1
|
Bale AA, Thammineni S, Bhargava R, Harley B. Hyaluronic Acid Influences Amino Acid Metabolism via Differential L-Type Amino Acid Transporter 1 Expression in the U87-Malignant Glioma Cell Line. ADVANCED NANOBIOMED RESEARCH 2024; 4:2400107. [PMID: 40017591 PMCID: PMC11864772 DOI: 10.1002/anbr.202400107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2025] Open
Abstract
The Glioblastoma (GBM) tumor microenvironment is heterogeneous, complex, and is being increasingly understood as a significant contributor to tumor progression. In brain tumors, the extracellular matrix contains a large concentration of Hyaluronic acid (HA) that makes it important to study its role in cancer progression. In particular, abnormal accumulation of HA is observed in gliomas and is often associated with poor prognosis. In addition, HA is a polymer and its molecular weight (MW) distribution may influence tumor cell activity. Here, we evaluate the influence of the molecular weight of HA on tumor cell metabolism. We use a 2D cell culture approach to expose the U87-MG cell line to different HA MWs (10, 60, and 500 kDa) and glucose concentrations (0, 5.5, and 25 mM). Notably, we found that HA influences GBM amino acid metabolism via reduction in LAT1 transporter protein expression. We also report an influence on mitochondrial respiration levels and a difference in the accumulation of some key products of cell metabolic activity (lactic acid, glutamic acid and succinic acid). Overall, these results indicate that HA MW can influence GBM metabolic state, with implications for cell invasion and tumor progression.
Collapse
Affiliation(s)
- Ashwin A. Bale
- Department of Chemical and Biomolecular Engineering, Urbana-Champaign, Urbana, 61802, USA
| | | | - Rohit Bhargava
- Department of Chemical and Biomolecular Engineering, Urbana-Champaign, Urbana, 61802, USA
- Department of Bioengineering, Urbana-Champaign, Urbana, 61802, USA
- Cancer Center at Illinois, Urbana-Champaign, Urbana, 61802, USA
- Departments of Electrical & Computer Engineering, Mechanical Science & Engineering, and Chemistry, Beckman Institute for Advanced Science and Technology, Urbana, 61802, USA
- Carl R. Woese Institute for Genomic Biology University of Illinois, Urbana-Champaign, Urbana, 61802, USA
| | - Brendan Harley
- Department of Chemical and Biomolecular Engineering, Urbana-Champaign, Urbana, 61802, USA
- Cancer Center at Illinois, Urbana-Champaign, Urbana, 61802, USA
- Carl R. Woese Institute for Genomic Biology University of Illinois, Urbana-Champaign, Urbana, 61802, USA
| |
Collapse
|
2
|
Kaneda-Nakashima K, Shirakami Y, Hisada K, Feng S, Kadonaga Y, Ooe K, Watabe T, Manabe Y, Shimoyama A, Murakami M, Toyoshima A, Haba H, Kanai Y, Fukase K. Development of LAT1-Selective Nuclear Medicine Therapeutics Using Astatine-211. Int J Mol Sci 2024; 25:12386. [PMID: 39596451 PMCID: PMC11594329 DOI: 10.3390/ijms252212386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 11/10/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024] Open
Abstract
We investigated nuclear medicine therapeutics targeting the L-type amino acid transporter 1 (LAT1). We previously reported that a nuclear medicine therapeutic drug using astatine 211 (211At), an alpha-emitting nuclide that can be produced in an accelerator and targets LAT1 as a molecular target, is effective. The seed compound was 3-[211At] Astato-α-methyl-L-tyrosine (211At-AAMT-OH-L). We used a unique labeling method. By changing the OH group of phenol to a methyl group, retention was successfully increased. It was also found that the amount of the L-isomer taken up by the D-isomer and L-isomer was clearly higher, and the L-isomer was superior as a therapeutic drug. Compounds in which the methyl group was replaced with an ethyl or propyl group were also examined, but their retention did not increase significantly. In fact, we observed increased non-specific accumulation and dynamics, suggesting that labeling may be off. In addition, 211At-AAMT-O-Me-L, which has a simple structure, was clearly superior in terms of uptake speed for several candidate compounds. As a result, we were able to develop a compound that can be easily labeled, has high specific radioactivity, is stable, and has a strong therapeutic effect.
Collapse
Affiliation(s)
- Kazuko Kaneda-Nakashima
- Radiation Biological Chemistry, MS-CORE, FRC, Graduate School of Science, Osaka University, 1-1 Machikaneyama-cho, Toyonaka, Osaka 560-0043, Japan
- Institute for Radiation Sciences, Osaka University, 2-4 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Yoshifumi Shirakami
- Institute for Radiation Sciences, Osaka University, 2-4 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Kentaro Hisada
- Radiation Biological Chemistry, MS-CORE, FRC, Graduate School of Science, Osaka University, 1-1 Machikaneyama-cho, Toyonaka, Osaka 560-0043, Japan
| | - Sifan Feng
- Radiation Biological Chemistry, MS-CORE, FRC, Graduate School of Science, Osaka University, 1-1 Machikaneyama-cho, Toyonaka, Osaka 560-0043, Japan
| | - Yuichiro Kadonaga
- Institute for Radiation Sciences, Osaka University, 2-4 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Kazuhiro Ooe
- Institute for Radiation Sciences, Osaka University, 2-4 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Tadashi Watabe
- Department of Radiology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Yoshiyuki Manabe
- Institute for Radiation Sciences, Osaka University, 2-4 Yamadaoka, Suita, Osaka 565-0871, Japan
- Natural Product Chemistry, Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama-cho, Toyonaka, Osaka 560-0043, Japan
| | - Atsushi Shimoyama
- Institute for Radiation Sciences, Osaka University, 2-4 Yamadaoka, Suita, Osaka 565-0871, Japan
- Natural Product Chemistry, Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama-cho, Toyonaka, Osaka 560-0043, Japan
| | - Masashi Murakami
- Institute for Radiation Sciences, Osaka University, 2-4 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Atsushi Toyoshima
- Institute for Radiation Sciences, Osaka University, 2-4 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Hiromitsu Haba
- Nishina Center, RIKEN, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Yoshikatsu Kanai
- Premium Research Institute for Human Metaverse Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Koichi Fukase
- Institute for Radiation Sciences, Osaka University, 2-4 Yamadaoka, Suita, Osaka 565-0871, Japan
- Natural Product Chemistry, Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama-cho, Toyonaka, Osaka 560-0043, Japan
| |
Collapse
|
3
|
Wang H, Xu M, Zhang T, Pan J, Li C, Pan B, Zhou L, Huang Y, Gao C, He M, Xue Y, Ji X, Zhang X, Wang N, Zhou H, Wang Q, Li JZ. PYCR1 promotes liver cancer cell growth and metastasis by regulating IRS1 expression through lactylation modification. Clin Transl Med 2024; 14:e70045. [PMID: 39422696 PMCID: PMC11488319 DOI: 10.1002/ctm2.70045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 09/13/2024] [Accepted: 09/22/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Liver cancer (LC) is among the deadliest cancers worldwide, with existing treatments showing limited efficacy. This study aimed to elucidate the role and underlying mechanisms of pyrroline-5-carboxylate reductase 1 (PYCR1) as a potential therapeutic target in LC. METHODS Immunohistochemistry and Western blot were used to analyse the expression of PYCR1 in LC cells and tissues. EdU assays, colony-forming assays, scratch wound healing assays, Transwell assays, nude mouse xenograft models and nude mouse lung metastasis models were used to detect the growth and metastasis abilities of LC cells. Transcriptome sequencing was used to search for downstream target genes regulated by PYCR1, and metabolomics was used to identify the downstream metabolites regulated by PYCR1. ChIP assays were used to analyse the enrichment of H3K18 lactylation in the IRS1 promoter region. RESULTS We found that the expression of PYCR1 was significantly increased in HCC and that this high expression was associated with poor prognosis in HCC patients. Knockout or inhibition of PYCR1 inhibited HCC cell proliferation, migration and invasion both in vivo and in vitro. In addition, we revealed that knocking out or inhibiting PYCR1 could inhibit glycolysis in HCC cells and reduce H3K18 lactylation of the IRS1 histone, thereby inhibiting IRS1 expression. CONCLUSIONS Our findings identify PYCR1 as a pivotal regulator of LC progression that influences tumour cell metabolism and gene expression. By demonstrating the potential of targeting PYCR1 to inhibit LC cell proliferation and metastasis, this study identified PYCR1 as a promising therapeutic target for LC. HIGHLIGHTS Pyrroline-5-carboxylate reductase 1 (PYCR1) promotes the proliferation and metastasis of liver cancer (LC) cells. The expression of PYCR1 in LC is regulated by DNA methylation. Knocking down or inhibiting PYCR1 inhibits glycolysis as well as the PI3K/AKT/mTOR and MAPK/ERK pathways in LC cells. PYCR1 regulates the transcriptional activity of IRS1 by affecting H3K18 lactylation in its promoter region.
Collapse
Affiliation(s)
- Haoyu Wang
- The Key Laboratory of Rare Metabolic DiseaseDepartment of Biochemistry and Molecular BiologyThe Key Laboratory of Human Functional Genomics of Jiangsu ProvinceKey Laboratory of Targeted Intervention of Cardiovascular DiseaseCollaborative Innovation Center for Cardiovascular Disease Translational MedicineNanjing Medical UniversityNanjingJiangsuChina
| | - Mu Xu
- Department of Laboratory MedicineNanjing First HospitalNanjing Medical UniversityNanjingJiangsuChina
| | - Tong Zhang
- The Key Laboratory of Rare Metabolic DiseaseDepartment of Biochemistry and Molecular BiologyThe Key Laboratory of Human Functional Genomics of Jiangsu ProvinceKey Laboratory of Targeted Intervention of Cardiovascular DiseaseCollaborative Innovation Center for Cardiovascular Disease Translational MedicineNanjing Medical UniversityNanjingJiangsuChina
| | - Jinkun Pan
- The Key Laboratory of Rare Metabolic DiseaseDepartment of Biochemistry and Molecular BiologyThe Key Laboratory of Human Functional Genomics of Jiangsu ProvinceKey Laboratory of Targeted Intervention of Cardiovascular DiseaseCollaborative Innovation Center for Cardiovascular Disease Translational MedicineNanjing Medical UniversityNanjingJiangsuChina
| | - Chaopu Li
- The Key Laboratory of Rare Metabolic DiseaseDepartment of Biochemistry and Molecular BiologyThe Key Laboratory of Human Functional Genomics of Jiangsu ProvinceKey Laboratory of Targeted Intervention of Cardiovascular DiseaseCollaborative Innovation Center for Cardiovascular Disease Translational MedicineNanjing Medical UniversityNanjingJiangsuChina
| | - Bei Pan
- Department of Laboratory MedicineNanjing First HospitalNanjing Medical UniversityNanjingJiangsuChina
| | - Linpeng Zhou
- School of Basic Medicine and Clinical PharmacyNanjing First HospitalChina Pharmaceutical UniversityNanjingJiangsuChina
| | - Yun Huang
- The Key Laboratory of Rare Metabolic DiseaseDepartment of Biochemistry and Molecular BiologyThe Key Laboratory of Human Functional Genomics of Jiangsu ProvinceKey Laboratory of Targeted Intervention of Cardiovascular DiseaseCollaborative Innovation Center for Cardiovascular Disease Translational MedicineNanjing Medical UniversityNanjingJiangsuChina
| | - Chenzi Gao
- The Key Laboratory of Rare Metabolic DiseaseDepartment of Biochemistry and Molecular BiologyThe Key Laboratory of Human Functional Genomics of Jiangsu ProvinceKey Laboratory of Targeted Intervention of Cardiovascular DiseaseCollaborative Innovation Center for Cardiovascular Disease Translational MedicineNanjing Medical UniversityNanjingJiangsuChina
| | - Mengping He
- The Key Laboratory of Rare Metabolic DiseaseDepartment of Biochemistry and Molecular BiologyThe Key Laboratory of Human Functional Genomics of Jiangsu ProvinceKey Laboratory of Targeted Intervention of Cardiovascular DiseaseCollaborative Innovation Center for Cardiovascular Disease Translational MedicineNanjing Medical UniversityNanjingJiangsuChina
| | - Yao Xue
- The Key Laboratory of Rare Metabolic DiseaseDepartment of Biochemistry and Molecular BiologyThe Key Laboratory of Human Functional Genomics of Jiangsu ProvinceKey Laboratory of Targeted Intervention of Cardiovascular DiseaseCollaborative Innovation Center for Cardiovascular Disease Translational MedicineNanjing Medical UniversityNanjingJiangsuChina
| | - Xuetao Ji
- The Key Laboratory of Rare Metabolic DiseaseDepartment of Biochemistry and Molecular BiologyThe Key Laboratory of Human Functional Genomics of Jiangsu ProvinceKey Laboratory of Targeted Intervention of Cardiovascular DiseaseCollaborative Innovation Center for Cardiovascular Disease Translational MedicineNanjing Medical UniversityNanjingJiangsuChina
| | - Xu Zhang
- The Key Laboratory of Rare Metabolic DiseaseDepartment of Biochemistry and Molecular BiologyThe Key Laboratory of Human Functional Genomics of Jiangsu ProvinceKey Laboratory of Targeted Intervention of Cardiovascular DiseaseCollaborative Innovation Center for Cardiovascular Disease Translational MedicineNanjing Medical UniversityNanjingJiangsuChina
| | - Ning Wang
- The Key Laboratory of Rare Metabolic DiseaseDepartment of Biochemistry and Molecular BiologyThe Key Laboratory of Human Functional Genomics of Jiangsu ProvinceKey Laboratory of Targeted Intervention of Cardiovascular DiseaseCollaborative Innovation Center for Cardiovascular Disease Translational MedicineNanjing Medical UniversityNanjingJiangsuChina
| | - Hongwen Zhou
- Department of EndocrinologyThe First affiliated Hospital of Nanjing Medical UniversityNanjingJiangsuChina
| | - Qian Wang
- The Key Laboratory of Rare Metabolic DiseaseDepartment of Biochemistry and Molecular BiologyThe Key Laboratory of Human Functional Genomics of Jiangsu ProvinceKey Laboratory of Targeted Intervention of Cardiovascular DiseaseCollaborative Innovation Center for Cardiovascular Disease Translational MedicineNanjing Medical UniversityNanjingJiangsuChina
| | - John Zhong Li
- The Key Laboratory of Rare Metabolic DiseaseDepartment of Biochemistry and Molecular BiologyThe Key Laboratory of Human Functional Genomics of Jiangsu ProvinceKey Laboratory of Targeted Intervention of Cardiovascular DiseaseCollaborative Innovation Center for Cardiovascular Disease Translational MedicineNanjing Medical UniversityNanjingJiangsuChina
- Department of EndocrinologyThe affiliated Huaian No.1 People's Hospital of Nanjing Medical UniversityNorthern Jiangsu Institute of Clinical MedicineHuaianJiangsuChina
- Tianjian Laboratory of Advanced Biomedical SciencesInstitute of Advanced Biomedical SciencesZhengzhou UniversityZhengzhouHenanChina
| |
Collapse
|
4
|
Shi X, Yang J, Wang M, Xia L, Zhang L, Qiao S. Hsa_circ_0050900 affects ferroptosis in intrahepatic cholangiocarcinoma cells by targeting hsa‑miR-605‑3p to regulate SLC3A2. Oncol Lett 2024; 27:2. [PMID: 38028176 PMCID: PMC10665981 DOI: 10.3892/ol.2023.14135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 09/20/2023] [Indexed: 12/01/2023] Open
Abstract
Intrahepatic cholangiocarcinoma (ICC) is a highly lethal hepatobiliary tumor with high aggressiveness. The role of circular RNA (circRNA) in ICC remains to be explored. The present study aimed to investigate whether hsa_circ_0050900 affected ferroptosis in ICC cells by regulating hsa-microRNA (miR)-605-3p/solute carrier family 3 member 2 (SLC3A2). Human ICC cells were cultured and hsa_circ_0050900 expression was evaluated by reverse transcription-quantitative PCR. hsa_circ_0050900 was knocked down and ferroptosis inhibitor ferrostatin-1 was added to HuCCT-1 cells. Following knockdown or overexpression of hsa-miR-605-3p, Fe2+, reactive oxygen species (ROS), glutathione peroxidase 4 and SLC3A2 levels were assessed using iron and ROS assay kit or RT-qPCR and western blotting, respectively. Cell function experiments were performed to examine proliferation and migration abilities. Dual-luciferase reporter gene and argonaute2-RNA immunoprecipitation assay verified the relationship among hsa_circ_0050900, hsa-miR-605-3p, and SLC3A2. hsa_circ_0050900 was derived from actinin alpha 4 gene and was elevated in ICC cells. Among HuCCT-1, QBC-939, HCCC-9810, and RBE cell lines, the highest expression was in HuCCT-1 cells. Inhibition of hsa_circ_0050900 inhibited proliferation and migration by facilitating ICC cell ferroptosis. hsa-miR-605-3p expression was elevated after knocking down hsa_circ_0050900 and hsa-miR-605-3p was negatively regulated by hsa_circ_0050900. In addition, hsa-miR-605-3p targeted SLC3A2. Overexpression of hsa-miR-605-3p regulated SLC3A2 to promote ICC cell ferroptosis and inhibit proliferation and migration. Taken together, knockdown of hsa_circ_0050900 inhibited SLC3A2 expression via sponging hsa-miR-605-3p to promote ICC cell ferroptosis, and finally suppressed proliferation and migration. The present study suggested that hsa_circ_0050900 was a potential therapeutic target for ICC.
Collapse
Affiliation(s)
- Xiangtian Shi
- Department of Hepatobiliary Surgery, Bayannur Hospital, Bayannur, Inner Mongolia Autonomous Region 015000, P.R. China
| | - Jiarui Yang
- Department of Hepatobiliary, Pancreatic and Splenic Surgery, Affiliated Dongguan Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan, Guangdong 523058, P.R. China
- Department of Pancreatic Hepato-Biliary-Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, P.R. China
| | - Meng Wang
- Department of Hepatobiliary Surgery, Bayannur Hospital, Bayannur, Inner Mongolia Autonomous Region 015000, P.R. China
| | - Long Xia
- Department of Hepatobiliary-Pancreatic-Splenic Surgery, Inner Mongolia Autonomous Region People's Hospital, Hohhot, Inner Mongolia Autonomous Region 010017, P.R. China
| | - Lei Zhang
- Department of Pancreatic Hepato-Biliary-Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, P.R. China
| | - Shan Qiao
- Department of Hepatobiliary Surgery, Bayannur Hospital, Bayannur, Inner Mongolia Autonomous Region 015000, P.R. China
| |
Collapse
|
5
|
Yang F, Hilakivi-Clarke L, Shaha A, Wang Y, Wang X, Deng Y, Lai J, Kang N. Metabolic reprogramming and its clinical implication for liver cancer. Hepatology 2023; 78:1602-1624. [PMID: 36626639 PMCID: PMC10315435 DOI: 10.1097/hep.0000000000000005] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 09/28/2022] [Indexed: 01/12/2023]
Abstract
Cancer cells often encounter hypoxic and hypo-nutrient conditions, which force them to make adaptive changes to meet their high demands for energy and various biomaterials for biomass synthesis. As a result, enhanced catabolism (breakdown of macromolecules for energy production) and anabolism (macromolecule synthesis from bio-precursors) are induced in cancer. This phenomenon is called "metabolic reprogramming," a cancer hallmark contributing to cancer development, metastasis, and drug resistance. HCC and cholangiocarcinoma (CCA) are 2 different liver cancers with high intertumoral heterogeneity in terms of etiologies, mutational landscapes, transcriptomes, and histological representations. In agreement, metabolism in HCC or CCA is remarkably heterogeneous, although changes in the glycolytic pathways and an increase in the generation of lactate (the Warburg effect) have been frequently detected in those tumors. For example, HCC tumors with activated β-catenin are addicted to fatty acid catabolism, whereas HCC tumors derived from fatty liver avoid using fatty acids. In this review, we describe common metabolic alterations in HCC and CCA as well as metabolic features unique for their subsets. We discuss metabolism of NAFLD as well, because NAFLD will likely become a leading etiology of liver cancer in the coming years due to the obesity epidemic in the Western world. Furthermore, we outline the clinical implication of liver cancer metabolism and highlight the computation and systems biology approaches, such as genome-wide metabolic models, as a valuable tool allowing us to identify therapeutic targets and develop personalized treatments for liver cancer patients.
Collapse
Affiliation(s)
- Flora Yang
- BA/MD Joint Admission Scholars Program, University of Minnesota, Minneapolis, Minnesota
| | - Leena Hilakivi-Clarke
- Food Science and Nutrition Section, The Hormel Institute, University of Minnesota, Austin, Minnesota
| | - Aurpita Shaha
- Tumor Microenvironment and Metastasis Section, the Hormel Institute, University of Minnesota, Austin, Minnesota
| | - Yuanguo Wang
- Tumor Microenvironment and Metastasis Section, the Hormel Institute, University of Minnesota, Austin, Minnesota
| | - Xianghu Wang
- Tumor Microenvironment and Metastasis Section, the Hormel Institute, University of Minnesota, Austin, Minnesota
| | - Yibin Deng
- Department of Urology, Masonic Cancer Center, The University of Minnesota Medical School, Minneapolis, Minnesota
| | - Jinping Lai
- Department of Pathology and Laboratory Medicine, Kaiser Permanente Sacramento Medical Center, Sacramento, California
| | - Ningling Kang
- Tumor Microenvironment and Metastasis Section, the Hormel Institute, University of Minnesota, Austin, Minnesota
| |
Collapse
|
6
|
HPV16 E6 and E7 Oncoproteins Stimulate the Glutamine Pathway Maintaining Cell Proliferation in a SNAT1-Dependent Fashion. Viruses 2023; 15:v15020324. [PMID: 36851539 PMCID: PMC9964736 DOI: 10.3390/v15020324] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/17/2023] [Accepted: 01/20/2023] [Indexed: 01/26/2023] Open
Abstract
Persistent high-risk human papillomavirus infection is the main risk factor for cervical cancer establishment, where the viral oncogenes E6 and E7 promote a cancerous phenotype. Metabolic reprogramming in cancer involves alterations in glutamine metabolism, also named glutaminolysis, to provide energy for supporting cancer processes including migration, proliferation, and production of reactive oxygen species, among others. The aim of this work was to analyze the effect of HPV16 E6 and E7 oncoproteins on the regulation of glutaminolysis and its contribution to cell proliferation. We found that the E6 and E7 oncoproteins exacerbate cell proliferation in a glutamine-dependent manner. Both oncoproteins increased the levels of transporter SNAT1, as well as GLS2 and GS enzymes; E6 also increased LAT1 transporter protein levels, while E7 increased ASCT2 and xCT. Some of these alterations are also regulated at a transcriptional level. Consistently, the amount of SNAT1 protein decreased in Ca Ski cells when E6 and E7 expression was knocked down. In addition, we demonstrated that cell proliferation was partially dependent on SNAT1 in the presence of glutamine. Interestingly, SNAT1 expression was higher in cervical cancer compared with normal cervical cells. The high expression of SNAT1 was associated with poor overall survival of cervical cancer patients. Our results indicate that HPV oncoproteins exacerbate glutaminolysis supporting the malignant phenotype.
Collapse
|
7
|
Raggi C, Taddei ML, Rae C, Braconi C, Marra F. Metabolic reprogramming in cholangiocarcinoma. J Hepatol 2022; 77:849-864. [PMID: 35594992 DOI: 10.1016/j.jhep.2022.04.038] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/16/2022] [Accepted: 04/28/2022] [Indexed: 12/25/2022]
Abstract
Metabolic reprogramming is a hallmark of cancer and allows tumour cells to meet the increased energy demands required for rapid proliferation, invasion, and metastasis. Indeed, many tumour cells acquire distinctive metabolic and bioenergetic features that enable them to survive in resource-limited conditions, mainly by harnessing alternative nutrients. Several recent studies have explored the metabolic plasticity of cancer cells with the aim of identifying new druggable targets, while therapeutic strategies to limit the access to nutrients have been successfully applied to the treatment of some tumours. Cholangiocarcinoma (CCA), a highly heterogeneous tumour, is the second most common form of primary liver cancer. It is characterised by resistance to chemotherapy and poor prognosis, with 5-year survival rates of below 20%. Deregulation of metabolic pathways have been described during the onset and progression of CCA. Increased aerobic glycolysis and glutamine anaplerosis provide CCA cells with the ability to generate biosynthetic intermediates. Other metabolic alterations involving carbohydrates, amino acids and lipids have been shown to sustain cancer cell growth and dissemination. In this review, we discuss the complex metabolic rewiring that occurs during CCA development and leads to unique nutrient addiction. The possible role of therapeutic interventions based on metabolic changes is also thoroughly discussed.
Collapse
Affiliation(s)
- Chiara Raggi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.
| | - Maria Letizia Taddei
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Colin Rae
- Institute of Cancer Sciences, The University of Glasgow, Glasgow, United Kingdom
| | - Chiara Braconi
- Institute of Cancer Sciences, The University of Glasgow, Glasgow, United Kingdom; Beatson West of Scotland Cancer Centre, Glasgow, United Kingdom
| | - Fabio Marra
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.
| |
Collapse
|
8
|
Gregorio JD, Petricca S, Iorio R, Toniato E, Flati V. MITOCHONDRIAL AND METABOLIC ALTERATIONS IN CANCER CELLS. Eur J Cell Biol 2022; 101:151225. [DOI: 10.1016/j.ejcb.2022.151225] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 04/11/2022] [Accepted: 04/11/2022] [Indexed: 02/07/2023] Open
|
9
|
Du D, Liu C, Qin M, Zhang X, Xi T, Yuan S, Hao H, Xiong J. Metabolic dysregulation and emerging therapeutical targets for hepatocellular carcinoma. Acta Pharm Sin B 2022; 12:558-580. [PMID: 35256934 PMCID: PMC8897153 DOI: 10.1016/j.apsb.2021.09.019] [Citation(s) in RCA: 328] [Impact Index Per Article: 109.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 08/31/2021] [Accepted: 09/01/2021] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is an aggressive human cancer with increasing incidence worldwide. Multiple efforts have been made to explore pharmaceutical therapies to treat HCC, such as targeted tyrosine kinase inhibitors, immune based therapies and combination of chemotherapy. However, limitations exist in current strategies including chemoresistance for instance. Tumor initiation and progression is driven by reprogramming of metabolism, in particular during HCC development. Recently, metabolic associated fatty liver disease (MAFLD), a reappraisal of new nomenclature for non-alcoholic fatty liver disease (NAFLD), indicates growing appreciation of metabolism in the pathogenesis of liver disease, including HCC, thereby suggesting new strategies by targeting abnormal metabolism for HCC treatment. In this review, we introduce directions by highlighting the metabolic targets in glucose, fatty acid, amino acid and glutamine metabolism, which are suitable for HCC pharmaceutical intervention. We also summarize and discuss current pharmaceutical agents and studies targeting deregulated metabolism during HCC treatment. Furthermore, opportunities and challenges in the discovery and development of HCC therapy targeting metabolism are discussed.
Collapse
Key Words
- 1,3-BPG, 1,3-bisphosphoglycerate
- 2-DG, 2-deoxy-d-glucose
- 3-BrPA, 3-bromopyruvic acid
- ACC, acetyl-CoA carboxylase
- ACLY, adenosine triphosphate (ATP) citrate lyase
- ACS, acyl-CoA synthease
- AKT, protein kinase B
- AML, acute myeloblastic leukemia
- AMPK, adenosine mono-phosphate-activated protein kinase
- ASS1, argininosuccinate synthase 1
- ATGL, adipose triacylglycerol lipase
- CANA, canagliflozin
- CPT, carnitine palmitoyl-transferase
- CYP4, cytochrome P450s (CYPs) 4 family
- Cancer therapy
- DNL, de novo lipogenesis
- EMT, epithelial-to-mesenchymal transition
- ER, endoplasmic reticulum
- ERK, extracellular-signal regulated kinase
- FABP1, fatty acid binding protein 1
- FASN, fatty acid synthase
- FBP1, fructose-1,6-bisphosphatase 1
- FFA, free fatty acid
- Fatty acid β-oxidation
- G6PD, glucose-6-phosphate dehydrogenase
- GAPDH, glyceraldehyde-3-phosphate dehydrogenase
- GLS1, renal-type glutaminase
- GLS2, liver-type glutaminase
- GLUT1, glucose transporter 1
- GOT1, glutamate oxaloacetate transaminase 1
- Glutamine metabolism
- Glycolysis
- HCC, hepatocellular carcinoma
- HIF-1α, hypoxia-inducible factor-1 alpha
- HK, hexokinase
- HMGCR, 3-hydroxy-3-methylglutaryl-CoA reductase
- HSCs, hepatic stellate cells
- Hepatocellular carcinoma
- IDH2, isocitrate dehydrogenase 2
- LCAD, long-chain acyl-CoA dehydrogenase
- LDH, lactate dehydrogenase
- LPL, lipid lipase
- LXR, liver X receptor
- MAFLD, metabolic associated fatty liver disease
- MAGL, monoacyglycerol lipase
- MCAD, medium-chain acyl-CoA dehydrogenase
- MEs, malic enzymes
- MMP9, matrix metallopeptidase 9
- Metabolic dysregulation
- NADPH, nicotinamide adenine nucleotide phosphate
- NAFLD, non-alcoholic fatty liver disease
- NASH, non-alcoholic steatohepatitis
- OTC, ornithine transcarbamylase
- PCK1, phosphoenolpyruvate carboxykinase 1
- PFK1, phosphofructokinase 1
- PGAM1, phosphoglycerate mutase 1
- PGK1, phosphoglycerate kinase 1
- PI3K, phosphoinositide 3-kinase
- PKM2, pyruvate kinase M2
- PPARα, peroxisome proliferator-activated receptor alpha
- PPP, pentose phosphate pathway
- Pentose phosphate pathway
- ROS, reactive oxygen species
- SCD1, stearoyl-CoA-desaturase 1
- SGLT2, sodium-glucose cotransporter 2
- SLC1A5/ASCT2, solute carrier family 1 member 5/alanine serine cysteine preferring transporter 2
- SLC7A5/LAT1, solute carrier family 7 member 5/L-type amino acid transporter 1
- SREBP1, sterol regulatory element-binding protein 1
- TAGs, triacylglycerols
- TCA cycle, tricarboxylic acid cycle
- TKIs, tyrosine kinase inhibitors
- TKT, transketolase
- Tricarboxylic acid cycle
- VEGFR, vascular endothelial growth factor receptor
- WD-fed MC4R-KO, Western diet (WD)-fed melanocortin 4 receptor-deficient (MC4R-KO)
- WNT, wingless-type MMTV integration site family
- mIDH, mutant IDH
- mTOR, mammalian target of rapamycin
Collapse
Affiliation(s)
- Danyu Du
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Chan Liu
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Mengyao Qin
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xiao Zhang
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Tao Xi
- Research Center of Biotechnology, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Shengtao Yuan
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Haiping Hao
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
- Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
- Corresponding authors.
| | - Jing Xiong
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
- Corresponding authors.
| |
Collapse
|
10
|
Zhao X, Sakamoto S, Maimaiti M, Anzai N, Ichikawa T. Contribution of LAT1-4F2hc in Urological Cancers via Toll-like Receptor and Other Vital Pathways. Cancers (Basel) 2022; 14:cancers14010229. [PMID: 35008399 PMCID: PMC8750950 DOI: 10.3390/cancers14010229] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 12/28/2021] [Accepted: 01/02/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary LAT1-4F2hc complex is an important amino acid transporter. It mainly transports specific amino acids through the cell membrane, provides nutrition for cells, and participates in a variety of metabolic pathways. LAT1 plays a role in transporting essential amino acids including leucine, which regulates the mTOR signaling pathway. However, the importance of SLCs is still not well known in the field of urological cancer. Therefore, the purpose of this review is to report the role of the LAT1-4F2hc complex in urological cancers, as well as their clinical significance and application. Moreover, the inhibitor of LAT1-4F2hc complex is a promising direction as a targeted therapy to improve the treatment and prognosis of urological cancers. Abstract Tumor cells are known for their ability to proliferate. Nutrients are essential for rapidly growing tumor cells. In particular, essential amino acids are essential for tumor cell growth. Tumor cell growth nutrition requires the regulation of membrane transport proteins. Nutritional processes require amino acid uptake across the cell membrane. Leucine, one of the essential amino acids, has recently been found to be closely associated with cancer, which activate mTOR signaling pathway. The transport of leucine into cells requires an L-type amino acid transporter protein 1, LAT1 (SLC7A5), which requires the 4F2 cell surface antigen heavy chain (4F2hc, SLC3A2) to form a heterodimeric amino acid transporter protein complex. Recent evidence identified 4F2hc as a specific downstream target of the androgen receptor splice variant 7 (AR-V7). We stressed the importance of the LAT1-4F2hc complex as a diagnostic and therapeutic target in urological cancers in this review, which covered the recent achievements in research on the involvement of the LAT1-4F2hc complex in urinary system tumors. In addition, JPH203, which is a selective LAT1 inhibitor, has shown excellent inhibitory effects on the proliferation in a variety of tumor cells. The current phase I clinical trials of JPH203 in patients with biliary tract cancer have also achieved good results, which is the future research direction for LAT1 targeted therapy drugs.
Collapse
Affiliation(s)
- Xue Zhao
- Department of Urology, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan; (X.Z.); (T.I.)
- Department of Urology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Shinichi Sakamoto
- Department of Urology, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan; (X.Z.); (T.I.)
- Correspondence: ; Tel.: +81-43-226-2134; Fax: +81-43-226-2136
| | - Maihulan Maimaiti
- Department of Tumor Pathology, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan;
| | - Naohiko Anzai
- Department of Pharmacology, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan;
| | - Tomohiko Ichikawa
- Department of Urology, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan; (X.Z.); (T.I.)
| |
Collapse
|
11
|
Loilome W, Dokduang H, Suksawat M, Padthaisong S. Therapeutic challenges at the preclinical level for targeted drug development for Opisthorchis viverrini-associated cholangiocarcinoma. Expert Opin Investig Drugs 2021; 30:985-1006. [PMID: 34292795 DOI: 10.1080/13543784.2021.1955102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Cholangiocarcinoma (CCA) is a malignant tumor of bile duct epithelium with the highest incidence found in Thailand. Some patients are considered suitable for adjuvant therapy and surgical resection is currently the curative treatment for CCA patients. Tumor recurrence is still a hurdle after treatment; hence, finding novel therapeutic strategies to combat CCA is necessary for improving outcome for patients. AREAS COVERED We discuss targeted therapies and other novel treatment approaches which include protein kinase inhibitors, natural products, amino acid transporter-based inhibitors, immunotherapy, and drug repurposing. We also examine the challenges of tumor heterogeneity, cancer stem cells (CSCs), the tumor microenvironment, exosomes, multiomics studies, and the potential of precision medicine. EXPERT OPINION Because CCA is difficult to diagnose at the early stage, the traditional treatment approaches are not effective for many patients and most tumors recur. Consequently, researchers are exploring multi-aspect molecular carcinogenesis to uncover molecular targets for further development of novel targeted drugs.
Collapse
Affiliation(s)
- Watcharin Loilome
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen Thailand.,Cholangiocarcinoma Screening and Care Program (CASCAP), Khon Kaen University, Khon Kaen, Thailand.,Cholangiocarcinoma Research Institute, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Hasaya Dokduang
- Cholangiocarcinoma Screening and Care Program (CASCAP), Khon Kaen University, Khon Kaen, Thailand.,Cholangiocarcinoma Research Institute, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Manida Suksawat
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen Thailand.,Cholangiocarcinoma Screening and Care Program (CASCAP), Khon Kaen University, Khon Kaen, Thailand.,Cholangiocarcinoma Research Institute, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Sureerat Padthaisong
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen Thailand.,Cholangiocarcinoma Screening and Care Program (CASCAP), Khon Kaen University, Khon Kaen, Thailand.,Cholangiocarcinoma Research Institute, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| |
Collapse
|
12
|
Highly Specific L-Type Amino Acid Transporter 1 Inhibition by JPH203 as a Potential Pan-Cancer Treatment. Processes (Basel) 2021. [DOI: 10.3390/pr9071170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Accelerated cancer cell growth requires a massive intake of amino acids. Overexpression of L-type (large) amino acid transporter 1 (LAT1) on the cancer cell membrane facilitates such a demand, which is limited in normal organs. Therefore, LAT1 overexpression is ideal as a molecular cancer therapeutic target. JPH203, a LAT1-selective non-transportable blocker, had demonstrated LAT1 inhibition in <10 µM IC50 values and effectively suppressed cancer cell growth in studies involving several types of cancer cell lines and tumor xenograft models. A limited phase I clinical trial was performed on five different solid tumors and showed that JPH203 is well-tolerated and has a promising activity for the treatment of bile duct cancer. This review details the development and prospect of JPH203 as a LAT1-targeting cancer therapy.
Collapse
|
13
|
Lopes C, Pereira C, Medeiros R. ASCT2 and LAT1 Contribution to the Hallmarks of Cancer: From a Molecular Perspective to Clinical Translation. Cancers (Basel) 2021; 13:E203. [PMID: 33429909 PMCID: PMC7828050 DOI: 10.3390/cancers13020203] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 12/31/2020] [Accepted: 01/07/2021] [Indexed: 02/06/2023] Open
Abstract
The role of the amino acid transporters ASCT2 and LAT1 in cancer has been explored throughout the years. In this review, we report their impact on the hallmarks of cancer, as well as their clinical significance. Overall, both proteins have been associated with cell death resistance through dysregulation of caspases and sustainment of proliferative signaling through mTOR activation. Furthermore, ASCT2 appears to play an important role in cellular energetics regulation, whereas LAT1 expression is associated with angiogenesis and invasion and metastasis activation. The molecular impact of these proteins on the hallmarks of cancer translates into various clinical applications and both transporters have been identified as prognostic factors in many types of cancer. Concerning their role as therapeutic targets, efforts have been undertaken to synthesize competitive or irreversible ASCT2 and LAT1 inhibitors. However, JHP203, a selective inhibitor of the latter, is, to the best of our knowledge, the only compound included in a Phase 1 clinical trial. In conclusion, considering the usefulness of ASCT2 and LAT1 in a variety of cancer-related pathways and cancer therapy/diagnosis, the development and testing of novel inhibitors for these transporters that could be evaluated in clinical trials represents a promising approach to cancer prognosis improvement.
Collapse
Affiliation(s)
- Catarina Lopes
- Molecular Oncology and Viral Pathology Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal; (C.L.); (R.M.)
| | - Carina Pereira
- Molecular Oncology and Viral Pathology Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal; (C.L.); (R.M.)
- CINTESIS—Center for Health Technology and Services Research, University of Porto, Rua Dr. Plácido da Costa, 4200-450 Porto, Portugal
| | - Rui Medeiros
- Molecular Oncology and Viral Pathology Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal; (C.L.); (R.M.)
- Research Department of the Portuguese League Against Cancer—North (LPCC-NRNorte), Estrada da Circunvalação, 4200-177 Porto, Portugal
| |
Collapse
|
14
|
The Harmonious Interplay of Amino Acid and Monocarboxylate Transporters Induces the Robustness of Cancer Cells. Metabolites 2021; 11:metabo11010027. [PMID: 33401672 PMCID: PMC7823946 DOI: 10.3390/metabo11010027] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/26/2020] [Accepted: 12/27/2020] [Indexed: 02/07/2023] Open
Abstract
There is a growing body of evidence that metabolic reprogramming contributes to the acquisition and maintenance of robustness associated with malignancy. The fine regulation of expression levels of amino acid and monocarboxylate transporters enables cancer cells to exhibit the metabolic reprogramming that is responsible for therapeutic resistance. Amino acid transporters characterized by xCT (SLC7A11), ASCT2 (SLC1A5), and LAT1 (SLC7A5) function in the uptake and export of amino acids such as cystine and glutamine, thereby regulating glutathione synthesis, autophagy, and glutaminolysis. CD44 variant, a cancer stem-like cell marker, stabilizes the xCT antiporter at the cellular membrane, and tumor cells positive for xCT and/or ASCT2 are susceptible to sulfasalazine, a system Xc(-) inhibitor. Inhibiting the interaction between LAT1 and CD98 heavy chain prevents activation of the mammalian target of rapamycin (mTOR) complex 1 by glutamine and leucine. mTOR signaling regulated by LAT1 is a sensor of dynamic alterations in the nutrient tumor microenvironment. LAT1 is overexpressed in various malignancies and positively correlated with poor clinical outcome. Metabolic reprogramming of glutamine occurs often in cancer cells and manifests as ASCT2-mediated glutamine addiction. Monocarboxylate transporters (MCTs) mediate metabolic symbiosis, by which lactate in cancer cells under hypoxia is exported through MCT4 and imported by MCT1 in less hypoxic regions, where it is used as an oxidative metabolite. Differential expression patterns of transporters cause functional intratumoral heterogeneity leading to the therapeutic resistance. Therefore, metabolic reprogramming based on these transporters may be a promising therapeutic target. This review highlights the pathological function and therapeutic targets of transporters including xCT, ASCT2, LAT1, and MCT.
Collapse
|
15
|
A Hierarchy of Proliferative and Migratory Keratinocytes Maintains the Tympanic Membrane. Cell Stem Cell 2020; 28:315-330.e5. [PMID: 33181078 DOI: 10.1016/j.stem.2020.10.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 08/30/2020] [Accepted: 10/14/2020] [Indexed: 12/29/2022]
Abstract
The tympanic membrane (TM) is critical for hearing and requires continuous clearing of cellular debris, but little is known about homeostatic mechanisms in the TM epidermis. Using single-cell RNA sequencing, lineage tracing, whole-organ explant, and live-cell imaging, we show that homeostatic TM epidermis is distinct from other epidermal sites and has discrete proliferative zones with a three-dimensional hierarchy of multiple keratinocyte populations. TM stem cells reside in a discrete location of the superior TM and generate long-lived clones and committed progenitors (CPs). CP clones exhibit lateral migration, and their proliferative capacity is supported by Pdgfra+ fibroblasts, generating migratory but non-proliferative progeny. Single-cell sequencing of the human TM revealed similar cell types and transcriptional programming. Thus, during homeostasis, TM keratinocytes transit through a proliferative CP state and exhibit directional lateral migration. This work forms a foundation for understanding TM disorders and modeling keratinocyte biology.
Collapse
|
16
|
Venkateswaran G, Dedhar S. Interplay of Carbonic Anhydrase IX With Amino Acid and Acid/Base Transporters in the Hypoxic Tumor Microenvironment. Front Cell Dev Biol 2020; 8:602668. [PMID: 33240897 PMCID: PMC7680889 DOI: 10.3389/fcell.2020.602668] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 10/09/2020] [Indexed: 01/13/2023] Open
Abstract
Solid tumors are challenged with a hypoxic and nutrient-deprived microenvironment. Hence, hypoxic tumor cells coordinatively increase the expression of nutrient transporters and pH regulators to adapt and meet their bioenergetic and biosynthetic demands. Carbonic Anhydrase IX (CAIX) is a membrane-bound enzyme that plays a vital role in pH regulation in the tumor microenvironment (TME). Numerous studies have established the importance of CAIX in mediating tumor progression and metastasis. To understand the mechanism of CAIX in mediating tumor progression, we performed an unbiased proteomic screen to identify the potential interactors of CAIX in the TME using the proximity-dependent biotin identification (BioID) technique. In this review, we focus on the interactors from this BioID screen that are crucial for nutrient and metabolite transport in the TME. We discuss the role of transport metabolon comprising CAIX and bicarbonate transporters in regulating intra- and extracellular pH of the tumor. We also discuss the role of amino acid transporters that are high confidence interactors of CAIX, in optimizing favorable metabolic state for tumor progression, and give our perspective on the coordinative interplay of CAIX with the amino acid transporters in the hypoxic TME.
Collapse
Affiliation(s)
- Geetha Venkateswaran
- Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC, Canada.,Interdisciplinary Oncology Program, The University of British Columbia, Vancouver, BC, Canada
| | - Shoukat Dedhar
- Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC, Canada.,Interdisciplinary Oncology Program, The University of British Columbia, Vancouver, BC, Canada.,Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
17
|
Zhang J, Xu Y, Li D, Fu L, Zhang X, Bao Y, Zheng L. Review of the Correlation of LAT1 With Diseases: Mechanism and Treatment. Front Chem 2020; 8:564809. [PMID: 33195053 PMCID: PMC7606929 DOI: 10.3389/fchem.2020.564809] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 09/17/2020] [Indexed: 12/17/2022] Open
Abstract
LAT1 is a member of the system L transporter family. The main role of the LAT1 is to transport specific amino acids through cell membranes to provide nutrients to cells and participate in several metabolic pathways. It also contributes to the transport of hormones and some drugs, which are essential for the development and treatment of some diseases. In recent years, many studies have shown that LAT1 is related to cancer, obesity, diabetes, and other diseases. However, the specific mechanism underlying the influence of LAT1 on such conditions remains unclear. Through the increasing number of studies on LAT1, we have obtained a preliminary understanding on the function of LAT1 in diseases. These studies also provide a theoretical basis for finding treatments for LAT1-related diseases, such as cancer. This review summarizes the function and mechanism of LAT1 in different diseases and the treatment of LAT1-related diseases. It also provides support for the development of novel and reliable disease treatments.
Collapse
Affiliation(s)
- Jingshun Zhang
- Reproductive Medical Center, Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Ying Xu
- Reproductive Medical Center, Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Dandan Li
- Reproductive Medical Center, Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Lulu Fu
- Reproductive Medical Center, Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Xueying Zhang
- Reproductive Medical Center, Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Yigang Bao
- Reproductive Medical Center, Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Lianwen Zheng
- Reproductive Medical Center, Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
18
|
Scalise M, Console L, Rovella F, Galluccio M, Pochini L, Indiveri C. Membrane Transporters for Amino Acids as Players of Cancer Metabolic Rewiring. Cells 2020; 9:cells9092028. [PMID: 32899180 PMCID: PMC7565710 DOI: 10.3390/cells9092028] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 08/31/2020] [Accepted: 09/01/2020] [Indexed: 02/06/2023] Open
Abstract
Cancer cells perform a metabolic rewiring to sustain an increased growth rate and compensate for the redox stress caused by augmented energy metabolism. The metabolic changes are not the same in all cancers. Some features, however, are considered hallmarks of this disease. As an example, all cancer cells rewire the amino acid metabolism for fulfilling both the energy demand and the changed signaling routes. In these altered conditions, some amino acids are more frequently used than others. In any case, the prerequisite for amino acid utilization is the presence of specific transporters in the cell membrane that can guarantee the absorption and the traffic of amino acids among tissues. Tumor cells preferentially use some of these transporters for satisfying their needs. The evidence for this phenomenon is the over-expression of selected transporters, associated with specific cancer types. The knowledge of the link between the over-expression and the metabolic rewiring is crucial for understanding the molecular mechanism of reprogramming in cancer cells. The continuous growth of information on structure-function relationships and the regulation of transporters will open novel perspectives in the fight against human cancers.
Collapse
Affiliation(s)
- Mariafrancesca Scalise
- Unit of Biochemistry and Molecular Biotechnology, Department DiBEST (Biologia, Ecologia, Scienze della Terra), University of Calabria, Via Bucci 4C, 87036 Arcavacata di Rende, Italy; (M.S.); (L.C.); (F.R.); (M.G.); (L.P.)
| | - Lara Console
- Unit of Biochemistry and Molecular Biotechnology, Department DiBEST (Biologia, Ecologia, Scienze della Terra), University of Calabria, Via Bucci 4C, 87036 Arcavacata di Rende, Italy; (M.S.); (L.C.); (F.R.); (M.G.); (L.P.)
| | - Filomena Rovella
- Unit of Biochemistry and Molecular Biotechnology, Department DiBEST (Biologia, Ecologia, Scienze della Terra), University of Calabria, Via Bucci 4C, 87036 Arcavacata di Rende, Italy; (M.S.); (L.C.); (F.R.); (M.G.); (L.P.)
| | - Michele Galluccio
- Unit of Biochemistry and Molecular Biotechnology, Department DiBEST (Biologia, Ecologia, Scienze della Terra), University of Calabria, Via Bucci 4C, 87036 Arcavacata di Rende, Italy; (M.S.); (L.C.); (F.R.); (M.G.); (L.P.)
| | - Lorena Pochini
- Unit of Biochemistry and Molecular Biotechnology, Department DiBEST (Biologia, Ecologia, Scienze della Terra), University of Calabria, Via Bucci 4C, 87036 Arcavacata di Rende, Italy; (M.S.); (L.C.); (F.R.); (M.G.); (L.P.)
| | - Cesare Indiveri
- Unit of Biochemistry and Molecular Biotechnology, Department DiBEST (Biologia, Ecologia, Scienze della Terra), University of Calabria, Via Bucci 4C, 87036 Arcavacata di Rende, Italy; (M.S.); (L.C.); (F.R.); (M.G.); (L.P.)
- CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM) via Amendola 122/O, 70126 Bari, Italy
- Correspondence: ; Tel.: +39-09-8449-2939
| |
Collapse
|
19
|
Xu J, Jiang C, Cai Y, Guo Y, Wang X, Zhang J, Xu J, Xu K, Zhu W, Wang S, Zhang F, Geng M, Han Y, Ning Q, Xu P, Meng L, Lu S. Intervening upregulated SLC7A5 could mitigate inflammatory mediator by mTOR-P70S6K signal in rheumatoid arthritis synoviocytes. Arthritis Res Ther 2020; 22:200. [PMID: 32867828 PMCID: PMC7457370 DOI: 10.1186/s13075-020-02296-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 08/19/2020] [Indexed: 12/12/2022] Open
Abstract
Objective The disruption of metabolic events and changes to nutrient and oxygen availability due to sustained inflammation in RA increases the demand of bioenergetic and biosynthetic processes within the damaged tissue. The current study aimed to understand the molecular mechanisms of SLC7A5 (amino acid transporter) in synoviocytes of RA patients. Methods Synovial tissues were obtained from OA and RA patients. Fibroblast-like synoviocytes (FLS) were isolated, and SLC7A5 expression was examined by using RT-qPCR, immunofluorescence, and Western blotting. RNAi and antibody blocking treatments were used to knockdown SLC7A5 expression or to block its transporter activities. mTOR activity assay and MMP expression levels were monitored in RA FLS under amino acid deprivation or nutrient-rich conditions. Results RA FLS displayed significantly upregulated expression of SLC7A5 compared to OA FLS. Cytokine IL-1β was found to play a crucial role in upregulating SLC7A5 expression via the NF-κB pathway. Intervening SLC7A5 expression with RNAi or blocking its function by monoclonal antibody ameliorated MMP3 and MMP13 protein expression. Conversely, upregulation of SLC7A5 or tryptophan supplementation enhanced mTOR-P70S6K signals which promoted the protein translation of MMP3 and MMP13 in RA FLS. Conclusion Activated NF-κB pathway upregulates SLC7A5, which enhances the mTOR-P70S6K activity and MMP3 and MMP13 expression in RA FLS.
Collapse
Affiliation(s)
- Jing Xu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, People's Republic of China.,Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Congshan Jiang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, People's Republic of China.,Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Yongsong Cai
- Department of Joint Surgery, Xi'an Hong Hui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Yuanxu Guo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, People's Republic of China.,Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Xipeng Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, People's Republic of China.,Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Jiaxiang Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, People's Republic of China.,Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Jiawen Xu
- Department of Joint Surgery, Xi'an Hong Hui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Ke Xu
- Department of Joint Surgery, Xi'an Hong Hui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Wenhua Zhu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, People's Republic of China.,Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Si Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, People's Republic of China.,Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Fujun Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, People's Republic of China.,Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Manman Geng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, People's Republic of China.,Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Yan Han
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, People's Republic of China.,Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Qilan Ning
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, People's Republic of China.,Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Peng Xu
- Department of Joint Surgery, Xi'an Hong Hui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Liesu Meng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, People's Republic of China.,Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Shemin Lu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, People's Republic of China. .,Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, 710061, Shaanxi, People's Republic of China.
| |
Collapse
|
20
|
Chatsirisupachai K, Kitdumrongthum S, Panvongsa W, Janpipatkul K, Worakitchanon W, Lertjintanakit S, Wongtrakoongate P, Chairoungdua A. Expression and roles of system L amino acid transporters in human embryonal carcinoma cells. Andrology 2020; 8:1844-1858. [PMID: 32741077 DOI: 10.1111/andr.12880] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 07/26/2020] [Accepted: 07/28/2020] [Indexed: 01/17/2023]
Abstract
BACKGROUND Testicular germ cell tumors (TGCTs) are the most common malignant cancer in young men. Although TGCTs are generally responsive to platinum-based chemotherapy particularly cisplatin, acquired resistance in patients with metastasis still occurs resulting in poor prognosis. Specifically, differentiation of embryonal carcinoma (EC) cells, the stem cells of TGCTs, can lead to the reduction of cisplatin responsiveness. Therefore, novel therapeutic strategies for TGCTs are needed. System L amino acid transporters have been reported to be up-regulated and to play an important role in tumorigenesis. However, expression and role of system L amino acid transporters in TGCTs remain elusive. MATERIALS AND METHODS Expression of system L amino acid transporters was analyzed in TGCT samples from The Cancer Genome Atlas (TCGA). Expression of LAT1, LAT2, and 4F2hc was examined in human embryonal carcinoma cell line NTERA2. Roles of system L amino acid transporters on NTERA2 cell survival, cell proliferation, pluripotency, and cisplatin sensitivity were evaluated. RESULTS Based upon TCGA datasets, we found that two isoforms of system L (LAT1 and LAT2) and their chaperone protein 4F2hc are highly expressed in EC samples compared with other groups. Treatment with the system L inhibitor BCH significantly suppressed leucine uptake into the pluripotent EC cell line NTERA2. The malignant phenotypes including cell viability, cell proliferation, and clonal ability were decreased following BCH treatment. Nonetheless, system L inhibition did not alter expression of stemness genes in NTERA2 cells. After NTERA2 differentiation, expressions of LAT1 and LAT2 were decreased. Finally, co-administration of BCH enhanced cisplatin sensitivity in both undifferentiated and differentiated cells. These effects were associated with the reduction in p70S6K phosphorylation. CONCLUSION Taken together, these results shed light on the roles of system L amino acid transporters in TGCTs. Therefore, system L amino acid transporters could provide novel therapeutic targets for treatment against TGCTs.
Collapse
Affiliation(s)
| | | | - Wittaya Panvongsa
- Toxicology Graduate Program, Faculty of Science, Mahidol University, Bangkok, Thailand
| | | | | | - Sarat Lertjintanakit
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Patompon Wongtrakoongate
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok, Thailand.,Center for Neuroscience, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Arthit Chairoungdua
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand.,Toxicology Graduate Program, Faculty of Science, Mahidol University, Bangkok, Thailand.,Excellent Center for Drug Discovery (ECDD), Mahidol University, Bangkok, Thailand
| |
Collapse
|
21
|
Enomoto K, Hotomi M. Amino Acid Transporters as Potential Therapeutic Targets in Thyroid Cancer. Endocrinol Metab (Seoul) 2020; 35:227-236. [PMID: 32615707 PMCID: PMC7386108 DOI: 10.3803/enm.2020.35.2.227] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 06/09/2020] [Indexed: 12/31/2022] Open
Abstract
Thyroid cancer cells have a high amino acid demand for proliferation, invasion, and metastasis. Amino acids are taken up by thyroid cancer cells, both thyroid follicular cell and thyroid parafollicular cells (commonly called "C-cells"), via amino acid transporters. Amino acid transporters up-regulate in many cancers, and their expression level associate with clinical aggressiveness and prognosis. This is the review to discuss the therapeutic potential of amino acid transporters and as molecular targets in thyroid cancer.
Collapse
Affiliation(s)
- Keisuke Enomoto
- Department of Otolaryngology-Head and Neck Surgery, Wakayama Medical University, Wakayama, Japan
| | - Muneki Hotomi
- Department of Otolaryngology-Head and Neck Surgery, Wakayama Medical University, Wakayama, Japan
| |
Collapse
|
22
|
Abstract
PURPOSE This study aims to explore whether 4-(2S,4R)-[18F]fluoroglutamine (4-[18F]FGln) positron emission tomography (PET) imaging is helpful in identifying and monitoring MYCN-amplified neuroblastoma by enhanced glutamine metabolism. PROCEDURES Cell uptake studies and dynamic small-animal PET studies of 4-[18F]FGln and 2-deoxy-2-[18F]fluoro-D-glucose ([18F]FDG) were conducted in human MYCN-amplified (IMR-32 and SK-N-BE (2) cells) and non-MYCN-amplified (SH-SY5Y cell) neuroblastoma cells and animal models. Subsequently, short hairpin RNA (shRNA) knockdown of alanine-serine-cysteine transporter 2 (ASCT2/SLC1A5) in IMR-32 cells and xenografts were investigated in vitro and in vivo. Western blot (WB), real-time polymerase chain reaction (RT-PCR), and immunofluorescence (IF) assays were used to measure the prevalence of ASCT2, Ki-67, and c-Caspase 3, respectively. RESULTS IMR-32 and SK-N-BE (2) cells showed high glutamine uptake in vitro (31.6 ± 1.7 and 21.6 ± 6.6 %ID/100 μg). In the in vivo study, 4-[18F]FGln was localized in IMR-32, SK-N-BE (2), and SH-SY5Y tumors with a high uptake (6.6 ± 0.3, 5.6 ± 0.2, and 3.7 ± 0.1 %ID/g). The maximum uptake (tumor-to-muscle, T/M) of the IMR-32 and SK-N-BE (2) tumors (3.71 and 2.63) was significantly higher than that of SH-SY5Y (1.54) tumors (P < 0.001, P < 0.001). The maximum uptake of 4-[18F]FGln in IMR-32 and SK-N-BE (2) tumors was 2.3-fold and 2.1-fold higher than that of [18F]FDG, respectively. Furthermore, in the in vitro and in vivo studies, the maximum uptake of 4-[18F]FGln in shASCT2-IMR-32 cells and tumors was 2.1-fold and 2.5-fold lower than that of the shControl-IMR-32. No significant difference in [18F]FDG uptake was found between shASCT2-IMR-32 and shControl-IMR-32 cells and tumors. CONCLUSION 4-[18F]FGln PET can provide a valuable clinical tool in the assessment of metabolic glutamine uptake in MYCN-amplified neuroblastoma. ASCT2-targeted therapy may provide a supplementary method in MYCN-amplified neuroblastoma treatment.
Collapse
|
23
|
Sakata T, Hana K, Mikami T, Yoshida T, Endou H, Okayasu I. Positive correlation of expression of L-type amino-acid transporter 1 with colorectal tumor progression and prognosis: Higher expression in sporadic colorectal tumors compared with ulcerative colitis-associated neoplasia. Pathol Res Pract 2020; 216:152972. [PMID: 32359697 DOI: 10.1016/j.prp.2020.152972] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 04/04/2020] [Accepted: 04/11/2020] [Indexed: 01/06/2023]
Abstract
The role of L-type amino-acid transporter 1 (LAT1), an oncofetal protein, in tumor progression is not well known, although it is important for the survival and proliferation of cancer cells. LAT1 expression was immunohistochemically analyzed and compared in sporadic (conventional) colorectal tumors and ulcerative colitis (UC)-associated neoplasia development and progression. LAT1 expression showed a significant stepwise increase in the order: conventional low-grade tubular adenoma, high-grade tubular adenoma, and invasive adenocarcinoma. Similarly, the same increasing trend in LAT1 expression was found in UC-associated low-grade dysplasia, high-grade dysplasia, and adenocarcinoma, whereas expression was significantly lower compared with that in an adenoma-adenocarcinoma series. LAT1 expression was predominant in the upper half of mucosal lesions in low-grade adenoma. This localized difference in LAT1 expression between the upper and lower halves of mucosal lesions disappeared in conventional high-grade adenoma and adenocarcinoma. LAT1 expression in the colorectal mucosa was significantly increased in the order: nontumor mucosa, quiescent phase of UC, and active phase of UC. Considering the histological pattern of Ki-67 labeling, LAT1 expression appeared partly related to cell proliferation, but this was not significant. In relation to the prognosis of patients with sporadic phase IV colorectal adenocarcinoma, this was significantly poorer in the group with high LAT1 expression compared with that with low LAT1 expression. This suggests LAT1 expression may be used as a companion biomarker for anti-cancer therapy targeting the LAT1 molecule in colorectal cancers.
Collapse
Affiliation(s)
| | | | - Tetuo Mikami
- Department of Pathology, Toho University School of Medicine, Tokyo, Japan
| | - Tutomu Yoshida
- Department of Pathology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | | | - Isao Okayasu
- Department of Pathology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan; Division of Nutrition, Faculty of Health Care, Kiryu University, Midori, Japan.
| |
Collapse
|
24
|
Okano N, Naruge D, Kawai K, Kobayashi T, Nagashima F, Endou H, Furuse J. First-in-human phase I study of JPH203, an L-type amino acid transporter 1 inhibitor, in patients with advanced solid tumors. Invest New Drugs 2020; 38:1495-1506. [PMID: 32198649 DOI: 10.1007/s10637-020-00924-3] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 03/11/2020] [Indexed: 11/28/2022]
Abstract
This open-label first-in-human study evaluated JPH203, which is a novel selective L-type amino acid transporter 1 inhibitor. We also evaluated the association between the N-acetyltransferase 2 phenotype and outcomes. Japanese patients with advanced solid tumors received daily intravenous JPH203 treatment for 7 days, followed by a 21-day rest period, at escalating doses of 12-85 mg/m2. Dose-limiting toxicities were evaluated during the first cycle using a 3 + 3 design. The study enrolled 17 patients, although grade 3 liver dysfunction was detected in one of six patients receiving 60 mg/m2 and in the first patient to receive 85 mg/m2. Further enrollment was terminated and the maximum tolerated dose was defined as 60 mg/m2. The AUC∞ increased between 12 mg/m2 and 25 mg/m2, although no differences were observed at 25-40 mg/m2. Partial response was observed for one patient with biliary tract cancer (BTC) at the 12 mg/m2 dose, and disease control was achieved by 3 of 6 patients at the 12 mg/m2 and 25 mg/m2 dose levels. Based on these results, we recommend a phase II dose of 25 mg/m2. The disease control rate for BTC was 60%. Two patients with grade 3 liver dysfunction had the rapid N-acetyltransferase 2 phenotype, and disease control was more common for the non-rapid phenotype (50% vs. 12.5%). It appears that JPH203 was well-tolerated and provided promising activity against BTC. The N-acetyltransferase 2 phenotype might help predict the safety and efficacy of JPH203. Clinical trial registration: UMIN000016546.
Collapse
Affiliation(s)
- Naohiro Okano
- Department of Medical Oncology, Kyorin University Faculty of Medicine, 6-20-2, Shinkawa, Mitaka-shi, Tokyo, 181-8611, Japan.
| | - Daisuke Naruge
- Department of Medical Oncology, Kyorin University Faculty of Medicine, 6-20-2, Shinkawa, Mitaka-shi, Tokyo, 181-8611, Japan
| | - Kirio Kawai
- Department of Medical Oncology, Kyorin University Faculty of Medicine, 6-20-2, Shinkawa, Mitaka-shi, Tokyo, 181-8611, Japan
| | - Takaaki Kobayashi
- Department of Medical Oncology, Kyorin University Faculty of Medicine, 6-20-2, Shinkawa, Mitaka-shi, Tokyo, 181-8611, Japan
| | - Fumio Nagashima
- Department of Medical Oncology, Kyorin University Faculty of Medicine, 6-20-2, Shinkawa, Mitaka-shi, Tokyo, 181-8611, Japan
| | | | - Junji Furuse
- Department of Medical Oncology, Kyorin University Faculty of Medicine, 6-20-2, Shinkawa, Mitaka-shi, Tokyo, 181-8611, Japan
| |
Collapse
|
25
|
Multifaceted Aspects of Metabolic Plasticity in Human Cholangiocarcinoma: An Overview of Current Perspectives. Cells 2020; 9:cells9030596. [PMID: 32138158 PMCID: PMC7140515 DOI: 10.3390/cells9030596] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 02/24/2020] [Accepted: 02/26/2020] [Indexed: 12/14/2022] Open
Abstract
Cholangiocarcinoma (CCA) is a deadly tumor without an effective therapy. Unique metabolic and bioenergetics features are important hallmarks of tumor cells. Metabolic plasticity allows cancer cells to survive in poor nutrient environments and maximize cell growth by sustaining survival, proliferation, and metastasis. In recent years, an increasing number of studies have shown that specific signaling networks contribute to malignant tumor onset by reprogramming metabolic traits. Several evidences demonstrate that numerous metabolic mediators represent key-players of CCA progression by regulating many signaling pathways. Besides the well-known Warburg effect, several other different pathways involving carbohydrates, proteins, lipids, and nucleic acids metabolism are altered in CCA. The goal of this review is to highlight the main metabolic processes involved in the cholangio-carcinogeneis that might be considered as potential novel druggable candidates for this disease.
Collapse
|
26
|
Plasma Membrane Transporters as Biomarkers and Molecular Targets in Cholangiocarcinoma. Cells 2020; 9:cells9020498. [PMID: 32098199 PMCID: PMC7072733 DOI: 10.3390/cells9020498] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Revised: 02/18/2020] [Accepted: 02/19/2020] [Indexed: 12/18/2022] Open
Abstract
The dismal prognosis of patients with advanced cholangiocarcinoma (CCA) is due, in part, to the extreme resistance of this type of liver cancer to available chemotherapeutic agents. Among the complex mechanisms accounting for CCA chemoresistance are those involving the impairment of drug uptake, which mainly occurs through transporters of the superfamily of solute carrier (SLC) proteins, and the active export of drugs from cancer cells, mainly through members of families B, C and G of ATP-binding cassette (ABC) proteins. Both mechanisms result in decreased amounts of active drugs able to reach their intracellular targets. Therefore, the “cancer transportome”, defined as the set of transporters expressed at a given moment in the tumor, is an essential element for defining the multidrug resistance (MDR) phenotype of cancer cells. For this reason, during the last two decades, plasma membrane transporters have been envisaged as targets for the development of strategies aimed at sensitizing cancer cells to chemotherapy, either by increasing the uptake or reducing the export of antitumor agents by modulating the expression/function of SLC and ABC proteins, respectively. Moreover, since some elements of the transportome are differentially expressed in CCA, their usefulness as biomarkers with diagnostic and prognostic purposes in CCA patients has been evaluated.
Collapse
|
27
|
Satriano L, Lewinska M, Rodrigues PM, Banales JM, Andersen JB. Metabolic rearrangements in primary liver cancers: cause and consequences. Nat Rev Gastroenterol Hepatol 2019; 16:748-766. [PMID: 31666728 DOI: 10.1038/s41575-019-0217-8] [Citation(s) in RCA: 163] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/19/2019] [Indexed: 02/07/2023]
Abstract
Primary liver cancer (PLC) is the fourth most frequent cause of cancer-related death. The high mortality rates arise from late diagnosis and the limited accuracy of diagnostic and prognostic biomarkers. The liver is a major regulator, orchestrating the clearance of toxins, balancing glucose, lipid and amino acid uptake, managing whole-body metabolism and maintaining metabolic homeostasis. Tumour onset and progression is frequently accompanied by rearrangements of metabolic pathways, leading to dysregulation of metabolism. The limitation of current therapies targeting PLCs, such as hepatocellular carcinoma and cholangiocarcinoma, points towards the importance of deciphering this metabolic complexity. In this Review, we discuss the role of metabolic liver disruptions and the implications of these processes in PLCs, emphasizing their clinical relevance and value in early diagnosis and prognosis and as putative therapeutic targets. We also describe system biology approaches able to reconstruct the metabolic complexity of liver diseases. We also discuss whether metabolic rearrangements are a cause or consequence of PLCs, emphasizing the opportunity to clinically exploit the rewired metabolism. In line with this idea, we discuss circulating metabolites as promising biomarkers for PLCs.
Collapse
Affiliation(s)
- Letizia Satriano
- Biotech Research and Innovation Centre (BRIC) Department of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Monika Lewinska
- Biotech Research and Innovation Centre (BRIC) Department of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Pedro M Rodrigues
- Biodonostia Health Research Institute, Donostia University Hospital, San Sebastian, Spain
| | - Jesus M Banales
- Biodonostia Health Research Institute, Donostia University Hospital, San Sebastian, Spain.,National Institute for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Carlos III National Institute of Health, Madrid, Spain.,IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Jesper B Andersen
- Biotech Research and Innovation Centre (BRIC) Department of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
28
|
Häfliger P, Graff J, Rubin M, Stooss A, Dettmer MS, Altmann KH, Gertsch J, Charles RP. The LAT1 inhibitor JPH203 reduces growth of thyroid carcinoma in a fully immunocompetent mouse model. J Exp Clin Cancer Res 2018; 37:234. [PMID: 30241549 PMCID: PMC6150977 DOI: 10.1186/s13046-018-0907-z] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 09/11/2018] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND The L-type amino acid transporter 1 (LAT1/SLC7A5) transports essential amino acids across the plasma membrane. While LAT1 is overexpressed in a variety of human neoplasms, its expression and its role in thyroid cancer is currently unknown. Anaplastic thyroid carcinoma (ATC) is a highly aggressive malignancy for which no effective therapy exists. The purpose of this study was to explore whether the inhibition of LAT1 in ATC would affect tumor growth both in vitro and in vivo. METHODS LAT1 was pharmacologically blocked by JPH203 in human ATC and papillary thyroid cancer (PTC) cell lines. The effects on proliferation and mTORC1 activity were addressed in vitro. A genetically engineered mouse model of ATC was used to address the effect of blocking LAT1 on tumor growth in vivo. SLC7A5 transcription was measured in patient-derived ATC samples to address the clinical relevance of the findings. RESULTS LAT1 block by JPH203 reduced proliferation and mTORC1 signaling in human thyroid cancer cell lines. SLC7A5 transcription was upregulated in ATC tissues derived from a genetically engineered mouse model and in ATC samples recovered from patients. JPH203 treatment induced thyroid tumor growth arrest in vivo in a fully immunocompetent mouse model of thyroid cancer. Additionally, analysis of publicly available datasets of thyroid carcinomas revealed that high LAT1 expression is associated with potentially untreatable PTC presenting reduced NIS/SLC5A5 transcription and with ATC. CONCLUSIONS These preclinical results show that LAT1 inhibition is a novel therapeutic approach in the context of thyroid cancers, and more interestingly in untreatable thyroid cancers.
Collapse
Affiliation(s)
- Pascal Häfliger
- Institute of Biochemistry and Molecular Medicine, and Swiss National Center of Competence in Research (NCCR) TransCure, University of Bern, Bühlstrasse 28, CH-3012 Bern, Switzerland
- Present address: Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, USA
| | - Julien Graff
- Institute of Pharmaceutical Sciences, and Swiss National Center of Competence in Research (NCCR) TransCure, ETH Zürich, Vladimir-Prelog-Weg 4, CH-8093 Zürich, Switzerland
| | - Matthias Rubin
- Institute of Biochemistry and Molecular Medicine, and Swiss National Center of Competence in Research (NCCR) TransCure, University of Bern, Bühlstrasse 28, CH-3012 Bern, Switzerland
| | - Amandine Stooss
- Institute of Biochemistry and Molecular Medicine, and Swiss National Center of Competence in Research (NCCR) TransCure, University of Bern, Bühlstrasse 28, CH-3012 Bern, Switzerland
| | - Matthias S. Dettmer
- Institute of Pathology, University of Bern, Murtenstrasse 31, CH-3008 Bern, Switzerland
| | - Karl-Heinz Altmann
- Institute of Pharmaceutical Sciences, and Swiss National Center of Competence in Research (NCCR) TransCure, ETH Zürich, Vladimir-Prelog-Weg 4, CH-8093 Zürich, Switzerland
| | - Jürg Gertsch
- Institute of Biochemistry and Molecular Medicine, and Swiss National Center of Competence in Research (NCCR) TransCure, University of Bern, Bühlstrasse 28, CH-3012 Bern, Switzerland
| | - Roch-Philippe Charles
- Institute of Biochemistry and Molecular Medicine, and Swiss National Center of Competence in Research (NCCR) TransCure, University of Bern, Bühlstrasse 28, CH-3012 Bern, Switzerland
| |
Collapse
|
29
|
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
|
30
|
|
31
|
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
|
32
|
Fibroblast-like synoviocyte migration is enhanced by IL-17-mediated overexpression of L-type amino acid transporter 1 (LAT1) via the mTOR/4E-BP1 pathway. Amino Acids 2017; 50:331-340. [PMID: 29198077 DOI: 10.1007/s00726-017-2520-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 11/28/2017] [Indexed: 01/25/2023]
Abstract
In rheumatoid arthritis (RA), activated synovial fibroblasts have the ability to invade joint cartilage, actively contributing to joint destruction in RA. The mechanisms underlying this cell migration and invasion remain unclear. Our previous results and data from the GEO profile indicate that the L-type amino acid transporter gene, LAT1, is overexpressed in the synovium of RA. To identify its potential role in RA, fibroblast-like synoviocytes (FLS) from patients with RA were used to determine the effects of suppressing the LAT1 genes using RNA interference and the LAT inhibitor, BCH. We found that BCH exposure reduced the phosphorylation of mTOR and its downstream target 4EBP1, radiolabeled leucine uptake, and migration of RA FLS. LAT1 silencing by siRNA presented effects similar to BCH inhibition. Treatment of cells with IL-17 stimulated the expression of LAT1. In contrast, applying an inhibitor of mTOR pathway, temsirolimus, or silencing eIF4E neutralized the stimulation of IL-17 on LAT1. BCH and siLAT1 also resulted in lower IL-17-stimulated leucine uptake and cell migration. These results suggest that the migration of RA FLS is aggravated by IL-17-mediated overexpression of LAT1 via mTOR/4E-BP1 pathway. In conclusion, further investigation is warranted into LAT1 as a potential target for drug therapies aimed at attenuating migration of transformed-appearing fibroblasts and subsequently preventing further erosion of bone and cartilage.
Collapse
|
33
|
Metabolic re-wiring of isogenic breast epithelial cell lines following epithelial to mesenchymal transition. Cancer Lett 2017; 396:117-129. [DOI: 10.1016/j.canlet.2017.03.019] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 03/03/2017] [Accepted: 03/12/2017] [Indexed: 12/22/2022]
|
34
|
Yothaisong S, Dokduang H, Anzai N, Hayashi K, Namwat N, Yongvanit P, Sangkhamanon S, Jutabha P, Endou H, Loilome W. Inhibition of l-type amino acid transporter 1 activity as a new therapeutic target for cholangiocarcinoma treatment. Tumour Biol 2017; 39:1010428317694545. [PMID: 28347255 DOI: 10.1177/1010428317694545] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Unlike normal cells, cancer cells undergo unlimited growth and multiplication, causing them to require massive amounts of amino acid to support their continuous metabolism. Among the amino acid transporters expressed on the plasma membrane, l-type amino acid transporter-1, a Na+-independent neutral amino acid transporter, is highly expressed in many types of human cancer including cholangiocarcinoma. Our previous study reported that l-type amino acid transporter-1 and its co-functional protein CD98 were highly expressed and implicated in cholangiocarcinoma progression and carcinogenesis. Therefore, this study determined the effect of JPH203, a selective inhibitor of l-type amino acid transporter-1 activity, on cholangiocarcinoma cell inhibition both in vitro and in vivo. JPH203 dramatically suppressed [14C]l-leucine uptake as well as cell growth in cholangiocarcinoma cell lines along with altering the expression of l-type amino acid transporter-1 and CD98 in response to amino acid depletion. We also demonstrated that JPH203 induced both G2/M and G0/G1 cell cycle arrest, as well as reduced the S phase accompanied by altered expression of the proteins in cell cycle progression: cyclin D1, CDK4, and CDK6. There was also cell cycle arrest of the related proteins, P21 and P27, in KKU-055 and KKU-213 cholangiocarcinoma cells. Apoptosis induction, detected by an increase in trypan blue-stained cells along with a cleaved caspase-3/caspase-3 ratio, occurred in JPH203-treated cholangiocarcinoma cells at the highest concentration tested (100 µM). As expected, daily intravenous administration of JPH203 (12.5 and 25 mg/kg) significantly inhibited tumor growth in KKU-213 cholangiocarcinoma cell xenografts in the nude mice model in a dose-dependent manner with no statistically significant change in the animal's body weight and with no differences in the histology and appearance of the internal organs compared with the control group. Our study demonstrates that suppression of l-type amino acid transporter-1 activity using JPH203 might be used as a new therapeutic strategy for cholangiocarcinoma treatment.
Collapse
Affiliation(s)
- Supak Yothaisong
- 1 Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,2 Liver Fluke and Cholangiocarcinoma Research Center, Khon Kaen University, Khon Kaen, Thailand.,3 Cholangiocarcinoma Screening and Care Program (CASCAP), Khon Kaen University, Khon Kaen, Thailand
| | - Hasaya Dokduang
- 2 Liver Fluke and Cholangiocarcinoma Research Center, Khon Kaen University, Khon Kaen, Thailand.,3 Cholangiocarcinoma Screening and Care Program (CASCAP), Khon Kaen University, Khon Kaen, Thailand
| | - Naohiko Anzai
- 4 Department of Pharmacology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Keitaro Hayashi
- 5 Department of Pharmacology and Toxicology, School of Medicine, Dokkyo Medical University, Mibu, Japan
| | - Nisana Namwat
- 1 Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,2 Liver Fluke and Cholangiocarcinoma Research Center, Khon Kaen University, Khon Kaen, Thailand.,3 Cholangiocarcinoma Screening and Care Program (CASCAP), Khon Kaen University, Khon Kaen, Thailand
| | - Puangrat Yongvanit
- 1 Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,2 Liver Fluke and Cholangiocarcinoma Research Center, Khon Kaen University, Khon Kaen, Thailand.,3 Cholangiocarcinoma Screening and Care Program (CASCAP), Khon Kaen University, Khon Kaen, Thailand
| | - Sakkarn Sangkhamanon
- 6 Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Promsuk Jutabha
- 5 Department of Pharmacology and Toxicology, School of Medicine, Dokkyo Medical University, Mibu, Japan
| | | | - Watcharin Loilome
- 1 Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,2 Liver Fluke and Cholangiocarcinoma Research Center, Khon Kaen University, Khon Kaen, Thailand.,3 Cholangiocarcinoma Screening and Care Program (CASCAP), Khon Kaen University, Khon Kaen, Thailand
| |
Collapse
|
35
|
Hayashi K, Anzai N. Novel therapeutic approaches targeting L-type amino acid transporters for cancer treatment. World J Gastrointest Oncol 2017; 9:21-29. [PMID: 28144396 PMCID: PMC5241523 DOI: 10.4251/wjgo.v9.i1.21] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 10/08/2016] [Accepted: 11/02/2016] [Indexed: 02/05/2023] Open
Abstract
L-type amino acid transporters (LATs) mainly assist the uptake of neutral amino acids into cells. Four LATs (LAT1, LAT2, LAT3 and LAT4) have so far been identified. LAT1 (SLC7A5) has been attracting much attention in the field of cancer research since it is commonly up-regulated in various cancers. Basic research has made it increasingly clear that LAT1 plays a predominant role in malignancy. The functional significance of LAT1 in cancer and the potential therapeutic application of the features of LAT1 to cancer management are described in this review.
Collapse
|
36
|
He B, Zhang N, Zhao R. Dexamethasone Downregulates SLC7A5 Expression and Promotes Cell Cycle Arrest, Autophagy and Apoptosis in BeWo Cells. J Cell Physiol 2016; 231:233-42. [PMID: 26094588 DOI: 10.1002/jcp.25076] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 06/08/2015] [Indexed: 12/14/2022]
Abstract
Synthetic glucocorticoids (GCs) such as dexamethasone (Dex) are widely given to pregnant women to induce maturation and improve viability of preterm infants. Despite the beneficial effects, synthetic GCs have adverse effects on placental growth and nutrient transport system. However, the molecular mechanisms involved in these events remain unknown. Here we use a human placental choriocarcinoma cell line (BeWo) as model to explore the pathway linking amino acids transport with cell viability under Dex challenge. BeWo cells treated with Dex (100 nM) for 24 h demonstrated G1/S cell cycle arrest together with enhanced autophagy and apoptosis. Concurrently, the amino acid carrier SLC7A5 was down-regulated in association with impaired cellular amino acids uptake and inhibition of mammalian target of rapamycin (mTOR) signaling. Similar cellular responses were observed in BeWo cells treated with BCH, a classical System L inhibitor which inactivates SLC7A5. The glucocorticoid receptor (GR) antagonist RU486 was able to diminish Dex-induced translocation of GR into nucleus and to abolish these effects. Furthermore, Dex treatment significantly promoted the binding of GR to the proximal promoter sequence of SLC7A5 gene. Taken together, our results show that Dex downregulates SLC7A5 expression via GR-mediated transrepression. The impaired amino acids uptake leads to inhibition of mTOR signaling which in turn causes inhibited proliferation and enhanced autophagy and apoptosis in BeWo cells. These findings indicate that SLC7A5 mediates the effect of Dex on cell viability, thus providing a novel molecular target for the prevention and treatment of Dex-induced cell cycle arrest and apoptosis in placental cells.
Collapse
Affiliation(s)
- Bin He
- Key Laboratory of Animal Physiology and Biochemistry, Ministry of Agriculture, Nanjing Agricultural University, Nanjing, China
| | - Nana Zhang
- Key Laboratory of Animal Physiology and Biochemistry, Ministry of Agriculture, Nanjing Agricultural University, Nanjing, China
| | - Ruqian Zhao
- Key Laboratory of Animal Physiology and Biochemistry, Ministry of Agriculture, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
37
|
Chen WQ, Hu L, Chen GX, Deng HX. Role of microRNA-7 in digestive system malignancy. World J Gastrointest Oncol 2016; 8:121-127. [PMID: 26798443 PMCID: PMC4714141 DOI: 10.4251/wjgo.v8.i1.121] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 11/19/2015] [Indexed: 02/05/2023] Open
Abstract
There are several malignancies of the digestive system (including gastric, pancreatic and colorectal cancers, and hepatocellular carcinoma), which are the most common types of cancer and a major cause of death worldwide. MicroRNA (miR)-7 is abundant in the pancreas, playing an important role in pancreatic development and endocrine function. Expression of miR-7 is downregulated in digestive system malignancies compared with normal tissue. Although there are contrasting results for miR-7 expression, almost all research reveals that miR-7 is a tumor suppressor, by targeting various genes in specific pathways. Moreover, miR-7 can target different genes simultaneously in different malignancies of the digestive system. By acting on many cytokines, miR-7 is also involved in many gastrointestinal inflammatory diseases as a significant carcinogenic factor. Consequently, miR-7 might be a biomarker or therapeutic target gene in digestive system malignancies.
Collapse
|
38
|
Yothaisong S, Namwat N, Yongvanit P, Khuntikeo N, Puapairoj A, Jutabha P, Anzai N, Tassaneeyakul W, Tangsucharit P, Loilome W. Increase in L-type amino acid transporter 1 expression during cholangiocarcinogenesis caused by liver fluke infection and its prognostic significance. Parasitol Int 2015; 66:471-478. [PMID: 26657242 DOI: 10.1016/j.parint.2015.11.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 10/29/2015] [Accepted: 11/30/2015] [Indexed: 11/18/2022]
Abstract
L-type amino acid transporter 1 (LAT1) is highly expressed in various human cancers, including cholangiocarcinoma (CCA), the most common cancer in Northeast Thailand. Chronic inflammation and oxidative stress induced by liver fluke, Opisthorchis viverrini, infection has been recognized as the major cause of CCA in this area. We show here that an increased expression of LAT1 and its co-functional protein CD98 are found during carcinogenesis induced by Ov in hamster CCA tissues. We also demonstrate that oxidative stress induced by H2O2 is time-dependent and dramatically activates LAT1 and CD98 expression in immortal cholangiocytes (MMNK1). In addition, H2O2 treatment increased LAT1 and CD98 expression, as well as an activated form of AKT and mTOR in MMNK1 and CCA cell lines (KKU-M055 and KKU-M213). We also show that suppression of PI3K/AKT pathway activity with a dual PI3K/mTOR inhibitor, BEZ235, causes a reduction in LAT1 and CD98 expression in KKU-M055 and KKU-M213 in parallel with a reduction of activated AKT and mTOR. Interestingly, high expression of LAT1 in human CCA tissues is a significant prognostic factor for shorter survival. Taken together, our data show that LAT1 expression is significantly associated with CCA progression and cholangiocarcinogenesis induced by oxidative stress. Moreover, the expression of LAT1 and CD98 in CCA is possibly regulated by the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Supak Yothaisong
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; Liver Fluke and Cholangiocarcinoma Research Center, Khon Kaen University, Khon Kaen 40002, Thailand; Cholangiocarcinoma Screening and Care Program (CASCAP), Khon Kaen University, Khon Kaen 40002, Thailand
| | - Nisana Namwat
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; Liver Fluke and Cholangiocarcinoma Research Center, Khon Kaen University, Khon Kaen 40002, Thailand; Cholangiocarcinoma Screening and Care Program (CASCAP), Khon Kaen University, Khon Kaen 40002, Thailand
| | - Puangrat Yongvanit
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; Liver Fluke and Cholangiocarcinoma Research Center, Khon Kaen University, Khon Kaen 40002, Thailand; Cholangiocarcinoma Screening and Care Program (CASCAP), Khon Kaen University, Khon Kaen 40002, Thailand
| | - Narong Khuntikeo
- Department of Surgery, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; Liver Fluke and Cholangiocarcinoma Research Center, Khon Kaen University, Khon Kaen 40002, Thailand; Cholangiocarcinoma Screening and Care Program (CASCAP), Khon Kaen University, Khon Kaen 40002, Thailand
| | - Anucha Puapairoj
- Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; Liver Fluke and Cholangiocarcinoma Research Center, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Promsuk Jutabha
- Department of Pharmacology and Toxicology, Dokkyo Medical University School of Medicine, Tochigi 321-0293, Japan
| | - Naohiko Anzai
- Department of Pharmacology and Toxicology, Dokkyo Medical University School of Medicine, Tochigi 321-0293, Japan
| | - Wichittra Tassaneeyakul
- Department of Pharmacology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Panot Tangsucharit
- Department of Pharmacology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Watcharin Loilome
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; Liver Fluke and Cholangiocarcinoma Research Center, Khon Kaen University, Khon Kaen 40002, Thailand; Cholangiocarcinoma Screening and Care Program (CASCAP), Khon Kaen University, Khon Kaen 40002, Thailand.
| |
Collapse
|
39
|
Zhao Y, Wang L, Pan J. The role of L-type amino acid transporter 1 in human tumors. Intractable Rare Dis Res 2015; 4:165-9. [PMID: 26668776 PMCID: PMC4660857 DOI: 10.5582/irdr.2015.01024] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Revised: 08/14/2015] [Accepted: 08/18/2015] [Indexed: 12/17/2022] Open
Abstract
L-type amino acid transporter 1 (LAT1) is an L-type amino acid transporter and transports large neutral amino acids such as leucine, isoleucine, valine, phenylalanine, tyrosine, tryptophan, methionine, and histidine. LAT1 was found to be highly expressed especially in human cancer tissues, and up-regulated LAT1 can lead to dysfunction in human tumor cells. These findings suggest that LAT1 plays an important role in human tumors. This review provides an overview of the current understanding of LAT1 expression and its clinical significance and function in tumors.
Collapse
Affiliation(s)
- Yu Zhao
- University of Ji'nan Shandong Academy of Medical Science School of Medicine and Life Science, Ji'nan, China
- Shandong Medicinal Biotechnology Center, Ji'nan, China
| | - Lin Wang
- University of Ji'nan Shandong Academy of Medical Science School of Medicine and Life Science, Ji'nan, China
- Shandong Medicinal Biotechnology Center, Ji'nan, China
| | - Jihong Pan
- University of Ji'nan Shandong Academy of Medical Science School of Medicine and Life Science, Ji'nan, China
- Shandong Medicinal Biotechnology Center, Ji'nan, China
- Key Laboratory for Rare Diseases of Shandong Province, Ji'nan, China
| |
Collapse
|
40
|
Chen L, Cui H. Targeting Glutamine Induces Apoptosis: A Cancer Therapy Approach. Int J Mol Sci 2015; 16:22830-55. [PMID: 26402672 PMCID: PMC4613338 DOI: 10.3390/ijms160922830] [Citation(s) in RCA: 129] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Revised: 09/11/2015] [Accepted: 09/15/2015] [Indexed: 02/06/2023] Open
Abstract
Glutamine metabolism has been proved to be dysregulated in many cancer cells, and is essential for proliferation of most cancer cells, which makes glutamine an appealing target for cancer therapy. In order to be well used by cells, glutamine must be transported to cells by specific transporters and converted to glutamate by glutaminase. There are currently several drugs that target glutaminase under development or clinical trials. Also, glutamine metabolism restriction has been proved to be effective in inhibiting tumor growth both in vivo and vitro through inducing apoptosis, growth arrest and/or autophagy. Here, we review recent researches about glutamine metabolism in cancer, and cell death induced by targeting glutamine, and their potential roles in cancer therapy.
Collapse
Affiliation(s)
- Lian Chen
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Ya'an 625014, China.
| | - Hengmin Cui
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Ya'an 625014, China.
- College of Veterinary Medicine, Sichuan Agricultural University, Ya'an 625014, China.
| |
Collapse
|
41
|
Dutta S, Reamtong O, Panvongsa W, Kitdumrongthum S, Janpipatkul K, Sangvanich P, Piyachaturawat P, Chairoungdua A. Proteomics profiling of cholangiocarcinoma exosomes: A potential role of oncogenic protein transferring in cancer progression. Biochim Biophys Acta Mol Basis Dis 2015; 1852:1989-99. [PMID: 26148937 DOI: 10.1016/j.bbadis.2015.06.024] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Revised: 06/27/2015] [Accepted: 06/29/2015] [Indexed: 02/06/2023]
Abstract
Cholangiocarcinoma (CCA), a common primary malignant tumor of bile duct epithelia, is highly prevalent in Asian countries and unresponsive to chemotherapeutic drugs. Thus, a newly recognized biological entity for early diagnosis and treatment is highly needed. Exosomes are small membrane bound vesicles found in body fluids and released by most cell types including cancer cells. The vesicles contain specific subset of proteins and nucleic acids corresponding to cell types and play essential roles in pathophysiological processes. The present study aimed to assess the protein profiles of CCA-derived exosomes and their potential roles. We have isolated exosomes from CCA cells namely KKU-M213 and KKU-100 derived from Thai patients and their roles were investigated by incubation with normal human cholangiocyte (H69) cells. Exosomes were internalized into H69 cells and had no effects on viability or proliferation of the host cells. Interestingly, the exosomes from KKU-M213 cells only induced migration and invasion of H69 cells. Proteomic analysis of the exosomes from KKU-M213 cells disclosed multiple cancer related proteins that are not present in H69 exosomes. Consistent with the protein profile, treatment with KKU-M213 exosomes induced β-catenin and reduced E-cadherin expressions in H69 cells. Collectively, our results suggest that a direct cell-to-cell transfer of oncogenic proteins via exosomal pathway may be a novel mechanism for CCA progression and metastasis.
Collapse
Affiliation(s)
- Suman Dutta
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand; Research Center of Transport Proteins for Medical Innovation, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Onrapak Reamtong
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Wittaya Panvongsa
- Research Center of Transport Proteins for Medical Innovation, Faculty of Science, Mahidol University, Bangkok, Thailand; Toxicology Graduate Program, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Sarunya Kitdumrongthum
- Research Center of Transport Proteins for Medical Innovation, Faculty of Science, Mahidol University, Bangkok, Thailand; Toxicology Graduate Program, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Keatdamrong Janpipatkul
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand; Research Center of Transport Proteins for Medical Innovation, Faculty of Science, Mahidol University, Bangkok, Thailand; Department of Basic Medical Science, Faculty of Medicine Vajira Hospital, Navamindradhiraj University, Bangkok, Thailand
| | - Polkit Sangvanich
- Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | - Pawinee Piyachaturawat
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand; Research Center of Transport Proteins for Medical Innovation, Faculty of Science, Mahidol University, Bangkok, Thailand; Toxicology Graduate Program, Faculty of Science, Mahidol University, Bangkok, Thailand; Chakri Naruebodindra Medical Institute, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Arthit Chairoungdua
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand; Research Center of Transport Proteins for Medical Innovation, Faculty of Science, Mahidol University, Bangkok, Thailand; Toxicology Graduate Program, Faculty of Science, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
42
|
Ogihara K, Naya Y, Sato R, Onda K, Ochiai H. Analysis of L-type amino acid transporter in canine hepatocellular carcinoma. J Vet Med Sci 2015; 77:527-34. [PMID: 25649314 PMCID: PMC4478731 DOI: 10.1292/jvms.14-0392] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Analysis of L-type amino acid transport expression of hepatocellular carcinoma cells
(HCCs) of the dog was performed. The leucine transport activity of canine HCCs was 0.628 ±
0.018 nmol/mg protein/min. The inhibitor of LAT 2-aminobicyclo[2.2.1]heptane-2-carboxylic
acid (BCH) reduced 90% of the activity at 1 mM. The deduced amino acid sequences of canine
LAT2, LAT3 and LAT4 were well conserved in mammalians, exhibiting 89, 88 and 77% homology,
respectively. RT-PCR revealed distinct LAT1 expression compared with normal hepatocytes.
Western blotting analysis confirmed the potent LAT1 expression in canine HCCs but not
hepatocytes, and real-time RT-PCR analysis indicated that canine HCCs possessed 28 times
higher LAT1 expression than hepatocytes. These results indicated that the leucine
transport activity of canine HCCs was due to LAT1.
Collapse
Affiliation(s)
- Kikumi Ogihara
- Laboratory of Pathology, School of Veterinary Medicine, Azabu University, 1-17-71 Fuchinobe, Chuo-ku, Sagamihara, Kanagawa 252-5201, Japan
| | | | | | | | | |
Collapse
|