Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Br J Pharmacol ; 173(23): 3292-3306, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27747870

RESUMO

BACKGROUND AND PURPOSE: Pancreatic cancer is a solid tumour that is often fatal. Hence, there is an urgent need to identify new drug targets for this disease. Highly proliferating cancer cells have an increased demand for nutrients and, therefore, need to up-regulate selective amino acid transporters. Here, we investigated which amino acid transporters are up-regulated in pancreatic cancer and whether any of these transporters has potential as a drug target for this fatal disease. EXPERIMENTAL APPROACH: The expression of amino acid transporters in pancreatic cancer was analysed using publicly available microarray datasets, and the findings with the transporter SLC6A14 were validated by mRNA and protein analysis. The potential of SLC6A14 as a drug target was evaluated using a pharmacological blocker in vitro and in vivo. KEY RESULTS: SLC6A14 was up-regulated several fold in patient-derived xenografts, primary tumour tissues and pancreatic cancer cells lines compared to normal pancreatic tissue or normal pancreatic epithelial cells. The magnitude of the up-regulation of SLC6A14 was the highest among the amino acid transporters examined. A pharmacological blocker of SLC6A14, α-methyltryptophan, induced amino acid starvation in pancreatic cancer cells and reduced the growth and proliferation of these cells, both in vitro and in vivo. CONCLUSION AND IMPLICATIONS: The salient features of this study are that SLC6A14 is markedly up-regulated in pancreatic cancer and that pharmacological blockade of this transporter interferes with amino acid nutrition and reduces growth and proliferation of pancreatic cancer cells. These findings identify SLC6A14 as a novel druggable target for pancreatic cancer.


Assuntos
Sistemas de Transporte de Aminoácidos Neutros/genética , Terapia de Alvo Molecular , Neoplasias Pancreáticas/tratamento farmacológico , Triptofano/análogos & derivados , Sistemas de Transporte de Aminoácidos , Sistemas de Transporte de Aminoácidos Neutros/antagonistas & inibidores , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Camundongos Nus , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , RNA Mensageiro/metabolismo , Triptofano/farmacologia , Regulação para Cima , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Indian J Cancer ; 50(1): 41-5, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23713043

RESUMO

BACKGROUND: Extra gastrointestinal stromal tumors (EGIST) are uncommon compared to their gastrointestinal counterparts. EGISTs involve omentum, mesentery, retroperitoneum, pancreas, and pelvis. MATERIALS AND METHODS: Ten EGISTs were analyzed in this study from January 1995 to November 2011. They were analyzed with respect to clinical features, imageological, histopathological, and immunohistochemical findings. The immunohistochemical stains used were Smooth muscle actin (SMA), Desmin, S-100 protein, CD34 and CD-117. RESULTS: There was slight female preponderance with wide age range. Four of the tumors were in retroperitoneum, three in mesentery, and two in omentum and one in pelvis. Histopathologically majority were spindle cell tumors. Immunohistochemically CD117 was consistently positive followed by CD34. Smooth muscle actin was positive in eight cases, S-100 protein and desmin were positive in two cases each. CONCLUSION: EGISTs are rare and should be considered in the differential diagnosis of the mesenchymal tumors and immunohistochemistry helps to confirm the diagnosis. Further study with better follow-up is desired to characterize these uncommon tumors.


Assuntos
Neoplasias de Tecido Conjuntivo/diagnóstico , Neoplasias Peritoneais/diagnóstico , Sarcoma/diagnóstico , Abdome/diagnóstico por imagem , Abdome/patologia , Actinas/metabolismo , Adulto , Idoso , Antígenos CD34/metabolismo , Diagnóstico Diferencial , Feminino , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Neoplasias de Tecido Conjuntivo/metabolismo , Neoplasias de Tecido Conjuntivo/patologia , Neoplasias Peritoneais/metabolismo , Neoplasias Peritoneais/patologia , Proteínas Proto-Oncogênicas c-kit/metabolismo , Radiografia Abdominal , Cintilografia , Sarcoma/metabolismo , Sarcoma/patologia , Centros de Atenção Terciária , Adulto Jovem
3.
Biochim Biophys Acta ; 1832(8): 1149-58, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23579073

RESUMO

Elevated levels of homocysteine produce detrimental effects in humans but its role in preterm birth is not known. Here we used a mouse model of hyperhomocysteinemia to examine the relevance of homocysteine to preterm birth. The mouse carries a heterozygous deletion of cystathionine ß-synthase (Cbs(+/-)). Gestational period was monitored in wild type and Cbs(+/-) female mice. Mouse uterine and placental tissues, human primary trophoblast cells, and human myometrial and placental cell lines were used to determine the influence of homocysteine on expression of specific genes in vitro. The activity of BKCa channel in the myometrial cell line was monitored using the patch-clamp technique. We found that hyperhomocysteinemia had detrimental effects on pregnancy and induced preterm birth in mice. Homocysteine increased the expression of oxytocin receptor and Cox-2 as well as PGE2 production in uterus and placenta, and initiated premature uterine contraction. A Cox-2 inhibitor reversed these effects. Gpr109a, a receptor for niacin, induced Cox-2 in uterus. Homocysteine upregulated GPR109A and suppressed BKCa channel activity in human myometrial cells. Deletion of Gpr109a in Cbs(+/-) mice reversed premature birth. We conclude that hyperhomocysteinemia causes preterm birth in mice through upregulation of the Gpr109a/Cox-2/PGE2 axis and that pharmacological blockade of Gpr109a may have potential in prevention of preterm birth.


Assuntos
Homocisteína/sangue , Hiper-Homocisteinemia/fisiopatologia , Complicações na Gravidez/sangue , Nascimento Prematuro/sangue , Animais , Linhagem Celular , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Inibidores de Ciclo-Oxigenase 2/farmacologia , Cistationina beta-Sintase/genética , Cistationina beta-Sintase/metabolismo , Dinoprostona/genética , Dinoprostona/metabolismo , Feminino , Homocisteína/genética , Homocisteína/metabolismo , Humanos , Hiper-Homocisteinemia/genética , Hiper-Homocisteinemia/metabolismo , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/genética , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Contração Muscular , Miométrio/metabolismo , Miométrio/fisiopatologia , Placenta/metabolismo , Placenta/fisiopatologia , Gravidez , Complicações na Gravidez/genética , Complicações na Gravidez/fisiopatologia , Nascimento Prematuro/genética , Nascimento Prematuro/fisiopatologia , Receptores Nicotínicos/genética , Receptores Nicotínicos/metabolismo , Receptores de Ocitocina/genética , Receptores de Ocitocina/metabolismo , Trofoblastos/metabolismo , Regulação para Cima , Útero/metabolismo , Útero/patologia , Útero/fisiopatologia
4.
Oncogene ; 30(38): 4026-37, 2011 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-21499304

RESUMO

There has been growing interest among the public and scientists in dichloroacetate (DCA) as a potential anticancer drug. Credible evidence exists for the antitumor activity of this compound, but high concentrations are needed for significant therapeutic effect. Unfortunately, these high concentrations produce detrimental side effects involving the nervous system, thereby precluding its use for cancer treatment. The mechanistic basis of the compound's antitumor activity is its ability to activate the pyruvate dehydrogenase complex through inhibition of pyruvate dehydrogenase kinase. As the compound inhibits the kinase at micromolar concentrations, it is not known why therapeutically prohibitive high doses are needed for suppression of tumor growth. We hypothesized that lack of effective mechanisms for the entry of DCA into tumor cells may underlie this phenomenon. Here we show that SLC5A8 transports DCA very effectively with high affinity. This transporter is expressed in normal cells, but expression is silenced in tumor cells by epigenetic mechanisms. The lack of the transporter makes tumor cells resistant to the antitumor activity of DCA. However, if the transporter is expressed in tumor cells ectopically, the cells become sensitive to the drug at low concentrations. This is evident in breast cancer cells, colon cancer cells and prostate cancer cells. Normal cells, which constitutively express the transporter, are however not affected by the compound, indicating tumor cell-selective therapeutic activity. The mechanism of the compound's antitumor activity still remains its ability to inhibit pyruvate dehydrogenase kinase and force mitochondrial oxidation of pyruvate. As silencing of SLC5A8 in tumors involves DNA methylation and its expression can be induced by treatment with DNA methylation inhibitors, our findings suggest that combining DCA with a DNA methylation inhibitor would offer a means to reduce the doses of DCA to avoid detrimental effects associated with high doses but without compromising antitumor activity.


Assuntos
Antineoplásicos/farmacologia , Proteínas de Transporte de Cátions/fisiologia , Ácido Dicloroacético/farmacologia , Proteínas Supressoras de Tumor/fisiologia , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Ácido Dicloroacético/farmacocinética , Humanos , Proteínas de Membrana Transportadoras/fisiologia , Transportadores de Ácidos Monocarboxílicos , Ácido Pirúvico/metabolismo , Sódio/metabolismo , Xenopus laevis
5.
Am J Physiol Cell Physiol ; 296(1): C142-50, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18987252

RESUMO

The activity of placental amino acid transporters is decreased in intrauterine growth restriction (IUGR), but the underlying regulatory mechanisms have not been established. Inhibition of the mammalian target of rapamycin (mTOR) signaling pathway has been shown to decrease the activity of the system L amino acid transporter in human placental villous fragments, and placental mTOR activity is decreased in IUGR. In the present study, we used cultured primary trophoblast cells to study mTOR regulation of placental amino acid transporters in more detail and to test the hypothesis that mTOR alters amino acid transport activity by changes in transporter expression. Inhibition of mTOR by rapamycin significantly reduced the activity of system A (-17%), system L (-28%), and taurine (-40%) amino acid transporters. mRNA expression of isoforms of the three amino acid transporter systems in response to mTOR inhibition was measured using quantitative real-time PCR. mRNA expression of l-type amino acid transporter 1 (LAT1; a system L isoform) and taurine transporter was reduced by 13% and 50%, respectively; however, mTOR inhibition did not alter the mRNA expression of system A isoforms (sodium-coupled neutral amino acid transporter-1, -2, and -4), LAT2, or 4F2hc. Rapamycin treatment did not significantly affect the protein expression of any of the transporter isoforms. We conclude that mTOR signaling regulates the activity of key placental amino acid transporters and that this effect is not due to a decrease in total protein expression. These data suggest that mTOR regulates placental amino acid transporters by posttranslational modifications or by affecting transporter translocation to the plasma membrane.


Assuntos
Sistemas de Transporte de Aminoácidos/metabolismo , Proteínas Quinases/metabolismo , Trofoblastos/metabolismo , Sistema A de Transporte de Aminoácidos/metabolismo , Sistema L de Transporte de Aminoácidos/metabolismo , Sistemas de Transporte de Aminoácidos/genética , Apoptose , Sobrevivência Celular , Células Cultivadas , Feminino , Humanos , Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Gravidez , Proteínas Quinases/efeitos dos fármacos , Processamento de Proteína Pós-Traducional , Transporte Proteico , RNA Mensageiro/metabolismo , Transdução de Sinais , Sirolimo/farmacologia , Serina-Treonina Quinases TOR , Fatores de Tempo , Trofoblastos/efeitos dos fármacos
6.
Biochim Biophys Acta ; 1768(11): 2690-7, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17692818

RESUMO

Recently, we cloned two Na(+)-coupled lactate transporters from mouse kidney, a high-affinity transporter (SMCT1 or slc5a8) and a low-affinity transporter (SMCT2 or slc5a12). Here we report on the cloning and functional characterization of human SMCT2 (SLC5A12) and compare the immunolocalization patterns of slc5a12 and slc5a8 in mouse kidney. The human SMCT2 cDNA codes for a protein consisting of 618 amino acids. When expressed in mammalian cells or Xenopus oocytes, human SMCT2 mediates Na(+) -coupled transport of lactate, pyruvate and nicotinate. The affinities of the transporter for these substrates are lower than those reported for human SMCT1. Several non-steroidal anti-inflammatory drugs inhibit human SMCT2-mediated nicotinate transport, suggesting that NSAIDs interact with the transporter as they do with human SMCT1. Immunofluorescence microscopy of mouse kidney sections with an antibody specific for SMCT2 shows that the transporter is expressed predominantly in the cortex. Similar studies with an anti-SMCT1 antibody demonstrate that SMCT1 is also expressed mostly in the cortex. Dual-labeling of SMCT1 and SMCT2 with 4F2hc (CD98), a marker for basolateral membrane of proximal tubular cells in the S1 and S2 segments of the nephron, shows that both SMCT1 and SMCT2 are expressed in the apical membrane of the tubular cells. These studies also show that while SMCT2 is broadly expressed along the entire length of the proximal tubule (S1/S2/S3 segments), the expression of SMCT1 is mostly limited to the S3 segment. These studies suggest that the low-affinity transporter SMCT2 initiates lactate absorption in the early parts of the proximal tubule followed by the participation of the high-affinity transporter SMCT1 in the latter parts of the proximal tubule.


Assuntos
Rim/química , Transportadores de Ácidos Monocarboxílicos/análise , Transportadores de Ácidos Monocarboxílicos/genética , Sequência de Aminoácidos , Animais , Clonagem Molecular , DNA Complementar/química , Imunofluorescência , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Transportadores de Ácidos Monocarboxílicos/fisiologia , Simportadores , Xenopus laevis
7.
Int J Pharm ; 336(1): 133-9, 2007 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-17184941

RESUMO

We have previously demonstrated that dopamine conjugation to glucose allows it to induce therapeutic effects against Parkinson's disease after intravenous administration. In this paper we demonstrate that, unlike dopamine, the prodrug glu-dopamine is a transportable substrate of glucose transporters. Towards this, the effect of glucose-conjugation on the affinity and uptake of dopamine have been assessed in vitro, using human retinal pigment epithelium (HRPE) cells. Glucose transporter-mediated uptake was measured using [(3)H]3-O-methylglucose ([(3)H]3-O-MG) as the tracer. The uptake was found to be rapid and hyperbolically related to its concentrations (K(t)=7.8+/-1.2mM and V(max)=54+/-2 nmol/min mg protein). Inhibition experiments showed that dopamine was able to interact with glucose carriers only when conjugated to glucose (IC(50)=2.6+/-0.6mM). HPLC analysis of HRPE cell extracts showed that both dopamine and the prodrug permeate the cell, but only the uptake of the prodrug is inhibitable by glucose. This confirms that glucose transporters mediate the transport of the prodrug glu-dopamine, but not of dopamine. HRPE cells is therefore proposed as a promising model for in vitro studies involving the glucose transporter-mediated transport of drugs and their conjugates.


Assuntos
Dopamina/metabolismo , Proteínas Facilitadoras de Transporte de Glucose/metabolismo , Glucose/metabolismo , Pró-Fármacos/metabolismo , 3-O-Metilglucose/metabolismo , Transporte Biológico , Células Cultivadas , Cromatografia Líquida de Alta Pressão , Dopamina/química , Dopamina/farmacocinética , Células Epiteliais/química , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Glucose/química , Glucose/farmacocinética , Humanos , Cinética , Estrutura Molecular , Epitélio Pigmentado Ocular/citologia , Pró-Fármacos/farmacocinética
8.
Placenta ; 27(6-7): 550-9, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16129486

RESUMO

NaS2 is a Na+-coupled transporter for sulfate that belongs to the SLC13 gene family. This transporter was originally cloned from high endothelial venule endothelial cells, but nothing is known about the functional characteristics of this transporter except that it transports sulfate in a Na+-coupled manner. Northern blot analysis indicates that NaS2 is expressed most robustly in placenta. In the present study, we cloned NaS2 from rat placenta and characterized its transport function in detail using the Xenopus laevis oocyte expression system. Rat NaS2 consists of 629 amino acids and is highly similar to human NaS2. In situ hybridization studies with mouse placental sections show that NaS2 transcripts are expressed primarily in trophoblasts of the labyrinth zone. The expression of the transporter is confirmed in primary cultures of trophoblasts isolated from human placenta. When expressed in X. laevis oocytes, rat NaS2 mediates Na+-coupled transport of sulfate. The transport of sulfate is inhibited by oxyanions of selenium, chromium, arsenic, molybdenum, and phosphorous, suggesting that the transporter may mediate the transport of these oxyanions in addition to sulfate. The Kt for sulfate is 153+/-30 microM and the Na+:sulfate stoichiometry is 3:1. The transport process is electrogenic as evidenced from the inhibition of the uptake process by K+-induced depolarization. We conclude that NaS2 is a placenta-specific Na+-coupled, electrogenic, transporter for sulfate expressed in trophoblasts and that it is also responsible for the transport of oxyanions of the micronutrients selenium and chromium.


Assuntos
Ânions/metabolismo , Cromo/metabolismo , Transportadores de Ânions Orgânicos Dependentes de Sódio/metabolismo , Selênio/metabolismo , Sulfatos/metabolismo , Simportadores/metabolismo , Trofoblastos/metabolismo , Sequência de Aminoácidos , Animais , Biblioteca Gênica , Humanos , Hibridização In Situ , Dados de Sequência Molecular , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/metabolismo , Ratos , Homologia de Sequência de Aminoácidos , Transportadores de Sulfato , Xenopus laevis
9.
Biochem Soc Trans ; 33(Pt 1): 237-40, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15667316

RESUMO

SLC5A8 is a candidate tumour suppressor gene that is silenced in colon cancer, gastric cancer and possibly other cancers in humans. This gene codes for a transporter belonging to the Na(+)/glucose co-transporter gene family (SLC5). The cancer-associated silencing of the gene involves hypermethylation of CpG islands present in exon 1 of the gene. SLC5A8 is expressed in colon, ileum, kidney and thyroid gland. The protein coded by the gene mediates the Na(+)-coupled and electrogenic transport of a variety of monocarboxylates, including short-chain fatty acids, lactate and nicotinate. It may also transport iodide. The normal physiological function of this transporter in the intestinal tract and kidney is likely to facilitate the active absorption of short-chain fatty acids, lactate and nicotinate. One of the short-chain fatty acids that serves as a substrate for SLC5A8 is butyrate. This fatty acid is an inhibitor of histone deacetylases and is known to induce apoptosis in a variety of tumours including colonic tumour. Since butyrate is produced in the colonic lumen at high concentrations by bacterial fermentation of dietary fibre, we speculate that the ability of SLC5A8 to mediate the entry of this short-chain fatty acid into colonic epithelial cells underlies the potential tumour suppressor function of this transporter.


Assuntos
Proteínas de Transporte de Cátions/fisiologia , Genes Supressores de Tumor , Proteínas de Transporte de Cátions/genética , Inativação Gênica , Humanos , Transportadores de Ácidos Monocarboxílicos
10.
Am J Physiol Cell Physiol ; 281(6): C1757-68, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11698233

RESUMO

We have cloned a new subtype of the amino acid transport system N2 (SN2 or second subtype of system N) from rat brain. Rat SN2 consists of 471 amino acids and belongs to the recently identified glutamine transporter gene family that consists of system N and system A. Rat SN2 exhibits 63% identity with rat SN1. It also shows considerable sequence identity (50-56%) with the members of the amino acid transporter A subfamily. In the rat, SN2 mRNA is most abundant in the liver but is detectable in the brain, lung, stomach, kidney, testis, and spleen. When expressed in Xenopus laevis oocytes and in mammalian cells, rat SN2 mediates Na(+)-dependent transport of several neutral amino acids, including glycine, asparagine, alanine, serine, glutamine, and histidine. The transport process is electrogenic, Li(+) tolerant, and pH sensitive. The transport mechanism involves the influx of Na(+) and amino acids coupled to the efflux of H(+), resulting in intracellular alkalization. Proline, alpha-(methylamino)isobutyric acid, and anionic and cationic amino acids are not recognized by rat SN2.


Assuntos
Sistemas de Transporte de Aminoácidos Básicos/metabolismo , Sistemas de Transporte de Aminoácidos Neutros , Aminoácidos/metabolismo , Proteínas de Transporte/metabolismo , Proteínas de Membrana Transportadoras , Simportadores , Sequência de Aminoácidos , Sistemas de Transporte de Aminoácidos Básicos/genética , Aminoácidos/farmacologia , Animais , Química Encefálica , Proteínas de Transporte/química , Proteínas de Transporte/genética , Clonagem Molecular , Humanos , Dados de Sequência Molecular , Oócitos/efeitos dos fármacos , Oócitos/fisiologia , Técnicas de Patch-Clamp , Transportador 1 de Peptídeos , Epitélio Pigmentado Ocular/citologia , Epitélio Pigmentado Ocular/metabolismo , Isoformas de Proteínas , Ratos , Alinhamento de Sequência , Cloreto de Sódio/farmacologia , Especificidade por Substrato , Distribuição Tecidual , Xenopus laevis
11.
Am J Physiol Cell Physiol ; 281(6): C1825-36, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11698241

RESUMO

Taurine is actively transported at the retinal pigment epithelial (RPE) apical membrane in an Na(+)- and Cl(-)-dependent manner. Diabetes may alter the function of the taurine transporter. Because nitric oxide (NO) is a molecule implicated in the pathogenesis of diabetes, we asked whether NO would alter the activity of the taurine transporter in cultured ARPE-19 cells. The activity of the transporter was stimulated in the presence of the NO donor 3-morpholinosydnonimine. The stimulatory effects of 3-morpholinosydnonimine were not observed during the initial 16-h treatment; however, stimulation of taurine uptake was elevated dramatically above control values with 20- and 24-h treatments. Kinetic analysis revealed that the stimulation was associated with an increase in the maximal velocity of the transporter with no significant change in the substrate affinity. The NO-induced increase in taurine uptake was inhibited by actinomycin D and cycloheximide. RT-PCR analysis and nuclear run-on assays provided evidence for upregulation of the transporter gene. This study provides the first evidence of an increase in taurine transporter gene expression in human RPE cells cultured under conditions of elevated levels of NO.


Assuntos
Proteínas de Transporte/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana Transportadoras , Molsidomina/análogos & derivados , Óxido Nítrico/metabolismo , Epitélio Pigmentado Ocular/metabolismo , Taurina/metabolismo , Tirosina/análogos & derivados , Animais , Antioxidantes/farmacologia , Ácido Ascórbico/farmacologia , Proteínas de Transporte/genética , Linhagem Celular , Diabetes Mellitus/metabolismo , Relação Dose-Resposta a Droga , Glutationa/farmacologia , Humanos , Imuno-Histoquímica , Glicoproteínas de Membrana/genética , Azul de Metileno/farmacologia , Camundongos , Camundongos Endogâmicos ICR , Molsidomina/farmacologia , Doadores de Óxido Nítrico/farmacologia , Nitroprussiato/farmacologia , Epitélio Pigmentado Ocular/citologia , Epitélio Pigmentado Ocular/efeitos dos fármacos , Inibidores da Síntese de Proteínas/farmacologia , Fatores de Tempo , Tirosina/metabolismo
12.
Pharm Res ; 18(7): 950-6, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11496954

RESUMO

PURPOSE: To investigate the potential for delivering large peptides orally by altering their absorptive transport pathways and improving intestinal permeability. The absorptive transport of retro-inverso (R.I.-) K-Tat9 and R.I.-K(biotin)-Tat9, novel peptidic inhibitors of the Tat protein of HIV-1, and their interactions with human SMVT (hSMVT), a high affinity, low capacity transporter, were investigated using Caco-2 and transfected CHO cells. METHODS: Following synthesis on a PAL resin using Fmoc chemistry, the transport of R.I.-K-Tat9 (0.01-25 microM) and R.I.-K(biotin)-Tat9 (0.1-25 microM) was evaluated across Caco-2 cells. The transport and kinetics of biotin, biocytin and desthiobiotin (positive controls for SMVT) were also determined. Uptake of R.I.-K-Tat9 and R.I.K(biotin)-Tat9 (both 0.1-10 microM) was determined in CHO/hSMVT and CHO/pSPORT (control) cells. RESULTS: The absorptive transport of R.I.-K-Tat9 was passive, low (Pm approximately 1 x 10(-6) cm/sec) and not concentration dependent. R.I.K(biotin)-Tat9 permeability was 3.2-fold higher than R.I.-K-Tat9 demonstrating active (Ea = 9.1 kcal/mole), concentration dependent and saturable transport (Km = 3.3 microM). R.I.-K(biotin)-Tat9 uptake in CHO/hSMVT cells (Km = 1.0 microM) was - 500-fold greater than R.I.-K-Tat9 (at 10 microM). R.I.-K(biotin)-Tat9 transport in Caco-2 and CHO/hSMVT cells was significantly inhibited by known substrates of SMVT including biotin, biocytin, and desthiobiotin. Passive uptake of R.I.-K(biotin)-Tat9 was significantly greater than R.I.-K-Tat9 uptake in CHO/pSPORT cells. CONCLUSIONS: These results demonstrate that the structural modification of R.I.-K-Tat9 to R.I.-K(biotin)-Tat9 altered its intestinal transport pathway resulting in a significant improvement in its absorptive permeability by enhancing nonspecific passive and carrier-mediated uptake by means of SMVT. The specific interactions between R.I.-K(biotin)-Tat9 and SMVT suggest that targeting approaches utilizing transporters such as SMVT may substantially improve the oral delivery of large peptides.


Assuntos
Fármacos Anti-HIV/farmacocinética , Biotina/análogos & derivados , Biotina/farmacocinética , Proteínas de Transporte/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Produtos do Gene tat/farmacocinética , Glicoproteínas de Membrana/metabolismo , Simportadores , Administração Oral , Animais , Transporte Biológico Ativo , Células CHO/metabolismo , Células CACO-2/metabolismo , Cricetinae , Humanos , Absorção Intestinal/efeitos dos fármacos , Absorção Intestinal/fisiologia , Produtos do Gene tat do Vírus da Imunodeficiência Humana
13.
Exp Eye Res ; 73(2): 159-65, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11446766

RESUMO

Vitamin C [ascorbic acid (AA)] is an important antioxidant present in m M amounts in the aqueous humor. Recently, two specific transporters for vitamin C (SVCT1, SVCT2) have been cloned in the rat and the human. The aim of the present study was to characterize vitamin C transport in an immortalized human lens epithelial cell line (HLE-B3). AA uptake was linear for 120 min in experiments conducted with 14C AA + 40 microM unlabelled AA. Uptake was measured at varying AA concentrations (0.04-1 m M) in Na+-containing and Na+-free buffers for 30 min at 37 degrees C. Effect of potential inhibitors of AA transport was also examined. Presence (or absence) of SVCT1 and SVCT2 was studied by RT-PCR of HLE-B3 poly (A)+ RNA using gene specific primers. Uptake studies revealed that AA uptake was highly Na+-dependent and exhibited saturation. Na+-dependent 14C-AA uptake was strongly inhibited (85-90%) by 10 m M unlabelled AA. Incubation of HLE-B3 cells with cAMP (0.1 m M), cytocholasin B (0.1 m M) and phorbol dibutyrate (1 microM) resulted in partial inhibition (36-51%) of AA uptake. Under similar conditions, D -glucose (10 m M) and staurosporine (0.1 microM) had no effect. RT-PCR showed the presence of SVCT2 while SVCT1 could not be amplified. Exposure to the chemical oxidant tert-butylhydroperoxide (TBH) up-regulated SVCT2 gene expression in HLE-B3 cells. Our data suggest that Na+-dependent transport of AA in normal lens epithelium is most likely mediated by SVCT2 rather than by SVCT1. This transport system may be subject to regulation by oxidant stress and by various second messenger signals.


Assuntos
Antioxidantes/farmacocinética , Ácido Ascórbico/farmacocinética , Proteínas de Transporte/metabolismo , Células Epiteliais/metabolismo , Cristalino/citologia , Transportadores de Ânions Orgânicos Dependentes de Sódio , Simportadores , Transporte Biológico/fisiologia , Butanóis/farmacologia , Células Cultivadas , AMP Cíclico/fisiologia , Citocalasina B/farmacologia , Glucose/farmacologia , Humanos , Estresse Oxidativo , Ésteres de Forbol/farmacologia , Proteínas/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sódio/metabolismo , Transportadores de Sódio Acoplados à Vitamina C , Estaurosporina/farmacologia
14.
Biochim Biophys Acta ; 1512(2): 299-307, 2001 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-11406107

RESUMO

The present study was undertaken to elucidate the functional characteristics of choline uptake and deduce the relationship between choline uptake and the expression of organic cation transporters in the rat brain microvessel endothelial cell line RBE4. Confluent RBE4 cells were found to express a high affinity choline uptake system. The system is Na(+)-independent and shows a Michaelis-Menten constant of approx. 20 microM for choline. The choline analogue hemicholinium-3 inhibits choline uptake in these cells with an inhibition constant of approx. 50 microM. The uptake system is also susceptible for inhibition by various organic cations, including 1-methyl-4-phenylpyridinium, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, clonidine, procainamide, and tetramethylammonium. The prototypical organic cation tetraethylammonium shows very little affinity for the choline uptake system in these cells. The inhibition of choline uptake by hemicholinium-3 is competitive. Northern analysis and RT-PCR show that these cells do not express the organic cation transporters OCT2 and OCT3. These cells do express, however, low levels of OCT1, but the functional characteristics of choline uptake in these cells are very different from the known properties of choline uptake via OCT1. The Na(+)-coupled high affinity choline transporter CHT1 is not expressed in these cells as evidenced by RT-PCR. This corroborates the Na(+)-independent nature of choline uptake in these cells. It is concluded that RBE4 cells express an organic cation transporter that is responsible for choline uptake in these cells and that this transporter is not identical to any of the organic cation transporters thus far identified at the molecular level in mammalian cells.


Assuntos
Proteínas de Transporte/metabolismo , Circulação Cerebrovascular/fisiologia , Colina/metabolismo , Endotélio Vascular/metabolismo , Proteínas de Membrana/metabolismo , Microcirculação/fisiologia , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/farmacologia , 1-Metil-4-fenilpiridínio/farmacologia , Animais , Transporte Biológico/efeitos dos fármacos , Encéfalo/irrigação sanguínea , Tronco Encefálico/metabolismo , Proteínas de Transporte/genética , Linhagem Celular , Clonidina/farmacologia , Endotélio Vascular/citologia , Hemicolínio 3/farmacologia , Rim/metabolismo , Cinética , Proteínas de Membrana/genética , Transportador 1 de Cátions Orgânicos , Compostos de Amônio Quaternário/farmacologia , RNA Mensageiro/genética , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sódio/metabolismo , Tetraetilamônio/farmacologia , Transcrição Gênica
15.
Biochim Biophys Acta ; 1512(1): 15-21, 2001 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-11334620

RESUMO

We investigated the molecular mechanism involved in the adaptive regulation of the amino acid transport system A, a process in which amino acid starvation induces the transport activity. These studies were done with rat C6 glioma cells. System A activity in these cells is mediated exclusively by the system A subtype, amino acid transporter A2 (ATA2). The other two known system A subtypes, ATA1 and ATA3, are not expressed in these cells. Exposure of these cells to an amino acid-free medium induces system A activity. This process consists of an acute phase and a chronic phase. Laser-scanning confocal microscopic immunolocalization of ATA2 reveals that the acute phase is associated with recruitment of preformed ATA2 from an intracellular pool to the plasma membrane. In contrast, the chronic phase is associated with an induction of ata2 gene expression as evidenced from the increase in the steady-state levels of ATA2 mRNA, restoration of the intracellular pool of ATA2 protein, and blockade of the induction by cycloheximide and actinomycin D. The increase in system A activity induced by amino acid starvation is blocked specifically by system A substrates, including the non-metabolizable alpha-(methylamino)isobutyric acid.


Assuntos
Proteínas de Transporte/genética , Regulação da Expressão Gênica , Adaptação Fisiológica , Sistemas de Transporte de Aminoácidos , Aminoácidos/biossíntese , Aminoácidos/deficiência , Animais , Transporte Biológico , Proteínas de Transporte/metabolismo , Membrana Celular/metabolismo , RNA Mensageiro/análise , Ratos , Especificidade por Substrato , Células Tumorais Cultivadas
16.
Biochim Biophys Acta ; 1510(1-2): 10-7, 2001 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-11342143

RESUMO

We report here on the cloning and functional characterization of the third subtype of amino acid transport system A, designated ATA3 (amino acid transporter A3), from a human liver cell line. This transporter consists of 547 amino acids and is structurally related to the members of the glutamine transporter family. The human ATA3 (hATA3) exhibits 88% identity in amino acid sequence with rat ATA3. The gene coding for hATA3 contains 16 exons and is located on human chromosome 12q13. It is expressed almost exclusively in the liver. hATA3 mediates the transport of neutral amino acids including alpha-(methylamino)isobutyric acid (MeAIB), the model substrate for system A, in a Na(+)-coupled manner and the transport of cationic amino acids in a Na(+)-independent manner. The affinity of hATA3 for cationic amino acids is higher than for neutral amino acids. The transport function of hATA3 is thus similar to that of system y(+)L. The ability of hATA3 to transport cationic amino acids with high affinity is unique among the members of the glutamine transporter family. hATA1 and hATA2, the other two known members of the system A subfamily, show little affinity toward cationic amino acids. hATA3 also differs from hATA1 and hATA2 in exhibiting low affinity for MeAIB. Since liver does not express any of the previously known high-affinity cationic amino acid transporters, ATA3 is likely to provide the major route for the uptake of arginine in this tissue.


Assuntos
Aminoácidos Neutros/metabolismo , Proteínas de Transporte/metabolismo , Fígado/metabolismo , beta-Alanina/análogos & derivados , Sistemas de Transporte de Aminoácidos , Arginina/metabolismo , Transporte Biológico , Northern Blotting , Proteínas de Transporte/genética , Linhagem Celular , Cromossomos Humanos Par 12 , Células Epiteliais/metabolismo , Éxons , Glicina/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Íntrons , RNA Mensageiro/análise , Especificidade por Substrato , beta-Alanina/metabolismo
17.
J Clin Invest ; 107(8): 1035-43, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11306607

RESUMO

Nitric oxide synthase (NOS) inhibitors have therapeutic potential in the management of numerous conditions in which NO overproduction plays a critical role. Identification of transport systems in the intestine that can mediate the uptake of NOS inhibitors is important to assess the oral bioavailability and therapeutic efficacy of these potential drugs. Here, we have cloned the Na+ - and Cl- -coupled amino acid transport system B(0,+) (ATB(0,+)) from the mouse colon and investigated its ability to transport NOS inhibitors. When expressed in mammalian cells, ATB(0,+) can transport a variety of zwitterionic and cationic amino acids in a Na+ - and Cl- -coupled manner. Each of the NOS inhibitors tested compete with glycine for uptake through this transport system. Furthermore, using a tritiated analog of the NOS inhibitor N(G)-nitro-L-arginine, we showed that Na+ - and Cl- -coupled transport occurs via ATB(0,+). We then studied transport of a wide variety of NOS inhibitors in Xenopus laevis oocytes expressing the cloned ATB(0,+) and found that ATB(0,+) can transport a broad range of zwitterionic or cationic NOS inhibitors. These data represent the first identification of an ion gradient-driven transport system for NOS inhibitors in the intestinal tract.


Assuntos
Sistema ASC de Transporte de Aminoácidos , Proteínas de Transporte/metabolismo , Cloretos/metabolismo , Óxido Nítrico Sintase/antagonistas & inibidores , Receptores Virais/metabolismo , Sódio/metabolismo , Animais , Transporte Biológico Ativo , Proteínas de Transporte/genética , Proteínas de Transporte/fisiologia , Perfilação da Expressão Gênica , Humanos , Intestino Delgado/metabolismo , Camundongos , Antígenos de Histocompatibilidade Menor , Proteínas de Transporte de Neurotransmissores , RNA Mensageiro , Receptores Virais/genética , Receptores Virais/fisiologia , Xenopus laevis
18.
J Physiol ; 532(Pt 2): 297-304, 2001 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-11306651

RESUMO

1. ATB(0,+) is an amino acid transporter energized by transmembrane gradients of Na+ and Cl(-) and membrane potential. We cloned this transporter from mouse colon and expressed the clone functionally in mammalian (human retinal pigment epithelial, HRPE) cells and Xenopus laevis oocytes to investigate the interaction of carnitine and its acyl esters with the transporter. 2. When expressed in mammalian cells, the cloned ATB(0,+) was able to transport carnitine, propionylcarnitine and acetylcarnitine. The transport process was Na(+) and Cl(-) dependent and inhibitable by the amino acid substrates of the transporter. The Michaelis constant for carnitine was 0.83 +/- 0.08 mM and the Hill coefficient for Na(+) activation was 1.6 +/- 0.1. 3. When expressed in Xenopus laevis oocytes, the cloned ATB(0,+) was able to induce inward currents in the presence of carnitine and propionylcarnitine under voltage-clamped conditions. There was no detectable current in the presence of acetylcarnitine. Carnitine-induced currents were obligatorily dependent on the presence of Na(+) and Cl(-). The currents were saturable with carnitine and the Michaelis constant was 1.8 +/- 0.4 mM. The analysis of Na(+)- and Cl(-)-activation kinetics revealed that 2 Na(+) and 1 Cl(-) were involved in the transport of carnitine via the transporter. 4. These studies describe the identification of a novel function for the amino acid transporter ATB(0,+). Since this transporter is expressed in the intestinal tract, lung and mammary gland, it is likely to play a significant role in the handling of carnitine in these tissues. 5. A Na(+)-dependent transport system for carnitine has already been described. This transporter, known as OCTN2 (novel organic cation transporter 2), is expressed in most tissues and transports carnitine with high affinity. It is energized, however, only by a Na(+) gradient and membrane potential. In contrast, ATB(0,+) is a low-affinity transporter for carnitine, but exhibits much higher concentrative capacity than OCTN2 because of its energization by transmembrane gradients of Na(+) and Cl(-) as well as by membrane potential.


Assuntos
Sistema ASC de Transporte de Aminoácidos , Carnitina/farmacocinética , Proteínas de Transporte/metabolismo , Cloretos/metabolismo , Colo/metabolismo , Oócitos/metabolismo , Epitélio Pigmentado Ocular/metabolismo , Receptores Virais/metabolismo , Sódio/metabolismo , Sequência de Aminoácidos/genética , Animais , Transporte Biológico Ativo/fisiologia , Proteínas de Transporte/genética , Camundongos , Antígenos de Histocompatibilidade Menor , Dados de Sequência Molecular , Epitélio Pigmentado Ocular/citologia , Receptores Virais/genética , Xenopus laevis
19.
Biochem Biophys Res Commun ; 281(5): 1343-8, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11243884

RESUMO

We have cloned a new subtype of the amino acid transport system N from a human liver cell line. This transporter, designated SN2, consists of 472 amino acids and exhibits 62% identity with human SN1 at the level of amino acid sequence. SN2-specific transcripts are expressed predominantly in the stomach, brain, liver, lung, and intestinal tract. The sizes of the transcripts vary in different tissues, indicating tissue-specific alternative splicing of the SN2 mRNA. In contrast, SN1 is expressed primarily in the brain and liver and there is no evidence for the presence of multiple transcripts of varying size for SN1. When expressed in mammalian cells, the cloned human SN2 mediates Na(+)-coupled transport of system N-specific amino acid substrates (glutamine, asparagine, and histidine). In addition, SN2 also transports serine, alanine, and glycine. Anionic amino acids, cationic amino acids, imino acids, and N-alkylated amino acids are not recognized as substrates by human SN2. The SN2-mediated transport process is Li(+)-tolerant and highly pH-dependent. The Michaelis-Menten constant for histidine uptake via human SN2 is 0.6 +/- 0.1 mM. The gene coding for SN2 is located on human chromosome Xp11.23. Successful cloning of SN2 provides the first molecular evidence for the existence of subtypes within the amino acid transport system N in mammalian tissues.


Assuntos
Sistemas de Transporte de Aminoácidos Neutros , Proteínas de Transporte/genética , Proteínas de Transporte/fisiologia , Proteínas de Membrana Transportadoras , Sequência de Aminoácidos , Aminoácidos/metabolismo , Transporte Biológico Ativo , Encéfalo/metabolismo , Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Linhagem Celular , Clonagem Molecular , Histidina/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Cinética , Fígado/metabolismo , Dados de Sequência Molecular , RNA Mensageiro/biossíntese , Homologia de Sequência de Aminoácidos , Especificidade por Substrato , Distribuição Tecidual , Transcrição Gênica , Transfecção
20.
Invest Ophthalmol Vis Sci ; 42(1): 47-54, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11133847

RESUMO

PURPOSE: The purpose of this investigation was to provide evidence for the expression of the cystine/glutamate transporter (x(c)(-)) in the human retinal pigment epithelial cell line ARPE-19, clone the light chain of the transporter from an ARPE-19 cell cDNA library and study its function, and investigate the regulation of this transporter by nitric oxide (NO) in ARPE-19 cells. METHODS: Uptake of radiolabeled cystine and glutamate was measured in ARPE-19 cells. The functional identity of x(c)(-) in these cells was established by substrate specificity and Na(+)-independence of the uptake process. The human x(c)(-) light chain (human xCT) was cloned from an ARPE-19 cell cDNA library. The functional identity of the cloned human xCT was investigated by heterologous coexpression of the light chain with the heavy chain (human 4F2hc) in HeLa cells. ARPE-19 cells were treated with or without the NO donor 3-nitroso-N:-acetylpenicillamine (SNAP) and the expression of x(c)(-) was studied at the functional and molecular levels. RESULTS: ARPE-19 cells take up cystine as well as glutamate in the absence of Na(+). Substrate specificity studies indicate that although the uptake of cystine in the absence of Na(+) is mediated by multiple amino acid transport systems including x(c)(-), the uptake of glutamate in the absence of Na(+) occurs exclusively via x(c)(-). The human xCT cloned from ARPE-19 cells is a protein of 501 amino acids. These cells express the heavy chain 4F2hc as evidenced from RT-PCR analysis. Coexpression of human xCT with 4F2hc in HeLa cells leads to the induction of cystine and glutamate uptake with characteristics similar to that of x(c)(-). The activity of x(c)(-) in ARPE-19 cells is upregulated by SNAP, and the process is associated with an increase in the expression of xCT with no detectable change in the expression of 4F2hc. CONCLUSIONS: ARPE-19 cells express the cystine/glutamate transporter x(c)(-) (the light chain xCT and the heavy chain 4F2hc) as is evident from functional and molecular studies. NO upregulates this transport system and the process is associated with an increase in xCT mRNA but with no change in 4F2hc mRNA.


Assuntos
Sistema y+ de Transporte de Aminoácidos , Proteínas de Transporte/química , Proteínas de Transporte/fisiologia , Cistina/metabolismo , Proteínas do Olho/química , Proteínas do Olho/fisiologia , Ácido Glutâmico/metabolismo , Penicilamina/análogos & derivados , Epitélio Pigmentado Ocular/metabolismo , Sequência de Aminoácidos , Células Cultivadas , Clonagem Molecular , Primers do DNA/química , Proteínas do Olho/genética , Expressão Gênica , Biblioteca Gênica , Gliceraldeído-3-Fosfato Desidrogenases/genética , Gliceraldeído-3-Fosfato Desidrogenases/metabolismo , Humanos , Dados de Sequência Molecular , Óxido Nítrico/metabolismo , Penicilamina/farmacologia , Epitélio Pigmentado Ocular/citologia , RNA Mensageiro/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência de Aminoácidos , Relação Estrutura-Atividade , Especificidade por Substrato
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...