Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
Add more filters










Publication year range
1.
PLoS Genet ; 17(11): e1009893, 2021 11.
Article in English | MEDLINE | ID: mdl-34780467

ABSTRACT

Identifying a common oncogenesis pathway among tumors with different oncogenic mutations is critical for developing anti-cancer strategies. Here, we performed transcriptome analyses on two different models of Drosophila malignant tumors caused by Ras activation with cell polarity defects (RasV12/scrib-/-) or by microRNA bantam overexpression with endocytic defects (bantam/rab5-/-), followed by an RNAi screen for genes commonly essential for tumor growth and malignancy. We identified that Juvenile hormone Inducible-21 (JhI-21), a Drosophila homolog of the L-amino acid transporter 1 (LAT1), is upregulated in these malignant tumors with different oncogenic mutations and knocking down of JhI-21 strongly blocked their growth and invasion. JhI-21 expression was induced by simultaneous activation of c-Jun N-terminal kinase (JNK) and Yorkie (Yki) in these tumors and thereby contributed to tumor growth and progression by activating the mTOR-S6 pathway. Pharmacological inhibition of LAT1 activity in Drosophila larvae significantly suppressed growth of RasV12/scrib-/- tumors. Intriguingly, LAT1 inhibitory drugs did not suppress growth of bantam/rab5-/- tumors and overexpression of bantam rendered RasV12/scrib-/- tumors unresponsive to LAT1 inhibitors. Further analyses with RNA sequencing of bantam-expressing clones followed by an RNAi screen suggested that bantam induces drug resistance against LAT1 inhibitors via downregulation of the TMEM135-like gene CG31157. Our observations unveil an evolutionarily conserved role of LAT1 induction in driving Drosophila tumor malignancy and provide a powerful genetic model for studying cancer progression and drug resistance.


Subject(s)
Amino Acid Transport Systems/metabolism , Carcinogenesis/genetics , Carcinogenesis/pathology , Drosophila Proteins/genetics , Drug Resistance, Neoplasm , MAP Kinase Kinase 4/metabolism , YAP-Signaling Proteins/metabolism , Amino Acid Transport Systems/antagonists & inhibitors , Amino Acid Transport Systems/genetics , Animals , Drosophila , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/metabolism , MAP Kinase Kinase 4/genetics , MicroRNAs/genetics , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , RNA Interference , Signal Transduction , Up-Regulation , YAP-Signaling Proteins/genetics
2.
Bioorg Med Chem Lett ; 33: 127728, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33346010

ABSTRACT

Triptolide (TP) is a diterpene epoxide component extracted from Tripterygium wilfordii and has been shown to possess an impressive anticancer effect. However, TP has not yet entered any clinic trials due to the severe adverse effects that resulted from the off-target absorption and distribution found in animal studies. In this study, we designed and synthesized three amino acids (tryptophan, valine, and lysine) based TP prodrugs to target ATB0,+ which are highly expressed in pancreatic cancer cells for more effective pancreatic cancer therapy. The stability, uptake profiles, uptake mechanism, and cancer-killing ability were studied in vitro. All three prodrugs showed increased uptake and enhanced cytotoxicity in pancreatic cancer cells, but not in normal pancreatic cells. The difference in killing effect on normal and cancer cells was attributed to pancreatic cancer over-expressed ATB0,+-mediated uptake. Specifically, tryptophan-conjugated TP prodrug (TP-Trp) showed the highest uptake and the best cancer cell killing effect, considered as the best candidate. The present study provided the proof-of-concept of exploiting TP prodrug to target ATB0,+ for pancreatic cancer-selective delivery and treatment.


Subject(s)
Amino Acid Transport Systems/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Diterpenes/pharmacology , Pancreatic Neoplasms/drug therapy , Phenanthrenes/pharmacology , Prodrugs/pharmacology , Amino Acid Transport Systems/metabolism , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Diterpenes/chemical synthesis , Diterpenes/chemistry , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Epoxy Compounds/chemical synthesis , Epoxy Compounds/chemistry , Epoxy Compounds/pharmacology , Humans , Molecular Conformation , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Phenanthrenes/chemical synthesis , Phenanthrenes/chemistry , Prodrugs/chemical synthesis , Prodrugs/chemistry , Structure-Activity Relationship
3.
Cells ; 9(8)2020 08 15.
Article in English | MEDLINE | ID: mdl-32824193

ABSTRACT

Amino acid metabolism promotes cancer cell proliferation and survival by supporting building block synthesis, producing reducing agents to mitigate oxidative stress, and generating immunosuppressive metabolites for immune evasion. Malignant cells rewire amino acid metabolism to maximize their access to nutrients. Amino acid transporter expression is upregulated to acquire amino acids from the extracellular environment. Under nutrient depleted conditions, macropinocytosis can be activated where proteins from the extracellular environment are engulfed and degraded into the constituent amino acids. The demand for non-essential amino acids (NEAAs) can be met through de novo synthesis pathways. Cancer cells can alter various signaling pathways to boost amino acid usage for the generation of nucleotides, reactive oxygen species (ROS) scavenging molecules, and oncometabolites. The importance of amino acid metabolism in cancer proliferation makes it a potential target for therapeutic intervention, including via small molecules and antibodies. In this review, we will delineate the targets related to amino acid metabolism and promising therapeutic approaches.


Subject(s)
Amino Acids/antagonists & inhibitors , Amino Acids/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Molecular Targeted Therapy/methods , Neoplasms/drug therapy , Neoplasms/metabolism , Signal Transduction/drug effects , Amino Acid Transport Systems/antagonists & inhibitors , Amino Acid Transport Systems/metabolism , Animals , Cell Proliferation/drug effects , Humans , Oxidative Stress/drug effects , Pinocytosis/drug effects , Reactive Oxygen Species/metabolism
4.
Endocrinol Metab (Seoul) ; 35(2): 227-236, 2020 06.
Article in English | MEDLINE | ID: mdl-32615707

ABSTRACT

Thyroid cancer cells have a high amino acid demand for proliferation, invasion, and metastasis. Amino acids are taken up by thyroid cancer cells, both thyroid follicular cell and thyroid parafollicular cells (commonly called "C-cells"), via amino acid transporters. Amino acid transporters up-regulate in many cancers, and their expression level associate with clinical aggressiveness and prognosis. This is the review to discuss the therapeutic potential of amino acid transporters and as molecular targets in thyroid cancer.


Subject(s)
Amino Acid Transport Systems/antagonists & inhibitors , Antineoplastic Agents/therapeutic use , Molecular Targeted Therapy , Thyroid Neoplasms/drug therapy , Animals , Humans , Proto-Oncogene Mas , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology
5.
Biochem J ; 477(10): 1923-1937, 2020 05 29.
Article in English | MEDLINE | ID: mdl-32379301

ABSTRACT

Gemcitabine is the first-line chemotherapy for pancreatic cancer. To overcome the often-acquired gemcitabine resistance, other drugs are used in combination with gemcitabine. It is well-known that cancer cells reprogram cellular metabolism, coupled with the up-regulation of selective nutrient transporters to feed into the altered metabolic pathways. Our previous studies have demonstrated that the amino acid transporter SLC6A14 is markedly up-regulated in pancreatic cancer and that it is a viable therapeutic target. α-Methyltryptophan (α-MT) is a blocker of SLC6A14 and is effective against pancreatic cancer in vitro and in vivo. In the present study, we tested the hypothesis that α-MT could synergize with gemcitabine in the treatment of pancreatic cancer. We investigated the effects of combination of α-MT and gemcitabine on proliferation, migration, and apoptosis in a human pancreatic cancer cell line, and examined the underlying mechanisms using 1H-NMR-based metabolomic analysis. These studies examined the intracellular metabolite profile and the extracellular metabolite profile separately. Combination of α-MT with gemcitabine elicited marked changes in a wide variety of metabolic pathways, particularly amino acid metabolism with notable alterations in pathways involving tryptophan, branched-chain amino acids, ketone bodies, and membrane phospholipids. The metabolomic profiles of untreated control cells and cells treated with gemcitabine or α-MT were distinctly separable, and the combination regimen showed a certain extent of overlap with the individual α-MT and gemcitabine groups. This represents the first study detailing the metabolomic basis of the anticancer efficacy of gemcitabine, α-MT and their combination.


Subject(s)
Deoxycytidine/analogs & derivatives , Drug Synergism , Pancreatic Neoplasms/drug therapy , Tryptophan/analogs & derivatives , Amino Acid Transport Systems/antagonists & inhibitors , Amino Acid Transport Systems/metabolism , Amino Acids/drug effects , Amino Acids/metabolism , Antineoplastic Agents , Antineoplastic Combined Chemotherapy Protocols , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Deoxycytidine/therapeutic use , Humans , Metabolomics , Pancreatic Neoplasms/pathology , Tryptophan/metabolism , Tryptophan/therapeutic use , Gemcitabine
6.
Curr Med Chem ; 26(36): 6636-6651, 2019.
Article in English | MEDLINE | ID: mdl-31218951

ABSTRACT

Amino acids and polyamines are involved in relevant processes for the parasite Trypanosoma cruzi, like protein synthesis, stress resistance, life cycle progression, infection establishment and redox balance, among others. In addition to the biosynthetic routes of amino acids, T. cruzi possesses transport systems that allow the active uptake from the extracellular medium; and in the case of polyamines, the uptake is the unique way to obtain these compounds. The TcAAAP protein family is absent in mammals and its members are responsible for amino acid and derivative uptake, thus the TcAAAP permeases are not only interesting and promising therapeutic targets but could also be used to direct the entry of toxic compounds into the parasite. Although there is a treatment available for Chagas disease, its limited efficacy in the chronic stage of the disease, as well as the side effects reported, highlight the urgent need to develop new therapies. Discovery of new drugs is a slow and cost-consuming process, and even during clinical trials the drugs can fail. In this context, drug repositioning is an interesting and recommended strategy by the World Health Organization since costs and time are significantly reduced. In this article, amino acids and polyamines transport and their potential as therapeutic targets will be revised, including examples of synthetic drugs and drug repurposing.


Subject(s)
Amino Acid Transport Systems/antagonists & inhibitors , Cation Transport Proteins/antagonists & inhibitors , Trypanocidal Agents/pharmacology , Trypanosoma cruzi/drug effects , Animals , Drug Repositioning , Polyamines/metabolism
7.
Am J Respir Cell Mol Biol ; 61(6): 755-764, 2019 12.
Article in English | MEDLINE | ID: mdl-31189070

ABSTRACT

SLC6A14-mediated l-arginine transport has been shown to augment the residual anion channel activity of the major mutant, F508del-CFTR, in the murine gastrointestinal tract. It is not yet known if this transporter augments residual and pharmacological corrected F508del-CFTR in primary airway epithelia. We sought to determine the role of l-arginine uptake via SLC6A14 in modifying F508del-CFTR channel activity in airway cells from patients with cystic fibrosis (CF). Human bronchial epithelial (HBE) cells from lung explants of patients without CF (HBE) and those with CF (CF-HBE) were used for H3-flux, airway surface liquid, and Ussing chamber studies. We used α-methyltryptophan as a specific inhibitor for SLC6A14. CFBE41o-, a commonly used CF airway cell line, was employed for studying the mechanism of the functional interaction between SLC6A14 and F508del-CFTR. SLC6A14 is functionally expressed in CF-HBE cells. l-arginine uptake via SLC6A14 augmented F508del-CFTR function at baseline and after treatment with lumacaftor. SLC6A14-mediated l-arginine uptake also increased the airway surface liquid in CF-HBE cells. Using CFBE41o cells, we showed that the positive SLC6A14 effect was mainly dependent on the nitric oxide (NO) synthase activity, nitrogen oxides, including NO, and phosphorylation by protein kinase G. These finding were confirmed in CF-HBE, as inducible NO synthase inhibition abrogated the functional interaction between SLC6A14 and pharmacological corrected F508del-CFTR. In summary, SLC6A14-mediated l-arginine transport augments residual F508del-CFTR channel function via a noncanonical, NO pathway. This effect is enhanced with increasing pharmacological rescue of F508del-CFTR to the membrane. The current study demonstrates how endogenous pathways can be used for the development of companion therapy in CF.


Subject(s)
Amino Acid Transport Systems/physiology , Arginine/metabolism , Bronchi/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Cystic Fibrosis/therapy , Amino Acid Transport Systems/antagonists & inhibitors , Amino Acid Transport Systems/genetics , Biological Transport , Bronchi/cytology , Cells, Cultured , Cystic Fibrosis/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/deficiency , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Genes, Reporter , Humans , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type II/metabolism , Recombinant Proteins/metabolism , Surface Properties , Transduction, Genetic , Tryptophan/analogs & derivatives , Tryptophan/pharmacology
8.
J Bacteriol ; 201(9)2019 05 01.
Article in English | MEDLINE | ID: mdl-30745372

ABSTRACT

Erwinia amylovora is the causal agent of fire blight of apple and pear trees. Several bacteria have been shown to produce antibiotics that antagonize E. amylovora, including pantocins, herbicolins, dapdiamides, and the vinylglycines, 4-formylaminooxyvinylglycine (FVG) and 4-aminoethoxyvinylglycine (AVG). Pantoea ananatis BRT175 was previously shown to exhibit antibiotic activity against E. amylovora via the production of Pantoea natural product 1 (PNP-1), later shown to be FVG; however, exposure of E. amylovora to FVG results in spontaneously resistant mutants. To identify the mechanism of resistance, we used genome variant analysis on spontaneous FVG-resistant mutants of E. amylovora and identified null mutations in the l-asparagine permease gene ansP Heterologous expression of ansP in normally resistant Escherichia coli was sufficient to impart FVG susceptibility, suggesting that FVG is imported through this permease. Because FVG and AVG are structurally similar, we hypothesized that resistance to AVG would also be conferred through inactivation of ansP; however, ansP mutants were not resistant to AVG. We found that spontaneously resistant Ea321 mutants also arise in the presence of AVG, with whole-genome variant analysis revealing that resistance was due to inactivation of the arginine ABC transporter permease subunit gene artQ Heterologous expression of the predicted lysE-like transporter encoded within the Pantoea ananatis BRT175 FVG biosynthetic cluster, which is likely responsible for antibiotic export, was sufficient to confer resistance to both FVG and AVG. This work highlights the important roles of amino acid transporters in antibiotic import into bacteria and the potential utility of antimicrobial amino acid analogs as antibiotics.IMPORTANCE The related antibiotics formylaminooxyvinylglycine (FVG) and aminoethoxyvinylglycine (AVG) have been shown to have activity against the fire blight pathogen Erwinia amylovora; however, E. amylovora can develop spontaneous resistance to these antibiotics. By comparing the genomes of mutants to those of the wild type, we found that inactivation of the l-asparagine transporter conferred resistance to FVG, while inactivation of the l-arginine transporter conferred resistance to AVG. We also show that the transporter encoded by the FVG biosynthetic cluster can confer resistance to both FVG and AVG. Our work indicates the important role that amino acid transporters play in the import of antibiotics and highlights the possible utility in designer antibiotics that enter the bacterial cell through amino acid transporters.


Subject(s)
Amino Acid Transport Systems/antagonists & inhibitors , Anti-Bacterial Agents/pharmacology , Enzyme Inhibitors/pharmacology , Erwinia amylovora/drug effects , Erwinia amylovora/enzymology , Glycine/analogs & derivatives , DNA Mutational Analysis , Drug Resistance, Bacterial , Glycine/pharmacology , Mutation
9.
Amino Acids ; 50(10): 1377-1390, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29978260

ABSTRACT

In yeast, toxicity of acetaminophen (APAP), a frequently used analgesic and antipyretic drug, depends on ubiquitin-controlled processes. Previously, we showed a remarkable overlap in toxicity profiles between APAP and tyrosine, and a similarity with drugs like rapamycin and quinine, which induce degradation of the amino acid permease Tat2. Therefore, we investigated in yeast whether APAP reduced the expression levels of amino acid permeases. The protein levels of Tat2, Tat1, Mup1 and Hip1 were reduced, while the expression of the general permease Gap1 was increased, consistent with a nutrient starvation response. Overexpression of Tat1 and Tat2, but not Mup1, Hip1 and Gap1 conferred resistance to APAP. A tryptophan auxotrophic strain trp1Δ was more sensitive to APAP than wild-type and addition of tryptophan completely restored the growth restriction of trp1∆ upon APAP exposure, while tyrosine had an additive effect on APAP toxicity. Furthermore, intracellular aromatic amino acid concentrations were reduced upon APAP exposure. This effect was less prominent in ubiquitin-deficient yeast strains that were APAP resistant and showed a reduced degradation of high affinity amino acid permeases. APAP-induced changes in intracellular amino acid concentrations were also detected in hepatoma HepG2 cells indicating significance for humans.


Subject(s)
Acetaminophen/toxicity , Enzyme Inhibitors/toxicity , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/enzymology , Tryptophan/metabolism , Amino Acid Transport Systems/antagonists & inhibitors , Amino Acid Transport Systems/genetics , Amino Acid Transport Systems/metabolism , Amino Acids/metabolism , Hep G2 Cells , Humans , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Ubiquitin/metabolism
10.
Neurotox Res ; 33(1): 1-5, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28470569

ABSTRACT

The study of the mechanism of ß-N-methylamino-L-alanine (BMAA) neurotoxicity originally focused on its effects at the N-methyl-D-aspartate (NMDA) receptor. In recent years, it has become clear that its mechanism of action is more complicated. First, there are certain cell types, such as motor neurons and cholinergic neurons, where the dominate mechanism of toxicity is through action at AMPA receptors. Second, even in cortical neurons where the primary mechanism of toxicity appears to be activation of NMDA receptors, there are other mechanisms involved. We found that along with NMDA receptors, activation of mGLuR5 receptors and effects on the cystine/glutamate antiporter (system xc-) were involved in the toxicity. The effects on system xc- are of particular interest. System xc- mediates the transport of cystine into the cell in exchange for releasing glutamate into the extracellular fluid. By releasing glutamate, system xc- can potentially cause excitotoxicity. However, through providing cystine to the cell, it regulates the levels of cellular glutathione (GSH), the main endogenous intracellular antioxidant, and in this way may protect cells against oxidative stress. We have previously published that BMAA inhibits cystine uptake leading to GSH depletion and had indirect evidence that BMAA is transported into the cells by system xc-. We now present direct evidence that BMAA is transported into both astrocytes and neurons through system xc-. The fact that BMAA is transported by system xc- also provides a mechanism for BMAA to enter brain cells potentially leading to misincorporation into proteins and protein misfolding.


Subject(s)
Amino Acid Transport Systems/physiology , Amino Acids, Diamino/metabolism , Astrocytes/metabolism , Neurons/metabolism , Amino Acid Transport Systems/antagonists & inhibitors , Animals , Aspartic Acid/pharmacology , Astrocytes/drug effects , Carbon Isotopes/metabolism , Cells, Cultured , Cerebral Cortex/cytology , Cyanobacteria Toxins , Cysteine/metabolism , Embryo, Mammalian , Female , Glutamic Acid/metabolism , Glutathione/metabolism , Mice , Neurons/drug effects , Neurotransmitter Uptake Inhibitors/pharmacology , Pregnancy , Sulfasalazine/pharmacology , Time Factors
11.
Leuk Lymphoma ; 59(2): 460-468, 2018 02.
Article in English | MEDLINE | ID: mdl-28641473

ABSTRACT

Cancer cells reprogram their metabolism to optimize their growth and proliferation in the host microenvironment. For this purpose, they enhance the uptake of extracellular nutrients and deal with the metabolic waste products through the overexpression of numerous membrane proteins including amino-acid transporters (LAT1) and acid-base regulating enzymes, such as carbonic anhydrases (CAs). Here we describe the anti-tumoral effects of a new class of CAXII inhibitors, the glycosyl coumarins on T-ALL/LL cells. These effects appeared to be mediated through inhibition of mTOR/Akt pathway and c-myc downregulation. Interestingly, we show that the combined targeting of amino acid fluxes and pH regulators provides a promising therapeutic strategy in the future of T-ALL/LL management.


Subject(s)
Amino Acids, Essential/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Amino Acid Transport Systems/antagonists & inhibitors , Animals , Cell Death/drug effects , Cell Line, Tumor , Coumarins/pharmacology , Hydrogen-Ion Concentration , Intracellular Space/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Phosphatidylinositol 3-Kinases/metabolism , Protein Binding , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-myc/metabolism
12.
Proc Natl Acad Sci U S A ; 114(9): 2373-2378, 2017 02 28.
Article in English | MEDLINE | ID: mdl-28193893

ABSTRACT

By selection of winners of dyadic fights for 35 generations, we have generated a hyperaggressive Bully line of flies that almost always win fights against the parental wild-type Canton-S stock. Maintenance of the Bully phenotype is temperature dependent during development, with the phenotype lost when flies are reared at 19 °C. No similar effect is seen with the parent line. This difference allowed us to carry out RNA-seq experiments and identify a limited number of genes that are differentially expressed by twofold or greater in the Bullies; one of these was a putative transmembrane transporter, CG13646, which showed consistent and reproducible twofold down-regulation in Bullies. We examined the causal effect of this gene on the phenotype with a mutant line for CG13646, and with an RNAi approach. In all cases, reduction in expression of CG13646 by approximately half led to a hyperaggressive phenotype partially resembling that seen in the Bully flies. This gene is a member of a very interesting family of solute carrier proteins (SLCs), some of which have been suggested as being involved in glutamine/glutamate and GABA cycles of metabolism in excitatory and inhibitory nerve terminals in mammalian systems.


Subject(s)
Aggression , Amino Acid Transport Systems/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , RNA, Messenger/genetics , Amino Acid Transport Systems/antagonists & inhibitors , Amino Acid Transport Systems/metabolism , Animals , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Gene Expression Regulation , Male , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Temperature
13.
Sci Rep ; 5: 15057, 2015 Oct 12.
Article in English | MEDLINE | ID: mdl-26455426

ABSTRACT

The purine salvage pathway plays a major role in the nucleotide production, relying on the supply of nucleobases and nucleosides from extracellular sources. Although specific transporters have been suggested to be involved in facilitating their transport across the plasma membrane in mammals, those which are specifically responsible for utilization of extracellular nucleobases remain unknown. Here we present the molecular and functional characterization of SLC43A3, an orphan transporter belonging to an amino acid transporter family, as a purine-selective nucleobase transporter. SLC43A3 was highly expressed in the liver, where it was localized to the sinusoidal membrane of hepatocytes, and the lung. In addition, SLC43A3 expressed in MDCKII cells mediated the uptake of purine nucleobases such as adenine, guanine, and hypoxanthine without requiring typical driving ions such as Na(+) and H(+), but it did not mediate the uptake of nucleosides. When SLC43A3 was expressed in APRT/HPRT1-deficient A9 cells, adenine uptake was found to be low. However, it was markedly enhanced by the introduction of SLC43A3 with APRT. In HeLa cells, knock-down of SLC43A3 markedly decreased adenine uptake. These data suggest that SLC43A3 is a facilitative and purine-selective nucleobase transporter that mediates the cellular uptake of extracellular purine nucleobases in cooperation with salvage enzymes.


Subject(s)
Amino Acid Transport Systems/genetics , Equilibrative Nucleoside Transporter 1/genetics , Purines/metabolism , Adenine/metabolism , Adenine Phosphoribosyltransferase/antagonists & inhibitors , Adenine Phosphoribosyltransferase/genetics , Adenine Phosphoribosyltransferase/metabolism , Adenosine/metabolism , Amino Acid Transport Systems/antagonists & inhibitors , Amino Acid Transport Systems/metabolism , Animals , Biological Transport , Dogs , Equilibrative Nucleoside Transporter 1/antagonists & inhibitors , Equilibrative Nucleoside Transporter 1/metabolism , HEK293 Cells , HeLa Cells , Hepatocytes/metabolism , Humans , Hypoxanthine/metabolism , Liver/metabolism , Lung/metabolism , Madin Darby Canine Kidney Cells , Mice , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Thymidine/metabolism , Uridine/metabolism
14.
PLoS One ; 8(10): e76028, 2013.
Article in English | MEDLINE | ID: mdl-24204588

ABSTRACT

Eugenol is an aromatic component of clove oil that has therapeutic potential as an antifungal drug, although its mode of action and precise cellular target(s) remain ambiguous. To address this knowledge gap, a chemical-genetic profile analysis of eugenol was done using ∼4700 haploid Saccharomyces cerevisiae gene deletion mutants to reveal 21 deletion mutants with the greatest degree of susceptibility. Cellular roles of deleted genes in the most susceptible mutants indicate that the main targets for eugenol include pathways involved in biosynthesis and transport of aromatic and branched-chain amino acids. Follow-up analyses showed inhibitory effects of eugenol on amino acid permeases in the yeast cytoplasmic membrane. Furthermore, phenotypic suppression analysis revealed that eugenol interferes with two permeases, Tat1p and Gap1p, which are both involved in dual transport of aromatic and branched-chain amino acids through the yeast cytoplasmic membrane. Perturbation of cytoplasmic permeases represents a novel antifungal target and may explain previous observations that exposure to eugenol results in leakage of cell contents. Eugenol exposure may also contribute to amino acid starvation and thus holds promise as an anticancer therapeutic drug. Finally, this study provides further evidence of the usefulness of the yeast Gene Deletion Array approach in uncovering the mode of action of natural health products.


Subject(s)
Amino Acid Transport Systems/antagonists & inhibitors , Amino Acids, Aromatic/metabolism , Amino Acids, Branched-Chain/metabolism , Antifungal Agents/pharmacology , Cell Membrane/metabolism , Eugenol/pharmacology , Yeasts/drug effects , Yeasts/metabolism , Gene Deletion , Metabolic Networks and Pathways/drug effects , Phenotype , Protein Biosynthesis/drug effects , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Yeasts/genetics
15.
Epilepsy Res ; 107(1-2): 20-36, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24070846

ABSTRACT

Rats were given unilateral kainate injection into hippocampal CA3 region, and the effect of chronic electrographic seizures on extracellular glutamine (GLNECF) was examined in those with low and steady levels of extracellular glutamate (GLUECF). GLNECF, collected by microdialysis in awake rats for 5h, decreased to 62±4.4% of the initial concentration (n=6). This change correlated with the frequency and magnitude of seizure activity, and occurred in the ipsilateral but not in contralateral hippocampus, nor in kainate-injected rats that did not undergo seizure (n=6). Hippocampal intracellular GLN did not differ between the Seizure and No-Seizure Groups. These results suggested an intriguing possibility that seizure-induced decrease of GLNECF reflects not decreased GLN efflux into the extracellular fluid, but increased uptake into neurons. To examine this possibility, neuronal uptake of GLNECF was inhibited in vivo by intrahippocampal perfusion of 2-(methylamino)isobutyrate, a competitive and reversible inhibitor of the sodium-coupled neutral amino acid transporter (SNAT) subtypes 1 and 2, as demonstrated by 1.8±0.17 fold elevation of GLNECF (n=7). The frequency of electrographic seizures during uptake inhibition was reduced to 35±7% (n=7) of the frequency in pre-perfusion period, and returned to 88±9% in the post-perfusion period. These novel in vivo results strongly suggest that, in this well-established animal model of temporal-lobe epilepsy, the observed seizure-induced decrease of GLNECF reflects its increased uptake into neurons to sustain enhanced glutamatergic epileptiform activity, thereby demonstrating a possible new target for anti-seizure therapies.


Subject(s)
Glutamine/metabolism , Hippocampus/drug effects , Neurons/drug effects , Seizures/drug therapy , Amino Acid Transport System A/antagonists & inhibitors , Amino Acid Transport Systems/antagonists & inhibitors , Aminoisobutyric Acids/pharmacology , Animals , Glutamic Acid/metabolism , Hippocampus/metabolism , Kainic Acid , Male , Microdialysis , Neurons/metabolism , Rats , Rats, Wistar , Seizures/chemically induced , Seizures/metabolism , gamma-Aminobutyric Acid/metabolism
16.
Br J Pharmacol ; 170(5): 1041-52, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23962042

ABSTRACT

BACKGROUND AND PURPOSE: Intestinal nutrient transporters may mediate the uptake of drugs. The aim of this study was to investigate whether sertraline interacts with the intestinal proton-coupled amino acid transporter 1 PAT1 (SLC36A1). EXPERIMENTAL APPROACH: In vitro investigations of interactions between sertraline and human (h)PAT1, hSGLT1 (sodium-glucose linked transporter 1) and hPepT1 (proton-coupled di-/tri-peptide transporter 1) were conducted in Caco-2 cells using radiolabelled substrates. In vivo pharmacokinetic investigations were conducted in male Sprague-Dawley rats using gaboxadol (10 mg·kg(-1), p.o.) as a PAT1 substrate and sertraline (0-30.6 mg·kg(-1)). Gaboxadol was quantified by hydrophilic interaction chromatography followed by MS/MS detection. KEY RESULTS: Sertraline inhibited hPAT1-mediated L-[(3)H]-Pro uptake in Caco-2 cells. This interaction between sertraline and PAT1 appeared to be non-competitive. The uptake of the hSGLT1 substrate [(14)C]-α-methyl-D-glycopyranoside and the hPepT1 substrate [(14)C]-Gly-Sar in Caco-2 cells was also decreased in the presence of 0.3 mM sertraline. In rats, the administration of sertraline (0.1-10 mM, corresponding to 0.3-30.6 mg·kg(-1), p.o.) significantly reduced the maximal gaboxadol plasma concentration and AUC after its administration p.o. CONCLUSIONS AND IMPLICATIONS: Sertraline is an apparent non-competitive inhibitor of hPAT1-mediated transport in vitro. This inhibitory effect of sertraline is not specific to hPAT1 as substrate transport via hPepT1 and hSGLT1 was also reduced in the presence of sertraline. In vivo, sertraline reduced the amount of gaboxadol absorbed, suggesting that the inhibitory effect of sertraline on PAT1 occurs both in vitro and in vivo. Hence, sertraline could alter the bioavailability of drugs absorbed via PAT1.


Subject(s)
Amino Acid Transport Systems, Neutral/antagonists & inhibitors , Amino Acid Transport Systems/antagonists & inhibitors , Intestinal Absorption/drug effects , Intestinal Mucosa/drug effects , Selective Serotonin Reuptake Inhibitors/pharmacology , Sertraline/pharmacology , Symporters/antagonists & inhibitors , Administration, Oral , Amino Acid Transport Systems/metabolism , Amino Acid Transport Systems, Neutral/metabolism , Animals , Area Under Curve , Biological Availability , Caco-2 Cells , Chromatography/methods , Dose-Response Relationship, Drug , Humans , Hydrophobic and Hydrophilic Interactions , Intestinal Mucosa/metabolism , Isoxazoles/administration & dosage , Isoxazoles/blood , Isoxazoles/pharmacokinetics , Male , Peptide Transporter 1 , Proline/metabolism , Rats , Rats, Sprague-Dawley , Sodium-Glucose Transporter 1/antagonists & inhibitors , Sodium-Glucose Transporter 1/metabolism , Symporters/metabolism , Tandem Mass Spectrometry , Xenopus laevis
17.
Psychopharmacology (Berl) ; 228(3): 419-26, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23503685

ABSTRACT

RATIONALE: Several studies suggest that repeated nicotine administration causes alterations in glutaminergic transmission that may play an important role in developing and maintaining nicotine addiction. Chronic nicotine administration in rats decreases the expression of the glutamate transporter-1 (GLT-1) and cysteine-glutamate exchanger (system xC-) in the nucleus accumbens. We hypothesized that ceftriaxone, a GLT-1 and system xC- activator, would decrease murine behavioral aspects of nicotine dependence. OBJECTIVE: This study aimed to investigate the effect of repeated ceftriaxone administration on the behavioral effects of nicotine using mouse models of conditioned reward and withdrawal. METHOD: Using male ICR mice, the ability of repeated ceftriaxone injections to modulate the development and reinstatement of a nicotine-conditioned place preference (CPP) was evaluated. Additionally, nicotine withdrawal-associated signs were assessed. These included both physical (somatic signs and hyperalgesia) and affective (anxiety-related behaviors) withdrawal signs in mice. Finally, the effects of ceftriaxone on nicotine-induced antinociception and hypothermia after acute nicotine injection were measured. RESULT: Ceftriaxone had no effect on the development of nicotine preference but significantly attenuated nicotine-induced reinstatement of CPP. Furthermore, ceftriaxone reversed all nicotine withdrawal signs measured in mice. CONCLUSION: Altogether, these findings show that a ß-lactam antibiotic reduces nicotine withdrawal and nicotine-seeking behavior. Our results suggest that the documented efficacy of ceftriaxone against cocaine and morphine dependence-related behaviors effects extends to nicotine.


Subject(s)
Anti-Bacterial Agents/pharmacology , Ceftriaxone/pharmacology , Nicotine/adverse effects , Reinforcement, Psychology , Substance Withdrawal Syndrome/prevention & control , Tobacco Use Disorder/psychology , Amino Acid Transport Systems/agonists , Amino Acid Transport Systems/antagonists & inhibitors , Animals , Anti-Bacterial Agents/therapeutic use , Ceftriaxone/therapeutic use , Conditioning, Psychological/drug effects , Cysteine/metabolism , Excitatory Amino Acid Transporter 2/agonists , Excitatory Amino Acid Transporter 2/antagonists & inhibitors , Extinction, Psychological , Glutamic Acid/metabolism , Male , Mice , Mice, Inbred ICR , Reward , Substance Withdrawal Syndrome/metabolism , Substance Withdrawal Syndrome/psychology , Tobacco Use Disorder/metabolism
18.
Mol Membr Biol ; 30(2): 184-94, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22908980

ABSTRACT

Membrane proteins are intrinsically involved in both human and pathogen physiology, and are the target of 60% of all marketed drugs. During the past decade, advances in the studies of membrane proteins using X-ray crystallography, electron microscopy and NMR-based techniques led to the elucidation of over 250 unique membrane protein crystal structures. The aim of the European Drug Initiative for Channels and Transporter (EDICT) project is to use the structures of clinically significant membrane proteins for the development of lead molecules. One of the approaches used to achieve this is a virtual high-throughput screening (vHTS) technique initially developed for soluble proteins. This paper describes application of this technique to the discovery of inhibitors of the leucine transporter (LeuT), a member of the neurotransmitter:sodium symporter (NSS) family.


Subject(s)
Amino Acid Transport Systems/antagonists & inhibitors , Bacterial Proteins/antagonists & inhibitors , High-Throughput Screening Assays/methods , Leucine/metabolism , Membrane Transport Proteins/metabolism , Amino Acid Transport Systems/metabolism , Bacterial Proteins/metabolism , Binding Sites , Biological Transport , Crystallography, X-Ray , Plasma Membrane Neurotransmitter Transport Proteins/metabolism
19.
Mol Pharmacol ; 83(1): 22-32, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23012257

ABSTRACT

4-(2-Butyl-6,7-dichloro-2-cyclopentyl-indan-1-on-5-yl) oxobutyric acid (DCPIB) was identified as the selective blocker of volume-regulated anion channels (VRAC). VRAC are permeable to small inorganic and organic anions, including the excitatory neurotransmitter glutamate. In recent years DCPIB has been increasingly used for probing the physiologic and pathologic roles of VRAC and was found to potently suppress pathologic glutamate release in cerebral ischemia. Because ischemic glutamate release can be mediated by a plethora of mechanisms, in this study we explored the selectivity of DCPIB toward the majority of previously identified glutamate transporters and permeability pathways. l-[(3)H]glutamate, d-[(3)H]aspartate, and l-[(14)C]cystine were used to trace amino acid release and uptake. We found that in addition to its well-characterized effect on VRAC, DCPIB potently inhibited glutamate release via connexin hemichannels and glutamate uptake via the glutamate transporter GLT-1 in rat glial cells. In contrast, DCPIB had no direct effect on vesicular glutamate release from rat brain synaptosomes or the cystine/glutamate exchange in astrocytes. The compound did not affect the astrocytic glutamate transporter GLAST, nor did it block glutamate release via the P2X(7)/pannexin permeability pathway. The ability of DCPIB to directly block connexin hemichannels was confirmed using a gene-specific siRNA knockdown approach. Overall, our data demonstrate that DCPIB influences several glutamate transport pathways and that its effects on VRAC in vivo should be verified using additional pharmacological controls.


Subject(s)
Amino Acid Transport Systems/physiology , Astrocytes/drug effects , Cyclopentanes/pharmacology , Glutamic Acid/metabolism , Indans/pharmacology , Microglia/drug effects , Adenosine Triphosphate/pharmacology , Amino Acid Transport System y+/antagonists & inhibitors , Amino Acid Transport System y+/physiology , Amino Acid Transport Systems/antagonists & inhibitors , Amino Acid Transport Systems, Acidic , Animals , Astrocytes/metabolism , Biological Transport , Cells, Cultured , Cerebral Cortex/cytology , Connexins/antagonists & inhibitors , Connexins/physiology , Excitatory Amino Acid Transporter 1/antagonists & inhibitors , Excitatory Amino Acid Transporter 1/physiology , Excitatory Amino Acid Transporter 2/antagonists & inhibitors , Excitatory Amino Acid Transporter 2/physiology , Microglia/metabolism , Permeability , Primary Cell Culture , Rats , Receptors, Purinergic P2X7/physiology , Synaptosomes/metabolism
20.
Eur J Pharmacol ; 695(1-3): 34-9, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-22975709

ABSTRACT

Estrogen has great potential as a therapeutic agent in focal ischemic brain injury. Amino acids as energy resources and neurotransmitters in the central nervous system are crucial for proper neuronal function and excitability. The proton-coupled amino acid transporter PAT1 has clear potential in drug absorption. In this paper, human brain PAT1 was cloned and expressed in Xenopus oocytes. The effects of estradiol on the activity of PAT1 were investigated. Glycine-induced membrane currents mediated by PAT1 were measured using the two-electrode voltage clamp technique. The amplitude of the glycine-elicited current was decreased progressively with increasing concentrations of ß-estradiol. A concentration-dependent outwards current of PAT1 was also detected by the presence of ß-estradiol. We conclude that estrogen attenuates the activity of PAT1 by directly closing PAT1 channel. Our results may provide an additional mechanism for estrogen on neurotransmission and neuronal metabolism during ischemic injury.


Subject(s)
Amino Acid Transport Systems/antagonists & inhibitors , Amino Acid Transport Systems/genetics , Estradiol/pharmacology , Estrogens/pharmacology , Oocytes/metabolism , Symporters/antagonists & inhibitors , Symporters/genetics , Xenopus laevis/genetics , Amino Acid Transport Systems/metabolism , Animals , Brain/drug effects , Brain/metabolism , Brain/physiology , Electric Conductivity , Electrophysiological Phenomena/drug effects , Female , Gene Expression , Glycine/pharmacology , Humans , Kinetics , Symporters/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...