51
|
Lord A, Ficz G. Corrupted devolution: how normal cells are reborn as cancer precursors. Int J Biochem Cell Biol 2022; 149:106263. [DOI: 10.1016/j.biocel.2022.106263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 06/27/2022] [Accepted: 07/03/2022] [Indexed: 11/26/2022]
|
52
|
Wang ZW, Pan JJ, Hu JF, Zhang JQ, Huang L, Huang Y, Liao CY, Yang C, Chen ZW, Wang YD, Shen BY, Tian YF, Chen S. SRSF3-mediated regulation of N6-methyladenosine modification-related lncRNA ANRIL splicing promotes resistance of pancreatic cancer to gemcitabine. Cell Rep 2022; 39:110813. [PMID: 35545048 DOI: 10.1016/j.celrep.2022.110813] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 03/21/2022] [Accepted: 04/21/2022] [Indexed: 02/06/2023] Open
Abstract
Serine/arginine-rich splicing factor 3 (SRSF3) regulates mRNA alternative splicing of more than 90% of protein-coding genes, providing an essential source for biological versatility. This study finds that SRSF3 expression is associated with drug resistance and poor prognosis in pancreatic cancer. We also find that SRSF3 regulates ANRIL splicing and m6A modification of ANRIL in pancreatic cancer cells. More importantly, we demonstrate that m6A methylation on lncRNA ANRIL is essential for the splicing. Moreover, our results show that SRSF3 promotes gemcitabine resistance by regulating ANRIL's splicing and ANRIL-208 (one of the ANRIL spliceosomes) can enhance DNA homologous recombination repair (HR) capacity by forming a complex with Ring1b and EZH2. In conclusion, this study establishes a link between SRSF3, m6A modification, lncRNA splicing, and DNA HR in pancreatic cancer and demonstrates that abnormal alternative splicing and m6A modification are closely related to chemotherapy resistance in pancreatic cancer.
Collapse
Affiliation(s)
- Zu-Wei Wang
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, Fuzhou 350001, China
| | - Jing-Jing Pan
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, Fuzhou 350001, China
| | - Jian-Fei Hu
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, Fuzhou 350001, China
| | - Jia-Qiang Zhang
- Department of General Surgery, Pancreatic Disease Center, Research Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Long Huang
- Department of Hepatopancreatobiliary Surgery, Fujian Provincial Hospital, Fuzhou 350001, China
| | - Yi Huang
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, Fuzhou 350001, China; Center for Experimental Research in Clinical Medicine, Fujian Provincial Hospital, Fuzhou 350001, China
| | - Cheng-Yu Liao
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, Fuzhou 350001, China
| | - Can Yang
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, Fuzhou 350001, China
| | - Zhi-Wen Chen
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, Fuzhou 350001, China
| | - Yao-Dong Wang
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, Fuzhou 350001, China; Department of Hepatopancreatobiliary Surgery, Fujian Provincial Hospital, Fuzhou 350001, China
| | - Bai-Yong Shen
- Department of General Surgery, Pancreatic Disease Center, Research Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yi-Feng Tian
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, Fuzhou 350001, China; Department of Hepatopancreatobiliary Surgery, Fujian Provincial Hospital, Fuzhou 350001, China.
| | - Shi Chen
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, Fuzhou 350001, China; Department of Hepatopancreatobiliary Surgery, Fujian Provincial Hospital, Fuzhou 350001, China.
| |
Collapse
|
53
|
Wu K, Liu Y, Liu L, Peng Y, Pang H, Sun X, Xia D. Emerging Trends and Research Foci in Tumor Microenvironment of Pancreatic Cancer: A Bibliometric and Visualized Study. Front Oncol 2022; 12:810774. [PMID: 35515122 PMCID: PMC9063039 DOI: 10.3389/fonc.2022.810774] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 03/21/2022] [Indexed: 01/07/2023] Open
Abstract
Background Pancreatic cancer (PC) is a serious disease with high mortality. The tumor microenvironment plays a key role in the occurrence and development of PC. The purpose of this study is to analyze trends by year, country, institution, journal, reference and keyword in publications on the PC microenvironment and to predict future research hotspots. Methods The Web of Science Core Collection was used to search for publications. We analyzed the contributions of various countries/regions, institutes, and authors and identified research hotspots and promising future trends using the CiteSpace and VOSviewer programs. We also summarized relevant completed clinical trials. Results A total of 2,155 papers on the PC microenvironment published between 2011 and 2021 were included in the study. The number of publications has increased every year. The average number of citations per article was 32.69. The USA had the most publications, followed by China, and a total of 50 influential articles were identified through co-citation analysis. Clustering analysis revealed two clusters of keywords: basic research and clinical application. The co-occurrence cluster analysis showed glutamine metabolism, carcinoma-associated fibroblasts, oxidative phosphorylation as the highly concerned research topics of basic research in recently. The three latest hot topics in clinical application are liposomes, endoscopic ultrasound and photodynamic therapy. Conclusion The number of publications and research interest have generally increased, and the USA has made prominent contributions to the study of the tumor microenvironment of PC. The current research hotspots mainly focus on energy metabolism in the hypoxic tumor microenvironment, cancer associated fibroblasts in regulating the tumor microenvironment, accurate diagnosis, drug delivery and new treatments.
Collapse
Affiliation(s)
- Kaiwen Wu
- Department of Gastroenterology, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, China.,Southwest Jiaotong University College of Medicine, Southwest Jiaotong University Affiliated Chengdu Third People's Hospital, Chengdu, China
| | - Ye Liu
- Naval Medical University, Shanghai, China
| | - Lei Liu
- Medical Research Center, Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
| | - Yunlan Peng
- Southwest Jiaotong University College of Medicine, Southwest Jiaotong University Affiliated Chengdu Third People's Hospital, Chengdu, China
| | - Honglin Pang
- Southwest Jiaotong University College of Medicine, Southwest Jiaotong University Affiliated Chengdu Third People's Hospital, Chengdu, China
| | - Xiaobin Sun
- Department of Gastroenterology, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
| | - Demeng Xia
- Luodian Clinical Drug Research Center, Shanghai Baoshan Luodian Hospital, Shanghai University, Shanghai, China
| |
Collapse
|
54
|
Wang SY, Hu QC, Wu T, Xia J, Tao XA, Cheng B. Abnormal lipid synthesis as a therapeutic target for cancer stem cells. World J Stem Cells 2022; 14:146-162. [PMID: 35432735 PMCID: PMC8963380 DOI: 10.4252/wjsc.v14.i2.146] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 05/19/2021] [Accepted: 02/20/2022] [Indexed: 02/06/2023] Open
Abstract
Cancer stem cells (CSCs) comprise a subpopulation of cancer cells with stem cell properties, which exhibit the characteristics of high tumorigenicity, self-renewal, and tumor initiation and are associated with the occurrence, metastasis, therapy resistance, and relapse of cancer. Compared with differentiated cells, CSCs have unique metabolic characteristics, and metabolic reprogramming contributes to the self-renewal and maintenance of stem cells. It has been reported that CSCs are highly dependent on lipid metabolism to maintain stemness and satisfy the requirements of biosynthesis and energy metabolism. In this review, we demonstrate that lipid anabolism alterations promote the survival of CSCs, including de novo lipogenesis, lipid desaturation, and cholesterol synthesis. In addition, we also emphasize the molecular mechanism underlying the relationship between lipid synthesis and stem cell survival, the signal trans-duction pathways involved, and the application prospect of lipid synthesis reprogramming in CSC therapy. It is demonstrated that the dependence on lipid synthesis makes targeting of lipid synthesis metabolism a promising therapeutic strategy for eliminating CSCs. Targeting key molecules in lipid synthesis will play an important role in anti-CSC therapy.
Collapse
Affiliation(s)
- Si-Yu Wang
- Department of Oral Medicine, Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510000, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510000, Guangdong Province, China
| | - Qin-Chao Hu
- Department of Oral Medicine, Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510000, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510000, Guangdong Province, China
| | - Tong Wu
- Department of Oral Medicine, Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510000, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510000, Guangdong Province, China
| | - Juan Xia
- Department of Oral Medicine, Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510000, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510000, Guangdong Province, China
| | - Xiao-An Tao
- Department of Oral Medicine, Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510000, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510000, Guangdong Province, China
| | - Bin Cheng
- Department of Oral Medicine, Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510000, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510000, Guangdong Province, China
| |
Collapse
|
55
|
Xue D, Xu Y, Kyani A, Roy J, Dai L, Sun D, Neamati N. Multiparameter Optimization of Oxidative Phosphorylation Inhibitors for the Treatment of Pancreatic Cancer. J Med Chem 2022; 65:3404-3419. [PMID: 35167303 DOI: 10.1021/acs.jmedchem.1c01934] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Targeting oxidative phosphorylation (OXPHOS) complexes is an emerging strategy to disrupt the metabolism of select cancer subtypes and to overcome resistance to targeted therapies. Here, we describe our lead optimization campaign on a series of benzene-1,4-disulfonamides as novel OXPHOS complex I inhibitors. This effort led to the discovery of compound 23 (DX3-213B) as one of the most potent complex I inhibitors reported to date. DX3-213B disrupts adenosine triphosphate (ATP) generation, inhibits complex I function, and results in the growth inhibition of pancreatic cancer cells in the low nanomolar range. Importantly, the oral administration of DX3-213B resulted in significant in vivo efficacy in a pancreatic cancer syngeneic model without obvious toxicity. Our data clearly demonstrate that OXPHOS inhibition can be a safe and efficacious strategy to treat pancreatic cancer.
Collapse
|
56
|
Nie S, Shi Z, Shi M, Li H, Qian X, Peng C, Ding X, Zhang S, Lv Y, Wang L, Kong B, Zou X, Shen S. PPARγ/SOD2 Protects Against Mitochondrial ROS-Dependent Apoptosis via Inhibiting ATG4D-Mediated Mitophagy to Promote Pancreatic Cancer Proliferation. Front Cell Dev Biol 2022; 9:745554. [PMID: 35186942 PMCID: PMC8847684 DOI: 10.3389/fcell.2021.745554] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 11/17/2021] [Indexed: 12/18/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an extremely aggressive disease with poor prognosis. Our previous study found that peroxisome proliferator activated receptor gamma (PPARγ) was capable of enhancing glycolysis in PDAC cells. However, whether PPARγ could promote PDAC progression remains unclear. In our present study, PPARγ was positively associated with tumor size and poor prognosis in PDAC patients. Functional assays demonstrated that PPARγ could promote the proliferation of pancreatic cancer cells in vitro and in vivo. Additionally, flow cytometry results showed that PPARγ decreased mitochondrial reactive oxygen species (mitochondrial ROS) production, stabilized mitochondrial membrane potential (MMP) and inhibited cell apoptosis via up-regulating superoxide dismutase 2 (SOD2), followed by the inhibition of ATG4D-mediated mitophagy. Meanwhile, the activation of PPARγ might reduce pancreatic cancer cell stemness to improve PDAC chemosensitivity via down-regulating ATG4D. Thus, these results revealed that PPARγ/SOD2 might protect against mitochondrial ROS-dependent apoptosis via inhibiting ATG4D-mediated mitophagy to promote pancreatic cancer proliferation, further improving PDAC chemosensitivity.
Collapse
Affiliation(s)
- Shuang Nie
- Department of Gastroenterology, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China.,Nanjing University Institute of Pancreatology, Nanjing, China
| | - Zhao Shi
- Nanjing University Institute of Pancreatology, Nanjing, China.,Department of Gastroenterology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China
| | - Mengyue Shi
- Department of Gastroenterology, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China.,Nanjing University Institute of Pancreatology, Nanjing, China
| | - Hongzhen Li
- Department of Gastroenterology, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China.,Nanjing University Institute of Pancreatology, Nanjing, China
| | - Xuetian Qian
- Nanjing University Institute of Pancreatology, Nanjing, China.,Department of Gastroenterology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China
| | - Chunyan Peng
- Department of Gastroenterology, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China.,Nanjing University Institute of Pancreatology, Nanjing, China
| | - Xiwei Ding
- Department of Gastroenterology, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China.,Nanjing University Institute of Pancreatology, Nanjing, China
| | - Shu Zhang
- Department of Gastroenterology, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China.,Nanjing University Institute of Pancreatology, Nanjing, China
| | - Ying Lv
- Department of Gastroenterology, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China.,Nanjing University Institute of Pancreatology, Nanjing, China.,Department of Gastroenterology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China
| | - Lei Wang
- Department of Gastroenterology, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China.,Nanjing University Institute of Pancreatology, Nanjing, China.,Department of Gastroenterology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China
| | - Bo Kong
- Department of Gastroenterology, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China.,Nanjing University Institute of Pancreatology, Nanjing, China.,Department of Surgery, Ulm University Hospital, Ulm University, Ulm, Germany
| | - Xiaoping Zou
- Department of Gastroenterology, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China.,Nanjing University Institute of Pancreatology, Nanjing, China.,Department of Gastroenterology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China
| | - Shanshan Shen
- Department of Gastroenterology, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China.,Nanjing University Institute of Pancreatology, Nanjing, China.,Department of Gastroenterology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China
| |
Collapse
|
57
|
Bedi M, Ray M, Ghosh A. Active mitochondrial respiration in cancer: a target for the drug. Mol Cell Biochem 2022; 477:345-361. [PMID: 34716860 DOI: 10.1007/s11010-021-04281-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 10/21/2021] [Indexed: 12/21/2022]
Abstract
The relative contribution of mitochondrial respiration and subsequent energy production in malignant cells has remained controversial to date. Enhanced aerobic glycolysis and impaired mitochondrial respiration have gained more attention in the metabolic study of cancer. In contrast to the popular concept, mitochondria of cancer cells oxidize a diverse array of metabolic fuels to generate a majority of the cellular energy by respiration. Several mitochondrial respiratory chain (MRC) subunits' expressions are critical for the growth, metastasis, and cancer cell invasion. Also, the assembly factors, which regulate the integration of individual MRC complexes into native super-complexes, are upregulated in cancer. Moreover, a series of anti-cancer drugs function by inhibiting respiration and ATP production. In this review, we have specified the roles of mitochondrial fuels, MRC subunits, and super-complex assembly factors that promote active respiration across different cancer types and discussed the potential roles of MRC inhibitor drugs in controlling cancer.
Collapse
Affiliation(s)
- Minakshi Bedi
- Department of Biochemistry, University of Calcutta, 35 Ballygunge Circular Road, Kolkata, West Bengal, 700019, India
| | - Manju Ray
- Department of Biophysics, Bose Institute, P 1/12, CIT Scheme VII M, Kolkata, West Bengal, 700054, India
- Department of Chemistry, Institute of Applied Science & Humanities GLA University Mathura, 17km Stone, NH-2, Mathura-Delhi Road, Mathura, UP, 281 406, India
| | - Alok Ghosh
- Department of Biochemistry, University of Calcutta, 35 Ballygunge Circular Road, Kolkata, West Bengal, 700019, India.
| |
Collapse
|
58
|
Reyes-Castellanos G, Abdel Hadi N, Carrier A. Autophagy Contributes to Metabolic Reprogramming and Therapeutic Resistance in Pancreatic Tumors. Cells 2022; 11:426. [PMID: 35159234 PMCID: PMC8834004 DOI: 10.3390/cells11030426] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/21/2022] [Accepted: 01/24/2022] [Indexed: 02/06/2023] Open
Abstract
Metabolic reprogramming is a feature of cancers for which recent research has been particularly active, providing numerous insights into the mechanisms involved. It occurs across the entire cancer process, from development to resistance to therapies. Established tumors exhibit dependencies for metabolic pathways, constituting vulnerabilities that can be targeted in the clinic. This knowledge is of particular importance for cancers that are refractory to any therapeutic approach, such as Pancreatic Ductal Adenocarcinoma (PDAC). One of the metabolic pathways dysregulated in PDAC is autophagy, a survival process that feeds the tumor with recycled intracellular components, through both cell-autonomous (in tumor cells) and nonautonomous (from the local and distant environment) mechanisms. Autophagy is elevated in established PDAC tumors, contributing to aberrant proliferation and growth even in a nutrient-poor context. Critical elements link autophagy to PDAC including genetic alterations, mitochondrial metabolism, the tumor microenvironment (TME), and the immune system. Moreover, high autophagic activity in PDAC is markedly related to resistance to current therapies. In this context, combining autophagy inhibition with standard chemotherapy, and/or drugs targeting other vulnerabilities such as metabolic pathways or the immune response, is an ongoing clinical strategy for which there is still much to do through translational and multidisciplinary research.
Collapse
Affiliation(s)
| | | | - Alice Carrier
- Centre de Recherche en Cancérologie de Marseille (CRCM), CNRS, INSERM, Institut Paoli-Calmettes, Aix Marseille Université, F-13009 Marseille, France; (G.R.-C.); (N.A.H.)
| |
Collapse
|
59
|
Hassan G, Ohara T, Afify SM, Kumon K, Zahra MH, Fu X, Al Kadi M, Seno A, Salomon DS, Seno M. Different pancreatic cancer microenvironments convert iPSCs into cancer stem cells exhibiting distinct plasticity with altered gene expression of metabolic pathways. J Exp Clin Cancer Res 2022; 41:29. [PMID: 35063003 PMCID: PMC8781112 DOI: 10.1186/s13046-021-02167-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 11/01/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Cancer stem cells (CSCs) are generated under irregular microenvironment in vivo, of which mimic is quite difficult due to the lack of enough information of the factors responsible for cancer initiation. Here, we demonstrated that mouse induced pluripotent cells (miPSCs) reprogrammed from normal embryonic fibroblasts were susceptible to the microenvironment affected by cancer cells to convert into CSCs in vivo. METHODS Three different pancreatic cancer line cells, BxPC3, PANC1, and PK8 cells were mixed with miPSCs and subcutaneously injected into immunodeficient mice. Tumors were evaluated by histological analysis and cells derived from iPSCs were isolated and selected from tumors. The isolated cells were characterized for cancer stem cell characters in vitro and in vivo as well as their responses to anticancer drugs. The impact of co-injection of iPSCs with cancer cells on transcriptome and signaling pathways of iPSCs was investigated. RESULTS The injection of miPSCs mixed with human pancreatic cancer cells into immunodeficient mice maintained the stemness of miPSCs and changed their phenotype. The miPSCs acquired CSC characteristics of tumorigenicity and self-renewal. The drug responses and the metastatic ability of CSCs converted from miPSCs varied depending on the microenvironment of cancer cells. Interestingly, transcriptome profiles of these cells indicated that the pathways related with aggressiveness and energy production were upregulated from the levels of miPSCs. CONCLUSIONS Our result suggests that cancer-inducing microenvironment in vivo could rewire the cell signaling and metabolic pathways to convert normal stem cells into CSCs.
Collapse
Affiliation(s)
- Ghmkin Hassan
- Department of Biotechnology and Drug Discovery, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, 3.1.1 Tsushima-Naka, Kita, Okayama, 700-8530, Japan
- Department of Genomic Oncology and Oral Medicine, Graduate School of Biomedical and Health Science, Hiroshima University, Hiroshima, 734-8553, Japan
| | - Toshiaki Ohara
- Department of Pathology and Experimental Medicine, Medical School, Okayama University, Okayama, 700-8558, Japan
| | - Said M Afify
- Department of Biotechnology and Drug Discovery, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, 3.1.1 Tsushima-Naka, Kita, Okayama, 700-8530, Japan
- Division of Biochemistry, Chemistry Department, Faculty of Science, Menoufia University, Shebin El Koum-Menoufia, 32511, Egypt
| | - Kazuki Kumon
- Department of Biotechnology and Drug Discovery, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, 3.1.1 Tsushima-Naka, Kita, Okayama, 700-8530, Japan
| | - Maram H Zahra
- Department of Biotechnology and Drug Discovery, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, 3.1.1 Tsushima-Naka, Kita, Okayama, 700-8530, Japan
| | - Xiaoying Fu
- Department of Biotechnology and Drug Discovery, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, 3.1.1 Tsushima-Naka, Kita, Okayama, 700-8530, Japan
- Department of Pathology, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Mohamad Al Kadi
- Department of Bacterial Infections, Research Institute for Microbial Diseases, Osaka University, Osaka, 565-0871, Japan
| | - Akimasa Seno
- Department of Biotechnology and Drug Discovery, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, 3.1.1 Tsushima-Naka, Kita, Okayama, 700-8530, Japan
- The Laboratory of Natural Food and Medicine, Co., Ltd., Okayama, 700-8530, Japan
| | - David S Salomon
- Mouse genetics program, Center for Cancer Research, National Cancer Institute, Frederick, MD, 21702-1201, USA
| | - Masaharu Seno
- Department of Biotechnology and Drug Discovery, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, 3.1.1 Tsushima-Naka, Kita, Okayama, 700-8530, Japan.
| |
Collapse
|
60
|
Moynihan E, Bassi G, Ruffini A, Panseri S, Montesi M, Velasco-Torrijos T, Montagner D. Click Pt(IV)-Carbohydrates Pro-Drugs for Treatment of Osteosarcoma. Front Chem 2021; 9:795997. [PMID: 34950638 PMCID: PMC8688915 DOI: 10.3389/fchem.2021.795997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 11/08/2021] [Indexed: 11/29/2022] Open
Abstract
The selectivity vs. cancer cells has always been a major challenge for chemotherapeutic agents and in particular for cisplatin, one of the most important anticancer drugs for the treatment of several types of tumors. One strategy to overtake this challenge is to modify the coordination sphere of the metallic center with specific vectors whose receptors are overexpressed in the tumoral cell membrane, such as monosaccharides. In this paper, we report the synthesis of four novel glyco-modified Pt(IV) pro-drugs, based on cisplatin scaffold, and their biological activity against osteosarcoma (OS), a malignant tumor affecting in particular adolescents and young adults. The sugar moiety and the Pt scaffold are linked exploiting the Copper Azide Alkyne Cycloaddition (CUAAC) reaction, which has become the flagship of click chemistry due to its versatility and mild conditions. Cytotoxicity and drug uptake on three different OS cell lines as well as CSCs (Cancer Stem Cell) are described.
Collapse
Affiliation(s)
- Eoin Moynihan
- Department of Chemistry, Maynooth University, Maynooth, Ireland
| | - Giada Bassi
- Institute of Science and Technology for Ceramics-National Research Council, Faenza, Italy
| | - Andrea Ruffini
- Institute of Science and Technology for Ceramics-National Research Council, Faenza, Italy
| | - Silvia Panseri
- Institute of Science and Technology for Ceramics-National Research Council, Faenza, Italy
| | - Monica Montesi
- Institute of Science and Technology for Ceramics-National Research Council, Faenza, Italy
| | - Trinidad Velasco-Torrijos
- Department of Chemistry, Maynooth University, Maynooth, Ireland.,Kathleen Londsdale Institute for Human Health Research, Maynooth University, Maynooth, Ireland
| | - Diego Montagner
- Department of Chemistry, Maynooth University, Maynooth, Ireland.,Kathleen Londsdale Institute for Human Health Research, Maynooth University, Maynooth, Ireland
| |
Collapse
|
61
|
Alors-Perez E, Blázquez-Encinas R, Alcalá S, Viyuela-García C, Pedraza-Arevalo S, Herrero-Aguayo V, Jiménez-Vacas JM, Mafficini A, Sánchez-Frías ME, Cano MT, Abollo-Jiménez F, Marín-Sanz JA, Cabezas-Sainz P, Lawlor RT, Luchini C, Sánchez L, Sánchez-Hidalgo JM, Ventura S, Martin-Hijano L, Gahete MD, Scarpa A, Arjona-Sánchez Á, Ibáñez-Costa A, Sainz B, Luque RM, Castaño JP. Dysregulated splicing factor SF3B1 unveils a dual therapeutic vulnerability to target pancreatic cancer cells and cancer stem cells with an anti-splicing drug. J Exp Clin Cancer Res 2021; 40:382. [PMID: 34857016 PMCID: PMC8638119 DOI: 10.1186/s13046-021-02153-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 10/23/2021] [Indexed: 12/13/2022] Open
Abstract
Background Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal cancer, requiring novel treatments to target both cancer cells and cancer stem cells (CSCs). Altered splicing is emerging as both a novel cancer hallmark and an attractive therapeutic target. The core splicing factor SF3B1 is heavily altered in cancer and can be inhibited by Pladienolide-B, but its actionability in PDAC is unknown. We explored the presence and role of SF3B1 in PDAC and interrogated its potential as an actionable target. Methods SF3B1 was analyzed in PDAC tissues, an RNA-seq dataset, and publicly available databases, examining associations with splicing alterations and key features/genes. Functional assays in PDAC cell lines and PDX-derived CSCs served to test Pladienolide-B treatment effects in vitro, and in vivo in zebrafish and mice. Results SF3B1 was overexpressed in human PDAC and associated with tumor grade and lymph-node involvement. SF3B1 levels closely associated with distinct splicing event profiles and expression of key PDAC players (KRAS, TP53). In PDAC cells, Pladienolide-B increased apoptosis and decreased multiple tumor-related features, including cell proliferation, migration, and colony/sphere formation, altering AKT and JNK signaling, and favoring proapoptotic splicing variants (BCL-XS/BCL-XL, KRASa/KRAS, Δ133TP53/TP53). Importantly, Pladienolide-B similarly impaired CSCs, reducing their stemness capacity and increasing their sensitivity to chemotherapy. Pladienolide-B also reduced PDAC/CSCs xenograft tumor growth in vivo in zebrafish and in mice. Conclusion SF3B1 overexpression represents a therapeutic vulnerability in PDAC, as altered splicing can be targeted with Pladienolide-B both in cancer cells and CSCs, paving the way for novel therapies for this lethal cancer. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-021-02153-9.
Collapse
Affiliation(s)
- Emilia Alors-Perez
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Córdoba, Spain.,Department of Cell Biology, Physiology, and Immunology, University of Cordoba, Córdoba, Spain.,Reina Sofia University Hospital, Córdoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Avenida Menéndez Pidal s/n, Edificio IMIBIC, 14004, Córdoba, Spain
| | - Ricardo Blázquez-Encinas
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Córdoba, Spain.,Department of Cell Biology, Physiology, and Immunology, University of Cordoba, Córdoba, Spain.,Reina Sofia University Hospital, Córdoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Avenida Menéndez Pidal s/n, Edificio IMIBIC, 14004, Córdoba, Spain
| | - Sonia Alcalá
- Department of Biochemistry, Universidad Autónoma de Madrid (UAM) and Department of Cancer Biology, Instituto de Investigaciones Biomédicas Alberto Sols (IIBM), CSIC-UAM, Madrid, Spain.,Department of Cancer Biology, Chronic Diseases and Cancer Area 3-Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Cristina Viyuela-García
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Córdoba, Spain.,Reina Sofia University Hospital, Córdoba, Spain.,Surgery Service, Reina Sofia University Hospital, Córdoba, Spain
| | - Sergio Pedraza-Arevalo
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Córdoba, Spain.,Department of Cell Biology, Physiology, and Immunology, University of Cordoba, Córdoba, Spain.,Reina Sofia University Hospital, Córdoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Avenida Menéndez Pidal s/n, Edificio IMIBIC, 14004, Córdoba, Spain
| | - Vicente Herrero-Aguayo
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Córdoba, Spain.,Department of Cell Biology, Physiology, and Immunology, University of Cordoba, Córdoba, Spain.,Reina Sofia University Hospital, Córdoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Avenida Menéndez Pidal s/n, Edificio IMIBIC, 14004, Córdoba, Spain
| | - Juan M Jiménez-Vacas
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Córdoba, Spain.,Department of Cell Biology, Physiology, and Immunology, University of Cordoba, Córdoba, Spain.,Reina Sofia University Hospital, Córdoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Avenida Menéndez Pidal s/n, Edificio IMIBIC, 14004, Córdoba, Spain
| | - Andrea Mafficini
- ARC-Net Research Centre, University and Hospital Trust of Verona, Verona, Italy
| | - Marina E Sánchez-Frías
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Córdoba, Spain.,Reina Sofia University Hospital, Córdoba, Spain.,Pathology Service, Reina Sofia University Hospital, Córdoba, Spain
| | - María T Cano
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Córdoba, Spain.,Reina Sofia University Hospital, Córdoba, Spain.,Medical Oncology Service, Reina Sofia University Hospital, Córdoba, Spain
| | - Fernando Abollo-Jiménez
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Córdoba, Spain.,Department of Computer Sciences, University of Cordoba, Córdoba, Spain
| | - Juan A Marín-Sanz
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Córdoba, Spain.,Department of Computer Sciences, University of Cordoba, Córdoba, Spain
| | - Pablo Cabezas-Sainz
- Department of Zoology, Genetics and Physical Anthropology, University of Santiago de Compostela, Lugo, Spain
| | - Rita T Lawlor
- ARC-Net Research Centre, University and Hospital Trust of Verona, Verona, Italy
| | - Claudio Luchini
- ARC-Net Research Centre, University and Hospital Trust of Verona, Verona, Italy.,Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy
| | - Laura Sánchez
- Department of Zoology, Genetics and Physical Anthropology, University of Santiago de Compostela, Lugo, Spain
| | - Juan M Sánchez-Hidalgo
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Córdoba, Spain.,Reina Sofia University Hospital, Córdoba, Spain.,Surgery Service, Reina Sofia University Hospital, Córdoba, Spain
| | - Sebastián Ventura
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Córdoba, Spain.,Department of Computer Sciences, University of Cordoba, Córdoba, Spain
| | - Laura Martin-Hijano
- Department of Biochemistry, Universidad Autónoma de Madrid (UAM) and Department of Cancer Biology, Instituto de Investigaciones Biomédicas Alberto Sols (IIBM), CSIC-UAM, Madrid, Spain.,Department of Cancer Biology, Chronic Diseases and Cancer Area 3-Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Manuel D Gahete
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Córdoba, Spain.,Department of Cell Biology, Physiology, and Immunology, University of Cordoba, Córdoba, Spain.,Reina Sofia University Hospital, Córdoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Avenida Menéndez Pidal s/n, Edificio IMIBIC, 14004, Córdoba, Spain
| | - Aldo Scarpa
- ARC-Net Research Centre, University and Hospital Trust of Verona, Verona, Italy.,Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy
| | - Álvaro Arjona-Sánchez
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Córdoba, Spain.,Reina Sofia University Hospital, Córdoba, Spain.,Surgery Service, Reina Sofia University Hospital, Córdoba, Spain
| | - Alejandro Ibáñez-Costa
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Córdoba, Spain.,Department of Cell Biology, Physiology, and Immunology, University of Cordoba, Córdoba, Spain.,Reina Sofia University Hospital, Córdoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Avenida Menéndez Pidal s/n, Edificio IMIBIC, 14004, Córdoba, Spain
| | - Bruno Sainz
- Department of Biochemistry, Universidad Autónoma de Madrid (UAM) and Department of Cancer Biology, Instituto de Investigaciones Biomédicas Alberto Sols (IIBM), CSIC-UAM, Madrid, Spain. .,Centro de Investigación Biomédica en Red, Área Cáncer, CIBERONC, ISCIII, Madrid, Spain.
| | - Raúl M Luque
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Córdoba, Spain. .,Department of Cell Biology, Physiology, and Immunology, University of Cordoba, Córdoba, Spain. .,Reina Sofia University Hospital, Córdoba, Spain. .,CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Avenida Menéndez Pidal s/n, Edificio IMIBIC, 14004, Córdoba, Spain.
| | - Justo P Castaño
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Córdoba, Spain. .,Department of Cell Biology, Physiology, and Immunology, University of Cordoba, Córdoba, Spain. .,Reina Sofia University Hospital, Córdoba, Spain. .,CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Avenida Menéndez Pidal s/n, Edificio IMIBIC, 14004, Córdoba, Spain.
| |
Collapse
|
62
|
Song IS, Jeong YJ, Jung Y, Park YH, Shim S, Kim SJ, Eom DW, Hong SM, Lee PCW, Kim SU, Jang SW. The sulfiredoxin-peroxiredoxin redox system regulates the stemness and survival of colon cancer stem cells. Redox Biol 2021; 48:102190. [PMID: 34798428 PMCID: PMC8605387 DOI: 10.1016/j.redox.2021.102190] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 11/14/2021] [Indexed: 12/11/2022] Open
Abstract
Cancer stem cells (CSCs) initiate tumor formation and are known to be resistant to chemotherapy. A metabolic alteration in CSCs plays a critical role in stemness and survival. However, the association between mitochondrial energy metabolism and the redox system remains undefined in colon CSCs. In this study, we assessed the role of the Sulfiredoxin-Peroxiredoxin (Srx-Prx) redox system and mitochondrial oxidative phosphorylation (OXPHOS) in maintaining the stemness and survival of colon CSCs. Notably, Srx contributed to the stability of PrxI, PrxII, and PrxIII proteins in colon CSCs. Increased Srx expression promoted the stemness and survival of CSCs and was important for the maintenance of the mitochondrial OXPHOS system. Furthermore, Nrf2 and FoxM1 led to OXPHOS activation and upregulated expression of Srx-Prx redox system-related genes. Therefore, the Nrf2/FoxM1-induced Srx-Prx redox system is a potential therapeutic target for eliminating CSCs in colon cancer.
Collapse
Affiliation(s)
- In-Sung Song
- Department of Biomedical Sciences, College of Medicine, Ulsan University, Asan Medical Center, Seoul, 138-736, Republic of Korea
| | - Yu Jeong Jeong
- Department of Biomedical Sciences, College of Medicine, Ulsan University, Asan Medical Center, Seoul, 138-736, Republic of Korea
| | - Yena Jung
- Futuristic Animal Resource &Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungchenongbuk-do, 28116, Republic of Korea
| | - Young-Ho Park
- Futuristic Animal Resource &Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungchenongbuk-do, 28116, Republic of Korea; Department of Functional Genomics, KRIBB, School of Bioscience, Korea University of Science and Technology, Republic of Korea
| | - Sungbo Shim
- Department of Biochemistry, College of Natural Sciences, Chungbuk National University, Cheongju, Republic of Korea
| | - Sung Joo Kim
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 138-736, Republic of Korea
| | - Dae-Woon Eom
- Department of Pathology, Gangneung Asan Hospital, Gangneung, Republic of Korea
| | - Seung-Mo Hong
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 138-736, Republic of Korea
| | - Peter C W Lee
- Department of Biomedical Sciences, College of Medicine, Ulsan University, Asan Medical Center, Seoul, 138-736, Republic of Korea
| | - Sun-Uk Kim
- Futuristic Animal Resource &Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungchenongbuk-do, 28116, Republic of Korea; Department of Functional Genomics, KRIBB, School of Bioscience, Korea University of Science and Technology, Republic of Korea.
| | - Sung-Wuk Jang
- Department of Biomedical Sciences, College of Medicine, Ulsan University, Asan Medical Center, Seoul, 138-736, Republic of Korea; Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 138-736, Republic of Korea; Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul, 138-736, Republic of Korea.
| |
Collapse
|
63
|
Targeting Cancer Stem Cells by Dietary Agents: An Important Therapeutic Strategy against Human Malignancies. Int J Mol Sci 2021; 22:ijms222111669. [PMID: 34769099 PMCID: PMC8584029 DOI: 10.3390/ijms222111669] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/23/2021] [Accepted: 10/23/2021] [Indexed: 02/07/2023] Open
Abstract
As a multifactorial disease, treatment of cancer depends on understanding unique mechanisms involved in its progression. The cancer stem cells (CSCs) are responsible for tumor stemness and by enhancing colony formation, proliferation as well as metastasis, and these cells can also mediate resistance to therapy. Furthermore, the presence of CSCs leads to cancer recurrence and therefore their complete eradication can have immense therapeutic benefits. The present review focuses on targeting CSCs by natural products in cancer therapy. The growth and colony formation capacities of CSCs have been reported can be attenuated by the dietary agents. These compounds can induce apoptosis in CSCs and reduce tumor migration and invasion via EMT inhibition. A variety of molecular pathways including STAT3, Wnt/β-catenin, Sonic Hedgehog, Gli1 and NF-κB undergo down-regulation by dietary agents in suppressing CSC features. Upon exposure to natural agents, a significant decrease occurs in levels of CSC markers including CD44, CD133, ALDH1, Oct4 and Nanog to impair cancer stemness. Furthermore, CSC suppression by dietary agents can enhance sensitivity of tumors to chemotherapy and radiotherapy. In addition to in vitro studies, as well as experiments on the different preclinical models have shown capacity of natural products in suppressing cancer stemness. Furthermore, use of nanostructures for improving therapeutic impact of dietary agents is recommended to rapidly translate preclinical findings for clinical use.
Collapse
|
64
|
Chisari A, Golán I, Campisano S, Gélabert C, Moustakas A, Sancho P, Caja L. Glucose and Amino Acid Metabolic Dependencies Linked to Stemness and Metastasis in Different Aggressive Cancer Types. Front Pharmacol 2021; 12:723798. [PMID: 34588983 PMCID: PMC8473699 DOI: 10.3389/fphar.2021.723798] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 08/20/2021] [Indexed: 12/26/2022] Open
Abstract
Malignant cells are commonly characterised by being capable of invading tissue, growing self-sufficiently and uncontrollably, being insensitive to apoptosis induction and controlling their environment, for example inducing angiogenesis. Amongst them, a subpopulation of cancer cells, called cancer stem cells (CSCs) shows sustained replicative potential, tumor-initiating properties and chemoresistance. These characteristics make CSCs responsible for therapy resistance, tumor relapse and growth in distant organs, causing metastatic dissemination. For these reasons, eliminating CSCs is necessary in order to achieve long-term survival of cancer patients. New insights in cancer metabolism have revealed that cellular metabolism in tumors is highly heterogeneous and that CSCs show specific metabolic traits supporting their unique functionality. Indeed, CSCs adapt differently to the deprivation of specific nutrients that represent potentially targetable vulnerabilities. This review focuses on three of the most aggressive tumor types: pancreatic ductal adenocarcinoma (PDAC), hepatocellular carcinoma (HCC) and glioblastoma (GBM). The aim is to prove whether CSCs from different tumour types share common metabolic requirements and responses to nutrient starvation, by outlining the diverse roles of glucose and amino acids within tumour cells and in the tumour microenvironment, as well as the consequences of their deprivation. Beyond their role in biosynthesis, they serve as energy sources and help maintain redox balance. In addition, glucose and amino acid derivatives contribute to immune responses linked to tumourigenesis and metastasis. Furthermore, potential metabolic liabilities are identified and discussed as targets for therapeutic intervention.
Collapse
Affiliation(s)
- Andrea Chisari
- Department of Chemistry, School of Sciences, National University of Mar del Plata, Mar del Plata, Argentina
| | - Irene Golán
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Biomedical Center, Uppsala University, Uppsala, Sweden
| | - Sabrina Campisano
- Department of Chemistry, School of Sciences, National University of Mar del Plata, Mar del Plata, Argentina
| | - Caroline Gélabert
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Biomedical Center, Uppsala University, Uppsala, Sweden
| | - Aristidis Moustakas
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Biomedical Center, Uppsala University, Uppsala, Sweden
| | - Patricia Sancho
- Translational Research Unit, Hospital Universitario Miguel Servet, IIS Aragon, Zaragoza, Spain
| | - Laia Caja
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Biomedical Center, Uppsala University, Uppsala, Sweden
| |
Collapse
|
65
|
Lenis-Rojas OA, Roma-Rodrigues C, Fernandes AR, Carvalho A, Cordeiro S, Guerra-Varela J, Sánchez L, Vázquez-García D, López-Torres M, Fernández A, Fernández JJ. Evaluation of the In Vitro and In Vivo Efficacy of Ruthenium Polypyridyl Compounds against Breast Cancer. Int J Mol Sci 2021; 22:ijms22168916. [PMID: 34445620 PMCID: PMC8396206 DOI: 10.3390/ijms22168916] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 07/31/2021] [Accepted: 08/10/2021] [Indexed: 11/16/2022] Open
Abstract
The clinical success of cisplatin, carboplatin, and oxaliplatin has sparked the interest of medicinal inorganic chemistry to synthesize and study compounds with non-platinum metal centers. Despite Ru(II)-polypyridyl complexes being widely studied and well established for their antitumor properties, there are not enough in vivo studies to establish the potentiality of this type of compound. Therefore, we report to the best of our knowledge the first in vivo study of Ru(II)-polypyridyl complexes against breast cancer with promising results. In order to conduct our study, we used MCF7 zebrafish xenografts and ruthenium complexes [Ru(bipy)2(C12H8N6-N,N)][CF3SO3]2Ru1 and [{Ru(bipy)2}2(μ-C12H8N6-N,N)][CF3SO3]4Ru2, which were recently developed by our group. Ru1 and Ru2 reduced the tumor size by an average of 30% without causing significant signs of lethality when administered at low doses of 1.25 mg·L-1. Moreover, the in vitro selectivity results were confirmed in vivo against MCF7 breast cancer cells. Surprisingly, this work suggests that both the mono- and the dinuclear Ru(II)-polypyridyl compounds have in vivo potential against breast cancer, since there were no significant differences between both treatments, highlighting Ru1 and Ru2 as promising chemotherapy agents in breast cancer therapy.
Collapse
Affiliation(s)
- Oscar A. Lenis-Rojas
- Instituto de Tecnologia Química e Biológica António Xavier, ITQB, Av. da República, EAN, 2780-157 Oeiras, Portugal;
| | - Catarina Roma-Rodrigues
- UCIBIO, Departamento Ciências da Vida, NOVA School of Science and Technology, Universidade Nova de Lisboa, Campus Caparica, 2829-516 Caparica, Portugal; (C.R.-R.); (A.C.); (S.C.)
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal
| | - Alexandra R. Fernandes
- UCIBIO, Departamento Ciências da Vida, NOVA School of Science and Technology, Universidade Nova de Lisboa, Campus Caparica, 2829-516 Caparica, Portugal; (C.R.-R.); (A.C.); (S.C.)
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal
- Correspondence: (A.R.F.); (J.J.F.)
| | - Andreia Carvalho
- UCIBIO, Departamento Ciências da Vida, NOVA School of Science and Technology, Universidade Nova de Lisboa, Campus Caparica, 2829-516 Caparica, Portugal; (C.R.-R.); (A.C.); (S.C.)
| | - Sandra Cordeiro
- UCIBIO, Departamento Ciências da Vida, NOVA School of Science and Technology, Universidade Nova de Lisboa, Campus Caparica, 2829-516 Caparica, Portugal; (C.R.-R.); (A.C.); (S.C.)
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal
| | - Jorge Guerra-Varela
- Departamento de Zoología, Genética y Antropología Física. Facultad de Veterinaria, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (J.G.-V.); (L.S.)
| | - Laura Sánchez
- Departamento de Zoología, Genética y Antropología Física. Facultad de Veterinaria, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (J.G.-V.); (L.S.)
- Preclinical Animal Models Group, Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Digna Vázquez-García
- Departamento de Química, Centro de Investigaciones Científicas Avanzadas (CICA), Universidade da Coruña, 15008 A Coruña, Spain; (D.V.-G.); (M.L.-T.); (A.F.)
| | - Margarita López-Torres
- Departamento de Química, Centro de Investigaciones Científicas Avanzadas (CICA), Universidade da Coruña, 15008 A Coruña, Spain; (D.V.-G.); (M.L.-T.); (A.F.)
| | - Alberto Fernández
- Departamento de Química, Centro de Investigaciones Científicas Avanzadas (CICA), Universidade da Coruña, 15008 A Coruña, Spain; (D.V.-G.); (M.L.-T.); (A.F.)
| | - Jesús J. Fernández
- Departamento de Química, Centro de Investigaciones Científicas Avanzadas (CICA), Universidade da Coruña, 15008 A Coruña, Spain; (D.V.-G.); (M.L.-T.); (A.F.)
- Correspondence: (A.R.F.); (J.J.F.)
| |
Collapse
|
66
|
Truong VNP, Nguyen YTK, Cho SK. Ampelopsin Suppresses Stem Cell Properties Accompanied by Attenuation of Oxidative Phosphorylation in Chemo- and Radio-Resistant MDA-MB-231 Breast Cancer Cells. Pharmaceuticals (Basel) 2021; 14:ph14080794. [PMID: 34451892 PMCID: PMC8400665 DOI: 10.3390/ph14080794] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/30/2021] [Accepted: 08/06/2021] [Indexed: 12/17/2022] Open
Abstract
Ampelopsin, also known as dihydromyricetin, is a commonly found flavonoid in medicinal plants. The cancer stem cell (CSC) population is a promising target for triple-negative breast cancer (TNBC). In this study, flavonoid screening was performed in the established MDA-MB-231/IR cell line, which is enriched in CSCs. Ampelopsin suppressed the proliferation and colony formation of stem cell-rich MDA-MB-231/IR, while inducing their apoptosis. Importantly, ampelopsin displayed an inhibitory impact on the stemness features of MDA-MB-231/IR cells, demonstrated by decreases in mammosphere formation, the CD44+/CD24-/low population, aldehyde dehydrogenase activity, and the levels of stem cell markers (e.g., CD44, MRP1, β-catenin, and KLF4). Ampelopsin also suppressed the epithelial-mesenchymal transition, as evidenced by decreases in migration, invasion capacity, and mesenchymal markers, as well as an increase in the epithelial marker E-cadherin. Moreover, ampelopsin significantly impaired oxidative phosphorylation by reducing the oxygen consumption rate and adenosine triphosphate production in MDA-MB-231/IR cells. Notably, ampelopsin treatment significantly reduced the levels of the phosphorylated forms of IκBα and NF-κB p65, as well as the levels of tumor necrosis factor (TNF)-α-stimulated phosphorylation of IκBα and NF-κB p65. These results demonstrated that ampelopsin prevents the TNF-α/NF-κB signaling axis in breast CSCs.
Collapse
Affiliation(s)
- Vi Nguyen-Phuong Truong
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju 63243, Korea; (V.N.-P.T.); (Y.T.-K.N.)
| | - Yen Thi-Kim Nguyen
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju 63243, Korea; (V.N.-P.T.); (Y.T.-K.N.)
| | - Somi-Kim Cho
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju 63243, Korea; (V.N.-P.T.); (Y.T.-K.N.)
- Subtropical/Tropical Organism Gene Bank, Jeju National University, Jeju 63243, Korea
- Correspondence: ; Tel.: +82-10-8660-1842
| |
Collapse
|
67
|
Shen YA, Chen CC, Chen BJ, Wu YT, Juan JR, Chen LY, Teng YC, Wei YH. Potential Therapies Targeting Metabolic Pathways in Cancer Stem Cells. Cells 2021; 10:1772. [PMID: 34359941 PMCID: PMC8304173 DOI: 10.3390/cells10071772] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 07/06/2021] [Accepted: 07/09/2021] [Indexed: 02/06/2023] Open
Abstract
Cancer stem cells (CSCs) are heterogeneous cells with stem cell-like properties that are responsible for therapeutic resistance, recurrence, and metastasis, and are the major cause for cancer treatment failure. Since CSCs have distinct metabolic characteristics that plays an important role in cancer development and progression, targeting metabolic pathways of CSCs appears to be a promising therapeutic approach for cancer treatment. Here we classify and discuss the unique metabolisms that CSCs rely on for energy production and survival, including mitochondrial respiration, glycolysis, glutaminolysis, and fatty acid metabolism. Because of metabolic plasticity, CSCs can switch between these metabolisms to acquire energy for tumor progression in different microenvironments compare to the rest of tumor bulk. Thus, we highlight the specific conditions and factors that promote or suppress CSCs properties to portray distinct metabolic phenotypes that attribute to CSCs in common cancers. Identification and characterization of the features in these metabolisms can offer new anticancer opportunities and improve the prognosis of cancer. However, the therapeutic window of metabolic inhibitors used alone or in combination may be rather narrow due to cytotoxicity to normal cells. In this review, we present current findings of potential targets in these four metabolic pathways for the development of more effective and alternative strategies to eradicate CSCs and treat cancer more effectively in the future.
Collapse
Affiliation(s)
- Yao-An Shen
- Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (Y.-A.S.); (C.-C.C.); (J.-R.J.); (L.-Y.C.); (Y.-C.T.)
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- International Master/Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Chang-Cyuan Chen
- Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (Y.-A.S.); (C.-C.C.); (J.-R.J.); (L.-Y.C.); (Y.-C.T.)
| | - Bo-Jung Chen
- Department of Pathology, Shuang-Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan;
| | - Yu-Ting Wu
- Center for Mitochondrial Medicine and Free Radical Research, Changhua Christian Hospital, Changhua City 50046, Taiwan;
| | - Jiun-Ru Juan
- Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (Y.-A.S.); (C.-C.C.); (J.-R.J.); (L.-Y.C.); (Y.-C.T.)
| | - Liang-Yun Chen
- Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (Y.-A.S.); (C.-C.C.); (J.-R.J.); (L.-Y.C.); (Y.-C.T.)
| | - Yueh-Chun Teng
- Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (Y.-A.S.); (C.-C.C.); (J.-R.J.); (L.-Y.C.); (Y.-C.T.)
| | - Yau-Huei Wei
- Center for Mitochondrial Medicine and Free Radical Research, Changhua Christian Hospital, Changhua City 50046, Taiwan;
| |
Collapse
|
68
|
Luna-Yolba R, Marmoiton J, Gigo V, Marechal X, Boet E, Sahal A, Alet N, Abramovich I, Gottlieb E, Visentin V, Paillasse MR, Sarry JE. Disrupting Mitochondrial Electron Transfer Chain Complex I Decreases Immune Checkpoints in Murine and Human Acute Myeloid Leukemic Cells. Cancers (Basel) 2021; 13:3499. [PMID: 34298712 PMCID: PMC8306173 DOI: 10.3390/cancers13143499] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 07/07/2021] [Accepted: 07/08/2021] [Indexed: 12/05/2022] Open
Abstract
Oxidative metabolism is crucial for leukemic stem cell (LSC) function and drug resistance in acute myeloid leukemia (AML). Mitochondrial metabolism also affects the immune system and therefore the anti-tumor response. The modulation of oxidative phosphorylation (OxPHOS) has emerged as a promising approach to improve the therapy outcome for AML patients. However, the effect of mitochondrial inhibitors on the immune compartment in the context of AML is yet to be explored. Immune checkpoints such as ectonucleotidase CD39 and programmed dead ligand 1 (PD-L1) have been reported to be expressed in AML and linked to chemo-resistance and a poor prognosis. In the present study, we first demonstrated that a novel selective electron transfer chain complex (ETC) I inhibitor, EVT-701, decreased the OxPHOS metabolism of murine and human cytarabine (AraC)-resistant leukemic cell lines. Furthermore, we showed that while AraC induced an immune response regulation by increasing CD39 expression and by reinforcing the interferon-γ/PD-L1 axis, EVT-701 reduced CD39 and PD-L1 expression in vitro in a panel of both murine and human AML cell lines, especially upon AraC treatment. Altogether, this work uncovers a non-canonical function of ETCI in controlling CD39 and PD-L1 immune checkpoints, thereby improving the anti-tumor response in AML.
Collapse
Affiliation(s)
- Raquel Luna-Yolba
- EVOTEC, Campus Curie, 31100 Toulouse, France; (R.L.-Y.); (J.M.); (V.G.); (X.M.); (N.A.); (V.V.)
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, CNRS, 31100 Toulouse, France; (E.B.); (A.S.)
- LabEx Toucan, 31100 Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, 31100 Toulouse, France
| | - Justine Marmoiton
- EVOTEC, Campus Curie, 31100 Toulouse, France; (R.L.-Y.); (J.M.); (V.G.); (X.M.); (N.A.); (V.V.)
| | - Véronique Gigo
- EVOTEC, Campus Curie, 31100 Toulouse, France; (R.L.-Y.); (J.M.); (V.G.); (X.M.); (N.A.); (V.V.)
| | - Xavier Marechal
- EVOTEC, Campus Curie, 31100 Toulouse, France; (R.L.-Y.); (J.M.); (V.G.); (X.M.); (N.A.); (V.V.)
| | - Emeline Boet
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, CNRS, 31100 Toulouse, France; (E.B.); (A.S.)
- LabEx Toucan, 31100 Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, 31100 Toulouse, France
| | - Ambrine Sahal
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, CNRS, 31100 Toulouse, France; (E.B.); (A.S.)
- LabEx Toucan, 31100 Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, 31100 Toulouse, France
| | - Nathalie Alet
- EVOTEC, Campus Curie, 31100 Toulouse, France; (R.L.-Y.); (J.M.); (V.G.); (X.M.); (N.A.); (V.V.)
| | - Ifat Abramovich
- Technion—Israel Institute of Technology, Haifa 32000, Israel; (I.A.); (E.G.)
| | - Eyal Gottlieb
- Technion—Israel Institute of Technology, Haifa 32000, Israel; (I.A.); (E.G.)
| | - Virgile Visentin
- EVOTEC, Campus Curie, 31100 Toulouse, France; (R.L.-Y.); (J.M.); (V.G.); (X.M.); (N.A.); (V.V.)
| | - Michael R. Paillasse
- EVOTEC, Campus Curie, 31100 Toulouse, France; (R.L.-Y.); (J.M.); (V.G.); (X.M.); (N.A.); (V.V.)
| | - Jean-Emmanuel Sarry
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, CNRS, 31100 Toulouse, France; (E.B.); (A.S.)
- LabEx Toucan, 31100 Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, 31100 Toulouse, France
| |
Collapse
|
69
|
Abstract
Cancer stem cells (CSCs) are heterogeneous cells with stem cell-like properties that are responsible for therapeutic resistance, recurrence, and metastasis, and are the major cause for cancer treatment failure. Since CSCs have distinct metabolic characteristics that plays an important role in cancer development and progression, targeting metabolic pathways of CSCs appears to be a promising therapeutic approach for cancer treatment. Here we classify and discuss the unique metabolisms that CSCs rely on for energy production and survival, including mitochondrial respiration, glycolysis, glutaminolysis, and fatty acid metabolism. Because of metabolic plasticity, CSCs can switch between these metabolisms to acquire energy for tumor progression in different microenvironments compare to the rest of tumor bulk. Thus, we highlight the specific conditions and factors that promote or suppress CSCs properties to portray distinct metabolic phenotypes that attribute to CSCs in common cancers. Identification and characterization of the features in these metabolisms can offer new anticancer opportunities and improve the prognosis of cancer. However, the therapeutic window of metabolic inhibitors used alone or in combination may be rather narrow due to cytotoxicity to normal cells. In this review, we present current findings of potential targets in these four metabolic pathways for the development of more effective and alternative strategies to eradicate CSCs and treat cancer more effectively in the future.
Collapse
|
70
|
Pensado-López A, Fernández-Rey J, Reimunde P, Crecente-Campo J, Sánchez L, Torres Andón F. Zebrafish Models for the Safety and Therapeutic Testing of Nanoparticles with a Focus on Macrophages. NANOMATERIALS 2021; 11:nano11071784. [PMID: 34361170 PMCID: PMC8308170 DOI: 10.3390/nano11071784] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/05/2021] [Accepted: 07/07/2021] [Indexed: 12/11/2022]
Abstract
New nanoparticles and biomaterials are increasingly being used in biomedical research for drug delivery, diagnostic applications, or vaccines, and they are also present in numerous commercial products, in the environment and workplaces. Thus, the evaluation of the safety and possible therapeutic application of these nanomaterials has become of foremost importance for the proper progress of nanotechnology. Due to economical and ethical issues, in vitro and in vivo methods are encouraged for the testing of new compounds and/or nanoparticles, however in vivo models are still needed. In this scenario, zebrafish (Danio rerio) has demonstrated potential for toxicological and pharmacological screenings. Zebrafish presents an innate immune system, from early developmental stages, with conserved macrophage phenotypes and functions with respect to humans. This fact, combined with the transparency of zebrafish, the availability of models with fluorescently labelled macrophages, as well as a broad variety of disease models offers great possibilities for the testing of new nanoparticles. Thus, with a particular focus on macrophage-nanoparticle interaction in vivo, here, we review the studies using zebrafish for toxicological and biodistribution testing of nanoparticles, and also the possibilities for their preclinical evaluation in various diseases, including cancer and autoimmune, neuroinflammatory, and infectious diseases.
Collapse
Affiliation(s)
- Alba Pensado-López
- Department of Zoology, Genetics and Physical Anthropology, Campus de Lugo, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (A.P.-L.); (J.F.-R.)
- Center for Research in Molecular Medicine & Chronic Diseases (CIMUS), Campus Vida, Universidade de Santiago de Compostela, 15706 Santiago de Compostela, Spain;
| | - Juan Fernández-Rey
- Department of Zoology, Genetics and Physical Anthropology, Campus de Lugo, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (A.P.-L.); (J.F.-R.)
- Center for Research in Molecular Medicine & Chronic Diseases (CIMUS), Campus Vida, Universidade de Santiago de Compostela, 15706 Santiago de Compostela, Spain;
| | - Pedro Reimunde
- Department of Physiotherapy, Medicine and Biomedical Sciences, Universidade da Coruña, Campus de Oza, 15006 A Coruña, Spain;
- Department of Neurosurgery, Hospital Universitario Lucus Augusti, 27003 Lugo, Spain
| | - José Crecente-Campo
- Center for Research in Molecular Medicine & Chronic Diseases (CIMUS), Campus Vida, Universidade de Santiago de Compostela, 15706 Santiago de Compostela, Spain;
| | - Laura Sánchez
- Department of Zoology, Genetics and Physical Anthropology, Campus de Lugo, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (A.P.-L.); (J.F.-R.)
- Correspondence: (L.S.); (F.T.A.)
| | - Fernando Torres Andón
- Center for Research in Molecular Medicine & Chronic Diseases (CIMUS), Campus Vida, Universidade de Santiago de Compostela, 15706 Santiago de Compostela, Spain;
- Correspondence: (L.S.); (F.T.A.)
| |
Collapse
|
71
|
Tuerhong A, Xu J, Shi S, Tan Z, Meng Q, Hua J, Liu J, Zhang B, Wang W, Yu X, Liang C. Overcoming chemoresistance by targeting reprogrammed metabolism: the Achilles' heel of pancreatic ductal adenocarcinoma. Cell Mol Life Sci 2021; 78:5505-5526. [PMID: 34131808 PMCID: PMC11072422 DOI: 10.1007/s00018-021-03866-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 05/04/2021] [Accepted: 05/27/2021] [Indexed: 02/07/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the leading causes of cancer-related death due to its late diagnosis that removes the opportunity for surgery and metabolic plasticity that leads to resistance to chemotherapy. Metabolic reprogramming related to glucose, lipid, and amino acid metabolism in PDAC not only enables the cancer to thrive and survive under hypovascular, nutrient-poor and hypoxic microenvironments, but also confers chemoresistance, which contributes to the poor prognosis of PDAC. In this review, we systematically elucidate the mechanism of chemotherapy resistance and the relationship of metabolic programming features with resistance to anticancer drugs in PDAC. Targeting the critical enzymes and/or transporters involved in glucose, lipid, and amino acid metabolism may be a promising approach to overcome chemoresistance in PDAC. Consequently, regulating metabolism could be used as a strategy against PDAC and could improve the prognosis of PDAC.
Collapse
Affiliation(s)
- Abudureyimu Tuerhong
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 270 Dong'An Road, Shanghai, 200032, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, People's Republic of China
| | - Jin Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 270 Dong'An Road, Shanghai, 200032, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, People's Republic of China
| | - Si Shi
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 270 Dong'An Road, Shanghai, 200032, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, People's Republic of China
| | - Zhen Tan
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 270 Dong'An Road, Shanghai, 200032, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, People's Republic of China
| | - Qingcai Meng
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 270 Dong'An Road, Shanghai, 200032, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, People's Republic of China
| | - Jie Hua
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 270 Dong'An Road, Shanghai, 200032, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, People's Republic of China
| | - Jiang Liu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 270 Dong'An Road, Shanghai, 200032, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, People's Republic of China
| | - Bo Zhang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 270 Dong'An Road, Shanghai, 200032, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, People's Republic of China
| | - Wei Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 270 Dong'An Road, Shanghai, 200032, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, People's Republic of China
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 270 Dong'An Road, Shanghai, 200032, People's Republic of China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, People's Republic of China.
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, People's Republic of China.
| | - Chen Liang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 270 Dong'An Road, Shanghai, 200032, People's Republic of China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, People's Republic of China.
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, People's Republic of China.
| |
Collapse
|
72
|
Integrated lipidomics and proteomics reveal cardiolipin alterations, upregulation of HADHA and long chain fatty acids in pancreatic cancer stem cells. Sci Rep 2021; 11:13297. [PMID: 34168259 PMCID: PMC8225828 DOI: 10.1038/s41598-021-92752-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 06/15/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer stem cells (PCSCs) play a key role in the aggressiveness of pancreatic ductal adenocarcinomas (PDAC); however, little is known about their signaling and metabolic pathways. Here we show that PCSCs have specific and common proteome and lipidome modulations. PCSCs displayed downregulation of lactate dehydrogenase A chain, and upregulation of trifunctional enzyme subunit alpha. The upregulated proteins of PCSCs are mainly involved in fatty acid (FA) elongation and biosynthesis of unsaturated FAs. Accordingly, lipidomics reveals an increase in long and very long-chain unsaturated FAs, which are products of fatty acid elongase-5 predicted as a key gene. Moreover, lipidomics showed the induction in PCSCs of molecular species of cardiolipin with mixed incorporation of 16:0, 18:1, and 18:2 acyl chains. Our data indicate a crucial role of FA elongation and alteration in cardiolipin acyl chain composition in PCSCs, representing attractive therapeutic targets in PDAC.
Collapse
|
73
|
Yasuda T, Ishimoto T, Baba H. Conflicting metabolic alterations in cancer stem cells and regulation by the stromal niche. Regen Ther 2021; 17:8-12. [PMID: 33598509 PMCID: PMC7851492 DOI: 10.1016/j.reth.2021.01.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 01/06/2021] [Indexed: 12/22/2022] Open
Abstract
Recent studies have revealed that cancer stem cells (CSCs) undergo metabolic alterations that differentiate them from non-CSCs. Inhibition of specific metabolic pathways in CSCs has been conducted to eliminate the CSC population in many types of cancer. However, there is conflicting evidence about whether CSCs depend on glycolysis or mitochondrial oxidative phosphorylation (OXPHOS) to maintain their stem cell properties. This review summarizes the latest knowledge regarding CSC-specific metabolic alterations and offers recent evidence that the surrounding microenvironments may play an important role in the maintenance of CSC properties.
Collapse
Key Words
- ALDH, aldehyde dehydrogenase
- ATP, adenosine triphosphate
- CD44v, CD44 variant isoform
- CSCs
- CSCs, cancer stem cells
- EMT, epithelial–mesenchymal transition
- EVs, extracellular vesicles
- FAO, fatty acid oxidation
- FBP1, fructose-1,6-biphosphatase 1
- GLUT1, glucose transporter 1
- GP6, glucose-6-phosphate
- Glycolysis
- HCC, hepatocellular carcinoma
- HIF1a, hypoxia inducible factor 1a
- IMP2, insulin-like growth factor 2
- IncRNAs, long noncoding RNAs
- LSCs, leukemia stem cells
- Mitochondrial OXPHOS
- NRF2, nuclear factor erythroid 2–related factor 2
- OXPHOS, oxidative phosphorylation
- PDK1, pyruvate dehydrogenase kinase 1
- PPP, pentose phosphate pathway
- ROS
- ROS, reactive oxygen species
- SOD2, superoxide dismutase 2
- Stromal niche
- TCA, tricarboxylic acid
- TICs, tumor initiating stem-like cells
- mTORC1, mammalian target of rapamycin complex 1
Collapse
Affiliation(s)
- Tadahito Yasuda
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
- Gastrointestinal Cancer Biology, International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan
| | - Takatsugu Ishimoto
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
- Gastrointestinal Cancer Biology, International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
- Center for Metabolic Regulation of Healthy Aging, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
74
|
Morein D, Rubinstein-Achiasaf L, Brayer H, Dorot O, Pichinuk E, Ben-Yaakov H, Meshel T, Pasmanik-Chor M, Ben-Baruch A. Continuous Inflammatory Stimulation Leads via Metabolic Plasticity to a Prometastatic Phenotype in Triple-Negative Breast Cancer Cells. Cells 2021; 10:cells10061356. [PMID: 34072893 PMCID: PMC8229065 DOI: 10.3390/cells10061356] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/23/2021] [Accepted: 05/28/2021] [Indexed: 12/14/2022] Open
Abstract
Chronic inflammation promotes cancer progression by affecting the tumor cells and their microenvironment. Here, we demonstrate that a continuous stimulation (~6 weeks) of triple-negative breast tumor cells (TNBC) by the proinflammatory cytokines tumor necrosis factor α (TNFα) + interleukin 1β (IL-1β) changed the expression of hundreds of genes, skewing the cells towards a proinflammatory phenotype. While not affecting stemness, the continuous TNFα + IL-1β stimulation has increased tumor cell dispersion and has induced a hybrid metabolic phenotype in TNBC cells; this phenotype was indicated by a transcription-independent elevation in glycolytic activity and by increased mitochondrial respiratory potential (OXPHOS) of TNBC cells, accompanied by elevated transcription of mitochondria-encoded OXPHOS genes and of active mitochondria area. The continuous TNFα + IL-1β stimulation has promoted in a glycolysis-dependent manner the activation of p65 (NF-κB), and the transcription and protein expression of the prometastatic and proinflammatory mediators sICAM-1, CCL2, CXCL8 and CXCL1. Moreover, when TNBC cells were stimulated continuously by TNFα + IL-1β in the presence of a glycolysis inhibitor, their conditioned media had reduced ability to recruit monocytes and neutrophils in vivo. Such inflammation-induced metabolic plasticity, which promotes prometastatic cascades in TNBC, may have important clinical implications in treatment of TNBC patients.
Collapse
Affiliation(s)
- Dina Morein
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel; (D.M.); (L.R.-A.); (H.B.); (H.B.-Y.); (T.M.)
| | - Linor Rubinstein-Achiasaf
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel; (D.M.); (L.R.-A.); (H.B.); (H.B.-Y.); (T.M.)
| | - Hadar Brayer
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel; (D.M.); (L.R.-A.); (H.B.); (H.B.-Y.); (T.M.)
| | - Orly Dorot
- Blavatnik Center for Drug Discovery, Tel Aviv University, Tel Aviv 6997801, Israel; (O.D.); (E.P.)
| | - Edward Pichinuk
- Blavatnik Center for Drug Discovery, Tel Aviv University, Tel Aviv 6997801, Israel; (O.D.); (E.P.)
| | - Hagar Ben-Yaakov
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel; (D.M.); (L.R.-A.); (H.B.); (H.B.-Y.); (T.M.)
| | - Tsipi Meshel
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel; (D.M.); (L.R.-A.); (H.B.); (H.B.-Y.); (T.M.)
| | - Metsada Pasmanik-Chor
- Bioinformatics Unit, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel;
| | - Adit Ben-Baruch
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel; (D.M.); (L.R.-A.); (H.B.); (H.B.-Y.); (T.M.)
- Correspondence: ; Tel.: +972-3-6405491; Fax: +972-3-6422046
| |
Collapse
|
75
|
Wu Y, Zeng H, Yu Q, Huang H, Fervers B, Chen ZS, Lu L. A Circulating Exosome RNA Signature Is a Potential Diagnostic Marker for Pancreatic Cancer, a Systematic Study. Cancers (Basel) 2021; 13:cancers13112565. [PMID: 34073722 PMCID: PMC8197236 DOI: 10.3390/cancers13112565] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 05/19/2021] [Indexed: 12/16/2022] Open
Abstract
Simple Summary Most patients with pancreatic cancer are diagnosed at an advanced stage due to the lack of tools with high sensitivity and specificity for early detection. Aberrant gene expression occurs in pancreatic cancer, which can be packaged into nanoparticles (also known as exosomes or nano-sized extracellular vesicles) and then released into blood. In this study, we aimed to evaluate the diagnostic value of a circulating exosome RNA signature in pancreatic cancer. Our findings indicate that the circulating exosome RNA signature is a potential marker for the early detection or diagnosis of pancreatic cancer. Abstract Several exosome proteins, miRNAs and KRAS mutations have been investigated in the hope of carrying out the early detection of pancreatic cancer with high sensitivity and specificity, but they have proven to be insufficient. Exosome RNAs, however, have not been extensively evaluated in the diagnosis of pancreatic cancer. The purpose of this study was to investigate the potential of circulating exosome RNAs in pancreatic cancer detection. By retrieving RNA-seq data from publicly accessed databases, differential expression and random-effects meta-analyses were performed. The results showed that pancreatic cancer had a distinct circulating exosome RNA signature in healthy individuals, and that the top 10 candidate exosome RNAs could distinguish patients from healthy individuals with an area under the curve (AUC) of 1.0. Three (HIST2H2AA3, LUZP6 and HLA-DRA) of the 10 genes in exosomes had similar differential patterns to those in tumor tissues based on RNA-seq data. In the validation dataset, the levels of these three genes in exosomes displayed good performance in distinguishing cancer from both chronic pancreatitis (AUC = 0.815) and healthy controls (AUC = 0.8558), whereas a slight difference existed between chronic pancreatitis and healthy controls (AUC = 0.586). Of the three genes, the level of HIST2H2AA3 was positively associated with KRAS status. However, there was no significant difference in the levels of the three genes across the disease stages (stages I–IV). These findings indicate that circulating exosome RNAs have a potential early detection value in pancreatic cancer, and that a distinct exosome RNA signature exists in distinguishing pancreatic cancer from healthy individuals.
Collapse
Affiliation(s)
- Yixing Wu
- Department of Endocrinology, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China;
| | - Hongmei Zeng
- National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China;
| | - Qing Yu
- Center for Cancer and Blood Disorders, Children’s National Medical Center, Washington, DC 20010, USA;
| | - Huatian Huang
- Department of Imaging, Guizhou Qianxinan People’s Hospital, Xingyi 652400, China;
| | - Beatrice Fervers
- Département Prévention Cancer Environnement, Centre Léon Bérard—Université Lyon 1, 69008 Lyon, France;
- UMR Inserm 1296 “Radiations: Défense, Santé, Environnement”, Centre Léon Bérard, 69008 Lyon, France
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, St. John’s University, New York, NY 11439, USA;
| | - Lingeng Lu
- Department of Chronic Disease Epidemiology, Yale School of Public Health, School of Medicine, New Haven, CT 06520, USA
- Center for Biomedical Data Science, Yale University, 60 College Street, New Haven, CT 06520, USA
- Yale Cancer Center, Yale University, 60 College Street, New Haven, CT 06520, USA
- Correspondence:
| |
Collapse
|
76
|
Pan H, Diao H, Zhong W, Wang T, Wen P, Wu C. A Cancer Cell Cluster Marked by LincRNA MEG3 Leads Pancreatic Ductal Adenocarcinoma Metastasis. Front Oncol 2021; 11:656564. [PMID: 34055623 PMCID: PMC8155708 DOI: 10.3389/fonc.2021.656564] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 04/19/2021] [Indexed: 12/25/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly devastating disease with poor prognosis and rising incidence worldwide. Late detection and particularly aggressive characteristics are the major challenges that lead to therapeutic failure of this disease. A well described gene program and core regulators are yet to be discovered to drive the metastasis of the PDAC cells. As the development of single cell omics technologies including single cell RNA-sequencing (scRNA-seq), detailed characterization of the cellular composition of solid tumors and their microenvironments are well elaborated. In the current study, we accessed a recently published scRNA-seq dataset on primary and metastatic PDAC tissues and subset the tumor cells. By comparative analysis, we profiled the differentially expressed gene programs of primary and metastatic PDAC and found several long intergenic non-coding RNAs (LincRNAs) in top genes. The PDAC cancer cells showed some heterogeneity and were divided into four major subclusters based on gene profiles, one of which was mostly contributed by metastatic PDAC. Interestingly, this subcluster was remarkably marked by one of the above LincRNAs, MEG3, and exhibited significantly increased Epithelial-Mesenchymal-Transition (EMT) signatures. Ingenuity Pathway Analysis (IPA) on the signature genes of this subcluster gave multiple cancer metastasis associated and EMT signaling pathways, suggesting a critical role of this cluster in leading tumor cell metastasis. Taken together, this study displayed a PDAC cancer subcluster and its marker gene, biologically targeting of which might significantly attenuate the metastasis of tumor and might be a potential strategy for the therapeutic treatment of cancer.
Collapse
Affiliation(s)
| | | | | | | | | | - Chunli Wu
- Department of Radiation Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
77
|
Chen S, Yang C, Wang ZW, Hu JF, Pan JJ, Liao CY, Zhang JQ, Chen JZ, Huang Y, Huang L, Zhan Q, Tian YF, Shen BY, Wang YD. CLK1/SRSF5 pathway induces aberrant exon skipping of METTL14 and Cyclin L2 and promotes growth and metastasis of pancreatic cancer. J Hematol Oncol 2021; 14:60. [PMID: 33849617 PMCID: PMC8045197 DOI: 10.1186/s13045-021-01072-8] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 03/29/2021] [Indexed: 12/11/2022] Open
Abstract
Background Both aberrant alternative splicing and m6A methylation play complicated roles in the development of pancreatic cancer (PC), while the relationship between these two RNA modifications remains unclear. Methods RNA sequencing (RNA-seq) was performed using 15 pairs of pancreatic ductal adenocarcinoma (PDAC) tissues and corresponding normal tissues, and Cdc2-like kinases 1 (CLK1) was identified as a significantly upregulated alternative splicing related gene. Real-time quantitative PCR (qPCR) and western blotting were applied to determine the CLK1 levels. The prognostic value of CLK1 was elucidated by Immunohistochemistry (IHC) analyses in two independent PDAC cohorts. The functional characterizations and mechanistic insights of CLK1 in PDAC growth and metastasis were evaluated with PDAC cell lines and nude mice. SR-like splicing factors5250-Ser (SRSF5250-Ser) was identified as an important target phosphorylation site by phosphorylation mass spectrometry. Through transcriptome sequencing, Methyltransferase-like 14exon10 (METTL14exon10) and Cyclin L2exon6.3 skipping were identified as key alternative splicing events regulated by the CLK1-SRSF5 axis. RIP assays, RNA-pulldown and CLIP-qPCR were performed to confirm molecular interactions and the precise binding sites. The roles of the shift of METTL14exon 10 and Cyclin L2exon6.3 skipping were surveyed. Results CLK1 expression was significantly increased in PDAC tissues at both the mRNA and protein levels. High CLK1 expression was associated with poor prognosis. Elevated CLK1 expression promoted growth and metastasis of PC cells in vitro and in vivo. Mechanistically, CLK1 enhanced phosphorylation on SRSF5250-Ser, which inhibited METTL14exon10 skipping while promoted Cyclin L2exon6.3 skipping. In addition, aberrant METTL14exon 10 skipping enhanced the N6-methyladenosine modification level and metastasis, while aberrant Cyclin L2exon6.3 promoted proliferation of PDAC cells. Conclusions The CLK1/SRSF5 pathway induces aberrant exon skipping of METTL14 and Cyclin L2, which promotes growth and metastasis and regulates m6A methylation of PDAC cells. This study suggests the potential prognostic value and therapeutic targeting of this pathway in PDAC patients. Supplementary Information The online version contains supplementary material available at 10.1186/s13045-021-01072-8.
Collapse
Affiliation(s)
- Shi Chen
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, No. 134, East Street, Fuzhou, 350001, Fujian Province, People's Republic of China. .,Department of Hepatobiliary Pancreatic Surgery, Fujian Provincial Hospital, Fuzhou, 350001, People's Republic of China.
| | - Can Yang
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, No. 134, East Street, Fuzhou, 350001, Fujian Province, People's Republic of China
| | - Zu-Wei Wang
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, No. 134, East Street, Fuzhou, 350001, Fujian Province, People's Republic of China
| | - Jian-Fei Hu
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, No. 134, East Street, Fuzhou, 350001, Fujian Province, People's Republic of China
| | - Jing-Jing Pan
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, No. 134, East Street, Fuzhou, 350001, Fujian Province, People's Republic of China
| | - Cheng-Yu Liao
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, No. 134, East Street, Fuzhou, 350001, Fujian Province, People's Republic of China
| | - Jia-Qiang Zhang
- Department of General Surgery, Pancreatic Disease Center, Research Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197 Ruijin Second Road, Shanghai, 200025, People's Republic of China
| | - Jiang-Zhi Chen
- Department of Hepatobiliary Surgery, Union Hospital, Fujian Medical University, Fuzhou, 350001, People's Republic of China
| | - Yi Huang
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, No. 134, East Street, Fuzhou, 350001, Fujian Province, People's Republic of China.,Center for Experimental Research in Clinical Medicine, Fujian Provincial Hospital, Fuzhou, 350001, People's Republic of China
| | - Long Huang
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, No. 134, East Street, Fuzhou, 350001, Fujian Province, People's Republic of China.,Department of Hepatobiliary Pancreatic Surgery, Fujian Provincial Hospital, Fuzhou, 350001, People's Republic of China
| | - Qian Zhan
- Department of General Surgery, Pancreatic Disease Center, Research Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197 Ruijin Second Road, Shanghai, 200025, People's Republic of China
| | - Yi-Feng Tian
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, No. 134, East Street, Fuzhou, 350001, Fujian Province, People's Republic of China.,Department of Hepatobiliary Pancreatic Surgery, Fujian Provincial Hospital, Fuzhou, 350001, People's Republic of China
| | - Bai-Yong Shen
- Department of General Surgery, Pancreatic Disease Center, Research Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197 Ruijin Second Road, Shanghai, 200025, People's Republic of China.
| | - Yao-Dong Wang
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, No. 134, East Street, Fuzhou, 350001, Fujian Province, People's Republic of China. .,Department of Hepatobiliary Pancreatic Surgery, Fujian Provincial Hospital, Fuzhou, 350001, People's Republic of China.
| |
Collapse
|
78
|
Courtois S, de Luxán-Delgado B, Penin-Peyta L, Royo-García A, Parejo-Alonso B, Jagust P, Alcalá S, Rubiolo JA, Sánchez L, Sainz B, Heeschen C, Sancho P. Inhibition of Mitochondrial Dynamics Preferentially Targets Pancreatic Cancer Cells with Enhanced Tumorigenic and Invasive Potential. Cancers (Basel) 2021; 13:698. [PMID: 33572276 PMCID: PMC7914708 DOI: 10.3390/cancers13040698] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 01/31/2021] [Accepted: 02/05/2021] [Indexed: 12/11/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest tumors, partly due to its intrinsic aggressiveness, metastatic potential, and chemoresistance of the contained cancer stem cells (CSCs). Pancreatic CSCs strongly rely on mitochondrial metabolism to maintain their stemness, therefore representing a putative target for their elimination. Since mitochondrial homeostasis depends on the tightly controlled balance between fusion and fission processes, namely mitochondrial dynamics, we aim to study this mechanism in the context of stemness. In human PDAC tissues, the mitochondrial fission gene DNM1L (DRP1) was overexpressed and positively correlated with the stemness signature. Moreover, we observe that primary human CSCs display smaller mitochondria and a higher DRP1/MFN2 expression ratio, indicating the activation of the mitochondrial fission. Interestingly, treatment with the DRP1 inhibitor mDivi-1 induced dose-dependent apoptosis, especially in CD133+ CSCs, due to the accumulation of dysfunctional mitochondria and the subsequent energy crisis in this subpopulation. Mechanistically, mDivi-1 inhibited stemness-related features, such as self-renewal, tumorigenicity, and invasiveness and chemosensitized the cells to the cytotoxic effects of Gemcitabine. In summary, mitochondrial fission is an essential process for pancreatic CSCs and represents an attractive target for designing novel multimodal treatments that will more efficiently eliminate cells with high tumorigenic potential.
Collapse
Affiliation(s)
- Sarah Courtois
- Translational Research Unit, Hospital Universitario Miguel Servet, IIS Aragon, 50009 Zaragoza, Spain; (S.C.); (A.R.-G.); (B.P.-A.)
| | - Beatriz de Luxán-Delgado
- Centre for Stem Cells in Cancer & Ageing, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK; (B.d.L.-D.); (L.P.-P.); (P.J.)
| | - Laure Penin-Peyta
- Centre for Stem Cells in Cancer & Ageing, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK; (B.d.L.-D.); (L.P.-P.); (P.J.)
| | - Alba Royo-García
- Translational Research Unit, Hospital Universitario Miguel Servet, IIS Aragon, 50009 Zaragoza, Spain; (S.C.); (A.R.-G.); (B.P.-A.)
| | - Beatriz Parejo-Alonso
- Translational Research Unit, Hospital Universitario Miguel Servet, IIS Aragon, 50009 Zaragoza, Spain; (S.C.); (A.R.-G.); (B.P.-A.)
| | - Petra Jagust
- Centre for Stem Cells in Cancer & Ageing, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK; (B.d.L.-D.); (L.P.-P.); (P.J.)
| | - Sonia Alcalá
- Department of Biochemistry, School of Medicine, Instituto de Investigaciones Biomédicas (IIBm) “Alberto Sols” CSIC-UAM, Autónoma University of Madrid (UAM), 28029 Madrid, Spain; (S.A.); (B.S.J.)
- Chronic Diseases and Cancer, Area 3, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28029 Madrid, Spain
| | - Juan A. Rubiolo
- Department of Zoology, Genetics and Physical Anthropology, Veterinary Faculty, Universidad de Santiago de Compostela, 27002 Lugo, Spain; (J.A.R.); (L.S.)
| | - Laura Sánchez
- Department of Zoology, Genetics and Physical Anthropology, Veterinary Faculty, Universidad de Santiago de Compostela, 27002 Lugo, Spain; (J.A.R.); (L.S.)
| | - Bruno Sainz
- Department of Biochemistry, School of Medicine, Instituto de Investigaciones Biomédicas (IIBm) “Alberto Sols” CSIC-UAM, Autónoma University of Madrid (UAM), 28029 Madrid, Spain; (S.A.); (B.S.J.)
- Chronic Diseases and Cancer, Area 3, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28029 Madrid, Spain
| | - Christopher Heeschen
- Center for Single-Cell Omics, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Patricia Sancho
- Translational Research Unit, Hospital Universitario Miguel Servet, IIS Aragon, 50009 Zaragoza, Spain; (S.C.); (A.R.-G.); (B.P.-A.)
| |
Collapse
|