1
|
Kato K, Yasui H, Sato-Akaba H, Emoto MC, Fujii HG, Kmiec MM, Kuppusamy P, Nagane M, Yamashita T, Inanami O. Non-invasive electron paramagnetic resonance imaging detects tumor redox imbalance induced by ferroptosis. Redox Rep 2025; 30:2454887. [PMID: 39836064 PMCID: PMC11753017 DOI: 10.1080/13510002.2025.2454887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025] Open
Abstract
Targeting ferroptosis, cell death caused by the iron-dependent accumulation of lipid peroxides, and disruption of the redox balance are promising strategies in cancer therapy owing to the physiological characteristics of cancer cells. However, the detection of ferroptosis using in vivo imaging remains challenging. We previously reported that redox maps showing the reduction power per unit time of implanted tumor tissues via non-invasive redox imaging using a novel, compact, and portable electron paramagnetic resonance imaging (EPRI) device could be compared with tumor tissue sections. This study aimed to apply the EPRI technique to the in vivo detection of ferroptosis. Notably, redox maps reflecting changes in the redox status of tumors induced by the ferroptosis-inducing agent imidazole ketone erastin (IKE) were compared with the immunohistochemical images of 4-hydroxynonenal (4-HNE) in tumor tissue sections. Our comparison revealed a negative correlation between the reducing power of tumor tissue and the number of 4-HNE-positive cells. Furthermore, the control and IKE-treated groups exhibited significantly different distributions on the correlation map. Therefore, redox imaging using EPRI may contribute to the non-invasive detection of ferroptosis in vivo.
Collapse
Affiliation(s)
- Kazuhiro Kato
- Laboratory of Radiation Biology, Department of Applied Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Hironobu Yasui
- Laboratory of Radiation Biology, Department of Applied Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
- One Health Research Center, Hokkaido University, Sapporo, Japan
| | - Hideo Sato-Akaba
- Department of Electrical and Electronic Engineering, Graduate School of Integrated Science and Technology, Shizuoka University, Hamamatsu, Japan
| | - Miho C. Emoto
- Department of Clinical Laboratory Science, School of Medical Technology, Health Sciences University of Hokkaido, Sapporo, Japan
| | - Hirotada G. Fujii
- Advanced Research Promotion Center, Health Sciences University of Hokkaido, Ishikari, Japan
| | - Maciej M. Kmiec
- Departments of Radiology and Radiation Oncology, Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| | - Periannan Kuppusamy
- Departments of Radiology and Radiation Oncology, Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| | - Masaki Nagane
- Laboratory of Biochemistry, School of Veterinary Medicine, Azabu University, Sagamihara, Japan
| | - Tadashi Yamashita
- Laboratory of Biochemistry, School of Veterinary Medicine, Azabu University, Sagamihara, Japan
| | - Osamu Inanami
- Laboratory of Radiation Biology, Department of Applied Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
2
|
Guo W, Duan Z, Wu J, Zhou BP. Epithelial-mesenchymal transition promotes metabolic reprogramming to suppress ferroptosis. Semin Cancer Biol 2025; 112:20-35. [PMID: 40058616 DOI: 10.1016/j.semcancer.2025.02.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 02/05/2025] [Accepted: 02/28/2025] [Indexed: 03/22/2025]
Abstract
Epithelial-mesenchymal transition (EMT) is a cellular de-differentiation process that provides cells with the increased plasticity and stem cell-like traits required during embryonic development, tissue remodeling, wound healing and metastasis. Morphologically, EMT confers tumor cells with fibroblast-like properties that lead to the rearrangement of cytoskeleton (loss of stiffness) and decrease of membrane rigidity by incorporating high level of poly-unsaturated fatty acids (PUFA) in their phospholipid membrane. Although large amounts of PUFA in membrane reduces rigidity and offers capabilities for tumor cells with the unbridled ability to stretch, bend and twist in metastasis, these PUFA are highly susceptible to lipid peroxidation, which leads to the breakdown of membrane integrity and, ultimately results in ferroptosis. To escape the ferroptotic risk, EMT also triggers the rewiring of metabolic program, particularly in lipid metabolism, to enforce the epigenetic regulation of EMT and mitigate the potential damages from ferroptosis. Thus, the interplay among EMT, lipid metabolism, and ferroptosis highlights a new layer of intricated regulation in cancer biology and metastasis. Here we summarize the latest findings and discuss these mutual interactions. Finally, we provide perspectives of how these interplays contribute to cellular plasticity and ferroptosis resistance in metastatic tumor cells that can be explored for innovative therapeutic interventions.
Collapse
Affiliation(s)
- Wenzheng Guo
- Departments of Molecular and Cellular Biochemistry, and the Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, KY 40506, United States
| | - Zhibing Duan
- Departments of Molecular and Cellular Biochemistry, and the Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, KY 40506, United States
| | - Jingjing Wu
- Departments of Molecular and Cellular Biochemistry, and the Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, KY 40506, United States
| | - Binhua P Zhou
- Departments of Molecular and Cellular Biochemistry, and the Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, KY 40506, United States.
| |
Collapse
|
3
|
Naderi S, Khodagholi F, Janahmadi M, Motamedi F, Torabi A, Batool Z, Heydarabadi MF, Pourbadie HG. Ferroptosis and cognitive impairment: Unraveling the link and potential therapeutic targets. Neuropharmacology 2025; 263:110210. [PMID: 39521042 DOI: 10.1016/j.neuropharm.2024.110210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 10/24/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Neurodegenerative disorders, such as Alzheimer's and Parkinson's diseases, share key characteristics, notably cognitive impairment and significant cell death in specific brain regions. Cognition, a complex mental process allowing individuals to perceive time and place, is disrupted in these conditions. This consistent disruption suggests the possibility of a shared underlying mechanism across all neurodegenerative diseases. One potential common factor is the activation of pathways leading to cell death. Despite significant progress in understanding cell death pathways, no definitive treatments have emerged. This has shifted focus towards less-explored mechanisms like ferroptosis, which holds potential due to its involvement in oxidative stress and iron metabolism. Unlike apoptosis or necrosis, ferroptosis offers a novel therapeutic avenue due to its distinct biochemical and genetic underpinnings, making it a promising target in neurodegenerative disease treatment. Ferroptosis is distinguished from other cellular death mechanisms, by distinctive characteristics such as an imbalance of iron hemostasis, peroxidation of lipids in the plasma membrane, and dysregulated glutathione metabolism. In this review, we discuss the potential role of ferroptosis in cognitive impairment. We then summarize the evidence linking ferroptosis biomarkers to cognitive impairment brought on by neurodegeneration while highlighting recent advancements in our understanding of the molecular and genetic mechanisms behind the condition. Finally, we discuss the prospective therapeutic implications of targeting ferroptosis for the treatment of cognitive abnormalities associated with neurodegeneration, including natural and synthetic substances that suppress ferroptosis via a variety of mechanisms. Promising therapeutic candidates, including antioxidants and iron chelators, are being explored to inhibit ferroptosis and mitigate cognitive decline.
Collapse
Affiliation(s)
- Soudabeh Naderi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fariba Khodagholi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahyar Janahmadi
- Neuroscience Research Center, Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fereshteh Motamedi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abolfazl Torabi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zehra Batool
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | | | - Hamid Gholami Pourbadie
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
4
|
Wang W, Hashimi B, Wang P. Targeting ferroptosis: the role of non-coding RNAs in hepatocellular carcinoma progression and therapy. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03791-y. [PMID: 39820644 DOI: 10.1007/s00210-025-03791-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 01/06/2025] [Indexed: 01/19/2025]
Abstract
One of the most common tumors is hepatocellular carcinoma (HCC), and the prognosis for late-stage HCC is still not good. It is anticipated that improved outcomes would result from a deeper comprehension of the pathophysiology of HCC. Ferroptosis as a new discovered cell death type is linked to the progression of HCC and may be crucial for its detection, prevention, therapy, and prognosis. Numerous studies suggest that epigenetic alterations mediated by non-coding RNAs (ncRNA) might influence cancer cell susceptibility to ferroptosis. This study elucidates the processes of ferroptosis and delineates the paths by which ncRNAs influence HCC by modulating ferroptosis. Furthermore, it offers significant insights into ferroptosis-associated ncRNAs, intending to discover novel therapeutic approaches for HCC. It also explores innovative concepts for the future use of ncRNA-based ferroptosis-targeted therapeutics.
Collapse
Affiliation(s)
- Weijia Wang
- Department of Oncology, Qilu Hospital of Shandong University Dezhou Hospital (Dezhou People's Hospital), Shandong Province, China
| | - Behishta Hashimi
- Department of Midwifery, Jahan Institute of Health Sciences, Kabul, Afghanistan
| | - Ping Wang
- Department of Oncology, Yantaishan Hospital Affiliated to Binzhou Medical University, Yantai, Shandong Province, China.
| |
Collapse
|
5
|
Derogar R, Nejadi Orang F, Abdoli Shadbad M. Competing endogenous RNA networks in ovarian cancer: from bench to bedside. EXCLI JOURNAL 2025; 24:86-112. [PMID: 39967908 PMCID: PMC11830916 DOI: 10.17179/excli2024-7827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 12/19/2024] [Indexed: 02/20/2025]
Abstract
Epithelial ovarian cancer is responsible for the majority of ovarian malignancies, and its highly invasive nature and chemoresistant development have been major obstacles to treating patients with mainstream treatments. In recent decades, the significance of microRNAs (miRNAs), circular RNAs (circRNAs), long non-coding RNAs (lncRNAs), and competing endogenous RNAs (ceRNAs) has been highlighted in ovarian cancer development. This hidden language between these RNAs has led to the discovery of enormous regulatory networks in ovarian cancer cells that substantially affect gene expression. Aside from providing ample opportunities for targeted therapies, circRNA- and lncRNA-mediated ceRNA network components provide invaluable biomarkers. The current study provides a comprehensive and up-to-date review of the recent findings on the significance of these ceRNA networks in the hallmarks of ovarian cancer oncogenesis, treatment, diagnosis, and prognosis. Also, it provides the authorship with future perspectives in the era of single-cell RNA sequencing and personalized medicine.
Collapse
Affiliation(s)
- Roghaiyeh Derogar
- Fellowship in Gynecologic Oncology, Department of Gynecology, Faculty of Medical Sciences, Tabriz Medical Sciences, Islamic Azad University, Tabriz, Iran
| | | | - Mahdi Abdoli Shadbad
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
6
|
Bandekar M, Panda D. Microtubule depolymerization induces ferroptosis in neuroblastoma cells. IUBMB Life 2024; 76:1186-1198. [PMID: 39038059 DOI: 10.1002/iub.2899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 06/06/2024] [Indexed: 07/24/2024]
Abstract
Estramustine (EM), a clinically successful hormone-refractory anti-prostate cancer drug, exhibited potent anti-proliferative activity, depolymerized microtubules, blocked cells at mitosis, and induced cell death in different cancer cells. Altered iron metabolism is a feature of cancer cells. Using EM, we examined the plausible relationship between microtubule depolymerization and induction of ferroptosis in human neuroblastoma (SH-SY5Y and IMR-32) cells. EM reduced glutathione (GSH) levels and induced reactive oxygen species (ROS) generation. The pre-treatment of neuroblastoma cells with ROS scavengers (N-acetyl cysteine and dithiothreitol) reduced the anti-proliferative effects of EM. EM treatment increased labile iron pool (LIP), depleted glutathione peroxidase 4 (GPX4) levels, and lipid peroxidation, hallmark features of ferroptosis, highlighting ferroptosis induction. Ferroptosis inhibitors (deferoxamine mesylate and liproxstatin-1) abrogated the cytotoxic effects of EM, further confirming ferroptosis induction. Vinblastine and nocodazole also increased LIP and induced lipid peroxidation in neuroblastoma cells. This study provides evidence for the coupling of microtubule integrity to ferroptosis. The results also suggest that microtubule-depolymerizing agents may be considered for developing pro-ferroptosis chemotherapeutics.
Collapse
Affiliation(s)
- Mayuri Bandekar
- Department of Biosciences & Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Dulal Panda
- Department of Biosciences & Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
- National Institute of Pharmaceutical Education and Research, Mohali, India
| |
Collapse
|
7
|
Herrera-Abreu MT, Guan J, Khalid U, Ning J, Costa MR, Chan J, Li Q, Fortin JP, Wong WR, Perampalam P, Biton A, Sandoval W, Vijay J, Hafner M, Cutts R, Wilson G, Frankum J, Roumeliotis TI, Alexander J, Hickman O, Brough R, Haider S, Choudhary J, Lord CJ, Swain A, Metcalfe C, Turner NC. Inhibition of GPX4 enhances CDK4/6 inhibitor and endocrine therapy activity in breast cancer. Nat Commun 2024; 15:9550. [PMID: 39500869 PMCID: PMC11538343 DOI: 10.1038/s41467-024-53837-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 10/18/2024] [Indexed: 11/08/2024] Open
Abstract
CDK4/6 inhibition in combination with endocrine therapy is the standard of care for estrogen receptor (ER+) breast cancer, and although cytostasis is frequently observed, new treatment strategies that enhance efficacy are required. Here, we perform two independent genome-wide CRISPR screens to identify genetic determinants of CDK4/6 and endocrine therapy sensitivity. Genes involved in oxidative stress and ferroptosis modulate sensitivity, with GPX4 as the top sensitiser in both screens. Depletion or inhibition of GPX4 increases sensitivity to palbociclib and giredestrant, and their combination, in ER+ breast cancer models, with GPX4 null xenografts being highly sensitive to palbociclib. GPX4 perturbation additionally sensitises triple negative breast cancer (TNBC) models to palbociclib. Palbociclib and giredestrant induced oxidative stress and disordered lipid metabolism, leading to a ferroptosis-sensitive state. Lipid peroxidation is promoted by a peroxisome AGPAT3-dependent pathway in ER+ breast cancer models, rather than the classical ACSL4 pathway. Our data demonstrate that CDK4/6 and ER inhibition creates vulnerability to ferroptosis induction, that could be exploited through combination with GPX4 inhibitors, to enhance sensitivity to the current therapies in breast cancer.
Collapse
Affiliation(s)
- M T Herrera-Abreu
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | - J Guan
- Genentech, 1 DNA Way, South San Francisco, CA, USA
| | - U Khalid
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | - J Ning
- Tumour Modelling Facility, Institute of Cancer Research, London, UK
| | - M R Costa
- Genentech, 1 DNA Way, South San Francisco, CA, USA
| | - J Chan
- Genentech, 1 DNA Way, South San Francisco, CA, USA
| | - Q Li
- Genentech, 1 DNA Way, South San Francisco, CA, USA
| | - J-P Fortin
- Genentech, 1 DNA Way, South San Francisco, CA, USA
| | - W R Wong
- Genentech, 1 DNA Way, South San Francisco, CA, USA
| | - P Perampalam
- ProCogia Inc. under contract to Hoffmann-La Roche Limited, Toronto, ON, Canada
| | - A Biton
- Genentech, 1 DNA Way, South San Francisco, CA, USA
| | - W Sandoval
- Genentech, 1 DNA Way, South San Francisco, CA, USA
| | - J Vijay
- Roche Informatics, Mississauga, ON, Canada
| | - M Hafner
- Genentech, 1 DNA Way, South San Francisco, CA, USA
| | - R Cutts
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | - G Wilson
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | - J Frankum
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
- CRUK Gene Function Laboratory, The Institute of Cancer Research, London, UK
| | - T I Roumeliotis
- Functional proteomics team, The Institute of Cancer Research, London, UK
| | - J Alexander
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | - O Hickman
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | - R Brough
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
- CRUK Gene Function Laboratory, The Institute of Cancer Research, London, UK
| | - S Haider
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | - J Choudhary
- Functional proteomics team, The Institute of Cancer Research, London, UK
| | - C J Lord
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
- CRUK Gene Function Laboratory, The Institute of Cancer Research, London, UK
| | - A Swain
- Tumour Modelling Facility, Institute of Cancer Research, London, UK
| | - C Metcalfe
- Genentech, 1 DNA Way, South San Francisco, CA, USA.
| | - N C Turner
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK.
- Breast Unit, The Royal Marsden Hospital, London, UK.
| |
Collapse
|
8
|
Xu Q, Ren L, Ren N, Yang Y, Pan J, Zheng Y, Wang G. Ferroptosis: a new promising target for hepatocellular carcinoma therapy. Mol Cell Biochem 2024; 479:2615-2636. [PMID: 38051404 DOI: 10.1007/s11010-023-04893-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 11/01/2023] [Indexed: 12/07/2023]
Abstract
Hepatocellular carcinoma (HCC) is the sixed most common malignant tumor in the world. The study for HCC is mired in the predicament confronted with the difficulty of early diagnosis and high drug resistance, the survival rate of patients with HCC being low. Ferroptosis, an iron-dependent cell death, has been discovered in recent years as a cell death means with tremendous potential to fight against cancer. The in-depth researches for iron metabolism, lipid peroxidation and dysregulation of antioxidant defense have brought about tangible progress in the firmament of ferroptosis with more and more results showing close connections between ferroptosis and HCC. The potential role of ferroptosis has been widely used in chemotherapy, immunotherapy, radiotherapy, and nanotherapy, with the development of various new drugs significantly improving the prognosis of patients. Based on the characteristics and mechanisms of ferroptosis, this article further focuses on the main signaling pathways and promising treatments of HCC, envisioning that existing problems in regard with ferroptosis and HCC could be grappled with in the foreseeable future.
Collapse
Affiliation(s)
- Qiaoping Xu
- Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Cancer Center, Westlake University School of Medical, Hangzhou, 310006, China
| | - Lanqi Ren
- Fourth Clinical Medical College of Zhejiang, Chinese Medical University, Hangzhou, 310051, China
| | - Ning Ren
- Fourth Clinical Medical College of Zhejiang, Chinese Medical University, Hangzhou, 310051, China
| | - Yibei Yang
- Fourth Clinical Medical College of Zhejiang, Chinese Medical University, Hangzhou, 310051, China
| | - Junjie Pan
- Fourth Clinical Medical College of Zhejiang, Chinese Medical University, Hangzhou, 310051, China
| | - Yu Zheng
- Second Clinical Medical College of Zhejiang, Chinese Medical University, Hangzhou, 310051, China
| | - Gang Wang
- Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Cancer Center, Westlake University School of Medical, Hangzhou, 310006, China.
| |
Collapse
|
9
|
Wang S, Wu Y, Yang F, Hsu F, Zhang K, Hung J. NCI677397 targeting USP24-mediated induction of lipid peroxidation induces ferroptosis in drug-resistant cancer cells. Mol Oncol 2024; 18:2255-2276. [PMID: 38140768 PMCID: PMC11467797 DOI: 10.1002/1878-0261.13574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 10/20/2023] [Accepted: 12/19/2023] [Indexed: 12/24/2023] Open
Abstract
Cancer represents a profound challenge to healthcare systems and individuals worldwide. The development of multiple drug resistance is a major problem in cancer therapy and can result in progression of the disease. In our previous studies, we developed small-molecule inhibitors targeting ubiquitin-specific peptidase 24 (USP24) to combat drug-resistant lung cancer. Recently, we found that the USP24 inhibitor NCI677397 induced ferroptosis, a type of programmed cell death, in drug-resistant cancer cells by increasing lipid reactive oxygen species (ROS) levels. In the present study, we investigated the molecular mechanisms and found that the targeting of USP24 by NCI677397 increased gene expression of most lipogenesis-related genes, such as acyl-CoA synthetase long-chain family member 4 (ACSL4), and activated autophagy. In addition, the activity of several antioxidant enzymes, such as glutathione peroxidase 4 (GPX4) and dihydrofolate reductase (DHFR), was inhibited by NCI677397 treatment via an increase in protein degradation, thereby inducing lipid ROS production and lipid peroxidation. In summary, we demonstrated that NCI677397 induced a marked increase in lipid ROS levels, subsequently causing lipid peroxidation and leading to the ferroptotic death of drug-resistant cancer cells. Our study provides new insights into the clinical use of USP24 inhibitors as ferroptosis inducers (FINs) to block drug resistance during chemotherapy.
Collapse
Affiliation(s)
- Shao‐An Wang
- School of Respiratory Therapy, College of MedicineTaipei Medical UniversityTaiwan
| | - Yu‐Chih Wu
- School of Respiratory Therapy, College of MedicineTaipei Medical UniversityTaiwan
| | - Feng‐Ming Yang
- School of Respiratory Therapy, College of MedicineTaipei Medical UniversityTaiwan
| | - Feng‐Lin Hsu
- School of Respiratory Therapy, College of MedicineTaipei Medical UniversityTaiwan
| | - Kuan Zhang
- Cardiovascular Research InstituteUniversity of California, San FranciscoCAUSA
| | - Jan‐Jong Hung
- Department of Biotechnology and Bioindustry SciencesNational Cheng Kung UniversityTainanTaiwan
| |
Collapse
|
10
|
Paramanantham A, Asfiya R, Manjunath Y, Xu L, McCully G, Das S, Yang H, Kaifi JT, Srivastava A. Induction of Ferroptosis by an Amalgam of Extracellular Vesicles and Iron Oxide Nanoparticles Overcomes Cisplatin Resistance in Lung Cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.19.608664. [PMID: 39229071 PMCID: PMC11370464 DOI: 10.1101/2024.08.19.608664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Extracellular vesicles (EVs) hold potential as effective carriers for drug delivery, providing a promising approach to resolving challenges in lung cancer treatment. Traditional treatments, such as with the chemotherapy drug cisplatin, encounter resistance in standard cell death pathways like apoptosis, prompting the need to explore alternative approaches. This study investigates the potential of iron oxide nanoparticles (IONP) and EVs to induce ferroptosis-a regulated cell death mechanism-in lung cancer cells. We formulated a novel EV and IONP-based system, namely 'ExoFeR', and observed that ExoFeR demonstrated efficient ferroptosis induction, evidenced by downregulation of ferroptosis markers (xCT/SLC7A11 and GPX4), increased intracellular and mitochondrial ferrous iron levels, and morphological changes in mitochondria. To enhance efficacy, tumor-targeting transferrin (TF)-conjugated ExoFeR (ExoFeR TF ) was developed. ExoFeR TF outperformed ExoFeR, exhibiting higher uptake and cell death in lung cancer cells. Mechanistically, nuclear factor erythroid 2-related factor 2 (Nrf2)-a key regulator of genes involved in glutathione biosynthesis, antioxidant responses, lipid metabolism, and iron metabolism-was found downregulated in the ferroptotic cells. Inhibition of Nrf2 intracellular translocation in ExoFeR TF -treated cells was also observed, emphasizing the role of Nrf2 in modulating ferroptosis-dependent cell death. Furthermore, ExoFeR and ExoFeR TF demonstrated the ability to sensitize chemo-resistant cancer cells, including cisplatin-resistant lung cancer patient-derived tumoroid organoids. In summary, ExoFeR TF presents a promising and multifaceted therapeutic approach for combating lung cancer by intrinsically inducing ferroptosis and sensitizing chemo-resistant cells.
Collapse
|
11
|
Fayed B, Shakartalla SB, Sabbah H, Dalle H, Tannira M, Senok A, Soliman SSM. Transcriptome Analysis of Human Dermal Cells Infected with Candida auris Identified Unique Pathogenesis/Defensive Mechanisms Particularly Ferroptosis. Mycopathologia 2024; 189:65. [PMID: 38990436 DOI: 10.1007/s11046-024-00868-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 06/10/2024] [Indexed: 07/12/2024]
Abstract
Candida auris is an emerging multi-drug resistant yeast that can cause life-threatening infections. A recent report clarified the ability of C. auris to form a biofilm with enhanced drug resistance properties in the host skin's deep layers. The formed biofilm may initiate further bloodstream spread and immune escape. Therefore, we propose that secreted chemicals from the biofilm may facilitate fungal pathogenesis. In response to this interaction, the host skin may develop potential defensive mechanisms. Comparative transcriptomics was performed on the host dermal cells in response to indirect interaction with C. auris biofilm through Transwell inserts compared to planktonic cells. Furthermore, the effect of antifungals including caspofungin and fluconazole was studied. The obtained data showed that the dermal cells exhibited different transcriptional responses. Kyoto Encyclopedia of Genes and Genomes and Reactome analyses identified potential defensive responses employed by the dermal cells and potential toxicity induced by C. auris. Additionally, our data indicated that the dominating toxic effect was mediated by ferroptosis; which was validated by qRT-PCR, cytotoxicity assay, and flow cytometry. On the other hand, the viability of C. auris biofilm was enhanced and accompanied by upregulation of MDR1, and KRE6 upon interaction with dermal cells; both genes play significant roles in drug resistance and biofilm maturation, respectively. This study for the first-time shed light on the dominating defensive responses of human dermal cells, microbe colonization site, to C. auris biofilm and its toxic effects. Further, it demonstrates how C. auris biofilm responds to the defensive mechanisms developed by the human dermal cells.
Collapse
Affiliation(s)
- Bahgat Fayed
- Research Institute for Medical and Health Sciences, University of Sharjah, P.O. Box 27272, Sharjah, UAE
- Chemistry of Natural and Microbial Products, National Research Centre, Cairo, Egypt
| | - Sarra B Shakartalla
- Research Institute for Medical and Health Sciences, University of Sharjah, P.O. Box 27272, Sharjah, UAE
- College of Medicine, University of Sharjah, P.O. Box 27272, Sharjah, UAE
- Faculty of Pharmacy, University of Gezira, P.O.Box. 21111, Wad Medani, Sudan
| | - Hassan Sabbah
- AbbVie BioPharmaceuticals, P.O. Box 118052, Dubai, UAE
| | - Hala Dalle
- AbbVie BioPharmaceuticals, Kuwait City, Kuwait
| | | | - Abiola Senok
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Building 14 Dubai Healthcare City, P.O.Box 505055, Dubai, UAE
| | - Sameh S M Soliman
- Research Institute for Medical and Health Sciences, University of Sharjah, P.O. Box 27272, Sharjah, UAE.
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, P.O. Box 27272, Sharjah, UAE.
| |
Collapse
|
12
|
Tang Z, Chen Y, Huang Y, Zhao J, Jia B. Novel ferroptosis signature for improving prediction of prognosis and indicating gene targets from single-cell level in oral squamous cell carcinoma. Heliyon 2024; 10:e31676. [PMID: 38845860 PMCID: PMC11153103 DOI: 10.1016/j.heliyon.2024.e31676] [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: 10/07/2023] [Revised: 05/18/2024] [Accepted: 05/20/2024] [Indexed: 06/09/2024] Open
Abstract
Background Oral squamous cell carcinoma (OSCC) is one of the most prevalent kinds of cancers. Therefore, there is a pressing need to create a new risk scoring model to personalize the prognosis of OSCC patients and screen for patient-specific therapeutic agents and molecular targets. Methods Firstly, A series of bioinformatics was performed to construct a novel ferroptosis-related prognostic model; Further, drug sensitivity analysis was used to screen for specific therapeutic agents for OSCC; Single-cell analysis was employed to investigate the enrichment of FRDEGs (ferroptosis-related differentially expressed genes) in the OSCC microenvironment; Finally, various experiments were conducted to screen and validate molecular therapeutic targets for OSCC. Results In this study, we constructed a novel 10-FRDEGs risk scoring model. Base on the risk scoring model, we founded three potential chemotherapeutic agents for OSCC: 5Z)-7-Oxozeaenol, AT-7519, KIN001-266; In addition, FRDEGs were enriched in the epithelial cells of OSCC. Finally, we found that CA9 and CAV1 could regulate OSCC proliferation, migration and ferroptosis in vitro. Conclusion A novel 10-FRDEGs risk scoring model can predict the prognosis of patients with OSCC.Further,5Z)-7-Oxozeaenol, AT-7519, KIN001-266 are potential chemotherapeutic agents for OSCC.Moreover, we identified CA9、CAV1 as potential molecular target for the treatment of OSCC.Our findings provide new directions for prognostic assessment and precise treatment of oral cell squamous carcinoma.
Collapse
Affiliation(s)
- Zhengming Tang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Yuanxin Chen
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Yisheng Huang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - JianJiang Zhao
- Shenzhen Stomatological Hospital, Southern Medical University, Shenzhen, China
| | - Bo Jia
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| |
Collapse
|
13
|
Chen B, Zhao L, Yang R, Xu T. The recent advancements of ferroptosis in the diagnosis, treatment and prognosis of ovarian cancer. Front Genet 2023; 14:1275154. [PMID: 38028615 PMCID: PMC10665572 DOI: 10.3389/fgene.2023.1275154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 10/26/2023] [Indexed: 12/01/2023] Open
Abstract
Ovarian cancer affects the female reproductive system and is the primary cause of cancer related mortality globally. The imprecise and non-specific nature of ovarian cancer symptoms often results in patients being diagnosed at an advanced stage, with metastatic lesions extending beyond the ovary. This presents a significant clinical challenge and imposes a substantial economic burden on both patients and society. Despite advancements in surgery, chemotherapy, and immunotherapy, the prognosis for most patients with ovarian cancer remains unsatisfactory. Therefore, the development of novel treatment strategies is imperative. Ferroptosis, a distinct form of regulated cell death, characterized by iron-dependent lipid peroxidation, differs from autophagy, apoptosis, and necrosis, and may hold promise as a novel cell death. Numerous studies have demonstrated the involvement of ferroptosis in various conventional signaling pathways and biological processes. Recent investigations have revealed the significant contribution of ferroptosis in the initiation, progression, and metastasis of diverse malignant tumors, including ovarian cancer. Moreover, ferroptosis exhibits a synergistic effect with chemotherapy, radiotherapy, and immunotherapy in restraining the proliferation of ovarian cancer cells. The aforementioned implies that ferroptosis holds considerable importance in the management of ovarian cancer and has the potential to serve as a novel therapeutic target. The present review provides a comprehensive overview of the salient features of ferroptosis, encompassing its underlying mechanisms and functional role in ovarian cancer, along with the associated signaling pathways and genes. Furthermore, the review highlights the prospective utility of ferroptosis in the treatment of ovarian cancer.
Collapse
Affiliation(s)
| | | | | | - Tianmin Xu
- The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
14
|
Park HB, Baek KH. Current and future directions of USP7 interactome in cancer study. Biochim Biophys Acta Rev Cancer 2023; 1878:188992. [PMID: 37775071 DOI: 10.1016/j.bbcan.2023.188992] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 09/14/2023] [Accepted: 09/23/2023] [Indexed: 10/01/2023]
Abstract
The ubiquitin-proteasome system (UPS) is an essential protein quality controller for regulating protein homeostasis and autophagy. Ubiquitination is a protein modification process that involves the binding of one or more ubiquitins to substrates through a series of enzymatic processes. These include ubiquitin-activating enzymes (E1), ubiquitin-conjugating enzymes (E2), and ubiquitin ligases (E3). Conversely, deubiquitination is a reverse process that removes ubiquitin from substrates via deubiquitinating enzymes (DUBs). Dysregulation of ubiquitination-related enzymes can lead to various human diseases, including cancer, through the modulation of protein ubiquitination. The most structurally and functionally studied DUB is the ubiquitin-specific protease 7 (USP7). Both the TRAF and UBL domains of USP7 are known to bind to the [P/A/E]-X-X-S or K-X-X-X-K motif of substrates. USP7 has been shown to be involved in cancer pathogenesis by binding with numerous substrates. Recently, a novel substrate of USP7 was discovered through a systemic analysis of its binding motif. This review summarizes the currently discovered substrates and cellular functions of USP7 in cancer and suggests putative substrates of USP7 through a comprehensive systemic analysis.
Collapse
Affiliation(s)
- Hong-Beom Park
- Department of Convergence, CHA University, Gyeonggi-Do 13488, Republic of Korea
| | - Kwang-Hyun Baek
- Department of Convergence, CHA University, Gyeonggi-Do 13488, Republic of Korea; International Ubiquitin Center(,) CHA University, Gyeonggi-Do 13488, Republic of Korea.
| |
Collapse
|
15
|
Hu S, Chu Y, Zhou X, Wang X. Recent advances of ferroptosis in tumor: From biological function to clinical application. Biomed Pharmacother 2023; 166:115419. [PMID: 37666176 DOI: 10.1016/j.biopha.2023.115419] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/21/2023] [Accepted: 08/30/2023] [Indexed: 09/06/2023] Open
Abstract
Ferroptosis is a recently recognized form of cell death with distinct features in terms of morphology, biochemistry, and molecular mechanisms. Unlike other types of cell death, ferroptosis is characterized by iron dependence, reactive oxygen species accumulation and lipid peroxidation. Recent studies have demonstrated that selective autophagy plays a vital role in the induction of ferroptosis, including ferritinophagy, lipophagy, clockophagy, and chaperone-mediated autophagy. Emerging evidence has indicated the involvement of ferroptosis in tumorigenesis through regulating various biological processes, including tumor growth, metastasis, stemness, drug resistance, and recurrence. Clinical and preclinical studies have found that novel therapies targeting ferroptosis exert great potential in the treatment of tumors. This review provides a comprehensive overview of the molecular mechanisms in ferroptosis, especially in autophagy-driven ferroptosis, discusses the recent advances in the biological roles of ferroptosis in tumorigenesis, and highlights the application of novel ferroptosis-targeted therapies in the clinical treatment of tumors.
Collapse
Affiliation(s)
- Shunfeng Hu
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China; Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Yurou Chu
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China
| | - Xiangxiang Zhou
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China; Taishan Scholars Program of Shandong Province, Jinan, Shandong 250021, China; Branch of National Clinical Research Center for Hematologic Diseases, Jinan, Shandong 250021, China; National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou 251006, China.
| | - Xin Wang
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China; Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China; Taishan Scholars Program of Shandong Province, Jinan, Shandong 250021, China; Branch of National Clinical Research Center for Hematologic Diseases, Jinan, Shandong 250021, China; National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou 251006, China.
| |
Collapse
|
16
|
Liu MR, Shi C, Song QY, Kang MJ, Jiang X, Liu H, Pei DS. Sorafenib induces ferroptosis by promoting TRIM54-mediated FSP1 ubiquitination and degradation in hepatocellular carcinoma. Hepatol Commun 2023; 7:e0246. [PMID: 37695069 PMCID: PMC10497252 DOI: 10.1097/hc9.0000000000000246] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 06/24/2023] [Indexed: 09/12/2023] Open
Abstract
BACKGROUND Ferroptosis is a unique form of regulated cell death that provided a new opportunity for cancer therapy. Ferroptosis suppressor protein 1 (FSP1) is a key regulator in the NAD(P)H/FSP1/CoQ10 antioxidant system, which sever as an oxide redox enzyme to scavenge harmful lipid hydroperoxides and escape from ferroptosis in cells. This study aimed to investigate the role of FSP1 on sorafenib-induced ferroptosis and disclosed the underlying mechanisms. METHODS Cell viability, malondialdehyde (MDA), glutathione (GSH), and lipid reactive oxygen species levels were assessed using indicated assay kits. The levels of FSP1 and glutathione peroxidase 4 (GPX4) in the patients with HCC were analyzed based on the database. Western blot and quantitative real-time PCR were performed to detect the protein and mRNA expression. Co-immunoprecipitation was applied to detect the interaction between proteins. Tumor xenograft experiments were used to evaluate whether overexpression of FSP1-inhibited sorafenib-induced ferroptosis in vivo. RESULTS We verified that sorafenib-induced ferroptosis in HCC. Furthermore, we found that sorafenib decreased the protein level of FSP1, and knockdown FSP1 rendered HCC cells susceptible to sorafenib-induced ferroptosis. Co-immunoprecipitation and ubiquitination assays showed that sorafenib accelerated the TRIM54-mediated FSP1 ubiquitination and degradation. Sorafenib-induced ferroptosis was abrogated by TRIM54 suppression. Mechanically, sorafenib-promoted TRIM54 ubiquitinated and degraded FSP1 by means of the ERK pathway. Moreover, FSP1 enhanced tumor development and decreased HCC cellular susceptibility to sorafenib in vivo. CONCLUSIONS Sorafenib facilitated the TRIM54-mediated FSP1 ubiquitination through the ERK pathway, thereby inducing ferroptosis in HCC cells.
Collapse
Affiliation(s)
- Man-ru Liu
- Department of Pathology, Xuzhou Medical University, Xuzhou, China
- Department of Human Anatomy, Nanchang University Fuzhou Medical College, Fuzhou, China
| | - Ce Shi
- Department of Pathology, Xuzhou Medical University, Xuzhou, China
- Department of Orthopedics, The Affiliated Suqian Hospital of Xuzhou Medical University, Suqian, China
| | - Qiu-ya Song
- Department of Pathology, Xuzhou Medical University, Xuzhou, China
| | - Meng-jie Kang
- Department of Pathology, Xuzhou Medical University, Xuzhou, China
| | - Xin Jiang
- Department of Pathology, Xuzhou Medical University, Xuzhou, China
| | - Hui Liu
- Department of Ultrasound Medicine, Suqian First Hospital, Suqian, China
| | - Dong-sheng Pei
- Department of Pathology, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
17
|
Favaron C, Gabano E, Zanellato I, Gaiaschi L, Casali C, Bottone MG, Ravera M. Effects of Ferrocene and Ferrocenium on MCF-7 Breast Cancer Cells and Interconnection with Regulated Cell Death Pathways. Molecules 2023; 28:6469. [PMID: 37764244 PMCID: PMC10537025 DOI: 10.3390/molecules28186469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 09/01/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
The effects of ferrocene (Fc) and ferrocenium (Fc+) induced in triple negative human breast cancer MCF-7 cells were explored by immunofluorescence, flow cytometry, and transmission electron microscopy analysis. The different abilities of Fc and Fc+ to produce reactive oxygen species and induce oxidative stress were clearly observed by activating apoptosis and morphological changes after treatment, but also after tests performed on the model organism D. discoideum, particularly in the case of Fc+. The induction of ferroptosis, an iron-dependent form of regulated cell death driven by an overload of lipid peroxides in cellular membranes, occurred after 2 h of treatment with Fc+ but not Fc. However, the more stable Fc showed its effects by activating necroptosis after a longer-lasting treatment. The differences observed in terms of cell death mechanisms and timing may be due to rapid interconversion between the two oxidative forms of internalized iron species (from Fe2+ to Fe3+ and vice versa). Potential limitations include the fact that iron metabolism and mitophagy have not been investigated. However, the ability of both Fc and Fc+ to trigger different and interregulated types of cell death makes them suitable to potentially overcome the shortcomings of traditional apoptosis-mediated anticancer therapies.
Collapse
Affiliation(s)
- Cristina Favaron
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, Via Ferrata 9, 27100 Pavia, Italy; (C.F.); (L.G.); (C.C.)
| | - Elisabetta Gabano
- Department of Sustainable Development and Ecological Transition, University of Piemonte Orientale, Piazza S. Eusebio 5, 13100 Vercelli, Italy;
| | - Ilaria Zanellato
- Department of Sciences and Technological Innovation, University of Piemonte Orientale, Viale Teresa Michel 11, 15121 Alessandria, Italy
| | - Ludovica Gaiaschi
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, Via Ferrata 9, 27100 Pavia, Italy; (C.F.); (L.G.); (C.C.)
| | - Claudio Casali
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, Via Ferrata 9, 27100 Pavia, Italy; (C.F.); (L.G.); (C.C.)
| | - Maria Grazia Bottone
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, Via Ferrata 9, 27100 Pavia, Italy; (C.F.); (L.G.); (C.C.)
| | - Mauro Ravera
- Department of Sciences and Technological Innovation, University of Piemonte Orientale, Viale Teresa Michel 11, 15121 Alessandria, Italy
| |
Collapse
|
18
|
Shan C, Liang Y, Wang K, Li P. Noncoding RNAs in cancer ferroptosis: From biology to clinical opportunity. Biomed Pharmacother 2023; 165:115053. [PMID: 37379641 DOI: 10.1016/j.biopha.2023.115053] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 06/16/2023] [Accepted: 06/21/2023] [Indexed: 06/30/2023] Open
Abstract
Ferroptosis is a recently discovered pattern of programmed cell death that is nonapoptotic and irondependent. It is involved in lipid peroxidation dependent on reactive oxygen species. Ferroptosis has been verified to play a crucial regulatory role in a variety of pathological courses of disease, in particularly cancer. Emerging research has highlighted the potential of ferroptosis in tumorigenesis, cancer development and resistance to chemotherapy. However, the regulatory mechanism of ferroptosis remains unclear, which limits the application of ferroptosis in cancer treatment. Noncoding RNAs (ncRNAs) are noncoding transcripts that regulate gene expression in various ways to affect the malignant phenotypes of cancer cells. At present, the biological function and underlying regulatory mechanism of ncRNAs in cancer ferroptosis have been partially elucidated. Herein, we summarize the current knowledge of the central regulatory network of ferroptosis, with a focus on the regulatory functions of ncRNAs in cancer ferroptosis. The clinical application and prospects of ferroptosis-related ncRNAs in cancer diagnosis, prognosis and anticancer therapies are also discussed. Elucidating the function and mechanism of ncRNAs in ferroptosis, along with assessing the clinical significance of ferroptosis-related ncRNAs, provides new perspectives for understanding cancer biology and treatment approaches, which may benefit numerous cancer patients in the future.
Collapse
Affiliation(s)
- Chan Shan
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China.
| | - Yan Liang
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266021, China
| | - Kun Wang
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Peifeng Li
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China.
| |
Collapse
|
19
|
Tkachenko A, Onishchenko A, Myasoedov V, Yefimova S, Havranek O. Assessing regulated cell death modalities as an efficient tool for in vitro nanotoxicity screening: a review. Nanotoxicology 2023; 17:218-248. [PMID: 37083543 DOI: 10.1080/17435390.2023.2203239] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2023]
Abstract
Nanomedicine is a fast-growing field of nanotechnology. One of the major obstacles for a wider use of nanomaterials for medical application is the lack of standardized toxicity screening protocols for assessing the safety of newly synthesized nanomaterials. In this review, we focus on less frequently studied nanomaterials-induced regulated cell death (RCD) modalities, including eryptosis, necroptosis, pyroptosis, and ferroptosis, as a tool for in vitro nanomaterials safety evaluation. We summarize the latest insights into the mechanisms that mediate these RCDs in response to nanomaterials exposure. Comprehensive data from reviewed studies suggest that ROS (reactive oxygen species) overproduction and ROS-mediated pathways play a central role in nanomaterials-induced RCDs activation. On the other hand, studies also suggest that individual properties of nanomaterials, including size, shape, or surface charge, could determine specific toxicity pathways with consequent RCD induction as well. We anticipate that the evaluation of RCDs can become one of the mechanism-based screening methods in nanotoxicology. In addition to the toxicity assessment, evaluation of necroptosis-, pyroptosis-, and ferroptosis-promoting capacity of nanomaterials could simultaneously provide useful information for specific medical applications as could be their anti-tumor potential. Moreover, a detailed understanding of molecular mechanisms driving nanomaterials-mediated induction of immunogenic RCDs will substantially aid novel anti-tumor nanodrugs development.
Collapse
Affiliation(s)
- Anton Tkachenko
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czechia
- Research Institute of Experimental and Clinical Medicine, Kharkiv National Medical University, Kharkiv, Ukraine
| | - Anatolii Onishchenko
- Research Institute of Experimental and Clinical Medicine, Kharkiv National Medical University, Kharkiv, Ukraine
| | - Valeriy Myasoedov
- Department of Medical Biology, Kharkiv National Medical University, Kharkiv, Ukraine
| | - Svetlana Yefimova
- Institute for Scintillation Materials, National Academy of Sciences of Ukraine, Kharkiv, Ukraine
| | - Ondrej Havranek
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czechia
- Department of Hematology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
| |
Collapse
|
20
|
Xu Z, Tan R, Li X, Pan L, Ji P, Tang H. Development of a classification model and an immune-related network based on ferroptosis in periodontitis. J Periodontal Res 2023; 58:403-413. [PMID: 36653725 DOI: 10.1111/jre.13100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 12/14/2022] [Accepted: 01/03/2023] [Indexed: 01/20/2023]
Abstract
BACKGROUND AND OBJECTIVES Periodontitis is an immunoinflammatory disease characterized by irreversible periodontal attachment loss and bone destruction. Ferroptosis is a kind of immunogenic cell death that depends on the participation of iron ions and is involved in various inflammatory and immune processes. However, information regarding the relationship between ferroptosis and immunomodulation processes in periodontitis is extremely limited. The purpose of this study was to investigate the correlation between ferroptosis and immune responses in periodontitis. METHODS Gene expression profiles of gingivae were collected from the Gene Expression Omnibus data portal. After detecting differentially expressed ferroptosis-related genes (FRGs), we used univariate logistic regression analysis followed by logistic least absolute shrinkage and selection operator (LASSO) regression to establish a ferroptosis-related classification model in an attempt to accurately distinguish periodontitis gingival tissues from healthy samples. The infiltration level of immunocytes in periodontitis was then assessed through single-sample gene-set enrichment analysis. Subsequently, we screened out immune-related genes by weighted correlation network analysis and protein-protein interaction (PPI) analysis and constructed an immune-related network based on FRGs and immune-related genes. RESULTS A total of 24 differentially expressed FRGs were detected, and an 8-FRG combined signature constituted the classification model. The established model showed outstanding discriminating ability according to the results of receiver operating characteristic (ROC) curve analysis. In addition, the periodontitis samples had a higher degree of immunocyte infiltration. Activated B cells had the strongest positive correlation while macrophages had a strong negative correlation with certain FRGs, and we found that XBP1, ALOX5 and their interacting genes might be crucial genes in the immune-related network. CONCLUSIONS The FRG-based classification model had a satisfactory determination ability, which could bring new insights into the pathogenesis of periodontitis. Those genes in the immune-related network, especially hub genes along with XBP1 and ALOX5, would have the potential to serve as promising targets of immunomodulatory treatments for periodontitis.
Collapse
Affiliation(s)
- Zhihong Xu
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, China.,The People's Hospital of Dadukou District, Chongqing, China
| | - Ruolan Tan
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaodong Li
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, China
| | - Lanlan Pan
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, China
| | - Ping Ji
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, China
| | - Han Tang
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
21
|
Wang L, Karges J, Wei F, Xie L, Chen Z, Gasser G, Ji L, Chao H. A mitochondria-localized iridium(iii) photosensitizer for two-photon photodynamic immunotherapy against melanoma. Chem Sci 2023; 14:1461-1471. [PMID: 36794192 PMCID: PMC9906708 DOI: 10.1039/d2sc06675k] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 01/12/2023] [Indexed: 01/13/2023] Open
Abstract
Conventional photodynamic therapy mainly causes a therapeutic effect on the primary tumor through the localized generation of reactive oxygen species, while metastatic tumors remain poorly affected. Complementary immunotherapy is effective in eliminating small, non-localized tumors distributed across multiple organs. Here, we report the Ir(iii) complex Ir-pbt-Bpa as a highly potent immunogenic cell death inducing photosensitizer for two-photon photodynamic immunotherapy against melanoma. Ir-pbt-Bpa can produce singlet oxygen and superoxide anion radicals upon light irradiation, causing cell death by a combination of ferroptosis and immunogenic cell death. In a mouse model with two physically separated melanoma tumors, although only one of the primary tumors was irradiated, a strong tumor reduction of both tumors was observed. Upon irradiation, Ir-pbt-Bpa not only induced the immune response of CD8+ T cells and the depletion of regulatory T cells, but also caused an increase in the number of the effector memory T cells to achieve long-term anti-tumor immunity.
Collapse
Affiliation(s)
- Lili Wang
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University Guangzhou 510006 P. R. China
- Public Research Center, Hainan Medical University Haikou 571199 P. R. China
| | - Johannes Karges
- Faculty of Chemistry and Biochemistry, Ruhr-University Bochum, Universitätsstrasse 150 44780 Bochum Germany
| | - Fangmian Wei
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University Guangzhou 510006 P. R. China
| | - Lina Xie
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University Guangzhou 510006 P. R. China
| | - Zhuoli Chen
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University Guangzhou 510006 P. R. China
| | - Gilles Gasser
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology Paris 75005 France
| | - Liangnian Ji
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University Guangzhou 510006 P. R. China
| | - Hui Chao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University Guangzhou 510006 P. R. China
- MOE Key Laboratory of Theoretical Organic Chemistry and Functional Molecule, School of Chemistry and Chemical Engineering, Hunan University of Science and Technology Xiangtan 400201 P. R. China
| |
Collapse
|
22
|
Sansone C, Pistelli L, Calabrone L, Del Mondo A, Fontana A, Festa M, Noonan DM, Albini A, Brunet C. The Carotenoid Diatoxanthin Modulates Inflammatory and Angiogenesis Pathways In Vitro in Prostate Cancer Cells. Antioxidants (Basel) 2023; 12:antiox12020359. [PMID: 36829917 PMCID: PMC9952135 DOI: 10.3390/antiox12020359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/23/2023] [Accepted: 01/27/2023] [Indexed: 02/05/2023] Open
Abstract
Xanthophylls, a group of carotenoids, have attracted attention as human health benefit compounds thanks to their functionality and bioavailability. The great antioxidant and anti-inflammatory abilities of diatoxanthin (Dt), a photoprotective xanthophyll synthetized by diatoms, were recently documented. This study investigates the capacity of Dt to intercept prostate cancer progression in vitro on different human cell lines, exploring its role against cancer proliferation and angiogenesis. Our results highlighted the chemopreventive role of Dt already at low concentration (44.1 pM) and suggest that the Dt-induced cancer cell death occurred through oxidative stress mechanisms. This hypothesis was supported by variations on the expression of key genes and proteins. Oxidative stress cell deaths (e.g., ferroptosis) are recently described types of cell death that are closely related to the pathophysiological processes of many diseases, such as tumors. Nonetheless, the interest of Dt was further strengthened by its ability to inhibit angiogenesis. The results are discussed considering the actual progress and requirements in cancer therapy, notably for prostate cancer.
Collapse
Affiliation(s)
- Clementina Sansone
- Stazione Zoologica Anton Dohrn, Sede Molosiglio Marina Acton, 80133 Napoli, Italy
- Laboratory of Vascular Biology and Angiogenesis, IRCCS MultiMedica, 20138 Milan, Italy
- Correspondence: (C.S.); (C.B.)
| | - Luigi Pistelli
- Stazione Zoologica Anton Dohrn, Sede Molosiglio Marina Acton, 80133 Napoli, Italy
| | - Luana Calabrone
- Laboratory of Vascular Biology and Angiogenesis, IRCCS MultiMedica, 20138 Milan, Italy
| | - Angelo Del Mondo
- Stazione Zoologica Anton Dohrn, Sede Molosiglio Marina Acton, 80133 Napoli, Italy
| | - Angelo Fontana
- Institute of Biomolecular Chemistry, Italian National Research Council (CNR), 80078 Pozzuoli, Italy
| | - Marco Festa
- Laboratory of Vascular Biology and Angiogenesis, IRCCS MultiMedica, 20138 Milan, Italy
| | - Douglas M. Noonan
- Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
- Unit of Molecular Pathology, Biochemistry and Immunology, IRCCS MultiMedica, 20138 Milan, Italy
| | - Adriana Albini
- IRCSS European Institute of Oncology (IEO), 20141 Milan, Italy
| | - Christophe Brunet
- Stazione Zoologica Anton Dohrn, Sede Molosiglio Marina Acton, 80133 Napoli, Italy
- Correspondence: (C.S.); (C.B.)
| |
Collapse
|
23
|
Fan G, Liu M, Liu J, Huang Y. The initiator of neuroexcitotoxicity and ferroptosis in ischemic stroke: Glutamate accumulation. Front Mol Neurosci 2023; 16:1113081. [PMID: 37033381 PMCID: PMC10076579 DOI: 10.3389/fnmol.2023.1113081] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 03/06/2023] [Indexed: 04/11/2023] Open
Abstract
Glutamate plays an important role in excitotoxicity and ferroptosis. Excitotoxicity occurs through over-stimulation of glutamate receptors, specifically NMDAR, while in the non-receptor-mediated pathway, high glutamate concentrations reduce cystine uptake by inhibiting the System Xc-, leading to intracellular glutathione depletion and resulting in ROS accumulation, which contributes to increased lipid peroxidation, mitochondrial damage, and ultimately ferroptosis. Oxidative stress appears to crosstalk between excitotoxicity and ferroptosis, and it is essential to maintain glutamate homeostasis and inhibit oxidative stress responses in vivo. As researchers work to develop natural compounds to further investigate the complex mechanisms and regulatory functions of ferroptosis and excitotoxicity, new avenues will be available for the effective treatment of ischaemic stroke. Therefore, this paper provides a review of the molecular mechanisms and treatment of glutamate-mediated excitotoxicity and ferroptosis.
Collapse
Affiliation(s)
- Genhao Fan
- Graduate School, Tianjin University of Chinese Medicine, Tianjin, China
| | - Menglin Liu
- Graduate School, Tianjin University of Chinese Medicine, Tianjin, China
| | - Jia Liu
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Tianjin University of Chinese Medicine, Tianjin, China
| | - Yuhong Huang
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Tianjin University of Chinese Medicine, Tianjin, China
- *Correspondence: Yuhong Huang,
| |
Collapse
|
24
|
Corti A, Dominici S, Piaggi S, Pompella A. Enhancement of ferroptosis by boric acid and its potential use as chemosensitizer in anticancer chemotherapy. Biofactors 2022; 49:405-414. [PMID: 36468437 DOI: 10.1002/biof.1919] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 11/15/2022] [Indexed: 12/12/2022]
Abstract
Ferroptosis is a form of regulated cell death (RCD) characterized by intracellular iron ion accumulation and reactive oxygen species (ROS)-induced lipid peroxidation. Ferroptosis in cancer and ferroptosis-related anticancer drugs have recently gained interest in the field of cancer treatment. Boron is an essential trace element playing an important role in several biological processes. Recent studies have described contrasting effects of boric acid (BA) in cancer cells, ranging from protective/mitogenic to damaging/antiproliferative. Interestingly, boron has been shown to interfere with critical factors involved in ferroptosis-intracellular glutathione and lipid peroxidation in the first place. Thus, the present study was aimed to verify the ability of boron to modulate the ferroptotic process in HepG2 cells, a model of hepatocellular carcinoma. Our results indicate that-when used at high, pharmacological concentrations-BA can increase intracellular ROS, glutathione, and TBARS levels, and enhance ferroptosis induced by RSL3 and erastin. Also, high BA concentrations can directly induce ferroptosis, and such BA-induced ferroptosis can add to the cytotoxic effects of anticancer drugs sorafenib, doxorubicin and cisplatin. These observations suggest that BA could be exploited as a chemo-sensitizer agent in order to overcome cancer drug resistance in selected conditions. However, the possibility of reaching suitably high concentrations of BA in the tumor microenvironment will need to be further investigated.
Collapse
Affiliation(s)
- Alessandro Corti
- Department of Translational Research NTMS, University of Pisa Medical School, Pisa, Italy
| | - Silvia Dominici
- Department of Translational Research NTMS, University of Pisa Medical School, Pisa, Italy
| | - Simona Piaggi
- Department of Translational Research NTMS, University of Pisa Medical School, Pisa, Italy
| | - Alfonso Pompella
- Department of Translational Research NTMS, University of Pisa Medical School, Pisa, Italy
| |
Collapse
|
25
|
Zhan S, Yung MMH, Siu MKY, Jiao P, Ngan HYS, Chan DW, Chan KKL. New Insights into Ferroptosis Initiating Therapies (FIT) by Targeting the Rewired Lipid Metabolism in Ovarian Cancer Peritoneal Metastases. Int J Mol Sci 2022; 23:ijms232315263. [PMID: 36499591 PMCID: PMC9737695 DOI: 10.3390/ijms232315263] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/04/2022] [Accepted: 11/24/2022] [Indexed: 12/11/2022] Open
Abstract
Ovarian cancer is one of the most lethal gynecological cancers worldwide. The poor prognosis of this malignancy is substantially attributed to the inadequate symptomatic biomarkers for early diagnosis and effective remedies to cure the disease against chemoresistance and metastasis. Ovarian cancer metastasis is often relatively passive, and the single clusters of ovarian cancer cells detached from the primary ovarian tumor are transcoelomic spread by the peritoneal fluid throughout the peritoneum cavity and omentum. Our earlier studies revealed that lipid-enriched ascitic/omental microenvironment enforced metastatic ovarian cancer cells to undertake metabolic reprogramming and utilize free fatty acids as the main energy source for tumor progression and aggression. Intriguingly, cell susceptibility to ferroptosis has been tightly correlated with the dysregulated fatty acid metabolism (FAM), and enhanced iron uptake as the prominent features of ferroptosis are attributed to the strengthened lipid peroxidation and aberrant iron accumulation, suggesting that ferroptosis induction is a targetable vulnerability to prevent cancer metastasis. Therefore, the standpoints about tackling altered FAM in combination with ferroptosis initiation as a dual-targeted therapy against advanced ovarian cancer were highlighted herein. Furthermore, a discussion on the prospect and challenge of inducing ferroptosis as an innovative therapeutic approach for reversing remedial resistance in cancer interventions was included. It is hoped this proof-of-concept review will indicate appropriate directions for speeding up the translational application of ferroptosis-inducing compounds (FINs) to improve the efficacy of ovarian cancer treatment.
Collapse
Affiliation(s)
- Shijie Zhan
- Department of Obstetrics & Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Mingo M. H. Yung
- Department of Obstetrics & Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Michelle K. Y. Siu
- Department of Obstetrics & Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Peili Jiao
- Department of Obstetrics & Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Hextan Y. S. Ngan
- Department of Obstetrics & Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - David W. Chan
- Department of Obstetrics & Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- School of Medicine, The Chinese University of Hong Kong-Shenzhen, Shenzhen 518172, China
- Correspondence: (D.W.C.); (K.K.L.C.); Tel.: +86-755-2351-6153 (D.W.C.); +852-2255-4260 (K.K.L.C.); Fax: +852-2255-0947 (K.K.L.C.)
| | - Karen K. L. Chan
- Department of Obstetrics & Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Correspondence: (D.W.C.); (K.K.L.C.); Tel.: +86-755-2351-6153 (D.W.C.); +852-2255-4260 (K.K.L.C.); Fax: +852-2255-0947 (K.K.L.C.)
| |
Collapse
|
26
|
Xie L, Chen W, Chen Q, Jiang Y, Song E, Zhu X, Song Y. Synergistic hydroxyl radical formation, system XC- inhibition and heat shock protein crosslinking tango in ferrotherapy: A prove-of-concept study of "sword and shield" theory. Mater Today Bio 2022; 16:100353. [PMID: 35865409 PMCID: PMC9294558 DOI: 10.1016/j.mtbio.2022.100353] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 06/23/2022] [Accepted: 07/04/2022] [Indexed: 01/18/2023] Open
Abstract
Ferroptosis provide new insights into designing nanomedicines for enhanced cancer therapy; however, its antitumor efficacy is relatively low, mainly due to self-protective mechanism of cancer cells, e.g., heat shock protein (HSP) overexpression. Since HSPs can be modified/inhibited by lipid peroxidation (LPO) ending products, we construct a nanoplatform, namely MPDA@Fe3O4-Era, to amplify intracellular reactive oxygen species (ROS) and LPO for synergistic ferrotherapy. Upon tumor acidic microenvironment and local near-infrared stimuli, this nanoplatform releases Fe3O4 and reacts with intracellular hydrogen peroxide (H2O2) to promote Fenton reaction, and yields significant intracellular ROS (specifically hydroxyl radical, •OH) and LPO. In turn, LPO ending products crosslink HSPs to destroy self-preservation pathways of cancer cells to enhance anticancer effect. Meanwhile, the released erastin inhibits system XC− signal pathway to depletes glutathione. Fe3O4 loading further provides magnetic resonance imaging T2-weighted signal to guide anti-tumor treatment. Together, this nanoplatform not only provides •OH (as a “sword” to attack tumor cells), but also inhibits system XC− signal pathway and crosslinks HSP (break down the “shield” of tumor cells) to maximize synergistic ferro-therapeutic effect. MPDA@Fe3O4-Era plus laser irradiation possessed highly efficient tumor suppression with magnified the levels of •OH and inactive glutathione peroxidase 4 (GPX4), which can promote the development of precise cooperative cancer therapy.
Collapse
Affiliation(s)
- Li Xie
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, College of Pharmaceutical Sciences, Southwest University, 2 Tiansheng Rd, Beibei District, Chongqing, 400715, China
| | - Wenjie Chen
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, College of Pharmaceutical Sciences, Southwest University, 2 Tiansheng Rd, Beibei District, Chongqing, 400715, China
| | - Qifang Chen
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, College of Pharmaceutical Sciences, Southwest University, 2 Tiansheng Rd, Beibei District, Chongqing, 400715, China
| | - Yang Jiang
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, College of Pharmaceutical Sciences, Southwest University, 2 Tiansheng Rd, Beibei District, Chongqing, 400715, China
| | - Erqun Song
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, College of Pharmaceutical Sciences, Southwest University, 2 Tiansheng Rd, Beibei District, Chongqing, 400715, China
| | - Xiaokang Zhu
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, College of Pharmaceutical Sciences, Southwest University, 2 Tiansheng Rd, Beibei District, Chongqing, 400715, China
| | - Yang Song
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, 18 Shuangqing Rd, Haidian District, Beijing, 100085, China
| |
Collapse
|
27
|
Kosim MY, Fukazawa T, Miyauchi M, Hirohashi N, Tanimoto K. p53 status modifies cytotoxic activity of lactoferrin under hypoxic conditions. Front Pharmacol 2022; 13:988335. [PMID: 36199689 PMCID: PMC9527284 DOI: 10.3389/fphar.2022.988335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 09/01/2022] [Indexed: 11/13/2022] Open
Abstract
Lactoferrin (LF) is an iron binding glycoprotein of the transferrin family with a wide spectrum of biological effects, including anti-cancer activity. However, the detailed molecular mechanisms of anti-cancer activity of LF have not been fully determined. In this study, we tried to clarify cytotoxic functions of LF on various cell lines under hypoxic conditions and elucidate those molecular mechanisms. Cytotoxic activity of LF on cell lines was found to have a range of sensitivities. Hypoxia decreased sensitivity to LF in KD (lip fibroblast) but increased that in HSC2 (oral squamous cell carcinoma). Expression analyses further revealed that LF treatments increased hypoxic HIF-1α, -2α and p53 proteins in KD but attenuated them in HSC2 cells, and decreased HIF-1 target gene, DEC2, in KD but increased it in HSC2, suggesting a possible relationship between LF-modified DEC2 expression and HIF-α protein. MTT assay strikingly demonstrated that cells expressing mutant-type p53 (MT5) were more sensitive to LF than control HepG2 (hepatoma), suggesting an important role of the p53 signal. Knock-down of TP53 (p53 gene) interestingly reduced sensitivity to LF in HepG2, suggesting that p53 may be a target of LF cytotoxic activity. Further analyses with a ferroptosis promoter or inhibitor demonstrated that LF increased ACSL4 in hypoxic MT5, suggesting LF-induced ferroptosis in cells expressing mutant-type p53. In conclusion, hypoxia was found to regulate cytotoxic activities of LF differently among various cell lines, possibly through the p53 signaling pathway. LF further appeared to regulate ferroptosis through a modification of ACSL4 expression.
Collapse
Affiliation(s)
- Maryami Yuliana Kosim
- Department of Radiation Disaster Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Takahiro Fukazawa
- Natural Science Center for Basic Research and Development, Hiroshima University, Hiroshima, Japan
| | - Mutsumi Miyauchi
- Department of Oral and Maxillofacial Pathology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima , Japan
| | - Nobuyuki Hirohashi
- Department of Radiation Disaster Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Keiji Tanimoto
- Department of Radiation Disaster Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
- *Correspondence: Keiji Tanimoto,
| |
Collapse
|
28
|
Ma H, Dong Y, Chu Y, Guo Y, Li L. The mechanisms of ferroptosis and its role in alzheimer’s disease. Front Mol Biosci 2022; 9:965064. [PMID: 36090039 PMCID: PMC9459389 DOI: 10.3389/fmolb.2022.965064] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 07/25/2022] [Indexed: 12/06/2022] Open
Abstract
Alzheimer’s disease (AD) accounts for two-thirds of all dementia cases, affecting 50 million people worldwide. Only four of the more than 100 AD drugs developed thus far have successfully improved AD symptoms. Furthermore, these improvements are only temporary, as no treatment can stop or reverse AD progression. A growing number of recent studies have demonstrated that iron-dependent programmed cell death, known as ferroptosis, contributes to AD-mediated nerve cell death. The ferroptosis pathways within nerve cells include iron homeostasis regulation, cystine/glutamate (Glu) reverse transporter (system xc−), glutathione (GSH)/glutathione peroxidase 4 (GPX4), and lipid peroxidation. In the regulation pathway of AD iron homeostasis, abnormal iron uptake, excretion and storage in nerve cells lead to increased intracellular free iron and Fenton reactions. Furthermore, decreased Glu transporter expression leads to Glu accumulation outside nerve cells, resulting in the inhibition of the system xc− pathway. GSH depletion causes abnormalities in GPX4, leading to excessive accumulation of lipid peroxides. Alterations in these specific pathways and amino acid metabolism eventually lead to ferroptosis. This review explores the connection between AD and the ferroptosis signaling pathways and amino acid metabolism, potentially informing future AD diagnosis and treatment methodologies.
Collapse
Affiliation(s)
- Hongyue Ma
- Department of Neurology, Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, China
| | - Yan Dong
- Department of Neurology, Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, China
| | - Yanhui Chu
- College of Life Sciences, Mudanjiang Medical University, Mudanjiang, China
- Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Mudanjiang, China
| | - Yanqin Guo
- Department of Neurology, Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, China
- Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Mudanjiang, China
- *Correspondence: Yanqin Guo, ; Luxin Li,
| | - Luxin Li
- College of Life Sciences, Mudanjiang Medical University, Mudanjiang, China
- Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Mudanjiang, China
- *Correspondence: Yanqin Guo, ; Luxin Li,
| |
Collapse
|
29
|
Yin J, Fu J, Zhao Y, Xu J, Chen C, Zheng L, Wang B. Comprehensive Analysis of the Significance of Ferroptosis-Related Genes in the Prognosis and Immunotherapy of Oral Squamous Cell Carcinoma. Bioinform Biol Insights 2022; 16:11779322221115548. [PMID: 35966810 PMCID: PMC9373167 DOI: 10.1177/11779322221115548] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 05/26/2022] [Indexed: 12/09/2022] Open
Abstract
Oral squamous cell carcinoma (OSCC) is a life-threatening disease, associated
with poor prognosis and the absence of specific biomarkers. Studies have shown
that the ferroptosis-related genes (FRGs) can be used as tumor prognostic
markers. However, FRGs’ prognostic value in OSCC needs further exploration. In
our study, gene expression profile and clinical data of OSCC patients were
collected from a public domain. We performed univariate and multivariate Cox
regression analyses to construct a multigene signature. The Kaplan-Meier and
receiver operating characteristic (ROC) methods were used to test the
effectiveness of the signature, followed by the expression analysis of human
leukocyte antigen (HLA) and immune checkpoints. The Cox regression analysis
identified 4 hubs from 103 FRGs expressed in OSCC that were associated with
overall survival (OS). A risk model based on the 4 FRGs was established to
classify patients into high-risk and low-risk groups. Compared with the low-risk
group, the survival time of the high-risk group was significantly reduced.
According to the multivariate Cox regression analysis, the risk score acted as
an independent predictor for OS. The accuracy of the 4 FRGs risk predictive
model was confirmed by ROC curve analysis. Moreover, the low-risk group had the
characteristics of higher expression of HLA and immune checkpoints, a lower
tumor purity, and a higher immune infiltration, indicating a more sensitive
response to immunotherapy. The novel FRGs-OSCC risk score system can be used to
predict the prognosis of OSCC patients and their response to immunotherapy.
Collapse
Affiliation(s)
- Junhao Yin
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,College of Stomatology, Shanghai Jiao Tong University, Shanghai, China.,National Center for Stomatology, Shanghai, China.,National Clinical Research Center for Oral Disease, Shanghai, China.,Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Jiayao Fu
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,College of Stomatology, Shanghai Jiao Tong University, Shanghai, China.,National Center for Stomatology, Shanghai, China.,National Clinical Research Center for Oral Disease, Shanghai, China.,Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Yijie Zhao
- Department of Oral and Maxillofacial Surgery, Shanghai Stomatological Hospital, Fudan University, Shanghai, China
| | - Jiabao Xu
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,College of Stomatology, Shanghai Jiao Tong University, Shanghai, China.,National Center for Stomatology, Shanghai, China.,National Clinical Research Center for Oral Disease, Shanghai, China.,Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Changyu Chen
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,College of Stomatology, Shanghai Jiao Tong University, Shanghai, China.,National Center for Stomatology, Shanghai, China.,National Clinical Research Center for Oral Disease, Shanghai, China.,Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Lingyan Zheng
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,College of Stomatology, Shanghai Jiao Tong University, Shanghai, China.,National Center for Stomatology, Shanghai, China.,National Clinical Research Center for Oral Disease, Shanghai, China.,Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Baoli Wang
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,College of Stomatology, Shanghai Jiao Tong University, Shanghai, China.,National Center for Stomatology, Shanghai, China.,National Clinical Research Center for Oral Disease, Shanghai, China.,Shanghai Key Laboratory of Stomatology, Shanghai, China
| |
Collapse
|
30
|
Sun Y, Xue Z, Huang T, Che X, Wu G. Lipid metabolism in ferroptosis and ferroptosis-based cancer therapy. Front Oncol 2022; 12:941618. [PMID: 35978815 PMCID: PMC9376317 DOI: 10.3389/fonc.2022.941618] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 07/08/2022] [Indexed: 11/28/2022] Open
Abstract
Ferroptosis refers to iron-dependent, specialized, and regulated-necrosis mediated by lipid peroxidation, which is closely related to a variety of diseases, including cancer. Tumor cells undergo extensive changes in lipid metabolism, including lipid peroxidation and ferroptosis. Changes in lipid metabolism are critical for the regulation of ferroptosis and thus have important roles in cancer therapy. In this review, we introduce the characteristics of ferroptosis and briefly analyze the links between several metabolic mechanisms and ferroptosis. The effects of lipid peroxides, several signaling pathways, and the molecules and pathways involved in lipid metabolism on ferroptosis were extensively analyzed. Finally, our review highlights some ferroptosis-based treatments and presents some methods and examples of how these treatments can be combined with other treatments.
Collapse
Affiliation(s)
- Yonghao Sun
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Zuoxing Xue
- Department of Urology, Dalian University Affiliated Xinhua Hospital, Dalian, China
| | - Tao Huang
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xiangyu Che
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Guangzhen Wu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
31
|
Song L, Wu J, Fu H, Wu C, Tong X, Zhang M. Abnormally Expressed Ferroptosis-Associated FANCD2 in Mediating the Temozolomide Resistance and Immune Response in Glioblastoma. Front Pharmacol 2022; 13:921963. [PMID: 35754466 PMCID: PMC9213730 DOI: 10.3389/fphar.2022.921963] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 05/20/2022] [Indexed: 11/25/2022] Open
Abstract
Ferroptosis-related genes (FRGs) have been identified as potential targets involved in oncogenesis and cancer therapeutic response. Nevertheless, the specific roles and underlying mechanisms of FRGs in GBM and temozolomide (TMZ) resistance remain unclear. Through comprehensive bioinformatics, we found that ferroptosis-related Fanconi anemia complementation group D2 (FANCD2) was significantly up-regulated in GBM tissues, and the high expression level of FANCD2 was related to the poor prognosis in primary and recurrent GBM patients. Furthermore, FANCD2 could promote TMZ resistance by attenuating ferroptosis in GBM cells. Knockdown of FANCD2 could increase reactive oxygen species (ROS) levels and inhibit cell survival. The two characteristics were associated with ferroptosis in TMZ-resistant GBM cells T98G-R and U118-R. The Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis indicated that aberrantly expressed FANCD2 was potentially linked with several cancer-associated signaling pathways, including chromosome segregation, DNA replication, and cell cycle transition. In addition, we demonstrated that FANCD2 expression was positively correlated with several tumor-infiltrating lymphocytes (TILs) and multiple immune-associated signatures in GBM. Therefore, up-regulated FANCD2 could protect GBM cells from ferroptosis and promote TMZ resistance. FANCD2 may be a novel therapeutic target in GBM.
Collapse
Affiliation(s)
- Liying Song
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Jiali Wu
- Department of Otolaryngology, Hunan Want Want Hospital, Changsha, China
| | - Hua Fu
- Department of Pathology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Cuifang Wu
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Xiaopei Tong
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Mingyu Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
32
|
Han D, Yao Y, Chen L, Miao Z, Xu S. Apigenin ameliorates di(2-ethylhexyl) phthalate-induced ferroptosis: The activation of glutathione peroxidase 4 and suppression of iron intake. Food Chem Toxicol 2022; 164:113089. [PMID: 35500696 DOI: 10.1016/j.fct.2022.113089] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 03/28/2022] [Accepted: 04/26/2022] [Indexed: 10/18/2022]
Abstract
Di(2-ethylhexyl) phthalate (DEHP) is a widely artificial persistent organic pollutant, the contamination of which infiltrates daily human life from many aspects, imperceptibly causing damage to multiple organs in the body, including the liver. Apigenin (APG) is widely distributed in vegetables and fruits and can relieve or prevent the injuries caused by exogenous chemicals through various pharmacological effects, such as antioxidant effects. To investigate the mechanism of DEHP-induced liver injury and the antagonistic effects of APG, we treated AML12 cells with 1 mM DEHP and/or APG. Ultrastructural morphology analysis indicated that DEHP induced typical ferroptosis-like damage. In addition, we found that DEHP exposure induced ferroptosis by enhancing reactive oxygen species (ROS) levels, disrupting iron homeostasis and lipid peroxidation, and regulating the expression of ferroptosis-related genes. Notably, supplementation with APG significantly inhibited these abnormal changes, and molecular docking further showed evidence of the activating effects of APG ligand on glutathione peroxidase 4 (GPX4). These results demonstrated that the protective effects of APG on DEHP-induced ferroptosis were achieved by activating GPX4 and suppressing intracellular iron accumulation. This information not only adds to DEHP toxicological data but also provides a basis for the practical application of APG.
Collapse
Affiliation(s)
- Dongxu Han
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Yujie Yao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Lu Chen
- College of Animal Science, Tarim University, Alar, Xinjiang Uygur Autonomous Region, 843300, PR China
| | - Zhiying Miao
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China
| | - Shiwen Xu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China; Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China.
| |
Collapse
|
33
|
Peng B, Peng J, Kang F, Zhang W, Peng E, He Q. Ferroptosis-Related Gene MT1G as a Novel Biomarker Correlated With Prognosis and Immune Infiltration in Colorectal Cancer. Front Cell Dev Biol 2022; 10:881447. [PMID: 35517502 PMCID: PMC9065264 DOI: 10.3389/fcell.2022.881447] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 03/22/2022] [Indexed: 12/13/2022] Open
Abstract
Ferroptosis, a newly discovered way of cell death, has been proved to be involved in the oncogenesis and development of cancers, including colorectal cancer (CRC). Here, by identifying the differentially expressed genes (DEGs) from three CRC transcriptome microarray datasets (GSE20842, GSE23878, and GSE25070), we found that the expression of MT1G was significantly decreased in CRC tissues, and the patients with a high level of MT1G displayed a poor prognosis. Quantitative PCR (qPCR) further confirmed the downregulated MT1G in two CRC cells, HCT8 and HCT116. The colony-forming assay indicated that the MT1G overexpression exhibited a remarkable inhibition of cell proliferation in HCT8 and HCT116 cells. In addition, we explored the co-expressed genes of MT1G to gain a better understanding of its potential signaling pathways. Aberrantly expressed MT1G also affected the immune response of CRC patients. Collectively, these findings might deepen our comprehension on the potential biological implications of MT1G in CRC.
Collapse
Affiliation(s)
- Bi Peng
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Jinwu Peng
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
- Department of Pathology, Xiangya Changde Hospital, Changde, China
- *Correspondence: Qingchun He, ; Jinwu Peng,
| | - Fanhua Kang
- Department of Pathology, Xiangya Changde Hospital, Changde, China
| | - Wenqin Zhang
- Department of Pathology, Xiangya Changde Hospital, Changde, China
| | - Emin Peng
- Xiangya International Medical Center, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Qingchun He
- Department of Emergency, Xiangya Hospital, Central South University, Changsha, China
- Department of Emergency, Xiangya Changde Hospital, Changde, China
- *Correspondence: Qingchun He, ; Jinwu Peng,
| |
Collapse
|
34
|
Feng S, Zhou Y, Huang H, Lin Y, Zeng Y, Han S, Huang K, Liu Q, Zhu W, Yuan Z, Liang B. Nobiletin Induces Ferroptosis in Human Skin Melanoma Cells Through the GSK3β-Mediated Keap1/Nrf2/HO-1 Signalling Pathway. Front Genet 2022; 13:865073. [PMID: 35350242 PMCID: PMC8957809 DOI: 10.3389/fgene.2022.865073] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 02/21/2022] [Indexed: 12/19/2022] Open
Abstract
Melanoma is an aggressive malignant skin tumour with an increasing global incidence. However, current treatments have limitations owing to the acquired tumour drug resistance. Ferroptosis is a recently discovered form of programmed cell death characterised by iron accumulation and lipid peroxidation and plays a critical role in tumour growth inhibition. Recently, ferroptosis inducers have been regarded as a promising therapeutic strategy to overcome apoptosis resistance in tumour cells. In this study, we reported that nobiletin, a natural product isolated from citrus peel, and exhibited antitumour activity by inducing ferroptosis in melanoma cells. Subsequently, we further explored the potential mechanism of nobiletin-induced ferroptosis, and found that the expression level of glycogen synthase kinase 3β (GSK3β) in the skin tissue of patients with melanoma was significantly reduced compared to that in the skin of normal tissue. Additionally, nobiletin increased GSK3β expression in melanoma cells. Moreover, the level of Kelch-like Ech-associated protein-1 (Keap1) was increased, while the level of nuclear factor erythroid 2-related factor 2 (Nrf2), and haem oxygenase-1 (HO-1) was decreased in nobiletin-treated melanoma cells, suggesting that the antioxidant defence system was downregulated. Furthermore, knockdown of GSK3β significantly reduced nobiletin-induced ferroptosis and upregulated the Keap1/Nrf2/HO-1 signalling pathway, while the opposite was observed in cells overexpressing GSK3β. In addition, molecular docking assay results indicated that nobiletin showed strong binding affinities for GSK3β, Keap1, Nrf2, and HO-1. Taken together, our results demonstrated that nobiletin could induce ferroptosis by regulating the GSK3β-mediated Keap1/Nrf2/HO-1 signalling pathway in human melanoma cells. Hence, nobiletin stands as a promising drug candidate for melanoma treatment with development prospects.
Collapse
Affiliation(s)
- Senling Feng
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, Department of Pharmacy, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yongheng Zhou
- Department of Pharmacy, The Fourth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hongliang Huang
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, Department of Pharmacy, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ying Lin
- Guangdong Provincial Clinical Research Center for Chinese Medicine Dermatology, Department of Dermatology, Guangdong Provincial Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Yifeng Zeng
- Department of Pharmacy, The Fourth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shanshan Han
- Guangdong Provincial Clinical Research Center for Chinese Medicine Dermatology, Department of Dermatology, Guangdong Provincial Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Kaikai Huang
- Guangdong Provincial Clinical Research Center for Chinese Medicine Dermatology, Department of Dermatology, Guangdong Provincial Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Quanzhi Liu
- Guangdong Provincial Clinical Research Center for Chinese Medicine Dermatology, Department of Dermatology, Guangdong Provincial Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Wenting Zhu
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, Department of Pharmacy, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhongwen Yuan
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, Department of Pharmacy, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Baoying Liang
- Guangdong Provincial Clinical Research Center for Chinese Medicine Dermatology, Department of Dermatology, Guangdong Provincial Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| |
Collapse
|
35
|
Huang C, Zhan L. Network Pharmacology Identifies Therapeutic Targets and the Mechanisms of Glutathione Action in Ferroptosis Occurring in Oral Cancer. Front Pharmacol 2022; 13:851540. [PMID: 35359830 PMCID: PMC8963897 DOI: 10.3389/fphar.2022.851540] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 02/23/2022] [Indexed: 01/06/2023] Open
Abstract
Oral cancer (OC) is one of the most pernicious cancers with increasing incidence and mortality worldwide. Surgery is the primary approach for the treatment of early-stage OC, which reduces the quality of life of the patients. Therefore, there is an urgent need to discover novel treatments for OC. Targeting ferroptosis to induce cell death through the modulation of lipid oxidation has been used as a new approach to treat many cancers. Glutathione (GSH) is a coenzyme factor of GSH peroxidase 4, and it carries potential applicability in treating OC. By using network pharmacology and molecular docking followed by systematic bioinformatic analysis, we aimed to study GSH-targeting ferroptosis to treat OC. We identified 14 core molecular targets, namely, EGFR, PTGS2, HIF1A, VEGFA, TFRC, SLC2A1, CAV1, CDKN2A, SLC3A2, IFNG, NOX4, DDIT4, CA9, and DUSP1, involved in ferroptosis that were targeted by GSH for OC treatment. Functional characterization of these molecular targets showed their importance in the control of cell apoptosis, cell proliferation, and immune responses through various kinase activities such as the mitogen-activated protein kinase activity (e.g., ERK1 and ERK2 cascades) and modulation of TOR signaling (e.g., the HIF-1 signaling pathway). Molecular docking further revealed the direct binding of GSH with EGFR, PTGS2, and HIF1A proteins. These findings provide a novel insight into the targets of GSH in ferroptosis as well as possible molecular mechanisms involved, suggesting the possible use of GSH as a combined therapy for treating OC.
Collapse
Affiliation(s)
- Chen Huang
- The Center for Data Science in Health and Medicine, Business School, Qingdao University, Qingdao, China
| | - Lei Zhan
- Department of Ophthalmology, The Second People’s Hospital of Guilin, Guilin, China
| |
Collapse
|