Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 92
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.
J Cell Mol Med ; 24(21): 12681-12693, 2020 11.
Article in English | MEDLINE | ID: mdl-33001560

ABSTRACT

The placenta supplies the foetus with critical nutrients such as essential amino acids (AA, eg leucine) for development and growth. It also represents a cellular barrier which is formed by a polarized, differentiated syncytiotrophoblast (STB) monolayer. Active Na+ -independent leucine transport across the placenta is mainly attributed to the System L transporters LAT1/SLC7A5 and LAT2/SLC7A8. This study explored the influence of trophoblast differentiation on the activity of LAT1/LAT2 and the relevance of LAT1/LAT2 in leucine uptake and transfer in trophoblasts by applying specific small molecule inhibitors (JPH203/JG336/JX009). L-leucine uptake (total dose = 167 µmol/L) was sensitive to LAT1-specific inhibition by JPH203 (EC50  = 2.55 µmol/L). The inhibition efficiency of JPH203 was increased by an additional methoxy group in the JPH203-derivate JG336 (EC50  = 1.99 µmol/L). Interestingly, JX009 showed efficient System L inhibition (EC50  = 2.35 µmol/L) and was the most potent inhibitor of leucine uptake in trophoblasts. The application of JPH203 and JX009 in Transwell® -based leucine transfer revealed LAT1 as the major accumulative transporter at the apical membrane, but other System L transporters such as LAT2 as rate-limiting for leucine efflux across the basal membrane. Therefore, differential specificity of the applied inhibitors allowed for estimation of the contribution of LAT1 and LAT2 in materno-foetal AA transfer and their potential impact in pregnancy diseases associated with impaired foetal growth.


Subject(s)
Amino Acid Transport System y+/metabolism , Fusion Regulatory Protein 1, Light Chains/metabolism , Large Neutral Amino Acid-Transporter 1/metabolism , Leucine/metabolism , Maternal-Fetal Exchange , Adult , Biological Transport/drug effects , Cell Differentiation/drug effects , Cell Line , Female , Fusion Regulatory Protein 1, Heavy Chain/metabolism , Humans , Infant, Newborn , Maternal-Fetal Exchange/drug effects , Placenta/metabolism , Pregnancy , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Sodium/metabolism , Trophoblasts/cytology , Trophoblasts/drug effects , Trophoblasts/metabolism , Up-Regulation/drug effects
6.
Breast Cancer Res Treat ; 181(1): 1-12, 2020 May.
Article in English | MEDLINE | ID: mdl-32200487

ABSTRACT

PURPOSE: Breast cancer (BC) is a heterogeneous disease consisting of various subtypes, with different prognostic and therapeutic outcomes. The amino acid transporter, SLC7A8, is overexpressed in oestrogen receptor-positive BC. However, the consequence of this overexpression, in terms of disease prognosis, is still obscure. This study aimed to evaluate the biological and prognostic value of SLC7A8 in BC with emphasis on the intrinsic molecular subtypes. METHODS: SLC7A8 was assessed at the genomic, using METABRIC data (n = 1980), and proteomic, using immunohistochemistry and TMA (n = 1562), levels in well-characterised primary BC cohorts. SLC7A8 expression was examined with clinicopathological parameters, molecular subtypes, and patient outcome. RESULTS: SLC7A8 mRNA and SLC7A8 protein expression were strongly associated with good prognostic features, including small tumour size, low tumour grade, and good Nottingham Prognostic Index (NPI) (all P < 0.05). Expression of SLC7A8 mRNA was higher in luminal tumours compared to other subtypes (P < 0.001). High expression of SLC7A8 mRNA and SLC7A8 protein was associated with good patient outcome (P ≤ 0.001) but only in the low proliferative ER+/luminal A tumours (P = 0.01). In multivariate analysis, SLC7A8 mRNA and SLC7A8 protein were independent factors for longer breast cancer specific survival (P = 0.01 and P = 0.03), respectively. CONCLUSION: SLC7A8 appears to play a role in BC and is a marker for favourable prognosis in the most predominant, ER+ low proliferative/luminal A, BC subtype. Functional assessment is necessary to reveal the specific role played by SLC7A8 in ER+ BC.


Subject(s)
Amino Acid Transport System y+/metabolism , Breast Neoplasms/metabolism , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Lobular/metabolism , Fusion Regulatory Protein 1, Light Chains/metabolism , Receptors, Estrogen/metabolism , Aged , Biomarkers, Tumor/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/surgery , Carcinoma, Ductal, Breast/pathology , Carcinoma, Ductal, Breast/surgery , Carcinoma, Lobular/pathology , Carcinoma, Lobular/surgery , Cell Proliferation/physiology , Female , Follow-Up Studies , Humans , Neoplasm Invasiveness , Prognosis , Receptor, ErbB-2/metabolism , Receptors, Progesterone/metabolism
7.
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
8.
Nutrients ; 11(11)2019 Nov 18.
Article in English | MEDLINE | ID: mdl-31752111

ABSTRACT

The intestine not only plays a role in fundamental processes in digestion and nutrient absorption, but it also has a role in eliminating ingested pathogenic bacteria and viruses. Paneth cells, which reside at the base of small intestinal crypts, secrete α-defensins and contribute to enteric innate immunity through potent microbicidal activities. However, the relationship between food factors and the innate immune functions of Paneth cells remains unknown. Here, we examined whether short-chain fatty acids and amino acids induce α-defensin secretion from Paneth cells in the isolated crypts of small intestine. Butyric acid and leucine elicit α-defensin secretion by Paneth cells, which kills Salmonella typhimurium. We further measured Paneth cell secretion in response to butyric acid and leucine using enteroids, a three-dimensional ex vivo culture system of small intestinal epithelial cells. Paneth cells expressed short-chain fatty acid receptors, Gpr41, Gpr43, and Gpr109a mRNAs for butyric acid, and amino acid transporter Slc7a8 mRNA for leucine. Antagonists of Gpr41 and Slc7a8 inhibited granule secretion by Paneth cells, indicating that these receptor and transporter on Paneth cells induce granule secretion. Our findings suggest that Paneth cells may contribute to intestinal homeostasis by secreting α-defensins in response to certain nutrients or metabolites.


Subject(s)
Butyric Acid/immunology , Intestine, Small/metabolism , Leucine/immunology , Paneth Cells/metabolism , alpha-Defensins/metabolism , Amino Acid Transport System y+/antagonists & inhibitors , Amino Acid Transport System y+/metabolism , Animals , Fusion Regulatory Protein 1, Light Chains/antagonists & inhibitors , Fusion Regulatory Protein 1, Light Chains/metabolism , Gene Expression , Homeostasis , Immunity, Innate , Mice , Mice, Inbred ICR , Microbiota , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/metabolism
9.
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
10.
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
11.
Br J Dermatol ; 180(1): 130-140, 2019 01.
Article in English | MEDLINE | ID: mdl-29938775

ABSTRACT

BACKGROUND: The incidence of basal cell carcinoma (BCC) is increasing and the costs for care rising. Therefore, the need for simplified and cost-effective treatment choices is substantial. Aberrant signalling in several pathways, induced by ultraviolet radiation, is of importance in the development of BCC. Alterations in tumour metabolic activity are part of general carcinogenesis; however, these alterations are only partially recognized in skin cancer. OBJECTIVES: To study expression profiles in BCCs compared with individually matched nontumour skin, with a focus on finding differences associated with tumour metabolism. MATERIALS AND METHODS: Gene expression in biopsies from BCCs (n = 14) compared with biopsies from nontumour gluteal skin was analysed with microarrays (n = 4 + 4) and/or quantitative real-time polymerase chain reaction (qPCR, n = 14 + 14). Protein expression and localization was assessed using immunohistochemistry (IHC) in formalin-fixed and paraffin-embedded BCC samples. RESULTS: Microarray analysis revealed increased expression of the amino acid transporters SLC7A5, SLC7A7 and SLC7A8 as well as the cytosolic enzyme tryptophan 2,3-dioxygenase (TDO) 2 in BCC. Higher expression of SLC7A5 (P < 0·001), SLC7A8 (P < 0·001) and TDO2 (P = 0·002), but not SLC7A7 (P = 0·50), was confirmed by qPCR, and IHC demonstrated correlating tumour cell protein expression of SLC7A5 and SLC7A8. Protein expression of SLC7A7 was observed in the stratum granulosum, and TDO2 in immune cells. CONCLUSIONS: This study highlights the upregulation of SLC7A5, SLC7A8 and TDO2 in BCC compared with nontumour skin. Our findings imply that amino acid transporters may be further explored as potential targets for future medical treatment.


Subject(s)
Carcinoma, Basal Cell/pathology , Skin Neoplasms/pathology , Aged , Aged, 80 and over , Amino Acid Transport System y+/metabolism , Amino Acid Transport System y+L , Cohort Studies , Female , Fusion Regulatory Protein 1, Light Chains/metabolism , Humans , Large Neutral Amino Acid-Transporter 1/metabolism , Male , Middle Aged , Skin/pathology , Tryptophan Oxygenase/metabolism , Up-Regulation
12.
J Exp Clin Cancer Res ; 37(1): 274, 2018 Nov 12.
Article in English | MEDLINE | ID: mdl-30419950

ABSTRACT

BACKGROUND: Reprogrammed energy metabolism has become an emerging hallmark of cancer in recent years. Transporters have been reported to be amino acid sensors involved in controlling mTOR recruitment and activation, which is crucial for the growth of both normal and tumor cells. L-type amino acid transporter 2 (LAT2), encoded by the SLC7A8 gene, is a Na+-independent neutral amino acid transporter and is responsible for transporting neutral amino acids, including glutamine, which can activate mTOR. Previous studies have shown that LAT2 was overexpressed in gemcitabine-resistant pancreatic cancer cells. However, the role of LAT2 in chemoresistance in pancreatic cancer remains uncertain and elusive. METHODS: The effects of LAT2 on biological behaviors were analyzed. LAT2 and LDHB levels in tissues were detected, and the clinical value was evaluated. RESULTS: We demonstrated that LAT2 emerged as an oncogenic protein and could decrease the gemcitabine sensitivity of pancreatic cancer cells in vitro and in vivo. The results of a survival analysis indicated that high expression levels of both LAT2 and LDHB predicted a poor prognosis in patients with pancreatic cancer. Furthermore, we found that LAT2 could promote proliferation, inhibit apoptosis, activate glycolysis and alter glutamine metabolism to activate mTOR in vitro and in vivo. Next, we found that gemcitabine combined with an mTOR inhibitor (RAD001) could reverse the decrease in chemosensitivity caused by LAT2 overexpression in pancreatic cancer cells. Mechanistically, we demonstrated that LAT2 could regulate two glutamine-dependent positive feedback loops (the LAT2/p-mTORSer2448 loop and the glutamine/p-mTORSer2448/glutamine synthetase loop) to promote glycolysis and decrease gemcitabine (GEM) sensitivity in pancreatic cancer. CONCLUSION: Taken together, our data reveal that LAT2 functions as an oncogenic protein and could regulate glutamine-dependent mTOR activation to promote glycolysis and decrease GEM sensitivity in pancreatic cancer. The LAT2-mTOR-LDHB pathway might be a promising therapeutic target in pancreatic cancer.


Subject(s)
Amino Acid Transport System y+/metabolism , Fusion Regulatory Protein 1, Light Chains/metabolism , Glutamine/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Apoptosis/drug effects , Apoptosis/physiology , Cell Line, Tumor , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Drug Resistance, Neoplasm , Female , Glycolysis , HEK293 Cells , Heterografts , Humans , Mice , Mice, Inbred BALB C , Transfection , Gemcitabine
13.
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
14.
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
15.
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
16.
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
17.
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
18.
Nutrients ; 10(1)2017 Dec 26.
Article in English | MEDLINE | ID: mdl-29278358

ABSTRACT

The branch chain amino acid leucine is a potent stimulator of protein synthesis in skeletal muscle. Leucine rapidly enters the cell via the L-Type Amino Acid Transporter 1 (LAT1); however, little is known regarding the localisation and distribution of this transporter in human skeletal muscle. Therefore, we applied immunofluorescence staining approaches to visualise LAT1 in wild type (WT) and LAT1 muscle-specific knockout (mKO) mice, in addition to basal human skeletal muscle samples. LAT1 positive staining was visually greater in WT muscles compared to mKO muscle. In human skeletal muscle, positive LAT1 staining was noted close to the sarcolemmal membrane (dystrophin positive staining), with a greater staining intensity for LAT1 observed in the sarcoplasmic regions of type II fibres (those not stained positively for myosin heavy-chain 1, Type II-25.07 ± 5.93, Type I-13.71 ± 1.98, p < 0.01), suggesting a greater abundance of this protein in these fibres. Finally, we observed association with LAT1 and endothelial nitric oxide synthase (eNOS), suggesting LAT1 association close to the microvasculature. This is the first study to visualise the distribution and localisation of LAT1 in human skeletal muscle. As such, this approach provides a validated experimental platform to study the role and regulation of LAT1 in human skeletal muscle in response to various physiological and pathophysiological models.


Subject(s)
Fluorescent Antibody Technique , Fusion Regulatory Protein 1, Light Chains/metabolism , Microscopy, Fluorescence , Muscle Fibers, Fast-Twitch/metabolism , Muscle Fibers, Slow-Twitch/metabolism , Quadriceps Muscle/metabolism , Amino Acid Transport System y+/genetics , Amino Acid Transport System y+/metabolism , Amino Acid Transport System y+L , Animals , Humans , Male , Mice, Knockout , Nitric Oxide Synthase Type III/metabolism , Quadriceps Muscle/cytology , Sarcolemma/metabolism , Young Adult
19.
Int J Mol Sci ; 18(11)2017 Nov 02.
Article in English | MEDLINE | ID: mdl-29099056

ABSTRACT

Previously we had shown that ammonia stimulates nitric oxide (NO) synthesis in astrocytes by increasing the uptake of the precursor amino acid, arginine via the heteromeric arginine/glutamine transporter y⁺LAT2. Ammonia also increases the concentration in the brain of the endogenous inhibitor of nitric oxide synthases (NOS), asymmetric dimethylarginine (ADMA), but distribution of ADMA surplus between the intraastrocytic and extracellular compartments of the brain has not been studied. Here we tested the hypothesis that ammonia modulates the distribution of ADMA and its analog symmetric dimethylarginine (SDMA) between the two compartments of the brain by competition with arginine for the y⁺LAT2 transporter. In extension of the hypothesis we analyzed the ADMA/Arg interaction in endothelial cells forming the blood-brain barrier. We measured by high-performance liquid chromatography (HPLC) and mass spectrometry (MS) technique the concentration of arginine, ADMA and SDMA in cultured cortical astrocytes and in a rat brain endothelial cell line (RBE-4) treated with ammonia and the effect of silencing the expression of a gene coding y⁺LAT2. We also tested the expression of ADMA metabolism enzymes: protein arginine methyltransferase (PRMT) and dimethylarginine dimethyl aminohydrolase (DDAH) and arginine uptake to astrocytes. Treatment for 48 h with 5 mM ammonia led to an almost 50% reduction of ADMA and SDMA concentration in both cell types, and the effect in astrocytes was substantially attenuated by silencing of the Slc7a6 gene. Moreover, the y⁺LAT2-dependent component of ammonia-evoked arginine uptake in astrocytes was reduced in the presence of ADMA in the medium. Our results suggest that increased ADMA efflux mediated by upregulated y⁺LAT2 may be a mechanism by which ammonia interferes with intra-astrocytic (and possibly intra-endothelial cell) ADMA content and subsequently, NO synthesis in both cell types.


Subject(s)
Amino Acid Transport System y+/metabolism , Ammonia/metabolism , Arginine/analogs & derivatives , Astrocytes/metabolism , Fusion Regulatory Protein 1, Light Chains/metabolism , Amidohydrolases/metabolism , Animals , Arginine/metabolism , Cell Line , Cells, Cultured , Nitric Oxide/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Rats , Rats, Wistar
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...