Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
J Vet Pharmacol Ther ; 40(6): 591-598, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28198024

RESUMEN

The ATP-binding cassette efflux transporter ABCG2 plays a key role in the mammary excretion of drugs and toxins in humans and animals. Aflatoxins (AF) are worldwide contaminants of food and feed commodities, while PCB 126 is a dioxin-like PCB which may contaminate milk and dairy products. Both compounds are known human carcinogens. The interactions between AF and bovine ABCG2 (bABCG2) as well as the effects of PCB 126 on its efflux activity have been investigated by means of the Hoechst H33342 transport assay in MDCKII cells stably expressing mammary bABCG2. Both AFB1 and its main milk metabolite AFM1 showed interaction with bABCG2 even at concentrations approaching the legal limits in feed and food commodities. Moreover, PCB 126 significantly enhanced bABCG2 functional activity. Specific inhibitors of either AhR (CH233191) or ABCG2 (Ko143) were able to reverse the PCB 126-induced increase in bABCG2 transport activity, showing the specific upregulation of the efflux protein by the AhR pathway. The incubation of PCB 126-pretreated cells with AFM1 was able to substantially reverse such effect, with still unknown mechanism(s). Overall, results from this study point to AFB1 and AFM1 as likely bABCG2 substrates. The PCB 126-dependent increased activity of the transporter could enhance the ABCG2-mediated excretion into dairy milk of chemicals (i.e., drugs and toxins) potentially harmful to neonates and consumers.


Asunto(s)
Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Aflatoxina B1/metabolismo , Glándulas Mamarias Animales/metabolismo , Bifenilos Policlorados/farmacología , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/efectos de los fármacos , Animales , Bencimidazoles/metabolismo , Bovinos , Perros , Femenino , Células de Riñón Canino Madin Darby/efectos de los fármacos , Células de Riñón Canino Madin Darby/metabolismo , Glándulas Mamarias Animales/efectos de los fármacos
2.
Eur J Histochem ; 55(1): e3, 2011 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-21556118

RESUMEN

The reduced folate carrier (Rfc1; Slc19a1) mediated transport of reduced folates and antifolate drugs such as methotrexate (MTX) play an essential role in physiological folate homeostasis and MTX cancer chemotherapy. As no systematic reports are as yet available correlating Rfc1 gene expression and protein levels in all tissues crucial for folate and antifolate uptake, storage or elimination, we investigated gene and protein expression of rat Rfc1 (rRfc1) in selected tissues. This included the generation of a specific anti-rRfc1 antibody. Rabbits were immunised with isolated rRfc1 peptides producing specific anti-rRfc1 antiserum targeted to the intracellular C-terminus of the carrier. Using RT-PCR analysis, high rRfc1 transcript levels were detected in colon, kidney, brain, thymus, and spleen. Moderate rRfc1 gene expression was observed in small intestine, liver, bone marrow, lung, and testes whereas transcript levels were negligible in heart, skeletal muscle or leukocytes. Immunohistochemical analyses revealed strong carrier expression in the apical membrane of tunica mucosa epithelial cells of small intestine and colon, in the brush-border membrane of choroid plexus epithelial cells or in endothelial cells of small vessels in brain and heart. Additionally, high rRfc1 protein levels were localized in the basolateral membrane of renal tubular epithelial cells, in the plasma membrane of periportal hepatocytes, and sertoli cells of the testes. Taken together, our results demonstrated that rRfc1 is expressed almost ubiquitously but to very different levels. The predominant tissue distribution supports the essential role of Rfc1 in physiological folate homeostasis. Moreover, our results may contribute to understand antifolate pharmacokinetics and selected organ toxicity associated with MTX chemotherapy.


Asunto(s)
Regulación de la Expresión Génica , Proteína Portadora de Folato Reducido/química , Animales , Células Cultivadas , Perros , Electroforesis en Gel de Poliacrilamida , Inmunohistoquímica , Riñón/química , Riñón/citología , Riñón/metabolismo , Masculino , Conejos , Ratas , Ratas Sprague-Dawley , Proteína Portadora de Folato Reducido/genética , Proteína Portadora de Folato Reducido/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Distribución Tisular
3.
Leukemia ; 23(6): 1087-97, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19212336

RESUMEN

Concurrent treatment with methotrexate (MTX) and antiepileptic drugs, such as phenobarbital (PB), reduces the efficacy of MTX chemotherapy in childhood acute lymphoblastic leukemia (ALL). This can result from defective Reduced folate carrier (Rfc1)-dependent cellular uptake of MTX. Indeed, we have shown that functional Rfc1 activity is significantly reduced by clinically relevant concentrations of the anticonvulsant drugs PB or carbamazepine in an adequate in vitro model. As PB is known to regulate carrier-associated transport by the nuclear receptor constitutive androstane receptor (CAR), we investigated the involvement of the CAR signaling cascade and the mode of PB-induced downregulation of Rfc1 activity. CAR activation by PB or the CAR agonist 1,4-bis[2-(3,5-dichloro- pyridyloxy)]-benzene resulted in translocation of Ca(2+)-dependent protein kinase Calpha (cPKCalpha) to the plasma membrane related to significantly elevated PKC activities. In contrast, subcellular localization of Ca(2+)-independent PKCdelta was only marginally altered. Studies on intracellular distribution of the Rfc1 protein indicated that PB-induced activation of cPKCalpha was associated with carrier internalization from the plasma membrane into the cytosol independent of the Rfc1 phosphorylation status. In conclusion, we identified for the first time the molecular mechanism of this clinically relevant drug resistance in patients with ALL concurrently receiving MTX chemotherapy and antiepileptic drugs.


Asunto(s)
Anticonvulsivantes/farmacología , Regulación hacia Abajo/efectos de los fármacos , Proteínas de Transporte de Membrana/metabolismo , Metotrexato/farmacología , Animales , Transporte Biológico , Línea Celular Tumoral , Receptor de Androstano Constitutivo , Antagonismo de Drogas , Antagonistas del Ácido Fólico/farmacología , Fenobarbital/farmacología , Proteína Quinasa C/metabolismo , Ratas , Receptores Citoplasmáticos y Nucleares/metabolismo , Proteína Portadora de Folato Reducido , Factores de Transcripción/metabolismo
4.
Hepatology ; 31(6): 1296-304, 2000 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10827155

RESUMEN

We have cloned two complementary DNAs (cDNAs), RL-Mtx-1 and RL-Mtx-2, corresponding to the bile acid- sensitive methotrexate carrier from rat liver by direct full-length rapid amplification of cDNA ends polymerase chain reaction (RACE-PCR) using degenerated primers that were deduced from published sequences of tumor cell methotrexate transporters. When expressed in Xenopus laevis oocytes and cosM6 cells, both clones mediate methotrexate and bumetanide transport. RL-Mtx-1 consists of 2,445 bp with an open reading frame of 1,536 bp. The corresponding protein with 512 amino acids has a molecular weight of 58 kd. RL-Mtx-2 (2,654 bp) differs by an additional insert of 203 bp. This insert is located in frame at position 1,196 of the RL-Mtx-1 and contains the typical splice junction sites at the 5' and 3' end, indicating that the RL-Mtx-2 messenger RNA (mRNA) is generated by alternative splicing. The insert contains a stop codon that shortens the RL-Mtx-2 protein to 330 amino acids (38 kd). Both cDNAs contain the binding site sequence for the dioxin/nuclear translocator responsive element (Ah/Arnt-receptor) in conjunction with a barbiturate recognition sequence (Barbie box). Preliminary results show that the Barbie box acts as a negative regulatory element. The two liver cDNA clones show homologies to the published sequences of folate and the reduced folate carriers, but no homology is found to the transport systems for organic anions like the Ntcp1, oatp1, OAT-K1, and OAT1. Expression of the mRNA for the methotrexate carrier is found in liver, kidney, heart, brain, spleen, lung, and skeletal muscle, but not in the testis as revealed by Northern blot analysis. The highest abundance of the mRNA is found in the kidney.


Asunto(s)
Transportadoras de Casetes de Unión a ATP , Ácidos y Sales Biliares/farmacología , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Clonación Molecular , Hígado/metabolismo , Proteínas de Transporte de Membrana , Proteínas de Neoplasias , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Secuencia de Aminoácidos/genética , Animales , Secuencia de Bases/genética , Bumetanida/metabolismo , Células COS/metabolismo , Células Cultivadas , ADN Complementario/genética , Femenino , Hígado/citología , Masculino , Metotrexato/metabolismo , Antígenos de Histocompatibilidad Menor , Datos de Secuencia Molecular , Oocitos/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Proteína Portadora de Folato Reducido , Distribución Tisular , Xenopus laevis
5.
Naunyn Schmiedebergs Arch Pharmacol ; 359(5): 411-9, 1999 May.
Artículo en Inglés | MEDLINE | ID: mdl-10498292

RESUMEN

The chemotherapeutic agent methotrexate is widely used in tumor therapy for different forms of leukemia and for the therapy of arthritis. Methotrexate is eliminated from systemic blood circulation by the liver and its transport into hepatocytes is therefore described in detail in this paper. Methotrexate uptake is energy- and sodium-dependent. The Km and the Vmax are 23 microM and 36 pmol/mg protein min, respectively. The apparent activation energy (E(app)) of methotrexate uptake (5 microM [3H]methotrexate) is 53.73 kJ/mol, which indicates an energy-dependent carrier-mediated process. Although methotrexate is a folate derivative, folate itself does not inhibit methotrexate uptake, whereas the reduced folates, dihydrofolate and tetrahydrofolate are weak uncompetitive inhibitors. In contrast, the bile acids taurocholate and cholate are effective competitive inhibitors of methotrexate uptake into hepatocytes. Further strong inhibitors are the loop diuretic bumetanide, the mycotoxin ochratoxin A and bromosulfophthalein. Because tumor patients develop drug resistance during methotrexate therapy, the uptake of methotrexate was tested in different hepatoma cell lines. In HepG2-cells and Reuber hepatoma Fao-cells the transport was non-existent or very small. However, the hepatocytoma fusion cell line HPCT-1E3, a hybrid cell line between primary rat hepatocytes and rat Reuber Fao-cells, shows an intermediate transport activity with a threefold increase of the methotrexate uptake. These results indicate the presence of a bile acid sensitive methotrexate carrier in hepatocytes which is absent in dedifferentiated hepatoma cells. The carrier differs from previously described transporters for the uptake of organic anions.


Asunto(s)
Ácidos y Sales Biliares/farmacología , Proteínas Portadoras/antagonistas & inhibidores , Hígado/metabolismo , Receptores de Superficie Celular , Animales , Tampones (Química) , Línea Celular , Membrana Celular/metabolismo , Células Cultivadas , Receptores de Folato Anclados a GPI , Ácido Fólico/análogos & derivados , Ácido Fólico/farmacología , Cinética , Hígado/efectos de los fármacos , Masculino , Metotrexato/metabolismo , Oligomicinas , Ratas , Ratas Wistar , Cloruro de Sodio , Sulfobromoftaleína/farmacología , Tetrahidrofolatos/farmacología , Tritio , Xenobióticos/farmacología
6.
Eur J Cell Biol ; 70(1): 54-60, 1996 May.
Artículo en Inglés | MEDLINE | ID: mdl-8738419

RESUMEN

Bile acids are taken up into liver parenchymal cells by active, carrier-mediated transport. This transport is lost during cell transformation in permanent growing liver tumor cell lines. In order to establish bile acid uptake in a permanent mammalian cell culture system, we transfected the cDNA from the cloned rat liver Na(+)-taurocholate cotransporting polypeptide (Ntcp) in Chinese hamster lung fibroblasts (V79 cells) and in a "hepatocyte-like" cell line HPCT-1F3 with three different gene transfer methods (calcium phosphate precipitation, lipofection, electroporation). A stable integration of the cDNA in both cell genomes was observed. However, in V79 fibroblasts, a permanent functional expression of taurocholate transport was not achieved. The sodium-dependent uptake of taurocholate was expressed permanently only in HPCT-1E3 cells, if the Ntcp was transfected by electroporation. In this cell line (HPCT-1E3-TC-6/2), substrate specificity, sodium- and energy dependence, as well as the kinetic parameters of the transfected single transporter were measured. The sodium-dependent taurocholate uptake was inhibited by addition of non-labeled bile acids, bumetanide, sulfobromophthalein and oligomycin. Pretreatment with 10 mM Na(+)-butyrate of this cell culture for 22 h stimulated taurocholate uptake twofold. Neither butyrate-stimulated cells nor unstimulated cells transport glycocholate or cholate. Besides taurocholate a fluorescence-labeled taurocholate derivative, NBD-taurocholate, was taken up by the HPCT-1E3-TC cells. In conclusion, the specific gene transfer with the electroporation technique in combination with the "right" cell line, HPCT-1E3, has been successful for the permanent and functional expression of the Ntcp. This allowed direct monitoring of the solitary sodium-dependent taurocholate transport system in a "liver cell-like" environment.


Asunto(s)
Línea Celular/citología , Fibroblastos/citología , Animales , Northern Blotting , Southern Blotting , Proteínas Portadoras/metabolismo , Técnica del Anticuerpo Fluorescente , Ratas , Sodio/farmacología , Ácido Taurocólico/metabolismo , Transfección
7.
Biochim Biophys Acta ; 1300(2): 114-8, 1996 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-8652636

RESUMEN

The loop diuretic bumetanide which inhibits hepatic bile acid uptake competitively according to its transport kinetics has been proposed to serve as a substrate of a multispecific bile acid transport system in liver parenchymal cells. However, when the in vitro transcripts of two cloned hepatic bile acid uptake carriers, the Ntcp (Na+/taurocholate cotransporting polypeptide) and the oatp (organic anion transporting polypeptide), was expressed for three days in Xenopus laevis oocytes [3H]bumetanide uptake was not increased although bile acid uptake was stimulated. The data presented show that bumetanide is taken up by a third organic anion transport system which is different from the cloned bile acid transporters.


Asunto(s)
Transportadoras de Casetes de Unión a ATP , Bumetanida/metabolismo , Proteínas Portadoras/metabolismo , Diuréticos/metabolismo , Hígado/metabolismo , Animales , Proteínas de Transporte de Anión , Transporte Biológico , Ácido Glicocólico/metabolismo , Transporte Iónico , Oocitos/metabolismo , ARN sin Sentido/metabolismo , ARN sin Sentido/farmacología , ARN Mensajero/metabolismo , Ratas , Sodio/farmacología , Xenopus laevis
8.
J Hepatol ; 24 Suppl 1: 42-6, 1996.
Artículo en Inglés | MEDLINE | ID: mdl-8926368

RESUMEN

Bumetanide is a weak organic acid which is transported into hepatocytes by a transport system that is related neither to the cloned sodium-dependent taurocholate cotransporting polypeptide Ntcp nor to the cloned organic anion transporting polypeptide oatp. Bumetanide is known to be transported in the kidney by a multispecific organic anion transporter which is the pAH-transporter from the proximal tubule cell. In the liver, bumetanide uptake competes with bile acid uptake, indicating a functionally related multispecific transporter for bile acids and drugs in hepatocytes. This multispecific bile acid transporter MBAT has not been cloned yet. When basolateral membranes were photoaffinity labeled with [3H]bumetanide, several bumetanide binding proteins were separated and identified after protein sequencing from two-dimensional electrophoresis gels.


Asunto(s)
Ácidos y Sales Biliares/metabolismo , Bumetanida/metabolismo , Proteínas Portadoras/metabolismo , Portadores de Fármacos/metabolismo , Hígado/metabolismo , Adenosina Trifosfato/análogos & derivados , Adenosina Trifosfato/metabolismo , Humanos , Masculino , Simportadores de Cloruro de Sodio-Potasio
9.
Biochem J ; 311 ( Pt 3): 975-9, 1995 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-7487959

RESUMEN

Recently two different bile-acid carriers for the hepatocellular sodium-dependent uptake of taurocholate have been described. The first transport system was isolated and characterized by functional expression cloning in Xenopus laevis oocytes. The corresponding cDNA clone, named Ntcp for Na+/taurocholate co-transporting polypeptide, codes for a protein of 362 amino acids and shows no similarity to previously known sequences. The transport function of this carrier system is well documented by expression in Xenopus laevis oocytes and by transient and stably transfected cell lines. In addition, several lines of evidence implied that the well-known xenobiotic-metabolizing enzyme microsomal epoxide hydrolase (mEH, EC 3.3.2.3) is also able to mediate sinusoidal uptake of taurocholate. Furthermore, it was claimed that the same enzyme also mediates the uptake of the conjugated bile acid into the smooth endoplasmic reticulum (ER). No direct proof of the transport function of mEH by its heterologous expression has yet been published. In the present work we used a stable transfected cell line that expressed high levels of heterologous mEH for uptake studies of various bile acids and the loop diuretic bumetanide. The uptake of the conjugated bile acid taurocholate, of the non-conjugated bile acid cholate and of the organic anion bumetanide was measured in the transfected as well as in the non-transfected parental cell line. These organic anions represent the main substrates of the known transport systems for organic anions in the rat liver. The results show that the microsomal epoxide hydrolase is unable to transport taurocholate, cholate or bumetanide. Furthermore, Western-blot analysis revealed the expression of mEH in hepatoma tumor cell lines, which show no transport activity for these organic anions. These results show that it is unlikely that mEH can mediate the transport of these substrates.


Asunto(s)
Bumetanida/farmacocinética , Ácidos Cólicos/farmacocinética , Epóxido Hidrolasas/metabolismo , Hígado/metabolismo , Ácido Taurocólico/farmacocinética , Animales , Transporte Biológico , Western Blotting , Línea Celular , Cricetinae , Hígado/enzimología , Masculino , Mesocricetus , Microsomas Hepáticos/enzimología , Ratas , Ratas Wistar , Transfección
10.
Eur J Pharmacol ; 208(1): 53-65, 1991 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-1936129

RESUMEN

To identify proteins involved in the hepatocellular uptake of loop diuretics, [3H]bumetanide was photoactivated by light flash in the presence of either intact isolated rat hepatocytes, rat liver basolateral plasma membranes or integral membrane proteins extracted from the basolateral plasma membranes. Proteins of 52-54, 48, 33, 27, 25 and 23 kDa in sodium dodecyl sulfate (SDS) gel electrophoresis were radiolabeled on intact hepatocytes. On liver basolateral plasma membranes a 50-52 kDa protein was the most intensely labeled protein. After separation into integral and associated membrane proteins by extraction with Triton X-114, radioactive labeling was only found in integral membrane proteins with a molecular weight of 50-52 kDa. Photoactivated bumetanide irreversibly inhibited the hepatocellular uptake of cholate, taurocholate but not of serine. Binding proteins for photoactivated bumetanide were absent on AS 30-D ascites hepatoma cells. Labeling of all proteins was sodium dependent in intact hepatocytes but was sodium independent in plasma membranes. Labeling was prevented by non-labeled bumetanide and by the loop diuretics piretanide and furosemide. Labeling protection was further achieved with organic anions such as bromosulfophthalein, rifampicin, probenecid and by the bile acids taurocholate, deoxycholate and dehydrocholate. The radiolabeled proteins did not belong to the bumetanide-sensitive NaCl/KCl co-transport system which apparently does not occur in intact isolated rat hepatocytes.


Asunto(s)
Bumetanida/farmacocinética , Hígado/metabolismo , Proteínas de la Membrana/metabolismo , Marcadores de Afinidad , Animales , Ácidos y Sales Biliares/metabolismo , Transporte Biológico Activo , Bumetanida/farmacología , Proteínas Portadoras/metabolismo , Membrana Celular/metabolismo , Luz , Hígado/citología , Masculino , Proteínas de la Membrana/química , Potasio/metabolismo , Ratas , Ratas Endogámicas , Serina/metabolismo , Sodio/metabolismo , Simportadores de Cloruro de Sodio-Potasio , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA