1
|
Shimada H, Powell TL, Jansson T. Regulation of placental amino acid transport in health and disease. Acta Physiol (Oxf) 2024; 240:e14157. [PMID: 38711335 PMCID: PMC11162343 DOI: 10.1111/apha.14157] [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: 01/29/2024] [Revised: 04/19/2024] [Accepted: 04/23/2024] [Indexed: 05/08/2024]
Abstract
Abnormal fetal growth, i.e., intrauterine growth restriction (IUGR) or fetal growth restriction (FGR) and fetal overgrowth, is associated with increased perinatal morbidity and mortality and is strongly linked to the development of metabolic and cardiovascular disease in childhood and later in life. Emerging evidence suggests that changes in placental amino acid transport may contribute to abnormal fetal growth. This review is focused on amino acid transport in the human placenta, however, relevant animal models will be discussed to add mechanistic insights. At least 25 distinct amino acid transporters with different characteristics and substrate preferences have been identified in the human placenta. Of these, System A, transporting neutral nonessential amino acids, and System L, mediating the transport of essential amino acids, have been studied in some detail. Importantly, decreased placental Systems A and L transporter activity is strongly associated with IUGR and increased placental activity of these two amino acid transporters has been linked to fetal overgrowth in human pregnancy. An array of factors in the maternal circulation, including insulin, IGF-1, and adiponectin, and placental signaling pathways such as mTOR, have been identified as key regulators of placental Systems A and L. Studies using trophoblast-specific gene targeting in mice have provided compelling evidence that changes in placental Systems A and L are mechanistically linked to altered fetal growth. It is possible that targeting specific placental amino acid transporters or their upstream regulators represents a novel intervention to alleviate the short- and long-term consequences of abnormal fetal growth in the future.
Collapse
Affiliation(s)
- Hiroshi Shimada
- Department of Obstetrics and Gynecology University of Colorado, Anschutz Medical Campus, Aurora, CO, US
- Departments of Obstetrics & Gynecology, Sapporo Medical University, Sapporo, Japan
| | - Theresa L Powell
- Department of Obstetrics and Gynecology University of Colorado, Anschutz Medical Campus, Aurora, CO, US
- Department of Pediatrics, University of Colorado, Anschutz Medical Campus, Aurora, CO, US
| | - Thomas Jansson
- Department of Obstetrics and Gynecology University of Colorado, Anschutz Medical Campus, Aurora, CO, US
| |
Collapse
|
2
|
Lyda BR, Leary GP, Farnsworth J, Seaver B, Silvius D, Kavanaugh MP, Esslinger CS, Natale NR. Discovery and Synthesis of Hydroxy-l-Proline Blockers of the Neutral Amino Acid Transporters SLC1A4 (ASCT1) and SLC1A5 (ASCT2). Molecules 2024; 29:2330. [PMID: 38792190 PMCID: PMC11123841 DOI: 10.3390/molecules29102330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 04/29/2024] [Accepted: 05/03/2024] [Indexed: 05/26/2024] Open
Abstract
As a conformationally restricted amino acid, hydroxy-l-proline is a versatile scaffold for the synthesis of diverse multi-functionalized pyrrolidines for probing the ligand binding sites of biological targets. With the goal to develop new inhibitors of the widely expressed amino acid transporters SLC1A4 and SLC1A5 (also known as ASCT1 and ASCT2), we synthesized and functionally screened synthetic hydroxy-l-proline derivatives using electrophysiological and radiolabeled uptake methods against amino acid transporters from the SLC1, SLC7, and SLC38 solute carrier families. We have discovered a novel class of alkoxy hydroxy-pyrrolidine carboxylic acids (AHPCs) that act as selective high-affinity inhibitors of the SLC1 family neutral amino acid transporters SLC1A4 and SLC1A5. AHPCs were computationally docked into a homology model and assessed with respect to predicted molecular orientation and functional activity. The series of hydroxyproline analogs identified here represent promising new agents to pharmacologically modulate SLC1A4 and SLC1A5 amino acid exchangers which are implicated in numerous pathophysiological processes such as cancer and neurological diseases.
Collapse
Affiliation(s)
- Brent R. Lyda
- Division of Biological Sciences, University of Montana, 32 Campus Dr., Missoula, MT 59812, USA
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Dr., Missoula, MT 59812, USA (B.S.); (D.S.)
| | - Gregory P. Leary
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Dr., Missoula, MT 59812, USA (B.S.); (D.S.)
| | - Jill Farnsworth
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Dr., Missoula, MT 59812, USA (B.S.); (D.S.)
| | - Benjamin Seaver
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Dr., Missoula, MT 59812, USA (B.S.); (D.S.)
| | - Derek Silvius
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Dr., Missoula, MT 59812, USA (B.S.); (D.S.)
| | - Michael P. Kavanaugh
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Dr., Missoula, MT 59812, USA (B.S.); (D.S.)
| | - C. Sean Esslinger
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Dr., Missoula, MT 59812, USA (B.S.); (D.S.)
| | - Nicholas R. Natale
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Dr., Missoula, MT 59812, USA (B.S.); (D.S.)
- Medicinal Chemistry Graduate Program, University of Montana, 32 Campus Dr., Missoula, MT 59812, USA
| |
Collapse
|
3
|
Zhao Q, Liu Y, Chuo Y, Wang X, Jiao Y, Shi W, Bao Y. Cuscuta chinensis flavonoids alleviate ovarian damage in offspring female mice induced by BPA exposure during pregnancy by regulating the central carbon metabolism pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 275:116253. [PMID: 38537475 DOI: 10.1016/j.ecoenv.2024.116253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 03/18/2024] [Accepted: 03/20/2024] [Indexed: 04/12/2024]
Abstract
Pregnancy is a sensitive window period for bisphenol A (BPA) exposure. BPA can pass through the placenta and cause reproductive damage in offspring female mice. Even BPA that is not metabolized during lactation can be passed through milk. Cuscuta chinensis flavonoids (CCFs) can alleviate reproductive damage caused by BPA, but the mechanism of action is unclear. To investigate the potential mitigating impact of CCFs on ovarian damage resulting from BPA exposure during pregnancy, we administered BPA and CCFs to pregnant mice during the gestational period spanning from 0.5 to 17.5 days. Aseptic collection of serum and ovaries from female mice was conducted on postnatal day 21 (PND21). Serum hormone levels and tissue receptor levels were quantified utilizing ELISA and PCR, while ovaries underwent sequencing and analysis through transcriptomics and metabolomics techniques. Additionally, the assessment of ovarian oxidative stress levels was carried out as part of the comprehensive analysis. The results showed that CCFs administration mitigated the adverse effects induced by BPA exposure on ovarian index, hormone levels, receptor expression, and mRNA expression levels in female offspring mice. The joint analysis of transcriptome and metabolome revealed 48 enriched pathways in positive ion mode and 44 enriched pathways in negative ion mode. Among them, the central carbon metabolism pathway is significantly regulated by BPA and CCFs. The screened sequencing results were verified through qPCR and biochemical kits. In this study, CCFs may participate in the central carbon metabolism pathway by reducing the expression of Kit proto-oncogene (Kit), hexokinase 1 gene (Hk1) and pyruvate kinase M (Pkm) mRNA and increasing the expression of h-ras proto-oncogene (Hras), sirtuin 3 (Sirt3), sirtuin 6 (Sirt6) and TP53 induced glycolysis regulatory phosphatase gene (Tigar) mRNA, thereby resisting the effects of BPA on the body. At the same time, the metabolic levels of D-Fructose 1,6-bisphosphate and L-Asparagine tend to be stable. Moreover, CCFs demonstrated a capacity to diminish the BPA-induced escalation in reactive oxygen species (ROS) and malondialdehyde (MDA). Simultaneously, it exhibited the ability to elevate levels of glutathione (GSH) and catalase (CAT), thereby effectively preventing peroxidation. In summary, CCFs alleviate BPA-induced ovarian damage in offspring female mice by regulating the central carbon metabolism pathway. This study will improve the information on BPA reproductive damage antagonist drugs and provide a theoretical basis for protecting animal reproductive health.
Collapse
Affiliation(s)
- Qianhui Zhao
- College of Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China
| | - Ying Liu
- College of Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China
| | - Yanan Chuo
- College of Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China
| | - Xiao Wang
- College of Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China
| | - Yulan Jiao
- College of Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China; Hebei Veterinary Biotenology Innovation Center, Baoding 071001, China; Ruipu (Baoding) Biological Pharmaceutical Co., Ltd., Baoding 071000, China
| | - Wanyu Shi
- College of Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China; Hebei Veterinary Biotenology Innovation Center, Baoding 071001, China.
| | - Yongzhan Bao
- College of Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China.
| |
Collapse
|
4
|
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
|
5
|
Mathew M, Nguyen NT, Bhutia YD, Sivaprakasam S, Ganapathy V. Metabolic Signature of Warburg Effect in Cancer: An Effective and Obligatory Interplay between Nutrient Transporters and Catabolic/Anabolic Pathways to Promote Tumor Growth. Cancers (Basel) 2024; 16:504. [PMID: 38339256 PMCID: PMC10854907 DOI: 10.3390/cancers16030504] [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: 12/17/2023] [Revised: 01/19/2024] [Accepted: 01/22/2024] [Indexed: 02/12/2024] Open
Abstract
Aerobic glycolysis in cancer cells, originally observed by Warburg 100 years ago, which involves the production of lactate as the end product of glucose breakdown even in the presence of adequate oxygen, is the foundation for the current interest in the cancer-cell-specific reprograming of metabolic pathways. The renewed interest in cancer cell metabolism has now gone well beyond the original Warburg effect related to glycolysis to other metabolic pathways that include amino acid metabolism, one-carbon metabolism, the pentose phosphate pathway, nucleotide synthesis, antioxidant machinery, etc. Since glucose and amino acids constitute the primary nutrients that fuel the altered metabolic pathways in cancer cells, the transporters that mediate the transfer of these nutrients and their metabolites not only across the plasma membrane but also across the mitochondrial and lysosomal membranes have become an integral component of the expansion of the Warburg effect. In this review, we focus on the interplay between these transporters and metabolic pathways that facilitates metabolic reprogramming, which has become a hallmark of cancer cells. The beneficial outcome of this recent understanding of the unique metabolic signature surrounding the Warburg effect is the identification of novel drug targets for the development of a new generation of therapeutics to treat cancer.
Collapse
Affiliation(s)
| | | | | | | | - Vadivel Ganapathy
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (M.M.); (N.T.N.); (Y.D.B.); (S.S.)
| |
Collapse
|
6
|
Chen P, Jiang Y, Liang J, Cai J, Zhuo Y, Fan H, Yuan R, Cheng S, Zhang Y. SLC1A5 is a novel biomarker associated with ferroptosis and the tumor microenvironment: a pancancer analysis. Aging (Albany NY) 2023; 15:7451-7475. [PMID: 37566748 PMCID: PMC10457057 DOI: 10.18632/aging.204911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 07/06/2023] [Indexed: 08/13/2023]
Abstract
Solute carrier family 1 member 5 (SLC1A5) is a member of the solute carrier (SLC) superfamily of transporters and plays an important role in tumors as a key transporter of glutamine into cells. However, the relationship between SLC1A5, which is involved in immune regulation, and immune cell infiltration in the tumor microenvironment has not been elucidated, and the relationship between SLC1A5 and ferroptosis is rarely reported. Therefore, we comprehensively analyzed the expression level of SLC1A5 across cancers and compared it with that in normal tissues. Then, the relationship between SLC1A5 expression and the tumor immune microenvironment was analyzed by single-cell analysis, gene set enrichment analysis (GSEA), and Tumor Immune Estimation Resource (TIMER). Next, the correlations of the SLC1A5 expression level with immunotherapy response, immunomodulator expression, tumor mutation burden (TMB) and microsatellite instability (MSI) were evaluated. Finally, in vitro experiments verified that SLC1A5 participates in ferroptosis of glioma cells to regulate tumor progression. Our results indicated that SLC1A5 is aberrantly expressed in most cancer types and closely associated with prognosis. The GSEA results showed that SLC1A5 is involved in immune activation processes and closely related to the infiltration levels of different immune cells in different cancer types. Upon further investigation, we found that SLC1A5 is a suppressor of ferroptosis in glioma, and SLC1A5 knockdown inhibited the proliferation and migration of glioma cells in vitro. In conclusion, we conducted a pancancer analysis of SLC1A5, demonstrated its role as a prognostic biomarker in cancer patients and explored its potential biological functions.
Collapse
Affiliation(s)
- Peng Chen
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
- Department of Medical, Nanchang University, Nanchang 330006, Jiangxi, P.R. China
| | - YongAn Jiang
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
- Department of Medical, Nanchang University, Nanchang 330006, Jiangxi, P.R. China
| | - JiaWei Liang
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
- Department of Medical, Nanchang University, Nanchang 330006, Jiangxi, P.R. China
| | - JiaHong Cai
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
- Department of Medical, Nanchang University, Nanchang 330006, Jiangxi, P.R. China
| | - Yi Zhuo
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
- Department of Medical, Nanchang University, Nanchang 330006, Jiangxi, P.R. China
| | - HengYi Fan
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
| | - RaoRao Yuan
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
| | - ShiQi Cheng
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
| | - Yan Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
| |
Collapse
|
7
|
Paladini A, Vallejo R, Guerrero M, Pasqualucci A, Peppin JF, Pergolizzi J, Varrassi G. Answering Big Questions in Pain Medicine. Cureus 2023; 15:e43561. [PMID: 37719539 PMCID: PMC10502917 DOI: 10.7759/cureus.43561] [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: 07/17/2023] [Accepted: 08/16/2023] [Indexed: 09/19/2023] Open
Abstract
The future of pain medicine is marked by many questions. What can other nations around the world learn from the opioid crisis that is still affecting the United States? The American opioid experience was mischaracterized and wrongly described, and its causes were misdiagnosed from the outset, leading to its mismanagement and the abandonment of many chronic pain patients to their suffering. There are a few new drugs in the analgesic armamentarium. What new targets do we have in pain medicine? There are many breakthroughs, discoveries, and potential new targets that could add to our analgesic prescribing choices. These include sigma receptors, d-amino acid oxidase, endoplasmic reticulum stress receptors, histone deacetylase, and others. Neuromodulation had been used with varying degrees of success for years, but with a simplistic approach based on the gate theory of pain. Despite our familiarity with neuromodulation and spinal cord stimulators, neuromodulation research indicates that the activation of glial cells may activate the immune system and enhance analgesia. Neuromodulation studies have concentrated on how electricity affects neuronal activity rather than how electrical activity could reduce pain. There are still more frontiers in our battle against pain and some promising avenues for treatments. This narrative review will try to summarize what can be done from the perspective of recent technological and pharmacological developments.
Collapse
Affiliation(s)
- Antonella Paladini
- Department of Life, Health & Environmental Sciences (MESVA), University of L'Aquila, L'Aquila, ITA
| | - Ricardo Vallejo
- Department of Research, Millennium Pain Center, Bloomington, USA
| | - Marixa Guerrero
- Department of Pain Medicine/ Pain Management, Clínica del Country, Bogota, COL
| | - Alberto Pasqualucci
- Department of Anesthesia and Critical Care, University of Perugia, Perugia, ITA
| | - John F Peppin
- Department of Osteopathic Medicine, Marian University, Indianapolis, USA
| | - Joseph Pergolizzi
- Department of Anesthesiology, Pain Medicine, and Critical Care Medicine, Nema Research, Naples, USA
| | | |
Collapse
|
8
|
Ai C, Sun X, Xiao S, Guo L, Shang M, Shi D, Meng D, Zhao Y, Wang X, Li J. CAFs targeted ultrasound-responsive nanodroplets loaded V9302 and GLULsiRNA to inhibit melanoma growth via glutamine metabolic reprogramming and tumor microenvironment remodeling. J Nanobiotechnology 2023; 21:214. [PMID: 37420266 DOI: 10.1186/s12951-023-01979-z] [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: 01/27/2023] [Accepted: 07/02/2023] [Indexed: 07/09/2023] Open
Abstract
Despite rapid advances in metabolic therapies over the past decade, their efficacy in melanoma has been modest, largely due to the interaction between cancer-associated fibroblasts (CAFs) and cancer cells to promote cancer growth. Altering the tumor microenvironment (TME) is challenging and elusive. CAFs is critical for glutamine deprivation survival in melanoma. In this research, we assembled a CAFs-targeted, controlled-release nanodroplets for the combined delivery of the amino acid transporter ASCT2 (SLC1A5) inhibitor V9302 and GLULsiRNA (siGLUL). The application of ultrasound-targeted microbubble disruption (UTMD) allows for rapid release of V9302 and siGLUL, jointly breaking the glutamine metabolism interaction between CAFs and cancer cells on one hand, on the other hand, blocking activated CAFs and reducing the expression of extracellular matrix (ECM) to facilitate drug penetration. In addition, ultrasound stimulation made siGLUL more accessible to tumor cells and CAFs, downregulating GLUL expression in both cell types. FH-V9302-siGLUL-NDs also serve as contrast-enhanced ultrasound imaging agents for tumor imaging. Our study developed and reported FH-NDs as nanocarriers for V9302 and siGLUL, demonstrating that FH-V9302-siGLUL-NDs have potential bright future applications for integrated diagnostic therapy. Graphical Abstract.
Collapse
Affiliation(s)
- Chen Ai
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Xiao Sun
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Shan Xiao
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Lu Guo
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Mengmeng Shang
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Dandan Shi
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Dong Meng
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Yading Zhao
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Xiaoxuan Wang
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Jie Li
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China.
| |
Collapse
|
9
|
Li S, Pei L, Zhou Q, Fu Z, Zhang L, Liu P, Yan N, Xi S. SLC1A5 regulates cell proliferation and self-renewal through β-catenin pathway mediated by redox signaling in arsenic-treated uroepithelial cells. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 262:115204. [PMID: 37393816 DOI: 10.1016/j.ecoenv.2023.115204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 06/13/2023] [Accepted: 06/27/2023] [Indexed: 07/04/2023]
Abstract
Arsenic exposure increases the risk of bladder cancer in humans, but its underlying mechanisms remain elusive. The alanine, serine, cysteine-preferring transporter 2 (ASCT2, SLC1A5) is frequently overexpressed in cancer cells. The aim of this study was to evaluate the effects of arsenic on SLC1A5, and to determine the role of SLC1A5 in the proliferation and self-renewal of uroepithelial cells. F344 rats were exposed to 87 mg/L NaAsO2 or 200 mg/L DMAV for 12 weeks. The SV-40 immortalized human uroepithelial (SV-HUC-1) cells were cultured in medium containing 0.5 μM NaAsO2 for 40 weeks. Arsenic increased the expression levels of SLC1A5 and β-catenin both in vivo and in vitro. SLC1A5 promoted cell proliferation and self-renewal by activating β-catenin, which in turn was dependent on maintaining GSH/ROS homeostasis. Our results suggest that SLC1A5 is a potential therapeutic target for arsenic-induced proliferation and self-renewal of uroepithelial cells.
Collapse
Affiliation(s)
- Sihao Li
- Health Sciences Institute, China Medical University, Shenyang, Liaoning Province, China
| | - Liang Pei
- Department of Pediatric, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Qing Zhou
- Department of Environmental and Occupational Health, School of Public Health, China Medical University, Shenyang, Liaoning Province, China
| | - Zhushan Fu
- Department of Environmental and Occupational Health, School of Public Health, China Medical University, Shenyang, Liaoning Province, China
| | - Lei Zhang
- Department of Environmental and Occupational Health, School of Public Health, China Medical University, Shenyang, Liaoning Province, China
| | - Pinya Liu
- Department of Environmental and Occupational Health, School of Public Health, China Medical University, Shenyang, Liaoning Province, China
| | - Nan Yan
- School of Medical Applied Technology, Shenyang Medical College, Shenyang, Liaoning Province, China
| | - Shuhua Xi
- Health Sciences Institute, China Medical University, Shenyang, Liaoning Province, China; Department of Environmental and Occupational Health, School of Public Health, China Medical University, Shenyang, Liaoning Province, China; School of Medical Applied Technology, Shenyang Medical College, Shenyang, Liaoning Province, China.
| |
Collapse
|
10
|
Fu S, Xu S, Zhang S. The role of amino acid metabolism alterations in pancreatic cancer: From mechanism to application. Biochim Biophys Acta Rev Cancer 2023; 1878:188893. [PMID: 37015314 DOI: 10.1016/j.bbcan.2023.188893] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 03/13/2023] [Accepted: 03/30/2023] [Indexed: 04/05/2023]
Abstract
The incidence of pancreatic cancer is increasing in both developed and developing Nations. In recent years, various research evidence suggested that reprogrammed metabolism may play a key role in pancreatic cancer tumorigenesis and development. Therefore, it has great potential as a diagnostic, prognostic and therapeutic target. Amino acid metabolism is deregulated in pancreatic cancer, and changes in amino acid metabolism can affect cancer cell status, systemic metabolism in malignant tumor patients and mistakenly involved in different biological processes including stemness, proliferation and growth, invasion and migration, redox state maintenance, autophagy, apoptosis and even tumor microenvironment interaction. Generally, the above effects are achieved through two pathways, energy metabolism and signal transduction. This review aims to highlight the current research progress on the abnormal alterations of amino acids metabolism in pancreatic cancer, how they affect tumorigenesis and development of pancreatic cancer and the application prospects of them as diagnostic, prognostic and therapeutic targets.
Collapse
Affiliation(s)
- Shenao Fu
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, Hunan 410013, PR China; Clinical Medicine Eight-Year Program, Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, PR China
| | - Shaokang Xu
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, Hunan 410013, PR China; Clinical Medicine Eight-Year Program, Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, PR China
| | - Shubing Zhang
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, Hunan 410013, PR China.
| |
Collapse
|
11
|
Jiang Q, Sherlock DN, Guyader J, Loor JJ. Abundance of Amino Acid Transporters and mTOR Pathway Components in the Gastrointestinal Tract of Lactating Holstein Cows. Animals (Basel) 2023; 13:ani13071189. [PMID: 37048445 PMCID: PMC10093496 DOI: 10.3390/ani13071189] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/25/2023] [Accepted: 03/27/2023] [Indexed: 03/31/2023] Open
Abstract
Data from non-ruminants indicate that amino acid (AA) transport into cells can regulate mTOR pathway activity and protein synthesis. Whether mTOR is expressed in the ruminant gastrointestinal tract (GIT) and how it may be related to AA transporters and the AA concentrations in the tissue is unknown. Ruminal papillae and the epithelia of the duodenum, jejunum, and ileum collected at slaughter from eight clinically healthy Holstein in mid-lactation were used. Metabolites and RNA were extracted from tissue for liquid chromatography–mass spectrometry and RT-qPCR analysis. The glycine and asparagine concentrations in the rumen were greater than those in the intestine (p < 0.05), but the concentrations of other AAs were greater in the small intestine than those in the rumen. Among the 20 AAs identified, the concentrations of glutamate, alanine, and glycine were the greatest. The mRNA abundances of AKT1 and MTOR were greater in the small intestine than those in the rumen (p < 0.05). Similarly, the SLC1A1, SLC6A6, SLC7A8, SLC38A1, SLC38A7, and SLC43A2 mRNA abundances were greater (p < 0.05) in the small intestine than those in the rumen. The mRNA abundances of SLC1A5, SLC3A2, and SLC7A5 were greater in the rumen than those in the small intestine (p < 0.05). Overall, the present study provides fundamental data on the relationship between mTOR pathway components and the transport of AAs in different sections of the gastrointestinal tract.
Collapse
Affiliation(s)
- Qianming Jiang
- Department of Animal Sciences, University of Illinois, Urbana, IL 61801, USA
| | | | - Jessie Guyader
- Evonik Operations GmbH, Hanau-Wolfgang, 63457 Essen, Germany
| | - Juan J. Loor
- Department of Animal Sciences, University of Illinois, Urbana, IL 61801, USA
- Division of Nutritional Sciences, University of Illinois, Urbana, IL 61801, USA
- Correspondence:
| |
Collapse
|
12
|
Liu Y, Liu T, Zhou Y, Li W, Wang M, Song N, Zhang W, Jiang J, Yuan S, Ding J, Hu G, Lu M. Impeding the combination of astrocytic ASCT2 and NLRP3 by talniflumate alleviates neuroinflammation in experimental models of Parkinson's disease. Acta Pharm Sin B 2023; 13:662-677. [PMID: 36873178 PMCID: PMC9978855 DOI: 10.1016/j.apsb.2022.07.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/02/2022] [Accepted: 06/16/2022] [Indexed: 11/27/2022] Open
Abstract
Alanine-serine-cysteine transporter 2 (ASCT2) is reported to participate in the progression of tumors and metabolic diseases. It is also considered to play a crucial role in the glutamate-glutamine shuttle of neuroglial network. However, it remains unclear the involvement of ASCT2 in neurological diseases such as Parkinson's disease (PD). In this study, we demonstrated that high expression of ASCT2 in the plasma samples of PD patients and the midbrain of MPTP mouse models is positively correlated with dyskinesia. We further illustrated that ASCT2 expressed in astrocytes rather than neurons significantly upregulated in response to either MPP+ or LPS/ATP challenge. Genetic ablation of astrocytic ASCT2 alleviated the neuroinflammation and rescued dopaminergic (DA) neuron damage in PD models in vitro and in vivo. Notably, the binding of ASCT2 to NLRP3 aggravates astrocytic inflammasome-triggered neuroinflammation. Then a panel of 2513 FDA-approved drugs were performed via virtual molecular screening based on the target ASCT2 and we succeed in getting the drug talniflumate. It is validated talniflumate impedes astrocytic inflammation and prevents degeneration of DA neurons in PD models. Collectively, these findings reveal the role of astrocytic ASCT2 in the pathogenesis of PD, broaden the therapeutic strategy and provide a promising candidate drug for PD treatment.
Collapse
Affiliation(s)
- Yang Liu
- Department of Pharmacology, School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Ting Liu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing 211166, China
| | - Yuanzhang Zhou
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing 211166, China
| | - Wenjie Li
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing 211166, China
| | - Min Wang
- Department of Geriatrics, Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Nanshan Song
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing 211166, China
| | - Wenbin Zhang
- Department of Neurosurgery, Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Jingwei Jiang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 211198, China
| | - Shengtao Yuan
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 211198, China
| | - Jianhua Ding
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing 211166, China
| | - Gang Hu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing 211166, China.,Department of Pharmacology, School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Ming Lu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing 211166, China
| |
Collapse
|
13
|
Circular RNA circ-AGFG1 contributes to esophageal squamous cell carcinoma progression and glutamine catabolism by targeting microRNA-497-5p/solute carrier family 1 member 5 axis. Anticancer Drugs 2023; 34:195-206. [PMID: 36206112 DOI: 10.1097/cad.0000000000001400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Circular RNAs (circRNAs) have been shown to play important regulatory roles in human malignancies. However, the role of circRNA ArfGAP with FG repeats 1 (circ-AGFG1) in esophageal squamous cell carcinoma (ESCC) progression and its associated mechanism are still largely undefined. Cell proliferation was analyzed by 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay and 5-ethynyl-2'-deoxyuridine assay. Cell apoptosis was assessed by flow cytometry analysis. Transwell assay and wound healing assay were used to analyze cell invasion and migration abilities. The uptake of glutamine and the production of α-ketoglutarate and glutamate were analyzed using Glutamine Determination Kit, α-ketoglutarate Assay Kit and Glutamate Determination Kit. A xenograft tumor model was used to analyze the biological role of circ-AGFG1 in vivo . The interaction between microRNA-497-5p (miR-497-5p) and circ-AGFG1 or solute carrier family 1 member 5 (SLC1A5) was verified by dual-luciferase reporter assay. Circ-AGFG1 expression was upregulated in ESCC tissues and cell lines. Circ-AGFG1 silencing suppressed the proliferation, migration, invasion and glutaminolysis and triggered the apoptosis of ESCC cells. Circ-AGFG1 knockdown significantly slowed down tumor growth in vivo . Circ-AGFG1 acted as a sponge for miR-497-5p, and miR-497-5p interacted with the 3' untranslated region (3'UTR) of SLC1A5. miR-497-5p silencing largely abolished circ-AGFG1 silencing-induced effects in ESCC cells. miR-497-5p overexpression-mediated influences in ESCC cells were largely reversed by the addition of SLC1A5 expressing plasmid. Circ-AGFG1 could upregulate SLC1A5 expression by sponging miR-497-5p. In summary, circ-AGFG1 acted as an oncogene to elevate the malignant potential and promote the glutamine catabolism of ESCC cells by targeting the miR-497-5p/SLC1A5 axis.
Collapse
|
14
|
Redeker KEM, Jensen O, Gebauer L, Meyer-Tönnies MJ, Brockmöller J. Atypical Substrates of the Organic Cation Transporter 1. Biomolecules 2022; 12:1664. [PMID: 36359014 PMCID: PMC9687798 DOI: 10.3390/biom12111664] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 10/27/2022] [Accepted: 11/02/2022] [Indexed: 10/29/2023] Open
Abstract
The human organic cation transporter 1 (OCT1) is expressed in the liver and mediates hepatocellular uptake of organic cations. However, some studies have indicated that OCT1 could transport neutral or even anionic substrates. This capability is interesting concerning protein-substrate interactions and the clinical relevance of OCT1. To better understand the transport of neutral, anionic, or zwitterionic substrates, we used HEK293 cells overexpressing wild-type OCT1 and a variant in which we changed the putative substrate binding site (aspartate474) to a neutral amino acid. The uncharged drugs trimethoprim, lamivudine, and emtricitabine were good substrates of hOCT1. However, the uncharged drugs zalcitabine and lamotrigine, and the anionic levofloxacin, and prostaglandins E2 and F2α, were transported with lower activity. Finally, we could detect only extremely weak transport rates of acyclovir, ganciclovir, and stachydrine. Deleting aspartate474 had a similar transport-lowering effect on anionic substrates as on cationic substrates, indicating that aspartate474 might be relevant for intra-protein, rather than substrate-protein, interactions. Cellular uptake of the atypical substrates by the naturally occurring frequent variants OCT1*2 (methionine420del) and OCT1*3 (arginine61cysteine) was similarly reduced, as it is known for typical organic cations. Thus, to comprehensively understand the substrate spectrum and transport mechanisms of OCT1, one should also look at organic anions.
Collapse
Affiliation(s)
- Kyra-Elisa Maria Redeker
- Institute of Clinical Pharmacology, University Medical Centre Göttingen, 37075 Göttingen, Germany
| | - Ole Jensen
- Institute of Clinical Pharmacology, University Medical Centre Göttingen, 37075 Göttingen, Germany
| | - Lukas Gebauer
- Institute of Clinical Pharmacology, University Medical Centre Göttingen, 37075 Göttingen, Germany
| | - Marleen Julia Meyer-Tönnies
- Department of General Pharmacology, Institute of Pharmacology, Centre of Drug Absorption and Transport (C-DAT), University Medical Centre Greifswald, 17487 Greifswald, Germany
| | - Jürgen Brockmöller
- Institute of Clinical Pharmacology, University Medical Centre Göttingen, 37075 Göttingen, Germany
| |
Collapse
|
15
|
Li Y, Feng R, Yu X, Li L, Liu Y, Zhang R, Chen X, Zhao Y, Liu Z. SLC35E2 promoter mutation as a prognostic marker of esophageal squamous cell carcinoma. Life Sci 2022; 296:120447. [PMID: 35247439 DOI: 10.1016/j.lfs.2022.120447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 01/29/2022] [Accepted: 02/26/2022] [Indexed: 12/24/2022]
Abstract
AIMS Esophageal squamous cell carcinoma (ESCC) is one of the deadliest digestive tract cancer with poor prognosis. In our previous comprehensive genomics study, we identified that hotspot mutations in the solute carrier family 35 member E2 (SLC35E2) promoter region was significantly associated with worse prognosis in patients with ESCC. However, the biological function and molecular mechanism of SLC35E2 remains unclear. This study was to investigate the malignant function and mechanism of SLC35E2 in ESCC. MAIN METHODS Western blotting and qRT-PCR were used to assess the expression of SLC35E2 in ESCC cell lines. Luciferase assay and chromatin immunoprecipitation (ChIP) assay were used to assess the transcriptional inhibition of KLF4. Incucyte cell proliferation assay, colony formation assay and subcutaneous tumor formation in nude mice were used to assess the malignant function of SLC35E2. KEY FINDINGS SLC35E2 can promote ESCC cell proliferation in vitro and in vivo. Krüppel-like factor 4 (KLF4), a transcriptional repressor in ESCC, binds to the SLC35E2 promoter and represses the expression of SLC35E2. The transcriptional suppression of KLF4 can be blocked by the mutation at -118 site of the SLC35E2 promoter. Besides, the accumulation of SLC35E2 expression contributes to the malignant phenotype of ESCC. SIGNIFICANCE These results indicate that SLC35E2 may be used as a biomarker for prognosis as well as a therapeutic target for patients with ESCC.
Collapse
Affiliation(s)
- Yang Li
- State Key Laboratory of Molecular Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Riyue Feng
- State Key Laboratory of Molecular Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Xiao Yu
- State Key Laboratory of Molecular Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Lei Li
- State Key Laboratory of Molecular Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yuhao Liu
- State Key Laboratory of Molecular Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Ruixiang Zhang
- State Key Laboratory of Molecular Oncology, Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xiankai Chen
- State Key Laboratory of Molecular Oncology, Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yahui Zhao
- State Key Laboratory of Molecular Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Zhihua Liu
- State Key Laboratory of Molecular Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| |
Collapse
|
16
|
Glutamine-Derived Aspartate Biosynthesis in Cancer Cells: Role of Mitochondrial Transporters and New Therapeutic Perspectives. Cancers (Basel) 2022; 14:cancers14010245. [PMID: 35008407 PMCID: PMC8750728 DOI: 10.3390/cancers14010245] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 12/30/2021] [Accepted: 01/01/2022] [Indexed: 12/20/2022] Open
Abstract
Simple Summary In recent years, aspartate has been increasingly acknowledged as a critical player in the metabolism of cancer cells which use this metabolite for nucleotide and protein synthesis and for redox homeostasis. Most intracellular aspartate derives from the mitochondrial catabolism of glutamine. To date at least four mitochondrial transporters have been involved in this metabolic pathway. Their involvement appears to be cancer type-specific and dependent on glutamine availability. Targeting these mitochondrial transporters may represent a new attractive strategy to fight cancer. The aim of this review is to dissect the role of each of these transporters in relation to the type of cancer and the availability of nutrients in the tumoral microenvironment. Abstract Aspartate has a central role in cancer cell metabolism. Aspartate cytosolic availability is crucial for protein and nucleotide biosynthesis as well as for redox homeostasis. Since tumor cells display poor aspartate uptake from the external environment, most of the cellular pool of aspartate derives from mitochondrial catabolism of glutamine. At least four transporters are involved in this metabolic pathway: the glutamine (SLC1A5_var), the aspartate/glutamate (AGC), the aspartate/phosphate (uncoupling protein 2, UCP2), and the glutamate (GC) carriers, the last three belonging to the mitochondrial carrier family (MCF). The loss of one of these transporters causes a paucity of cytosolic aspartate and an arrest of cell proliferation in many different cancer types. The aim of this review is to clarify why different cancers have varying dependencies on metabolite transporters to support cytosolic glutamine-derived aspartate availability. Dissecting the precise metabolic routes that glutamine undergoes in specific tumor types is of upmost importance as it promises to unveil the best metabolic target for therapeutic intervention.
Collapse
|
17
|
Scalise M, Console L, Cosco J, Pochini L, Galluccio M, Indiveri C. ASCT1 and ASCT2: Brother and Sister? SLAS DISCOVERY 2021; 26:1148-1163. [PMID: 34269129 DOI: 10.1177/24725552211030288] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The SLC1 family includes seven members divided into two groups, namely, EAATs and ASCTs, that share similar 3D architecture; the first one includes high-affinity glutamate transporters, and the second one includes SLC1A4 and SLC1A5, known as ASCT1 and ASCT2, respectively, responsible for the traffic of neutral amino acids across the cell plasma membrane. The physiological role of ASCT1 and ASCT2 has been investigated over the years, revealing different properties in terms of substrate specificities, affinities, and regulation by physiological effectors and posttranslational modifications. Furthermore, ASCT1 and ASCT2 are involved in pathological conditions, such as neurodegenerative disorders and cancer. This has driven research in the pharmaceutical field aimed to find drugs able to target the two proteins.This review focuses on structural, functional, and regulatory aspects of ASCT1 and ASCT2, highlighting similarities and differences.
Collapse
Affiliation(s)
- Mariafrancesca Scalise
- Department DiBEST (Biologia, Ecologia e Scienze della Terra), Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Arcavacata di Rende, Italy
| | - Lara Console
- Department DiBEST (Biologia, Ecologia e Scienze della Terra), Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Arcavacata di Rende, Italy
| | - Jessica Cosco
- Department DiBEST (Biologia, Ecologia e Scienze della Terra), Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Arcavacata di Rende, Italy
| | - Lorena Pochini
- Department DiBEST (Biologia, Ecologia e Scienze della Terra), Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Arcavacata di Rende, Italy
| | - Michele Galluccio
- Department DiBEST (Biologia, Ecologia e Scienze della Terra), Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Arcavacata di Rende, Italy
| | - Cesare Indiveri
- Department DiBEST (Biologia, Ecologia e Scienze della Terra), Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Arcavacata di Rende, Italy.,CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnology (IBIOM), Bari, Italy
| |
Collapse
|
18
|
Guo H, Gao J, Qian Y, Wang H, Liu J, Peng Q, Zhou Y, Wang K. miR-125b-5p inhibits cell proliferation by targeting ASCT2 and regulating the PI3K/AKT/mTOR pathway in an LPS-induced intestinal mucosa cell injury model. Exp Ther Med 2021; 22:838. [PMID: 34149884 PMCID: PMC8210225 DOI: 10.3892/etm.2021.10270] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 05/17/2021] [Indexed: 12/11/2022] Open
Abstract
Intestinal barrier injury is an important cause of death in patients with acquired immune deficiency syndrome (AIDS). Therefore, it is of great significance to identify a therapeutic target for intestinal barrier injury to delay the progression of AIDS. microRNA (miRNA/miR)-125b-5p has an extensive role in cancer and controlling intestinal epithelial barrier function, but its role in human immunodeficiency virus-related intestinal mucosal damage remains unknown. The present study was designed to explore the effects of miR-125b-5p on lipopolysaccharide (LPS)-induced intestinal mucosal injury and the underlying mechanism. The expression of miR-125b-5p and ASCT2 mRNA was detected in colon biopsy samples of 10 patients with AIDS and 10 control healthy subjects. Human intestinal embryonic mucosa cells (CCC-HIE-2) were used to establish an LPS-induced intestinal mucosa cell injury model in vitro. Cell proliferation and apoptosis were determined by MTT assays and flow cytometry, respectively. miR-125b-5p levels and ASCT2 mRNA and protein expression levels in the LPS-induced intestinal mucosa cell injury model were detected by reverse transcription-quantitative PCR (RT-qPCR) and western blotting. The interaction between miR-125b-5p and ASCT2 was analyzed using a dual luciferase reporter assay. The results demonstrated that miR-125b-5p levels were increased and ASCT2 mRNA expression levels were decreased in colon samples from patients with AIDS and in LPS-induced intestinal mucosa cells. In the LPS-induced intestinal mucosa cell injury model, transfection with miR-125b-5p mimic inhibited cell proliferation and promoted cell apoptosis, while transfection with a miR-125b-5p inhibitor increased cell proliferation and attenuated cell apoptosis. Furthermore, miR-125b-5p mimic transfection resulted in a decrease of ASCT2 mRNA and protein expression, whereas the inhibitor increased ASCT2 mRNA and protein expression. Dual luciferase reporter assays suggested that ASCT2 was a direct target of miR-125b-5p, and its restoration weakened the effect of miR-125b-5p on LPS-induced intestinal mucosa cell injury. Transfection with the miR-125b-5p mimic also exhibited a suppressive effect on the PI3K/AKT/mTOR pathway in the LPS-induced intestinal mucosal cell injury model. Overall, the present study indicated that miR-125b-5p accelerated LPS-induced intestinal mucosa cell injury by targeting ASCT2 and upregulating the PI3K/AKT/mTOR pathway. The current findings may provide novel targets for the treatment of intestinal barrier injury in patients with AIDS.
Collapse
Affiliation(s)
- Huiming Guo
- Department of Gynaecology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China.,NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Jianyuan Gao
- NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Yuan Qian
- NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Huawei Wang
- NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Jiang Liu
- NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Qingyan Peng
- NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, Yunnan 650500, P.R. China.,The Scientific Research Laboratory Center, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Yong Zhou
- NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, Yunnan 650500, P.R. China.,The Scientific Research Laboratory Center, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Kunhua Wang
- NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| |
Collapse
|
19
|
Teixeira E, Silva C, Martel F. The role of the glutamine transporter ASCT2 in antineoplastic therapy. Cancer Chemother Pharmacol 2021; 87:447-464. [PMID: 33464409 DOI: 10.1007/s00280-020-04218-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 12/14/2020] [Indexed: 12/12/2022]
Abstract
Cancer cells are metabolically reprogrammed to support their high rates of proliferation, continuous growth, survival, invasion, metastasis, and resistance to cancer treatments. Among changes in cancer cell bioenergetics, the role of glutamine metabolism has been receiving increasing attention. Increased glutaminolysis in cancer cells is associated with increased expression of membrane transporters that mediate the cellular uptake of glutamine. ASCT2 (Alanine, Serine, Cysteine Transporter 2) is a Na+-dependent transmembrane transporter overexpressed in cancer cells and considered to be the primary transporter for glutamine in these cells. The possibility of inhibiting ASCT2 for antineoplastic therapy is currently under investigation. In this article, we will present the pharmacological agents currently known to act on ASCT2, which have been attracting attention in antineoplastic therapy research. We will also address the impact of ASCT2 inhibition on the prognosis of some cancers. We conclude that ASCT2 inhibition and combination of ASCT2 inhibitors with other anti-tumor therapies may be a promising antineoplastic strategy. However, more research is needed in this area.
Collapse
Affiliation(s)
- Estefânia Teixeira
- Department of Biomedicine, Unit of Biochemistry, Faculty of Medicine, University of Porto, Al Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| | - Cláudia Silva
- Department of Biomedicine, Unit of Biochemistry, Faculty of Medicine, University of Porto, Al Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
- Instituto de Investigação E Inovação Em Saúde (i3S), University of Porto, Porto, Portugal
| | - Fátima Martel
- Department of Biomedicine, Unit of Biochemistry, Faculty of Medicine, University of Porto, Al Prof. Hernâni Monteiro, 4200-319, Porto, Portugal.
- Instituto de Investigação E Inovação Em Saúde (i3S), University of Porto, Porto, Portugal.
| |
Collapse
|
20
|
Li S, Zhou Q, Liu W, Fu Z, Zhao H, Xi S. Targeting SLC1A5 blocks cell proliferation through inhibition of mTORC1 in arsenite-treated human uroepithelial cells. Toxicol Lett 2021; 345:1-11. [PMID: 33781819 DOI: 10.1016/j.toxlet.2021.03.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 03/14/2021] [Accepted: 03/24/2021] [Indexed: 10/21/2022]
Abstract
Arsenic is an environmental contaminant, which is widely distributed in soil, air, and water. There is sufficient evidence to indicate that arsenic increases the risk of bladder cancer in humans. However, its underlying mechanisms remain elusive. Glutamine (Gln) has multiple functions that promote carcinogenesis. Indeed, Gln transporters on cancer cells surface are often upregulated. Elevated expression levels of Alanine, serine, cysteine-preferring transporter 2 (ASCT2; SLC1A5) have been reported in many types of human tumors. This study characterized the role of SLC1A5 in cell proliferation in arsenite-treated cells. In short-term experiments, SV-40 immortalized human uroepithelial (SV-HUC-1) cells were treated with Sodium arsenite (NaAsO2) (0, 0.5, 1, 2, 4, 8 μM) for 24 h. In long-term experiments, SV-HUC-1 cells were exposed to 0.5 μM NaAsO2 for 40 weeks. In both short-term and long-term experiments, arsenite increased expression of SLC1A5 by 1.89-fold and 2.25-fold, respectively. Arsenite increased Gln consumption of SV-HUC-1 cells, and Gln starvation inhibited cell proliferation in long-term arsenite-treated cells. Importantly, inhibiting SLC1A5 blocked cell proliferation by downregulating mTORC1 in long-term arsenite-treated cells. Moreover, SLC1A5 regulated mTORC1 in an αKG-dependent manner. Our results suggest that SLC1A5 plays an important role in cell proliferation of arsenite-treated SV-HUC-1 cells.
Collapse
Affiliation(s)
- Sihao Li
- Department of Environmental and Occupational Health, School of Public Health, China Medical University, China.
| | - Qing Zhou
- Department of Environmental and Occupational Health, School of Public Health, China Medical University, China.
| | - Weijue Liu
- Department of Environmental and Occupational Health, School of Public Health, China Medical University, China.
| | - Zhushan Fu
- Department of Environmental and Occupational Health, School of Public Health, China Medical University, China.
| | - Hanqing Zhao
- Department of Environmental and Occupational Health, School of Public Health, China Medical University, China.
| | - Shuhua Xi
- Department of Environmental and Occupational Health, School of Public Health, China Medical University, China.
| |
Collapse
|
21
|
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
|
22
|
Transport of L-Arginine Related Cardiovascular Risk Markers. J Clin Med 2020; 9:jcm9123975. [PMID: 33302555 PMCID: PMC7764698 DOI: 10.3390/jcm9123975] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/30/2020] [Accepted: 12/01/2020] [Indexed: 12/15/2022] Open
Abstract
L-arginine and its derivatives, asymmetric and symmetric dimethylarginine (ADMA and SDMA) and L-homoarginine, have emerged as cardiovascular biomarkers linked to cardiovascular outcomes and various metabolic and functional pathways such as NO-mediated endothelial function. Cellular uptake and efflux of L-arginine and its derivatives are facilitated by transport proteins. In this respect the cationic amino acid transporters CAT1 and CAT2 (SLC7A1 and SLC7A2) and the system y+L amino acid transporters (SLC7A6 and SLC7A7) have been most extensively investigated, so far, but the number of transporters shown to mediate the transport of L-arginine and its derivatives is constantly increasing. In the present review we assess the growing body of evidence regarding the function, expression, and clinical relevance of these transporters and their possible relation to cardiovascular diseases.
Collapse
|
23
|
Ding M, Bu X, Li Z, Xu H, Feng L, Hu J, Wei X, Gao J, Tao Y, Cai B, Liu Y, Qu X, Shen L. NDRG2 ablation reprograms metastatic cancer cells towards glutamine dependence via the induction of ASCT2. Int J Biol Sci 2020; 16:3100-3115. [PMID: 33162818 PMCID: PMC7645990 DOI: 10.7150/ijbs.48066] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 09/21/2020] [Indexed: 01/06/2023] Open
Abstract
Background: Metastasis is the most common cause of lethal outcome in various types of cancers. Although the cell proliferation related metabolism rewiring has been well characterized, less is known about the association of metabolic changes with tumor metastasis. Herein, we demonstrate that metastatic tumor obtained a mesenchymal phenotype, which is obtained by the loss of tumor suppressor NDRG2 triggered metabolic switch to glutamine metabolism. Methods: mRNA-seq and gene expression profile analysis were performed to define the differential gene expressions in primary MEC1 and metastatic MC3 cells and the downstream pathways of NDRG2. NDRG2 regulation of Fbw7-dependent c-Myc stability were determined by immunoprecipitation and protein half-life assay. Luciferase reporter and ChIP assays were used to determine the roles of Akt and c-Myc in mediating NDRG2-dependent regulation of ASCT2 in in both tumor and NDRG2-knockout MEF cells. Finally, the effect of the NDRG2/Akt/c-Myc/ASCT2 signaling on glutaminolysis and tumor metastasis were evaluated by functional experiments and clinical samples. Results: Based on the gene expression profile analysis, we identified metastatic tumor cells acquired the mesenchymal-like characteristics and displayed the increased dependency on glutamine utilization. Further, the gain of NDRG2 function blocked epithelial-mesenchymal transition (EMT) and glutaminolysis, potentially through suppression of glutamine transporter ASCT2 expression. The ASCT2 restoration reversed NDRG2 inhibitory effect on EMT program and tumor metastasis. Mechanistic study indicates that NDRG2 promoted Fbw7-dependent c-Myc degradation by inhibiting Akt activation, and subsequently decreased c-Myc-mediated ASCT2 transcription, in both tumor and NDRG2-knockout MEF cells. Supporting the biological significance, the reciprocal relationship between NDRG2 and ASCT2 were observed in multiple types of tumor tissues, and associated with tumor malignancy. Conclusions: NDRG2-dependent repression of ASCT2 presumably is the predominant route by which NDRG2 rewires glutaminolysis and blocks metastatic tumor survival. Targeting glutaminolytic pathway may provide a new strategy for the treatment of metastatic tumors.
Collapse
Affiliation(s)
- Mingchao Ding
- The State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, The Fourth Military Medical University, Xi'an, 710032, China.,State Key Laboratory of Military Stomatology &National Clinical Research Center for Oral Diseases&Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, No. 145 Changle Xi Road, Xi'an, 710032, China
| | - Xin Bu
- The State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Zhehao Li
- The State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, The Fourth Military Medical University, Xi'an, 710032, China.,Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Jiamusi University, Jiamusi, 154002, China
| | - Haokun Xu
- State Key Laboratory of Military Stomatology &National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Department of Oral Anatomy and Physiology and TMD, the Fourth Military Medical University, Xi'an 710032, China
| | - Lin Feng
- Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Junbi Hu
- Department of Gastroenterology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Xinxin Wei
- The State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, The Fourth Military Medical University, Xi'an, 710032, China.,Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Jiamusi University, Jiamusi, 154002, China
| | - Jiwei Gao
- Department of General Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Yanyan Tao
- Xi'an Peihua University, Xi'an, 710125, China
| | - Bolei Cai
- State Key Laboratory of Military Stomatology &National Clinical Research Center for Oral Diseases&Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, No. 145 Changle Xi Road, Xi'an, 710032, China
| | - Yanpu Liu
- State Key Laboratory of Military Stomatology &National Clinical Research Center for Oral Diseases&Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, No. 145 Changle Xi Road, Xi'an, 710032, China
| | - Xuan Qu
- Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Liangliang Shen
- The State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, The Fourth Military Medical University, Xi'an, 710032, China
| |
Collapse
|
24
|
Scalise M, Console L, Rovella F, Galluccio M, Pochini L, Indiveri C. Membrane Transporters for Amino Acids as Players of Cancer Metabolic Rewiring. Cells 2020; 9:cells9092028. [PMID: 32899180 PMCID: PMC7565710 DOI: 10.3390/cells9092028] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 08/31/2020] [Accepted: 09/01/2020] [Indexed: 02/06/2023] Open
Abstract
Cancer cells perform a metabolic rewiring to sustain an increased growth rate and compensate for the redox stress caused by augmented energy metabolism. The metabolic changes are not the same in all cancers. Some features, however, are considered hallmarks of this disease. As an example, all cancer cells rewire the amino acid metabolism for fulfilling both the energy demand and the changed signaling routes. In these altered conditions, some amino acids are more frequently used than others. In any case, the prerequisite for amino acid utilization is the presence of specific transporters in the cell membrane that can guarantee the absorption and the traffic of amino acids among tissues. Tumor cells preferentially use some of these transporters for satisfying their needs. The evidence for this phenomenon is the over-expression of selected transporters, associated with specific cancer types. The knowledge of the link between the over-expression and the metabolic rewiring is crucial for understanding the molecular mechanism of reprogramming in cancer cells. The continuous growth of information on structure-function relationships and the regulation of transporters will open novel perspectives in the fight against human cancers.
Collapse
Affiliation(s)
- Mariafrancesca Scalise
- Unit of Biochemistry and Molecular Biotechnology, Department DiBEST (Biologia, Ecologia, Scienze della Terra), University of Calabria, Via Bucci 4C, 87036 Arcavacata di Rende, Italy; (M.S.); (L.C.); (F.R.); (M.G.); (L.P.)
| | - Lara Console
- Unit of Biochemistry and Molecular Biotechnology, Department DiBEST (Biologia, Ecologia, Scienze della Terra), University of Calabria, Via Bucci 4C, 87036 Arcavacata di Rende, Italy; (M.S.); (L.C.); (F.R.); (M.G.); (L.P.)
| | - Filomena Rovella
- Unit of Biochemistry and Molecular Biotechnology, Department DiBEST (Biologia, Ecologia, Scienze della Terra), University of Calabria, Via Bucci 4C, 87036 Arcavacata di Rende, Italy; (M.S.); (L.C.); (F.R.); (M.G.); (L.P.)
| | - Michele Galluccio
- Unit of Biochemistry and Molecular Biotechnology, Department DiBEST (Biologia, Ecologia, Scienze della Terra), University of Calabria, Via Bucci 4C, 87036 Arcavacata di Rende, Italy; (M.S.); (L.C.); (F.R.); (M.G.); (L.P.)
| | - Lorena Pochini
- Unit of Biochemistry and Molecular Biotechnology, Department DiBEST (Biologia, Ecologia, Scienze della Terra), University of Calabria, Via Bucci 4C, 87036 Arcavacata di Rende, Italy; (M.S.); (L.C.); (F.R.); (M.G.); (L.P.)
| | - Cesare Indiveri
- Unit of Biochemistry and Molecular Biotechnology, Department DiBEST (Biologia, Ecologia, Scienze della Terra), University of Calabria, Via Bucci 4C, 87036 Arcavacata di Rende, Italy; (M.S.); (L.C.); (F.R.); (M.G.); (L.P.)
- CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM) via Amendola 122/O, 70126 Bari, Italy
- Correspondence: ; Tel.: +39-09-8449-2939
| |
Collapse
|
25
|
Rajeeve K, Vollmuth N, Janaki-Raman S, Wulff TF, Baluapuri A, Dejure FR, Huber C, Fink J, Schmalhofer M, Schmitz W, Sivadasan R, Eilers M, Wolf E, Eisenreich W, Schulze A, Seibel J, Rudel T. Reprogramming of host glutamine metabolism during Chlamydia trachomatis infection and its key role in peptidoglycan synthesis. Nat Microbiol 2020; 5:1390-1402. [PMID: 32747796 DOI: 10.1038/s41564-020-0762-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 06/26/2020] [Indexed: 12/31/2022]
Abstract
Obligate intracellular bacteria such as Chlamydia trachomatis undergo a complex developmental cycle between infectious, non-replicative elementary-body and non-infectious, replicative reticulate-body forms. Elementary bodies transform to reticulate bodies shortly after entering a host cell, a crucial process in infection, initiating chlamydial replication. As Chlamydia fail to replicate outside the host cell, it is unknown how the replicative part of the developmental cycle is initiated. Here we show, using a cell-free approach in axenic media, that the uptake of glutamine by the bacteria is crucial for peptidoglycan synthesis, which has a role in Chlamydia replication. The increased requirement for glutamine in infected cells is satisfied by reprogramming the glutamine metabolism in a c-Myc-dependent manner. Glutamine is effectively taken up by the glutamine transporter SLC1A5 and metabolized via glutaminase. Interference with this metabolic reprogramming limits the growth of Chlamydia. Intriguingly, Chlamydia failed to produce progeny in SLC1A5-knockout organoids and mice. Thus, we report on the central role of glutamine for the development of an obligate intracellular pathogenic bacterium and the reprogramming of host glutamine metabolism, which may provide a basis for innovative anti-infection strategies.
Collapse
Affiliation(s)
- Karthika Rajeeve
- Department of Microbiology, Biocenter, University of Würzburg, Würzburg, Germany. .,Department of Biomedicine, Aarhus University, Aarhus C, Denmark.
| | - Nadine Vollmuth
- Department of Microbiology, Biocenter, University of Würzburg, Würzburg, Germany
| | - Sudha Janaki-Raman
- Department of Biochemistry and Molecular Biology, Biocenter, University of Würzburg, Würzburg, Germany
| | - Thomas F Wulff
- Department of Microbiology, Biocenter, University of Würzburg, Würzburg, Germany
| | - Apoorva Baluapuri
- Department of Biochemistry and Molecular Biology, Biocenter, University of Würzburg, Würzburg, Germany
| | - Francesca R Dejure
- Department of Biochemistry and Molecular Biology, Biocenter, University of Würzburg, Würzburg, Germany.,BioMed X Institute, Heidelberg, Germany
| | - Claudia Huber
- Chair of Biochemistry, Technical University of Munich, Garching, Germany
| | - Julian Fink
- Institute of Organic Chemistry, University of Würzburg, Würzburg, Germany
| | | | - Werner Schmitz
- Department of Biochemistry and Molecular Biology, Biocenter, University of Würzburg, Würzburg, Germany
| | - Rajeeve Sivadasan
- RNA Biology and Cancer, German Cancer Research Center, Heidelberg, Germany
| | - Martin Eilers
- Department of Biochemistry and Molecular Biology, Biocenter, University of Würzburg, Würzburg, Germany
| | - Elmar Wolf
- Department of Biochemistry and Molecular Biology, Biocenter, University of Würzburg, Würzburg, Germany
| | | | - Almut Schulze
- Department of Biochemistry and Molecular Biology, Biocenter, University of Würzburg, Würzburg, Germany.,Division of Tumour Metabolism and Microenvironment, German Cancer Research Center, Heidelberg, Germany
| | - Jürgen Seibel
- Institute of Organic Chemistry, University of Würzburg, Würzburg, Germany
| | - Thomas Rudel
- Department of Microbiology, Biocenter, University of Würzburg, Würzburg, Germany. .,Helmholtz Institute for RNA-based Infection Research (HIRI), Würzburg, Germany.
| |
Collapse
|
26
|
Target the human Alanine/Serine/Cysteine Transporter 2(ASCT2): Achievement and Future for Novel Cancer Therapy. Pharmacol Res 2020; 158:104844. [DOI: 10.1016/j.phrs.2020.104844] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 04/12/2020] [Accepted: 04/13/2020] [Indexed: 12/11/2022]
|
27
|
Sun NY, Yang MH. Metabolic Reprogramming and Epithelial-Mesenchymal Plasticity: Opportunities and Challenges for Cancer Therapy. Front Oncol 2020; 10:792. [PMID: 32509584 PMCID: PMC7252305 DOI: 10.3389/fonc.2020.00792] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 04/22/2020] [Indexed: 12/27/2022] Open
Abstract
Metabolic reprogramming and epithelial-mesenchymal plasticity are both hallmarks of the adaptation of cancer cells for tumor growth and progression. For metabolic changes, cancer cells alter metabolism by utilizing glucose, lipids, and amino acids to meet the requirement of rapid proliferation and to endure stressful environments. Dynamic changes between the epithelial and mesenchymal phenotypes through epithelial-mesenchymal transition (EMT) and mesenchymal-epithelial transition (MET) are critical steps for cancer invasion and metastatic colonization. Compared to the extensively studied metabolic reprogramming in tumorigenesis, the metabolic changes in metastasis are relatively unclear. Here, we review metabolic reprogramming, epithelial-mesenchymal plasticity, and their mutual influences on tumor cells. We also review the developing treatments for targeting cancer metabolism and the impact of metabolic targeting on EMT. In summary, understanding the metabolic adaption and phenotypic plasticity will be mandatory for developing new strategies to target metastatic and refractory cancers that are intractable to current treatments.
Collapse
Affiliation(s)
- Nai-Yun Sun
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan.,Cancer Progression Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Muh-Hwa Yang
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan.,Cancer Progression Research Center, National Yang-Ming University, Taipei, Taiwan.,Division of Medical Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
| |
Collapse
|
28
|
Singh S, Arthur S, Sundaram U. Mechanisms of Regulation of Transporters of Amino Acid Absorption in Inflammatory Bowel Diseases. Compr Physiol 2020; 10:673-686. [PMID: 32163200 DOI: 10.1002/cphy.c190016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Intestinal absorption of dietary amino acids/peptides is essential for protein homeostasis, which in turn is crucial for maintaining health as well as restoration of health from significant diseases. Dietary amino acids/peptides are absorbed by unique transporter processes present in the brush border membrane of absorptive villus cells, which line the entire length of the intestine. To date, the only nutrient absorptive system described in the secretory crypt cells in the mammalian intestine is the one that absorbs the amino acid glutamine. Majority of the amino acid transporters are sodium dependent and therefore require basolateral membrane Na-K-ATPase to maintain an efficient transcellular Na gradient for their activity. These transport processes are tightly regulated by various cellular and molecular mechanisms that facilitate their optimal activity during normal physiological processes. Malabsorption of amino acids, recently described in pathophysiological states such as in inflammatory bowel disease (IBD), is undoubtedly responsible for the debilitating symptoms of IBD such as malnutrition, weight loss and ultimately a failure to thrive. Also recently, in vivo models of IBD and in vitro studies have demonstrated that specific immune-inflammatory mediators/pathways regulate specific amino acid transporters. This provides possibilities to derive novel nutrition and immune-based treatment options for conditions such as IBD. © 2020 American Physiological Society. Compr Physiol 10:673-686, 2020.
Collapse
Affiliation(s)
- Soudamani Singh
- Department of Clinical and Translational Sciences and Appalachian Clinical and Translational Science Institute, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia, USA
| | - Subha Arthur
- Department of Clinical and Translational Sciences and Appalachian Clinical and Translational Science Institute, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia, USA
| | - Uma Sundaram
- Department of Clinical and Translational Sciences and Appalachian Clinical and Translational Science Institute, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia, USA
| |
Collapse
|
29
|
Scalise M, Pochini L, Galluccio M, Console L, Indiveri C. Glutamine transporters as pharmacological targets: From function to drug design. Asian J Pharm Sci 2020; 15:207-219. [PMID: 32373200 PMCID: PMC7193454 DOI: 10.1016/j.ajps.2020.02.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 02/18/2020] [Accepted: 02/29/2020] [Indexed: 12/17/2022] Open
Abstract
Among the different targets of administered drugs, there are membrane transporters that play also a role in drug delivery and disposition. Moreover, drug-transporter interactions are responsible for off-target effects of drugs underlying their toxicity. The improvement of the drug design process is subjected to the identification of those membrane transporters mostly relevant for drug absorption, delivery and side effect production. A peculiar group of proteins with great relevance to pharmacology is constituted by the membrane transporters responsible for managing glutamine traffic in different body districts. The interest around glutamine metabolism lies in its physio-pathological role; glutamine is considered a conditionally essential amino acid because highly proliferative cells have an increased request of glutamine that cannot be satisfied only by endogenous synthesis. Then, glutamine transporters provide cells with this special nutrient. Among the glutamine transporters, SLC1A5, SLC6A14, SLC6A19, SLC7A5, SLC7A8 and some members of SLC38 family are the best characterized, so far, in both physiological and pathological conditions. Few 3D structures have been solved by CryoEM; other structural data on these transporters have been obtained by computational analysis. Interactions with drugs have been described for several transporters of this group. For some of them, the studies are at an advanced stage, for others, the studies are still in nuce and novel biochemical findings open intriguing perspectives.
Collapse
Affiliation(s)
- Mariafrancesca Scalise
- Department of DiBEST (Biologia, Ecologia e Scienze della Terra), University of Calabria, Arcavacata di Rende (CS) 87036, Italy
| | - Lorena Pochini
- Department of DiBEST (Biologia, Ecologia e Scienze della Terra), University of Calabria, Arcavacata di Rende (CS) 87036, Italy
| | - Michele Galluccio
- Department of DiBEST (Biologia, Ecologia e Scienze della Terra), University of Calabria, Arcavacata di Rende (CS) 87036, Italy
| | - Lara Console
- Department of DiBEST (Biologia, Ecologia e Scienze della Terra), University of Calabria, Arcavacata di Rende (CS) 87036, Italy
| | - Cesare Indiveri
- Department of DiBEST (Biologia, Ecologia e Scienze della Terra), University of Calabria, Arcavacata di Rende (CS) 87036, Italy
| |
Collapse
|
30
|
Wang R, Xiang W, Xu Y, Han L, Li Q, Dai W, Cai G. Enhanced glutamine utilization mediated by SLC1A5 and GPT2 is an essential metabolic feature of colorectal signet ring cell carcinoma with therapeutic potential. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:302. [PMID: 32355746 PMCID: PMC7186745 DOI: 10.21037/atm.2020.03.31] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background Colorectal signet-ring cell carcinoma (SRCC) is characterized as a rare subset of colorectal cancer with extremely poor prognosis and it is known to have low or negative 18F fluorodeoxyglucose (18F-FDG) uptake. To date, no in-depth study revealing the metabolic features of colorectal SRCC has been conducted for the lack of reliable study model. The aim of this study was to explore the distinct characteristics of energy utilization for colorectal SRCC based on organoid model. Methods Three organoids were derived from colorectal SRCC patients with low or negative FDG uptake and three organoids were derived from colorectal adenocarcinoma (AC) patients. Glucose, fatty acid and glutamine uptake assays were performed to reveal the different metabolic features of SRCC organoids. Immunohistochemistry (IHC), western blotting and real-time PCR were used to test the expression of critical transporters and enzymes of energy metabolism. Glutamine deprivation analyses were used to confirm the dependence of colorectal SRCC on glutamine. Results Glucose, fatty acid and glutamine uptake assays showed that only glutamine uptake was significantly increased in colorectal SRCC organoids compared with paired normal organoids. Comparing SRCC organoids with AC organoids indicated that glucose and fatty acid uptake were strikingly higher in AC organoids while glutamine uptake was notably lower. Gene expression analyses confirmed that the glutamine transporter SLC1A5 and glutaminolysis enzyme GPT2 were significantly unregulated in colorectal SRCC. Silencing of SLC1A5 or GPT2 could suppress the proliferation of SRCC organoids but attenuating the sensitivity of SRCC to glutamine deprivation. Administration of SLC1A5 or GPT2 inhibitor could prohibit SRCC growth and significantly enhance the sensitivity of SRCC to the treatment of 5-fu and L-OHP. Conclusions This study highlights enhanced glutamine uptake and glutaminolysis as a metabolic feature of colorectal SRCC and a potential therapeutic target.
Collapse
Affiliation(s)
- Renjie Wang
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Wenqiang Xiang
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Ye Xu
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Lingyu Han
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Qingguo Li
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Weixing Dai
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Guoxiang Cai
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| |
Collapse
|
31
|
A Variant of SLC1A5 Is a Mitochondrial Glutamine Transporter for Metabolic Reprogramming in Cancer Cells. Cell Metab 2020; 31:267-283.e12. [PMID: 31866442 DOI: 10.1016/j.cmet.2019.11.020] [Citation(s) in RCA: 237] [Impact Index Per Article: 59.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 08/05/2019] [Accepted: 11/27/2019] [Indexed: 12/27/2022]
Abstract
Glutamine is an essential nutrient that regulates energy production, redox homeostasis, and signaling in cancer cells. Despite the importance of glutamine in mitochondrial metabolism, the mitochondrial glutamine transporter has long been unknown. Here, we show that the SLC1A5 variant plays a critical role in cancer metabolic reprogramming by transporting glutamine into mitochondria. The SLC1A5 variant has an N-terminal targeting signal for mitochondrial localization. Hypoxia-induced gene expression of the SLC1A5 variant is mediated by HIF-2α. Overexpression of the SLC1A5 variant mediates glutamine-induced ATP production and glutathione synthesis and confers gemcitabine resistance to pancreatic cancer cells. SLC1A5 variant knockdown and overexpression alter cancer cell and tumor growth, supporting an oncogenic role. This work demonstrates that the SLC1A5 variant is a mitochondrial glutamine transporter for cancer metabolic reprogramming.
Collapse
|
32
|
Solute carrier transporters: the metabolic gatekeepers of immune cells. Acta Pharm Sin B 2020; 10:61-78. [PMID: 31993307 PMCID: PMC6977534 DOI: 10.1016/j.apsb.2019.12.006] [Citation(s) in RCA: 116] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 09/29/2019] [Accepted: 10/31/2019] [Indexed: 02/06/2023] Open
Abstract
Solute carrier (SLC) transporters meditate many essential physiological functions, including nutrient uptake, ion influx/efflux, and waste disposal. In its protective role against tumors and infections, the mammalian immune system coordinates complex signals to support the proliferation, differentiation, and effector function of individual cell subsets. Recent research in this area has yielded surprising findings on the roles of solute carrier transporters, which were discovered to regulate lymphocyte signaling and control their differentiation, function, and fate by modulating diverse metabolic pathways and balanced levels of different metabolites. In this review, we present current information mainly on glucose transporters, amino-acid transporters, and metal ion transporters, which are critically important for mediating immune cell homeostasis in many different pathological conditions.
Collapse
Key Words
- 3-PG, 3-phosphoglyceric acid
- ABC, ATP-binding cassette
- AIF, apoptosis-inducing factor
- AP-1, activator protein 1
- ASCT2, alanine serine and cysteine transporter system 2
- ATP, adenosine triphosphate
- BCR, B cell receptor
- BMDMs, bone marrow-derived macrophages
- CD45R, a receptor-type protein tyrosine phosphatase
- CTL, cytotoxic T lymphocytes
- DC, dendritic cells
- EAATs, excitatory amino acid transporters
- ER, endoplasmic reticulum
- ERRα, estrogen related receptor alpha
- FFA, free fatty acids
- G-6-P, glucose 6-phosphate
- GLUT, glucose transporters
- GSH, glutathione
- Glucose
- Glutamine
- HIF-1α, hypoxia-inducible factor 1-alpha
- HIV-1, human immunodeficiency virus type 1
- Hk1, hexokinase-1
- IFNβ, interferon beta
- IFNγ, interferon gamma
- IKK, IκB kinase
- IKKβ, IκB kinase beta subunit
- IL, interleukin
- LDHA, lactate dehydrogenase A
- LPS, lipopolysaccharide
- Lymphocytes
- Lyn, tyrosine-protein kinase
- MAPK, mitogen-activated protein kinase
- MCT, monocarboxylate transporters
- MS, multiple sclerosis
- Metal ion
- NADPH, nicotinamide adenine dinucleotide phosphate
- NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells
- NO, nitric oxide
- NOD2, nucleotide-binding oligomerization domain containing 2
- PEG2, prostaglandin E2
- PI-3K/AKT, phosphatidylinositol-3-OH kinase/serine–threonine kinase
- PPP, pentose phosphate pathway
- Pfk, phosphofructokinase
- RA, rheumatoid arthritis
- RLR, RIG-I-like receptor
- ROS, reactive oxygen species
- SLC, solute carrier
- SLE, systemic lupus erythematosus
- SNAT, sodium-coupled neutral amino acid transporters
- STAT, signal transducers and activators of transcription
- Solute carrier
- TAMs, tumor-associated macrophages
- TCA, tricarboxylic acid
- TCR, T cell receptor
- TLR, toll-like receptor
- TNF, tumor necrosis factor
- TRPM7, transient receptor potential cation channel subfamily M member 7
- Teffs, effector T cells
- Th1/2/17, type 1/2/17 helper T cells
- Tregs, regulatory T cells
- VEGF, vascular endothelial growth factor
- ZIP, zrt/irt-like proteins
- iNOS, inducible nitric oxide synthase
- iTregs, induced regulatory T cells
- mTORC1, mammalian target of rapamycin complex 1
- α-KG, α-ketoglutaric acid
Collapse
|
33
|
Zhang Z, Liu R, Shuai Y, Huang Y, Jin R, Wang X, Luo J. ASCT2 (SLC1A5)-dependent glutamine uptake is involved in the progression of head and neck squamous cell carcinoma. Br J Cancer 2019; 122:82-93. [PMID: 31819178 PMCID: PMC6964701 DOI: 10.1038/s41416-019-0637-9] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Revised: 07/10/2019] [Accepted: 10/28/2019] [Indexed: 01/20/2023] Open
Abstract
Background Glutamine is an abundant and versatile nutrient in cancer cells. Head and neck squamous cell carcinoma (HNSCC) was reported to be dependent on mainly glucose, not glutamine, for producing the energy required for survival and proliferation. Methods The roles of ASCT2 (SLC1A5) and associated glutamine metabolism were determined by the MTT, colony formation, glutamine uptake, intracellular glutathione, ROS detection, immunofluorescence, immunohistochemistry, and apoptosis enzyme-linked immunosorbent assays as well as animal studies. Results We found that glutamine is also critical for HNSCC. In this study, ASCT2, an amino acid transporter responsible for glutamine transport, in addition to LAT1 and GLS, is overexpressed in HNSCC and associated with poor survival. Using both in vivo and in vitro models, we found that knocking down ASCT2 by shRNAs or miR-137 or the combination of silencing ASCT2 and pharmacologically inhibiting SNAT2 via a small-molecule antagonist called V-9302 significantly suppressed intracellular glutamine levels and downstream glutamine metabolism, including glutathione production; these effects attenuated growth and proliferation, increased apoptosis and autophagy, and increased oxidative stress and mTORC1 pathway suppression in HNSCC. Additionally, silencing ASCT2 improved the response to cetuximab in HNSCC. Conclusions In summary, ASCT2-dependent glutamine uptake and subsequent glutamine metabolism are essential for HNSCC tumorigenesis, and the combination of glutamine uptake inhibitors and cetuximab presents a promising strategy for improving the outcomes of HNSCC patients.
Collapse
Affiliation(s)
- Ze Zhang
- Department of Maxillofacial and Otorhinolaryngology Oncology and Department of Head and Neck Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Ruoyan Liu
- Department of Gynecologic Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Yanjie Shuai
- Department of Maxillofacial and Otorhinolaryngology Oncology and Department of Head and Neck Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Yuting Huang
- Cancer and Immunology, Children's Research Institute, Children's National Medical Center, Washington, DC, 20010, USA
| | - Rui Jin
- Department of Maxillofacial and Otorhinolaryngology Oncology and Department of Head and Neck Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Xudong Wang
- Department of Maxillofacial and Otorhinolaryngology Oncology and Department of Head and Neck Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Jingtao Luo
- Department of Maxillofacial and Otorhinolaryngology Oncology and Department of Head and Neck Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.
| |
Collapse
|
34
|
Agarwal AK, Tunison K, Mitsche MA, McDonald JG, Garg A. Insights into lipid accumulation in skeletal muscle in dysferlin-deficient mice. J Lipid Res 2019; 60:2057-2073. [PMID: 31653658 PMCID: PMC6889719 DOI: 10.1194/jlr.ra119000399] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 10/21/2019] [Indexed: 12/15/2022] Open
Abstract
Loss of dysferlin (DYSF) protein in humans results in limb-girdle muscular dystrophy 2B, characterized by progressive loss of muscles in the distal limbs with impaired locomotion. The DYSF-null (Bla/J) mouse develops severe steatotic muscles upon aging. Here, we report a marked increase in adipocytes, especially in the psoas and gluteus muscles but not in the soleus and tibialis anterior muscles in aged Bla/J mice compared with WT mice. There was a robust upregulation in the mRNA expression of enzymes involved in lipogenesis and triacylglycerol (TAG) synthesis pathways in the steatotic skeletal muscles. Lipidomic analysis of the steatotic skeletal muscles revealed an increase in several molecular species of TAG, although it is unclear whether it was at the expense of phosphatidylcholine and phosphatidylserine. The adipocytes in steatotic muscles were extramyocellular, as determined by the increased expression of caveolin 1 (a cellular marker for adipocytes) and lipid-droplet protein, perilipin 1. This increase in adipocytes occured as a consequence of the loss of myocytes.
Collapse
Affiliation(s)
- Anil K Agarwal
- Division of Nutrition and Metabolic Diseases Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Katie Tunison
- Division of Nutrition and Metabolic Diseases Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Matthew A Mitsche
- Center for Human Nutrition, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Jeffrey G McDonald
- Center for Human Nutrition, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Abhimanyu Garg
- Division of Nutrition and Metabolic Diseases Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
| |
Collapse
|
35
|
Scalise M, Pochini L, Cosco J, Aloe E, Mazza T, Console L, Esposito A, Indiveri C. Interaction of Cholesterol With the Human SLC1A5 (ASCT2): Insights Into Structure/Function Relationships. Front Mol Biosci 2019; 6:110. [PMID: 31709262 PMCID: PMC6819821 DOI: 10.3389/fmolb.2019.00110] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 10/07/2019] [Indexed: 01/01/2023] Open
Abstract
The human SLC1A5 commonly known as ASCT2 is a sodium-dependent neutral amino acid antiporter involved in transmembrane traffic of glutamine that is exchanged through the cell membrane with smaller amino acids such as serine or threonine. Due to the strong overexpression in human cancers, ASCT2 is widely studied for its relevance to human health. Of special interest are the aspects related to the regulation of its function. The role of cholesterol as a modulator of the transport activity has been studied using a combined strategy of computational and experimental approaches. The effect of cholesterol on theNa ex + -[3H]glutamineex/glutaminein antiport in proteoliposomes has been evaluated by adding cholesteryl hemisuccinate. A strong stimulation of transport activity was observed in the presence of 75 μg cholesteryl hemisuccinate per mg total lipids. The presence of cholesterol did not influence the proteoliposome volume, in a wide range of tested concentration, excluding that the stimulation could be due to effects on the vesicles. cholesteryl hemisuccinate, indeed, improved the incorporation of the protein into the phospholipid bilayer to some extent and increased about three times the Vmax of transport without affecting the Km for glutamine. Docking of cholesterol into the hASCT2 trimer was performed. Six poses were obtained some of which overlapped the hypothetical cholesterol molecules observed in the available 3D structures. Additional poses were docked close to CARC/CRAC motifs (Cholesterol Recognition/interaction Amino acid Consensus sequence). To test the direct binding of cholesterol to the protein, a strategy based on the specific targeting of tryptophan and cysteine residues located in the neighborhood of cholesterol poses was employed. On the one hand, cholesterol binding was impaired by modification of tryptophan residues by the Koshland's reagent. On the other hand, the presence of cholesterol impaired the interaction of thiol reagents with the protein. Altogether, these results confirmed that cholesterol molecules interacted with the protein in correspondence of the poses predicted by the docking analysis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Cesare Indiveri
- Unit of Biochemistry and Molecular Biotechnology, Department DiBEST (Biologia, Ecologia, Scienze della Terra), University of Calabria, Cosenza, Italy
| |
Collapse
|
36
|
Zhuang X, Tong H, Ding Y, Wu L, Cai J, Si Y, Zhang H, Shen M. Long noncoding RNA ABHD11-AS1 functions as a competing endogenous RNA to regulate papillary thyroid cancer progression by miR-199a-5p/SLC1A5 axis. Cell Death Dis 2019; 10:620. [PMID: 31409775 PMCID: PMC6692390 DOI: 10.1038/s41419-019-1850-4] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 07/15/2019] [Accepted: 07/29/2019] [Indexed: 12/25/2022]
Abstract
With the increasing incidence of papillary thyroid cancer (PTC), more attention has been paid to exploring the mechanism of PTC initiation and progression. In addition, ectopic expression of long noncoding RNAs (lncRNAs) is reported to play a pivotal role in multiple human cancers. Based on these findings, we examined lncRNA ABHD11 antisense RNA 1 (ABHD11-AS1) expression and its clinical significance, biological function and mechanism in PTC. First, we analyzed thyroid ABHD11-AS1 expression in The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. Then, qRT-PCR was applied to detect the expression in paired PTC tissues and adjacent normal tissues, as well as in PTC cell lines (TPC-1 and K-1) and a normal thyroid follicular epithelium cell line (Nthy-ori3-1). In addition, we validated the relationship between ABHD11-AS1 expression and clinicopathological features by the Pearson X2 test. The oncogenic role of ABHD11-AS1 and its regulation of miR-199a-5p in PTC were examined by biological assays. Finally, bioinformatics analysis and mechanism assays were used to elucidate the underlying mechanism. We found that ABHD11-AS1 was remarkably overexpressed in PTC, and high expression was related to tumor size, lymph node metastasis, extrathyroidal extension and advanced TNM stage. Moreover, ABHD11-AS1 enhanced the abilities of cell proliferation, migration, and invasion, inhibited apoptosis in vitro, promoted tumorigenesis in vivo via sponging miR-199a-5p and then induced SLC1A5 activation. In addition, rescue assays were performed to confirm the ABHD11-AS1/miR-199a-5p/SLC1A5 axis. Taken together, the data show that ABHD11-AS1 acts as a competing endogenous RNA (ceRNA) to exert malignant properties in PTC through the miR-199a-5p/SLC1A5 axis. Therefore, our study may shed light on PTC diagnosis and therapies.
Collapse
Affiliation(s)
- Xi Zhuang
- Department of General Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, PR China
| | - Houchao Tong
- Department of General Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, PR China
| | - Yu Ding
- Department of General Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, PR China
| | - Luyao Wu
- Department of General Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, PR China
| | - Jingsheng Cai
- Department of General Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, PR China
| | - Yan Si
- Department of General Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, PR China
| | - Hao Zhang
- Department of General Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, PR China
| | - Meiping Shen
- Department of General Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, PR China.
| |
Collapse
|
37
|
Punekar S, Cho DC. Novel Therapeutics Affecting Metabolic Pathways. Am Soc Clin Oncol Educ Book 2019; 39:e79-e87. [PMID: 31099667 DOI: 10.1200/edbk_238499] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cancer cells are known to have distinct metabolic characteristics compared with normal cells, given the catabolic and anabolic demands of increased cell growth and proliferation. This altered metabolism in cancer cells imbues differential dependencies, and substantial effort has been invested in developing therapeutic strategies to exploit these potential vulnerabilities. Parallel to these efforts has been a growing appreciation for the presence of notable intratumoral metabolic heterogeneity. Although many novel agents are showing some promising results in targeting specific metabolic processes, the challenge moving forward will be to develop combination strategies to address the aforementioned metabolic heterogeneity and its interplay with both epigenetic and immune factors in the tumor microenvironment. In this review, we discuss recent developments in targeting tumor catabolism, lipid biosynthesis, glycolysis, and the citric acid cycle as well as efforts to combine these approaches with immunotherapy.
Collapse
Affiliation(s)
| | - Daniel C Cho
- 1 Perlmutter Cancer Center at NYU Langone, New York, NY
| |
Collapse
|
38
|
Ni F, Yu WM, Li Z, Graham DK, Jin L, Kang S, Rossi MR, Li S, Broxmeyer HE, Qu CK. Critical role of ASCT2-mediated amino acid metabolism in promoting leukaemia development and progression. Nat Metab 2019; 1:390-403. [PMID: 31535081 PMCID: PMC6750232 DOI: 10.1038/s42255-019-0039-6] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Amino acid (AA) metabolism is involved in diverse cellular functions, including cell survival and growth, however it remains unclear how it regulates normal hematopoiesis versus leukemogenesis. Here, we report that knockout of Slc1a5 (ASCT2), a transporter of neutral AAs, especially glutamine, results in mild to moderate defects in bone marrow and mature blood cell development under steady state conditions. In contrast, constitutive or induced deletion of Slc1a5 decreases leukemia initiation and maintenance driven by the oncogene MLL-AF9 or Pten deficiency. Survival of leukemic mice is prolonged following Slc1a5 deletion, and pharmacological inhibition of ASCT2 also decreases leukemia development and progression in xenograft models of human acute myeloid leukemia. Mechanistically, loss of ASCT2 generates a global effect on cellular metabolism, disrupts leucine influx and mTOR signaling, and induces apoptosis in leukemic cells. Given the substantial difference in reliance on ASCT2-mediated AA metabolism between normal and malignant blood cells, this in vivo study suggests ASCT2 as a promising therapeutic target for the treatment of leukemia.
Collapse
Affiliation(s)
- Fang Ni
- Department of Pediatrics, Division of Hematology/Oncology, Aflac Cancer and Blood Disorders Center, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Wen-Mei Yu
- Department of Pediatrics, Division of Hematology/Oncology, Aflac Cancer and Blood Disorders Center, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Zhiguo Li
- Department of Pediatrics, Division of Hematology/Oncology, Aflac Cancer and Blood Disorders Center, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Douglas K Graham
- Department of Pediatrics, Division of Hematology/Oncology, Aflac Cancer and Blood Disorders Center, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Lingtao Jin
- Department of Hematology/Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Sumin Kang
- Department of Hematology/Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Michael R Rossi
- Department of Radiation Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Shiyong Li
- Department of Pathology and Laboratory Medicine, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Hal E Broxmeyer
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Cheng-Kui Qu
- Department of Pediatrics, Division of Hematology/Oncology, Aflac Cancer and Blood Disorders Center, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
39
|
Abstract
The small intestine mediates the absorption of amino acids after ingestion of protein and sustains the supply of amino acids to all tissues. The small intestine is an important contributor to plasma amino acid homeostasis, while amino acid transport in the large intestine is more relevant for bacterial metabolites and fluid secretion. A number of rare inherited disorders have contributed to the identification of amino acid transporters in epithelial cells of the small intestine, in particular cystinuria, lysinuric protein intolerance, Hartnup disorder, iminoglycinuria, and dicarboxylic aminoaciduria. These are most readily detected by analysis of urine amino acids, but typically also affect intestinal transport. The genes underlying these disorders have all been identified. The remaining transporters were identified through molecular cloning techniques to the extent that a comprehensive portrait of functional cooperation among transporters of intestinal epithelial cells is now available for both the basolateral and apical membranes. Mouse models of most intestinal transporters illustrate their contribution to amino acid homeostasis and systemic physiology. Intestinal amino acid transport activities can vary between species, but these can now be explained as differences of amino acid transporter distribution along the intestine. © 2019 American Physiological Society. Compr Physiol 9:343-373, 2019.
Collapse
Affiliation(s)
- Stefan Bröer
- Research School of Biology, The Australian National University, Canberra, ACT 2601, Australia
| | - Stephen J Fairweather
- Research School of Biology, The Australian National University, Canberra, ACT 2601, Australia
| |
Collapse
|
40
|
The role of ASCT2 in cancer: A review. Eur J Pharmacol 2018; 837:81-87. [DOI: 10.1016/j.ejphar.2018.07.007] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Revised: 06/10/2018] [Accepted: 07/09/2018] [Indexed: 01/18/2023]
|
41
|
Scalise M, Pochini L, Console L, Losso MA, Indiveri C. The Human SLC1A5 (ASCT2) Amino Acid Transporter: From Function to Structure and Role in Cell Biology. Front Cell Dev Biol 2018; 6:96. [PMID: 30234109 PMCID: PMC6131531 DOI: 10.3389/fcell.2018.00096] [Citation(s) in RCA: 161] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 08/08/2018] [Indexed: 12/30/2022] Open
Abstract
SLC1A5, known as ASCT2, is a neutral amino acid transporter belonging to the SLC1 family and localized in the plasma membrane of several body districts. ASCT2 is an acronym standing for Alanine, Serine, Cysteine Transporter 2 even if the preferred substrate is the conditionally essential amino acid glutamine, with cysteine being a modulator and not a substrate. The studies around amino acid transport in cells and tissues began in the '60s by using radiolabeled compounds and competition assays. After identification of murine and human genes, the function of the coded protein has been studied in cell system and in proteoliposomes revealing that this transporter is a Na+ dependent antiporter of neutral amino acids, some of which are only inwardly transported and others are bi-directionally exchanged. The functional asymmetry merged with the kinetic asymmetry in line with the physiological role of amino acid pool harmonization. An intriguing function has been described for ASCT2 that is exploited as a receptor by a group of retroviruses to infect human cells. Interactions with scaffold proteins and post-translational modifications regulate ASCT2 stability, trafficking and transport activity. Two asparagine residues, namely N163 and N212, are the sites of glycosylation that is responsible for the definitive localization into the plasma membrane. ASCT2 expression increases in highly proliferative cells such as inflammatory and stem cells to fulfill the augmented glutamine demand. Interestingly, for the same reason, the expression of ASCT2 is greatly enhanced in many human cancers. This finding has generated interest in its candidacy as a pharmacological target for new anticancer drugs. The recently solved 3D structure of ASCT2 will aid in the rational design of such therapeutic compounds.
Collapse
Affiliation(s)
- Mariafrancesca Scalise
- Department DiBEST (Biologia, Ecologia, Scienze Della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Cosenza, Italy
| | - Lorena Pochini
- Department DiBEST (Biologia, Ecologia, Scienze Della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Cosenza, Italy
| | - Lara Console
- Department DiBEST (Biologia, Ecologia, Scienze Della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Cosenza, Italy
| | - Maria A Losso
- Department DiBEST (Biologia, Ecologia, Scienze Della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Cosenza, Italy
| | - Cesare Indiveri
- Department DiBEST (Biologia, Ecologia, Scienze Della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Cosenza, Italy.,CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnology, Bari, Italy
| |
Collapse
|
42
|
Lin W, Liu Z, Zheng X, Chen M, Gao D, Tian Z. High-salt diet affects amino acid metabolism in plasma and muscle of Dahl salt-sensitive rats. Amino Acids 2018; 50:1407-1414. [DOI: 10.1007/s00726-018-2615-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 07/03/2018] [Indexed: 10/28/2022]
|
43
|
D'Aquila P, Crocco P, De Rango F, Indiveri C, Bellizzi D, Rose G, Passarino G. A Genetic Variant of ASCT2 Hampers In Vitro RNA Splicing and Correlates with Human Longevity. Rejuvenation Res 2018; 21:193-199. [DOI: 10.1089/rej.2017.1948] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Affiliation(s)
- Patrizia D'Aquila
- Department of Biology, Ecology and Earth Science, University of Calabria, Rende, Italy
| | - Paolina Crocco
- Department of Biology, Ecology and Earth Science, University of Calabria, Rende, Italy
| | - Francesco De Rango
- Department of Biology, Ecology and Earth Science, University of Calabria, Rende, Italy
| | - Cesare Indiveri
- Department of Biology, Ecology and Earth Science, University of Calabria, Rende, Italy
| | - Dina Bellizzi
- Department of Biology, Ecology and Earth Science, University of Calabria, Rende, Italy
| | - Giuseppina Rose
- Department of Biology, Ecology and Earth Science, University of Calabria, Rende, Italy
| | - Giuseppe Passarino
- Department of Biology, Ecology and Earth Science, University of Calabria, Rende, Italy
| |
Collapse
|
44
|
Abstract
With the global population rising, the need for sustainable and resource-efficiently produced proteins with nutritional and health promoting qualities has become urgent. Proteins are important macronutrients and are involved in most, if not all, biological processes in the human body. This review discusses these absorption mechanisms in the small intestine. To study intestinal transport and predict bioavailability, cell lines are widely applied as screening models and often concern Caco-2, HT-29, HT-29/MTX and T84 cells. Here, we provide an overview of the presence and activities of peptide- and amino acid transporters in these cell models. Further, inter-laboratory differences are discussed as well as the culture micro-environment, both of which may influence cell culture phenotype and performance. Finally, the value of new developments in the field, including culturing cells in 3-dimensional systems under shear stress (i.e., gut-on-chips), is highlighted. In particular, their suitability in screening novel food proteins and prediction of the nutritional quality needed for inclusion in the human diet of the future is addressed.
Collapse
|
45
|
Liu H, Han Y, Li J, Qin M, Fu Q, Wang C, Liu Z. 18F-Alanine Derivative Serves as an ASCT2 Marker for Cancer Imaging. Mol Pharm 2018; 15:947-954. [DOI: 10.1021/acs.molpharmaceut.7b00884] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Hui Liu
- Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Yuxiang Han
- Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Jiyuan Li
- Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Ming Qin
- Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Qunfeng Fu
- Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Chunhong Wang
- Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Zhibo Liu
- Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| |
Collapse
|
46
|
Edwards DN, Ngwa VM, Wang S, Shiuan E, Brantley-Sieders DM, Kim LC, Reynolds AB, Chen J. The receptor tyrosine kinase EphA2 promotes glutamine metabolism in tumors by activating the transcriptional coactivators YAP and TAZ. Sci Signal 2017; 10:eaan4667. [PMID: 29208682 PMCID: PMC5819349 DOI: 10.1126/scisignal.aan4667] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Malignant tumors reprogram cellular metabolism to support cancer cell proliferation and survival. Although most cancers depend on a high rate of aerobic glycolysis, many cancer cells also display addiction to glutamine. Glutamine transporters and glutaminase activity are critical for glutamine metabolism in tumor cells. We found that the receptor tyrosine kinase EphA2 activated the TEAD family transcriptional coactivators YAP and TAZ (YAP/TAZ), likely in a ligand-independent manner, to promote glutamine metabolism in cells and mouse models of HER2-positive breast cancer. Overexpression of EphA2 induced the nuclear accumulation of YAP and TAZ and increased the expression of YAP/TAZ target genes. Inhibition of the GTPase Rho or the kinase ROCK abolished EphA2-dependent YAP/TAZ nuclear localization. Silencing YAP or TAZ substantially reduced the amount of intracellular glutamate through decreased expression of SLC1A5 and GLS, respectively, genes that encode proteins that promote glutamine uptake and metabolism. The regulatory DNA elements of both SLC1A5 and GLS contain TEAD binding sites and were bound by TEAD4 in an EphA2-dependent manner. In patient breast cancer tissues, EphA2 expression positively correlated with that of YAP and TAZ, as well as that of GLS and SLC1A5 Although high expression of EphA2 predicted enhanced metastatic potential and poor patient survival, it also rendered HER2-positive breast cancer cells more sensitive to glutaminase inhibition. The findings define a previously unknown mechanism of EphA2-mediated glutaminolysis through YAP/TAZ activation in HER2-positive breast cancer and identify potential therapeutic targets in patients.
Collapse
Affiliation(s)
- Deanna N Edwards
- Division of Rheumatology and Immunology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Verra M Ngwa
- Department of Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Shan Wang
- Division of Rheumatology and Immunology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Eileen Shiuan
- Department of Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA
- Medical Scientist Training Program, Vanderbilt University, Nashville, TN 37232, USA
| | - Dana M Brantley-Sieders
- Division of Rheumatology and Immunology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Laura C Kim
- Department of Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Albert B Reynolds
- Department of Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Jin Chen
- Division of Rheumatology and Immunology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
- Department of Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Veterans Affairs Medical Center, Tennessee Valley Healthcare System, Nashville, TN 37212, USA
| |
Collapse
|
47
|
AP1G1 is involved in cetuximab-mediated downregulation of ASCT2-EGFR complex and sensitization of human head and neck squamous cell carcinoma cells to ROS-induced apoptosis. Cancer Lett 2017; 408:33-42. [PMID: 28823958 DOI: 10.1016/j.canlet.2017.08.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 08/07/2017] [Accepted: 08/09/2017] [Indexed: 12/12/2022]
Abstract
In this study, we expanded our recent work showing that ASCT2, a Na+-dependent neutral amino acid transporter that plays a major role in glutamine uptake in cancer cells, is physically associated with EGFR in human head and neck squamous cell carcinoma cells and in several other types of cancer cells. We found in our current study that ASCT2 can be downregulated by cetuximab, an approved anti-EGFR therapeutic antibody, via cetuximab-induced EGFR endocytosis independently of cetuximab-mediated inhibition of EGFR tyrosine kinase. We further found that ASCT2-EGFR association involves the adaptor-related protein complex 1 gamma 1 subunit (AP1G1), a subunit of clathrin-associated adaptor protein complex 1, which plays a role in membrane protein sorting in endosomes after receptor-mediated endocytosis. We found that AP1G1 is physically associated with both ASCT2 and EGFR and, together with those molecules, forms a heterotrimeric molecular complex. Knockdown of AP1G1 lowered the level of ASCT2-EGFR association, inhibited cetuximab-mediated internalization of ASCT2-EGFR complex, and decreased intracellular glutamine uptake and glutathione biosynthesis. These findings suggest a new therapeutic strategy to overcome cetuximab resistance in cancer cells through combination of cetuximab, which co-targets ASCT2 along with EGFR, with an ROS-inducing agent.
Collapse
|
48
|
Suzuki M, Toki H, Furuya A, Ando H. Establishment of monoclonal antibodies against cell surface domains of ASCT2/SLC1A5 and their inhibition of glutamine-dependent tumor cell growth. Biochem Biophys Res Commun 2016; 482:651-657. [PMID: 27865832 DOI: 10.1016/j.bbrc.2016.11.089] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 11/15/2016] [Indexed: 12/20/2022]
Abstract
Human alanine-serine-cysteine transporter 2 (ASCT2; SLC1A5) is a major transporter of the amino acid glutamine that is known to be overexpressed in certain malignant tumors. In this study, we generated specific monoclonal antibodies (MAbs) against ASCT2 by establishing an ASCT2-expressing Chinese hamster ovary cell line that was used to immunize mice and rats. The MAbs KM4008, KM4012, and KM4018 against ASCT2 were isolated through a cell-based screen; these specifically bound to ASCT2-positive cells, as determined by flow cytometry and immunoprecipitation. In addition, the antibodies suppressed glutamine-dependent growth of WiDr colorectal cancer cells. These results provide evidence supporting the use of MAbs against ASCT2 as an effective therapeutic strategy for cancer treatment.
Collapse
Affiliation(s)
- Masayo Suzuki
- R&D Division, Tokyo Research Park, Kyowa Hakko Kirin Co., Ltd, Machida-shi, Tokyo, Japan
| | - Hiroe Toki
- R&D Division, Tokyo Research Park, Kyowa Hakko Kirin Co., Ltd, Machida-shi, Tokyo, Japan
| | - Akiko Furuya
- R&D Division, Tokyo Research Park, Kyowa Hakko Kirin Co., Ltd, Machida-shi, Tokyo, Japan
| | - Hiroshi Ando
- R&D Division, Tokyo Research Park, Kyowa Hakko Kirin Co., Ltd, Machida-shi, Tokyo, Japan.
| |
Collapse
|
49
|
Timosenko E, Ghadbane H, Silk JD, Shepherd D, Gileadi U, Howson LJ, Laynes R, Zhao Q, Strausberg RL, Olsen LR, Taylor S, Buffa FM, Boyd R, Cerundolo V. Nutritional Stress Induced by Tryptophan-Degrading Enzymes Results in ATF4-Dependent Reprogramming of the Amino Acid Transporter Profile in Tumor Cells. Cancer Res 2016; 76:6193-6204. [PMID: 27651314 PMCID: PMC5096689 DOI: 10.1158/0008-5472.can-15-3502] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 08/26/2016] [Indexed: 01/08/2023]
Abstract
Tryptophan degradation is an immune escape strategy shared by many tumors. However, cancer cells' compensatory mechanisms remain unclear. We demonstrate here that a shortage of tryptophan caused by expression of indoleamine 2,3-dioxygenase (IDO) and tryptophan 2,3-dioxygenase (TDO) resulted in ATF4-dependent upregulation of several amino acid transporters, including SLC1A5 and its truncated isoforms, which in turn enhanced tryptophan and glutamine uptake. Importantly, SLC1A5 failed to be upregulated in resting human T cells kept under low tryptophan conditions but was enhanced upon cognate antigen T-cell receptor engagement. Our results highlight key differences in the ability of tumor and T cells to adapt to tryptophan starvation and provide important insights into the poor prognosis of tumors coexpressing IDO and SLC1A5. Cancer Res; 76(21); 6193-204. ©2016 AACR.
Collapse
Affiliation(s)
- Elina Timosenko
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Hemza Ghadbane
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Jonathan D Silk
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Dawn Shepherd
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Uzi Gileadi
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Lauren J Howson
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Robert Laynes
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Qi Zhao
- Ludwig Cancer Research, New York, New York
| | | | - Lars R Olsen
- Center for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, Lyngby, Denmark
| | - Stephen Taylor
- The Computational Biology Research Group (CBRG), Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Francesca M Buffa
- Department of Oncology, Old Road Campus Research Building, University of Oxford, Oxford, United Kingdom
| | - Richard Boyd
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Vincenzo Cerundolo
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom.
| |
Collapse
|
50
|
Chen Z, Huang W, Srinivas SR, Jones CR, Ganapathy V, Prasad PD. Serine Racemase and D-Serine Transport in Human Placenta and Evidence for a Transplacental Gradient for D-Serine in Humans. ACTA ACUST UNITED AC 2016; 11:294-303. [PMID: 15219883 DOI: 10.1016/j.jsgi.2004.02.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
OBJECTIVE To investigate the possible role of human placenta in providing D-serine to the developing fetus. METHODS Expression of serine racemase in placenta was determined by reverse transcriptase polymerase chain reaction and northern analysis and confirmed by subsequent cloning. The transport of D-serine by human ATB(0) was characterized by expressing the cloned cDNA transiently in mammalian cells using the vaccinia virus expression system. D-serine levels in maternal and fetal blood were measured by fluorescence high-performance liquid chromatography (HPLC) after derivatization of the amino acids with o-phthaldialdehyde and N-tertiary-butyloxycarbonyl-L-cysteine. RESULTS mRNA for serine racemase was detected in placenta. ATB(0) was capable of d-serine transport, and the transport process is obligatorily dependent on sodium (Na+) with a Na(+):substrate stoichiometry of 1:1 and saturable with a Michaelis-Menten constant of 310 +/- 30 microM. Furthermore, studies have shown that ATB(0) is not expressed in the maternal-facing brush border membrane of human placental syncytiotrophoblast. The circulating concentration of D-serine in maternal serum is 5.8 +/- 0.5 microM, and the corresponding value in the fetal serum is 14.6 +/- 1.2 microM, indicating a two- to three-fold higher concentration of D-serine in the fetus than in the mother. CONCLUSION We speculate that D-serine is synthesized in human placenta by the racemization of L-serine and that ATB(0), expressed on the basal membrane of the syncytiotrophoblast, mediates the efflux of D-serine into the fetal circulation in exchange for other amino acids in fetal blood.
Collapse
Affiliation(s)
- Zhong Chen
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, 1120 Laney Walker Boulevard, Augusta, GA 30912-2100, USA
| | | | | | | | | | | |
Collapse
|