Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 100
Filter
1.
JCI Insight ; 6(17)2021 09 08.
Article in English | MEDLINE | ID: mdl-34494553

ABSTRACT

BACKGROUNDTargeted arterial infusion of verapamil combined with chemotherapy (TVCC) is an effective clinical interventional therapy for esophageal squamous cell carcinoma (ESCC), but multidrug resistance (MDR) remains the major cause of relapse or poor prognosis, and the underlying molecular mechanisms of MDR, temporal intratumoral heterogeneity, and clonal evolutionary processes of resistance have not been determined.METHODSTo elucidate the roles of genetic and epigenetic alterations in the evolution of acquired resistance during therapies, we performed whole-exome sequencing on 16 serial specimens from 7 patients with ESCC at every cycle of therapeutic intervention from 3 groups, complete response, partial response, and progressive disease, and we performed whole-genome bisulfite sequencing for 3 of these 7 patients, 1 patient from each group.RESULTSPatients with progressive disease exhibited a substantially higher genomic and epigenomic temporal heterogeneity. Subclonal expansions driven by the beneficial new mutations were observed during combined therapies, which explained the emergence of MDR. Notably, SLC7A8 was identified as a potentially novel MDR gene, and functional assays demonstrated that mutant SLC7A8 promoted the resistance phenotypes of ESCC cell lines. Promoter methylation dynamics during treatments revealed 8 drug resistance protein-coding genes characterized by hypomethylation in promoter regions. Intriguingly, promoter hypomethylation of SLC8A3 and mutant SLC7A8 were enriched in an identical pathway, protein digestion and absorption, indicating a potentially novel MDR mechanism during treatments.CONCLUSIONOur integrated multiomics investigations revealed the dynamics of temporal genetic and epigenetic inter- and intratumoral heterogeneity, clonal evolutionary processes, and epigenomic changes, providing potential MDR therapeutic targets in treatment-resistant patients with ESCC during combined therapies.FUNDINGNational Natural Science Foundation of China, Science Foundation of Peking University Cancer Hospital, CAMS Innovation Fund for Medical Sciences, Major Program of Shenzhen Bay Laboratory, Guangdong Basic and Applied Basic Research Foundation, and the third round of public welfare development and reform pilot projects of Beijing Municipal Medical Research Institutes.


Subject(s)
Amino Acid Transport System y+/genetics , Antineoplastic Agents/therapeutic use , Drug Resistance, Neoplasm/genetics , Epigenomics/methods , Esophageal Neoplasms/genetics , Esophageal Squamous Cell Carcinoma/genetics , Fusion Regulatory Protein 1, Light Chains/genetics , Mutation , Amino Acid Transport System y+/metabolism , Combined Modality Therapy , DNA Methylation , DNA, Neoplasm/genetics , Esophageal Neoplasms/pathology , Esophageal Neoplasms/therapy , Esophageal Squamous Cell Carcinoma/pathology , Esophageal Squamous Cell Carcinoma/therapy , Female , Fusion Regulatory Protein 1, Light Chains/metabolism , Humans , Male , Exome Sequencing
2.
J Physiol Sci ; 71(1): 11, 2021 Mar 31.
Article in English | MEDLINE | ID: mdl-33789576

ABSTRACT

L-type amino acid transporter 2 (LAT2) is a Na+-independent neutral amino acid transporter, whose function regulation system remains unclarified. Since protein kinase C (PKC) is known to regulate the functions of various transporters, we investigated whether human LAT2 (hLAT2) function is regulated by PKC. In mouse proximal tubule S2 cells, hLAT2 transport activity was upregulated by PKC activation. However, we found that the mRNA and protein expression of hLAT2 was not affected by PKC activation and that the upregulation was independent of the three potential PKC consensus sites in the hLAT2 amino acid sequence. Moreover, we found that PKC activation upregulated the Vmax value for hLAT2-mediated alanine transport, which was not accompanied by the induction of hLAT2 membrane insertion. In conclusion, we showed that hLAT2 function is upregulated by PKC activation, which is not related to either the de novo synthesis, the phosphorylation or the membrane insertion of hLAT2.


Subject(s)
Amino Acid Transport System y+/metabolism , Fusion Regulatory Protein 1, Light Chains/metabolism , Protein Kinase C/metabolism , Amino Acid Transport System y+/genetics , Animals , Cell Line , Cell Survival , Cloning, Molecular , Fusion Regulatory Protein 1, Light Chains/genetics , Gene Expression Regulation/drug effects , Humans , Indoles/pharmacology , Maleimides/pharmacology , Mice , Mutagenesis, Site-Directed , Up-Regulation
3.
Placenta ; 103: 188-198, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33160252

ABSTRACT

INTRODUCTION: Amino acid transport across the placenta is crucial for fetal growth. In rodent models, the visceral yolk sac (referred to as yolk sac hereafter) is also likely to contribute to fetal amino acid provision. System L amino acid transporters mediate the transport of essential amino acids. System L activity is mediated by light chains LAT1 (Slc7a5) and LAT2 (Slc7a8) which form functional complexes by heterodimeric linkage to CD98 (Slc3a2). LAT4 (Slc43a2) is monomeric, possessing overlapping amino acid substrate specificity with LAT1 and LAT2. METHODS: This study investigates the expression of these LAT subtypes in fetus-matched rat placenta and yolk sac. RESULTS: Slc7a5, Slc7a8 and Slc43a2 transcripts were expressed in placenta and yolk sac with similar expression patterns between sexes. LAT1 expression was significantly higher in placenta than yolk sac. Conversely, LAT2 and LAT4 expression was significantly higher in yolk sac than placenta; CD98 expression was comparable. LAT1, LAT2, LAT4 and CD98 were distributed to rat placental labyrinth zone (LZ) and junctional zone (JZ). LAT1 and LAT4 demonstrated higher expression in LZ, whilst LAT2 was more intensely distributed to JZ. LAT1, LAT2, LAT4 and CD98 were expressed in yolk sac, with punctate LAT1 staining to endodermal cell cytoplasm, contrasting with the intense LAT2, LAT4 and CD98 endodermal cell basolateral distribution, accounting for greater LAT2 and LAT4 expression in yolk sac compared to placenta. CONCLUSION: LAT1, LAT2 and LAT4 are expressed in rat placenta and yolk sac implicating a combined role for these LAT subtypes in supporting fetal growth and development.


Subject(s)
Amino Acid Transport System L/genetics , Placenta/metabolism , Yolk Sac/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Transport System L/classification , Amino Acid Transport System L/metabolism , Amino Acid Transport System y+/genetics , Amino Acid Transport System y+/metabolism , Animals , Female , Fusion Regulatory Protein 1, Light Chains/genetics , Fusion Regulatory Protein 1, Light Chains/metabolism , Gene Expression Regulation, Developmental , Large Neutral Amino Acid-Transporter 1/genetics , Large Neutral Amino Acid-Transporter 1/metabolism , Male , Pregnancy , Rats , Rats, Wistar
4.
Int J Mol Sci ; 21(20)2020 Oct 14.
Article in English | MEDLINE | ID: mdl-33066406

ABSTRACT

The human L-type amino acid transporters LAT1 and LAT2 mediate the transport of amino acids and amino acid derivatives across plasma membranes in a sodium-independent, obligatory antiport mode. In mammalian cells, LAT1 and LAT2 associate with the type-II membrane N-glycoprotein 4F2hc to form heteromeric amino acid transporters (HATs). The glycosylated ancillary protein 4F2hc is known to be important for successful trafficking of the unglycosylated transporters to the plasma membrane. The heavy (i.e., 4F2hc) and light (i.e., LAT1 and LAT2) chains belong to the solute carrier (SLC) families SLC3 and SLC7, and are covalently linked by a conserved disulfide bridge. Overexpression, absence, or malfunction of certain HATs is associated with human diseases and HATs are therefore considered therapeutic targets. Here, we present a comparative, functional characterization of the HATs 4F2hc-LAT1 and 4F2hc-LAT2, and their light chains LAT1 and LAT2. For this purpose, the HATs and the light chains were expressed in the methylotrophic yeast Pichia pastoris and a radiolabel transport assay was established. Importantly and in contrast to mammalian cells, P. pastoris has proven useful as eukaryotic expression system to successfully express human LAT1 and LAT2 in the plasma membrane without the requirement of co-expressed trafficking chaperone 4F2hc. Our results show a novel function of the heavy chain 4F2hc that impacts transport by modulating the substrate affinity and specificity of corresponding LATs. In addition, the presented data confirm that the light chains LAT1 and LAT2 constitute the substrate-transporting subunits of the HATs, and that light chains are also functional in the absence of the ancillary protein 4F2hc.


Subject(s)
Amino Acid Transport System y+/metabolism , Fusion Regulatory Protein 1, Heavy Chain/metabolism , Fusion Regulatory Protein 1, Light Chains/metabolism , Large Neutral Amino Acid-Transporter 1/metabolism , Amino Acid Transport System y+/genetics , Fusion Regulatory Protein 1, Light Chains/genetics , Histidine/metabolism , Humans , Large Neutral Amino Acid-Transporter 1/genetics , Leucine/metabolism , Pichia , Protein Binding , Protein Transport , Substrate Specificity
5.
Fluids Barriers CNS ; 17(1): 17, 2020 Feb 11.
Article in English | MEDLINE | ID: mdl-32046769

ABSTRACT

BACKGROUND: Cerebrospinal fluid (CSF) is mainly produced by the choroid plexus (CP) located in brain ventricles. Although derived from blood plasma, it is nearly protein-free (~ 250-fold less) and contains about 2-20-fold less free amino acids, with the exception of glutamine (Gln) which is nearly equal. The aim of this study was to determine which amino acid transporters are expressed in mouse CP epithelium in order to gain understanding about how this barrier maintains the observed amino acid concentration gradient. METHODS: Expression of amino acid transporters was assessed in isolated choroid plexuses (CPs) by qRT-PCR followed by localization studies using immunofluorescence with specific antibodies. The impact of LAT2 (Slc7a8) antiporter deletion on CSF amino acids was determined. RESULTS: The purity of isolated choroid plexuses was tested on the mRNA level using specific markers, in particular transthyretin (Ttr) that was enriched 330-fold in CP compared to cerebral tissue. In a first experimental round, 14 out of 32 Slc amino acid transporters tested on the mRNA level by qPCR were selected for further investigation. Out of these, five were considered highly expressed, SNAT1 (Slc38a1), SNAT3 (Slc38a3), LAT2 (Slc7a8), ASC1 (Slc7a10) and SIT1 (Slc6a20b). Three of them were visualized by immunofluorescence: SNAT1 (Slc38a1), a neutral amino acid-Na+ symporter, found at the blood side basolateral membrane of CP epithelium, while SNAT3 (Slc38a3), an amino acid-Na+ symporter and H+ antiporter, as well as LAT2 (Slc7a8), a neutral amino acid antiporter, were localized at the CSF-facing luminal membrane. In a LAT2 knock-out mouse model, CSF Gln was unchanged, whereas other amino acids normally 2-20-fold lower than in plasma, were increased, in particular the LAT2 uptake substrates leucine (Leu), valine (Val) and tryptophan (Trp) and some other amino acids such as glutamate (Glu), glycine (Gly) and proline (Pro). CONCLUSION: These results suggest that Gln is actively transported by SNAT1 from the blood into CP epithelial cells and then released luminally into CSF via SNAT3 and LAT2. Its efflux via LAT2 may drive the reuptake from the CSF of essential amino acid substrates of this antiporter and thereby participates to maintaining the amino acid gradient between plasma and CSF.


Subject(s)
Amino Acid Transport System y+/metabolism , Amino Acid Transport Systems, Neutral/metabolism , Amino Acids/cerebrospinal fluid , Epithelial Cells/metabolism , Fusion Regulatory Protein 1, Light Chains/metabolism , Homeostasis , Amino Acid Transport System y+/genetics , Animals , Biological Transport/genetics , Biological Transport/physiology , Cells, Cultured , Choroid Plexus/metabolism , Female , Fusion Regulatory Protein 1, Light Chains/genetics , Glutamic Acid/metabolism , Homeostasis/physiology , Male , Mice, Knockout , Prealbumin/metabolism
6.
Mol Genet Genomic Med ; 8(1): e1036, 2020 01.
Article in English | MEDLINE | ID: mdl-31701662

ABSTRACT

BACKGROUND: Autism spectrum disorder (ASD) is a common neurodevelopmental disorder whose molecular mechanisms are largely unknown. Several studies have shown an association between ASD and abnormalities in the metabolism of amino acids, specifically tryptophan and branched-chain amino acids (BCAAs). METHODS: Ninety-seven patients with ASD were screened by Sanger sequencing the genes encoding the heavy (SLC3A2) and light subunits (SLC7A5 and SLC7A8) of the large amino acid transporters (LAT) 1 and 2. LAT1 and 2 are responsible for the transportation of tryptophan and BCAA across the blood-brain barrier and are expressed both in blood and brain. Functional studies were performed employing the Biolog Phenotype Microarray Mammalian (PM-M) technology to investigate the metabolic profiling in lymphoblastoid cell lines from 43 patients with ASD and 50 controls with particular focus on the amino acid substrates of LATs. RESULTS: We detected nine likely pathogenic variants in 11 of 97 patients (11.3%): three in SLC3A2, three in SLC7A5, and three in SLC7A8. Six variants of unknown significance were detected in eight patients, two of which also carrying a likely pathogenic variant. The functional studies showed a consistently reduced utilization of tryptophan, accompanied by evidence of reduced utilization of other large aromatic amino acids (LAAs), either alone or as part of a dipeptide. CONCLUSION: Coding variants in the LAT genes were detected in 17 of 97 patients with ASD (17.5%). Metabolic assays indicate that such abnormalities affect the utilization of certain amino acids, particularly tryptophan and other LAAs, with potential consequences on their transport across the blood barrier and their availability during brain development. Therefore, abnormalities in the LAT1 and two transporters are likely associated with an increased risk of developing ASD.


Subject(s)
Amino Acid Transport System y+/genetics , Autism Spectrum Disorder/genetics , Fusion Regulatory Protein 1, Heavy Chain/genetics , Fusion Regulatory Protein 1, Light Chains/genetics , Large Neutral Amino Acid-Transporter 1/genetics , Adolescent , Adult , Autism Spectrum Disorder/metabolism , Cell Line , Child , Child, Preschool , Female , Humans , Male , Mutation , Tryptophan/metabolism
7.
J Hum Genet ; 64(9): 849-858, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31213652

ABSTRACT

Lysinuric protein intolerance (LPI) is caused by dysfunction of the dibasic amino acid membrane transport owing to the functional abnormality of y+L amino acid transporter-1 (y+ LAT-1). LPI is associated with autosomal recessive inheritance and pathological variants in the responsible gene SLC7A7 are also observed. The pathophysiology of this disease had earlier been understood as a transport defect in polarized cells (e.g., intestinal or renal tubular epithelium); however, in recent years, transport defects in non-polarized cells such as lymphocytes and macrophages have also been recognized as important. Although the former can cause death, malnutrition, and urea cycle dysfunction (hyperammonemia), the latter can induce renal, pulmonary, and immune disorders. Furthermore, although therapeutic interventions can prevent hyperammonemic episodes to some extent, progression of pulmonary and renal complications cannot be prevented, thereby influencing prognosis. Such pathological conditions are currently being explored and further investigation would prove beneficial. In this study, we have summarized the basic pathology as revealed in recent years, along with the clinical aspects and genetic features.


Subject(s)
Amino Acid Metabolism, Inborn Errors , Fusion Regulatory Protein 1, Light Chains , Kidney , Large Neutral Amino Acid-Transporter 1 , Mutation , Amino Acid Metabolism, Inborn Errors/genetics , Amino Acid Metabolism, Inborn Errors/metabolism , Amino Acid Metabolism, Inborn Errors/pathology , Amino Acid Metabolism, Inborn Errors/therapy , Amino Acid Transport System y+L , Fusion Regulatory Protein 1, Light Chains/genetics , Fusion Regulatory Protein 1, Light Chains/metabolism , Humans , Kidney/metabolism , Kidney/pathology , Large Neutral Amino Acid-Transporter 1/genetics , Large Neutral Amino Acid-Transporter 1/metabolism
8.
Zhongguo Dang Dai Er Ke Za Zhi ; 21(4): 375-380, 2019 Apr.
Article in Chinese | MEDLINE | ID: mdl-31014432

ABSTRACT

Lysinuric protein intolerance (LPI) is an autosomal recessive disorder caused by SLC7A7 gene mutation and often involves severe lesions in multiple systems. Lung involvement is frequently seen in children with LPI and such children tend to have a poor prognosis. This article summarizes the clinical manifestations and gene mutation characteristics of three children diagnosed with LPI by SLC7A7 gene analysis. All three children had the manifestations of aversion to protein-rich food after weaning, delayed development, anemia, hepatosplenomegaly, and osteoporosis, as well as an increase in orotic acid in urine. In addition, interstitial pneumonia and diffuse pulmonary interstitial lesions were observed in two children. SLC7A7 gene detection showed three pathogenic mutations in these children, namely c.1387delG(p.V463CfsX56), c.1215G>A(p.W405X) and homozygous c.625+1G>A. After a definite diagnosis was made, all three children were given a low-protein diet and oral administration of citrulline [100 mg/(kg.d)], iron protein succinylate [4 mg/(kg.d)], calcium and zinc gluconates oral solution (10 mL/day) and vitamin D (400 IU/day). In addition, patient 3 was given prednisone acetate (5 mg/day). The children had varying degrees of improvement in symptoms and signs. It is hard to distinguish LPI from urea cycle disorder due to the features of amino acid and organic acid metabolism in LPI, and SLC7A7 gene analysis is the basis for a definite diagnosis of LPI.


Subject(s)
Amino Acid Metabolism, Inborn Errors , Fusion Regulatory Protein 1, Light Chains/genetics , Amino Acid Metabolism, Inborn Errors/genetics , Amino Acid Transport System y+L , Child , Citrulline , Humans , Lysine , Mutation
9.
Orphanet J Rare Dis ; 14(1): 63, 2019 03 04.
Article in English | MEDLINE | ID: mdl-30832686

ABSTRACT

BACKGROUND: y+LAT1, encoded by SCL7A7, is the protein mutated in Lysinuric Protein Intolerance (LPI), a rare metabolic disease caused by a defective cationic amino acid (CAA, arginine, lysine, ornithine) transport at the basolateral membrane of intestinal and renal tubular cells. The disease is characterized by protein-rich food intolerance with secondary urea cycle disorder, but symptoms are heterogeneous with lung and immunological complications that are not explainable by the CAA transport defect. With the exception of the Finnish founder mutation (c.895-2A > T, LPIFin), LPI-causative mutations are heterogeneous and genotype-phenotype correlations have not been found. Here we addressed system y+L-mediated arginine uptake in monocytes from three LPI Italian patients and in lymphoblasts carrying the same mutations; in parallel, the genetic defects carried by the patients were reproduced as eGFP-tagged y+LAT1 mutants in transfected CHO cells to define the function and localization protein. RESULTS: System y+L activity is impaired in monocytes isolated from all LPI patients, and in CHO cells transfected with the three eGFP-y+LAT1 mutants, but not in lymphoblasts bearing the same mutations. The analysis of protein localization with confocal microscopy revealed that the eGFP-tagged mutants were retained inside the cytosol, with a pattern of expression quite heterogeneous among the mutants. CONCLUSIONS: The three mutations studied of y+LAT1 transporter result in a defective arginine transport both in ex vivo (monocytes) and in vitro (CHO transfected cells) models, likely caused by the retention of the mutated proteins in the cytosol. The different effect of y+LAT1 mutation on arginine transport in monocytes and lymphoblasts is supposed to be due to the different expression of SLC7A7 mRNA in the two models, supporting the hypothesis that the impact of LPI defect largely depends on the relative abundance of LPI target gene in each cell type.


Subject(s)
Amino Acid Metabolism, Inborn Errors/genetics , Fusion Regulatory Protein 1, Light Chains/genetics , Fusion Regulatory Protein 1, Light Chains/metabolism , Mutation , Protein Transport/genetics , Adult , Amino Acid Transport System y+L , Animals , Arginine/metabolism , CHO Cells , Cells, Cultured , Child , Child, Preschool , Cricetulus , Cytosol/metabolism , Female , Humans , Male , Monocytes
10.
J Physiol ; 597(2): 521-542, 2019 01.
Article in English | MEDLINE | ID: mdl-30379325

ABSTRACT

KEY POINTS: Amino acid absorption requires luminal uptake into and subsequent basolateral efflux out of epithelial cells, with the latter step being critical to regulate the intracellular concentration of the amino acids. The basolateral essential neutral amino acid uniporter LAT4 (SLC43A2) has been suggested to drive the net efflux of non-essential and cationic amino acids via parallel amino acid antiporters by recycling some of their substrates; its deletion has been shown to cause defective postnatal growth and death in mice. Here we test the regulatory function of LAT4 phosphorylation sites by mimicking their phosphorylated and dephosphorylated states in Xenopus laevis oocytes and show that dephosphorylation of S274 and phosphorylation of S297 increase LAT4 membrane localization and function. Using new phosphorylation site-specific antibodies, we observe changes in LAT4 phosphorylation in mouse small intestine that correspond to its upregulation at the expected feeding time. These results strongly suggest that LAT4 phosphorylation participates in the regulation of transepithelial amino acid absorption. ABSTRACT: The essential amino acid uniporters LAT4 and TAT1 are located at the basolateral side of intestinal and kidney epithelial cells and their transport function has been suggested to control the transepithelial (re)absorption of neutral and possibly also cationic amino acids. Uniporter LAT4 selectively transports the branched chain amino acids leucine, isoleucine and valine, and additionally methionine and phenylalanine. Its deletion leads to a postnatal growth failure and early death in mice. Since LAT4 has been reported to be phosphorylated in vivo, we hypothesized that phosphorylation regulates its function. Using Xenopus laevis oocytes, we tested the impact of LAT4 phosphorylation at Ser274 and Ser297 by expressing mutant constructs mimicking phosphorylated and dephosphorylated states. We then investigated the in vivo regulation of LAT4 in mouse small intestine using new phosphorylation site-specific antibodies and a time-restricted diet. In Xenopus oocytes, mimicking non-phosphorylation of Ser274 led to an increase in affinity and apparent surface membrane localization of LAT4, stimulating its transport activity, while the same mutation of Ser297 decreased LAT4's apparent surface expression and transport rate. In wild-type mice, LAT4 phosphorylation on Ser274 was uniform at the beginning of the inactive phase (ZT0). In contrast, at the beginning of the active phase (ZT12), corresponding to the anticipated feeding time, Ser274 phosphorylation was decreased and restricted to relatively large patches of cells, while Ser297 phosphorylation was increased. We conclude that phosphorylation of small intestinal LAT4 is under food-entrained circadian control, leading presumably to an upregulation of LAT4 function at the anticipated feeding time.


Subject(s)
Amino Acid Transport System y+/physiology , Eating/physiology , Fusion Regulatory Protein 1, Light Chains/physiology , Intestine, Small/physiology , Amino Acid Transport System y+/genetics , Animals , Female , Fusion Regulatory Protein 1, Light Chains/genetics , Mice , Mice, Transgenic , Oocytes , Phosphorylation , Xenopus laevis
11.
Pharm Res ; 35(12): 246, 2018 Oct 29.
Article in English | MEDLINE | ID: mdl-30374619

ABSTRACT

PURPOSE: The anti-epileptic drug pregabalin crosses the blood-brain barrier (BBB) in spite of its low lipophilicity. This study was performed to determine whether L-type amino acid transporters (LAT1/SLC7A5 and LAT2/SLC7A8) contribute to the uptake of pregabalin. METHODS: Pregabalin uptake by LATs-transfected HEK293 cells or hCMEC/D3 cells, an in vitro human BBB model, was measured by LC-MS/MS analysis. Expression of LAT1 mRNA in hCMEC/D3 cells was determined by quantitative RT-PCR analysis. RESULTS: Overexpression of LAT1, but not LAT2, in HEK293 cells significantly increased the cellular uptake of pregabalin, and the LAT1-mediated uptake was saturable with a Km of 0.288 mM. LAT1-mediated amino acid uptake was inhibited specifically and almost completely in the presence of 1 mM pregabalin. The uptake of pregabalin by hCMEC/D3 cells was sodium-independent, saturable (Km = 0.854 mM), and strongly inhibited by large amino acids at 1 mM, 2-aminobicyclo-(2,2,1)-heptane-2-carboxylic acid, a specific system L inhibitor, at 1 mM and by JPH203, a LAT1-selective inhibitor, at 10 µM. Pregabalin uptake in hCMEC/D3 cells was also decreased by 75% by the silencing of LAT1 gene using LAT1 siRNA. CONCLUSIONS: Our results indicate that LAT1, but not LAT2, recognizes pregabalin as a substrate. It is suggested that LAT1 mediates pregabalin transport at the BBB.


Subject(s)
Anticonvulsants/pharmacokinetics , Blood-Brain Barrier/metabolism , Endothelial Cells/drug effects , Large Neutral Amino Acid-Transporter 1/metabolism , Pregabalin/pharmacokinetics , Amino Acid Transport System y+/genetics , Amino Acid Transport System y+/metabolism , Animals , Anticonvulsants/metabolism , Biological Transport , Brain/blood supply , Cell Line , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Fusion Regulatory Protein 1, Light Chains/genetics , Fusion Regulatory Protein 1, Light Chains/metabolism , Humans , Large Neutral Amino Acid-Transporter 1/genetics , Leucine/metabolism , Permeability , Pregabalin/metabolism , RNA, Small Interfering/genetics , Rats
12.
Cell Physiol Biochem ; 48(2): 731-740, 2018.
Article in English | MEDLINE | ID: mdl-30025393

ABSTRACT

BACKGROUND/AIMS: Y+LAT1 protein, encoded by the SLC7A7 gene (a member of the SLC7 family), forms the cationic amino acid transport system y+L (system y+L). This system transports cationic amino acids such as arginine and lysine out of the cell. Arginine, in particular, is critical for T-cell activation and function in the immune response. METHODS: We analyzed the role of the SLC7A7 gene in the cellular activities of Jurkat cells, specifically the cell cycle and cell proliferation, apoptosis, migration, and invasion. Cell proliferation was assessed using the Cell Counting Kit-8. Apoptosis and the cell cycle were determined with a FACSCalibur flow cytometer. A Transwell chamber was used to measure cell invasion and migration. RESULTS: The proliferative ability of Jurkat cells was not significantly altered by transfection with SLC7A7 overexpression vectors. However, SLC7A7 overexpression significantly decreased the percentage of apoptotic Jurkat cells (P = 0.007) but significantly increased the proportion of G1 phase cells (P = 0.029) and cell migration (P < 0.001) and invasion (P < 0.001). Knockdown of SLC7A7 increased the cell apoptosis rate (P = 0.006) but decreased the G1 phase ratio (P = 0.002) and cell migration (P < 0.001) and invasion (P < 0.001). CONCLUSIONS: SLC7A7 plays a significant role in the pathogenesis of T-cell acute lymphoblastic leukemia.


Subject(s)
Fusion Regulatory Protein 1, Light Chains/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Adolescent , Amino Acid Transport System y+L , Apoptosis , Arginine/analysis , Bone Marrow/metabolism , Bone Marrow/pathology , Cell Movement , Cell Proliferation , Child , Child, Preschool , Enzyme-Linked Immunosorbent Assay , Female , Fusion Regulatory Protein 1, Light Chains/antagonists & inhibitors , Fusion Regulatory Protein 1, Light Chains/genetics , G1 Phase Cell Cycle Checkpoints , Humans , Infant , Jurkat Cells , Male , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , RNA Interference , RNA, Small Interfering/metabolism , TOR Serine-Threonine Kinases/metabolism
13.
J Am Soc Nephrol ; 29(6): 1624-1635, 2018 06.
Article in English | MEDLINE | ID: mdl-29610403

ABSTRACT

Background Reabsorption of amino acids (AAs) across the renal proximal tubule is crucial for intracellular and whole organism AA homeostasis. Although the luminal transport step is well understood, with several diseases caused by dysregulation of this process, the basolateral transport step is not understood. In humans, only cationic aminoaciduria due to malfunction of the basolateral transporter y+LAT1/CD98hc (SLC7A7/SLC3A2), which mediates the export of cationic AAs, has been described. Thus, the physiologic roles of basolateral transporters of neutral AAs, such as the antiporter LAT2/CD98hc (SLC7A8/SLC3A2), a heterodimer that exports most neutral AAs, and the uniporter TAT1 (SLC16A10), which exports only aromatic AAs, remain unclear. Functional cooperation between TAT1 and LAT2/CD98hc has been suggested by in vitro studies but has not been evaluated in vivoMethods To study the functional relationship of TAT1 and LAT2/CD98hc in vivo, we generated a double-knockout mouse model lacking TAT1 and LAT2, the catalytic subunit of LAT2/CD98hc (dKO LAT2-TAT1 mice).Results Compared with mice lacking only TAT1 or LAT2, dKO LAT2-TAT1 mice lost larger amounts of aromatic and other neutral AAs in their urine due to a tubular reabsorption defect. Notably, dKO mice also displayed decreased tubular reabsorption of cationic AAs and increased expression of y+LAT1/CD98hc.Conclusions The LAT2/CD98hc and TAT1 transporters functionally cooperate in vivo, and y+LAT1/CD98hc may compensate for the loss of LAT2/CD98hc and TAT1, functioning as a neutral AA exporter at the expense of some urinary loss of cationic AAs. Cooperative and compensatory mechanisms of AA transporters may explain the lack of basolateral neutral aminoacidurias in humans.


Subject(s)
Amino Acid Transport System y+/genetics , Amino Acid Transport Systems, Neutral/genetics , Amino Acids, Neutral/metabolism , Fusion Regulatory Protein 1, Light Chains/genetics , Renal Reabsorption , Amino Acid Transport System y+/metabolism , Amino Acid Transport Systems, Neutral/metabolism , Amino Acids, Neutral/urine , Animals , Female , Fusion Regulatory Protein 1, Heavy Chain/metabolism , Fusion Regulatory Protein 1, Light Chains/metabolism , Kidney Tubules/physiology , Male , Mice, Knockout
14.
Front Immunol ; 9: 508, 2018.
Article in English | MEDLINE | ID: mdl-29616026

ABSTRACT

Lysinuric protein intolerance (LPI) is a recessively inherited aminoaciduria caused by mutations of SLC7A7, the gene encoding y+LAT1 light chain of system y+L for cationic amino acid transport. The pathogenesis of LPI is still unknown. In this study, we have utilized a gene silencing approach in macrophages and airway epithelial cells to investigate whether complications affecting lung and immune system are directly ascribable to the lack of SLC7A7 or, rather, mediated by an abnormal accumulation of arginine in mutated cells. When SLC7A7/y+LAT1 was silenced in human THP-1 macrophages and A549 airway epithelial cells by means of short interference RNA (siRNA), a significant induction of the expression and release of the inflammatory mediators IL1ß and TNFα was observed, no matter the intracellular arginine availability. This effect was mainly regulated at transcriptional level through the activation of NFκB signaling pathway. Moreover, since respiratory epithelial cells are the important sources of chemokines in response to pro-inflammatory stimuli, the effect of IL1ß has been addressed on SLC7A7 silenced A549 cells. Results obtained indicated that the downregulation of SLC7A7/y+LAT1 markedly strengthened the stimulatory effect of the cytokine on CCL5/RANTES expression and release without affecting the levels of CXCL8/IL8. Consistently, also the conditioned medium of silenced THP-1 macrophages activated airway epithelial cells in terms of CCL5/RANTES expression due to the presence of elevated amount of proinflammatory cytokines. In conclusion, our results point to a novel thus far unknown function of SLC7A7/y+LAT1, that, under physiological conditions, besides transporting arginine, may act as a brake to restrain inflammation.


Subject(s)
Amino Acid Metabolism, Inborn Errors/immunology , Fusion Regulatory Protein 1, Light Chains/metabolism , Inflammation/immunology , Macrophages/immunology , Renal Aminoacidurias/immunology , Respiratory Mucosa/immunology , A549 Cells , Amino Acid Metabolism, Inborn Errors/genetics , Amino Acid Transport System y+L , Chemokine CCL5/metabolism , Fusion Regulatory Protein 1, Light Chains/genetics , Gene Silencing , Humans , Inflammation/genetics , Interleukin-1beta/metabolism , Mutation/genetics , NF-kappa B/metabolism , Phenotype , RNA, Small Interfering/genetics , Renal Aminoacidurias/genetics , THP-1 Cells , Tumor Necrosis Factor-alpha/metabolism
15.
BMC Genomics ; 19(1): 173, 2018 03 02.
Article in English | MEDLINE | ID: mdl-29499643

ABSTRACT

BACKGROUND: Gestational disorders such as intrauterine growth restriction (IUGR) and pre-eclampsia (PE) are main causes of poor perinatal outcomes worldwide. Both diseases are related with impaired materno-fetal nutrient transfer, but the crucial transport mechanisms underlying IUGR and PE are not fully elucidated. In this study, we aimed to identify membrane transporters highly associated with transplacental nutrient deficiencies in IUGR/PE. RESULTS: In silico analyses on the identification of differentially expressed nutrient transporters were conducted using seven eligible microarray datasets (from Gene Expression Omnibus), encompassing control and IUGR/PE placental samples. Thereby 46 out of 434 genes were identified as potentially interesting targets. They are involved in the fetal provision with amino acids, carbohydrates, lipids, vitamins and microelements. Targets of interest were clustered into a substrate-specific interaction network by using Search Tool for the Retrieval of Interacting Genes. The subsequent wet-lab validation was performed using quantitative RT-PCR on placentas from clinically well-characterized IUGR/PE patients (IUGR, n = 8; PE, n = 5; PE+IUGR, n = 10) and controls (term, n = 13; preterm, n = 7), followed by 2D-hierarchical heatmap generation. Statistical evaluation using Kruskal-Wallis tests was then applied to detect significantly different expression patterns, while scatter plot analysis indicated which transporters were predominantly influenced by IUGR or PE, or equally affected by both diseases. Identified by both methods, three overlapping targets, SLC7A7, SLC38A5 (amino acid transporters), and ABCA1 (cholesterol transporter), were further investigated at the protein level by western blotting. Protein analyses in total placental tissue lysates and membrane fractions isolated from disease and control placentas indicated an altered functional activity of those three nutrient transporters in IUGR/PE. CONCLUSIONS: Combining bioinformatic analysis, molecular biological experiments and mathematical diagramming, this study has demonstrated systematic alterations of nutrient transporter expressions in IUGR/PE. Among 46 initially targeted transporters, three significantly regulated genes were further investigated based on the severity and the disease specificity for IUGR and PE. Confirmed by mRNA and protein expression, the amino acid transporters SLC7A7 and SLC38A5 showed marked differences between controls and IUGR/PE and were regulated by both diseases. In contrast, ABCA1 may play an exclusive role in the development of PE.


Subject(s)
ATP Binding Cassette Transporter 1/metabolism , Amino Acid Transport Systems, Neutral/metabolism , Fetal Growth Retardation/pathology , Fusion Regulatory Protein 1, Light Chains/metabolism , Placenta/pathology , Pre-Eclampsia/pathology , ATP Binding Cassette Transporter 1/genetics , Adult , Amino Acid Transport System y+L , Amino Acid Transport Systems, Neutral/genetics , Case-Control Studies , Computational Biology/methods , Female , Fetal Growth Retardation/genetics , Fetal Growth Retardation/metabolism , Fusion Regulatory Protein 1, Light Chains/genetics , Gene Expression Regulation, Developmental , Humans , Infant, Newborn , Placenta/metabolism , Pre-Eclampsia/genetics , Pre-Eclampsia/metabolism , Pregnancy , Young Adult
16.
Elife ; 72018 01 22.
Article in English | MEDLINE | ID: mdl-29355479

ABSTRACT

Age-related hearing loss (ARHL) is the most common sensory deficit in the elderly. The disease has a multifactorial etiology with both environmental and genetic factors involved being largely unknown. SLC7A8/SLC3A2 heterodimer is a neutral amino acid exchanger. Here, we demonstrated that SLC7A8 is expressed in the mouse inner ear and that its ablation resulted in ARHL, due to the damage of different cochlear structures. These findings make SLC7A8 transporter a strong candidate for ARHL in humans. Thus, a screening of a cohort of ARHL patients and controls was carried out revealing several variants in SLC7A8, whose role was further investigated by in vitro functional studies. Significant decreases in SLC7A8 transport activity was detected for patient's variants (p.Val302Ile, p.Arg418His, p.Thr402Met and p.Val460Glu) further supporting a causative role for SLC7A8 in ARHL. Moreover, our preliminary data suggest that a relevant proportion of ARHL cases could be explained by SLC7A8 mutations.


Subject(s)
Mutation , Presbycusis/genetics , Presbycusis/pathology , Amino Acid Transport System y+/deficiency , Amino Acid Transport System y+/genetics , Animals , Fusion Regulatory Protein 1, Light Chains/deficiency , Fusion Regulatory Protein 1, Light Chains/genetics , Gene Deletion , Genetic Testing , Humans , Mice
17.
Pediatr Pulmonol ; 52(11): E94-E96, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29058386

ABSTRACT

Lysinuric protein intolerance (LPI) is an inherited aminoaciduria with an autosomal recessive mode of inheritance.The first two cases of sisters being diagnosed with LPI in China is contained within this report. In our cases, there were two heterozygous mutations in the SLC7A7 gene of the two sisters: deletion of c.1387: del C and IVS4+1C>T. One patient was treated with inhaled rGM-CSF for 1.5 years at 5 µg/kg two times a day. Her condition is improving with no side effects.


Subject(s)
Amino Acid Metabolism, Inborn Errors/genetics , Fusion Regulatory Protein 1, Light Chains/genetics , Amino Acid Metabolism, Inborn Errors/drug therapy , Amino Acid Transport System y+L , Asian People/genetics , Child , Female , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Heterozygote , Humans , Mutation , Sequence Deletion , Siblings
18.
Mol Med Rep ; 16(1): 887-893, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28560461

ABSTRACT

The kidney serves a central role in the control of blood pressure through the release of vasoactive substances and the urinary excretion of Na+. Patients with essential hypertension usually exhibit persistent high blood pressure accompanied by Na+ retention. L-dihydroxyphenylalanine (L­DOPA) is an amino acid, converted by the enzyme aromatic L­amino acid decarboxylase to dopamine. The uptake of L­DOPA by cells of the proximal tubular epithelium of the kidney is controlled by the L­type amino acid transporter 2 (LAT2). LAT2 belongs to the solute carrier family 7 (SLC7) of amino acid transporters and is coded by the SLC7A8 gene. SLC7A8 expression is increased in the second­order mesenteric arteries and kidneys of spontaneously hypertensive rats. The present study aimed to investigate the physiological role of the SLC7A8 gene in L­DOPA handling by kidney cells. Selective upregulation of SLC7A8 mRNA and protein levels was achieved by adenoviral transduction of NRK­52E cells, which retain several properties of proximal tubular epithelial cells. In addition, L­DOPA uptake was determined using high performance liquid chromatography; NRK­52E cells expressing SLC7A8 exhibited increased uptake of L­DOPA. The results of the present study suggested that SLC7A8 may serve a critical role in blood pressure control through regulating L­DOPA uptake in renal epithelial cells of the proximal tubule.


Subject(s)
Amino Acid Transport System y+/genetics , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Fusion Regulatory Protein 1, Light Chains/genetics , Gene Expression Regulation , Kidney Tubules/cytology , Levodopa/metabolism , Amino Acid Transport System y+/metabolism , Animals , Biological Transport , Blood Pressure/drug effects , Blood Pressure/genetics , Cell Line , Fusion Regulatory Protein 1, Light Chains/metabolism , Gene Expression , Gene Expression Profiling , Levodopa/pharmacology , Male , Rats , Rats, Inbred SHR , Transduction, Genetic
19.
Cell Microbiol ; 19(11)2017 11.
Article in English | MEDLINE | ID: mdl-28655069

ABSTRACT

Cryptosporidial infection causes dysregulated transcription of host genes key to intestinal epithelial homeostasis, but the underlying mechanisms remain obscure. Previous studies demonstrate that several Cryptosporidium parvum (C. parvum) RNA transcripts are selectively delivered into epithelial cells during host cell invasion and may modulate gene transcription in infected cells. We report here that C. parvum infection suppresses the transcription of LRP5, SLC7A8, and IL33 genes in infected intestinal epithelium. Trans-suppression of these genes in infected host cells is associated with promoter enrichment of suppressive epigenetic markers (i.e., H3K9me3). Cdg7_FLc_0990, a C. parvum RNA that has previously demonstrated to be delivered into the nuclei of infected epithelial cells, is recruited to the promoter regions of LRP5, SLC7A8, and IL33 genes. Cdg7_FLc_0990 appears to be recruited to their promoter regions together with G9a, a histone methyltransferase for H3K9 methylation. The PR domain zinc finger protein 1, a G9a-interacting protein, is required for the assembly of Cdg7_FLc_0990 to the G9a complex and gene-specific enrichment of H3K9 methylation. Our data demonstrate that cryptosporidial infection induces epigenetic histone methylations in infected cells through nuclear transfer of parasite Cdg7_Flc_0990 RNA transcript, resulting in transcriptional suppression of the LRP5, SLC7A8, and IL33 genes.


Subject(s)
Amino Acid Transport System y+/biosynthesis , Cryptosporidium parvum/genetics , Fusion Regulatory Protein 1, Light Chains/biosynthesis , Interleukin-33/biosynthesis , Intestinal Mucosa/parasitology , Low Density Lipoprotein Receptor-Related Protein-5/biosynthesis , Transcription, Genetic/genetics , Amino Acid Transport System y+/genetics , Animals , Cell Line , Cryptosporidiosis/parasitology , Cryptosporidiosis/pathology , Cryptosporidium parvum/pathogenicity , Epigenesis, Genetic , Epithelial Cells/parasitology , Fusion Regulatory Protein 1, Light Chains/genetics , HSP72 Heat-Shock Proteins/genetics , Histocompatibility Antigens/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Humans , Interleukin-33/genetics , Low Density Lipoprotein Receptor-Related Protein-5/genetics , Methylation , Mice , Positive Regulatory Domain I-Binding Factor 1/genetics , Promoter Regions, Genetic/genetics , RNA Interference , RNA, Protozoan/genetics , RNA, Small Interfering/genetics
20.
Genet Mol Res ; 16(2)2017 May 10.
Article in English | MEDLINE | ID: mdl-28510245

ABSTRACT

Gliomas are brain tumors that can be seriously damaging to human health. The SLC7 family is involved in amino acid or peptide transportation. The relationship between SLC7A7 polymorphisms and the development of glioma has been reported previously by a few studies. Therefore, we performed a hospital based case-control study to investigate the association of three common SNPs (rs12888930, rs12436190, and rs2065134) of SLC7A7 with the development of glioma in a Chinese population. From January 2014 to December 2015, 122 patients with glioma and 252 individuals (controls) were recruited from the department of neurosurgery of Tangshan People's Hospital affiliated to North China University of Science and Technology. SLC7A7 rs12888930, rs12436190, and rs2065134 genotyping was performed by the polymerase chain reaction-restriction fragment length polymorphism method. Multiple logistic regression analysis showed that a significantly higher risk of glioma was harbored by the GG and AG + GG genotypes than by the AA genotype; OR (95%CI) was 2.24 (1.18-4.22) and 1.59 (1.01-2.60), respectively. However, no significant relationship was observed between SLC7A7 rs12888930 and rs2065134 and the risk of glioma. In conclusion, this study reports a significant association between SLC7A7 rs12436190 and the risk of glioma in a Chinese population.


Subject(s)
Brain Neoplasms/genetics , Fusion Regulatory Protein 1, Light Chains/genetics , Glioma/genetics , Polymorphism, Single Nucleotide , Amino Acid Transport System y+L , Case-Control Studies , China , Female , Humans , Male , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL
...