1
|
You S, Han X, Xu Y, Yao Q. Research progress on the role of cationic amino acid transporter (CAT) family members in malignant tumors and immune microenvironment. Amino Acids 2023; 55:1213-1222. [PMID: 37572157 DOI: 10.1007/s00726-023-03313-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 08/02/2023] [Indexed: 08/14/2023]
Abstract
Amino acids are essential for the survival of all living organisms and living cells. Amino acid transporters mediate the transport and absorption of amino acids, and the dysfunction of these proteins can induce human diseases. Cationic amino acid transporters (CAT family, SLC7A1-4, and SLC7A14) are considered to be a group of transmembrane transporters, of which SLC7A1-3 are essential for arginine transport in mammals. Numerous studies have shown that CAT family-mediated arginine transport is involved in signal crosstalk between malignant tumor cells and immune cells, especially T cells. The modulation of extracellular arginine concentration has entered a number of clinical trials and achieved certain therapeutic effects. Here, we review the role of CAT family on tumor cells and immune infiltrating cells in malignant tumors and explore the therapeutic strategies to interfere with extracellular arginine concentration, to elaborate its application prospects. CAT family members may be used as biomarkers for certain cancer entities and might be included in new ideas for immunotherapy of malignant tumors.
Collapse
Affiliation(s)
- Shijing You
- Department of Obstetrics and Gynaecology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong, China
| | - Xiahui Han
- Department of Obstetrics and Gynaecology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong, China
| | - Yuance Xu
- Department of Obstetrics and Gynaecology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong, China
| | - Qin Yao
- Department of Obstetrics and Gynaecology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong, China.
| |
Collapse
|
2
|
Afshinpour M, Parsi P, Mahdiuni H. Investigation of molecular details of a bacterial cationic amino acid transporter (GkApcT) during arginine transportation using molecular dynamics simulation and umbrella sampling techniques. J Mol Model 2023; 29:260. [PMID: 37479900 DOI: 10.1007/s00894-023-05670-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 07/17/2023] [Indexed: 07/23/2023]
Abstract
CONTEXT Cationic amino acid transporters (CATs) facilitate arginine transport across membranes and maintain its levels in various tissues and organs, but their overexpression has been associated with severe cancers. A recent study identified the alternating access mechanism and critical residues involved in arginine transportation in a cationic amino acid transporter from Geobacillus kaustophilus (GkApcT). Here, we used molecular dynamics (MD) simulation methods to investigate the transportation mechanism of arginine (Arg) through GkApcT. The results revealed that arginine strongly interacts with specific binding site residues (Thr43, Asp111, Glu115, Lys191, Phe231, Ile234, and Asp237). Based on the umbrella sampling, the main driving force for arginine transport is the polar interactions of the arginine with channel-lining residues. An in-depth description of the dissociation mechanism and binding energy analysis brings valuable insight into the interactions between arginine and transporter residues, facilitating the design of effective CAT inhibitors in cancer cells. METHODS The membrane-protein system was constructed by uploading the prokaryotic CAT (PDB ID: 6F34) to the CHARMM-GUI web server. Molecular dynamics simulations were done using the GROMACS package, version 5.1.4, with the CHARMM36 force field and TIP3P water model. The MM-PBSA approach was performed for determining the arginine binding free energy. Furthermore, the hotspot residues were identified through per-residue decomposition analysis. The characteristics of the channel such as bottleneck radius and channel length were analyzed using the CaverWeb 1.1 web server. The proton wire inside the transporter was investigated based on the classic Grotthuss mechanism. We also investigated the atomistic details of arginine transportation using the path-based free energy umbrella sampling technique (US).
Collapse
Affiliation(s)
- Maral Afshinpour
- Bioinformatics Lab, Department of Biology, School of Sciences, Razi University, P.O. Box, Kermanshah, 67149-67346, Iran
- Department of Chemistry and Biochemistry, South Dakota State University (SDSU), Brookings, SD, USA
| | - Parinaz Parsi
- Bioinformatics Lab, Department of Biology, School of Sciences, Razi University, P.O. Box, Kermanshah, 67149-67346, Iran
| | - Hamid Mahdiuni
- Bioinformatics Lab, Department of Biology, School of Sciences, Razi University, P.O. Box, Kermanshah, 67149-67346, Iran.
| |
Collapse
|
3
|
McColl ER, Henderson JT, Piquette-Miller M. Dysregulation of Amino Acid Transporters in a Rat Model of TLR7-Mediated Maternal Immune Activation. Pharmaceutics 2023; 15:1857. [PMID: 37514044 PMCID: PMC10385561 DOI: 10.3390/pharmaceutics15071857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 06/16/2023] [Accepted: 06/26/2023] [Indexed: 07/30/2023] Open
Abstract
Maternal immune activation (MIA) during pregnancy is linked to neurodevelopmental disorders in humans. Similarly, the TLR7 agonist imiquimod alters neurodevelopment in rodents. While the mechanisms underlying MIA-mediated neurodevelopmental changes are unknown, they could involve dysregulation of amino acid transporters essential for neurodevelopment. Therefore, we sought to determine the nature of such transporter changes in both imiquimod-treated rats and human placentas during infection. Pregnant rats received imiquimod on gestational day (GD)14. Transporter expression was measured in placentas and fetal brains via qPCR (GD14.5) and immunoblotting (GD16). To monitor function, fetal brain amino acid levels were measured by HPLC on GD16. Gene expression in the cortex of female fetal brains was further examined by RNAseq on GD19. In human placentas, suspected active infection was associated with decreased ASCT1 and SNAT2 protein expression. Similarly, in imiquimod-treated rats, ASCT1 and SNAT2 protein was also decreased in male placentas, while EAAT2 was decreased in female placentas. CAT3 was increased in female fetal brains. Consistent with this, imiquimod altered amino acid levels in fetal brains, while RNAseq demonstrated changes in expression of several genes implicated in autism. Thus, imiquimod alters amino acid transporter levels in pregnant rats, and similar changes occur in human placentas during active infection. This suggests that changes in expression of amino acid transporters may contribute to effects mediated by MIA toward altered neurodevelopment.
Collapse
Affiliation(s)
- Eliza R McColl
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College St, Toronto, ON M5S 3M2, Canada
| | - Jeffrey T Henderson
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College St, Toronto, ON M5S 3M2, Canada
| | - Micheline Piquette-Miller
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College St, Toronto, ON M5S 3M2, Canada
| |
Collapse
|
4
|
Tongta S, Daendee S, Kalandakanond-Thongsong S. Anxiety-like behavior and GABAergic system in ovariectomized rats exposed to chronic mild stress. Physiol Behav 2023; 258:114014. [PMID: 36328075 DOI: 10.1016/j.physbeh.2022.114014] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 10/14/2022] [Accepted: 10/27/2022] [Indexed: 11/06/2022]
Abstract
Stress or low level of estrogen could promote anxiety and depression; thus, it is of interest to investigate the combined effect of mild stress and the lack of estrogen on mental disorders by utilizing an animal model. This study was conducted to assess anxiety- and depressive- like behaviors in ovariectomized (Ovx) rats exposed to chronic mild stress (CMS) and determine the alteration in gamma-aminobutyric acid (GABA)-related transmission. Ovx rats were randomly assigned into four groups: (1) estrogen replacement (E2-NoCMS), (2) estrogen replacement and exposure to CMS (E2-CMS), (3) vehicle (VEH-NoCMS), and (4) vehicle and exposure to CMS (VEH-CMS). Following 4-week CMS, VEH groups (VEH-NoCMS and VEH-CMS) showed a similar level of anxiety-like behavior in elevated T-maze, whereas E2-CMS, VEH-NoCMS and VEH-CMS showed anxiety-like behavior in open field. The depressive-like behavior in the force swimming test tended to be affected by estrogen deprivation than CMS. The alteration of the GABAergic system as determined from the GABA level and mRNA expression of GABA-related transmission (i.e., glutamic acid decarboxylase, GABA transporter and GABAA subunits) showed that the GABA level in the amygdala and frontal cortex was affected by CMS. For mRNA expression, the mRNA profile in the amygdala and hippocampus of VEH-NoCMS and E2-CMS was the same but different from those of VEH-NoCMS and E2-CMS. In addition, compared with E2-NoCMS, the mRNA profile in the frontal cortex was similar in VEH-NoCMS, E2-CMS, and VEH-CMS. These findings indicated that the underlying mechanism of the GABAergic system was differently modified, although VEH-NoCMS and VEH-CMS showed anxiety-like behavior. The findings of this study may provide a comprehensive understanding of the modulation of the GABAergic system during estrogen deprivation under CMS, as observed in menopausal women who were daily exposed to stress.
Collapse
Affiliation(s)
- Sushawadee Tongta
- Department of Veterinary Physiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Suwaporn Daendee
- College of Medicine and Public Health, Ubon Ratchathani University, Ubon Ratchathani 34190, Thailand
| | | |
Collapse
|
5
|
Yan Y, Chen C, Li Z, Zhang J, Park N, Qu CK. Extracellular arginine is required but the arginine transporter CAT3 (Slc7a3) is dispensable for mouse normal and malignant hematopoiesis. Sci Rep 2022; 12:21832. [PMID: 36528691 PMCID: PMC9759514 DOI: 10.1038/s41598-022-24554-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 11/16/2022] [Indexed: 12/23/2022] Open
Abstract
Amino acid-mediated metabolism is one of the key catabolic and anabolic processes involved in diverse cellular functions. However, the role of the semi-essential amino acid arginine in normal and malignant hematopoietic cell development is poorly understood. Here we report that a continuous supply of exogenous arginine is required for the maintenance/function of normal hematopoietic stem cells (HSCs). Surprisingly, knockout of Slc7a3 (CAT3), a major L-arginine transporter, does not affect HSCs in steady-state or under stress. Although Slc7a3 is highly expressed in naïve and activated CD8 T cells, neither T cell development nor activation/proliferation is impacted by Slc7a3 depletion. Furthermore, the Slc7a3 deletion does not attenuate leukemia development driven by Pten loss or the oncogenic Ptpn11E76K mutation. Arginine uptake assays reveal that L-arginine uptake is not disrupted in Slc7a3 knockout cells. These data suggest that extracellular arginine is critically important for HSCs, but CAT3 is dispensable for normal hematopoiesis and leukemogenesis.
Collapse
Affiliation(s)
- Yuhan Yan
- grid.189967.80000 0001 0941 6502Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Winship Cancer Institute, Children’s Healthcare of Atlanta, Emory University School of Medicine, 1760 Haygood Drive NE, HSRB E302, Atlanta, GA 30322 USA
| | - Chao Chen
- grid.189967.80000 0001 0941 6502Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Winship Cancer Institute, Children’s Healthcare of Atlanta, Emory University School of Medicine, 1760 Haygood Drive NE, HSRB E302, Atlanta, GA 30322 USA
| | - Zhiguo Li
- grid.189967.80000 0001 0941 6502Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Winship Cancer Institute, Children’s Healthcare of Atlanta, Emory University School of Medicine, 1760 Haygood Drive NE, HSRB E302, Atlanta, GA 30322 USA
| | - Jing Zhang
- grid.189967.80000 0001 0941 6502Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Winship Cancer Institute, Children’s Healthcare of Atlanta, Emory University School of Medicine, 1760 Haygood Drive NE, HSRB E302, Atlanta, GA 30322 USA
| | - Narin Park
- grid.189967.80000 0001 0941 6502Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Winship Cancer Institute, Children’s Healthcare of Atlanta, Emory University School of Medicine, 1760 Haygood Drive NE, HSRB E302, Atlanta, GA 30322 USA
| | - Cheng-Kui Qu
- grid.189967.80000 0001 0941 6502Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Winship Cancer Institute, Children’s Healthcare of Atlanta, Emory University School of Medicine, 1760 Haygood Drive NE, HSRB E302, Atlanta, GA 30322 USA
| |
Collapse
|
6
|
Bangsumruaj J, Kijtawornrat A, Kalandakanond-Thongsong S. Effects of chronic mild stress on GABAergic system in the paraventricular nucleus of hypothalamus associated with cardiac autonomic activity. Behav Brain Res 2022; 432:113985. [PMID: 35787398 DOI: 10.1016/j.bbr.2022.113985] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 04/26/2022] [Accepted: 06/28/2022] [Indexed: 11/29/2022]
Abstract
Stress is associated with cardiovascular diseases. One possible mechanism is the reduction in gamma-aminobutyric acid (GABA)ergic transmission at the paraventricular nucleus (PVN), which contributes to the disinhibition of sympathoexcitatory circuits and activates sympathetic outflow. At present, the mechanism of chronic mild stress (CMS) on GABAergic transmission at the PVN and cardiac autonomic activity is not yet fully clarified. Therefore, this study was designed to investigate the effects of CMS on the GABAergic system at the PVN and on the cardiac autonomic activity. Adult male Sprague-Dawley rats were randomly assigned to control (left undisturbed in their home cage) or CMS (subjected to various mild stressors for 4 weeks). Cardiac autonomic activities were determined by heart rate variability (HRV) analysis, and GABAergic alterations at the PVN were determined from GABA levels and mRNA expression of GABA-related activities. Results showed that the CMS group had decreased HRV as determined by the standard deviation of all R-R intervals (SDNN). The low frequency (LF) and high frequency (HF) powers of the CMS group were higher than those of the control. Hence, the LF/HF ratio was consequently unaffected. These findings indicated that despite the increase in sympathetic and parasympathetic activities, the autonomic balance was preserved at 4 weeks post CMS. For the GABAergic-related parameters, the CMS group had decreased mRNA expression of glutamic acid decarboxylase-65 (GAD-65), the GABA-synthesizing enzyme, and increased mRNA expression of gamma-aminobutyric acid transporter-1 (GAT-1). Moreover, the GAD-65 mRNA expression was negatively correlated with LF. In conclusion, 4-week CMS exposure in male rats could attenuate GABAergic transmission at the PVN and alter cardiac autonomic activities.
Collapse
Affiliation(s)
- Janpen Bangsumruaj
- Interdisciplinary Program in Physiology, Graduate School, Chulalongkorn University, Bangkok 10330, Thailand.
| | - Anusak Kijtawornrat
- Department of Veterinary Physiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand.
| | | |
Collapse
|
7
|
Maeda K, Tasaki M, Ando Y, Ohtsubo K. Galectin-lattice sustains function of cationic amino acid transporter and insulin secretion of pancreatic β cells. J Biochem 2021; 167:587-596. [PMID: 31960919 DOI: 10.1093/jb/mvaa007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 01/09/2020] [Indexed: 01/01/2023] Open
Abstract
Maintenance of cell surface residency and function of glycoproteins by lectins are essential for regulating cellular functions. Galectins are β-galactoside-binding lectins and form a galectin-lattice, which regulates stability, clustering, membrane sub-domain localization and endocytosis of plasmalemmal glycoproteins. We have previously reported that galectin-2 (Gal-2) forms a complex with cationic amino acid transporter 3 (CAT3) in pancreatic β cells, although the biological significance of the molecular interaction between Gal-2 and CAT3 has not been elucidated. In this study, we demonstrated that the structure of N-glycan of CAT3 was either tetra- or tri-antennary branch structure carrying β-galactosides, which works as galectin-ligands. Indeed, CAT3 bound to Gal-2 using β-galactoside epitope. Moreover, the disruption of the glycan-mediated bindings between galectins and CAT3 significantly reduced cell surface expression levels of CAT3. The reduced cell surface residency of CAT3 attenuated the cellular arginine uptake activities and subsequently reduced nitric oxide production, and thus impaired the arginine-stimulated insulin secretion of pancreatic β cells. These results indicate that galectin-lattice stabilizes CAT3 by preventing endocytosis to sustain the arginine-stimulated insulin secretion of pancreatic β cells. This provides a novel cell biological insight into the endocrinological mechanism of nutrition metabolism and homeostasis.
Collapse
Affiliation(s)
- Kento Maeda
- Department of Analytical Biochemistry;, Graduate School of Health Sciences, Kumamoto University, 4-24-1 Kuhonji, Chuo-Ku, Kumamoto 862-0976, Japan
| | - Masayoshi Tasaki
- Department of Morphological and Physiological Sciences, Graduate School of Health Sciences, Kumamoto University, 4-24-1 Kuhonji, Chuo-Ku, Kumamoto 862-0976, Japan.,Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-0811, Japan
| | - Yukio Ando
- Depatment of Amyloidosis Research, Nagasaki International University, Nagasaki 859-3243, Japan
| | - Kazuaki Ohtsubo
- Department of Analytical Biochemistry;, Graduate School of Health Sciences, Kumamoto University, 4-24-1 Kuhonji, Chuo-Ku, Kumamoto 862-0976, Japan.,Department of Analytical Biochemistry, Faculty of Life Sciences, Kumamoto University, 4-24-1 Kuhonji, Chuo-Ku, Kumamoto 862-0976, Japan
| |
Collapse
|
8
|
Transport of L-Arginine Related Cardiovascular Risk Markers. J Clin Med 2020; 9:jcm9123975. [PMID: 33302555 PMCID: PMC7764698 DOI: 10.3390/jcm9123975] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/30/2020] [Accepted: 12/01/2020] [Indexed: 12/15/2022] Open
Abstract
L-arginine and its derivatives, asymmetric and symmetric dimethylarginine (ADMA and SDMA) and L-homoarginine, have emerged as cardiovascular biomarkers linked to cardiovascular outcomes and various metabolic and functional pathways such as NO-mediated endothelial function. Cellular uptake and efflux of L-arginine and its derivatives are facilitated by transport proteins. In this respect the cationic amino acid transporters CAT1 and CAT2 (SLC7A1 and SLC7A2) and the system y+L amino acid transporters (SLC7A6 and SLC7A7) have been most extensively investigated, so far, but the number of transporters shown to mediate the transport of L-arginine and its derivatives is constantly increasing. In the present review we assess the growing body of evidence regarding the function, expression, and clinical relevance of these transporters and their possible relation to cardiovascular diseases.
Collapse
|
9
|
Dawson HD, Chen C, Gaynor B, Shao J, Urban JF. The porcine translational research database: a manually curated, genomics and proteomics-based research resource. BMC Genomics 2017; 18:643. [PMID: 28830355 PMCID: PMC5568366 DOI: 10.1186/s12864-017-4009-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 08/02/2017] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The use of swine in biomedical research has increased dramatically in the last decade. Diverse genomic- and proteomic databases have been developed to facilitate research using human and rodent models. Current porcine gene databases, however, lack the robust annotation to study pig models that are relevant to human studies and for comparative evaluation with rodent models. Furthermore, they contain a significant number of errors due to their primary reliance on machine-based annotation. To address these deficiencies, a comprehensive literature-based survey was conducted to identify certain selected genes that have demonstrated function in humans, mice or pigs. RESULTS The process identified 13,054 candidate human, bovine, mouse or rat genes/proteins used to select potential porcine homologs by searching multiple online sources of porcine gene information. The data in the Porcine Translational Research Database (( http://www.ars.usda.gov/Services/docs.htm?docid=6065 ) is supported by >5800 references, and contains 65 data fields for each entry, including >9700 full length (5' and 3') unambiguous pig sequences, >2400 real time PCR assays and reactivity information on >1700 antibodies. It also contains gene and/or protein expression data for >2200 genes and identifies and corrects 8187 errors (gene duplications artifacts, mis-assemblies, mis-annotations, and incorrect species assignments) for 5337 porcine genes. CONCLUSIONS This database is the largest manually curated database for any single veterinary species and is unique among porcine gene databases in regard to linking gene expression to gene function, identifying related gene pathways, and connecting data with other porcine gene databases. This database provides the first comprehensive description of three major Super-families or functionally related groups of proteins (Cluster of Differentiation (CD) Marker genes, Solute Carrier Superfamily, ATP binding Cassette Superfamily), and a comparative description of porcine microRNAs.
Collapse
Affiliation(s)
- Harry D Dawson
- United States Department of Agriculture, Agricultural Research Service, Beltsville Human Nutrition Research Center, Diet, Genomics and Immunology Laboratory, Beltsville, MD, USA.
| | - Celine Chen
- United States Department of Agriculture, Agricultural Research Service, Beltsville Human Nutrition Research Center, Diet, Genomics and Immunology Laboratory, Beltsville, MD, USA
| | - Brady Gaynor
- United States Department of Agriculture, Agricultural Research Service, Beltsville Agricultural Research Center, Molecular Plant Pathology Lab, Beltsville, MD, 20705, USA
| | - Jonathan Shao
- United States Department of Agriculture, Agricultural Research Service, Beltsville Agricultural Research Center, Molecular Plant Pathology Lab, Beltsville, MD, 20705, USA
| | - Joseph F Urban
- United States Department of Agriculture, Agricultural Research Service, Beltsville Human Nutrition Research Center, Diet, Genomics and Immunology Laboratory, Beltsville, MD, USA
| |
Collapse
|
10
|
Ishida A, Ashihara A, Nakashima K, Katsumata M. Expression of cationic amino acid transporters in pig skeletal muscles during postnatal development. Amino Acids 2017; 49:1805-1814. [PMID: 28803359 DOI: 10.1007/s00726-017-2478-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 08/01/2017] [Indexed: 11/24/2022]
Abstract
The cationic amino acid transporter (CAT) protein family transports lysine and arginine in cellular amino acid pools. We hypothesized that CAT expression changes in pig skeletal muscles during rapid pig postnatal development. We aimed to investigate the tissue distribution and changes in the ontogenic expression of CATs in pig skeletal muscles during postnatal development. Six piglets at 1, 12, 26, 45, and 75 days old were selected from six litters, and their longissimus dorsi (LD), biceps femoris (BF), and rhomboideus (RH) muscles, and their stomach, duodenum, jejunum, ileum, colon, liver, kidney, heart, and cerebrum were collected. CAT-1 was expressed in all the 12 tissues investigated. CAT-2 (CAT-2A isoform) expression was highest in the skeletal muscle and liver and lowest in the jejunum, ileum, kidney, and heart. CAT-3 was expressed mainly in the colon and detected in the jejunum, ileum, and cerebrum. The CAT-1 expression was higher in the skeletal muscle of day 1 pigs than in that of older pigs (P < 0.05). The CAT-2 mRNA level was lowest at day 1, but increased with postnatal development (P < 0.05). There was no significant change in CAT-1 expression among the LD, BF, and RH during postnatal development (P > 0.05); however, there was a change in CAT-2 expression. The CAT-2 expression was highest in the LD of 12-, 26-, 45-, and 75-day-old pigs, followed by the BF and RH (P < 0.05). These results suggest that CAT-1 and CAT-2 play different roles in pig skeletal muscles during postnatal development.
Collapse
Affiliation(s)
- Aiko Ishida
- Institute of Livestock and Grassland Science, NARO, Tsukuba, Ibaraki, 305-0901, Japan.
| | - Akane Ashihara
- Institute of Livestock and Grassland Science, NARO, Tsukuba, Ibaraki, 305-0901, Japan
| | - Kazuki Nakashima
- Institute of Livestock and Grassland Science, NARO, Tsukuba, Ibaraki, 305-0901, Japan
| | - Masaya Katsumata
- Institute of Livestock and Grassland Science, NARO, Tsukuba, Ibaraki, 305-0901, Japan.,School of Veterinary Science, Azabu University, Sagamihara, Kanagawa, 252-5201, Japan
| |
Collapse
|
11
|
de la Ballina LR, Cano-Crespo S, González-Muñoz E, Bial S, Estrach S, Cailleteau L, Tissot F, Daniel H, Zorzano A, Ginsberg MH, Palacín M, Féral CC. Amino Acid Transport Associated to Cluster of Differentiation 98 Heavy Chain (CD98hc) Is at the Cross-road of Oxidative Stress and Amino Acid Availability. J Biol Chem 2016; 291:9700-11. [PMID: 26945935 DOI: 10.1074/jbc.m115.704254] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Indexed: 01/08/2023] Open
Abstract
CD98hc functions as an amino acid (AA) transporter (together with another subunit) and integrin signaling enhancer. It is overexpressed in highly proliferative cells in both physiological and pathological conditions. CD98hc deletion induces strong impairment of cell proliferation in vivo and in vitro Here, we investigate CD98hc-associated AA transport in cell survival and proliferation. By using chimeric versions of CD98hc, the two functions of the protein can be uncoupled. Although recovering the CD98hc AA transport capacity restores the in vivo and in vitro proliferation of CD98hc-null cells, reconstitution of the integrin signaling function of CD98hc is unable to restore in vitro proliferation of those cells. CD98hc-associated transporters (i.e. xCT, LAT1, and y(+)LAT2 in wild-type cells) are crucial to control reactive oxygen species and intracellular AA levels, thus sustaining cell survival and proliferation. Moreover, in CD98hc-null cells the deficiency of CD98hc/xCT cannot be compensated, leading to cell death by ferroptosis. Supplementation of culture media with β-mercaptoethanol rescues CD98hc-deficient cell survival. Under such conditions null cells show oxidative stress and intracellular AA imbalance and, consequently, limited proliferation. CD98hc-null cells also present reduced intracellular levels of branched-chain and aromatic amino acids (BCAAs and ARO AAs, respectively) and induced expression of peptide transporter 1 (PEPT1). Interestingly, external supply of dipeptides containing BCAAs and ARO AAs rescues cell proliferation and compensates for impaired uptake of CD98hc/LAT1 and CD98hc/y(+)LAT2. Our data establish CD98hc as a master protective gene at the cross-road of redox control and AA availability, making it a relevant therapeutic target in cancer.
Collapse
Affiliation(s)
- Laura R de la Ballina
- From the Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology, Baldiri Reixac 10, 08028 Barcelona, Spain and Department of Biochemistry and Molecular Biology, University of Barcelona, 08028 Barcelona, Spain, INSERM, U1081, Institute for Research on Cancer and Aging, Nice (IRCAN), University of Nice Sophia-Antipolis, CNRS UMR 7284, 06107 Nice, France,
| | - Sara Cano-Crespo
- From the Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology, Baldiri Reixac 10, 08028 Barcelona, Spain and Department of Biochemistry and Molecular Biology, University of Barcelona, 08028 Barcelona, Spain, Spanish Biomedical Research Network in Rare Diseases (CIBERER U-731), 08028 Barcelona, Spain
| | - Elena González-Muñoz
- From the Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology, Baldiri Reixac 10, 08028 Barcelona, Spain and Department of Biochemistry and Molecular Biology, University of Barcelona, 08028 Barcelona, Spain, The Andalusian Cellular Reprogramming Laboratory (LARCEL), Fundación Progreso y Salud, 41092 Seville, Spain
| | - Susanna Bial
- From the Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology, Baldiri Reixac 10, 08028 Barcelona, Spain and Department of Biochemistry and Molecular Biology, University of Barcelona, 08028 Barcelona, Spain, Spanish Biomedical Research Network in Rare Diseases (CIBERER U-731), 08028 Barcelona, Spain
| | - Soline Estrach
- INSERM, U1081, Institute for Research on Cancer and Aging, Nice (IRCAN), University of Nice Sophia-Antipolis, CNRS UMR 7284, 06107 Nice, France
| | - Laurence Cailleteau
- INSERM, U1081, Institute for Research on Cancer and Aging, Nice (IRCAN), University of Nice Sophia-Antipolis, CNRS UMR 7284, 06107 Nice, France
| | - Floriane Tissot
- INSERM, U1081, Institute for Research on Cancer and Aging, Nice (IRCAN), University of Nice Sophia-Antipolis, CNRS UMR 7284, 06107 Nice, France
| | - Hannelore Daniel
- ZIEL Research Center of Nutrition and Food Sciences, Molecular Nutrition and Biochemistry Unit, Technische Universität München, Gregor-Mendel-Strasse 2, 85350 Freising, Germany
| | - Antonio Zorzano
- From the Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology, Baldiri Reixac 10, 08028 Barcelona, Spain and Department of Biochemistry and Molecular Biology, University of Barcelona, 08028 Barcelona, Spain, Spanish Biomedical Research Network in Diabetes and Associated Metabolic Diseases (CIBERDEM), 08028 Barcelona, Spain, and
| | - Mark H Ginsberg
- Department of Medicine, University of California, San Diego, La Jolla, California 92093
| | - Manuel Palacín
- From the Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology, Baldiri Reixac 10, 08028 Barcelona, Spain and Department of Biochemistry and Molecular Biology, University of Barcelona, 08028 Barcelona, Spain, Spanish Biomedical Research Network in Rare Diseases (CIBERER U-731), 08028 Barcelona, Spain,
| | - Chloé C Féral
- INSERM, U1081, Institute for Research on Cancer and Aging, Nice (IRCAN), University of Nice Sophia-Antipolis, CNRS UMR 7284, 06107 Nice, France,
| |
Collapse
|
12
|
Closs EI. Putative role of cationic amino acid transporter-3 in murine liver metabolism. Hepatology 2015; 62:1326-7. [PMID: 25712160 DOI: 10.1002/hep.27763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/07/2022]
Affiliation(s)
- Ellen I Closs
- Institute of Pharmacology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
13
|
Gu Q, Cui Z. Reply: To PMID 25130427. Hepatology 2015; 62:1327. [PMID: 25711644 DOI: 10.1002/hep.27759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/07/2022]
Affiliation(s)
- Qilin Gu
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Zongbin Cui
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| |
Collapse
|
14
|
Nava C, Rupp J, Boissel JP, Mignot C, Rastetter A, Amiet C, Jacquette A, Dupuits C, Bouteiller D, Keren B, Ruberg M, Faudet A, Doummar D, Philippe A, Périsse D, Laurent C, Lebrun N, Guillemot V, Chelly J, Cohen D, Héron D, Brice A, Closs EI, Depienne C. Hypomorphic variants of cationic amino acid transporter 3 in males with autism spectrum disorders. Amino Acids 2015. [PMID: 26215737 PMCID: PMC4633447 DOI: 10.1007/s00726-015-2057-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Cationic amino acid transporters (CATs) mediate the entry of L-type cationic amino acids (arginine, ornithine and lysine) into the cells including neurons. CAT-3, encoded by the SLC7A3 gene on chromosome X, is one of the three CATs present in the human genome, with selective expression in brain. SLC7A3 is highly intolerant to variation in humans, as attested by the low frequency of deleterious variants in available databases, but the impact on variants in this gene in humans remains undefined. In this study, we identified a missense variant in SLC7A3, encoding the CAT-3 cationic amino acid transporter, on chromosome X by exome sequencing in two brothers with autism spectrum disorder (ASD). We then sequenced the SLC7A3 coding sequence in 148 male patients with ASD and identified three additional rare missense variants in unrelated patients. Functional analyses of the mutant transporters showed that two of the four identified variants cause severe or moderate loss of CAT-3 function due to altered protein stability or abnormal trafficking to the plasma membrane. The patient with the most deleterious SLC7A3 variant had high-functioning autism and epilepsy, and also carries a de novo 16p11.2 duplication possibly contributing to his phenotype. This study shows that rare hypomorphic variants of SLC7A3 exist in male individuals and suggest that SLC7A3 variants possibly contribute to the etiology of ASD in male subjects in association with other genetic factors.
Collapse
Affiliation(s)
- Caroline Nava
- Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, ICM, 75013, Paris, France.,INSERM, U 1127, 75013, Paris, France.,CNRS, UMR 7225, 75013, Paris, France.,Institut du cerveau et de la moelle épinière (ICM), 75013, Paris, France.,Département de Génétique et de Cytogénétique, Hôpital de la Pitié-Salpêtrière, AP-HP, 75013, Paris, France
| | - Johanna Rupp
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Jean-Paul Boissel
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Cyril Mignot
- Département de Génétique et de Cytogénétique, Hôpital de la Pitié-Salpêtrière, AP-HP, 75013, Paris, France.,Centre de Référence "déficiences intellectuelles de causes rares", Paris, France.,Groupe de Recherche Clinique (GRC) "déficience intellectuelle et autisme" UPMC, Paris, France.,Service de neuropédiatrie, Hôpital Trousseau, AP-HP, Paris, France
| | - Agnès Rastetter
- Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, ICM, 75013, Paris, France.,INSERM, U 1127, 75013, Paris, France.,CNRS, UMR 7225, 75013, Paris, France.,Institut du cerveau et de la moelle épinière (ICM), 75013, Paris, France
| | - Claire Amiet
- Service de psychiatrie de l'enfant et de l'adolescent, Hôpital Pitié-Salpêtrière, AP-HP, 75013, Paris, France
| | - Aurélia Jacquette
- Département de Génétique et de Cytogénétique, Hôpital de la Pitié-Salpêtrière, AP-HP, 75013, Paris, France.,Centre de Référence "déficiences intellectuelles de causes rares", Paris, France.,Groupe de Recherche Clinique (GRC) "déficience intellectuelle et autisme" UPMC, Paris, France
| | - Céline Dupuits
- Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, ICM, 75013, Paris, France.,INSERM, U 1127, 75013, Paris, France.,CNRS, UMR 7225, 75013, Paris, France.,Institut du cerveau et de la moelle épinière (ICM), 75013, Paris, France
| | - Delphine Bouteiller
- Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, ICM, 75013, Paris, France.,INSERM, U 1127, 75013, Paris, France.,CNRS, UMR 7225, 75013, Paris, France.,Institut du cerveau et de la moelle épinière (ICM), 75013, Paris, France
| | - Boris Keren
- Département de Génétique et de Cytogénétique, Hôpital de la Pitié-Salpêtrière, AP-HP, 75013, Paris, France
| | - Merle Ruberg
- Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, ICM, 75013, Paris, France.,INSERM, U 1127, 75013, Paris, France.,CNRS, UMR 7225, 75013, Paris, France.,Institut du cerveau et de la moelle épinière (ICM), 75013, Paris, France
| | - Anne Faudet
- Département de Génétique et de Cytogénétique, Hôpital de la Pitié-Salpêtrière, AP-HP, 75013, Paris, France
| | - Diane Doummar
- Service de neuropédiatrie, Hôpital Trousseau, AP-HP, Paris, France
| | - Anne Philippe
- Service de psychiatrie de l'enfant et de l'adolescent, Hôpital Pitié-Salpêtrière, AP-HP, 75013, Paris, France
| | - Didier Périsse
- Service de psychiatrie de l'enfant et de l'adolescent, Hôpital Pitié-Salpêtrière, AP-HP, 75013, Paris, France.,Centre Diagnostic Autisme de l'Hôpital Pitié-Salpêtrière, 75013, Paris, France
| | - Claudine Laurent
- Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, ICM, 75013, Paris, France.,INSERM, U 1127, 75013, Paris, France.,CNRS, UMR 7225, 75013, Paris, France.,Institut du cerveau et de la moelle épinière (ICM), 75013, Paris, France.,Service de psychiatrie de l'enfant et de l'adolescent, Hôpital Pitié-Salpêtrière, AP-HP, 75013, Paris, France
| | - Nicolas Lebrun
- Institut Cochin, Inserm U567, UMR 8104, Université René Descartes, Paris 5, France
| | - Vincent Guillemot
- Bioinformatics and Biostatistics Core Facility (iCONICS), Institut du cerveau et de la moelle épinière (ICM), Paris, France
| | - Jamel Chelly
- Institut Cochin, Inserm U567, UMR 8104, Université René Descartes, Paris 5, France
| | - David Cohen
- Service de psychiatrie de l'enfant et de l'adolescent, Hôpital Pitié-Salpêtrière, AP-HP, 75013, Paris, France.,Institut des Systèmes Intelligents et Robotiques, CNRS UMR 7222, UPMC-Paris-6, Paris, France
| | - Delphine Héron
- Département de Génétique et de Cytogénétique, Hôpital de la Pitié-Salpêtrière, AP-HP, 75013, Paris, France.,Centre de Référence "déficiences intellectuelles de causes rares", Paris, France.,Groupe de Recherche Clinique (GRC) "déficience intellectuelle et autisme" UPMC, Paris, France.,Service de neuropédiatrie, Hôpital Trousseau, AP-HP, Paris, France
| | - Alexis Brice
- Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, ICM, 75013, Paris, France.,INSERM, U 1127, 75013, Paris, France.,CNRS, UMR 7225, 75013, Paris, France.,Institut du cerveau et de la moelle épinière (ICM), 75013, Paris, France.,Département de Génétique et de Cytogénétique, Hôpital de la Pitié-Salpêtrière, AP-HP, 75013, Paris, France
| | - Ellen I Closs
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Christel Depienne
- Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, ICM, 75013, Paris, France. .,INSERM, U 1127, 75013, Paris, France. .,CNRS, UMR 7225, 75013, Paris, France. .,Institut du cerveau et de la moelle épinière (ICM), 75013, Paris, France. .,Département de Génétique et de Cytogénétique, Hôpital de la Pitié-Salpêtrière, AP-HP, 75013, Paris, France.
| |
Collapse
|
15
|
The SLC3 and SLC7 families of amino acid transporters. Mol Aspects Med 2013; 34:139-58. [PMID: 23506863 DOI: 10.1016/j.mam.2012.10.007] [Citation(s) in RCA: 503] [Impact Index Per Article: 41.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Accepted: 08/15/2012] [Indexed: 01/18/2023]
Abstract
Amino acids are necessary for all living cells and organisms. Specialized transporters mediate the transfer of amino acids across plasma membranes. Malfunction of these proteins can affect whole-body homoeostasis giving raise to diverse human diseases. Here, we review the main features of the SLC3 and SLC7 families of amino acid transporters. The SLC7 family is divided into two subfamilies, the cationic amino acid transporters (CATs), and the L-type amino acid transporters (LATs). The latter are the light or catalytic subunits of the heteromeric amino acid transporters (HATs), which are associated by a disulfide bridge with the heavy subunits 4F2hc or rBAT. These two subunits are glycoproteins and form the SLC3 family. Most CAT subfamily members were functionally characterized and shown to function as facilitated diffusers mediating the entry and efflux of cationic amino acids. In certain cells, CATs play an important role in the delivery of L-arginine for the synthesis of nitric oxide. HATs are mostly exchangers with a broad spectrum of substrates and are crucial in renal and intestinal re-absorption and cell redox balance. Furthermore, the role of the HAT 4F2hc/LAT1 in tumor growth and the application of LAT1 inhibitors and PET tracers for reduction of tumor progression and imaging of tumors are discussed. Finally, we describe the link between specific mutations in HATs and the primary inherited aminoacidurias, cystinuria and lysinuric protein intolerance.
Collapse
|
16
|
Beyer SR, Mallmann RT, Jaenecke I, Habermeier A, Boissel JP, Closs EI. Identification of cysteine residues in human cationic amino acid transporter hCAT-2A that are targets for inhibition by N-ethylmaleimide. J Biol Chem 2013; 288:30411-30419. [PMID: 24019517 DOI: 10.1074/jbc.m113.490698] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
In most cells, cationic amino acids such as l-arginine, l-lysine, and l-ornithine are transported by cationic (CAT) and y(+)L (y(+)LAT) amino acid transporters. In human erythrocytes, the cysteine-modifying agent N-ethylmaleimide (NEM) has been shown to inhibit system y(+) (most likely CAT-1), but not system y(+)L (Devés, R., Angelo, S., and Chávez, P. (1993) J. Physiol. 468, 753-766). We thus wondered if sensitivity to NEM distinguishes generally all CAT and y(+)LAT isoforms. Transport assays in Xenopus laevis oocytes established that indeed all human CATs (including the low affinity hCAT-2A), but neither y(+)LAT isoform, are inhibited by NEM. hCAT-2A inhibition was not due to reduced transporter expression in the plasma membrane, indicating that NEM reduces the intrinsic transporter activity. Individual mutation of each of the seven cysteine residues conserved in all CAT isoforms did not lead to NEM insensitivity of hCAT-2A. However, a cysteine-less mutant was no longer inhibited by NEM, suggesting that inhibition occurs through modification of more than one cysteine in hCAT-2A. Indeed, also the double mutant C33A/C273A was insensitive to NEM inhibition, whereas reintroduction of a cysteine at either position 33 or 273 in the cysteine-less mutant led to NEM sensitivity. We thus identified Cys-33 and Cys-273 in hCAT-2A as the targets of NEM inhibition. In addition, all proteins with Cys-33 mutations showed a pronounced reduction in transport activity, suggesting that, surprisingly, this residue, located in the cytoplasmic N terminus, is important for transporter function.
Collapse
Affiliation(s)
- Sarah R Beyer
- From the Department of Pharmacology, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany
| | - Robert T Mallmann
- From the Department of Pharmacology, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany
| | - Isabel Jaenecke
- From the Department of Pharmacology, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany
| | - Alice Habermeier
- From the Department of Pharmacology, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany
| | - Jean-Paul Boissel
- From the Department of Pharmacology, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany
| | - Ellen I Closs
- From the Department of Pharmacology, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany.
| |
Collapse
|
17
|
Iino I, Kikuchi H, Miyazaki S, Hiramatsu Y, Ohta M, Kamiya K, Kusama Y, Baba S, Setou M, Konno H. Effect of miR-122 and its target gene cationic amino acid transporter 1 on colorectal liver metastasis. Cancer Sci 2013; 104:624-30. [PMID: 23373973 PMCID: PMC7657140 DOI: 10.1111/cas.12122] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Revised: 01/14/2013] [Accepted: 02/04/2013] [Indexed: 12/22/2022] Open
Abstract
Control of liver metastasis is an important issue in the treatment of colorectal cancer (CRC). MicroRNAs have been shown to be involved in the development of many cancers, but little is known about their role in the process of colorectal liver metastasis. We compared miRNA expression between primary colorectal tumors and liver metastasis to identify those involved in the process of metastasis. Cancer cells were isolated from formalin-fixed paraffin-embedded primary CRC samples and their corresponding metastatic liver tumors in six patients using laser capture microdissection, and miRNA expression was analyzed using TaqMan miRNA arrays. The most abundant miRNA in liver metastasis compared with primary tumors was miR-122. Immunohistochemical analysis revealed that the expression levels of cationic amino acid transporter 1 (CAT1), a negative target gene of miR-122, were lower in liver metastases than primary tumors (P < 0.001). Expression levels of CAT1 in 132 primary tumors were negatively correlated with the existence of synchronous liver metastasis (P = 0.0333) and tumor stage (P < 0.0001). In an analysis of 121 colon cancer patients without synchronous liver metastasis, patients with CAT1-low colon cancer had significantly shorter liver metastasis-free survival (P = 0.0258) but not overall survival or disease-free survival. Overexpression of miR-122 and concomitant suppression of CAT1 in the primary tumor appears to play important roles in the development of colorectal liver metastasis. Expression of CAT1 in the primary CRC has the potential to be a novel biomarker to predict the risk of postoperative liver metastasis of CRC patients.
Collapse
Affiliation(s)
- Ichirota Iino
- Second Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Differential topochemistry of three cationic amino acid transporter proteins, hCAT1, hCAT2 and hCAT3, in the adult human brain. Amino Acids 2012; 44:423-33. [DOI: 10.1007/s00726-012-1348-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Accepted: 06/20/2012] [Indexed: 11/26/2022]
|
19
|
Abdelmagid SA, Rickard JA, McDonald WJ, Thomas LN, Too CKL. CAT-1-mediated arginine uptake and regulation of nitric oxide synthases for the survival of human breast cancer cell lines. J Cell Biochem 2011; 112:1084-92. [PMID: 21308737 DOI: 10.1002/jcb.23022] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Growth of the human MCF-7 breast cancer cell line is highly dependent on L-arginine. We have reported that L-arginine, released from extracellular substrates by prolactin (PRL)- and 17β-estradiol (E2)-induced carboxypeptidase-D in the cell membrane, promotes nitric oxide (NO) production for MCF-7 cell survival. Arginine uptake is mediated by members of the cationic amino acid transporter (CAT) family and may coincide with induction of nitric oxide synthase (NOS) for the production of NO. The present study investigated the CAT isoforms and PRL/E2 regulation of CAT and NOS in breast cancer cell lines. Using RT-PCR analysis, CAT-1, CAT-2A, and CAT-2B transcripts were detected in MCF-7, T47D, and MDA-MB-231 cells. The CAT-4 transcript was detected in MDA-MB-231 only. CAT-3 was not detected in any of these cells. PRL and E2 did not significantly alter levels of CAT-1 mRNA and protein, nor CAT-2A and CAT-2B mRNAs in MCF-7 and T47D cells. PRL and E2 also had no effect on the overall uptake of L-[2,3,4,5-H(3)] arginine into these cells. However, confocal immunofluorescent microscopy showed that PRL and E2 upregulated eNOS and iNOS proteins, which distributed in the cytoplasm and/or nucleus of MCF-7 cells. Knockdown of CAT-1 gene expression using small interfering RNA significantly decreased L-[2,3,4,5-H(3)]-arginine uptake, decreased viability and increased apoptosis of MCF-7 and T47D cells. In summary, several CAT isoforms are expressed in breast cancer cells. The CAT-1 isoform plays a role in arginine uptake and, together with PRL/E2-induced NOS, contribute to NO production for the survival of MCF-7 and T47D cells.
Collapse
Affiliation(s)
- Salma A Abdelmagid
- Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, Nova Scotia B3H 1X5, Canada
| | | | | | | | | |
Collapse
|
20
|
McConathy J, Yu W, Jarkas N, Seo W, Schuster DM, Goodman MM. Radiohalogenated nonnatural amino acids as PET and SPECT tumor imaging agents. Med Res Rev 2011; 32:868-905. [DOI: 10.1002/med.20250] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Jonathan McConathy
- Mallinckrodt Institute of Radiology; Washington University School of Medicine; St. Louis Missouri
| | - Weiping Yu
- Department of Radiology and Imaging Sciences; School of Medicine, Emory University; Atlanta Georgia
| | - Nachwa Jarkas
- Department of Radiology and Imaging Sciences; School of Medicine, Emory University; Atlanta Georgia
| | - Wonewoo Seo
- Department of Radiology and Imaging Sciences; School of Medicine, Emory University; Atlanta Georgia
| | - David M. Schuster
- Department of Radiology and Imaging Sciences; School of Medicine, Emory University; Atlanta Georgia
| | - Mark M. Goodman
- Department of Radiology and Imaging Sciences; School of Medicine, Emory University; Atlanta Georgia
| |
Collapse
|
21
|
Griselda CM. d-Arginine action against neurotoxicity induced by glucocorticoids in the brain. Neurosci Biobehav Rev 2011; 35:1353-62. [DOI: 10.1016/j.neubiorev.2011.02.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2010] [Revised: 01/13/2011] [Accepted: 02/15/2011] [Indexed: 11/30/2022]
|
22
|
Inbar E, Canepa GE, Carrillo C, Glaser F, Suter Grotemeyer M, Rentsch D, Zilberstein D, Pereira CA. Lysine transporters in human trypanosomatid pathogens. Amino Acids 2010; 42:347-60. [PMID: 21170560 DOI: 10.1007/s00726-010-0812-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2010] [Accepted: 11/10/2010] [Indexed: 11/30/2022]
Abstract
In previous studies we characterized arginine transporter genes from Trypanosoma cruzi and Leishmania donovani, the etiological agents of chagas disease and kala azar, respectively, both fatal diseases in humans. Unlike arginine transporters in higher eukaryotes that transport also lysine, these parasite transporters translocate only arginine. This phenomenon prompted us to identify and characterize parasite lysine transporters. Here we demonstrate that LdAAP7 and TcAAP7 encode lysine-specific permeases in L. donovani and T. cruzi, respectively. These two lysine permeases are both members of the large amino acid/auxin permease family and share certain biochemical properties, such as specificity and Km. However, we evidence that LdAAP7 and TcAAP7 differ in their regulation and localization, such differences are likely a reflection of the dissimilar L. donovani and T. cruzi life cycles. Failed attempts to delete both alleles of LdAAP7 support the premise that this is an essential gene that encodes the only lysine permeases expressed in L. donovani promastigotes and T. cruzi epimastigotes, respectively.
Collapse
Affiliation(s)
- Ehud Inbar
- Faculty of Biology, Technion-Israel Institute of Technology, 32000, Haifa, Israel
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Functional characterization of the chicken cationic amino acid transporter-2 isoforms. Comp Biochem Physiol B Biochem Mol Biol 2010; 156:279-86. [DOI: 10.1016/j.cbpb.2010.04.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2009] [Revised: 03/09/2010] [Accepted: 04/13/2010] [Indexed: 11/18/2022]
|
24
|
Meyer E, Manahan DT. Nutrient uptake by marine invertebrates: cloning and functional analysis of amino acid transporter genes in developing sea urchins (Strongylocentrotus purpuratus). THE BIOLOGICAL BULLETIN 2009; 217:6-24. [PMID: 19679719 DOI: 10.1086/bblv217n1p6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Transport of amino acids from low concentrations in seawater by marine invertebrates has been extensively studied, but few of the genes involved in this physiological process have been identified. We have characterized three amino acid transporter genes cloned from embryos of the sea urchin Strongylocentrotus purpuratus. These genes show phylogenetic proximity to classical amino acid transport systems, including Gly and B0+, and the inebriated gene (INE). Heterologous expression of these genes in frog oocytes induced a 40-fold increase in alanine transport above endogenous levels, demonstrating that these genes mediate alanine transport. Antibodies specific to one of these genes (Sp-AT1) inhibited alanine transport, confirming the physiological activity of this gene in larvae. Whole-mount antibody staining of larvae revealed expression of Sp-AT1 in the ectodermal tissues associated with amino acid transport, as independently demonstrated by autoradiographic localization of radioactive alanine. Maximum rates of alanine transport increased 6-fold during early development, from embryonic to larval stages. Analysis of gene expression during this developmental period revealed that Sp-AT1 transcript abundance remained nearly constant, while that of another transporter gene (Sp-AT2) increased 11-fold. The functional characterization of these genes establishes a molecular biological basis for amino acid transport by developmental stages of marine invertebrates.
Collapse
Affiliation(s)
- Eli Meyer
- Department of Biological Sciences, University of Southern California, Los Angeles, California 90089-0371, USA
| | | |
Collapse
|
25
|
Chan CH, Ramirez-Montealegre D, Pearce DA. Altered arginine metabolism in the central nervous system (CNS) of the Cln3-/- mouse model of juvenile Batten disease. Neuropathol Appl Neurobiol 2009; 35:189-207. [PMID: 19284480 DOI: 10.1111/j.1365-2990.2008.00984.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND Juvenile neuronal ceroid lipofuscinoses (JNCL) or juvenile Batten disease is a recessively inherited childhood neurodegenerative disorder resulting from a mutation in CLN3, which encodes a putative lysosomal protein of unknown function. AIM Recent evidence suggests that a disruption in CLN3 function results in altered regulation of arginine transport into lysosomes, and may influence intracellular arginine levels. We sought to investigate the possible consequences of arginine dysregulation in the brain of the Cln3(-/-) mouse model of JNCL. METHODS Using a combination of enzyme assays, metabolite profiling, quantitative reverse-transcription polymerase chain reaction and Western blotting, we analysed the activities and expression of enzymes involved in arginine metabolism in the cerebral cortex and cerebellum of Cln3(-/-) mice over several developmental time points. RESULTS We report subtle, but significant changes in the activities of enzymes involved in the citrulline-NO recycling pathway, and altered regulation of neuronal nitric oxide synthase in the cortex and cerebellum of Cln3(-/-) mice. In addition, a significant decrease in arginine transport into cerebellar granule cells was observed, despite an apparent upregulation of the cationic amino acid transporter-1 transporter at the cell surface. Our results provide further evidence that CLN3 function and arginine homeostasis are intricately related, and that cellular mechanisms may act to compensate for the loss of this protein. CONCLUSIONS This and other studies indicate that CLN3 dysfunction in JNCL may result in multiple disturbances in metabolism that together contribute to the pathophysiological processes underlying this disease.
Collapse
Affiliation(s)
- C-H Chan
- Center for Neural Development and Disease, University of Rochester School of Medicine and Dentistry, Rochester 14642, USA
| | | | | |
Collapse
|
26
|
Humphrey BD, Kirsch S, Morris D. Molecular cloning and characterization of the chicken cationic amino acid transporter-2 gene. Comp Biochem Physiol B Biochem Mol Biol 2008; 150:301-11. [DOI: 10.1016/j.cbpb.2008.03.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2007] [Revised: 03/24/2008] [Accepted: 03/31/2008] [Indexed: 11/26/2022]
|
27
|
Eros D, Orfi L, Csuka I, Kéri G, Hrabák A. Binding specificity of the L-arginine transport systems in mouse macrophages and human cells overexpressing the cationic amino acid transporter hCAT-1. Amino Acids 2008; 36:483-92. [PMID: 18504524 DOI: 10.1007/s00726-008-0106-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2007] [Accepted: 05/06/2008] [Indexed: 11/29/2022]
Abstract
The uptake of L-arginine into mouse peritoneal macrophages can be inhibited by numerous amino acids and derivatives. Kinetic studies showed an almost entirely competitive inhibition for both cationic and neutral amino acids and derivatives suggesting that the comparison of their binding specificity by using a quantitative structure-activity relationship (QSAR) study is reasonable. The properties of the most efficient inhibitors were the following: the length of the aliphatic side chain, a general structural similarity to L-arginine (>0.79), cationic character, L-configuration, the presence of an alpha-amino group (with a mean pK(a) of 9.41), the van der Waals volume (mean 225 A(3)) and a low logP value (mean: -2.99). The significance of four other descriptors (neutral character, presence and the pK(a) of an alpha-carboxyl group, and the presence of a modified guanidino group) is much lower. Similar results were obtained for the hCAT-1 cell line, but the significance of the descriptors was slightly different. The L-configuration, van der Waals volume, the low logP value and the length of aliphatic side chain were the most significant, while the pK(a) value of the side chain (mean pK(a)=11.6) was found to be more important than that of the alpha-amino group. In addition, the general similarity to L-arginine, the presence of an amino group in the terminal position of the side chain (Orn, Lys) and the basic character were significant descriptors, while the significance of the acidity is negligibly low. As a final conclusion, the following descriptors were found to be important generally for the cationic transporters: the van der Waals volume, hydrophobicity (log P); L-configuration; the size of the side chain; the general similarity to L-arginine; the presence of an alpha-amino group; the general basicity of the molecule; the pK(a) values of the alpha-amino group (in macrophages) or that of the side chain (in CAT-1 cells). These descriptors can be regarded as the general structurally important binding characteristics of the cationic amino transporters.
Collapse
Affiliation(s)
- Dániel Eros
- Vichem Ltd., II. Herman Ottó u. 15, 1022 Budapest, Hungary
| | | | | | | | | |
Collapse
|
28
|
Levin-Iaina N, Schwartz I, Chernichovsky T, Davidovitch A, Iaina A, Schwartz D. Tubular and glomerular L-arginine transport (uptake and transporters) and the nitric oxide synthases in ischemic acute renal failure (iARF) in streptozotocin-induced diabetic rats (STZ-DM). Ren Fail 2008; 29:1031-8. [PMID: 18067052 DOI: 10.1080/08860220701641744] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND L-arginine or its metabolites may be important pathogenetic factors in ischemic acute renal failure (iARF) in rats. It was found that the L-arginine-nitric oxide synthase-nitric oxide system plays an important role in the renal hemodynamic alterations in the early stages of diabetes. The iARF in diabetic rats is much more severe than the normal rats exposed to a same ischemia time. The purpose of the present study was to evaluated L-arginine uptake and its transporters and nitric oxide synthase isoform expression in tubuli and glomeruli of STZ-induced diabetic rats with iARF. METHODS iARF was induced by right nephrectomy and left renal artery clamping for 60 min followed by a 60 min reflow period. iARF was induced in STZ diabetes rats two weeks after intraperitoneal streptozotocin (60 mg/kg body weight) and in normal control rats. L-arginine uptake, L-arginine transporters (CAT1 and CAT2) and nitric oxide synthases (iNOS, eNOS, and bNOS) were determined by RT-PCR) in both glomeruli and tubuli preparations. RESULTS The STZ diabetic rats compared with the non diabetic normal rats have a higher glomerular L-arginine uptake, higher iNOS mRNA, lower eNOS mRNA, and lower tubular CAT1 mRNA, eNOS mRNA, and bNOS mRNA. The diabetic iARF after one hour of reperfusion had lower glomerular L-arginine uptake, lower CAT1 mRNA, lower eNOS mRNA, lower bNOS, and higher tubular iNOS mRNA compared with iARF in normal rats. CONCLUSIONS Our findings suggest a prolonged and more severe post-glomerular vasoconstriction very early after the reflow in the iARF of STZ diabetic rats compared with the iARF in the normal control rats. That may be a plausible explanation to the very significant decline in GFR and tubular necrosis that characterize the iARF in diabetic rats.
Collapse
Affiliation(s)
- Nomy Levin-Iaina
- Department of Nephrology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.
| | | | | | | | | | | |
Collapse
|
29
|
Closs EI, Boissel JP, Habermeier A, Rotmann A. Structure and Function of Cationic Amino Acid Transporters (CATs). J Membr Biol 2007; 213:67-77. [PMID: 17417706 DOI: 10.1007/s00232-006-0875-7] [Citation(s) in RCA: 177] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2006] [Indexed: 11/29/2022]
Abstract
The CAT proteins (CAT for cationic amino acid transporter) are amongst the first mammalian amino acid transporters identified on the molecular level and seem to be the major entry path for cationic amino acids in most cells. However, CAT proteins mediate also efflux of their substrates and thus may also deplete cells from cationic amino acids under certain circumstances. The CAT proteins form a subfamily of the solute carrier family 7 (SLC7) that consists of four confirmed transport proteins for cationic amino acids: CAT-1 (SLC7A1), CAT-2A (SLC7A2A), CAT-2B (SLC7A2B), and CAT-3 (SLC7A3). SLC7A4 and SLC7A14 are two related proteins with yet unknown function. One focus of this review lies on structural and functional differences between the different CAT isoforms. The expression of the CAT proteins is highly regulated on the level of transcription, mRNA stability, translation and subcellular localization. Recent advances toward a better understanding of these mechanisms provide a second focus of this review.
Collapse
Affiliation(s)
- E I Closs
- Department of Pharmacology, Johannes Gutenberg University, Obere Zahlbacher Str. 67, D-55101 Mainz, Germany.
| | | | | | | |
Collapse
|
30
|
Rotmann A, Simon A, Martiné U, Habermeier A, Closs EI. Activation of classical protein kinase C decreases transport via systems y+ and y+L. Am J Physiol Cell Physiol 2007; 292:C2259-68. [PMID: 17329401 DOI: 10.1152/ajpcell.00323.2006] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Activation of protein kinase C (PKC) downregulates the human cationic amino acid transporters hCAT-1 (SLC7A1) and hCAT-3 (SLC7A3) (Rotmann A, Strand D, Martiné U, Closs EI. J Biol Chem 279: 54185-54192, 2004; Rotmann A, Vekony N, Gassner D, Niegisch G, Strand D, Martine U, Closs EI. Biochem J 395: 117-123, 2006). However, others found that PKC increased arginine transport in various mammalian cell types, suggesting that the expression of different arginine transporters might be responsible for the opposite PKC effects. We thus investigated the consequence of PKC activation by phorbol-12-myristate-13-acetate (PMA) in various human cell lines expressing leucine-insensitive system y(+) [hCAT-1, hCAT-2B (SLC7A2), or hCAT-3] as well as leucine-sensitive system y(+)L [y(+)LAT1 (SLC7A7) or y(+)LAT2 (SLC7A6)] arginine transporters. PMA reduced system y(+) activity in all cell lines tested, independent of the hCAT isoform expressed, while mRNAs encoding the individual hCAT isoforms were either unchanged or increased. System y(+)L activity was also inhibited by PMA. The extent and onset of inhibition varied between cell lines; however, a PMA-induced increase in arginine transport was never observed. In addition, when expressed in Xenopus laevis oocytes, y(+)LAT1 and y(+)LAT2 activity was reduced by PMA, and this inhibition could be prevented by the PKC inhibitor bisindolylmaleimide I. In ECV304 cells, PMA-induced inhibition of systems y(+) and y(+)L could be prevented by Gö6976, a specific inhibitor of conventional PKCs. Thymelea toxin, which activates preferentially classical PKC, had a similar inhibitory effect as PMA. In contrast, phosphatidylinositol-3,4,5-triphosphate-dipalmitoyl, an activator of atypical PKC, had no effect. These data demonstrate that systems y(+) and y(+)L are both downregulated by classical PKC.
Collapse
Affiliation(s)
- Alexander Rotmann
- Dept. of Pharmacology, Johannes Gutenberg University, Obere Zahlbacher Str. 67, 55101 Mainz, Germany
| | | | | | | | | |
Collapse
|
31
|
Huang Y, Kang BN, Tian J, Liu Y, Luo HR, Hester L, Snyder SH. The cationic amino acid transporters CAT1 and CAT3 mediate NMDA receptor activation-dependent changes in elaboration of neuronal processes via the mammalian target of rapamycin mTOR pathway. J Neurosci 2007; 27:449-58. [PMID: 17234578 PMCID: PMC6672784 DOI: 10.1523/jneurosci.4489-06.2007] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Neuronal activity influences protein synthesis and neuronal growth. Availability of nutrients, especially leucine and arginine, regulates the mammalian target of rapamycin (mTOR) pathway that controls cell growth. We show that NMDA receptor activation markedly reduces arginine transport by decreasing surface expression of the cationic amino acid transporters (CAT) 1 and 3. Depletion of CAT1 and CAT3 by RNA interference blocks influences of NMDA receptor activation on the mTOR pathway and neuronal process formation. Thus, the CATs mediate influences of NMDA receptor activation on the mTOR pathway that regulates neuronal processes.
Collapse
Affiliation(s)
- Yunfei Huang
- The Solomon H. Snyder Department of Neuroscience and
| | - Bingnan N. Kang
- The Solomon H. Snyder Department of Neuroscience and
- Departments of Pharmacology and Molecular Sciences and
| | - Jing Tian
- The Solomon H. Snyder Department of Neuroscience and
| | - Yi Liu
- The Solomon H. Snyder Department of Neuroscience and
| | - Hongbo R. Luo
- Department of Pathology, Harvard Medical School, Boston, Massachusetts 02115
| | - Lynda Hester
- The Solomon H. Snyder Department of Neuroscience and
| | - Solomon H. Snyder
- The Solomon H. Snyder Department of Neuroscience and
- Departments of Pharmacology and Molecular Sciences and
- Psychiatry and Behavioral Sciences, Johns Hopkins University, School of Medicine, Baltimore, Maryland 21205, and
| |
Collapse
|
32
|
Cérec V, Piquet-Pellorce C, Aly HAA, Touzalin AM, Jégou B, Bauché F. Multiple Pathways for Cationic Amino Acid Transport in Rat Seminiferous Tubule Cells1. Biol Reprod 2007; 76:241-9. [PMID: 17065601 DOI: 10.1095/biolreprod.106.056168] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Arginine and ornithine are known to be important for various biological processes in the testis, but the delivery of extracellular cationic amino acids to the seminiferous tubule cells remains poorly understood. We investigated the activity and expression of cationic amino acid transporters in isolated rat Sertoli cells, peritubular cells, pachytene spermatocytes, and early spermatids. We assessed the l-arginine uptake kinetics, Na(+) dependence of transport, profiles of cis inhibition of uptake by cationic and neutral amino acids, and sensitivity to trans stimulation of cationic amino acid transporters, and studied the expression of the genes encoding them by RT-PCR. Our data suggest that l-arginine is taken up by Sertoli cells and peritubular cells, principally via system y(+)L (SLC3A2/SLC7A6) and system y(+) (SLC7A1 and SLC7A2), with system B(0+) making a minor contribution. By contrast, system B(0+), associated with system y(+)L (SLC3A2/SLC7A7 and SLC7A6), made a major contribution to the transport of cationic amino acids in pachytene spermatocytes and early spermatids. Sertoli cells had higher rates of l-arginine transport than the other seminiferous tubule cells. This high efficiency of arginine transport in Sertoli cells and the properties of the y(+)L system predominating in these cells strongly suggest that Sertoli cells play a key role in supplying germ cells with l-arginine and other cationic amino acids. Furthermore, whereas cytokines induce nitric oxide (NO) production in peritubular and Sertoli cells, little or no upregulation of arginine transport by cytokines was observed in these cells. Thus, NO synthesis does not depend on the stimulation of arginine transport in these somatic tubular cells.
Collapse
|
33
|
Yeramian A, Martin L, Arpa L, Bertran J, Soler C, McLeod C, Modolell M, Palacín M, Lloberas J, Celada A. Macrophages require distinct arginine catabolism and transport systems for proliferation and for activation. Eur J Immunol 2006; 36:1516-26. [PMID: 16703566 DOI: 10.1002/eji.200535694] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
In murine macrophages, as a result of arginine catabolism during activation, citruline is produced under the effect of IFN-gamma and LPS, and ornithine and polyamines by IL-4 and IL-10. For proliferation, arginine is required from the extracellular medium and is used for protein synthesis. During activation, most arginine (>95% in 6 h) was metabolized, while under proliferation only half was incorporated into proteins. Under basal conditions, this amino acid was preferentially transported by y(+)L activity. During activation, arginine transport increased drastically (4-5-fold) through y(+) cationic amino acid transporter (CAT) activity. By contrast, M-CSF induced only a modest increase in uptake (0.5-fold). The increase in arginine transport during activation, but not proliferation, was mediated by the SLC7A2/Cat2 gene. SLC7A1/Cat1 is constitutively expressed, and is not modified by proliferating or activating agents. M-CSF-dependent proliferation was not affected in the macrophages of SLC7A2 knockout mice; however, these cells showed a drastic reduction in the production of citruline or ornithine and polyamines during activation. The data show that a large increase in a specific transport system (CAT2) is necessary for activation-induced arginine metabolism, while arginine is in excess for the requirements of proliferation and a modest increase in transport occurs.
Collapse
Affiliation(s)
- Andrée Yeramian
- Macrophage Biology Group, Institute of Research in Biomedicine, Barcelona Science Park, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Yeramian A, Martin L, Serrat N, Arpa L, Soler C, Bertran J, McLeod C, Palacín M, Modolell M, Lloberas J, Celada A. Arginine transport via cationic amino acid transporter 2 plays a critical regulatory role in classical or alternative activation of macrophages. THE JOURNAL OF IMMUNOLOGY 2006; 176:5918-24. [PMID: 16670299 DOI: 10.4049/jimmunol.176.10.5918] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Arginine is processed by macrophages in response to the cytokines to which these cells are exposed. Th1-type cytokines induce NO synthase 2, which metabolizes arginine into nitrites, while the Th2-type cytokines produce arginase, which converts arginine into polyamines and proline. Activation of bone marrow-derived macrophages by these two types of cytokines increases L-arginine transport only through the y(+) system. Analysis of the expression of the genes involved in this system showed that Slc7A1, encoding cationic amino acid transporters (CAT)1, is constitutively expressed and is not modified by activating agents, while Slc7A2, encoding CAT2, is induced during both classical and alternative activation. Macrophages from Slc7A2 knockout mice showed a decrease in L-arginine transport in response to the two kinds of cytokines. However, while NO synthase 2 and arginase expression were unmodified in these cells, the catabolism of arginine was impaired by both pathways, producing smaller amounts of nitrites and also of polyamines and proline. In addition, the induction of Slc7A2 expression was independent of the arginine available and of the enzymes that metabolize it. In conclusion, the increased arginine transport mediated by activators is strongly regulated by CAT2 expression, which could limit the function of macrophages.
Collapse
Affiliation(s)
- Andrée Yeramian
- Macrophage Biology Group, Institute of Biomedical Research, Barcelona Science Park, Josep Samitier 1-5, E-08028 Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Rotmann A, Vékony N, Gassner D, Niegisch G, Strand D, Martiné U, Closs E. Activation of classical protein kinase C reduces the expression of human cationic amino acid transporter 3 (hCAT-3) in the plasma membrane. Biochem J 2006; 395:117-23. [PMID: 16332251 PMCID: PMC1409692 DOI: 10.1042/bj20051558] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
We have previously shown that activation of PKC (protein kinase C) results in internalization of hCAT-1 [human CAT-1 (cationic amino acid transporter 1)] and a decrease in arginine transport [Rotmann, Strand, Martiné and Closs (2004) J. Biol. Chem. 279, 54185-54192]. However, others found increased transport rates for arginine in response to PKC activation, suggesting a differential effect of PKC on different CAT isoforms. Therefore we investigated the effect of PKC on hCAT-3, an isoform expressed in thymus, brain, ovary, uterus and mammary gland. In Xenopus laevis oocytes and human U373MG glioblastoma cells, hCAT-3-mediated L-arginine transport was significantly reduced upon treatment with compounds that activate classical PKC. In contrast, inactive phorbol esters and an activator of novel PKC isoforms had no effect. PKC inhibitors (including the PKCalpha-preferring Ro 31-8280) reduced the inhibitory effect of the PKC-activating compounds. Microscopic analyses revealed a PMA-induced reduction in the cell-surface expression of fusion proteins between hCAT-3 and enhanced green fluorescent protein expressed in X. laevis oocytes and glioblastoma cells. Western-blot analysis of biotinylated surface proteins demonstrated a PMA-induced decrease in hCAT-3 in the plasma membrane, but not in total protein lysates. Pretreatment with a PKC inhibitor also reduced this PMA effect. It is concluded that similar to hCAT-1, hCAT-3 activity is decreased by PKC via reduction of transporter molecules in the plasma membrane. Classical PKC isoforms seem to be responsible for this effect.
Collapse
Affiliation(s)
- Alexander Rotmann
- *Department of Pharmacology, Johannes Gutenberg University, Obere Zahlbacher Strasse 67, 55101 Mainz, Germany
| | - Nicole Vékony
- *Department of Pharmacology, Johannes Gutenberg University, Obere Zahlbacher Strasse 67, 55101 Mainz, Germany
| | - Davina Gassner
- *Department of Pharmacology, Johannes Gutenberg University, Obere Zahlbacher Strasse 67, 55101 Mainz, Germany
| | - Günter Niegisch
- *Department of Pharmacology, Johannes Gutenberg University, Obere Zahlbacher Strasse 67, 55101 Mainz, Germany
| | - Dennis Strand
- †First Department of Internal Medicine, Johannes Gutenberg University, Obere Zahlbacher Strasse 63, 55101 Mainz, Germany
| | - Ursula Martiné
- *Department of Pharmacology, Johannes Gutenberg University, Obere Zahlbacher Strasse 67, 55101 Mainz, Germany
| | - Ellen I. Closs
- *Department of Pharmacology, Johannes Gutenberg University, Obere Zahlbacher Strasse 67, 55101 Mainz, Germany
- To whom correspondence should be addressed (email )
| |
Collapse
|
36
|
Martín L, Comalada M, Marti L, Closs EI, MacLeod CL, Martín del Río R, Zorzano A, Modolell M, Celada A, Palacín M, Bertran J. Granulocyte-macrophage colony-stimulating factor increases L-arginine transport through the induction of CAT2 in bone marrow-derived macrophages. Am J Physiol Cell Physiol 2005; 290:C1364-72. [PMID: 16371438 DOI: 10.1152/ajpcell.00520.2005] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
L-arginine transport is crucial for macrophage activation because it supplies substrate for the key enzymes nitric oxide synthase 2 and arginase I. These enzymes participate in classic and alternative activation of macrophages, respectively. Classic activation of macrophages is induced by type I cytokines, and alternative activation is induced by type II cytokines. The granulocyte macrophage colony-stimulating factor (GM-CSF), in addition to inducing proliferation and differentiation of macrophages, activates arginase I, but its action on L-arginine transport is unknown. We studied the L-arginine transporters that are active in mouse primary bone marrow-derived macrophages (BMM) and examined the effect of GM-CSF treatment on transport activities. Under basal conditions, L-arginine entered mainly through system y(+)L (>75%). The remaining transport was explained by system y(+) (<10%) and a diffusion component (10-15%). In response to GM-CSF treatment, transport activity increased mostly through system y(+) (>10-fold), accounting for about 40% of the total L-arginine transport. The increase in y(+) activity correlated with a rise in cationic amino acid transporter (CAT)-2 mRNA and protein. Furthermore, GM-CSF induced an increase in arginase activity and in the conversion of L-arginine to ornithine, citrulline, glutamate, proline, and polyamines. BMM obtained from CAT2-knockout mice responded to GM-CSF by increasing arginase activity and the expression of CAT1 mRNA, which also encodes system y(+) activity. Nonetheless, the increase in CAT1 activity only partially compensated the lack of CAT2 and L-arginine metabolism was hardly stimulated. We conclude that BMM present mainly y(+)L activity and that, in response to GM-CSF, l-arginine transport augments through CAT2, thereby increasing the availability of this amino acid to the cell.
Collapse
Affiliation(s)
- Lorena Martín
- Department of Biochemistry and Molecular Biology, University of Barcelona, Avenida Diagonal 645, Barcelona E-08028, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Cui Z, Zharikov S, Xia SL, Anderson SI, Law AS, Archibald AL, Block ER. Molecular cloning, characterization, and chromosomal assignment of porcine cationic amino acid transporter-1. Genomics 2005; 85:352-9. [PMID: 15718102 DOI: 10.1016/j.ygeno.2004.11.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2004] [Accepted: 11/10/2004] [Indexed: 10/26/2022]
Abstract
We have cloned and characterized the gene encoding the porcine cationic amino acid transporter, member 1 (CAT-1) (HGMW-approved gene symbol SLC7A1) from porcine pulmonary artery endothelial cells. The porcine SLC7A1 encodes 629 deduced amino acid residues showing a higher degree of sequence similarity with the human counterpart (91.1%) than with the rat (87.3%) and mouse (87.6%) counterparts. Confocal microscopic examination of porcine CAT-1-GFP-expressing HEK293 cells revealed that porcine CAT-1 localizes on the plasma membrane. Amino acid uptake studies in Xenopus oocytes injected with cRNA encoding this protein demonstrated transport properties consistent with system y(+). Radiation hybrid mapping data indicate that the porcine SLC7A1 maps to the distal end of the short arm of pig chromosome 11 (SSC11). This map location is consistent with the known conservation of genome organization between human and pig and provides further confirmation that we have characterized the porcine orthologue of the human SLC7A1.
Collapse
Affiliation(s)
- Zhaoqiang Cui
- Department of Medicine, University of Florida, Gainesville, FL 32610, USA
| | | | | | | | | | | | | |
Collapse
|
38
|
Gilles W, Vulcu SD, Liewald JF, Habermeier A, Vékony N, Closs EI, Rupp J, Nawrath H. Monovalent cation conductance in Xenopus laevis oocytes expressing hCAT-3. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2005; 1668:234-9. [PMID: 15737334 DOI: 10.1016/j.bbamem.2004.12.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2004] [Revised: 12/17/2004] [Accepted: 12/20/2004] [Indexed: 11/18/2022]
Abstract
hCAT-3 (human cationic amino acid transporter type three) was investigated with both the two-electrode voltage clamp method and tracer experiments. Oocytes expressing hCAT-3 displayed less negative membrane potentials and larger voltage-dependent currents than native or water-injected oocytes did. Ion substitution experiments in hCAT-3-expressing oocytes revealed a large conductance for Na+ and K+. In the presence of L-Arg, voltage-dependent inward and outward currents were observed. At symmetrical (inside/outside) concentrations of L-Arg, the conductance of the transporter increased monoexponentially with the L-Arg concentrations; the calculated Vmax and KM values amounted to 8.3 microS and 0.36 mM, respectively. The time constants of influx and efflux of [3H]L-Arg, at symmetrically inside/outside L-Arg concentrations (1 mM), amounted to 79 and 77 min, respectively. The flux data and electrophysiological experiments suggest that the transport of L-Arg through hCAT-3 is symmetric, when the steady state of L-Arg flux has been reached. It is concluded that hCAT-3 is a passive transport system that conducts monovalent cations including L-Arg. The particular role of hCAT-3 in the diverse tissues remains to be elucidated.
Collapse
Affiliation(s)
- Wolfgang Gilles
- Department of Pharmacology, Johannes-Gutenberg University, Obere Zahlbacher Str. 67, 55101 Mainz, Germany
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Armer RE, Miller DJ. Advances in potential therapeutic uses of inhibitors of CNS selective amino acid transporters. Expert Opin Ther Pat 2005. [DOI: 10.1517/13543776.11.4.563] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
40
|
Bae SY, Xu Q, Hutchinson D, Colton CA. Y+ and y+ L arginine transporters in neuronal cells expressing tyrosine hydroxylase. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2005; 1745:65-73. [PMID: 16085056 DOI: 10.1016/j.bbamcr.2004.12.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2004] [Revised: 12/28/2004] [Accepted: 12/28/2004] [Indexed: 11/18/2022]
Abstract
Arginine is a semi-essential amino acid that serves as sole substrate for enzymes involved in diverse cell processes including redox balance via nitric oxide synthase (NOS) and cell proliferation via arginase. Neurons that express nNOS require intracellular arginine to generate nitric oxide (NO). Using a TH+ neuronal cell line (CAD cells), we show that neuronal NO production is largely dependent on extracellular arginine. Although a small intracellular pool exists in CAD cells, the lack of mRNA for argininosuccinate synthase (AS), a rate limiting enzyme for arginine recycling, suggests that intracellular pools are not re-supplied by this mechanism in this sub-class of neurons. Rather, arginine is taken up from the extracellular media by two primary transport systems, the y+ and the y+ L systems. The expression of CAT1, CAT3, y+ LAT1 and y+ LAT2 mRNAs supports the presence of each system. CAD cell arginine transport is depressed by increased extracellular K+ levels and demonstrates that variations in membrane potential control neuronal arginine uptake. Short term exposure to the oxidizing agents, rotenone and Angeli's salt, but not FeSO4, increases arginine transport. The regulation of arginine uptake by physiological factors suggests that arginine supply adapts in a moment-to-moment fashion to the changing needs of the neuron.
Collapse
Affiliation(s)
- S Y Bae
- Division of Neurology, Box 2900, Bryan Research Bldg, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | |
Collapse
|
41
|
Cui Z, Tuladhar R, Hart SL, Marber MS, Pearson JD, Baydoun AR. Rate of transport of l-arginine is independent of the expression of inducible nitric oxide synthase in HEK 293 cells. Nitric Oxide 2004; 12:21-30. [PMID: 15631944 DOI: 10.1016/j.niox.2004.11.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2004] [Revised: 10/19/2004] [Accepted: 11/08/2004] [Indexed: 11/24/2022]
Abstract
Expression of inducible nitric oxide synthase (iNOS) is generally accompanied by a parallel upregulation in l-arginine transport which is dependent, at least in part, on the synthesis of new carrier proteins. It is not clear however whether the induction of iNOS and its subsequent utilisation of l-arginine for NO synthesis contribute to the enhancement in l-arginine transport rates observed following induction of cells with pro-inflammatory mediators. To address this issue, we have transfected an iNOS construct in a pEGFP-N1 vector into HEK-293 cells and investigated the effects this has on l-arginine transport. The expression of iNOS through transfection resulted in the production of significant quantities of NO as detected by the standard Griess assay. Under these conditions, the transport of l-arginine was found to be unaltered, with rate of uptake being comparable in both transfected and non-transfected cells. Characterisation of the transporter(s) involved with uptake of l-arginine revealed features characteristic of the classical cationic amino acid transport system y(+). Further analysis of the expression profile of the cationic amino acid transporter (CAT) involved revealed the presence of transcripts for CAT-1 and CAT-2B. These data demonstrate that iNOS activity does not drive or enhance l-arginine transport despite the fact that HEK-293 cells transport l-arginine via the CATs, including CAT-2B which is thought to be critical for supply of substrate to iNOS.
Collapse
Affiliation(s)
- Zhaoqiang Cui
- Department of Biosciences, University of Hertfordshire, College Lane, Herts AL10 9AB, UK
| | | | | | | | | | | |
Collapse
|
42
|
Hatzoglou M, Fernandez J, Yaman I, Closs E. Regulation of cationic amino acid transport: the story of the CAT-1 transporter. Annu Rev Nutr 2004; 24:377-99. [PMID: 15459982 DOI: 10.1146/annurev.nutr.23.011702.073120] [Citation(s) in RCA: 150] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The discovery of the function of the receptor for the ecotropic retrovirus as a membrane transporter for the essential amino acids lysine and arginine was a landmark finding in the field of molecular nutrition. This finding indicated that cationic amino acid transporters (CATs) act pathologically as viral receptors. The importance of this transporter was further supported by knockout mice that were not viable after birth. CAT-1 was the first amino acid transporter to be cloned; several other CATs were later characterized biochemically and molecularly. These transporters mediate the bidirectional transport of cationic amino acids, thus supporting important metabolic functions, such as synthesis of proteins, nitric oxide (NO) synthesis, polyamine biosynthesis, and interorgan amino acid flow. This review briefly describes the advances in the regulation of cationic amino acid transport, focusing on the molecular mechanisms that regulate the CAT-1 transporter. Of particular interest to this review is the regulation of CAT-1 by nutritional stresses, such as amino acid availability. The studies that are reviewed conclude that the CAT-1 gene is essential for cell survival during stress because it allows cells to resume growth as soon as amino acids become available.
Collapse
Affiliation(s)
- Maria Hatzoglou
- Department of Nutrition, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106, USA.
| | | | | | | |
Collapse
|
43
|
Schwartz IF, Iaina A, Benedict Y, Wollman Y, Chernichovski T, Brasowski E, Misonzhnik F, Ben-Dor A, Blum M, Levo Y, Schwartz D. Augmented arginine uptake, through modulation of cationic amino acid transporter-1, increases GFR in diabetic rats. Kidney Int 2004; 65:1311-9. [PMID: 15086470 DOI: 10.1111/j.1523-1755.2004.00508.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND It is suggested that either arginine or its metabolites, nitric oxide and polyamines play a role in the renal hemodynamic alterations observed in the early stages of diabetes. Yet, the regulation of arginine transport in diabetic kidneys has never been studied. METHODS Arginine uptake was determined in glomeruli harvested from control rats; diabetic rats (2 weeks following an intraperitoneal injection of streptozotocin, 60 mg/kg body weight); rats, 4 days following left nephrectomy (a nondiabetic model of hyperfiltration); diabetes + lysine (0.5% in the drinking water to attenuate arginine uptake); and control + lysine. RESULTS Glomerular arginine transport was significantly increased in diabetic rats, but remained unchanged following uninephrectomy. Lysine abolished the increase in arginine uptake in diabetic rats but had no effect in controls. The increase in creatinine clearance observed in diabetes was completely abolished by lysine. Using reverse transcription-polymerase chain reaction (RT-PCR), Northern blotting, and immunohistochemistry, we found a significant increase in glomerular cationic amino acid transporter-1 (CAT-1) expression in diabetic animals, which was unaffected by lysine. When human endothelial cells were incubated with arginine end products no effect on arginine transport was observed. However, only in the presence of 0.5 mM/L sodium nitroprusside (SNP) an augmented steady-state CAT-1 mRNA was demonstrated by RT-PCR. CONCLUSION In a rat model of early diabetes, glomerular arginine uptake is elevated through modulation of CAT-1 expression, thus, contributing to the pathogenesis of hyperfiltration. Increased nitric oxide formation may play a role in this process.
Collapse
Affiliation(s)
- Idit F Schwartz
- Nephrology Department, Tel Aviv Sourasky Medical Center, Tel Aviv University, Sackler School of Medicine, Tel Aviv, Israel.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Closs EI, Simon A, Vékony N, Rotmann A. Plasma membrane transporters for arginine. J Nutr 2004; 134:2752S-2759S; discussion 2765S-2767S. [PMID: 15465780 DOI: 10.1093/jn/134.10.2752s] [Citation(s) in RCA: 197] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The supply of arginine may become rate limiting for enzymatic reactions that use this semiessential amino acid as a substrate (e.g., nitric oxide, agmatine, creatine, and urea synthesis), particularly under conditions of high demand such as growth, sepsis, or wound healing. In addition, arginine acts as a signaling molecule that regulates essential cellular functions such as protein synthesis, apoptosis, and growth. In the past decade, a number of carrier proteins for amino acids have been identified on the molecular level. They belong to different gene families, exhibit overlapping but distinctive substrate specificities, and can further be distinguished by their requirement for the cotransport or countertransport of inorganic ions. A number of these transporters function as exchangers rather than uniporters. Uptake of amino acids by these transporters therefore depends largely on the intracellular substrate composition. Hence, there is a complex crosstalk between transporters for cationic and neutral amino acids as well as for peptides. This article briefly reviews current knowledge regarding mammalian plasma membrane transporters that accept arginine as a substrate.
Collapse
Affiliation(s)
- Ellen I Closs
- Department of Pharmacology, Johannes Gutenberg University, 55101 Mainz, Germany.
| | | | | | | |
Collapse
|
45
|
Humphrey BD, Stephensen CB, Calvert CC, Klasing KC. Glucose and cationic amino acid transporter expression in growing chickens (Gallus gallus domesticus). Comp Biochem Physiol A Mol Integr Physiol 2004; 138:515-25. [PMID: 15369841 DOI: 10.1016/j.cbpb.2004.06.016] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2004] [Revised: 06/23/2004] [Accepted: 06/23/2004] [Indexed: 11/25/2022]
Abstract
Tissue glucose transporter (GLUT1-3) and cationic amino acid transporter (CAT1-3) mRNA expression was determined in growing broiler chicks posthatch. In two experiments, tissues were either collected on days 1, 3 and 7 or days 1 and 14 posthatch. Heart and liver were the only tissues expressing a GLUT isoform on day 1. All tissues expressed a GLUT isoform on day 7 except for the thymus. Most tissues expressing a CAT isoform on day 1 decreased mRNA levels through day 7 (P<0.05), except for bursa CAT-1 which tended to increase (P=0.05). The thymus and spleen did not express any CAT isoform mRNA until day 7. The liver was the only tissue expressing GLUT-2 mRNA through day 14. On day 14, GLUT-1, CAT-1 and CAT-2 mRNA were differentially expressed across tissues (P<0.05). High-affinity GLUT and CAT mRNA expression was highest in the heart and bursa, respectively (P<0.05). Total CAT mRNA expression was greatest in the bursa (P<0.05). The thymus had the lowest high affinity GLUT and total CAT mRNA expression on day 14 posthatch. Therefore, T lymphocytes within the thymus may be most susceptible to glucose and cationic amino acid supply.
Collapse
Affiliation(s)
- Brooke D Humphrey
- Department of Animal Science, University of California, 1 Shields Avenue, Davis, CA 95616, USA
| | | | | | | |
Collapse
|
46
|
Tailor CS, Lavillette D, Marin M, Kabat D. Cell surface receptors for gammaretroviruses. Curr Top Microbiol Immunol 2003; 281:29-106. [PMID: 12932075 DOI: 10.1007/978-3-642-19012-4_2] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Evidence obtained during the last few years has greatly extended our understanding of the cell surface receptors that mediate infections of retroviruses and has provided many surprising insights. In contrast to other cell surface components such as lectins or proteoglycans that influence infections indirectly by enhancing virus adsorption onto specific cells, the true receptors induce conformational changes in the viral envelope glycoproteins that are essential for infection. One surprise is that all of the cell surface receptors for gamma-retroviruses are proteins that have multiple transmembrane (TM) sequences, compatible with their identification in known instances as transporters for important solutes. In striking contrast, almost all other animal viruses use receptors that exclusively have single TM sequences, with the sole proven exception we know of being the coreceptors used by lentiviruses. This evidence strongly suggests that virus genera have been prevented because of their previous evolutionary adaptations from switching their specificities between single-TM and multi-TM receptors. This evidence also implies that gamma-retroviruses formed by divergent evolution from a common origin millions of years ago and that individual viruses have occasionally jumped between species (zoonoses) while retaining their commitment to using the orthologous receptor of the new host. Another surprise is that many gamma-retroviruses use not just one receptor but pairs of closely related receptors as alternatives. This appears to have enhanced viral survival by severely limiting the likelihood of host escape mutations. All of the receptors used by gamma-retroviruses contain hypervariable regions that are often heavily glycosylated and that control the viral host range properties, consistent with the idea that these sequences are battlegrounds of virus-host coevolution. However, in contrast to previous assumptions, we propose that gamma-retroviruses have become adapted to recognize conserved sites that are important for the receptor's natural function and that the hypervariable sequences have been elaborated by the hosts as defense bulwarks that surround the conserved viral attachment sites. Previously, it was believed that binding to receptors directly triggers a series of conformational changes in the viral envelope glycoproteins that culminate in fusion of the viral and cellular membranes. However, new evidence suggests that gamma-retroviral association with receptors triggers an obligatory interaction or cross-talk between envelope glycoproteins on the viral surface. If this intermediate step is prevented, infection fails. Conversely, in several circumstances this cross-talk can be induced in the absence of a cell surface receptor for the virus, in which case infection can proceed efficiently. This new evidence strongly implies that the role of cell surface receptors in infections of gamma-retroviruses (and perhaps of other enveloped animal viruses) is more complex and interesting than was previously imagined. Recently, another gammaretroviral receptor with multiple transmembrane sequences was cloned. See Prassolov, Y., Zhang, D., Ivanov, D., Lohler, J., Ross, S.R., and Stocking, C. Sodium-dependent myo-inositol transporter 1 is a receptor for Mus cervicolor M813 murine leukemia virus.
Collapse
Affiliation(s)
- C S Tailor
- Infection, Immunity Injury and Repair Program, Hospital for Sick Children, Toronto, ON M5G 1XB, Canada
| | | | | | | |
Collapse
|
47
|
Rodriguez PC, Zea AH, DeSalvo J, Culotta KS, Zabaleta J, Quiceno DG, Ochoa JB, Ochoa AC. L-arginine consumption by macrophages modulates the expression of CD3 zeta chain in T lymphocytes. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 171:1232-9. [PMID: 12874210 DOI: 10.4049/jimmunol.171.3.1232] [Citation(s) in RCA: 376] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
L-Arginine plays a central role in the normal function of several organs including the immune system. It is metabolized in macrophages by inducible nitric oxide synthase to produce nitric oxide, important in the cytotoxic mechanisms, and by arginase I (ASE I) and arginase II (ASE II) to synthesize L-ornithine and urea, the first being the precursor for the production of polyamines needed for cell proliferation. L-Arginine availability can modulate T cell function. Human T cells stimulated and cultured in the absence of L-arginine lose the expression of the TCR zeta-chain (CD3zeta) and have an impaired proliferation and a decreased cytokine production. The aim of this work was to test whether activated macrophages could modulate extracellular levels of L-arginine and alter T cell function, and to determine which metabolic pathway was responsible for this event. The results show that macrophages stimulated with IL-4 + IL-13 up-regulate ASE I and cationic amino acid transporter 2B, causing a rapid reduction of extracellular levels of L-arginine and inducing decreased expression of CD3zeta and diminished proliferation in normal T lymphocytes. Competitive inhibitors of ASE I or the addition of excess L-arginine lead to the re-expression of CD3zeta and recovery of T cell proliferation. In contrast, inducible nitric oxide synthase or ASE II failed to significantly reduce the extracellular levels of L-arginine and modulate CD3zeta expression. These results may provide new insights into the mechanisms leading to T cell dysfunction and the down-regulation of CD3zeta in cancer and chronic infectious diseases.
Collapse
Affiliation(s)
- Paulo C Rodriguez
- Tumor Immunology Program, Stanley S Scott Cancer Center, Louisiana State University, Health Sciences Center, New Orleans, LA 70112, USA
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Verrey F, Closs EI, Wagner CA, Palacin M, Endou H, Kanai Y. CATs and HATs: the SLC7 family of amino acid transporters. Pflugers Arch 2003; 447:532-42. [PMID: 14770310 DOI: 10.1007/s00424-003-1086-z] [Citation(s) in RCA: 533] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2002] [Accepted: 03/17/2003] [Indexed: 02/07/2023]
Abstract
The SLC7 family is divided into two subgroups, the cationic amino acid transporters (the CAT family, SLC7A1-4) and the glycoprotein-associated amino acid transporters (the gpaAT family, SLC7A5-11), also called light chains or catalytic chains of the hetero(di)meric amino acid transporters (HAT). The associated glycoproteins (heavy chains) 4F2hc (CD98) or rBAT (D2, NBAT) form the SLC3 family. Members of the CAT family transport essentially cationic amino acids by facilitated diffusion with differential trans-stimulation by intracellular substrates. In some cells, they may regulate the rate of NO synthesis by controlling the uptake of l-arginine as the substrate for nitric oxide synthase (NOS). The heterodimeric amino acid transporters are, in contrast, quite diverse in terms of substrate selectivity and function (mostly) as obligatory exchangers. Their selectivity ranges from large neutral amino acids (system L) to small neutral amino acids (ala, ser, cys-preferring, system asc), negatively charged amino acid (system x(c)(-)) and cationic amino acids plus neutral amino acids (system y(+)L and b(0,+)-like). Cotransport of Na(+) is observed only for the y(+)L transporters when they carry neutral amino acids. Mutations in b(0,+)-like and y(+)L transporters lead to the hereditary diseases cystinuria and lysinuric protein intolerance (LPI), respectively.
Collapse
Affiliation(s)
- François Verrey
- Institute of Physiology, University of Zürich, Winterthurerstrasse 190, 8057, Zürich, Switzerland,
| | | | | | | | | | | |
Collapse
|
49
|
Habermeier A, Wolf S, Martiné U, Gräf P, Closs EI. Two amino acid residues determine the low substrate affinity of human cationic amino acid transporter-2A. J Biol Chem 2003; 278:19492-9. [PMID: 12637504 DOI: 10.1074/jbc.m210254200] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Mammalian cationic amino acid transporters (CAT) differ in their substrate affinity and sensitivity to trans-stimulation. The apparent Km values for cationic amino acids and the sensitivity to trans-stimulation of CAT-1, -2B, and -3 are characteristic of system y+. In contrast, CAT-2A exhibits a 10-fold lower substrate affinity and is largely independent of substrate at the trans-side of the membrane. CAT-2A and -2B demonstrate such divergent transport properties, even though their amino acid sequences differ only in a stretch of 42 amino acids. Here, we identify two amino acid residues within this 42-amino acid domain of the human CAT-2A protein that are responsible for the apparent low affinity of both the extracellular and intracellular substrate-binding sites. These residues are located in the fourth intracellular loop, suggesting that they are not part of the translocation pathway. Rather, they may be responsible for the low affinity conformation of the substrate-binding sites. The sensitivity to trans-stimulation is not determined by the same amino acid residues as the substrate affinity and must involve a more complex interaction between individual amino acid residues. In addition to the 42-amino acid domain, the adjacent transmembrane domain X seems to be involved in this function.
Collapse
Affiliation(s)
- Alice Habermeier
- Department of Pharmacology, Johannes Gutenberg University, Obere Zahlbacher Strasse 67, 55101 Mainz, Germany
| | | | | | | | | |
Collapse
|
50
|
Schwartz D, Schwartz IF, Gnessin E, Wollman Y, Chernichovsky T, Blum M, Iaina A. Differential regulation of glomerular arginine transporters (CAT-1 and CAT-2) in lipopolysaccharide-treated rats. Am J Physiol Renal Physiol 2003; 284:F788-95. [PMID: 12475743 DOI: 10.1152/ajprenal.00221.2002] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The decrease in glomerular filtration rate (GFR) that is characteristic of sepsis has been shown to result from inhibition of glomerular endothelial nitric oxide synthase (eNOS) by nitric oxide (NO) generated from the inducible isoform of NOS (iNOS). Although l-arginine is the sole precursor for NO biosynthesis, its intracellular availability in glomeruli from septic animals has never been investigated. Arginine uptake was measured in freshly harvested glomeruli from the following experimental groups: 1) untreated rats; 2) rats pretreated with LPS (4 mg/kg body wt, 4 h before experiments); 3) rats treated with LPS as above with either l-N(6)-(1-iminoethyl)lysine hydrochloride (l-NIL), a selective iNOS antagonist, or 7-nitroindazole, a selective neuronal NOS antagonist; and 4) rats treated with l-NIL only. Both glomeular and mesangial arginine transport characteristics were found compatible with a y(+) system. Arginine uptake was augmented in glomeruli from LPS-treated rats. Treatment with l-NIL completely abolished this effect whereas l-NIL alone had no effect. Similar results were obtained when primary cultures of rat mesangial cells were preincubated with LPS (10 microg/ml for 24 h) with or without l-NIL. Using RT-PCR, we found that in vivo administration of LPS resulted in a significant increase in glomerular cationic amino acid transporter-2 (CAT-2) mRNA expression whereas CAT-1 mRNA was undetected. Northern blotting further confirmed a significant increase in glomerular CAT-2 by LPS. In mesangial cells, the expression of both CAT-1 and CAT-2 mRNA was augmented after incubation with LPS. In conclusion, in vivo administration of LPS augments glomerular arginine transport through upregulation of steady-state CAT-2 mRNA while downregulating CAT-1 mRNA. These results may correspond to the changes in glomerular iNOS and eNOS activity in sepsis.
Collapse
Affiliation(s)
- Doron Schwartz
- Nephrology Department, The Tel Aviv Sourasky Medical Center, Tel Aviv 64239, Israel.
| | | | | | | | | | | | | |
Collapse
|