51
|
Zhang L, Sui C, Yang W, Luo Q. Amino acid transporters: Emerging roles in drug delivery for tumor-targeting therapy. Asian J Pharm Sci 2020; 15:192-206. [PMID: 32373199 PMCID: PMC7193455 DOI: 10.1016/j.ajps.2019.12.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 11/22/2019] [Accepted: 12/22/2019] [Indexed: 12/16/2022] Open
Abstract
Amino acid transporters, which play a vital role in transporting amino acids for the biosynthesis of mammalian cells, are highly expressed in types of tumors. Increasing studies have shown the feasibility of amino acid transporters as a component of tumor-targeting therapy. In this review, we focus on tumor-related amino acid transporters and their potential use in tumor-targeting therapy. Firstly, the expression characteristics of amino acid transporters in cancer and their relationship with tumor growth are reviewed. Secondly, the recognition requirements are discussed, focusing on the "acid-base" properties, conformational isomerism and structural analogues. Finally, recent developments in amino acid transporter-targeting drug delivery strategies are highlighted, including prodrugs and nanocarriers, with special attention to the latest findings of molecular mechanisms and targeting efficiency of transporter-mediated endocytosis. We aim to offer related clues that might lead to valuable tumor-targeting strategies by the utilization of amino acid transporters.
Collapse
Affiliation(s)
- Ling Zhang
- Department of Biotherapy, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Chengguang Sui
- Department of Biotherapy, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Wenhan Yang
- Department of Pharmacy, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
- Department of Pharmacy, China Medical University, Shenyang 110001, China
| | - Qiuhua Luo
- Department of Pharmacy, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
- Department of Pharmacy, China Medical University, Shenyang 110001, China
| |
Collapse
|
52
|
Asc-1 Transporter (SLC7A10): Homology Models And Molecular Dynamics Insights Into The First Steps Of The Transport Mechanism. Sci Rep 2020; 10:3731. [PMID: 32111919 PMCID: PMC7048771 DOI: 10.1038/s41598-020-60617-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Accepted: 02/14/2020] [Indexed: 12/02/2022] Open
Abstract
The alanine-serine-cysteine transporter Asc-1 regulates the synaptic availability of d-serine and glycine (the two co-agonists of the NMDA receptor) and is regarded as an important drug target. To shuttle the substrate from the extracellular space to the cytoplasm, this transporter undergoes multiple distinct conformational states. In this work, homology modeling, substrate docking and molecular dynamics simulations were carried out to learn more about the transition between the “outward-open” and “outward-open occluded” states. We identified a transition state involving the highly-conserved unwound TM6 region in which the Phe243 flips close to the d-serine substrate without major movements of TM6. This feature and those of other key residues are proposed to control the binding site and substrate translocation. Competitive inhibitors ACPP, LuAE00527 and SMLC were docked and their binding modes at the substrate binding site corroborated the key role played by Phe243 of TM6. For ACPP and LuAE00527, strong hydrophobic interactions with this residue hinder its mobility and prevent the uptake and the efflux of substrates. As for SMLC, the weaker interactions maintain the flexibility of Phe243 and the efflux process. Overall, we propose a molecular basis for the inhibition of substrate translocation of the Asc-1 transporter that should be valuable for rational drug design.
Collapse
|
53
|
Hobbach AJ, Closs EI. Human cationic amino acid transporters are not affected by direct nitros(yl)ation. Amino Acids 2020; 52:499-503. [PMID: 32008093 DOI: 10.1007/s00726-020-02819-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 01/20/2020] [Indexed: 11/26/2022]
Abstract
A direct inhibiting effect of NO on the function of CAT-1 and -2A has been postulated to occur via nitrosylation of cysteine residues in the transporters. Neither the NO donor SNAP nor a mixture of SIN-1 and Spermine NONOate, that generates the strong nitrosating agent N2O3, reduced CAT-mediated L-arginine transport. Direct nitros(yl)ation does either not occur in CATs or does not affect their transport function. A regulatory effect of NO or nitrosating agents on CAT-mediated transport under physiological conditions seems, therefore, unlikely.
Collapse
Affiliation(s)
- Anastasia J Hobbach
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Ellen I Closs
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstraße 1, 55131, Mainz, Germany.
| |
Collapse
|
54
|
Fang T, Jia G, Zhao H, Chen X, Wu C, Xue B, Cai J, Tian G, Wang J, Liu G. Effects of spermine supplementation on blood biochemical parameters, amino acid profile and ileum expression of amino acid transporters in piglets. JOURNAL OF ANIMAL AND FEED SCIENCES 2019. [DOI: 10.22358/jafs/114433/2019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
55
|
Lu X. The Role of Large Neutral Amino Acid Transporter (LAT1) in Cancer. Curr Cancer Drug Targets 2019; 19:863-876. [DOI: 10.2174/1568009619666190802135714] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 05/24/2019] [Accepted: 05/31/2019] [Indexed: 12/11/2022]
Abstract
Background:
The solute carrier family 7 (SLC7) can be categorically divided into two
subfamilies, the L-type amino acid transporters (LATs) including SLC7A5-13, and SLC7A15, and
the cationic amino acid transporters (CATs) including SLC7A1-4 and SLC7A14. Members of the
CAT family transport predominantly cationic amino acids by facilitating diffusion with intracellular
substrates. LAT1 (also known as SLC7A5), is defined as a heteromeric amino acid transporter
(HAT) interacting with the glycoprotein CD98 (SLC3A2) through a conserved disulfide to uptake
not only large neutral amino acids, but also several pharmaceutical drugs to cells.
Methods:
In this review, we provide an overview of the interaction of the structure-function of
LAT1 and its essential role in cancer, specifically, its role at the blood-brain barrier (BBB) to facilitate
the transport of thyroid hormones, pharmaceuticals (e.g., I-DOPA, gabapentin), and metabolites
into the brain.
Results:
LAT1 expression increases as cancers progress, leading to higher expression levels in highgrade
tumors and metastases. In addition, LAT1 plays a crucial role in cancer-associated
reprogrammed metabolic networks by supplying tumor cells with essential amino acids.
Conclusion:
The increasing understanding of the role of LAT1 in cancer has led to an increase in
interest surrounding its potential as a drug target for cancer treatment.
Collapse
Affiliation(s)
- Xinjie Lu
- The Mary and Garry Weston Molecular Immunology Laboratory, Thrombosis Research Institute, London, SW3 6LR, United Kingdom
| |
Collapse
|
56
|
Metabolomic Profile of BALB/c Macrophages Infected with Leishmania amazonensis: Deciphering L-Arginine Metabolism. Int J Mol Sci 2019; 20:ijms20246248. [PMID: 31835767 PMCID: PMC6940984 DOI: 10.3390/ijms20246248] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 12/06/2019] [Accepted: 12/08/2019] [Indexed: 12/18/2022] Open
Abstract
Background: Leishmaniases are neglected tropical diseases that are caused by Leishmania, being endemic worldwide. L-arginine is an essential amino acid that is required for polyamines production on mammal cells. During Leishmania infection of macrophages, L-arginine is used by host and parasite arginase to produce polyamines, leading to parasite survival; or, by nitric oxide synthase 2 to produce nitric oxide leading to parasite killing. Here, we determined the metabolomic profile of BALB/c macrophages that were infected with L. amazonensis wild type or with L. amazonensis arginase knockout, correlating the regulation of L-arginine metabolism from both host and parasite. Methods: The metabolites of infected macrophages were analyzed by capillary electrophoresis coupled with mass spectrometry (CE-MS). The metabolic fingerprints analysis provided the dual profile from the host and parasite. Results: We observed increased levels of proline, glutamic acid, glutamine, L-arginine, ornithine, and putrescine in infected-L. amazonensis wild type macrophages, which indicated that this infection induces the polyamine production. Despite this, we observed reduced levels of ornithine, proline, and trypanothione in infected-L. amazonensis arginase knockout macrophages, indicating that this infection reduces the polyamine production. Conclusions: The metabolome fingerprint indicated that Leishmania infection alters the L-arginine/polyamines/trypanothione metabolism inside the host cell and the parasite arginase impacts on L-arginine metabolism and polyamine production, defining the infection fate.
Collapse
|
57
|
Wu R, Liu S, Liu Y, Sun Y, Cheng X, Huang Y, Yang Z, Wu Z. Synthesis and biological evaluation of [18F](2S,4S)4-(3-fluoropropyl) arginine as a tumor imaging agent. Eur J Med Chem 2019; 183:111730. [DOI: 10.1016/j.ejmech.2019.111730] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 09/21/2019] [Accepted: 09/21/2019] [Indexed: 12/31/2022]
|
58
|
Werner A, Pieh D, Echchannaoui H, Rupp J, Rajalingam K, Theobald M, Closs EI, Munder M. Cationic Amino Acid Transporter-1-Mediated Arginine Uptake Is Essential for Chronic Lymphocytic Leukemia Cell Proliferation and Viability. Front Oncol 2019; 9:1268. [PMID: 31824848 PMCID: PMC6879669 DOI: 10.3389/fonc.2019.01268] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 11/01/2019] [Indexed: 12/22/2022] Open
Abstract
Interfering with tumor metabolism by specifically restricting the availability of extracellular nutrients is a rapidly emerging field of cancer research. A variety of tumor entities depend on the uptake of the amino acid arginine since they have lost the ability to synthesize it endogenously, that is they do not express the rate limiting enzyme for arginine synthesis, argininosuccinate synthase (ASS). Arginine transport through the plasma membrane of mammalian cells is mediated by eight different transporters that belong to two solute carrier (SLC) families. In the present study we found that the proliferation of primary as well as immortalized chronic lymphocytic leukemia (CLL) cells depends on the availability of extracellular arginine and that primary CLL cells do not express ASS and are therefore arginine-auxotrophic. The cationic amino acid transporter-1 (CAT-1) was the only arginine importer expressed in CLL cells. Lentiviral-mediated downregulation of the CAT-1 transporter in HG3 CLL cells significantly reduced arginine uptake, abolished cell proliferation and impaired cell viability. In a murine CLL xenograft model, tumor growth was significantly suppressed upon induced downregulation of CAT-1 in the CLL cells. Our results suggest that inhibition of CAT-1 is a promising new therapeutic approach for CLL.
Collapse
Affiliation(s)
- Anke Werner
- Third Department of Medicine (Hematology, Oncology, and Pneumology), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.,Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Daniel Pieh
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Hakim Echchannaoui
- Third Department of Medicine (Hematology, Oncology, and Pneumology), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Johanna Rupp
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Krishnaraj Rajalingam
- Cell Biology Unit, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Matthias Theobald
- Third Department of Medicine (Hematology, Oncology, and Pneumology), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.,Research Center for Immune Therapy, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.,German Cancer Consortium (DKTK), Mainz, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ellen I Closs
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Markus Munder
- Third Department of Medicine (Hematology, Oncology, and Pneumology), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.,Research Center for Immune Therapy, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
59
|
Bai L, Sato H, Kubo Y, Wada S, Aida Y. CAT1/SLC7A1 acts as a cellular receptor for bovine leukemia virus infection. FASEB J 2019; 33:14516-14527. [PMID: 31648581 PMCID: PMC6894071 DOI: 10.1096/fj.201901528r] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Bovine leukemia virus (BLV) is the causative agent of enzootic bovine leukosis, the most common neoplastic disease of cattle, which is closely related to human T-cell leukemia viruses. BLV has spread worldwide and causes a serious problem for the cattle industry. The cellular receptor specifically binds with viral envelope glycoprotein (Env), and this attachment mediates cell fusion to lead virus entry. BLV Env reportedly binds to cationic amino acid transporter 1 (CAT1)/solute carrier family 7 member 1 (SLC7A1), but whether the CAT1/SLC7A1 is an actual receptor for BLV remains unknown. Here, we showed that CAT1 functioned as an infection receptor, interacting with BLV particles. Cells expressing undetectable CAT1 levels were resistant to BLV infection but became highly susceptible upon CAT1 overexpression. CAT1 exhibited specific binding to BLV particles on the cell surface and colocalized with the Env in endomembrane compartments and membrane. Knockdown of CAT1 in permissive cells significantly reduced binding to BLV particles and BLV infection. Expression of CAT1 from various species demonstrated no species specificity for BLV infection, implicating CAT1 as a functional BLV receptor responsible for its broad host range. These findings provide insights for BLV infection and for developing new strategies for treating BLV and preventing its spread.-Bai, L., Sato, H., Kubo, Y., Wada, S., Aida, Y. CAT1/SLC7A1 acts as a cellular receptor for bovine leukemia virus infection.
Collapse
Affiliation(s)
- Lanlan Bai
- Viral Infectious Disease Unit, RIKEN.,Photonics Control Technology Team, RIKEN Center for Advanced Photonics
| | - Hirotaka Sato
- Viral Infectious Disease Unit, RIKEN.,Nakamura Laboratory, RIKEN Cluster for Science, Technology, and Innovation Hub, Wako, Japan; and
| | - Yoshinao Kubo
- Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Satoshi Wada
- Photonics Control Technology Team, RIKEN Center for Advanced Photonics
| | - Yoko Aida
- Viral Infectious Disease Unit, RIKEN.,Nakamura Laboratory, RIKEN Cluster for Science, Technology, and Innovation Hub, Wako, Japan; and
| |
Collapse
|
60
|
Wu Z, Alibay I, Newstead S, Biggin PC. Proton Control of Transitions in an Amino Acid Transporter. Biophys J 2019; 117:1342-1351. [PMID: 31500802 PMCID: PMC6818167 DOI: 10.1016/j.bpj.2019.07.056] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 07/03/2019] [Accepted: 07/30/2019] [Indexed: 12/19/2022] Open
Abstract
Amino acid transport into the cell is often coupled to the proton electrochemical gradient, as found in the solute carrier 36 family of proton-coupled amino acid transporters. Although no structure of a human proton-coupled amino acid transporter exists, the crystal structure of a related homolog from bacteria, GkApcT, has recently been solved in an inward-occluded state and allows an opportunity to examine how protons are coupled to amino acid transport. Our working hypothesis is that release of the amino acid substrate is facilitated by the deprotonation of a key glutamate residue (E115) located at the bottom of the binding pocket, which forms part of the intracellular gate, allowing the protein to transition from an inward-occluded to an inward-open conformation. During unbiased molecular dynamics simulations, we observed a transition from the inward-occluded state captured in the crystal structure to a much more open state, which we consider likely to be representative of the inward-open state associated with substrate release. To explore this and the role of protons in these transitions, we have used umbrella sampling to demonstrate that the transition from inward occluded to inward open is more energetically favorable when E115 is deprotonated. That E115 is likely to be protonated in the inward-occluded state and deprotonated in the inward-open state is further confirmed via the use of absolute binding free energies. Finally, we also show, via the use of absolute binding free energy calculations, that the affinity of the protein for alanine is very similar regardless of either the conformational state or the protonation of E115, presumably reflecting the fact that all the key interactions are deep within the binding cavity. Together, our results give a detailed picture of the role of protons in driving one of the major transitions in this transporter.
Collapse
Affiliation(s)
- Zhiyi Wu
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| | - Irfan Alibay
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| | - Simon Newstead
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| | - Philip C Biggin
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
61
|
Zhang B, Gan L, Shahid MS, Lv Z, Fan H, Liu D, Guo Y. In vivo and in vitro protective effect of arginine against intestinal inflammatory response induced by Clostridium perfringens in broiler chickens. J Anim Sci Biotechnol 2019; 10:73. [PMID: 31428367 PMCID: PMC6697915 DOI: 10.1186/s40104-019-0371-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Accepted: 06/07/2019] [Indexed: 01/29/2023] Open
Abstract
Background Necrotic enteritis is a widespread disease in poultry caused by Clostridium perfringens. We previously reported that dietary arginine supplementation protected the intestinal mucosa of broiler chickens with necrotic enteritis, but the related protective mechanisms remain unclear. The in vivo trial was designed as a 2 × 2 factorial arrangement to evaluated the effects of arginine supplementation on inflammatory responses, arginine transporters, arginine catabolism and JAK-STAT signalling pathway in broiler chickens challenged with C. perfringens or without C. perfringens. Furthermore, we validated the in vivo results using intestinal epithelial cells of chicken embryos. Results C. perfringens infection markedly increased gut gross pathological and histopathological lesion scores, promoted liver C. perfringens invasion, reduced serum arginine levels, and elevated jejunal mucosal lysozyme activities (P < 0.05), but these effects were significantly reversed by arginine supplementation in vivo (P < 0.05). The challenge significantly increased serum procalcitonin levels, jejunal mucosal iNOS activities and jejunal IL-6, TGF-β3, cationic amino acid transporter (CAT)-1, and CAT-3 mRNA expression (P < 0.05), whereas arginine supplementation significantly reduced jejunal IFN-γ, IL-1β, IL-6, IL-10, TGF-β3, and CAT-3 mRNA expression (P < 0.05). Arginine supplementation significantly attenuated the C. perfringens challenge-induced increases in jejunal iNOS, arginase 2, arginine decarboxylase, arginine:glycine amidinotransferase, JAK1, JAK3, STAT1, and STAT6 mRNA expression (P < 0.05). The in vitro experiment showed that C. perfringens challenge markedly increased cellular cytotoxicity and the mRNA expression of IL-1β, IL-8, IL-10, CAT-1 and CAT-3 (P < 0.05), which were significantly reversed by 50 μmol/L and/or 400 μmol/L arginine pre-treatment (P < 0.05). Conclusions Arginine prevented C. perfringens challenge-induced circulated arginine deficiency, normalized intestinal arginine transport and catabolism, down-regulated JAK-STAT signalling pathway and attenuated the inflammatory response, which exerted protective effects on the intestine of broiler chickens.
Collapse
Affiliation(s)
- Beibei Zhang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 People's Republic of China
| | - Liping Gan
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 People's Republic of China
| | - Muhammad Suhaib Shahid
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 People's Republic of China
| | - Zengpeng Lv
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 People's Republic of China
| | - Hao Fan
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 People's Republic of China
| | - Dan Liu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 People's Republic of China
| | - Yuming Guo
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 People's Republic of China
| |
Collapse
|
62
|
Motahari Z, Moody SA, Maynard TM, LaMantia AS. In the line-up: deleted genes associated with DiGeorge/22q11.2 deletion syndrome: are they all suspects? J Neurodev Disord 2019; 11:7. [PMID: 31174463 PMCID: PMC6554986 DOI: 10.1186/s11689-019-9267-z] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 04/21/2019] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND 22q11.2 deletion syndrome (22q11DS), a copy number variation (CNV) disorder, occurs in approximately 1:4000 live births due to a heterozygous microdeletion at position 11.2 (proximal) on the q arm of human chromosome 22 (hChr22) (McDonald-McGinn and Sullivan, Medicine 90:1-18, 2011). This disorder was known as DiGeorge syndrome, Velo-cardio-facial syndrome (VCFS) or conotruncal anomaly face syndrome (CTAF) based upon diagnostic cardiovascular, pharyngeal, and craniofacial anomalies (McDonald-McGinn and Sullivan, Medicine 90:1-18, 2011; Burn et al., J Med Genet 30:822-4, 1993) before this phenotypic spectrum was associated with 22q11.2 CNVs. Subsequently, 22q11.2 deletion emerged as a major genomic lesion associated with vulnerability for several clinically defined behavioral deficits common to a number of neurodevelopmental disorders (Fernandez et al., Principles of Developmental Genetics, 2015; Robin and Shprintzen, J Pediatr 147:90-6, 2005; Schneider et al., Am J Psychiatry 171:627-39, 2014). RESULTS The mechanistic relationships between heterozygously deleted 22q11.2 genes and 22q11DS phenotypes are still unknown. We assembled a comprehensive "line-up" of the 36 protein coding loci in the 1.5 Mb minimal critical deleted region on hChr22q11.2, plus 20 protein coding loci in the distal 1.5 Mb that defines the 3 Mb typical 22q11DS deletion. We categorized candidates based upon apparent primary cell biological functions. We analyzed 41 of these genes that encode known proteins to determine whether haploinsufficiency of any single 22q11.2 gene-a one gene to one phenotype correspondence due to heterozygous deletion restricted to that locus-versus complex multigenic interactions can account for single or multiple 22q11DS phenotypes. CONCLUSIONS Our 22q11.2 functional genomic assessment does not support current theories of single gene haploinsufficiency for one or all 22q11DS phenotypes. Shared molecular functions, convergence on fundamental cell biological processes, and related consequences of individual 22q11.2 genes point to a matrix of multigenic interactions due to diminished 22q11.2 gene dosage. These interactions target fundamental cellular mechanisms essential for development, maturation, or homeostasis at subsets of 22q11DS phenotypic sites.
Collapse
Affiliation(s)
- Zahra Motahari
- The Institute for Neuroscience, and Department of Anatomy and Cell Biology, The George Washington University School of Medicine and Health Sciences, Washington DC, 20037 USA
| | - Sally Ann Moody
- The Institute for Neuroscience, and Department of Anatomy and Cell Biology, The George Washington University School of Medicine and Health Sciences, Washington DC, 20037 USA
| | - Thomas Michael Maynard
- The Institute for Neuroscience, and Department of Anatomy and Cell Biology, The George Washington University School of Medicine and Health Sciences, Washington DC, 20037 USA
| | - Anthony-Samuel LaMantia
- The Institute for Neuroscience, and Department of Anatomy and Cell Biology, The George Washington University School of Medicine and Health Sciences, Washington DC, 20037 USA
| |
Collapse
|
63
|
Torrecillas IR, Conde-Ceide S, de Lucas AI, Garcı́a Molina A, Trabanco AA, Lavreysen H, Pardo L, Tresadern G. Inhibition of the Alanine-Serine-Cysteine-1 Transporter by BMS-466442. ACS Chem Neurosci 2019; 10:2510-2517. [PMID: 30821959 DOI: 10.1021/acschemneuro.9b00019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Experiment and modeling were combined to understand inhibition of the alanine-serine-cysteine-1 (asc1) transporter. The structure-activity relationship (SAR) was explored with synthesis of analogues of BMS-466442. Direct target interaction and binding site location between TM helices 6 and 10 were confirmed via site directed mutagenesis. Computational modeling suggested the inhibitor binds via competitive occupation of the orthosteric site while also blocking the movement of TM helices that are required for transport.
Collapse
Affiliation(s)
- Ivan R. Torrecillas
- Laboratori de Medicina Computacional Unitat de Bioestadistica, Facultat de Medicina, Universitat Autonoma de Barcelona, 08193, Bellaterra, Spain
| | - Susana Conde-Ceide
- Medicinal Chemistry, Janssen Research & Development, Calle Jarama 75A, Toledo 45007, Spain
| | - Ana Isabel de Lucas
- Medicinal Chemistry, Janssen Research & Development, Calle Jarama 75A, Toledo 45007, Spain
| | | | - Andrés A. Trabanco
- Medicinal Chemistry, Janssen Research & Development, Calle Jarama 75A, Toledo 45007, Spain
| | | | - Leonardo Pardo
- Laboratori de Medicina Computacional Unitat de Bioestadistica, Facultat de Medicina, Universitat Autonoma de Barcelona, 08193, Bellaterra, Spain
| | | |
Collapse
|
64
|
Yahyaoui R, Blasco-Alonso J, Benito C, Rodríguez-García E, Andrade F, Aldámiz-Echevarría L, Muñoz-Hernández MC, Vega AI, Pérez-Cerdá C, García-Martín ML, Pérez B. A new metabolic disorder in human cationic amino acid transporter-2 that mimics arginase 1 deficiency in newborn screening. J Inherit Metab Dis 2019; 42:407-413. [PMID: 30671984 DOI: 10.1002/jimd.12063] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 11/30/2018] [Indexed: 11/06/2022]
Abstract
PURPOSE We report a patient with a human cationic amino acid transporter 2 (CAT-2) defect discovered due to a suspected arginase 1 deficiency observed in newborn screening (NBS). METHODS A NBS sample was analyzed using tandem mass spectrometry. Screen results were confirmed by plasma and urine amino acid quantification. Molecular diagnosis was done using clinical exome sequencing. Dimethylated arginines were determined by HPLC and nitrate/nitrite levels by a colorimetric assay. The metabolomic profile was analyzed using 1D nuclear magnetic resonance spectroscopy. RESULTS A Spanish boy of nonconsanguineous parents had high arginine levels in a NBS blood sample. Plasma and urinary cationic amino acids were high. Arginase enzyme activity in erythrocytes was normal and no pathogenic mutations were identified in the ARG1 gene. Massive parallel sequencing detected two loss-of-function mutations in the SLC7A2 gene. Currently, the child receives a protein-controlled diet of 1.2 g/kg/day with protein-and amino-acid free infant formula, 30 g/day, and is asymptomatic. CONCLUSION We identified a novel defect in human CAT-2 due to biallelic pathogenic variants in the SLC7A2 gene. The characteristic biochemical profile includes high plasma and urine arginine, ornithine, and lysine levels. NBS centers should know of this disorder since it can be detected in arginase 1 deficiency screening.
Collapse
Affiliation(s)
- Raquel Yahyaoui
- Laboratory of Metabolic Disorders and Newborn Screening Center of Eastern Andalusia, Málaga Regional University Hospital, Málaga, Spain
- Grupo Endocrinología y Nutrición, Diabetes y Obesidad, Instituto de Investigación Biomédica de Málaga-IBIMA
| | | | - Carmen Benito
- Department of Genetics, Málaga Regional University Hospital, Málaga, Spain
| | - Enrique Rodríguez-García
- Grupo Endocrinología y Nutrición, Diabetes y Obesidad, Instituto de Investigación Biomédica de Málaga-IBIMA
- Laboratorios Vircell, SL, Granada, Spain
| | - Fernando Andrade
- Metabolomics Platform, Metabolism Group, BioCruces Health Research Institute, CIBERER, Barakaldo, Spain
| | - Luis Aldámiz-Echevarría
- Metabolomics Platform, Metabolism Group, BioCruces Health Research Institute, CIBERER, Barakaldo, Spain
| | - María C Muñoz-Hernández
- BIONAND, Andalusian Center for Nanomedicine and Biotechnology, Junta de Andalucía, Universidad de Málaga, Málaga, Spain
| | - Ana I Vega
- Centro Diagnóstico de Enfermedades Moleculares (CEDEM), Centro de Biología Molecular, Universidad Autónoma de Madrid, CIBERER, idiPAZ, Madrid, Spain
| | - Celia Pérez-Cerdá
- Centro Diagnóstico de Enfermedades Moleculares (CEDEM), Centro de Biología Molecular, Universidad Autónoma de Madrid, CIBERER, idiPAZ, Madrid, Spain
| | - María L García-Martín
- BIONAND, Andalusian Center for Nanomedicine and Biotechnology, Junta de Andalucía, Universidad de Málaga, Málaga, Spain
| | - Belén Pérez
- Centro Diagnóstico de Enfermedades Moleculares (CEDEM), Centro de Biología Molecular, Universidad Autónoma de Madrid, CIBERER, idiPAZ, Madrid, Spain
| |
Collapse
|
65
|
Ma YF, Batistel F, Xu TL, Han LQ, Bucktrout R, Liang Y, Coleman DN, Parys C, Loor JJ. Phosphorylation of AKT serine/threonine kinase and abundance of milk protein synthesis gene networks in mammary tissue in response to supply of methionine in periparturient Holstein cows. J Dairy Sci 2019; 102:4264-4274. [PMID: 30879806 DOI: 10.3168/jds.2018-15451] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 01/25/2019] [Indexed: 12/17/2022]
Abstract
The main objective was to evaluate the effect of increasing the supply of Met around parturition on abundance and phosphorylation of insulin- and mechanistic target of rapamycin complex 1 (mTORC1)-related signaling proteins along with mRNA abundance of milk protein and fat synthesis-related genes in postpartal mammary tissue. A basal control diet (control) or the basal diet plus ethyl-cellulose rumen-protected Met (0.9 g/kg of dry matter intake; Mepron, Evonik Nutrition & Care GmbH, Hanau-Wolfgang, Germany) were fed (n = 30 cows/diet) from d -28 to 60 relative to parturition. Mammary tissue and blood plasma were harvested from the same cows (n = 5/diet) in the control and Met groups at d 21 postpartum for mRNA, protein, and AA analysis. Increasing the supply of Met led to greater milk protein percentage and milk yield along with greater ratio of phosphorylated (p-)AKT to total AKT. The ratio of p-mTORC1 to total mTORC1 did not differ, but ratio of p-RPS6 to total ribosomal protein S6 (RPS6) was lower in response to Met supply. These responses were associated with greater mRNA abundance of the signaling proteins Janus kinase 2 (JAK2) and insulin receptor substrate 1 (IRS1). Greater Met supply also upregulated mRNA abundance of high-affinity cationic (SLC7A1) and sodium-coupled AA transporters (SLC38A1, SLC38A2); leucyl-tRNA (LARS), valyl-tRNA (VARS), and isoleucyl-tRNA synthetases (IARS); glucose transport solute carrier family 2 member 3 (SLC2A1); glucose transport solute carrier family 2 member 3 (SLC2A3); and casein α-s1 (CSN1S1). The mRNA abundance of components of the unfolded protein response, such as x-box binding protein 1 (XBP1) and activating transcription factor 6 (ATF6), were upregulated, and protein phosphatase 1, regulatory subunit 15A (PPP1R15A) was downregulated in response to greater Met supply. Overall, the data suggest that increased dry matter intake, greater phosphorylation status of AKT, upregulation of glucose and AA transporters, and transcripts of tRNases in response to enhanced Met supply might have compensated for a reduction in ribosome biogenesis due to a lower ratio of p-RPS6 to total RPS6. Together, these cellular responses constitute a mechanism whereby Met supply can regulate milk protein synthesis in early lactation.
Collapse
Affiliation(s)
- Y F Ma
- Institute of Animal Nutrition and Feed, Inner Mongolia Academy of Agriculture and Animal Husbandry Sciences, Hohhot 010031, P. R. China; Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801
| | - F Batistel
- Department of Animal, Dairy & Veterinary Science, Utah State University, Logan 84322
| | - T L Xu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, P. R. China
| | - L Q Han
- Department of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, P. R. China
| | - R Bucktrout
- Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801
| | - Y Liang
- Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801
| | - D N Coleman
- Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801
| | - C Parys
- Evonik Nutrition & Care GmbH, Hanau-Wolfgang, 63457, Germany
| | - J J Loor
- Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801.
| |
Collapse
|
66
|
Exchange-mode glutamine transport across CNS cell membranes. Neuropharmacology 2019; 161:107560. [PMID: 30853601 DOI: 10.1016/j.neuropharm.2019.03.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 02/28/2019] [Accepted: 03/02/2019] [Indexed: 12/18/2022]
Abstract
CNS cell membranes possess four transporters capable of exchanging Lglutamine (Gln) for other amino acids: the large neutral amino acid (LNAA) transporters LAT1 and LAT2, the hybrid basic amino acid (L-arginine (Arg), L-leucine (Leu)/LNAA transporter y+LAT2, and the L-alanine/L-serine/L-cysteine transporter 2 (ASCT2). LAT1/LAT2 and y+LAT2 are present in astrocytes, neurons and the blood brain barrier (BBB) - forming cerebral vascular endothelial cells (CVEC), while the location of ASCT2 in the individual cell types is a matter of debate. In the healthy brain, contribution of the exchangers to Gln shuttling from astrocytes to neurons and thus their role in controlling the conversion of Gln to the amino acid neurotransmitters l-glutamate (Glu) and γ-aminobutyric acid (GABA) and Gln flux across the BBB appears negligible as compared to the system A and system N uniporters. Insofar, except for the contribution of LAT1 to the maintenance of Gln homeostasis in the interstitial fluid (ISF), no well-defined CNS-specific function has been established for either of the three transporters in the healthy brain. The Gln-accepting amino acid exchangers appear to gain significance under conditions of excessive brain Gln load (glutaminosis). Excess Gln efflux across the BBB enhances influx into the brain of L-tryptophan (Trp). Excess of Trp is responsible for overloading the brain with neuroactive compounds: serotonin, kynurenic acid, quinolinic acid and/or oxindole, which contribute to neurotransmission imbalance accompanying hyperammonemia. In turn, alterations of y+LAT2-mediated Gln/Arg exchange and Arg uptake in astrocyte, modulate astrocytic nitric oxide synthesis and oxidative/nitrosative stress in ammonia-overexposed brain. This article is part of the issue entitled 'Special Issue on Neurotransmitter Transporters'.
Collapse
|
67
|
Subiabre M, Villalobos-Labra R, Silva L, Fuentes G, Toledo F, Sobrevia L. Role of insulin, adenosine, and adipokine receptors in the foetoplacental vascular dysfunction in gestational diabetes mellitus. Biochim Biophys Acta Mol Basis Dis 2019; 1866:165370. [PMID: 30660686 DOI: 10.1016/j.bbadis.2018.12.021] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 12/18/2018] [Accepted: 12/21/2018] [Indexed: 12/11/2022]
Abstract
Gestational diabetes mellitus (GDM) is a disease of pregnancy associated with maternal and foetal hyperglycaemia and altered foetoplacental vascular function. Human foetoplacental microvascular and macrovascular endothelium from GDM pregnancy show increased maximal l-arginine transport capacity via the human cationic amino acid transporter 1 (hCAT-1) isoform and nitric oxide (NO) synthesis by the endothelial NO synthase (eNOS). These alterations are paralleled by lower maximal transport activity of the endogenous nucleoside adenosine via the human equilibrative nucleoside transporter 1 (hENT1) and activation of adenosine receptors. A causal relationship has been described for adenosine-activation of A2A adenosine receptors, hCAT-1, and eNOS activity (i.e. the Adenosine/l-Arginine/Nitric Oxide, ALANO, signalling pathway). Insulin restores these alterations in GDM via activation of insulin receptor A (IR-A) form in the macrovascular but IR-A and IR-B forms in the microcirculation of the human placenta. Adipokines are secreted from adipocytes influencing the foetoplacental metabolic and vascular function. Various adipokines are dysregulated in GDM, with adiponectin and leptin playing major roles. Abnormal plasma concentration of these adipokines and the activation or their receptors are involved in the pathophysiology of GDM. However, involvement of adipokines, adenosine, and insulin receptors and membrane transporters in the aetiology of this disease of pregnancy is unknown. This review focuses on the pathophysiology of insulin and adenosine receptors and l-arginine and adenosine membranes transporters giving an overview of the key adipokines leptin and adiponectin in the foetoplacental vasculature in GDM. This article is part of a Special Issue entitled: Membrane Transporters and Receptors in Pregnancy Metabolic Complications edited by Luis Sobrevia.
Collapse
Affiliation(s)
- Mario Subiabre
- Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile.
| | - Roberto Villalobos-Labra
- Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile
| | - Luis Silva
- Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen, University Medical Centre Groningen (UMCG), Groningen 9700 RB, the Netherlands
| | - Gonzalo Fuentes
- Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; Cell Physiology Laboratory, Biomedical Department, Faculty of Health Sciences, Universidad de Antofagasta, Antofagasta 1270300, Chile
| | - Fernando Toledo
- Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; Department of Basic Sciences, Faculty of Sciences, Universidad del Bío Bío, Chillán 3780000, Chile
| | - Luis Sobrevia
- Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; Department of Physiology, Faculty of Pharmacy, Universidad de Sevilla, Seville E-41012, Spain; University of Queensland Centre for Clinical Research (UQCCR), Faculty of Medicine and Biomedical Sciences, University of Queensland, Herston 4029, Queensland, Australia.
| |
Collapse
|
68
|
Shen L, Qian C, Cao H, Wang Z, Luo T, Liang C. Upregulation of the solute carrier family 7 genes is indicative of poor prognosis in papillary thyroid carcinoma. World J Surg Oncol 2018; 16:235. [PMID: 30558624 PMCID: PMC6297957 DOI: 10.1186/s12957-018-1535-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 11/30/2018] [Indexed: 11/22/2022] Open
Abstract
Background The solute carrier (SLC) 7 family genes comprise 14 members and function as cationic amino acid/glycoprotein transporters in many cells, they are essential for the maintenance of amino acid nutrition and survival of tumor cells. This study was conducted to analyze the associations of SLC7 family gene expression with mortality in papillary thyroid carcinoma (PTC). Methods Clinical features, somatic mutations, and SLC7 family gene expression data were downloaded from The Cancer Genome Atlas database. Linear regression model analysis was performed to analyze the correlations between SLC7 family gene expression and clinicopathologic features. Kaplan-Meier survival and logistic regression analyses were performed to characterize the associations between gene expression and patients’ overall survival. Results Patient mortality was negatively associated with age and tumor size but positively increased cancer stage and absence of thyroiditis in PTC patients. Kaplan-Meier survival analysis indicated that patients with high SLC7A3, SLC7A5, and SLC7A11 expression levels exhibited poorer survival than those with low SLC7A3, SLC7A5, and SLC7A11 expression levels (P < 0.05 for all cases). Logistic regression analysis showed that SLC7A3, SLC7A5, and SLC7A11 were associated with increased mortality (odds ratio [OR] 8.61, 95% confidence interval [CI] 2.3–55.91; OR 3.87, 95% CI 1.18–17.31; and OR 3.87, 95% CI 1.18–17.31, respectively. Conclusion Upregulation of SLC7A3, SLC7A5, and SLC7A11 expression was associated with poor prognosis in PTC patients, and SLC7 gene expression levels are potentially useful prognostic biomarkers.
Collapse
Affiliation(s)
- Lei Shen
- Department of Thyroid Breast Oncology, Shanghai East Hospital, School of Medicine, Tongji University School of Medicine, No.150 Jimo Road, Shanghai, 200120, China
| | - Chunhua Qian
- Department of Endocrinolgy and Metabolism, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Huimin Cao
- Department of Thyroid and Breast, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhongrui Wang
- Department of Thyroid Breast Oncology, Shanghai East Hospital, School of Medicine, Tongji University School of Medicine, No.150 Jimo Road, Shanghai, 200120, China
| | - Tingxian Luo
- Department of Thyroid Breast Oncology, Shanghai East Hospital, School of Medicine, Tongji University School of Medicine, No.150 Jimo Road, Shanghai, 200120, China
| | - Chunli Liang
- Department of Thyroid Breast Oncology, Shanghai East Hospital, School of Medicine, Tongji University School of Medicine, No.150 Jimo Road, Shanghai, 200120, China.
| |
Collapse
|
69
|
Zhu X, Jiao J, Zhou C, Tang S, Wang M, Kang J, Han X, Tan Z. Effects of dietary methionine and lysine supplementation on nutrients digestion, serum parameters and mRNA expression of related amino acid sensing and transporting genes in growing goats. Small Rumin Res 2018. [DOI: 10.1016/j.smallrumres.2018.07.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
70
|
Crouse MS, McLean KJ, Greseth NP, Crosswhite MR, Pereira NN, Ward AK, Reynolds LP, Dahlen CR, Neville BW, Borowicz PP, Caton JS. Maternal nutrition and stage of early pregnancy in beef heifers: Impacts on expression of glucose, fructose, and cationic amino acid transporters in utero-placental tissues. J Anim Sci 2018; 95:5563-5572. [PMID: 29293768 DOI: 10.2527/jas2017.1983] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
We hypothesized that maternal nutrition and day of gestation would impact utero-placental mRNA expression of the nutrient transporters , , , , and in beef heifers. Crossbred Angus heifers (n = 49) were estrous synchronized, bred via AI, assigned to nutritional treatment (CON = 100% of NRC requirements for 0.45 kg/d gain and RES = 60% of CON) and ovariohysterectomized on d 16, 34, or 50 of gestation (n = 6 to 9/d); Non-bred, non-pregnant (NB-NP) controls were fed the CON diet, not bred, and were ovariohysterectomized on d 16 of the synchronized estrous cycle = 6). The resulting arrangement of treatments was a 2 × 3 factorial + 1 (CON vs. RES × d 16, 34, or 50 + NB-NP controls). Caruncle (CAR), intercaruncular endometrium (ICAR), and fetal membranes (FM [chorioallantois]), were obtained from the pregnant uterine horn (the uterine horn containing the conceptus) immediately after ovariohysterectomy. On d 50 cotyledons (COT), intercotyledonary placenta (ICOT) and amnion (AMN) were also collected. Relative expression of nutrient transporters was determined for each tissue utilizing NB-NP-CAR and NB-NP-ICAR tissues as the baseline. For FM, NB-NP endometrium served as the baseline. There was no interaction of day × treatment ( ≥ 0.20) for any genes in CAR. However, CAR expression of was greater ( < 0.01) on d 16 compared with d 34 and 50, and , , and were greater ( ≤ 0.05) on d 34 compared with d 16 and 50. In ICAR, was the only gene to be influenced by the day × treatment interaction ( = 0.01), being greater in d 50 CON compared with d 34 CON and d 16 and 50 RES. In ICAR, expression of was greater ( < 0.01) on d 16 compared with d 34, and expression of was greater ( < 0.01) on d 34 and 50 compared with d 16. In FM, expression of was greater ( = 0.04) on d 16 compared with d 50 of gestation, and expression of was greater ( < 0.01) on d 34 and 50 compared with d 16. On d 50, expression of , , and expression were all greater ( < 0.05) in AMN compared with COT and ICOT, and expression of was greater ( < 0.01) in ICOT compared with COT and AMN. These data indicate that day was a more influential factor for mRNA expression of utero-placental glucose and cationic AA transporters than maternal nutritional status in heifers during early pregnancy.
Collapse
|
71
|
Jungnickel KEJ, Parker JL, Newstead S. Structural basis for amino acid transport by the CAT family of SLC7 transporters. Nat Commun 2018; 9:550. [PMID: 29416041 PMCID: PMC5803215 DOI: 10.1038/s41467-018-03066-6] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 01/17/2018] [Indexed: 12/12/2022] Open
Abstract
Amino acids play essential roles in cell biology as regulators of metabolic pathways. Arginine in particular is a major signalling molecule inside the cell, being a precursor for both l-ornithine and nitric oxide (NO) synthesis and a key regulator of the mTORC1 pathway. In mammals, cellular arginine availability is determined by members of the solute carrier (SLC) 7 family of cationic amino acid transporters. Whereas CAT-1 functions to supply cationic amino acids for cellular metabolism, CAT-2A and -2B are required for macrophage activation and play important roles in regulating inflammation. Here, we present the crystal structure of a close homologue of the mammalian CAT transporters that reveals how these proteins specifically recognise arginine. Our structural and functional data provide a model for cationic amino acid transport in mammalian cells and reveals mechanistic insights into proton-coupled, sodium-independent amino acid transport in the wider APC superfamily. Cationic amino acid transporters (CATs) belong to the physiologically important solute carrier (SLC) 7 family. Here, the authors present the structure of the mammalian CAT transporter homologue Geobacillus kaustophilus GkApcT, which reveals how arginine is recognized, and propose a model for proton-coupled amino acid transport.
Collapse
Affiliation(s)
| | - Joanne L Parker
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, UK
| | - Simon Newstead
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, UK.
| |
Collapse
|
72
|
Muxel SM, Aoki JI, Fernandes JCR, Laranjeira-Silva MF, Zampieri RA, Acuña SM, Müller KE, Vanderlinde RH, Floeter-Winter LM. Arginine and Polyamines Fate in Leishmania Infection. Front Microbiol 2018; 8:2682. [PMID: 29379478 PMCID: PMC5775291 DOI: 10.3389/fmicb.2017.02682] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 12/22/2017] [Indexed: 01/22/2023] Open
Abstract
Leishmania is a protozoan parasite that alternates its life cycle between the sand fly and the mammalian host macrophages, involving several environmental changes. The parasite responds to these changes by promoting a rapid metabolic adaptation through cellular signaling modifications that lead to transcriptional and post-transcriptional gene expression regulation and morphological modifications. Molecular approaches such as gene expression regulation, next-generation sequencing (NGS), microRNA (miRNA) expression profiling, in cell Western blot analyses and enzymatic activity profiling, have been used to characterize the infection of murine BALB/c and C57BL/6 macrophages, as well as the human monocytic cell-lineage THP-1, with Leishmania amazonensis wild type (La-WT) or arginase knockout (La-arg-). These models are being used to elucidate physiological roles of arginine and polyamines pathways and the importance of arginase for the establishment of the infection. In this review, we will describe the main aspects of Leishmania-host interaction, focusing on the arginine and polyamines pathways and pointing to possible targets to be used for prognosis and/or in the control of the infection. The parasite enzymes, arginase and nitric oxide synthase-like, have essential roles in the parasite survival and in the maintenance of infection. On the other hand, in mammalian macrophages, defense mechanisms are activated inducing alterations in the mRNA, miRNA and enzymatic profiles that lead to the control of infection. Furthermore, the genetic background of both parasite and host are also important to define the fate of infection.
Collapse
Affiliation(s)
- Sandra M Muxel
- Department of Physiology, Institute of Biosciences, University of São Paulo, São Paulo, Brazil
| | - Juliana I Aoki
- Department of Physiology, Institute of Biosciences, University of São Paulo, São Paulo, Brazil
| | - Juliane C R Fernandes
- Department of Physiology, Institute of Biosciences, University of São Paulo, São Paulo, Brazil
| | | | - Ricardo A Zampieri
- Department of Physiology, Institute of Biosciences, University of São Paulo, São Paulo, Brazil
| | - Stephanie M Acuña
- Department of Physiology, Institute of Biosciences, University of São Paulo, São Paulo, Brazil
| | - Karl E Müller
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Rubia H Vanderlinde
- Department of Physiology, Institute of Biosciences, University of São Paulo, São Paulo, Brazil
| | - Lucile M Floeter-Winter
- Department of Physiology, Institute of Biosciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
73
|
Morais M, Ferreira VFC, Figueira F, Mendes F, Raposinho P, Santos I, Oliveira BL, Correia JDG. Technetium-99m complexes of l-arginine derivatives for targeting amino acid transporters. Dalton Trans 2017; 46:14537-14547. [PMID: 28612866 DOI: 10.1039/c7dt01146f] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Although relevant from the clinical point of view, radiotracers targeting cationic amino acid transporters are relatively unexplored and, in particular, no metal-based radiotracers are known. The rare examples of complexes recognized by amino acid transporters, namely by the Na+-independent neutral l-type amino acid transporter 1 (LAT1), are 99mTc(i)/Re(i) compounds. Herein, we describe conjugates comprising a pyrazolyl-diamine chelating unit and the cationic amino acid l-arginine (l-Arg) linked by a propyl (L1) or hexyl linker (L2), which allowed the preparation of stable complexes of the type fac-[99mTc(CO)3(k3-L)]+ (Tc1, L = L1; Tc2, L = L2) and of the respective surrogates Re1 and Re2. Interestingly, complex Tc2 exhibited moderate levels of time-dependent internalization in three human tumoural cell lines, with approximately 3% of total applied activity internalized, corresponding to 21% of the cell-associated activity. A putative mechanism of retention in the cytoplasm of cells could be the interaction of the complex with inducible nitric oxide synthase (iNOS), which is the enzyme responsible for the catalytic oxidation of l-Arg to citrulline and nitric oxide. However, the surrogate complex Re2 does not recognize iNOS, as demonstrated by the in vitro assays with purified iNOS and in studies with lipopolysaccharide(LPS)-activated macrophages. Preliminary mechanistic studies suggest that the internalization of Tc2 is linked to the cationic amino acid transporters, namely system y+. This finding might open the way towards the development of novel families of metal-based radiotracers for probing metabolically active cancer cells.
Collapse
Affiliation(s)
- Maurício Morais
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10 (km 139.7), 2695-066 Bobadela LRS, Portugal.
| | | | | | | | | | | | | | | |
Collapse
|
74
|
Fluorine-18 labeled amino acids for tumor PET/CT imaging. Oncotarget 2017; 8:60581-60588. [PMID: 28947996 PMCID: PMC5601164 DOI: 10.18632/oncotarget.19943] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 07/25/2017] [Indexed: 12/19/2022] Open
Abstract
Tumor glucose metabolism and amino acid metabolism are usually enhanced, 18F-FDG for tumor glucose metabolism PET imaging has been clinically well known, but tumor amino acid metabolism PET imaging is not clinically familiar. Radiolabeled amino acids (AAs) are an important class of PET/CT tracers that target the upregulated amino acid transporters to show elevated amino acid metabolism in tumor cells. Radiolabeled amino acids were observed to have high uptake in tumor cells but low in normal tissues and inflammatory tissues. The radionuclides used in labeling amino acids include 15O, 13N, 11C, 123I, 18F and 68Ga, among which the most commonly used is 18F [1]. Available data support the use of certain 18F-labeled AAs for PET/CT imaging of gliomas, neuroendocrine tumors, prostate cancer and breast cancer [2, 3]. With the progress of the method of 18F labeling AAs [4-6], 18F-labeled AAs are well established for tumor PET/CT imaging. This review focuses on the current status of key clinical applications of 18F-labeled AAs in tumor PET/CT imaging.
Collapse
|
75
|
Dinkeloo K, Boyd S, Pilot G. Update on amino acid transporter functions and on possible amino acid sensing mechanisms in plants. Semin Cell Dev Biol 2017; 74:105-113. [PMID: 28705659 DOI: 10.1016/j.semcdb.2017.07.010] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 06/30/2017] [Accepted: 07/07/2017] [Indexed: 12/25/2022]
Abstract
Amino acids are essential components of plant metabolism, not only as constituents of proteins, but also as precursors of important secondary metabolites and as carriers of organic nitrogen between the organs of the plant. Transport across intracellular membranes and translocation of amino acids within the plant is mediated by membrane amino acid transporters. The past few years have seen the identification of a new family of amino acid transporters in Arabidopsis, the characterization of intracellular amino acid transporters, and the discovery of new roles for already known proteins. While amino acid metabolism needs to be tightly coordinated with amino acid transport activity and carbohydrate metabolism, no gene involved in amino acid sensing in plants has been unequivocally identified to date. This review aims at summarizing the recent data accumulated on the identity and function of amino acid transporters in plants, and discussing the possible identity of amino acid sensors based on data from other organisms.
Collapse
Affiliation(s)
- Kasia Dinkeloo
- Department of Plant Pathology, Physiology and Weed Science, Virginia Tech, Blacksburg, VA 24060, USA
| | - Shelton Boyd
- Department of Plant Pathology, Physiology and Weed Science, Virginia Tech, Blacksburg, VA 24060, USA
| | - Guillaume Pilot
- Department of Plant Pathology, Physiology and Weed Science, Virginia Tech, Blacksburg, VA 24060, USA.
| |
Collapse
|
76
|
The prognostic biomarker L-homoarginine is a substrate of the cationic amino acid transporters CAT1, CAT2A and CAT2B. Sci Rep 2017; 7:4767. [PMID: 28684763 PMCID: PMC5500509 DOI: 10.1038/s41598-017-04965-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 05/22/2017] [Indexed: 02/05/2023] Open
Abstract
Low plasma concentration of L-homoarginine is an independent predictor of cardiovascular events and total mortality. Experimental data indicate that supplementation of L-homoarginine may have protective effects. We aimed to elucidate the mechanisms involved in the cellular uptake of L-homoarginine, which are little understood, so far. Using human embryonic kidney (HEK293) cell lines stably overexpressing the human cationic amino acid transporters CAT1 [solute carrier family 7 (SLC7A1)], CAT2A (SLC7A2A) or CAT2B (SLC7A2B) we assessed the transport kinetics of L-homoarginine and interactions with the CAT substrates L-arginine and asymmetric dimethylarginine (ADMA). Significant uptake of L-homoarginine was observed for all three CATs with apparent KM-values of 175 ± 7 µM for CAT1 and 523 ± 35 µM for CAT2B. Saturation of CAT2A-mediated L-homoarginine uptake could not be reached. Uptake of L-homoarginine by any of the three CATs could be inhibited by L-arginine and ADMA. Significant inhibition of CAT1-mediated uptake of L-homoarginine by L-arginine already occurred in the physiological concentration range. Taken together these data demonstrate that L-homoarginine is a substrate of CAT1, CAT2A and CAT2B and that CAT1 is a key site with regard to physiological relevance and interactions with related substrates such as L-arginine.
Collapse
|
77
|
Abstract
Prior to infecting erythrocytes and causing malaria symptoms, Plasmodium parasites undergo an obligatory phase of invasion and extensive replication inside their mammalian host's liver cells that depends on the parasite's ability to obtain the nutrients it requires for its intra-hepatic growth and multiplication. Here, we show that L-arginine (Arg) uptake through the host cell's SLC7A2-encoded transporters is essential for the parasite's development and maturation in the liver. Our data suggest that the Arg that is taken up is primarily metabolized by the arginase pathway to produce the polyamines required for Plasmodium growth. Although the parasite may hijack the host's biosynthesis pathway, it relies mainly upon its own arginase-AdoMetDC/ODC pathway to acquire the polyamines it needs to develop. These results identify for the first time a pivotal role for Arg-dependent polyamine production during Plasmodium's hepatic development and pave the way to the exploitation of strategies to impact liver infection by the malaria parasite through the modulation of Arg uptake and polyamine synthesis.
Collapse
|
78
|
Wu Y, Yin Q, Lin S, Huang X, Xia Q, Chen Z, Zhang X, Yang D. Increased SLC7A8 expression mediates L-DOPA uptake by renal tubular epithelial cells. Mol Med Rep 2017; 16:887-893. [DOI: 10.3892/mmr.2017.6620] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Accepted: 01/17/2017] [Indexed: 11/05/2022] Open
|
79
|
Dai R, Peng F, Xiao X, Gong X, Jiang Y, Zhang M, Tian Y, Xu Y, Ma J, Li M, Luo Y, Gong G. Hepatitis B virus X protein-induced upregulation of CAT-1 stimulates proliferation and inhibits apoptosis in hepatocellular carcinoma cells. Oncotarget 2017; 8:60962-60974. [PMID: 28977838 PMCID: PMC5617398 DOI: 10.18632/oncotarget.17631] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 04/23/2017] [Indexed: 12/21/2022] Open
Abstract
The HBx protein of hepatitis B virus (HBV) is widely recognized to be a critical oncoprotein contributing to the development of HBV-related hepatocellular carcinoma (HCC). In addition, cationic amino acid transporter 1 (CAT-1) gene is a target of miR-122. In this study, we found that CAT-1 protein levels were higher in HBV-related HCC carcinomatous tissues than in para-cancerous tumor tissues, and that CAT-1 promoted HCC cell growth, proliferation, and metastasis. Moreover, HBx-induced decreases in Gld2 and miR-122 levels that contributed to the upregulation of CAT-1 in HCC. These results indicate that a Gld2/miR-122/CAT-1 pathway regulated by HBx likely participates in HBV-related hepatocellular carcinogenesis.
Collapse
Affiliation(s)
- Rongjuan Dai
- Department of Infectious Diseases, Institute of Hepatology Central South University, Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, PR China
| | - Feng Peng
- Department of Infectious Diseases, Institute of Hepatology Central South University, Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, PR China
| | - Xinqiang Xiao
- Department of Infectious Diseases, Institute of Hepatology Central South University, Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, PR China
| | - Xing Gong
- Department of Infectious Diseases, Institute of Hepatology Central South University, Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, PR China
| | - Yongfang Jiang
- Department of Infectious Diseases, Institute of Hepatology Central South University, Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, PR China
| | - Min Zhang
- Department of Infectious Diseases, Institute of Hepatology Central South University, Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, PR China
| | - Yi Tian
- Department of Infectious Diseases, Institute of Hepatology Central South University, Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, PR China
| | - Yun Xu
- Department of Infectious Diseases, Institute of Hepatology Central South University, Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, PR China
| | - Jing Ma
- Department of Infectious Diseases, Institute of Hepatology Central South University, Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, PR China
| | - Mingming Li
- Department of Infectious Diseases, Institute of Hepatology Central South University, Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, PR China
| | - Yue Luo
- Department of Infectious Diseases, Institute of Hepatology Central South University, Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, PR China
| | - Guozhong Gong
- Department of Infectious Diseases, Institute of Hepatology Central South University, Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, PR China
| |
Collapse
|
80
|
Crouse MS, McLean KJ, Crosswhite MR, Reynolds LP, Dahlen CR, Neville BW, Borowicz PP, Caton JS. Nutrient transporters in bovine uteroplacental tissues on days sixteen to fifty of gestation. J Anim Sci 2017; 94:4738-4747. [PMID: 27898936 DOI: 10.2527/jas.2016-0857] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
During early gestation, nutrients are transported to the developing embryo via transporters in the uterine endometrium and chorioallantois. In the present study, we examined glucose transporters and and the cationic AA transporters , , and to test the hypotheses that 1) relative mRNA expression of transporters would be different among uteroplacental tissue type as gestation progresses and 2) concentrations of glucose and cationic AA would be different among target sites (placental compartments, serum, and histotrophic) and days of gestation. To test these hypotheses, crossbred Angus heifers ( = 46) were synchronized, bred via AI, and then ovariohysterectomized on d 16, 22, 28, 34, 40, or 50 of gestation (5 to 9/d) or not bred and ovariohysterectomized on d 16 of the synchronized estrous cycle ( = 7) to serve as nonpregnant (NP) controls. Uteroplacental tissues (maternal caruncle [CAR], intercaruncular endometrium [ICAR], and fetal membranes [FM; chorioallantois, d 22 and later]) were collected from the uterine horn ipsilateral to the corpus luteum immediately following ovariohysterectomy. Relative mRNA expression of the glucose transporters and cationic AA transporters was determined for each tissue from d 16 to 50 of gestation and from NP controls. Chorioallantoic, amniotic, and plasma fluids were collected from heifers on d 40 and 50 of gestation to determine concentrations of glucose and cationic AA. Expression of and showed a tendency ( < 0.10) toward being greater in d 16 ICAR and d 34 ICAR, respectively. Day × tissue interactions ( < 0.05) were present for , , and . Expression of was greater in d 50 CAR, expression of was greater on d 34 in ICAR, and expression of was greater in CAR tissue on d 34 compared with all other tissues and days of gestation. Glucose concentrations tended ( = 0.10) to be impacted by a day × fluid interaction. A day × fluid interaction ( = 0.01) for arginine concentration was observed, with greater concentrations in allantoic fluid on d 40 compared with all other days and fluid types. These data support our hypothesis that glucose and cationic AA transporters differ in their level of mRNA expression due to day of gestation and uteroplacental tissue type. In addition, concentrations of nutrients were differentially impacted by day, target site, and/or their respective interaction.
Collapse
|
81
|
Rajendran E, Hapuarachchi SV, Miller CM, Fairweather SJ, Cai Y, Smith NC, Cockburn IA, Bröer S, Kirk K, van Dooren GG. Cationic amino acid transporters play key roles in the survival and transmission of apicomplexan parasites. Nat Commun 2017; 8:14455. [PMID: 28205520 PMCID: PMC5316894 DOI: 10.1038/ncomms14455] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 12/30/2016] [Indexed: 11/09/2022] Open
Abstract
Apicomplexans are obligate intracellular parasites that scavenge essential nutrients from their hosts via transporter proteins on their plasma membrane. The identities of the transporters that mediate amino acid uptake into apicomplexans are unknown. Here we demonstrate that members of an apicomplexan-specific protein family-the Novel Putative Transporters (NPTs)-play key roles in the uptake of cationic amino acids. We show that an NPT from Toxoplasma gondii (TgNPT1) is a selective arginine transporter that is essential for parasite survival and virulence. We also demonstrate that a homologue of TgNPT1 from the malaria parasite Plasmodium berghei (PbNPT1), shown previously to be essential for the sexual gametocyte stage of the parasite, is a cationic amino acid transporter. This reveals a role for cationic amino acid scavenging in gametocyte biology. Our study demonstrates a critical role for amino acid transporters in the survival, virulence and life cycle progression of these parasites.
Collapse
Affiliation(s)
- Esther Rajendran
- Research School of Biology, Australian National University, Canberra, Australian Capital Territory 2601, Australia
| | - Sanduni V Hapuarachchi
- Research School of Biology, Australian National University, Canberra, Australian Capital Territory 2601, Australia
| | - Catherine M Miller
- College of Public Health, Medical and Veterinary Sciences, James Cook University, Smithfield, Queensland 4878, Australia
| | - Stephen J Fairweather
- Research School of Biology, Australian National University, Canberra, Australian Capital Territory 2601, Australia
| | - Yeping Cai
- John Curtin School of Medical Research, Australian National University, Canberra, Australian Capital Territory 2601, Australia
| | - Nicholas C Smith
- Queensland Tropical Health Alliance Research Laboratory, Australian Institute of Tropical Health and Medicine, James Cook University, Smithfield, Queensland 4878, Australia
| | - Ian A Cockburn
- John Curtin School of Medical Research, Australian National University, Canberra, Australian Capital Territory 2601, Australia
| | - Stefan Bröer
- Research School of Biology, Australian National University, Canberra, Australian Capital Territory 2601, Australia
| | - Kiaran Kirk
- Research School of Biology, Australian National University, Canberra, Australian Capital Territory 2601, Australia
| | - Giel G van Dooren
- Research School of Biology, Australian National University, Canberra, Australian Capital Territory 2601, Australia
| |
Collapse
|
82
|
Morris SM. Arginine Metabolism Revisited. J Nutr 2016; 146:2579S-2586S. [PMID: 27934648 DOI: 10.3945/jn.115.226621] [Citation(s) in RCA: 251] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 01/22/2016] [Accepted: 02/05/2016] [Indexed: 01/20/2023] Open
Abstract
Mammalian arginine metabolism is complex due to the expression of multiple enzymes that utilize arginine as substrate and to interactions or competition between specific enzymes involved in arginine metabolism. Moreover, cells may contain multiple intracellular arginine pools that are not equally accessible to all arginine metabolic enzymes, thus presenting additional challenges to more fully understanding arginine metabolism. At the whole-body level, arginine metabolism ultimately results in the production of a biochemically diverse range of products, including nitric oxide, urea, creatine, polyamines, proline, glutamate, agmatine, and homoarginine. Included in this group of compounds are the methylated arginines (e.g., asymmetric dimethylarginine), which are released upon degradation of proteins containing methylated arginine residues. Changes in arginine concentration also can regulate cellular metabolism and function via a variety of arginine sensors. Although much is known about arginine metabolism, elucidation of the physiologic or pathophysiologic roles for all of the pathways and their metabolites remains an active area of investigation, as exemplified by current findings highlighted in this review.
Collapse
Affiliation(s)
- Sidney M Morris
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA
| |
Collapse
|
83
|
Singh K, Al-Greene NT, Verriere TG, Coburn LA, Asim M, Barry DP, Allaman MM, Hardbower DM, Delgado AG, Piazuelo MB, Vallance BA, Gobert AP, Wilson KT. The L-Arginine Transporter Solute Carrier Family 7 Member 2 Mediates the Immunopathogenesis of Attaching and Effacing Bacteria. PLoS Pathog 2016; 12:e1005984. [PMID: 27783672 PMCID: PMC5081186 DOI: 10.1371/journal.ppat.1005984] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 10/06/2016] [Indexed: 12/17/2022] Open
Abstract
Solute carrier family 7 member 2 (SLC7A2) is an inducible transporter of the semi-essential amino acid L-arginine (L-Arg), which has been implicated in immune responses to pathogens. We assessed the role of SLC7A2 in murine infection with Citrobacter rodentium, an attaching and effacing enteric pathogen that causes colitis. Induction of SLC7A2 was upregulated in colitis tissues, and localized predominantly to colonic epithelial cells. Compared to wild-type mice, Slc7a2–/–mice infected with C. rodentium had improved survival and decreased weight loss, colon weight, and histologic injury; this was associated with decreased colonic macrophages, dendritic cells, granulocytes, and Th1 and Th17 cells. In infected Slc7a2–/–mice, there were decreased levels of the proinflammatory cytokines G-CSF, TNF-α, IL-1α, IL-1β, and the chemokines CXCL1, CCL2, CCL3, CCL4, CXCL2, and CCL5. In bone marrow chimeras, the recipient genotype drove the colitis phenotype, indicative of the importance of epithelial, rather than myeloid SLC7A2. Mice lacking Slc7a2 exhibited reduced adherence of C. rodentium to the colonic epithelium and decreased expression of Talin-1, a focal adhesion protein involved in the attachment of the bacterium. The importance of SLC7A2 and Talin-1 in the intimate attachment of C. rodentium and induction of inflammatory response was confirmed in vitro, using conditionally-immortalized young adult mouse colon (YAMC) cells with shRNA knockdown of Slc7a2 or Tln1. Inhibition of L-Arg uptake with the competitive inhibitor, L-lysine (L-Lys), also prevented attachment of C. rodentium and chemokine expression. L-Lys and siRNA knockdown confirmed the role of L-Arg and SLC7A2 in human Caco-2 cells co-cultured with enteropathogenic Escherichia coli. Overexpression of SLC7A2 in human embryonic kidney cells increased bacterial adherence and chemokine expression. Taken together, our data indicate that C. rodentium enhances its own pathogenicity by inducing the expression of SLC7A2 to favor its attachment to the epithelium and thus create its ecological niche. Intestinal infections by attaching and effacing (A/E) bacteria widely impact human health, with major social and economic repercussions. Mucosal immunity plays a critical role in determining the outcome of these infections. The amino acid L-arginine regulates inflammatory responses to bacterial pathogens. We studied the role of the L-arginine transporter solute carrier family 7 member 2 (SLC7A2) during infection with the A/E pathogen Citrobacter rodentium. SLC7A2 is induced in colonic epithelial cells during the infection and facilitates the intimate attachment of the bacteria, thus initiating the inflammatory response of the infected mucosa. These data were confirmed in vitro using C. rodentium-infected mouse cells and human colonic epithelial cells infected with enteropathogenic Escherichia coli. Our work describes a mechanism by which A/E bacteria manipulate host response to favor their colonization, thereby positioning SLC7A2 as an unrecognized therapeutic target to limit infection with enterobacteria.
Collapse
Affiliation(s)
- Kshipra Singh
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Nicole T. Al-Greene
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Thomas G. Verriere
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Lori A. Coburn
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee, United States of America
| | - Mohammad Asim
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Daniel P. Barry
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Margaret M. Allaman
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Dana M. Hardbower
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Alberto G. Delgado
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - M. Blanca Piazuelo
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Bruce A. Vallance
- Division of Gastroenterology, Department of Pediatrics, Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Alain P. Gobert
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Keith T. Wilson
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee, United States of America
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- * E-mail:
| |
Collapse
|
84
|
Crespo K, Ménard A, Deng AY. Retinoblastoma-associated protein 140 as a candidate for a novel etiological gene to hypertension. Clin Exp Hypertens 2016; 38:533-40. [PMID: 27391979 DOI: 10.3109/10641963.2016.1163373] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Gene discovery in animal models may lead to the revelation of therapeutic targets for essential hypertension as well as mechanistic insights into blood pressure (BP) regulation. Our aim was to identify a disease-causing gene for a component of polygenic hypertension contrasting inbred hypertensive Dahl salt-sensitive (DSS) and normotensive Lewis rats. The chromosome segment harboring a quantitative trait locus (QTL), C16QTL, was first isolated from the rat genome via congenic strains. A candidate gene responsible for C16QTL causing a BP difference between DSS and Lewis rats was then identified using molecular analyses combining our independently-conducted total genome and gene-specific sequencings. The retinoblastoma-associated protein 140 (Rap140)/family with sequence similarity 208 member A (Fam208a) is the only candidate gene supported to be C16QTL among three genes in genome block 1 present in the C16QTL-residing interval. A mode of its actions could be to influence the expressions of genes that are downstream in a pathway potentially leading to BP regulation such as that encoding the solute carrier family 7 (cationic amino acid transporter, y+ system) member 12 (Slc7a12), which is specifically expressed in kidneys. Thus, Rap140/Fam208a probably encoding a transcription factor is the strongest candidate for a novel BP QTL that acts via a putative Rap140/Fam208a-Slc7a12-BP pathway. These data implicate a premier physiological role for Rap140/Fam208 beyond development and a first biological function for the Slc7a12 protein in any organism.
Collapse
Affiliation(s)
- Kimberley Crespo
- a Department of Medicine , Université de Montréal, Research Center, CRCHUM (Center Hospitalier de l'Université de Montréal) , Montréal , Québec , Canada
| | - Annie Ménard
- a Department of Medicine , Université de Montréal, Research Center, CRCHUM (Center Hospitalier de l'Université de Montréal) , Montréal , Québec , Canada
| | - Alan Y Deng
- a Department of Medicine , Université de Montréal, Research Center, CRCHUM (Center Hospitalier de l'Université de Montréal) , Montréal , Québec , Canada
| |
Collapse
|
85
|
Heteromeric amino acid transporters. In search of the molecular bases of transport cycle mechanisms1. Biochem Soc Trans 2016; 44:745-52. [DOI: 10.1042/bst20150294] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Indexed: 01/18/2023]
Abstract
Heteromeric amino acid transporters (HATs) are relevant targets for structural studies. On the one hand, HATs are involved in inherited and acquired human pathologies. On the other hand, these molecules are the only known examples of solute transporters composed of two subunits (heavy and light) linked by a disulfide bridge. Unfortunately, structural knowledge of HATs is scarce and limited to the atomic structure of the ectodomain of a heavy subunit (human 4F2hc-ED) and distant prokaryotic homologues of the light subunits that share a LeuT-fold. Recent data on human 4F2hc/LAT2 at nanometer resolution revealed 4F2hc-ED positioned on top of the external loops of the light subunit LAT2. Improved resolution of the structure of HATs, combined with conformational studies, is essential to establish the structural bases for light subunit recognition and to evaluate the functional relevance of heavy and light subunit interactions for the amino acid transport cycle.
Collapse
|
86
|
Cremades A, Del Rio-Garcia J, Lambertos A, López-Garcia C, Peñafiel R. Tissue-specific regulation of potassium homeostasis by high doses of cationic amino acids. SPRINGERPLUS 2016; 5:616. [PMID: 27330882 PMCID: PMC4870509 DOI: 10.1186/s40064-016-2224-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 04/22/2016] [Indexed: 01/19/2023]
Abstract
The administration of l-arginine hydrochloride has been used for testing pituitary secretion in humans, and as an experimental model for induction of acute pancreatitis in rats and mice. Whereas in the first case, the administration of the amino acid is associated with hiperkalemia, in the model of acute pancreatitis no data are available on possible changes in potassium homeostasis. The present study shows that the acute administration to mice of l-arginine hydrochloride or other cationic amino acids almost duplicate plasma potassium levels. This effect was associated to a marked decrease of tissue potassium in both pancreas and liver. No changes were found in other tissues. These changes cannot be ascribed to the large load of chloride ions, since similar effects were produced when l-ornithine aspartate was administered. The changes in potassium levels were dependent on the dose. The displacement of intracellular potassium from the liver and pancreas to the extracellular compartment appears to be dependent on the entry of the cationic amino acid, since the administration of an equivalent dose of alfa-difluoromethyl ornithine HCl (DFMO), a non physiological analog of l-ornithine, which is poorly taken by the tissues in comparison with the physiological cationic amino acids, did not produce any change in potassium levels in pancreas and liver. The analyses of the expression of cationic amino acid transporters (CAT) suggest that the CAT-2 transporter may be implicated in the potassium/cationic amino acid interchange in liver and pancreas. The possible physiological or pathological relevance of these findings is discussed.
Collapse
Affiliation(s)
- Asunción Cremades
- Department of Pharmacology, Faculty of Medicine, University of Murcia, Murcia, Spain ; Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
| | - Jesús Del Rio-Garcia
- Department of Pharmacology, Faculty of Medicine, University of Murcia, Murcia, Spain
| | - Ana Lambertos
- Department of Biochemistry, Molecular Biology B and Immunology, School of Medicine, University of Murcia, 30100 Murcia, Spain
| | - Carlos López-Garcia
- Department of Pharmacology, Faculty of Medicine, University of Murcia, Murcia, Spain
| | - Rafael Peñafiel
- Department of Biochemistry, Molecular Biology B and Immunology, School of Medicine, University of Murcia, 30100 Murcia, Spain ; Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
| |
Collapse
|
87
|
Advances in Autophagy Regulatory Mechanisms. Cells 2016; 5:cells5020024. [PMID: 27187479 PMCID: PMC4931673 DOI: 10.3390/cells5020024] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Revised: 04/20/2016] [Accepted: 05/05/2016] [Indexed: 12/19/2022] Open
Abstract
Autophagy plays a critical role in cell metabolism by degrading and recycling internal components when challenged with limited nutrients. This fundamental and conserved mechanism is based on a membrane trafficking pathway in which nascent autophagosomes engulf cytoplasmic cargo to form vesicles that transport their content to the lysosome for degradation. Based on this simple scheme, autophagy modulates cellular metabolism and cytoplasmic quality control to influence an unexpectedly wide range of normal mammalian physiology and pathophysiology. In this review, we summarise recent advancements in three broad areas of autophagy regulation. We discuss current models on how autophagosomes are initiated from endogenous membranes. We detail how the uncoordinated 51-like kinase (ULK) complex becomes activated downstream of mechanistic target of rapamycin complex 1 (MTORC1). Finally, we summarise the upstream signalling mechanisms that can sense amino acid availability leading to activation of MTORC1.
Collapse
|
88
|
Dubó S, Gallegos D, Cabrera L, Sobrevia L, Zúñiga L, González M. Cardiovascular Action of Insulin in Health and Disease: Endothelial L-Arginine Transport and Cardiac Voltage-Dependent Potassium Channels. Front Physiol 2016; 7:74. [PMID: 27014078 PMCID: PMC4791397 DOI: 10.3389/fphys.2016.00074] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 02/15/2016] [Indexed: 12/19/2022] Open
Abstract
Impairment of insulin signaling on diabetes mellitus has been related to cardiovascular dysfunction, heart failure, and sudden death. In human endothelium, cationic amino acid transporter 1 (hCAT-1) is related to the synthesis of nitric oxide (NO) and insulin has a vascular effect in endothelial cells through a signaling pathway that involves increases in hCAT-1 expression and L-arginine transport. This mechanism is disrupted in diabetes, a phenomenon potentiated by excessive accumulation of reactive oxygen species (ROS), which contribute to lower availability of NO and endothelial dysfunction. On the other hand, electrical remodeling in cardiomyocytes is considered a key factor in heart failure progression associated to diabetes mellitus. This generates a challenge to understand the specific role of insulin and the pathways involved in cardiac function. Studies on isolated mammalian cardiomyocytes have shown prolongated action potential in ventricular repolarization phase that produces a long QT interval, which is well explained by attenuation in the repolarizing potassium currents in cardiac ventricles. Impaired insulin signaling causes specific changes in these currents, such a decrease amplitude of the transient outward K(+) (Ito) and the ultra-rapid delayed rectifier (IKur) currents where, together, a reduction of mRNA and protein expression levels of α-subunits (Ito, fast; Kv 4.2 and IKs; Kv 1.5) or β-subunits (KChIP2 and MiRP) of K(+) channels involved in these currents in a MAPK mediated pathway process have been described. These results support the hypothesis that lack of insulin signaling can produce an abnormal repolarization in cardiomyocytes. Furthermore, the arrhythmogenic potential due to reduced Ito current can contribute to an increase in the incidence of sudden death in heart failure. This review aims to show, based on pathophysiological models, the regulatory function that would have insulin in vascular system and in cardiac electrophysiology.
Collapse
Affiliation(s)
- Sebastián Dubó
- Department of Kinesiology, Faculty of Medicine, Universidad de Concepción Concepción, Chile
| | - David Gallegos
- Vascular Physiology Laboratory, Department of Physiology, Faculty of Biological Sciences, Universidad de Concepción Concepción, Chile
| | - Lissette Cabrera
- Vascular Physiology Laboratory, Department of Physiology, Faculty of Biological Sciences, Universidad de ConcepciónConcepción, Chile; Department of Morphophysiology, Faculty of Medicine, Universidad Diego PortalesSantiago, Chile
| | - Luis Sobrevia
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynecology, Faculty of Medicine, School of Medicine, Pontificia Universidad Católica de ChileSantiago, Chile; Department of Physiology, Faculty of Pharmacy, Universidad de SevillaSeville, Spain; Faculty of Medicine and Biomedical Sciences, University of Queensland Centre for Clinical Research (UQCCR), University of QueenslandHerston, QLD, Queensland, Australia
| | - Leandro Zúñiga
- Centro de Investigaciones Médicas, Escuela de Medicina, Universidad de Talca Talca, Chile
| | - Marcelo González
- Vascular Physiology Laboratory, Department of Physiology, Faculty of Biological Sciences, Universidad de ConcepciónConcepción, Chile; Group of Research and Innovation in Vascular Health (GRIVAS-Health)Chillán, Chile
| |
Collapse
|
89
|
Chen MX, Li XG, Yang JX, Gao CQ, Wang B, Wang XQ, Yan HC. Growth of embryo and gene expression of nutrient transporters in the small intestine of the domestic pigeon (Columba livia). J Zhejiang Univ Sci B 2016; 16:511-23. [PMID: 26055913 DOI: 10.1631/jzus.b1400340] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The objective of this study was to investigate the relationship between gene expression of nutrient (amino acid, peptide, sodium and proton) transporters in the small intestine and embryonic growth in domestic pigeons (Columba livia). One hundred and twenty-five fertilized eggs were randomly assigned into five groups and were incubated under optimal conditions (temperature of 38.1 °C and relative humidity of 55%). Twenty embryos/birds from each group were sacrificed by cervical dislocation on embryonic day (E) 9, 11, 13, 15 and day of hatch (DOH). The eggs, embryos (without yolk sac), and organs (head, brain, heart, liver, lungs, kidney, gizzard, small intestine, legs, and thorax) were dissected, cleaned, and weighed. Small intestine samples were collected for RNA isolation. The mRNA abundance of intestinal nutrient transporters was evaluated by real-time reverse transcription-polymerase chain reaction (RT-PCR). We classified these ten organs into four types according to the changes in relative weight during embryonic development. In addition, the gene expression of nutrient transporters was differentially regulated by embryonic day. The mRNA abundances of b(0,+)AT, EAAT3, y(+)LAT2, PepT1, LAT4, NHE2, and NHE3 increased linearly with age, whereas mRNA abundances of CAT1, CAT2, LAT1, EAAT2, SNAT1, and SNAT2 were increased to higher levels on E9 or E11 and then decreased to lower levels until DOH. The results of correlation analysis showed that the gene expressions of b(0,+)AT, EAAT3, PepT1, LAT4, NHE2, NHE3, and y(+)LAT2 had positive correlations with body weight (0.71<correlation coefficient (CC)<0.82, P<0.0001), while CAT1, CAT2, EAAT2, SNAT1, and SNAT2 had negative correlations with body weight (-0.86<CC<-0.64, P<0.0001). The gene expressions of b(0,+)AT, EAAT3, LAT4, PepT1, NHE2, NHE3, and y(+)LAT2 showed positive correlations with intestinal weight (0.80<CC<0.91, P<0.0001), while CAT1, CAT2, and EAAT2 showed negative correlations with intestinal weight (-0.84<CC<-0.67, P<0.0001). It was concluded that the differences between growth trajectories of organs and gene expression of nutrient transporters in small intestine were due to their functional and physiological properties, which provided a comprehensive study of amino acid and peptide transporter mRNA in the small intestine during embryonic growth of pigeons.
Collapse
Affiliation(s)
- Ming-xia Chen
- College of Animal Science, South China Agricultural University / Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture / Guangdong Provincial Key Laboratory of Agro-Animal Genomics / South China Collaborative Innovatio Center for Poultry Disease Control and Product Safety, Guangzhou 510642, China; Guangdong Fengli Agricultural Comprehensive Development Co., Ltd., Maoming 525000, China
| | | | | | | | | | | | | |
Collapse
|
90
|
Polarization of immune responses in fish: The ‘macrophages first’ point of view. Mol Immunol 2016; 69:146-56. [DOI: 10.1016/j.molimm.2015.09.026] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 09/30/2015] [Accepted: 09/30/2015] [Indexed: 01/01/2023]
|
91
|
Real-time functional characterization of cationic amino acid transporters using a new FRET sensor. Pflugers Arch 2015; 468:563-72. [PMID: 26555760 DOI: 10.1007/s00424-015-1754-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 10/26/2015] [Accepted: 10/29/2015] [Indexed: 10/22/2022]
Abstract
L-arginine is a semi-essential amino acid that serves as precursor for the production of urea, nitric oxide (NO), polyamines, and other biologically important metabolites. Hence, a fast and reliable assessment of its intracellular concentration changes is highly desirable. Here, we report on a genetically encoded Förster resonance energy transfer (FRET)-based arginine nanosensor that employs the arginine repressor/activator ahrC gene from Bacillus subtilis. This new nanosensor was expressed in HEK293T cells, and experiments with cell lysate showed that it binds L-arginine with high specificity and with a K d of ∼177 μM. Live imaging experiments showed that the nanosensor was expressed throughout the cytoplasm and displayed a half maximal FRET increase at an extracellular L-arginine concentration of ∼22 μM. By expressing the nanosensor together with SLC7A1, SLC7A2B, or SLC7A3 cationic amino acid transporters (CAT1-3), it was shown that L-arginine was imported at a similar rate via SLC7A1 and SLC7A2B and slower via SLC7A3. In contrast, upon withdrawal of extracellular L-arginine, intracellular levels decreased as fast in SLC7A3-expressing cells compared with SLC7A1, but the efflux was slower via SLC7A2B. SLC7A4 (CAT4) could not be convincingly shown to transport L-arginine. We also demonstrated the impact of membrane potential on L-arginine transport and showed that physiological concentrations of symmetrical and asymmetrical dimethylarginine do not significantly interfere with L-arginine transport through SLC7A1. Our results demonstrate that the FRET nanosensor can be used to assess L-arginine transport through plasma membrane in real time.
Collapse
|
92
|
Zielińska M, Milewski K, Skowrońska M, Gajos A, Ziemińska E, Beręsewicz A, Albrecht J. Induction of inducible nitric oxide synthase expression in ammonia-exposed cultured astrocytes is coupled to increased arginine transport by upregulated y(+)LAT2 transporter. J Neurochem 2015; 135:1272-81. [PMID: 26448619 DOI: 10.1111/jnc.13387] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 09/10/2015] [Accepted: 09/25/2015] [Indexed: 12/17/2022]
Abstract
One of the aspects of ammonia toxicity to brain cells is increased production of nitric oxide (NO) by NO synthases (NOSs). Previously we showed that ammonia increases arginine (Arg) uptake in cultured rat cortical astrocytes specifically via y(+)L amino acid transport system, by activation of its member, a heteromeric y(+)LAT2 transporter. Here, we tested the hypothesis that up-regulation of y(+)LAT2 underlies ammonia-dependent increase of NO production via inducible NOS (iNOS) induction, and protein nitration. Treatment of rat cortical astrocytes for 48 with 5 mM ammonium chloride ('ammonia') (i) increased the y(+)L-mediated Arg uptake, (ii) raised the expression of iNOS and endothelial NOS (eNOS), (iii) stimulated NO production, as manifested by increased nitrite+nitrate (Griess) and/or nitrite alone (chemiluminescence), and consequently, (iv) evoked nitration of tyrosine residues of proteins in astrocytes. Except for the increase of eNOS, all the above described effects of ammonia were abrogated by pre-treatment of astrocytes with either siRNA silencing of the Slc7a6 gene coding for y(+)LAT2 protein, or antibody to y(+)LAT2, indicating their strict coupling to y(+)LAT2 activity. Moreover, induction of y(+)LAT2 expression by ammonia was sensitive to Nf-κB inhibitor, BAY 11-7085, linking y(+)LAT2 upregulation to the Nf-κB activation in this experimental setting as reported earlier and here confirmed. Importantly, ammonia did not affect y(+)LAT2 expression nor y(+)L-mediated Arg uptake activity in the cultured cerebellar neurons, suggesting astroglia-specificity of the above described mechanism. The described coupling of up-regulation of y(+)LAT2 transporter with iNOS in ammonia-exposed astrocytes may be considered as a mechanism to ensure NO supply for protein nitration. Ammonia (NH4(+)) increases the expression and activity of the L-arginine (Arg) transporter (Arg/neutral amino acids [NAA] exchanger) y(+)LAT2 in cultured rat cortical astrocytes by a mechanism involving activation (nuclear translocation) of the transcription factor nuclear factor-Nuclear factor-κB (Nf-κB-p65). Up-regulation of y(+)LAT2 transporter is coupled with increased inducible nitric oxide synthase (iNOS) expression, which leads to increase nitric oxide (NO) synthesis and protein nitration.
Collapse
Affiliation(s)
- Magdalena Zielińska
- Department of Neurotoxicology, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Krzysztof Milewski
- Department of Neurotoxicology, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Marta Skowrońska
- Department of Neurotoxicology, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Anna Gajos
- Medical Center of Postgraduate Education, Warsaw, Poland
| | - Elżbieta Ziemińska
- Department of Neurochemistry, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | | | - Jan Albrecht
- Department of Neurotoxicology, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
93
|
Werner A, Amann E, Schnitzius V, Habermeier A, Luckner-Minden C, Leuchtner N, Rupp J, Closs EI, Munder M. Induced arginine transport via cationic amino acid transporter-1 is necessary for human T-cell proliferation. Eur J Immunol 2015; 46:92-103. [PMID: 26449889 DOI: 10.1002/eji.201546047] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Revised: 09/03/2015] [Accepted: 10/05/2015] [Indexed: 11/10/2022]
Abstract
Availability of the semiessential amino acid arginine is fundamental for the efficient function of human T lymphocytes. Tumor-associated arginine deprivation, mainly induced by myeloid-derived suppressor cells, is a central mechanism of tumor immune escape from T-cell-mediated antitumor immune responses. We thus assumed that transmembranous transport of arginine must be crucial for T-cell function and studied which transporters are responsible for arginine influx into primary human T lymphocytes. Here, we show that activation via CD3 and CD28 induces arginine transport into primary human T cells. Both naïve and memory CD4(+) T cells as well as CD8(+) T cells specifically upregulated the human cationic amino acid transporter-1 (hCAT-1), with an enhanced and persistent expression under arginine starvation. When hCAT-1 induction was suppressed via siRNA transfection, arginine uptake, and cellular proliferation were impaired. In summary, our results demonstrate that hCAT-1 is a key component of efficient T-cell activation and a novel potential target structure to modulate adaptive immune responses in tumor immunity or inflammation.
Collapse
Affiliation(s)
- Anke Werner
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Eva Amann
- Third Department of Medicine (Hematology, Oncology, and Pneumology), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Vanessa Schnitzius
- Third Department of Medicine (Hematology, Oncology, and Pneumology), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Alice Habermeier
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Claudia Luckner-Minden
- Third Department of Medicine (Hematology, Oncology, and Pneumology), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Nadine Leuchtner
- Third Department of Medicine (Hematology, Oncology, and Pneumology), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Johanna Rupp
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Ellen I Closs
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Markus Munder
- Third Department of Medicine (Hematology, Oncology, and Pneumology), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.,Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
94
|
Substrate specificity and transport mechanism of amino-acid transceptor Slimfast from Aedes aegypti. Nat Commun 2015; 6:8546. [PMID: 26449545 PMCID: PMC4608377 DOI: 10.1038/ncomms9546] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 09/03/2015] [Indexed: 12/11/2022] Open
Abstract
Anautogenous mosquitoes depend on vertebrate blood as nutrient source for their eggs. A highly efficient set of membrane transporters mediates the massive movement of nutrient amino acids between mosquito tissues after a blood meal. Here we report the characterization of the amino-acid transporter Slimfast (Slif) from the yellow-fever mosquito Aedes aegypti using codon-optimized heterologous expression. Slif is a well-known component of the target-of-rapamycin signalling pathway and fat body nutrient sensor, but its substrate specificity and transport mechanism were unknown. We found that Slif transports essential cationic and neutral amino acids with preference for arginine. It has an unusual dual-affinity mechanism with only the high affinity being Na+ dependent. Tissue-specific expression and blood meal-dependent regulation of Slif are consistent with conveyance of essential amino acids from gut to fat body. Slif represents a novel transport system and type of transceptor for sensing and transporting essential amino acids during mosquito reproduction. Anautogenous mosquitoes need to obtain essential amino acids from a blood meal for reproduction. Here, the authors examine the amino acid transporter Slimfast from the yellow-fever mosquito and describe both its specificity and mechanism of action.
Collapse
|
95
|
Guo L, Tian S, Chen Y, Mao Y, Cui S, Hu A, Zhang J, Xia SL, Su Y, Du J, Block ER, Wang XL, Cui Z. CAT-1 as a novel CAM stabilizes endothelial integrity and mediates the protective actions of l-Arg via a NO-independent mechanism. J Mol Cell Cardiol 2015; 87:180-91. [DOI: 10.1016/j.yjmcc.2015.08.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 08/10/2015] [Accepted: 08/12/2015] [Indexed: 12/21/2022]
|
96
|
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
|
97
|
Wijnands KAP, Meesters DM, van Barneveld KWY, Visschers RGJ, Briedé JJ, Vandendriessche B, van Eijk HMH, Bessems BAFM, van den Hoven N, von Wintersdorff CJH, Brouckaert P, Bouvy ND, Lamers WH, Cauwels A, Poeze M. Citrulline Supplementation Improves Organ Perfusion and Arginine Availability under Conditions with Enhanced Arginase Activity. Nutrients 2015; 7:5217-38. [PMID: 26132994 PMCID: PMC4516994 DOI: 10.3390/nu7075217] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 05/15/2015] [Accepted: 06/18/2015] [Indexed: 01/10/2023] Open
Abstract
Enhanced arginase-induced arginine consumption is believed to play a key role in the pathogenesis of sickle cell disease-induced end organ failure. Enhancement of arginine availability with l-arginine supplementation exhibited less consistent results; however, l-citrulline, the precursor of l-arginine, may be a promising alternative. In this study, we determined the effects of l-citrulline compared to l-arginine supplementation on arginine-nitric oxide (NO) metabolism, arginine availability and microcirculation in a murine model with acutely-enhanced arginase activity. The effects were measured in six groups of mice (n = 8 each) injected intraperitoneally with sterile saline or arginase (1000 IE/mouse) with or without being separately injected with l-citrulline or l-arginine 1 h prior to assessment of the microcirculation with side stream dark-field (SDF)-imaging or in vivo NO-production with electron spin resonance (ESR) spectroscopy. Arginase injection caused a decrease in plasma and tissue arginine concentrations. l-arginine and l-citrulline supplementation both enhanced plasma and tissue arginine concentrations in arginase-injected mice. However, only the citrulline supplementation increased NO production and improved microcirculatory flow in arginase-injected mice. In conclusion, the present study provides for the first time in vivo experimental evidence that l-citrulline, and not l-arginine supplementation, improves the end organ microcirculation during conditions with acute arginase-induced arginine deficiency by increasing the NO concentration in tissues.
Collapse
Affiliation(s)
- Karolina A P Wijnands
- Department of Surgery, NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University Medical Center, Maastricht 6200 MD, The Netherlands.
| | - Dennis M Meesters
- Department of Surgery, NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University Medical Center, Maastricht 6200 MD, The Netherlands.
| | - Kevin W Y van Barneveld
- Department of Surgery, NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University Medical Center, Maastricht 6200 MD, The Netherlands.
| | - Ruben G J Visschers
- Department of Surgery, NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University Medical Center, Maastricht 6200 MD, The Netherlands.
| | - Jacob J Briedé
- Department of Toxicogenomics, GROW School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht 6200, The Netherlands.
| | - Benjamin Vandendriessche
- Department of Molecular Biomedical Research, VIB, Ghent B-9000, Belgium.
- Department of Biomedical Molecular Biology, Ghent University, Ghent B-9000, Belgium.
| | - Hans M H van Eijk
- Department of Surgery, NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University Medical Center, Maastricht 6200 MD, The Netherlands.
| | - Babs A F M Bessems
- Department of Surgery, NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University Medical Center, Maastricht 6200 MD, The Netherlands.
| | - Nadine van den Hoven
- Department of Surgery, NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University Medical Center, Maastricht 6200 MD, The Netherlands.
| | - Christian J H von Wintersdorff
- Department of Surgery, NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University Medical Center, Maastricht 6200 MD, The Netherlands.
| | - Peter Brouckaert
- Department of Molecular Biomedical Research, VIB, Ghent B-9000, Belgium.
- Department of Biomedical Molecular Biology, Ghent University, Ghent B-9000, Belgium.
| | - Nicole D Bouvy
- Department of Surgery, NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University Medical Center, Maastricht 6200 MD, The Netherlands.
| | - Wouter H Lamers
- Department of Anatomy & Embryology, Maastricht University Medical Center, Maastricht 6200, The Netherlands.
| | - Anje Cauwels
- Department of Molecular Biomedical Research, VIB, Ghent B-9000, Belgium.
- Department of Biomedical Molecular Biology, Ghent University, Ghent B-9000, Belgium.
| | - Martijn Poeze
- Department of Surgery, NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University Medical Center, Maastricht 6200 MD, The Netherlands.
| |
Collapse
|
98
|
Abstract
The pathogenesis of Alzheimer's disease (AD) is a critical unsolved question; and although recent studies have demonstrated a strong association between altered brain immune responses and disease progression, the mechanistic cause of neuronal dysfunction and death is unknown. We have previously described the unique CVN-AD mouse model of AD, in which immune-mediated nitric oxide is lowered to mimic human levels, resulting in a mouse model that demonstrates the cardinal features of AD, including amyloid deposition, hyperphosphorylated and aggregated tau, behavioral changes, and age-dependent hippocampal neuronal loss. Using this mouse model, we studied longitudinal changes in brain immunity in relation to neuronal loss and, contrary to the predominant view that AD pathology is driven by proinflammatory factors, we find that the pathology in CVN-AD mice is driven by local immune suppression. Areas of hippocampal neuronal death are associated with the presence of immunosuppressive CD11c(+) microglia and extracellular arginase, resulting in arginine catabolism and reduced levels of total brain arginine. Pharmacologic disruption of the arginine utilization pathway by an inhibitor of arginase and ornithine decarboxylase protected the mice from AD-like pathology and significantly decreased CD11c expression. Our findings strongly implicate local immune-mediated amino acid catabolism as a novel and potentially critical mechanism mediating the age-dependent and regional loss of neurons in humans with AD.
Collapse
|
99
|
Glushko AA, Voronkov AV, Chernikov MV. [Molecular targets for searching of endothelial-protective substances]. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2015; 40:515-27. [PMID: 25895347 DOI: 10.1134/s1068162014050069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Endothelial dysfunction underlies the development of many cardiovascular diseases. Thus endothelium becomes an independent therapeutic target, and the search of new substances with endothelial-protective action to date is one of the promising tasks for pharmacotherapy and medicinal chemistry. Molecular modeling is an effective tool for solving this problem. Computer chemistry methods use is only possible in combination with detailed information on three dimensional structure and functions of molecular targets: receptors and enzymes, involved in signal transduction inside and outside of endothelial cells. Information on structure and function of various macromolecules involved in vascular tone regulation is collected in the review. The structure of endothelial NO-synthase (EC 1.14.13.39) (eNOS)--enzyme, responsible for the nitric oxide synthesis and involved in vascular tone regulation process is reviewed. The importance of eNOS substrate--L-arginine is underlined in the review in terms of this enzyme activity, regulation, the information on structure and functions of L-arginine transport system is provided. Also different ways of eNOS activity regulation are reviewed, among which are enzyme activation and concurrent inhibition by substances interaction with active center of enzyme, inhibition by caveoline binding with oxigenase domain, and also regulation by phosphorylation of certain amino acids of eNOS by proteinkinase and dephoshphorylation of them by phosphatases. The importance of membrane receptors of endothelial cells as targets for endothelial-protective substances is underlined. Among them are receptors of endothelin, platelet activation factor, prostanoids, bradykinin, histamine, serotonin and protease activated receptors. The important role of potassium and calcium ion channels of vascular cells in endothelial-protective activity is underlined. Macromolecules presented in the review finally are considered as targets for searching for medicinal substances with endothelial-protective activity using proposed ways and methods of molecular modeling.
Collapse
|
100
|
Ramond E, Gesbert G, Guerrera IC, Chhuon C, Dupuis M, Rigard M, Henry T, Barel M, Charbit A. Importance of host cell arginine uptake in Francisella phagosomal escape and ribosomal protein amounts. Mol Cell Proteomics 2015; 14:870-81. [PMID: 25616868 PMCID: PMC4390266 DOI: 10.1074/mcp.m114.044552] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 01/19/2015] [Indexed: 11/06/2022] Open
Abstract
Upon entry into mammalian host cells, the pathogenic bacterium Francisella must import host cell arginine to multiply actively in the host cytoplasm. We identified and functionally characterized an arginine transporter (hereafter designated ArgP) whose inactivation considerably delayed bacterial phagosomal escape and intracellular multiplication. Intramacrophagic growth of the ΔargP mutant was fully restored upon supplementation of the growth medium with excess arginine, in both F. tularensis subsp. novicida and F. tularensis subsp. holarctica LVS, demonstrating the importance of arginine acquisition in these two subspecies. High-resolution mass spectrometry revealed that arginine limitation reduced the amount of most of the ribosomal proteins in the ΔargP mutant. In response to stresses such as nutritional limitation, repression of ribosomal protein synthesis has been observed in all kingdoms of life. Arginine availability may thus contribute to the sensing of the intracellular stage of the pathogen and to trigger phagosomal egress. All MS data have been deposited in the ProteomeXchange database with identifier PXD001584 (http://proteomecentral.proteomexchange.org/dataset/PXD001584).
Collapse
Affiliation(s)
- Elodie Ramond
- From the ‡Université Paris Descartes, Sorbonne Paris Cité, Bâtiment Leriche; §INSERM U1151-CNRS UMR 8253, Institut Necker-Enfants Malades, Team 11. Paris, France
| | - Gael Gesbert
- From the ‡Université Paris Descartes, Sorbonne Paris Cité, Bâtiment Leriche; §INSERM U1151-CNRS UMR 8253, Institut Necker-Enfants Malades, Team 11. Paris, France
| | - Ida Chiara Guerrera
- From the ‡Université Paris Descartes, Sorbonne Paris Cité, Bâtiment Leriche; ¶Plateforme Protéome Institut Necker, PPN, Structure Fédérative de Recherche SFR Necker, Université Paris Descartes, Paris 75015 France
| | - Cerina Chhuon
- From the ‡Université Paris Descartes, Sorbonne Paris Cité, Bâtiment Leriche; ¶Plateforme Protéome Institut Necker, PPN, Structure Fédérative de Recherche SFR Necker, Université Paris Descartes, Paris 75015 France
| | - Marion Dupuis
- From the ‡Université Paris Descartes, Sorbonne Paris Cité, Bâtiment Leriche; §INSERM U1151-CNRS UMR 8253, Institut Necker-Enfants Malades, Team 11. Paris, France
| | - Mélanie Rigard
- ‖CIRI, Centre International de Recherche en Infectiologie, Lyon, France
| | - Thomas Henry
- ‖CIRI, Centre International de Recherche en Infectiologie, Lyon, France
| | - Monique Barel
- From the ‡Université Paris Descartes, Sorbonne Paris Cité, Bâtiment Leriche; §INSERM U1151-CNRS UMR 8253, Institut Necker-Enfants Malades, Team 11. Paris, France
| | - Alain Charbit
- From the ‡Université Paris Descartes, Sorbonne Paris Cité, Bâtiment Leriche; §INSERM U1151-CNRS UMR 8253, Institut Necker-Enfants Malades, Team 11. Paris, France;
| |
Collapse
|