18
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Inhibition of NPC1L1 disrupts adaptive responses of drug‐tolerant persister cells to chemotherapy

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Entering a drug‐tolerant persister (DTP) state of cancer cells is a transient self‐adaptive mechanism by which a residual cell subpopulation accelerates tumor progression. Here, we identified the acquisition of a DTP phenotype in multidrug‐resistant (MDR) cancer cells as a tolerance response to routine combination treatment. Characterization of MDR cancer cells with a DTP state by RNA‐seq revealed that these cells partially prevented chemotherapy‐triggered oxidative stress by promoting NPC1L1‐regulated uptake of vitamin E. Treatment with the NPC1L1 inhibitor ezetimibe further enhanced the therapeutic effect of combinatorial therapy by inducing methuosis. Mechanistically, we demonstrated that NRF2 was involved in transcriptional regulation of NPC1L1 by binding to the −205 to −215 bp site on its promoter. Decreased DNA methylation was also related partially to this process. Furthermore, we confirmed that a triple‐combination of chemotherapeutic agents, verapamil, and ezetimibe, had a significant anti‐tumor effect and prevented tumor recurrence in mice. Together, our study provides a novel insight into the role of DTP state and emphasizes the importance of disrupting redox homeostasis during cancer therapy.

          Abstract

          Drug‐tolerant persister (DTP) state is a driver of therapy failure and cancer relapse. This study identified a key role for NPC1L1 in multidrug‐resistant (MDR) cancer cells with the DTP state, where NPC1L1 orchestrated a redox signaling against the harsh environment caused by cancer therapy.

          Related collections

          Most cited references49

          • Record: found
          • Abstract: found
          • Article: not found

          Targeting cancer cells by ROS-mediated mechanisms: a radical therapeutic approach?

          Increased generation of reactive oxygen species (ROS) and an altered redox status have long been observed in cancer cells, and recent studies suggest that this biochemical property of cancer cells can be exploited for therapeutic benefits. Cancer cells in advanced stage tumours frequently exhibit multiple genetic alterations and high oxidative stress, suggesting that it might be possible to preferentially eliminate these cells by pharmacological ROS insults. However, the upregulation of antioxidant capacity in adaptation to intrinsic oxidative stress in cancer cells can confer drug resistance. Abrogation of such drug-resistant mechanisms by redox modulation could have significant therapeutic implications. We argue that modulating the unique redox regulatory mechanisms of cancer cells might be an effective strategy to eliminate these cells.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: found

            Dependency of a therapy-resistant state of cancer cells on a lipid peroxidase pathway

            Plasticity of the cell state has been proposed to drive resistance to multiple classes of cancer therapies, thereby limiting their effectiveness. A high-mesenchymal cell state observed in human tumours and cancer cell lines has been associated with resistance to multiple treatment modalities across diverse cancer lineages, but the mechanistic underpinning for this state has remained incompletely understood. Here we molecularly characterize this therapy-resistant high-mesenchymal cell state in human cancer cell lines and organoids and show that it depends on a druggable lipid-peroxidase pathway that protects against ferroptosis, a non-apoptotic form of cell death induced by the build-up of toxic lipid peroxides. We show that this cell state is characterized by activity of enzymes that promote the synthesis of polyunsaturated lipids. These lipids are the substrates for lipid peroxidation by lipoxygenase enzymes. This lipid metabolism creates a dependency on pathways converging on the phospholipid glutathione peroxidase (GPX4), a selenocysteine-containing enzyme that dissipates lipid peroxides and thereby prevents the iron-mediated reactions of peroxides that induce ferroptotic cell death. Dependency on GPX4 was found to exist across diverse therapy-resistant states characterized by high expression of ZEB1, including epithelial–mesenchymal transition in epithelial-derived carcinomas, TGFβ-mediated therapy-resistance in melanoma, treatment-induced neuroendocrine transdifferentiation in prostate cancer, and sarcomas, which are fixed in a mesenchymal state owing to their cells of origin. We identify vulnerability to ferroptic cell death induced by inhibition of a lipid peroxidase pathway as a feature of therapy-resistant cancer cells across diverse mesenchymal cell-state contexts.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Oxidative Stress in Cancer

              Contingent upon concentration, reactive oxygen species (ROS) influence cancer evolution in apparently contradictory ways, either initiating/stimulating tumorigenesis and supporting transformation/proliferation of cancer cells or causing cell death. To accommodate high ROS levels, tumor cells modify sulfur-based metabolism, NADPH generation, and the activity of antioxidant transcription factors. During initiation, genetic changes enable cell survival under high ROS levels by activating antioxidant transcription factors or increasing NADPH via the pentose phosphate pathway (PPP). During progression and metastasis, tumor cells adapt to oxidative stress by increasing NADPH in various ways, including activation of AMPK, the PPP, and reductive glutamine and folate metabolism.
                Bookmark

                Author and article information

                Contributors
                hnchen@scu.edu.cn
                hcanhua@scu.edu.cn
                xiaweiwei@scu.edu.cn
                Journal
                EMBO Mol Med
                EMBO Mol Med
                10.1002/(ISSN)1757-4684
                EMMM
                embomm
                EMBO Molecular Medicine
                John Wiley and Sons Inc. (Hoboken )
                1757-4676
                1757-4684
                13 January 2022
                07 February 2022
                : 14
                : 2 ( doiID: 10.1002/emmm.v14.2 )
                : e14903
                Affiliations
                [ 1 ] Laboratory of Aging Research and Cancer Drug Target State Key Laboratory of Biotherapy and Cancer Center National Clinical Research Center for Geriatrics, West China Hospital Sichuan University Chengdu China
                [ 2 ] State Key Laboratory of Biotherapy and Cancer Center West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine Sichuan University, and Collaborative Innovation Center for Biotherapy Chengdu China
                [ 3 ] Department of Pediatric Surgery West China Hospital Sichuan University Chengdu China
                [ 4 ] School of Medicine Southern University of Science and Technology Shenzhen Guangdong China
                [ 5 ] Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research Shenzhen China
                [ 6 ] Department of Urology Graduate School of Medical Sciences Kanazawa University Kanazawa Japan
                [ 7 ] Department of Biochemistry and Molecular Biology Monash University Clayton Vic Australia
                [ 8 ] Department of Gastrointestinal Surgery State Key Laboratory of Biotherapy and Cancer Center West China Hospital Sichuan University, and Collaborative Innovation Center for Biotherapy Chengdu China
                Author notes
                [*] [* ] Corresponding author. Tel: +86 18980606468; E‐mail: hnchen@ 123456scu.edu.cn

                Corresponding author. Tel: +86 13258370346; E‐mail: hcanhua@ 123456scu.edu.cn

                Corresponding author. Tel: +86 18081954096; E‐mail: xiaweiwei@ 123456scu.edu.cn

                [ † ]

                These authors contributed equally to this work

                Author information
                https://orcid.org/0000-0001-7509-6965
                https://orcid.org/0000-0002-0423-6358
                https://orcid.org/0000-0003-0104-8498
                https://orcid.org/0000-0003-2247-7750
                https://orcid.org/0000-0002-6513-6422
                Article
                EMMM202114903
                10.15252/emmm.202114903
                8819355
                35023619
                444710bf-4d4e-4a05-9154-036a3b97df94
                © 2022 The Authors. Published under the terms of the CC BY 4.0 license

                This is an open access article under the terms of the http://creativecommons.org/licenses/by/4.0/ License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.

                History
                : 10 December 2021
                : 26 July 2021
                : 14 December 2021
                Page count
                Figures: 13, Tables: 6, Pages: 28, Words: 14086
                Funding
                Funded by: National Natural Science Foundation of China (NSFC) , doi 10.13039/501100001809;
                Award ID: 32122052
                Award ID: 81821002
                Award ID: 81790251
                Award ID: 82130082
                Award ID: 82003113
                Award ID: 82073246
                Award ID: 81972766
                Award ID: 82173336
                Award ID: U19A2003
                Funded by: MOST | National Key Research and Development Program of China (973 Program) , doi 10.13039/501100012166;
                Award ID: 2020YFA0509400
                Funded by: Guangdong Provincial Key Laboratory of Urology (Guangdong Key Laboratory of Urology) , doi 10.13039/100013261;
                Award ID: 2017B030301018
                Funded by: CAS | Guangdong Academy of Sciences (GDAS) , doi 10.13039/501100009075;
                Award ID: 2019B030302012
                Categories
                Article
                Articles
                Custom metadata
                2.0
                7 February 2022
                Converter:WILEY_ML3GV2_TO_JATSPMC version:6.1.1 mode:remove_FC converted:07.02.2022

                Molecular medicine
                cancer therapy,drug‐tolerant persister state,multidrug resistance,npc1l1,oxidative stress,cancer

                Comments

                Comment on this article