1
|
Gupta I, Badrzadeh F, Tsentalovich Y, Gaykalova DA. Connecting the dots: investigating the link between environmental, genetic, and epigenetic influences in metabolomic alterations in oral squamous cell carcinoma. J Exp Clin Cancer Res 2024; 43:239. [PMID: 39169426 PMCID: PMC11337877 DOI: 10.1186/s13046-024-03141-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 07/28/2024] [Indexed: 08/23/2024] Open
Abstract
Oral squamous cell carcinoma (OSCC) accounts for around 90% of all oral cancers and is the eighth most common cancer worldwide. Despite progress in managing OSCC, the overall prognosis remains poor, with a survival rate of around 50-60%, largely due to tumor size and recurrence. The challenges of late-stage diagnosis and limitations in current methods emphasize the urgent need for less invasive techniques to enable early detection and treatment, crucial for improving outcomes in this aggressive form of oral cancer. Research is currently aimed at unraveling tumor-specific metabolite profiles to identify candidate biomarkers as well as discover underlying pathways involved in the onset and progression of cancer that could be used as new targets for diagnostic and therapeutic purposes. Metabolomics is an advanced technological approach to identify metabolites in different sample types (biological fluids and tissues). Since OSCC promotes metabolic reprogramming influenced by a combination of genetic predisposition and environmental factors, including tobacco and alcohol consumption, and viral infections, the identification of distinct metabolites through screening may aid in the diagnosis of this condition. Moreover, studies have shown the use of metabolites during the catalysis of epigenetic modification, indicating a link between epigenetics and metabolism. In this review, we will focus on the link between environmental, genetic, and epigenetic influences in metabolomic alterations in OSCC. In addition, we will discuss therapeutic targets of tumor metabolism, which may prevent oral tumor growth, metastasis, and drug resistance.
Collapse
Affiliation(s)
- Ishita Gupta
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Otorhinolaryngology-Head and Neck Surgery, Marlene & Stewart Greenebaum Comprehensive Cancer Center, University of Maryland Medical Center, Baltimore, MD, USA
| | - Fariba Badrzadeh
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Otorhinolaryngology-Head and Neck Surgery, Marlene & Stewart Greenebaum Comprehensive Cancer Center, University of Maryland Medical Center, Baltimore, MD, USA
| | - Yuri Tsentalovich
- International tomography center CB RAS, Institutskaya str. 3a, Novosibirsk, 630090, Russia
| | - Daria A Gaykalova
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA.
- Department of Otorhinolaryngology-Head and Neck Surgery, Marlene & Stewart Greenebaum Comprehensive Cancer Center, University of Maryland Medical Center, Baltimore, MD, USA.
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA.
- Institute for Genome Sciences, 670 West Baltimore Street, Baltimore, MD, 21201, USA.
| |
Collapse
|
2
|
Lin GC, Tevini J, Mair L, Friedl HP, Fuchs D, Felder T, Gostner JM, Neuhaus W. Investigations Towards Tryptophan Uptake and Transport Across an In Vitro Model of the Oral Mucosa Epithelium. Int J Tryptophan Res 2024; 17:11786469241266312. [PMID: 39092002 PMCID: PMC11292681 DOI: 10.1177/11786469241266312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 06/03/2024] [Indexed: 08/04/2024] Open
Abstract
Tryptophan is an essential amino acid and plays an important role in several metabolic processes relevant for the human health. As the main metabolic pathway for tryptophan along the kynurenine axis is involved in inflammatory responses, changed metabolite levels can be used to monitor inflammatory diseases such as ulcerative colitis. As a progenitor of serotonin, altered tryptophan levels have been related to several neurogenerative diseases as well as depression or anxiety. While tryptophan concentrations are commonly evaluated in serum, a non-invasive detection approach using saliva might offer significant advantages, especially during long-term treatments of patients or elderly. In order to estimate whether active transport processes for tryptophan might contribute to a potential correlation between blood and saliva tryptophan concentrations, we investigated tryptophan's transport across an established oral mucosa in vitro model. Interestingly, treatment with tryptophan revealed a concentration dependent secretion of tryptophan and the presence of a saturable transporter while transport studies with deuterated tryptophan displayed increased permeability from the saliva to the blood compartment. Protein analysis demonstrated a distinct expression of L-type amino acid transporter 1 (LAT1), the major transporter for tryptophan, and exposure to inhibitors (2 -amino-2-norbornanecarboxylic acid (BCH), L-leucine) led to increased tryptophan levels on the saliva side. Additionally, exposure to tryptophan in equilibrium studies resulted in a regulation of LAT1 at the mRNA level. The data collected in this study suggest the participation of active transport mechanisms for tryptophan across the oral mucosa epithelium. Future studies should investigate the transport of tryptophan across salivary gland epithelia in order to enable a comprehensive understanding of tryptophan exchange at the blood-saliva barrier.
Collapse
Affiliation(s)
- Grace C. Lin
- AIT – Austrian Institute of Technology GmbH, Competence Unit Molecular Diagnostics, Center for Health and Bioresources, Vienna, Austria
| | - Julia Tevini
- Medical University of Innsbruck, Biocenter, Institute of Medical Biochemistry, Austria
- Paracelsus Medical University, Department of Laboratory Medicine, Salzburg, Austria
| | - Lisa Mair
- Medical University of Innsbruck, Biocenter, Institute of Medical Biochemistry, Austria
| | - Heinz-Peter Friedl
- AIT – Austrian Institute of Technology GmbH, Competence Unit Molecular Diagnostics, Center for Health and Bioresources, Vienna, Austria
| | - Dietmar Fuchs
- Medical University of Innsbruck, Biocenter, Institute of Biological Chemistry, Austria
| | - Thomas Felder
- Paracelsus Medical University, Department of Laboratory Medicine, Salzburg, Austria
| | - Johanna M. Gostner
- Medical University of Innsbruck, Biocenter, Institute of Medical Biochemistry, Austria
| | - Winfried Neuhaus
- AIT – Austrian Institute of Technology GmbH, Competence Unit Molecular Diagnostics, Center for Health and Bioresources, Vienna, Austria
- Department of Medicine, Faculty of Medicine and Dentistry, Danube Private University, Krems, Austria
| |
Collapse
|
3
|
Jakobsen S, Nielsen CU. Exploring Amino Acid Transporters as Therapeutic Targets for Cancer: An Examination of Inhibitor Structures, Selectivity Issues, and Discovery Approaches. Pharmaceutics 2024; 16:197. [PMID: 38399253 PMCID: PMC10893028 DOI: 10.3390/pharmaceutics16020197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/18/2024] [Accepted: 01/28/2024] [Indexed: 02/25/2024] Open
Abstract
Amino acid transporters are abundant amongst the solute carrier family and have an important role in facilitating the transfer of amino acids across cell membranes. Because of their impact on cell nutrient distribution, they also appear to have an important role in the growth and development of cancer. Naturally, this has made amino acid transporters a novel target of interest for the development of new anticancer drugs. Many attempts have been made to develop inhibitors of amino acid transporters to slow down cancer cell growth, and some have even reached clinical trials. The purpose of this review is to help organize the available information on the efforts to discover amino acid transporter inhibitors by focusing on the amino acid transporters ASCT2 (SLC1A5), LAT1 (SLC7A5), xCT (SLC7A11), SNAT1 (SLC38A1), SNAT2 (SLC38A2), and PAT1 (SLC36A1). We discuss the function of the transporters, their implication in cancer, their known inhibitors, issues regarding selective inhibitors, and the efforts and strategies of discovering inhibitors. The goal is to encourage researchers to continue the search and development within the field of cancer treatment research targeting amino acid transporters.
Collapse
Affiliation(s)
- Sebastian Jakobsen
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, DK-5230 Odense, Denmark
| | - Carsten Uhd Nielsen
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, DK-5230 Odense, Denmark
| |
Collapse
|
4
|
Misawa N, Honda S. Increased sodium fluorescein transport by corticosteroids is inhibited by a LAT-1 specific inhibitor in retinal pigment epithelial cells in vitro. Sci Rep 2023; 13:22981. [PMID: 38151501 PMCID: PMC10752866 DOI: 10.1038/s41598-023-50196-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 12/16/2023] [Indexed: 12/29/2023] Open
Abstract
To investigate whether aldosterone (ALD) and hydrocortisone (HC) change the gene expression of SLC7A5, which encodes the large neutral amino acid transporter small subunit 1 (LAT1), and the transport activity of LAT1 in the retinal pigment epithelium (RPE) in vitro. ARPE-19 cells were grown to confluence. After withdrawing the serum, ALD or HC was added with several doses and incubated, and SLC7A5 gene expression was measured. The influx and efflux transport of sodium fluorescein (Na-F) were evaluated using the Transwell culture system. SLC7A5 gene expression was upregulated by ALD and downregulated by HC in a dose-dependent manner. Both ALD and HC significantly increased the influx and efflux Na-F transport of RPE cells at a dose that did not change the expression of SLC7A5. JPH203, a specific inhibitor of LAT1, significantly reduced accelerated Na-F transport. Both ALD and HC increased the gene expression of zonula occludin-1 (ZO-1) although they did not change the immunoreactivity of ZO-1 in RPE cells. LAT1 may play an important role in increasing Na-F transport associated with ALD and HC administration. A specific LAT1 inhibitor may effectively regulate the increased material transport of RPE induced by ALD and HC.
Collapse
Affiliation(s)
- Norihiko Misawa
- Department of Ophthalmology and Visual Sciences, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan
| | - Shigeru Honda
- Department of Ophthalmology and Visual Sciences, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan.
| |
Collapse
|
5
|
Hou Y, Zhang H, Zhu Y, He X, Li W, Su L, Liu M, Chen X, Shen F, Chen X, Jiang W, Zou C, Xu Z. Targeting upregulation of the immunosuppressive activity of MDSCs with indirubin as a novel strategy to alleviate psoriasis. Int Immunopharmacol 2023; 123:110710. [PMID: 37531829 DOI: 10.1016/j.intimp.2023.110710] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 07/19/2023] [Accepted: 07/23/2023] [Indexed: 08/04/2023]
Abstract
BACKGROUND Psoriasis is a chronic and incurable skin disorder that causes inflammation. There is an urgent clinical need for new treatments. We identified the natural compound indirubin as a potential potent agent for the treatment of psoriasis, but it's therapeutic effect and underlying mechanisms were not well understood. METHODS Peripheral blood and skin tissues from psoriasis patients and healthy individuals were collected. Bioinformatics analysis was performed to investigate LAT1 expression and associated signal pathways in psoriasis skin lesions. A mouse model of psoriasis was established. Indirubin was administered separately or in combination with MDSCs depletion or adoptively transferred MDSCs. JPH203, rapamycin, siRNA, and NV5138 were further used to investigate the potential mechanism by which indirubin regulates MDSCs. RESULTS Psoriasis patients had increased numbers of MDSCs in their blood and skin lesions, with high expression of Lat1. The upregulation of LAT1 expression and the arginine synthesis pathway was observed in psoriasis skin lesions. The number of MDSCs was increased, while their inhibitory effect on psoriatic T cells was decreased. Indirubin decreased Lat1 expression on the surface of MDSCs, inhibited mTOR pathway activation, upregulated Arg1 expression in MDSCs, and enhanced the immunosuppressive activity of MDSCs while inhibiting CD4+CCR6+ T cells. CONCLUSION This study demonstrates indirubin's pharmacological and therapeutic effects, providing a basis for future clinical application in treating psoriasis.
Collapse
Affiliation(s)
- Yifei Hou
- Shanghai Key Laboratory of Health Identification and Assessment, School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Huimin Zhang
- Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200021, China
| | - Yangzhuangzhuang Zhu
- Shanghai Key Laboratory of Health Identification and Assessment, School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xufeng He
- Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200021, China
| | - Wen Li
- Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200021, China
| | - Lin Su
- Shanghai Key Laboratory of Health Identification and Assessment, School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Mingxi Liu
- Shanghai Key Laboratory of Health Identification and Assessment, School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xi Chen
- Department of Traditional Chinese Medicine Dermatology, Shanghai Skin Disease Hospital, Shanghai 200443, China
| | - Fang Shen
- Department of Traditional Chinese Medicine Dermatology, Shanghai Skin Disease Hospital, Shanghai 200443, China
| | - Xiao Chen
- Shanghai Key Laboratory of Health Identification and Assessment, School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Wencheng Jiang
- Department of Traditional Chinese Medicine Dermatology, Shanghai Skin Disease Hospital, Shanghai 200443, China.
| | - Chunpu Zou
- Shanghai Key Laboratory of Health Identification and Assessment, School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Zihang Xu
- Shanghai Key Laboratory of Health Identification and Assessment, School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
6
|
Tahara T, Takatani S, Tsuji M, Shibata N, Hosaka N, Inoue M, Ohno M, Ozaki D, Mawatari A, Watanabe Y, Doi H, Onoe H. Characteristic Evaluation of a 11C-Labeled Leucine Analog, l-α-[5- 11C]methylleucine, as a Tracer for Brain Tumor Imaging by Positron Emission Tomography. Mol Pharm 2023; 20:1842-1849. [PMID: 36802622 DOI: 10.1021/acs.molpharmaceut.2c01069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Abstract
Amino acid transporters are upregulated in many cancer cells, and system L amino acid transporters (LAT1-4), in particular, LAT1, which preferentially transports large, neutral, and branched side-chain amino acids, are considered a primary target for cancer positron emission tomography (PET) tracer development. Recently, we developed a 11C-labeled leucine analog, l-α-[5-11C]methylleucine ([5-11C]MeLeu), via a continuous two-step reaction of Pd0-mediated 11C-methylation and microfluidic hydrogenation. In this study, we evaluated the characteristics of [5-11C]MeLeu and also compared the sensitivity to brain tumors and inflammation with l-[11C]methionine ([11C]Met) to determine its potential for brain tumor imaging. Competitive inhibition experiments, protein incorporation, and cytotoxicity experiments of [5-11C]MeLeu were performed in vitro. Further, metabolic analyses of [5-11C]MeLeu were performed using a thin-layer chromatogram. The accumulation of [5-11C]MeLeu in tumor and inflamed regions of the brain was compared with [11C]Met and 11C-labeled (S)-ketoprofen methyl ester by PET imaging, respectively. Transporter assay with various inhibitors revealed that [5-11C]MeLeu is mainly transported via system L amino acid transporters, especially LAT1, into A431 cells. The protein incorporation assay and metabolic assay in vivo demonstrated that [5-11C]MeLeu was neither used for protein synthesis nor metabolized. These results indicate that MeLeu is very stable in vivo. Furthermore, the treatment of A431 cells with various concentrations of MeLeu did not change their viability, even at high concentrations (∼10 mM). In brain tumors, the tumor-to-normal ratio of [5-11C]MeLeu was more elevated than that of [11C]Met. However, the accumulation levels of [5-11C]MeLeu were lower than those of [11C]Met (the standardized uptake value (SUV) of [5-11C]MeLeu and [11C]Met was 0.48 ± 0.08 and 0.63 ± 0.06, respectively). In brain inflammation, no significant accumulation of [5-11C]MeLeu was observed at the inflamed brain area. These data suggested that [5-11C]MeLeu was identified as a stable and safe agent for PET tracers and could help detect brain tumors, which overexpress the LAT1 transporter.
Collapse
Affiliation(s)
- Tsuyoshi Tahara
- RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan.,RIKEN Center for Life Science Technologies, 6-7-3 Minatojima Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan.,Department of In Vivo Imaging, Tokushima University, 3-18-15 Kuramoto-Cho, Tokushima, Tokushima 770-8503, Japan
| | - Shuhei Takatani
- RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan.,RIKEN Center for Life Science Technologies, 6-7-3 Minatojima Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Mieko Tsuji
- RIKEN Center for Life Science Technologies, 6-7-3 Minatojima Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Nina Shibata
- RIKEN Center for Life Science Technologies, 6-7-3 Minatojima Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Nami Hosaka
- RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan.,RIKEN Center for Life Science Technologies, 6-7-3 Minatojima Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Michiko Inoue
- RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan.,RIKEN Center for Life Science Technologies, 6-7-3 Minatojima Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Masahiro Ohno
- RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan.,RIKEN Center for Life Science Technologies, 6-7-3 Minatojima Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Daiki Ozaki
- RIKEN Center for Life Science Technologies, 6-7-3 Minatojima Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Aya Mawatari
- RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan.,RIKEN Center for Life Science Technologies, 6-7-3 Minatojima Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Yasuyoshi Watanabe
- RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan.,RIKEN Center for Life Science Technologies, 6-7-3 Minatojima Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Hisashi Doi
- RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan.,RIKEN Center for Life Science Technologies, 6-7-3 Minatojima Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Hirotaka Onoe
- RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan.,RIKEN Center for Life Science Technologies, 6-7-3 Minatojima Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan.,Human Brain Research Center, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawahara-Cho, Sakyo-ku, Kyoto 606-8507, Japan
| |
Collapse
|
7
|
Kantipudi S, Harder D, Fotiadis D. Characterization of substrates and inhibitors of the human heterodimeric transporter 4F2hc-LAT1 using purified protein and the scintillation proximity radioligand binding assay. Front Physiol 2023; 14:1148055. [PMID: 36895635 PMCID: PMC9989278 DOI: 10.3389/fphys.2023.1148055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 02/06/2023] [Indexed: 02/25/2023] Open
Abstract
Amino acids have diverse and essential roles in many cellular functions such as in protein synthesis, metabolism and as precursors of different hormones. Translocation of amino acids and derivatives thereof across biological membranes is mediated by amino acid transporters. 4F2hc-LAT1 is a heterodimeric amino acid transporter that is composed of two subunits belonging to the SLC3 (4F2hc) and SLC7 (LAT1) solute carrier families. The ancillary protein 4F2hc is responsible for the correct trafficking and regulation of the transporter LAT1. Preclinical studies have identified 4F2hc-LAT1 as a valid anticancer target due to its importance in tumor progression. The scintillation proximity assay (SPA) is a valuable radioligand binding assay that allows the identification and characterization of ligands of membrane proteins. Here, we present a SPA ligand binding study using purified recombinant human 4F2hc-LAT1 protein and the radioligand [3H]L-leucine as tracer. Binding affinities of different 4F2hc-LAT1 substrates and inhibitors determined by SPA are comparable with previously reported K m and IC 50 values from 4F2hc-LAT1 cell-based uptake assays. In summary, the SPA is a valuable method for the identification and characterization of ligands of membrane transporters including inhibitors. In contrast to cell-based assays, where the potential interference with other proteins such as endogenous transporters persists, the SPA uses purified protein making target engagement and characterization of ligands highly reliable.
Collapse
Affiliation(s)
- Satish Kantipudi
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
| | - Daniel Harder
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
| | - Dimitrios Fotiadis
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
| |
Collapse
|
8
|
Hutchinson K, Silva DB, Bohlke J, Clausen C, Thomas AA, Bonomi M, Schlessinger A. Describing inhibitor specificity for the amino acid transporter LAT1 from metainference simulations. Biophys J 2022; 121:4476-4491. [PMID: 36369754 PMCID: PMC9748366 DOI: 10.1016/j.bpj.2022.11.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 09/09/2022] [Accepted: 10/31/2022] [Indexed: 11/13/2022] Open
Abstract
The human L-type amino acid transporter 1 (LAT1; SLC7A5) is a membrane transporter of amino acids, thyroid hormones, and drugs such as the Parkinson's disease drug levodopa (L-Dopa). LAT1 is found in the blood-brain barrier, testis, bone marrow, and placenta, and its dysregulation has been associated with various neurological diseases, such as autism and epilepsy, as well as cancer. In this study, we combine metainference molecular dynamics simulations, molecular docking, and experimental testing, to characterize LAT1-inhibitor interactions. We first conducted a series of molecular docking experiments to identify the most relevant interactions between LAT1's substrate-binding site and ligands, including both inhibitors and substrates. We then performed metainference molecular dynamics simulations using cryoelectron microscopy structures in different conformations of LAT1 with the electron density map as a spatial restraint, to explore the inherent heterogeneity in the structures. We analyzed the LAT1 substrate-binding site to map important LAT1-ligand interactions as well as newly described druggable pockets. Finally, this analysis guided the discovery of previously unknown LAT1 ligands using virtual screening and cellular uptake experiments. Our results improve our understanding of LAT1-inhibitor recognition, providing a framework for rational design of future lead compounds targeting this key drug target.
Collapse
Affiliation(s)
- Keino Hutchinson
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Dina Buitrago Silva
- Department of Bioengineering and Therapeutic Sciences University of California, San Francisco, San Francisco, California
| | - Joshua Bohlke
- Department of Chemistry, University of Nebraska at Kearney, Kearney, Nebraska
| | - Chase Clausen
- Department of Chemistry, University of Nebraska at Kearney, Kearney, Nebraska
| | - Allen A Thomas
- Department of Chemistry, University of Nebraska at Kearney, Kearney, Nebraska
| | - Massimiliano Bonomi
- Department of Structural Biology and Chemistry, Institut Pasteur, Université Paris Cité, CNRS UMR 3528, Paris, France.
| | - Avner Schlessinger
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York.
| |
Collapse
|
9
|
Okanishi H, Ohgaki R, Xu M, Endou H, Kanai Y. Phosphoproteomics revealed cellular signals immediately responding to disruption of cancer amino acid homeostasis induced by inhibition of l-type amino acid transporter 1. Cancer Metab 2022; 10:18. [PMID: 36357940 PMCID: PMC9650822 DOI: 10.1186/s40170-022-00295-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 10/26/2022] [Indexed: 11/12/2022] Open
Abstract
Background Cancer-upregulated l-type amino acid transporter 1 (LAT1; SLC7A5) supplies essential amino acids to cancer cells. LAT1 substrates are not only needed for cancer rapid growth, but involved in cellular signaling. LAT1 has been proposed as a potential target for cancer treatment—its inhibitor, JPH203, is currently in clinical trials and targets biliary tract cancer (BTC). Here, we revealed to what extent LAT1 inhibitor affects intracellular amino acid content and what kind of cellular signals are directly triggered by LAT1 inhibition. Methods Liquid chromatography assay combined with o-phthalaldehyde- and 9-fluorenyl-methylchloroformate-based derivatization revealed changes in intracellular amino acid levels induced by LAT1 inhibition with JPH203 treatment in three BTC cell lines. Tandem mass tag-based quantitative phosphoproteomics characterized the effect of JPH203 treatment on BTC cells, and suggested key regulators in LAT1-inhibited cells. We further studied one of the key regulators, CK2 protein kinase, by using Western blot, enzymatic activity assay, and co-immunoprecipitation. We evaluated anticancer effects of combination of JPH203 with CK2 inhibitor using cell growth and would healing assay. Results JPH203 treatment decreased intracellular levels of LAT1 substrates including essential amino acids of three BTC cell lines, immediately and drastically. We also found levels of some of these amino acids were partially recovered after longer-time treatment. Therefore, we performed phosphoproteomics with short-time JPH203 treatment prior to the cellular compensatory response, and revealed hundreds of differentially phosphorylated sites. Commonly downregulated phosphorylation sites were found on proteins involved in the cell cycle and RNA splicing. Our phosphoproteomics also suggested key regulators immediately responding to LAT1 inhibition. Focusing on one of these regulators, protein kinase CK2, we revealed LAT1 inhibition decreased phosphorylation of CK2 substrate without changing CK2 enzymatic activity. Furthermore, LAT1 inhibition abolished interaction between CK2 and its regulatory protein NOLC1, which suggests regulatory mechanism of CK2 substrate protein specificity controlled by LAT1 inhibition. Moreover, we revealed that the combination of JPH203 with CK2 inhibitor resulted in the enhanced inhibition of proliferation and migration of BTC cells. Conclusion This study provides new perspectives on LAT1-dependent cellular processes and a rationale for therapeutics targeting reprogrammed cancer metabolism. Supplementary Information The online version contains supplementary material available at 10.1186/s40170-022-00295-8.
Collapse
|
10
|
Cappoli N, Jenkinson MD, Russo CD, Dickens D. LAT1, a novel pharmacological target for the treatment of glioblastoma. Biochem Pharmacol 2022; 201:115103. [PMID: 35618000 DOI: 10.1016/j.bcp.2022.115103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 05/18/2022] [Accepted: 05/18/2022] [Indexed: 11/02/2022]
Abstract
The L-Type Amino Acid transporter, LAT1 (SLC7A5), has a crucial role in mediating amino acid uptake into the cells, thus modulating cell growth and proliferation as well as other intracellular functions. Different studies have reported a central role of LAT1 in glioblastoma development and progression, suggesting that the modulation of its activity could be a novel therapeutic strategy. LAT1 also has an important role in the peripheral immune system, by regulating the activation status of several immune cells through modulation of the mechanistic target of rapamycin kinase. In glioblastoma (GBM), the blood-brain barrier is disrupted, which allows the recruitment of peripheral immune cells to the tumour site. These cells, together with resident microglia, contribute to cancer growth and progression. Currently, little is known about the function of LAT1 in the reprogramming of the immune component of the tumour microenvironment in the context of GBM. In this article, we review the available data on the role of LAT1 in the regulation of GBM biology, including its potential role in the tumour microenvironment, particularly in infiltrating-peripheral immune cells and resident microglial cells. In addition, we review the available data on the main pharmacological inhibitors of LAT1, aiming to evaluate their possible role as novel therapeutics for GBM.
Collapse
Affiliation(s)
- Natalia Cappoli
- Department of Healthcare Surveillance and Bioethics, Section of Pharmacology, Università Cattolica del Sacro Cuore-Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Michael D Jenkinson
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology (ISMIB), University of Liverpool, Liverpool, United Kingdom; Department of Neurosurgery, The Walton Centre NHS Foundation Trust, Liverpool, United Kingdom
| | - Cinzia Dello Russo
- Department of Healthcare Surveillance and Bioethics, Section of Pharmacology, Università Cattolica del Sacro Cuore-Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy; Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology (ISMIB), University of Liverpool, Liverpool, United Kingdom.
| | - David Dickens
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology (ISMIB), University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
11
|
Wang Y, Qin L, Chen W, Chen Q, Sun J, Wang G. Novel strategies to improve tumour therapy by targeting the proteins MCT1, MCT4 and LAT1. Eur J Med Chem 2021; 226:113806. [PMID: 34517305 DOI: 10.1016/j.ejmech.2021.113806] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 08/22/2021] [Accepted: 08/24/2021] [Indexed: 02/08/2023]
Abstract
Poor selectivity, potential systemic toxicity and drug resistance are the main challenges associated with chemotherapeutic drugs. MCT1 and MCT4 and LAT1 play vital roles in tumour metabolism and growth by taking up nutrients and are thus potential targets for tumour therapy. An increasing number of studies have shown the feasibility of including these transporters as components of tumour-targeting therapy. Here, we summarize the recent progress in MCT1-, MCT4-and LAT1-based therapeutic strategies. First, protein structures, expression, relationships with cancer, and substrate characteristics are introduced. Then, different drug targeting and delivery strategies using these proteins have been reviewed, including designing protein inhibitors, prodrugs and nanoparticles. Finally, a dual targeted strategy is discussed because these proteins exert a synergistic effect on tumour proliferation. This article concentrates on tumour treatments targeting MCT1, MCT4 and LAT1 and delivery techniques for improving the antitumour effect. These innovative tactics represent current state-of-the-art developments in transporter-based antitumour drugs.
Collapse
Affiliation(s)
- Yang Wang
- Personnel Department, Guang Xi University of Chinese Medicine, Nanning, 530200, PR China
| | - Liuxin Qin
- School of Pharmacy, Guang Xi University of Chinese Medicine, Nanning, 530200, PR China
| | - Weiwei Chen
- School of Pharmacy, Guang Xi University of Chinese Medicine, Nanning, 530200, PR China
| | - Qing Chen
- Zhuang Yao Medicine Center of Engineering and Technology, Guang Xi University of Chinese Medicine, Nanning, 530200, PR China
| | - Jin Sun
- Key Laboratory of Structure-Based Drug Design and Discovery, Shenyang Pharmaceutical University, Ministry of Education, China
| | - Gang Wang
- Zhuang Yao Medicine Center of Engineering and Technology, Guang Xi University of Chinese Medicine, Nanning, 530200, PR China.
| |
Collapse
|
12
|
Bo T, Kobayashi S, Inanami O, Fujii J, Nakajima O, Ito T, Yasui H. LAT1 inhibitor JPH203 sensitizes cancer cells to radiation by enhancing radiation-induced cellular senescence. Transl Oncol 2021; 14:101212. [PMID: 34461558 PMCID: PMC8405945 DOI: 10.1016/j.tranon.2021.101212] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/26/2021] [Accepted: 08/24/2021] [Indexed: 11/23/2022] Open
Abstract
X-irradiation increased cellular neutral amino acid uptake via LAT1. JPH203 inhibited the radiation-induced increase in neutral amino acid uptake. JPH203 significantly sensitized cancer cells to radiation. JPH203 downregulated mTOR activity after irradiation. JPH203 enhanced cellular senescence after irradiation.
L-type amino acid transporter 1 (LAT1) is important for transporting neutral amino acids into cells. LAT1 expression is correlated with cancer malignancy, suggesting that LAT1 is a promising target for cancer therapy. JPH203, a potential novel drug targeting LAT1, has been shown to suppress tumor growth in various cancer cell lines. However, a combination study of JPH203 and radiation therapy has not been reported. Here, we examined the effects of JPH203 on radiosensitivity after irradiation in A549 and MIA Paca-2 cells. We showed that X-irradiation increased cellular neutral amino acid uptake via LAT1 in both cell lines. JPH203 inhibited the radiation-induced increase in neutral amino acid uptake. We demonstrated that JPH203, at minimally toxic concentrations, significantly sensitized cancer cells to radiation. JPH203 significantly downregulated mTOR activity and enhanced cellular senescence post-irradiation without reducing ATP and GSH levels. These results indicate that LAT1 inhibition by JPH203 sensitizes cancer cells to radiation by enhancing cellular senescence via mTOR downregulation. Thus, JPH203 may be a potent anti-cancer drug in combination with radiation therapy.
Collapse
Affiliation(s)
- Tomoki Bo
- Laboratory Animal Center, Institute for Promotion of Medical Science Research, Yamagata University Faculty of Medicine, Japan.
| | - Sho Kobayashi
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Yamagata University, Japan
| | - Osamu Inanami
- Laboratory of Radiation Biology, Department of Applied Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Junichi Fujii
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Yamagata University, Japan
| | - Osamu Nakajima
- Research Center for Molecular Genetics, Institute for Promotion of Medical Science Research, Yamagata University Faculty of Medicine, Japan
| | - Tsunekata Ito
- Laboratory Animal Center, Institute for Promotion of Medical Science Research, Yamagata University Faculty of Medicine, Japan
| | - Hironobu Yasui
- Laboratory of Radiation Biology, Department of Applied Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan.
| |
Collapse
|
13
|
Targeting Amino Acid Metabolic Reprogramming via L-Type Amino Acid Transporter 1 (LAT1) for Endocrine-Resistant Breast Cancer. Cancers (Basel) 2021; 13:cancers13174375. [PMID: 34503187 PMCID: PMC8431153 DOI: 10.3390/cancers13174375] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 08/23/2021] [Accepted: 08/25/2021] [Indexed: 12/22/2022] Open
Abstract
The PI3K/Akt/mTOR pathway has been well known to interact with the estrogen receptor (ER)-pathway and to be also frequently upregulated in aromatase inhibitor (AI)-resistant breast cancer patients. Intracellular levels of free amino acids, especially leucine, regulate the mammalian target of rapamycin complex 1 (mTORC1) activation. L-type amino acid transporters such as LAT1 and LAT3 are associated with the uptake of essential amino acids. LAT1 expression could mediate leucine uptake, mTORC1 signaling, and cell proliferation. Therefore, in this study, we explored amino acid metabolism, including LAT1, in breast cancer and clarified the potential roles of LAT1 in the development of therapeutic resistance and the eventual clinical outcome of the patients. We evaluated LAT1 and LAT3 expression before and after neoadjuvant hormone therapy (NAH) and examined LAT1 function and expression in estrogen deprivation-resistant (EDR) breast carcinoma cell lines. Tumors tended to be in advanced stages in the cases whose LAT1 expression was high. LAT1 expression in the EDR cell lines was upregulated. JPH203, a selective LAT1 inhibitor, demonstrated inhibitory effects on cell proliferation in EDR cells. Hormone therapy changed the tumor microenvironment and resulted in metabolic reprogramming through inducing LAT1 expression. LAT1 expression then mediated leucine uptake, enhanced mTORC1 signaling, and eventually resulted in AI resistance. Therefore, LAT1 could be the potential therapeutic target in AI-resistant breast cancer patients.
Collapse
|
14
|
Zhang C, Xu J, Xue S, Ye J. Prognostic Value of L-Type Amino Acid Transporter 1 (LAT1) in Various Cancers: A Meta-Analysis. Mol Diagn Ther 2021; 24:523-536. [PMID: 32410110 DOI: 10.1007/s40291-020-00470-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
BACKGROUND AND OBJECTIVE The L-type amino acid transporter 1 (LAT1, SLC7A5) is overexpressed in various types of cancer and has been thought to assist cancer progression through its uptake of neutral amino acids. However, the prognostic role of LAT1 in human cancers remains uncharacterized. Therefore, we conducted this meta-analysis to determine the prognostic significance of LAT1 in various cancers. METHODS We systematically searched the PubMed, Web of Science, EMBASE, Chinese National Knowledge Infrastructure, and WanFang databases to collect relevant cohort studies investigating the prognostic value of LAT1 expression in patients with cancer. Hazard ratios (HRs) with corresponding 95% confidence intervals (CIs) were pooled to clarify the association between the LAT1 expression and the survival of patients with cancer. Odds ratios (ORs) with 95% CIs were calculated to appraise the correlation between LAT1 and the clinicopathological characteristics in patients with cancer. RESULTS A total of 32 eligible articles, including 34 cohorts and 6410 patients, were enrolled in this meta-analysis. Our results demonstrated that high LAT1 expression was significantly associated with poor overall survival (HR = 1.66, 95% CI 1.41-1.96, P < 0.001), cancer-specific survival (HR = 1.64, 95% CI 1.31-2.05, P < 0.001), disease-free survival (HR = 1.55, 95% CI 1.31-1.83, P < 0.001), and progression-free survival (HR = 1.18, 95% CI 1.02-1.37, P = 0.026) in patients with cancer. In addition, we found that the elevated expression level of LAT1 was significantly related to certain phenotypes of tumor aggressiveness, such as tumor size, clinical stage, T stage, lymphatic invasion, vascular invasion, tumor differentiation, Ki-67, CD34, CD98, p53, and system ASC amino acid transporter-2. CONCLUSIONS Elevated expression of LAT1 is associated with poor prognosis in human cancers and may serve as a potential prognostic marker and therapeutic target for patients with malignancies.
Collapse
Affiliation(s)
- Chuanmeng Zhang
- The Center for Translational Medicine, Taizhou People's Hospital, Affiliated 5 to Nantong University, Taizhou, 225300, Jiangsu Province, China
| | - Jie Xu
- The Center for Translational Medicine, Taizhou People's Hospital, Affiliated 5 to Nantong University, Taizhou, 225300, Jiangsu Province, China
| | - Shanshan Xue
- Department of Clinical Laboratory, Taizhou People's Hospital, Affiliated 5 to Nantong University, Taizhou, 225300, Jiangsu Province, China
| | - Jun Ye
- The Center for Translational Medicine, Taizhou People's Hospital, Affiliated 5 to Nantong University, Taizhou, 225300, Jiangsu Province, China.
| |
Collapse
|
15
|
Loilome W, Dokduang H, Suksawat M, Padthaisong S. Therapeutic challenges at the preclinical level for targeted drug development for Opisthorchis viverrini-associated cholangiocarcinoma. Expert Opin Investig Drugs 2021; 30:985-1006. [PMID: 34292795 DOI: 10.1080/13543784.2021.1955102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Cholangiocarcinoma (CCA) is a malignant tumor of bile duct epithelium with the highest incidence found in Thailand. Some patients are considered suitable for adjuvant therapy and surgical resection is currently the curative treatment for CCA patients. Tumor recurrence is still a hurdle after treatment; hence, finding novel therapeutic strategies to combat CCA is necessary for improving outcome for patients. AREAS COVERED We discuss targeted therapies and other novel treatment approaches which include protein kinase inhibitors, natural products, amino acid transporter-based inhibitors, immunotherapy, and drug repurposing. We also examine the challenges of tumor heterogeneity, cancer stem cells (CSCs), the tumor microenvironment, exosomes, multiomics studies, and the potential of precision medicine. EXPERT OPINION Because CCA is difficult to diagnose at the early stage, the traditional treatment approaches are not effective for many patients and most tumors recur. Consequently, researchers are exploring multi-aspect molecular carcinogenesis to uncover molecular targets for further development of novel targeted drugs.
Collapse
Affiliation(s)
- Watcharin Loilome
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen Thailand.,Cholangiocarcinoma Screening and Care Program (CASCAP), Khon Kaen University, Khon Kaen, Thailand.,Cholangiocarcinoma Research Institute, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Hasaya Dokduang
- Cholangiocarcinoma Screening and Care Program (CASCAP), Khon Kaen University, Khon Kaen, Thailand.,Cholangiocarcinoma Research Institute, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Manida Suksawat
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen Thailand.,Cholangiocarcinoma Screening and Care Program (CASCAP), Khon Kaen University, Khon Kaen, Thailand.,Cholangiocarcinoma Research Institute, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Sureerat Padthaisong
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen Thailand.,Cholangiocarcinoma Screening and Care Program (CASCAP), Khon Kaen University, Khon Kaen, Thailand.,Cholangiocarcinoma Research Institute, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| |
Collapse
|
16
|
Rii J, Sakamoto S, Sugiura M, Kanesaka M, Fujimoto A, Yamada Y, Maimaiti M, Ando K, Wakai K, Xu M, Imamura Y, Shindo N, Hirota T, Kaneda A, Kanai Y, Ikehara Y, Anzai N, Ichikawa T. Functional analysis of LAT3 in prostate cancer: Its downstream target and relationship with androgen receptor. Cancer Sci 2021; 112:3871-3883. [PMID: 34050700 PMCID: PMC8409400 DOI: 10.1111/cas.14991] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 05/05/2021] [Accepted: 05/18/2021] [Indexed: 12/11/2022] Open
Abstract
L‐type amino acid transporter 3 (LAT3, SLC43A1) is abundantly expressed in prostate cancer (PC) and is thought to play an essential role in PC progression through the cellular uptake of essential amino acids. Here, we analyzed the expression, function, and downstream target of LAT3 in PC. LAT3 was highly expressed in PC cells expressing androgen receptor (AR), and its expression was increased by dihydrotestosterone treatment and decreased by bicalutamide treatment. In chromatin immunoprecipitation sequencing of AR, binding of AR to the SLC43A1 region was increased by dihydrotestosterone stimulation. Knockdown of LAT3 inhibited cell proliferation, migration, and invasion, and the phosphorylation of p70S6K and 4EBP‐1. Separase (ESPL1) was identified as a downstream target of LAT3 by RNA sequencing analysis. In addition, immunostaining of prostatectomy specimens was performed. In the multivariate analysis, high expression of LAT3 was an independent prognostic factor for recurrence‐free survival (hazard ratio: 3.24; P = .0018). High LAT3 expression was correlated with the pathological T stage and a high International Society of Urological Pathology grade. In summary, our results suggest that LAT3 plays an important role in the progression of PC.
Collapse
Affiliation(s)
- Junryo Rii
- Department of Urology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Shinichi Sakamoto
- Department of Urology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Masahiro Sugiura
- Department of Urology, Chiba University Graduate School of Medicine, Chiba, Japan.,Department of Molecular Oncology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Manato Kanesaka
- Department of Urology, Chiba University Graduate School of Medicine, Chiba, Japan.,Department of Molecular Oncology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Ayumu Fujimoto
- Department of Urology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Yasutaka Yamada
- Department of Urology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Maihulan Maimaiti
- Department of Tumor Pathology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Keisuke Ando
- Department of Urology, Chiba University Graduate School of Medicine, Chiba, Japan.,Department of Pharmacology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Ken Wakai
- Department of Urology, Chiba University Graduate School of Medicine, Chiba, Japan.,Department of Tumor Pathology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Minhui Xu
- Bio-system Pharmacology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yusuke Imamura
- Department of Urology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Norihisa Shindo
- Division of Experimental Pathology, Cancer Institute of the Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Toru Hirota
- Division of Experimental Pathology, Cancer Institute of the Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Atsushi Kaneda
- Department of Molecular Oncology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Yoshikatsu Kanai
- Bio-system Pharmacology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yuzuru Ikehara
- Department of Tumor Pathology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Naohiko Anzai
- Department of Pharmacology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Tomohiko Ichikawa
- Department of Urology, Chiba University Graduate School of Medicine, Chiba, Japan
| |
Collapse
|
17
|
Highly Specific L-Type Amino Acid Transporter 1 Inhibition by JPH203 as a Potential Pan-Cancer Treatment. Processes (Basel) 2021. [DOI: 10.3390/pr9071170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Accelerated cancer cell growth requires a massive intake of amino acids. Overexpression of L-type (large) amino acid transporter 1 (LAT1) on the cancer cell membrane facilitates such a demand, which is limited in normal organs. Therefore, LAT1 overexpression is ideal as a molecular cancer therapeutic target. JPH203, a LAT1-selective non-transportable blocker, had demonstrated LAT1 inhibition in <10 µM IC50 values and effectively suppressed cancer cell growth in studies involving several types of cancer cell lines and tumor xenograft models. A limited phase I clinical trial was performed on five different solid tumors and showed that JPH203 is well-tolerated and has a promising activity for the treatment of bile duct cancer. This review details the development and prospect of JPH203 as a LAT1-targeting cancer therapy.
Collapse
|
18
|
Saito Y, Soga T. Amino acid transporters as emerging therapeutic targets in cancer. Cancer Sci 2021; 112:2958-2965. [PMID: 34091991 PMCID: PMC8353895 DOI: 10.1111/cas.15006] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/13/2021] [Accepted: 05/31/2021] [Indexed: 01/17/2023] Open
Abstract
Amino acids are indispensable nutrients for both normal and cancer cells. Cancer cells are unable to synthesize essential amino acids as well as some non‐essential amino acids adequately to support rapid proliferation, and must take up amino acids from the surroundings. To meet the increased demand for the amino acid needed for proliferation, high levels of amino acid transporters are expressed on the surface of cancer cells. Cancer cells utilize amino acids to synthesize proteins and nucleotides, as well as to obtain energy. In addition, amino acids are known to play pathological roles in cancer cells. Interestingly, breast cancer cells limit the use of amino acids for cell proliferation based on amino acid availability, which depends on estrogen receptor status. Here, we present a summarized literature review of novel amino acid functions in cancer cells. This review organizes the available knowledge on 2 amino acid transporters, SLC7A5 and SLC7A11, which are considered essential for breast cancer cell growth in a cell‐dependent manner. In particular, we propose the glutamine recycling model to clarify the mechanism underlying aberrant SLC7A5 activation. Finally, we overview the pathological significances of SLC7A5 and SLC7A11 in cancer tissues.
Collapse
Affiliation(s)
- Yasuhiro Saito
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Japan
| | - Tomoyoshi Soga
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Japan
| |
Collapse
|
19
|
Markowicz-Piasecka M, Huttunen J, Montaser A, Huttunen KM. Hemocompatible LAT1-inhibitor can induce apoptosis in cancer cells without affecting brain amino acid homeostasis. Apoptosis 2021; 25:426-440. [PMID: 32405891 PMCID: PMC7244471 DOI: 10.1007/s10495-020-01603-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Increased amounts of amino acids are essential for cancer cells to support their sustained growth and survival. Therefore, inhibitors of amino acid transporters, such as l-type amino acid transporter 1 (LAT1) have been developed. In this study, a previously reported LAT1-inhibitor (KMH-233) was studied for its hemocompatibility and toxicity towards human umbilical vein endothelial cells (HUVEC) and human aortic smooth muscle cells (AoSMCs). Furthermore, the cytotoxic effects against human breast adenocarcinoma cells (MCF-7) and its ability to affect mammalian (or mechanistic) target of rapamycin (mTOR) and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling were evaluated. Moreover, the effects of this inhibitor to modulate LAT1 function on the cell surface and the brain amino acid homeostasis were evaluated after intraperitoneal (i.p.) administration of LAT1-inhibitor (23 µmol/kg) in mice. The results showed that LAT1-inhibitor (KMH-233) is hemocompatible at concentrations below 25 µM and it does not affect coagulation in plasma. However, it can reduce the total protein amount of mTOR and NF-κB, resulting in increased apoptosis in LAT1-expressing cancer cells. Most importantly, the inhibitor did not affect mouse brain levels of l-Leu, l-Tyr or l-Trp or modulate the function of LAT1 on the MCF-7 cell surface. Therefore, this inhibitor can be considered as a safe but effective anti-cancer agent. However, due to the compensative mechanism of cancer cells for their increased amino acid demand, this compound is most effective inducing apoptosis when used in combinations with other chemotherapeutics, such as protease inhibitor, bestatin, as demonstrated in this study.
Collapse
Affiliation(s)
- Magdalena Markowicz-Piasecka
- Laboratory of Bioanalysis, Department of Pharmaceutical Chemistry, Drug Analysis and Radiopharmacy, Medical University of Lodz, ul. Muszyńskiego 1, Lodz, 90-151, Poland
| | - Johanna Huttunen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, Kuopio, 70211, Finland
| | - Ahmed Montaser
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, Kuopio, 70211, Finland
| | - Kristiina M Huttunen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, Kuopio, 70211, Finland.
| |
Collapse
|
20
|
Kantipudi S, Fotiadis D. Yeast Cell-Based Transport Assay for the Functional Characterization of Human 4F2hc-LAT1 and -LAT2, and LAT1 and LAT2 Substrates and Inhibitors. Front Mol Biosci 2021; 8:676854. [PMID: 34124158 PMCID: PMC8193492 DOI: 10.3389/fmolb.2021.676854] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 05/06/2021] [Indexed: 01/04/2023] Open
Abstract
In mammalian cells, the L-type amino acid transporters (LATs) LAT1 (SLC7A5) and LAT2 (SLC7A8) form heterodimeric amino acid transporters (HATs) with the ancillary protein 4F2hc and are involved in the cellular uptake of specific amino acids. The HAT 4F2hc-LAT1 is found upregulated in various cancer cell types, while 4F2hc-LAT2 is a transporter for non-cancer cells. Preclinical studies have highlighted that 4F2hc-LAT1 plays an important role in tumor progression representing a valid anticancer target. Consequently, current research is focusing on the development of potent and specific human 4F2hc-LAT1 inhibitors. On the other hand, 4F2hc-LAT2 is emerging as target of other diseases, thus also gaining clinical interest. To determine affinity and specificity of substrates and inhibitors for 4F2hc-LAT1 or 4F2hc-LAT2, robust transport cell assays are indispensable. We have optimized and validated a transport assay using cells of the methylotrophic yeast Pichia pastoris stably overexpressing the human HATs 4F2hc-LAT1 or -LAT2, and the LATs LAT1 or LAT2 alone. The radioligand [3H]L-leucine was used as reporter and the substrates L-leucine, triiodothyronine (T3) and thyroxine (T4) as well as the inhibitors BCH and JPH203 (KYT-0353) for assay validation. Obtained half-maximal inhibitory concentrations also provided new insights, e.g., into the LAT specificity of the potent inhibitor JPH203 and on the potency of the thyroid hormones T3 and T4 to inhibit transport through human 4F2hc-LAT2. The LAT1 and LAT2 assays are of particular interest to determine possible implications and influences of 4F2hc in ligand binding and transport. In summary, the presented assays are valuable for characterization of ligands, e.g., towards 4F2hc-LAT1 specificity, and can also be applied for compound screening. Finally, our established approach and assay would also be applicable to other HATs and LATs of interest.
Collapse
Affiliation(s)
- Satish Kantipudi
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
| | - Dimitrios Fotiadis
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
| |
Collapse
|
21
|
Wei Z, Liu X, Cheng C, Yu W, Yi P. Metabolism of Amino Acids in Cancer. Front Cell Dev Biol 2021; 8:603837. [PMID: 33511116 PMCID: PMC7835483 DOI: 10.3389/fcell.2020.603837] [Citation(s) in RCA: 179] [Impact Index Per Article: 59.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 11/30/2020] [Indexed: 12/13/2022] Open
Abstract
Metabolic reprogramming has been widely recognized as a hallmark of malignancy. The uptake and metabolism of amino acids are aberrantly upregulated in many cancers that display addiction to particular amino acids. Amino acids facilitate the survival and proliferation of cancer cells under genotoxic, oxidative, and nutritional stress. Thus, targeting amino acid metabolism is becoming a potential therapeutic strategy for cancer patients. In this review, we will systematically summarize the recent progress of amino acid metabolism in malignancy and discuss their interconnection with mammalian target of rapamycin complex 1 (mTORC1) signaling, epigenetic modification, tumor growth and immunity, and ferroptosis. Finally, we will highlight the potential therapeutic applications.
Collapse
Affiliation(s)
- Zhen Wei
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Brain Science and Advanced Technology Institute, Wuhan University of Science and Technology, Wuhan, China
| | - Xiaoyi Liu
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chunming Cheng
- Department of Radiation Oncology, James Comprehensive Cancer Center and College of Medicine at The Ohio State University, Columbus, OH, United States
| | - Wei Yu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ping Yi
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
22
|
Lopes C, Pereira C, Medeiros R. ASCT2 and LAT1 Contribution to the Hallmarks of Cancer: From a Molecular Perspective to Clinical Translation. Cancers (Basel) 2021; 13:E203. [PMID: 33429909 PMCID: PMC7828050 DOI: 10.3390/cancers13020203] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 12/31/2020] [Accepted: 01/07/2021] [Indexed: 02/06/2023] Open
Abstract
The role of the amino acid transporters ASCT2 and LAT1 in cancer has been explored throughout the years. In this review, we report their impact on the hallmarks of cancer, as well as their clinical significance. Overall, both proteins have been associated with cell death resistance through dysregulation of caspases and sustainment of proliferative signaling through mTOR activation. Furthermore, ASCT2 appears to play an important role in cellular energetics regulation, whereas LAT1 expression is associated with angiogenesis and invasion and metastasis activation. The molecular impact of these proteins on the hallmarks of cancer translates into various clinical applications and both transporters have been identified as prognostic factors in many types of cancer. Concerning their role as therapeutic targets, efforts have been undertaken to synthesize competitive or irreversible ASCT2 and LAT1 inhibitors. However, JHP203, a selective inhibitor of the latter, is, to the best of our knowledge, the only compound included in a Phase 1 clinical trial. In conclusion, considering the usefulness of ASCT2 and LAT1 in a variety of cancer-related pathways and cancer therapy/diagnosis, the development and testing of novel inhibitors for these transporters that could be evaluated in clinical trials represents a promising approach to cancer prognosis improvement.
Collapse
Affiliation(s)
- Catarina Lopes
- Molecular Oncology and Viral Pathology Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal; (C.L.); (R.M.)
| | - Carina Pereira
- Molecular Oncology and Viral Pathology Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal; (C.L.); (R.M.)
- CINTESIS—Center for Health Technology and Services Research, University of Porto, Rua Dr. Plácido da Costa, 4200-450 Porto, Portugal
| | - Rui Medeiros
- Molecular Oncology and Viral Pathology Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal; (C.L.); (R.M.)
- Research Department of the Portuguese League Against Cancer—North (LPCC-NRNorte), Estrada da Circunvalação, 4200-177 Porto, Portugal
| |
Collapse
|
23
|
Kahya U, Köseer AS, Dubrovska A. Amino Acid Transporters on the Guard of Cell Genome and Epigenome. Cancers (Basel) 2021; 13:E125. [PMID: 33401748 PMCID: PMC7796306 DOI: 10.3390/cancers13010125] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/26/2020] [Accepted: 12/27/2020] [Indexed: 02/06/2023] Open
Abstract
Tumorigenesis is driven by metabolic reprogramming. Oncogenic mutations and epigenetic alterations that cause metabolic rewiring may also upregulate the reactive oxygen species (ROS). Precise regulation of the intracellular ROS levels is critical for tumor cell growth and survival. High ROS production leads to the damage of vital macromolecules, such as DNA, proteins, and lipids, causing genomic instability and further tumor evolution. One of the hallmarks of cancer metabolism is deregulated amino acid uptake. In fast-growing tumors, amino acids are not only the source of energy and building intermediates but also critical regulators of redox homeostasis. Amino acid uptake regulates the intracellular glutathione (GSH) levels, endoplasmic reticulum stress, unfolded protein response signaling, mTOR-mediated antioxidant defense, and epigenetic adaptations of tumor cells to oxidative stress. This review summarizes the role of amino acid transporters as the defender of tumor antioxidant system and genome integrity and discusses them as promising therapeutic targets and tumor imaging tools.
Collapse
Affiliation(s)
- Uğur Kahya
- OncoRay–National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, 01309 Dresden, Germany; (U.K.); (A.S.K.)
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology-OncoRay, 01328 Dresden, Germany
| | - Ayşe Sedef Köseer
- OncoRay–National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, 01309 Dresden, Germany; (U.K.); (A.S.K.)
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology-OncoRay, 01328 Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Anna Dubrovska
- OncoRay–National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, 01309 Dresden, Germany; (U.K.); (A.S.K.)
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology-OncoRay, 01328 Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| |
Collapse
|
24
|
Zhang B, Chen Y, Shi X, Zhou M, Bao L, Hatanpaa KJ, Patel T, DeBerardinis RJ, Wang Y, Luo W. Regulation of branched-chain amino acid metabolism by hypoxia-inducible factor in glioblastoma. Cell Mol Life Sci 2021; 78:195-206. [PMID: 32088728 PMCID: PMC8112551 DOI: 10.1007/s00018-020-03483-1] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 12/30/2019] [Accepted: 02/12/2020] [Indexed: 02/07/2023]
Abstract
Hypoxia-inducible factors (HIFs) mediate metabolic reprogramming in response to hypoxia. However, the role of HIFs in branched-chain amino acid (BCAA) metabolism remains unknown. Here we show that hypoxia upregulates mRNA and protein levels of the BCAA transporter LAT1 and the BCAA metabolic enzyme BCAT1, but not their paralogs LAT2-4 and BCAT2, in human glioblastoma (GBM) cell lines as well as primary GBM cells. Hypoxia-induced LAT1 protein upregulation is mediated by both HIF-1 and HIF-2 in GBM cells. Although both HIF-1α and HIF-2α directly bind to the hypoxia response element at the first intron of the human BCAT1 gene, HIF-1α is exclusively responsible for hypoxia-induced BCAT1 expression in GBM cells. Knockout of HIF-1α and HIF-2α significantly reduces glutamate labeling from BCAAs in GBM cells under hypoxia, which provides functional evidence for HIF-mediated reprogramming of BCAA metabolism. Genetic or pharmacological inhibition of BCAT1 inhibits GBM cell growth under hypoxia. Together, these findings uncover a previously unrecognized HIF-dependent metabolic pathway that increases GBM cell growth under conditions of hypoxic stress.
Collapse
Affiliation(s)
- Bo Zhang
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, 75390-9072, USA
| | - Yan Chen
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, 75390-9072, USA
| | - Xiaolei Shi
- Children's Medical Center Research Institute, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Mi Zhou
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, 75390-9072, USA
| | - Lei Bao
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, 75390-9072, USA
| | - Kimmo J Hatanpaa
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, 75390-9072, USA
| | - Toral Patel
- Department of Neurological Surgery, UT Southwestern Medical Center, Dallas, TX, 75390, USA
- Department of Neurology and Neurotherapeutics, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Ralph J DeBerardinis
- Children's Medical Center Research Institute, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Yingfei Wang
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, 75390-9072, USA.
- Department of Neurology and Neurotherapeutics, UT Southwestern Medical Center, Dallas, TX, 75390, USA.
| | - Weibo Luo
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, 75390-9072, USA.
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
25
|
Okanishi H, Ohgaki R, Okuda S, Endou H, Kanai Y. Proteomics and phosphoproteomics reveal key regulators associated with cytostatic effect of amino acid transporter LAT1 inhibitor. Cancer Sci 2020; 112:871-883. [PMID: 33264461 PMCID: PMC7893994 DOI: 10.1111/cas.14756] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 11/28/2020] [Accepted: 11/30/2020] [Indexed: 12/13/2022] Open
Abstract
L‐type amino acid transporter 1 (LAT1) is highly expressed in various cancers and plays important roles not only in the amino acid uptake necessary for cancer growth but also in cellular signaling. Recent research studies have reported anticancer effects of LAT1 inhibitors and demonstrated their potential for cancer therapy. Here, we characterized the proteome and phosphoproteome in LAT1‐inhibited cancer cells. We used JPH203, a selective LAT1 inhibitor, and performed tandem mass tag–based quantitative proteomics and phosphoproteomics on four biliary tract cancer cell lines sensitive to JPH203. Our analysis identified hundreds to thousands of differentially expressed proteins and phosphorylated sites, demonstrating the broad influence of LAT1 inhibition. Our findings showed various functional pathways altered by LAT1 inhibition, and provided possible regulators and key kinases in LAT1‐inhibited cells. Comparison of these changes among cell lines provides insights into general pathways and regulators associated with LAT1 inhibition and particularly suggests the importance of cell cycle–related pathways and kinases. Moreover, we evaluated the anticancer effects of the combinations of JPH203 with cell cycle–related kinase inhibitors and demonstrated their potential for cancer therapy. This is the first study providing the proteome‐wide scope of both protein expression and phosphorylation signaling perturbed by LAT1 inhibition in cancer cells.
Collapse
Affiliation(s)
- Hiroki Okanishi
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Ryuichi Ohgaki
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, Osaka, Japan.,Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiative (OTRI), Osaka University, Osaka, Japan
| | - Suguru Okuda
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | | | - Yoshikatsu Kanai
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, Osaka, Japan.,Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiative (OTRI), Osaka University, Osaka, Japan
| |
Collapse
|
26
|
Venteicher B, Merklin K, Ngo HX, Chien HC, Hutchinson K, Campbell J, Way H, Griffith J, Alvarado C, Chandra S, Hill E, Schlessinger A, Thomas AA. The Effects of Prodrug Size and a Carbonyl Linker on l-Type Amino Acid Transporter 1-Targeted Cellular and Brain Uptake. ChemMedChem 2020; 16:869-880. [PMID: 33230949 DOI: 10.1002/cmdc.202000824] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/23/2020] [Indexed: 11/08/2022]
Abstract
The l-type amino acid transporter 1 (LAT1, SLC7A5) imports dietary amino acids and amino acid drugs (e. g., l-DOPA) into the brain, and plays a role in cancer metabolism. Though there have been numerous reports of LAT1-targeted amino acid-drug conjugates (prodrugs), identifying the structural determinants to enhance substrate activity has been challenging. In this work, we investigated the position and orientation of a carbonyl group in linking hydrophobic moieties including the anti-inflammatory drug ketoprofen to l-tyrosine and l-phenylalanine. We found that esters of meta-carboxyl l-phenylalanine had better LAT1 transport rates than the corresponding acylated l-tyrosine analogues. However, as the size of the hydrophobic moiety increased, we observed a decrease in LAT1 transport rate with a concomitant increase in potency of inhibition. Our results have important implications for designing amino acid prodrugs that target LAT1 at the blood-brain barrier or on cancer cells.
Collapse
Affiliation(s)
- Brooklynn Venteicher
- Department of Chemistry, University of Nebraska at Kearney, 2401 11th Ave, Bruner Hall of Science, Kearney, NE 68849, USA
| | - Kasey Merklin
- Department of Chemistry, University of Nebraska at Kearney, 2401 11th Ave, Bruner Hall of Science, Kearney, NE 68849, USA
| | - Huy X Ngo
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, 1550 4th St, Rm RH581, San Francisco, CA 94143, USA
| | - Huan-Chieh Chien
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, 1550 4th St, Rm RH581, San Francisco, CA 94143, USA
| | - Keino Hutchinson
- Department of Pharmacological Sciences, Icahn School of Medicine at Mt. Sinai, 1468 Madison Ave, Annenberg Building Floor 19, New York, NY 10029, USA
| | - Jerome Campbell
- Department of Chemistry, University of Nebraska at Kearney, 2401 11th Ave, Bruner Hall of Science, Kearney, NE 68849, USA
| | - Hannah Way
- Department of Chemistry, University of Nebraska at Kearney, 2401 11th Ave, Bruner Hall of Science, Kearney, NE 68849, USA
| | - Joseph Griffith
- Department of Chemistry, University of Nebraska at Kearney, 2401 11th Ave, Bruner Hall of Science, Kearney, NE 68849, USA
| | - Cesar Alvarado
- Department of Chemistry, University of Nebraska at Kearney, 2401 11th Ave, Bruner Hall of Science, Kearney, NE 68849, USA
| | - Surabhi Chandra
- Department of Biology, University of Nebraska at Kearney, 2401 11th Ave, Bruner Hall of Science, Kearney, NE 68849, USA
| | - Evan Hill
- Department of Psychology, University of Nebraska, at Kearney 2507 11th Ave, Copeland Hall, Kearney, NE, 68849, (USA)
| | - Avner Schlessinger
- Department of Pharmacological Sciences, Icahn School of Medicine at Mt. Sinai, 1468 Madison Ave, Annenberg Building Floor 19, New York, NY 10029, USA
| | - Allen A Thomas
- Department of Chemistry, University of Nebraska at Kearney, 2401 11th Ave, Bruner Hall of Science, Kearney, NE 68849, USA
| |
Collapse
|
27
|
Zhang J, Xu Y, Li D, Fu L, Zhang X, Bao Y, Zheng L. Review of the Correlation of LAT1 With Diseases: Mechanism and Treatment. Front Chem 2020; 8:564809. [PMID: 33195053 PMCID: PMC7606929 DOI: 10.3389/fchem.2020.564809] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 09/17/2020] [Indexed: 12/17/2022] Open
Abstract
LAT1 is a member of the system L transporter family. The main role of the LAT1 is to transport specific amino acids through cell membranes to provide nutrients to cells and participate in several metabolic pathways. It also contributes to the transport of hormones and some drugs, which are essential for the development and treatment of some diseases. In recent years, many studies have shown that LAT1 is related to cancer, obesity, diabetes, and other diseases. However, the specific mechanism underlying the influence of LAT1 on such conditions remains unclear. Through the increasing number of studies on LAT1, we have obtained a preliminary understanding on the function of LAT1 in diseases. These studies also provide a theoretical basis for finding treatments for LAT1-related diseases, such as cancer. This review summarizes the function and mechanism of LAT1 in different diseases and the treatment of LAT1-related diseases. It also provides support for the development of novel and reliable disease treatments.
Collapse
Affiliation(s)
- Jingshun Zhang
- Reproductive Medical Center, Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Ying Xu
- Reproductive Medical Center, Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Dandan Li
- Reproductive Medical Center, Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Lulu Fu
- Reproductive Medical Center, Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Xueying Zhang
- Reproductive Medical Center, Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Yigang Bao
- Reproductive Medical Center, Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Lianwen Zheng
- Reproductive Medical Center, Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
28
|
Satou M, Wang J, Nakano-Tateno T, Teramachi M, Suzuki T, Hayashi K, Lamothe S, Hao Y, Kurata H, Sugimoto H, Chik C, Tateno T. L-type amino acid transporter 1, LAT1, in growth hormone-producing pituitary tumor cells. Mol Cell Endocrinol 2020; 515:110868. [PMID: 32579901 DOI: 10.1016/j.mce.2020.110868] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 05/03/2020] [Accepted: 05/11/2020] [Indexed: 11/22/2022]
Abstract
Pituitary tumors (PTs) can cause significant mortality and morbidity due to limited therapeutic options. L-type amino acid transporters (LATs), in particular, the LAT1 isoform, is expressed in a variety of tumor cells. Pharmacological inhibition or genetic ablation of LAT1 can suppress leucine transport into cancer cells, resulting in suppression of cancer cell growth. However, roles of LAT1 in PTs have not been elucidated. Therefore, we assessed LAT1 expression in PTs and evaluated a LAT1-specific inhibitor, JPH203, on rat somatomammotroph tumor cells, GH4 cells. GH4 cells dominantly express LAT1 mRNA rather than other LAT isoforms, whereas LAT2 transcripts were most abundant in normal rat pituitary tissues. JPH203 inhibited leucine uptake and cell growth in GH4 cells in a concentration-dependent manner, and appeared to be independent of the mechanistic target, the rapamycin pathway. Although JPH203 did not induce apoptosis, it suppressed growth hormone production in GH4 cells. Also, genetic downregulation of LAT1 showed similar effects on cell growth and hormone production. These results indicated that restriction of LAT1 substrates by JPH203 modulated both cell growth and hormone production. In conclusion, LAT1 may be a new therapeutic target for PTs because its inhibition leads to suppression of cell growth as well as hormone production. JPH203 may represent a promising drug for clinical use in patients with PTs, with the potential of hormonal control and tumor suppression.
Collapse
Affiliation(s)
- Motoyasu Satou
- Division of Endocrinology and Metabolism, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada; Department of Biochemistry, Dokkyo Medical University School of Medicine, Mibu, Tochigi, Japan
| | - Jason Wang
- Division of Endocrinology and Metabolism, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Tae Nakano-Tateno
- Division of Endocrinology and Metabolism, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Mariko Teramachi
- Division of Endocrinology and Metabolism, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | | | - Keitaro Hayashi
- Department of Pharmacology and Toxicology, Dokkyo Medical University School of Medicine, Mibu, Tochigi, Japan
| | - Shawn Lamothe
- Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
| | - Yubin Hao
- Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
| | - Harley Kurata
- Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
| | - Hiroyuki Sugimoto
- Department of Biochemistry, Dokkyo Medical University School of Medicine, Mibu, Tochigi, Japan
| | - Constance Chik
- Division of Endocrinology and Metabolism, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Toru Tateno
- Division of Endocrinology and Metabolism, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
29
|
Ichinoe M, Mikami T, Yanagisawa N, Yoshida T, Hana K, Endou H, Okayasu I, Sengoku N, Ogata H, Saegusa M, Shibuya K, Murakumo Y. Prognostic values of L-type amino acid transporter 1 and CD98hc expression in breast cancer. J Clin Pathol 2020; 74:589-595. [PMID: 32907912 PMCID: PMC8380907 DOI: 10.1136/jclinpath-2020-206457] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 06/23/2020] [Accepted: 08/04/2020] [Indexed: 11/03/2022]
Abstract
AIMS L-type amino acid transporter 1 (LAT1) is a major Na+-independent neutral amino acid transporter, forming a complex with CD98hc. The aim of this study is to investigate the significance of LAT1 and CD98hc in invasive breast cancer. METHODS LAT1 and CD98hc expression was immunohistochemically assessed in 280 invasive breast cancers and analysed for association with clinicopathological features. RESULTS High levels of LAT1 and CD98hc were observed in triple-negative breast cancers (TNBCs) possessing negative immunoreactivity with oestrogen receptor, progesterone receptor and human epidermal growth factor receptor 2, compared with non-TNBCs (NTNBCs), and were associated with lymph-node metastasis and higher nuclear grade. The high-LAT1-expression group showed a poor prognosis in NTNBC and TNBC, however, high-CD98hc-expression group showed a poor prognosis only in NTNBC. LAT1 and CD98hc expression could be the prognostic factors in univariate analyses, but not in multivariate analyses. Further, we found that invasive tumour components showed higher LAT1 and CD98hc expression than non-invasive tumour components. CONCLUSIONS LAT1 and CD98hc may possess prognostic values in invasive breast cancer. LAT1 may be linked with cancer cell activities and disease progression in breast cancer.
Collapse
Affiliation(s)
- Masaaki Ichinoe
- Department of Pathology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Tetuo Mikami
- Department of Pathology, Toho University School of Medicine, Ota-ku, Tokyo, Japan
| | - Nobuyuki Yanagisawa
- Department of Pathology, St. Marianna University School of Medicine Yokohama-City Seibu Hospital, Yokohama, Kanagawa, Japan
| | - Tsutomu Yoshida
- Department of Pathology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | | | | | - Isao Okayasu
- Department of Pathology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Norihiko Sengoku
- Department of Breast and Thyroid Surgery, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Hideaki Ogata
- Department of Surgery, Toho University Omori Medical Center, Ota-ku, Tokyo, Japan
| | - Makoto Saegusa
- Department of Pathology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Kazutoshi Shibuya
- Department of Surgical Pathology, Toho University School of Medicine, Ota-ku, Tokyo, Japan
| | - Yoshiki Murakumo
- Department of Pathology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| |
Collapse
|
30
|
Oncology Therapeutics Targeting the Metabolism of Amino Acids. Cells 2020; 9:cells9081904. [PMID: 32824193 PMCID: PMC7463463 DOI: 10.3390/cells9081904] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 08/12/2020] [Accepted: 08/13/2020] [Indexed: 12/19/2022] Open
Abstract
Amino acid metabolism promotes cancer cell proliferation and survival by supporting building block synthesis, producing reducing agents to mitigate oxidative stress, and generating immunosuppressive metabolites for immune evasion. Malignant cells rewire amino acid metabolism to maximize their access to nutrients. Amino acid transporter expression is upregulated to acquire amino acids from the extracellular environment. Under nutrient depleted conditions, macropinocytosis can be activated where proteins from the extracellular environment are engulfed and degraded into the constituent amino acids. The demand for non-essential amino acids (NEAAs) can be met through de novo synthesis pathways. Cancer cells can alter various signaling pathways to boost amino acid usage for the generation of nucleotides, reactive oxygen species (ROS) scavenging molecules, and oncometabolites. The importance of amino acid metabolism in cancer proliferation makes it a potential target for therapeutic intervention, including via small molecules and antibodies. In this review, we will delineate the targets related to amino acid metabolism and promising therapeutic approaches.
Collapse
|
31
|
Okunushi K, Furihata T, Morio H, Muto Y, Higuchi K, Kaneko M, Otsuka Y, Ohno Y, Watanabe Y, Reien Y, Nakagawa K, Sakamoto S, Wakashin H, Shimojo N, Anzai N. JPH203, a newly developed anti-cancer drug, shows a preincubation inhibitory effect on L-type amino acid transporter 1 function. J Pharmacol Sci 2020; 144:16-22. [PMID: 32653341 DOI: 10.1016/j.jphs.2020.06.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 03/26/2020] [Accepted: 06/07/2020] [Indexed: 12/26/2022] Open
Abstract
JPH203 is a novel anti-cancer drug targeting L-type amino acid transporter 1 (LAT1), which plays a primary role in the uptake of essential amino acids in tumor cells. Although a co-incubation inhibitory effect of JPH203 has been shown in a conventional uptake assay, its preincubation inhibitory effects have remained undetermined. Therefore, we aimed to characterize the preincubation inhibitory effects of JPH203 on LAT1 function using leucine uptake assays in LAT1-positive human colon cancer HT-29 cells. Preincubation of the cells with JPH203 (0.3 μM for 120 min) decreased the activity level to 30% of that in dimethylsulfoxide-treated cells. Similarly, in time-dependency analysis, preincubation of HT-29 cells with 10 μM JPH203 for 30, 60, and 120 min decreased the leucine uptake activity (42%, 32%, and 28% of that in control cells, respectively). Furthermore, the IC50 value of the combination of preincubation and co-incubation effects was lower than that of co-incubation inhibition alone (34.2 ± 3.6 nM vs. 99.2 ± 11.0 nM). In conclusion, we revealed that JPH203 has the capability to inhibit LAT1 function through preincubation effects. Moreover, preincubation synergistically enhances the co-incubation inhibitory effects. These findings provide a novel insight into the anti-cancer effects of JPH203 in cancer therapy.
Collapse
Affiliation(s)
- Kentaro Okunushi
- Department of Pharmacology, Chiba University Graduate School of Medicine, Chiba, Japan; Department of Pediatrics, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Tomomi Furihata
- Department of Pharmacology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Hanae Morio
- Department of Pharmacology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Yasuhide Muto
- Department of Pharmacology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Kosuke Higuchi
- Department of Pharmacology, Chiba University Graduate School of Medicine, Chiba, Japan; Department of Urology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Meika Kaneko
- Department of Pharmacology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Yusuke Otsuka
- Department of Pharmacology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Yuta Ohno
- Department of Pharmacology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Yasuhiro Watanabe
- Department of Pharmacology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Yoshie Reien
- Department of Pharmacology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Kiyoshi Nakagawa
- Department of Pharmacology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Shinichi Sakamoto
- Department of Urology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Hidefumi Wakashin
- Department of Regulatory Physiology, Dokkyo Medical University School of Medicine, Tochigi, Japan
| | - Naoki Shimojo
- Department of Pediatrics, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Naohiko Anzai
- Department of Pharmacology, Chiba University Graduate School of Medicine, Chiba, Japan; Department of Pharmacology and Toxicology, Dokkyo Medical University School of Medicine, Tochigi, Japan.
| |
Collapse
|
32
|
Maimaiti M, Sakamoto S, Yamada Y, Sugiura M, Rii J, Takeuchi N, Imamura Y, Furihata T, Ando K, Higuchi K, Xu M, Sazuka T, Nakamura K, Kaneda A, Kanai Y, Kyprianou N, Ikehara Y, Anzai N, Ichikawa T. Expression of L-type amino acid transporter 1 as a molecular target for prognostic and therapeutic indicators in bladder carcinoma. Sci Rep 2020; 10:1292. [PMID: 31992742 PMCID: PMC6987139 DOI: 10.1038/s41598-020-58136-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 01/09/2020] [Indexed: 12/24/2022] Open
Abstract
L-type amino acid transporter 1 (LAT1) plays a role in transporting essential amino acids including leucine, which regulates the mTOR signaling pathway. Here, we studied the expression profile and functional role of LAT1 in bladder cancer. Furthermore, the pharmacological activity of JPH203, a specific inhibitor of LAT1, was studied in bladder cancer. LAT1 expression in bladder cancer cells was higher than that in normal cells. SiLAT1 and JPH203 suppressed cell proliferative and migratory and invasive abilities in bladder cancer cells. JPH203 inhibited leucine uptake by > 90%. RNA-seq analysis identified insulin-like growth factor-binding protein-5 (IGFBP-5) as a downstream target of JPH203. JPH203 inhibited phosphorylation of MAPK / Erk, AKT, p70S6K and 4EBP-1. Multivariate analysis revealed that high LAT1 expression was found as an independent prognostic factor for overall survival (HR3.46 P = 0.0204). Patients with high LAT1 and IGFBP-5 expression had significantly shorter overall survival periods than those with low expression (P = 0.0005). High LAT1 was related to the high Grade, pathological T stage, LDH, and NLR. Collectively, LAT1 significantly contributed to bladder cancer progression. Targeting LAT1 by JPH203 may represent a novel therapeutic option in bladder cancer treatment.
Collapse
Affiliation(s)
- Maihulan Maimaiti
- Department of Urology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Shinichi Sakamoto
- Department of Urology, Chiba University Graduate School of Medicine, Chiba, Japan.
| | - Yasutaka Yamada
- Department of Urology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Masahiro Sugiura
- Department of Urology, Chiba University Graduate School of Medicine, Chiba, Japan
- Department of Molecular Oncology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Junryo Rii
- Department of Urology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Nobuyoshi Takeuchi
- Department of Urology, Chiba University Graduate School of Medicine, Chiba, Japan
- Department of Tumor Pathology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Yusuke Imamura
- Department of Urology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Tomomi Furihata
- Department of Pharmacology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Keisuke Ando
- Department of Urology, Chiba University Graduate School of Medicine, Chiba, Japan
- Department of Pharmacology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Kosuke Higuchi
- Department of Urology, Chiba University Graduate School of Medicine, Chiba, Japan
- Department of Pharmacology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Minhui Xu
- Bio-system Pharmacology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tomokazu Sazuka
- Department of Urology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Kazuyoshi Nakamura
- Department of Urology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Atsushi Kaneda
- Department of Molecular Oncology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Yoshikatsu Kanai
- Bio-system Pharmacology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Natasha Kyprianou
- Department of Urology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Yuzuru Ikehara
- Department of Tumor Pathology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Naohiko Anzai
- Department of Pharmacology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Tomohiko Ichikawa
- Department of Urology, Chiba University Graduate School of Medicine, Chiba, Japan
| |
Collapse
|
33
|
Lieu EL, Nguyen T, Rhyne S, Kim J. Amino acids in cancer. Exp Mol Med 2020; 52:15-30. [PMID: 31980738 PMCID: PMC7000687 DOI: 10.1038/s12276-020-0375-3] [Citation(s) in RCA: 400] [Impact Index Per Article: 100.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 11/24/2019] [Accepted: 12/02/2019] [Indexed: 01/22/2023] Open
Abstract
Over 90 years ago, Otto Warburg's seminal discovery of aerobic glycolysis established metabolic reprogramming as one of the first distinguishing characteristics of cancer1. The field of cancer metabolism subsequently revealed additional metabolic alterations in cancer by focusing on central carbon metabolism, including the citric acid cycle and pentose phosphate pathway. Recent reports have, however, uncovered substantial non-carbon metabolism contributions to cancer cell viability and growth. Amino acids, nutrients vital to the survival of all cell types, experience reprogrammed metabolism in cancer. This review outlines the diverse roles of amino acids within the tumor and in the tumor microenvironment. Beyond their role in biosynthesis, they serve as energy sources and help maintain redox balance. In addition, amino acid derivatives contribute to epigenetic regulation and immune responses linked to tumorigenesis and metastasis. Furthermore, in discussing the transporters and transaminases that mediate amino acid uptake and synthesis, we identify potential metabolic liabilities as targets for therapeutic intervention.
Collapse
Affiliation(s)
- Elizabeth L. Lieu
- 0000 0001 2175 0319grid.185648.6Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL USA
| | - Tu Nguyen
- 0000 0001 2175 0319grid.185648.6Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL USA
| | - Shawn Rhyne
- 0000 0001 2175 0319grid.185648.6Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL USA
| | - Jiyeon Kim
- 0000 0001 2175 0319grid.185648.6Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL USA
| |
Collapse
|
34
|
Characterization of the expression of LAT1 as a prognostic indicator and a therapeutic target in renal cell carcinoma. Sci Rep 2019; 9:16776. [PMID: 31748583 PMCID: PMC6868143 DOI: 10.1038/s41598-019-53397-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Accepted: 10/30/2019] [Indexed: 01/02/2023] Open
Abstract
Large neutral amino acid transporter 1 (LAT1, SLC7A5) is abundantly expressed in various types of cancer, and it has been thought to assist cancer progression through its activity for uptake of neutral amino acids. However, the roles of LAT1 in renal cell carcinoma (RCC) prognosis and treatment remain uncharacterized. Therefore, we first retrospectively examined the LAT1 expression profile and its associations with clinical factors in RCC tissues (n = 92). The results of immunohistochemistry showed that most of the tissues examined (92%) had cancer-associated LAT1 expression. Furthermore, the overall survival (OS) and progression-free survival (PFS) were shorter in patients with high LAT1 expression levels than in those with low LAT1 expression levels (P = 0.018 and 0.014, respectively), and these associations were further strengthened by the results of univariate and multivariate analyses. Next, we tested the effects of JPH203, which is a selective LAT1 inhibitor, on RCC-derived Caki-1 and ACHN cells. It was found that JPH203 inhibited the growth of these cell types in a dose-dependent manner. Moreover, JPH203 clearly suppressed their migration and invasion activities. Thus, our results show that LAT1 has a great potential to become not only a prognosis biomarker but also a therapeutic target in RCC clinical settings.
Collapse
|
35
|
A novel therapeutic approach for anaplastic thyroid cancer through inhibition of LAT1. Sci Rep 2019; 9:14616. [PMID: 31601917 PMCID: PMC6787004 DOI: 10.1038/s41598-019-51144-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 09/20/2019] [Indexed: 12/13/2022] Open
Abstract
A novel therapeutic approach is urgently needed for patients with anaplastic thyroid cancer (ATC) due to its fatal and rapid progress. We recently reported that ATC highly expressed MYC protein and blocking of MYC through its selective inhibitor, JQ1, decreased ATC growth and improved survival in preclinical models. One of the important roles of MYC is regulation of L-neutral amino acid transporter 1 (LAT1) protein and inhibition of LAT1 would provide similar anti-tumor effect. We first identified that while the human ATC expresses LAT1 protein, it is little or not detected in non-cancerous thyroidal tissue, further supporting LAT1 as a good target. Then we evaluated the efficacy of JPH203, a LAT1 inhibitor, against ATC by using the in vitro cell-based studies and in vivo xenograft model bearing human ATC cells. JPH203 markedly inhibited proliferation of three ATC cell lines through suppression of mTOR signals and blocked cell cycle progression from the G0/G1 phase to the S phase. The tumor growth inhibition and decrease in size by JPH203 via inhibition of mTOR signaling and G0/G1 cell cycle associated proteins were further confirmed in xenograft models. These preclinical findings suggest that LAT1 inhibitors are strong candidates to control ATC, for which current treatment options are highly limited.
Collapse
|
36
|
Zhang BK, Moran AM, Bailey CG, Rasko JEJ, Holst J, Wang Q. EGF-activated PI3K/Akt signalling coordinates leucine uptake by regulating LAT3 expression in prostate cancer. Cell Commun Signal 2019; 17:83. [PMID: 31345230 PMCID: PMC6659227 DOI: 10.1186/s12964-019-0400-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 07/16/2019] [Indexed: 02/06/2023] Open
Abstract
Background Growth factors, such as EGF, activate the PI3K/Akt/mTORC1 signalling pathway, which regulates a distinct program of protein synthesis leading to cell growth. This pathway relies on mTORC1 sensing sufficient levels of intracellular amino acids, such as leucine, which are required for mTORC1 activation. However, it is currently unknown whether there is a direct link between these external growth signals and intracellular amino acid levels. In primary prostate cancer cells, intracellular leucine levels are regulated by L-type amino acid transporter 3 (LAT3/SLC43A1), and we therefore investigated whether LAT3 is regulated by growth factor signalling. Methods To investigate how PI3K/Akt signalling regulates leucine transport, prostate cancer cells were treated with different PI3K/Akt inhibitors, or stable knock down of LAT3 by shRNA, followed by analysis of leucine uptake, western blotting, immunofluorescent staining and proximity ligation assay. Results Inhibition of PI3K/Akt signalling significantly reduced leucine transport in LNCaP and PC-3 human prostate cancer cell lines, while growth factor addition significantly increased leucine uptake. These effects appeared to be mediated by LAT3 transport, as LAT3 knockdown blocked leucine uptake, and was not rescued by growth factor activation or further inhibited by signalling pathway inhibition. We further demonstrated that EGF significantly increased LAT3 protein levels when Akt was phosphorylated, and that Akt and LAT3 co-localised on the plasma membrane in EGF-activated LNCaP cells. These effects were likely due to stabilisation of LAT3 protein levels on the plasma membrane, with EGF treatment preventing ubiquitin-mediated LAT3 degradation. Conclusion Growth factor-activated PI3K/Akt signalling pathway regulates leucine transport through LAT3 in prostate cancer cell lines. These data support a direct link between growth factor and amino acid uptake, providing a mechanism by which the cells rapidly coordinate amino acid uptake for cell growth. Electronic supplementary material The online version of this article (10.1186/s12964-019-0400-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Blake K Zhang
- Centenary Institute, University of Sydney, Camperdown, Australia.,Sydney Medical School, University of Sydney, Camperdown, Australia
| | - Anne M Moran
- Centenary Institute, University of Sydney, Camperdown, Australia.,Sydney Medical School, University of Sydney, Camperdown, Australia
| | - Charles G Bailey
- Sydney Medical School, University of Sydney, Camperdown, Australia.,Gene & Stem Cell Therapy Program Centenary Institute, University of Sydney, Camperdown, Australia
| | - John E J Rasko
- Sydney Medical School, University of Sydney, Camperdown, Australia.,Gene & Stem Cell Therapy Program Centenary Institute, University of Sydney, Camperdown, Australia.,Cell and Molecular Therapies, Royal Prince Alfred Hospital, Camperdown, Australia
| | - Jeff Holst
- Translational Cancer Metabolism Laboratory, Lowy Cancer Research Centre, School of Medical Sciences and Prince of Wales Clinical School, University of New South Wales, Sydney, Australia. .,Origins of Cancer Program Centenary Institute, University of Sydney, Camperdown, Australia.
| | - Qian Wang
- Sydney Medical School, University of Sydney, Camperdown, Australia. .,Translational Cancer Metabolism Laboratory, Lowy Cancer Research Centre, School of Medical Sciences and Prince of Wales Clinical School, University of New South Wales, Sydney, Australia.
| |
Collapse
|
37
|
Katada T, Sakurai H. Xenopus slc7a5 is essential for notochord function and eye development. Mech Dev 2019; 155:48-59. [DOI: 10.1016/j.mod.2019.01.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 12/26/2018] [Accepted: 01/03/2019] [Indexed: 10/27/2022]
|
38
|
Chien HC, Colas C, Finke K, Springer S, Stoner L, Zur AA, Venteicher B, Campbell J, Hall C, Flint A, Augustyn E, Hernandez C, Heeren N, Hansen L, Anthony A, Bauer J, Fotiadis D, Schlessinger A, Giacomini KM, Thomas AA. Reevaluating the Substrate Specificity of the L-Type Amino Acid Transporter (LAT1). J Med Chem 2018; 61:7358-7373. [PMID: 30048132 DOI: 10.1021/acs.jmedchem.8b01007] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The L-type amino acid transporter 1 (LAT1, SLC7A5) transports essential amino acids across the blood-brain barrier (BBB) and into cancer cells. To utilize LAT1 for drug delivery, potent amino acid promoieties are desired, as prodrugs must compete with millimolar concentrations of endogenous amino acids. To better understand ligand-transporter interactions that could improve potency, we developed structural LAT1 models to guide the design of substituted analogues of phenylalanine and histidine. Furthermore, we evaluated the structure-activity relationship (SAR) for both enantiomers of naturally occurring LAT1 substrates. Analogues were tested in cis-inhibition and trans-stimulation cell assays to determine potency and uptake rate. Surprisingly, LAT1 can transport amino acid-like substrates with wide-ranging polarities including those containing ionizable substituents. Additionally, the rate of LAT1 transport was generally nonstereoselective even though enantiomers likely exhibit different binding modes. Our findings have broad implications to the development of new treatments for brain disorders and cancer.
Collapse
Affiliation(s)
- Huan-Chieh Chien
- Department of Bioengineering and Therapeutic Sciences , University of California, San Francisco , San Francisco , California 94158 , United States
| | - Claire Colas
- Department of Pharmacological Sciences , Icahn School of Medicine at Mount Sinai , New York , New York 10029 , United States
| | - Karissa Finke
- Department of Chemistry , University of Nebraska at Kearney , Kearney , Nebraska 68849 , United States
| | - Seth Springer
- Department of Chemistry , University of Nebraska at Kearney , Kearney , Nebraska 68849 , United States
| | - Laura Stoner
- Department of Chemistry , University of Nebraska at Kearney , Kearney , Nebraska 68849 , United States
| | - Arik A Zur
- Department of Bioengineering and Therapeutic Sciences , University of California, San Francisco , San Francisco , California 94158 , United States
| | - Brooklynn Venteicher
- Department of Chemistry , University of Nebraska at Kearney , Kearney , Nebraska 68849 , United States
| | - Jerome Campbell
- Department of Chemistry , University of Nebraska at Kearney , Kearney , Nebraska 68849 , United States
| | - Colton Hall
- Department of Chemistry , University of Nebraska at Kearney , Kearney , Nebraska 68849 , United States
| | - Andrew Flint
- Department of Chemistry , University of Nebraska at Kearney , Kearney , Nebraska 68849 , United States
| | - Evan Augustyn
- Department of Chemistry , University of Nebraska at Kearney , Kearney , Nebraska 68849 , United States
| | - Christopher Hernandez
- Department of Chemistry , University of Nebraska at Kearney , Kearney , Nebraska 68849 , United States
| | - Nathan Heeren
- Department of Chemistry , University of Nebraska at Kearney , Kearney , Nebraska 68849 , United States
| | - Logan Hansen
- Department of Chemistry , University of Nebraska at Kearney , Kearney , Nebraska 68849 , United States
| | - Abby Anthony
- Department of Chemistry , University of Nebraska at Kearney , Kearney , Nebraska 68849 , United States
| | - Justine Bauer
- Department of Chemistry , University of Nebraska at Kearney , Kearney , Nebraska 68849 , United States
| | - Dimitrios Fotiadis
- Institute of Biochemistry and Molecular Medicine, and Swiss National Centre of Competence in Research (NCCR) TransCure , University of Bern , 3012 Bern , Switzerland
| | - Avner Schlessinger
- Department of Pharmacological Sciences , Icahn School of Medicine at Mount Sinai , New York , New York 10029 , United States
| | - Kathleen M Giacomini
- Department of Bioengineering and Therapeutic Sciences , University of California, San Francisco , San Francisco , California 94158 , United States
| | - Allen A Thomas
- Department of Chemistry , University of Nebraska at Kearney , Kearney , Nebraska 68849 , United States
| |
Collapse
|
39
|
Singh N, Ecker GF. Insights into the Structure, Function, and Ligand Discovery of the Large Neutral Amino Acid Transporter 1, LAT1. Int J Mol Sci 2018; 19:E1278. [PMID: 29695141 PMCID: PMC5983779 DOI: 10.3390/ijms19051278] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 04/18/2018] [Accepted: 04/19/2018] [Indexed: 12/31/2022] Open
Abstract
The large neutral amino acid transporter 1 (LAT1, or SLC7A5) is a sodium- and pH-independent transporter, which supplies essential amino acids (e.g., leucine, phenylalanine) to cells. It plays an important role at the Blood⁻Brain Barrier (BBB) where it facilitates the transport of thyroid hormones, pharmaceuticals (e.g., l-DOPA, gabapentin), and metabolites into the brain. Moreover, its expression is highly upregulated in various types of human cancer that are characterized by an intense demand for amino acids for growth and proliferation. Therefore, LAT1 is believed to be an important drug target for cancer treatment. With the crystallization of the arginine/agmatine antiporter (AdiC) from Escherichia Coli, numerous homology models of LAT1 have been built to elucidate the substrate binding site, ligand⁻transporter interaction, and structure⁻function relationship. The use of these models in combination with molecular docking and experimental testing has identified novel chemotypes of ligands of LAT1. Here, we highlight the structure, function, transport mechanism, and homology modeling of LAT1. Additionally, results from structure⁻function studies performed on LAT1 are addressed, which have enhanced our knowledge of the mechanism of substrate binding and translocation. This is followed by a discussion on ligand- and structure-based approaches, with an emphasis on elucidating the molecular basis of LAT1 inhibition. Finally, we provide an exhaustive summary of different LAT1 inhibitors that have been identified so far, including the recently discovered irreversible covalent inhibitors.
Collapse
Affiliation(s)
- Natesh Singh
- Department of Pharmaceutical Chemistry, University of Vienna, Althanstrasse 14, 1090 Wien, Austria.
| | - Gerhard F Ecker
- Department of Pharmaceutical Chemistry, University of Vienna, Althanstrasse 14, 1090 Wien, Austria.
| |
Collapse
|
40
|
Thongon N, Zucal C, D'Agostino VG, Tebaldi T, Ravera S, Zamporlini F, Piacente F, Moschoi R, Raffaelli N, Quattrone A, Nencioni A, Peyron JF, Provenzani A. Cancer cell metabolic plasticity allows resistance to NAMPT inhibition but invariably induces dependence on LDHA. Cancer Metab 2018. [PMID: 29541451 PMCID: PMC5844108 DOI: 10.1186/s40170-018-0174-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Background Inhibitors of nicotinamide phosphoribosyltransferase (NAMPT), the rate-limiting enzyme in NAD+ biosynthesis from nicotinamide, exhibit anticancer effects in preclinical models. However, continuous exposure to NAMPT inhibitors, such as FK866, can induce acquired resistance. Methods We developed FK866-resistant CCRF-CEM (T cell acute lymphoblastic leukemia) and MDA MB231 (breast cancer) models, and by exploiting an integrated approach based on genetic, biochemical, and genome wide analyses, we annotated the drug resistance mechanisms. Results Acquired resistance to FK866 was independent of NAMPT mutations but rather was based on a shift towards a glycolytic metabolism and on lactate dehydrogenase A (LDHA) activity. In addition, resistant CCRF-CEM cells, which exhibit high quinolinate phosphoribosyltransferase (QPRT) activity, also exploited amino acid catabolism as an alternative source for NAD+ production, becoming addicted to tryptophan and glutamine and sensitive to treatment with the amino acid transport inhibitor JPH203 and with l-asparaginase, which affects glutamine exploitation. Vice versa, in line with their low QPRT expression, FK866-resistant MDA MB231 did not rely on amino acids for their resistance phenotype. Conclusions Our study identifies novel mechanisms of resistance to NAMPT inhibition, which may be useful to design more rational strategies for targeting cancer metabolism.
Collapse
Affiliation(s)
- Natthakan Thongon
- 1Center For Integrative Biology (CIBIO), University of Trento, via Sommarive 9, Trento, Italy
| | - Chiara Zucal
- 1Center For Integrative Biology (CIBIO), University of Trento, via Sommarive 9, Trento, Italy
| | | | - Toma Tebaldi
- 1Center For Integrative Biology (CIBIO), University of Trento, via Sommarive 9, Trento, Italy
| | - Silvia Ravera
- 2Department of Pharmacy, Biochemistry Laboratory, University of Genova, Genova, Italy
| | - Federica Zamporlini
- 3Department of Agricultural, Food and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy
| | | | - Ruxanda Moschoi
- 5Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), INSERM U1065, Nice, France
| | - Nadia Raffaelli
- 3Department of Agricultural, Food and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Alessandro Quattrone
- 1Center For Integrative Biology (CIBIO), University of Trento, via Sommarive 9, Trento, Italy
| | - Alessio Nencioni
- 4Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Jean-Francois Peyron
- 5Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), INSERM U1065, Nice, France
| | - Alessandro Provenzani
- 1Center For Integrative Biology (CIBIO), University of Trento, via Sommarive 9, Trento, Italy
| |
Collapse
|
41
|
Yoon BR, Oh YJ, Kang SW, Lee EB, Lee WW. Role of SLC7A5 in Metabolic Reprogramming of Human Monocyte/Macrophage Immune Responses. Front Immunol 2018; 9:53. [PMID: 29422900 PMCID: PMC5788887 DOI: 10.3389/fimmu.2018.00053] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 01/09/2018] [Indexed: 12/20/2022] Open
Abstract
Amino acids (AAs) are necessary nutrients which act not only as building blocks in protein synthesis but also in crucial anabolic cellular signaling pathways. It has been demonstrated that SLC7A5 is a critical transporter that mediates uptake of several essential amino acids in highly proliferative tumors and activated T cells. However, the dynamics and relevance of SLC7A5 activity in monocytes/macrophages is still poorly understood. We provide evidence that SLC7A5-mediated leucine influx contributes to pro-inflammatory cytokine production via mTOR complex 1 (mTORC1)-induced glycolytic reprograming in activated human monocytes/macrophages. Moreover, expression of SLC7A5 is significantly elevated in monocytes derived from patients with rheumatoid arthritis (RA), a chronic inflammatory disease, and was also markedly induced by LPS stimulation of both monocytes and macrophages from healthy individuals. Further, pharmacological blockade or silencing of SLC7A5 led to a significant reduction of IL-1β downstream of leucine-mediated mTORC1 activation. Inhibition of SLC7A5-mediated leucine influx was linked to downregulation of glycolytic metabolism as evidenced by the decreased extracellular acidification rate, suggesting a regulatory role for this molecule in glycolytic reprograming. Furthermore, the expression of SLC7A5 on circulating monocytes from RA patients positively correlated with clinical parameters, suggesting that SLC7A5-mediated AA influx is related to inflammatory conditions.
Collapse
Affiliation(s)
- Bo Ruem Yoon
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, South Korea
| | - Yoon-Jeong Oh
- Division of Rheumatology, Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | - Seong Wook Kang
- Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Eun Bong Lee
- Division of Rheumatology, Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | - Won-Woo Lee
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, South Korea.,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea.,Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, South Korea.,Institute of Infectious Diseases, Seoul National University College of Medicine, Seoul, South Korea.,Seoul National University Hospital Biomedical Research Institute, Seoul, South Korea
| |
Collapse
|
42
|
Choi DW, Kim DK, Kanai Y, Wempe MF, Endou H, Kim JK. JPH203, a selective L-type amino acid transporter 1 inhibitor, induces mitochondria-dependent apoptosis in Saos2 human osteosarcoma cells. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2017; 21:599-607. [PMID: 29200902 PMCID: PMC5709476 DOI: 10.4196/kjpp.2017.21.6.599] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 08/11/2017] [Accepted: 09/08/2017] [Indexed: 11/30/2022]
Abstract
Most normal cells express L-type amino acid transporter 2 (LAT2). However, L-type amino acid transporter 1 (LAT1) is highly expressed in many tumor cells and presumed to support their increased growth and proliferation. This study examined the effects of JPH203, a selective LAT1 inhibitor, on cell growth and its mechanism for cell death in Saos2 human osteosarcoma cells. FOB human osteoblastic cells and Saos2 cells expressed LAT1 and LAT2 together with their associating protein 4F2 heavy chain, but the expression of LAT2 in the Saos2 cells was especially weak. JPH203 and BCH, a non-selective L-type amino acid transporter inhibitor, potently inhibited L-leucine uptake in Saos2 cells. As expected, the intrinsic ability of JPH203 to inhibit L-leucine uptake was far more efficient than that of BCH in Saos2 cells. Likewise, JPH203 and BCH inhibited Saos2 cell growth with JPH203 being superior to BCH in this regard. Furthermore, JPH203 increased apoptosis rates and formed DNA ladder in Saos2 cells. Moreover, JPH203 activated the mitochondria-dependent apoptotic signaling pathway by upregulating pro-apoptotic factors, such as Bad, Bax, and Bak, and the active form of caspase-9, and downregulating anti-apoptotic factors, such as Bcl-2 and Bcl-xL. These results suggest that the inhibition of LAT1 activity via JPH203, which may act as a potential novel anti-cancer agent, leads to apoptosis mediated by the mitochondria-dependent intrinsic apoptotic signaling pathway by inducing the intracellular depletion of neutral amino acids essential for cell growth in Saos2 human osteosarcoma cells.
Collapse
Affiliation(s)
- Dae Woo Choi
- Department of Pharmacology, Chonnam National University Medical School, Gwangju 61469, Korea
| | - Do Kyung Kim
- Department of Oral Physiology, Chosun University School of Dentistry, Gwangju 61452, Korea
| | - Yoshikatsu Kanai
- Department of Bio-system Pharmacology, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Michael F Wempe
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado 80045, USA
| | - Hitoshi Endou
- Department of Pharmacology and Toxicology, Kyorin University School of Medicine, Tokyo 181-8611, Japan.,J-Pharma Co., Ltd., Yokohama, Kanagawa 230-0046, Japan
| | - Jong-Keun Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju 61469, Korea
| |
Collapse
|
43
|
Yothaisong S, Dokduang H, Anzai N, Hayashi K, Namwat N, Yongvanit P, Sangkhamanon S, Jutabha P, Endou H, Loilome W. Inhibition of l-type amino acid transporter 1 activity as a new therapeutic target for cholangiocarcinoma treatment. Tumour Biol 2017; 39:1010428317694545. [PMID: 28347255 DOI: 10.1177/1010428317694545] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Unlike normal cells, cancer cells undergo unlimited growth and multiplication, causing them to require massive amounts of amino acid to support their continuous metabolism. Among the amino acid transporters expressed on the plasma membrane, l-type amino acid transporter-1, a Na+-independent neutral amino acid transporter, is highly expressed in many types of human cancer including cholangiocarcinoma. Our previous study reported that l-type amino acid transporter-1 and its co-functional protein CD98 were highly expressed and implicated in cholangiocarcinoma progression and carcinogenesis. Therefore, this study determined the effect of JPH203, a selective inhibitor of l-type amino acid transporter-1 activity, on cholangiocarcinoma cell inhibition both in vitro and in vivo. JPH203 dramatically suppressed [14C]l-leucine uptake as well as cell growth in cholangiocarcinoma cell lines along with altering the expression of l-type amino acid transporter-1 and CD98 in response to amino acid depletion. We also demonstrated that JPH203 induced both G2/M and G0/G1 cell cycle arrest, as well as reduced the S phase accompanied by altered expression of the proteins in cell cycle progression: cyclin D1, CDK4, and CDK6. There was also cell cycle arrest of the related proteins, P21 and P27, in KKU-055 and KKU-213 cholangiocarcinoma cells. Apoptosis induction, detected by an increase in trypan blue-stained cells along with a cleaved caspase-3/caspase-3 ratio, occurred in JPH203-treated cholangiocarcinoma cells at the highest concentration tested (100 µM). As expected, daily intravenous administration of JPH203 (12.5 and 25 mg/kg) significantly inhibited tumor growth in KKU-213 cholangiocarcinoma cell xenografts in the nude mice model in a dose-dependent manner with no statistically significant change in the animal's body weight and with no differences in the histology and appearance of the internal organs compared with the control group. Our study demonstrates that suppression of l-type amino acid transporter-1 activity using JPH203 might be used as a new therapeutic strategy for cholangiocarcinoma treatment.
Collapse
Affiliation(s)
- Supak Yothaisong
- 1 Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,2 Liver Fluke and Cholangiocarcinoma Research Center, Khon Kaen University, Khon Kaen, Thailand.,3 Cholangiocarcinoma Screening and Care Program (CASCAP), Khon Kaen University, Khon Kaen, Thailand
| | - Hasaya Dokduang
- 2 Liver Fluke and Cholangiocarcinoma Research Center, Khon Kaen University, Khon Kaen, Thailand.,3 Cholangiocarcinoma Screening and Care Program (CASCAP), Khon Kaen University, Khon Kaen, Thailand
| | - Naohiko Anzai
- 4 Department of Pharmacology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Keitaro Hayashi
- 5 Department of Pharmacology and Toxicology, School of Medicine, Dokkyo Medical University, Mibu, Japan
| | - Nisana Namwat
- 1 Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,2 Liver Fluke and Cholangiocarcinoma Research Center, Khon Kaen University, Khon Kaen, Thailand.,3 Cholangiocarcinoma Screening and Care Program (CASCAP), Khon Kaen University, Khon Kaen, Thailand
| | - Puangrat Yongvanit
- 1 Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,2 Liver Fluke and Cholangiocarcinoma Research Center, Khon Kaen University, Khon Kaen, Thailand.,3 Cholangiocarcinoma Screening and Care Program (CASCAP), Khon Kaen University, Khon Kaen, Thailand
| | - Sakkarn Sangkhamanon
- 6 Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Promsuk Jutabha
- 5 Department of Pharmacology and Toxicology, School of Medicine, Dokkyo Medical University, Mibu, Japan
| | | | - Watcharin Loilome
- 1 Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,2 Liver Fluke and Cholangiocarcinoma Research Center, Khon Kaen University, Khon Kaen, Thailand.,3 Cholangiocarcinoma Screening and Care Program (CASCAP), Khon Kaen University, Khon Kaen, Thailand
| |
Collapse
|
44
|
Kagawa S, Nishii R, Higashi T, Yamauchi H, Ogawa E, Okudaira H, Kobayashi M, Yoshimoto M, Shikano N, Kawai K. Relationship between [ 14C]MeAIB uptake and amino acid transporter family gene expression levels or proliferative activity in a pilot study in human carcinoma cells: Comparison with [ 3H]methionine uptake. Nucl Med Biol 2017; 49:8-15. [PMID: 28284101 DOI: 10.1016/j.nucmedbio.2017.01.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 01/18/2017] [Accepted: 01/18/2017] [Indexed: 01/09/2023]
Abstract
INTRODUCTION To clarify the difference between system A and L amino acid transport imaging in PET clinical imaging, we focused on the use of α-[N-methyl-11C]-methylaminoisobutyric acid ([11C]MeAIB), and compared it with [S-methyl-11C]-L-methionine ([11C]MET). The aim of this study was to assess the correlation of accumulation of these two radioactive amino acid analogs with expression of amino acid transporters and cell proliferative activity in carcinoma cells. METHODS Amino acid uptake inhibitor studies were performed in four human carcinoma cells (epidermal carcinoma A431, colorectal carcinoma LS180, and lung carcinomas PC14/GL and H441/GL) using the radioisotope analogs [3H]MET and [14C]MeAIB. MeAIB was used to inhibit the A system and 2-amino-2-norbornane-carboxylic acid (BCH) was used to inhibit the L system. The carcinoma gene expression levels of a number of amino acid transporters were measured by microarray and quantitative polymerase chain reaction. Carcinoma proliferative activity was assessed using accumulation of [methyl-3H]-3'-deoxy-3'-fluorothymidine ([3H]FLT). RESULTS AND CONCLUSION [14C]MeAIB uptake occurred principally via a Na+-dependent A type mechanism whereas [3H]MET uptake occurred predominantly via a Na+-independent L type mechanism although other transporters were also utilized depending on cell type. There was no correlation between [3H]MET uptake and total system L amino acid transporter (LAT) expression. In contrast, [14C]MeAIB uptake strongly correlated with total system A amino acid transporter (SNAT) expression and proliferative activity in this preliminary study using four human carcinoma cell lines. Carcinoma proliferative activity also correlated with total SNAT expression. Advances in Knowledge and Implications for Patient Care: Because there is a significant correlation between the accumulation of [14C]MeAIB and the gene expression level of total SNAT as well as the accumulation of [3H]FLT, it is suggested that use of the analog [11C]MeAIB in PET may provide an indication of tumor cell proliferative activity. [11C]MeAIB is therefore expected to be very useful in PET imaging.
Collapse
Affiliation(s)
- Shinya Kagawa
- Division of PET Imaging, Shiga Medical Center Research Institute, Shiga, Japan; Division of Health Sciences, Graduate School of Medical Sciences, Kanazawa University, Ishikawa, Japan
| | - Ryuichi Nishii
- Division of PET Imaging, Shiga Medical Center Research Institute, Shiga, Japan; Department of Molecular Imaging and Theranostics, National Institute of Radiological Sciences, Chiba, Japan
| | - Tatsuya Higashi
- Division of PET Imaging, Shiga Medical Center Research Institute, Shiga, Japan; Department of Molecular Imaging and Theranostics, National Institute of Radiological Sciences, Chiba, Japan
| | - Hiroshi Yamauchi
- Division of PET Imaging, Shiga Medical Center Research Institute, Shiga, Japan
| | - Emi Ogawa
- Department of Radiology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | | | - Masato Kobayashi
- Wellness Promotion Science Center, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Ishikawa, Japan
| | - Mitsuyoshi Yoshimoto
- Division of Functional Imaging, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Chiba, Japan
| | - Naoto Shikano
- Department of Radiological Sciences, Ibaraki Prefectural University of Health Sciences, Ibaraki, Japan
| | - Keiichi Kawai
- Division of Health Sciences, Graduate School of Medical Sciences, Kanazawa University, Ishikawa, Japan.
| |
Collapse
|
45
|
Zur AA, Chien HC, Augustyn E, Flint A, Heeren N, Finke K, Hernandez C, Hansen L, Miller S, Lin L, Giacomini KM, Colas C, Schlessinger A, Thomas AA. LAT1 activity of carboxylic acid bioisosteres: Evaluation of hydroxamic acids as substrates. Bioorg Med Chem Lett 2016; 26:5000-5006. [PMID: 27624080 DOI: 10.1016/j.bmcl.2016.09.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 08/27/2016] [Accepted: 09/01/2016] [Indexed: 01/23/2023]
Abstract
Large neutral amino acid transporter 1 (LAT1) is a solute carrier protein located primarily in the blood-brain barrier (BBB) that offers the potential to deliver drugs to the brain. It is also up-regulated in cancer cells, as part of a tumor's increased metabolic demands. Previously, amino acid prodrugs have been shown to be transported by LAT1. Carboxylic acid bioisosteres may afford prodrugs with an altered physicochemical and pharmacokinetic profile than those derived from natural amino acids, allowing for higher brain or tumor levels of drug and/or lower toxicity. The effect of replacing phenylalanine's carboxylic acid with a tetrazole, acylsulfonamide and hydroxamic acid (HA) bioisostere was examined. Compounds were tested for their ability to be LAT1 substrates using both cis-inhibition and trans-stimulation cell assays. As HA-Phe demonstrated weak substrate activity, its structure-activity relationship (SAR) was further explored by synthesis and testing of HA derivatives of other LAT1 amino acid substrates (i.e., Tyr, Leu, Ile, and Met). The potential for a false positive in the trans-stimulation assay caused by parent amino acid was evaluated by conducting compound stability experiments for both HA-Leu and the corresponding methyl ester derivative. We concluded that HA's are transported by LAT1. In addition, our results lend support to a recent account that amino acid esters are LAT1 substrates, and that hydrogen bonding may be as important as charge for interaction with the transporter binding site.
Collapse
Affiliation(s)
- Arik A Zur
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, CA 94158, United States.
| | - Huan-Chieh Chien
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, CA 94158, United States
| | - Evan Augustyn
- Department of Chemistry, University of Nebraska Kearney, Kearney, NE 68849, United States
| | - Andrew Flint
- Department of Chemistry, University of Nebraska Kearney, Kearney, NE 68849, United States
| | - Nathan Heeren
- Department of Chemistry, University of Nebraska Kearney, Kearney, NE 68849, United States
| | - Karissa Finke
- Department of Chemistry, University of Nebraska Kearney, Kearney, NE 68849, United States
| | - Christopher Hernandez
- Department of Chemistry, University of Nebraska Kearney, Kearney, NE 68849, United States
| | - Logan Hansen
- Department of Chemistry, University of Nebraska Kearney, Kearney, NE 68849, United States
| | - Sydney Miller
- Department of Chemistry, University of Nebraska Kearney, Kearney, NE 68849, United States
| | - Lawrence Lin
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, CA 94158, United States
| | - Kathleen M Giacomini
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, CA 94158, United States
| | - Claire Colas
- Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Department of Structural and Chemical Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Avner Schlessinger
- Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Department of Structural and Chemical Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Allen A Thomas
- Department of Chemistry, University of Nebraska Kearney, Kearney, NE 68849, United States.
| |
Collapse
|
46
|
Huttunen KM, Gynther M, Huttunen J, Puris E, Spicer JA, Denny WA. A Selective and Slowly Reversible Inhibitor of l-Type Amino Acid Transporter 1 (LAT1) Potentiates Antiproliferative Drug Efficacy in Cancer Cells. J Med Chem 2016; 59:5740-51. [PMID: 27253989 DOI: 10.1021/acs.jmedchem.6b00190] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The l-type amino acid transporter 1 (LAT1) is a transmembrane protein carrying bulky and neutral amino acids into cells. LAT1 is overexpressed in several types of tumors, and its inhibition can result in reduced cancer cell growth. However, known LAT1 inhibitors lack selectivity over other transporters. In the present study, we designed and synthesized a novel selective LAT1 inhibitor (1), which inhibited the uptake of LAT1 substrate, l-leucin as well as cell growth. It also significantly potentiated the efficacy of bestatin and cisplatin even at low concentrations (25 μM). Inhibition was slowly reversible, as the inhibitor was able to be detached from the cell surface and blood-brain barrier. Moreover, the inhibitor was metabolically stable and selective toward LAT1. Since the inhibitor was readily accumulated into the prostate after intraperitoneal injection to the healthy mice, this compound may be a promising agent or adjuvant especially for the treatment of prostate cancer.
Collapse
Affiliation(s)
- Kristiina M Huttunen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland , P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Mikko Gynther
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland , P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Johanna Huttunen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland , P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Elena Puris
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland , P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Julie A Spicer
- Auckland Cancer Society Research Centre, The University of Auckland , Private Bag 92019, Auckland 1142, New Zealand
| | - William A Denny
- Auckland Cancer Society Research Centre, The University of Auckland , Private Bag 92019, Auckland 1142, New Zealand
| |
Collapse
|
47
|
Ueno S, Kimura T, Yamaga T, Kawada A, Ochiai T, Endou H, Sakurai H. Metformin enhances anti-tumor effect of L-type amino acid transporter 1 (LAT1) inhibitor. J Pharmacol Sci 2016; 131:110-7. [PMID: 27262901 DOI: 10.1016/j.jphs.2016.04.021] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 03/17/2016] [Accepted: 04/24/2016] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND In many cancer cells, L-type amino acid transporter 1 (LAT1) transports neutral amino acids with bulky side chain, which activate mammalian target of rapamycin (mTOR) to cause cell proliferation. An anti-diabetic drug, metformin, has been shown to activate AMP-activated protein kinase (AMPK), which leads to inhibition of mTOR. LAT1 inhibition in combination with metformin could result in more prominent suppression of mTOR activity. PURPOSE Anti-proliferative effect of a newly developed LAT1 specific inhibitor JPH203 in combination with metformin is evaluated in 2 head and neck cancer cell lines, Ca9-22 and HEp-2 cells and in nude mice inoculated with Ca9-22 cells. RESULTS AND DISCUSSION By MTT assay, 0.5 mM metformin inhibited proliferation of Ca9-22 cells to 70% of control. In the presence of 100 μM JPH203, proliferation of Ca9-22 cells was inhibited to 60% of control. By combining these 2 drugs, proliferation of Ca9-22 was significantly inhibited to 40% of control. However, this regimen was not very effective against HEp-2 cells. This combination also suppressed in vivo growth of Ca9-22 cells in a xenotransplant model. A combination of anti-LAT1 drug with metformin may be an effective anti-proliferative therapy for certain subsets of cancers.
Collapse
Affiliation(s)
- Seiji Ueno
- Department of Pharmacology and Toxicology, Kyorin University School of Medicine, Mitaka, Tokyo, 181-8611, Japan
| | - Toru Kimura
- Department of Pharmacology and Toxicology, Kyorin University School of Medicine, Mitaka, Tokyo, 181-8611, Japan
| | - Takashi Yamaga
- Department of Pharmacology and Toxicology, Kyorin University School of Medicine, Mitaka, Tokyo, 181-8611, Japan
| | | | | | - Hitoshi Endou
- Department of Pharmacology and Toxicology, Kyorin University School of Medicine, Mitaka, Tokyo, 181-8611, Japan; J-Pharma Co. Ltd., Yokohama, Kanagawa, 230-0046, Japan
| | - Hiroyuki Sakurai
- Department of Pharmacology and Toxicology, Kyorin University School of Medicine, Mitaka, Tokyo, 181-8611, Japan.
| |
Collapse
|
48
|
Inhibition of Large Neutral Amino Acid Transporters Suppresses Kynurenic Acid Production Via Inhibition of Kynurenine Uptake in Rodent Brain. Neurochem Res 2016; 41:2256-66. [DOI: 10.1007/s11064-016-1940-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 04/18/2016] [Accepted: 04/28/2016] [Indexed: 12/11/2022]
|
49
|
Augustyn E, Finke K, Zur AA, Hansen L, Heeren N, Chien HC, Lin L, Giacomini KM, Colas C, Schlessinger A, Thomas AA. LAT-1 activity of meta-substituted phenylalanine and tyrosine analogs. Bioorg Med Chem Lett 2016; 26:2616-2621. [PMID: 27106710 DOI: 10.1016/j.bmcl.2016.04.023] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 04/08/2016] [Accepted: 04/09/2016] [Indexed: 12/23/2022]
Abstract
The transporter protein Large-neutral Amino Acid Transporter 1 (LAT-1, SLC7A5) is responsible for transporting amino acids such as tyrosine and phenylalanine as well as thyroid hormones, and it has been exploited as a drug delivery mechanism. Recently its role in cancer has become increasingly appreciated, as it has been found to be up-regulated in many different tumor types, and its expression levels have been correlated with prognosis. Substitution at the meta position of aromatic amino acids has been reported to increase affinity for LAT-1; however, the SAR for this position has not previously been explored. Guided by newly refined computational models of the binding site, we hypothesized that groups capable of filling a hydrophobic pocket would increase binding to LAT-1, resulting in improved substrates relative to parent amino acid. Tyrosine and phenylalanine analogs substituted at the meta position with halogens, alkyl and aryl groups were synthesized and tested in cis-inhibition and trans-stimulation cell assays to determine activity. Contrary to our initial hypothesis we found that lipophilicity was correlated with diminished substrate activity and increased inhibition of the transporter. The synthesis and SAR of meta-substituted phenylalanine and tyrosine analogs is described.
Collapse
Affiliation(s)
- Evan Augustyn
- Department of Chemistry, University of Nebraska Kearney, Kearney, NE, 68849
| | - Karissa Finke
- Department of Chemistry, University of Nebraska Kearney, Kearney, NE, 68849
| | - Arik A Zur
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, CA 94158
| | - Logan Hansen
- Department of Chemistry, University of Nebraska Kearney, Kearney, NE, 68849
| | - Nathan Heeren
- Department of Chemistry, University of Nebraska Kearney, Kearney, NE, 68849
| | - Huan-Chieh Chien
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, CA 94158
| | - Lawrence Lin
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, CA 94158
| | - Kathleen M Giacomini
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, CA 94158
| | - Claire Colas
- Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Avner Schlessinger
- Department of Structural and Chemical Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Allen A Thomas
- Department of Chemistry, University of Nebraska Kearney, Kearney, NE, 68849
| |
Collapse
|
50
|
Hypoxia optimises tumour growth by controlling nutrient import and acidic metabolite export. Mol Aspects Med 2016; 47-48:3-14. [DOI: 10.1016/j.mam.2015.12.001] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|