Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Free Radic Res ; 54(7): 525-534, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32873097

RESUMEN

Slc7a11 (xCT) and Slc3a1 (rBAT) are cystine uptake transporters that maintain intracellular concentrations of cysteine, the rate-limiting amino acid in glutathione synthesis. This study was conducted to first determine the tissue distribution of the two transporters in male and female mice. Because Slc3a1 was the primary cystine transporter in liver, its sex-divergent expression, ontogeny, diurnal rhythm and whether its mRNA expression is altered by transcription factors (AhR, CAR, PXR, PPARα, and Nrf2) was also investigated. Slc7a11 was expressed highest in brain and gonads. Slc3a1 was expressed highest in kidney and intestine, followed by liver. Duodenal and hepatic Slc3a1 was higher in females than males. Hepatic Slc3a1 was high during darkness and low during daytime. Hepatic Scl3a1 was lowest pre-birth, increased to near maximal levels at birth, decreased back to pre-birth levels between Days 3-10, and then returned to peak levels by Day 45. Except for CAR, activation of transcription factors did not increase hepatic mRNA expression of Slc3a1. Chemical activation of CAR significantly induced Slc3a1 1.4-fold in wild-type but not CAR-null mice. Slc3a1 mRNA was higher in livers of AhR- and Nrf2-null mice compared to wild-type mice. High doses of diquat but not acetaminophen induced Slc3a1, suggesting Slc3a1 may respond to oxidative stress but not necessarily to GSH depletion. Overall, Slc7a11 is mainly expressed in brain and gonads, whereas Slc3a1 is mainly expressed in kidney, small intestine and liver, and its hepatic expression is regulated by diurnal rhythm and certain xenobiotic treatments.


Asunto(s)
Sistemas de Transporte de Aminoácidos Básicos/metabolismo , Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Cistina/metabolismo , Sistemas de Transporte de Aminoácidos Básicos/biosíntesis , Sistemas de Transporte de Aminoácidos Básicos/genética , Sistemas de Transporte de Aminoácidos Neutros/biosíntesis , Sistemas de Transporte de Aminoácidos Neutros/genética , Animales , Femenino , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Estrés Oxidativo/fisiología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Distribución Tisular
2.
PLoS One ; 12(9): e0184845, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28915252

RESUMEN

Absorption of neutral amino acids across the luminal membrane of intestinal enterocytes is mediated by the broad neutral amino acid transporter B0AT1 (SLC6A19). Its intestinal expression depends on co-expression of the membrane-anchored peptidase angiotensin converting enzyme 2 (ACE2) and is additionally enhanced by aminopeptidase N (CD13). We investigated in this study the expression of B0AT1 and its auxiliary peptidases as well as its transport function along the rat small intestine. Additionally, we tested its possible short- and long-term regulation by dietary proteins and amino acids. We showed by immunofluorescence that B0AT1, ACE2 and CD13 co-localize on the luminal membrane of small intestinal villi and by Western blotting that their protein expression increases in distal direction. Furthermore, we observed an elevated transport activity of the neutral amino acid L-isoleucine during the nocturnal active phase compared to the inactive one. Gastric emptying was delayed by intragastric application of an amino acid cocktail but we observed no acute dietary regulation of B0AT1 protein expression and L-isoleucine transport. Investigation of the chronic dietary regulation of B0AT1, ACE2 and CD13 by different diets revealed an increased B0AT1 protein expression under amino acid-supplemented diet in the proximal section but not in the distal one and for ACE2 protein expression a reverse localization of the effect. Dietary regulation for CD13 protein expression was not as distinct as for the two other proteins. Ring uptake experiments showed a tendency for increased L-isoleucine uptake under amino acid-supplemented diet and in vivo L-isoleucine absorption was more efficient under high protein and amino acid-supplemented diet. Additionally, plasma levels of branched-chain amino acids were elevated under high protein and amino acid diet. Taken together, our experiments did not reveal an acute amino acid-induced regulation of B0AT1 but revealed a chronic dietary adaptation mainly restricted to the proximal segment of the small intestine.


Asunto(s)
Sistemas de Transporte de Aminoácidos Neutros/biosíntesis , Antígenos CD13/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Intestino Delgado/metabolismo , Isoleucina/farmacología , Peptidil-Dipeptidasa A/metabolismo , Enzima Convertidora de Angiotensina 2 , Animales , Suplementos Dietéticos , Masculino , Ratas , Ratas Wistar
3.
J Biosci Bioeng ; 123(4): 444-450, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28057466

RESUMEN

The alaE gene in Escherichia coli encodes an l-alanine exporter that catalyzes the active export of l-alanine using proton electrochemical potential. In our previous study, alaE expression was shown to increase in the presence of l-alanyl-l-alanine (Ala-Ala). In this study, the global regulator leucine-responsive regulatory protein (Lrp) was identified as an activator of the alaE gene. A promoter less ß-galactosidase gene was fused to an alaE upstream region (240 nucleotides). Cells that were lacZ-deficient and harbored this reporter plasmid showed significant induction of ß-galactosidase activity (approximately 17-fold) in the presence of 6 mM l-alanine, l-leucine, and Ala-Ala. However, a reporter plasmid possessing a smaller alaE upstream region (180 nucleotides) yielded transformants with strikingly low enzyme activity under the same conditions. In contrast, lrp-deficient cells showed almost no ß-galactosidase induction, indicating that Lrp positively regulates alaE expression. We next performed an electrophoretic mobility shift assay (EMSA) and a DNase I footprinting assay using purified hexahistidine-tagged Lrp (Lrp-His). Consequently, we found that Lrp-His binds to the alaE upstream region spanning nucleotide -161 to -83 with a physiologically relevant affinity (apparent KD, 288.7 ± 83.8 nM). Furthermore, the binding affinity of Lrp-His toward its cis-element was increased by l-alanine and l-leucine, but not by Ala-Ala and d-alanine. Based on these results, we concluded that the gene expression of the alaE is regulated by Lrp in response to intracellular levels of l-alanine, which eventually leads to intracellular homeostasis of l-alanine concentrations.


Asunto(s)
Alanina/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Regulación Bacteriana de la Expresión Génica , Proteína Reguladora de Respuesta a la Leucina/metabolismo , Alanina/farmacología , Sistemas de Transporte de Aminoácidos Neutros/biosíntesis , Sistemas de Transporte de Aminoácidos Neutros/genética , Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Secuencia de Bases , Huella de ADN , Desoxirribonucleasa I/metabolismo , Dipéptidos/metabolismo , Dipéptidos/farmacología , Ensayo de Cambio de Movilidad Electroforética , Escherichia coli/efectos de los fármacos , Proteínas de Escherichia coli/biosíntesis , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Genes Reporteros/genética , Leucina/metabolismo , Leucina/farmacología , Proteína Reguladora de Respuesta a la Leucina/deficiencia , Operón/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Secuencias Reguladoras de Ácidos Nucleicos/genética , Regulación hacia Arriba/efectos de los fármacos , beta-Galactosidasa/genética , beta-Galactosidasa/metabolismo
4.
Amino Acids ; 48(6): 1491-508, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26984322

RESUMEN

Gut apical amino acid (AA) transport activity is high at birth and during suckling, thus being essential to maintain luminal nutrient-dependent mucosal growth through providing AA as essential metabolic fuel, substrates and nutrient stimuli for cellular growth. Because system-B(0) Na(+)-neutral AA co-transporter (B(0)AT1, encoded by the SLC6A19 gene) plays a dominant role for apical uptake of large neutral AA including L-Gln, we hypothesized that high apical Na(+)-Gln co-transport activity, and B(0)AT1 (SLC6A19) in co-expression with angiotensin-converting enzyme 2 (ACE2) were expressed along the entire small intestinal crypt-villus axis in young animals via unique control mechanisms. Kinetics of Na(+)-Gln co-transport activity in the apical membrane vesicles, prepared from epithelial cells sequentially isolated along the jejunal crypt-villus axis from liquid formula-fed young pigs, were measured with the membrane potential being clamped to zero using thiocyanate. Apical maximal Na(+)-Gln co-transport activity was much higher (p < 0.05) in the upper villus cells than in the middle villus (by 29 %) and the crypt (by 30 %) cells, whereas Na(+)-Gln co-transport affinity was lower (p < 0.05) in the upper villus cells than in the middle villus and the crypt cells. The B(0)AT1 (SLC6A19) mRNA abundance was lower (p < 0.05) in the crypt (by 40-47 %) than in the villus cells. There were no significant differences in B(0)AT1 and ACE2 protein abundances on the apical membrane among the upper villus, the middle villus and the crypt cells. Our study suggests that piglet fast growth is associated with very high intestinal apical Na(+)-neutral AA uptake activities via abundantly co-expressing B(0)AT1 and ACE2 proteins in the apical membrane and by transcribing the B(0)AT1 (SLC6A19) gene in the epithelia along the entire crypt-villus axis.


Asunto(s)
Sistemas de Transporte de Aminoácidos Básicos/biosíntesis , Sistemas de Transporte de Aminoácidos Neutros/biosíntesis , Alimentación Animal , Regulación de la Expresión Génica , Mucosa Intestinal/metabolismo , Yeyuno/metabolismo , Peptidil-Dipeptidasa A/biosíntesis , Enzima Convertidora de Angiotensina 2 , Animales , Femenino , Masculino , Porcinos
5.
Biomed Res Int ; 2015: 593572, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26106611

RESUMEN

We used a target-centric strategy to identify transporter proteins upregulated in pancreatic ductal adenocarcinoma (PDAC) as potential targets for a functional imaging probe to complement existing anatomical imaging approaches. We performed transcriptomic profiling (microarray and RNASeq) on histologically confirmed primary PDAC tumors and normal pancreas tissue from 33 patients, including five patients whose tumors were not visible on computed tomography. Target expression was confirmed with immunohistochemistry on tissue microarrays from 94 PDAC patients. The best imaging target identified was SLC6A14 (a neutral and basic amino acid transporter). SLC6A14 was overexpressed at the transcriptional level in all patients and expressed at the protein level in 95% of PDAC tumors. Very little is known about the role of SLC6A14 in PDAC and our results demonstrate that this target merits further investigation as a candidate transporter for functional imaging of PDAC.


Asunto(s)
Adenocarcinoma/genética , Sistemas de Transporte de Aminoácidos Neutros/biosíntesis , Biomarcadores de Tumor/biosíntesis , Carcinoma Ductal Pancreático/genética , Adenocarcinoma/patología , Anciano , Sistemas de Transporte de Aminoácidos , Sistemas de Transporte de Aminoácidos Neutros/genética , Biomarcadores de Tumor/genética , Carcinoma Ductal Pancreático/patología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad
6.
Neurochem Int ; 88: 32-7, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25842041

RESUMEN

Hepatic encephalopathy (HE) is a neuropsychiatric disorder that occurs due to acute and chronic liver diseases, the hallmark of which is the increased levels of ammonia and subsequent alterations in glutamine synthesis, i.e. conditions associated with the pathophysiology of HE. Under physiological conditions, glutamine is fundamental for replenishment of the neurotransmitter pools of glutamate and GABA. The different isoforms of glutamine transporters play an important role in the transfer of this amino acid between astrocytes and neurons. A disturbance in the GABA biosynthetic pathways has been described in bile duct ligated (BDL) rats, a well characterized model of chronic HE. Considering that glutamine is important for GABA biosynthesis, altered glutamine transport and the subsequent glutamate/GABA-glutamine cycle efficacy might influence these pathways. Given this potential outcome, the aim of the present study was to investigate whether the expression of the glutamine transporters SAT1, SAT2, SN1 and SN2 would be affected in chronic HE. We verified that mRNA expression of the neuronal glutamine transporters SAT1 and SAT2 was found unaltered in the cerebral cortex of BDL rats. Similarly, no changes were found in the mRNA level for the astrocytic transporter SN1, whereas the gene expression of SN2 was increased by two-fold in animals with chronic HE. However, SN2 protein immuno-reactivity did not correspond with the increase in gene transcription since it remained unaltered. These data indicate that the expression of the glutamine transporter isoforms is unchanged during chronic HE, and thus likely not to participate in the pathological mechanisms related to the imbalance in the GABAergic neurotransmitter system observed in this neurologic condition.


Asunto(s)
Sistema de Transporte de Aminoácidos A/biosíntesis , Sistemas de Transporte de Aminoácidos Neutros/biosíntesis , Corteza Cerebral/metabolismo , Encefalopatía Hepática/metabolismo , Sistema de Transporte de Aminoácidos A/genética , Sistemas de Transporte de Aminoácidos Neutros/genética , Animales , Proteínas Portadoras/biosíntesis , Corteza Cerebral/patología , Enfermedad Crónica , Regulación de la Expresión Génica , Encefalopatía Hepática/genética , Encefalopatía Hepática/patología , Masculino , Isoformas de Proteínas/biosíntesis , Ratas , Ratas Wistar
7.
Cell Physiol Biochem ; 33(5): 1591-606, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24854847

RESUMEN

BACKGROUND: Despite homeostatic pH regulation, systemic and cellular pH changes take place and strongly influence metabolic processes. Transcription of the glutamine transporter SNAT3 (Slc38a3) for instance is highly up-regulated in the kidney during metabolic acidosis to provide glutamine for ammonia production. METHODS: Slc38a3 promoter activity and messenger RNA stability were measured in cultured cells in response to different extracellular pH values. RESULTS: Up-regulation of SNAT3 mRNA was mediated both by the stabilization of its mRNA and by the up-regulation of gene transcription. Stabilisation of the mRNA involved a pH-response element, while enhanced transcription made use of a second pH-sensitive Sp1 binding site in addition to a constitutive Sp1 binding site. Transcriptional regulation dominated the early response to acidosis, while mRNA stability was more important for chronic adaptation. Tissue-specific expression of SNAT3, by contrast, appeared to be controlled by promoter methylation and histone modifications. CONCLUSIONS: Regulation of SNAT3 gene expression by extracellular pH involves post-transcriptional and transcriptional mechanisms, the latter being distinct from the mechanisms that control the tissue-specific expression of the gene.


Asunto(s)
Acidosis/metabolismo , Sistemas de Transporte de Aminoácidos Neutros/biosíntesis , Sistemas de Transporte de Aminoácidos Neutros/genética , Acidosis/genética , Animales , Células Cultivadas , Células HEK293 , Células HeLa , Células Hep G2 , Humanos , Concentración de Iones de Hidrógeno , Ratones , Especificidad de Órganos/genética , Regiones Promotoras Genéticas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transcripción Genética/genética
8.
Nephrol Dial Transplant ; 29(3): 522-8, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23861466

RESUMEN

Cystinosis is an autosomal recessive metabolic disease that belongs to the family of lysosomal storage disorders. The defective gene is CTNS, which encodes the lysosomal cystine transporter, cystinosin. Cystine accumulates in all tissues and leads to organ damage including end-stage renal disease. In this review, we outline the studies that support that genetic rescue of cystinosis could be an achievable goal, even though cystinosis is a multi-compartmental disease and cystinosin an intracellular transmembrane protein. Using the mouse model of cystinosis, the Ctns(-/-) mice, we showed that transplanted hematopoietic stem cells (HSCs) were able to act as vehicles for the delivery of a functional Ctns gene to the different organs and led to the significant decrease of the tissue cystine content and tissue preservation. Ex vivo gene-modified Ctns(-/-) HSC transplantation using a lentiviral vector containing CTNS complementary DNA (cDNA) was also successful in the Ctns(-/-) mice and built the foundations for a clinical trial for autologous HSC transplantation for cystinosis. The capacity of HSCs for rescuing non-hematopoietic disease is controversial, and new insights into regenerative medicine could be gained from unraveling the underlying mechanism of action.


Asunto(s)
Sistemas de Transporte de Aminoácidos Neutros/genética , Cistinosis/terapia , Terapia Genética , Sistemas de Transporte de Aminoácidos Neutros/biosíntesis , Animales , Cistinosis/genética , Técnicas de Transferencia de Gen , Objetivos , Trasplante de Células Madre Hematopoyéticas , Humanos , Riñón/metabolismo , Riñón/patología , Ratones Noqueados
9.
J Neurosci ; 33(44): 17429-34, 2013 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-24174676

RESUMEN

The mechanisms by which the excitatory neurotransmitter glutamate is recycled at synapses are currently unknown. By examining the functional expression of plasma membrane transporters at presynaptic terminals, we aim to elucidate some of the mechanisms of glutamate recycling. Using whole-cell voltage-clamp recordings from rat calyx of Held presynaptic terminals, our data show, for the first time, that the glutamate precursor glutamine causes the direct activation of an electrogenic, sodium-dependent presynaptic transporter, which supplies glutamine for generation of presynaptic glutamate and helps sustain synaptic transmission. Interestingly, the functional expression of this transporter at the presynaptic plasma membrane is dynamically controlled by electrical activity of the terminal, indicating that uptake of neurotransmitter precursors is controlled by the demand at an individual terminal. Induction of the transporter current is calcium-dependent and inhibited by botulinum neurotoxin C, demonstrating the involvement of SNARE-dependent exocytosis in inserting transporters into the plasma membrane when the terminal is active. Conversely, inactivity of the presynaptic terminal results in removal of transporters via clathrin-mediated endocytosis. To investigate whether the presynaptic glutamine transporter supplies the precursor for generating the synaptically released glutamate, we measured miniature EPSCs to assess vesicular glutamate content. When the presynaptic glutamate pool was turned over by synaptic activity, inhibiting the presynaptic glutamine transporters with MeAIB reduced the miniature EPSC amplitude significantly. This demonstrates that presynaptic glutamine transport is centrally involved in the production of glutamate and assists in maintaining excitatory neurotransmission.


Asunto(s)
Sistemas de Transporte de Aminoácidos Neutros/biosíntesis , Tronco Encefálico/fisiología , Ácido Glutámico/fisiología , Glutamina/biosíntesis , Terminales Presinápticos/metabolismo , Sinapsis/metabolismo , Transmisión Sináptica/fisiología , Sistemas de Transporte de Aminoácidos Neutros/antagonistas & inhibidores , Sistemas de Transporte de Aminoácidos Neutros/fisiología , Animales , Transporte Biológico Activo/fisiología , Tronco Encefálico/metabolismo , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Femenino , Ácido Glutámico/metabolismo , Glutamina/metabolismo , Glutamina/fisiología , Masculino , Técnicas de Cultivo de Órganos , Ratas , Ratas Wistar , Sinapsis/fisiología , Transmisión Sináptica/efectos de los fármacos , beta-Alanina/análogos & derivados , beta-Alanina/farmacología
10.
J Biol Chem ; 288(47): 33813-33823, 2013 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-24121511

RESUMEN

Enterocytes are specialized to absorb nutrients from the lumen of the small intestine by expressing a select set of genes to maximize the uptake of nutrients. They develop from stem cells in the crypt and differentiate into mature enterocytes while moving along the crypt-villus axis. Using the Slc6a19 gene as an example, encoding the neutral amino acid transporter B(0)AT1, we studied regulation of the gene by transcription factors and epigenetic factors in the intestine. To investigate this question, we used a fractionation method to separate mature enterocytes from crypt cells and analyzed gene expression. Transcription factors HNF1a and HNF4a activate transcription of the Slc6a19 gene in villus enterocytes, whereas high levels of SOX9 repress expression in the crypts. CpG dinucleotides in the proximal promoter were highly methylated in the crypt and fully de-methylated in the villus. Furthermore, histone modification H3K27Ac, indicating an active promoter, was prevalent in villus cells but barely detectable in crypt cells. The results suggest that Slc6a19 expression in the intestine is regulated at three different levels involving promoter methylation, histone modification, and opposing transcription factors.


Asunto(s)
Sistemas de Transporte de Aminoácidos Neutros/biosíntesis , Enterocitos/metabolismo , Epigénesis Genética/fisiología , Factor Nuclear 1-alfa del Hepatocito/metabolismo , Factor Nuclear 4 del Hepatocito/metabolismo , Regiones Promotoras Genéticas/fisiología , Factor de Transcripción SOX9/metabolismo , Transcripción Genética/fisiología , Sistemas de Transporte de Aminoácidos Neutros/genética , Animales , Células Cultivadas , Metilación de ADN/fisiología , Enterocitos/citología , Factor Nuclear 1-alfa del Hepatocito/genética , Factor Nuclear 4 del Hepatocito/genética , Histonas/genética , Histonas/metabolismo , Ratones , Procesamiento Proteico-Postraduccional/fisiología , Factor de Transcripción SOX9/genética
11.
Pancreatology ; 13(5): 475-85, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24075511

RESUMEN

Pancreatic acinar cells accumulate amino acids against a marked concentration gradient to synthesize digestive enzymes. Thus, the function of acinar cells depends on amino acid uptake mediated by active transport. Despite the importance of this process, pancreatic amino acid transporter expression and cellular localization is still unclear. We screened mouse pancreas for the expression of genes encoding amino acid transporters. We showed that the most highly expressed transporters, namely sodium dependent SNAT3 (Slc38a3) and SNAT5 (Slc38a5) and sodium independent neutral amino acids transporters LAT1 (Slc7a5) and LAT2 (Slc7a8), are expressed in the basolateral membrane of acinar cells. SNAT3 and SNAT5, LAT1 and LAT2 are expressed in acinar cells. Additional evidence that these transporters are expressed in mature acinar cells was gained using acinar cell culture and acute pancreatitis models. In the acute phase of pancreatic injury, when acinar cell loss occurs, and in an acinar cell culture model, which mimics changes occurring during pancreatitis, SNAT3 and SNAT5 are strongly down-regulated. LAT1 and LAT2 were down-regulated only in the in vitro model. At protein level, SNAT3 and SNAT5 expression was also reduced during pancreatitis. Expression of other amino acid transporters was also modified in both models of pancreatitis. The subset of transporters with differential expression patterns during acute pancreatitis might be involved in the injury/regeneration phases. Further expression, localization and functional studies will follow to better understand changes occurring during acute pancreatitis. These findings provide insight into pancreatic amino acid transport in healthy pancreas and during acute pancreatitis injury.


Asunto(s)
Células Acinares/metabolismo , Sistemas de Transporte de Aminoácidos/biosíntesis , Páncreas/fisiología , Pancreatitis/fisiopatología , Enfermedad Aguda , Sistema de Transporte de Aminoácidos y+/biosíntesis , Sistemas de Transporte de Aminoácidos Neutros/biosíntesis , Animales , Células Cultivadas , Cadenas Ligeras de la Proteína-1 Reguladora de Fusión/biosíntesis , Transportador de Aminoácidos Neutros Grandes 1/biosíntesis , Masculino , Ratones , Páncreas/fisiopatología
12.
Food Chem Toxicol ; 59: 137-44, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23747718

RESUMEN

We investigated the underlying mechanisms of L-leucine and L-isoleucine mediated promotion of bladder carcinogenesis using an initiation-promotion model. Rats were administered N-butyl-N-(4-hydroxybutyl) nitrosamine for 4 weeks and then fed AIN-93G basal diet or diet supplemented with L-leucine or L-isoleucine for 8 weeks followed by the basal diet for another 8 weeks. At the end of the experiment, week 20, there was a significant elevation of papillary and nodular (PN) hyperplasia multiplicity in the amino acid groups. L-Leucine and L-isoleucine transporters were up-regulated in PN hyperplasias and/or bladder tumors compared with concomitant normal-appearing bladder urothelium at weeks 12 and/or 20 in all groups. In addition, in normal-appearing bladder urothelium, significantly increased mRNA levels of y+LAT1, LAT2, LAT4, and 4F2hc were observed in the amino acid groups compared with the BBN control group at both weeks 12 and 20, and increased mRNA levels of LAT1 were observed at week 20. Furthermore, up-regulation of TNF-α, c-fos, ß-catenin, p53, p21(Cip1/WAF1), cdk4, cyclin D1 and caspase 3 in the amino acid groups was detected in normal-appearing bladder urothelium. Overall, our results indicate that supplementation with l-leucine or l-isoleucine enhanced growth of bladder urothelial tumors by triggering expression of amino acid transporters and tumorigenesis-associated genes.


Asunto(s)
Sistemas de Transporte de Aminoácidos/metabolismo , Aminoácidos de Cadena Ramificada/efectos adversos , Suplementos Dietéticos/efectos adversos , Regulación Neoplásica de la Expresión Génica , Proteínas de Neoplasias/metabolismo , Neoplasias de la Vejiga Urinaria/metabolismo , Urotelio/metabolismo , Sistema de Transporte de Aminoácidos y+/biosíntesis , Sistema de Transporte de Aminoácidos y+/genética , Sistema de Transporte de Aminoácidos y+/metabolismo , Sistemas de Transporte de Aminoácidos/biosíntesis , Sistemas de Transporte de Aminoácidos/genética , Sistemas de Transporte de Aminoácidos Neutros/biosíntesis , Sistemas de Transporte de Aminoácidos Neutros/genética , Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Aminoácidos de Cadena Ramificada/metabolismo , Animales , Carcinogénesis/inducido químicamente , Carcinogénesis/metabolismo , Cadena Pesada de la Proteína-1 Reguladora de Fusión/biosíntesis , Cadena Pesada de la Proteína-1 Reguladora de Fusión/genética , Cadena Pesada de la Proteína-1 Reguladora de Fusión/metabolismo , Cadenas Ligeras de la Proteína-1 Reguladora de Fusión/biosíntesis , Cadenas Ligeras de la Proteína-1 Reguladora de Fusión/genética , Cadenas Ligeras de la Proteína-1 Reguladora de Fusión/metabolismo , Hiperplasia , Isoleucina/efectos adversos , Isoleucina/metabolismo , Transportador de Aminoácidos Neutros Grandes 1/biosíntesis , Transportador de Aminoácidos Neutros Grandes 1/genética , Transportador de Aminoácidos Neutros Grandes 1/metabolismo , Leucina/efectos adversos , Leucina/metabolismo , Masculino , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Distribución Aleatoria , Ratas , Ratas Endogámicas F344 , Carga Tumoral , Vejiga Urinaria/efectos de los fármacos , Vejiga Urinaria/metabolismo , Vejiga Urinaria/patología , Neoplasias de la Vejiga Urinaria/inducido químicamente , Neoplasias de la Vejiga Urinaria/patología , Urotelio/efectos de los fármacos , Urotelio/patología
13.
Am J Transplant ; 10(10): 2241-51, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20883558

RESUMEN

T cell-mediated rejection of kidney allografts causes epithelial deterioration, manifested by tubulitis, but the mechanism remains unclear. We hypothesized that interstitial inflammation triggers a stereotyped epithelial response similar to that triggered by other types of injury such as ischemia-reperfusion. We identified solute carrier transcripts with decreased expression in mouse allografts, and compared their behavior in T cell-mediated rejection to native kidneys with ischemic acute tubular necrosis (ATN). Average loss of solute carrier expression was similar in ATN (77%) and T cell-mediated rejection (75%) with high correlation of individual transcripts. Immunostaining of SLC6A19 confirmed loss of proteins. Analysis of human kidney transplant biopsies confirmed that T cell-mediated rejection and ATN showed similar loss of solute carrier mRNAs. The loss of solute carrier expression was weakly correlated with interstitial inflammation, but kidneys with ATN showed decreased solute carriers despite minimal inflammation. Loss of renal function correlated better with decreased solute carrier expression than with histologic lesions (r = 0.396, p < 0.001). Thus the loss of epithelial transcripts in rejection is not a unique consequence of T cell-mediated rejection but an active injury-repair response of epithelium, triggered by rejection but also by other injury mechanisms.


Asunto(s)
Rechazo de Injerto/metabolismo , Necrosis Tubular Aguda/patología , Proteínas de Transporte de Membrana/fisiología , Sistemas de Transporte de Aminoácidos Neutros/biosíntesis , Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Animales , Rechazo de Injerto/inmunología , Rechazo de Injerto/patología , Humanos , Riñón/patología , Trasplante de Riñón/patología , Trasplante de Riñón/fisiología , Necrosis Tubular Aguda/metabolismo , Túbulos Renales/patología , Ratones , Ratones Endogámicos CBA , Cicatrización de Heridas/inmunología
14.
Placenta ; 31(4): 295-304, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20167367

RESUMEN

Thyroid hormones (TH) are important for the development of the human fetus and placenta from very early gestation. The transplacental passage of TH from mother to fetus and the supply of TH into trophoblasts require the expression of placental TH plasma membrane transporters. We describe the ontogeny of the TH transporters MCT8, MCT10, LAT1, LAT2, OATP1A2 and OATP4A1 in a large series (n = 110) of normal human placentae across gestation and describe their expression changes with intrauterine fetal growth restriction (IUGR n = 22). Quantitative RT-PCR revealed that all the mRNAs encoding TH transporters are expressed in human placenta from 6 weeks gestation and throughout pregnancy. MCT8, MCT10, OATP1A2 and LAT1 mRNA expression increased with gestation. OATP4A1 and CD98 (LATs obligatory associated protein) mRNA expression reached a nadir in mid-gestation before increasing towards term. LAT2 mRNA expression did not alter throughout gestation. Immunohistochemistry localised MCT10 and OATP1A2 to villous cytotrophoblasts and syncytiotrophoblasts, and extravillous trophoblasts while OATP4A1 was preferentially expressed in the villous syncytiotrophoblasts. Whilst MCT8 protein expression was increased, MCT10 mRNA expression was decreased in placentae from IUGR pregnancies delivered in the early 3rd trimester compared to age matched appropriately grown for gestational age controls. No significant change was found in the mRNA expression of the other transporters with IUGR. In conclusion, several TH transporters are present in the human placenta from early 1st trimester with varying patterns of expression throughout gestation. Their coordinated effects may regulate both transplacental TH passage and TH supply to trophoblasts, which are critical for the normal development of the fetus and placenta. Increased MCT8 and decreased MCT10 expression within placentae of pregnancies complicated by IUGR may contribute to aberrant development of the fetoplacental unit.


Asunto(s)
Sistemas de Transporte de Aminoácidos Neutros/biosíntesis , Retardo del Crecimiento Fetal/metabolismo , Transportadores de Ácidos Monocarboxílicos/biosíntesis , Hormonas Tiroideas/metabolismo , Femenino , Humanos , Transportador de Aminoácidos Neutros Grandes 1/biosíntesis , Transportadores de Anión Orgánico/biosíntesis , Placenta/metabolismo , Embarazo , ARN Mensajero/metabolismo , Simportadores , Trofoblastos/metabolismo
15.
J Anim Sci ; 88(3): 1028-33, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19933436

RESUMEN

To test the hypothesis that AA transporter transcripts are present in the large intestine and similarly expressed along the intestinal tract, mRNA abundance of candidate AA transporter genes solute carrier (SLC) family 7, member 9 (SLC7A9), SLC7A1, SLC7A8, and SLC43A1 encoding for b(0,+)-type AA transporter (b(0,+)AT), cationic AA transporter-1 (CAT-1), L-type AA transporter-2 (LAT-2), and L-type AA transporter-3 (LAT-3), respectively, was determined in small and large intestinal segments of the horse. Mucosa was collected from the equine small (jejunum and ileum) and large intestine (cecum, left ventral colon, and left dorsal colon), flash frozen in liquid nitrogen, and stored at -80 degrees C. Messenger RNA was isolated from tissue samples, followed by manufacture of cDNA. Relative quantitative reverse transcription-PCR was conducted using the 2(-DeltaDeltaCT) method, with glyceraldehyde-3-phosphate dehydrogenase serving as the housekeeping gene. Compared with the jejunum, cationic and neutral AA transporter SLC7A9 mRNA abundance was similar in the ileum, cecum, and large intestinal segments. Compared with the jejunum, cationic AA transporter SLC7A1 mRNA abundance was similar in the ileum and decreased in the cecum, left ventral colon, and left dorsal colon (P < 0.001). Neutral AA transporter SLC7A8 mRNA abundance decreased from the cranial to caudal end of the intestinal tract (P < 0.001). Neutral AA transporter SLC43A1 mRNA abundance was similar in the ileum and left dorsal colon and increased in the cecum (P < 0.01) and left ventral colon (P < 0.1) compared with the jejunum. Cationic and neutral AA transporter SLC7A9 mRNA abundance was similarly expressed in the large compared with small intestine, whereas cationic AA transporter SLC7A1 was of low abundance in the large intestine; neutral AA transporters SLC7A8 and SLC43A1 were differentially expressed with decreased abundance of SLC7A8 and increased abundance of SLC43A1 in the large intestine. Results indicate that the large intestine might contribute to both cationic and neutral AA uptake and absorption predominantly via transporters LAT-3 and b(0,+)AT.


Asunto(s)
Sistemas de Transporte de Aminoácidos Básicos/análisis , Sistemas de Transporte de Aminoácidos Neutros/análisis , Caballos/metabolismo , Intestinos/química , Sistemas de Transporte de Aminoácidos Básicos/biosíntesis , Sistemas de Transporte de Aminoácidos Neutros/biosíntesis , Animales , Transportador de Aminoácidos Catiónicos 1/análisis , Transportador de Aminoácidos Catiónicos 1/biosíntesis , Ciego/química , Ciego/metabolismo , Colon/química , Colon/metabolismo , Caballos/fisiología , Íleon/química , Íleon/metabolismo , Mucosa Intestinal/metabolismo , Yeyuno/química , Yeyuno/metabolismo , ARN Mensajero/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
16.
Eur J Endocrinol ; 161(2): 243-50, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19439506

RESUMEN

OBJECTIVE: Prolonged critically ill patients have low circulating thyroid hormone (TH) levels without a rise in TSH, a condition labeled 'the low tri-iodothyronine (T(3)) syndrome'. Currently, it is not clear whether this represents an adaptive response. We examined the role of TH transporters monocarboxylate transporter 8 (MCT8, also known as SLC16A2) and MCT10 in the pathogenesis of the low T(3) syndrome in prolonged critical illness. METHODS: A clinical observational study in critically ill patients and an intervention study in an in vivo animal model of critical illness. Gene expression levels of MCT8 and MCT10 were measured by real-time PCR. RESULTS: In prolonged critically ill patients, we measured increased MCT8 but not MCT10 gene expression levels in liver and skeletal muscle as compared with patients undergoing acute surgical stress. In a rabbit model of prolonged critical illness, gene expression levels of MCT8 in liver and of MCT10 in skeletal muscle were increased as compared with healthy controls. Treatment of prolonged critically ill rabbits with TH (thyroxine+T(3)) resulted in a downregulation of gene expression levels of MCT8 in liver and of MCT10 in muscle. Transporter expression levels correlated inversely with circulating TH parameters. CONCLUSIONS: These data suggest that alterations in the expression of TH transporters do not play a major role in the pathogenesis of the 'low T(3) syndrome' but rather reflect a compensatory effort in response to hypothyroidism.


Asunto(s)
Sistemas de Transporte de Aminoácidos Neutros/biosíntesis , Enfermedad Crítica , Síndromes del Eutiroideo Enfermo/metabolismo , Transportadores de Ácidos Monocarboxílicos/biosíntesis , Triyodotironina/sangre , Anciano , Sistemas de Transporte de Aminoácidos Neutros/genética , Animales , Secuencia de Bases , Modelos Animales de Enfermedad , Síndromes del Eutiroideo Enfermo/sangre , Síndromes del Eutiroideo Enfermo/genética , Femenino , Humanos , Hígado/metabolismo , Masculino , Datos de Secuencia Molecular , Transportadores de Ácidos Monocarboxílicos/genética , Músculo Esquelético/metabolismo , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Conejos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Simportadores , Tiroxina/administración & dosificación , Tiroxina/sangre , Triyodotironina/administración & dosificación , Triyodotironina/deficiencia , Triyodotironina Inversa/sangre
17.
J Neurochem ; 110(3): 822-30, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19457077

RESUMEN

Glutamine (Gln) plays an important role in brain energy metabolism and as a precursor for the synthesis of neurotransmitter glutamate and GABA. Previous studies have shown that astrocytic Gln transport is impaired following manganese (Mn) exposure. The present studies were performed to identify the transport routes and the respective Gln transporters contributing to the impairment. Rat neonatal cortical primary astrocytes treated with Mn displayed a significant decrease in Gln uptake mediated by the principle Gln transporting systems, N and ASC. Moreover, systems N, ASC and L were less efficient in Gln export after Mn treatment. Mn treatment caused a significant reduction of both in mRNA expression and protein levels of SNAT3 (system N), SNAT2 (system A) and LAT2 (system L), and lowered the protein but not mRNA expression of ASCT2 (system ASC). Mn exposure did not affect the expression of the less abundant systems N transporter SNAT5 and the system L transporter LAT1, at either the mRNA or protein level. Hence, Mn-induced decrease of inward and outward Gln transport can be largely ascribed to the loss of the specific Gln transporters. Consequently, deregulation of glutamate homeostasis and its diminished availability to neurons may lead to impairment in glutamatergic neurotransmission, a phenomenon characteristic of Mn-induced neurotoxicity.


Asunto(s)
Sistema de Transporte de Aminoácidos ASC/antagonistas & inhibidores , Sistema de Transporte de Aminoácidos ASC/metabolismo , Sistemas de Transporte de Aminoácidos Neutros/antagonistas & inhibidores , Astrocitos/metabolismo , Proteínas Portadoras/antagonistas & inhibidores , Cloruros/toxicidad , Regulación de la Expresión Génica/fisiología , Sistema de Transporte de Aminoácidos ASC/biosíntesis , Sistema de Transporte de Aminoácidos ASC/fisiología , Sistemas de Transporte de Aminoácidos Neutros/biosíntesis , Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Sistemas de Transporte de Aminoácidos Neutros/fisiología , Animales , Animales Recién Nacidos , Astrocitos/efectos de los fármacos , Astrocitos/fisiología , Proteínas Portadoras/biosíntesis , Proteínas Portadoras/metabolismo , Proteínas Portadoras/fisiología , Células Cultivadas , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Corteza Cerebral/fisiología , Compuestos de Manganeso , Antígenos de Histocompatibilidad Menor , Ratas , Ratas Sprague-Dawley
18.
Biochem Biophys Res Commun ; 381(3): 378-82, 2009 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-19233140

RESUMEN

Insulin and glucagon secretion from the islets of Langerhans is highly regulated. Although an increased plasma glucose level is the major stimulus for insulin exocytosis, roles for glutamine and glutamate have been suggested. Interestingly, the islet cells display elements associated with synaptic transmission. In the central nervous system (CNS), glutamine transport by SN1 and SAT2 sustain the generation of neurotransmitter glutamate. We hypothesized that the same transporters are essential for glutamine transport into the islet cells and for subsequent formation of glutamate acting as an intracellular signaling molecule. We demonstrate that islet cells express several transporters which can mediate glutamine transport. In particular, we show pronounced expression of SN1 and SAT2 in B-cells and A-cells, respectively. The cell-specific expression of these transporters together with their functional characteristics suggest an important role for glutamine in the regulation of insulin secretion.


Asunto(s)
Acetiltransferasas/biosíntesis , Sistemas de Transporte de Aminoácidos Neutros/biosíntesis , Células Secretoras de Glucagón/metabolismo , Glutamina/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Acetiltransferasas/genética , Sistemas de Transporte de Aminoácidos Neutros/genética , Animales , Transporte Biológico , Secreción de Insulina , Ratones , Ratas , Ratas Wistar
19.
Invest Ophthalmol Vis Sci ; 49(11): 5151-60, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18689705

RESUMEN

PURPOSE: Glutamine transport is essential for the glutamate-glutamine cycle, which occurs between neurons and glia. System N, consisting of SN1 (SNAT3) and SN2 (SNAT5), is the principal mediator of glutamine transport in retinal Müller cells. Mediators of glutamine transport in retinal ganglion cells were investigated. METHODS: The relative contributions of various transport systems for glutamine uptake (systems N, A, L, y+L, ASCT, and ATB(0,+)) were examined in RGC-5 cells based on differential features of the individual transport systems. mRNA for the genes encoding members of these transport systems were analyzed by RT-PCR. Based on these data, SN1 and SN2 were analyzed in mouse retina, RGC-5 cells, and primary mouse ganglion cells (GCs) by in situ hybridization (ISH), immunofluorescence (IF), and Western blotting. RESULTS: Three transport systems--N, A, and L--participated in glutamine uptake in RGC-5 cells. System N was the principal contributor; systems A and L contributed considerably less. ISH and IF revealed SN1 and SN2 expression in the ganglion, inner nuclear, and photoreceptor cell layers. SN1 and SN2 colocalized with the ganglion cell marker Thy 1.2 and with the Müller cell marker vimentin, confirming their presence in both retinal cell types. SN1 and SN2 proteins were detected in primary mouse GCs. CONCLUSIONS: These findings suggest that in addition to its role in glutamine uptake in retinal glial cells, system N contributes significantly to glutamine uptake in ganglion cells and, hence, contributes to the retinal glutamate-glutamine cycle.


Asunto(s)
Sistemas de Transporte de Aminoácidos Neutros/genética , Expresión Génica , ARN Mensajero/genética , Células Ganglionares de la Retina/metabolismo , Sistemas de Transporte de Aminoácidos Neutros/biosíntesis , Animales , Western Blotting , Células Cultivadas , Hibridación in Situ , Ratones , Neurotransmisores , Células Ganglionares de la Retina/citología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal
20.
Physiol Genomics ; 32(3): 322-34, 2008 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-18056784

RESUMEN

Production and excretion of acids are balanced to maintain systemic acid-base homeostasis. During metabolic acidosis (MA) excess acid accumulates and is removed from the body, a process achieved, at least in part, by increasing renal acid excretion. This acid-secretory process requires the concerted regulation of metabolic and transport pathways, which are only partially understood. Chronic MA causes also morphological remodeling of the kidney. Therefore, we characterized transcriptional changes in mammalian kidney during MA to gain insights into adaptive pathways. Total kidney RNA from control and 2- and 7-days NH(4)Cl treated mice was subjected to microarray gene profiling. We identified 4,075 transcripts significantly (P < 0.05) regulated after 2 and/or 7 days of treatment. Microarray results were confirmed by qRT-PCR. Analysis of candidate genes revealed that a large group of regulated transcripts was represented by different solute carrier transporters, genes involved in cell growth, proliferation, apoptosis, water homeostasis, and ammoniagenesis. Pathway analysis revealed that oxidative phosphorylation was the most affected pathway. Interestingly, the majority of acutely regulated genes after 2 days, returned to normal values after 7 days suggesting that adaptation had occurred. Besides these temporal changes, we detected also differential regulation of selected genes (SNAT3, PEPCK, PDG) between early and late proximal tubule. In conclusion, the mammalian kidney responds to MA by temporally and spatially altering the expression of a large number of genes. Our analysis suggests that many of these genes may participate in various processes leading to adaptation and restoration of normal systemic acid-base and electrolyte homeostasis.


Asunto(s)
Acidosis Tubular Renal/genética , Adaptación Fisiológica/genética , Perfilación de la Expresión Génica , Túbulos Renales Proximales/metabolismo , Acidosis Tubular Renal/inducido químicamente , Acidosis Tubular Renal/metabolismo , Sistemas de Transporte de Aminoácidos Neutros/biosíntesis , Sistemas de Transporte de Aminoácidos Neutros/genética , Cloruro de Amonio/toxicidad , Animales , Arginina/metabolismo , Cloruros/sangre , Regulación de la Expresión Génica , Redes Reguladoras de Genes/genética , Glutamina/metabolismo , Riñón/química , Masculino , Ratones , Ratones Endogámicos C57BL , Fosforilación Oxidativa , Fosfoenolpiruvato Carboxilasa/biosíntesis , Fosfoenolpiruvato Carboxilasa/genética , ARN Mensajero/biosíntesis , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...