Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Asian J Pharm Sci ; 15(2): 237-251, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32373202

RESUMEN

Based on the evidence that hemochromatosis, an iron-overload disease, drives hepatocellular carcinoma, we hypothesized that chronic exposure to excess iron, either due to genetic or environmental causes, predisposes an individual to cancer. Using pancreatic cancer as our primary focus, we employed cell culture studies to interrogate the connection between excess iron and cancer, and combined in vitro and in vivo studies to explore the connection further. Ferric ammonium citrate was used as an exogenous iron source. Chronic exposure to excess iron induced epithelial-mesenchymal transition (EMT) in normal and cancer cell lines, loss of p53, and suppression of p53 transcriptional activity evidenced from decreased expression of p53 target genes (p21, cyclin D1, Bax, SLC7A11). To further extrapolate our cell culture data, we generated EL-KrasG12D (EL-Kras) mouse (pancreatic neoplastic mouse model) expressing Hfe+/+ and Hfe-/- genetic background. p53 target gene expression decreased in EL-Kras/Hfe-/- mouse pancreas compared to EL-Kras/Hfe+/+ mouse pancreas. Interestingly, the incidence of acinar-to-ductal metaplasia and cystic pancreatic neoplasms (CPN) decreased in EL-Kras/Hfe-/- mice, but the CPNs that did develop were larger in these mice than in EL-Kras/Hfe+/+ mice. In conclusion, these in vitro and in vivo studies support a potential role for chronic exposure to excess iron as a promoter of more aggressive disease via p53 loss and SLC7A11 upregulation within pancreatic epithelial cells.

2.
Pharm Res ; 36(6): 84, 2019 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-30997560

RESUMEN

PURPOSE: ß-Hydroxy-ß-methylbutyrate (HMB), a nutritional supplement, elicits anabolic activity in muscle. Here we investigated the mechanism of HMB uptake in muscle cells. METHODS: Murine muscle cells (C2C12) and human mammary epithelial cells (MCF7) were used for uptake. As HMB is a monocarboxylate, focus was on monocarboxylate transporters, monitoring interaction of HMB with H+-coupled lactate uptake, and influence of H+ directly on HMB uptake. Involvement of MCT1-4 was studied using selective inhibitors and gene silencing. Involvement of human Na+/monocarboxylate transporter SMCT1 was also assessed using Xenopus oocytes. RESULTS: H+-coupled lactate uptake was inhibited by HMB in both mammalian cells. HMB uptake was H+-coupled and inhibited by lactate. C2C12 cells expressed MCT1 and MCT4; MCF7 cells expressed MCT1-4; undifferentiated C2C12 cells expressed SMCT1. SMCT1 mediated Na+-coupled HMB transport. Inhibitors of MCT1/4, siRNA-mediated gene silencing, and expression pattern showed that MCT1-4 were responsible only for a small portion of HMB uptake in these cells. CONCLUSION: HMB uptake in C2C12 and MCF7 cells is primarily H+-coupled and inhibited by lactate, but MCT1-4 are only partly responsible for HMB uptake. SMCT1 also transports HMB, but in a Na+-coupled manner. Other, yet unidentified, transporters mediate the major portion of HMB uptake in C2C12 and MCF7 cells.


Asunto(s)
Suplementos Dietéticos , Transportadores de Ácidos Monocarboxílicos/metabolismo , Valeratos/metabolismo , Animales , Transporte Biológico , Línea Celular , Células Epiteliales/metabolismo , Silenciador del Gen , Humanos , Ácido Láctico/metabolismo , Células MCF-7 , Ratones , Transportadores de Ácidos Monocarboxílicos/antagonistas & inhibidores , Células Musculares/metabolismo , ARN Interferente Pequeño , Transducción de Señal , Sodio/metabolismo , Xenopus laevis
4.
Sci Rep ; 8(1): 2519, 2018 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-29410496

RESUMEN

NaCT is a Na+-coupled transporter for citrate expressed in hepatocytes and neurons. It is the mammalian ortholog of INDY (I'm Not Dead Yet), a transporter which modifies lifespan in Drosophila. Here we describe a hitherto unknown transport system for citrate in mammalian cells. When liver and mammary epithelial cells were pretreated with the iron supplement ferric ammonium citrate (FAC), uptake of citrate increased >10-fold. Iron chelators abrogated the stimulation of citrate uptake in FAC-treated cells. The iron exporter ferroportin had no role in this process. The stimulation of citrate uptake also occurred when Fe3+ was added during uptake without pretreatment. Similarly, uptake of Fe3+ was enhanced by citrate. The Fe3+-citrate uptake was coupled to Na+. This transport system was detectable in primary hepatocytes and neuronal cell lines. The functional features of this citrate transport system distinguish it from NaCT. Loss-of-function mutations in NaCT cause early-onset epilepsy and encephalopathy; the newly discovered Na+-coupled Fe3+-citrate transport system might offer a novel treatment strategy for these patients to deliver citrate into affected neurons independent of NaCT. It also has implications to iron-overload conditions where circulating free iron increases, which would stimulate cellular uptake of citrate and consequently affect multiple metabolic pathways.

5.
Nutrients ; 8(11)2016 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-27801863

RESUMEN

Nutrient tranters (NT) facilitate nutrient absorption and contribute to the regulation of circulating nutrients. In this cross-sectional study, we determined the associations between the level of obesity; mRNA abundance for NTs; and serum concentrations of amino acids, short-chain fatty acids, and glucose in patients with morbid obesity undergoing a Roux-en-Y gastric bypass. Proximal jejunal samples were obtained at the time of surgery from 42 patients (90% female, age = 42.6 ± 11.9 years, pre-operative body mass index (BMI) = 55.5 ± 11.3 kg/m²) undergoing a Roux-en-Y gastric bypass. RNA was extracted from the jejunal mucosa and quantitative real-time-PCR was performed for the NTs studied. BMI negatively correlated with jejunal mRNA abundance of the amino acid NTs TauT (r = -0.625, p < 0.0001), ASCT2 (r = -0.320, p = 0.039), LAT1 (r = -0.304, p = 0.05). BMI positively correlated with jejunal mRNA abundance of the lactate/short-chain fatty acid NT SMCT1 (r = 0.543, p = 0.0002). Serum concentrations of the short-chain fatty acids, butyric, valeric, and isocaproic acid correlated positively with BMI (n = 30) (r = 0.45, r = 0.44, r = 0.36, p ≤ 0.05; respectively). Lower jejunal mRNA abundance for the amino acid NTs TauT, ASCT2, and LAT1 could protect against further obesity-related elevations in circulating amino acids. The positive correlation between BMI and the jejunal mRNA abundance of the high-affinity short-chain fatty acid/monocarboxylate transporter SMCT1 is intriguing and requires further investigation.


Asunto(s)
Ácidos Grasos Volátiles/metabolismo , Regulación de la Expresión Génica , Mucosa Intestinal/metabolismo , Yeyuno/metabolismo , Transportadores de Ácidos Monocarboxílicos/metabolismo , Obesidad Mórbida/metabolismo , Obesidad/metabolismo , Adulto , Índice de Masa Corporal , Estudios de Cohortes , Comorbilidad , Estudios Transversales , Ácidos Grasos Volátiles/sangre , Femenino , Derivación Gástrica , Humanos , Mucosa Intestinal/cirugía , Yeyuno/cirugía , Masculino , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Persona de Mediana Edad , Transportadores de Ácidos Monocarboxílicos/genética , Obesidad/sangre , Obesidad/epidemiología , Obesidad/cirugía , Obesidad Mórbida/sangre , Obesidad Mórbida/patología , Obesidad Mórbida/cirugía , ARN Mensajero/metabolismo , Circunferencia de la Cintura
6.
Biochem J ; 473(11): 1503-6, 2016 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-27234586

RESUMEN

Tumour cell metabolism is very different from normal cell metabolism; cancer cells re-programme the metabolic pathways that occur in normal cells in such a manner that it optimizes their proliferation, growth and survival. Although this metabolic re-programming obviously operates to the advantage of the tumour, it also offers unique opportunities for effective cancer therapy. Molecules that target the tumour cell-specific metabolic pathways have potential as novel anti-cancer drugs. Lonidamine belongs to this group of molecules and is already in use in some countries for cancer treatment. It has been known for a long time that lonidamine interferes with energy production in tumour cells by inhibiting hexokinase II (HKII), a glycolytic enzyme. However, subsequent studies have uncovered additional pharmacological targets for the drug, which include the electron transport chain and the mitochondrial permeability transition pore, thus expanding the pharmacological effects of the drug on tumour cell metabolism. A study by Nancolas et al. in a recent issue of the Biochemical Journal identifies two additional new targets for lonidamine: the pyruvate transporter in the mitochondria and the H(+)-coupled monocarboxylate transporters in the plasma membrane (PM). It is thus becoming increasingly apparent that the anti-cancer effects of lonidamine do not occur through a single target; the drug works at multiple sites. Irrespective of the molecular targets, what lonidamine does in the end is to undo what the tumour cells have done in terms of re-programming cellular metabolism and mitochondrial function.


Asunto(s)
Antineoplásicos/uso terapéutico , Indazoles/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Glucólisis/efectos de los fármacos , Humanos , Transducción de Señal/efectos de los fármacos
7.
Biochem J ; 473(9): 1113-24, 2016 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-27118869

RESUMEN

The role of plasma membrane transporters in cancer is receiving increasing attention in recent years. Several transporters for essential nutrients are up-regulated in cancer and serve as tumour promoters. Transporters could also function as tumour suppressors. To date, four transporters belonging to the SLC gene family have been identified as tumour suppressors. SLC5A8 is a Na(+)-coupled transporter for monocarboxylates. Among its substrates are the bacterial fermentation products butyrate and propionate and the ubiquitous metabolite pyruvate. The tumour-suppressive function of this transporter relates to the ability of butyrate, propionate and pyruvate to inhibit histone deacetylases (HDAC). SLC5A8 functions as a tumour suppressor in most tissues studied thus far, and provides a molecular link to Warburg effect, a characteristic feature in most cancers. It also links colonic bacteria and dietary fibre to the host. SLC26A3 as a tumour suppressor is restricted to colon; it is a Cl(-)/HCO(-) 3 exchanger, facilitating the efflux of HCO(-) 3 The likely mechanism for the tumour-suppressive function of SLC26A3 is related to intracellular pH regulation. SLC39A1 is a Zn(2+) transporter and its role in tumour suppression has been shown in prostate. Zn(2+) is present at high concentrations in normal prostate where it elicits its tumour-suppressive function. SLC22A18 is possibly an organic cation transporter, but the identity of its physiological substrates is unknown. As such, there is no information on molecular pathways responsible for the tumour-suppressive function of this transporter. It is likely that additional SLC transporters will be discovered as tumour suppressors in the future.


Asunto(s)
Antiportadores de Cloruro-Bicarbonato/metabolismo , Transportadores de Ácidos Monocarboxílicos/metabolismo , Neoplasias/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Antiportadores de Cloruro-Bicarbonato/genética , Humanos , Transporte Iónico/genética , Transportadores de Ácidos Monocarboxílicos/genética , Neoplasias/genética , Transportadores de Sulfato , Proteínas Supresoras de Tumor/genética
8.
Biochem J ; 469(1): 17-23, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-26173258

RESUMEN

SLC6A14 mediates Na(+)/Cl(-)-coupled concentrative uptake of a broad-spectrum of amino acids. It is expressed at low levels in many tissues but up-regulated in certain cancers. Pharmacological blockade of SLC6A14 causes amino acid starvation in estrogen receptor positive (ER+) breast cancer cells and suppresses their proliferation in vitro and in vivo. In the present study, we interrogated the role of this transporter in breast cancer by deleting Slc6a14 in mice and monitoring the consequences of this deletion in models of spontaneous breast cancer (Polyoma middle T oncogene-transgenic mouse and mouse mammary tumour virus promoter-Neu-transgenic mouse). Slc6a14-knockout mice are viable, fertile and phenotypically normal. The plasma amino acids were similar in wild-type and knockout mice and there were no major compensatory changes in the expression of other amino acid transporter mRNAs. There was also no change in mammary gland development in the knockout mouse. However, when crossed with PyMT-Tg mice or MMTV/Neu (mouse mammary tumour virus promoter-Neu)-Tg mice, the development and progression of breast cancer were markedly decreased on Slc6a14(-/-) background. Analysis of transcriptomes in tumour tissues from wild-type mice and Slc6a14-null mice indicated no compensatory changes in the expression of any other amino acid transporter mRNA. However, the tumours from the null mice showed evidence of amino acid starvation, decreased mTOR signalling and decreased cell proliferation. These studies demonstrate that SLC6A14 is critical for the maintenance of amino acid nutrition and optimal mammalian target of rapamycin (mTOR) signalling in ER+ breast cancer and that the transporter is a potential target for development of a novel class of anti-cancer drugs targeting amino acid nutrition in tumour cells.


Asunto(s)
Sistemas de Transporte de Aminoácidos , Proliferación Celular , Eliminación de Gen , Neoplasias Mamarias Experimentales/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas de Transporte de Neurotransmisores en la Membrana Plasmática , Transducción de Señal , Animales , Sistemas de Liberación de Medicamentos , Femenino , Neoplasias Mamarias Experimentales/dietoterapia , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Noqueados , Proteínas de Neoplasias/genética , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo
9.
Cancer Res ; 75(9): 1782-8, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25855379

RESUMEN

Tumor cells have an increased demand for amino acids because of their rapid proliferation rate. In addition to their need in protein synthesis, several amino acids have other roles in supporting cancer growth. There are approximately two-dozen amino acid transporters in humans, and tumor cells must upregulate one or more of these transporters to satisfy their demand for amino acids. If the transporters that specifically serve this purpose in tumor cells are identified, they can be targeted for the development of a brand new class of anticancer drugs; the logical basis of such a strategy would be to starve the tumor cells of an important class of nutrients. To date, four amino acid transporters have been found to be expressed at high levels in cancer: SLC1A5, SLC7A5, SLC7A11, and SLC6A14. Their induction occurs in a cancer type-specific manner with a direct or indirect involvement of the oncogene c-Myc. Further, these transporters are functionally coupled, thus maximizing their ability to promote cancer growth and chemoresistance. Progress has been made in preclinical studies, exploiting these transporters as drug targets in cancer therapy. These transporters also show promise in development of new tumor-imaging probes and in tumor-specific delivery of appropriately designed chemotherapeutic agents.


Asunto(s)
Sistemas de Transporte de Aminoácidos/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Glutamina/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Animales , Humanos , Terapia Molecular Dirigida
10.
J Pharmacol Exp Ther ; 353(1): 17-26, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25617245

RESUMEN

NaCT (SLC13A5) is a Na(+)-coupled transporter for Krebs cycle intermediates and is expressed predominantly in the liver. Human NaCT is relatively specific for citrate compared with other Krebs cycle intermediates. The transport activity of human NaCT is stimulated by Li(+), whereas that of rat NaCT is inhibited by Li(+). We studied the influence of Li(+) on NaCTs cloned from eight different species. Li(+) stimulated the activity of only NaCTs from primates (human, chimpanzee, and monkey); by contrast, NaCTs from nonprimate species (mouse, rat, dog, and zebrafish) were inhibited by Li(+). Caenorhabditis elegans NaCT was not affected by Li(+). With human NaCT, the Li(+)-induced increase in transport activity was associated with the conversion of the transporter from a low-affinity/high-capacity type to a high-affinity/low-capacity type. H(+) was able to substitute for Li(+) in eliciting the stimulatory effect. The amino acid Phe500 in human NaCT was critical for Li(+)/H(+)-induced stimulation. Mutation of this amino acid to tryptophan (F500W) markedly increased the basal transport activity of human NaCT in the absence of Li(+), but the ability of Li(+) to stimulate the transporter was almost completely lost with this mutant. Substitution of Phe500 with tryptophan in human NaCT converted the transporter from a low-affinity/high-capacity type to a high-affinity/low-capacity type, an effect similar to that of Li(+) on the wild-type NaCT. These studies show that Li(+)-induced activation of NaCT is specific for the transporter in primates and that the region surrounding Phe500 in primate NaCTs is important for the Li(+) effect.


Asunto(s)
Compuestos de Litio/farmacología , Simportadores/metabolismo , Animales , Transporte Biológico , Caenorhabditis elegans , Línea Celular , Citratos/metabolismo , Perros , Femenino , Humanos , Macaca mulatta , Ratones , Mutación , Oocitos/metabolismo , Pan troglodytes , Ratas , Especificidad de la Especie , Simportadores/genética , Xenopus laevis , Pez Cebra
11.
Biochim Biophys Acta ; 1848(2): 453-62, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25450809

RESUMEN

IDO1, which encodes the immunosuppressive and tryptophan-catabolizing enzyme indoleamine 2,3-dioxygenase-1 (IDO1), is a target for interferon-γ (IFN-γ). IDO1-mediated tryptophan catabolism in dendritic cells and macrophages arrests T cell proliferation, thereby providing a molecular basis for the immunosuppressive function of IDO1. Whether the entry of tryptophan into IDO1-expressing cells is also regulated by IFN-γ is not known. Here we used a human colonic epithelial cell line (CCD841) and a mouse dendritic cell line (DC2.4) to test the hypothesis that IFN-γ, which induces IDO1, also induces a tryptophan transporter to promote substrate availability to IDO1. Upon treatment with IFN-γ, there was a marked increase in IDO1 mRNA and a concomitant increase in tryptophan uptake in both cell lines. The induced uptake system was selective for tryptophan and saturable with a Michaelis constant of 36±3 µM in CCD841 cells and 0.5±0.1 µM in DC2.4 cells. The induction by IFN-γ and the tryptophan-selectivity of the induced transport system were demonstrable even in the presence of physiologic concentrations of all other amino acids. Since kynurenine, the catabolic end product of IDO1, is a signaling molecule as an agonist for the aryl hydrocarbon receptor (AhR), we examined if AhR signaling induces the tryptophan-selective transporter. Treatment of the cells with kynurenine and other AhR agonists increased tryptophan uptake. The present studies demonstrate that IFN-γ coordinately induces IDO1 and a tryptophan-selective transporter to maximize tryptophan depletion in IDO1-expressing cells and that the process involves a positive feedback mechanism via kynurenine-AhR signaling.


Asunto(s)
Sistemas de Transporte de Aminoácidos/genética , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Interferón gamma/farmacología , Receptores de Hidrocarburo de Aril/genética , Triptófano/metabolismo , Sistemas de Transporte de Aminoácidos/agonistas , Sistemas de Transporte de Aminoácidos/metabolismo , Animales , Transporte Biológico , Línea Celular , Colon/citología , Colon/efectos de los fármacos , Colon/metabolismo , Células Dendríticas/citología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Retroalimentación Fisiológica , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Interferón gamma/metabolismo , Cinética , Quinurenina/metabolismo , Ratones , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Transducción de Señal
12.
Mol Cell Biol ; 33(19): 3920-35, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23918800

RESUMEN

SLC5A8 is a putative tumor suppressor that is inactivated in more than 10 different types of cancer, but neither the oncogenic signaling responsible for SLC5A8 inactivation nor the functional relevance of SLC5A8 loss to tumor growth has been elucidated. Here, we identify oncogenic HRAS (HRAS(G12V)) as a potent mediator of SLC5A8 silencing in human nontransformed normal mammary epithelial cell lines and in mouse mammary tumors through DNMT1. Further, we demonstrate that loss of Slc5a8 increases cancer-initiating stem cell formation and promotes mammary tumorigenesis and lung metastasis in an HRAS-driven murine model of mammary tumors. Mammary-gland-specific overexpression of Slc5a8 (mouse mammary tumor virus-Slc5a8 transgenic mice), as well as induction of endogenous Slc5a8 in mice with inhibitors of DNA methylation, protects against HRAS-driven mammary tumors. Collectively, our results provide the tumor-suppressive role of SLC5A8 and identify the oncogenic HRAS as a mediator of tumor-associated silencing of this tumor suppressor in mammary glands. These findings suggest that pharmacological approaches to reactivate SLC5A8 expression in tumor cells have potential as a novel therapeutic strategy for breast cancer treatment.


Asunto(s)
Neoplasias de la Mama/genética , Proteínas de Transporte de Catión/genética , Regulación Neoplásica de la Expresión Génica , Proteínas Proto-Oncogénicas p21(ras)/genética , Animales , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Carcinogénesis/genética , Carcinogénesis/metabolismo , Proteínas de Transporte de Catión/metabolismo , Línea Celular , Línea Celular Tumoral , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Femenino , Células HCT116 , Humanos , Immunoblotting , Células MCF-7 , Masculino , Ratones , Ratones Noqueados , Ratones Desnudos , Ratones Transgénicos , Transportadores de Ácidos Monocarboxílicos , Mutación , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Trasplante Heterólogo
13.
Invest Ophthalmol Vis Sci ; 54(3): 1592-602, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23404113

RESUMEN

PURPOSE: Oxidative stress is a common pathological factor in degenerative retinal diseases; therefore, identifying novel strategies for its limitation is critically important and highly relevant clinically. Along these lines, our present goal was to evaluate the effect(s) of the fumarate ester and antipsoriatic agent monomethylfumarate (MMF) on the expression and functional activity of the cystine/glutamate exchanger SLC7A11 (system xc(-)), a transport system critical to potentiation of antioxidant signaling in retina. METHODS: ARPE-19 and primary mouse RPE cells were cultured in the presence or absence of varying concentrations of MMF (0-5000 µM) for 0 to 24 hours. MMF (10 mM) was also delivered intravitreally to mouse eyes. RT-PCR, radiolabeled uptake, Western blotting, and glutathione (GSH) assays were then used to evaluate the effects of MMF on endogenous antioxidant machinery. RESULTS: MMF induced system xc(-), Nrf2, and hypoxia-inducible factor 1α (Hif-1α) in cultured RPE cells. Additionally, the compound was recognized as a transportable substrate by the Na(+)-coupled monocarboxylate transporter SLC5A8 (SMCT1). In vivo these factors were evidenced by a significant increase in retinal levels of GSH. CONCLUSIONS: MMF stimulates multiple pathways in retinal cells that potentiate cellular events leading to the upregulation of genes/mechanisms that function to protect retina against various forms of insult; upregulation of system xc(-) is one such consequence. To our knowledge, this is the first report that fumarate esters, compounds already employed clinically for other indications, are effective in retina via xc(-) induction. This novel, hitherto unknown mechanism helps to explain the antioxidant feature of these compounds and highlights their therapeutic potential in retina.


Asunto(s)
Sistema de Transporte de Aminoácidos y+/metabolismo , Fármacos Dermatológicos/farmacología , Células Epiteliales/metabolismo , Fumaratos/farmacología , Maleatos/farmacología , Epitelio Pigmentado de la Retina/citología , Sistema de Transporte de Aminoácidos y+/genética , Animales , Células Cultivadas , Células Epiteliales/efectos de los fármacos , Ojo/metabolismo , Glutatión/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia , Ratones , Ratones Endogámicos BALB C , Factor 2 Relacionado con NF-E2/metabolismo , ARN Mensajero/metabolismo , Regulación hacia Arriba
14.
Drug Metab Dispos ; 40(6): 1138-43, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22396231

RESUMEN

Plasma membrane monoamine transporter (PMAT) is a polyspecific organic cation (OC) transporter that transports a variety of endogenous biogenic amines and xenobiotic cations. Previous radiotracer uptake studies showed that PMAT-mediated OC transport is sensitive to changes in membrane potential and extracellular pH, but the precise role of membrane potential and protons on PMAT-mediated OC transport is unknown. Here, we characterized the electrophysiological properties of PMAT in Xenopus laevis oocytes using a two-microelectrode voltage-clamp approach. PMAT-mediated histamine uptake is associated with inward currents under voltage-clamp conditions, and the currents increased in magnitude as the holding membrane potential became more negative. A similar effect was also observed for another cation, nicotine. Substrate-induced currents were largely independent of Na+ but showed strong dependence on membrane potential and pH of the perfusate. Detailed kinetic analysis of histamine uptake revealed that the energizing effect of membrane potentials on PMAT transport is mainly due to an augmentation of Imax with little effect on K0.5. At most holding membrane potentials, Imax at pH 6.0 is approximately 3- to 4-fold higher than that at pH 7.5, whereas K0.5 is not dependent on pH. Together, these data unequivocally demonstrate PMAT as an electrogenic transporter and establish the physiological inside-negative membrane potential as a driving force for PMAT-mediated OC transport. The important role of membrane potential and pH in modulating the transport activity of PMAT toward OCs suggests that the in vivo activity of PMAT could be regulated by pathophysiological processes that alter physiological pH or membrane potential.


Asunto(s)
Membrana Celular/fisiología , Proteínas de Transporte de Nucleósido Equilibrativas/fisiología , Proteínas de Transporte de Catión Orgánico/fisiología , Proteínas de Xenopus/fisiología , Animales , Fenómenos Electrofisiológicos/fisiología , Femenino , Humanos , Potenciales de la Membrana/fisiología , Xenopus laevis
15.
J Biol Chem ; 286(36): 31830-8, 2011 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-21771784

RESUMEN

SLC6A14, also known as ATB(0,+), is an amino acid transporter with unique characteristics. It transports 18 of the 20 proteinogenic amino acids. However, this transporter is expressed only at low levels in normal tissues. Here, we show that the transporter is up-regulated specifically in estrogen receptor (ER)-positive breast cancer, demonstrable with primary human breast cancer tissues and human breast cancer cell lines. SLC6A14 is an estrogen/ER target. The transport features of SLC6A14 include concentrative transport of leucine (an activator of mTOR), glutamine (an essential amino acid for nucleotide biosynthesis and substrate for glutaminolysis), and arginine (an essential amino acid for tumor cells), suggesting that ER-positive breast cancer cells up-regulate SLC6A14 to meet their increased demand for these amino acids. Consequently, treatment of ER-positive breast cancer cells in vitro with α-methyl-DL-tryptophan (α-MT), a selective blocker of SLC6A14, induces amino acid deprivation, inhibits mTOR, and activates autophagy. Prolongation of the treatment with α-MT causes apoptosis. Addition of an autophagy inhibitor (3-methyladenine) during α-MT treatment also induces apoptosis. These effects of α-MT are specific to ER-positive breast cancer cells, which express the transporter. The ability of α-MT to cause amino acid deprivation is significantly attenuated in MCF-7 cells, an ER-positive breast cancer cell line, when SLC6A14 is silenced with shRNA. In mouse xenograft studies, α-MT by itself is able to reduce the growth of the ER-positive ZR-75-1 breast cancer cells. These studies identify SLC6A14 as a novel and effective drug target for the treatment of ER-positive breast cancer.


Asunto(s)
Sistemas de Transporte de Aminoácidos Neutros/antagonistas & inhibidores , Neoplasias de la Mama/tratamiento farmacológico , Sistemas de Transporte de Aminoácidos , Sistemas de Transporte de Aminoácidos Neutros/genética , Animales , Autofagia/efectos de los fármacos , Neoplasias de la Mama/patología , Femenino , Humanos , Ratones , Terapia Molecular Dirigida/métodos , Receptores de Estrógenos , Trasplante Heterólogo , Triptófano/análogos & derivados , Triptófano/farmacología , Células Tumorales Cultivadas
16.
Invest Ophthalmol Vis Sci ; 52(8): 5749-57, 2011 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-21508099

RESUMEN

PURPOSE: To evaluate the role of SLC5A8 in the transport of 2-oxothiazolidine-4-carboxylate (OTC) and to determine whether OTC augments glutathione production in RPE cells, thereby providing protection against oxidative stress. METHODS: SLC5A8-mediated transport of OTC was monitored in Xenopus laevis oocytes by electrophysiological means. Saturation kinetics, Na(+)-activation kinetics, and inhibition by ibuprofen were analyzed by monitoring OTC-induced currents as a measure of transport activity. Oxidative stress was induced in ARPE-19 cells and primary RPE cells isolated from wild type and Slc5a8(-/-) mouse retinas using H(2)O(2), and the effects of OTC on cell death and intracellular glutathione concentration were examined. RESULTS: Heterologous expression of human SLC5A8 in X. laevis oocytes induced Na(+)-dependent inward currents in the presence of OTC under voltage-clamp conditions. The transport of OTC via SLC5A8 was saturable, with a K(t) of 104 ± 3 µM. The Na(+)-activation kinetics was sigmoidal with a Hill coefficient of 1.9 ± 0.1, suggesting involvement of two Na(+) in the activation process. Ibuprofen, a blocker of SLC5A8, inhibited SLC5A8-mediated OTC transport; the concentration necessary for half-maximal inhibition was 17 ± 1 µM. OTC increased glutathione levels and protected ARPE-19 and primary RPE cells isolated from wild type mouse retinas from H(2)O(2)-induced cell death. These effects were abolished in primary RPE isolated from Slc5a8(-/-) mouse retinas. CONCLUSIONS: OTC is a transportable substrate for SLC5A8. OTC augments glutathione production in RPE cells, thereby protecting them from oxidative damage. Transport via SLC5A8 is obligatory for this process.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Glutatión/metabolismo , Ácido Pirrolidona Carboxílico/farmacocinética , Epitelio Pigmentado de la Retina/efectos de los fármacos , Epitelio Pigmentado de la Retina/metabolismo , Tiazolidinas/farmacocinética , Animales , Apoptosis , Transporte Biológico/fisiología , Proteínas de Transporte de Catión/genética , Supervivencia Celular/efectos de los fármacos , Inhibidores de la Ciclooxigenasa/farmacología , Interacciones Farmacológicas , Humanos , Peróxido de Hidrógeno/toxicidad , Ibuprofeno/farmacología , Cinética , Ratones , Ratones Mutantes , Transportadores de Ácidos Monocarboxílicos , Oocitos/fisiología , Oxidantes/toxicidad , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Ácido Pirrolidona Carboxílico/química , Epitelio Pigmentado de la Retina/citología , Tiazolidinas/química , Xenopus laevis
17.
J Pharmacol Sci ; 113(2): 192-6, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20508390

RESUMEN

Aristolochic acids (AAs), contained in Chinese herbal preparations, have been considered to induce nephropathy. In order to elucidate the molecular mechanisms of AA-induced nephrotoxicity, we have elucidated the interaction of human organic anion transporters (hOATs) with AAs using their stable cell lines. AA-I and AA-II inhibited organic anion uptake by hOAT1, hOAT3, and hOAT4 in dose-dependent manners. Treatment of hOAT3 with AA-I resulted in a significant reduction in viability compared with that of mock, which was rescued by the organic anion transport inhibitor probenecid. In conclusion, hOAT3-mediated AA-I uptake may be associated with the induction of nephrotoxicity.


Asunto(s)
Aniones/metabolismo , Ácidos Aristolóquicos/toxicidad , Transportadores de Anión Orgánico/farmacología , Animales , Ácidos Aristolóquicos/antagonistas & inhibidores , Ácidos Aristolóquicos/farmacología , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Interacciones Farmacológicas , Medicamentos Herbarios Chinos , Probenecid/farmacología
18.
Biochim Biophys Acta ; 1798(6): 1164-71, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20211600

RESUMEN

Pyroglutamate, also known as 5-oxoproline, is a structural analog of proline. This amino acid derivative is a byproduct of glutathione metabolism, and is reabsorbed efficiently in kidney by Na(+)-coupled transport mechanisms. Previous studies have focused on potential participation of amino acid transport systems in renal reabsorption of this compound. Here we show that it is not the amino acid transport systems but instead the Na(+)-coupled monocarboxylate transporter SLC5A8 that plays a predominant role in this reabsorptive process. Expression of cloned human and mouse SLC5A8 in mammalian cells induces Na(+)-dependent transport of pyroglutamate that is inhibitable by various SLC5A8 substrates. SLC5A8-mediated transport of pyroglutamate is saturable with a Michaelis constant of 0.36+/-0.04mM. Na(+)-activation of the transport process exhibits sigmoidal kinetics with a Hill coefficient of 1.8+/-0.4, indicating involvement of more than one Na(+) in the activation process. Expression of SLC5A8 in Xenopuslaevis oocytes induces Na(+)-dependent inward currents in the presence of pyroglutamate under voltage-clamp conditions. The concentration of pyroglutamate necessary for induction of half-maximal current is 0.19+/-0.01mM. The Na(+)-activation kinetics is sigmoidal with a Hill coefficient of 2.3+/-0.2. Ibuprofen, a blocker of SLC5A8, suppressed pyroglutamate-induced currents in SLC5A8-expressing oocytes; the concentration of the blocker necessary for causing half-maximal inhibition is 14+/-1microM. The involvement of SLC5A8 can be demonstrated in rabbit renal brush border membrane vesicles by showing that the Na(+)-dependent uptake of pyroglutamate in these vesicles is inhibitable by known substrates of SLC5A8. The Na(+) gradient-driven pyroglutamate uptake was stimulated by an inside-negative K(+) diffusion potential induced by valinomycin, showing that the uptake process is electrogenic.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Membrana Celular/metabolismo , Ácido Pirrolidona Carboxílico/metabolismo , Sodio/metabolismo , Animales , Transporte Biológico/efectos de los fármacos , Transporte Biológico/fisiología , Proteínas de Transporte de Catión/genética , Línea Celular , Membrana Celular/genética , Expresión Génica , Humanos , Ionóforos/farmacología , Riñón/metabolismo , Cinética , Ratones , Microvellosidades/genética , Microvellosidades/metabolismo , Transportadores de Ácidos Monocarboxílicos , Oocitos , Técnicas de Placa-Clamp , Potasio/metabolismo , Conejos , Ratas , Valinomicina/farmacología , Xenopus laevis
19.
J Am Soc Nephrol ; 20(7): 1586-96, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19443642

RESUMEN

LAT3 is a Na+-independent neutral l-amino acid transporter recently isolated from a human hepatocellular carcinoma cell line. Although liver, skeletal muscle, and pancreas are known to express LAT3, the tissue distribution and physiologic function of this transporter are not completely understood. Here, we observed that glomeruli express LAT3. Immunofluorescence, confocal microscopy, and immunoelectron microscopy revealed that LAT3 localizes to the apical plasma membrane of podocyte foot processes. In mice, starvation upregulated glomerular LAT3, phosphorylated AKT1, reconstituted the actin network, and elongated foot processes. In the fetal kidney, we observed intense LAT3 expression at the capillary loops stage of renal development. Finally, zebrafish morphants lacking lat3 function showed collapsed glomeruli with thickened glomerular basement membranes. Permeability studies of the glomerular filtration barrier in these zebrafish morphants demonstrated a disruption of selective glomerular permeability. Our data suggest that LAT3 may play a crucial role in the development and maintenance of podocyte structure and function by regulating protein synthesis and the actin cytoskeleton.


Asunto(s)
Sistemas de Transporte de Aminoácidos Básicos/metabolismo , Diferenciación Celular/fisiología , Glomérulos Renales/metabolismo , Podocitos/metabolismo , Actinas/metabolismo , Sistemas de Transporte de Aminoácidos Básicos/genética , Animales , Membrana Celular/metabolismo , Citoesqueleto/metabolismo , Femenino , Membrana Basal Glomerular/metabolismo , Tasa de Filtración Glomerular/fisiología , Humanos , Glomérulos Renales/citología , Glomérulos Renales/embriología , Masculino , Ratones , Ratones Endogámicos ICR , Fosforilación , Podocitos/citología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Pez Cebra , Proteínas de Pez Cebra/metabolismo
20.
J Biol Chem ; 283(40): 26834-8, 2008 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-18701466

RESUMEN

Hyperuricemia is a significant factor in a variety of diseases, including gout and cardiovascular diseases. Although renal excretion largely determines plasma urate concentration, the molecular mechanism of renal urate handling remains elusive. Previously, we identified a major urate reabsorptive transporter, URAT1 (SLC22A12), on the apical side of the renal proximal tubular cells. However, it is not known how urate taken up by URAT1 exits from the tubular cell to the systemic circulation. Here, we report that a sugar transport facilitator family member protein GLUT9 (SLC2A9) functions as an efflux transporter of urate from the tubular cell. GLUT9-expressed Xenopus oocytes mediated saturable urate transport (K(m): 365+/-42 microm). The transport was Na(+)-independent and enhanced at high concentrations of extracellular potassium favoring negative to positive potential direction. Substrate specificity and pyrazinoate sensitivity of GLUT9 was distinct from those of URAT1. The in vivo role of GLUT9 is supported by the fact that a renal hypouricemia patient without any mutations in SLC22A12 was found to have a missense mutation in SLC2A9, which reduced urate transport activity in vitro. Based on these data, we propose a novel model of transcellular urate transport in the kidney; urate [corrected] is taken up via apically located URAT1 and exits the cell via basolaterally located GLUT9, which we suggest be renamed URATv1 (voltage-driven urate transporter 1).


Asunto(s)
Proteínas Facilitadoras del Transporte de la Glucosa/metabolismo , Hiperuricemia/metabolismo , Túbulos Renales Proximales/metabolismo , Modelos Biológicos , Transportadores de Anión Orgánico/metabolismo , Proteínas de Transporte de Catión Orgánico/metabolismo , Ácido Úrico/metabolismo , Animales , Enfermedades Cardiovasculares/complicaciones , Enfermedades Cardiovasculares/genética , Enfermedades Cardiovasculares/metabolismo , Femenino , Expresión Génica , Proteínas Facilitadoras del Transporte de la Glucosa/genética , Humanos , Hiperuricemia/etiología , Hiperuricemia/genética , Transporte Iónico/genética , Mutación Missense , Oocitos , Transportadores de Anión Orgánico/genética , Proteínas de Transporte de Catión Orgánico/genética , Xenopus laevis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...