Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
World J Gastroenterol ; 28(10): 1024-1054, 2022 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-35431492

RESUMEN

BACKGROUND: The expression of amino acid transporters is known to vary during acute pancreatitis (AP) except for LAT1 (slc7a5), the expression of which remains stable. LAT1 supports cell growth by importing leucine and thereby stimulates mammalian target of rapamycin (mTOR) activity, a phenomenon often observed in cancer cells. The mechanisms by which LAT1 influences physiological and pathophysiological processes and affects disease progression in the pancreas are not yet known. AIM: To evaluate the role of LAT1 in the development of and recovery from AP. METHODS: AP was induced with caerulein (cae) injections in female and male mice expressing LAT1 or after its knockout (LAT1 Cre/LoxP). The development of the initial AP injury and its recovery were followed for seven days after cae injections by daily measuring body weight, assessing microscopical tissue architecture, mRNA and protein expression, protein synthesis, and enzyme activity levels, as well as by testing the recruitment of immune cells by FACS and ELISA. RESULTS: The initial injury, evaluated by measurements of plasma amylase, lipase, and trypsin activity, as well as the gene expression of dedifferentiation markers, did not differ between the groups. However, early metabolic adaptations that support regeneration at later stages were blunted in LAT1 knockout mice. Especially in females, we observed less mTOR reactivation and dysfunctional autophagy. The later regeneration phase was clearly delayed in female LAT1 knockout mice, which did not regain normal expression of the pancreas-specific differentiation markers recombining binding protein suppressor of hairless-like protein (rbpjl) and basic helix-loop-helix family member A15 (mist1). Amylase mRNA and protein levels remained lower, and, strikingly, female LAT1 knockout mice presented signs of fibrosis lasting until day seven. In contrast, pancreas morphology had returned to normal in wild-type littermates. CONCLUSION: LAT1 supports the regeneration of acinar cells after AP. Female mice lacking LAT1 exhibited more pronounced alterations than male mice, indicating a sexual dimorphism of amino acid metabolism.


Asunto(s)
Ceruletida , Pancreatitis , Enfermedad Aguda , Amilasas , Animales , Ceruletida/toxicidad , Femenino , Transportador de Aminoácidos Neutros Grandes 1/genética , Masculino , Mamíferos/genética , Ratones , Ratones Noqueados , Páncreas/metabolismo , Pancreatitis/inducido químicamente , Pancreatitis/genética , Pancreatitis/metabolismo , ARN Mensajero , Serina-Treonina Quinasas TOR
2.
Int J Mol Sci ; 22(1)2021 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-33466458

RESUMEN

The thyroid gland is both a thyroid hormone (TH) generating as well as a TH responsive organ. It is hence crucial that cathepsin-mediated proteolytic cleavage of the precursor thyroglobulin is regulated and integrated with the subsequent export of TH into the blood circulation, which is enabled by TH transporters such as monocarboxylate transporters Mct8 and Mct10. Previously, we showed that cathepsin K-deficient mice exhibit the phenomenon of functional compensation through cathepsin L upregulation, which is independent of the canonical hypothalamus-pituitary-thyroid axis, thus, due to auto-regulation. Since these animals also feature enhanced Mct8 expression, we aimed to understand if TH transporters are part of the thyroid auto-regulatory mechanisms. Therefore, we analyzed phenotypic differences in thyroid function arising from combined cathepsin K and TH transporter deficiencies, i.e., in Ctsk-/-/Mct10-/-, Ctsk-/-/Mct8-/y, and Ctsk-/-/Mct8-/y/Mct10-/-. Despite the impaired TH export, thyroglobulin degradation was enhanced in the mice lacking Mct8, particularly in the triple-deficient genotype, due to increased cathepsin amounts and enhanced cysteine peptidase activities, leading to ongoing thyroglobulin proteolysis for TH liberation, eventually causing self-thyrotoxic thyroid states. The increased cathepsin amounts were a consequence of autophagy-mediated lysosomal biogenesis that is possibly triggered due to the stress accompanying intrathyroidal TH accumulation, in particular in the Ctsk-/-/Mct8-/y/Mct10-/- animals. Collectively, our data points to the notion that the absence of cathepsin K and Mct8 leads to excessive thyroglobulin degradation and TH liberation in a non-classical pathway of thyroid auto-regulation.


Asunto(s)
Autofagia/fisiología , Catepsina K/metabolismo , Transportadores de Ácidos Monocarboxílicos/metabolismo , Simportadores/metabolismo , Tiroglobulina/metabolismo , Glándula Tiroides/metabolismo , Hormonas Tiroideas/metabolismo , Animales , Transporte Biológico , Catepsina L/metabolismo , Hipotálamo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Hipófisis/metabolismo
3.
PLoS One ; 15(5): e0233863, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32470053

RESUMEN

Adaptive regulation of epithelial transporters to nutrient intake is essential to decrease energy costs of their synthesis and maintenance, however such regulation is understudied. Previously we demonstrated that the transport function of the basolateral amino acid uniporter LAT4 (Slc43a2) is increased by dephosphorylation of serine 274 (S274) and nearly abolished by dephosphorylation of serine 297 (S297) when expressed in Xenopus oocytes. Phosphorylation changes in the jejunum of food-entrained mice suggested an increase in LAT4 transport function during food expectation. Thus, we investigated further how phosphorylation, expression and localization of mouse intestinal LAT4 respond to food-entrained diurnal rhythm and dietary protein content. In mice entrained with 18% protein diet, LAT4 mRNA was not submitted to diurnal regulation, unlike mRNAs of luminal symporters and antiporters. Only in duodenum, LAT4 protein expression increased during food intake. Concurrently, S274 phosphorylation was decreased in all three small intestinal segments, whereas S297 phosphorylation was increased only in jejunum. Interestingly, during food intake, S274 phosphorylation was nearly absent in ileum and accompanied by strong phosphorylation of mTORC1 target S6. Entraining mice with 8% protein diet provoked a shift in jejunal LAT4 localization from the cell surface to intracellular stores and increased S274 phosphorylation in both jejunum and ileum during food anticipation, suggesting decreased transport function. In contrast, 40% dietary protein content led to increased LAT4 expression in jejunum and its internalization in ileum. Ex vivo treatments of isolated intestinal villi fraction demonstrated that S274 phosphorylation was stimulated by protein kinase A. Rapamycin-sensitive insulin treatment and amino acids increased S297 phosphorylation, suggesting that the response to food intake might be regulated via the insulin-mTORC1 pathway. Ghrelin, an oscillating orexigenic hormone, did not affect phosphorylation of intestinal LAT4. Overall, we show that phosphorylation, expression and localization of intestinal mouse LAT4 responds to diurnal and dietary stimuli in location-specific manner.


Asunto(s)
Sistema de Transporte de Aminoácidos L/metabolismo , Sistema de Transporte de Aminoácidos y+/metabolismo , Ritmo Circadiano , Proteínas en la Dieta/farmacología , Alimentos , Intestinos/fisiología , Aminoácidos/metabolismo , Animales , Antiportadores/metabolismo , Ritmo Circadiano/efectos de los fármacos , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Ghrelina/administración & dosificación , Ghrelina/farmacología , Insulina/metabolismo , Intestino Delgado/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones Endogámicos C57BL , Microvellosidades/efectos de los fármacos , Microvellosidades/metabolismo , Fosforilación/efectos de los fármacos , Fosfoserina/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Fracciones Subcelulares/metabolismo , Simportadores/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
4.
Fluids Barriers CNS ; 17(1): 17, 2020 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-32046769

RESUMEN

BACKGROUND: Cerebrospinal fluid (CSF) is mainly produced by the choroid plexus (CP) located in brain ventricles. Although derived from blood plasma, it is nearly protein-free (~ 250-fold less) and contains about 2-20-fold less free amino acids, with the exception of glutamine (Gln) which is nearly equal. The aim of this study was to determine which amino acid transporters are expressed in mouse CP epithelium in order to gain understanding about how this barrier maintains the observed amino acid concentration gradient. METHODS: Expression of amino acid transporters was assessed in isolated choroid plexuses (CPs) by qRT-PCR followed by localization studies using immunofluorescence with specific antibodies. The impact of LAT2 (Slc7a8) antiporter deletion on CSF amino acids was determined. RESULTS: The purity of isolated choroid plexuses was tested on the mRNA level using specific markers, in particular transthyretin (Ttr) that was enriched 330-fold in CP compared to cerebral tissue. In a first experimental round, 14 out of 32 Slc amino acid transporters tested on the mRNA level by qPCR were selected for further investigation. Out of these, five were considered highly expressed, SNAT1 (Slc38a1), SNAT3 (Slc38a3), LAT2 (Slc7a8), ASC1 (Slc7a10) and SIT1 (Slc6a20b). Three of them were visualized by immunofluorescence: SNAT1 (Slc38a1), a neutral amino acid-Na+ symporter, found at the blood side basolateral membrane of CP epithelium, while SNAT3 (Slc38a3), an amino acid-Na+ symporter and H+ antiporter, as well as LAT2 (Slc7a8), a neutral amino acid antiporter, were localized at the CSF-facing luminal membrane. In a LAT2 knock-out mouse model, CSF Gln was unchanged, whereas other amino acids normally 2-20-fold lower than in plasma, were increased, in particular the LAT2 uptake substrates leucine (Leu), valine (Val) and tryptophan (Trp) and some other amino acids such as glutamate (Glu), glycine (Gly) and proline (Pro). CONCLUSION: These results suggest that Gln is actively transported by SNAT1 from the blood into CP epithelial cells and then released luminally into CSF via SNAT3 and LAT2. Its efflux via LAT2 may drive the reuptake from the CSF of essential amino acid substrates of this antiporter and thereby participates to maintaining the amino acid gradient between plasma and CSF.


Asunto(s)
Sistema de Transporte de Aminoácidos y+/metabolismo , Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Aminoácidos/líquido cefalorraquídeo , Células Epiteliales/metabolismo , Cadenas Ligeras de la Proteína-1 Reguladora de Fusión/metabolismo , Homeostasis , Sistema de Transporte de Aminoácidos y+/genética , Animales , Transporte Biológico/genética , Transporte Biológico/fisiología , Células Cultivadas , Plexo Coroideo/metabolismo , Femenino , Cadenas Ligeras de la Proteína-1 Reguladora de Fusión/genética , Ácido Glutámico/metabolismo , Homeostasis/fisiología , Masculino , Ratones Noqueados , Prealbúmina/metabolismo
5.
FASEB J ; 32(10): 5520-5531, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29718707

RESUMEN

Type-II l-arginine:ureahydrolase, arginase-II (Arg-II), is abundantly expressed in the kidney. The physiologic role played by Arg-II in the kidney remains unknown. Herein, we report that in mice that are deficient in Arg-II (Arg-II-/-), total and membrane-associated aquaporin-2 (AQP2) protein levels were significantly higher compared with wild-type (WT) controls. Water deprivation enhanced Arg-II expression, AQP2 levels, and membrane association in collecting ducts. Effects of water deprivation on AQP2 were stronger in Arg-II-/- mice than in WT mice. Accordingly, a decrease in urine volume and an increase in urine osmolality under water deprivation were more pronounced in Arg-II-/- mice than in WT mice, which correlated with a weaker increase in plasma osmolality in Arg-II-/- mice. There was no difference in vasopressin release under water deprivation conditions between either genotype of mice. Although total AQP2 and phosphorylated AQP2-S256 levels (mediated by PKA) in kidneys under water deprivation conditions were significantly higher in Arg-II-/- mice compared with WT animals, there is no difference in the ratio of AQP2-S256:AQP2. In cultured mouse collecting duct principal mCCDcl1 cells, expression of both Arg-II and AQP2 were enhanced by the vasopressin type 2 receptor agonist, desamino- d-arginine vasopressin (dDAVP). Silencing Arg-II enhanced the expression and membrane association of AQP2 by dDAVP without influencing cAMP levels. In conclusion, in vivo and in vitro experiments demonstrate that Arg-II negatively regulates AQP2 and the urine-concentrating capability in kidneys via a mechanism that is not associated with the modulation of the cAMP pathway.-Huang, J., Montani, J.-P., Verrey, F., Feraille, E., Ming, X.-F., Yang, Z. Arginase-II negatively regulates renal aquaporin-2 and water reabsorption.


Asunto(s)
Acuaporina 2/metabolismo , Arginasa/metabolismo , Túbulos Renales Colectores/metabolismo , Agua/metabolismo , Animales , Acuaporina 2/genética , Arginasa/genética , Arginina Vasopresina/farmacología , Línea Celular , AMP Cíclico/genética , AMP Cíclico/metabolismo , Túbulos Renales Colectores/citología , Ratones , Ratones Noqueados , Receptores de Vasopresinas/agonistas , Receptores de Vasopresinas/genética , Receptores de Vasopresinas/metabolismo
6.
Sci Rep ; 8(1): 5629, 2018 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-29618784

RESUMEN

Expression of the glutamine transporter SNAT3 increases in kidney during metabolic acidosis, suggesting a role during ammoniagenesis. Microarray analysis of Nrf2 knock-out (KO) mouse kidney identified Snat3 as the most significantly down-regulated transcript compared to wild-type (WT). We hypothesized that in the absence of NRF2 the kidney would be unable to induce SNAT3 under conditions of metabolic acidosis and therefore reduce the availability of glutamine for ammoniagenesis. Metabolic acidosis was induced for 7 days in WT and Nrf2 KO mice. Nrf2 KO mice failed to induce Snat3 mRNA and protein expression during metabolic acidosis. However, there were no differences in blood pH, bicarbonate, pCO2, chloride and calcium or urinary pH, ammonium and phosphate levels. Normal induction of ammoniagenic enzymes was observed whereas several amino acid transporters showed differential regulation. Moreover, Nrf2 KO mice during acidosis showed increased expression of renal markers of oxidative stress and injury and NRF2 activity was increased during metabolic acidosis in WT kidney. We conclude that NRF2 is required to adapt the levels of SNAT3 in response to metabolic acidosis. In the absence of NRF2 and SNAT3, the kidney does not have any major acid handling defect; however, increased oxidative stress and renal injury may occur.


Asunto(s)
Acidosis/fisiopatología , Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Túbulos Renales/patología , Factor 2 Relacionado con NF-E2/fisiología , Sistemas de Transporte de Aminoácidos Neutros/genética , Aminoácidos/análisis , Animales , Glutatión/metabolismo , Túbulos Renales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
7.
J Am Heart Assoc ; 7(5)2018 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-29478971

RESUMEN

BACKGROUND: Uninephrectomy (UNX) is performed for various reasons, including kidney cancer or donation. Kidneys being the main site of l-arginine production in the body, we tested whether UNX mediated kidney mass reduction impacts l-arginine metabolism and thereby nitric oxide production and blood pressure regulation in mice. METHODS AND RESULTS: In a first series of experiments, we observed a significant increase in arterial blood pressure 8 days post-UNX in female and not in male mice. Further experimental series were performed in female mice, and the blood pressure increase was confirmed by telemetry. l-citrulline, that is used in the kidney to produce l-arginine, was elevated post-UNX as was also asymmetric dimethylarginine, an inhibitor of nitric oxide synthase that competes with l-arginine and is a marker for renal failure. Interestingly, the UNX-induced blood pressure increase was prevented by supplementation of the diet with 5% of the l-arginine precursor, l-citrulline. Because l-arginine is metabolized in the kidney and other peripheral tissues by arginase-2, we tested whether the lack of this metabolic pathway also compensates for decreased l-arginine production in the kidney and/or for local nitric oxide synthase inhibition and consecutive blood pressure increase. Indeed, upon uninephrectomy, arginase-2 knockout mice (Arg-2-/-) neither displayed an increase in asymmetric dimethylarginine and l-citrulline plasma levels nor a significant increase in blood pressure. CONCLUSIONS: UNX leads to a small increase in blood pressure that is prevented by l-citrulline supplementation or arginase deficiency, 2 measures that appear to compensate for the impact of kidney mass reduction on l-arginine metabolism.


Asunto(s)
Arginina/metabolismo , Presión Sanguínea , Riñón/cirugía , Nefrectomía/efectos adversos , Animales , Arginasa/genética , Arginasa/metabolismo , Arginina/análogos & derivados , Arginina/sangre , Presión Sanguínea/efectos de los fármacos , Citrulina/administración & dosificación , Citrulina/sangre , Femenino , Riñón/metabolismo , Riñón/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico/metabolismo , Tamaño de los Órganos
8.
Nat Commun ; 6: 7250, 2015 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-25998567

RESUMEN

Mammalian target of rapamycin 1 (mTORC1), a master regulator of cellular growth, is activated downstream of growth factors, energy signalling and intracellular essential amino acids (EAAs) such as Leu. mTORC1 activation occurs at the lysosomal membrane, and involves V-ATPase stimulation by intra-lysosomal EAA (inside-out activation), leading to activation of the Ragulator, RagA/B-GTP and mTORC1 via Rheb-GTP. How Leu enters the lysosomes is unknown. Here we identified the lysosomal protein LAPTM4b as a binding partner for the Leu transporter, LAT1-4F2hc (SLC7A5-SLAC3A2). We show that LAPTM4b recruits LAT1-4F2hc to lysosomes, leading to uptake of Leu into lysosomes, and is required for mTORC1 activation via V-ATPase following EAA or Leu stimulation. These results demonstrate a functional Leu transporter at the lysosome, and help explain the inside-out lysosomal activation of mTORC1 by Leu/EAA.


Asunto(s)
Transportador de Aminoácidos Neutros Grandes 1/metabolismo , Leucina/metabolismo , Lisosomas/metabolismo , Proteínas de la Membrana/metabolismo , Complejos Multiproteicos/metabolismo , Proteínas Oncogénicas/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Células HeLa , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina , ATPasas de Translocación de Protón Vacuolares/metabolismo
9.
Am J Physiol Regul Integr Comp Physiol ; 307(7): R869-78, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25100072

RESUMEN

Diabetes mellitus is a disease characterized by elevated blood glucose levels and represents a worldwide health issue. Postprandial hyperglycemia is considered a major predictor of diabetic complications, and its reduction represents a specific treatment target in Type 1 and 2 diabetes. Since postprandial glucose excursions depend to a large extent on gastric secretion and emptying, amylin and glucagon-like peptide 1 analogs are prescribed to reduce them. Although gastric function is considered mainly sensitive to ingested calories, its chemospecificity is not well understood. To identify ingestible nutrients reducing postprandial hyperglycemia, we applied intragastrically more than 40 individual nutrients at an isomolar dose to rats and quantified their impact on gastric secretion and emptying using a novel in vivo computed tomography imaging method. We identified l-tryptophan, l-arginine, l-cysteine, and l-lysine as the most potent modulators with effective strength comparable to a supraphysiological dose of amylin. Importantly, all identified candidates reduced postprandial glucose excursion within an oral glucose tolerance test in healthy and diabetic rats. This clinical beneficial effect originated predominantly from their impact on gastric function, as none of the candidates altered plasma glucose concentrations induced by intraperitoneal or intraduodenal glucose tolerance tests. Overall, these data demonstrate a remarkable chemospecificity of stomach function, unveil a strong role of the stomach for glycemic control and identifies nutrients with antidiabetic potential.


Asunto(s)
Glucemia/metabolismo , Diabetes Mellitus Experimental/metabolismo , Vaciamiento Gástrico/fisiología , Hiperglucemia/metabolismo , Hipoglucemiantes/farmacología , Animales , Diabetes Mellitus Experimental/tratamiento farmacológico , Modelos Animales de Enfermedad , Alimentos , Glucagón/sangre , Insulina/metabolismo , Masculino , Periodo Posprandial/fisiología , Ratas Wistar
10.
PLoS One ; 9(5): e96823, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24797296

RESUMEN

Gene expression and cell growth rely on the intracellular concentration of amino acids, which in metazoans depends on extracellular amino acid availability and transmembrane transport. To investigate the impact of extracellular amino acid concentrations on the expression of a concentrative amino acid transporter, we overexpressed the main kidney proximal tubule luminal neutral amino acid transporter B0AT1-collectrin (SLC6A19-TMEM27) in MDCK cell epithelia. Exogenously expressed proteins co-localized at the luminal membrane and mediated neutral amino acid uptake. However, the transgenes were lost over few cell culture passages. In contrast, the expression of a control transgene remained stable. To test whether this loss was due to inappropriately high amino acid uptake, freshly transduced MDCK cell lines were cultivated either with physiological amounts of amino acids or with the high concentration found in standard cell culture media. Expression of exogenous transporters was unaffected by physiological amino acid concentration in the media. Interestingly, mycoplasma infection resulted in a significant increase in transgene expression and correlated with the rapid metabolism of L-arginine. However, L-arginine metabolites were shown to play no role in transgene expression. In contrast, activation of the GCN2 pathway revealed by an increase in eIF2α phosphorylation may trigger transgene derepression. Taken together, high extracellular amino acid concentration provided by cell culture media appears to inhibit the constitutive expression of concentrative amino acid transporters whereas L-arginine depletion by mycoplasma induces the expression of transgenes possibly via stimulation of the GCN2 pathway.


Asunto(s)
Sistemas de Transporte de Aminoácidos Neutros/genética , Aminoácidos/metabolismo , Regulación de la Expresión Génica , Glicoproteínas de Membrana/genética , Transgenes , Animales , Arginina/metabolismo , Medios de Cultivo , Perros , Células Epiteliales/metabolismo , Técnicas Genéticas , Células HEK293 , Humanos , Riñón/metabolismo , Células de Riñón Canino Madin Darby , Mycoplasma/metabolismo , Fosforilación , Reacción en Cadena en Tiempo Real de la Polimerasa
11.
J Physiol ; 590(24): 6413-24, 2012 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-23045339

RESUMEN

The uniporter TAT1 (Slc16a10) mediates the facilitated diffusion of aromatic amino acids (AAAs) across basolateral membranes of kidney, small intestine and liver epithelial cells, and across the plasma membrane of non-epithelial cells like skeletal myocytes. Its role for body AA homeostasis has now been investigated using newly generated TAT1 (Slc16a10) defective mice (tat1(-/-)). These mice grow and reproduce normally, show no gross phenotype and no obvious neurological defect. Histological analysis did not reveal abnormalities and there is no compensatory change in any tested AA transporter mRNA. TAT1 null mice, however, display increased plasma, muscle and kidney AAA concentration under both normal and high protein diet, although this concentration remains normal in the liver. A major aromatic aminoaciduria and a smaller urinary loss of all substrates additionally transported by l-type AA antiporter Lat2-4F2hc (Slc7a8) were revealed under a high protein diet. This suggests an epithelial transport defect as also shown by the accumulation of intravenously injected (123)I-2-I-l-Phe in kidney and l-[(3)H]Phe in ex vivo everted gut sac enterocytes. Taken together, these data indicate that the uniporter TAT1 is required to equilibrate the concentration of AAAs across specific membranes. For instance, it enables hepatocytes to function as a sink that controls the extracellular AAAs concentration. Additionally, it facilitates the release of AAAs across the basolateral membrane of small intestine and proximal kidney tubule epithelial cells, thereby allowing the efflux of other neutral AAs presumably via Lat2-4F2hc.


Asunto(s)
Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Aminoácidos Aromáticos/metabolismo , Proteínas en la Dieta/metabolismo , Sistema de Transporte de Aminoácidos y+/metabolismo , Sistemas de Transporte de Aminoácidos Neutros/deficiencia , Sistemas de Transporte de Aminoácidos Neutros/genética , Aminoácidos Aromáticos/sangre , Animales , Proteínas en la Dieta/sangre , Células Epiteliales/metabolismo , Cadena Pesada de la Proteína-1 Reguladora de Fusión/metabolismo , Cadenas Ligeras de la Proteína-1 Reguladora de Fusión/metabolismo , Genotipo , Homeostasis , Intestino Delgado/metabolismo , Riñón/metabolismo , Hígado/metabolismo , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Esquelético/metabolismo , Fenotipo , ARN Mensajero/metabolismo
12.
Am J Physiol Renal Physiol ; 301(1): F189-96, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21478478

RESUMEN

Regulation of the epithelial Na(+) channel (ENaC) by ubiquitylation is controlled by the activity of two counteracting enzymes, the E3 ubiquitin-protein ligase Nedd4-2 (mouse ortholog of human Nedd4L) and the ubiquitin-specific protease Usp2-45. Previously, Usp2-45 was shown to decrease ubiquitylation and to increase surface function of ENaC in Xenopus laevis oocytes, whereas the splice variant Usp2-69, which has a different N-terminal domain, was inactive toward ENaC. It is shown here that the catalytic core of Usp2 lacking the N-terminal domain has a reduced ability relative to Usp2-45 to enhance ENaC activity in Xenopus oocytes. In contrast, its catalytic activity toward the artificial substrate ubiquitin-AMC is fully maintained. The interaction of Usp2-45 with ENaC exogenously expressed in HEK293 cells was tested by coimmunoprecipitation. The data indicate that different combinations of ENaC subunits, as well as the α-ENaC cytoplasmic N-terminal but not C-terminal domain, coprecipitate with Usp2-45. This interaction is decreased but not abolished when the cytoplasmic ubiquitylation sites of ENaC are mutated. Importantly, coimmunoprecipitation in HEK293 cells and GST pull-down of purified recombinant proteins show that both the catalytic domain and the N-terminal tail of Usp2-45 physically interact with the HECT domain of Nedd4-2. Taken together, the data support the conclusion that Usp2-45 action on ENaC is promoted by various interactions, including through binding to Nedd4-2 that is suggested to position Usp2-45 favorably for ENaC deubiquitylation.


Asunto(s)
Endopeptidasas/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Canales Epiteliales de Sodio/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Western Blotting , Catálisis , ADN/genética , Endopeptidasas/biosíntesis , Endopeptidasas/genética , Complejos de Clasificación Endosomal Requeridos para el Transporte/biosíntesis , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Escherichia coli/metabolismo , Glutatión/metabolismo , Células HEK293 , Humanos , Inmunoprecipitación , Cinética , Ratones , Ubiquitina-Proteína Ligasas Nedd4 , Oocitos/metabolismo , Técnicas de Placa-Clamp , Unión Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Transfección , Ubiquitina Tiolesterasa , Ubiquitina-Proteína Ligasas/biosíntesis , Ubiquitina-Proteína Ligasas/genética , Proteínas de Xenopus , Xenopus laevis
13.
J Cereb Blood Flow Metab ; 29(9): 1491-502, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19491922

RESUMEN

Tight homeostatic control of brain amino acids (AA) depends on transport by solute carrier family proteins expressed by the blood-brain barrier (BBB) microvascular endothelial cells (BMEC). To characterize the mouse BMEC transcriptome and probe culture-induced changes, microarray analyses of platelet endothelial cell adhesion molecule-1-positive (PECAM1(+)) endothelial cells (ppMBMECs) were compared with primary MBMECs (pMBMEC) cultured in the presence or absence of glial cells and with b.End5 endothelioma cell line. Selected cell marker and AA transporter mRNA levels were further verified by reverse transcription real-time PCR. Regardless of glial coculture, expression of a large subset of genes was strongly altered by a brief culture step. This is consistent with the known dependence of BMECs on in vivo interactions to maintain physiologic functions, for example, tight barrier formation, and their consequent dedifferentiation in culture. Seven (4F2hc, Lat1, Taut, Snat3, Snat5, Xpct, and Cat1) of nine AA transporter mRNAs highly expressed in freshly isolated ppMBMECs were strongly downregulated for all cultures and two (Snat2 and Eaat3) were variably regulated. In contrast, five AA transporter mRNAs with low expression in ppMBMECs, including y(+)Lat2, xCT, and Snat1, were upregulated by culture. We hypothesized that the AA transporters highly expressed in ppMBMECs and downregulated in culture have a major in vivo function for BBB transendothelial transport.


Asunto(s)
Sistemas de Transporte de Aminoácidos/metabolismo , Barrera Hematoencefálica/fisiología , Técnicas de Cultivo de Célula , Perfilación de la Expresión Génica , Sistemas de Transporte de Aminoácidos/genética , Animales , Biomarcadores/metabolismo , Células Cultivadas , Circulación Cerebrovascular , Células Endoteliales/citología , Células Endoteliales/fisiología , Femenino , Homeostasis , Ratones , Ratones Endogámicos C57BL , Análisis por Micromatrices , Microcirculación , Datos de Secuencia Molecular , Neuroglía/citología , Neuroglía/metabolismo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/genética , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo
14.
Biochemistry ; 48(25): 5801-12, 2009 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-19425588

RESUMEN

The three-component AcrA/AcrB/TolC efflux system of Escherichia coli catalyzes the proton motive force-driven extrusion of a variety of cytotoxic compounds. The inner membrane pump component AcrB belongs to the resistance nodulation and cell division (RND) superfamily and is responsible for drug specificity and energy transduction of the entire tripartite efflux system. Systematic mutational analysis of titratable and polar membrane-located amino acids revealed four residues, D407, D408, K940, and, R971, to be of prime importance for AcrB function. Using matrix-assisted laser desorption ionization time-of-flight (MALDI-TOF) mass spectrometry, D408 was shown to specifically react with dicyclohexylcarbodiimide (DCCD) in a pH-dependent manner. The apparent pK(a) of D408 of 7.4 would enable binding and release of protons under physiological conditions. In contrast to other secondary transporters, D408 was not protected from carbodiimide modification in the presence of drugs, which supports the notion of spatially separated transport pathways for drugs and protons. This study provides evidence for a substantial role of membrane-located carboxylates as a central element of the proton translocation pathway in AcrB and other members of the RND superfamily.


Asunto(s)
Ácido Aspártico/genética , Ácido Aspártico/fisiología , Carbodiimidas/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/fisiología , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/fisiología , ATPasas de Translocación de Protón/genética , ATPasas de Translocación de Protón/fisiología , Secuencia de Aminoácidos , Ácido Aspártico/metabolismo , Proteínas Portadoras/metabolismo , Proteínas Portadoras/fisiología , División Celular/genética , Proteínas de Escherichia coli/metabolismo , Datos de Secuencia Molecular , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Familia de Multigenes , Mutagénesis Sitio-Dirigida/métodos , ATPasas de Translocación de Protón/metabolismo , Transducción de Señal/genética , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Coloración y Etiquetado/métodos
15.
Curr Drug Targets ; 9(9): 729-49, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18781920

RESUMEN

Antimicrobial resistance of human pathogenic bacteria is an emerging problem for global public health. This resistance is often associated with the overproduction of membrane transport proteins that are capable to pump chemotherapeutics, antibiotics, detergents, dyes and organic solvents out of the cell. In Gram-negative bacteria such as Escherichia coli and Pseudomonas aeruginosa, tripartite multidrug efflux systems extrude a large variety of cytotoxic substances from the cell membrane directly into the medium bypassing the periplasm and the outer membrane. In E. coli, the tripartite efflux system AcrA/AcrB/TolC is the pump in charge of the efflux of multiple antibiotics, dyes, bile salts and detergents. The trimeric outer membrane factor (OMF) TolC forms a beta-barrel pore in the outer membrane and exhibits a long periplasmic alpha-helical conduit. The periplasmic membrane fusion protein (MFP) AcrA serves as a linker between TolC and the trimeric resistance nodulation cell division (RND) pump AcrB, located in the inner membrane acting as a proton/drug antiporter. The newly elucidated asymmetric structure of trimeric AcrB reveals three different monomer conformations representing consecutive states in a transport cycle. The monomers show tunnels with occlusions at different sites leading from the lateral side through the periplasmic porter (pore) domains towards the funnel of the trimer and TolC. The structural changes create a hydrophobic pocket in one monomer, which is not present in the other two monomers. Minocyclin and doxorubicin, both AcrB substrates, specifically bind to this pocket substantiating its role as drug binding pocket. The energy transduction from the proton motive force into drug efflux includes proton binding in (and release from) the transmembrane part. The conformational changes observed within a triad of essential, titratable residues (Asp407/Asp408/Lys940) residing in the hydrophobic transmembrane domain appear to be transduced by transmembrane helix 8 and associated with the conformational changes seen in the periplasmic domain. From the asymmetric structure a possible peristaltic pump transport mechanism based on a functional rotation of the AcrB trimer has been postulated. The novel transport model merges Jardetzky's alternate access pump mechanism with the rotating site catalysis of F(1)F(0) ATPase and suggests a working hypothesis for the transport mechanism of RND transporters in general.


Asunto(s)
Antibacterianos/metabolismo , Farmacorresistencia Bacteriana Múltiple/fisiología , Proteínas de Escherichia coli/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Quinasas/metabolismo , Transporte Biológico , Membrana Celular/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Modelos Moleculares , Unión Proteica , Conformación Proteica , ATPasas de Translocación de Protón/metabolismo
16.
Nat Struct Mol Biol ; 15(2): 199-205, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18223659

RESUMEN

The AcrA-AcrB-TolC complex is the major multidrug efflux pump in Escherichia coli. The asymmetric structure of the trimeric inner-membrane component AcrB implies functional rotation of the monomers and a peristaltic mode of drug efflux. This mechanism suggests the occurrence of conformational changes in the periplasmic pore domain through the movements of subdomains during cycling of the monomers through the different states loose (L), tight (T) and open (O). We introduced cysteines at the interfaces of potentially moving subdomains, leading to disulfide bond formation as quantified by alkylation of free cysteines and MALDI-TOF analysis. Inhibition of pump function as a result of cross-linking caused increased susceptibility to noxious compounds and reduction of N-phenylnaphthylamine efflux. Regain of function for impaired mutants was obtained upon exposure to the reducing agent DTT. The results support the presence of the asymmetric AcrB trimer in E. coli membranes and the functional rotation mechanism.


Asunto(s)
Disulfuros/química , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Escherichia coli/efectos de los fármacos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/química , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , 1-Naftilamina/análogos & derivados , 1-Naftilamina/metabolismo , Sustitución de Aminoácidos/genética , Antibacterianos/farmacología , Cisteína/genética , Ditiotreitol/farmacología , Electroforesis en Gel de Poliacrilamida , Escherichia coli/química , Escherichia coli/genética , Eliminación de Gen , Pruebas de Sensibilidad Microbiana , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Estructura Terciaria de Proteína , Sustancias Reductoras/farmacología
17.
Brain Res ; 1147: 25-33, 2007 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-17343835

RESUMEN

Amino acids are valuable tracers for brain tumor imaging with positron emission tomography (PET). In this study the transport of O-(2-[(18)F]fluoroethyl)-D-tyrosine (D-FET) across the blood-brain barrier (BBB) was studied with PET in anesthetized piglets and patients after subtotal resection of brain tumors and compared with O-(2-[(18)F]fluoroethyl)-L-tyrosine (L-FET) and 3-O-methyl-6-[(18)F]fluoro-L-DOPA (L-OMFD). In piglets, compartmental modeling of PET data was used to calculate the rate constants for the blood-brain (K(1)) and the brain-blood (k(2)) transfer of D-FET, L-FET and L-OMFD. In patients standardized uptake values (SUVs) were calculated in brain cortex and lesions. Additionally, affinity determinations on various amino acid transporters (LAT1, LAT2, PAT1, XPCT) were performed in vitro using unlabeled D-FET, L-FET and L-OMFD. The initial brain uptake of D-FET in piglets was more than two-fold higher than that of l-FET, whereas the initial brain uptake of D-FET in patients was similar to that of L-FET. Calculation of K(1) and k(2) from the brain uptake curves and the plasma input data in piglets revealed about 4- and 2-fold higher values for D-FET compared to L-FET and L-OMFD, respectively. The distribution volume of D-FET in the piglet brain was slightly higher than that of L-FET as it was also found for most other organs. In brain tumor patients, initial D-FET uptake in the brain was similar to that of L-FET but showed faster tracer washout. L-FET uptake remained rather constant and provided a better delineation of residual tumor than D-FET. In conclusion, our data indicate considerable differences of stereoselective amino acid transport at the BBB in different species. Therefore, the results from animal experiments concerning BBB amino acid transport may not be transferable to humans.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Neoplasias Encefálicas/diagnóstico por imagen , Encéfalo/diagnóstico por imagen , Radioisótopos de Flúor/farmacocinética , Tirosina/análogos & derivados , Adulto , Animales , Transporte Biológico , Encéfalo/metabolismo , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/cirugía , Medios de Contraste/farmacocinética , Dihidroxifenilalanina/análogos & derivados , Dihidroxifenilalanina/farmacocinética , Femenino , Humanos , Masculino , Persona de Mediana Edad , Tomografía de Emisión de Positrones , Estereoisomerismo , Porcinos , Distribución Tisular , Tirosina/farmacocinética
19.
Pflugers Arch ; 451(6): 738-48, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16308696

RESUMEN

Gastric acid secretion is regulated by a variety of stimuli, in particular histamine and acetyl choline. In addition, dietary factors such as the acute intake of a protein-rich diet and the subsequent increase in serum amino acids can stimulate gastric acid secretion only through partially characterized pathways. Recently, we described in mouse stomach parietal cells the expression of the system L heteromeric amino acid transporter comprised of the LAT2-4F2hc dimer. Here we address the potential role of the system L amino acid transporter in gastric acid secretion by parietal cells in freshly isolated rat gastric glands. RT-PCR, western blotting and immunohistochemistry confirmed the expression of 4F2-LAT2 amino acid transporters in rat parietal cells. In addition, mRNA was detected for the B(0)AT1, ASCT2, and ATB(0+) amino acid transporters. Intracellular pH measurements in parietal cells showed histamine-induced and omeprazole-sensitive H+-extrusion which was enhanced by about 50% in the presence of glutamine or cysteine (1 mM), two substrates of system L amino acid transporters. BCH, a non-metabolizable substrate and a competitive inhibitor of system L amino acid transport, abolished the stimulation of acid secretion by glutamine or cysteine suggesting that this stimulation required the uptake of amino acids by system L. In the absence of histamine glutamine also stimulated H+-extrusion, whereas glutamate did not. Also, phenylalanine was effective in stimulating H+/K+-ATPase activity. Glutamine did not increase intracellular Ca2+ levels indicating that it did not act via the recently described amino acid modulated Ca2+-sensing receptor. These data suggest a novel role for heterodimeric amino acid transporters and may elucidate a pathway by which protein-rich diets stimulate gastric acid secretion.


Asunto(s)
Sistemas de Transporte de Aminoácidos/metabolismo , Ácido Gástrico/metabolismo , Mucosa Gástrica/metabolismo , Potenciales de Acción , Sistemas de Transporte de Aminoácidos/genética , Aminoácidos/metabolismo , Aminoácidos Cíclicos/farmacología , Animales , Calcio/metabolismo , Cimetidina/farmacología , Inhibidores Enzimáticos/farmacología , ATPasa Intercambiadora de Hidrógeno-Potásio/metabolismo , Histamina/farmacología , Concentración de Iones de Hidrógeno , Masculino , Omeprazol/farmacología , Células Parietales Gástricas/fisiología , Inhibidores de la Bomba de Protones , Protones , Ratas , Ratas Wistar
20.
Biochem J ; 388(Pt 2): 435-43, 2005 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-15679469

RESUMEN

The heteromeric amino acid transporter glycoprotein subunits rBAT and 4F2hc (heavy chains) form, with different catalytic subunits (light chains), functional heterodimers that are covalently stabilized by a disulphide bridge. Whereas rBAT associates with b(0,+)AT to form the cystine and cationic amino acid transporter defective in cystinuria, 4F2hc associates with other homologous light chains, for instance with LAT1 to form a system L neutral amino acid transporter. To identify within the heavy chains the domain(s) involved in recognition of and functional interaction with partner light chains, chimaeric and truncated forms of rBAT and 4F2hc were co-expressed in Xenopus laevis oocytes with b(0,+)AT or LAT1. Heavy chain-light chain association was analysed by co-immunoprecipitation, and transport function was tested by tracer uptake experiments. The results indicate that the cytoplasmic tail and transmembrane domain of rBAT together play a dominant role in selective functional interaction with b(0,+)AT, whereas the extracellular domain of rBAT appears to facilitate specifically L-cystine uptake. For 4F2hc, functional interaction with LAT1 was mediated by the N-terminal part, comprising cytoplasmic tail, transmembrane segment and neck, even in the absence of the extracellular domain. Alternatively, functional association with LAT1 was also supported by the extracellular part of 4F2hc comprising neck and glycosidase-like domain linked to the complementary part of rBAT. In conclusion, the cytoplasmic tail and the transmembrane segment together play a determinant role for the functional interaction of rBAT with b(0,+)AT, whereas either cytoplasmic or extracellular glycosidase-like domains are dispensable for the functional interaction of 4F2hc with LAT1.


Asunto(s)
Sistemas de Transporte de Aminoácidos/fisiología , Glicoproteínas/fisiología , Sistemas de Transporte de Aminoácidos/química , Aminoácidos/metabolismo , Animales , Transporte Biológico Activo , Expresión Génica , Glicoproteínas/química , Oocitos , Conformación Proteica , Estructura Terciaria de Proteína , Subunidades de Proteína , Proteínas Recombinantes de Fusión , Relación Estructura-Actividad , Xenopus laevis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA