Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Amino Acids ; 48(6): 1491-508, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26984322

RESUMO

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.


Assuntos
Sistemas de Transporte de Aminoácidos Básicos/biossíntese , Sistemas de Transporte de Aminoácidos Neutros/biossíntese , Ração Animal , Regulação da Expressão Gênica , Mucosa Intestinal/metabolismo , Jejuno/metabolismo , Peptidil Dipeptidase A/biossíntese , Enzima de Conversão de Angiotensina 2 , Animais , Feminino , Masculino , Suínos
2.
J Neurosci ; 33(44): 17429-34, 2013 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-24174676

RESUMO

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.


Assuntos
Sistemas de Transporte de Aminoácidos Neutros/biossíntese , Tronco Encefálico/fisiologia , Ácido Glutâmico/fisiologia , Glutamina/biossíntese , Terminações Pré-Sinápticas/metabolismo , Sinapses/metabolismo , Transmissão Sináptica/fisiologia , Sistemas de Transporte de Aminoácidos Neutros/antagonistas & inibidores , Sistemas de Transporte de Aminoácidos Neutros/fisiologia , Animais , Transporte Biológico Ativo/fisiologia , Tronco Encefálico/metabolismo , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Feminino , Ácido Glutâmico/metabolismo , Glutamina/metabolismo , Glutamina/fisiologia , Masculino , Técnicas de Cultura de Órgãos , Ratos , Ratos Wistar , Sinapses/fisiologia , Transmissão Sináptica/efeitos dos fármacos , beta-Alanina/análogos & derivados , beta-Alanina/farmacologia
3.
J Biol Chem ; 288(47): 33813-33823, 2013 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-24121511

RESUMO

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.


Assuntos
Sistemas de Transporte de Aminoácidos Neutros/biossíntese , Enterócitos/metabolismo , Epigênese Genética/fisiologia , Fator 1-alfa Nuclear de Hepatócito/metabolismo , Fator 4 Nuclear de Hepatócito/metabolismo , Regiões Promotoras Genéticas/fisiologia , Fatores de Transcrição SOX9/metabolismo , Transcrição Gênica/fisiologia , Sistemas de Transporte de Aminoácidos Neutros/genética , Animais , Células Cultivadas , Metilação de DNA/fisiologia , Enterócitos/citologia , Fator 1-alfa Nuclear de Hepatócito/genética , Fator 4 Nuclear de Hepatócito/genética , Histonas/genética , Histonas/metabolismo , Camundongos , Processamento de Proteína Pós-Traducional/fisiologia , Fatores de Transcrição SOX9/genética
4.
Cell Physiol Biochem ; 33(5): 1591-606, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24854847

RESUMO

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.


Assuntos
Acidose/metabolismo , Sistemas de Transporte de Aminoácidos Neutros/biossíntese , Sistemas de Transporte de Aminoácidos Neutros/genética , Acidose/genética , Animais , Células Cultivadas , Células HEK293 , Células HeLa , Células Hep G2 , Humanos , Concentração de Íons de Hidrogênio , Camundongos , Especificidade de Órgãos/genética , Regiões Promotoras Genéticas/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transcrição Gênica/genética
5.
Nephrol Dial Transplant ; 29(3): 522-8, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23861466

RESUMO

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.


Assuntos
Sistemas de Transporte de Aminoácidos Neutros/genética , Cistinose/terapia , Terapia Genética , Sistemas de Transporte de Aminoácidos Neutros/biossíntese , Animais , Cistinose/genética , Técnicas de Transferência de Genes , Objetivos , Transplante de Células-Tronco Hematopoéticas , Humanos , Rim/metabolismo , Rim/patologia , Camundongos Knockout
6.
Pancreatology ; 13(5): 475-85, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24075511

RESUMO

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.


Assuntos
Células Acinares/metabolismo , Sistemas de Transporte de Aminoácidos/biossíntese , Pâncreas/fisiologia , Pancreatite/fisiopatologia , Doença Aguda , Sistema y+ de Transporte de Aminoácidos/biossíntese , Sistemas de Transporte de Aminoácidos Neutros/biossíntese , Animais , Células Cultivadas , Cadeias Leves da Proteína-1 Reguladora de Fusão/biossíntese , Transportador 1 de Aminoácidos Neutros Grandes/biossíntese , Masculino , Camundongos , Pâncreas/fisiopatologia
7.
Am J Transplant ; 10(10): 2241-51, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20883558

RESUMO

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.


Assuntos
Rejeição de Enxerto/metabolismo , Necrose Tubular Aguda/patologia , Proteínas de Membrana Transportadoras/fisiologia , Sistemas de Transporte de Aminoácidos Neutros/biossíntese , Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Animais , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/patologia , Humanos , Rim/patologia , Transplante de Rim/patologia , Transplante de Rim/fisiologia , Necrose Tubular Aguda/metabolismo , Túbulos Renais/patologia , Camundongos , Camundongos Endogâmicos CBA , Cicatrização/imunologia
8.
Free Radic Res ; 54(7): 525-534, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32873097

RESUMO

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.


Assuntos
Sistemas de Transporte de Aminoácidos Básicos/metabolismo , Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Cistina/metabolismo , Sistemas de Transporte de Aminoácidos Básicos/biossíntese , Sistemas de Transporte de Aminoácidos Básicos/genética , Sistemas de Transporte de Aminoácidos Neutros/biossíntese , Sistemas de Transporte de Aminoácidos Neutros/genética , Animais , Feminino , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Estresse Oxidativo/fisiologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Distribuição Tecidual
9.
J Neurochem ; 110(3): 822-30, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19457077

RESUMO

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.


Assuntos
Sistema ASC de Transporte de Aminoácidos/antagonistas & inibidores , Sistema ASC de Transporte de Aminoácidos/metabolismo , Sistemas de Transporte de Aminoácidos Neutros/antagonistas & inibidores , Astrócitos/metabolismo , Proteínas de Transporte/antagonistas & inibidores , Cloretos/toxicidade , Regulação da Expressão Gênica/fisiologia , Sistema ASC de Transporte de Aminoácidos/biossíntese , Sistema ASC de Transporte de Aminoácidos/fisiologia , Sistemas de Transporte de Aminoácidos Neutros/biossíntese , Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Sistemas de Transporte de Aminoácidos Neutros/fisiologia , Animais , Animais Recém-Nascidos , Astrócitos/efeitos dos fármacos , Astrócitos/fisiologia , Proteínas de Transporte/biossíntese , Proteínas de Transporte/metabolismo , Proteínas de Transporte/fisiologia , Células Cultivadas , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Córtex Cerebral/fisiologia , Compostos de Manganês , Antígenos de Histocompatibilidade Menor , Ratos , Ratos Sprague-Dawley
10.
Biochem Biophys Res Commun ; 381(3): 378-82, 2009 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-19233140

RESUMO

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.


Assuntos
Acetiltransferases/biossíntese , Sistemas de Transporte de Aminoácidos Neutros/biossíntese , Células Secretoras de Glucagon/metabolismo , Glutamina/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Acetiltransferases/genética , Sistemas de Transporte de Aminoácidos Neutros/genética , Animais , Transporte Biológico , Secreção de Insulina , Camundongos , Ratos , Ratos Wistar
11.
Physiol Genomics ; 32(3): 322-34, 2008 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-18056784

RESUMO

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.


Assuntos
Acidose Tubular Renal/genética , Adaptação Fisiológica/genética , Perfilação da Expressão Gênica , Túbulos Renais Proximais/metabolismo , Acidose Tubular Renal/induzido quimicamente , Acidose Tubular Renal/metabolismo , Sistemas de Transporte de Aminoácidos Neutros/biossíntese , Sistemas de Transporte de Aminoácidos Neutros/genética , Cloreto de Amônio/toxicidade , Animais , Arginina/metabolismo , Cloretos/sangue , Regulação da Expressão Gênica , Redes Reguladoras de Genes/genética , Glutamina/metabolismo , Rim/química , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação Oxidativa , Fosfoenolpiruvato Carboxilase/biossíntese , Fosfoenolpiruvato Carboxilase/genética , RNA Mensageiro/biossíntese , Transcrição Gênica
12.
Physiol Genomics ; 31(2): 183-92, 2007 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-17536021

RESUMO

Gastric enterochromaffin-like (ECL) cells release histamine in response to food because of elevation of gastrin and neural release of pituitary adenylate cyclase-activating peptide (PACAP). Acid secretion is at a basal level in the absence of food but is rapidly stimulated with feeding. Rats fasted for 24 h showed a significant decrease of mucosal histamine despite steady-state expression of the histamine-synthesizing enzyme histidine decarboxylase (HDC). Comparative transcriptomal analysis using gene expression oligonucleotide microarrays of 95% pure ECL cells from fed and 24-h fasted rats, thereby eliminating mRNA contamination from other gastric mucosal cell types, identified significantly increased gene expression of the enzymes histidase and urocanase catabolizing the HDC substrate L-histidine but significantly decreased expression of the cellular L-histidine uptake transporter SN2 and of the vesicular monoamine transporter 2 (VMAT-2) responsible for histamine uptake into secretory vesicles. This was confirmed by reverse transcriptase-quantitative polymerase chain reaction of gastric fundic mucosal samples from fed and 24-h fasted rats. The decrease of VMAT-2 gene expression was also shown by a decrease in VMAT-2 protein content in protein extracts from fed and 24-h fasted rats compared with equal amounts of HDC protein and Na-K-ATPase alpha(1)-subunit protein content. These results indicate that rat gastric ECL cells regulate their histamine content during 24-h fasting not by a change in HDC gene or protein expression but by regulation of substrate concentration for HDC and a decreased histamine secretory pool.


Assuntos
Células Enterocromafins/metabolismo , Jejum/metabolismo , Regulação da Expressão Gênica/fisiologia , Liberação de Histamina/genética , Sistemas de Transporte de Aminoácidos Neutros/biossíntese , Sistemas de Transporte de Aminoácidos Neutros/genética , Animais , Contagem de Células , Indução Enzimática , Perfilação da Expressão Gênica , Histamina/metabolismo , Liberação de Histamina/fisiologia , Histidina Amônia-Liase/biossíntese , Histidina Amônia-Liase/genética , Histidina Descarboxilase/metabolismo , Masculino , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ratos , Ratos Sprague-Dawley , ATPase Trocadora de Sódio-Potássio/biossíntese , ATPase Trocadora de Sódio-Potássio/genética , Transcrição Gênica , Urocanato Hidratase/biossíntese , Urocanato Hidratase/genética , Proteínas Vesiculares de Transporte de Monoamina/biossíntese , Proteínas Vesiculares de Transporte de Monoamina/genética
13.
Toxicol Sci ; 98(1): 198-205, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17468184

RESUMO

Excessive free radical formation has been implicated as a causative factor in neurotoxic damage associated with exposures to a variety of metals, including manganese (Mn). It is well established that Mn accumulates in astrocytes, affecting their ability to indirectly induce and/or exacerbate neuronal dysfunction. The present study examined the effects of Mn treatment on the following endpoints in primary astrocyte cultures: (1) oxidative injury, (2) alterations in high-energy phosphate (adenosine 5'-triphosphate, ATP) levels, (3) mitochondrial inner membrane potential, and (4) glutamine uptake and the expression of glutamine transporters. We quantified astrocyte cerebral oxidative damage by measuring F(2)-isoprostanes (F(2)-IsoPs) using stable isotope dilution methods followed by gas chromatography-mass spectrometry with selective ion monitoring. Our data showed a significant (p < 0.01) elevation in F(2)-IsoPs levels at 2 h following exposure to Mn (100 microM, 500 microM, or 1 mM). Consistent with this observation, Mn induced a concentration-dependent reduction in ATP and the inner mitochondrial membrane potential (DeltaPsi(m)), measured by the high pressure liquid chromatography method and the potentiometric dye, tetramethyl rhodamine ethyl ester, respectively. Moreover, 30 min of pretreatment with Mn (100 microM, 500 microM, or 1 mM) inhibited the net uptake of glutamine (GLN) ((3)H-glutamine) measured at 1 and 5 min. Expression of the messenger RNA coding the GLN transporters, SNAT3/SN1 and SNAT1, was inhibited after 100 and 500 microM Mn treatment for 24 h. Our results demonstrate that induction of oxidative stress, associated mitochondrial dysfunction, and alterations in GLN/glutamate cycling in astrocytes represent key mechanisms by which Mn exerts its neurotoxicity.


Assuntos
Astrócitos/patologia , Intoxicação por Manganês/metabolismo , Intoxicação por Manganês/patologia , Estresse Oxidativo/efeitos dos fármacos , Trifosfato de Adenosina/metabolismo , Sistema ASC de Transporte de Aminoácidos/biossíntese , Sistemas de Transporte de Aminoácidos Neutros/biossíntese , Animais , Animais Recém-Nascidos , Astrócitos/efeitos dos fármacos , Células Cultivadas , Espaço Extracelular/efeitos dos fármacos , F2-Isoprostanos/metabolismo , Glutamina/metabolismo , L-Lactato Desidrogenase/metabolismo , Potenciais da Membrana/efeitos dos fármacos , Antígenos de Histocompatibilidade Menor , Membranas Mitocondriais/efeitos dos fármacos , RNA Mensageiro/biossíntese , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sais de Tetrazólio/metabolismo , Tiazóis/metabolismo
14.
PLoS One ; 12(9): e0184845, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28915252

RESUMO

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.


Assuntos
Sistemas de Transporte de Aminoácidos Neutros/biossíntese , Antígenos CD13/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Intestino Delgado/metabolismo , Isoleucina/farmacologia , Peptidil Dipeptidase A/metabolismo , Enzima de Conversão de Angiotensina 2 , Animais , Suplementos Nutricionais , Masculino , Ratos , Ratos Wistar
15.
J Biosci Bioeng ; 123(4): 444-450, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28057466

RESUMO

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.


Assuntos
Alanina/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Regulação Bacteriana da Expressão Gênica , Proteína Reguladora de Resposta a Leucina/metabolismo , Alanina/farmacologia , Sistemas de Transporte de Aminoácidos Neutros/biossíntese , Sistemas de Transporte de Aminoácidos Neutros/genética , Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Sequência de Bases , Pegada de DNA , Desoxirribonuclease I/metabolismo , Dipeptídeos/metabolismo , Dipeptídeos/farmacologia , Ensaio de Desvio de Mobilidade Eletroforética , Escherichia coli/efeitos dos fármacos , Proteínas de Escherichia coli/biossíntese , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Genes Reporter/genética , Leucina/metabolismo , Leucina/farmacologia , Proteína Reguladora de Resposta a Leucina/deficiência , Óperon/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Sequências Reguladoras de Ácido Nucleico/genética , Regulação para Cima/efeitos dos fármacos , beta-Galactosidase/genética , beta-Galactosidase/metabolismo
16.
Neurochem Int ; 48(6-7): 547-52, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16513216

RESUMO

The system N glutamine (Gln) transporter SN1(SNAT3) is overexpressed in human malignant glioma cells in situ as compared to the adjacent brain tissue or metastases from different organs [Sidoryk, M., Matyja, E., Dybel, A., Zielinska, M., Bogucki, J., Jaskólski, D.J., Liberski, P.P., Kowalczyk, P., Albrecht, J., 2004]. Increased expression of a glutamine transporter SNAT3 is a marker of malignant gliomas. NeuroReport 15, 575-578], but its role in tumor growth as compared to the other Gln transporters is unknown. One of the profound, growth-promoting effects of glial tumor in situ is acidification of the extracellular space. In the kidney SN1(SNAT3) mRNA participates in the adaptation to acidosis. In this study therefore, expression of mRNAs coding for SN1(SNAT3) and other Gln transporters was measured in human (T98G) and rat (C6) glioma cells incubated for 4h in an acidic medium (AI) (pH 6.5). MTT assay revealed no cell loss in AI cells, and intracellular pH (pHi) as measured by a fluorescent probe (BCECF-AM) was slightly alkaline in C6 and T98G cells, indicating that the cells have adapted to AI. AI significantly decreased the SN1(SNAT3) mRNA expression in C6 (a 60% decrease) and T98G cells (a 50% decrease). The decrease retreated in C6 cells 4h after transferring them back to the neutral medium. The expression of ASCT2 mRNA (system ASC), ATA1 mRNA (system A) and SN2(SNAT5) mRNA (system N) were not affected by AI in either of the cell lines. [(3)H]Gln uptake in C6 or T98G cells grown in neutral medium was mainly mediated by system ASCT2: system N contributed to only approximately 7% of the uptake. AI did not affect the total Gln uptake, and only slightly decreased the system N-mediated component of the uptake. Hence, SN1(SNAT3) does not seem to be involved in the adaptation of cultured glioma cells to acidic millieu.


Assuntos
Sistemas de Transporte de Aminoácidos Neutros/biossíntese , Biomarcadores Tumorais/biossíntese , RNA Mensageiro/biossíntese , Sistema A de Transporte de Aminoácidos/biossíntese , Sistema A de Transporte de Aminoácidos/genética , Sistema ASC de Transporte de Aminoácidos/biossíntese , Sistema ASC de Transporte de Aminoácidos/genética , Sistemas de Transporte de Aminoácidos Básicos/biossíntese , Sistemas de Transporte de Aminoácidos Básicos/genética , Sistemas de Transporte de Aminoácidos Neutros/genética , Animais , Biomarcadores Tumorais/genética , Linhagem Celular Tumoral , Sobrevivência Celular , Meios de Cultura , Glioma , Glutamina/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Líquido Intracelular/metabolismo , Antígenos de Histocompatibilidade Menor , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
17.
Ann N Y Acad Sci ; 1053: 435-43, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16179550

RESUMO

High demand of neoplastic tissues for glutamine (Gln) is met by its active transport across cell membranes. Chronic treatment with acrylamide in rodents is associated with an increased incidence of neoplasms, including astrocytomas. In this study, 24-h acrylamide treatment significantly increased the initial rate of l-[G-3H]glutamine uptake in astrocyte cultures derived from the acrylamide-sensitive Fischer 344 rat, and this effect could be fully inhibited by histidine, a model substrate for the amino acid transport system N. RT-PCR analysis revealed that acrylamide treatment caused a significant increase in the astrocytic expression of the mRNA coding for the major system N protein, SNAT3, which is specifically overexpressed in malignant gliomas in situ. The acrylamide-induced upregulation of astrocytic Gln transport via system N is likely to affect Gln homeostasis in these cells and may be causally related to the increased astrocytoma incidence observed in Fischer 344 rats.


Assuntos
Acrilamida/farmacologia , Sistemas de Transporte de Aminoácidos Neutros/biossíntese , Sistemas de Transporte de Aminoácidos/biossíntese , Astrócitos/metabolismo , Glutamina/metabolismo , Animais , Astrócitos/efeitos dos fármacos , Células Cultivadas , Histidina/fisiologia , RNA Mensageiro/biossíntese , Ratos , Ratos Endogâmicos F344 , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Regulação para Cima/efeitos dos fármacos
18.
Neurosci Lett ; 373(1): 79-84, 2005 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-15555781

RESUMO

The occupation of the glycine binding-site is a prerequisite for NMDA receptor activation by glutamate. To analyze the regulation of NMDA receptor function by the glycine transporter 1 (GlyT1), we generated heterozygous constitutive GlyT1 knockout mice (GlyT1tm1.1(+/-)). These animals were fully viable. Using a newly generated antibody, the pattern of GlyT1 expression in brain was found to be unaltered in the mutants while the level of expression was strongly reduced in all brain regions, as shown immunohistochemically. In hippocampal slices the ratio of the peak amplitude of NMDA and AMPA receptor evoked excitatory postsynaptic currents (EPSCs), recorded in CA1 pyramidal cells, was significantly enhanced by 36% in Glyt1tm1.1(+/-) compared to wild-type slices. The frequency and amplitude of AMPA miniature events in Glyt1tm1.1(+/-) mice were indistinguishable from those recorded in wild type. These results provide proof that the NMDA receptor function is enhanced by a reduction of GlyT1 expression. Thus, GlyT1 function is a controlling factor for an enhancement of the NMDA receptor response. These findings are of relevance for the development of GlyT1 inhibitory drugs.


Assuntos
Sistemas de Transporte de Aminoácidos Neutros/biossíntese , Encéfalo/metabolismo , Potenciais Pós-Sinápticos Excitadores/fisiologia , Receptores de N-Metil-D-Aspartato/metabolismo , Sistemas de Transporte de Aminoácidos Neutros/efeitos dos fármacos , Sistemas de Transporte de Aminoácidos Neutros/genética , Animais , Southern Blotting , Western Blotting , Encéfalo/efeitos dos fármacos , Antagonistas de Aminoácidos Excitatórios/farmacologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Glicina/metabolismo , Proteínas da Membrana Plasmática de Transporte de Glicina , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Técnicas de Cultura de Órgãos , Reação em Cadeia da Polimerase , Receptores de AMPA/efeitos dos fármacos , Receptores de AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/efeitos dos fármacos
19.
Arch Pharm Res ; 28(4): 421-32, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15918515

RESUMO

In order to understand the renal reabsorption mechanism of neutral amino acids via amino acid transporters, we have isolated human L-type amino acid transporter 2 (hLAT2) and human T-type amino acid transporter 1 (hTAT1) in human, then, we have examined and compared the gene structures, the functional characterizations and the localization in human kidney. Northern blot analysis showed that hLAT2 mRNA was expressed at high levels in the heart, brain, placenta, kidney, spleen, prostate, testis, ovary, lymph node and the fetal liver. The hTAT1 mRNA was detected at high levels in the heart, placenta, liver, skeletal muscle, kidney, pancreas, spleen, thymus and prostate. Immunohistochemical analysis on the human kidney revealed that the hLAT2 and hTAT1 proteins coexist in the basolateral membrane of the renal proximal tubules. The hLAT2 transports all neutral amino acids and hTAT1 transports aromatic amino acids. The basolateral location of the hLAT2 and hTAT1 proteins in the renal proximal tubule as well as the amino acid transport activity of hLAT2 and hTAT1 suggests that these transporters contribute to the renal reabsorption of neutral and aromatic amino acids in the basolateral domain of epithelial proximal tubule cells, respectively. Therefore, LAT2 and TAT1 play essential roles in the reabsorption of neutral amino acids from the epithelial cells to the blood stream in the kidney. Because LAT2 and TAT1 are essential to the efficient absorption of neutral amino acids from the kidney, their defects might be involved in the pathogenesis of disorders caused by a disruption in amino acid absorption such as blue diaper syndrome.


Assuntos
Sistema y+ de Transporte de Aminoácidos/biossíntese , Sistemas de Transporte de Aminoácidos Neutros/biossíntese , Aminoácidos Neutros/metabolismo , Cadeias Leves da Proteína-1 Reguladora de Fusão/biossíntese , Túbulos Renais Proximais/metabolismo , Absorção , Sequência de Aminoácidos , Sistema y+ de Transporte de Aminoácidos/química , Sistema y+ de Transporte de Aminoácidos/genética , Sistemas de Transporte de Aminoácidos Neutros/química , Sistemas de Transporte de Aminoácidos Neutros/genética , Animais , Sequência de Bases , Transporte Biológico , Northern Blotting , Mapeamento Cromossômico , Cromossomos Humanos Par 14/genética , Clonagem Molecular , Cadeias Leves da Proteína-1 Reguladora de Fusão/química , Cadeias Leves da Proteína-1 Reguladora de Fusão/genética , Humanos , Imuno-Histoquímica , Hibridização in Situ Fluorescente , Técnicas In Vitro , Dados de Sequência Molecular , Oócitos/metabolismo , Xenopus laevis
20.
Neurochem Int ; 88: 32-7, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25842041

RESUMO

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.


Assuntos
Sistema A de Transporte de Aminoácidos/biossíntese , Sistemas de Transporte de Aminoácidos Neutros/biossíntese , Córtex Cerebral/metabolismo , Encefalopatia Hepática/metabolismo , Sistema A de Transporte de Aminoácidos/genética , Sistemas de Transporte de Aminoácidos Neutros/genética , Animais , Proteínas de Transporte/biossíntese , Córtex Cerebral/patologia , Doença Crônica , Regulação da Expressão Gênica , Encefalopatia Hepática/genética , Encefalopatia Hepática/patologia , Masculino , Isoformas de Proteínas/biossíntese , Ratos , Ratos Wistar
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA