármacos , Absorción Intestinal/fisiología , Intestino Delgado/efectos de los fármacos , Intestino Delgado/metabolismo , Ratones , Especificidad por Sustrato/fisiología
4.
Exp Cell Res ; 318(6): 716-22, 2012 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-22309779

RESUMEN

Transforming Growth Factor Beta (TGF-ß) is involved in regulating many biological processes and disease states. Cells secrete cytokine as a latent complex that must be activated for it to exert its biological functions. We previously discovered that the epithelial-restricted integrin α(v)ß(6) activates TGF-ß and that this process is important in a number of in vivo models of disease. Here, we show that agonists of G-protein coupled receptors (Sphingosine-1-Phosphate and Lysophosphatidic Acid) which are ligated under conditions of epithelial injury directly stimulate primary airway epithelial cells to activate latent TGF-ß through a pathway that involves Rho Kinase, non-muscle myosin, the α(v)ß(6) integrin, and the generation of mechanical tension. Interestingly, lung epithelial cells appear to exert force on latent TGF-ß using sub-cortical actin/myosin rather than the stress fibers utilized by fibroblasts and other traditionally "contractile" cells. These findings extend recent evidence suggesting TGF-ß can be activated by integrin-mediated mechanical force and suggest that this mechanism is important for an integrin (α(v)ß(6)) and a cell type (epithelial cells) that have important roles in biologically relevant TGF-ß activation in vivo.


Asunto(s)
Actinas/metabolismo , Antígenos de Neoplasias/metabolismo , Células Epiteliales/metabolismo , Integrinas/metabolismo , Miosinas/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Células Cultivadas , Células Epiteliales/citología , Humanos , Lisofosfolípidos/metabolismo , Transducción de Señal , Esfingosina/análogos & derivados , Esfingosina/metabolismo
6.
Clin Transl Sci ; 12(4): 388-399, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30982223

RESUMEN

Understanding transporter-mediated drug-drug interactions is an integral part of risk assessment in drug development. Recent studies support the use of hexadecanedioate (HDA), tetradecanedioate (TDA), coproporphyrin (CP)-I, and CP-III as clinical biomarkers for evaluating organic anion-transporting polypeptide (OATP)1B1 (SLCO1B1) inhibition. The current study investigated the effect of OATP1B1 genotype c.521T>C (OATP1B1-Val174Ala) on the extent of interaction between cyclosporin A (CsA) and pravastatin, and associated endogenous biomarkers of the transporter (HDA, TDA, CP-I, and CP-III), in 20 healthy volunteers. The results show that the levels of each clinical biomarker and pravastatin were significantly increased in plasma samples of the volunteers following administration of pravastatin plus CsA compared with pravastatin plus placebo. The overall fold change in the area under the concentration-time curve (AUC) and maximum plasma concentration (Cmax ) was similar among the four biomarkers (1.8-2.5-fold, paired t-test P value < 0.05) in individuals who were homozygotes or heterozygotes of the major allele, c.521T. However, the fold change in AUC and Cmax for HDA and TDA was significantly abolished in the subjects who were c.521-CC, whereas the respective fold change in AUC and Cmax for pravastatin and CP-I and CP-III were slightly weaker in individuals who were c.521-CC compared with c.521-TT/TC genotypes. In addition, this study provides the first evidence that SLCO1B1 c.521T>C genotype is significantly associated with CP-I but not CP-III levels. Overall, these results suggest that OATP1B1 genotype can modulate the effects of CsA on biomarker levels; the extent of modulation differs among the biomarkers.


Asunto(s)
Interacciones Farmacológicas , Voluntarios Sanos , Transportador 1 de Anión Orgánico Específico del Hígado/genética , Polimorfismo de Nucleótido Simple/genética , Alelos , Área Bajo la Curva , Biomarcadores/sangre , Coproporfirinas/sangre , Ciclosporina/administración & dosificación , Femenino , Heterocigoto , Humanos , Masculino , Pravastatina/sangre , Pravastatina/farmacocinética
7.
Nat Med ; 19(12): 1617-24, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24216753

RESUMEN

Myofibroblasts are the major source of extracellular matrix components that accumulate during tissue fibrosis, and hepatic stellate cells (HSCs) are believed to be the major source of myofibroblasts in the liver. To date, robust systems to genetically manipulate these cells have not been developed. We report that Cre under control of the promoter of Pdgfrb (Pdgfrb-Cre) inactivates loxP-flanked genes in mouse HSCs with high efficiency. We used this system to delete the gene encoding α(v) integrin subunit because various α(v)-containing integrins have been suggested as central mediators of fibrosis in multiple organs. Such depletion protected mice from carbon tetrachloride-induced hepatic fibrosis, whereas global loss of ß3, ß5 or ß6 integrins or conditional loss of ß8 integrins in HSCs did not. We also found that Pdgfrb-Cre effectively targeted myofibroblasts in multiple organs, and depletion of the α(v) integrin subunit using this system was protective in other models of organ fibrosis, including pulmonary and renal fibrosis. Pharmacological blockade of α(v)-containing integrins by a small molecule (CWHM 12) attenuated both liver and lung fibrosis, including in a therapeutic manner. These data identify a core pathway that regulates fibrosis and suggest that pharmacological targeting of all α(v) integrins may have clinical utility in the treatment of patients with a broad range of fibrotic diseases.


Asunto(s)
Integrina alfaV/metabolismo , Enfermedades Renales/genética , Riñón/patología , Cirrosis Hepática/genética , Fibrosis Pulmonar/genética , Animales , Células Cultivadas , Evaluación Preclínica de Medicamentos , Femenino , Fibrosis/genética , Marcación de Gen , Integrina alfaV/genética , Riñón/metabolismo , Enfermedades Renales/patología , Cirrosis Hepática/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Miofibroblastos/metabolismo , Miofibroblastos/patología , Fibrosis Pulmonar/patología , Transducción de Señal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA