Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 346
Filtrar
1.
Front Immunol ; 15: 1357072, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38638435

RESUMO

Introduction: Clostridium perfringens α toxin is a main virulence factor responsible for gut damage in animals. Arginine is a functional amino acid exhibiting significant immunoregulatory activities. However, the effects and immunoregulatory mechanisms of arginine supplementation on α toxin-induced intestinal injury remain unclear. Methods: In vivo, 256 male Arbor Acres chickens were randomly assigned to a 2×2 factorial arrangement, involving diet treatments (with or without 0.3% arginine supplementation) and immunological stress (with or without α toxin challenge). In vitro, IEC-6 cells were treated with or without arginine in the presence or absence of α toxin. Moreover, IEC-6 cells were transfected with siRNA targeting mTOR and SLC38A9 to explore the underlying mechanisms. Results and discussion: The results showed that in vivo, arginine supplementation significantly alleviated the α toxin-induced growth performance impairment, decreases in serum immunoglobulin (Ig)A and IgG levels, and intestinal morphology damage. Arginine supplementation also significantly reduced the α toxin-induced increase in jejunal proinflammatory cytokines interleukin (IL)-1ß, IL-6 and IL-17 mRNA expression. Clostridium perfringens α toxin significantly decreased jejunal mechanistic target of rapamycin (mTOR) and solute carrier family 38 member 9 (SLC38A9) mRNA expression, while arginine supplementation significantly increased mTOR and SLC38A9 mRNA expression. In vitro, arginine pretreatment mitigated the α toxin-induced decrease in cell viability and the increase in cytotoxicity and apoptosis. Arginine pretreatment also alleviated the α toxin-induced upregulation of mRNA expression of inflammation-related cytokines IL-6, C-X-C motif chemokine ligand (CXCL)10, CXCL11 and transforming growth factor-ß (TGF-ß), as well as apoptosis-related genes B-cell lymphoma-2 associated X protein (Bax), B-cell lymphoma-2 (Bcl-2), B-cell lymphoma-extra large (Bcl-XL) and cysteinyl aspartate specific proteinase 3 (Caspase-3) and the ratio of Bax to Bcl-2. Arginine pretreatment significantly increased the α toxin-induced decrease in mTOR, SLC38A9, eukaryotic translation initiation factor 4E (eIF4E)-binding protein 1 (4EBP1) and ribosomal protein S6 kinase (S6K) mRNA expression. Knockdown SLC38A9 and mTOR largely abrogated the positive effects of arginine pretreatment on α toxin-induced intracellular changes. Furthermore, SLC38A9 silencing abolished the increased mTOR mRNA expression caused by arginine pretreatment. In conclusion, arginine administration attenuated α toxin-induced intestinal injury in vivo and in vitro, which could be associated with the downregulation of inflammation via regulating SLC38A9/mTORC1 pathway.


Assuntos
Arginina , Toxinas Bacterianas , Proteínas de Ligação ao Cálcio , Interleucina-6 , Fosfolipases Tipo C , Animais , Masculino , Arginina/farmacologia , Toxinas Bacterianas/toxicidade , Proteína X Associada a bcl-2 , Galinhas/genética , Inflamação , Alvo Mecanístico do Complexo 1 de Rapamicina , RNA Mensageiro/genética , Serina-Treonina Quinases TOR/metabolismo , Sistemas de Transporte de Aminoácidos/metabolismo
2.
Biol Pharm Bull ; 47(3): 652-659, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38508745

RESUMO

Platelets have been reported to exert diverse actions besides hemostasis and thrombus formation in the body. However, whether platelets affect transporter activity remains to be determined. In this study, we examined the effects of platelets on the activity of amino acid transporter system A, which is known to be changed by various factors, and we clarified the mechanism by which platelets affect system A activity. Among system A subtypes, we found that sodium-coupled neutral amino acid transporter (SNAT) 4 played a central role in the transport activity of system A in HuH-7 human hepatoma cells. Interestingly, platelets showed a biphasic effect on system A activity: activated platelet supernatants (APS) including the granule contents released from platelets downregulated system A activity at lower concentrations and the downregulation was suppressed at higher concentrations. The downregulation was due to a decrease in the affinity of SNAT4 for its substrate and not a decrease in the SNAT4 abundance on the plasma membrane. In addition, APS did not decrease the expression level of SNAT4 mRNA. On the other hand, platelets did not affect system A activity when the platelet suspension was added to HuH-7 cells. These results indicate that platelets indirectly affect the transport activity of system A by releasing bioactive substances but do not directly affect it by binding to HuH-7 cells.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Sistemas de Transporte de Aminoácidos/metabolismo , Plaquetas/metabolismo , Membrana Celular/metabolismo , RNA Mensageiro/genética
3.
Curr Opin Biotechnol ; 85: 103022, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38056204

RESUMO

Amino acid transporters (AATs) facilitate nutrient uptake and nutrient exchange between cancer and stromal cells. The posttranslational modification (PTM) of transporters is an important mechanism that tumor-associated cells use to dynamically regulate their function and stability in response to microenvironmental cues. In this review, we summarize recent findings that demonstrate the significance of N-glycosylation, phosphorylation, and ubiquitylation for the function of AATs. We also highlight powerful approaches that hijack the PTM machinery that could be used as therapeutics or tools to modulate transporter activity.


Assuntos
Processamento de Proteína Pós-Traducional , Microambiente Tumoral , Fosforilação , Glicosilação , Sistemas de Transporte de Aminoácidos/genética , Sistemas de Transporte de Aminoácidos/metabolismo
4.
Int J Mol Sci ; 24(7)2023 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-37047148

RESUMO

The L-type amino acid transporter (LAT) family contains four members, LAT1~4, which are important amino acid transporters. They mainly transport specific amino acids through cell membranes, provide nutrients to cells, and are involved in a variety of metabolic pathways. They regulate the mTOR signaling pathway which has been found to be strongly linked to cancer in recent years. However, in the field of prostate cancer (PCa), the LAT family is still in the nascent stage of research, and the importance of LATs in the diagnosis and treatment of prostate cancer is still unknown. Therefore, this article aims to report the role of LATs in prostate cancer and their clinical significance and application. LATs promote the progression of prostate cancer by increasing amino acid uptake, activating the mammalian target of rapamycin (mTOR) pathway and downstream signals, mediating castration-resistance, promoting tumor angiogenesis, and enhancing chemotherapy resistance. The importance of LATs as diagnostic and therapeutic targets for prostate cancer was emphasized and the latest research results were introduced. In addition, we introduced selective LAT1 inhibitors, including JPH203 and OKY034, which showed excellent inhibitory effects on the proliferation of various tumor cells. This is the future direction of amino acid transporter targeting therapy drugs.


Assuntos
Neoplasias da Próstata , Masculino , Humanos , Neoplasias da Próstata/diagnóstico , Neoplasias da Próstata/genética , Neoplasias da Próstata/terapia , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo , Aminoácidos/metabolismo , Transdução de Sinais , Sistemas de Transporte de Aminoácidos/genética , Sistemas de Transporte de Aminoácidos/metabolismo
5.
Placenta ; 135: 33-42, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36913807

RESUMO

INTRODUCTION: During pregnancy, the growth of the fetus is supported by the exchange of nutrients, waste, and other molecules between maternal and fetal circulations in the utero-placental unit. Nutrient transfer, in particular, is mediated by solute transporters such as solute carrier (SLC) and adenosine triphosphate-binding cassette (ABC) proteins. While nutrient transport has been extensively studied in the placenta, the role of human fetal membranes (FM), which was recently reported to have a role in drug transport, in nutrient uptake remains unknown. OBJECTIVES: This study determined nutrient transport expression in human FM and FM cells and compared expression with placental tissues and BeWo cells. METHODS: RNA sequencing (RNA-Seq) of placental and FM tissues and cells was done. Genes of major solute transporter groups, such as SLC and ABC, were identified. Proteomic analysis of cell lysates was performed via nano-liquid chromatography-tandem mass spectrometry (nanoLC-MS/MS) to confirm expression at a protein level. RESULTS: We determined that FM tissues and cells derived from the fetal membrane tissues express nutrient transporter genes, and their expression is similar to that seen in the placenta or BeWo cells. In particular, transporters involved in macronutrient and micronutrient transfer were identified in both placental and FM cells. Consistent with RNA-Seq findings, carbohydrate transporters (3), vitamin transport-related proteins (8), amino acid transporters (21), fatty acid transport-related proteins (9), cholesterol transport-related proteins (6) and nucleoside transporters (3) were identified in BeWo and FM cells, with both groups sharing similar nutrient transporter expression. CONCLUSION: This study determined the expression of nutrient transporters in human FMs. This knowledge is the first step in improving our understanding of nutrient uptake kinetics during pregnancy. Functional studies are required to determine the properties of nutrient transporters in human FMs.


Assuntos
Placenta , Espectrometria de Massas em Tandem , Gravidez , Feminino , Humanos , Placenta/metabolismo , Proteômica , Proteínas de Transporte , Sistemas de Transporte de Aminoácidos/metabolismo , Nutrientes
6.
Cell Mol Life Sci ; 80(1): 30, 2023 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-36609617

RESUMO

Tumor cells have an increased demand for nutrients to sustain their growth, but how these increased metabolic needs are ensured or how this influences tumor formation and progression remains unclear. To unravel tumor metabolic dependencies, particularly from extracellular metabolites, we have analyzed the role of plasma membrane metabolic transporters in Drosophila brain tumors. Using a well-established neural stem cell-derived tumor model, caused by brat knockdown, we have found that 13 plasma membrane metabolic transporters, including amino acid, carbohydrate and monocarboxylate transporters, are upregulated in tumors and are required for tumor growth. We identified CD98hc and several of the light chains with which it can form heterodimeric amino acid transporters, as crucial players in brat RNAi (brat IR) tumor progression. Knockdown of these components of CD98 heterodimers caused a dramatic reduction in tumor growth. Our data also reveal that the oncogene dMyc is required and sufficient for the upregulation of CD98 transporter subunits in these tumors. Furthermore, tumor-upregulated dmyc and CD98 transporters orchestrate the overactivation of the growth-promoting signaling pathway TOR, forming a core growth regulatory network to support brat IR tumor progression. Our findings highlight the important link between oncogenes, metabolism, and signaling pathways in the regulation of tumor growth and allow for a better understanding of the mechanisms necessary for tumor progression.


Assuntos
Neoplasias Encefálicas , Proteínas de Drosophila , Animais , Sistemas de Transporte de Aminoácidos/genética , Sistemas de Transporte de Aminoácidos/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Membrana Celular/metabolismo , Proteínas de Ligação a DNA/genética , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo , Regulação para Cima , Proteína-1 Reguladora de Fusão/metabolismo
7.
Basic Clin Pharmacol Toxicol ; 133(5): 459-472, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36460306

RESUMO

LAT1 and 4F2hc form a heterodimeric membrane protein complex, which functions as one of the best characterized amino acid transporters. Since LAT1-4F2hc is required for the efficient uptake of essential amino acids and hormones, it promotes cellular growth, in part, by stimulating mTORC1 (mechanistic target of rapamycin complex 1) signalling and by repressing the integrated stress response (ISR). Gain or loss of LAT1-4F2hc function is associated with cancer, diabetes, and immunological and neurological diseases. Hence, LAT1-4F2hc represents an attractive drug target for disease treatment. Specific targeting of LAT1-4F2hc will be facilitated by the increasingly detailed understanding of its molecular architecture, which provides important concepts for its function and regulation. Here, we summarize (i) structural insights that help to explain how LAT1 and 4F2hc assemble to transport amino acids across membranes, (ii) the role of LAT1-4F2hc in key metabolic signalling pathways, and (iii) how derailing these processes could contribute to diseases.


Assuntos
Sistemas de Transporte de Aminoácidos , Cadeia Pesada da Proteína-1 Reguladora de Fusão , Transportador 1 de Aminoácidos Neutros Grandes , Humanos , Sistemas de Transporte de Aminoácidos/metabolismo , Aminoácidos/metabolismo , Transporte Biológico , Cadeia Pesada da Proteína-1 Reguladora de Fusão/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo
8.
Microbiol Spectr ; 10(6): e0250122, 2022 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-36377953

RESUMO

Escherichia coli serine hydroxymethyltransferase (GlyA) converts serine to glycine, and glyA mutants are auxotrophic for glycine. CycA is a transporter that mediates glycine uptake. Deleting glyA in E. coli strain W3110 led to activation of CysB, which was related to novobiocin (NOV) susceptibility. Moreover, deleting glyA resulted in increased sensitivity to NOV, and this could be reversed by high concentrations of glycine. Reverse mutants of ΔglyA were selected and one of them had a mutation in yrdC, the gene encoding threonylcarbamoyl-AMP synthase. Subsequent proteome analysis showed that deleting glyA led to increased expression of TcyP and TdcB, making this bacterium dependent on CycA for glycine assimilation. Furthermore, deleting cycA in a ΔglyA background caused a severe growth defect on Luria-Bertani medium, which could be complemented by high concentrations of exogenous glycine. Mutation of yrdC led to decreased expression of TdcB but increased expression of ThrA/B/C and LtaE, which favored the conversion of threonine to glycine and thus avoided the dependence on CycA. Correspondingly, deleting of tcyP, tdcB, or gshA could reverse the NOV-sensitive phenotype of ΔglyA mutants. Overexpression of cycA resulted in increased sensitivity to NOV, whereas deleting this gene caused NOV resistance. Moreover, overexpression of cycA led to increased accumulation of NOV upon drug treatment. Therefore, inactivation of glyA in E. coli led to CycA-dependent glycine assimilation, which enhanced the accumulation of NOV and then made the bacterium more sensitive to this drug. These findings broaden our understanding of glycine metabolism and mechanisms of NOV susceptibility. IMPORTANCE Novobiocin (NOV) has been used in clinical practice as an ATPase inhibitor for decades. However, because it has been withdrawn from the market, pharmaceutical companies are searching for other ATPase inhibitors. Thus, probing the mechanisms of susceptibility to NOV will be beneficial to those efforts. In this study, we showed that inactivation of glyA in E. coli led to CycA-dependent glycine assimilation, which accompanied the accumulation of NOV and thereby increased the sensitivity to this drug. To date, this is the first report demonstrating the linkage between glycine assimilation and NOV susceptibility, and it is also the first report showing that YrdC is able to modulate the metabolic flux of threonine.


Assuntos
Sistemas de Transporte de Aminoácidos , Proteínas de Escherichia coli , Glicina , Adenosina Trifosfatases/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Glicina/metabolismo , Novobiocina/farmacologia , Treonina/metabolismo , Sistemas de Transporte de Aminoácidos/genética , Sistemas de Transporte de Aminoácidos/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo
9.
Biomolecules ; 12(10)2022 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-36291613

RESUMO

SLC6A14 (ATB0,+) is unique among SLC proteins in its ability to transport 18 of the 20 proteinogenic (dipolar and cationic) amino acids and naturally occurring and synthetic analogues (including anti-viral prodrugs and nitric oxide synthase (NOS) inhibitors). SLC6A14 mediates amino acid uptake in multiple cell types where increased expression is associated with pathophysiological conditions including some cancers. Here, we investigated how a key position within the core LeuT-fold structure of SLC6A14 influences substrate specificity. Homology modelling and sequence analysis identified the transmembrane domain 3 residue V128 as equivalent to a position known to influence substrate specificity in distantly related SLC36 and SLC38 amino acid transporters. SLC6A14, with and without V128 mutations, was heterologously expressed and function determined by radiotracer solute uptake and electrophysiological measurement of transporter-associated current. Substituting the amino acid residue occupying the SLC6A14 128 position modified the binding pocket environment and selectively disrupted transport of cationic (but not dipolar) amino acids and related NOS inhibitors. By understanding the molecular basis of amino acid transporter substrate specificity we can improve knowledge of how this multi-functional transporter can be targeted and how the LeuT-fold facilitates such diversity in function among the SLC6 family and other SLC amino acid transporters.


Assuntos
Aminoácidos , Pró-Fármacos , Aminoácidos/metabolismo , Sistemas de Transporte de Aminoácidos/genética , Sistemas de Transporte de Aminoácidos/metabolismo , Óxido Nítrico Sintase/metabolismo , Neurotransmissores
10.
Pathol Res Pract ; 239: 154169, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36257236

RESUMO

OBJECTIVE: In the mouse model, manipulations of assisted reproductive technology (ART) can lead to enlarged placentas and influence the expression of amino acid transporters in placentas during mid-to late-gestation. However, it is uncertain whether those abnormal changes presented in ART mouse placentas also occur in human ART placentas. METHODS: We choose the placenta tissue of pregnant woman by ART in term birth (ART group) and by natural pregnancy in term birth (control group) to compare the birth weight of the baby, placental weight and ratio of the fetal/placental weight of these two groups. We then detect the mRNA and protein expression of placental amino acid transporter genes (SNAT1, SNAT2, SNAT4, LAT1 and LAT2) in these two groups by real-time quantitative PCR, Western blot and immunofluorescence staining. Additionally, the PI3K-AKT-mTOR signaling activity was also analyzed. RESULTS: There were no statistical differences in new birth weight between the ART group and the control group. Compared with the control group, the placenta weight of baby was significantly higher in ART group and ratio of the fetal/placental weight was significantly lower in ART group. We found that the mRNA and protein expression of A system of amino acid transporter SNAT1, SNAT2 and SNAT4 in placenta were significantly down-regulated in ART group compared with placentas from natural pregnancies. Additionally, there were no statistical differences of the mRNA and protein expression of L system of amino acid transporter LAT1 between these two groups, but amino acid transporter LAT2 in placenta was significantly down-regulated in ART group. Furthermore, the PI3K-AKT-mTOR signaling activity was inhibited in ART group. CONCLUSIONS: ART leads to the expressions of amino acid transporter genes SNAT1, SNAT2, SNAT4 and LAT2 significantly down-regulated in placenta accompanied by the PI3K-AKT-mTOR signaling inhibition, which suggests that ART may affect the function of placenta amino acid transporter during the late pregnancy, leading to enlarged placentas and probably low birth weight.


Assuntos
Placenta , Proteínas Proto-Oncogênicas c-akt , Camundongos , Animais , Gravidez , Humanos , Feminino , Placenta/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Peso ao Nascer , Fosfatidilinositol 3-Quinases/metabolismo , Sistemas de Transporte de Aminoácidos/genética , Sistemas de Transporte de Aminoácidos/metabolismo , Serina-Treonina Quinases TOR/metabolismo , RNA Mensageiro/metabolismo , Técnicas de Reprodução Assistida
11.
Proc Natl Acad Sci U S A ; 119(41): e2205874119, 2022 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-36191186

RESUMO

ATB[Formula: see text] (SLC6A14) is a member of the amino acid transporter branch of the SLC6 family along with GlyT1 (SLC6A9) and GlyT2 (SLC6A5), two glycine-specific transporters coupled to 2:1 and 3:1 Na[Formula: see text]:Cl[Formula: see text], respectively. In contrast, ATB[Formula: see text] exhibits broad substrate specificity for all neutral and cationic amino acids, and its ionic coupling remains unsettled. Using the reversal potential slope method, we demonstrate a 3:1:1 Na[Formula: see text]:Cl[Formula: see text]:Gly stoichiometry for ATB[Formula: see text] that is consistent with its 2.1 e/Gly charge coupling. Like GlyT2, ATB[Formula: see text] behaves as a unidirectional transporter with virtually no glycine efflux at negative potentials after uptake, except by heteroexchange as remarkably shown by leucine activation of NMDARs in Xenopus oocytes coexpressing both membrane proteins. Analysis and computational modeling of the charge movement of ATB[Formula: see text] reveal a higher affinity for sodium in the absence of substrate than GlyT2 and a gating mechanism that locks Na[Formula: see text] into the apo-transporter at depolarized potentials. A 3:1 Na[Formula: see text]:Cl[Formula: see text] stoichiometry justifies the concentrative transport properties of ATB[Formula: see text] and explains its trophic role in tumor growth, while rationalizing its phylogenetic proximity to GlyT2 despite their extreme divergence in specificity.


Assuntos
Proteínas da Membrana Plasmática de Transporte de Glicina , Sódio , Sistemas de Transporte de Aminoácidos/metabolismo , Aminoácidos , Glicina/metabolismo , Proteínas da Membrana Plasmática de Transporte de Glicina/química , Íons/metabolismo , Leucina , Filogenia , Sódio/metabolismo
12.
Cell ; 185(20): 3739-3752.e18, 2022 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-36113465

RESUMO

Lysosomal amino acid efflux by proton-driven transporters is essential for lysosomal homeostasis, amino acid recycling, mTOR signaling, and maintaining lysosomal pH. To unravel the mechanisms of these transporters, we focus on cystinosin, a prototypical lysosomal amino acid transporter that exports cystine to the cytosol, where its reduction to cysteine supplies this limiting amino acid for diverse fundamental processes and controlling nutrient adaptation. Cystinosin mutations cause cystinosis, a devastating lysosomal storage disease. Here, we present structures of human cystinosin in lumen-open, cytosol-open, and cystine-bound states, which uncover the cystine recognition mechanism and capture the key conformational states of the transport cycle. Our structures, along with functional studies and double electron-electron resonance spectroscopic investigations, reveal the molecular basis for the transporter's conformational transitions and protonation switch, show conformation-dependent Ragulator-Rag complex engagement, and demonstrate an unexpected activation mechanism. These findings provide molecular insights into lysosomal amino acid efflux and a potential therapeutic strategy.


Assuntos
Cistina , Prótons , Sistemas de Transporte de Aminoácidos/metabolismo , Cisteína/metabolismo , Cistina/metabolismo , Humanos , Lisossomos/metabolismo , Serina-Treonina Quinases TOR/metabolismo
13.
J Pharmacol Exp Ther ; 382(3): 335-345, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35798387

RESUMO

6-Mercaptopurine (6-MP) is used extensively in the treatment of acute lymphoblastic leukemia (ALL) and inflammatory bowel diseases. Our laboratory determined previously, using a recombinant HEK293 cell model, that the SLC43A3-encoded equilibrative nucleobase transporter 1 (ENBT1) transports 6-MP into cells and significantly impacts the cytotoxicity of 6-MP in that model. To further investigate the clinical relevance of this finding, we now extend this work to an analysis of the impact of SLC43A3/ENBT1 expression and function on 6-MP uptake and cytotoxicity in leukemic lymphoblasts, the therapeutic target of 6-MP in ALL. A panel of ALL cell lines was assessed for SLC43A3/ENBT1 expression, ENBT1 function, and sensitivity to 6-MP. There was a significant difference in SLC43A3 expression among the cell lines that positively correlated with the rate of ENBT1-mediated 6-MP uptake. Cells with the lowest expression of SLC43A3 (SUP-B15: Vmax = 22± 5 pmol/µl per second) were also significantly less sensitive to 6-MP-induced cytotoxicity than were the highest expressing cells (ALL-1: Vmax = 69 ± 10 pmol/µl per second). Furthermore, knockdown of ENBT1 using short hairpin RNA interference (shRNAi) in RS4;11 cells caused a significant decrease in ENBT1-mediated 6-MP uptake (Vmax: RS4;11 = 40 ± 4 pmol/µl per second; RS4;11 shRNAi = 26 ± 3 pmol/µl per Second) and 6-MP cytotoxicity (EC50: RS4;11 = 0.58 ± 0.05 µM; RS4;11 shRNAi =1.44 ± 0.59 µM). This study showed that ENBT1 is a major contributor to 6-MP uptake in leukemia cell lines and may prove to be a biomarker for the therapeutic efficacy of 6-MP in patients with ALL. SIGNIFICANCE STATEMENT: This study shows that SLC43A3-encoded equilibrative nucleobase transporter 1 is responsible for the transport of 6-mercaptopurine (6-MP) into leukemia cells and that its level of expression can impact the cytotoxicity of 6-MP. Further studies are warranted to investigate the therapeutic implications in patient populations.


Assuntos
Mercaptopurina , Leucemia-Linfoma Linfoblástico de Células Precursoras , Sistemas de Transporte de Aminoácidos/metabolismo , Transporte Biológico , Células HEK293 , Humanos , Mercaptopurina/farmacologia , Mercaptopurina/uso terapêutico , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico
14.
IUBMB Life ; 74(7): 573-591, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35730628

RESUMO

S-adenosyl-L-methionine (SAM) is a coenzyme and the most commonly used methyl-group donor for the modification of metabolites, DNA, RNA and proteins. SAM biosynthesis and SAM regeneration from the methylation reaction product S-adenosyl-L-homocysteine (SAH) take place in the cytoplasm. Therefore, the intramitochondrial SAM-dependent methyltransferases require the import of SAM and export of SAH for recycling. Orthologous mitochondrial transporters belonging to the mitochondrial carrier family have been identified to catalyze this antiport transport step: Sam5p in yeast, SLC25A26 (SAMC) in humans, and SAMC1-2 in plants. In mitochondria SAM is used by a vast number of enzymes implicated in the following processes: the regulation of replication, transcription, translation, and enzymatic activities; the maturation and assembly of mitochondrial tRNAs, ribosomes and protein complexes; and the biosynthesis of cofactors, such as ubiquinone, lipoate, and molybdopterin. Mutations in SLC25A26 and mitochondrial SAM-dependent enzymes have been found to cause human diseases, which emphasizes the physiological importance of these proteins.


Assuntos
Mitocôndrias , S-Adenosilmetionina , Sistemas de Transporte de Aminoácidos/genética , Sistemas de Transporte de Aminoácidos/metabolismo , Transporte Biológico , Proteínas de Ligação ao Cálcio/metabolismo , Humanos , Mitocôndrias/genética , Mitocôndrias/metabolismo , S-Adenosilmetionina/metabolismo
15.
Curr Opin Struct Biol ; 74: 102389, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35605357

RESUMO

Heteromeric amino acid transporters (HATs) are one of the ten types of amino acid transporters present in the human body. Growing interest in the pathophysiological role of this group of transporters in rare and complex diseases and cancer has brought about the recent resolution of various structures of human HATs and bacterial homologues at atomic level. This knowledge sheds light on the mechanisms of transport used by these molecules. Here, we discuss the molecular bases underlying substrate specificity, binding asymmetry, and the impact of disease-causing mutations on transporter biogenesis and function.


Assuntos
Sistemas de Transporte de Aminoácidos , Biologia Molecular , Sistemas de Transporte de Aminoácidos/química , Sistemas de Transporte de Aminoácidos/genética , Sistemas de Transporte de Aminoácidos/metabolismo , Transporte Biológico/fisiologia , Humanos , Especificidade por Substrato
16.
Nat Commun ; 13(1): 2708, 2022 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-35577790

RESUMO

Cystinuria is a genetic disorder characterized by overexcretion of dibasic amino acids and cystine, causing recurrent kidney stones and kidney failure. Mutations of the regulatory glycoprotein rBAT and the amino acid transporter b0,+AT, which constitute system b0,+, are linked to type I and non-type I cystinuria respectively and they exhibit distinct phenotypes due to protein trafficking defects or catalytic inactivation. Here, using electron cryo-microscopy and biochemistry, we discover that Ca2+ mediates higher-order assembly of system b0,+. Ca2+ stabilizes the interface between two rBAT molecules, leading to super-dimerization of b0,+AT-rBAT, which in turn facilitates N-glycan maturation and protein trafficking. A cystinuria mutant T216M and mutations of the Ca2+ site of rBAT cause the loss of higher-order assemblies, resulting in protein trapping at the ER and the loss of function. These results provide the molecular basis of system b0,+ biogenesis and type I cystinuria and serve as a guide to develop new therapeutic strategies against it. More broadly, our findings reveal an unprecedented link between transporter oligomeric assembly and protein-trafficking diseases.


Assuntos
Sistemas de Transporte de Aminoácidos Básicos , Cálcio , Cistinúria , Sistemas de Transporte de Aminoácidos/metabolismo , Sistemas de Transporte de Aminoácidos Básicos/metabolismo , Sistemas de Transporte de Aminoácidos Básicos/ultraestrutura , Cálcio/química , Cálcio/metabolismo , Cistina/metabolismo , Cistinúria/genética , Cistinúria/metabolismo , Humanos
17.
Biochem Biophys Res Commun ; 614: 41-46, 2022 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-35569376

RESUMO

SLC6A14 is a plasma membrane transporter specific for neutral and basic amino acids, upregulated in many tumors. This study focused on breast cancer cell lines, showing the fully glycosylated band, known to be at the cell surface, in estrogen receptor positive lines. Inhibition of heat shock protein 90ß (HSP90ß) decreased the level of this band, what correlated with a decrease of SLC6A14 transport activity. A direct interaction between SLC6A14 and HSP90ß was confirmed in proximity ligation assay, pointing to the role of HSP90 in folding control in endoplasmic reticulum and affecting farther transporter trafficking to the cell surface. Either inhibitor of SLC6A14 (α-methyltryptophan) or of HSP90 (radicicol) had the cytotoxic effect, when added alone, while treatment with both compounds had a synergistic effect. This points to SLC6A14 as a druggable target in breast cancer and a combination therapy being more efficient in killing cancer cells.


Assuntos
Sistemas de Transporte de Aminoácidos , Neoplasias da Mama , Sistemas de Transporte de Aminoácidos/metabolismo , Neoplasias da Mama/metabolismo , Retículo Endoplasmático/metabolismo , Feminino , Proteínas de Choque Térmico HSP90/metabolismo , Proteínas de Choque Térmico/metabolismo , Humanos
18.
Am J Hum Genet ; 109(2): 253-269, 2022 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-35065708

RESUMO

Mucus obstruction is a central feature in the cystic fibrosis (CF) airways. A genome-wide association study (GWAS) of lung disease by the CF Gene Modifier Consortium (CFGMC) identified a significant locus containing two mucin genes, MUC20 and MUC4. Expression quantitative trait locus (eQTL) analysis using human nasal epithelia (HNE) from 94 CF-affected Canadians in the CFGMC demonstrated MUC4 eQTLs that mirrored the lung association pattern in the region, suggesting that MUC4 expression may mediate CF lung disease. Complications arose, however, with colocalization testing using existing methods: the locus is complex and the associated SNPs span a 0.2 Mb region with high linkage disequilibrium (LD) and evidence of allelic heterogeneity. We previously developed the Simple Sum (SS), a powerful colocalization test in regions with allelic heterogeneity, but SS assumed eQTLs to be present to achieve type I error control. Here we propose a two-stage SS (SS2) colocalization test that avoids a priori eQTL assumptions, accounts for multiple hypothesis testing and the composite null hypothesis, and enables meta-analysis. We compare SS2 to published approaches through simulation and demonstrate type I error control for all settings with the greatest power in the presence of high LD and allelic heterogeneity. Applying SS2 to the MUC20/MUC4 CF lung disease locus with eQTLs from CF HNE revealed significant colocalization with MUC4 (p = 1.31 × 10-5) rather than with MUC20. The SS2 is a powerful method to inform the responsible gene(s) at a locus and guide future functional studies. SS2 has been implemented in the application LocusFocus.


Assuntos
Sistemas de Transporte de Aminoácidos/genética , Fibrose Cística/genética , Modelos Estatísticos , Mucina-4/genética , Mucinas/genética , Locos de Características Quantitativas , Alelos , Sistemas de Transporte de Aminoácidos/metabolismo , Fibrose Cística/metabolismo , Fibrose Cística/patologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Heterogeneidade Genética , Genoma Humano , Estudo de Associação Genômica Ampla , Humanos , Desequilíbrio de Ligação , Pulmão/metabolismo , Pulmão/patologia , Mucina-4/metabolismo , Mucinas/metabolismo , Mucosa Nasal/metabolismo , Mucosa Nasal/patologia , Polimorfismo de Nucleotídeo Único
19.
Neurochem Res ; 47(1): 23-36, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33606172

RESUMO

In humans, more than 50 transporters are responsible for the traffic and balance of amino acids within and between cells and tissues, and half of them have been associated with disease [1]. Covering all common amino acids, Heteromeric Amino acid Transporters (HATs) are one class of such transporters. This review first highlights structural and functional studies that solved the atomic structure of HATs and revealed molecular clues on substrate interaction. Moreover, this review focuses on HATs that have a role in the central nervous system (CNS) and that are related to neurological diseases, including: (i) LAT1/CD98hc and its role in the uptake of branched chain amino acids trough the blood brain barrier and autism. (ii) LAT2/CD98hc and its potential role in the transport of glutamine between plasma and cerebrospinal fluid. (iii) y+LAT2/CD98hc that is emerging as a key player in hepatic encephalopathy. xCT/CD98hc as a potential therapeutic target in glioblastoma, and (iv) Asc-1/CD98hc as a potential therapeutic target in pathologies with alterations in NMDA glutamate receptors.


Assuntos
Sistemas de Transporte de Aminoácidos , Sistemas de Transporte de Aminoácidos/metabolismo , Aminoácidos/metabolismo , Transporte Biológico , Barreira Hematoencefálica/metabolismo , Humanos
20.
Cells ; 10(12)2021 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-34943927

RESUMO

Induced Pluripotent Stem Cells (iPSCs) can be differentiated into epithelial organoids that recapitulate the relevant context for CFTR and enable testing of therapies targeting Cystic Fibrosis (CF)-causing mutant proteins. However, to date, CF-iPSC-derived organoids have only been used to study pharmacological modulation of mutant CFTR channel activity and not the activity of other disease-relevant membrane protein constituents. In the current work, we describe a high-throughput, fluorescence-based assay of CFTR channel activity in iPSC-derived intestinal organoids and describe how this method can be adapted to study other apical membrane proteins. Specifically, we show how this assay can be employed to study CFTR and ENaC channels and an electrogenic acid transporter in the same iPSC-derived intestinal tissue. This phenotypic platform promises to expand CF therapy discovery to include strategies that target multiple determinants of epithelial fluid transport.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Intestinos/metabolismo , Organoides/metabolismo , Sistemas de Transporte de Aminoácidos/metabolismo , Animais , Diferenciação Celular , Cães , Canais Epiteliais de Sódio/metabolismo , Edição de Genes , Humanos , Células Madin Darby de Rim Canino
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA