201
|
Bröer A, Gauthier-Coles G, Rahimi F, van Geldermalsen M, Dorsch D, Wegener A, Holst J, Bröer S. Ablation of the ASCT2 ( SLC1A5) gene encoding a neutral amino acid transporter reveals transporter plasticity and redundancy in cancer cells. J Biol Chem 2019; 294:4012-4026. [PMID: 30635397 DOI: 10.1074/jbc.ra118.006378] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Revised: 01/08/2019] [Indexed: 12/21/2022] Open
Abstract
The neutral amino acid transporter solute carrier family 1 member 5 (SLC1A5 or ASCT2) is overexpressed in many cancers. To identify its roles in tumors, we employed 143B osteosarcoma cells and HCC1806 triple-negative breast cancer cells with or without ASCT2 deletion. ASCT2ko 143B cells grew well in standard culture media, but ASCT2 was required for optimal growth at <0.5 mm glutamine, with tumor spheroid growth and monolayer migration of 143B ASCT2ko cells being strongly impaired at lower glutamine concentrations. However, the ASCT2 deletion did not affect matrix-dependent invasion. ASCT2ko 143B xenografts in nude mice exhibited a slower onset of growth and a higher number of small tumors than ASCT2wt 143B xenografts, but did not differ in average tumor size 25 days after xenotransplantation. ASCT2 deficiency was compensated by increased levels of sodium neutral amino acid transporter 1 (SNAT1 or SLC38A1) and SNAT2 (SLC38A2) in ASCT2ko 143B cells, mediated by a GCN2 EIF2α kinase (GCN2)-dependent pathway, but this compensation was not observed in ASCT2ko HCC1806 cells. Combined SNAT1 silencing and GCN2 inhibition significantly inhibited growth of ASCT2ko HCC1806 cells, but not of ASCT2ko 143B cells. Similarly, pharmacological inhibition of l-type amino acid transporter 1 (LAT1) and GCN2 significantly inhibited growth of ASCT2ko HCC1806 cells, but not of ASCT2ko 143B cells. We conclude that cancer cells with reduced transporter plasticity are more vulnerable to disruption of amino acid homeostasis than cells with a full capacity to up-regulate redundant transporters by an integrated stress response.
Collapse
Affiliation(s)
- Angelika Bröer
- From the Research School of Biology, The Australian National University, Canberra, Australian Capital Territory 2601, Australia
| | - Gregory Gauthier-Coles
- From the Research School of Biology, The Australian National University, Canberra, Australian Capital Territory 2601, Australia
| | - Farid Rahimi
- From the Research School of Biology, The Australian National University, Canberra, Australian Capital Territory 2601, Australia
| | - Michelle van Geldermalsen
- Origins of Cancer Program, Centenary Institute, University of Sydney, Sydney, New South Wales 2050, Australia.,Sydney Medical School, University of Sydney, Sydney, New South Wales 2006, Australia
| | | | | | - Jeff Holst
- School of Medical Sciences and Prince of Wales Clinical School, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Stefan Bröer
- From the Research School of Biology, The Australian National University, Canberra, Australian Capital Territory 2601, Australia,
| |
Collapse
|
202
|
Abstract
Amino acids are an alternate energy source to glucose, and amino acid metabolism is up-regulated in multiple malignancies, including breast cancers. Multiple amino acid radiotracers have been used to image breast cancer with unique strengths and weaknesses. 11C-methionine uptake correlates with S-phase fraction in breast cancer and may be useful for evaluation of treatment response. Invasive lobular breast cancers may demonstrate greater 18F-fluciclovine avidity than 18F-fluorodeoxyglucose. Thus, different histologic subtypes of breast cancer may use diverse metabolic pathways and may be better imaged by different tracers.
Collapse
Affiliation(s)
- Gary A Ulaner
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, Box 77, New York, NY 10065, USA; Department of Radiology, Weill Cornell Medical School, 525 East 68th Street, New York, NY 10065, USA.
| | - David M Schuster
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology and Imaging Sciences, Emory University Hospital, Room E152, 1364 Clifton Road, Atlanta, GA 30322, USA
| |
Collapse
|
203
|
Bai M, Liu H, Xu K, Yu R, Oso AO, Deng J, Yin Y. Effects of coated cysteamine hydrochloride on muscle fiber characteristics and amino acid composition of finishing pigs. ASIAN-AUSTRALASIAN JOURNAL OF ANIMAL SCIENCES 2018; 32:1430-1438. [PMID: 30381744 PMCID: PMC6722302 DOI: 10.5713/ajas.18.0414] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 09/28/2018] [Indexed: 02/07/2023]
Abstract
Objective This experiment was designed to determine the effects of coated cysteamine hydrochloride (CC) on muscle fiber characteristics, amino acid composition and transporters gene expression in the longissimus dorsi muscle (LDM) of finishing pigs. Methods Two hundred and sixteen Duroc/Landrace/Yorkshire cross-bred male finishing pigs were fed with a corn-soybean basal diet supplemented with 0, 70, and 140 mg/kg cysteamine. Each group contained eight replicates of nine pigs per replicate. After 29 days, one pig was randomly selected from each replicate and slaughtered. Blood and LDM samples were collected and analyzed. Results The results showed that supplemental dietary CC increased (p<0.05) the muscle fiber density. And CC supplementation also up-regulated (p<0.05) the expression of myosin heavy chain 1 (MyHC1) and MyHC2x mRNA levels, and down-regulated (p<0.05) MyHC2b expression in the LDM. Additionally, supplemental dietary CC reduced (p<0.05) the concentration of total cholesterol in the plasma and enhanced (p<0.05) the concentrations of essential amino acid and total amino acid in the LDM. The relative expression levels of chloramphenicol acetyltransferase 2, b0,+ amino acid transporter, and y+-L-type amino acid transporter 1 were up-regulated (p<0.05) in the LDM when pigs were fed with the dietary CC of 70 mg/kg. Conclusion Cysteamine supplementation could increase fiber density and distribution of fiber types. It also improved the deposition of protein in the LDM by up-regulated the expression of amino acid transporters.
Collapse
Affiliation(s)
- Miaomiao Bai
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center for Healthy Breeding of Livestock and Poultry, Key Laboratory of Agro-ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China.,College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Hongnan Liu
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center for Healthy Breeding of Livestock and Poultry, Key Laboratory of Agro-ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China.,Hangzhou King Techina Technology Company Academician Expert Workstation, Hangzhou King Techina Technology Co., Ltd., Hangzhou 311107, China.,Hunan Co-Innovation Center of Animal Production Safety, CICAPS, Changsha, Hunan 410128, China
| | - Kang Xu
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center for Healthy Breeding of Livestock and Poultry, Key Laboratory of Agro-ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China.,Hangzhou King Techina Technology Company Academician Expert Workstation, Hangzhou King Techina Technology Co., Ltd., Hangzhou 311107, China.,Hunan Co-Innovation Center of Animal Production Safety, CICAPS, Changsha, Hunan 410128, China
| | - Rong Yu
- Hangzhou King Techina Technology Company Academician Expert Workstation, Hangzhou King Techina Technology Co., Ltd., Hangzhou 311107, China
| | - Abimbola Oladele Oso
- Department of Animal Nutrition, College of Animal Science and Livestock Production, Federal University of Agriculture, Abeokuta PMB 2240, Nigeria
| | - Jinping Deng
- College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Yulong Yin
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center for Healthy Breeding of Livestock and Poultry, Key Laboratory of Agro-ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China.,College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China.,Hangzhou King Techina Technology Company Academician Expert Workstation, Hangzhou King Techina Technology Co., Ltd., Hangzhou 311107, China.,Hunan Co-Innovation Center of Animal Production Safety, CICAPS, Changsha, Hunan 410128, China
| |
Collapse
|
204
|
Sannananja B, Shah HU, Behnia F. 18F-Fluciclovine Uptake by an Incidentally Detected Hepatocellular Carcinoma in a Case of Biochemically Recurrent Prostate Cancer. Clin Nucl Med 2018; 43:695-696. [PMID: 29939954 DOI: 10.1097/rlu.0000000000002176] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Prostate cancer is one of the most common cancers affecting men worldwide with a high recurrence rate following therapy. F-fluciclovine, is a US Food and Drug Administration-approved radiopharmaceutical for PET imaging in biochemically recurrent prostate cancer. It targets increased amino acid transporters in the cell membrane of cancer cells. We report a case of incidentally detected hepatocellular carcinoma showing F-fluciclovine uptake in a 71-year-old man with biochemically recurrent prostate cancer.
Collapse
Affiliation(s)
- Bhagya Sannananja
- From the Division of Nuclear Medicine, Department of Radiology, University of Washington School of Medicine, Seattle, WA
| | | | | |
Collapse
|
205
|
Miranda-Gonçalves V, Lameirinhas A, Henrique R, Jerónimo C. Metabolism and Epigenetic Interplay in Cancer: Regulation and Putative Therapeutic Targets. Front Genet 2018; 9:427. [PMID: 30356832 PMCID: PMC6190739 DOI: 10.3389/fgene.2018.00427] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Accepted: 09/10/2018] [Indexed: 12/31/2022] Open
Abstract
Alterations in the epigenome and metabolism affect molecular rewiring of cancer cells facilitating cancer development and progression. Modulation of histone and DNA modification enzymes occurs owing to metabolic reprogramming driven by oncogenes and expression of metabolism-associated genes is, in turn, epigenetically regulated, promoting the well-known metabolic reprogramming of cancer cells and, consequently, altering the metabolome. Thus, several malignant traits are supported by the interplay between metabolomics and epigenetics, promoting neoplastic transformation. In this review we emphasize the importance of tumour metabolites in the activity of most chromatin-modifying enzymes and implication in neoplastic transformation. Furthermore, candidate targets deriving from metabolism of cancer cells and altered epigenetic factors is emphasized, focusing on compounds that counteract the epigenomic-metabolic interplay in cancer.
Collapse
Affiliation(s)
- Vera Miranda-Gonçalves
- Cancer Biology and Epigenetics Group, Research Center (CI-IPOP), Portuguese Oncology Institute of Porto, Porto, Portugal
| | - Ana Lameirinhas
- Cancer Biology and Epigenetics Group, Research Center (CI-IPOP), Portuguese Oncology Institute of Porto, Porto, Portugal.,Master in Oncology, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Rui Henrique
- Cancer Biology and Epigenetics Group, Research Center (CI-IPOP), Portuguese Oncology Institute of Porto, Porto, Portugal.,Department of Pathology, Portuguese Oncology Institute of Porto, Porto, Portugal.,Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Carmen Jerónimo
- Cancer Biology and Epigenetics Group, Research Center (CI-IPOP), Portuguese Oncology Institute of Porto, Porto, Portugal.,Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| |
Collapse
|
206
|
Kaira K, Kawashima O, Endoh H, Imaizumi K, Goto Y, Kamiyoshihara M, Sugano M, Yamamoto R, Osaki T, Tanaka S, Fujita A, Imai H, Kogure Y, Seki Y, Shimizu K, Mogi A, Shitara Y, Oyama T, Kanai Y, Asao T. Expression of amino acid transporter (LAT1 and 4F2hc) in pulmonary pleomorphic carcinoma. Hum Pathol 2018; 84:142-149. [PMID: 30300664 DOI: 10.1016/j.humpath.2018.09.020] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 09/26/2018] [Accepted: 09/29/2018] [Indexed: 10/28/2022]
Abstract
Amino acid transporters are necessary for tumor growth, metastasis, and survival of various neoplasms; however, the clinicopathological significance of L-type amino acid transporter 1 (LAT1) and 4F2 cell surface antigen (4F2hc) in patients with pulmonary pleomorphic carcinoma (PPC) remainsunknown. The aim of this study is to clarify the prognostic impact of these amino acid transporters in PPC. One hundred five patients with surgically resected PPC were assessed by immunohistochemistry. The expression of LAT1 and 4F2hc, and Ki-67 labeling index were investigated using specimens of the resected tumors. LAT1 and 4F2hc were highly expressed in 35% and 53% of all patients (n = 105, P < .01), 25% and 48% of patients with an adenocarcinoma component (n = 48, P = .02), and 44% and 58% of patients with a nonadenocarcinoma component (n = 57, P = .18), respectively. A high LAT1 expression was significantly related to advanced disease, lymphatic permeation, tumor cell proliferation, and 4F2hc expression. By multivariate analysis, LAT1 and 4F2hc were identified as significant independent markers for predicting a worse prognosis. LAT1 is highly expressed in PPC, and high LAT1 expression can serve as a significant predictor linked to a worse prognosis in patients with PPC.
Collapse
Affiliation(s)
- Kyoichi Kaira
- Department of Oncology Clinical Development, Gunma University Graduate School of Medicine, Gunma 371-8511, Japan.
| | - Osamu Kawashima
- Department of Respiratory Surgery, Shibukawa Medical Center, Gunma 377-0280, Japan
| | - Hedeki Endoh
- Department of Thoracic Surgery, Saku Central Hospital Advanced Care Center, Nagano 385-0051, Japan
| | - Kazuyoshi Imaizumi
- Department of Respiratory Medicine, Fujita Health University, Aichi 470-1192, Japan
| | - Yasuhiro Goto
- Department of Respiratory Medicine, Fujita Health University, Aichi 470-1192, Japan
| | - Mitsuhiro Kamiyoshihara
- Department of Respiratory Surgery, Japanese Red Cross Maebashi Hospital, Gunma 371-0014, Japan
| | - Masayuki Sugano
- Department of Respiratory Surgery, Takasaki Medical Center, Gunma 370-0829, Japan
| | - Ryohei Yamamoto
- Department of Thoracic Surgery, Saku Central Hospital Advanced Care Center, Nagano 385-0051, Japan
| | - Takashi Osaki
- Department of Respiratory Medicine, Shibukawa Medical Center, Gunma 377-0280, Japan
| | - Shigefumi Tanaka
- Department of Respiratory Surgery, Isesaki Municipal Hospital, Gunma 372-0817, Japan
| | - Atsushi Fujita
- Department of Respiratory Surgery, Gunma Prefectural Cancer Center, Gunma 373-8550, Japan
| | - Hisao Imai
- Department of Respiratory Medicine, Gunma Prefectural Cancer Center, Gunma 373-8550, Japan
| | - Yoshihito Kogure
- Department of Respiratory Medicine, Nagoya Medical Center, Aichi 460-0001, Japan
| | - Yukio Seki
- Department of Thoracic Surgery, Nagoya Medical Center, Aichi 460-0001, Japan
| | - Kimihiro Shimizu
- Department of General Surgical Science, Gunma University, Graduate School of Medicine, Gunma 371-8511, Japan
| | - Akira Mogi
- Department of General Surgical Science, Gunma University, Graduate School of Medicine, Gunma 371-8511, Japan
| | - Yoshinori Shitara
- Department of Respiratory Surgery, Fujioka General Hospital, Gunma 375-0015, Japan
| | - Tetsunari Oyama
- Department of Diagnostic Pathology, Gunma University Graduate School of Medicine, Gunma 371-8511, Japan
| | - Yoshikatsu Kanai
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Takayuki Asao
- Big Data Center for Integrative Analysis, Gunma University Initiative for Advance Research, Gunma 371-8511, Japan
| |
Collapse
|
207
|
Geoghegan D, Arnall C, Hatton D, Noble-Longster J, Sellick C, Senussi T, James DC. Control of amino acid transport into Chinese hamster ovary cells. Biotechnol Bioeng 2018; 115:2908-2929. [PMID: 29987891 DOI: 10.1002/bit.26794] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 06/06/2018] [Accepted: 07/04/2018] [Indexed: 12/21/2022]
Abstract
Amino acid transporters (AATs) represent a key interface between the cell and its environment, critical for all cellular processes: Energy generation, redox control, and synthesis of cell and product biomass. However, very little is known about the activity of different functional classes of AATs in Chinese hamster ovary (CHO) cells, how they support cell growth and productivity, and the potential for engineering their activity and/or the composition of amino acids in growth media to improve CHO cell performance in vitro. In this study, we have comparatively characterized AAT expression in untransfected and monoclonal antibody (MAb)-producing CHO cells using transcriptome analysis by RNA-seq, and mechanistically dissected AAT function using a variety of transporter-specific chemical inhibitors, comparing their effect on cell proliferation, recombinant protein production, and amino acid transport. Of a possible 56 mammalian plasma membrane AATs, 16 AAT messenger RNAs (mRNAs) were relatively abundant across all CHO cell populations. Of these, a subset of nine AAT mRNAs were more abundant in CHO cells engineered to produce a recombinant MAb. Together, upregulated AATs provide additional supply of specific amino acids overrepresented in MAb biomass compared to CHO host cell biomass, enable transport of synthetic substrates for glutathione synthesis, facilitate transport of essential amino acids to maintain active protein synthesis, and provide amino acid substrates for coordinated antiport systems to maintain supplies of proteinogenic and essential amino acids.
Collapse
Affiliation(s)
- Darren Geoghegan
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield, UK
| | - Claire Arnall
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield, UK
| | | | - Joanne Noble-Longster
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield, UK
| | | | | | - David C James
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield, UK
| |
Collapse
|
208
|
Md Fuzi AA, Omar SZ, Mohamed Z, Mat Adenan NA, Mokhtar NM. High throughput silencing identifies novel genes in endometrioid endometrial cancer. Taiwan J Obstet Gynecol 2018; 57:217-226. [PMID: 29673664 DOI: 10.1016/j.tjog.2018.02.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/17/2017] [Indexed: 01/18/2023] Open
Abstract
OBJECTIVE To validate the gene expression profile obtained from the previous microarray analysis and to further study the biological functions of these genes in endometrial cancer. From our previous study, we identified 621 differentially expressed genes in laser-captured microdissected endometrioid endometrial cancer as compared to normal endometrial cells. Among these genes, 146 were significantly up-regulated in endometrial cancer. MATERIALS AND METHODS A total of 20 genes were selected from the list of up-regulated genes for the validation assay. The qPCR confirmed that 19 out of the 20 genes were up-regulated in endometrial cancer compared with normal endometrium. RNA interference (RNAi) was used to knockdown the expression of the upregulated genes in ECC-1 and HEC-1A endometrial cancer cell lines and its effect on proliferation, migration and invasion were examined. RESULTS Knockdown of MIF, SOD2, HIF1A and SLC7A5 by RNAi significantly decreased the proliferation of ECC-1 cells (p < 0.05). Our results also showed that the knockdown of MIF, SOD2 and SLC7A5 by RNAi significantly decreased the proliferation and migration abilities of HEC-1A cells (p < 0.05). Moreover, the knockdown of SLC38A1 and HIF1A by RNAi resulted in a significant decrease in the proliferation of HEC1A cells (p < 0.05). CONCLUSION We have identified the biological roles of SLC38A1, MIF, SOD2, HIF1A and SLC7A5 in endometrial cancer, which opens up the possibility of using the RNAi silencing approach to design therapeutic strategies for treatment of endometrial cancer.
Collapse
Affiliation(s)
- Afiqah Alyaa Md Fuzi
- Department of Pharmacology, Faculty of Medicine, University Malaya, 50603 Kuala Lumpur, Malaysia
| | - Siti Zawiah Omar
- Department of Obstetrics and Gynecology, Faculty of Medicine, University Malaya, 50603 Kuala Lumpur, Malaysia
| | - Zahurin Mohamed
- Department of Pharmacology, Faculty of Medicine, University Malaya, 50603 Kuala Lumpur, Malaysia
| | - Noor Azmi Mat Adenan
- Department of Obstetrics and Gynecology, Faculty of Medicine, University Malaya, 50603 Kuala Lumpur, Malaysia
| | - Norfilza Mohd Mokhtar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, 56000 Cheras, Kuala Lumpur, Malaysia.
| |
Collapse
|
209
|
Xia W, Osorio JS, Yang Y, Liu D, Jiang MF. Short communication: Characterization of gene expression profiles related to yak milk protein synthesis during the lactation cycle. J Dairy Sci 2018; 101:11150-11158. [PMID: 30268611 DOI: 10.3168/jds.2018-14715] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 08/06/2018] [Indexed: 12/17/2022]
Abstract
This research assessed the gene expression patterns related to the synthesis of milk in yak, which is characterized by high fat and protein content but low yield. The yak (Bos grunniens) is one of the most crucial domestic animals in Tibetan life; however, the genetic and molecular factors underlying yak milk protein synthesis remain understudied. Yak mammary biopsies harvested during late-pregnancy (d -15) through the end of subsequent lactation (d 1, 15, 30, 60, 180, and 240) were used to evaluate gene expression via real-time quantitative PCR. The expression pattern of 41 genes encompassing multiple pathways integral to milk protein synthesis including insulin, mammalian target of rapamycin (mTOR), 5' AMP-activated protein kinase, Jak2-Stat5 signaling, and the expression of glucose and AA transporters was evaluated. Our results confirmed that most upregulated genes increased from d -15 and peaked at d 30 or 60 and then remained relatively highly expressed. Specifically, there was an increased expression of mTOR-related amino acid transporters (SLC1A5, SLC7A5, and SLC36A1), glucose transporters (SLC2A1, SLC2A3, and SLC2A8), Jak2-Stat5 pathway (ELF5), and insulin signaling pathway components (IRS1, PDPK1, and AKT1). For activation of proteins synthesis, MTOR was significantly increased only at d 1. Among inhibitors of mTOR signaling, TSC1 and PRKAA2 were significantly upregulated during lactation. The RPL23 was downregulated among ribosomal components. In conclusion, a critical role for AA and glucose transporters and insulin signaling through mTOR for regulation of yak milk protein synthesis was revealed in this study of the yak mammary gland.
Collapse
Affiliation(s)
- Wei Xia
- College of Life Science and Technology, Southwest Minzu University, Chengdu 610041, China; Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Conservation and Exploitation, Key Laboratory of Animal Genetics & Breeding of State Ethnic Affairs Commission and Ministry of Education, Southwest University for Nationalities, Chengdu 610041, China
| | - Johan S Osorio
- Dairy and Food Science Department, South Dakota State University, Brookings 57007
| | - Yuanxiao Yang
- College of Life Science and Technology, Southwest Minzu University, Chengdu 610041, China; Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Conservation and Exploitation, Key Laboratory of Animal Genetics & Breeding of State Ethnic Affairs Commission and Ministry of Education, Southwest University for Nationalities, Chengdu 610041, China
| | | | - Ming Feng Jiang
- College of Life Science and Technology, Southwest Minzu University, Chengdu 610041, China; Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Conservation and Exploitation, Key Laboratory of Animal Genetics & Breeding of State Ethnic Affairs Commission and Ministry of Education, Southwest University for Nationalities, Chengdu 610041, China.
| |
Collapse
|
210
|
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: 177] [Impact Index Per Article: 25.3] [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
|
211
|
Wang C, Wu J, Wang Z, Yang Z, Li Z, Deng H, Li L, Peng X, Feng M. Glutamine addiction activates polyglutamine-based nanocarriers delivering therapeutic siRNAs to orthotopic lung tumor mediated by glutamine transporter SLC1A5. Biomaterials 2018; 183:77-92. [PMID: 30149232 DOI: 10.1016/j.biomaterials.2018.08.035] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 08/03/2018] [Accepted: 08/18/2018] [Indexed: 01/09/2023]
Abstract
Many human cancer cells exhibit an oncogenetic-driven addiction to glutamine (Gln) as rapidly proliferating cancer cells consume Gln at a dramatically increased rate compared to normal cells. Tumor cells, therefore, compete with host cells for Gln, which causes Gln to flux from normal tissues to the tumor. We have developed and characterized a Gln macromolecular analog polyglutamine (PGS) for the delivery of gene regulators, such as siRNAs, in our previous works. Here, we hypothesize that PGS can utilize the Gln transporter SLC1A5 to specifically deliver therapeutic compounds to Gln-addicted cancer cells. Compared to human lung fibroblast HLF cells, cisplatin-resistant human lung adenocarcinoma A549/DDP cells significantly overexpress SLC1A5, which has a high binding affinity to PGS, as confirmed through molecular docking analysis. Due to the differences in Gln metabolism between malignant and normal cells, PGS/siRNA complexes were remarkably increased in cancer cells, especially when cells were deprived of Gln, which mirrors the conditions that are commonly found in a tumor microenvironment. Furthermore, we identified that chemical and genetic inhibition of Gln transporter SLC1A5 reduced the cellular internalization of PGS/siRNA complexes, suggesting a critical role for SLC1A5 in PGS uptake in cells. In turn, PGS upregulated SLC1A5 expression. Increased uptake of PGS complexes profoundly decreased intracellular Gln levels. Decreased Gln caused a moderate reduction in cell growth. To restore drug sensitivity and further enhance anti-tumor effects, the hybrid siRNAs anti-Survivin and anti-MDR1 (siSM), as model therapeutics, were administered through the PGS delivery system, which resulted in knockdown of Survivin and MDR1 and further sensitized cancer cells to the drug cisplatin (DDP). Since PGS complexes administered i.v. mostly accumulated in the lung parenchyma, a lung orthotopic tumor model was established to evaluate their inhibitory effects on tumors in the lungs. PGS/siSM comparably decreased the rate of tumor growth, while concurrent administration of PGS/siSM and DDP enhanced this effect and insignificantly improved life span. Consistent with our hypothesis, this study demonstrated that PGS mimicked Gln in the SLC1A5 pathway and selectively ferried therapeutics to Gln-addicted cancer cells. Our findings identified a new lung cancer targeting strategy based on Gln metabolism and can be used as a drug/gene delivery system.
Collapse
Affiliation(s)
- Cuifeng Wang
- School of Pharmaceutical Sciences, Sun Yat-sen University, University Town, Guangzhou 510006, PR China.
| | - Jiamin Wu
- School of Pharmaceutical Sciences, Sun Yat-sen University, University Town, Guangzhou 510006, PR China; School of Pharmacy, Guangdong Medical University, Dongguan 523808, PR China
| | - Zhongjuan Wang
- School of Pharmaceutical Sciences, Sun Yat-sen University, University Town, Guangzhou 510006, PR China; Department of Pharmacy, Yan'an Hospital Affiliated to Kunming Medical University, Kunming 650051, PR China
| | - Zeping Yang
- School of Pharmaceutical Sciences, Sun Yat-sen University, University Town, Guangzhou 510006, PR China
| | - Zhi Li
- School of Pharmaceutical Sciences, Sun Yat-sen University, University Town, Guangzhou 510006, PR China
| | - Huihui Deng
- School of Pharmaceutical Sciences, Sun Yat-sen University, University Town, Guangzhou 510006, PR China
| | - Long Li
- School of Pharmaceutical Sciences, Sun Yat-sen University, University Town, Guangzhou 510006, PR China
| | - Xiao Peng
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, PR China
| | - Min Feng
- School of Pharmaceutical Sciences, Sun Yat-sen University, University Town, Guangzhou 510006, PR China.
| |
Collapse
|
212
|
|
213
|
Bröer A, Fairweather S, Bröer S. Disruption of Amino Acid Homeostasis by Novel ASCT2 Inhibitors Involves Multiple Targets. Front Pharmacol 2018; 9:785. [PMID: 30072900 PMCID: PMC6060247 DOI: 10.3389/fphar.2018.00785] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 06/27/2018] [Indexed: 12/02/2022] Open
Abstract
The glutamine transporter ASCT2 (SLC1A5) is actively investigated as an oncological target, but the field lacks efficient ASCT2 inhibitors. A new group of ASCT2 inhibitors, 2-amino-4-bis(aryloxybenzyl)aminobutanoic acids (AABA), were developed recently and shown to suppress tumor growth in preclinical in vivo models. To test its specificity, we deleted ASCT2 in two human cancer cell lines. Surprisingly, growth of parental and ASCT2-knockout cells was equally sensitive to AABA compounds. AABA compounds inhibited glutamine transport in cells lacking ASCT2, but not in parental cells. Deletion of ASCT2 and amino acid (AA) depletion induced expression of SNAT2 (SLC38A2), the activity of which was inhibited by AABA compounds. They also potently inhibited isoleucine uptake via LAT1 (SLC7A5), a transporter that is upregulated in cancer cells together with ASCT2. Inhibition of SNAT2 and LAT1 was confirmed by recombinant expression in Xenopus laevis oocytes. The reported reduction of tumor growth in pre-clinical models may be explained by a significant disruption of AA homeostasis.
Collapse
Affiliation(s)
- Angelika Bröer
- Research School of Biology, Australian National University, Canberra, ACT, Australia
| | - Stephen Fairweather
- Research School of Biology, Australian National University, Canberra, ACT, Australia
| | - Stefan Bröer
- Research School of Biology, Australian National University, Canberra, ACT, Australia
| |
Collapse
|
214
|
Bothwell PJ, Kron CD, Wittke EF, Czerniak BN, Bode BP. Targeted Suppression and Knockout of ASCT2 or LAT1 in Epithelial and Mesenchymal Human Liver Cancer Cells Fail to Inhibit Growth. Int J Mol Sci 2018; 19:ijms19072093. [PMID: 30029480 PMCID: PMC6073291 DOI: 10.3390/ijms19072093] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 07/05/2018] [Indexed: 02/02/2023] Open
Abstract
Amino acid transporters alanine-serine-cysteine transporter 2 (ASCT2) and L-Type Amino Acid Transporter 1 (LAT1) are coordinately enhanced in human cancers where among other roles, they are thought to drive mechanistic target-of-rapamycin (mTOR) growth signaling. To assess ASCT2 and LAT1 as therapeutic targets, nine unique short hairpin RNA (shRNA) vectors were used to stably suppress transporter expression in human epithelial (Hep3B) and mesenchymal (SK-Hep1) hepatocellular carcinoma (HCC) cell lines. In addition, six unique CRISPR-Cas9 vectors were used to edit the ASCT2 (SLC1A5) and LAT1 (SLC7A5) genes in epithelial (HUH7) and mesenchymal (SK-Hep1) HCC cells. Both approaches successfully diminished glutamine (ASCT2) and leucine (LAT1) initial-rate transport proportional to transporter protein suppression. In spite of profoundly reduced glutamine or leucine transport (up to 90%), transporter suppression or knockout failed to substantially affect cellular proliferation or basal and amino acid-stimulated mTORC1 growth signaling in either HCC cell type. Only LAT1 knockout in HUH7 slightly reduced growth rate. However, intracellular accumulation of radiolabeled glutamine and leucine over longer time periods largely recovered to control levels in ASCT2 and LAT1 knockout cells, respectively, which partially explains the lack of an impaired growth phenotype. These data collectively establish that in an in vitro context, human epithelial and mesenchymal HCC cell lines adapt to ASCT2 or LAT1 knockout. These results comport with an emerging model of amino acid exchangers like ASCT2 and LAT1 as “harmonizers”, not drivers, of amino acid accumulation and signaling, despite their long-established dominant role in initial-rate amino acid transport.
Collapse
Affiliation(s)
- Paige J Bothwell
- Department of Biological Sciences/Center for Biochemical and Biophysical Studies, Northern Illinois University, DeKalb, IL 60115, USA.
| | - Clare D Kron
- Department of Biological Sciences/Center for Biochemical and Biophysical Studies, Northern Illinois University, DeKalb, IL 60115, USA.
| | - Evan F Wittke
- Department of Biological Sciences/Center for Biochemical and Biophysical Studies, Northern Illinois University, DeKalb, IL 60115, USA.
| | - Bradley N Czerniak
- Department of Biological Sciences/Center for Biochemical and Biophysical Studies, Northern Illinois University, DeKalb, IL 60115, USA.
| | - Barrie P Bode
- Department of Biological Sciences/Center for Biochemical and Biophysical Studies, Northern Illinois University, DeKalb, IL 60115, USA.
| |
Collapse
|
215
|
Prakash S, Hazari PP, Meena VK, Mishra AK. Radiolabeling and Preclinical Evaluation of a New S-Alkylated Cysteine Derivative Conjugated to C-Substituted Macrocycle for Positron Emission Tomography. ACS OMEGA 2018; 3:6497-6505. [PMID: 30023950 PMCID: PMC6045381 DOI: 10.1021/acsomega.8b00059] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 06/05/2018] [Indexed: 06/08/2023]
Abstract
A new S-alkylated cysteine-derivatized tumor targeting agent, 2,2'-(12-(2-((2-acetamido-2-carboxyethyl)thio)acetamido)-11,13-dioxo-1,4,7,10-tetraazacyclotridecane-4,7-diyl)diacetic acid was developed for positron emission tomography (PET) imaging. N-Acetyl cysteine (NAC) was conjugated to ATRIDAT as a specific targeting agent toward L-type and ASC amino acid transporter systems in the oncogenic cells. NAC was attached via S-alkylation to prevent its incorporation at undesired recognition sites affecting the signal-to-noise ratio. NAC-ATRIDAT was subjected to gallium-68 complexation with >75% radiolabeling yield. The radiocomplex was purified through the tc18 cartridge to obtain 99.89% radiochemical yield. IC-50 of the NAC-ATRIDAT conjugate was 0.8 mM in A549 cells as evaluated through 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazonium bromide assay. Binding affinity experiments on A549 cells showed noteworthy binding with KD in the nanomolar range. A time course study showed a Km value of 0.19 μM and Vmax value of 0.49 pmol/μg protein/min showing reasonable tumor kinetics. Efflux studies showed that the synthesized radioligand is transported majorly by LAT followed by the ASC system. Clearance was found to be renal with 7.67 ± 1.48% ID/g uptake at 30 min which substantially declined to 0.52 ± 0.% ID/g at 4 h. A significant uptake of 10.06 ± 1.056% ID/g was observed at the tumor site in mice at 1 h. μPET images revealed a high contrast with a tumor-to-kidney ratio of 4.8 and a tumor-to-liver ratio of 35.85 at 1 h after injection. These preclinical in vitro and in vivo evaluation supports its potential on the way of becoming a successful 68Ga-radiolabeled amino acid-based PET imaging agent.
Collapse
|
216
|
Scalise M, Galluccio M, Console L, Pochini L, Indiveri C. The Human SLC7A5 (LAT1): The Intriguing Histidine/Large Neutral Amino Acid Transporter and Its Relevance to Human Health. Front Chem 2018; 6:243. [PMID: 29988369 PMCID: PMC6023973 DOI: 10.3389/fchem.2018.00243] [Citation(s) in RCA: 197] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 06/06/2018] [Indexed: 12/22/2022] Open
Abstract
SLC7A5, known as LAT1, belongs to the APC superfamily and forms a heterodimeric amino acid transporter interacting with the glycoprotein CD98 (SLC3A2) through a conserved disulfide. The complex is responsible for uptake of essential amino acids in crucial body districts such as placenta and blood brain barrier. LAT1/CD98 heterodimer has been studied over the years to unravel the transport mechanism and the role of each subunit. Studies conducted in intact cells demonstrated that LAT1/CD98 mediates a Na+ and pH independent antiport of amino acids. Some novel insights into the function of LAT1 derived from studies conducted in proteoliposomes reconstituted with the recombinant human LAT1. Using this experimental tool, it has been demonstrated that the preferred substrate is histidine and that CD98 is not required for transport being, plausibly, involved in routing LAT1 to the plasma membrane. Since a 3D structure of LAT1 is not available, homology models have been built on the basis of the AdiC transporter from E.coli. Crucial residues for substrate recognition and gating have been identified using a combined approach of bioinformatics and site-directed mutagenesis coupled to functional assays. Over the years, the interest around LAT1 increased because this transporter is involved in important human diseases such as neurological disorders and cancer. Therefore, LAT1 became an important pharmacological target together with other nutrient membrane transporters. Moving from knowledge on structure/function relationships, two cysteine residues, lying on the substrate binding site, have been exploited for designing thiol reacting covalent inhibitors. Some lead compounds have been characterized whose efficacy has been tested in a cancer cell line.
Collapse
Affiliation(s)
- Mariafrancesca Scalise
- Unit of Biochemistry and Molecular Biotechnology, Department DiBEST (Biologia, Ecologia, Scienze della Terra), University of Calabria, Rende, Italy
| | - Michele Galluccio
- Unit of Biochemistry and Molecular Biotechnology, Department DiBEST (Biologia, Ecologia, Scienze della Terra), University of Calabria, Rende, Italy
| | - Lara Console
- Unit of Biochemistry and Molecular Biotechnology, Department DiBEST (Biologia, Ecologia, Scienze della Terra), University of Calabria, Rende, Italy
| | - Lorena Pochini
- Unit of Biochemistry and Molecular Biotechnology, Department DiBEST (Biologia, Ecologia, Scienze della Terra), University of Calabria, Rende, Italy
| | - Cesare Indiveri
- Unit of Biochemistry and Molecular Biotechnology, Department DiBEST (Biologia, Ecologia, Scienze della Terra), University of Calabria, Rende, Italy.,CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnology, Bari, Italy
| |
Collapse
|
217
|
Yang W, Zhu G, Zhou G, Liu Y, Shao J, Xie B. Alterations of glutamine and glutamate levels in patients and rats with hepatocellular carcinoma. J LIQ CHROMATOGR R T 2018. [DOI: 10.1080/10826076.2018.1485034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
- Wenlong Yang
- Department of Infectious Diseases, The Second Affiliated Hospital of Nanchang University, School of Pharmaceutical Science, Nanchang University, Nanchang, PR China
| | - Guojian Zhu
- Department of Infectious Diseases, The Second Affiliated Hospital of Nanchang University, School of Pharmaceutical Science, Nanchang University, Nanchang, PR China
- Department of Infectious Diseases, The Fifth People’s Hospital, Ganzhou, PR China
| | - Guanlin Zhou
- Department of Infectious Diseases, The Second Affiliated Hospital of Nanchang University, School of Pharmaceutical Science, Nanchang University, Nanchang, PR China
| | - Yan Liu
- Department of Infectious Diseases, The Second Affiliated Hospital of Nanchang University, School of Pharmaceutical Science, Nanchang University, Nanchang, PR China
| | - Jianghua Shao
- Jiangxi Provincial Key Laboratory of Molecular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, PR China
| | - Baogang Xie
- Department of Infectious Diseases, The Second Affiliated Hospital of Nanchang University, School of Pharmaceutical Science, Nanchang University, Nanchang, PR China
| |
Collapse
|
218
|
The Nutrient-Sensing Hexosamine Biosynthetic Pathway as the Hub of Cancer Metabolic Rewiring. Cells 2018; 7:cells7060053. [PMID: 29865240 PMCID: PMC6025041 DOI: 10.3390/cells7060053] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 05/29/2018] [Accepted: 05/31/2018] [Indexed: 12/12/2022] Open
Abstract
Alterations in glucose and glutamine utilizing pathways and in fatty acid metabolism are currently considered the most significant and prevalent metabolic changes observed in almost all types of tumors. Glucose, glutamine and fatty acids are the substrates for the hexosamine biosynthetic pathway (HBP). This metabolic pathway generates the “sensing molecule” UDP-N-Acetylglucosamine (UDP-GlcNAc). UDP-GlcNAc is the substrate for the enzymes involved in protein N- and O-glycosylation, two important post-translational modifications (PTMs) identified in several proteins localized in the extracellular space, on the cell membrane and in the cytoplasm, nucleus and mitochondria. Since protein glycosylation controls several key aspects of cell physiology, aberrant protein glycosylation has been associated with different human diseases, including cancer. Here we review recent evidence indicating the tight association between the HBP flux and cell metabolism, with particular emphasis on the post-transcriptional and transcriptional mechanisms regulated by the HBP that may cause the metabolic rewiring observed in cancer. We describe the implications of both protein O- and N-glycosylation in cancer cell metabolism and bioenergetics; focusing our attention on the effect of these PTMs on nutrient transport and on the transcriptional regulation and function of cancer-specific metabolic pathways.
Collapse
|
219
|
Matsuura M, Ohshima M, Hiruta Y, Nishimura T, Nagase K, Kanazawa H. LAT1-Targeting Thermoresponsive Fluorescent Polymer Probes for Cancer Cell Imaging. Int J Mol Sci 2018; 19:E1646. [PMID: 29865203 PMCID: PMC6032285 DOI: 10.3390/ijms19061646] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 05/24/2018] [Accepted: 05/25/2018] [Indexed: 01/31/2023] Open
Abstract
L-type amino acid transporter 1 (LAT1) is more highly expressed in cancer cells compared with normal cells. LAT1 targeting probes would therefore be a promising tool for cancer cell imaging. In this study, LAT1-targeting thermoresponsive fluorescent polymer probes based on poly(N-isopropylacrylamide-co-N,N-dimethylacrylamide) (P(NIPAAm-co-DMAAm)) were synthesized and their affinity for LAT1 was evaluated. The synthesized polymer probes interacted with LAT1 on HeLa cells, and inhibition of l-[³H]-leucine, one of the substrates for LAT1 uptake, was investigated. l-Tyrosine-conjugated P(NIPAAm-co-DMAAm) inhibited the uptake of l-[³H]-leucine, while P(NIPAAm-co-DMAAm) and l-phenylalanine-conjugated P(NIPAAm-co-DMAAm) did not. This result indicated that l-tyrosine-conjugated polymer has a high affinity for LAT1. The fluorescent polymer probes were prepared by modification of a terminal polymer group with fluorescein-5-maleimide (FL). Above the polymer transition temperature, cellular uptake of the polymer probes was observed because the polymers became hydrophobic, which enhanced the interaction with the cell membrane. Furthermore, quantitative analysis of the fluorescent probe using flow cytometry indicated that l-tyrosine-conjugated P(NIPAAm-co-DMAAm)-FL shows higher fluorescence intensity earlier than P(NIPAAm-co-DMAAm)-FL. The result suggested that cellular uptake was promoted by the LAT1 affinity site. The developed LAT1-targeting thermoresponsive fluorescent polymer probes are expected to be useful for cancer cell imaging.
Collapse
Affiliation(s)
- Minami Matsuura
- Faculty of Pharmacy, Keio University, 1-5-30, Shibakoen, Minato-ku, Tokyo 105-8012, Japan.
| | - Mariko Ohshima
- Faculty of Pharmacy, Keio University, 1-5-30, Shibakoen, Minato-ku, Tokyo 105-8012, Japan.
| | - Yuki Hiruta
- Department of Applied Chemistry, Faculty of Science and Technology, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama 223-8522, Japan.
| | - Tomohiro Nishimura
- Faculty of Pharmacy, Keio University, 1-5-30, Shibakoen, Minato-ku, Tokyo 105-8012, Japan.
| | - Kenichi Nagase
- Faculty of Pharmacy, Keio University, 1-5-30, Shibakoen, Minato-ku, Tokyo 105-8012, Japan.
| | - Hideko Kanazawa
- Faculty of Pharmacy, Keio University, 1-5-30, Shibakoen, Minato-ku, Tokyo 105-8012, Japan.
| |
Collapse
|
220
|
Kärkkäinen J, Gynther M, Kokkola T, Petsalo A, Auriola S, Lahtela-Kakkonen M, Laine K, Rautio J, Huttunen KM. Structural properties for selective and efficient l-type amino acid transporter 1 (LAT1) mediated cellular uptake. Int J Pharm 2018; 544:91-99. [DOI: 10.1016/j.ijpharm.2018.04.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 04/13/2018] [Accepted: 04/13/2018] [Indexed: 12/24/2022]
|
221
|
The receptor protein tyrosine phosphatase PTPRJ negatively modulates the CD98hc oncoprotein in lung cancer cells. Oncotarget 2018; 9:23334-23348. [PMID: 29805737 PMCID: PMC5955124 DOI: 10.18632/oncotarget.25101] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 03/24/2018] [Indexed: 01/16/2023] Open
Abstract
PTPRJ, a receptor protein tyrosine phosphatase strongly downregulated in human cancer, displays tumor suppressor activity by negatively modulating several proteins involved in proliferating signals. Here, through a proteomic-based approach, we identified a list of potential PTPRJ-interacting proteins and among them we focused on CD98hc, a type II glycosylated integral membrane protein encoded by SLC3A2, corresponding to the heavy chain of a heterodimeric transmembrane amino-acid transporter, including LAT1. CD98hc is widely overexpressed in several types of cancers and contributes to the process of tumorigenesis by interfering with cell proliferation, adhesion, and migration. We first validated PTPRJ-CD98hc interaction, then demonstrated that PTPRJ overexpression dramatically reduces CD98hc protein levels in A549 lung cancer cells. In addition, following to the treatment of PTPRJ-transduced cells with MG132, a proteasome inhibitor, CD98hc levels did not decrease compared to controls, indicating that PTPRJ is involved in the regulation of CD98hc proteasomal degradation. Moreover, PTPRJ overexpression combined with CD98hc silencing consistently reduced cell proliferation and triggered apoptosis of lung cancer cells. Interestingly, by interrogating the can Evolve database, we observed an inverse correlation between PTPRJ and SLC3A2 gene expression. Indeed, the non-small cell lung cancers (NSCLCs) of patients showing a short survival rate express the lowest and the highest levels of PTPRJ and SLC3A2, respectively. Therefore, the results reported here contribute to shed lights on PTPRJ signaling in cancer cells: moreover, our findings also support the development of a novel anticancer therapeutic approach by targeting the pathway of PTPRJ that is usually downregulated in highly malignant human neoplasias.
Collapse
|
222
|
Improved Radiosynthesis and Biological Evaluations of L- and D-1-[ 18F]Fluoroethyl-Tryptophan for PET Imaging of IDO-Mediated Kynurenine Pathway of Tryptophan Metabolism. Mol Imaging Biol 2018; 19:589-598. [PMID: 27815661 DOI: 10.1007/s11307-016-1024-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
PURPOSE Tryptophan metabolism via indoleamine 2,3-dioxygenase (IDO)-mediated kynurenine pathway plays a role in immunomodulation and has been emerging as a plausible target for cancer immunotherapy. Imaging IDO-mediated kynurenine pathway of tryptophan metabolism with positron emission tomography (PET) could provide valuable information for noninvasive assessment of cancer immunotherapy response. In this work, radiotracer 1-(2-[18F]fluoroethyl)-L-tryptophan (1-L-[18F]FETrp) and its enantioisomer 1-D-[18F]FETrp were synthesized and evaluated for PET imaging of IDO-mediated kynurenine pathway of tryptophan metabolism. PROCEDURES Enantiopure 1-L-[18F]FETrp and 1-D-[18F]FETrp were prepared by a nucleophilic reaction of N-boc-1-(2-tosylethyl) tryptophan tert-butyl ester with [18F]Fluoride, followed by acid hydrolysis in a GE Tracerlab FX-N module. In vitro cell uptake assays were performed with a breast cancer cell line MDA-MB-231. Small animal PET/computed tomography (CT) imaging was carried out in a mouse model bearing MDA-MB-231 xenografts. RESULTS Automatic radiosynthesis of 1-L-[18F]FETrp and 1-D-[18F]FETrp was achieved by a one-pot two-step approach in 19.0 ± 7.0 and 9.0 ± 3.0 % (n = 3) decay-corrected yield with radiochemical purity over 99 %, respectively. In vitro cell uptake study indicated the uptake of 1-D-[18F]FETrp in MDA-MB-231 cells was 0.73 ± 0.07 %/mg of protein at 60 min, while, the corresponding uptake of 1-L-[18F]FETrp was 6.60 ± 0.77 %/mg. Further mechanistic assays revealed that amino acid transport systems L-tpye amino acid transporter (LAT) and alanine-, serine-, and cysteine-preferring (ASC), and enzyme IDO expression were involved in cell uptake of 1-L-[18F]FETrp. Small animal PET/CT imaging study showed the tumor uptake of 1-L-[18F]FETrp was 4.6 ± 0.4 % ID/g, while, the tumor uptake of 1-D-[18F]FETrp was low to 1.0 ± 0.2 % ID/g, which were confirmed by ex vivo biodistribution study. CONCLUSIONS We have developed a practical method for the automatic radiosynthesis of 1-L-[18F]FETrp and 1-D-[18F]FETrp. Our biological evaluation results suggest that 1-L-[18F]FETrp is a promising radiotracer for PET imaging of IDO-mediated kynurenine pathway of tryptophan metabolism in cancer.
Collapse
|
223
|
Chen Q, Kirk K, Shurubor YI, Zhao D, Arreguin AJ, Shahi I, Valsecchi F, Primiano G, Calder EL, Carelli V, Denton TT, Beal MF, Gross SS, Manfredi G, D'Aurelio M. Rewiring of Glutamine Metabolism Is a Bioenergetic Adaptation of Human Cells with Mitochondrial DNA Mutations. Cell Metab 2018; 27:1007-1025.e5. [PMID: 29657030 PMCID: PMC5932217 DOI: 10.1016/j.cmet.2018.03.002] [Citation(s) in RCA: 123] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 01/03/2018] [Accepted: 03/12/2018] [Indexed: 01/05/2023]
Abstract
Using molecular, biochemical, and untargeted stable isotope tracing approaches, we identify a previously unappreciated glutamine-derived α-ketoglutarate (αKG) energy-generating anaplerotic flux to be critical in mitochondrial DNA (mtDNA) mutant cells that harbor human disease-associated oxidative phosphorylation defects. Stimulating this flux with αKG supplementation enables the survival of diverse mtDNA mutant cells under otherwise lethal obligatory oxidative conditions. Strikingly, we demonstrate that when residual mitochondrial respiration in mtDNA mutant cells exceeds 45% of control levels, αKG oxidative flux prevails over reductive carboxylation. Furthermore, in a mouse model of mitochondrial myopathy, we show that increased oxidative αKG flux in muscle arises from enhanced alanine synthesis and release into blood, concomitant with accelerated amino acid catabolism from protein breakdown. Importantly, in this mouse model of mitochondriopathy, muscle amino acid imbalance is normalized by αKG supplementation. Taken together, our findings provide a rationale for αKG supplementation as a therapeutic strategy for mitochondrial myopathies.
Collapse
Affiliation(s)
- Qiuying Chen
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Kathryne Kirk
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Yevgeniya I Shurubor
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Dazhi Zhao
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Andrea J Arreguin
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Ifrah Shahi
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Federica Valsecchi
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Guido Primiano
- Institute of Neurology, Catholic University of the Sacred Heart, Rome, Italy
| | - Elizabeth L Calder
- Center for Stem Cell Biology and Developmental Biology Program, Sloan-Kettering Institute for Cancer Research, New York, NY 10065, USA
| | - Valerio Carelli
- IRCCS, Institute of Neurological Sciences of Bologna, Bellaria Hospital, Bologna, Italy; Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| | - Travis T Denton
- Department of Pharmaceutical Sciences, Washington State University, College of Pharmacy, Spokane, WA 99210, USA
| | - M Flint Beal
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Steven S Gross
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Giovanni Manfredi
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA.
| | - Marilena D'Aurelio
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA.
| |
Collapse
|
224
|
Singh N, Ecker GF. Insights into the Structure, Function, and Ligand Discovery of the Large Neutral Amino Acid Transporter 1, LAT1. Int J Mol Sci 2018; 19:E1278. [PMID: 29695141 PMCID: PMC5983779 DOI: 10.3390/ijms19051278] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 04/18/2018] [Accepted: 04/19/2018] [Indexed: 12/31/2022] Open
Abstract
The large neutral amino acid transporter 1 (LAT1, or SLC7A5) is a sodium- and pH-independent transporter, which supplies essential amino acids (e.g., leucine, phenylalanine) to cells. It plays an important role at the Blood⁻Brain Barrier (BBB) where it facilitates the transport of thyroid hormones, pharmaceuticals (e.g., l-DOPA, gabapentin), and metabolites into the brain. Moreover, its expression is highly upregulated in various types of human cancer that are characterized by an intense demand for amino acids for growth and proliferation. Therefore, LAT1 is believed to be an important drug target for cancer treatment. With the crystallization of the arginine/agmatine antiporter (AdiC) from Escherichia Coli, numerous homology models of LAT1 have been built to elucidate the substrate binding site, ligand⁻transporter interaction, and structure⁻function relationship. The use of these models in combination with molecular docking and experimental testing has identified novel chemotypes of ligands of LAT1. Here, we highlight the structure, function, transport mechanism, and homology modeling of LAT1. Additionally, results from structure⁻function studies performed on LAT1 are addressed, which have enhanced our knowledge of the mechanism of substrate binding and translocation. This is followed by a discussion on ligand- and structure-based approaches, with an emphasis on elucidating the molecular basis of LAT1 inhibition. Finally, we provide an exhaustive summary of different LAT1 inhibitors that have been identified so far, including the recently discovered irreversible covalent inhibitors.
Collapse
Affiliation(s)
- Natesh Singh
- Department of Pharmaceutical Chemistry, University of Vienna, Althanstrasse 14, 1090 Wien, Austria.
| | - Gerhard F Ecker
- Department of Pharmaceutical Chemistry, University of Vienna, Althanstrasse 14, 1090 Wien, Austria.
| |
Collapse
|
225
|
Chitgupi U, Lovell JF, Rajendiran V. Assessing Photosensitizer Targeting Using Meso-Tetra(Carboxyphenyl) Porphyrin. Molecules 2018; 23:molecules23040892. [PMID: 29649139 PMCID: PMC6017280 DOI: 10.3390/molecules23040892] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 03/28/2018] [Accepted: 04/10/2018] [Indexed: 11/16/2022] Open
Abstract
Mesotetra(4-carboxyphenyl)porphyrin (mTCPP) is a commercially available small molecule fluorophore and photosensitizer with four free carboxylic acid groups. mTCPP can readily be conjugated with amines for facile attachment of functional groups. In this work, we synthesized and assessed tetravalent, lysine-conjugated mTCPP, for its potential applications in targeted imaging and photodynamic therapy. Fmoc-protected d-lysine or l-lysine was conjugated to mTCPP via amide coupling with the epsilon amine group of lysine, followed by Fmoc deprotection. The resulting compounds did not dissolve well in aqueous solvent, but could be solubilized with the assistance of surfactants, including cholic acid. The l-amino acid transporter (LAT1) can uptake diverse neutral l-amino acids. In vitro studies with U87 cells revealed a non-specific uptake of the hydrophobic Fmoc-protected lysine-conjugated mTCPP precursors, but not d- or l-lysine mTCPP. Likewise, only the Fmoc-protected compounds induced substantial phototoxicty in cells following incubation and irradiation with blue light. These experimental results do not provide evidence to suggest that lysine-mTCPP is able to specifically target cancer cells. However, they do highlight mTCPP as a convenient and accessible framework for assessing molecular targeting of photosensitizers.
Collapse
Affiliation(s)
- Upendra Chitgupi
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, New York, NY 14260, USA.
| | - Jonathan F Lovell
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, New York, NY 14260, USA.
| | - Venugopal Rajendiran
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, New York, NY 14260, USA.
- Department of Chemistry, School of Basic and Applied Sciences, Central University of Tamil Nadu, Thiruvarur 610005, India.
| |
Collapse
|
226
|
Chiu M, Taurino G, Bianchi MG, Ottaviani L, Andreoli R, Ciociola T, Lagrasta CAM, Tardito S, Bussolati O. Oligodendroglioma Cells Lack Glutamine Synthetase and Are Auxotrophic for Glutamine, but Do not Depend on Glutamine Anaplerosis for Growth. Int J Mol Sci 2018; 19:E1099. [PMID: 29642388 PMCID: PMC5979401 DOI: 10.3390/ijms19041099] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 03/29/2018] [Accepted: 04/04/2018] [Indexed: 12/31/2022] Open
Abstract
In cells derived from several types of cancer, a transcriptional program drives high consumption of glutamine (Gln), which is used for anaplerosis, leading to a metabolic addiction for the amino acid. Low or absent expression of Glutamine Synthetase (GS), the only enzyme that catalyzes de novo Gln synthesis, has been considered a marker of Gln-addicted cancers. In this study, two human cell lines derived from brain tumors with oligodendroglioma features, HOG and Hs683, have been shown to be GS-negative. Viability of both lines depends from extracellular Gln with EC50 of 0.175 ± 0.056 mM (Hs683) and 0.086 ± 0.043 mM (HOG), thus suggesting that small amounts of extracellular Gln are sufficient for OD cell growth. Gln starvation does not significantly affect the cell content of anaplerotic substrates, which, consistently, are not able to rescue cell growth, but causes hindrance of the Wnt/β-catenin pathway and protein synthesis attenuation, which is mitigated by transient GS expression. Gln transport inhibitors cause partial depletion of intracellular Gln and cell growth inhibition, but do not lower cell viability. Therefore, GS-negative human oligodendroglioma cells are Gln-auxotrophic but do not use the amino acid for anaplerosis and, hence, are not Gln addicted, exhibiting only limited Gln requirements for survival and growth.
Collapse
Affiliation(s)
- Martina Chiu
- Laboratory of General Pathology, Department of Medicine and Surgery, University of Parma, Via Volturno 39, 43125 Parma, Italy.
| | - Giuseppe Taurino
- Laboratory of General Pathology, Department of Medicine and Surgery, University of Parma, Via Volturno 39, 43125 Parma, Italy.
| | - Massimiliano G Bianchi
- Laboratory of General Pathology, Department of Medicine and Surgery, University of Parma, Via Volturno 39, 43125 Parma, Italy.
| | - Laura Ottaviani
- Laboratory of General Pathology, Department of Medicine and Surgery, University of Parma, Via Volturno 39, 43125 Parma, Italy.
| | - Roberta Andreoli
- Laboratory of Industrial Toxicology, Department of Medicine and Surgery, University of Parma, Via Gramsci 14, 43126 Parma, Italy.
| | - Tecla Ciociola
- Laboratory of Medical Microbiology and Virology, Department of Medicine and Surgery, University of Parma, Via Volturno 39, 43125 Parma, Italy.
| | - Costanza A M Lagrasta
- Laboratory of Anatomical Pathology, Department of Medicine and Surgery, University of Parma, Via Gramsci 14, 43126 Parma, Italy.
| | - Saverio Tardito
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback road, Glasgow G611BD, UK.
| | - Ovidio Bussolati
- Laboratory of General Pathology, Department of Medicine and Surgery, University of Parma, Via Volturno 39, 43125 Parma, Italy.
| |
Collapse
|
227
|
Liu H, Xing R, Cheng X, Li Q, Liu F, Ye H, Zhao M, Wang H, Wang G, Hao H. De-novo NAD+ synthesis regulates SIRT1-FOXO1 apoptotic pathway in response to NQO1 substrates in lung cancer cells. Oncotarget 2018; 7:62503-62519. [PMID: 27566573 PMCID: PMC5308742 DOI: 10.18632/oncotarget.11526] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 08/08/2016] [Indexed: 12/19/2022] Open
Abstract
Tryptophan metabolism is essential in diverse kinds of tumors via regulating tumor immunology. However, the direct role of tryptophan metabolism and its signaling pathway in cancer cells remain largely elusive. Here, we establish a mechanistic link from L-type amino acid transporter 1 (LAT1) mediated transport of tryptophan and the subsequent de-novo NAD+ synthesis to SIRT1-FOXO1 regulated apoptotic signaling in A549 cells in response to NQO1 activation. In response to NQO1 activation, SIRT1 is repressed leading to the increased cellular accumulation of acetylated FOXO1 that transcriptionally activates apoptotic signaling. Decreased uptake of tryptophan due to the downregulation of LAT1 coordinates with PARP-1 hyperactivation to induce rapid depletion of NAD+ pool. Particularly, the LAT1-NAD+-SIRT1 signaling is activated in tumor tissues of patients with non-small cell lung cancer. Because NQO1 activation is characterized with oxidative challenge induced DNA damage, these results suggest that LAT1 and de-novo NAD+ synthesis in NSCLC cells may play essential roles in sensing excessive oxidative stress.
Collapse
Affiliation(s)
- Huiying Liu
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China.,Department of Physiology and Pathophysiology, Basic Medical College of Peking University, Beijing 100191, China
| | - Rong Xing
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China.,Department of Pharmacy, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, China
| | - Xuefang Cheng
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| | - Qingran Li
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| | - Fang Liu
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| | - Hui Ye
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| | - Min Zhao
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| | - Hong Wang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| | - Guangji Wang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| | - Haiping Hao
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
228
|
El Ansari R, Craze ML, Miligy I, Diez-Rodriguez M, Nolan CC, Ellis IO, Rakha EA, Green AR. The amino acid transporter SLC7A5 confers a poor prognosis in the highly proliferative breast cancer subtypes and is a key therapeutic target in luminal B tumours. Breast Cancer Res 2018; 20:21. [PMID: 29566741 PMCID: PMC5863851 DOI: 10.1186/s13058-018-0946-6] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 02/26/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Breast cancer (BC) is a heterogeneous disease characterised by variant biology and patient outcome. The amino acid transporter, SLC7A5, plays a role in BC although its impact on patient outcome in different BC subtypes remains to be validated. This study aimed to determine whether the clinicopathological and prognostic value of SLC7A5 is different within the molecular classes of BC. METHODS SLC7A5 was assessed at the genomic level, using Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) data (n = 1980), and proteomic level, using immunohistochemical analysis and tissue microarray (TMA) (n = 2664; 1110 training and 1554 validation sets) in well-characterised primary BC cohorts. SLC7A5 expression correlated with clinicopathological and biological parameters, molecular subtypes and patient outcome. RESULTS SLC7A5 mRNA and protein expression were strongly correlated with larger tumour size and higher grade. High expression was observed in triple negative (TN), human epidermal growth factor receptor 2 (HER2)+, and luminal B subtypes. SLC7A5 mRNA and protein expression was significantly associated with the expression of the key regulator of tumour cell metabolism, c-MYC, specifically in luminal B tumours only (p = 0.001). High expression of SLC7A5 mRNA and protein was associated with poor patient outcome (p < 0.001) but only in the highly proliferative oestrogen receptor (ER)+/ luminal B (p = 0.007) and HER2+ classes of BC (p = 0.03). In multivariate analysis, SLC7A5 protein was an independent risk factor for shorter breast-cancer-specific survival only in ER+ high-proliferation tumours (p = 0.02). CONCLUSIONS SLC7A5 appears to play a role in the aggressive highly proliferative ER+ subtype driven by MYC and could act as a potential therapeutic target. Functional assessment is necessary to reveal the specific role played by this transporter in the ER+ highly proliferative subclass and HER2+ subclass of BC.
Collapse
Affiliation(s)
- Rokaya El Ansari
- Academic Pathology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham City Hospital, Hucknall Road, Nottingham, NG5 1PB UK
| | - Madeleine L. Craze
- Academic Pathology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham City Hospital, Hucknall Road, Nottingham, NG5 1PB UK
| | - Islam Miligy
- Academic Pathology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham City Hospital, Hucknall Road, Nottingham, NG5 1PB UK
| | - Maria Diez-Rodriguez
- Academic Pathology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham City Hospital, Hucknall Road, Nottingham, NG5 1PB UK
| | - Christopher C. Nolan
- Academic Pathology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham City Hospital, Hucknall Road, Nottingham, NG5 1PB UK
| | - Ian O. Ellis
- Academic Pathology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham City Hospital, Hucknall Road, Nottingham, NG5 1PB UK
- Breast Institute, Nottingham University Hospitals NHS Trust, Hucknall Road, Nottingham, NG5 1PB UK
| | - Emad A. Rakha
- Academic Pathology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham City Hospital, Hucknall Road, Nottingham, NG5 1PB UK
- Breast Institute, Nottingham University Hospitals NHS Trust, Hucknall Road, Nottingham, NG5 1PB UK
| | - Andrew R. Green
- Academic Pathology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham City Hospital, Hucknall Road, Nottingham, NG5 1PB UK
| |
Collapse
|
229
|
Shiromizu S, Kusunose N, Matsunaga N, Koyanagi S, Ohdo S. Optimizing the dosing schedule of l-asparaginase improves its anti-tumor activity in breast tumor-bearing mice. J Pharmacol Sci 2018; 136:228-233. [PMID: 29605274 DOI: 10.1016/j.jphs.2018.01.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 01/08/2018] [Accepted: 01/09/2018] [Indexed: 12/18/2022] Open
Abstract
Proliferation of acute lymphoblastic leukemic cells is nutritionally dependent on the external supply of asparagine. l-asparaginase, an enzyme hydrolyzing l-asparagine in blood, is used for treatment of acute lymphoblastic leukemic and other related blood cancers. Although previous studies demonstrated that l-asparaginase suppresses the proliferation of cultured solid tumor cells, it remains unclear whether this enzyme prevents the growth of solid tumors in vivo. In this study, we demonstrated the importance of optimizing dosing schedules for the anti-tumor activity of l-asparaginase in 4T1 breast tumor-bearing mice. Cultures of several types of murine solid tumor cells were dependent on the external supply of asparagine. Among them, we selected murine 4T1 breast cancer cells and implanted them into BALB/c female mice kept under standardized light/dark cycle conditions. The growth of 4T1 tumor cells implanted in mice was significantly suppressed by intravenous administration of l-asparaginase during the light phase, whereas its administration during the dark phase failed to show significant anti-tumor activity. Decreases in plasma asparagine levels due to the administration of l-asparaginase were closely related to the dosing time-dependency of its anti-tumor effects. These results suggest that the anti-tumor efficacy of l-asparaginase in breast tumor-bearing mice is improved by optimizing the dosing schedule.
Collapse
Affiliation(s)
- Shoya Shiromizu
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Naoki Kusunose
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Naoya Matsunaga
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan; Department of Glocal Healthcare, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Satoru Koyanagi
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan; Department of Glocal Healthcare, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Shigehiro Ohdo
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
| |
Collapse
|
230
|
Kumar A, Giri S, Shaha C. Sestrin2 facilitates glutamine-dependent transcription of PGC-1α and survival of liver cancer cells under glucose limitation. FEBS J 2018; 285:1326-1345. [PMID: 29436167 DOI: 10.1111/febs.14406] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 01/22/2018] [Accepted: 02/07/2018] [Indexed: 12/13/2022]
Abstract
Differential utilization of metabolites and metabolic plasticity can confer adaptation to cancer cells under metabolic stress. Glutamine is one of the vital and versatile nutrients that cancer cells consume avidly for their proliferation, and therefore mechanisms related to glutamine metabolism have been identified as targets. Recently, sestrin2 (SESN2), a stress-inducible protein, has been reported to regulate survival in glutamine-depleted cancer cells; based on this, we explored if SESN2 could regulate glutamine metabolism during glucose starvation. This report highlights the role of SESN2 in the regulation of glutamine-dependent activation of the mitochondrial biogenesis marker peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α) under glucose scarcity in liver cancer cells (HepG2). We demonstrate that down-regulation of SESN2 induces a decrease in the levels of intracellular glutamine and PGC-1α under glucose deprivation, concomitant with a decline in cell survival, but no effect was observed on the invasive or migration potential of the cells. Under similar metabolic conditions, SESN2 forms a complex with c-Jun N-terminal kinase (JNK) and forkhead box protein O1 (FOXO1). Absence of SESN2 or inhibition of JNK reduces nuclear translocation of FOXO1, consequently causing transcriptional inhibition of PGC-1α. Notably, our observations demonstrate a reduction in cell viability under high glutamine and low glucose conditions during SESN2 down-regulation that could be rescued on JNK inhibition. To recover from acetaminophen-induced mitochondrial damage, SESN2 was crucial for glutamine-mediated activation of PGC-1α in HepG2 cells. Collectively, we demonstrate a novel role of SESN2 in mediating activation of PGC-1α by modulating glutamine metabolism that facilitates cancer cell survival under glucose-limited metabolic conditions.
Collapse
Affiliation(s)
- Ashish Kumar
- Cell Death and Differentiation Research Laboratory, National Institute of Immunology, New Delhi, India
| | - Sagnik Giri
- Cell Death and Differentiation Research Laboratory, National Institute of Immunology, New Delhi, India
| | - Chandrima Shaha
- Cell Death and Differentiation Research Laboratory, National Institute of Immunology, New Delhi, India
| |
Collapse
|
231
|
Cys Site-Directed Mutagenesis of the Human SLC1A5 (ASCT2) Transporter: Structure/Function Relationships and Crucial Role of Cys467 for Redox Sensing and Glutamine Transport. Int J Mol Sci 2018; 19:ijms19030648. [PMID: 29495336 PMCID: PMC5877509 DOI: 10.3390/ijms19030648] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 02/12/2018] [Accepted: 02/23/2018] [Indexed: 01/17/2023] Open
Abstract
The human plasma membrane transporter ASCT2 is responsible for mediating Na- dependent antiport of neutral amino acids. New insights into structure/function relationships were unveiled by a combined approach of recombinant over-expression, site-directed mutagenesis, transport assays in proteoliposomes and bioinformatics. WT and Cys mutants of hASCT2 were produced in P. pastoris and purified for functional assay. The reactivity towards SH reducing and oxidizing agents of WT protein was investigated and opposite effects were revealed; transport activity increased upon treatment with the Cys reducing agent DTE, i.e., when Cys residues were in thiol (reduced) state. Methyl-Hg, which binds to SH groups, was able to inhibit WT and seven out of eight Cys to Ala mutants. On the contrary, C467A loses the sensitivity to both DTE activation and Methyl-Hg inhibition. The C467A mutant showed a Km for Gln one order of magnitude higher than that of WT. Moreover, the C467 residue is localized in the substrate binding region of the protein, as suggested by bioinformatics on the basis of the EAAT1 structure comparison. Taken together, the experimental data allowed identifying C467 residue as crucial for substrate binding and for transport activity modulation of hASCT2.
Collapse
|
232
|
Zama K, Mitsutake S, Okazaki T, Igarashi Y. Sphingomyelin in microdomains of the plasma membrane regulates amino acid-stimulated mTOR signal activation. Cell Biol Int 2018; 42:823-831. [PMID: 29369436 DOI: 10.1002/cbin.10941] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 01/21/2018] [Indexed: 11/11/2022]
Abstract
Sphingomyelin (SM) is required for cells to proliferate, but the reason is not fully understood. In order to asses this question, we employed a cell line, ZS, which lacks both SMS1 and SMS2, isolated from mouse embryonic fibroblasts in SMS1 and 2 double knockout mouse, and SMS1 or SMS2 re-expressing cells, ZS/SMS1 or ZS/SMS2, respectively. We investigated regulation of SM in activating the mammalian target of rapamycin (mTOR) signal induced by essential amino acids (EAA), using these cells. EAA-stimulated mTOR signal was more activated in ZS/SMS1 and ZS/SMS2 cells than in controls. Treatment with methyl-b-cyclodextrin dramatically inhibited the activation. Interestingly, we found that the expression of CD98, LAT-1 and ASCT-2, amino acid transporters concerned with mTOR activation, was down-regulated in ZS cells. Transporters localized in microdomains and formed a functional complex. Our results indicate that SM affect proliferation through the transport of amino acids via SM-enriched microdomains.
Collapse
Affiliation(s)
- Kota Zama
- Faculty of Advanced Life Science, Department of Biomembrane and Biofunctional Chemistry, Hokkaido University, Sapporo, 001-0021, Japan
| | - Susumu Mitsutake
- Faculty of Agriculture, Department of Biochemistry and Food Science, Saga University, Honjo-machi 1, Saga, 840-8502, Japan
| | - Toshiro Okazaki
- Department of Hematology and Immunology, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Ishikawa, 920-0293, Japan
| | - Yasuyuki Igarashi
- Faculty of Advanced Life Science, Department of Biomembrane and Biofunctional Chemistry, Hokkaido University, Sapporo, 001-0021, Japan
| |
Collapse
|
233
|
CD44 variant inhibits insulin secretion in pancreatic β cells by attenuating LAT1-mediated amino acid uptake. Sci Rep 2018; 8:2785. [PMID: 29434323 PMCID: PMC5809395 DOI: 10.1038/s41598-018-20973-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 01/18/2018] [Indexed: 12/18/2022] Open
Abstract
CD44 variant (CD44v) contributes to cancer stemness by stabilizing the xCT subunit of system xc(−) and thereby promoting its glutamate-cystine antiporter activity. CD44 has also been implicated in autoimmune insulitis and inflammation in diabetic islets, but whether CD44v regulates insulin secretion has remained unclear. Here we show that CD44v inhibits insulin secretion by attenuating amino acid transport mediated by the L-type amino acid transporter LAT1. CD44v expression level was inversely related to insulin content in islets of normal and diabetic model mice. Knockdown of CD44 increased insulin secretion, the intracellular insulin level, and the transport of neutral amino acids mediated by LAT1 in Min6 cells. Attenuation of the uptake of neutral amino acids with a LAT inhibitor reduced insulin secretion and insulin content in Min6 cells, whereas overexpression of LAT1 increased insulin secretion. Moreover, inhibition of LAT1 prevented the increase in insulin secretion and content induced by CD44 depletion in Min6 cells. Our results thus implicate CD44v in the regulation of insulin secretion and reveal that amino acid transport is rate limiting for such secretion. They further suggest that amino acid transport mediated by LAT1 is a potential therapeutic target for diabetes.
Collapse
|
234
|
Goodman MM, Yu W, Jarkas N. Synthesis and biological properties of radiohalogenated α,α-disubstituted amino acids for PET and SPECT imaging of amino acid transporters (AATs). J Labelled Comp Radiopharm 2018; 61:272-290. [PMID: 29143354 DOI: 10.1002/jlcr.3584] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 09/27/2017] [Accepted: 10/30/2017] [Indexed: 11/08/2022]
Abstract
Fluorine-18 and iodine-123 labeled nonnatural alicyclic and methyl branched disubstituted α,α-amino acids are a diverse and useful class of tumor imaging agents suitable for positron emission tomography and single photon emission computed tomography. These tracers target the increased expression of the cell membrane amino acid transporter systems L, ASC, and A exhibited by many human tumor cells. The most established clinical use for these radiolabeled amino acids is imaging primary and recurrent gliomas and primary, recurrent, and metastatic prostate cancer. This review focuses on the synthesis, radiolabeling, and amino acid transport mechanism of a series of nonnatural fluorine-18 and iodine-123 labeled analogs of 1-aminocyclobutane-1-carboxylic acid, 1-aminocyclopentane-1-carboxylic acid, α-aminoisobutyric acid, and α-methylaminoisobutyric acid.
Collapse
Affiliation(s)
- Mark M Goodman
- Department of Radiology and Imaging Sciences, Center for Systems Imaging, Emory University, Atlanta, GA, USA
| | - Weiping Yu
- Department of Radiology and Imaging Sciences, Center for Systems Imaging, Emory University, Atlanta, GA, USA
| | - Nashwa Jarkas
- Department of Radiology and Imaging Sciences, Center for Systems Imaging, Emory University, Atlanta, GA, USA
| |
Collapse
|
235
|
Schulte ML, Fu A, Zhao P, Li J, Geng L, Smith ST, Kondo J, Coffey RJ, Johnson MO, Rathmell JC, Sharick JT, Skala MC, Smith JA, Berlin J, Washington MK, Nickels ML, Manning HC. Pharmacological blockade of ASCT2-dependent glutamine transport leads to antitumor efficacy in preclinical models. Nat Med 2018; 24:194-202. [PMID: 29334372 PMCID: PMC5803339 DOI: 10.1038/nm.4464] [Citation(s) in RCA: 330] [Impact Index Per Article: 47.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 11/29/2017] [Indexed: 12/11/2022]
Abstract
The unique metabolic demands of cancer cells underscore potentially fruitful opportunities for drug discovery in the era of precision medicine. However, therapeutic targeting of cancer metabolism has led to surprisingly few new drugs to date. The neutral amino acid glutamine serves as a key intermediate in numerous metabolic processes leveraged by cancer cells, including biosynthesis, cell signaling, and oxidative protection. Herein we report the preclinical development of V-9302, a competitive small molecule antagonist of transmembrane glutamine flux that selectively and potently targets the amino acid transporter ASCT2. Pharmacological blockade of ASCT2 with V-9302 resulted in attenuated cancer cell growth and proliferation, increased cell death, and increased oxidative stress, which collectively contributed to antitumor responses in vitro and in vivo. This is the first study, to our knowledge, to demonstrate the utility of a pharmacological inhibitor of glutamine transport in oncology, representing a new class of targeted therapy and laying a framework for paradigm-shifting therapies targeting cancer cell metabolism.
Collapse
Affiliation(s)
- Michael L. Schulte
- Vanderbilt Center for Molecular Probes, Vanderbilt University Medical Center, Nashville, TN 37232, United States
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN 37232, United States
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Allie Fu
- Vanderbilt Center for Molecular Probes, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Ping Zhao
- Vanderbilt Center for Molecular Probes, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Jun Li
- Vanderbilt Center for Molecular Probes, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Ling Geng
- Vanderbilt Center for Molecular Probes, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Shannon T. Smith
- Vanderbilt Center for Molecular Probes, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Jumpei Kondo
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Robert J. Coffey
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, United States
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232, United States
- Veterans Health Administration, Tennessee Valley Healthcare System, Nashville, TN, 37212, United States
| | - Marc O. Johnson
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Jeffrey C. Rathmell
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Joe T. Sharick
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37232, United States
| | - Melissa C. Skala
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37232, United States
| | - Jarrod A. Smith
- Vanderbilt Center for Structural Biology, Vanderbilt University, Nashville, TN 37232, United States
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37232, United States
| | - Jordan Berlin
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - M. Kay Washington
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232, United States
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Michael L. Nickels
- Vanderbilt Center for Molecular Probes, Vanderbilt University Medical Center, Nashville, TN 37232, United States
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN 37232, United States
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - H. Charles Manning
- Vanderbilt Center for Molecular Probes, Vanderbilt University Medical Center, Nashville, TN 37232, United States
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN 37232, United States
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, United States
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232, United States
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37232, United States
- Department of Neurosurgery, Vanderbilt University Medical Center, Nashville, TN 37232, United States
- Department of Chemistry, Vanderbilt University, Nashville, TN 37232, United States
| |
Collapse
|
236
|
Zanoni L, Bossert I, Matti A, Schiavina R, Pultrone C, Fanti S, Nanni C. A review discussing fluciclovine ( 18F) PET/CT imaging in the detection of recurrent prostate cancer. Future Oncol 2018; 14:1101-1115. [PMID: 29359581 DOI: 10.2217/fon-2017-0446] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
A significant number of patients radically treated for prostate cancer (PCa) will develop prostate-specific antigen recurrence (27-53%). Localizing the anatomical site of relapse is critical, in order to achieve the optimal treatment management. To date the diagnostic accuracy of standard imaging is low. Several desirable features have been identified for the amino-acid-based PET agent, fluciclovine (18F) including: long 18F half-life which allows more practical use in centers without a cyclotron onsite; acting as a substrate for amino acid transporters upregulated in PCa or associated with malignant phenotype; lacking of incorporation into protein; and limited urinary excretion. Fluciclovine (18F) is currently approved both in USA and Europe with specific indication in adult men with suspected recurrent PCa based on elevated prostate-specific antigen following prior treatment.
Collapse
Affiliation(s)
- Lucia Zanoni
- Nuclear Medicine, Azienda Ospedaliero Universitaria Policlinico S.Orsola-Malpighi, Bologna, Italy
| | - Irene Bossert
- Nuclear Medicine, Istituti Clinici Scientifici Maugeri SpA SB IRCCS, Pavia, Italy
| | - Antonella Matti
- Nuclear Medicine, Azienda Ospedaliero Universitaria Policlinico S.Orsola-Malpighi, Bologna, Italy
| | - Riccardo Schiavina
- Department of Urology, Azienda Ospedaliero Universitaria Policlinico S.Orsola-Malpighi, Bologna, Italy
| | - Cristian Pultrone
- Department of Urology, Azienda Ospedaliero Universitaria Policlinico S.Orsola-Malpighi, Bologna, Italy
| | - Stefano Fanti
- Nuclear Medicine, Azienda Ospedaliero Universitaria Policlinico S.Orsola-Malpighi, Bologna, Italy
| | - Cristina Nanni
- Nuclear Medicine, Azienda Ospedaliero Universitaria Policlinico S.Orsola-Malpighi, Bologna, Italy
| |
Collapse
|
237
|
Tomita R, Todoroki K, Maruoka H, Yoshida H, Fujioka T, Nakashima M, Yamaguchi M, Nohta H. Amino Acid Metabolomics Using LC-MS/MS: Assessment of Cancer-Cell Resistance in a Simulated Tumor Microenvironment. ANAL SCI 2018; 32:893-900. [PMID: 27506717 DOI: 10.2116/analsci.32.893] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
We performed a comprehensive quantification of 20 amino acids in RPMI 1640 medium-cultured human colorectal adenocarcinoma cells to evaluate the efficacy of 5-fluorouracil treatment under hypoxic and hypoglycemic conditions, which mimic the tumor microenvironment. In this study, we developed a simple and comprehensive analytical method by using LC-MS/MS connected to the Intrada amino acid column, which eluted amino acids within 9 min. The present method covered a linearity range of 3.6 - 1818 μM, except for Gly (227 - 1818 μM), Ala, Asp, His (7.1 - 1818 μM each), and Trp (3.6 - 909 μM). The limits of detection were in the range of 0.02 - 38.0 pmol per injection in a standard solution. Amino acid concentration data were analyzed using principal-component analysis to represent samples on two-dimensional graphs. Linear discriminant analysis was used to classify samples on the score plots. Using this approach, the effect of 5-fluorouracil treatment could be successfully discriminated at high discrimination rates. Moreover, several amino acids were extracted from corresponding loading plots as candidate markers for distinguishing the effects of the 5-fluorouracil treatment or tumor microenvironmental conditions. These results suggest that our proposed method might be a useful tool for evaluating the efficacy of anticancer drugs in the tumor microenvironment.
Collapse
Affiliation(s)
- Ryoko Tomita
- Faculty of Pharmaceutical Sciences, Fukuoka University
| | | | | | | | | | | | | | | |
Collapse
|
238
|
Sun A, Liu X, Tang G. Carbon-11 and Fluorine-18 Labeled Amino Acid Tracers for Positron Emission Tomography Imaging of Tumors. Front Chem 2018; 5:124. [PMID: 29379780 PMCID: PMC5775220 DOI: 10.3389/fchem.2017.00124] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 12/12/2017] [Indexed: 12/12/2022] Open
Abstract
Tumor cells have an increased nutritional demand for amino acids (AAs) to satisfy their rapid proliferation. Positron-emitting nuclide labeled AAs are interesting probes and are of great importance for imaging tumors using positron emission tomography (PET). Carbon-11 and fluorine-18 labeled AAs include the [1-11C] AAs, labeling alpha-C- AAs, the branched-chain of AAs and N-substituted carbon-11 labeled AAs. These tracers target protein synthesis or amino acid (AA) transport, and their uptake mechanism mainly involves AA transport. AA PET tracers have been widely used in clinical settings to image brain tumors, neuroendocrine tumors, prostate cancer, breast cancer, non-small cell lung cancer (NSCLC) and hepatocellular carcinoma. This review focuses on the fundamental concepts and the uptake mechanism of AAs, AA PET tracers and their clinical applications.
Collapse
Affiliation(s)
- Aixia Sun
- Guangdong Engineering Research Center for Translational Application of Medical Radiopharmaceuticals and Department of Nuclear Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiang Liu
- Department of Anesthesiology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ganghua Tang
- Guangdong Engineering Research Center for Translational Application of Medical Radiopharmaceuticals and Department of Nuclear Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
239
|
Cormerais Y, Massard PA, Vucetic M, Giuliano S, Tambutté E, Durivault J, Vial V, Endou H, Wempe MF, Parks SK, Pouyssegur J. The glutamine transporter ASCT2 (SLC1A5) promotes tumor growth independently of the amino acid transporter LAT1 (SLC7A5). J Biol Chem 2018; 293:2877-2887. [PMID: 29326164 PMCID: PMC5827425 DOI: 10.1074/jbc.ra117.001342] [Citation(s) in RCA: 127] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Revised: 12/27/2017] [Indexed: 12/12/2022] Open
Abstract
The transporters for glutamine and essential amino acids, ASCT2 (solute carrier family 1 member 5, SLC1A5) and LAT1 (solute carrier family 7 member 5, SLC7A5), respectively, are overexpressed in aggressive cancers and have been identified as cancer-promoting targets. Moreover, previous work has suggested that glutamine influx via ASCT2 triggers essential amino acids entry via the LAT1 exchanger, thus activating mechanistic target of rapamycin complex 1 (mTORC1) and stimulating growth. Here, to further investigate whether these two transporters are functionally coupled, we compared the respective knockout (KO) of either LAT1 or ASCT2 in colon (LS174T) and lung (A549) adenocarcinoma cell lines. Although ASCT2KO significantly reduced glutamine import (>60% reduction), no impact on leucine uptake was observed in both cell lines. Although an in vitro growth-reduction phenotype was observed in A549-ASCT2KO cells only, we found that genetic disruption of ASCT2 strongly decreased tumor growth in both cell lines. However, in sharp contrast to LAT1KO cells, ASCT2KO cells displayed no amino acid (AA) stress response (GCN2/EIF2a/ATF4) or altered mTORC1 activity (S6K1/S6). We therefore conclude that ASCT2KO reduces tumor growth by limiting AA import, but that this effect is independent of LAT1 activity. These data were further supported by in vitro cell proliferation experiments performed in the absence of glutamine. Together these results confirm and extend ASCT2's pro-tumoral role and indicate that the proposed functional coupling model of ASCT2 and LAT1 is not universal across different cancer types.
Collapse
Affiliation(s)
- Yann Cormerais
- Medical Biology Department, Centre Scientifique de Monaco (CSM), MC 98000 Monaco
| | - Pierre André Massard
- Medical Biology Department, Centre Scientifique de Monaco (CSM), MC 98000 Monaco
| | - Milica Vucetic
- Medical Biology Department, Centre Scientifique de Monaco (CSM), MC 98000 Monaco
| | - Sandy Giuliano
- Medical Biology Department, Centre Scientifique de Monaco (CSM), MC 98000 Monaco
| | - Eric Tambutté
- Medical Biology Department, Centre Scientifique de Monaco (CSM), MC 98000 Monaco
| | - Jerome Durivault
- Medical Biology Department, Centre Scientifique de Monaco (CSM), MC 98000 Monaco
| | - Valérie Vial
- Medical Biology Department, Centre Scientifique de Monaco (CSM), MC 98000 Monaco
| | | | - Michael F Wempe
- School of Pharmacy, Anschutz Medical Campus, University of Colorado Denver, Aurora, Colorado 80045
| | - Scott K Parks
- Medical Biology Department, Centre Scientifique de Monaco (CSM), MC 98000 Monaco.
| | - Jacques Pouyssegur
- Medical Biology Department, Centre Scientifique de Monaco (CSM), MC 98000 Monaco; Institute for Research on Cancer and Aging (IRCAN), CNRS, INSERM, Centre A. Lacassagne, University of Nice Sophia Antipolis, 06088 Nice, France.
| |
Collapse
|
240
|
Morita T, Kurihara H, Hiroi K, Honda N, Igaki H, Hatazawa J, Arai Y, Itami J. Dynamic changes in 18F-borono-L-phenylalanine uptake in unresectable, advanced, or recurrent squamous cell carcinoma of the head and neck and malignant melanoma during boron neutron capture therapy patient selection. Radiat Oncol 2018; 13:4. [PMID: 29325590 PMCID: PMC5765671 DOI: 10.1186/s13014-017-0949-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 12/28/2017] [Indexed: 12/27/2022] Open
Abstract
Background We evaluated dynamic changes in 18F–borono-L-phenylalanine (18F–BPA) uptake in unresectable, advanced, or recurrent squamous cell carcinoma of the head and neck (SCC) and malignant melanoma (MM) during boron neutron capture therapy (BNCT) patient selection. Methods Dynamic changes in the maximum standardized uptake value (SUVmax), tumor-to-normal tissue ratio (TNR), and tumor-to-blood pool ratio (TBR) for 18F–BPA were evaluated in 20 patients with SCC and 8 patients with MM. Results SUVmax in SCC tumors decreased significantly from 30 to 120 min. There was a non-statistically significant decrease in SUVmax for SCC tumors from 30 to 60 min and from 60 to 120 min. Patients with MM had nonsignificant SUVmax changes in 18F–BPA uptake on delayed imaging. Nonsignificant 18F–BPA TNR and TBR changes were seen in patients with SCC and MM. Conclusions Dynamic changes in SUVmax for 18F–BPA uptake had a washout pattern in SCC and a persistent pattern in MM. Dynamic 18F–BPA -PET studies should be performed to investigate the pharmacokinetics of 18F–BPA in humans and select appropriate candidates who may benefit from BNCT.
Collapse
Affiliation(s)
- Takahiro Morita
- Department of Diagnostic Radiology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan. .,Department of Nuclear Medicine and Tracer Kinetics, Osaka University Graduate School of Medicine, Osaka, Japan.
| | - Hiroaki Kurihara
- Department of Diagnostic Radiology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Kenta Hiroi
- Department of Diagnostic Radiology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Natsuki Honda
- Department of Diagnostic Radiology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Hiroshi Igaki
- Department of Radiation Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Jun Hatazawa
- Department of Nuclear Medicine and Tracer Kinetics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yasuaki Arai
- Department of Diagnostic Radiology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Jun Itami
- Department of Radiation Oncology, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
241
|
|
242
|
Goetzman ES, Prochownik EV. The Role for Myc in Coordinating Glycolysis, Oxidative Phosphorylation, Glutaminolysis, and Fatty Acid Metabolism in Normal and Neoplastic Tissues. Front Endocrinol (Lausanne) 2018; 9:129. [PMID: 29706933 PMCID: PMC5907532 DOI: 10.3389/fendo.2018.00129] [Citation(s) in RCA: 143] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 03/13/2018] [Indexed: 12/24/2022] Open
Abstract
That cancer cells show patterns of metabolism different from normal cells has been known for over 50 years. Yet, it is only in the past decade or so that an appreciation of the benefits of these changes has begun to emerge. Altered cancer cell metabolism was initially attributed to defective mitochondria. However, we now realize that most cancers do not have mitochondrial mutations and that normal cells can transiently adopt cancer-like metabolism during periods of rapid proliferation. Indeed, an encompassing, albeit somewhat simplified, conceptual framework to explain both normal and cancer cell metabolism rests on several simple premises. First, the metabolic pathways used by cancer cells and their normal counterparts are the same. Second, normal quiescent cells use their metabolic pathways and the energy they generate largely to maintain cellular health and organelle turnover and, in some cases, to provide secreted products necessary for the survival of the intact organism. By contrast, undifferentiated cancer cells minimize the latter functions and devote their energy to producing the anabolic substrates necessary to maintain high rates of unremitting cellular proliferation. Third, as a result of the uncontrolled proliferation of cancer cells, a larger fraction of the metabolic intermediates normally used by quiescent cells purely as a source of energy are instead channeled into competing proliferation-focused and energy-consuming anabolic pathways. Fourth, cancer cell clones with the most plastic and rapidly adaptable metabolism will eventually outcompete their less well-adapted brethren during tumor progression and evolution. This attribute becomes increasingly important as tumors grow and as their individual cells compete in a constantly changing and inimical environment marked by nutrient, oxygen, and growth factor deficits. Here, we review some of the metabolic pathways whose importance has gained center stage for tumor growth, particularly those under the control of the c-Myc (Myc) oncoprotein. We discuss how these pathways differ functionally between quiescent and proliferating normal cells, how they are kidnapped and corrupted during the course of transformation, and consider potential therapeutic strategies that take advantage of common features of neoplastic and metabolic disorders.
Collapse
Affiliation(s)
- Eric S. Goetzman
- Division of Medical Genetics, Children’s Hospital of Pittsburgh of UPMC, Pittsburgh, PA, United States
| | - Edward V. Prochownik
- Division of Hematology/Oncology, Children’s Hospital of Pittsburgh of UPMC, Pittsburgh, PA, United States
- Department of Microbiology and Molecular Genetics, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
- University of Pittsburgh Hillman Cancer Center, Pittsburgh, PA, United States
- *Correspondence: Edward V. Prochownik,
| |
Collapse
|
243
|
N-ω-chloroacetyl-L-ornithine has in-vitro activity against cancer cell lines and in-vivo activity against ascitic and solid tumors. Anticancer Drugs 2017; 27:508-18. [PMID: 26918391 DOI: 10.1097/cad.0000000000000353] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
N-ω-chloroacetyl-L-ornithine (NCAO) is an ornithine decarboxylase (ODC) inhibitor that is known to exert cytotoxic and antiproliferative effects on three neoplastic human cancer cell lines (HeLa, MCF-7, and HepG2). Here, we show that NCAO has antiproliferative activity in 13 cancer cell lines, of diverse tissue origin from human and mice, and in a mouse cancer model in vivo. All cell lines were sensitive to NCAO after 72 h of treatment (the EC50 ranged from 1 to 50.6 µmol/l). The Ca Ski cell line was the most sensitive (EC50=1.18±0.07 µmol/l) and MDA-MB-231 was the least sensitive (EC50=50.6±0.3 µmol/l). This ODC inhibitor showed selectivity for cancer cells, exerting almost no cytotoxic effect on the normal Vero cell line (EC50>1000 µmol/l). NCAO induced apoptosis and inhibited tumor cell migration in vitro. Furthermore, in vivo, this compound (at 50 and 100 mg/kg, daily intraperitoneal injection for 7 days) exerted potent antitumor activity against both solid and ascitic tumors in a mouse model using the myeloma (Ag8) cell line. At these same two doses, the toxicological evaluation showed that NCAO has no obvious systemic toxicity. The current results suggest that the antitumor activity is exerted by apoptosis related not only to a local but also a systemic cytotoxic effect exerted by NCAO on tumor cells. The applications for NCAO as an antitumor agent may be extensive; however, further studies are needed to ascertain the antitumor activity on other types of tumor in vivo and to determine the precise molecular mechanism of its activity.
Collapse
|
244
|
Scalise M, Pochini L, Galluccio M, Console L, Indiveri C. Glutamine Transport and Mitochondrial Metabolism in Cancer Cell Growth. Front Oncol 2017; 7:306. [PMID: 29376023 PMCID: PMC5770653 DOI: 10.3389/fonc.2017.00306] [Citation(s) in RCA: 141] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 11/28/2017] [Indexed: 12/20/2022] Open
Abstract
The concept that cancer is a metabolic disease is now well acknowledged: many cancer cell types rely mostly on glucose and some amino acids, especially glutamine for energy supply. These findings were corroborated by overexpression of plasma membrane nutrient transporters, such as the glucose transporters (GLUTs) and some amino acid transporters such as ASCT2, LAT1, and ATB0,+, which became promising targets for pharmacological intervention. On the basis of their sodium-dependent transport modes, ASCT2 and ATB0+ have the capacity to sustain glutamine need of cancer cells; while LAT1, which is sodium independent will have the role of providing cancer cells with some amino acids with plausible signaling roles. According to the metabolic reprogramming of many types of cancer cells, glucose is mainly catabolized by aerobic glycolysis in tumors, while the fate of Glutamine is completed at mitochondrial level where the enzyme Glutaminase converts Glutamine to Glutamate. Glutamine rewiring in cancer cells is heterogeneous. For example, Glutamate is converted to α-Ketoglutarate giving rise to a truncated form of Krebs cycle. This reprogrammed pathway leads to the production of ATP mainly at substrate level and regeneration of reducing equivalents needed for cells growth, redox balance, and metabolic energy. Few studies on hypothetical mitochondrial transporter for Glutamine are reported and indirect evidences suggested its presence. Pharmacological compounds able to inhibit Glutamine metabolism may represent novel drugs for cancer treatments. Interestingly, well acknowledged targets for drugs are the Glutamine transporters of plasma membrane and the key enzyme Glutaminase.
Collapse
Affiliation(s)
- Mariafrancesca Scalise
- Department DiBEST (Biologia, Ecologia, Scienze della Terra), Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Arcavacata di Rende, Italy
| | - Lorena Pochini
- Department DiBEST (Biologia, Ecologia, Scienze della Terra), Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Arcavacata di Rende, Italy
| | - Michele Galluccio
- Department DiBEST (Biologia, Ecologia, Scienze della Terra), Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Arcavacata di Rende, Italy
| | - Lara Console
- Department DiBEST (Biologia, Ecologia, Scienze della Terra), Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Arcavacata di Rende, Italy
| | - Cesare Indiveri
- Department DiBEST (Biologia, Ecologia, Scienze della Terra), Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Arcavacata di Rende, Italy.,CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnology, Bari, Italy
| |
Collapse
|
245
|
Na + entry through heteromeric TRPC4/C1 channels mediates (-)Englerin A-induced cytotoxicity in synovial sarcoma cells. Sci Rep 2017; 7:16988. [PMID: 29209034 PMCID: PMC5717101 DOI: 10.1038/s41598-017-17303-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 11/22/2017] [Indexed: 12/22/2022] Open
Abstract
The sesquiterpene (-)Englerin A (EA) is an organic compound from the plant Phyllanthus engleri which acts via heteromeric TRPC4/C1 channels to cause cytotoxicity in some types of cancer cell but not normal cells. Here we identified selective cytotoxicity of EA in human synovial sarcoma cells (SW982 cells) and investigated the mechanism. EA induced cation channel current (Icat) in SW982 cells with biophysical characteristics of heteromeric TRPC4/C1 channels. Inhibitors of homomeric TRPC4 channels were weak inhibitors of the Icat and EA-induced cytotoxicity whereas a potent inhibitor of TRPC4/C1 channels (Pico145) strongly inhibited Icat and cytotoxicity. Depletion of TRPC1 converted Icat into a current with biophysical and pharmacological properties of homomeric TRPC4 channels and depletion of TRPC1 or TRPC4 suppressed the cytotoxicity of EA. A Na+/K+-ATPase inhibitor (ouabain) potentiated EA-induced cytotoxicity and direct Na+ loading by gramicidin-A caused Pico145-resistant cytotoxicity in the absence of EA. We conclude that EA has a potent cytotoxic effect on human synovial sarcoma cells which is mediated by heteromeric TRPC4/C1 channels and Na+ loading.
Collapse
|
246
|
Tian Y, Du W, Cao S, Wu Y, Dong N, Wang Y, Xu Y. Systematic analyses of glutamine and glutamate metabolisms across different cancer types. CHINESE JOURNAL OF CANCER 2017; 36:88. [PMID: 29116024 PMCID: PMC5678792 DOI: 10.1186/s40880-017-0255-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 10/26/2017] [Indexed: 12/23/2022]
Abstract
BACKGROUND Glutamine and glutamate are known to play important roles in cancer biology. However, no detailed information is available in terms of their levels of involvement in various biological processes across different cancer types, whereas such knowledge could be critical for understanding the distinct characteristics of different cancer types. Our computational study aimed to examine the functional roles of glutamine and glutamate across different cancer types. METHODS We conducted a comparative analysis of gene expression data of cancer tissues versus normal control tissues of 11 cancer types to understand glutamine and glutamate metabolisms in cancer. Specifically, we developed a linear regression model to assess differential contributions by glutamine and/or glutamate to each of seven biological processes in cancer versus control tissues. RESULTS While our computational predictions were consistent with some of the previous observations, multiple novel predictions were made: (1) glutamine is generally not involved in purine synthesis in cancer except for breast cancer, and is similarly not involved in pyridine synthesis except for kidney cancer; (2) glutamine is generally not involved in ATP production in cancer; (3) glutamine's contribution to nucleotide synthesis is minimal if any in cancer; (4) glutamine is not involved in asparagine synthesis in cancer except for bladder and lung cancers; and (5) glutamate does not contribute to serine synthesis except for bladder cancer. CONCLUSIONS We comprehensively predicted the roles of glutamine and glutamate metabolisms in selected metabolic pathways in cancer tissues versus control tissues, which may lead to novel approaches to therapeutic development targeted at glutamine and/or glutamate metabolism. However, our predictions need further functional validation.
Collapse
Affiliation(s)
- Yuan Tian
- College of Computer Science and Technology, Jilin University, Changchun, 130012 Jilin P. R. China
| | - Wei Du
- College of Computer Science and Technology, Jilin University, Changchun, 130012 Jilin P. R. China
- Computational Systems Biology Lab, Department of Biochemistry and Molecular Biology and Institute of Bioinformatics, University of Georgia, 120 E Green St, Athens, GA 30602 USA
| | - Sha Cao
- Computational Systems Biology Lab, Department of Biochemistry and Molecular Biology and Institute of Bioinformatics, University of Georgia, 120 E Green St, Athens, GA 30602 USA
| | - Yue Wu
- Computational Systems Biology Lab, Department of Biochemistry and Molecular Biology and Institute of Bioinformatics, University of Georgia, 120 E Green St, Athens, GA 30602 USA
| | - Ning Dong
- The First Hospital, Jilin University, Changchun, 130012 Jilin P. R. China
- Computational Systems Biology Lab, Department of Biochemistry and Molecular Biology and Institute of Bioinformatics, University of Georgia, 120 E Green St, Athens, GA 30602 USA
| | - Yan Wang
- College of Computer Science and Technology, Jilin University, Changchun, 130012 Jilin P. R. China
| | - Ying Xu
- College of Computer Science and Technology, Jilin University, Changchun, 130012 Jilin P. R. China
- College of Public Health, Jilin University, Changchun, 130012 Jilin P. R. China
- Computational Systems Biology Lab, Department of Biochemistry and Molecular Biology and Institute of Bioinformatics, University of Georgia, 120 E Green St, Athens, GA 30602 USA
| |
Collapse
|
247
|
Wen S, Zhan B, Feng J, Hu W, Lin X, Bai J, Huang H. Non-invasively predicting differentiation of pancreatic cancer through comparative serum metabonomic profiling. BMC Cancer 2017; 17:708. [PMID: 29096620 PMCID: PMC5668965 DOI: 10.1186/s12885-017-3703-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Accepted: 10/25/2017] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The differentiation of pancreatic ductal adenocarcinoma (PDAC) could be associated with prognosis and may influence the choices of clinical management. No applicable methods could reliably predict the tumor differentiation preoperatively. Thus, the aim of this study was to compare the metabonomic profiling of pancreatic ductal adenocarcinoma with different differentiations and assess the feasibility of predicting tumor differentiations through metabonomic strategy based on nuclear magnetic resonance spectroscopy. METHODS By implanting pancreatic cancer cell strains Panc-1, Bxpc-3 and SW1990 in nude mice in situ, we successfully established the orthotopic xenograft models of PDAC with different differentiations. The metabonomic profiling of serum from different PDAC was achieved and analyzed by using 1H nuclear magnetic resonance (NMR) spectroscopy combined with the multivariate statistical analysis. Then, the differential metabolites acquired were used for enrichment analysis of metabolic pathways to get a deep insight. RESULTS An obvious metabonomic difference was demonstrated between all groups and the pattern recognition models were established successfully. The higher concentrations of amino acids, glycolytic and glutaminolytic participators in SW1990 and choline-contain metabolites in Panc-1 relative to other PDAC cells were demonstrated, which may be served as potential indicators for tumor differentiation. The metabolic pathways and differential metabolites identified in current study may be associated with specific pathways such as serine-glycine-one-carbon and glutaminolytic pathways, which can regulate tumorous proliferation and epigenetic regulation. CONCLUSION The NMR-based metabonomic strategy may be served as a non-invasive detection method for predicting tumor differentiation preoperatively.
Collapse
MESH Headings
- Animals
- Biomarkers, Tumor/blood
- Biomarkers, Tumor/metabolism
- Carcinoma, Pancreatic Ductal/blood
- Carcinoma, Pancreatic Ductal/metabolism
- Carcinoma, Pancreatic Ductal/pathology
- Cell Line, Tumor
- Feasibility Studies
- Humans
- Metabolomics/methods
- Mice, Inbred BALB C
- Mice, Nude
- Nuclear Magnetic Resonance, Biomolecular
- Pancreatic Neoplasms/blood
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/pathology
- Prognosis
- Reproducibility of Results
- Transplantation, Heterologous
Collapse
Affiliation(s)
- Shi Wen
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001 China
| | - Bohan Zhan
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance, Xiamen University, Xiamen, 361005 China
| | - Jianghua Feng
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance, Xiamen University, Xiamen, 361005 China
| | - Weize Hu
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001 China
| | - Xianchao Lin
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001 China
| | - Jianxi Bai
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001 China
| | - Heguang Huang
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001 China
| |
Collapse
|
248
|
Potent inhibitors of human LAT1 (SLC7A5) transporter based on dithiazole and dithiazine compounds for development of anticancer drugs. Biochem Pharmacol 2017; 143:39-52. [DOI: 10.1016/j.bcp.2017.07.006] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 07/07/2017] [Indexed: 12/11/2022]
|
249
|
El Ansari R, McIntyre A, Craze ML, Ellis IO, Rakha EA, Green AR. Altered glutamine metabolism in breast cancer; subtype dependencies and alternative adaptations. Histopathology 2017; 72:183-190. [DOI: 10.1111/his.13334] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 07/26/2017] [Accepted: 07/29/2017] [Indexed: 12/28/2022]
Affiliation(s)
- Rokaya El Ansari
- Academic Pathology; University of Nottingham; Nottingham City Hospital; Nottingham UK
| | - Alan McIntyre
- Cancer Biology Unit; Division of Cancer and Stem Cells; School of Medicine; University of Nottingham; Nottingham City Hospital; Nottingham UK
| | - Madeleine L Craze
- Academic Pathology; University of Nottingham; Nottingham City Hospital; Nottingham UK
| | - Ian O Ellis
- Academic Pathology; University of Nottingham; Nottingham City Hospital; Nottingham UK
- Cellular Pathology; Nottingham University Hospitals NHS Trust; Nottingham UK
| | - Emad A Rakha
- Academic Pathology; University of Nottingham; Nottingham City Hospital; Nottingham UK
- Cellular Pathology; Nottingham University Hospitals NHS Trust; Nottingham UK
| | - Andrew R Green
- Academic Pathology; University of Nottingham; Nottingham City Hospital; Nottingham UK
| |
Collapse
|
250
|
Mello-Andrade F, da Costa WL, Pires WC, Pereira FDC, Cardoso CG, Lino-Junior RDS, Irusta VRC, Carneiro CC, de Melo-Reis PR, Castro CH, Almeida MAP, Batista AA, Silveira-Lacerda EDP. Antitumor effectiveness and mechanism of action of Ru(II)/amino acid/diphosphine complexes in the peritoneal carcinomatosis progression. Tumour Biol 2017; 39:1010428317695933. [DOI: 10.1177/1010428317695933] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Francyelli Mello-Andrade
- Laboratório de Genética Molecular e Citogenética, Departamento de Genética, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, Brazil
| | - Wanderson Lucas da Costa
- Laboratório de Genética Molecular e Citogenética, Departamento de Genética, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, Brazil
| | - Wanessa Carvalho Pires
- Laboratório de Genética Molecular e Citogenética, Departamento de Genética, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, Brazil
| | - Flávia de Castro Pereira
- Laboratório de Genética Molecular e Citogenética, Departamento de Genética, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, Brazil
| | - Clever Gomes Cardoso
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, Brazil
| | - Ruy de Souza Lino-Junior
- Laboratório de Patologia Geral, Departamento de Microbiologia, Imunologia, Parasitologia e Patologia, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Brazil
| | | | - Cristiene Costa Carneiro
- Laboratório de Radiobiologia de Microrganismos e Mutagênese, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, Brazil
| | - Paulo Roberto de Melo-Reis
- Laboratório de Estudos Experimentais em Biotecnologia, Departamento de Biomedicina, Pontifícia Universidade Católica de Goiás, Goiânia, Brazil
| | - Carlos Henrique Castro
- Laboratório de Fisiologia Autonômica e Cardíaca, Departamento de Fisiologia e Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, Brazil
| | | | | | - Elisângela de Paula Silveira-Lacerda
- Laboratório de Genética Molecular e Citogenética, Departamento de Genética, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, Brazil
| |
Collapse
|