Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Neurochem Int ; 129: 104505, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31310779

RESUMO

The green tea amino acid theanine is abundant in green tea rather than black and oolong teas, which are all made of the identical tea plant "Chanoki" (Camellia sinensis). Theanine has a molecular structure close to glutamine (GLN) compared to glutamic acid (Glu), in terms of the absence of a free carboxylic acid moiety from the gamma carbon position. Theanine efficiently inhibits [3H]GLN uptake without affecting [3H]Glu uptake in rat brain synaptosomes. In contrast to GLN, however, theanine markedly stimulates the abilities to replicate and to commit to a neuronal lineage following prolonged exposure in cultured neural progenitor cells (NPCs) prepared from embryonic and adult rodent brains. Upregulation of transcript expression is found for one of the GLN transporter isoforms, Slc38a1, besides the promotion of both proliferation and neuronal commitment along with acceleration of the phosphorylation of mechanistic target of rapamycin (mTOR) and relevant downstream proteins, in murine NPCs cultured with theanine. Stable overexpression of Slc38a1 similarly facilitates both cellular replication and neuronal commitment in pluripotent embryonic carcinoma P19 cells. In P19 cells with stable overexpression of Slc38a1, marked phosphorylation is seen for mTOR and downstream proteins in a manner insensitive to further additional phosphorylation by theanine. Taken together, theanine would exhibit a novel pharmacological property to up-regulate Slc38a1 expression for activation of the intracellular mTOR signaling pathway required for neurogenesis after sustained exposure in undifferentiated NPCs in the brain. In this review, a novel neurogenic property of the green tea amino acid theanine is summarized for embryonic and adult neurogenesis with a focus on the endogenous amino acid GLN on the basis of our accumulating evidence to date.


Assuntos
Encéfalo/efeitos dos fármacos , Glutamatos/farmacologia , Glutamina/fisiologia , Células-Tronco Neurais/efeitos dos fármacos , Neurogênese/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Sistema A de Transporte de Aminoácidos/fisiologia , Animais , Transtornos Cognitivos/tratamento farmacológico , Transtornos Cognitivos/prevenção & controle , Método Duplo-Cego , Previsões , Glutamatos/química , Glutamatos/uso terapêutico , Glutamina/química , Hipocampo/efeitos dos fármacos , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Camundongos , Camundongos Transgênicos , Transtornos do Humor/tratamento farmacológico , Células-Tronco Neurais/fisiologia , Neurogênese/fisiologia , Fosforilação/efeitos dos fármacos , Fitoterapia , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Ensaios Clínicos Controlados Aleatórios como Assunto , Ratos , Transtornos do Sono-Vigília/tratamento farmacológico , Serina-Treonina Quinases TOR/metabolismo , Chá/química
2.
Taiwan J Obstet Gynecol ; 57(2): 217-226, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29673664

RESUMO

OBJECTIVE: To validate the gene expression profile obtained from the previous microarray analysis and to further study the biological functions of these genes in endometrial cancer. From our previous study, we identified 621 differentially expressed genes in laser-captured microdissected endometrioid endometrial cancer as compared to normal endometrial cells. Among these genes, 146 were significantly up-regulated in endometrial cancer. MATERIALS AND METHODS: A total of 20 genes were selected from the list of up-regulated genes for the validation assay. The qPCR confirmed that 19 out of the 20 genes were up-regulated in endometrial cancer compared with normal endometrium. RNA interference (RNAi) was used to knockdown the expression of the upregulated genes in ECC-1 and HEC-1A endometrial cancer cell lines and its effect on proliferation, migration and invasion were examined. RESULTS: Knockdown of MIF, SOD2, HIF1A and SLC7A5 by RNAi significantly decreased the proliferation of ECC-1 cells (p < 0.05). Our results also showed that the knockdown of MIF, SOD2 and SLC7A5 by RNAi significantly decreased the proliferation and migration abilities of HEC-1A cells (p < 0.05). Moreover, the knockdown of SLC38A1 and HIF1A by RNAi resulted in a significant decrease in the proliferation of HEC1A cells (p < 0.05). CONCLUSION: We have identified the biological roles of SLC38A1, MIF, SOD2, HIF1A and SLC7A5 in endometrial cancer, which opens up the possibility of using the RNAi silencing approach to design therapeutic strategies for treatment of endometrial cancer.


Assuntos
Neoplasias do Endométrio/genética , Inativação Gênica , Sistema A de Transporte de Aminoácidos/genética , Sistema A de Transporte de Aminoácidos/fisiologia , Linhagem Celular Tumoral , Proliferação de Células/genética , Neoplasias do Endométrio/patologia , Feminino , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/fisiologia , Oxirredutases Intramoleculares/genética , Oxirredutases Intramoleculares/fisiologia , Transportador 1 de Aminoácidos Neutros Grandes/genética , Transportador 1 de Aminoácidos Neutros Grandes/fisiologia , Fatores Inibidores da Migração de Macrófagos/genética , Fatores Inibidores da Migração de Macrófagos/fisiologia , Interferência de RNA , RNA Interferente Pequeno , Reação em Cadeia da Polimerase em Tempo Real , Superóxido Dismutase/genética , Superóxido Dismutase/fisiologia , Regulação para Cima
3.
J Biol Chem ; 291(25): 13194-205, 2016 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-27129276

RESUMO

Many cancer cells depend on glutamine as they use the glutaminolysis pathway to generate building blocks and energy for anabolic purposes. As a result, glutamine transporters are essential for cancer growth and are potential targets for cancer chemotherapy with ASCT2 (SLC1A5) being investigated most intensively. Here we show that HeLa epithelial cervical cancer cells and 143B osteosarcoma cells express a set of glutamine transporters including SNAT1 (SLC38A1), SNAT2 (SLC38A2), SNAT4 (SLC38A4), LAT1 (SLC7A5), and ASCT2 (SLC1A5). Net glutamine uptake did not depend on ASCT2 but required expression of SNAT1 and SNAT2. Deletion of ASCT2 did not reduce cell growth but caused an amino acid starvation response and up-regulation of SNAT1 to replace ASCT2 functionally. Silencing of GCN2 in the ASCT2(-/-) background reduced cell growth, showing that a combined targeted approach would inhibit growth of glutamine-dependent cancer cells.


Assuntos
Sistema ASC de Transporte de Aminoácidos/genética , Sistema A de Transporte de Aminoácidos/fisiologia , Proliferação de Células , Deleção de Genes , Expressão Gênica , Glutamina/metabolismo , Células HeLa , Homeostase , Humanos , Transporte de Íons , Redes e Vias Metabólicas , Antígenos de Histocompatibilidade Menor
4.
Placenta ; 31(11): 1003-9, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20801504

RESUMO

The purpose of this study was to clarify the cytoprotective mechanism(s) induced in a conditionally immortalized syncytiotrophoblast cell line (TR-TBT 18d-1) exposed to hypertonic conditions. Hypertonicity-induced apoptosis of TR-TBT 18d-1 cells, but this was blocked by addition of 1 mM taurine to the culture medium. TauT-knockdown using siRNA revealed that TauT is a major contributor to taurine uptake by TR-TBT 18d-1 cells, at least under normal conditions. Cellular uptake of [(3)H]taurine and [(14)C]betaine by TR-TBT 18d-1 cells cultured under hypertonic conditions was increased compared to that under normal conditions. TauT, BGT-1, ATA2 and HSP70 mRNAs were upregulated by hypertonicity, while OCTN2, ENT1 and CNT1 mRNAs were downregulated. [(3)H]Taurine uptake was strongly inhibited by TauT inhibitors such as hypotaurine and ß-alanine. MeAIB, a system A specific substrate, inhibited hypertonic stress-induced [(14)C]betaine uptake. These results suggest that TauT and system A play cytoprotective roles in syncytiotrophoblasts exposed to hypertonic stress.


Assuntos
Sistema A de Transporte de Aminoácidos/fisiologia , Citoproteção , Glicoproteínas de Membrana/fisiologia , Proteínas de Membrana Transportadoras/fisiologia , Estresse Fisiológico , Taurina/metabolismo , Trofoblastos/patologia , Sistema A de Transporte de Aminoácidos/antagonistas & inibidores , Sistema A de Transporte de Aminoácidos/genética , Animais , Apoptose , Betaína/metabolismo , Transporte Biológico/efeitos dos fármacos , Linhagem Celular , Regulação da Expressão Gênica no Desenvolvimento , Inativação Gênica , Soluções Hipertônicas , Glicoproteínas de Membrana/antagonistas & inibidores , Glicoproteínas de Membrana/genética , Moduladores de Transporte de Membrana/farmacologia , Proteínas de Membrana Transportadoras/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno , Ratos , Taurina/análogos & derivados , Trofoblastos/efeitos dos fármacos , Trofoblastos/metabolismo , beta-Alanina/análogos & derivados , beta-Alanina/metabolismo , beta-Alanina/farmacologia
5.
Am J Physiol Cell Physiol ; 297(5): C1228-35, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19741197

RESUMO

Changes in placental nutrient transport are closely associated with abnormal fetal growth. However, the molecular mechanisms underlying the regulation of placental amino acid transporters are unknown. We demonstrate that physiological concentrations of the proinflammatory cytokines interleukin (IL)-6 and tumor necrosis factor (TNF)-alpha stimulate the activity of amino acid transporter system A, but not system L, in cultured human primary trophoblast cells. Both cytokines increased the gene and protein expression of the Na(+)-coupled neutral amino acid transporter (SNAT)2 isoform and upregulated SNAT1 protein expression. IL-6 increased Tyr705 phosphorylation of signal transducer and activator of transcription 3 (STAT3). In cells transfected with small interfering RNA (siRNA) targeting STAT3, the RNA and protein expression of SNAT2, but not SNAT1, was reduced and the stimulating effect of IL-6 on system A activity was abolished. Despite eliciting similar responses in amino acid transport activity and transporter expression, TNF-alpha effects on system A activity were not mediated through the JAK/STAT pathway. In conclusion, we have identified a novel regulatory pathway involving increased gene expression of the SNAT2 isoform mediated by a STAT-dependent pathway, which links IL-6 to increased activity of system A, a ubiquitously expressed transporter of neutral amino acids. From these new findings, we propose that upregulation of amino acid transporters by cytokines may contribute to increased placental nutrient transport and fetal overgrowth, which are commonly found in pregnancies complicated by maternal diabetes and obesity.


Assuntos
Sistema A de Transporte de Aminoácidos/metabolismo , Regulação da Expressão Gênica , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/fisiologia , Trofoblastos/metabolismo , Sistema A de Transporte de Aminoácidos/genética , Sistema A de Transporte de Aminoácidos/fisiologia , Western Blotting , Células Cultivadas , Ensaio de Imunoadsorção Enzimática , Feminino , Expressão Gênica , Humanos , Interleucina-6 , Gravidez , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Necrose Tumoral alfa/metabolismo , Regulação para Cima
6.
Am J Physiol Gastrointest Liver Physiol ; 295(2): G260-72, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18556422

RESUMO

Mechanisms by which gut luminal content regulates secretion and motility are ill understood. We evaluated whether neuroendocrine enterochromaffin (EC) cells act as luminal sensors for a wide variety of nutrients and defined the secretory mechanisms of this process. Pure (98-99%) FACS-sorted human EC cells and neoplastic EC cells (KRJ-I) were studied. RT-PCR identified transcripts for T2R1 (bitter), OR1G1 (class II olfactory) and trace amine (TAR1) G protein-coupled receptors (GPCRs) and transporters for glutamine (SNAT1/2), glucose (GLUT1/3/SGLT1), and bile salts (ABST). Glutamine and sodium deoxycholate stimulated 5-HT release (EC(50) = 0.002-0.2 microM; 2-fold release) but were 10-100 times more potent in neoplastic EC cells, which also secreted 6-13 times more 5-HT. Tastants (caffeine, tyramine, octopamine) and olfactants (thymol and eugenol) also stimulated normal and neoplastic EC cell 5-HT secretion (EC(50) = 1.2 nM to 2.1 microM and 0.05 nM to 0.1 microM release, respectively); 2-deoxyglucose and the artificial sweetener sucralose also stimulated (EC(50) = 9.2 and 0.38 nM). 5-HT release was associated with ERK phosphorylation (1.5-fold, P < 0.02) and could be inhibited by a somatostatin analog (IC(50) = 1 pM). Eleven secretory associated genes including the vesicle docking inhibitor STXBP3 were upregulated in response to glutamine and bile salt stimulation in neoplastic EC cells. Targeting STXBP3 expression by use of antisense knockdown significantly (P < 0.05) reduced 5-HT secretion. In conclusion, EC cells express GPCRs and transporters for luminal tastants, olfactants, glutamine, glucose, and bile salts. Activation includes a panel of secretory genes, ERK phosphorylation, and 5-HT secretion. Luminal EC cell regulation is likely to be as important as G cell regulation in gastric acid secretion; development of agents to target EC cell function is therefore a critical therapeutic goal.


Assuntos
Ácidos e Sais Biliares/fisiologia , Tumor Carcinoide/metabolismo , Células Enterocromafins/metabolismo , Neoplasias Intestinais/metabolismo , Serotonina/metabolismo , Sistema A de Transporte de Aminoácidos/fisiologia , Cafeína/farmacologia , Tumor Carcinoide/genética , Linhagem Celular Tumoral , Ácido Desoxicólico/farmacologia , Desoxiglucose/farmacologia , Células Enterocromafins/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Proteínas Facilitadoras de Transporte de Glucose/fisiologia , Glutamina/farmacologia , Humanos , Neoplasias Intestinais/genética , Intestino Delgado , Transportadores de Ânions Orgânicos Dependentes de Sódio/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Receptores de Glutamato Metabotrópico/fisiologia , Receptores Odorantes/fisiologia , Transportador 1 de Glucose-Sódio/fisiologia , Somatostatina/farmacologia , Sacarose/análogos & derivados , Sacarose/farmacologia , Simportadores/fisiologia , Tirosina/farmacologia
7.
Arch Biochem Biophys ; 446(2): 119-30, 2006 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-16455044

RESUMO

Hyperhomocysteinemia is an independent risk factor for cardiovascular disease. Transport of L-homocysteine into and out of the human vascular endothelium is poorly understood. We hypothesized that cultured human aortic endothelial cells (HAEC) would import L-homocysteine on one or more of the L-cysteine transport systems. Inhibitors of the transporters were used to characterize the uptake of [35S]L-homocysteine, [35S]L-homocystine, and [35S]L-cysteine. We found that L-homocysteine uptake is mediated by the sodium-dependent cysteine transport systems X(AG), ASC, and A, and the sodium-independent transport system L. Thus, HAEC utilize multiple cysteine transporters (X(AG) > or = L > ASC > A) to import L-homocysteine. Kinetic analysis supported the uptake results. Michaelis-Menten constants (Km) for the four systems yielded values of 19.0, 27.1, 112, and 1000 microM for systems L, X(AG), ASC, and A, respectively. The binding and uptake of [35S]L-homocystine, the disulfide homodimer of L-homocysteine, was mediated by systems X(AG), L, and ASC but not by system A. In contrast to [35S]L-homocysteine, system x(c) was active for [35S]L-homocystine uptake. A similar pattern was observed for [35S]L-cysteine. Thus, L-homocysteine and L-homocystine found in hyperhomocysteinemic subjects can gain entry into the vascular endothelium by way of multiple L-cysteine transporters.


Assuntos
Células Endoteliais/metabolismo , Homocisteína/metabolismo , Sistema A de Transporte de Aminoácidos/fisiologia , Sistema ASC de Transporte de Aminoácidos/fisiologia , Sistema L de Transporte de Aminoácidos/fisiologia , Sistema X-AG de Transporte de Aminoácidos/fisiologia , Aorta Torácica/citologia , Transporte Biológico Ativo , Células Cultivadas , Cisteína/metabolismo , Cisteína/farmacocinética , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Homocisteína/farmacocinética , Humanos , Ligação Proteica , Radioisótopos de Enxofre
8.
J Soc Gynecol Investig ; 12(7): 495-503, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16202926

RESUMO

OBJECTIVE: The current study sought to investigate the influence of interleukin-1beta (IL-1beta) on the function of the amino acid transport system A in trophoblasts. METHODS: BeWo choriocarcinoma cells were exposed to recombinant human IL-1beta in serum-free medium. Cells incubated with serum-free medium in the absence of IL-1beta were used as control. System A activity was determined in control and treated cells by measuring the uptake of alpha-(methylamino)isobutyric acid. The results obtained were confirmed by measuring system A activity in placental brush border membrane vesicles isolated from pregnant rats injected with IL-1beta. RESULTS: Treatment of BeWo cells with IL-1beta resulted in a time- and dose- dependent inhibition of system A. Treatment with IL-1beta also inhibited the uptake of arginine, and glutamate but had no significant effect on the uptake of leucine, tryptophan, and ascorbate. The inhibition of system A activity by IL-1beta was abolished in the presence of IL-1beta receptor antagonist. The inhibitory effect was associated with a decrease in the maximal velocity of the transport system with no effect on the substrate affinity. Steady-state levels of both SNAT1 and SNAT2 mRNA were reduced by IL-1beta treatment as evidenced by semiquantitative reverse transcriptase-polymerase chain reaction (RT-PCR) analysis. In rat placental brush border membrane vesicles isolated from IL-1beta-treated pregnant rats, system A activity was found to be decreased by approximately 40% compared to activity in control membrane vesicles. CONCLUSIONS: IL-1beta decreases SNAT1 and SNAT2 mRNA levels in trophoblasts, which is associated with a decrease in system A-mediated transport activity at the functional level. These findings may have important consequences under both physiologic conditions and pathologic conditions during pregnancy that are associated with elevated levels of IL-1beta.


Assuntos
Sistema A de Transporte de Aminoácidos/fisiologia , Interleucina-1/fisiologia , Gravidez/fisiologia , Trofoblastos/fisiologia , Sistema ASC de Transporte de Aminoácidos/fisiologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Humanos , RNA Mensageiro/análise , Células Tumorais Cultivadas/patologia , Neoplasias Uterinas/patologia
9.
Yakugaku Zasshi ; 124(11): 791-802, 2004 Nov.
Artigo em Japonês | MEDLINE | ID: mdl-15516806

RESUMO

The blood-brain barrier (BBB) segregates the circulating blood from interstitial fluid in the brain and restricts drug permeability into the brain. Our latest studies have revealed that the BBB transporters play important physiological roles in maintaining the brain environment. For an energy-storing system, the creatine transporter localized at the brain capillary endothelial cells (BCECs) mediates the supply of creatine from the blood to the brain. The BBB is involved in the brain-to-blood efflux transport of gamma-aminobutyric acid, and GAT2/BGT-1 mediates this transport process. BCECs also express serotonin and norepinephrine transporters. Organic anion transporter 3 (OAT3) and ASCT2 are localized at the abluminal membrane of the BCECs. OAT3 is involved in the brain-to-blood efflux of a dopamine metabolite, a uremic toxin, and thiopurine nucleobase analogues. ASCT2 plays a role in L-isomer-selective aspartic acid efflux transport at the BBB. Dehydroepiandrosterone sulfate and small neutral amino acids undergo brain-to-blood efflux transport mediated by organic anion transporting polypeptide 2 and ATA2, respectively. The BBB transporters are regulated by various factors: ATA2 by osmolarity, taurine transporter by tumor necrosis factor-alpha, and L-cystine/L-glutamic acid exchange transporter by oxidative stress. Clarifying the physiological roles of BBB transport systems should give important information allowing the development of better central nervous system (CNS) drugs and improving our understanding of the relationship between CNS disorders and BBB function.


Assuntos
Barreira Hematoencefálica/fisiologia , Proteínas de Membrana Transportadoras/fisiologia , Neurotransmissores/metabolismo , Sistema A de Transporte de Aminoácidos/fisiologia , Sistema ASC de Transporte de Aminoácidos/fisiologia , Aminoácidos/metabolismo , Animais , Transporte Biológico , Barreira Hematoencefálica/metabolismo , Encéfalo/irrigação sanguínea , Fármacos do Sistema Nervoso Central , Desenho de Fármacos , Células Endoteliais/metabolismo , Proteínas da Membrana Plasmática de Transporte de GABA , Humanos , Transportador 1 de Ânion Orgânico Específico do Fígado/fisiologia , Proteínas de Membrana Transportadoras/metabolismo , Antígenos de Histocompatibilidade Menor , Transportadores de Ânions Orgânicos Sódio-Independentes/fisiologia , Concentração Osmolar , Estresse Oxidativo/fisiologia , Fator de Necrose Tumoral alfa/fisiologia
10.
Pflugers Arch ; 447(5): 784-95, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12845534

RESUMO

The sodium-coupled neutral amino acid transporters (SNAT) of the SLC38 gene family resemble the classically-described System A and System N transport activities in terms of their functional properties and patterns of regulation. Transport of small, aliphatic amino acids by System A subtypes (SNAT1, SNAT2, and SNAT4) is rheogenic and pH sensitive. The System N subtypes SNAT3 and SNAT5 also countertransport H(+), which may be key to their operation in reverse, and have narrower substrate profiles than do the System A subtypes. Glutamine emerges as a favored substrate throughout the family, except for SNAT4. The SLC38 transporters undoubtedly play many physiological roles including the transfer of glutamine from astrocyte to neuron in the CNS, ammonia detoxification and gluconeogenesis in the liver, and the renal response to acidosis. Probing their regulation has revealed additional roles, and recent work has considered SLC38 transporters as therapeutic targets in neoplasia.


Assuntos
Sistema A de Transporte de Aminoácidos/fisiologia , Aminoácidos Neutros/metabolismo , Sódio/metabolismo , Sequência de Aminoácidos , Sistema A de Transporte de Aminoácidos/química , Sistema A de Transporte de Aminoácidos/genética , Animais , Transporte Biológico/fisiologia , Humanos , Dados de Sequência Molecular , Família Multigênica/fisiologia
11.
J Biol Chem ; 278(26): 23720-30, 2003 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-12684517

RESUMO

Glutamine, the preferred precursor for neurotransmitter glutamate and GABA, is likely to be the principal substrate for the neuronal System A transporter SNAT1 in vivo. We explored the functional properties of SNAT1 (the product of the rat Slc38a1 gene) by measuring radiotracer uptake and currents associated with SNAT1 expression in Xenopus oocytes and determined the neuronal-phenotypic and cellular distribution of SNAT1 by confocal laser-scanning microscopy alongside other markers. We found that SNAT1 mediates transport of small, neutral, aliphatic amino acids including glutamine (K0.5 approximately 0.3 mm), alanine, and the System A-specific analogue 2-(methylamino)isobutyrate. Amino acid transport is driven by the Na+ electrochemical gradient. The voltage-dependent binding of Na+ precedes that of the amino acid in a simultaneous transport mechanism. Li+ (but not H+) can substitute for Na+ but results in reduced Vmax. In the absence of amino acid, SNAT1 mediates Na+-dependent presteady-state currents (Qmax approximately 9 nC) and a nonsaturable cation leak with selectivity Na+, Li+ >> H+, K+. Simultaneous flux and current measurements indicate coupling stoichiometry of 1 Na+ per 1 amino acid. SNAT1 protein was detected in somata and proximal dendrites but not nerve terminals of glutamatergic and GABAergic neurons throughout the adult CNS. We did not detect SNAT1 expression in astrocytes but detected its expression on the luminal membranes of the ependyma. The functional properties and cellular distribution of SNAT1 support a primary role for SNAT1 in glutamine transport serving the glutamate/GABA-glutamine cycle in central neurons. Localization of SNAT1 to certain dopaminergic neurons of the substantia nigra and cholinergic motoneurons suggests that SNAT1 may play additional specialized roles, providing metabolic fuel (via alpha-ketoglutarate) or precursors (cysteine, glycine) for glutathione synthesis.


Assuntos
Sistema A de Transporte de Aminoácidos/metabolismo , Sistema Nervoso Central/citologia , Neurônios/química , Sistema A de Transporte de Aminoácidos/fisiologia , Aminoácidos Neutros/metabolismo , Animais , Cátions Monovalentes , Sistema Nervoso Central/química , DNA Complementar , Glutamina/metabolismo , Cinética , Microinjeções , Microscopia de Fluorescência , Oócitos , Técnicas de Patch-Clamp , Ratos , Distribuição Tecidual , Xenopus
12.
Int J Mol Med ; 9(3): 271-9, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11836633

RESUMO

The transport of amino acids across the plasma membrane is a process of fundamental physiological importance. If this process is modified by estrogens and if estrogen receptors play a role in this regulation, then the alteration of metabolic events will be of significant importance in cancer cells which have high estrogen receptor content because estrogens modify cellular physiology through transactivational effects of their cognate receptors. In the present study, we investigated the role of 17 beta-estradiol on the regulation of different amino acid transport systems, in particular, Systems A, ASC and y+ in estrogen receptor positive (MCF-7, T47D, H-301) and estrogen receptor negative (MCF-10, SKBR-3, MDA-MB-231) cell lines. The cells were treated with 17 beta-estradiol (10 nM), ICI 182780 (1 microM), or 17 beta-estradiol plus ICI 182780. We discovered that 17 beta-estradiol specifically stimulates System A activity by 2- to 4-fold in estrogen receptor positive cell lines with a maximal stimulation 48 h after estrogen-treatment while no stimulation was observed in estrogen receptor negative cell lines. Estrogen-dependent activation of System A was inhibited by co-treatment with the antiestrogen ICI 182780. Northern blot analysis showed that System A mRNA levels are also increased following estrogen treatment, and this induction of mRNA transcript levels by estrogen can be inhibited by co-treatment with antiestrogen ICI 182780. The increase in System A transport activity following estrogen treatment was abbrogated when estrogen receptor positive cells were stably transfected with human antisense ER alpha cDNA. Kinetic analysis demonstrated that estrogen-induced stimulation results in a doubling of Vmax with no changes in Km. However, 17 beta-estradiol did not stimulate the activation of transport systems responsible for the transport of arginine (y+) or threonine (ASC). In summary, we have provided evidence that estrogen receptors play a role in the activation of System A by estrogen. This adaptation may have important physiological and nutritional significance on estrogen dependent growth of breast tumors.


Assuntos
Sistema A de Transporte de Aminoácidos/fisiologia , Neoplasias da Mama/fisiopatologia , Estradiol/fisiologia , Receptores de Estrogênio/fisiologia , Células Tumorais Cultivadas/fisiologia , Sistema y+ de Transporte de Aminoácidos/fisiologia , Animais , Cricetinae , Receptor alfa de Estrogênio , Feminino , Humanos , Rim/metabolismo
13.
Langenbecks Arch Chir Suppl Kongressbd ; 115(Suppl I): 635-9, 1998.
Artigo em Alemão | MEDLINE | ID: mdl-14518332

RESUMO

Positron emission tomography (PET) with fluorine-18-fluorodeoxyglucose (FDG), carbon-11-labeled aminoisobutyric acid (AIB) and oxygen-15-labeled water (15O-labeled water) was evaluated for imaging soft tissue sarcomas (STS). Eleven pathologically confirmed STS were evaluated by PET using FDG, AIB and 15O-labeled water. All radiotracers showed an increased activity in viable tumor with a significant differentiation from normal tissue (muscle). An increased glucose metabolism in STS allowed a differentiation from normal tissue (muscle) with FDG. Furthermore, STS showed an increased AIB activity suggesting an increased "A-type" aminoacid transport. AIB therefore can be used for evaluation of STS. 15O-labeled water demonstrated an increased uptake with an overlap with blood vessel activity. These radiotracers supply information about biological properties of STS. Five patients with suspicious lesions in conventional radiodiagnostic but negative PET scans were free of local recurrence, diagnosed either by surgery or by follow-up. Two patients with PET findings suggesting local failure, ultimately demonstrated local recurrence. The differentiation of local recurrence from benign postoperative tissue alterations is encouraging and requires further evaluation.


Assuntos
Sistema A de Transporte de Aminoácidos/fisiologia , Glicemia/metabolismo , Proteínas de Neoplasias/metabolismo , Sarcoma/diagnóstico por imagem , Neoplasias de Tecidos Moles/diagnóstico por imagem , Tomografia Computadorizada de Emissão/métodos , Ácidos Aminoisobutíricos , Radioisótopos de Carbono , Fluordesoxiglucose F18 , Humanos , Infusões Intravenosas , Radioisótopos de Oxigênio , Fluxo Sanguíneo Regional/fisiologia , Sarcoma/irrigação sanguínea , Sarcoma/fisiopatologia , Sensibilidade e Especificidade , Neoplasias de Tecidos Moles/irrigação sanguínea , Neoplasias de Tecidos Moles/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA