Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 367
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 112(21): 6601-6, 2015 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-25964343

RESUMEN

Despite technological advances in metabolomics, large parts of the human metabolome are still unexplored. In an untargeted metabolomics screen aiming to identify substrates of the orphan transporter ATP-binding cassette subfamily C member 5 (ABCC5), we identified a class of mammalian metabolites, N-lactoyl-amino acids. Using parallel protein fractionation in conjunction with shotgun proteomics on fractions containing N-lactoyl-Phe-forming activity, we unexpectedly found that a protease, cytosolic nonspecific dipeptidase 2 (CNDP2), catalyzes their formation. N-lactoyl-amino acids are ubiquitous pseudodipeptides of lactic acid and amino acids that are rapidly formed by reverse proteolysis, a process previously considered to be negligible in vivo. The plasma levels of these metabolites strongly correlate with plasma levels of lactate and amino acid, as shown by increased levels after physical exercise and in patients with phenylketonuria who suffer from elevated Phe levels. Our approach to identify unknown metabolites and their biosynthesis has general applicability in the further exploration of the human metabolome.


Asunto(s)
Aminoácidos/metabolismo , Dipeptidasas/metabolismo , Lactatos/metabolismo , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Aminoácidos/sangre , Citosol/metabolismo , Ejercicio Físico/fisiología , Células HEK293 , Humanos , Lactatos/sangre , Metaboloma , Proteolisis
2.
Proc Natl Acad Sci U S A ; 110(50): 20206-11, 2013 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-24277820

RESUMEN

Pseudoxanthoma elasticum (PXE) is an autosomal recessive disease characterized by progressive ectopic mineralization of the skin, eyes, and arteries, for which no effective treatment exists. PXE is caused by inactivating mutations in the gene encoding ATP-binding cassette sub-family C member 6 (ABCC6), an ATP-dependent efflux transporter present mainly in the liver. Abcc6(-/-) mice have been instrumental in demonstrating that PXE is a metabolic disease caused by the absence of an unknown factor in the circulation, the presence of which depends on ABCC6 in the liver. Why absence of this factor results in PXE has remained a mystery. Here we report that medium from HEK293 cells overexpressing either human or rat ABCC6 potently inhibits mineralization in vitro, whereas medium from HEK293 control cells does not. Untargeted metabolomics revealed that cells expressing ABCC6 excrete large amounts of nucleoside triphosphates, even though ABCC6 itself does not transport nucleoside triphosphates. Extracellularly, ectonucleotidases hydrolyze the excreted nucleoside triphosphates to nucleoside monophosphates and inorganic pyrophosphate (PPi), a strong inhibitor of mineralization that plays a pivotal role in several mineralization disorders similar to PXE. The in vivo relevance of our data are demonstrated in Abcc6(-/-) mice, which had plasma PPi levels <40% of those found in WT mice. This study provides insight into how ABCC6 affects PXE. Our data indicate that the factor that normally prevents PXE is PPi, which is provided to the circulation in the form of nucleoside triphosphates via an as-yet unidentified but ABCC6-dependent mechanism.


Asunto(s)
Difosfatos/sangre , Enfermedades Metabólicas/genética , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Seudoxantoma Elástico/genética , Animales , Cartilla de ADN/genética , ADN Complementario/genética , Fosfatos de Dinucleósidos/metabolismo , Células HEK293 , Humanos , Enfermedades Metabólicas/metabolismo , Enfermedades Metabólicas/patología , Metabolómica , Ratones , Ratones Noqueados , Mutagénesis Sitio-Dirigida , Mutación/genética , Seudoxantoma Elástico/metabolismo , Seudoxantoma Elástico/patología , Ratas
3.
Nat Med ; 3(11): 1275-9, 1997 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-9359705

RESUMEN

The multidrug resistance-associated protein (MRP) mediates the cellular excretion of many drugs, glutathione S-conjugates (GS-X) of lipophilic xenobiotics and endogenous cysteinyl leukotrienes. Increased MRP levels in tumor cells can cause multidrug resistance (MDR) by decreasing the intracellular drug concentration. The physiological role or roles of MRP remain ill-defined, however. We have generated MRP-deficient mice by using embryonic stem cell technology. Mice homozygous for the mrp mutant allele, mrp-/-, are viable and fertile, but their response to an inflammatory stimulus is impaired. We attribute this defect to a decreased secretion of leukotriene C4 (LTC4) from leukotriene-synthesizing cells. Moreover, the mrp-/- mice are hypersensitive to the anticancer drug etoposide. The phenotype of mrp-/- mice is consistent with a role for MRP as the main LTC4-exporter in leukotriene-synthesizing cells, and as an important drug exporter in drug-sensitive cells. Our results suggest that this ubiquitous GS-X pump is dispensable in mice, making treatment of MDR with MRP-specific reversal agents potentially feasible.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/fisiología , Antineoplásicos/uso terapéutico , Resistencia a Múltiples Medicamentos/fisiología , Inflamación/inmunología , Proteínas de Neoplasias/fisiología , Neoplasias Experimentales/tratamiento farmacológico , Transportadoras de Casetes de Unión a ATP/genética , Adulto , Animales , Antineoplásicos/efectos adversos , Antineoplásicos/farmacocinética , Antineoplásicos Fitogénicos/efectos adversos , Antineoplásicos Fitogénicos/farmacocinética , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Proteínas Portadoras/fisiología , Resistencia a Antineoplásicos , Etopósido/efectos adversos , Etopósido/farmacocinética , Humanos , Inflamación/inducido químicamente , Leucotrieno C4/metabolismo , Proteínas de Transporte de Membrana , Ratones , Ratones Noqueados , Proteínas Asociadas a Resistencia a Múltiples Medicamentos , Neoplasias Experimentales/inmunología , Células Tumorales Cultivadas
4.
J Exp Med ; 188(5): 797-808, 1998 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-9730882

RESUMEN

The multidrug resistance protein 1 (MRP1) gene encodes a transporter protein that helps to protect cells against xenobiotics. Elevated levels of MRP1 in tumor cells can result in active extrusion of a wide range of (anticancer) drugs with different cellular targets, a phenomenon called multidrug resistance (MDR). To explore the protective function of the mouse mrp1 protein during drug treatment, we investigated the toxicity caused by the anticancer drug etoposide-phosphate (ETOPOPHOS) in mice lacking the mrp1 gene (mrp1(-/-) mice). We show here that the lack of mrp1 protein results in increased etoposide-induced damage to the mucosa of the oropharyngeal cavity and to the seminiferous tubules of the testis. The high concentrations of mrp1 that we find in the basal layers of the oropharyngeal mucosa and in the basal membrane of the Sertoli cells in the testis apparently protect wild-type mice against this tissue damage. We also find drug-induced polyuria in mrp1(-/-) mice, which correlates with the presence of mrp1 protein in the urinary collecting tubules, the major site of kidney water reabsorption. Our results indicate that specific inhibitors of MRP1 used to reverse MDR, in combination with carcinostatic drugs transported by MRP1, might lead to drug-induced mucositis, (temporary) infertility, and diabetes insipidus.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/fisiología , Antineoplásicos/efectos adversos , Etopósido/análogos & derivados , Mucosa Bucal/patología , Compuestos Organofosforados/efectos adversos , Orofaringe/patología , Túbulos Seminíferos/patología , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/antagonistas & inhibidores , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Animales , Etopósido/efectos adversos , Riñón/efectos de los fármacos , Riñón/patología , Masculino , Ratones , Ratones Endogámicos , Ratones Noqueados , Mucosa Bucal/efectos de los fármacos , Orofaringe/efectos de los fármacos , Túbulos Seminíferos/efectos de los fármacos , Estomatitis/inducido químicamente , Estomatitis/patología , Testículo/efectos de los fármacos , Testículo/patología , Lengua/efectos de los fármacos , Lengua/patología
5.
Perioper Med (Lond) ; 9(1): 39, 2020 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-33292504

RESUMEN

BACKGROUND: Increasing numbers of patients receiving oral anticoagulants are undergoing elective surgery. Low molecular weight heparin (LMWH) is frequently applied as bridging therapy during perioperative interruption of anticoagulation. The aim of this study was to explore the postoperative bleeding risk of patients receiving surgery under bridging anticoagulation. METHODS: We performed a monocentric retrospective two-arm matched cohort study. Patients that received perioperative bridging anticoagulation were compared to a matched control group with identical surgical procedure, age, and sex. Emergency and vascular operations were excluded. The primary endpoint was the incidence of major postoperative bleeding. Secondary endpoints were minor postoperative bleeding, thromboembolic events, length of stay, and in-hospital mortality. Multivariate analysis explored risk factors of major postoperative bleeding. RESULTS: A total of 263 patients in each study arm were analyzed. The patient cohort included the entire field of general and visceral surgery including a large proportion of major oncological resections. Bridging anticoagulation increased the postoperative incidence of major bleeding events (8% vs. 1%; p < 0.001) as well as minor bleeding events (14% vs. 5%; p < 0.001). Thromboembolic events were equally rare in both groups (1% vs. 2%; p = 0.45). No effect on mortality was observed (1.5% vs. 1.9%). Independent risk factors of major postoperative bleeding were full-therapeutic dose of LMWH, renal insufficiency, and the procedure-specific bleeding risk. CONCLUSION: Perioperative bridging anticoagulation, especially full-therapeutic dose LMWH, markedly increases the risk of postoperative bleeding complications in general and visceral surgery. Surgeons should carefully consider the practice of routine bridging.

6.
J Cell Biol ; 61(2): 383-97, 1974 May.
Artículo en Inglés | MEDLINE | ID: mdl-4208072

RESUMEN

The effect of growth of Tetrahymena pyriformis in ethidium bromide (EthBr) on the structure and synthesis of mitochondrial DNA (mtDNA) has been investigated. During the first 5 h of growth in EthBr, mtDNA synthesis is inhibited 95% or more. After 10-15 h, this block is partially released and large numbers of replicating molecules accumulate, indicating that inhibition by EthBr primarily affects the rate of chain growth and not the initiation of new rounds of replication. The accumulated molecules sediment more rapidly than normal Tetrahymena mtDNA and do not contain enough single-strand regions to distinguish them from normal Tetrahymena mtDNA when banded in buoyant CsCl or NaI gradients. Electron microscopy shows that the predominant species in this rapidly sedimenting DNA is a linear molecule containing one symmetrical double-stranded replication loop of varying size located at its center. No degradation of mtDNA from cells grown in EthBr was detected in alkaline velocity gradients.


Asunto(s)
ADN/biosíntesis , Etidio/farmacología , Mitocondrias/metabolismo , Tetrahymena pyriformis/metabolismo , Animales , Bromuros , Fraccionamiento Celular , Centrifugación por Gradiente de Densidad , Cromatografía DEAE-Celulosa , Medios de Cultivo , ADN/análisis , Replicación del ADN/efectos de los fármacos , Microscopía Electrónica , Modelos Químicos , Radioisótopos de Fósforo , Tetrahymena pyriformis/análisis , Tetrahymena pyriformis/crecimiento & desarrollo , Timidina/metabolismo , Factores de Tiempo , Tritio
7.
J Cell Biol ; 76(2): 293-309, 1978 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-10605439

RESUMEN

We have used restriction endonucleases PstI, EcoRI, HapII, HhaI, and S1 nuclease to demonstrate the presence of a large complex component, the maxi-circle, in addition to the major mini-circle component in kinetoplast DNA (kDNA) networks of Trypanosoma brucei (East African Trypanosomiasis Research Organization [EATRO] 427). Endonuclease PstI and S1 nuclease cut the maxi-circle at a single site, allowing its isolation in a linear form with a mol wt of 12.2 x 10(6), determined by electron microscopy. The other enzymes give multiple maxi-circle fragments, whose added mol wt is 12-13 x 10(6), determined by gel electrophoresis. The maxi-circle in another T. brucei isolate (EATRO 1125) yields similar fragments but appears to contain a deletion of about 0.7 x 10(6) daltons. Electron microscopy of kDNA shows the presence of DNA considerably longer than the mini-circle contour length (0.3 micron) either in the network or as loops extending from the edge. This long DNA never exceeds the maxi-circle length (6.3 microns) and is completely removed by digestion with endonuclease PstI. 5-10% of the networks are doublets with up to 40 loops of DNA clustered between the two halves of the mini-circle network and probably represent a division stage of the kDNA. Digestion with PstI selectively removes these loops without markedly altering the mini-circle network. We conclude that the long DNA in both single and double networks represents maxi-circles and that long tandemly repeated oligomers of mini-circles are (virtually) absent. kDNA from Trypanosoma equiperdum, a trypanosome species incapable of synthesizing a fully functional mitochondrion, contains single and double networks of dimensions similar to those from T. brucei but without any DNA longer than mini-circle contour length. We conclude that the maxi-circle of trypanosomes is the genetic equivalent of the mitochondrial DNA (mtDNA) of other organisms.


Asunto(s)
ADN de Cinetoplasto/química , ADN de Cinetoplasto/ultraestructura , Trypanosoma brucei brucei/genética , Animales , Enzimas de Restricción del ADN , ADN de Cinetoplasto/aislamiento & purificación , Microscopía Electrónica , Ratas , Mapeo Restrictivo , Trypanosoma brucei brucei/aislamiento & purificación , Tripanosomiasis Africana/sangre
8.
J Cell Biol ; 132(4): 701-16, 1996 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-8647899

RESUMEN

We have generated mice transgenic for a human multidrug resistance (MDR)3 mini-gene driven by a hamster vimentin promoter. The MDR3 gene encodes a P-Glycoprotein that resembles the mouse multidrug resistance 2 P-Glycoprotein shown to be involved in the translocation of the phospholipid phosphatidylcholine through the hepatocyte canalicular membrane (Smit et al., 1993. Cell. 75:451-462). The vimentin promoter drives expression of the MDR3 transgene in mesenchymal tissues and in the eye lens. We show here that the presence of human multidrug resistance 3 P-Glycoprotein in the lens results in a severe lenticular pathology. Lens structural abnormalities initiate at a late embryonic stage and increase during postnatal lens development. Differentiation of the primary fibers is affected, and the terminal differentiation of the lens epithelium into secondary fibers is also perturbed. The ultrastructural alterations, particularly of the lens plasma membranes, resemble those identified in congenital mouse osmotic cataract.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/biosíntesis , Subfamilia B de Transportador de Casetes de Unión a ATP , Transportadoras de Casetes de Unión a ATP/biosíntesis , Catarata/etiología , Ojo/patología , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/genética , Animales , Animales Recién Nacidos , Catarata/metabolismo , Catarata/patología , Resistencia a Múltiples Medicamentos , Ojo/embriología , Ojo/metabolismo , Anomalías del Ojo/embriología , Técnica de Fractura por Congelación , Humanos , Ratones , Ratones Transgénicos , Microscopía Electrónica
9.
Science ; 235(4789): 658-67, 1987 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-3544215

RESUMEN

Programmed gene rearrangements are used in nature to to alter gene copy number (gene amplification and deletion), to create diversity by reassorting gene segments (as in the formation of mammalian immunoglobulin genes), or to control the expression of a set of genes that code for the same function (such as surface antigens). Two major mechanisms for expression control are DNA inversion and DNA transposition. In DNA inversion a DNA segment flips around and is rejoined by site-specific recombination, disconnecting or connecting a gene to sequences required for its expression. In DNA transposition a gene moves into an expression site where it displaces its predecessor by gene conversion. Gene rearrangements altering gene expression have mainly been found in some unicellular organisms. They allow a fraction of the organisms to preadapt to sudden changes in environment, that is, to alter properties such as surface antigens in the absence of an inducing stimulus. The antigenic variation that helps the causative agents of African trypanosomiasis, gonorrhea, and relapsing fever to elude host defense is controlled in this way.


Asunto(s)
Amplificación de Genes , Regulación de la Expresión Génica , Recombinación Genética , Animales , Antígenos/genética , Linfocitos B/fisiología , Borrelia/inmunología , Deleción Cromosómica , Glicoproteínas/genética , Humanos , Inmunoglobulinas/genética , Factor de Apareamiento , Neisseria/inmunología , Péptidos/genética , Saccharomyces cerevisiae/genética , Linfocitos T/fisiología , Trypanosoma/genética , Glicoproteínas Variantes de Superficie de Trypanosoma , Zea mays/genética
10.
Science ; 271(5252): 1126-8, 1996 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-8599091

RESUMEN

The human Dubin-Johnson syndrome and its animal model, the TR(-) rat, are characterized by a chronic conjugated hyperbilirubinemia. TR(-) rats are defective in the canalicular multispecific organic anion transporter (cMOAT), which mediates hepatobiliary excretion of numerous organic anions. The complementary DNA for rat cmoat, a homolog of the human multidrug resistance gene (hMRP1), was isolated and shown to be expressed in the canalicular membrane of hepatocytes. In the TR(-) rat, a single-nucleotide deletion in this gene resulted in a reduced messenger RNA level and absence of the protein. It is likely that this mutation accounts for the TR(-) phenotype.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Proteínas Portadoras/genética , Hiperbilirrubinemia Hereditaria/genética , Hígado/química , Transportadoras de Casetes de Unión a ATP/análisis , Transportadoras de Casetes de Unión a ATP/química , Secuencia de Aminoácidos , Animales , Proteínas de Transporte de Anión , Secuencia de Bases , Proteínas Portadoras/análisis , Proteínas Portadoras/química , Membrana Celular/química , ADN Complementario/genética , Mutación del Sistema de Lectura , Humanos , Hígado/citología , Datos de Secuencia Molecular , Peso Molecular , Proteínas Asociadas a Resistencia a Múltiples Medicamentos , Fenotipo , Ratas , Ratas Wistar , Alineación de Secuencia , Eliminación de Secuencia
11.
Trends Genet ; 13(6): 217-22, 1997 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-9196326

RESUMEN

Mammalian P-glycoproteins are plasma membrane proteins belonging to the superfamily of ATP-binding cassette transporters. They were discovered as drug pumps in multidrug-resistant cancer cells, but are also present in many normal tissues. Genetic approaches have helped to dissect the physiological functions and mode of action of P-glycoproteins. Disruption of both genes for the drug-transporting P-glycoproteins in mice has no effect on the normal sheltered life of these mice, but renders them hypersensitive to many drugs. P-glycoprotein appears to be especially important in protecting the brain and in limiting uptake of hydrophobic drugs from the gut. Recent experiments with polarized cells support the idea that drug-transporting P-glycoproteins act by flipping drugs from the inner to the outer leaflet of the plasma membrane.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/fisiología , Resistencia a Medicamentos/fisiología , Mamíferos/genética , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/química , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/clasificación , Animales , Sitios de Unión , Transporte Biológico , Canales de Cloruro/metabolismo , Ratones , Ratones Noqueados/genética , Preparaciones Farmacéuticas/metabolismo , Especificidad por Sustrato
12.
J Clin Invest ; 96(4): 1698-705, 1995 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-7560060

RESUMEN

We have previously shown that absence of the mouse mdr1a (also called mdr3) P-glycoprotein in mdr1a (-/-) "knockout" mice has a profound effect on the tissue distribution and elimination of vinblastine and ivermectin, and hence on the toxicity of these compounds. We show here that the mouse mdr1a and the human MDR1 P-glycoprotein actively transport ivermectin, dexamethasone, digoxin, and cyclosporin A and, to a lesser extent, morphine across a polarized kidney epithelial cell layer in vitro. Injection of these radio-labeled drugs in mdr1a (-/-) and wild-type mice resulted in markedly (20- to 50-fold) higher levels of radioactivity in mdr1a (-/-) brain for digoxin and cyclosporin A, with more moderate effects for dexamethasone (2- to 3-fold) and morphine (1.7-fold). Digoxin and cyclosporin A were also more slowly eliminated from mdr1a (-/-) mice. Our findings show that P-glycoprotein can be a major determinant for the pharmacology of several medically important drugs other than anti-cancer agents, especially in the blood-brain barrier. These results may explain a range of pharmacological interactions observed between various drugs in patients.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/fisiología , Cardiotónicos/farmacocinética , Ciclosporina/farmacocinética , Dexametasona/farmacocinética , Digoxina/farmacocinética , Glucocorticoides/farmacocinética , Inmunosupresores/farmacocinética , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/análisis , Animales , Transporte Biológico , Humanos , Masculino , Ratones , Morfina/farmacocinética , Porcinos , Distribución Tisular
13.
J Clin Invest ; 79(2): 344-50, 1987 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-3805272

RESUMEN

Thrombin, collagen, and Ca2+-ionophore A23187 aggregate platelets in the presence of inhibitors of the first (ADP-mediated) and second (cyclooxygenase-dependent) pathway of platelet activation. This aggregation, via a third pathway, was hypothesized to be mediated by the alkoxyether lipid platelet-activating factor (PAF). We recently demonstrated virtual absence of plasmalogen-type alkoxyether lipids and deficiency in key enzymes of their biosynthesis in Zellweger patients. We hypothesized that PAF synthesis might also be impaired. We report two Zellweger patients with an undetectable A23187-induced PAF synthesis of leukocytes (patients, less than 3 pmol PAF/10(8) granulocytes (PMN); four age-matched controls, 249-2,757 pmol PAF/10(8) PMN; five adult controls, 291-5,433 pmol PAF/10(8) PMN). In a third patient, residual PAF synthesis was detected. However in all patients the thrombin-induced third mechanism of platelet aggregation was present. We therefore conclude that PAF may not be the mediator of the third pathway.


Asunto(s)
Anomalías Múltiples/sangre , Leucocitos/metabolismo , Factor de Activación Plaquetaria/fisiología , Encéfalo/anomalías , Femenino , Humanos , Técnicas In Vitro , Lactante , Recién Nacido , Riñón/anomalías , Hígado/anomalías , Neutrófilos/metabolismo , Factor de Activación Plaquetaria/biosíntesis , Factor de Activación Plaquetaria/aislamiento & purificación , Agregación Plaquetaria , Trombina/fisiología
14.
J Clin Invest ; 100(10): 2430-6, 1997 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-9366556

RESUMEN

Mice lacking mdr1-type P-glycoproteins (mdr1a/1b [-/-] mice) display large changes in the pharmacokinetics of digoxin and other drugs. Using the kinetics of digoxin in mdr1a/1b (-/-) mice as a model representing a complete block of P-glycoprotein activity, we investigated the activity and specificity of the reversal agent SDZ PSC833 in inhibiting mdr1-type P-glycoproteins in vivo. Oral PSC833 was coadministered with intravenous [3H]digoxin to wild-type and mdr1a/1b (-/-) mice. The direct excretion of [3H]digoxin mediated by P-glycoprotein in the intestinal mucosa of wild-type mice was abolished by administration of PSC833. Hepatobiliary excretion of [3H]digoxin was markedly decreased in both wild-type and mdr1a/1b (-/-) mice by PSC833, the latter effect indicating that in vivo, PSC833 inhibits not only mdr1-type P-glycoproteins, but also other drug transporters. Upon coadministration of PSC833, brain levels of [3H]digoxin in wild-type mice showed a large increase, approaching (but not equaling) the levels found in brains of PSC833-treated mdr1a/1b (-/-) mice. Thus, orally administered PSC833 can inhibit blood-brain barrier P-glycoprotein extensively, and intestinal P-glycoprotein completely. These profound pharmacokinetic effects of PSC833 treatment imply potential risks, but also promising pharmacological applications of the use of effective reversal agents.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/antagonistas & inhibidores , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/deficiencia , Barrera Hematoencefálica/fisiología , Encéfalo/metabolismo , Ciclosporinas/farmacología , Digoxina/farmacocinética , Contenido Digestivo/química , Administración Oral , Animales , Bilis/efectos de los fármacos , Bilis/metabolismo , Barrera Hematoencefálica/efectos de los fármacos , Ciclosporinas/administración & dosificación , Digoxina/sangre , Resistencia a Múltiples Medicamentos , Intestinos/efectos de los fármacos , Intestinos/fisiología , Hígado/fisiología , Ratones , Ratones Noqueados , Factores de Tiempo , Distribución Tisular
15.
J Clin Invest ; 100(10): 2562-7, 1997 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-9366571

RESUMEN

Hepatocellular secretion of bile salts into the biliary space induces phospholipid and cholesterol secretion, but the mechanism for integrated lipid secretion is poorly understood. Knockout mice unable to make the canalicular membrane mdr2 P-glycoprotein exhibit normal rates of bile salt secretion, yet are virtually incapable of secreting biliary phospholipid and cholesterol. As the mdr2 P-glycoprotein is thought to mediate transmembrane movement of phospholipid molecules, this mouse model was used to examine the mechanism for biliary phospholipid secretion. In wild-type mdr2 (+/+) mice, ultrarapid cryofixation of livers in situ revealed abundant unilamellar lipid vesicles within bile canalicular lumina. Although 74% of vesicles were adherent to the external aspect of the canalicular plasma membrane, bilayer exocytosis was not observed. Vesicle numbers in mdr2 (+/-) and (-/-) mice were 55 and 12% of wild-type levels, respectively. In a strain of mdr2 (-/-) mice which had been "rescued" by heterozygous genomic insertion of the MDR3 gene, the human homologue of the murine mdr2 gene, vesicle numbers returned to 95% of wild-type levels. Our findings indicate that biliary phospholipid is secreted as vesicles by a process largely dependent on the action of the murine mdr2 P-glycoprotein or human MDR3 P-glycoprotein. We conclude that mdr2-mediated phospholipid translocation from the internal to external hemileaflet of the canalicular membrane permits exovesiculation of the external hemileaflet, a vesiculation process promoted by the detergent environment of the bile canalicular lumen.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/biosíntesis , Subfamilia B de Transportador de Casetes de Unión a ATP , Transportadoras de Casetes de Unión a ATP/biosíntesis , Ácidos y Sales Biliares/metabolismo , Canalículos Biliares/fisiología , Bilis/metabolismo , Hígado/fisiología , Fosfolípidos/metabolismo , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/genética , Animales , Canalículos Biliares/ultraestructura , Colesterol/metabolismo , Cruzamientos Genéticos , Resistencia a Múltiples Medicamentos/genética , Femenino , Humanos , Hígado/ultraestructura , Masculino , Ratones , Ratones Mutantes , Modelos Biológicos
16.
J Clin Invest ; 101(7): 1310-9, 1998 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-9525973

RESUMEN

The canalicular (apical) membrane of the hepatocyte contains an ATP-dependent transport system for organic anions, known as the multispecific organic anion transporter (cMOAT). The deduced amino acid sequence of cMOAT is 49% identical to that of the human multidrug resistance- associated protein (MRP) MRP1, and cMOAT and MRP1 are members of the same sub-family of adenine nucleotide binding cassette transporters. In contrast to MRP1, cMOAT was predominantly found intracellularly in nonpolarized cells, suggesting that cMOAT requires a polarized cell for plasma membrane routing. Therefore, we expressed cMOAT cDNA in polarized kidney epithelial MDCK cell lines. When these cells are grown in a monolayer, cMOAT localizes to the apical plasma membrane. We demonstrate that cMOAT causes transport of the organic anions S-(2,4-dinitrophenyl)-glutathione, the glutathione conjugate of ethacrynic acid, and S-(PGA1)-glutathione, a substrate not shown to be transported by organic anion transporters previously. Transport is inhibited only inefficiently by compounds known to block MRP1. We also show that cMOAT causes transport of the anticancer drug vinblastine to the apical side of a cell monolayer. We conclude that cMOAT is a 5'-adenosine triphosphate binding cassette transporter that potentially might be involved in drug resistance in mammalian cells.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Proteínas Portadoras/metabolismo , Células Epiteliales/metabolismo , Proteínas de Transporte de Anión , Transporte Biológico Activo , Compartimento Celular , Línea Celular , Membrana Celular/metabolismo , Polaridad Celular , Técnica del Anticuerpo Fluorescente Indirecta , Glutatión/análogos & derivados , Glutatión/metabolismo , Humanos , Inmunohistoquímica , Riñón/citología , Riñón/metabolismo , Transfección , Vinblastina/metabolismo
17.
J Clin Invest ; 97(5): 1211-8, 1996 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-8636432

RESUMEN

The human multidrug resistance-associated protein MRP confers resistance to various cytotoxic drugs by lowering the intracellular drug concentration. Recent evidence indicates that MRP can also transport glutathione S-conjugates across membranes. To study the transport properties of MRP in intact cells, we have expressed human MRP cDNA in the polarized pig kidney epithelial cell line LLC-PK1. MRP mainly localized to the basolateral plasma membrane of these cells, and not to the apical membrane, as determined by immunocytochemistry using confocal laser scanning and electron microscopy. In accordance with this localization, MRP caused increased transport of the glutathione S-conjugate S-(2, 4-dinitrophenyl)-glutathione and of the anticancer drug daunorubicin to the basal side of the epithelial cell layer. Sulfinpyrazone and probenecid, known inhibitors of multispecific organic anion transport, inhibited this basolateral transport, but not the apical transport of daunorubicin mediated by the apically localized human MDR1 P-glycoprotein in MDR1-transfected LLC-PK1 cells. Probenecid and sulfinpyrazone may therefore be useful lead compounds for the development of clinical reversal agents specific for MRP-mediated drug resistance.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/análisis , Resistencia a Múltiples Medicamentos , Riñón/química , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Transporte Biológico , Línea Celular , Daunorrubicina/farmacocinética , Glutatión/análogos & derivados , Glutatión/metabolismo , Humanos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos , Porcinos
18.
J Clin Invest ; 105(3): 279-85, 2000 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-10675353

RESUMEN

Multidrug resistance protein 1 (MRP1) is a transporter protein that helps to protect normal cells and tumor cells against the influx of certain xenobiotics. We previously showed that Mrp1 protects against cytotoxic drugs at the testis-blood barrier, the oral epithelium, and the kidney urinary collecting duct tubules. Here, we generated Mrp1/Mdr1a/Mdr1b triple-knockout (TKO) mice, and used them together with Mdr1a/Mdr1b double-knockout (DKO) mice to study the contribution of Mrp1 to the tissue distribution and pharmacokinetics of etoposide. We observed increased toxicity in the TKO mice, which accumulated etoposide in brown adipose tissue, colon, salivary gland, heart, and the female urogenital system. Immunohistochemical staining revealed the presence of Mrp1 in the oviduct, uterus, salivary gland, and choroid plexus (CP) epithelium. To explore the transport function of Mrp1 in the CP epithelium, we used TKO and DKO mice cannulated for cerebrospinal fluid (CSF). We show here that the lack of Mrp1 protein causes etoposide levels to increase about 10-fold in the CSF after intravenous administration of the drug. Our results indicate that Mrp1 helps to limit tissue distribution of certain drugs and contributes to the blood-CSF drug-permeability barrier.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/fisiología , Barrera Hematoencefálica/genética , Plexo Coroideo/metabolismo , Etopósido/farmacocinética , Genes MDR , Animales , Líquido Cefalorraquídeo/metabolismo , Plexo Coroideo/patología , Plexo Coroideo/fisiopatología , Células Epiteliales/metabolismo , Células Epiteliales/patología , Femenino , Ratones , Ratones Noqueados
19.
Mol Cell Biol ; 5(3): 545-53, 1985 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-3879972

RESUMEN

The gene encoding variant surface glycoprotein 221 in Trypanosoma brucei is located adjacent to a chromosome end and can be activated with or without a concomitant gene duplication. To test whether transcription initiates within the cloned segment of the 221 gene, we analyzed nascent and stable transcripts. We show here that the 221 coding region and 8.5 kilobases of adjacent upstream DNA are transcribed into nascent RNA at a similar rate when gene 221 is activated without duplication. Since only part of this transcribed upstream segment is transferred with the coding region to another telomere upon duplicative activation of gene 221, we infer that initiation of variant surface glycoprotein gene transcription occurs outside the gene segment that moves into an expression site by gene conversion. Our analysis shows that part of the variant surface glycoprotein 221 transcription unit consists of an unusual 3.5-kilobase tandem array of ca. 50 repeat segments and that a rearrangement in this array accompanies the nonduplicative activation of gene 221. A variant surface glycoprotein pseudogene is located within the transcription unit of gene 221, and we discuss models that account for this unusual situation.


Asunto(s)
Antígenos de Superficie/genética , Glicoproteínas/genética , Trypanosoma brucei brucei/genética , Animales , Secuencia de Bases , Regulación de la Expresión Génica , Genes , Proteínas de la Membrana/genética , Ratones , Modelos Genéticos , Ratas , Transcripción Genética , Trypanosoma brucei brucei/inmunología , Glicoproteínas Variantes de Superficie de Trypanosoma
20.
Mol Cell Biol ; 18(1): 198-205, 1998 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-9418867

RESUMEN

African trypanosomes undergo antigenic variation of their variant surface glycoprotein (VSG) coat to avoid being killed by their mammalian hosts. The active VSG gene is located in one of many telomeric expression sites. Replacement of the VSG gene in the active site or switching between expression sites can give rise to a new VSG coat. To study Trypanosoma brucei VSG expression site inactivation rather than VSG gene switching, it is useful to have an in vitro negative-selection system independent of the VSG. We have achieved this aim by using a viral thymidine kinase (TK) gene. Following integration of the TK gene downstream of the 221a VSG expression site promoter, transformant cell lines became sensitive to the nucleoside analog 1-(2-deoxy-2-fluoro-8-D-arabinofuranosyl)-5-iodouracil. These TK trypanosomes were able to revert to resistance at a rate approaching 10(-5) per cell per generation. The majority of revertants expressed a new VSG gene even though there had been no selection against the VSG itself. Analysis of these switched variants showed that some had shut down TK expression via an in situ expression site switch. However, most variants had the complete 221 expression site deleted and another VSG expression site activated. We speculate that a new VSG expression site cannot switch on without inactivation of the old site.


Asunto(s)
Antígenos de Protozoos/genética , Regulación de la Expresión Génica , Glicoproteínas de Membrana/genética , Trypanosoma brucei brucei/genética , Animales
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA