1
|
Shen Z, Yu N, Zhang Y, Jia M, Sun Y, Li Y, Zhao L. The potential roles of HIF-1α in epithelial-mesenchymal transition and ferroptosis in tumor cells. Cell Signal 2024; 122:111345. [PMID: 39134249 DOI: 10.1016/j.cellsig.2024.111345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/03/2024] [Accepted: 08/09/2024] [Indexed: 08/15/2024]
Abstract
In tumors, the rapid proliferation of cells and the imperfect blood supply system lead to hypoxia, which can regulate the adaptation of tumor cells to the hypoxic environment through hypoxia-inducible factor-1α (HIF-1α) and promote tumor development in multiple ways. Recent studies have found that epithelial-mesenchymal transition (EMT) and ferroptosis play important roles in the progression of tumor cells. The activation of HIF-1α is considered a key factor in inducing EMT in tumor cells. When HIF-1α is activated, it can regulate EMT-related genes, causing tumor cells to gradually lose their epithelial characteristics and acquire more invasive mesenchymal traits. The occurrence of EMT allows tumor cells to better adapt to changes in the surrounding tissue, enhancing their migratory and invasive capabilities, thus promoting tumor progression. At the same time, HIF-1α also plays a crucial regulatory role in ferroptosis in tumor cells. In a hypoxic environment, HIF-1α may affect processes such as iron metabolism and oxidative stress responses, inducing ferroptosis in tumor cells. This article briefly reviews the dual role of HIF-1α in EMT and ferroptosis in tumor cells, helping to gain a deeper understanding of the regulatory pathways of HIF-1α in the development of tumor cells, providing a new perspective for understanding the pathogenesis of tumors. The regulation of HIF-1α may become an important strategy for future tumor therapy.
Collapse
Affiliation(s)
- Zhongjun Shen
- Department of Blood Transfusion, Second Hospital of Jilin University, Changchun, 130041 Jilin, China
| | - Na Yu
- Department of Blood Transfusion, Second Hospital of Jilin University, Changchun, 130041 Jilin, China
| | - Yanfeng Zhang
- Department of Blood Transfusion, Second Hospital of Jilin University, Changchun, 130041 Jilin, China
| | - Mingbo Jia
- Department of Blood Transfusion, Second Hospital of Jilin University, Changchun, 130041 Jilin, China
| | - Ying Sun
- Department of Blood Transfusion, Second Hospital of Jilin University, Changchun, 130041 Jilin, China
| | - Yao Li
- Department of Blood Transfusion, Second Hospital of Jilin University, Changchun, 130041 Jilin, China
| | - Liyan Zhao
- Department of Blood Transfusion, Second Hospital of Jilin University, Changchun, 130041 Jilin, China.
| |
Collapse
|
2
|
Kerdkumthong K, Nanarong S, Roytrakul S, Pitakpornpreecha T, Tantimetta P, Runsaeng P, Obchoei S. Quantitative proteomics analysis reveals possible anticancer mechanisms of 5'-deoxy-5'-methylthioadenosine in cholangiocarcinoma cells. PLoS One 2024; 19:e0306060. [PMID: 38923999 PMCID: PMC11206958 DOI: 10.1371/journal.pone.0306060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
Cholangiocarcinoma (CCA) is an aggressive cancer originating from bile duct epithelium, particularly prevalent in Asian countries with liver fluke infections. Current chemotherapy for CCA often fails due to drug resistance, necessitating novel anticancer agents. This study investigates the potential of 5'-deoxy-5'-methylthioadenosine (MTA), a naturally occurring nucleoside, against CCA. While MTA has shown promise against various cancers, its effects on CCA remain unexplored. We evaluated MTA's anticancer activity in CCA cell lines and drug-resistant sub-lines, assessing cell viability, migration, invasion, and apoptosis. The potential anticancer mechanisms of MTA were explored through proteomic analysis using LC-MS/MS and bioinformatic analysis. The results show a dose-dependent reduction in CCA cell viability, with enhanced effects on cancer cells compared to normal cells. Moreover, MTA inhibits growth, induces apoptosis, and suppresses cell migration and invasion. Additionally, MTA enhanced the anticancer effects of gemcitabine on drug-resistant CCA cells. Proteomics revealed the down-regulation of multiple proteins by MTA, affecting various molecular functions, biological processes, and cellular components. Network analysis highlighted MTA's role in inhibiting proteins related to mitochondrial function and energy derivation, crucial for cell growth and survival. Additionally, MTA suppressed proteins involved in cell morphology and cytoskeleton organization, important for cancer cell motility and metastasis. Six candidate genes, including ZNF860, KLC1, GRAMD1C, MAMSTR, TANC1, and TTC13, were selected from the top 10 most down-regulated proteins identified in the proteomics results and were subsequently verified through RT-qPCR. Further, KLC1 protein suppression by MTA treatment was confirmed through Western blotting. Additionally, based on TCGA data, KLC1 mRNA was found to be upregulated in the tissue of CCA patients compared to that of normal adjacent tissues. In summary, MTA shows promising anticancer potential against CCA by inhibiting growth, inducing apoptosis, and suppressing migration and invasion, while enhancing gemcitabine's effects. Proteomic analysis elucidates possible molecular mechanisms underlying MTA's anticancer activity, laying the groundwork for future research and development of MTA as a treatment for advanced CCA.
Collapse
Affiliation(s)
- Kankamol Kerdkumthong
- Faculty of Science, Division of Health and Applied Sciences, Biochemistry Graduate Program, Prince of Songkla University, Hatyai, Songkhla, Thailand
| | - Sutthipong Nanarong
- Faculty of Science, Division of Health and Applied Sciences, Biochemistry Graduate Program, Prince of Songkla University, Hatyai, Songkhla, Thailand
- Faculty of Pharmaceutical Sciences, Department of Pharmacognosy and Pharmaceutical Botany, Prince of Songkla University, Hatyai, Songkhla, Thailand
| | - Sittiruk Roytrakul
- National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathumtani, Thailand
| | - Thanawat Pitakpornpreecha
- Faculty of Science, Division of Health and Applied Sciences, Biochemistry Graduate Program, Prince of Songkla University, Hatyai, Songkhla, Thailand
- Faculty of Science, Center of Excellence for Biochemistry, Prince of Songkla University, Hatyai, Songkhla, Thailand
| | - Phonprapavee Tantimetta
- Faculty of Science, Division of Health and Applied Sciences, Biochemistry Graduate Program, Prince of Songkla University, Hatyai, Songkhla, Thailand
| | - Phanthipha Runsaeng
- Faculty of Science, Division of Health and Applied Sciences, Biochemistry Graduate Program, Prince of Songkla University, Hatyai, Songkhla, Thailand
- Faculty of Science, Center of Excellence for Biochemistry, Prince of Songkla University, Hatyai, Songkhla, Thailand
| | - Sumalee Obchoei
- Faculty of Science, Division of Health and Applied Sciences, Biochemistry Graduate Program, Prince of Songkla University, Hatyai, Songkhla, Thailand
- Faculty of Science, Center of Excellence for Biochemistry, Prince of Songkla University, Hatyai, Songkhla, Thailand
| |
Collapse
|
3
|
Lyu Q, Chen RA, Chuang HL, Zou HB, Liu L, Sung LK, Liu PY, Wu HY, Chang HY, Cheng WJ, Wu WK, Wu MS, Hsu CC. Bifidobacterium alleviate metabolic disorders via converting methionine to 5'-methylthioadenosine. Gut Microbes 2024; 16:2300847. [PMID: 38439565 PMCID: PMC10936671 DOI: 10.1080/19490976.2023.2300847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 12/27/2023] [Indexed: 03/06/2024] Open
Abstract
Dietary patterns and corresponding gut microbiota profiles are associated with various health conditions. A diet rich in polyphenols, primarily plant-based, has been shown to promote the growth of probiotic bacteria in the gastrointestinal tract, subsequently reducing the risk of metabolic disorders in the host. The beneficial effects of these bacteria are largely due to the specific metabolites they produce, such as short-chain fatty acids and membrane proteins. In this study, we employed a metabolomics-guided bioactive metabolite identification platform that included bioactivity testing using in vitro and in vivo assays to discover a bioactive metabolite produced from probiotic bacteria. Through this approach, we identified 5'-methylthioadenosine (MTA) as a probiotic bacterial-derived metabolite with anti-obesity properties. Furthermore, our findings indicate that MTA administration has several regulatory impacts on liver functions, including modulating fatty acid synthesis and glucose metabolism. The present study elucidates the intricate interplay between dietary habits, gut microbiota, and their resultant metabolites.
Collapse
Affiliation(s)
- Qiang Lyu
- Department of Chemistry, National Taiwan University, Taipei, Taiwan
| | - Rou-An Chen
- Department of Chemistry, National Taiwan University, Taipei, Taiwan
- Leeuwenhoek Laboratories Co. Ltd, Taipei, Taiwan
| | - Hsiao-Li Chuang
- National Laboratory Animal Center, National Applied Research Laboratories Research Institute, Taipei, Taiwan
| | - Hsin-Bai Zou
- Department of Chemistry, National Taiwan University, Taipei, Taiwan
- Leeuwenhoek Laboratories Co. Ltd, Taipei, Taiwan
| | - Lihong Liu
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Li-Kang Sung
- Department of Chemistry, National Taiwan University, Taipei, Taiwan
| | - Po-Yu Liu
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hsin-Yi Wu
- Department of Chemistry, National Taiwan University, Taipei, Taiwan
| | - Hsin-Yuan Chang
- Department of Chemistry, National Taiwan University, Taipei, Taiwan
| | - Wan-Ju Cheng
- Department of Chemistry, National Taiwan University, Taipei, Taiwan
| | - Wei-Kai Wu
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Ming-Shiang Wu
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Cheng-Chih Hsu
- Department of Chemistry, National Taiwan University, Taipei, Taiwan
- Leeuwenhoek Laboratories Co. Ltd, Taipei, Taiwan
| |
Collapse
|
4
|
Ren Y, Mao X, Xu H, Dang Q, Weng S, Zhang Y, Chen S, Liu S, Ba Y, Zhou Z, Han X, Liu Z, Zhang G. Ferroptosis and EMT: key targets for combating cancer progression and therapy resistance. Cell Mol Life Sci 2023; 80:263. [PMID: 37598126 PMCID: PMC10439860 DOI: 10.1007/s00018-023-04907-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 07/31/2023] [Accepted: 08/01/2023] [Indexed: 08/21/2023]
Abstract
Iron-dependent lipid peroxidation causes ferroptosis, a form of regulated cell death. Crucial steps in the formation of ferroptosis include the accumulation of ferrous ions (Fe2+) and lipid peroxidation, of which are controlled by glutathione peroxidase 4 (GPX4). Its crucial role in stopping the spread of cancer has been shown by numerous studies undertaken in the last ten years. Epithelial-mesenchymal transition (EMT) is the process by which epithelial cells acquire mesenchymal characteristics. EMT is connected to carcinogenesis, invasiveness, metastasis, and therapeutic resistance in cancer. It is controlled by a range of internal and external signals and changes the phenotype from epithelial to mesenchymal like. Studies have shown that mesenchymal cancer cells tend to be more ferroptotic than their epithelial counterparts. Drug-resistant cancer cells are more easily killed by inducers of ferroptosis when they undergo EMT. Therefore, understanding the interaction between ferroptosis and EMT will help identify novel cancer treatment targets. In-depth discussion is given to the regulation of ferroptosis, the potential application of EMT in the treatment of cancer, and the relationships between ferroptosis, EMT, and signaling pathways associated with tumors. Invasion, metastasis, and inflammation in cancer all include ferroptosis and EMT. The goal of this review is to provide suggestions for future research and practical guidance for applying ferroptosis and EMT in clinical practice.
Collapse
Affiliation(s)
- Yuqing Ren
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Xiangrong Mao
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Hui Xu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Qin Dang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Siyuan Weng
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Yuyuan Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Shuang Chen
- Center of Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shutong Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Yuhao Ba
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Zhaokai Zhou
- Department of Pediatric Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| | - Guojun Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
5
|
Cao K, Lyu Y, Chen J, He C, Lyu X, Zhang Y, Chen L, Jiang Y, Xiang J, Liu B, Wu C. Prognostic Implication of Plasma Metabolites in Gastric Cancer. Int J Mol Sci 2023; 24:12774. [PMID: 37628957 PMCID: PMC10454100 DOI: 10.3390/ijms241612774] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 08/02/2023] [Accepted: 08/07/2023] [Indexed: 08/27/2023] Open
Abstract
Gastric cancer (GC) typically carries a poor prognosis as it is often diagnosed at a late stage. Altered metabolism has been found to impact cancer outcomes and affect patients' quality of life, and the role of metabolites in gastric cancer prognosis has not been sufficiently understood. We aimed to establish a prognostic prediction model for GC patients based on a metabolism-associated signature and identify the unique role of metabolites in the prognosis of GC. Thus, we conducted untargeted metabolomics to detect the plasma metabolites of 218 patients with gastric adenocarcinoma and explored the metabolites related to the survival of patients with gastric cancer. Firstly, we divided patients into two groups based on the cutoff value of the abundance of each of the 60 metabolites and compared the differences using Kaplan-Meier (K-M) survival analysis. As a result, 23 metabolites associated with gastric cancer survival were identified. To establish a risk score model, we performed LASSO regression and Cox regression analysis on the 60 metabolites and identified 8 metabolites as an independent prognostic factor. Furthermore, a nomogram incorporating clinical parameters and the metabolic signature was constructed to help individualize outcome predictions. The results of the ROC curve and nomogram plot showed good predictive performance of metabolic risk features. Finally, we performed pathway analysis on the 24 metabolites identified in the two parts, and the results indicated that purine metabolism and arachidonic acid metabolism play important roles in gastric cancer prognosis. Our study highlights the important role of metabolites in the progression of gastric cancer and newly identified metabolites could be potential biomarkers or therapeutic targets for gastric cancer patients.
Collapse
Affiliation(s)
- Kang Cao
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou 350122, China; (K.C.)
- The Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Yanping Lyu
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou 350122, China; (K.C.)
- The Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Jingwen Chen
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou 350122, China; (K.C.)
- The Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Chenzhou He
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou 350122, China; (K.C.)
- The Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Xuejie Lyu
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou 350122, China; (K.C.)
- The Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Yuling Zhang
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou 350122, China; (K.C.)
- The Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Liangping Chen
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou 350122, China; (K.C.)
- The Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Yu Jiang
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou 350122, China; (K.C.)
- The Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Jianjun Xiang
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou 350122, China; (K.C.)
- The Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Baoying Liu
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou 350122, China; (K.C.)
- The Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Chuancheng Wu
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou 350122, China; (K.C.)
- The Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| |
Collapse
|
6
|
Heo S, Cho S, Dinh PTN, Park J, Jin DH, Cha J, Kim YK, Koh YJ, Lee SH, Lee JH. A genome-wide association study for eumelanin pigmentation in chicken plumage using a computer vision approach. Anim Genet 2023; 54:355-362. [PMID: 36855963 DOI: 10.1111/age.13303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 11/28/2022] [Accepted: 01/26/2023] [Indexed: 03/02/2023]
Abstract
Chicken plumage colouration is an important trait related to productivity in poultry industry. Therefore, the genetic basis for pigmentation in chicken plumage is an area of great interest. However, the colour trait is generally regarded as a qualitative trait and representing colour variations is difficult. In this study, we developed a method to quantify and classify colour using an F2 population crossed from two pure lines: White Leghorn and the Korean indigenous breed Yeonsan Ogye. Using red, green, and blue values in the cropped body region, we identified significant genomic regions on chromosomes 33:3 160 480-7 447 197 and Z:78 748 287-79 173 793. Furthermore, we identified two potential candidate genes (PMEL and MTAP) that might have significant effects on melanin-based plumage pigmentation. Our study presents a new phenotyping method using a computer vision approach and provides new insights into the genetic basis of melanin-based feather colouration in chickens.
Collapse
Affiliation(s)
- Seonyeong Heo
- Department of Bio-AI Convergence, Chungnam National University, Daejeon, South Korea
| | - Sunghyun Cho
- Research and Development Center, Insilicogen Inc., Yongin, South Korea
| | | | - Jongho Park
- Department of Bio-AI Convergence, Chungnam National University, Daejeon, South Korea
| | - Dae-Hyeok Jin
- Animal Genetic Resources Research Center, National Institute of Animal Science, Rural Development Administration, Hamyang, South Korea
| | - Jihye Cha
- Animal Genome & Bioinformatics, National Institute of Animal Science, Rural Development Administration, Wanju, South Korea
| | - Young-Kuk Kim
- Department of Bio-AI Convergence, Chungnam National University, Daejeon, South Korea.,Department of Computer Science & Engineering, Chungnam National University, Daejeon, South Korea
| | - Yeong Jun Koh
- Department of Bio-AI Convergence, Chungnam National University, Daejeon, South Korea.,Department of Computer Science & Engineering, Chungnam National University, Daejeon, South Korea
| | - Seung Hwan Lee
- Department of Bio-AI Convergence, Chungnam National University, Daejeon, South Korea.,Division of Animal and Dairy Science, Chungnam National University, Daejeon, South Korea
| | - Jun Heon Lee
- Department of Bio-AI Convergence, Chungnam National University, Daejeon, South Korea.,Division of Animal and Dairy Science, Chungnam National University, Daejeon, South Korea
| |
Collapse
|
7
|
McGrail K, Granado-Martínez P, Esteve-Puig R, García-Ortega S, Ding Y, Sánchez-Redondo S, Ferrer B, Hernandez-Losa J, Canals F, Manzano A, Navarro-Sabaté A, Bartrons R, Yanes O, Pérez-Alea M, Muñoz-Couselo E, Garcia-Patos V, Recio JA. BRAF activation by metabolic stress promotes glycolysis sensitizing NRAS Q61-mutated melanomas to targeted therapy. Nat Commun 2022; 13:7113. [PMID: 36402789 PMCID: PMC9675737 DOI: 10.1038/s41467-022-34907-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 11/08/2022] [Indexed: 11/21/2022] Open
Abstract
NRAS-mutated melanoma lacks a specific line of treatment. Metabolic reprogramming is considered a novel target to control cancer; however, NRAS-oncogene contribution to this cancer hallmark is mostly unknown. Here, we show that NRASQ61-mutated melanomas specific metabolic settings mediate cell sensitivity to sorafenib upon metabolic stress. Mechanistically, these cells are dependent on glucose metabolism, in which glucose deprivation promotes a switch from CRAF to BRAF signaling. This scenario contributes to cell survival and sustains glucose metabolism through BRAF-mediated phosphorylation of 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase-2/3 (PFKFB2/PFKFB3). In turn, this favors the allosteric activation of phosphofructokinase-1 (PFK1), generating a feedback loop that couples glycolytic flux and the RAS signaling pathway. An in vivo treatment of NRASQ61 mutant melanomas, including patient-derived xenografts, with 2-deoxy-D-glucose (2-DG) and sorafenib effectively inhibits tumor growth. Thus, we provide evidence for NRAS-oncogene contributions to metabolic rewiring and a proof-of-principle for the treatment of NRASQ61-mutated melanoma combining metabolic stress (glycolysis inhibitors) and previously approved drugs, such as sorafenib.
Collapse
Affiliation(s)
- Kimberley McGrail
- grid.430994.30000 0004 1763 0287Biomedical Research in Melanoma-Animal Models and Cancer Laboratory, Vall d’Hebron Research Institute (VHIR), Vall d’Hebron Hospital Barcelona-UAB, Barcelona, 08035 Spain
| | - Paula Granado-Martínez
- grid.430994.30000 0004 1763 0287Biomedical Research in Melanoma-Animal Models and Cancer Laboratory, Vall d’Hebron Research Institute (VHIR), Vall d’Hebron Hospital Barcelona-UAB, Barcelona, 08035 Spain
| | - Rosaura Esteve-Puig
- grid.430994.30000 0004 1763 0287Biomedical Research in Melanoma-Animal Models and Cancer Laboratory, Vall d’Hebron Research Institute (VHIR), Vall d’Hebron Hospital Barcelona-UAB, Barcelona, 08035 Spain ,Present Address: MAJ3 Capital S.L, Barcelona, 08018 Spain
| | - Sara García-Ortega
- grid.430994.30000 0004 1763 0287Biomedical Research in Melanoma-Animal Models and Cancer Laboratory, Vall d’Hebron Research Institute (VHIR), Vall d’Hebron Hospital Barcelona-UAB, Barcelona, 08035 Spain
| | - Yuxin Ding
- grid.430994.30000 0004 1763 0287Biomedical Research in Melanoma-Animal Models and Cancer Laboratory, Vall d’Hebron Research Institute (VHIR), Vall d’Hebron Hospital Barcelona-UAB, Barcelona, 08035 Spain
| | - Sara Sánchez-Redondo
- grid.430994.30000 0004 1763 0287Biomedical Research in Melanoma-Animal Models and Cancer Laboratory, Vall d’Hebron Research Institute (VHIR), Vall d’Hebron Hospital Barcelona-UAB, Barcelona, 08035 Spain ,grid.7719.80000 0000 8700 1153Present Address: Microenvironment & Metastasis Group, Molecular Oncology Program, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Berta Ferrer
- grid.430994.30000 0004 1763 0287Biomedical Research in Melanoma-Animal Models and Cancer Laboratory, Vall d’Hebron Research Institute (VHIR), Vall d’Hebron Hospital Barcelona-UAB, Barcelona, 08035 Spain ,grid.411083.f0000 0001 0675 8654Anatomy Pathology Department, Vall d’Hebron Hospital Barcelona-UAB, Barcelona, 08035 Spain
| | - Javier Hernandez-Losa
- grid.411083.f0000 0001 0675 8654Anatomy Pathology Department, Vall d’Hebron Hospital Barcelona-UAB, Barcelona, 08035 Spain
| | - Francesc Canals
- grid.411083.f0000 0001 0675 8654Proteomics Laboratory, Vall d’Hebron Institute of Oncology (VHIO), Barcelona, 08035 Spain
| | - Anna Manzano
- grid.418284.30000 0004 0427 2257Department of Physiological Sciences, University of Barcelona, Bellvitge Biomedical Research Institute, Barcelona, Spain
| | - Aura Navarro-Sabaté
- grid.418284.30000 0004 0427 2257Department of Physiological Sciences, University of Barcelona, Bellvitge Biomedical Research Institute, Barcelona, Spain
| | - Ramón Bartrons
- grid.418284.30000 0004 0427 2257Department of Physiological Sciences, University of Barcelona, Bellvitge Biomedical Research Institute, Barcelona, Spain
| | - Oscar Yanes
- grid.410367.70000 0001 2284 9230Universitat Rovira i Virgili, Department of Electronic Engineering, IISPV, Tarragona, Spain ,grid.413448.e0000 0000 9314 1427CIBER on Diabetes and Associated Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Mileidys Pérez-Alea
- grid.430994.30000 0004 1763 0287Biomedical Research in Melanoma-Animal Models and Cancer Laboratory, Vall d’Hebron Research Institute (VHIR), Vall d’Hebron Hospital Barcelona-UAB, Barcelona, 08035 Spain ,Present Address: Advance Biodesign, 69800 Saint-Priest, France
| | - Eva Muñoz-Couselo
- grid.430994.30000 0004 1763 0287Biomedical Research in Melanoma-Animal Models and Cancer Laboratory, Vall d’Hebron Research Institute (VHIR), Vall d’Hebron Hospital Barcelona-UAB, Barcelona, 08035 Spain ,grid.411083.f0000 0001 0675 8654Clinical Oncology Program, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Hospital Barcelona-UAB, Barcelona, 08035 Spain
| | - Vicenç Garcia-Patos
- grid.430994.30000 0004 1763 0287Biomedical Research in Melanoma-Animal Models and Cancer Laboratory, Vall d’Hebron Research Institute (VHIR), Vall d’Hebron Hospital Barcelona-UAB, Barcelona, 08035 Spain ,grid.411083.f0000 0001 0675 8654Dermatology Department, Vall d’Hebron Hospital Barcelona-UAB, Barcelona, 08035 Spain
| | - Juan A. Recio
- grid.430994.30000 0004 1763 0287Biomedical Research in Melanoma-Animal Models and Cancer Laboratory, Vall d’Hebron Research Institute (VHIR), Vall d’Hebron Hospital Barcelona-UAB, Barcelona, 08035 Spain
| |
Collapse
|
8
|
Yang R, Dong S, Zhang J, Zhu S, Miao G, Zhang B. Downregulation of PRMT5 by AMI-1 enhances therapeutic efficacy of compound kushen injection in lung carcinoma in vitro and in vivo. Mol Cell Biochem 2022; 478:1031-1044. [PMID: 36214894 DOI: 10.1007/s11010-022-04577-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 09/26/2022] [Indexed: 11/28/2022]
Abstract
Protein arginine methyltransferase 5 (PRMT5) is overexpressed in lung carcinoma, which promotes tumor cell proliferation, survival, migration and invasion. Compound Kushen injection (CKI) is a mixture of natural compounds extracted from Kushen and Baituling, which are mainly used to stop in cancer pain and bleeding. Here we found that cell viability and colony formation were inhibited after the incubation of AMI-1. Meanwhile, AMI-1 suppressed cell migration, enhanced apoptosis, induced cell cycle arrest, inhibited PRMT5 expression and histone H3R8 and H4R3 symmetric di-methylation (H3R8me2s and H4R3me2s) accumulation, down-regulated the expression of eukaryotic translation initiation factor 4E (eIF4E) in lung carcinoma cells. Moreover, AMI-1 suppressed tumor growth, decreased H3R8me2s and H4R3me2s accumulation, down-regulated eIF4E expression and increased p53 expression in lung carcinoma xenografts of BALB/c nude mice. Of note, combined and CKI markedly enhanced the anticancer efficacy CKI in lung carcinoma. The above findings demonstrated that AMI-1 has established antineoplastic activity and this role may be associated with affecting the function of eIF4E via inhibiting PRMT5 activity or protein levels in lung carcinoma. This study highlights evidence of novel selective anticancer activity of AMI-1 in combination with CKI in lung carcinoma.
Collapse
Affiliation(s)
- Ruiying Yang
- Department of Pharmacology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Shuhong Dong
- Department of Pharmacology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Jinghui Zhang
- College of Pharmacy, Gansu University of Traditional Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Shihao Zhu
- Department of Pharmacology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Guoliang Miao
- Department of Pharmacology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Baolai Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China.
| |
Collapse
|
9
|
Jacobs B, Schlögl S, Strobl CD, Völkl S, Stoll A, Mougiakakos D, Malmberg KJ, Mackensen A, Aigner M. The Oncometabolite 5'-Deoxy-5'-Methylthioadenosine Blocks Multiple Signaling Pathways of NK Cell Activation. Front Immunol 2020; 11:2128. [PMID: 33123121 PMCID: PMC7573074 DOI: 10.3389/fimmu.2020.02128] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 08/05/2020] [Indexed: 11/13/2022] Open
Abstract
Tumor cells develop various mechanisms to escape immune surveillance. In this context, oncometabolites secreted by tumor cells due to deregulated metabolic pathways, have been in the spotlight of researchers during the last years. 5'-Deoxy-5'-methylthioadenosine (MTA) phosphorylase (MTAP) deficiency in tumors results in the accumulation of MTA within the tumor microenvironment and thereby negatively influencing immune functions of various immune cells, including T and NK cells. The influence of MTA on T cell activation has been recently described in more detail, while its impact on NK cells is still largely unknown. Therefore, we aimed to illuminate the molecular mechanism of MTA-induced NK cell dysfunction. NK cell cytotoxicity against target cells was reduced in the presence of MTA in a dose-dependent manner, while NK cell viability remained unaffected. Furthermore, we revealed that MTA blocks NK cell degranulation and cytokine production upon target cell engagement as well as upon antibody stimulation. Interestingly, the immune-suppressive effect of MTA was less pronounced in healthy donors harboring an expansion of NKG2C+ NK cells. Finally, we demonstrated that MTA interferes with various signaling pathways downstream of the CD16 receptor upon NK cell activation, including the PI3K/AKT/S6, MAPK/ERK, and NF-κB pathways. In summary, we revealed that MTA blocks NK cell functions like cytotoxicity and cytokine production by interfering with the signaling cascade of activating NK cell receptors. Specific targeting of MTA metabolism in MTAP-deficient tumors therefore could offer a promising new strategy to reverse immune dysfunction of NK cells within the tumor microenvironment.
Collapse
Affiliation(s)
- Benedikt Jacobs
- Department of Internal Medicine 5, Hematology and Oncology, Friedrich Alexander University Erlangen-Nuremberg (FAU), University Hospital Erlangen, Erlangen, Germany
| | - Sebastian Schlögl
- Department of Anesthesiology, Intensive Care and Pain Therapy, General Hospital Fürth, Fürth, Germany
| | - Carolin Dorothea Strobl
- Department of Internal Medicine 5, Hematology and Oncology, Friedrich Alexander University Erlangen-Nuremberg (FAU), University Hospital Erlangen, Erlangen, Germany
| | - Simon Völkl
- Department of Internal Medicine 5, Hematology and Oncology, Friedrich Alexander University Erlangen-Nuremberg (FAU), University Hospital Erlangen, Erlangen, Germany
| | - Andrej Stoll
- Department of Internal Medicine 5, Hematology and Oncology, Friedrich Alexander University Erlangen-Nuremberg (FAU), University Hospital Erlangen, Erlangen, Germany
| | - Dimitrios Mougiakakos
- Department of Internal Medicine 5, Hematology and Oncology, Friedrich Alexander University Erlangen-Nuremberg (FAU), University Hospital Erlangen, Erlangen, Germany
| | - Karl-Johan Malmberg
- Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Sweden.,K.G. Jebsen Center for Cancer Immunotherapy, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Andreas Mackensen
- Department of Internal Medicine 5, Hematology and Oncology, Friedrich Alexander University Erlangen-Nuremberg (FAU), University Hospital Erlangen, Erlangen, Germany
| | - Michael Aigner
- Department of Internal Medicine 5, Hematology and Oncology, Friedrich Alexander University Erlangen-Nuremberg (FAU), University Hospital Erlangen, Erlangen, Germany
| |
Collapse
|
10
|
Zhang T, Bauer C, Newman AC, Uribe AH, Athineos D, Blyth K, Maddocks ODK. Polyamine pathway activity promotes cysteine essentiality in cancer cells. Nat Metab 2020; 2:1062-1076. [PMID: 32747794 PMCID: PMC7614128 DOI: 10.1038/s42255-020-0253-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 06/26/2020] [Indexed: 02/05/2023]
Abstract
Cancer cells have high demands for non-essential amino acids (NEAAs), which are precursors for anabolic and antioxidant pathways that support cell survival and proliferation. It is well-established that cancer cells consume the NEAA cysteine, and that cysteine deprivation can induce cell death; however, the specific factors governing acute sensitivity to cysteine starvation are poorly characterized. Here, we show that that neither expression of enzymes for cysteine synthesis nor availability of the primary precursor methionine correlated with acute sensitivity to cysteine starvation. We observed a strong correlation between efflux of the methionine-derived metabolite methylthioadenosine (MTA) and sensitivity to cysteine starvation. MTA efflux results from genetic deletion of methylthioadenosine phosphorylase (MTAP), which is frequently deleted in cancers. We show that MTAP loss upregulates polyamine metabolism which, concurrently with cysteine withdrawal, promotes elevated reactive oxygen species and prevents cell survival. Our results reveal an unexplored metabolic weakness at the intersection of polyamine and cysteine metabolism.
Collapse
Affiliation(s)
- Tong Zhang
- Institute of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, UK
- Novartis Institutes for BioMedical Research, Shanghai, China
| | - Christin Bauer
- Institute of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, UK
- AstraZeneca R&D, Cambridge, UK
| | - Alice C Newman
- Institute of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, UK
| | - Alejandro Huerta Uribe
- Institute of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, UK
| | | | - Karen Blyth
- Institute of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, UK
- Cancer Research UK Beatson Institute, Glasgow, UK
| | - Oliver D K Maddocks
- Institute of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, UK.
| |
Collapse
|
11
|
Granado-Martínez P, Garcia-Ortega S, González-Sánchez E, McGrail K, Selgas R, Grueso J, Gil R, Naldaiz-Gastesi N, Rhodes AC, Hernandez-Losa J, Ferrer B, Canals F, Villanueva J, Méndez O, Espinosa-Gil S, Lizcano JM, Muñoz-Couselo E, García-Patos V, Recio JA. STK11 (LKB1) missense somatic mutant isoforms promote tumor growth, motility and inflammation. Commun Biol 2020; 3:366. [PMID: 32647375 PMCID: PMC7347935 DOI: 10.1038/s42003-020-1092-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 06/19/2020] [Indexed: 02/07/2023] Open
Abstract
Elucidating the contribution of somatic mutations to cancer is essential for personalized medicine. STK11 (LKB1) appears to be inactivated in human cancer. However, somatic missense mutations also occur, and the role/s of these alterations to this disease remain unknown. Here, we investigated the contribution of four missense LKB1 somatic mutations in tumor biology. Three out of the four mutants lost their tumor suppressor capabilities and showed deficient kinase activity. The remaining mutant retained the enzymatic activity of wild type LKB1, but induced increased cell motility. Mechanistically, LKB1 mutants resulted in differential gene expression of genes encoding vesicle trafficking regulating molecules, adhesion molecules and cytokines. The differentially regulated genes correlated with protein networks identified through comparative secretome analysis. Notably, three mutant isoforms promoted tumor growth, and one induced inflammation-like features together with dysregulated levels of cytokines. These findings uncover oncogenic roles of LKB1 somatic mutations, and will aid in further understanding their contributions to cancer development and progression. Paula Granado-Martínez, Sara Ortega, Elena González-Sánchez et al. report a functional analysis of four cancer-associated mutant isoforms of the gene STK11 using cell-based and animal models. They find the mutant isoforms no longer show tumor suppressor activity, promote tumor growth, and affect the regulation of cytokines and genes involved in vesicle trafficking.
Collapse
Affiliation(s)
- Paula Granado-Martínez
- Biomedical Research in Melanoma-Animal Models and Cancer Laboratory- Vall d'Hebron Research Institute VHIR-Vall d'Hebron Hospital Barcelona-UAB, Barcelona, 08035, Spain
| | - Sara Garcia-Ortega
- Biomedical Research in Melanoma-Animal Models and Cancer Laboratory- Vall d'Hebron Research Institute VHIR-Vall d'Hebron Hospital Barcelona-UAB, Barcelona, 08035, Spain
| | - Elena González-Sánchez
- Biomedical Research in Melanoma-Animal Models and Cancer Laboratory- Vall d'Hebron Research Institute VHIR-Vall d'Hebron Hospital Barcelona-UAB, Barcelona, 08035, Spain
| | - Kimberley McGrail
- Biomedical Research in Melanoma-Animal Models and Cancer Laboratory- Vall d'Hebron Research Institute VHIR-Vall d'Hebron Hospital Barcelona-UAB, Barcelona, 08035, Spain
| | - Rafael Selgas
- Biomedical Research in Melanoma-Animal Models and Cancer Laboratory- Vall d'Hebron Research Institute VHIR-Vall d'Hebron Hospital Barcelona-UAB, Barcelona, 08035, Spain
| | - Judit Grueso
- Biomedical Research in Melanoma-Animal Models and Cancer Laboratory- Vall d'Hebron Research Institute VHIR-Vall d'Hebron Hospital Barcelona-UAB, Barcelona, 08035, Spain.,Experimental Therapeutics Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, 08035, Spain
| | - Rosa Gil
- Biomedical Research in Melanoma-Animal Models and Cancer Laboratory- Vall d'Hebron Research Institute VHIR-Vall d'Hebron Hospital Barcelona-UAB, Barcelona, 08035, Spain
| | - Neia Naldaiz-Gastesi
- Biomedical Research in Melanoma-Animal Models and Cancer Laboratory- Vall d'Hebron Research Institute VHIR-Vall d'Hebron Hospital Barcelona-UAB, Barcelona, 08035, Spain.,Biodonostia, Neurosciences Area, Group of Neuromuscular Diseases, San Sebastian, 20014, Spain
| | - Ana C Rhodes
- Biomedical Research in Melanoma-Animal Models and Cancer Laboratory- Vall d'Hebron Research Institute VHIR-Vall d'Hebron Hospital Barcelona-UAB, Barcelona, 08035, Spain.,Barcelona Clinic Liver Cancer (BCLC) Group, Liver Unit, Hospital Clínic of Barcelona, University of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, 08036, Spain
| | - Javier Hernandez-Losa
- Anatomy Pathology Department, Vall d'Hebron Hospital Barcelona-UAB, Barcelona, 08035, Spain
| | - Berta Ferrer
- Biomedical Research in Melanoma-Animal Models and Cancer Laboratory- Vall d'Hebron Research Institute VHIR-Vall d'Hebron Hospital Barcelona-UAB, Barcelona, 08035, Spain.,Anatomy Pathology Department, Vall d'Hebron Hospital Barcelona-UAB, Barcelona, 08035, Spain
| | - Francesc Canals
- Proteomics Laboratory, Vall d'Hebron Institute of Oncology (VHIO, Barcelona, 08035, Spain
| | - Josep Villanueva
- Preclinical Research Program, Vall d'Hebron Institute of Oncology (VHIO, Barcelona, 08035, Spain
| | - Olga Méndez
- Preclinical Research Program, Vall d'Hebron Institute of Oncology (VHIO, Barcelona, 08035, Spain
| | - Sergio Espinosa-Gil
- Protein Kinases and Signal Transduction Laboratory, Neuroscience Institute and Molecular Biology and Biochemistry Department, UAB, Bellaterra, Barcelona, 08193, Spain
| | - José M Lizcano
- Protein Kinases and Signal Transduction Laboratory, Neuroscience Institute and Molecular Biology and Biochemistry Department, UAB, Bellaterra, Barcelona, 08193, Spain
| | - Eva Muñoz-Couselo
- Biomedical Research in Melanoma-Animal Models and Cancer Laboratory- Vall d'Hebron Research Institute VHIR-Vall d'Hebron Hospital Barcelona-UAB, Barcelona, 08035, Spain.,Clinical Oncology Program, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Hospital, Barcelona-UAB, Barcelona, 08035, Spain
| | - Vicenç García-Patos
- Biomedical Research in Melanoma-Animal Models and Cancer Laboratory- Vall d'Hebron Research Institute VHIR-Vall d'Hebron Hospital Barcelona-UAB, Barcelona, 08035, Spain.,Dermatology Department, Vall d'Hebron Hospital Barcelona-UAB, Barcelona, 08035, Spain
| | - Juan A Recio
- Biomedical Research in Melanoma-Animal Models and Cancer Laboratory- Vall d'Hebron Research Institute VHIR-Vall d'Hebron Hospital Barcelona-UAB, Barcelona, 08035, Spain.
| |
Collapse
|
12
|
Li Y, Wang Y, Wu P. 5'-Methylthioadenosine and Cancer: old molecules, new understanding. J Cancer 2019; 10:927-936. [PMID: 30854099 PMCID: PMC6400808 DOI: 10.7150/jca.27160] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 12/17/2018] [Indexed: 12/19/2022] Open
Abstract
While the metabolic changes in cancer tissues were first observed by Warburg Otto almost a century ago, altered metabolism has recently returned as a focus of cancer research. 5'-Methylthioadenosine (MTA) is a naturally occurring sulfur-containing nucleoside found in numerous species. While MTA was first isolated several decades ago, a lack of sensitive and specific analytical methodologies designed for its direct quantification has hampered the study of its physiological and pathophysiological features. Many studies indicate that MTA suppresses tumors by inhibiting tumor cell proliferation, invasion, and the induction of apoptosis while controlling the inflammatory micro-environments of tumor tissue. In this review, we assessed the effects of MTA and of related materials on the growth and functions of normal and malignant cells.
Collapse
Affiliation(s)
- Yaofeng Li
- Department of Pathophysiology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yubo Wang
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Ping Wu
- Department of Pathophysiology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
13
|
Zhang B, Chen X, Ge S, Peng C, Zhang S, Chen X, Liu T, Zhang W. Arginine methyltransferase inhibitor-1 inhibits sarcoma viability in vitro and in vivo. Oncol Lett 2018; 16:2161-2166. [PMID: 30008914 PMCID: PMC6036477 DOI: 10.3892/ol.2018.8929] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 04/16/2018] [Indexed: 01/02/2023] Open
Abstract
Protein arginine methyltransferases (PRMTs) are a class of epigenetic modified enzymes that are overexpressed in a various types of cancer and serve pivotal functions in malignant transformation. Arginine methyltransferase inhibitor-1 (AMI-1) is a symmetrical sulfonated urea that inhibits the activity of type I PRMT in vitro. However, previous studies demonstrated that AMI-1 may also inhibit the activity of type II PRMT5 in vitro. To the best of our knowledge, the present study provides the first evidence that AMI-1 may significantly inhibit the viability of mouse sarcoma 180 (S180) and human osteosarcoma U2OS cells. Additionally, the results demonstrated that AMI-1 downregulated the activities of PRMT5, the symmetric dimethylation of histone 4 and histone 3 (a PRMT5-specific epigenetic mark) in a mouse xenograft model of S180 and induced apoptosis in S180 cells. Taken together, the results suggest that AMI-1 may exhibit antitumor effects against sarcoma cells by targeting PRMT5.
Collapse
Affiliation(s)
- Baolai Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu 730000, P.R. China.,Key Lab of Preclinical Study for New Drugs of Gansu Province, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Xue Chen
- Department of Pharmacology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu 730000, P.R. China.,Key Lab of Preclinical Study for New Drugs of Gansu Province, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Suyin Ge
- Department of Pharmacology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu 730000, P.R. China.,Key Lab of Preclinical Study for New Drugs of Gansu Province, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Caili Peng
- Day-Care Unit, Gansu Provincial People's Hospital, Lanzhou, Gansu 730000, P.R. China
| | - Su Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu 730000, P.R. China.,Key Lab of Preclinical Study for New Drugs of Gansu Province, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Xu Chen
- Department of Pharmacology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu 730000, P.R. China.,Key Lab of Preclinical Study for New Drugs of Gansu Province, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Tao Liu
- Department of Pharmacology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu 730000, P.R. China.,Key Lab of Preclinical Study for New Drugs of Gansu Province, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Wenkai Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu 730000, P.R. China.,Key Lab of Preclinical Study for New Drugs of Gansu Province, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| |
Collapse
|
14
|
Pérez-Alea M, McGrail K, Sánchez-Redondo S, Ferrer B, Fournet G, Cortés J, Muñoz E, Hernandez-Losa J, Tenbaum S, Martin G, Costello R, Ceylan I, Garcia-Patos V, Recio JA. ALDH1A3 is epigenetically regulated during melanocyte transformation and is a target for melanoma treatment. Oncogene 2017; 36:5695-5708. [PMID: 28581514 DOI: 10.1038/onc.2017.160] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 04/18/2017] [Accepted: 04/18/2017] [Indexed: 12/12/2022]
Abstract
Despite the promising targeted and immune-based interventions in melanoma treatment, long-lasting responses are limited. Melanoma cells present an aberrant redox state that leads to the production of toxic aldehydes that must be converted into less reactive molecules. Targeting the detoxification machinery constitutes a novel therapeutic avenue for melanoma. Here, using 56 cell lines representing nine different tumor types, we demonstrate that melanoma cells exhibit a strong correlation between reactive oxygen species amounts and aldehyde dehydrogenase 1 (ALDH1) activity. We found that ALDH1A3 is upregulated by epigenetic mechanisms in melanoma cells compared with normal melanocytes. Furthermore, it is highly expressed in a large percentage of human nevi and melanomas during melanocyte transformation, which is consistent with the data from the TCGA, CCLE and protein atlas databases. Melanoma treatment with the novel irreversible isoform-specific ALDH1 inhibitor [4-dimethylamino-4-methyl-pent-2-ynthioic acid-S methylester] di-methyl-ampal-thio-ester (DIMATE) or depletion of ALDH1A1 and/or ALDH1A3, promoted the accumulation of apoptogenic aldehydes leading to apoptosis and tumor growth inhibition in immunocompetent, immunosuppressed and patient-derived xenograft mouse models. Interestingly, DIMATE also targeted the slow cycling label-retaining tumor cell population containing the tumorigenic and chemoresistant cells. Our findings suggest that aldehyde detoxification is relevant metabolic mechanism in melanoma cells, which can be used as a novel approach for melanoma treatment.
Collapse
Affiliation(s)
- M Pérez-Alea
- Biomedical Research in Melanoma-Animal Models and Cancer Laboratory-Oncology Program, Vall d'Hebron Research institute VHIR-Vall d'Hebron Hospital, Barcelona-UAB, Spain
| | - K McGrail
- Biomedical Research in Melanoma-Animal Models and Cancer Laboratory-Oncology Program, Vall d'Hebron Research institute VHIR-Vall d'Hebron Hospital, Barcelona-UAB, Spain
| | - S Sánchez-Redondo
- Biomedical Research in Melanoma-Animal Models and Cancer Laboratory-Oncology Program, Vall d'Hebron Research institute VHIR-Vall d'Hebron Hospital, Barcelona-UAB, Spain
| | - B Ferrer
- Biomedical Research in Melanoma-Animal Models and Cancer Laboratory-Oncology Program, Vall d'Hebron Research institute VHIR-Vall d'Hebron Hospital, Barcelona-UAB, Spain.,Anatomy Pathology Department, Vall d'Hebron Hospital, Barcelona-UAB, Spain
| | - G Fournet
- Institut de Chimie et Biochimie Moléculaire et Supramoléculaire, UMR-CNRS 5246, Université de Lyon, Université Claude Bernard-Lyon1, Villeurbanne, France
| | - J Cortés
- Ramon y Cajal University Hospital, Madrid, Spain.,Clinical Oncology Program, Vall d'Hebron Institute of Oncology-VHIO, Vall d'Hebron Hospital, Barcelona-UAB, Spain
| | - E Muñoz
- Clinical Oncology Program, Vall d'Hebron Institute of Oncology-VHIO, Vall d'Hebron Hospital, Barcelona-UAB, Spain
| | - J Hernandez-Losa
- Anatomy Pathology Department, Vall d'Hebron Hospital, Barcelona-UAB, Spain
| | - S Tenbaum
- Stem Cells and Cancer Laboratory, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - G Martin
- Advanced BioDesign, Parc Technologique de Lyon, Woodstock - Bâtiment Cèdre 1, Saint Priest, France
| | - R Costello
- Service d'Hématologie et Thérapie Cellulaire, Centre Hospitalier Universitaire La Conception, Marseille, France
| | - I Ceylan
- Advanced BioDesign, Parc Technologique de Lyon, Woodstock - Bâtiment Cèdre 1, Saint Priest, France
| | - V Garcia-Patos
- Biomedical Research in Melanoma-Animal Models and Cancer Laboratory-Oncology Program, Vall d'Hebron Research institute VHIR-Vall d'Hebron Hospital, Barcelona-UAB, Spain.,Dermatology Department, Vall d'Hebron Hospital, Barcelona-UAB, Spain
| | - J A Recio
- Biomedical Research in Melanoma-Animal Models and Cancer Laboratory-Oncology Program, Vall d'Hebron Research institute VHIR-Vall d'Hebron Hospital, Barcelona-UAB, Spain
| |
Collapse
|
15
|
Rodzik Ł, Lewandowska-Łańcucka J, Szuwarzyński M, Szczubiałka K, Nowakowska M. Novel fluorescent CdTe quantum dot-thymine conjugate-synthesis, properties and possible application. NANOTECHNOLOGY 2017; 28:045701. [PMID: 27977416 DOI: 10.1088/1361-6528/28/4/045701] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Novel, highly fluorescent cadmium telluride quantum dots conjugated with thymine and stabilized with thioglycolic acid were obtained and characterized. Successful formation of the conjugate was confirmed by elemental analysis, and UV-vis, fluorescence and Fourier transform infrared spectroscopies. Crystal structure and composition of the conjugates were characterized with xray diffraction and x-ray photoelectron spectroscopy. The size of the conjugates was 4-6 nm as demonstrated using atomic force microscopy and high resolution transmission electron microscopy imaging. The plasmon resonance fluorescence band at 540 nm on excitation at 351 nm was observed for these nanoparticles. The intensity of this band increased with the increase in the amount of conjugated thymine with no shift in its position. Based on the fluorescence measurements it was found that the CdTe-thymine conjugate interacted efficiently and selectively not only with adenine, a nucleobase complementary to thymine, but also with adenine-containing modified nucleosides, i.e., 5'-deoxy-5'-(methylthio)adenosine and 2'-O-methyladenosine, the urinary tumor markers which allow monitoring of the disease progression. To the best of our knowledge, as yet, there have been no studies presented in literature on that type of the interaction with CdTe-thymine conjugates. Therefore, the system presented can be considered as a working component of a selective adenine/adenosine biosensor with potential application in cancer diagnosis.
Collapse
Affiliation(s)
- Łucja Rodzik
- Faculty of Chemistry, Jagiellonian University, Ingardena 3, 30-060 Kraków, Poland
| | | | | | | | | |
Collapse
|
16
|
Daghir-Wojtkowiak E, Wiczling P, Waszczuk-Jankowska M, Kaliszan R, Markuszewski MJ. Multilevel pharmacokinetics-driven modeling of metabolomics data. Metabolomics 2017; 13:31. [PMID: 28255294 PMCID: PMC5306155 DOI: 10.1007/s11306-017-1164-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 01/12/2017] [Indexed: 10/28/2022]
Abstract
INTRODUCTION Multilevel modeling is a quantitative statistical method to investigate variability and relationships between variables of interest, taking into account population structure and dependencies. It can be used for prediction, data reduction and causal inference from experiments and observational studies allowing for more efficient elucidation of knowledge. OBJECTIVES In this study we introduced the concept of multilevel pharmacokinetics (PK)-driven modelling for large-sample, unbalanced and unadjusted metabolomics data comprising nucleoside and creatinine concentration measurements in urine of healthy and cancer patients. METHODS A Bayesian multilevel model was proposed to describe the nucleoside and creatinine concentration ratio considering age, sex and health status as covariates. The predictive performance of the proposed model was summarized via area under the ROC, sensitivity and specificity using external validation. RESULTS Cancer was associated with an increase in methylthioadenosine/creatinine excretion rate by a factor of 1.42 (1.09-2.03) which constituted the highest increase among all nucleosides. Age influenced nucleosides/creatinine excretion rates for all nucleosides in the same direction which was likely caused by a decrease in creatinine clearance with age. There was a small evidence of sex-related differences for methylthioadenosine. The individual a posteriori prediction of patient classification as area under the ROC with 5th and 95th percentile was 0.57(0.5-0.67) with sensitivity and specificity of 0.59(0.42-0.76) and 0.57(0.45-0.7), respectively suggesting limited usefulness of 13 nucleosides/creatinine urine concentration measurements in predicting disease in this population. CONCLUSION Bayesian multilevel pharmacokinetics-driven modeling in metabolomics may be useful in understanding the data and may constitute a new tool for searching towards potential candidates of disease indicators.
Collapse
Affiliation(s)
- Emilia Daghir-Wojtkowiak
- 0000 0001 0531 3426grid.11451.30Department of Biopharmaceutics and Pharmacodynamics, Medical University of Gdańsk, Al. Gen. Hallera 107, 80-416 Gdańsk, Poland
| | - Paweł Wiczling
- 0000 0001 0531 3426grid.11451.30Department of Biopharmaceutics and Pharmacodynamics, Medical University of Gdańsk, Al. Gen. Hallera 107, 80-416 Gdańsk, Poland
| | - Małgorzata Waszczuk-Jankowska
- 0000 0001 0531 3426grid.11451.30Department of Biopharmaceutics and Pharmacodynamics, Medical University of Gdańsk, Al. Gen. Hallera 107, 80-416 Gdańsk, Poland
| | - Roman Kaliszan
- 0000 0001 0531 3426grid.11451.30Department of Biopharmaceutics and Pharmacodynamics, Medical University of Gdańsk, Al. Gen. Hallera 107, 80-416 Gdańsk, Poland
| | - Michał Jan Markuszewski
- 0000 0001 0531 3426grid.11451.30Department of Biopharmaceutics and Pharmacodynamics, Medical University of Gdańsk, Al. Gen. Hallera 107, 80-416 Gdańsk, Poland
| |
Collapse
|
17
|
Limm K, Dettmer K, Reinders J, Oefner PJ, Bosserhoff AK. Characterization of the Methylthioadenosine Phosphorylase Polymorphism rs7023954 - Incidence and Effects on Enzymatic Function in Malignant Melanoma. PLoS One 2016; 11:e0160348. [PMID: 27479139 PMCID: PMC4968798 DOI: 10.1371/journal.pone.0160348] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 07/18/2016] [Indexed: 02/07/2023] Open
Abstract
Deficiency of methylthioadenosine phosphorylase (MTAP) supports melanoma development and progression through accumulation of its substrate 5’-methylthioadenosine (MTA), which leads amongst others to a constitutive inhibition of protein arginine methyltransferases (PRMTs) and activation of the transcription factor AP-1 via the receptor ADORA2B. Genetic association studies have also suggested that genetic polymorphism in MTAP may modulate the risk of melanoma. Here, we investigated the only globally common non-synonymous single nucleotide polymorphism (SNP) reported to date for MTAP. The SNP rs7023954 is located in exon 3 (c.166G>A), and leads to the conservative substitution of one branched-chain amino acid residue (valine) for another (isoleucine) at position 56 (p.Val56Ile). Whereas genotype frequencies in normal and primary melanoma tissues or cell lines were in Hardy-Weinberg equilibrium based on cDNA amplicon sequencing, a marked (P = 0.00019) deviation was observed in metastatic melanoma tissues and cell lines due to a deficit of heterozygotes. Enzyme assays conducted on the co-dominantly expressed alleles revealed no difference in the conversion rate of MTA to adenine and 5-methylthioribose-1-phosphate, indicating that this known enzymatic activity does not modulate the tumor suppressive function of MTAP.
Collapse
Affiliation(s)
- Katharina Limm
- Institute of Biochemistry, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Katja Dettmer
- Institute of Functional Genomics, University of Regensburg, Regensburg, Germany
| | - Jörg Reinders
- Institute of Functional Genomics, University of Regensburg, Regensburg, Germany
| | - Peter J. Oefner
- Institute of Functional Genomics, University of Regensburg, Regensburg, Germany
| | - Anja-Katrin Bosserhoff
- Institute of Biochemistry, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen, CCC Erlangen-EMN, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
- * E-mail:
| |
Collapse
|
18
|
Iwanowska A, Yusa SI, Nowakowska M, Szczubiałka K. Selective adsorption of modified nucleoside cancer biomarkers by hybrid molecularly imprinted adsorbents. J Sep Sci 2016; 39:3072-80. [DOI: 10.1002/jssc.201600132] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Revised: 05/31/2016] [Accepted: 06/01/2016] [Indexed: 12/21/2022]
Affiliation(s)
| | - Shin-Ichi Yusa
- Graduate School of Engineering, Department of Materials Science and Chemistry; University of Hyogo; Himeji Hyogo Japan
| | | | | |
Collapse
|
19
|
Henrich FC, Singer K, Poller K, Bernhardt L, Strobl CD, Limm K, Ritter AP, Gottfried E, Völkl S, Jacobs B, Peter K, Mougiakakos D, Dettmer K, Oefner PJ, Bosserhoff AK, Kreutz MP, Aigner M, Mackensen A. Suppressive effects of tumor cell-derived 5'-deoxy-5'-methylthioadenosine on human T cells. Oncoimmunology 2016; 5:e1184802. [PMID: 27622058 DOI: 10.1080/2162402x.2016.1184802] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 04/13/2016] [Accepted: 04/26/2016] [Indexed: 02/01/2023] Open
Abstract
The immunosuppressive tumor microenvironment represents one of the main obstacles for immunotherapy of cancer. The tumor milieu is among others shaped by tumor metabolites such as 5'-deoxy-5'-methylthioadenosine (MTA). Increased intratumoral MTA levels result from a lack of the MTA-catabolizing enzyme methylthioadenosine phosphorylase (MTAP) in tumor cells and are found in various tumor entities. Here, we demonstrate that MTA suppresses proliferation, activation, differentiation, and effector function of antigen-specific T cells without eliciting cell death. Conversely, if MTA is added to highly activated T cells, MTA exerts cytotoxic effects on T cells. We identified the Akt pathway, a critical signal pathway for T cell activation, as a target of MTA, while, for example, p38 remained unaffected. Next, we provide evidence that MTA exerts its immunosuppressive effects by interfering with protein methylation in T cells. To confirm the relevance of the suppressive effects of exogenously added MTA on human T cells, we used an MTAP-deficient tumor cell-line that was stably transfected with the MTAP-coding sequence. We observed that T cells stimulated with MTAP-transfected tumor cells revealed a higher proliferative capacity compared to T cells stimulated with Mock-transfected cells. In conclusion, our findings reveal a novel immune evasion strategy of human tumor cells that could be of interest for therapeutic targeting.
Collapse
Affiliation(s)
- Frederik C Henrich
- Department of Internal Medicine 5 - Hematology and Oncology, University Hospital of Erlangen , Erlangen, Germany
| | - Katrin Singer
- Department of Internal Medicine 5 - Hematology and Oncology, University Hospital of Erlangen, Erlangen, Germany; Department of Internal Medicine 3 - Hematology and Oncology, University Hospital of Regensburg, Regensburg, Germany
| | - Kerstin Poller
- Department of Internal Medicine 5 - Hematology and Oncology, University Hospital of Erlangen , Erlangen, Germany
| | - Luise Bernhardt
- Department of Internal Medicine 5 - Hematology and Oncology, University Hospital of Erlangen , Erlangen, Germany
| | - Carolin D Strobl
- Department of Internal Medicine 5 - Hematology and Oncology, University Hospital of Erlangen , Erlangen, Germany
| | - Katharina Limm
- Institute of Biochemistry - Emil-Fischer-Zentrum, Friedrich-Alexander University of Erlangen-Nuremberg , Erlangen, Germany
| | - Axel P Ritter
- Institute of Functional Genomics, University of Regensburg , Regensburg, Germany
| | - Eva Gottfried
- Department of Internal Medicine 3 - Hematology and Oncology, University Hospital of Regensburg , Regensburg, Germany
| | - Simon Völkl
- Department of Internal Medicine 5 - Hematology and Oncology, University Hospital of Erlangen , Erlangen, Germany
| | - Benedikt Jacobs
- Department of Internal Medicine 5 - Hematology and Oncology, University Hospital of Erlangen, Erlangen, Germany; Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Radiumhospital, Oslo, Norway; The KG Jebsen Center for Cancer Immunotherapy, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Katrin Peter
- Department of Internal Medicine 3 - Hematology and Oncology, University Hospital of Regensburg , Regensburg, Germany
| | - Dimitrios Mougiakakos
- Department of Internal Medicine 5 - Hematology and Oncology, University Hospital of Erlangen , Erlangen, Germany
| | - Katja Dettmer
- Institute of Functional Genomics, University of Regensburg , Regensburg, Germany
| | - Peter J Oefner
- Institute of Functional Genomics, University of Regensburg , Regensburg, Germany
| | - Anja-Katrin Bosserhoff
- Institute of Biochemistry - Emil-Fischer-Zentrum, Friedrich-Alexander University of Erlangen-Nuremberg , Erlangen, Germany
| | - Marina P Kreutz
- Department of Internal Medicine 3 - Hematology and Oncology, University Hospital of Regensburg , Regensburg, Germany
| | - Michael Aigner
- Department of Internal Medicine 5 - Hematology and Oncology, University Hospital of Erlangen , Erlangen, Germany
| | - Andreas Mackensen
- Department of Internal Medicine 5 - Hematology and Oncology, University Hospital of Erlangen , Erlangen, Germany
| |
Collapse
|
20
|
Baines A, Martin P, Rorie C. Current and Emerging Targeting Strategies for Treatment of Pancreatic Cancer. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2016; 144:277-320. [DOI: 10.1016/bs.pmbts.2016.09.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
21
|
He HL, Lee YE, Shiue YL, Lee SW, Chen TJ, Li CF. Characterization and Prognostic Significance of Methylthioadenosine Phosphorylase Deficiency in Nasopharyngeal Carcinoma. Medicine (Baltimore) 2015; 94:e2271. [PMID: 26656376 PMCID: PMC5008521 DOI: 10.1097/md.0000000000002271] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Identification of cancer-associated genes by genomic profiling contributes to the elucidation of tumor development and progression. The methylthioadenosine phosphorylase (MTAP) gene, located at chromosome 9p21, plays a critical role in tumorigenicity and disease progression in a wide variety of cancers. However, the prognostic impact of MTAP in patients with nasopharyngeal carcinoma (NPC) remains obscured. Through data mining from published transcriptomic database, MTAP was first identified as a differentially downregulated gene in NPC. In this study, our aim was to evaluate the expression of MTAP in NPC and to clarify its prognostic significance.MTAP immunohistochemistry was retrospectively performed and analyzed in biopsy specimens from 124 NPC patients who received standard treatment without distant metastasis at initial diagnosis. The immunoexpression status was correlated with the clinicopathological variables, disease-specific survival (DSS), distant metastasis-free survival (DMFS), and local recurrence-free survival (LRFS). Real-time quantitative polymerase chain reaction (PCR) was used to measure MTAP gene dosage. In some cases, we also performed methylation-specific PCR and pyrosequencing to assess the status of promoter methylation.MTAP deficiency was significantly associated with advanced tumor stages (P = 0.023) and univariately predictive of adverse outcomes for DSS, DMFS, and LRFS. In the multivariate comparison, MTAP deficiency still remained prognostically independent to portend worse DSS (P = 0.021, hazard ratio = 1.870) and DMFS (P = 0.009, hazard ratio = 2.154), together with advanced AJCC stages III to IV. Homozygous deletion or promoter methylation of MTAP gene were identified to be significantly associated with MTAP protein deficiency (P < 0.001).MTAP deficiency was correlated with an aggressive phenotype and independently predictive of worse DSS and DMFS, suggesting its role in disease progression and as an independent prognostic biomarker of NPC, which potentially offers new strategy of targeted treatment for patients lacking MTAP expression.
Collapse
Affiliation(s)
- Hong-Lin He
- From the Department of Pathology, E-DA Hospital, I-Shou University, Kaohsiung, Taiwan (H-LH); Institute of Biomedical Science, National Sun Yat-Sen University, Kaohsiung, Taiwan (H-LH, Y-LS); Department of Anesthesiology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan (Y-EL); Department of Radiation Oncology, Chi-Mei Medical Center, Liouying, Tainan, Taiwan (S-WL); Department of Pathology, Chi-Mei Medical Center, Tainan, Taiwan (T-JC, C-FL); National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan (C-FL); Department of Biotechnology, Southern Taiwan University of Science and Technology, Tainan, Taiwan (C-FL); and Institute of Clinical Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan (C-FL)
| | | | | | | | | | | |
Collapse
|
22
|
Zhang B, Dong S, Li Z, Lu L, Zhang S, Chen X, Cen X, Wu Y. Targeting protein arginine methyltransferase 5 inhibits human hepatocellular carcinoma growth via the downregulation of beta-catenin. J Transl Med 2015; 13:349. [PMID: 26541651 PMCID: PMC4635578 DOI: 10.1186/s12967-015-0721-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 11/02/2015] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Protein arginine methyltransferase 5 (PRMT5), a type II PRMT, is highly expressed in some tumors, but its role in hepatocellular carcinoma (HCC) is still unknown. METHODS PRMT5 level in HCC specimens was determined by immunohistochemical staining and the association with clinicopathologic features was evaluated. PRMT5 was inhibited by AMI-1 (a small molecule inhibitor of PRMTs) or small interference RNA (siRNA). The proliferation of HCC cells was tested by Cell Counting Kit-8, cell migration was evaluated by Transwell assay and cell cycle and apoptosis were analyzed by flow cytometry. The effect of AMI-1 on HCC in vivo was examined by mouse xenograft model. RESULTS PRMT5 expression was markedly upregulated in HCC tissues, and correlated inversely with overall patient survival. Knockdown of PRMT5 significantly reduced the proliferation of HCC cells, but did not affect the growth of normal liver cells. Furthermore, β-catenin was identified as a target of PRMT5. Silencing PRMT5 significantly down-regulated the expression of β-catenin and the downstream effector Cyclin D1 in HCC cells. AMI-1 strongly inhibited HCC growth in vivo, increased the ratio of Bax/Bcl-2, and led to apoptosis and loss of migratory activity in several HCC cells. Meanwhile, AMI-1 decreased the expression levels of symmetric dimethylation of H4 (H4R3me2s), a histone mark of PRMT5. CONCLUSIONS PRMT5 plays an important role in HCC. PRMT5 may be a promising target for HCC therapy.
Collapse
Affiliation(s)
- Baolai Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Lanzhou University; Key Lab of Preclinical Study for New Drugs of Gansu Province, No 199, Dongang West Road, Lanzhou, 730000, Gansu, China.
| | - Shuhong Dong
- Department of Pharmacology, School of Basic Medical Sciences, Lanzhou University; Key Lab of Preclinical Study for New Drugs of Gansu Province, No 199, Dongang West Road, Lanzhou, 730000, Gansu, China.
| | - Zhongxin Li
- Gansu Provincial Second People's Hospital, Lanzhou, 730000, China.
| | - Li Lu
- Department of Pharmacology, School of Basic Medical Sciences, Lanzhou University; Key Lab of Preclinical Study for New Drugs of Gansu Province, No 199, Dongang West Road, Lanzhou, 730000, Gansu, China.
| | - Su Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Lanzhou University; Key Lab of Preclinical Study for New Drugs of Gansu Province, No 199, Dongang West Road, Lanzhou, 730000, Gansu, China.
| | - Xue Chen
- Department of Pharmacology, School of Basic Medical Sciences, Lanzhou University; Key Lab of Preclinical Study for New Drugs of Gansu Province, No 199, Dongang West Road, Lanzhou, 730000, Gansu, China.
| | - Xiaobo Cen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| | - Yongjie Wu
- Department of Pharmacology, School of Basic Medical Sciences, Lanzhou University; Key Lab of Preclinical Study for New Drugs of Gansu Province, No 199, Dongang West Road, Lanzhou, 730000, Gansu, China.
| |
Collapse
|
23
|
Immunomodulatory metabolites released by the frog-killing fungus Batrachochytrium dendrobatidis. Infect Immun 2015; 83:4565-70. [PMID: 26371122 DOI: 10.1128/iai.00877-15] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 09/04/2015] [Indexed: 11/20/2022] Open
Abstract
Batrachochytrium dendrobatidis is a fungal pathogen in the phylum Chytridiomycota that causes the skin disease chytridiomycosis. Chytridiomycosis is considered an emerging infectious disease linked to worldwide amphibian declines and extinctions. Although amphibians have well-developed immune defenses, clearance of this pathogen from the skin is often impaired. Previously, we showed that the adaptive immune system is involved in the control of the pathogen, but B. dendrobatidis releases factors that inhibit in vitro and in vivo lymphocyte responses and induce lymphocyte apoptosis. Little is known about the nature of the inhibitory factors released by this fungus. Here, we describe the isolation and characterization of three fungal metabolites produced by B. dendrobatidis but not by the closely related nonpathogenic chytrid Homolaphlyctis polyrhiza. These metabolites are methylthioadenosine (MTA), tryptophan, and an oxidized product of tryptophan, kynurenine (Kyn). Independently, both MTA and Kyn inhibit the survival and proliferation of amphibian lymphocytes and the Jurkat human T cell leukemia cell line. However, working together, they become effective at much lower concentrations. We hypothesize that B. dendrobatidis can adapt its metabolism to release products that alter the local environment in the skin to inhibit immunity and enhance the survival of the pathogen.
Collapse
|
24
|
Clinch K, Evans GB, Fröhlich RFG, Gulab SA, Gutierrez JA, Mason JM, Schramm VL, Tyler PC, Woolhouse AD. Transition state analogue inhibitors of human methylthioadenosine phosphorylase and bacterial methylthioadenosine/S-adenosylhomocysteine nucleosidase incorporating acyclic ribooxacarbenium ion mimics. Bioorg Med Chem 2012; 20:5181-7. [PMID: 22854195 DOI: 10.1016/j.bmc.2012.07.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Revised: 06/28/2012] [Accepted: 07/04/2012] [Indexed: 11/24/2022]
Abstract
Several acyclic hydroxy-methylthio-amines with 3-5 carbon atoms were prepared and coupled via a methylene link to 9-deazaadenine. The products were tested for inhibition against human MTAP and Escherichia coli and Neisseria meningitidis MTANs and gave K(i) values as low as 0.23 nM. These results were compared to those obtained with 1st and 2nd generation inhibitors (1S)-1-(9-deazaadenin-9-yl)-1,4-dideoxy-1,4-imino-5-methylthio-D-ribitol (MT-Immucillin-A, 3) and (3R,4S)-1-[9-deazaadenin-9-yl)methyl]3-hydroxy-4-methylthiomethylpyrrolidine (MT-DADMe-Immucillin-A, 4). The best inhibitors were found to exhibit binding affinities of approximately 2- to 4-fold those of 3 but were significantly weaker than 4. Cleavage of the 2,3 carbon-carbon bond in MT-Immucillin-A (3) gave an acyclic product (79) with a 21,500 fold loss of activity against E. coli MTAN. In another case, N-methylation of a side chain secondary amine resulted in a 250-fold loss of activity against the same enzyme [(±)-65 vs (±)-68]. The inhibition results were also contrasted with those acyclic derivatives previously prepared as inhibitors for a related enzyme, purine nucleoside phosphorylase (PNP), where some inhibitors in the latter case were found to be more potent than their cyclic counterparts.
Collapse
Affiliation(s)
- Keith Clinch
- Carbohydrate Chemistry, Industrial Research Limited, PO Box 31310, Lower Hutt 5040, New Zealand.
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Benight NM, Stoll B, Marini JC, Burrin DG. Preventative oral methylthioadenosine is anti-inflammatory and reduces DSS-induced colitis in mice. Am J Physiol Gastrointest Liver Physiol 2012; 303:G71-82. [PMID: 22556140 PMCID: PMC3404577 DOI: 10.1152/ajpgi.00549.2011] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Methylthioadenosine (MTA) is a precursor of the methionine salvage pathway and has been shown to have anti-inflammatory properties in various models of acute and chronic inflammation. However, the anti-inflammatory properties of MTA in models of intestinal inflammation are not defined. We hypothesized that orally administered MTA would be bioavailable and reduce morbidity associated with experimental colitis. We examined clinical, histological, and molecular markers of disease in mice provided oral MTA before (preventative) or after (therapy) the induction of colitis with 3% dextran sulfate sodium (DSS). We found a reduction in disease activity, weight loss, myeloperoxidase activity, and histological damage in mice given preventative MTA compared with DSS alone. We also found that equivalent supplementation with methionine could not reproduce the anti-inflammatory effects of MTA, and that MTA had no detectable adverse effects in control or DSS mice. Expression microarray analysis of colonic tissue showed several dominant pathways related to inflammatory cytokines/chemokines and extracellular matrix remodeling were upregulation by DSS and suppressed in MTA-supplemented mice. MTA is rapidly absorbed in the gastrointestinal tract and disseminated throughout the body, based on a time course analysis of an oral bolus of MTA. This effect is transient, with MTA levels falling to near baseline within 90 min in most organs. Moreover, MTA did not lead to increased blood or tissue methionine levels, suggesting that its effects are specific. However, MTA provided limited therapeutic benefit when administered after the onset of colitis. Our results show that oral MTA supplementation is a safe and effective strategy to prevent inflammation and tissue injury associated with DSS colitis in mice. Additional studies in chronic inflammatory models are necessary to determine if MTA is a safe and beneficial option for the maintenance of remission in human inflammatory bowel disease.
Collapse
Affiliation(s)
- Nancy M. Benight
- 1US Department of Agriculture/Agricltural Research Service, Children's Nutrition Research Center, Baylor College of Medicine, Houston, Texas; and
| | - Barbara Stoll
- 1US Department of Agriculture/Agricltural Research Service, Children's Nutrition Research Center, Baylor College of Medicine, Houston, Texas; and
| | - Juan C. Marini
- 1US Department of Agriculture/Agricltural Research Service, Children's Nutrition Research Center, Baylor College of Medicine, Houston, Texas; and
| | - Douglas G. Burrin
- 1US Department of Agriculture/Agricltural Research Service, Children's Nutrition Research Center, Baylor College of Medicine, Houston, Texas; and ,2Section of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
26
|
Li TWH, Yang H, Peng H, Xia M, Mato JM, Lu SC. Effects of S-adenosylmethionine and methylthioadenosine on inflammation-induced colon cancer in mice. Carcinogenesis 2011; 33:427-35. [PMID: 22159228 DOI: 10.1093/carcin/bgr295] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Chronic inflammation is an underlying risk factor for colon cancer. Tumor necrosis factor alpha (TNF-α) plays a critical role in the development of inflammation-induced colon cancer in a mouse model. S-adenosylmethionine (SAMe) and its metabolite methylthioadenosine (MTA) can inhibit lipopolysaccharide-induced TNF-α expression in macrophages. The aim of this work was to examine whether SAMe and MTA are effective in preventing inflammation-induced colon cancer and if so identify signaling pathways affected. Balb/c mice were treated with azoxymethane (AOM) and dextran sulfate sodium to induce colon cancer. Two days after AOM treatment, mice were divided into three groups: vehicle control, SAMe or MTA. Tumor load, histology, immunohistochemistry, gene and protein expression were determined. SAMe and MTA treatment reduced tumor load by ∼40%. Both treatments raised SAMe and MTA levels but MTA also raised S-adenosylhomocysteine levels. MTA treatment prevented the induction of many genes known to play pathogenetic roles in this model except for TNF-α and inducible nitric oxide synthase (iNOS). SAMe also had no effect on TNF-α or iNOS and was less inhibitory than MTA on the other genes. In vivo, both treatments induced apoptosis but inhibited proliferation, β-catenin, nuclear factor kappa B activation and interleukin (IL) 6 signaling. Effect of SAMe and MTA on IL-6 signaling was examined using Colo 205 colon cancer cells. In these cells, SAMe and MTA inhibited IL-6-induced IL-10 expression. MTA also inhibited IL-10 transcription and signal transducer and activator of transcription 3 activation. In conclusion, SAMe and MTA reduced inflammation-induced colon cancer and inhibited several pathways important in colon carcinogenesis.
Collapse
Affiliation(s)
- Tony W H Li
- Division of Gastroenterology and Liver Diseases, University of Southern California Research Center for Liver Diseases, University of Southern California-University of California, Los Angeles, CA 90033, USA
| | | | | | | | | | | |
Collapse
|
27
|
Pommerrenig B, Feussner K, Zierer W, Rabinovych V, Klebl F, Feussner I, Sauer N. Phloem-specific expression of Yang cycle genes and identification of novel Yang cycle enzymes in Plantago and Arabidopsis. THE PLANT CELL 2011; 23:1904-19. [PMID: 21540433 PMCID: PMC3123959 DOI: 10.1105/tpc.110.079657] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2010] [Revised: 03/14/2011] [Accepted: 04/15/2011] [Indexed: 05/19/2023]
Abstract
The 5-methylthioadenosine (MTA) or Yang cycle is a set of reactions that recycle MTA to Met. In plants, MTA is a byproduct of polyamine, ethylene, and nicotianamine biosynthesis. Vascular transcriptome analyses revealed phloem-specific expression of the Yang cycle gene 5-METHYLTHIORIBOSE KINASE1 (MTK1) in Plantago major and Arabidopsis thaliana. As Arabidopsis has only a single MTK gene, we hypothesized that the expression of other Yang cycle genes might also be vascular specific. Reporter gene studies and quantitative analyses of mRNA levels for all Yang cycle genes confirmed this hypothesis for Arabidopsis and Plantago. This includes the Yang cycle genes 5-METHYLTHIORIBOSE-1-PHOSPHATE ISOMERASE1 and DEHYDRATASE-ENOLASE-PHOSPHATASE-COMPLEX1. We show that these two enzymes are sufficient for the conversion of methylthioribose-1-phosphate to 1,2-dihydroxy-3-keto-5-methylthiopentene. In bacteria, fungi, and animals, the same conversion is catalyzed in three to four separate enzymatic steps. Furthermore, comparative analyses of vascular and nonvascular metabolites identified Met, S-adenosyl Met, and MTA preferentially or almost exclusively in the vascular tissue. Our data represent a comprehensive characterization of the Yang cycle in higher plants and demonstrate that the Yang cycle works primarily in the vasculature. Finally, expression analyses of polyamine biosynthetic genes suggest that the Yang cycle in leaves recycles MTA derived primarily from polyamine biosynthesis.
Collapse
Affiliation(s)
- Benjamin Pommerrenig
- Molekulare Pflanzenphysiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91058 Erlangen, Germany
- Erlangen Center of Plant Science, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91058 Erlangen, Germany
| | - Kirstin Feussner
- Abteilung Biochemie der Pflanze, Albrecht-von-Haller-Institut für Pflanzenwissenschaften, Georg-August-Universität Göttingen, 37077 Gottingen, Germany
| | - Wolfgang Zierer
- Molekulare Pflanzenphysiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91058 Erlangen, Germany
| | - Valentyna Rabinovych
- Molekulare Pflanzenphysiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91058 Erlangen, Germany
| | - Franz Klebl
- Molekulare Pflanzenphysiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91058 Erlangen, Germany
| | - Ivo Feussner
- Abteilung Biochemie der Pflanze, Albrecht-von-Haller-Institut für Pflanzenwissenschaften, Georg-August-Universität Göttingen, 37077 Gottingen, Germany
| | - Norbert Sauer
- Molekulare Pflanzenphysiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91058 Erlangen, Germany
- Erlangen Center of Plant Science, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91058 Erlangen, Germany
| |
Collapse
|
28
|
Bertino JR, Waud WR, Parker WB, Lubin M. Targeting tumors that lack methylthioadenosine phosphorylase (MTAP) activity: current strategies. Cancer Biol Ther 2011; 11:627-32. [PMID: 21301207 DOI: 10.4161/cbt.11.7.14948] [Citation(s) in RCA: 102] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Many solid tumors and hematologic malignancies lack expression of the enzyme methylthioadenosine phosphorylase (MTAP), due either to deletion of the MTAP gene or to methylation of the MTAP promoter. In cells that have MTAP, its natural substrate, methylthioadenosine (MTA), generated during polyamine biosynthesis, is cleaved to adenine and 5-methylthioribose-1-phosphate. The latter compound is further metabolized to methionine. Adenine and methionine are further metabolized and hence salvaged. In MTAP-deficient cells, however, MTA is not cleaved and the salvage pathway for adenine and methionine is absent. As a result, MTAP-deficient cells are more sensitive than MTAP-positive cells to inhibitors of de novo purine synthesis and to methionine deprivation. The challenge has been to take advantage of MTAP deficiency, and the changes in metabolism that follow, to design a strategy for targeted treatment. In this review, the frequency of MTAP-deficiency is presented and past and recent strategies to target such deficient cells are discussed, including one in which MTA is administered, followed by very high doses of a toxic purine or pyrimidine analog. In normal host cells, adenine, generated from MTA, blocks conversion of the analog to its toxic nucleotide. In MTAP-deficient tumor cells, conversion proceeds and the tumor cells are selectively killed. Successful mouse studies using this novel strategy were recently reported.
Collapse
Affiliation(s)
- Joseph R Bertino
- Departments of Medicine and Pharmacology, The Cancer Institute of NJ, Robert Wood Johnson Medical School, New Brunswick, USA.
| | | | | | | |
Collapse
|
29
|
Latasa MU, Gil-Puig C, Fernández-Barrena MG, Rodríguez-Ortigosa CM, Banales JM, Urtasun R, Goñi S, Méndez M, Arcelus S, Juanarena N, Recio JA, Lotersztajn S, Prieto J, Berasain C, Corrales FJ, Lecanda J, Ávila MA. Oral methylthioadenosine administration attenuates fibrosis and chronic liver disease progression in Mdr2-/- mice. PLoS One 2010; 5:e15690. [PMID: 21209952 PMCID: PMC3012093 DOI: 10.1371/journal.pone.0015690] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2010] [Accepted: 11/21/2010] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Inflammation and fibrogenesis are directly related to chronic liver disease progression, including hepatocellular carcinoma (HCC) development. Currently there are few therapeutic options available to inhibit liver fibrosis. We have evaluated the hepatoprotective and anti-fibrotic potential of orally-administered 5'-methylthioadenosine (MTA) in Mdr2(-/-) mice, a clinically relevant model of sclerosing cholangitis and spontaneous biliary fibrosis, followed at later stages by HCC development. METHODOLOGY MTA was administered daily by gavage to wild type and Mdr2(-/-) mice for three weeks. MTA anti-inflammatory and anti-fibrotic effects and potential mechanisms of action were examined in the liver of Mdr2(-/-) mice with ongoing fibrogenesis and in cultured liver fibrogenic cells (myofibroblasts). PRINCIPAL FINDINGS MTA treatment reduced hepatomegaly and liver injury. α-Smooth muscle actin immunoreactivity and collagen deposition were also significantly decreased. Inflammatory infiltrate, the expression of the cytokines IL6 and Mcp-1, pro-fibrogenic factors like TGFβ2 and tenascin-C, as well as pro-fibrogenic intracellular signalling pathways were reduced by MTA in vivo. MTA inhibited the activation and proliferation of isolated myofibroblasts and down-regulated cyclin D1 gene expression at the transcriptional level. The expression of JunD, a key transcription factor in liver fibrogenesis, was also reduced by MTA in activated myofibroblasts. CONCLUSIONS/SIGNIFICANCE Oral MTA administration was well tolerated and proved its efficacy in reducing liver inflammation and fibrosis. MTA may have multiple molecular and cellular targets. These include the inhibition of inflammatory and pro-fibrogenic cytokines, as well as the attenuation of myofibroblast activation and proliferation. Downregulation of JunD and cyclin D1 expression in myofibroblasts may be important regarding the mechanism of action of MTA. This compound could be a good candidate to be tested for the treatment of (biliary) liver fibrosis.
Collapse
Affiliation(s)
- M. Ujue Latasa
- Division of Hepatology and Gene Therapy, CIMA, University of Navarra, Pamplona, Spain
| | - Carmen Gil-Puig
- Division of Hepatology and Gene Therapy, CIMA, University of Navarra, Pamplona, Spain
- Digna Biotech, Madrid, Spain
| | - Maite G. Fernández-Barrena
- Division of Hepatology and Gene Therapy, CIMA, University of Navarra, Pamplona, Spain
- CIBERehd, University Clinic, University of Navarra, Pamplona, Spain
| | - Carlos M. Rodríguez-Ortigosa
- Division of Hepatology and Gene Therapy, CIMA, University of Navarra, Pamplona, Spain
- CIBERehd, University Clinic, University of Navarra, Pamplona, Spain
| | - Jesús M. Banales
- Division of Hepatology and Gene Therapy, CIMA, University of Navarra, Pamplona, Spain
- CIBERehd, University Clinic, University of Navarra, Pamplona, Spain
| | - Raquel Urtasun
- Division of Hepatology and Gene Therapy, CIMA, University of Navarra, Pamplona, Spain
| | - Saioa Goñi
- Division of Hepatology and Gene Therapy, CIMA, University of Navarra, Pamplona, Spain
| | - Miriam Méndez
- Division of Hepatology and Gene Therapy, CIMA, University of Navarra, Pamplona, Spain
| | - Sara Arcelus
- CIBERehd, University Clinic, University of Navarra, Pamplona, Spain
| | - Nerea Juanarena
- Division of Hepatology and Gene Therapy, CIMA, University of Navarra, Pamplona, Spain
| | - Juan A. Recio
- Vall d'Hebron Research Institute, Institute of Oncology and Hospital, Barcelona, Spain
| | - Sophie Lotersztajn
- Inserm, U955, Créteil, France
- Université Paris-Est, Faculté de Médecine, UMR-S955, Créteil, France
| | - Jesús Prieto
- Division of Hepatology and Gene Therapy, CIMA, University of Navarra, Pamplona, Spain
- CIBERehd, University Clinic, University of Navarra, Pamplona, Spain
| | - Carmen Berasain
- Division of Hepatology and Gene Therapy, CIMA, University of Navarra, Pamplona, Spain
| | - Fernando J. Corrales
- Division of Hepatology and Gene Therapy, CIMA, University of Navarra, Pamplona, Spain
| | - Jon Lecanda
- Division of Hepatology and Gene Therapy, CIMA, University of Navarra, Pamplona, Spain
- Digna Biotech, Madrid, Spain
| | - Matías A. Ávila
- Division of Hepatology and Gene Therapy, CIMA, University of Navarra, Pamplona, Spain
| |
Collapse
|