51
|
Chen Q, Wang C, Zhao FQ, Liu J, Liu H. Effects of methionine partially replaced by methionyl-methionine dipeptide on intestinal function in methionine-deficient pregnant mice. J Anim Physiol Anim Nutr (Berl) 2019; 103:1610-1618. [PMID: 31106911 DOI: 10.1111/jpn.13126] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 04/24/2019] [Accepted: 05/03/2019] [Indexed: 12/17/2022]
Abstract
This study was to compare the effects of parenteral supplementation of methionyl-methionine (Met-Met) or Met on intestinal barrier function in Met-deficient pregnant mice. Pregnant mice were randomly divided into three groups. The Control group was provided a diet containing Met and received i.p. injection of saline. The Met group was fed the same diet but without Met and received daily i.p. injection of 35% of the Met contained in the control diet. The Met-Met group was treated the same as the Met group, except that 25% of the Met injected was replaced with Met-Met. Met-Met promoted villus surface area in ileum compared with Met alone. In addition, the mRNA abundance of amino acid and glucose transporters in the small intestine was altered with Met-Met. Moreover, Met-Met increased tight junction protein and decreased apoptosis-related proteins expression in the jejunum and ileum. These results suggest that Met-Met can promote intestinal function over Met alone in Met-deficient mice.
Collapse
Affiliation(s)
- Qiong Chen
- Institute of Dairy Science, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Caihong Wang
- Institute of Dairy Science, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Feng-Qi Zhao
- Institute of Dairy Science, College of Animal Sciences, Zhejiang University, Hangzhou, China.,Department of Animal and Veterinary Sciences, University of Vermont, Burlington, Vermont
| | - Jianxin Liu
- Institute of Dairy Science, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Hongyun Liu
- Institute of Dairy Science, College of Animal Sciences, Zhejiang University, Hangzhou, China
| |
Collapse
|
52
|
The L-Type Amino Acid Transporter LAT1-An Emerging Target in Cancer. Int J Mol Sci 2019; 20:ijms20102428. [PMID: 31100853 PMCID: PMC6566973 DOI: 10.3390/ijms20102428] [Citation(s) in RCA: 137] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 05/10/2019] [Accepted: 05/14/2019] [Indexed: 12/11/2022] Open
Abstract
Chronic proliferation is a major hallmark of tumor cells. Rapidly proliferating cancer cells are highly dependent on nutrients in order to duplicate their cell mass during each cell division. In particular, essential amino acids are indispensable for proliferating cancer cells. Their uptake across the cell membrane is tightly controlled by membrane transporters. Among those, the L-type amino acid transporter LAT1 (SLC7A5) has been repeatedly found overexpressed in a vast variety of cancers. In this review, we summarize the most recent advances in our understanding of the role of LAT1 in cancer and highlight preclinical studies and drug developments underlying the potential of LAT1 as therapeutic target.
Collapse
|
53
|
Abstract
The small intestine mediates the absorption of amino acids after ingestion of protein and sustains the supply of amino acids to all tissues. The small intestine is an important contributor to plasma amino acid homeostasis, while amino acid transport in the large intestine is more relevant for bacterial metabolites and fluid secretion. A number of rare inherited disorders have contributed to the identification of amino acid transporters in epithelial cells of the small intestine, in particular cystinuria, lysinuric protein intolerance, Hartnup disorder, iminoglycinuria, and dicarboxylic aminoaciduria. These are most readily detected by analysis of urine amino acids, but typically also affect intestinal transport. The genes underlying these disorders have all been identified. The remaining transporters were identified through molecular cloning techniques to the extent that a comprehensive portrait of functional cooperation among transporters of intestinal epithelial cells is now available for both the basolateral and apical membranes. Mouse models of most intestinal transporters illustrate their contribution to amino acid homeostasis and systemic physiology. Intestinal amino acid transport activities can vary between species, but these can now be explained as differences of amino acid transporter distribution along the intestine. © 2019 American Physiological Society. Compr Physiol 9:343-373, 2019.
Collapse
Affiliation(s)
- Stefan Bröer
- Research School of Biology, The Australian National University, Canberra, ACT 2601, Australia
| | - Stephen J Fairweather
- Research School of Biology, The Australian National University, Canberra, ACT 2601, Australia
| |
Collapse
|
54
|
Oparija L, Rajendran A, Poncet N, Verrey F. Anticipation of food intake induces phosphorylation switch to regulate basolateral amino acid transporter LAT4 (SLC43A2) function. J Physiol 2018; 597:521-542. [PMID: 30379325 DOI: 10.1113/jp276714] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Accepted: 10/29/2018] [Indexed: 12/30/2022] Open
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.
Collapse
Affiliation(s)
- Lalita Oparija
- Institute of Physiology and Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| | - Anuradha Rajendran
- Institute of Physiology and Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| | - Nadège Poncet
- Institute of Physiology and Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| | - François Verrey
- Institute of Physiology and Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland.,NCCR Kidney.CH, University of Zurich, Zurich, Switzerland
| |
Collapse
|
55
|
Transport of Pregabalin Via L-Type Amino Acid Transporter 1 (SLC7A5) in Human Brain Capillary Endothelial Cell Line. Pharm Res 2018; 35:246. [PMID: 30374619 PMCID: PMC6208607 DOI: 10.1007/s11095-018-2532-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Accepted: 10/21/2018] [Indexed: 12/21/2022]
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. Electronic supplementary material The online version of this article (10.1007/s11095-018-2532-0) contains supplementary material, which is available to authorized users.
Collapse
|
56
|
Iresjö BM, Engström C, Smedh U, Lundholm K. Overnight Steady-State Infusions of Parenteral Nutrition on Myosin Heavy Chain Transcripts in Rectus Abdominis Muscle Related to Amino Acid Transporters, Insulin-like Growth Factor 1, and Blood Amino Acids in Patients Aimed at Major Surgery. JPEN J Parenter Enteral Nutr 2018; 43:497-507. [PMID: 30350380 DOI: 10.1002/jpen.1458] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 09/12/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND Evaluation of improvements by nutrition support to severely ill patients requires sensitive methods to demonstrate activation of protein synthesis in various tissues from groups with a limited number of patients to be statistically efficient. This study examines effects of standard parenteral nutrition (PN) on abdominal muscle transcripts of amino acid (AA) transporters, myosin heavy chains (MHCs), and the insulin-like growth factor 1 and its receptor (IGF-1/IGF-1R) in patients aimed at major surgery. METHODS Twenty-two randomized patients received steady-state PN (0.16 gN/kg/d, 30 kcal/kg/d) or saline infusions for 12 hours before operation. Blood samples and muscle biopsies were obtained at operation start. Muscle messenger RNA (mRNA) levels of AA transporters (solute carrier family members SNAT2, LAT1, LAT3, LAT4, TAUT, PAT1, CD98), IGF-1, IGF-1R, MHC isoforms (MHC1, MHC2A, MHC2X), and LAT3 protein were quantified and related to concentrations of AA, IGF-1, insulin, and metabolic substrates in blood. RESULTS Muscle mRNA LAT3, LAT4, IGF-1R, and MHC2A increased by PN infusion, with correlations to specific AA transporters and MHC isoforms (P < .01-.05). TAUT and LAT3 correlated to slow (MHC1) and fast (MHC2A, MHC2X) isoforms (P < .001-.02). Muscle IGF-1 mRNA correlated to plasma essential AAs, whereas IGF-1R mRNA was related to LAT3, MHC2A, and serum IGF-1 (P < .001-.03). CONCLUSIONS The results confirm that short-term preoperative PN activates transcription of AA transporters and myosin isoforms. Thus, combinations of methods on gene transcription and translation of muscle proteins can be applied to define efficient combinations of nutrition and hormones to catabolic patients in preoperative and postoperative settings.
Collapse
Affiliation(s)
- Britt-Marie Iresjö
- Surgical Metabolic Research Lab, Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Cecilia Engström
- Surgical Metabolic Research Lab, Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Ulrika Smedh
- Surgical Metabolic Research Lab, Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Kent Lundholm
- Surgical Metabolic Research Lab, Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Sweden
| |
Collapse
|
57
|
Geoghegan D, Arnall C, Hatton D, Noble-Longster J, Sellick C, Senussi T, James DC. Control of amino acid transport into Chinese hamster ovary cells. Biotechnol Bioeng 2018; 115:2908-2929. [PMID: 29987891 DOI: 10.1002/bit.26794] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 06/06/2018] [Accepted: 07/04/2018] [Indexed: 12/21/2022]
Abstract
Amino acid transporters (AATs) represent a key interface between the cell and its environment, critical for all cellular processes: Energy generation, redox control, and synthesis of cell and product biomass. However, very little is known about the activity of different functional classes of AATs in Chinese hamster ovary (CHO) cells, how they support cell growth and productivity, and the potential for engineering their activity and/or the composition of amino acids in growth media to improve CHO cell performance in vitro. In this study, we have comparatively characterized AAT expression in untransfected and monoclonal antibody (MAb)-producing CHO cells using transcriptome analysis by RNA-seq, and mechanistically dissected AAT function using a variety of transporter-specific chemical inhibitors, comparing their effect on cell proliferation, recombinant protein production, and amino acid transport. Of a possible 56 mammalian plasma membrane AATs, 16 AAT messenger RNAs (mRNAs) were relatively abundant across all CHO cell populations. Of these, a subset of nine AAT mRNAs were more abundant in CHO cells engineered to produce a recombinant MAb. Together, upregulated AATs provide additional supply of specific amino acids overrepresented in MAb biomass compared to CHO host cell biomass, enable transport of synthetic substrates for glutathione synthesis, facilitate transport of essential amino acids to maintain active protein synthesis, and provide amino acid substrates for coordinated antiport systems to maintain supplies of proteinogenic and essential amino acids.
Collapse
Affiliation(s)
- Darren Geoghegan
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield, UK
| | - Claire Arnall
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield, UK
| | | | - Joanne Noble-Longster
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield, UK
| | | | | | - David C James
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield, UK
| |
Collapse
|
58
|
van Geldermalsen M, Quek LE, Turner N, Freidman N, Pang A, Guan YF, Krycer JR, Ryan R, Wang Q, Holst J. Benzylserine inhibits breast cancer cell growth by disrupting intracellular amino acid homeostasis and triggering amino acid response pathways. BMC Cancer 2018; 18:689. [PMID: 29940911 PMCID: PMC6019833 DOI: 10.1186/s12885-018-4599-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 06/15/2018] [Indexed: 01/22/2023] Open
Abstract
Background Cancer cells require increased levels of nutrients such as amino acids to sustain their rapid growth. In particular, leucine and glutamine have been shown to be important for growth and proliferation of some breast cancers, and therefore targeting the primary cell-surface transporters that mediate their uptake, L-type amino acid transporter 1 (LAT1) and alanine, serine, cysteine-preferring transporter 2 (ASCT2), is a potential therapeutic strategy. Methods The ASCT2 inhibitor, benzylserine (BenSer), is also able to block LAT1 activity, thus inhibiting both leucine and glutamine uptake. We therefore aimed to investigate the effects of BenSer in breast cancer cell lines to determine whether combined LAT1 and ASCT2 inhibition could inhibit cell growth and proliferation. Results BenSer treatment significantly inhibited both leucine and glutamine uptake in MCF-7, HCC1806 and MDA-MB-231 breast cancer cells, causing decreased cell viability and cell cycle progression. These effects were not primarily leucine-mediated, as BenSer was more cytostatic than the LAT family inhibitor, BCH. Oocyte uptake assays with ectopically expressed amino acid transporters identified four additional targets of BenSer, and gas chromatography-mass spectrometry (GCMS) analysis of intracellular amino acid concentrations revealed that this BenSer-mediated inhibition of amino acid uptake was sufficient to disrupt multiple pathways of amino acid metabolism, causing reduced lactate production and activation of an amino acid response (AAR) through activating transcription factor 4 (ATF4). Conclusions Together these data showed that BenSer blockade inhibited breast cancer cell growth and viability through disruption of intracellular amino acid homeostasis and inhibition of downstream metabolic and growth pathways. Electronic supplementary material The online version of this article (10.1186/s12885-018-4599-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Michelle van Geldermalsen
- Origins of Cancer Program, Centenary Institute, University of Sydney, Locked Bag 6, Newtown, NSW, 2042, Australia.,Sydney Medical School, University of Sydney, Sydney, Australia
| | - Lake-Ee Quek
- School of Mathematics and Statistics, University of Sydney, Sydney, Australia
| | - Nigel Turner
- School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Natasha Freidman
- Transporter Biology Group, Discipline of Pharmacology, Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Angel Pang
- Origins of Cancer Program, Centenary Institute, University of Sydney, Locked Bag 6, Newtown, NSW, 2042, Australia.,Sydney Medical School, University of Sydney, Sydney, Australia
| | - Yi Fang Guan
- Origins of Cancer Program, Centenary Institute, University of Sydney, Locked Bag 6, Newtown, NSW, 2042, Australia.,Sydney Medical School, University of Sydney, Sydney, Australia
| | - James R Krycer
- School of Life and Environmental Sciences, University of Sydney, Sydney, Australia.,Charles Perkins Centre, University of Sydney, Sydney, Australia
| | - Renae Ryan
- Transporter Biology Group, Discipline of Pharmacology, Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Qian Wang
- Origins of Cancer Program, Centenary Institute, University of Sydney, Locked Bag 6, Newtown, NSW, 2042, Australia.,Sydney Medical School, University of Sydney, Sydney, Australia
| | - Jeff Holst
- Origins of Cancer Program, Centenary Institute, University of Sydney, Locked Bag 6, Newtown, NSW, 2042, Australia. .,Sydney Medical School, University of Sydney, Sydney, Australia.
| |
Collapse
|
59
|
Habashy WS, Milfort MC, Adomako K, Attia YA, Rekaya R, Aggrey SE. Effect of heat stress on amino acid digestibility and transporters in meat-type chickens. Poult Sci 2018; 96:2312-2319. [PMID: 28339933 DOI: 10.3382/ps/pex027] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 02/24/2017] [Indexed: 12/19/2022] Open
Abstract
The present study was conducted to investigate the effect of heat stress (HS) on performance, digestibility, and molecular transporters of amino acids in broilers. Cobb 500 chicks were raised from hatch till 13 d in floor pens. At d 14, 48 birds were randomly and equally divided between a control group (25°C) and a HS treatment group (35°C). Birds in both treatment classes were individually caged and fed ad libitum on a diet containing 18.7% CP and 3,560 Kcal ME/Kg. Five birds per treatment at one and 12 d post treatment were euthanized and the Pectoralis major (P. major) and ileum were sampled for gene expression analysis. At d 33, ileal contents were collected and used for digestibility analysis. Broilers under HS had reduced growth and feed intake compared to controls. Although the apparent ileal digestibility (AID) was consistently higher for all amino acids in the HS group, it was not significant except for hydroxylysine. The amino acid consumption and retention were significantly lower in the HS group when compared to the control group. Meanwhile, the retention of amino acids per BWG was higher in the HS group when compared to the control group except for hydroxylysine and ornithine. The dynamics of amino acid transporters in the P. major and ileum was influenced by HS. In P. major and ileum tissues at d one, transporters SNAT1, SNAT2, SNAT7, TAT1, and b0,+AT, were down-regulated in the HS group. Meanwhile, LAT4 and B0AT were down-regulated only in the P. major in the treatment group. The amino acid transporters B0AT and SNAT7 at d 12 post HS were down-regulated in the P. major and ileum, but SNAT2 was down-regulated only in the ileum and TAT1 was down-regulated only in the P. major compared with the control group. These changes in amino acid transporters may explain the reduced growth in meat type chickens under heat stress.
Collapse
Affiliation(s)
- W S Habashy
- NutriGenomics Laboratory, Department of Poultry Science, University of Georgia, Athens 30602.,Department of Animal and Poultry Production, Damanhour University, Damanhour, Al-Behira, Egypt
| | - M C Milfort
- NutriGenomics Laboratory, Department of Poultry Science, University of Georgia, Athens 30602
| | - K Adomako
- Department of Animal Science, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Y A Attia
- Department of Animal and Poultry Production, Damanhour University, Damanhour, Al-Behira, Egypt.,Arid Land Agriculture Department, King Abdulaziz University, Jeddah, Saudi Arabia
| | - R Rekaya
- Department of Animal and Dairy Sciences, University of Georgia, Athens 30602
| | - S E Aggrey
- NutriGenomics Laboratory, Department of Poultry Science, University of Georgia, Athens 30602
| |
Collapse
|
60
|
Abstract
Nucleobases are water-soluble compounds that need specific transporters to cross biological membranes. Cumulative evidence based on studies using animal tissues and cells indicates that the carrier-mediated transport systems for purine and pyrimidine nucleobases can be classified into the following two types: concentrative transport systems that mediate nucleobase transport depending on the sodium ion concentration gradient; and other systems that mediate facilitated diffusion depending on the concentration gradient of the substrate. Recently, several molecular transporters that are involved in both transport systems have been identified. The function and activity of these transporters could be of pharmacological significance considering the roles that they play not only in nucleotide synthesis and metabolism but also in the pharmacokinetics and delivery of a variety of nucleobase analogues used in anticancer and antiviral drug therapy. The present review provides an overview of the recent advances in our understanding of the molecular basis of nucleobase transport systems, focusing on the transporters that mediate purine nucleobases, and discusses the involvement of intracellular metabolism in purine nucleobase transport and chemotherapy using ganciclovir.
Collapse
Affiliation(s)
- Katsuhisa Inoue
- Department of Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences
| |
Collapse
|
61
|
Akanuma SI, Yamakoshi A, Sugouchi T, Kubo Y, Hartz AMS, Bauer B, Hosoya KI. Role of l-Type Amino Acid Transporter 1 at the Inner Blood-Retinal Barrier in the Blood-to-Retina Transport of Gabapentin. Mol Pharm 2018; 15:2327-2337. [PMID: 29688723 DOI: 10.1021/acs.molpharmaceut.8b00179] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Gabapentin is an antiseizure drug that is known to also have beneficial effects on the retinal cells. To use gabapentin in retinal pharmacotherapy, it is critical to understand gabapentin distribution in the retina. The purpose of this study was to clarify the kinetics of gabapentin influx transport across the inner and outer blood-retinal barrier (BRB), which regulates the exchange of compounds/drugs between the circulating blood and the retina. In vivo blood-to-retina gabapentin transfer was evaluated by the rat carotid artery injection technique. In addition, gabapentin transport was examined using in vitro models of the inner (TR-iBRB2 cells) and outer BRB (RPE-J cells). The in vivo [3H]gabapentin transfer to the rat retina across the BRB was significantly reduced in the presence of unlabeled gabapentin, suggesting transporter-mediated blood-to-retina distribution of gabapentin. Substrates of the Na+-independent l-type amino acid transporter 1 (LAT1), such as 2-aminobicyclo[2.2.1]heptane-2-carboxylic acid (BCH), also significantly inhibited the in vivo [3H]gabapentin transfer. [3H]Gabapentin uptake in TR-iBRB2 and RPE-J cells exhibited Na+-independent and saturable kinetics with a Km of 735 and 507 μM, respectively. Regarding the effect of various transporter substrates/inhibitors on gabapentin transport in these cells, LAT1 substrates significantly inhibited [3H]gabapentin uptake in TR-iBRB2 and RPE-J cells. In addition, preloaded [3H]gabapentin release from TR-iBRB2 and RPE-J cells was trans-stimulated by LAT1 substrates through the obligatory exchange mechanism as LAT1. Immunoblot analysis indicates the protein expression of LAT1 in TR-iBRB2 and RPE-J cells. These results imply that LAT1 at the inner and outer BRB takes part in gabapentin transport between the circulating blood and retina. Moreover, treatment of LAT1-targeted small interfering RNA to TR-iBRB2 cells significantly reduced both the level of LAT1 protein expression and [3H]gabapentin uptake activities in TR-iBRB2 cells. In conclusion, data from the present study indicate that LAT1 at the inner BRB is involved in retinal gabapentin transfer, and also suggest that LAT1 mediates gabapentin transport in the RPE cells.
Collapse
Affiliation(s)
- Shin-Ichi Akanuma
- Department of Pharmaceutics, Graduate School of Medicine and Pharmaceutical Sciences , University of Toyama , Sugitani , 2630 Toyama , Japan
| | - Atsuko Yamakoshi
- Department of Pharmaceutics, Graduate School of Medicine and Pharmaceutical Sciences , University of Toyama , Sugitani , 2630 Toyama , Japan
| | - Takeshi Sugouchi
- Department of Pharmaceutics, Graduate School of Medicine and Pharmaceutical Sciences , University of Toyama , Sugitani , 2630 Toyama , Japan
| | - Yoshiyuki Kubo
- Department of Pharmaceutics, Graduate School of Medicine and Pharmaceutical Sciences , University of Toyama , Sugitani , 2630 Toyama , Japan
| | - Anika M S Hartz
- Sanders-Brown Center on Aging , University of Kentucky , Lexington , KY 40536 , United States.,Department of Pharmacology and Nutritional Sciences, College of Medicine , University of Kentucky , Lexington , Kentucky 40536 , United States
| | - Björn Bauer
- Department of Pharmaceutical Sciences, College of Pharmacy , University of Kentucky , Lexington , Kentucky 40536 , United States
| | - Ken-Ichi Hosoya
- Department of Pharmaceutics, Graduate School of Medicine and Pharmaceutical Sciences , University of Toyama , Sugitani , 2630 Toyama , Japan
| |
Collapse
|
62
|
Yoshimoto M, Honda N, Kurihara H, Hiroi K, Nakamura S, Ito M, Shikano N, Itami J, Fujii H. Non-invasive estimation of 10 B-4-borono-L-phenylalanine-derived boron concentration in tumors by PET using 4-borono-2- 18 F-fluoro-phenylalanine. Cancer Sci 2018; 109:1617-1626. [PMID: 29498142 PMCID: PMC5980255 DOI: 10.1111/cas.13553] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Revised: 02/19/2018] [Accepted: 02/21/2018] [Indexed: 01/02/2023] Open
Abstract
In boron neutron capture therapy (BNCT), 10 B-4-borono-L-phenylalanine (BPA) is commonly used as a 10 B carrier. PET using 4-borono-2-18 F-fluoro-phenylalanine (18 F-FBPA PET) has been performed to estimate boron concentration and predict the therapeutic effects of BNCT; however, the association between tumor uptake of 18 F-FBPA and boron concentration in tumors remains unclear. The present study investigated the transport mechanism of 18 F-FBPA and BPA, and evaluated the utility of 18 F-FBPA PET in predicting boron concentration in tumors. The transporter assay revealed that 2-aminobicyclo-(2.2.1)-heptane-2-carboxylic acid, an inhibitor of the L-type amino acid transporter, significantly inhibited 18 F-FBPA and 14 C-4-borono-L-phenylalanine (14 C-BPA) uptake in FaDu and LN-229 human cancer cells. 18 F-FBPA uptake strongly correlated with 14 C-BPA uptake in 7 human tumor cell lines (r = .93; P < .01). PET experiments demonstrated that tumor uptake of 18 F-FBPA was independent of the administration method, and uptake of 18 F-FBPA by bolus injection correlated well with BPA uptake by continuous intravenous infusion. The results of this study revealed that evaluating tumor uptake of 18 F-FBPA by PET was useful for estimating 10 B concentration in tumors.
Collapse
Affiliation(s)
- Mitsuyoshi Yoshimoto
- Division of Functional ImagingExploratory Oncology Research & Clinical Trial CenterNational Cancer CenterKashiwaChibaJapan
| | - Natsuki Honda
- Department of PharmacyNational Cancer Center HospitalTokyoJapan
| | - Hiroaki Kurihara
- Department of Diagnostic RadiologyNational Cancer Center HospitalTokyoJapan
| | - Kenta Hiroi
- Department of Diagnostic RadiologyNational Cancer Center HospitalTokyoJapan
| | - Satoshi Nakamura
- Department of Radiation OncologyNational Cancer Center HospitalTokyoJapan
| | - Masashi Ito
- Department of Radiation OncologyNational Cancer Center HospitalTokyoJapan
| | - Naoto Shikano
- Department of Radiological SciencesIbaraki Prefectural University of Health SciencesAmiIbarakiJapan
| | - Jun Itami
- Department of Radiation OncologyNational Cancer Center HospitalTokyoJapan
| | - Hirofumi Fujii
- Division of Functional ImagingExploratory Oncology Research & Clinical Trial CenterNational Cancer CenterKashiwaChibaJapan
| |
Collapse
|
63
|
Singh N, Ecker GF. Insights into the Structure, Function, and Ligand Discovery of the Large Neutral Amino Acid Transporter 1, LAT1. Int J Mol Sci 2018; 19:E1278. [PMID: 29695141 PMCID: PMC5983779 DOI: 10.3390/ijms19051278] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 04/18/2018] [Accepted: 04/19/2018] [Indexed: 12/31/2022] Open
Abstract
The large neutral amino acid transporter 1 (LAT1, or SLC7A5) is a sodium- and pH-independent transporter, which supplies essential amino acids (e.g., leucine, phenylalanine) to cells. It plays an important role at the Blood⁻Brain Barrier (BBB) where it facilitates the transport of thyroid hormones, pharmaceuticals (e.g., l-DOPA, gabapentin), and metabolites into the brain. Moreover, its expression is highly upregulated in various types of human cancer that are characterized by an intense demand for amino acids for growth and proliferation. Therefore, LAT1 is believed to be an important drug target for cancer treatment. With the crystallization of the arginine/agmatine antiporter (AdiC) from Escherichia Coli, numerous homology models of LAT1 have been built to elucidate the substrate binding site, ligand⁻transporter interaction, and structure⁻function relationship. The use of these models in combination with molecular docking and experimental testing has identified novel chemotypes of ligands of LAT1. Here, we highlight the structure, function, transport mechanism, and homology modeling of LAT1. Additionally, results from structure⁻function studies performed on LAT1 are addressed, which have enhanced our knowledge of the mechanism of substrate binding and translocation. This is followed by a discussion on ligand- and structure-based approaches, with an emphasis on elucidating the molecular basis of LAT1 inhibition. Finally, we provide an exhaustive summary of different LAT1 inhibitors that have been identified so far, including the recently discovered irreversible covalent inhibitors.
Collapse
Affiliation(s)
- Natesh Singh
- Department of Pharmaceutical Chemistry, University of Vienna, Althanstrasse 14, 1090 Wien, Austria.
| | - Gerhard F Ecker
- Department of Pharmaceutical Chemistry, University of Vienna, Althanstrasse 14, 1090 Wien, Austria.
| |
Collapse
|
64
|
Vilches C, Boiadjieva-Knöpfel E, Bodoy S, Camargo S, López de Heredia M, Prat E, Ormazabal A, Artuch R, Zorzano A, Verrey F, Nunes V, Palacín M. Cooperation of Antiporter LAT2/CD98hc with Uniporter TAT1 for Renal Reabsorption of Neutral Amino Acids. J Am Soc Nephrol 2018; 29:1624-1635. [PMID: 29610403 DOI: 10.1681/asn.2017111205] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 02/24/2018] [Indexed: 01/01/2023] Open
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.
Collapse
Affiliation(s)
- Clara Vilches
- Molecular Genetics Laboratory, Genes Disease and Therapy Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Spain
| | - Emilia Boiadjieva-Knöpfel
- Department of Physiology.,Zurich Center for Integrative Human Physiology (ZIHP), and.,Swiss National Centre of Competence in Research (NCCR), Kidney Control of Homeostasis (Kidney.CH), University of Zurich, Zurich, Switzerland
| | - Susanna Bodoy
- Department of Biochemistry and Molecular Medicine, Biology Faculty, University of Barcelona, Barcelona, Spain.,Molecular Medicine Unit, Amino acid transporters and disease group, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Simone Camargo
- Department of Physiology.,Zurich Center for Integrative Human Physiology (ZIHP), and.,Swiss National Centre of Competence in Research (NCCR), Kidney Control of Homeostasis (Kidney.CH), University of Zurich, Zurich, Switzerland
| | - Miguel López de Heredia
- Molecular Genetics Laboratory, Genes Disease and Therapy Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) - U730, U731, U703, and
| | - Esther Prat
- Molecular Genetics Laboratory, Genes Disease and Therapy Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) - U730, U731, U703, and.,Genetics Section, Physiological Sciences Department, Health Sciences and Medicine Faculty, University of Barcelona, Barcelona, Spain; and
| | - Aida Ormazabal
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) - U730, U731, U703, and.,Clinical Biochemistry Department, Institut de Recerca Sant Joan de Déu, Hospital Sant Joan de Déu, Esplugues de Llobregat, Spain
| | - Rafael Artuch
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) - U730, U731, U703, and.,Clinical Biochemistry Department, Institut de Recerca Sant Joan de Déu, Hospital Sant Joan de Déu, Esplugues de Llobregat, Spain
| | - Antonio Zorzano
- Department of Biochemistry and Molecular Medicine, Biology Faculty, University of Barcelona, Barcelona, Spain.,Molecular Medicine Unit, Amino acid transporters and disease group, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM) - CB07/08/0017, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - François Verrey
- Department of Physiology.,Zurich Center for Integrative Human Physiology (ZIHP), and.,Swiss National Centre of Competence in Research (NCCR), Kidney Control of Homeostasis (Kidney.CH), University of Zurich, Zurich, Switzerland
| | - Virginia Nunes
- Molecular Genetics Laboratory, Genes Disease and Therapy Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Spain; .,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) - U730, U731, U703, and.,Genetics Section, Physiological Sciences Department, Health Sciences and Medicine Faculty, University of Barcelona, Barcelona, Spain; and
| | - Manuel Palacín
- Department of Biochemistry and Molecular Medicine, Biology Faculty, University of Barcelona, Barcelona, Spain; .,Molecular Medicine Unit, Amino acid transporters and disease group, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) - U730, U731, U703, and
| |
Collapse
|
65
|
Blazejewski SM, Bennison SA, Smith TH, Toyo-Oka K. Neurodevelopmental Genetic Diseases Associated With Microdeletions and Microduplications of Chromosome 17p13.3. Front Genet 2018; 9:80. [PMID: 29628935 PMCID: PMC5876250 DOI: 10.3389/fgene.2018.00080] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 02/26/2018] [Indexed: 01/24/2023] Open
Abstract
Chromosome 17p13.3 is a region of genomic instability that is linked to different rare neurodevelopmental genetic diseases, depending on whether a deletion or duplication of the region has occurred. Chromosome microdeletions within 17p13.3 can result in either isolated lissencephaly sequence (ILS) or Miller-Dieker syndrome (MDS). Both conditions are associated with a smooth cerebral cortex, or lissencephaly, which leads to developmental delay, intellectual disability, and seizures. However, patients with MDS have larger deletions than patients with ILS, resulting in additional symptoms such as poor muscle tone, congenital anomalies, abnormal spasticity, and craniofacial dysmorphisms. In contrast to microdeletions in 17p13.3, recent studies have attracted considerable attention to a condition known as a 17p13.3 microduplication syndrome. Depending on the genes involved in their microduplication, patients with 17p13.3 microduplication syndrome may be categorized into either class I or class II. Individuals in class I have microduplications of the YWHAE gene encoding 14-3-3ε, as well as other genes in the region. However, the PAFAH1B1 gene encoding LIS1 is never duplicated in these patients. Class I microduplications generally result in learning disabilities, autism, and developmental delays, among other disorders. Individuals in class II always have microduplications of the PAFAH1B1 gene, which may include YWHAE and other genetic microduplications. Class II microduplications generally result in smaller body size, developmental delays, microcephaly, and other brain malformations. Here, we review the phenotypes associated with copy number variations (CNVs) of chromosome 17p13.3 and detail their developmental connection to particular microdeletions or microduplications. We also focus on existing single and double knockout mouse models that have been used to study human phenotypes, since the highly limited number of patients makes a study of these conditions difficult in humans. These models are also crucial for the study of brain development at a mechanistic level since this cannot be accomplished in humans. Finally, we emphasize the usefulness of the CRISPR/Cas9 system and next generation sequencing in the study of neurodevelopmental diseases.
Collapse
Affiliation(s)
- Sara M Blazejewski
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Sarah A Bennison
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Trevor H Smith
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Kazuhito Toyo-Oka
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, United States
| |
Collapse
|
66
|
Characterisation of L-Type Amino Acid Transporter 1 (LAT1) Expression in Human Skeletal Muscle by Immunofluorescent Microscopy. Nutrients 2017; 10:nu10010023. [PMID: 29278358 PMCID: PMC5793251 DOI: 10.3390/nu10010023] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 11/16/2017] [Accepted: 12/13/2017] [Indexed: 12/21/2022] Open
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.
Collapse
|
67
|
Lin Y, Duan X, Lv H, Yang Y, Liu Y, Gao X, Hou X. The effects of L-type amino acid transporter 1 on milk protein synthesis in mammary glands of dairy cows. J Dairy Sci 2017; 101:1687-1696. [PMID: 29224866 DOI: 10.3168/jds.2017-13201] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 10/12/2017] [Indexed: 01/23/2023]
Abstract
The mammary gland requires the uptake of AA for milk protein synthesis during lactation. The L-type amino acid transporter 1 (LAT1, encoded by SLC7A5), found in many different types of mammalian cells, is indispensable as a transporter of essential AA to maintain cell growth and protein synthesis. However, the function of LAT1 in regulating milk protein synthesis in the mammary gland of the dairy cow remains largely unknown. For the current study, we characterized the relationship between LAT1 expression and milk protein synthesis in lactating dairy cows and investigated whether the mammalian target of rapamycin complex 1 (mTORC1) signaling controls the expression of LAT1 in their mammary glands. We found that LAT1 and the heavy chain of its chaperone, 4F2, were expressed in mammary tissues of lactating cows, with the expression levels of LAT1 and the 4F2 heavy chain being significantly greater in lactating mammary tissues with high-milk protein content (milk yield, 33.8 ± 2.1 kg/d; milk protein concentration >3%, wt/vol,; n = 3) than in tissues from cows with low-milk protein content (milk yield, 33.7 ± 0.5 kg/d; milk protein concentration <3%, wt/vol; n = 3). Immunofluorescence staining of sectioned mammary tissues from cows with high and low milk protein content showed that LAT1 was located on the whole plasma membrane of alveolar epithelial cells, suggesting that LAT1 provides essential AA to the mammary gland. In cultured mammary epithelial cells from the dairy cows with high-milk protein content, knockdown of LAT1 expression decreased cell viability and β-casein expression; in contrast, overexpression of LAT1 had the opposite effect. Inhibition of mTORC1 by rapamycin attenuated the phosphorylation of molecules related to mTORC1 signaling and caused a marked decrease in LAT1 expression in the cultured cells; expression of β-casein also decreased significantly. These results suggest that LAT1 is involved in milk protein synthesis in the mammary glands of lactating dairy cows and that the mTORC1 signaling pathway might be a control point for regulation of LAT1 expression, which could ultimately be used to alter milk protein synthesis.
Collapse
Affiliation(s)
- Ye Lin
- College of Life Science, Northeast Agricultural University, Harbin 150030, China; Key Laboratory of Dairy Science of Education Ministry, and Northeast Agricultural University, Harbin 150030, China
| | - Xiaoyu Duan
- College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - He Lv
- College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Yang Yang
- College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Ying Liu
- College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Xuejun Gao
- College of Life Science, Northeast Agricultural University, Harbin 150030, China; Key Laboratory of Dairy Science of Education Ministry, and Northeast Agricultural University, Harbin 150030, China
| | - Xiaoming Hou
- College of Life Science, Northeast Agricultural University, Harbin 150030, China; Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin 150030, China.
| |
Collapse
|
68
|
Fibroblast-like synoviocyte migration is enhanced by IL-17-mediated overexpression of L-type amino acid transporter 1 (LAT1) via the mTOR/4E-BP1 pathway. Amino Acids 2017; 50:331-340. [PMID: 29198077 DOI: 10.1007/s00726-017-2520-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 11/28/2017] [Indexed: 01/25/2023]
Abstract
In rheumatoid arthritis (RA), activated synovial fibroblasts have the ability to invade joint cartilage, actively contributing to joint destruction in RA. The mechanisms underlying this cell migration and invasion remain unclear. Our previous results and data from the GEO profile indicate that the L-type amino acid transporter gene, LAT1, is overexpressed in the synovium of RA. To identify its potential role in RA, fibroblast-like synoviocytes (FLS) from patients with RA were used to determine the effects of suppressing the LAT1 genes using RNA interference and the LAT inhibitor, BCH. We found that BCH exposure reduced the phosphorylation of mTOR and its downstream target 4EBP1, radiolabeled leucine uptake, and migration of RA FLS. LAT1 silencing by siRNA presented effects similar to BCH inhibition. Treatment of cells with IL-17 stimulated the expression of LAT1. In contrast, applying an inhibitor of mTOR pathway, temsirolimus, or silencing eIF4E neutralized the stimulation of IL-17 on LAT1. BCH and siLAT1 also resulted in lower IL-17-stimulated leucine uptake and cell migration. These results suggest that the migration of RA FLS is aggravated by IL-17-mediated overexpression of LAT1 via mTOR/4E-BP1 pathway. In conclusion, further investigation is warranted into LAT1 as a potential target for drug therapies aimed at attenuating migration of transformed-appearing fibroblasts and subsequently preventing further erosion of bone and cartilage.
Collapse
|
69
|
Ikeda K, Kinoshita M, Kayama H, Nagamori S, Kongpracha P, Umemoto E, Okumura R, Kurakawa T, Murakami M, Mikami N, Shintani Y, Ueno S, Andou A, Ito M, Tsumura H, Yasutomo K, Ozono K, Takashima S, Sakaguchi S, Kanai Y, Takeda K. Slc3a2 Mediates Branched-Chain Amino-Acid-Dependent Maintenance of Regulatory T Cells. Cell Rep 2017; 21:1824-1838. [DOI: 10.1016/j.celrep.2017.10.082] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 08/06/2017] [Accepted: 10/20/2017] [Indexed: 12/23/2022] Open
|
70
|
Choi DW, Kim DK, Kanai Y, Wempe MF, Endou H, Kim JK. JPH203, a selective L-type amino acid transporter 1 inhibitor, induces mitochondria-dependent apoptosis in Saos2 human osteosarcoma cells. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2017; 21:599-607. [PMID: 29200902 PMCID: PMC5709476 DOI: 10.4196/kjpp.2017.21.6.599] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 08/11/2017] [Accepted: 09/08/2017] [Indexed: 11/30/2022]
Abstract
Most normal cells express L-type amino acid transporter 2 (LAT2). However, L-type amino acid transporter 1 (LAT1) is highly expressed in many tumor cells and presumed to support their increased growth and proliferation. This study examined the effects of JPH203, a selective LAT1 inhibitor, on cell growth and its mechanism for cell death in Saos2 human osteosarcoma cells. FOB human osteoblastic cells and Saos2 cells expressed LAT1 and LAT2 together with their associating protein 4F2 heavy chain, but the expression of LAT2 in the Saos2 cells was especially weak. JPH203 and BCH, a non-selective L-type amino acid transporter inhibitor, potently inhibited L-leucine uptake in Saos2 cells. As expected, the intrinsic ability of JPH203 to inhibit L-leucine uptake was far more efficient than that of BCH in Saos2 cells. Likewise, JPH203 and BCH inhibited Saos2 cell growth with JPH203 being superior to BCH in this regard. Furthermore, JPH203 increased apoptosis rates and formed DNA ladder in Saos2 cells. Moreover, JPH203 activated the mitochondria-dependent apoptotic signaling pathway by upregulating pro-apoptotic factors, such as Bad, Bax, and Bak, and the active form of caspase-9, and downregulating anti-apoptotic factors, such as Bcl-2 and Bcl-xL. These results suggest that the inhibition of LAT1 activity via JPH203, which may act as a potential novel anti-cancer agent, leads to apoptosis mediated by the mitochondria-dependent intrinsic apoptotic signaling pathway by inducing the intracellular depletion of neutral amino acids essential for cell growth in Saos2 human osteosarcoma cells.
Collapse
Affiliation(s)
- Dae Woo Choi
- Department of Pharmacology, Chonnam National University Medical School, Gwangju 61469, Korea
| | - Do Kyung Kim
- Department of Oral Physiology, Chosun University School of Dentistry, Gwangju 61452, Korea
| | - Yoshikatsu Kanai
- Department of Bio-system Pharmacology, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Michael F Wempe
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado 80045, USA
| | - Hitoshi Endou
- Department of Pharmacology and Toxicology, Kyorin University School of Medicine, Tokyo 181-8611, Japan.,J-Pharma Co., Ltd., Yokohama, Kanagawa 230-0046, Japan
| | - Jong-Keun Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju 61469, Korea
| |
Collapse
|
71
|
Badziong J, Ting S, Synoracki S, Tiedje V, Brix K, Brabant G, Moeller LC, Schmid KW, Fuhrer D, Zwanziger D. Differential regulation of monocarboxylate transporter 8 expression in thyroid cancer and hyperthyroidism. Eur J Endocrinol 2017; 177:243-250. [PMID: 28576880 DOI: 10.1530/eje-17-0279] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 05/08/2017] [Accepted: 06/02/2017] [Indexed: 11/08/2022]
Abstract
OBJECTIVE Thyroid hormone (TH) transporters are expressed in thyrocytes and most play a role in TH release. We asked whether expression of the monocarboxylate transporter 8 (MCT8) and the L-type amino acid transporters LAT2 and LAT4 is changed with thyrocyte dedifferentiation and in hyperfunctioning thyroid tissues. DESIGN AND METHODS Protein expression and localization of transporters was determined by immunohistochemistry in human thyroid specimen including normal thyroid tissue (NT, n = 19), follicular adenoma (FA, n = 44), follicular thyroid carcinoma (FTC, n = 45), papillary thyroid carcinoma (PTC, n = 40), anaplastic thyroid carcinoma (ATC, n = 40) and Graves' disease (GD, n = 50) by calculating the 'hybrid' (H) score. Regulation of transporter expression was investigated in the rat follicular thyroid cell line PCCL3 under basal and thyroid stimulating hormone (TSH) conditions. RESULTS MCT8 and LAT4 were localized at the plasma membrane, while LAT2 transporter showed cytoplasmic localization. MCT8 expression was downregulated in benign and malignant thyroid tumours as compared to NT. In contrast, significant upregulation of MCT8, LAT2 and LAT4 was found in GD. Furthermore, a stronger expression of MCT8 was demonstrated in PCCL3 cells after TSH stimulation. CONCLUSIONS Downregulation of MCT8 in thyroid cancers qualifies MCT8 as a marker of thyroid differentiation. The more variable expression of LATs in distinct thyroid malignancies may be linked with other transporter properties relevant to altered metabolism in cancer cells, i.e. amino acid transport. Consistent upregulation of MCT8 in GD is in line with increased TH release in hyperthyroidism, an assumption supported by our in vitro results showing TSH-dependent upregulation of MCT8.
Collapse
Affiliation(s)
- Julia Badziong
- Department of Endocrinology and Metabolism and Division of Laboratory Research, University of Duisburg-Essen, Essen, Germany
| | - Saskia Ting
- University Hospital Essen, Institute of Pathology, Essen, Germany
| | - Sarah Synoracki
- University Hospital Essen, Institute of Pathology, Essen, Germany
| | - Vera Tiedje
- Department of Endocrinology and Metabolism and Division of Laboratory Research, University of Duisburg-Essen, Essen, Germany
| | - Klaudia Brix
- Department of Life Sciences and Chemistry, Jacobs University Bremen, Bremen, Germany
| | - Georg Brabant
- University Hospital Schleswig-Holstein, Experimental and Clinical Endocrinology, Lübeck, Germany
| | - Lars Christian Moeller
- Department of Endocrinology and Metabolism and Division of Laboratory Research, University of Duisburg-Essen, Essen, Germany
| | | | - Dagmar Fuhrer
- Department of Endocrinology and Metabolism and Division of Laboratory Research, University of Duisburg-Essen, Essen, Germany
| | - Denise Zwanziger
- Department of Endocrinology and Metabolism and Division of Laboratory Research, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
72
|
Werner A, Koschke M, Leuchtner N, Luckner-Minden C, Habermeier A, Rupp J, Heinrich C, Conradi R, Closs EI, Munder M. Reconstitution of T Cell Proliferation under Arginine Limitation: Activated Human T Cells Take Up Citrulline via L-Type Amino Acid Transporter 1 and Use It to Regenerate Arginine after Induction of Argininosuccinate Synthase Expression. Front Immunol 2017; 8:864. [PMID: 28791021 PMCID: PMC5523021 DOI: 10.3389/fimmu.2017.00864] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 07/07/2017] [Indexed: 11/13/2022] Open
Abstract
In the tumor microenvironment, arginine is metabolized by arginase-expressing myeloid cells. This arginine depletion profoundly inhibits T cell functions and is crucially involved in tumor-induced immunosuppression. Reconstitution of adaptive immune functions in the context of arginase-mediated tumor immune escape is a promising therapeutic strategy to boost the immunological antitumor response. Arginine can be recycled in certain mammalian tissues from citrulline via argininosuccinate (ASA) in a two-step enzymatic process involving the enzymes argininosuccinate synthase (ASS) and argininosuccinate lyase (ASL). Here, we demonstrate that anti-CD3/anti-CD28-activated human primary CD4+ and CD8+ T cells upregulate ASS expression in response to low extracellular arginine concentrations, while ASL is expressed constitutively. ASS expression peaked under moderate arginine restriction (20 µM), but no relevant induction was detectable in the complete absence of extracellular arginine. The upregulated ASS correlated with a reconstitution of T cell proliferation upon supplementation of citrulline, while the suppressed production of IFN-γ was refractory to citrulline substitution. In contrast, ASA reconstituted proliferation and cytokine synthesis even in the complete absence of arginine. By direct quantification of intracellular metabolites we show that activated primary human T cells import citrulline but only metabolize it further to ASA and arginine when ASS is expressed in the context of low amounts of extracellular arginine. We then clarify that citrulline transport is largely mediated by the L-type amino acid transporter 1 (LAT1), induced upon human T cell activation. Upon siRNA-mediated knockdown of LAT1, activated T cells lost the ability to import citrulline. These data underline the potential of citrulline substitution as a promising pharmacological way to treat immunosuppression in settings of arginine deprivation.
Collapse
Affiliation(s)
- Anke Werner
- Third Department of Medicine (Hematology, Oncology, and Pneumology), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.,Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Miriam Koschke
- Third Department of Medicine (Hematology, Oncology, and Pneumology), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Nadine Leuchtner
- Third Department of Medicine (Hematology, Oncology, and Pneumology), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Claudia Luckner-Minden
- Third Department of Medicine (Hematology, Oncology, and Pneumology), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Alice Habermeier
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Johanna Rupp
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Christin Heinrich
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Roland Conradi
- Transfusion Center, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Ellen I Closs
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Markus Munder
- Third Department of Medicine (Hematology, Oncology, and Pneumology), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.,Research Center for Immunotherapy, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
73
|
Comasco E, Vumma R, Toffoletto S, Johansson J, Flyckt L, Lewander T, Oreland L, Bjerkenstedt L, Andreou D, Söderman E, Terenius L, Agartz I, Jönsson EG, Venizelos N. Genetic and Functional Study of L-Type Amino Acid Transporter 1 in Schizophrenia. Neuropsychobiology 2017; 74:96-103. [PMID: 28190014 DOI: 10.1159/000455234] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2016] [Accepted: 12/20/2016] [Indexed: 12/27/2022]
Abstract
Schizophrenia involves neural catecholaminergic dysregulation. Tyrosine is the precursor of catecholamines, and its major transporter, according to studies on fibroblasts, in the brain is the L-type amino acid transporter 1 (LAT1). The present study assessed haplotype tag single-nucleotide polymorphisms (SNPs) of the SLC7A5/LAT1 gene in 315 patients with psychosis within the schizophrenia spectrum and 233 healthy controls to investigate genetic vulnerability to the disorder as well as genetic relationships to homovanillic acid (HVA) and 3-methoxy-4-hydroxyphenylglycol (MHPG), the major catecholamine metabolites in the cerebrospinal fluid (CSF). Moreover, the involvement of the different isoforms of the system L in tyrosine uptake and LAT1 tyrosine kinetics were studied in fibroblast cell lines of 10 patients with schizophrenia and 10 healthy controls. The results provide suggestive evidence of individual vulnerability to schizophrenia related to the LAT1 SNP rs9936204 genotype. A number of SNPs were nominally associated with CSF HVA and MHPG concentrations but did not survive correction for multiple testing. The LAT1 isoform was confirmed as the major tyrosine transporter in patients with schizophrenia. However, the kinetic parameters (maximal transport capacity, affinity of the binding sites, and diffusion constant of tyrosine transport through the LAT1 isoform) did not differ between patients with schizophrenia and controls. The present genetic findings call for independent replication in larger samples, while the functional study seems to exclude a role of LAT1 in the aberrant transport of tyrosine in fibroblasts of patients with schizophrenia.
Collapse
Affiliation(s)
- Erika Comasco
- Department of Neuroscience, Uppsala University, Uppsala, Sweden
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
74
|
Yothaisong S, Dokduang H, Anzai N, Hayashi K, Namwat N, Yongvanit P, Sangkhamanon S, Jutabha P, Endou H, Loilome W. Inhibition of l-type amino acid transporter 1 activity as a new therapeutic target for cholangiocarcinoma treatment. Tumour Biol 2017; 39:1010428317694545. [PMID: 28347255 DOI: 10.1177/1010428317694545] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Unlike normal cells, cancer cells undergo unlimited growth and multiplication, causing them to require massive amounts of amino acid to support their continuous metabolism. Among the amino acid transporters expressed on the plasma membrane, l-type amino acid transporter-1, a Na+-independent neutral amino acid transporter, is highly expressed in many types of human cancer including cholangiocarcinoma. Our previous study reported that l-type amino acid transporter-1 and its co-functional protein CD98 were highly expressed and implicated in cholangiocarcinoma progression and carcinogenesis. Therefore, this study determined the effect of JPH203, a selective inhibitor of l-type amino acid transporter-1 activity, on cholangiocarcinoma cell inhibition both in vitro and in vivo. JPH203 dramatically suppressed [14C]l-leucine uptake as well as cell growth in cholangiocarcinoma cell lines along with altering the expression of l-type amino acid transporter-1 and CD98 in response to amino acid depletion. We also demonstrated that JPH203 induced both G2/M and G0/G1 cell cycle arrest, as well as reduced the S phase accompanied by altered expression of the proteins in cell cycle progression: cyclin D1, CDK4, and CDK6. There was also cell cycle arrest of the related proteins, P21 and P27, in KKU-055 and KKU-213 cholangiocarcinoma cells. Apoptosis induction, detected by an increase in trypan blue-stained cells along with a cleaved caspase-3/caspase-3 ratio, occurred in JPH203-treated cholangiocarcinoma cells at the highest concentration tested (100 µM). As expected, daily intravenous administration of JPH203 (12.5 and 25 mg/kg) significantly inhibited tumor growth in KKU-213 cholangiocarcinoma cell xenografts in the nude mice model in a dose-dependent manner with no statistically significant change in the animal's body weight and with no differences in the histology and appearance of the internal organs compared with the control group. Our study demonstrates that suppression of l-type amino acid transporter-1 activity using JPH203 might be used as a new therapeutic strategy for cholangiocarcinoma treatment.
Collapse
Affiliation(s)
- Supak Yothaisong
- 1 Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,2 Liver Fluke and Cholangiocarcinoma Research Center, Khon Kaen University, Khon Kaen, Thailand.,3 Cholangiocarcinoma Screening and Care Program (CASCAP), Khon Kaen University, Khon Kaen, Thailand
| | - Hasaya Dokduang
- 2 Liver Fluke and Cholangiocarcinoma Research Center, Khon Kaen University, Khon Kaen, Thailand.,3 Cholangiocarcinoma Screening and Care Program (CASCAP), Khon Kaen University, Khon Kaen, Thailand
| | - Naohiko Anzai
- 4 Department of Pharmacology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Keitaro Hayashi
- 5 Department of Pharmacology and Toxicology, School of Medicine, Dokkyo Medical University, Mibu, Japan
| | - Nisana Namwat
- 1 Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,2 Liver Fluke and Cholangiocarcinoma Research Center, Khon Kaen University, Khon Kaen, Thailand.,3 Cholangiocarcinoma Screening and Care Program (CASCAP), Khon Kaen University, Khon Kaen, Thailand
| | - Puangrat Yongvanit
- 1 Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,2 Liver Fluke and Cholangiocarcinoma Research Center, Khon Kaen University, Khon Kaen, Thailand.,3 Cholangiocarcinoma Screening and Care Program (CASCAP), Khon Kaen University, Khon Kaen, Thailand
| | - Sakkarn Sangkhamanon
- 6 Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Promsuk Jutabha
- 5 Department of Pharmacology and Toxicology, School of Medicine, Dokkyo Medical University, Mibu, Japan
| | | | - Watcharin Loilome
- 1 Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,2 Liver Fluke and Cholangiocarcinoma Research Center, Khon Kaen University, Khon Kaen, Thailand.,3 Cholangiocarcinoma Screening and Care Program (CASCAP), Khon Kaen University, Khon Kaen, Thailand
| |
Collapse
|
75
|
Zhenyukh O, Civantos E, Ruiz-Ortega M, Sánchez MS, Vázquez C, Peiró C, Egido J, Mas S. High concentration of branched-chain amino acids promotes oxidative stress, inflammation and migration of human peripheral blood mononuclear cells via mTORC1 activation. Free Radic Biol Med 2017; 104:165-177. [PMID: 28089725 DOI: 10.1016/j.freeradbiomed.2017.01.009] [Citation(s) in RCA: 239] [Impact Index Per Article: 34.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 12/23/2016] [Accepted: 01/06/2017] [Indexed: 12/30/2022]
Abstract
Leucine, isoleucine and valine are essential aminoacids termed branched-chain amino acids (BCAA) due to its aliphatic side-chain. In several pathological and physiological conditions increased BCAA plasma concentrations have been described. Elevated BCAA levels predict insulin resistance development. Moreover, BCAA levels higher than 2mmol/L are neurotoxic by inducing microglial activation in maple syrup urine disease. However, there are no studies about the direct effects of BCAA in circulating cells. We have explored whether BCAA could promote oxidative stress and pro-inflammatory status in peripheral blood mononuclear cells (PBMCs) obtained from healthy donors. In cultured PBMCs, 10mmol/L BCAA increased the production of reactive oxygen species (ROS) via both NADPH oxidase and the mitochondria, and activated Akt-mTOR signalling. By using several inhibitors and activators of these molecular pathways we have described that mTOR activation by BCAA is linked to ROS production and mitochondrial dysfunction. BCAA stimulated the activation of the redox-sensitive transcription factor NF-κB, which resulted in the release of pro-inflammatory molecules, such as interleukin-6, tumor necrosis factor-α, intracellular adhesion molecule-1 or CD40L, and the migration of PBMCs. In conclusion, elevated BCAA blood levels can promote the activation of circulating PBMCs, by a mechanism that involving ROS production and NF-κB pathway activation. These data suggest that high concentrations of BCAA could exert deleterious effects on circulating blood cells and therefore contribute to the pro-inflammatory and oxidative status observed in several pathophysiological conditions.
Collapse
Affiliation(s)
- Olha Zhenyukh
- Renal, Vascular and Diabetes Research Laboratory, Instituto de Investigación Sanitaria Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Spain and Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Spain.
| | - Esther Civantos
- Renal, Vascular and Diabetes Research Laboratory, Instituto de Investigación Sanitaria Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Spain and Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Spain.
| | - Marta Ruiz-Ortega
- Renal, Vascular and Diabetes Research Laboratory, Instituto de Investigación Sanitaria Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Spain and Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Spain.
| | | | - Clotilde Vázquez
- Division of Endocrinology, Fundación Jiménez Díaz, Madrid, Spain.
| | - Concepción Peiró
- Department of Pharmacology, Faculty of Medicine, Universidad Autónoma de Madrid, Spain.
| | - Jesús Egido
- Renal, Vascular and Diabetes Research Laboratory, Instituto de Investigación Sanitaria Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Spain and Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Spain.
| | - Sebastián Mas
- Renal, Vascular and Diabetes Research Laboratory, Instituto de Investigación Sanitaria Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Spain and Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Spain.
| |
Collapse
|
76
|
Taslimifar M, Oparija L, Verrey F, Kurtcuoglu V, Olgac U, Makrides V. Quantifying the relative contributions of different solute carriers to aggregate substrate transport. Sci Rep 2017; 7:40628. [PMID: 28091567 PMCID: PMC5238446 DOI: 10.1038/srep40628] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 12/07/2016] [Indexed: 02/07/2023] Open
Abstract
Determining the contributions of different transporter species to overall cellular transport is fundamental for understanding the physiological regulation of solutes. We calculated the relative activities of Solute Carrier (SLC) transporters using the Michaelis-Menten equation and global fitting to estimate the normalized maximum transport rate for each transporter (Vmax). Data input were the normalized measured uptake of the essential neutral amino acid (AA) L-leucine (Leu) from concentration-dependence assays performed using Xenopus laevis oocytes. Our methodology was verified by calculating Leu and L-phenylalanine (Phe) data in the presence of competitive substrates and/or inhibitors. Among 9 potentially expressed endogenous X. laevis oocyte Leu transporter species, activities of only the uniporters SLC43A2/LAT4 (and/or SLC43A1/LAT3) and the sodium symporter SLC6A19/B0AT1 were required to account for total uptake. Furthermore, Leu and Phe uptake by heterologously expressed human SLC6A14/ATB0,+ and SLC43A2/LAT4 was accurately calculated. This versatile systems biology approach is useful for analyses where the kinetics of each active protein species can be represented by the Hill equation. Furthermore, its applicable even in the absence of protein expression data. It could potentially be applied, for example, to quantify drug transporter activities in target cells to improve specificity.
Collapse
Affiliation(s)
- Mehdi Taslimifar
- The Interface Group, Institute of Physiology, University of Zurich, Switzerland.,Epithelial Transport Group, Institute of Physiology, University of Zurich, Switzerland
| | - Lalita Oparija
- Epithelial Transport Group, Institute of Physiology, University of Zurich, Switzerland.,Zurich Center for Integrative Human Physiology, University of Zurich, Switzerland
| | - Francois Verrey
- Epithelial Transport Group, Institute of Physiology, University of Zurich, Switzerland.,Zurich Center for Integrative Human Physiology, University of Zurich, Switzerland.,National Center of Competence in Research, Kidney CH, Switzerland
| | - Vartan Kurtcuoglu
- The Interface Group, Institute of Physiology, University of Zurich, Switzerland.,Zurich Center for Integrative Human Physiology, University of Zurich, Switzerland.,National Center of Competence in Research, Kidney CH, Switzerland
| | - Ufuk Olgac
- The Interface Group, Institute of Physiology, University of Zurich, Switzerland.,National Center of Competence in Research, Kidney CH, Switzerland
| | - Victoria Makrides
- Epithelial Transport Group, Institute of Physiology, University of Zurich, Switzerland
| |
Collapse
|
77
|
Hayashi K, Anzai N. Novel therapeutic approaches targeting L-type amino acid transporters for cancer treatment. World J Gastrointest Oncol 2017; 9:21-29. [PMID: 28144396 PMCID: PMC5241523 DOI: 10.4251/wjgo.v9.i1.21] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 10/08/2016] [Accepted: 11/02/2016] [Indexed: 02/05/2023] Open
Abstract
L-type amino acid transporters (LATs) mainly assist the uptake of neutral amino acids into cells. Four LATs (LAT1, LAT2, LAT3 and LAT4) have so far been identified. LAT1 (SLC7A5) has been attracting much attention in the field of cancer research since it is commonly up-regulated in various cancers. Basic research has made it increasingly clear that LAT1 plays a predominant role in malignancy. The functional significance of LAT1 in cancer and the potential therapeutic application of the features of LAT1 to cancer management are described in this review.
Collapse
|
78
|
Mastrototaro L, Sponder G, Saremi B, Aschenbach JR. Gastrointestinal methionine shuttle: Priority handling of precious goods. IUBMB Life 2016; 68:924-934. [DOI: 10.1002/iub.1571] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 09/22/2016] [Indexed: 01/05/2023]
Affiliation(s)
- Lucia Mastrototaro
- Institute of Veterinary Physiology, Department of Veterinary Medicine, Free University of Berlin; Berlin Germany
| | - Gerhard Sponder
- Institute of Veterinary Physiology, Department of Veterinary Medicine, Free University of Berlin; Berlin Germany
| | - Behnam Saremi
- Evonik Nutrition & Care GmbH; Animal Nutrition-Animal Nutrition Services; Hanau Germany
| | - Jörg R. Aschenbach
- Institute of Veterinary Physiology, Department of Veterinary Medicine, Free University of Berlin; Berlin Germany
| |
Collapse
|
79
|
Wingelhofer B, Kreis K, Mairinger S, Muchitsch V, Stanek J, Wanek T, Langer O, Kuntner C. Preloading with L-BPA, L-tyrosine and L-DOPA enhances the uptake of [ 18F]FBPA in human and mouse tumour cell lines. Appl Radiat Isot 2016; 118:67-72. [PMID: 27619946 DOI: 10.1016/j.apradiso.2016.08.026] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 07/29/2016] [Accepted: 08/31/2016] [Indexed: 11/26/2022]
Abstract
Aim of this study was to investigate if cellular [18F]FBPA uptake can be increased upon preloading with amino acids. [18F]FBPA uptake was assessed in HuH-7, CaCo-2 and B16-F1 cells pretreated with different concentrations or incubation times of L-BPA, L-tyrosine or L-DOPA. Without preloading, highest uptake of [18F]FBPA was observed in B16-F1 cells, followed by CaCo-2 cells and HuH-7 cells. In all cell lines higher [18F]FBPA accumulation (up to 1.65-fold) was obtained with increasing L-BPA, L-DOPA and L-tyrosine concentrations.
Collapse
Affiliation(s)
| | - Katharina Kreis
- Biomedical Systems, Health & Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Severin Mairinger
- Biomedical Systems, Health & Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Viktoria Muchitsch
- Biomedical Systems, Health & Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Johann Stanek
- Biomedical Systems, Health & Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Thomas Wanek
- Biomedical Systems, Health & Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Oliver Langer
- Biomedical Systems, Health & Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria; Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Claudia Kuntner
- Biomedical Systems, Health & Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria.
| |
Collapse
|
80
|
Marshall AD, van Geldermalsen M, Otte NJ, Anderson LA, Lum T, Vellozzi MA, Zhang BK, Thoeng A, Wang Q, Rasko JEJ, Holst J. LAT1 is a putative therapeutic target in endometrioid endometrial carcinoma. Int J Cancer 2016; 139:2529-39. [PMID: 27486861 DOI: 10.1002/ijc.30371] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 06/15/2016] [Accepted: 06/21/2016] [Indexed: 12/12/2022]
Abstract
l-type amino acid transporters (LAT1-4) are expressed in various cancer types and are involved in the uptake of essential amino acids such as leucine. Here we investigated the expression of LAT1-4 in endometrial adenocarcinoma and evaluated the contribution of LATs to endometrial cancer cell growth. Analysis of human gene expression data showed that all four LAT family members are expressed in endometrial adenocarcinomas. LAT1 was the most highly expressed, and showed a significant increase in both serous and endometrioid subtypes compared to normal endometrium. Endometrioid patients with the highest LAT1 levels exhibited the lowest disease-free survival. The pan-LAT inhibitor BCH led to a significant decrease in cell growth and spheroid area in four endometrial cancer cell lines tested in vitro. Knockdown of LAT1 by shRNA inhibited cell growth in HEC1A and Ishikawa cells, as well as inhibiting spheroid area in HEC1A cells. These data show that LAT1 plays an important role in regulating the uptake of essential amino acids such as leucine into endometrial cancer cells. Increased ability of BCH compared to LAT1 shRNA at inhibiting Ishikawa spheroid area suggests that other LAT family members may also contribute to cell growth. LAT1 inhibition may offer an effective therapeutic strategy in endometrial cancer patients whose tumours exhibit high LAT1 expression.
Collapse
Affiliation(s)
- Amy D Marshall
- Gene and Stem Cell Therapy Program, Centenary Institute, University of Sydney, Camperdown, New South Wales, Australia.,Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
| | - Michelle van Geldermalsen
- Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia.,Origins of Cancer Program, Centenary Institute, University of Sydney, Camperdown, New South Wales, Australia
| | - Nicholas J Otte
- Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia.,Origins of Cancer Program, Centenary Institute, University of Sydney, Camperdown, New South Wales, Australia
| | - Lyndal A Anderson
- Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia.,Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Trina Lum
- Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Melissa A Vellozzi
- Gene and Stem Cell Therapy Program, Centenary Institute, University of Sydney, Camperdown, New South Wales, Australia.,Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
| | - Blake K Zhang
- Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia.,Origins of Cancer Program, Centenary Institute, University of Sydney, Camperdown, New South Wales, Australia
| | - Annora Thoeng
- Gene and Stem Cell Therapy Program, Centenary Institute, University of Sydney, Camperdown, New South Wales, Australia.,Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
| | - Qian Wang
- Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia.,Origins of Cancer Program, Centenary Institute, University of Sydney, Camperdown, New South Wales, Australia
| | - John E J Rasko
- Gene and Stem Cell Therapy Program, Centenary Institute, University of Sydney, Camperdown, New South Wales, Australia.,Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia.,Cell and Molecular Therapies, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Jeff Holst
- Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia. .,Origins of Cancer Program, Centenary Institute, University of Sydney, Camperdown, New South Wales, Australia.
| |
Collapse
|
81
|
Huttunen KM, Gynther M, Huttunen J, Puris E, Spicer JA, Denny WA. A Selective and Slowly Reversible Inhibitor of l-Type Amino Acid Transporter 1 (LAT1) Potentiates Antiproliferative Drug Efficacy in Cancer Cells. J Med Chem 2016; 59:5740-51. [PMID: 27253989 DOI: 10.1021/acs.jmedchem.6b00190] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The l-type amino acid transporter 1 (LAT1) is a transmembrane protein carrying bulky and neutral amino acids into cells. LAT1 is overexpressed in several types of tumors, and its inhibition can result in reduced cancer cell growth. However, known LAT1 inhibitors lack selectivity over other transporters. In the present study, we designed and synthesized a novel selective LAT1 inhibitor (1), which inhibited the uptake of LAT1 substrate, l-leucin as well as cell growth. It also significantly potentiated the efficacy of bestatin and cisplatin even at low concentrations (25 μM). Inhibition was slowly reversible, as the inhibitor was able to be detached from the cell surface and blood-brain barrier. Moreover, the inhibitor was metabolically stable and selective toward LAT1. Since the inhibitor was readily accumulated into the prostate after intraperitoneal injection to the healthy mice, this compound may be a promising agent or adjuvant especially for the treatment of prostate cancer.
Collapse
Affiliation(s)
- Kristiina M Huttunen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland , P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Mikko Gynther
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland , P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Johanna Huttunen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland , P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Elena Puris
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland , P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Julie A Spicer
- Auckland Cancer Society Research Centre, The University of Auckland , Private Bag 92019, Auckland 1142, New Zealand
| | - William A Denny
- Auckland Cancer Society Research Centre, The University of Auckland , Private Bag 92019, Auckland 1142, New Zealand
| |
Collapse
|
82
|
Cheng Q, Beltran VD, Chan SMH, Brown JR, Bevington A, Herbert TP. System-L amino acid transporters play a key role in pancreatic β-cell signalling and function. J Mol Endocrinol 2016; 56:175-87. [PMID: 26647387 DOI: 10.1530/jme-15-0212] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The branched-chain amino acids (BCAA) leucine, isoleucine and valine, are essential amino acids that play a critical role in cellular signalling and metabolism. They acutely stimulate insulin secretion and activate the regulatory serine/threonine kinase mammalian target of rapamycin complex 1 (mTORC1), a kinase that promotes increased β-cell mass and function. The effects of BCAA on cellular function are dependent on their active transport into the mammalian cells via amino acid transporters and thus the expression and activity of these transporters likely influence β-cell signalling and function. In this report, we show that the System-L transporters are required for BCAA uptake into clonal β-cell lines and pancreatic islets, and that these are essential for signalling to mTORC1. Further investigation revealed that the System-L amino acid transporter 1 (LAT1) is abundantly expressed in the islets, and that knockdown of LAT1 using siRNA inhibits mTORC1 signalling, leucine-stimulated insulin secretion and islet cell proliferation. In summary, we show that the LAT1 is required for regulating β-cell signalling and function in islets and thus may be a novel pharmacological/nutritional target for the treatment and prevention of type 2 diabetes.
Collapse
Affiliation(s)
- Qi Cheng
- Research School of BiologyAustralian National University, Acton, Australia
| | - Violeta D Beltran
- Department of InfectionImmunity and Inflammation, University of Leicester, UK
| | - Stanley M H Chan
- Health Innovations Research Institute (HIRI)School of Medical Sciences, RMIT University, Bundoora, Australia
| | - Jeremy R Brown
- Department of InfectionImmunity and Inflammation, University of Leicester, UK
| | - Alan Bevington
- Department of InfectionImmunity and Inflammation, University of Leicester, UK
| | - Terence P Herbert
- Health Innovations Research Institute (HIRI)School of Medical Sciences, RMIT University, Bundoora, Australia
| |
Collapse
|
83
|
Yang C, Yang X, Lackeyram D, Rideout TC, Wang Z, Stoll B, Yin Y, Burrin DG, Fan MZ. Expression of apical Na(+)-L-glutamine co-transport activity, B(0)-system neutral amino acid co-transporter (B(0)AT1) and angiotensin-converting enzyme 2 along the jejunal crypt-villus axis in young pigs fed a liquid formula. Amino Acids 2016; 48:1491-508. [PMID: 26984322 DOI: 10.1007/s00726-016-2210-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 02/29/2016] [Indexed: 01/11/2023]
Abstract
Gut apical amino acid (AA) transport activity is high at birth and during suckling, thus being essential to maintain luminal nutrient-dependent mucosal growth through providing AA as essential metabolic fuel, substrates and nutrient stimuli for cellular growth. Because system-B(0) Na(+)-neutral AA co-transporter (B(0)AT1, encoded by the SLC6A19 gene) plays a dominant role for apical uptake of large neutral AA including L-Gln, we hypothesized that high apical Na(+)-Gln co-transport activity, and B(0)AT1 (SLC6A19) in co-expression with angiotensin-converting enzyme 2 (ACE2) were expressed along the entire small intestinal crypt-villus axis in young animals via unique control mechanisms. Kinetics of Na(+)-Gln co-transport activity in the apical membrane vesicles, prepared from epithelial cells sequentially isolated along the jejunal crypt-villus axis from liquid formula-fed young pigs, were measured with the membrane potential being clamped to zero using thiocyanate. Apical maximal Na(+)-Gln co-transport activity was much higher (p < 0.05) in the upper villus cells than in the middle villus (by 29 %) and the crypt (by 30 %) cells, whereas Na(+)-Gln co-transport affinity was lower (p < 0.05) in the upper villus cells than in the middle villus and the crypt cells. The B(0)AT1 (SLC6A19) mRNA abundance was lower (p < 0.05) in the crypt (by 40-47 %) than in the villus cells. There were no significant differences in B(0)AT1 and ACE2 protein abundances on the apical membrane among the upper villus, the middle villus and the crypt cells. Our study suggests that piglet fast growth is associated with very high intestinal apical Na(+)-neutral AA uptake activities via abundantly co-expressing B(0)AT1 and ACE2 proteins in the apical membrane and by transcribing the B(0)AT1 (SLC6A19) gene in the epithelia along the entire crypt-villus axis.
Collapse
Affiliation(s)
- Chengbo Yang
- Department of Animal Biosciences, University of Guelph, Guelph, ON, N1G 2W1, Canada. .,Department of Animal Science, Faculty of Agricultural and Food Sciences, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada.
| | - Xiaojian Yang
- Department of Animal Biosciences, University of Guelph, Guelph, ON, N1G 2W1, Canada.,Southern Research and Outreach Center, University of Minnesota, Waseca, MN, 56093, USA
| | - Dale Lackeyram
- Department of Animal Biosciences, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Todd C Rideout
- Department of Animal Biosciences, University of Guelph, Guelph, ON, N1G 2W1, Canada.,Department of Exercise and Nutrition Sciences, the State University of New York at Buffalo, New York, 14214, USA
| | - Zirong Wang
- College of Animal Science, Xinjiang Agricultural University, Urumqi, 830052, Xinjiang, China
| | - Barbara Stoll
- US Department of Agriculture/Agricultural Research Service, Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Yulong Yin
- Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, 410125, Hunan, China
| | - Douglas G Burrin
- US Department of Agriculture/Agricultural Research Service, Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Ming Z Fan
- Department of Animal Biosciences, University of Guelph, Guelph, ON, N1G 2W1, Canada.
| |
Collapse
|
84
|
Li Y, Wei H, Li F, Chen S, Duan Y, Guo Q, Liu Y, Yin Y. Supplementation of branched-chain amino acids in protein-restricted diets modulates the expression levels of amino acid transporters and energy metabolism associated regulators in the adipose tissue of growing pigs. ACTA ACUST UNITED AC 2016; 2:24-32. [PMID: 29767034 PMCID: PMC5940986 DOI: 10.1016/j.aninu.2016.01.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Revised: 01/29/2016] [Accepted: 01/31/2016] [Indexed: 12/17/2022]
Abstract
This experiment was conducted to investigate the effects of branched-chain amino acids (BCAA) supplemented in protein-restricted diets on the growth performance and the expression profile of amino acid transporters and energy metabolism related regulators in the white adipose tissue (WAT) of different regional depots including dorsal subcutaneous adipose (DSA) and abdominal subcutaneous adipose (ASA). A total of 24 crossbred barrows (7.40 ± 0.70 kg) were randomly divided into 4 groups and were fed the following isocaloric diets for 33 days: 1) a recommended adequate protein diet (AP, 20% CP, as a positive control); 2) a low protein diet (LP, 17% CP); 3) the LP diet supplemented with BCAA (LP + B, 17% CP) to reach the same level of the AP diet group; 4) the LP diet supplemented with 2 times the amount of BCAA (LP + 2B, 17% CP). The daily gain and daily feed intake of the LP diet group were the lowest among all the treatments (P < 0.01). The feed conversion was improved markedly in the group of LP + B compared with the LP diet group (P < 0.05). No significant difference was noted for the serum biochemical parameter concentrations of glucose, triglyceride, nonesterified fatty acid and insulin among the groups (P > 0.05). Moreover, BCAA supplementation down-regulated the expression levels of amino acid transporters including L-type amino acid transporter 1 and sodium-coupled neutral amino acid transporter 2 in DSA, but up-regulated the expression level of L-type amino acid transporter 4 in ASA (P < 0.05). Meanwhile, the energy sensor AMP-activated protein kinase α was activated in the DSA of pigs fed LP diet and in the ASA of the pigs fed AP or LP + 2B diets (P < 0.05). The mRNA expression profile of the selected mitochondrial component and mitochondrial biogenesis associated regulators in DSA and ASA also responded differently to dietary BCAA supplementation. These results suggested that the growth performance of growing pigs fed protein restricted diets supplemented with BCAA could catch up to that of the pigs fed AP diets. The results also partly demonstrated that the regulation mechanisms of BCAA are different in the adipose tissues of different depots.
Collapse
Affiliation(s)
- Yinghui Li
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center of Healthy Livestock, Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- University of Chinese Academy of Sciences, Beijing 100039, China
| | - Hongkui Wei
- College of Animal Sciences, Huazhong Agricultural University, Wuhan 430070, China
| | - Fengna Li
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center of Healthy Livestock, Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- Hunan Co-Innovation Center of Animal Production Safety (CICAPS), Changsha 410128, China
- Corresponding authors.
| | - Shuai Chen
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center of Healthy Livestock, Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- University of Chinese Academy of Sciences, Beijing 100039, China
| | - Yehui Duan
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center of Healthy Livestock, Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- University of Chinese Academy of Sciences, Beijing 100039, China
| | - Qiuping Guo
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center of Healthy Livestock, Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- University of Chinese Academy of Sciences, Beijing 100039, China
| | - Yingying Liu
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center of Healthy Livestock, Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- University of Chinese Academy of Sciences, Beijing 100039, China
| | - Yulong Yin
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center of Healthy Livestock, Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- Changsha Lvye Biotechnology Limited Company Academician Expert Workstation, Changsha 410126, China
- Hang Zhou King Techina Limited Company Academician Expert Workstation, Hangzhou 311107, China
- Corresponding authors.
| |
Collapse
|
85
|
Zevenbergen C, Meima ME, Lima de Souza EC, Peeters RP, Kinne A, Krause G, Visser WE, Visser TJ. Transport of Iodothyronines by Human L-Type Amino Acid Transporters. Endocrinology 2015; 156:4345-55. [PMID: 26305885 DOI: 10.1210/en.2015-1140] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Thyroid hormone (TH) transporters facilitate cellular TH influx and efflux, which is paramount for normal physiology. The L-type amino acid transporters LAT1 and LAT2 are known to facilitate TH transport. However, the role of LAT3, LAT4, and LAT5 is still unclear. Therefore, the aim of this study was to further characterize TH transport by LAT1 and LAT2 and to explore possible TH transport by LAT3, LAT4, and LAT5. FLAG-LAT1-5 constructs were transiently expressed in COS1 cells. LAT1 and LAT2 were cotransfected with the CD98 heavy chain. Cellular transport was measured using 10 nM (125)I-labeled T4, T3, rT3, 3,3'-T2, and 10 μM [(125)I]3'-iodotyrosine (MIT) as substrates. Intracellular metabolism of these substrates was determined in cells cotransfected with either of the LATs with type 1 or type 3 deiodinase. LAT1 facilitated cellular uptake of all substrates and LAT2 showed a net uptake of T3, 3,3'-T2, and MIT. Expression of LAT3 or LAT4 did not affect transport of T4 and T3 but resulted in the decreased cellular accumulation of 3,3'-T2 and MIT. LAT5 did not facilitate the transport of any substrate. Cotransfection with LAT3 or LAT4 strongly diminished the cellular accumulation of 3,3'-T2 and MIT by LAT1 and LAT2. These data were confirmed by metabolism studies. LAT1 and LAT2 show distinct preferences for the uptake of the different iodocompounds, whereas LAT3 and LAT4 specifically facilitate the 3,3'-T2 and MIT efflux. Together our findings suggest that different sets of transporters with specific influx or efflux capacities may cooperate to regulate the cellular thyroid state.
Collapse
Affiliation(s)
- Chantal Zevenbergen
- Department of Internal Medicine and Rotterdam Thyroid Center (C.Z., M.E.M., E.C.L.d.S., R.P.P., W.E.V., T.J.V.), Erasmus University Medical Center, 3015 CN, Rotterdam, The Netherlands; and Department of Nuclear Magnetic Resonance-Supported Structural Biology (A.K., G.K.), Leibniz-Institut für Molekulare Pharmakologie, 13125 Berlin, Germany
| | - Marcel E Meima
- Department of Internal Medicine and Rotterdam Thyroid Center (C.Z., M.E.M., E.C.L.d.S., R.P.P., W.E.V., T.J.V.), Erasmus University Medical Center, 3015 CN, Rotterdam, The Netherlands; and Department of Nuclear Magnetic Resonance-Supported Structural Biology (A.K., G.K.), Leibniz-Institut für Molekulare Pharmakologie, 13125 Berlin, Germany
| | - Elaine C Lima de Souza
- Department of Internal Medicine and Rotterdam Thyroid Center (C.Z., M.E.M., E.C.L.d.S., R.P.P., W.E.V., T.J.V.), Erasmus University Medical Center, 3015 CN, Rotterdam, The Netherlands; and Department of Nuclear Magnetic Resonance-Supported Structural Biology (A.K., G.K.), Leibniz-Institut für Molekulare Pharmakologie, 13125 Berlin, Germany
| | - Robin P Peeters
- Department of Internal Medicine and Rotterdam Thyroid Center (C.Z., M.E.M., E.C.L.d.S., R.P.P., W.E.V., T.J.V.), Erasmus University Medical Center, 3015 CN, Rotterdam, The Netherlands; and Department of Nuclear Magnetic Resonance-Supported Structural Biology (A.K., G.K.), Leibniz-Institut für Molekulare Pharmakologie, 13125 Berlin, Germany
| | - Anita Kinne
- Department of Internal Medicine and Rotterdam Thyroid Center (C.Z., M.E.M., E.C.L.d.S., R.P.P., W.E.V., T.J.V.), Erasmus University Medical Center, 3015 CN, Rotterdam, The Netherlands; and Department of Nuclear Magnetic Resonance-Supported Structural Biology (A.K., G.K.), Leibniz-Institut für Molekulare Pharmakologie, 13125 Berlin, Germany
| | - Gerd Krause
- Department of Internal Medicine and Rotterdam Thyroid Center (C.Z., M.E.M., E.C.L.d.S., R.P.P., W.E.V., T.J.V.), Erasmus University Medical Center, 3015 CN, Rotterdam, The Netherlands; and Department of Nuclear Magnetic Resonance-Supported Structural Biology (A.K., G.K.), Leibniz-Institut für Molekulare Pharmakologie, 13125 Berlin, Germany
| | - W Edward Visser
- Department of Internal Medicine and Rotterdam Thyroid Center (C.Z., M.E.M., E.C.L.d.S., R.P.P., W.E.V., T.J.V.), Erasmus University Medical Center, 3015 CN, Rotterdam, The Netherlands; and Department of Nuclear Magnetic Resonance-Supported Structural Biology (A.K., G.K.), Leibniz-Institut für Molekulare Pharmakologie, 13125 Berlin, Germany
| | - Theo J Visser
- Department of Internal Medicine and Rotterdam Thyroid Center (C.Z., M.E.M., E.C.L.d.S., R.P.P., W.E.V., T.J.V.), Erasmus University Medical Center, 3015 CN, Rotterdam, The Netherlands; and Department of Nuclear Magnetic Resonance-Supported Structural Biology (A.K., G.K.), Leibniz-Institut für Molekulare Pharmakologie, 13125 Berlin, Germany
| |
Collapse
|
86
|
Furukawa J, Inoue K, Maeda J, Yasujima T, Ohta K, Kanai Y, Takada T, Matsuo H, Yuasa H. Functional identification of SLC43A3 as an equilibrative nucleobase transporter involved in purine salvage in mammals. Sci Rep 2015; 5:15057. [PMID: 26455426 PMCID: PMC4796657 DOI: 10.1038/srep15057] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 09/16/2015] [Indexed: 12/25/2022] Open
Abstract
The purine salvage pathway plays a major role in the nucleotide production, relying on the supply of nucleobases and nucleosides from extracellular sources. Although specific transporters have been suggested to be involved in facilitating their transport across the plasma membrane in mammals, those which are specifically responsible for utilization of extracellular nucleobases remain unknown. Here we present the molecular and functional characterization of SLC43A3, an orphan transporter belonging to an amino acid transporter family, as a purine-selective nucleobase transporter. SLC43A3 was highly expressed in the liver, where it was localized to the sinusoidal membrane of hepatocytes, and the lung. In addition, SLC43A3 expressed in MDCKII cells mediated the uptake of purine nucleobases such as adenine, guanine, and hypoxanthine without requiring typical driving ions such as Na(+) and H(+), but it did not mediate the uptake of nucleosides. When SLC43A3 was expressed in APRT/HPRT1-deficient A9 cells, adenine uptake was found to be low. However, it was markedly enhanced by the introduction of SLC43A3 with APRT. In HeLa cells, knock-down of SLC43A3 markedly decreased adenine uptake. These data suggest that SLC43A3 is a facilitative and purine-selective nucleobase transporter that mediates the cellular uptake of extracellular purine nucleobases in cooperation with salvage enzymes.
Collapse
Affiliation(s)
- Junji Furukawa
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Katsuhisa Inoue
- Department of Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Junya Maeda
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Tomoya Yasujima
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Kinya Ohta
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Yoshikatsu Kanai
- Division of Bio-system Pharmacology, Department of Pharmacology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Tappei Takada
- Department of Pharmacy, The University of Tokyo Hospital, Tokyo, Japan
| | - Hirotaka Matsuo
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Saitama, Japan
| | - Hiroaki Yuasa
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| |
Collapse
|
87
|
Yamane S, Nomura R, Yanagihara M, Nakamura H, Fujino H, Matsumoto K, Horie S, Murayama T. L-cysteine/d,L-homocysteine-regulated ileum motility via system L and B°,+ transporter: Modification by inhibitors of hydrogen sulfide synthesis and dietary treatments. Eur J Pharmacol 2015. [DOI: 10.1016/j.ejphar.2015.07.042] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
88
|
Glycosylation of solute carriers: mechanisms and functional consequences. Pflugers Arch 2015; 468:159-76. [PMID: 26383868 DOI: 10.1007/s00424-015-1730-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 08/19/2015] [Accepted: 08/21/2015] [Indexed: 12/21/2022]
Abstract
Solute carriers (SLCs) are one of the largest groups of multi-spanning membrane proteins in mammals and include ubiquitously expressed proteins as well as proteins with highly restricted tissue expression. A vast number of studies have addressed the function and organization of SLCs as well as their posttranslational regulation, but only relatively little is known about the role of SLC glycosylation. Glycosylation is one of the most abundant posttranslational modifications of animal proteins and through recent advances in our understanding of protein-glycan interactions, the functional roles of SLC glycosylation are slowly emerging. The purpose of this review is to provide a concise overview of the aspects of glycobiology most relevant to SLCs, to discuss the roles of glycosylation in the regulation and function of SLCs, and to outline the major open questions in this field, which can now be addressed given major technical advances in this and related fields of study in recent years.
Collapse
|
89
|
Preclinical evaluation of 2-amino-2-[11C]methyl-butanoic acid as a potential tumor-imaging agent in a mouse model. Nucl Med Commun 2015; 36:1107-12. [PMID: 26259115 DOI: 10.1097/mnm.0000000000000364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE C-labeled 2-amino-2-methyl-butanoic acid (Iva) was previously reported to provide high tumor uptake; however, the pharmacokinetic properties of C-labeled Iva have not been characterized. In the present study, we evaluated the potential of [C]Iva as a PET probe for tumor imaging. METHODS [C]Iva was incubated in mouse serum for 60 min at 37°C and then analyzed by high-performance liquid chromatography and thin-layer chromatography. In-vitro cellular uptake of [C]Iva was determined in PBS and sodium-free buffer at 37°C using SY human small-cell lung cancer cells. The effects of inhibitors of amino acid transporters on [C]Iva uptake were also determined in PBS. In-vivo distribution and dynamic PET studies were conducted in SY tumor-bearing mice. RESULTS [C]Iva was stable in mouse serum in vitro for 60 min. The cellular uptake of [C]Iva was linearly increased for 20 min in both PBS and sodium-free buffer and almost completely inhibited by an inhibitor of system L amino acid transporters and another of LAT1, a transporter of system L. In-vivo distribution and dynamic PET studies showed that [C]Iva was highly accumulated in tumor, but not in normal tissues, except for the pancreas and kidneys. The [C]Iva uptake ratio of tumor to several normal tissues, such as the lung, muscle, and brain, was high. CONCLUSION [C]Iva was stable in mouse serum and transported through system L amino acid transporters including LAT1, which is highly expressed in several tumors. [C]Iva is a promising PET probe for noninvasive tumor imaging.
Collapse
|
90
|
Zhen H, Nakamura K, Kitaura Y, Kadota Y, Ishikawa T, Kondo Y, Xu M, Shimomura Y. Regulation of the plasma amino acid profile by leucine via the system L amino acid transporter. Biosci Biotechnol Biochem 2015; 79:2057-62. [PMID: 26125295 DOI: 10.1080/09168451.2015.1060845] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Plasma concentrations of amino acids reflect the intracellular amino acid pool in mammals. However, the regulatory mechanism requires clarification. In this study, we examined the effect of leucine administration on plasma amino acid profiles in mice with and without the treatment of 2-aminobicyclo-(2,2,1)-heptane-2-carboxylic acid (BCH) or rapamycin as an inhibitor of system L or mammalian target of rapamycin complex 1, respectively. The elevation of plasma leucine concentration after leucine administration was associated with a significant decrease in the plasma concentrations of isoleucine, valine, methionine, phenylalanine, and tyrosine; BCH treatment almost completely blocked the leucine-induced decrease in plasma amino acid concentrations. Rapamycin treatment had much less effects on the actions of leucine than BCH treatment. These results suggest that leucine regulates the plasma concentrations of branched-chain amino acids, methionine, phenylalanine, and tyrosine, and that system L amino acid transporters are involved in the leucine action.
Collapse
Affiliation(s)
- Hongmin Zhen
- a Laboratory of Nutritional Biochemistry, Department of Applied Molecular Biosciences , Graduate School of Bioagricultural Sciences, Nagoya University , Nagoya , Japan
| | - Koichi Nakamura
- a Laboratory of Nutritional Biochemistry, Department of Applied Molecular Biosciences , Graduate School of Bioagricultural Sciences, Nagoya University , Nagoya , Japan
| | - Yasuyuki Kitaura
- a Laboratory of Nutritional Biochemistry, Department of Applied Molecular Biosciences , Graduate School of Bioagricultural Sciences, Nagoya University , Nagoya , Japan
| | - Yoshihiro Kadota
- a Laboratory of Nutritional Biochemistry, Department of Applied Molecular Biosciences , Graduate School of Bioagricultural Sciences, Nagoya University , Nagoya , Japan
| | - Takuya Ishikawa
- a Laboratory of Nutritional Biochemistry, Department of Applied Molecular Biosciences , Graduate School of Bioagricultural Sciences, Nagoya University , Nagoya , Japan
| | - Yusuke Kondo
- a Laboratory of Nutritional Biochemistry, Department of Applied Molecular Biosciences , Graduate School of Bioagricultural Sciences, Nagoya University , Nagoya , Japan
| | - Minjun Xu
- a Laboratory of Nutritional Biochemistry, Department of Applied Molecular Biosciences , Graduate School of Bioagricultural Sciences, Nagoya University , Nagoya , Japan
| | - Yoshiharu Shimomura
- a Laboratory of Nutritional Biochemistry, Department of Applied Molecular Biosciences , Graduate School of Bioagricultural Sciences, Nagoya University , Nagoya , Japan
| |
Collapse
|
91
|
Vogel KR, Ainslie GR, Phillips B, Arning E, Bottiglieri T, Shen DD, Gibson KM. Physiological Competition of Brain Phenylalanine Accretion: Initial Pharmacokinetic Analyses of Aminoisobutyric and Methylaminoisobutyric Acids in Pahenu2-/- Mice. Mol Genet Metab Rep 2015; 3:80-87. [PMID: 26120559 PMCID: PMC4478616 DOI: 10.1016/j.ymgmr.2015.03.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Objective Initial studies on the use of non-physiological amino acids (NPAAs) to block the accretion of Phe in the brain of Pahenu2 −/− mice revealed that 2-aminoisobutyrate (AIB) and N-methyl-2-aminoisobutyrate (MAIB) were promising lead compounds whose pharmacokinetic parameters warranted investigation. Methods Control and Pahenu2 −/− mice received intraperitoneal NPAA treatments as test compounds (150, 300 and 500 mg/kg, 1 or 7 days) followed by collection of sera, liver and brain. LC–MS analysis was developed to quantify both AIB and MAIB in all matrices, and pharmacokinetic parameters for distribution, partitioning, accumulation and MAIB demethylation were determined. Results MAIB was partially converted to AIB in vivo. AIB and MAIB partitioned similarly from sera to the brain and liver, with an approximate 10-fold higher accumulation in the liver compared to the brain. In comparison to MAIB, AIB accumulated to approximately 3 to 7-fold higher concentration in the brain. Analysis of the brain and liver revealed a trend toward decreased Phe with increased MAIB serum concentration. Conclusions Our data support further pharmacokinetic characterization of MAIB and AIB in preparation for additional preclinical safety, toxicity and tolerability studies of both AIB and MAIB.
Collapse
Affiliation(s)
- Kara R Vogel
- Section of Experimental and Systems Pharmacology, College of Pharmacy, Washington State University, Spokane, WA
| | - Garrett R Ainslie
- Section of Experimental and Systems Pharmacology, College of Pharmacy, Washington State University, Spokane, WA
| | - Brian Phillips
- Departments of Pharmaceutics and Pharmacy, University of Washington School of Pharmacy, Seattle, WA
| | - Erland Arning
- Institute for Metabolic Disease, Baylor Research Institute, Dallas, TX
| | | | - Danny D Shen
- Departments of Pharmaceutics and Pharmacy, University of Washington School of Pharmacy, Seattle, WA
| | - K Michael Gibson
- Section of Experimental and Systems Pharmacology, College of Pharmacy, Washington State University, Spokane, WA
| |
Collapse
|
92
|
Chung J, Bauer DE, Ghamari A, Nizzi CP, Deck KM, Kingsley PD, Yien YY, Huston NC, Chen C, Schultz IJ, Dalton AJ, Wittig JG, Palis J, Orkin SH, Lodish HF, Eisenstein RS, Cantor AB, Paw BH. The mTORC1/4E-BP pathway coordinates hemoglobin production with L-leucine availability. Sci Signal 2015; 8:ra34. [PMID: 25872869 PMCID: PMC4402725 DOI: 10.1126/scisignal.aaa5903] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
In multicellular organisms, the mechanisms by which diverse cell types acquire distinct amino acids and how cellular function adapts to their availability are fundamental questions in biology. We found that increased neutral essential amino acid (NEAA) uptake was a critical component of erythropoiesis. As red blood cells matured, expression of the amino acid transporter gene Lat3 increased, which increased NEAA import. Inadequate NEAA uptake by pharmacologic inhibition or RNAi-mediated knockdown of LAT3 triggered a specific reduction in hemoglobin production in zebrafish embryos and murine erythroid cells through the mTORC1 (mammalian target of rapamycin complex 1)/4E-BP (eukaryotic translation initiation factor 4E-binding protein) pathway. CRISPR-mediated deletion of members of the 4E-BP family in murine erythroid cells rendered them resistant to mTORC1 and LAT3 inhibition and restored hemoglobin production. These results identify a developmental role for LAT3 in red blood cells and demonstrate that mTORC1 serves as a homeostatic sensor that couples hemoglobin production at the translational level to sufficient uptake of NEAAs, particularly L-leucine.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing
- Amino Acid Transport Systems, Basic/genetics
- Amino Acid Transport Systems, Basic/metabolism
- Animals
- Animals, Genetically Modified
- CRISPR-Cas Systems
- Carrier Proteins/genetics
- Carrier Proteins/metabolism
- Cell Cycle Proteins
- Cell Line, Tumor
- Cells, Cultured
- Embryo, Mammalian/blood supply
- Embryo, Mammalian/embryology
- Embryo, Mammalian/metabolism
- Embryo, Nonmammalian/embryology
- Embryo, Nonmammalian/metabolism
- Erythroid Cells/metabolism
- Erythropoiesis/genetics
- Eukaryotic Initiation Factors/genetics
- Eukaryotic Initiation Factors/metabolism
- Gene Expression Regulation, Developmental
- HEK293 Cells
- Hemoglobins/genetics
- Hemoglobins/metabolism
- Humans
- Immunoblotting
- Leucine/metabolism
- Mechanistic Target of Rapamycin Complex 1
- Mice
- Microscopy, Confocal
- Multiprotein Complexes/genetics
- Multiprotein Complexes/metabolism
- Phosphoproteins/genetics
- Phosphoproteins/metabolism
- RNA Interference
- Reverse Transcriptase Polymerase Chain Reaction
- Signal Transduction/genetics
- TOR Serine-Threonine Kinases/genetics
- TOR Serine-Threonine Kinases/metabolism
- Zebrafish
Collapse
Affiliation(s)
- Jacky Chung
- Division of Hematology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Daniel E Bauer
- Division of Hematology-Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA. Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Alireza Ghamari
- Division of Hematology-Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA. Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Christopher P Nizzi
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Kathryn M Deck
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Paul D Kingsley
- Department of Pediatrics, Center for Pediatric Biomedical Research, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Yvette Y Yien
- Division of Hematology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Nicholas C Huston
- Division of Hematology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Caiyong Chen
- Division of Hematology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Iman J Schultz
- Division of Hematology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Arthur J Dalton
- Division of Hematology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Johannes G Wittig
- Division of Hematology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - James Palis
- Department of Pediatrics, Center for Pediatric Biomedical Research, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Stuart H Orkin
- Division of Hematology-Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA. Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Harvey F Lodish
- Whitehead Institute for Biomedical Research, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Richard S Eisenstein
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Alan B Cantor
- Division of Hematology-Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA. Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Barry H Paw
- Division of Hematology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA. Division of Hematology-Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA. Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
93
|
Juhász C, Dwivedi S, Kamson DO, Michelhaugh SK, Mittal S. Comparison of amino acid positron emission tomographic radiotracers for molecular imaging of primary and metastatic brain tumors. Mol Imaging 2015; 13. [PMID: 24825818 DOI: 10.2310/7290.2014.00015] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Positron emission tomography (PET) is an imaging technology that can detect and characterize tumors based on their molecular and biochemical properties, such as altered glucose, nucleoside, or amino acid metabolism. PET plays a significant role in the diagnosis, prognostication, and treatment of various cancers, including brain tumors. In this article, we compare uptake mechanisms and the clinical performance of the amino acid PET radiotracers (l-[methyl-11C]methionine [MET], 18F-fluoroethyl-tyrosine [FET], 18F-fluoro-l-dihydroxy-phenylalanine [FDOPA], and 11C-alpha-methyl-l-tryptophan [AMT]) most commonly used for brain tumor imaging. First, we discuss and compare the mechanisms of tumoral transport and accumulation, the basis of differential performance of these radioligands in clinical studies. Then we summarize studies that provided direct comparisons among these amino acid tracers and to clinically used 2-deoxy-2[18F]fluoro-d-glucose [FDG] PET imaging. We also discuss how tracer kinetic analysis can enhance the clinical information obtained from amino acid PET images. We discuss both similarities and differences in potential clinical value for each radioligand. This comparative review can guide which radiotracer to favor in future clinical trials aimed at defining the role of these molecular imaging modalities in the clinical management of brain tumor patients.
Collapse
|
94
|
Bae JH, Kim JG, Heo K, Yang K, Kim TO, Yi JM. Identification of radiation-induced aberrant hypomethylation in colon cancer. BMC Genomics 2015; 16:56. [PMID: 25887185 PMCID: PMC4342812 DOI: 10.1186/s12864-015-1229-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2014] [Accepted: 01/09/2015] [Indexed: 12/22/2022] Open
Abstract
Background Exposure to ionizing radiation (IR) results in the simultaneous activation or downregulation of multiple signaling pathways that play critical roles in cell type-specific control of survival or death. IR is a well-known genotoxic agent and human carcinogen that induces cellular damage through direct and indirect mechanisms. However, its impact on epigenetic mechanisms has not been elucidated, and more specifically, little information is available regarding genome-wide DNA methylation changes in cancer cells after IR exposure. Recently, genome-wide DNA methylation profiling technology using the Illumina HumanMethylation450K platform has emerged that allows us to query >450,000 loci within the genome. This improved technology is capable of identifying genome-wide DNA methylation changes in CpG islands and other CpG island-associated regions. Results In this study, we employed this technology to test the hypothesis that exposure to IR not only induces differential DNA methylation patterns at a genome-wide level, but also results in locus- and gene-specific DNA methylation changes. We screened for differential DNA methylation changes in colorectal cancer cells after IR exposure with 2 and 5 Gy. Twenty-nine genes showed radiation-induced hypomethylation in colon cancer cells, and of those, seven genes showed a corresponding increase in gene expression by reverse transcriptase polymerase chain reaction (RT-PCR). In addition, we performed chromatin immunoprecipitation (ChIP) to confirm that the DNA-methyltransferase 1 (DNMT1) level associated with the promoter regions of these genes correlated with their methylation level and gene expression changes. Finally, we used a gene ontology (GO) database to show that a handful of hypomethylated genes induced by IR are associated with a variety of biological pathways related to cancer. Conclusion We identified alterations in global DNA methylation patterns and hypomethylation at specific cancer-related genes following IR exposure, which suggests that radiation exposure plays a critical role in conferring epigenetic alterations in cancer. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-1229-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jin-Han Bae
- Research Center, Dongnam Institute of Radiological & Medical Sciences (DIRAMS), Busan, 619-953, South Korea.
| | - Joong-Gook Kim
- Research Center, Dongnam Institute of Radiological & Medical Sciences (DIRAMS), Busan, 619-953, South Korea.
| | - Kyu Heo
- Research Center, Dongnam Institute of Radiological & Medical Sciences (DIRAMS), Busan, 619-953, South Korea.
| | - Kwangmo Yang
- Research Center, Dongnam Institute of Radiological & Medical Sciences (DIRAMS), Busan, 619-953, South Korea. .,Department of Radiation Oncology, Korea Institute of Radiological and Medical Sciences, Seoul, 139-709, Korea.
| | - Tae-Oh Kim
- Department of Internal Medicine, Inje University Haeundae Paik hospital, Busan, 612-896, South Korea.
| | - Joo Mi Yi
- Research Center, Dongnam Institute of Radiological & Medical Sciences (DIRAMS), Busan, 619-953, South Korea.
| |
Collapse
|
95
|
Ogihara K, Naya Y, Sato R, Onda K, Ochiai H. Analysis of L-type amino acid transporter in canine hepatocellular carcinoma. J Vet Med Sci 2015; 77:527-34. [PMID: 25649314 PMCID: PMC4478731 DOI: 10.1292/jvms.14-0392] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Analysis of L-type amino acid transport expression of hepatocellular carcinoma cells
(HCCs) of the dog was performed. The leucine transport activity of canine HCCs was 0.628 ±
0.018 nmol/mg protein/min. The inhibitor of LAT 2-aminobicyclo[2.2.1]heptane-2-carboxylic
acid (BCH) reduced 90% of the activity at 1 mM. The deduced amino acid sequences of canine
LAT2, LAT3 and LAT4 were well conserved in mammalians, exhibiting 89, 88 and 77% homology,
respectively. RT-PCR revealed distinct LAT1 expression compared with normal hepatocytes.
Western blotting analysis confirmed the potent LAT1 expression in canine HCCs but not
hepatocytes, and real-time RT-PCR analysis indicated that canine HCCs possessed 28 times
higher LAT1 expression than hepatocytes. These results indicated that the leucine
transport activity of canine HCCs was due to LAT1.
Collapse
Affiliation(s)
- Kikumi Ogihara
- Laboratory of Pathology, School of Veterinary Medicine, Azabu University, 1-17-71 Fuchinobe, Chuo-ku, Sagamihara, Kanagawa 252-5201, Japan
| | | | | | | | | |
Collapse
|
96
|
Guetg A, Mariotta L, Bock L, Herzog B, Fingerhut R, Camargo SMR, Verrey F. Essential amino acid transporter Lat4 (Slc43a2) is required for mouse development. J Physiol 2015; 593:1273-89. [PMID: 25480797 DOI: 10.1113/jphysiol.2014.283960] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 11/25/2014] [Indexed: 12/23/2022] Open
Abstract
Amino acid (AA) uniporter Lat4 (Slc43a2) mediates facilitated diffusion of branched-chain AAs, methionine and phenylalanine, although its physiological role and subcellular localization are not known. We report that Slc43a2 knockout mice were born at expected Mendelian frequency but displayed an ∼10% intrauterine growth retardation and low amniotic fluid AAs, suggesting defective transplacental transport. Postnatal growth was strongly reduced, with premature death occurring within 9 days such that further investigations were made within 3 days of birth. Lat4 immunofluorescence showed a strong basolateral signal in the small intestine, kidney proximal tubule and thick ascending limb epithelial cells of wild-type but not Slc43a2 null littermates and no signal in liver and skeletal muscle. Experiments using Xenopus laevis oocytes demonstrated that Lat4 functioned as a symmetrical low affinity uniporter with a K₀.₅ of ∼5 mm for both in- and efflux. Plasma AA concentration was decreased in Slc43a2 null pups, in particular that of non-essential AAs alanine, serine, histidine and proline. Together with an increased level of plasma long chain acylcarnitines and a strong alteration of liver gene expression, this indicates malnutrition. Attempts to rescue pups by decreasing the litter size or by nutrients injected i.p. did not succeed. Radioactively labelled leucine but not lysine given per os accumulated in the small intestine of Slc43a2null pups, suggesting the defective transcellular transport of Lat4 substrates. In summary, Lat4 is a symmetrical uniporter for neutral essential AAs localizing at the basolateral side of (re)absorbing epithelia and is necessary for early nutrition and development.
Collapse
Affiliation(s)
- Adriano Guetg
- Institute of Physiology and Zurich Center of Integrative Human Physiology, University of Zurich, Switzerland
| | | | | | | | | | | | | |
Collapse
|
97
|
Zhang S, Regnault TRH, Barker PL, Botting KJ, McMillen IC, McMillan CM, Roberts CT, Morrison JL. Placental adaptations in growth restriction. Nutrients 2015; 7:360-89. [PMID: 25580812 PMCID: PMC4303845 DOI: 10.3390/nu7010360] [Citation(s) in RCA: 140] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 12/22/2014] [Indexed: 12/17/2022] Open
Abstract
The placenta is the primary interface between the fetus and mother and plays an important role in maintaining fetal development and growth by facilitating the transfer of substrates and participating in modulating the maternal immune response to prevent immunological rejection of the conceptus. The major substrates required for fetal growth include oxygen, glucose, amino acids and fatty acids, and their transport processes depend on morphological characteristics of the placenta, such as placental size, morphology, blood flow and vascularity. Other factors including insulin-like growth factors, apoptosis, autophagy and glucocorticoid exposure also affect placental growth and substrate transport capacity. Intrauterine growth restriction (IUGR) is often a consequence of insufficiency, and is associated with a high incidence of perinatal morbidity and mortality, as well as increased risk of cardiovascular and metabolic diseases in later life. Several different experimental methods have been used to induce placental insufficiency and IUGR in animal models and a range of factors that regulate placental growth and substrate transport capacity have been demonstrated. While no model system completely recapitulates human IUGR, these animal models allow us to carefully dissect cellular and molecular mechanisms to improve our understanding and facilitate development of therapeutic interventions.
Collapse
Affiliation(s)
- Song Zhang
- Early Origins of Adult Health Research Group, Sansom Institute for Health Research, University of South Australia, Adelaide, SA 5001, Australia.
| | - Timothy R H Regnault
- Departments of Obstetrics and Gynecology, University of Western Ontario, London, ON N6A 5C1, Canada.
| | - Paige L Barker
- Early Origins of Adult Health Research Group, Sansom Institute for Health Research, University of South Australia, Adelaide, SA 5001, Australia.
| | - Kimberley J Botting
- Early Origins of Adult Health Research Group, Sansom Institute for Health Research, University of South Australia, Adelaide, SA 5001, Australia.
| | - Isabella C McMillen
- Early Origins of Adult Health Research Group, Sansom Institute for Health Research, University of South Australia, Adelaide, SA 5001, Australia.
| | - Christine M McMillan
- Early Origins of Adult Health Research Group, Sansom Institute for Health Research, University of South Australia, Adelaide, SA 5001, Australia.
| | - Claire T Roberts
- The Robinson Research Institute, University of Adelaide, Adelaide, SA 5005, Australia.
| | - Janna L Morrison
- Early Origins of Adult Health Research Group, Sansom Institute for Health Research, University of South Australia, Adelaide, SA 5001, Australia.
| |
Collapse
|
98
|
Kadiyala KG, Tyagi T, Kakkar D, Chadha N, Chuttani K, Roy BG, Thirumal M, Mishra AK, Datta A. Picolinic acid based acyclic bifunctional chelating agent and its methionine conjugate as potential SPECT imaging agents: syntheses and preclinical evaluation. RSC Adv 2015. [DOI: 10.1039/c4ra13690j] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Syntheses and preclinical evaluation of picolinic acid based acyclic bifunctional chelating agent and its methionine conjugate as SPECT imaging agents.
Collapse
Affiliation(s)
- K. Ganesh Kadiyala
- Division of Cyclotron and Radiopharmaceutical Sciences
- Institute of Nuclear Medicine and Allied Sciences
- Defence Research and Development Organization
- Delhi-110054
- India
| | - Tulika Tyagi
- Division of Cyclotron and Radiopharmaceutical Sciences
- Institute of Nuclear Medicine and Allied Sciences
- Defence Research and Development Organization
- Delhi-110054
- India
| | - Dipti Kakkar
- Division of Cyclotron and Radiopharmaceutical Sciences
- Institute of Nuclear Medicine and Allied Sciences
- Defence Research and Development Organization
- Delhi-110054
- India
| | - Nidhi Chadha
- Division of Cyclotron and Radiopharmaceutical Sciences
- Institute of Nuclear Medicine and Allied Sciences
- Defence Research and Development Organization
- Delhi-110054
- India
| | - Krishna Chuttani
- Division of Cyclotron and Radiopharmaceutical Sciences
- Institute of Nuclear Medicine and Allied Sciences
- Defence Research and Development Organization
- Delhi-110054
- India
| | - Bal Gangadhar Roy
- Division of Cyclotron and Radiopharmaceutical Sciences
- Institute of Nuclear Medicine and Allied Sciences
- Defence Research and Development Organization
- Delhi-110054
- India
| | | | - Anil K. Mishra
- Division of Cyclotron and Radiopharmaceutical Sciences
- Institute of Nuclear Medicine and Allied Sciences
- Defence Research and Development Organization
- Delhi-110054
- India
| | - Anupama Datta
- Division of Cyclotron and Radiopharmaceutical Sciences
- Institute of Nuclear Medicine and Allied Sciences
- Defence Research and Development Organization
- Delhi-110054
- India
| |
Collapse
|
99
|
Kharlyngdoh JB, Pradhan A, Asnake S, Walstad A, Ivarsson P, Olsson PE. Identification of a group of brominated flame retardants as novel androgen receptor antagonists and potential neuronal and endocrine disrupters. ENVIRONMENT INTERNATIONAL 2015; 74:60-70. [PMID: 25454221 DOI: 10.1016/j.envint.2014.09.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 08/26/2014] [Accepted: 09/06/2014] [Indexed: 06/04/2023]
Abstract
Brominated flame-retardants (BFRs) are used in industrial products to reduce the risk of fire. However, their continuous release into the environment is a concern as they are often persistent, bioaccumulating and toxic. Information on the impact these compounds have on human health and wildlife is limited and only a few of them have been identified to disrupt hormone receptor functions. In the present study we used in silico modeling to determine the interactions of selected BFRs with the human androgen receptor (AR). Three compounds were found to dock into the ligand-binding domain of the human AR and these were further tested using in vitro analysis. Allyl 2,4,6-tribromophenyl ether (ATE), 2-bromoallyl 2,4,6-tribromophenyl ether (BATE) and 2,3-dibromopropyl-2,4,6-tribromophenyl ether (DPTE) were observed to act as AR antagonists. These BFRs have recently been detected in the environment, in house dust and in aquatic animals. The compounds have been detected at high concentrations in both blubber and brain of seals and we therefore also assessed their impact on the expression of L-type amino acid transporter system (LAT) genes, that are needed for amino acid uptake across the blood-brain barrier, as disruption of LAT gene function has been implicated in several brain disorders. The three BFRs down-regulated the expression of AR target genes that encode for prostate specific antigen (PSA), 5α-reductases and β-microseminoprotein. The potency of PSA inhibition was of the same magnitude as the common prostate cancer drugs, demonstrating that these compounds are strong AR antagonists. Western blot analysis of AR protein showed that ATE, BATE and DPTE decreased the 5α-dihydrotestosterone-induced AR protein levels, further confirming that these BFRs act as AR antagonists. The transcription of the LAT genes was altered by the three BFRs, indicating an effect on amino-acid uptake across cellular membranes and blood-brain barrier. This study demonstrated that ATE, BATE and DPTE are potent AR antagonists and the alterations in LAT gene transcription suggest that these compounds can affect neuronal functions and should be considered as potential neurotoxic and endocrine disrupting compounds.
Collapse
Affiliation(s)
- Joubert Banjop Kharlyngdoh
- Biology, Örebro Life Science Center, School of Science and Technology, Örebro University, SE-701 82 Örebro, Sweden
| | - Ajay Pradhan
- Biology, Örebro Life Science Center, School of Science and Technology, Örebro University, SE-701 82 Örebro, Sweden
| | - Solomon Asnake
- Biology, Örebro Life Science Center, School of Science and Technology, Örebro University, SE-701 82 Örebro, Sweden
| | - Anders Walstad
- Biology, Örebro Life Science Center, School of Science and Technology, Örebro University, SE-701 82 Örebro, Sweden
| | - Per Ivarsson
- ALS Laboratory Group, Analytical Chemistry & Testing Services, Stockholm, Sweden
| | - Per-Erik Olsson
- Biology, Örebro Life Science Center, School of Science and Technology, Örebro University, SE-701 82 Örebro, Sweden.
| |
Collapse
|
100
|
|