1
|
Gao Y, Song Z, Gan W, Zou X, Bai Y, Zhao X, Chen D, Qiao M. Selective and iron-independent ferroptosis in cancer cells induced by manipulation of mitochondrial fatty acid oxidation. Biomaterials 2025; 320:123259. [PMID: 40112511 DOI: 10.1016/j.biomaterials.2025.123259] [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: 11/21/2024] [Revised: 02/20/2025] [Accepted: 03/14/2025] [Indexed: 03/22/2025]
Abstract
Despite the promise of ferroptosis in cancer therapy, selectively inducing robust ferroptosis in cancer cells remains a significant challenge. In this study, manipulation of fatty acids β-oxidation (FAO) by combination of mild photodynamic therapy (PDT) and inhibition of triglycerides (TGs) synthesis was found to induce robust and iron-independent ferroptosis in cancer cells with dysregulated lipid metabolism for the first time. To achieve that, TGs synthesis inhibitor of xanthohumol (Xan) and FAO initiator of tetrakis (4-carboxyphenyl) porphyrin (TCPP) were co-delivered by a nanoplexes composed of pH-responsive amphiphilic lipopeptide C18-pHis10 and DSPE-PEG2000. TCPP was found to rapidly increase the intracellular ROS under laser irradiation without inducing antioxidant response and apoptosis, activating the AMPK in cancer cells and accelerating mitochondrial FAO. Xan fueled the mitochondrial FAO with substrates by suppressing the conversion of fatty acids (FAs) to TGs. This also led to augmented intracellular polyunsaturated fatty acids (PUFAs) and PUFAs-phospholipids levels, increasing the intrinsic susceptibility of cancer cells to lipid peroxidization. As a result, the excessive ROS generated from the sustained mitochondrial FAO caused remarkably lipid peroxidation and ultimately ferroptosis. Collectively, our study provides a new approach to selectively induce iron-independent ferroptosis in cancer cells by taking advantage of dysregulated lipid metabolism.
Collapse
Affiliation(s)
- Yan Gao
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Zilin Song
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Wenxin Gan
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Xue Zou
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yaning Bai
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Xiuli Zhao
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Dawei Chen
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Mingxi Qiao
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| |
Collapse
|
2
|
Wang Y, He Y, Liang W, Kan S, Gao Y, Yang C, Han S, Ren Y, Nie K. Huangqin Decoction alleviates chemotherapy-induced intestinal injury by inhibiting ferroptosis via modulation of P53/SLC7A11/GPX4 pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 351:120135. [PMID: 40513923 DOI: 10.1016/j.jep.2025.120135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2025] [Revised: 06/03/2025] [Accepted: 06/09/2025] [Indexed: 06/16/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Huangqin Decoction (HQD), a traditional Chinese medicine formula for the treatment of heat-induced diarrhea and dysentery. HQD can significantly reduce chemotherapy induced intestinal toxicity. However, the underlying mechanism of HQD in alleviating chemotherapy-induced intestinal injury remains unclear. AIMS OF THE STUDY To explore the underlying mechanism of HQD against chemotherapy-induced intestinal injury through the P53/SLC7A11/GPX4 signaling pathway-mediated ferroptosis. MATERIALS AND METHODS Irinotecan (CPT-11) was used to establish chemotherapy-induced intestinal injury model in mice. The inflammatory damage of the jejunum and colon is evaluated by HE staining and ELISA. The degree of ferroptosis is validated by the iron deposition, the levels of ROS, MDA, SOD and GSH, and the changes in the distribution of 4-HNE and iron in the jejunum and colon. Expression of intestinal tight junction protein TJP-1 and Occludin was detected by immunofluorescence. The P53/SLC7A11/GPX4 signal pathway was examined by RT-qPCR and Western blot. RESULTS HQD significantly decreased diarrhea scores in chemotherapy mice, and improved the inflammatory pathological damage, with decreased levels of IL-1β and TNF-α. HQD also restored intestinal barrier function and intestinal tight junction protein TJP-1 and Occluding expression. Furthermore, HQD significantly suppressed ferroptosis, as indicated by the improvement of iron deposition, and oxidative stress. The PCR and Western blot results indicated that HQD could activate the P53/SLC7A11/GPX4 signaling pathway. CONCLUSION The occurrence of chemotherapy-induced intestinal injury is associated with ferroptosis-induced intestinal inflammation; HQD prevent chemotherapy-induced intestinal injury by activating the p53/SLC7A11/GPX4 signaling pathway, inhibiting ferroptosis, and alleviating intestinal inflammatory injury.
Collapse
Affiliation(s)
- Yusu Wang
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yunjing He
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Wan Liang
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Shaojun Kan
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yujie Gao
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Chenglu Yang
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Siyu Han
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yuke Ren
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Ke Nie
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|
3
|
Zhang Y, Zheng L, Che T, Meng N. Dysregulation of Calcium Homeostasis: An Important Factor Leading to Ferroptosis in Cardiovascular Diseases. Cell Biol Int 2025; 49:589-605. [PMID: 40042118 DOI: 10.1002/cbin.70012] [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: 07/11/2024] [Revised: 10/31/2024] [Accepted: 02/16/2025] [Indexed: 05/14/2025]
Abstract
Cardiovascular disease is a circulatory system disease involving the heart and blood vessels, which is one of the main causes of human health loss and even life-threatening. Ca2+ is an important signal molecule. Free calcium ions in the cytoplasm are involved in various physiological and biochemical reactions of cells. Ferroptosis is a programmed cell death driven by lipid peroxidation dependent on free ferrous ions. The essence of ferroptosis is the accumulation of lipid peroxide caused by the increase of intracellular ferrous ion content, which leads to the damage of the phospholipid membrane and eventually cell death. Studies have shown that calcium homeostasis and ferroptosis are involved in the occurrence and development of cardiovascular diseases, but the relationship between them remains to be clarified. This article reviews the various pathways regulating calcium homeostasis in cells and the occurrence and development mechanism of ferroptosis, and discusses the relationship between the two in cardiovascular diseases, which is expected to provide novel and important strategies for alleviating and treating cardiovascular diseases.
Collapse
Affiliation(s)
- Yifan Zhang
- School of Biological Science and Technology, University of Jinan, Jinan, China
| | - Leiyin Zheng
- School of Biological Science and Technology, University of Jinan, Jinan, China
| | - Tongtong Che
- School of Biological Science and Technology, University of Jinan, Jinan, China
| | - Ning Meng
- School of Biological Science and Technology, University of Jinan, Jinan, China
| |
Collapse
|
4
|
Ajam-Hosseini M, Babashah S. Exploring ferroptosis and miRNAs: implications for cancer modulation and therapy. Mol Cell Biochem 2025; 480:3455-3476. [PMID: 39869280 DOI: 10.1007/s11010-024-05169-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 11/16/2024] [Indexed: 01/28/2025]
Abstract
Ferroptosis is a novel, iron-dependent form of non-apoptotic cell death characterized by the accumulation of lipid reactive oxygen species (ROS) and mitochondrial shrinkage. It is closely associated with the onset and progression of various diseases, especially cancer, at all stages, making it a key focus of research for developing therapeutic strategies. Numerous studies have explored the role of microRNAs (miRNAs) in regulating ferroptosis by modulating the expression of critical genes involved in iron metabolism and lipid peroxidation. Due to their diversity, unique properties, and dynamic expression patterns in diseases, exosomal miRNAs are emerging as promising biomarkers. Exosomes act as biological messengers, delivering miRNAs to target cells through specific internalization, thus influencing the ferroptosis response in recipient cells. This review summarizes the roles of miRNAs, with particular focus on exosomal miRNAs, in ferroptosis and their implications for cancer pathology. By examining the molecular mechanisms of miRNAs, we aim to provide valuable insights into potential therapeutic approaches.
Collapse
Affiliation(s)
- Mobarakeh Ajam-Hosseini
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, P.O. Box: 14115-154, Tehran, Iran
| | - Sadegh Babashah
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, P.O. Box: 14115-154, Tehran, Iran.
| |
Collapse
|
5
|
Ji G, Yuan W, Wang X, Li W, Sun Z, Wei Z, Zhou L, Hu H. 5-Fluorouracil induces ferroptosis in breast cancer cells via targeting SLC7A11. Biochem Biophys Res Commun 2025; 770:151972. [PMID: 40378619 DOI: 10.1016/j.bbrc.2025.151972] [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: 01/31/2025] [Revised: 04/03/2025] [Accepted: 05/07/2025] [Indexed: 05/19/2025]
Abstract
Breast cancer (BC) is one of the major causes of cancer mortality worldwide among women. 5-Fluorouracil (5-FU) is a widely used chemotherapy drug to treat breast cancer, which is unclear that the mechanism of inhibiting BC. Ferroptosis is a mode of programmed cell death determined by iron-dependent lipid peroxidation. The aim of the study was to investigate whether ferroptosis is involved in 5-FU-induced BC cell injury. In the current study, we found that iron metabolism and SLC7A11/GPX4 signaling may play a key role in cell death of BC induced by 5-FU in vitro. In vitro experiments, we found that 5-FU exposure significantly increased the levels of iron and reactive oxygen species (ROS) in MCF-7 and MDA-MB-231 cells. Furthermore, ferrostatin-1, the ferroptosis inhibitor, inhibited cell death induced by 5-FU. Subsequent western blotting, qRT-PCR, and measurement of various kits, fluorescence staining as well as cellular thermal shift assay, confirmed the results that 5-FU induces ferroptosis by targeting SLC7A11 in BC cells. In conclusion, the results in our study reveals that 5-FU exposure leads to ferroptosis in BC cells via targeting inhibition of SLC7A11/GPX4 signaling pathway, which offers novel insight in pharmacodynamic effect and mechanism of 5-FU in therapeutic avenues of BC.
Collapse
Affiliation(s)
- Guojie Ji
- Key Laboratory of Fertility Preservation, School of Life Sciences and Technologies, Sanquan College of Xinxiang Medical University, Xinxiang, 453003, Henan Province, PR China.
| | - Wenzheng Yuan
- Key Laboratory of Fertility Preservation, School of Life Sciences and Technologies, Sanquan College of Xinxiang Medical University, Xinxiang, 453003, Henan Province, PR China.
| | - Xiaoyi Wang
- Key Laboratory of Fertility Preservation, School of Life Sciences and Technologies, Sanquan College of Xinxiang Medical University, Xinxiang, 453003, Henan Province, PR China.
| | - Wenmi Li
- Key Laboratory of Fertility Preservation, School of Life Sciences and Technologies, Sanquan College of Xinxiang Medical University, Xinxiang, 453003, Henan Province, PR China.
| | - Zhibin Sun
- Key Laboratory of Fertility Preservation, School of Life Sciences and Technologies, Sanquan College of Xinxiang Medical University, Xinxiang, 453003, Henan Province, PR China.
| | - Ziyu Wei
- Key Laboratory of Fertility Preservation, School of Life Sciences and Technologies, Sanquan College of Xinxiang Medical University, Xinxiang, 453003, Henan Province, PR China.
| | - Liuyang Zhou
- Key Laboratory of Fertility Preservation, School of Life Sciences and Technologies, Sanquan College of Xinxiang Medical University, Xinxiang, 453003, Henan Province, PR China.
| | - Huanhuan Hu
- Key Laboratory of Fertility Preservation, School of Life Sciences and Technologies, Sanquan College of Xinxiang Medical University, Xinxiang, 453003, Henan Province, PR China.
| |
Collapse
|
6
|
Kumari R, Banerjee S. Regulation of Different Types of Cell Death by Noncoding RNAs: Molecular Insights and Therapeutic Implications. ACS Pharmacol Transl Sci 2025; 8:1205-1226. [PMID: 40370994 PMCID: PMC12070317 DOI: 10.1021/acsptsci.4c00681] [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: 11/25/2024] [Revised: 04/23/2025] [Accepted: 04/24/2025] [Indexed: 05/16/2025]
Abstract
Noncoding RNAs (ncRNAs) are crucial regulatory molecules in various biological processes, despite not coding for proteins. ncRNAs are further divided into long noncoding RNAs (lncRNAs), microRNAs (miRNAs), and circular RNAs (circRNAs) based on the size of their nucleotides. These ncRNAs play crucial roles in transcriptional, post-transcriptional, and epigenetic regulation. The regulatory roles of noncoding RNAs, including lncRNAs, miRNAs, and circRNAs, are essential in various modalities of cellular death, such as apoptosis, ferroptosis, cuproptosis, pyroptosis, disulfidptosis, and necroptosis. These noncoding RNAs are integral to modulating gene expression and protein functionality during cellular death mechanisms. In apoptosis, lncRNAs, miRNAs, and circRNAs influence the transcription of apoptotic genes. In ferroptosis, these noncoding RNAs target genes and proteins involved in iron homeostasis and oxidative stress responses. For cuproptosis, noncoding RNAs regulate pathways associated with the accumulation of copper ions, leading to cellular death. During pyroptosis, noncoding RNAs modulate inflammatory mediators and caspases, affecting the proinflammatory cell death pathway. In necroptosis, noncoding RNAs oversee the formation and functionality of necrosomes, thereby influencing the balance between cellular survival and death. Disulfidptosis is a unique type of regulated cell death caused by the excessive formation of disulfide bonds within cells, leading to cytoskeletal collapse and oxidative stress, especially under glucose-limited conditions. This investigation highlights the complex mechanisms through which noncoding RNAs coordinate cellular death, emphasizing their therapeutic promise as potential targets, particularly in the domain of cancer treatment.
Collapse
Affiliation(s)
- Reshmi Kumari
- Department of Biotechnology, School
of Biosciences and Technology, VIT University, Vellore 632014, Tamil Nadu, India
| | - Satarupa Banerjee
- Department of Biotechnology, School
of Biosciences and Technology, VIT University, Vellore 632014, Tamil Nadu, India
| |
Collapse
|
7
|
Pourhabib Mamaghani M, Mousavikia SN, Azimian H. Ferroptosis in cancer: Mechanisms, therapeutic strategies, and clinical implications. Pathol Res Pract 2025; 269:155907. [PMID: 40101548 DOI: 10.1016/j.prp.2025.155907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 02/10/2025] [Accepted: 03/08/2025] [Indexed: 03/20/2025]
Abstract
The resistance of cancer cells to existing treatments has become a major challenge for researchers despite advancements in cancer treatment. Studies have shown that this resistance is due to cancer cells evading apoptosis. Moreover, the most common form of cell death induced by chemotherapy and radiotherapy is apoptosis. One of the most essential mechanisms cancer cells escape apoptosis is the excessive expression of tumors' apoptosis inhibitors. Therefore, finding a non-apoptotic pathway that bypasses apoptosis could be a hopeful strategy for cancer treatment. Ferroptosis has been identified as a non-apoptotic and regulated cell death process characterized by the accumulation of lipid peroxides and iron-dependent reactive oxygen species (ROS). Although studies have shown that ferroptosis plays a role in the development of many diseases, including cancer, it also has the potential to decrease resistance to current treatments, such as chemotherapy. Additionally, research has shown that ferroptosis successfully kills cancer cells, such as breast, stem, and lung cancer cells. Therefore, ferroptosis can be identified as a beneficial therapeutic mechanism for cancer treatment. Although ferroptosis has been introduced as an effective treatment path for cancer, its role, along with its therapeutic inducers, in increasing the therapeutic effect has not been investigated. In this review, we aim to introduce ferroptosis, compare it with other cell deaths known so far, and explain its role in cancer treatment. We believe that ferroptosis can be widely used to overcome cancer cells.
Collapse
Affiliation(s)
- Mina Pourhabib Mamaghani
- Department of Medical Physics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyedeh Nasibeh Mousavikia
- Department of Medical Physics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Medical Physics Research Center, Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hosein Azimian
- Department of Medical Physics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Medical Physics Research Center, Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
8
|
Xu Y, Ren M, Deng R, Meng J, Xu L, Zhao W, Ni Y, Mao C, Zhang S. UCNPs@PVP-Hemin-GOx@CaCO 3 Nanoplatform for Ferroptosis Self-Amplification Combined with Calcium Overload. Adv Healthc Mater 2025; 14:e2404215. [PMID: 40072332 DOI: 10.1002/adhm.202404215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 02/27/2025] [Indexed: 04/26/2025]
Abstract
Due to the complexity of the tumor microenvironment (TME), current tumor treatments cannot achieve satisfactory results. A nanocomposite material, UCNPs@PVP-Hemin-GOx@CaCO3 (UPHGC NPs) is developed that responds to the TME and controls release to achieve multimodal synergistic therapy in tumor tissues. UPHGC NPs mediate photodynamic therapy (PDT), chemodynamic therapy (CDT), and starvation therapy (ST) synergistically, ultimately inducing self-amplification of ferroptosis. The Hemin loaded in UPHGC NPs exhibits peroxidase (POD) activity, which can react with H2O2 to produce ·OH (CDT) and generate the maximum amount of ·O2 - (PDT) under UV excitation from upconversion materials. Hemin can also consume glutathione (GSH), downregulate glutathione peroxidase 4 (GPX4), and combine with PDT/CDT to induce lipid peroxidation (LPO), leading to ferroptosis. In addition, Glucose oxidase (GOx) provides sufficient H2O2 for the ·OH production, amplifying ROS generation to further enhance ferroptosis. The gluconic acid produced by GOx during the ST process synergizes with the TME's acidic conditions to promote Ca2+ release, induce intracellular calcium overload, enhance oxidative stress, lead to mitochondrial dysfunction, and ultimately kill tumor cells through mitochondrial damage. Furthermore, the externally mineralized calcium carbonate can prevent premature drug release in normal tissues.
Collapse
Affiliation(s)
- Yuping Xu
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China
| | - Mingming Ren
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China
| | - Runzhi Deng
- Nanjing Stomatological Hospital, Affliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Jiajia Meng
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China
| | - Lingxia Xu
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China
| | - Wenbo Zhao
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China
| | - Yanhong Ni
- Nanjing Stomatological Hospital, Affliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Chun Mao
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China
| | - Shirong Zhang
- Molecular Diagnostic Center, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Hangzhou First People's Hospital, Hangzhou, 310006, China
| |
Collapse
|
9
|
Cui W, Hao M, Yang X, Yin C, Chu B. Gut microbial metabolism in ferroptosis and colorectal cancer. Trends Cell Biol 2025; 35:341-351. [PMID: 39261152 DOI: 10.1016/j.tcb.2024.08.006] [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: 05/15/2024] [Revised: 08/14/2024] [Accepted: 08/16/2024] [Indexed: 09/13/2024]
Abstract
Ferroptosis is programmed cell death induced by iron-driven lipid peroxidation. Numerous studies have shown that ferroptosis is implicated in the progression of colorectal cancer (CRC) and has emerged as a promising strategy to combat therapy-resistant CRC. While the intrinsic antiferroptotic and proferroptotic pathways in CRC cells have been well characterized, extrinsic metabolism pathways regulating ferroptosis in CRC pathogenesis remain less understood. Emerging evidence shows that gut microbial metabolism is tightly correlated with the progression of CRC. This review provides an overview of gut microbial metabolism and discusses how these metabolites derived from intestinal microflora contribute to cancer plasticity through ferroptosis. Targeting gut microbe-mediated ferroptosis is a potential approach for CRC treatment.
Collapse
Affiliation(s)
- Weiwei Cui
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Meng Hao
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen 518107, China
| | - Xin Yang
- Jiangsu Key Laboratory of Infection and Immunity, The Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215123, China.
| | - Chengqian Yin
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen 518107, China.
| | - Bo Chu
- Department of Cell Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China.
| |
Collapse
|
10
|
Li H, Li JX, Zeng YD, Zheng CX, Dai SS, Yi J, Song XD, Liu T, Liu WH. Luteolin ameliorates ischemic/reperfusion injury by inhibiting ferroptosis. Metab Brain Dis 2025; 40:159. [PMID: 40138029 DOI: 10.1007/s11011-025-01588-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 03/13/2025] [Indexed: 03/29/2025]
Abstract
Ischaemic stroke is a large disease burden worldwide. Thrombolysis and thrombectomy are the main treatment methods for cerebral ischemia-reperfusion (I/R) injury. Luteolin, as a flavonoid compound, has an antagonistic effect on inflammation, oxidative stress, and tumorigenesis in disease. Therefore, the primary objective of this study is to determine the role of luteolin in cerebral I/R injury. Oxygen glucose deprivation/reoxygenation (OGD/R)-treated BV2 cells and a cerebral I/R rat model were established. Cell viability and death were verified using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay and propidium iodide staining. The glutathione/oxidized glutathione (GSH/GSSG) ratio, superoxide dismutase (SOD) activity, and reactive oxygen species (ROS) and malondialdehyde (MDA) levels were determined using corresponding kits. Solute Carrier Family 7 Member 11 (SLC7A11), nuclear transcription factor erythroid 2-related factor 2 (NRF2), and glutathione peroxidase 4 (GPX4) levels were analyzed by western blotting. In addition, the infarct volume of brain tissues was examined by tetrazolium chloride (TTC) staining. Luteolin treatment significantly enhanced cell viability, decreased LDH release and intracellular ROS and MDA levels, and increased the GSH/GSSG ratio and SOD activity in OGD/R-treated BV2 cells. PI staining demonstrated that cell death was inhibited after luteolin treatment. Additionally, luteolin treatment significantly increased the SLC7A11, NRF2, and GPX4 protein levels. After treatment with ML385, an NRF2 inhibitor, the influence of luteolin on OGD/R-treated BV2 cells was reversed. Moreover, luteolin treatment significantly decreased the neurological score and infarct area in the brain tissues of cerebral I/R rats. Our research demonstrated that luteolin treatment inhibited ferroptosis by enhancing antioxidant functions through the NRF2 pathway, which provides a promising method for treating cerebral I/R injury.
Collapse
Affiliation(s)
- Hua Li
- Hunan University of Traditional Chinese Medicine Cultivates World First-class Disciplines of Traditional Chinese Medicine, Changsha, Hunan, 410208, China
- The Key Laboratory of Diagnostics of TCM in Hunan Province, Changsha, Hunan, 410208, China
- Key Laboratory of TCM Heart and Lung Syndrome Differentiation & Medicated Diet and Dietotherapy, Changsha, Hunan, 410208, China
| | - Jin-Xia Li
- Hunan University of Traditional Chinese Medicine Cultivates World First-class Disciplines of Traditional Chinese Medicine, Changsha, Hunan, 410208, China
- The Key Laboratory of Diagnostics of TCM in Hunan Province, Changsha, Hunan, 410208, China
| | - Yi-di Zeng
- Hunan University of Traditional Chinese Medicine Cultivates World First-class Disciplines of Traditional Chinese Medicine, Changsha, Hunan, 410208, China
- The Key Laboratory of Diagnostics of TCM in Hunan Province, Changsha, Hunan, 410208, China
| | - Cai-Xing Zheng
- Hunan University of Traditional Chinese Medicine Cultivates World First-class Disciplines of Traditional Chinese Medicine, Changsha, Hunan, 410208, China
- The Key Laboratory of Diagnostics of TCM in Hunan Province, Changsha, Hunan, 410208, China
| | - Si-Si Dai
- Hunan University of Traditional Chinese Medicine Cultivates World First-class Disciplines of Traditional Chinese Medicine, Changsha, Hunan, 410208, China
- The Key Laboratory of Diagnostics of TCM in Hunan Province, Changsha, Hunan, 410208, China
| | - Jian Yi
- Hunan University of Traditional Chinese Medicine Cultivates World First-class Disciplines of Traditional Chinese Medicine, Changsha, Hunan, 410208, China
| | - Xu-Dong Song
- Hunan University of Traditional Chinese Medicine Cultivates World First-class Disciplines of Traditional Chinese Medicine, Changsha, Hunan, 410208, China
- The Key Laboratory of Diagnostics of TCM in Hunan Province, Changsha, Hunan, 410208, China
| | - Ting Liu
- Hunan University of Traditional Chinese Medicine Cultivates World First-class Disciplines of Traditional Chinese Medicine, Changsha, Hunan, 410208, China
- The Key Laboratory of Diagnostics of TCM in Hunan Province, Changsha, Hunan, 410208, China
| | - Wang-Hua Liu
- Hunan University of Traditional Chinese Medicine Cultivates World First-class Disciplines of Traditional Chinese Medicine, Changsha, Hunan, 410208, China.
- The Key Laboratory of Diagnostics of TCM in Hunan Province, Changsha, Hunan, 410208, China.
- Key Laboratory of TCM Heart and Lung Syndrome Differentiation & Medicated Diet and Dietotherapy, Changsha, Hunan, 410208, China.
- Hunan Engineering Technology Research Center For Medicinal and Functional Food, Hunan University of Traditional Chinese Medicine, XueShi Road, Changsha, Hunan, 410208, China.
| |
Collapse
|
11
|
Scattolin T, Cavarzerani E, Alessi D, Mauceri M, Botter E, Tonon G, Caligiuri I, Repetto O, Kamensek U, Brezar SK, Dalla Pozza M, Palazzolo S, Cemazar M, Canzonieri V, Demitri N, Nolan SP, Gasser G, Visentin F, Rizzolio F. Unlocking the potential of organopalladium complexes for high-grade serous ovarian cancer therapy. Dalton Trans 2025; 54:4685-4696. [PMID: 39967474 DOI: 10.1039/d5dt00194c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2025]
Abstract
High-Grade Serous Ovarian Cancer (HGSOC) is the most common and lethal subtype of ovarian cancer, known for its high aggressiveness and extensive genomic alterations. Typically diagnosed at an advanced stage, HGSOC presents formidable challenges in drug therapy. The limited efficacy of standard treatments, development of chemoresistance, scarcity of targeted therapies, and significant tumor heterogeneity render this disease incurable with current treatment options, highlighting the urgent need for novel therapeutic approaches to improve patient outcomes. In this study we report a straightforward and stereoselective synthetic route to novel Pd(II)-vinyl and -butadienyl complexes bearing a wide range of monodentate and bidentate ligands. Most of the synthesized complexes exhibited good to excellent in vitro anticancer activity against ovarian cancer cells. Particularly promising is the water-soluble complex bearing two PTA (1,3,5-triaza-7-phosphaadamantane) ligands and the Pd(II)-butadienyl fragment. This compound combines excellent cytotoxicity towards cancer cells with substantial inactivity towards non-cancerous ones. This derivative was selected for further studies on ex vivo tumor organoids and in vivo mouse models, which demonstrate its remarkable efficacy with surprisingly low collateral toxicity even at high dosages. Moreover, this class of compounds appears to operate through a ferroptotic mechanism, thus representing the first such example for an organopalladium compound.
Collapse
Affiliation(s)
- Thomas Scattolin
- Dipartimento di Scienze Chimiche, Università degli Studi di Padova, via Marzolo 1, 35131 Padova, Italy.
| | - Enrico Cavarzerani
- Dipartimento di Scienze Molecolari e Nanosistemi, Università Ca' Foscari, Campus Scientifico Via Torino 155, 30174 Venezia-Mestre, Italy.
| | - Dario Alessi
- Dipartimento di Scienze Chimiche, Università degli Studi di Padova, via Marzolo 1, 35131 Padova, Italy.
| | - Matteo Mauceri
- Dipartimento di Scienze Chimiche, Università degli Studi di Padova, via Marzolo 1, 35131 Padova, Italy.
| | - Eleonora Botter
- Dipartimento di Scienze Molecolari e Nanosistemi, Università Ca' Foscari, Campus Scientifico Via Torino 155, 30174 Venezia-Mestre, Italy.
| | - Giovanni Tonon
- Dipartimento di Scienze Molecolari e Nanosistemi, Università Ca' Foscari, Campus Scientifico Via Torino 155, 30174 Venezia-Mestre, Italy.
| | - Isabella Caligiuri
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (C.R.O.) IRCCS, via Franco Gallini 2, 33081, Aviano, Italy
| | - Ombretta Repetto
- Immunopathology and Cancer Biomarkers, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, via Franco Gallini 2, 33081, Aviano, Italy
| | - Urska Kamensek
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Ljubljana, 1000, Slovenia
| | - Simona Kranjc Brezar
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Ljubljana, 1000, Slovenia
| | - Maria Dalla Pozza
- ChimieParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology, 75005, Paris, France
| | - Stefano Palazzolo
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (C.R.O.) IRCCS, via Franco Gallini 2, 33081, Aviano, Italy
| | - Maja Cemazar
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Ljubljana, 1000, Slovenia
| | - Vincenzo Canzonieri
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (C.R.O.) IRCCS, via Franco Gallini 2, 33081, Aviano, Italy
- Department of Medical, Surgical and Health Sciences, Università degli Studi di Trieste, Strada di Fiume 447, Trieste, Italy
| | - Nicola Demitri
- Area Science Park, Elettra-Sincrotrone Trieste, S.S. 14 Km 163.5, Basovizza, 34149, Trieste, Italy
| | - Steven P Nolan
- Department of Chemistry and Centre for Sustainable Chemistry, Ghent University Krijgslaan 281, S-3, 9000 Ghent, Belgium
| | - Gilles Gasser
- ChimieParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology, 75005, Paris, France
| | - Fabiano Visentin
- Dipartimento di Scienze Molecolari e Nanosistemi, Università Ca' Foscari, Campus Scientifico Via Torino 155, 30174 Venezia-Mestre, Italy.
| | - Flavio Rizzolio
- Dipartimento di Scienze Molecolari e Nanosistemi, Università Ca' Foscari, Campus Scientifico Via Torino 155, 30174 Venezia-Mestre, Italy.
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (C.R.O.) IRCCS, via Franco Gallini 2, 33081, Aviano, Italy
| |
Collapse
|
12
|
Chatterjee T, Zarjou A. Navigating the Complex Pathogenesis of Acute Kidney Injury: Exploring Macrophage Dynamics, Mitochondrial Dysfunction, and Ferroptosis Pathways. ADVANCES IN KIDNEY DISEASE AND HEALTH 2025; 32:122-132. [PMID: 40222799 PMCID: PMC11999248 DOI: 10.1053/j.akdh.2024.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 12/02/2024] [Accepted: 12/13/2024] [Indexed: 04/15/2025]
Abstract
Acute kidney injury, a rapid decline in kidney function coupled with physiological and homeostatic perturbations, is an independent risk factor for both short-term and long-term health outcomes. As incidence of acute kidney injury continues to rise globally, the significant clinical and economic challenge of acute kidney injury underscores the need for its prompt recognition and application of novel and germane strategies to reduce its severity and facilitate recovery. Understanding the multifaceted cascade of events engaged in pathogenesis of acute kidney injury is pivotal for the development of effective preventive and therapeutic strategies. To facilitate an in-depth discussion on emerging therapeutic targets, this review will examine the role of macrophages in kidney injury and repair, explore the alterations in mitochondrial biogenesis dynamics induced by acute kidney injury, and provide insights into the molecular mechanisms underlying the contribution of ferroptosis to kidney injury.
Collapse
Affiliation(s)
- Tanima Chatterjee
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Abolfazl Zarjou
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL.
| |
Collapse
|
13
|
Yang G, Qian B, He L, Zhang C, Wang J, Qian X, Wang Y. Application prospects of ferroptosis in colorectal cancer. Cancer Cell Int 2025; 25:59. [PMID: 39984914 PMCID: PMC11846473 DOI: 10.1186/s12935-025-03641-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 01/07/2025] [Indexed: 02/23/2025] Open
Abstract
Colorectal cancer (CRC) is a serious threat to human health with the third morbidity and the second cancer-related mortality worldwide. It is urgent to explore more effective strategy for CRC because of the acquired treatment resistance from the non-surgical conventional therapies, including radiation, chemotherapy, targeted therapy and immunotherapy. Ferroptosis is a novel form of programmed cell death characterized by iron-dependent lipid peroxidation species (ROS) accumulation and has been identified as a promising target for cancer treatment, especially for those with treatment resistance. In this review, we mainly summarize the recent studies on the influence and regulation of ferroptosis by which (including gut microbiota) modulating the metabolism of iron, amino acid and lipid. Thus this analysis may provide potential targets for inducing CRC ferroptosis and shed lights on the future application of ferroptosis in CRC.
Collapse
Affiliation(s)
- Gen Yang
- Department of Pathology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Boning Qian
- Department of Pathology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Liya He
- Department of Pathology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Chi Zhang
- Department of Pathology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Jianqiang Wang
- Department of Pathology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Xinlai Qian
- Department of Pathology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453003, Henan, China.
- Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, Henan, China.
- Henan Provincial Key Laboratory of Molecular Oncologic Pathology, Xinxiang, Henan, China.
| | - Yongxia Wang
- Department of Pathology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453003, Henan, China.
- Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, Henan, China.
- Henan Provincial Key Laboratory of Molecular Oncologic Pathology, Xinxiang, Henan, China.
| |
Collapse
|
14
|
Wang Y, Wang W, Zhang Y, Fleishman JS, Wang H. Targeting ferroptosis offers therapy choice in sepsis-associated acute lung injury. Eur J Med Chem 2025; 283:117152. [PMID: 39657462 DOI: 10.1016/j.ejmech.2024.117152] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 10/06/2024] [Accepted: 12/06/2024] [Indexed: 12/12/2024]
Abstract
Sepsis-associated acute lung injury (SALI) is a common complication of sepsis, consisting of a dysfunctional host response to infection-mediated heterogenous complexes. SALI is reported in up to 50 % of patients with sepsis and causes poor outcomes. Despite high incidence, there is a lack of understanding in its pathogenesis and optimal treatment. A better understanding of the molecular mechanisms underlying SALI may help produce better therapeutics. The effects of altered cell-death mechanisms, such as non-apoptotic regulated cell death (RCD) (i.e., ferroptosis), on the development of SALI are beginning to be discovered, while targeting ferroptosis as a meaningful target in SALI is increasingly being recognized. Here, we outline how a susceptible lung alveoli may develop SALI. Then we discuss the general mechanisms underlying ferroptosis, and how it contributes to SALI. We then outline the chemical structures of the emerging agents or compounds that can protect against SALI by inhibiting ferroptosis, summarizing their potential pharmacological effects. Finally, we highlight key limitations and possible strategies to overcome them. This review suggests that a detailed mechanistic and biological understanding of ferroptosis can foster the development of pharmacological antagonists in the treatment of SALI.
Collapse
Affiliation(s)
- Yu Wang
- Department of Geriatrics, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, China
| | - Weixue Wang
- Department of Geriatrics, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, China
| | - Yi Zhang
- Department of Geriatrics, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, China
| | - Joshua S Fleishman
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA.
| | - Hongquan Wang
- Department of Geriatrics, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, China.
| |
Collapse
|
15
|
Tao R, Li Y, Gong S, Zhang Q, Zhu Z. Unveiling intricating roles and mechanisms of ferroptosis in melanoma. Biochim Biophys Acta Rev Cancer 2025; 1880:189234. [PMID: 39644939 DOI: 10.1016/j.bbcan.2024.189234] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 11/29/2024] [Accepted: 12/01/2024] [Indexed: 12/09/2024]
Abstract
Melanoma is a highly invasive malignant tumor originating from melanocytes, with increasing incidence in recent years. Ferroptosis is an iron-dependent and non-apoptotic form of programmed cell death characterized by the accumulation of lipid peroxides and reactive oxygen species. Morphologically, ferroptosis exhibits the alteration in cells, such as reduced mitochondrial volume, increased density of bilayer membrane, and a decrease or disappearance of mitochondrial cristae. Ferroptosis has shown tremendous potential and applicability in regulating the development of melanoma. As melanoma progresses, certain biomarkers associated with ferroptosis display characteristic patterns of expression. These changes not only reveal the sensitivity of tumor cells to ferroptosis but also provide potential targets for diagnosis and treatment. Besides, inducing ferroptosis has been well-documented to inhibit the growth of melanoma and enhance the efficacy of tumor immunotherapy. Hence, this review emphasizes the roles and regulatory mechanisms of ferroptosis in melanoma development, the involved immune regulation, as well as the potential for diagnosis and treatment of melanoma. The continuous explorations will endow novel strategies for developing ferroptosis-based therapies for melanoma.
Collapse
Affiliation(s)
- Rui Tao
- Department of Plastic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Yichuan Li
- Department of Dermatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Song Gong
- Division of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China.
| | - Qi Zhang
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China; Xianning Medical College, Hubei University of Science & Technology, Xianning 437000, Hubei Province, China.
| | - Zhanyong Zhu
- Department of Plastic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China.
| |
Collapse
|
16
|
Sae-Fung A, Vinayavekhin N, Fadeel B, Jitkaew S. ACSL3 is an unfavorable prognostic marker in cholangiocarcinoma patients and confers ferroptosis resistance in cholangiocarcinoma cells. NPJ Precis Oncol 2024; 8:284. [PMID: 39706856 DOI: 10.1038/s41698-024-00783-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 12/10/2024] [Indexed: 12/23/2024] Open
Abstract
Cholangiocarcinoma (CCA) is a bile duct malignancy. Our previous comprehensive analysis showed that ferroptosis-related genes can stratify CCA patients into low-risk and high-risk groups based on survival time. Here, we explored the role of ferroptosis in CCA by analyzing mRNA expression in CCA patients from public databases. We identified acyl-CoA synthetase long chain family member 3 (ACSL3) as a potential ferroptosis suppressor in high-risk CCA patients. Using a panel of CCA cell lines, we confirmed ACSL3 upregulation in CCA cell lines associated with high-risk CCA, correlating this with resistance to the ferroptosis inducer RSL3. Lipidomic analysis revealed increased monounsaturated fatty acid (MUFA)-containing phospholipids in resistant cell lines. ACSL3 silencing sensitized these cells to RSL3. Resistance to ferroptosis was also dependent on exogenous MUFAs and was enhanced by lipid droplet biogenesis inhibition. These findings highlight ACSL3 as a promising target for therapeutic strategies aimed at overcoming ferroptosis resistance in CCA.
Collapse
Affiliation(s)
- Apiwit Sae-Fung
- Graduate Program in Clinical Biochemistry and Molecular Medicine, Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
- Division of Molecular Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Nawaporn Vinayavekhin
- Center of Excellence in Natural Products Chemistry, Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | - Bengt Fadeel
- Division of Molecular Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Siriporn Jitkaew
- Center of Excellence for Cancer and Inflammation, Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand.
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand.
| |
Collapse
|
17
|
Selyutina OY, Ul’yanova MA, Chinak OA, Timoshnikov VA, Fedenok LG, Stepanov AA, Yanshole VV, Kulik LV, Vasilevsky SF, Polyakov NE, Kontoghiorghes GJ. Novel Anthraquinone Derivatives and Their Complexes with Metal Ions with Anticancer Activity: Structure/Redox and Chelation Activity Correlations. Pharmaceuticals (Basel) 2024; 17:1717. [PMID: 39770559 PMCID: PMC11678833 DOI: 10.3390/ph17121717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 12/13/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
Background/Objectives: Some specific anthraquinone derivatives (AQs) are known to be used widely as effective chemotherapeutic agents in the treatment of cancer. However, their fundamental shortcoming is the high rate of cardiotoxicity observed in treated patients, which is thought to be caused by the increase in production of reactive oxygen species (ROS) catalyzed by iron and copper. The development of improved AQs and other anticancer drugs with enhanced efficacy but reduced toxicity remains a high priority. The aim of this study was to evaluate the cytotoxic and ROS production effects of chelate iron and copper complexes of two novel AQs, namely 4-hydroxynaphto[2,3-h]cinnoline-7,12-dione (Q2) and 3-(hydroxymethyl)naphto[2,3-h]cinnoline-4,7,12(1H)-trione (Q3). Methods: The chelation ability of Q2 and Q3 was studied using NMR and UV-Vis spectroscopy. Cytotoxicity studies were carried out using the MTT assay. The influence of chelation on ROS production was studied using NMR spectroscopy in linoleic acid micelles. Results: It was found that only Q3 forms complexes with Fe(III) and Cu(II) ions, whereas Q2 does not demonstrate chelating properties. A cytotoxicity study revealed that Fe[Q3]3 significantly decreased the viability of lung cancer A549 cells, while Q3 and Cu[Q3]2 did not demonstrate cytotoxic properties in this cell line. Furthermore, the presence of Q3 lowered the rate of iron-induced lipid peroxidation in linoleic acid micelles. By contrast, Q2 did not influence the rate of lipid peroxidation, probably due to the absence of effective metal chelating ability. Conclusions: The high cytotoxic effects observed with the iron complex of Q3 against cancer cells in combination with a reduced rate of iron induced lipid peroxidation in the presence of Q3, make Q3 and its iron complex promising for further evaluation and use as chemotherapeutic agents in cancer.
Collapse
Affiliation(s)
- Olga Yu. Selyutina
- Institute of Chemical Kinetics and Combustion, Institutskaya Str. 3, Novosibirsk 630090, Russia; (O.Y.S.); (M.A.U.); (O.A.C.); (V.A.T.); (L.G.F.); (A.A.S.); (L.V.K.); (N.E.P.)
| | - Maya A. Ul’yanova
- Institute of Chemical Kinetics and Combustion, Institutskaya Str. 3, Novosibirsk 630090, Russia; (O.Y.S.); (M.A.U.); (O.A.C.); (V.A.T.); (L.G.F.); (A.A.S.); (L.V.K.); (N.E.P.)
| | - Olga A. Chinak
- Institute of Chemical Kinetics and Combustion, Institutskaya Str. 3, Novosibirsk 630090, Russia; (O.Y.S.); (M.A.U.); (O.A.C.); (V.A.T.); (L.G.F.); (A.A.S.); (L.V.K.); (N.E.P.)
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Lavrentyev Ave. 8, Novosibirsk 630090, Russia
| | - Viktor A. Timoshnikov
- Institute of Chemical Kinetics and Combustion, Institutskaya Str. 3, Novosibirsk 630090, Russia; (O.Y.S.); (M.A.U.); (O.A.C.); (V.A.T.); (L.G.F.); (A.A.S.); (L.V.K.); (N.E.P.)
| | - Lidiya G. Fedenok
- Institute of Chemical Kinetics and Combustion, Institutskaya Str. 3, Novosibirsk 630090, Russia; (O.Y.S.); (M.A.U.); (O.A.C.); (V.A.T.); (L.G.F.); (A.A.S.); (L.V.K.); (N.E.P.)
| | - Alexander A. Stepanov
- Institute of Chemical Kinetics and Combustion, Institutskaya Str. 3, Novosibirsk 630090, Russia; (O.Y.S.); (M.A.U.); (O.A.C.); (V.A.T.); (L.G.F.); (A.A.S.); (L.V.K.); (N.E.P.)
| | - Vadim V. Yanshole
- International Tomography Center SB RAS, Institutskaya Str. 3a, Novosibirsk 630090, Russia;
| | - Leonid V. Kulik
- Institute of Chemical Kinetics and Combustion, Institutskaya Str. 3, Novosibirsk 630090, Russia; (O.Y.S.); (M.A.U.); (O.A.C.); (V.A.T.); (L.G.F.); (A.A.S.); (L.V.K.); (N.E.P.)
| | - Sergey F. Vasilevsky
- Institute of Chemical Kinetics and Combustion, Institutskaya Str. 3, Novosibirsk 630090, Russia; (O.Y.S.); (M.A.U.); (O.A.C.); (V.A.T.); (L.G.F.); (A.A.S.); (L.V.K.); (N.E.P.)
| | - Nikolay E. Polyakov
- Institute of Chemical Kinetics and Combustion, Institutskaya Str. 3, Novosibirsk 630090, Russia; (O.Y.S.); (M.A.U.); (O.A.C.); (V.A.T.); (L.G.F.); (A.A.S.); (L.V.K.); (N.E.P.)
| | - George J. Kontoghiorghes
- Postgraduate Research Institute of Science, Technology, Environment and Medicine, Limassol CY-3021, Cyprus
| |
Collapse
|
18
|
Hushmandi K, Klionsky DJ, Aref AR, Bonyadi M, Reiter RJ, Nabavi N, Salimimoghadam S, Saadat SH. Ferroptosis contributes to the progression of female-specific neoplasms, from breast cancer to gynecological malignancies in a manner regulated by non-coding RNAs: Mechanistic implications. Noncoding RNA Res 2024; 9:1159-1177. [PMID: 39022677 PMCID: PMC11250880 DOI: 10.1016/j.ncrna.2024.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 04/27/2024] [Accepted: 05/19/2024] [Indexed: 07/20/2024] Open
Abstract
Ferroptosis, a recently identified type of non-apoptotic cell death, triggers the elimination of cells in the presence of lipid peroxidation and in an iron-dependent manner. Indeed, ferroptosis-stimulating factors have the ability of suppressing antioxidant capacity, leading to the accumulation of reactive oxygen species (ROS) and the subsequent oxidative death of the cells. Ferroptosis is involved in the pathophysiological basis of different maladies, such as multiple cancers, among which female-oriented malignancies have attracted much attention in recent years. In this context, it has also been unveiled that non-coding RNA transcripts, including microRNAs, long non-coding RNAs, and circular RNAs have regulatory interconnections with the ferroptotic flux, which controls the pathogenic development of diseases. Furthermore, the potential of employing these RNA transcripts as therapeutic targets during the onset of female-specific neoplasms to modulate ferroptosis has become a research hotspot; however, the molecular mechanisms and functional alterations of ferroptosis still require further investigation. The current review comprehensively highlights ferroptosis and its association with non-coding RNAs with a focus on how this crosstalk affects the pathogenesis of female-oriented malignancies, from breast cancer to ovarian, cervical, and endometrial neoplasms, suggesting novel therapeutic targets to decelerate and even block the expansion and development of these tumors.
Collapse
Affiliation(s)
- Kiavash Hushmandi
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Daniel J. Klionsky
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Amir Reza Aref
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Translational Sciences, Xsphera Biosciences Inc., Boston, MA, USA
| | - Mojtaba Bonyadi
- Department of Food Hygiene and Quality Control, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Russel J. Reiter
- Department of Cell Systems and Anatomy, UT Health San Antonio, Long School of Medicine, San Antonio, TX, USA
| | - Noushin Nabavi
- Department of Urologic Sciences and Vancouver Prostate Centre, University of British Columbia, V6H3Z6, Vancouver, BC, Canada
| | - Shokooh Salimimoghadam
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Seyed Hassan Saadat
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
19
|
Lu Y, Xie X, Luo L. Ferroptosis crosstalk in anti-tumor immunotherapy: molecular mechanisms, tumor microenvironment, application prospects. Apoptosis 2024; 29:1914-1943. [PMID: 39008197 DOI: 10.1007/s10495-024-01997-8] [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] [Accepted: 06/24/2024] [Indexed: 07/16/2024]
Abstract
Immunotherapies for cancer, specifically immune checkpoint inhibition (ICI), have shown potential in reactivating the body's immune response against tumors. However, there are challenges to overcome in addressing drug resistance and improving the effectiveness of these treatments. Recent research has highlighted the relationship between ferroptosis and the immune system within immune cells and the tumor microenvironment (TME), suggesting that combining targeted ferroptosis with immunotherapy could enhance anti-tumor effects. This review explores the potential of using immunotherapy to target ferroptosis either alone or in conjunction with other therapies like immune checkpoint blockade (ICB) therapy, radiotherapy, and nanomedicine synergistic treatments. It also delves into the roles of different immune cell types in promoting anti-tumor immune responses through ferroptosis. Together, these findings provide a comprehensive understanding of synergistic immunotherapy focused on ferroptosis and offer innovative strategies for cancer treatment.
Collapse
Affiliation(s)
- Yining Lu
- The First Clinical College, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China
| | - Xiaoting Xie
- The First Clinical College, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China
| | - Lianxiang Luo
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, Guangdong, 524023, China.
| |
Collapse
|
20
|
Zhu Y, Jin Y, He X, Chen J, Zhang Y, Wang J. ALKBH5 insufficiency protects against ferroptosis-driven cisplatin-induced renal cytotoxicity. Cell Biol Toxicol 2024; 40:99. [PMID: 39557743 PMCID: PMC11573822 DOI: 10.1007/s10565-024-09947-5] [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: 05/03/2024] [Accepted: 11/07/2024] [Indexed: 11/20/2024]
Abstract
In the clinical setting, cisplatin-induced nephrotoxicity primarily manifests as acute kidney injury (AKI). Recent studies have indicated that ferroptosis, a type of iron-dependent cell death, is closely involved in the cisplatin nephrotoxicity. AlkB homologue 5 (ALKBH5), an N6-methyladenosine (m6A) eraser protein expressed in various tissues, including the kidneys, has been implicated in this process. However, the specific role of ALKBH5 in cisplatin-induced nephrotoxicity remains unknown. Our findings indicated that ALKBH5 was upregulated in cisplatin-induced AKI, and the in vivo study results were consistent with the results of the in vitro study. Additionally, ALKBH5 knockout in transgenic animals was found to mitigate cisplatin-induced renal dysfunction, whereas its knock-in exacerbated the effects. Our study revealed that ALKBH5 controls the traditional ferroptosis metabolic pathway, leading to worsening of AKI in experiments conducted both in vivo and in vitro. The efficacy of pharmacological intervention targeting ALKBH5 in AKI animal models was demonstrated, and ALKBH5-based gene therapy confirmed these findings and displayed renoprotective effects against AKI. In conclusion, this study highlighted the crucial role of ALKBH5 as a key regulator of AKI. Overall, our research demonstrates the significant impact of ALKBH5 in controlling ferroptosis in cisplatin-induced AKI, suggesting that focusing on ALKBH5 could be a promising approach for treating cisplatin-related kidney damage.
Collapse
Affiliation(s)
- Yu Zhu
- Department of Traditional Chinese Medicine, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310005, China
| | - Yanyan Jin
- Department of Nephrology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310005, China
| | - Xue He
- Department of Nephrology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310005, China
| | - JunYi Chen
- Department of Nephrology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310005, China
| | - Yao Zhang
- Department of Traditional Chinese Medicine, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310005, China
| | - JingJing Wang
- Department of Nephrology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310005, China.
| |
Collapse
|
21
|
Veglia Tranchese R, Battista S, Cerchia L, Fedele M. Ferroptosis in Cancer: Epigenetic Control and Therapeutic Opportunities. Biomolecules 2024; 14:1443. [PMID: 39595619 PMCID: PMC11592303 DOI: 10.3390/biom14111443] [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: 10/16/2024] [Revised: 11/06/2024] [Accepted: 11/10/2024] [Indexed: 11/28/2024] Open
Abstract
Ferroptosis, an iron-dependent form of regulated cell death driven by lipid peroxidation, has emerged as a critical pathway in cancer biology. This review delves into the epigenetic mechanisms that modulate ferroptosis in cancer cells, focusing on how DNA methylation, histone modifications, and non-coding RNAs influence the expression and function of essential genes involved in this process. By unraveling the complex interplay between these epigenetic mechanisms and ferroptosis, the article sheds light on novel gene targets and functional insights that could pave the way for innovative cancer treatments to enhance therapeutic efficacy and overcome resistance in cancer therapy.
Collapse
Affiliation(s)
| | | | | | - Monica Fedele
- Institute of Experimental Endocrinology and Oncology “G. Salvatore” (IEOS), National Research Council—CNR, 80131 Naples, Italy; (R.V.T.); (S.B.); (L.C.)
| |
Collapse
|
22
|
Shi J, Ma W, Deng J, Zheng S, Xia F, Liu X, Kikkawa A, Tanaka K, Kamei KI, Tian C. Self-assembled hyaluronic acid nanomicelle for enhanced cascade cancer chemotherapy via self-sensitized ferroptosis. Carbohydr Polym 2024; 343:122489. [PMID: 39174141 DOI: 10.1016/j.carbpol.2024.122489] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 07/08/2024] [Accepted: 07/11/2024] [Indexed: 08/24/2024]
Abstract
The clinical utility of chemotherapy is often compromised by its limited efficacy and significant side effects. Addressing these concerns, we have developed a self-assembled nanomicelle, namely SANTA FE OXA, which consists of hyaluronic acid (HA) conjugated with ferrocene methanol (FC), oxaliplatin prodrug (OXA(IV)) and ethylene glycol-coupled linoleic acid (EG-LA). Targeted delivery is achieved by HA binding to the CD44 receptors that are overexpressed on tumor cells, facilitating drug uptake. Once internalized, hyaluronidase (HAase) catalyzes the digestion of the SANTA FE OXA, releasing FC and reducing OXA(IV) into an active form. The active oxaliplatin (OXA) induces DNA damage and increases intracellular hydrogen peroxide (H2O2) levels via cascade reactions. Simultaneously, FC disrupts the redox balance within tumor cells, inducing ferroptosis. Both in vivo and in vitro experiments confirmed that SANTA FE OXA inhibited tumor growth by combining cascade chemotherapy and self-sensitized ferroptosis, achieving a tumor inhibition rate of up to 76.61 %. Moreover, this SANTA FE OXA significantly mitigates the systemic toxicity commonly associated with platinum-based chemotherapeutics. Our findings represent a compelling advancement in nanomedicine for enhanced cascade cancer therapy.
Collapse
Affiliation(s)
- Jianbin Shi
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Wenjing Ma
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jia Deng
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Shunzhe Zheng
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Fengli Xia
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xinying Liu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Ayumi Kikkawa
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto 606-8501, Japan
| | - Kaho Tanaka
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto 606-8501, Japan
| | - Ken-Ichiro Kamei
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang 110016, China; Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto 606-8501, Japan; Program of Biology, Division of Science, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates; Program of Bioengineering, Division of Engineering, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates; Department of Biomedical Engineering, Tandon School of Engineering, New York University, MetroTech, Brooklyn, NY 11201, United States of America.
| | - Chutong Tian
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang 110016, China; Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, Hangzhou 310058, China.
| |
Collapse
|
23
|
Cao PHA, Dominic A, Lujan FE, Senthilkumar S, Bhattacharya PK, Frigo DE, Subramani E. Unlocking ferroptosis in prostate cancer - the road to novel therapies and imaging markers. Nat Rev Urol 2024; 21:615-637. [PMID: 38627553 PMCID: PMC12067944 DOI: 10.1038/s41585-024-00869-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/04/2024] [Indexed: 04/19/2024]
Abstract
Ferroptosis is a distinct form of regulated cell death that is predominantly driven by the build-up of intracellular iron and lipid peroxides. Ferroptosis suppression is widely accepted to contribute to the pathogenesis of several tumours including prostate cancer. Results from some studies reported that prostate cancer cells can be highly susceptible to ferroptosis inducers, providing potential for an interesting new avenue of therapeutic intervention for advanced prostate cancer. In this Perspective, we describe novel molecular underpinnings and metabolic drivers of ferroptosis, analyse the functions and mechanisms of ferroptosis in tumours, and highlight prostate cancer-specific susceptibilities to ferroptosis by connecting ferroptosis pathways to the distinctive metabolic reprogramming of prostate cancer cells. Leveraging these novel mechanistic insights could provide innovative therapeutic opportunities in which ferroptosis induction augments the efficacy of currently available prostate cancer treatment regimens, pending the elimination of major bottlenecks for the clinical translation of these treatment combinations, such as the development of clinical-grade inhibitors of the anti-ferroptotic enzymes as well as non-invasive biomarkers of ferroptosis. These biomarkers could be exploited for diagnostic imaging and treatment decision-making.
Collapse
Affiliation(s)
- Pham Hong Anh Cao
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Abishai Dominic
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Fabiola Ester Lujan
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Sanjanaa Senthilkumar
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Mayo Clinic Alix School of Medicine, Rochester, MN, USA
| | - Pratip K Bhattacharya
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Daniel E Frigo
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Center for Nuclear Receptors and Cell Signalling, University of Houston, Houston, TX, USA.
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA.
| | - Elavarasan Subramani
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
24
|
Vinik Y, Maimon A, Raj H, Dubey V, Geist F, Wienke D, Lev S. Computational pipeline predicting cell death suppressors as targets for cancer therapy. iScience 2024; 27:110859. [PMID: 39310772 PMCID: PMC11416655 DOI: 10.1016/j.isci.2024.110859] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 06/24/2024] [Accepted: 08/28/2024] [Indexed: 09/25/2024] Open
Abstract
Identification of promising targets for cancer therapy is a global effort in precision medicine. Here, we describe a computational pipeline integrating transcriptomic and vulnerability responses to cell-death inducing drugs, to predict cell-death suppressors as candidate targets for cancer therapy. The prediction is based on two modules; the transcriptomic similarity module to identify genes whose targeting results in similar transcriptomic responses of the death-inducing drugs, and the correlation module to identify candidate genes whose expression correlates to the vulnerability of cancer cells to the same death-inducers. The combined predictors of these two modules were integrated into a single metric. As a proof-of-concept, we selected ferroptosis inducers as death-inducing drugs in triple negative breast cancer. The pipeline reliably predicted candidate genes as ferroptosis suppressors, as validated by computational methods and cellular assays. The described pipeline might be used to identify repressors of various cell-death pathways as potential therapeutic targets for different cancer types.
Collapse
Affiliation(s)
- Yaron Vinik
- Molecular Cell Biology Department, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Avi Maimon
- Molecular Cell Biology Department, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Harsha Raj
- Molecular Cell Biology Department, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Vinay Dubey
- Molecular Cell Biology Department, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Felix Geist
- The Healthcare Business of Merck KGaA, Darmstadt, Germany
| | - Dirk Wienke
- The Healthcare Business of Merck KGaA, Darmstadt, Germany
| | - Sima Lev
- Molecular Cell Biology Department, Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
25
|
Sun L, Cao H, Wang Y, Wang H. Regulating ferroptosis by non-coding RNAs in hepatocellular carcinoma. Biol Direct 2024; 19:80. [PMID: 39267124 PMCID: PMC11391853 DOI: 10.1186/s13062-024-00530-w] [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: 08/08/2024] [Accepted: 09/05/2024] [Indexed: 09/14/2024] Open
Abstract
Ferroptosis, a unique type of regulated cell death plays a vital role in inhibiting tumour malignancy and has presented new opportunities for treatment of therapy in hepatocellular carcinoma. Accumulating studies indicate that epigenetic modifications by non-coding RNAs, including microRNAs, long noncoding RNAs, and circular RNAs, can determine cancer cell vulnerability to ferroptosis in HCC. The present review first summarize the updated core molecular mechanisms of ferroptosis. We then provide a concised overview of epigenetic modification of ferroptosis in HCC. Finally, we review the recent progress in understanding of the ncRNA-mediated regulated mechanisms on ferroptosis in HCC. The review will promote our understanding of the ncRNA-mediated epigenetic regulatory mechanisms modulating ferroptosis in malignancy of HCC, highlighting a novel strategies for treatment of HCC through targeting ncRNA-ferroptosis axis.
Collapse
Affiliation(s)
- Lijie Sun
- Department of Gastroenterology, The Affiliated Hospital of Chifeng University, Chifeng, 024005, China.
| | - Hongfei Cao
- Department of Gastroenterology, The Affiliated Hospital of Chifeng University, Chifeng, 024005, China.
| | - Yanzhe Wang
- Department of Gastroenterology, The Affiliated Hospital of Chifeng University, Chifeng, 024005, China
| | - Hongquan Wang
- Department of Geriatrics, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, China
| |
Collapse
|
26
|
Wang S, Cheng M, Wang S, Jiang W, Yang F, Shen X, Zhang L, Yan X, Jiang B, Fan K. A Self-Catalytic NO/O 2 Gas-Releasing Nanozyme for Radiotherapy Sensitization through Vascular Normalization and Hypoxia Relief. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2403921. [PMID: 39101290 DOI: 10.1002/adma.202403921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 06/29/2024] [Indexed: 08/06/2024]
Abstract
Radiotherapy (RT), essential for treating various cancers, faces challenges from tumor hypoxia, which induces radioresistance. A tumor-targeted "prosthetic-Arginine" coassembled nanozyme system, engineered to catalytically generate nitric oxide (NO) and oxygen (O2) in the tumor microenvironment (TME), overcoming hypoxia and enhancing radiosensitivity is presented. This system integrates the prosthetic heme of nitric oxide synthase (NOS) and catalase (CAT) with NO-donating Fmoc-protected Arginine and Ru3+ ions, creating HRRu nanozymes that merge NOS and CAT functionalities. Surface modification with human heavy chain ferritin (HFn) improves the targeting ability of nanozymes (HRRu-HFn) to tumor tissues. In the TME, strategic arginine incorporation within the nanozyme allows autonomous O2 and NO release, triggered by endogenous hydrogen peroxide, elevating NO and O2 levels to normalize vasculature and improve blood perfusion, thus mitigating hypoxia. Employing the intrinsic O2-transporting ability of heme, HRRu-HFn nanozymes also deliver O2 directly to the tumor site. Utilizing esophageal squamous cell carcinoma as a tumor model, the studies reveal that the synergistic functions of NO and O2 production, alongside targeted delivery, enable the HRRu-HFn nanozymes to combat tumor hypoxia and potentiate radiotherapy. This HRRu-HFn nanozyme based approach holds the potential to reduce the radiation dose required and minimize side effects associated with conventional radiotherapy.
Collapse
Affiliation(s)
- Shuyu Wang
- Nanozyme Laboratory in Zhongyuan, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Miaomiao Cheng
- Nanozyme Laboratory in Zhongyuan, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Shenghui Wang
- Nanozyme Laboratory in Zhongyuan, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Wei Jiang
- Nanozyme Laboratory in Zhongyuan, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Feifei Yang
- College of Chemistry and Chemical Engineering, Jiangxi Normal University, Nanchang, 330022, China
| | - Xiaomei Shen
- College of Chemistry and Chemical Engineering, Jiangxi Normal University, Nanchang, 330022, China
| | - Lirong Zhang
- State Key Laboratory of Esophageal Cancer Prevention &Treatment, Henan, 450001, China
| | - Xiyun Yan
- Nanozyme Laboratory in Zhongyuan, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
- Nanozyme Laboratory in Zhongyuan, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, 451163, China
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Bing Jiang
- Nanozyme Laboratory in Zhongyuan, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
- Nanozyme Laboratory in Zhongyuan, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, 451163, China
| | - Kelong Fan
- Nanozyme Laboratory in Zhongyuan, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, 451163, China
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| |
Collapse
|
27
|
Delgado-Martín S, Martínez-Ruiz A. The role of ferroptosis as a regulator of oxidative stress in the pathogenesis of ischemic stroke. FEBS Lett 2024; 598:2160-2173. [PMID: 38676284 DOI: 10.1002/1873-3468.14894] [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: 11/10/2023] [Revised: 03/25/2024] [Accepted: 04/02/2024] [Indexed: 04/28/2024]
Abstract
Ferroptosis is a unique form of cell death that was first described in 2012 and plays a significant role in various diseases, including neurodegenerative conditions. It depends on a dysregulation of cellular iron metabolism, which increases free, redox-active, iron that can trigger Fenton reactions, generating hydroxyl radicals that damage cells through oxidative stress and lipid peroxidation. Lipid peroxides, resulting mainly from unsaturated fatty acids, damage cells by disrupting membrane integrity and propagating cell death signals. Moreover, lipid peroxide degradation products can further affect cellular components such as DNA, proteins, and amines. In ischemic stroke, where blood flow to the brain is restricted, there is increased iron absorption, oxidative stress, and compromised blood-brain barrier integrity. Imbalances in iron-transport and -storage proteins increase lipid oxidation and contribute to neuronal damage, thus pointing to the possibility of brain cells, especially neurons, dying from ferroptosis. Here, we review the evidence showing a role of ferroptosis in ischemic stroke, both in recent studies directly assessing this type of cell death, as well as in previous studies showing evidence that can now be revisited with our new knowledge on ferroptosis mechanisms. We also review the efforts made to target ferroptosis in ischemic stroke as a possible treatment to mitigate cellular damage and death.
Collapse
Affiliation(s)
- Susana Delgado-Martín
- Unidad de Investigación, Hospital Santa Cristina, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
| | - Antonio Martínez-Ruiz
- Unidad de Investigación, Hospital Santa Cristina, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad Complutense de Madrid, Spain
| |
Collapse
|
28
|
Wang H, Fleishman JS, Cheng S, Wang W, Wu F, Wang Y, Wang Y. Epigenetic modification of ferroptosis by non-coding RNAs in cancer drug resistance. Mol Cancer 2024; 23:177. [PMID: 39192329 DOI: 10.1186/s12943-024-02088-7] [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: 07/09/2024] [Accepted: 08/13/2024] [Indexed: 08/29/2024] Open
Abstract
The development of drug resistance remains a major challenge in cancer treatment. Ferroptosis, a unique type of regulated cell death, plays a pivotal role in inhibiting tumour growth, presenting new opportunities in treating chemotherapeutic resistance. Accumulating studies indicate that epigenetic modifications by non-coding RNAs (ncRNA) can determine cancer cell vulnerability to ferroptosis. In this review, we first summarize the role of chemotherapeutic resistance in cancer growth/development. Then, we summarize the core molecular mechanisms of ferroptosis, its upstream epigenetic regulation, and its downstream effects on chemotherapeutic resistance. Finally, we review recent advances in understanding how ncRNAs regulate ferroptosis and from such modulate chemotherapeutic resistance. This review aims to enhance general understanding of the ncRNA-mediated epigenetic regulatory mechanisms which modulate ferroptosis, highlighting the ncRNA-ferroptosis axis as a key druggable target in overcoming chemotherapeutic resistance.
Collapse
Affiliation(s)
- Hongquan Wang
- Department of Geriatrics, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, China.
| | - Joshua S Fleishman
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Sihang Cheng
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Weixue Wang
- Department of Geriatrics, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, China
| | - Fan Wu
- Department of Hepatobiliary Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Yumin Wang
- Department of Respiratory and Critical Care Medicine, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, China.
| | - Yu Wang
- Department of Geriatrics, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, China.
| |
Collapse
|
29
|
Wang S, Cheng M, Wang S, Jiang W, Yang F, Shen X, Zhang L, Yan X, Jiang B, Fan K. A Self‐Catalytic NO/O 2 Gas‐Releasing Nanozyme for Radiotherapy Sensitization through Vascular Normalization and Hypoxia Relief. ADVANCED MATERIALS 2024. [DOI: doi:10.1002/adma.202403921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Indexed: 04/16/2025]
Abstract
AbstractRadiotherapy (RT), essential for treating various cancers, faces challenges from tumor hypoxia, which induces radioresistance. A tumor‐targeted “prosthetic‐Arginine” coassembled nanozyme system, engineered to catalytically generate nitric oxide (NO) and oxygen (O2) in the tumor microenvironment (TME), overcoming hypoxia and enhancing radiosensitivity is presented. This system integrates the prosthetic heme of nitric oxide synthase (NOS) and catalase (CAT) with NO‐donating Fmoc‐protected Arginine and Ru3+ ions, creating HRRu nanozymes that merge NOS and CAT functionalities. Surface modification with human heavy chain ferritin (HFn) improves the targeting ability of nanozymes (HRRu‐HFn) to tumor tissues. In the TME, strategic arginine incorporation within the nanozyme allows autonomous O2 and NO release, triggered by endogenous hydrogen peroxide, elevating NO and O2 levels to normalize vasculature and improve blood perfusion, thus mitigating hypoxia. Employing the intrinsic O2‐transporting ability of heme, HRRu‐HFn nanozymes also deliver O2 directly to the tumor site. Utilizing esophageal squamous cell carcinoma as a tumor model, the studies reveal that the synergistic functions of NO and O2 production, alongside targeted delivery, enable the HRRu‐HFn nanozymes to combat tumor hypoxia and potentiate radiotherapy. This HRRu‐HFn nanozyme based approach holds the potential to reduce the radiation dose required and minimize side effects associated with conventional radiotherapy.
Collapse
Affiliation(s)
- Shuyu Wang
- Nanozyme Laboratory in Zhongyuan School of Basic Medical Sciences Zhengzhou University Zhengzhou Henan 450001 China
| | - Miaomiao Cheng
- Nanozyme Laboratory in Zhongyuan School of Basic Medical Sciences Zhengzhou University Zhengzhou Henan 450001 China
| | - Shenghui Wang
- Nanozyme Laboratory in Zhongyuan School of Basic Medical Sciences Zhengzhou University Zhengzhou Henan 450001 China
| | - Wei Jiang
- Nanozyme Laboratory in Zhongyuan School of Basic Medical Sciences Zhengzhou University Zhengzhou Henan 450001 China
| | - Feifei Yang
- College of Chemistry and Chemical Engineering Jiangxi Normal University Nanchang 330022 China
| | - Xiaomei Shen
- College of Chemistry and Chemical Engineering Jiangxi Normal University Nanchang 330022 China
| | - Lirong Zhang
- State Key Laboratory of Esophageal Cancer Prevention &Treatment Henan 450001 China
| | - Xiyun Yan
- Nanozyme Laboratory in Zhongyuan School of Basic Medical Sciences Zhengzhou University Zhengzhou Henan 450001 China
- Nanozyme Laboratory in Zhongyuan Henan Academy of Innovations in Medical Science Zhengzhou Henan 451163 China
- CAS Engineering Laboratory for Nanozyme Key Laboratory of Biomacromolecules (CAS) CAS Center for Excellence in Biomacromolecules Institute of Biophysics Chinese Academy of Sciences Beijing 100101 China
| | - Bing Jiang
- Nanozyme Laboratory in Zhongyuan School of Basic Medical Sciences Zhengzhou University Zhengzhou Henan 450001 China
- Nanozyme Laboratory in Zhongyuan Henan Academy of Innovations in Medical Science Zhengzhou Henan 451163 China
| | - Kelong Fan
- Nanozyme Laboratory in Zhongyuan Henan Academy of Innovations in Medical Science Zhengzhou Henan 451163 China
- CAS Engineering Laboratory for Nanozyme Key Laboratory of Biomacromolecules (CAS) CAS Center for Excellence in Biomacromolecules Institute of Biophysics Chinese Academy of Sciences Beijing 100101 China
| |
Collapse
|
30
|
Dai Q, Wei X, Zhao J, Zhang D, Luo Y, Yang Y, Xiang Y, Liu X. Inhibition of FSP1: A new strategy for the treatment of tumors (Review). Oncol Rep 2024; 52:105. [PMID: 38940330 PMCID: PMC11228423 DOI: 10.3892/or.2024.8764] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 05/10/2024] [Indexed: 06/29/2024] Open
Abstract
Ferroptosis, a regulated form of cell death, is intricately linked to iron‑dependent lipid peroxidation. Recent evidence strongly supports the induction of ferroptosis as a promising strategy for treating cancers resistant to conventional therapies. A key player in ferroptosis regulation is ferroptosis suppressor protein 1 (FSP1), which promotes cancer cell resistance by promoting the production of the antioxidant form of coenzyme Q10. Of note, FSP1 confers resistance to ferroptosis independently of the glutathione (GSH) and glutathione peroxidase‑4 pathway. Therefore, targeting FSP1 to weaken its inhibition of ferroptosis may be a viable strategy for treating refractory cancer. This review aims to clarify the molecular mechanisms underlying ferroptosis, the specific pathway by which FSP1 suppresses ferroptosis and the effect of FSP1 inhibitors on cancer cells.
Collapse
Affiliation(s)
- Qiangfang Dai
- School of Medicine, Yan'an University, Yan'an, Shaanxi 716000, P.R. China
| | - Xiaoli Wei
- School of Medicine, Yan'an University, Yan'an, Shaanxi 716000, P.R. China
| | - Jumei Zhao
- School of Medicine, Yan'an University, Yan'an, Shaanxi 716000, P.R. China
| | - Die Zhang
- School of Medicine, Yan'an University, Yan'an, Shaanxi 716000, P.R. China
| | - Yidan Luo
- School of Medicine, Yan'an University, Yan'an, Shaanxi 716000, P.R. China
| | - Yue Yang
- School of Medicine, Yan'an University, Yan'an, Shaanxi 716000, P.R. China
| | - Yang Xiang
- School of Medicine, Yan'an University, Yan'an, Shaanxi 716000, P.R. China
- College of Physical Education, Yan'an University, Yan'an, Shaanxi 716000, P.R. China
| | - Xiaolong Liu
- School of Medicine, Yan'an University, Yan'an, Shaanxi 716000, P.R. China
| |
Collapse
|
31
|
Yang T, Liu Z, Zhang T, Liu Y. Hybrid nano-stimulator for specific amplification of oxidative stress and precise tumour treatment. J Drug Target 2024; 32:756-769. [PMID: 38832845 DOI: 10.1080/1061186x.2024.2349112] [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: 12/01/2023] [Revised: 04/23/2024] [Accepted: 04/23/2024] [Indexed: 06/06/2024]
Abstract
BACKGROUND The use of reactive oxygen species (ROS) to target cancer cells has become a hot topic in tumor therapy. PURPOSE Although ROS has strong cytotoxicity against tumor cells, the key issue currently is how to generate a large amount of ROS within tumor cells. METHODS Organic/inorganic hybrid nanoreactor materials combine the advantages of organic and inorganic components and can amplify cancer treatment by increasing targeting and material self-action. The multifunctional organic / inorganic hybrid nanoreactor is helpful to overcome the shortcomings of current reactive oxygen species in cancer treatment. It can realize the combination of in situ dynamic therapy and immunotherapy strategies, and has a synergistic anti-tumor effect. RESULTS This paper reviews the research progress of organic/inorganic hybrid nanoreactor materials using tumor components to amplify reactive oxygen species for cancer treatment. The article reviews the tumor treatment strategies of nanohybrids from the perspectives of cancer cells, immune cells, tumor microenvironment, as well as 3D printing and electrospinning techniques, which are different from traditional nanomaterial technologies, and will arouse interest among scientists in tumor therapy and nanomedicine.
Collapse
Affiliation(s)
- Ting Yang
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Zihan Liu
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Tong Zhang
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Yanhua Liu
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, China
- Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area, Ministry of Education, Yinchuan, China
| |
Collapse
|
32
|
Robert M, Kennedy BK, Crasta KC. Therapy-induced senescence through the redox lens. Redox Biol 2024; 74:103228. [PMID: 38865902 PMCID: PMC11215421 DOI: 10.1016/j.redox.2024.103228] [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: 05/08/2024] [Revised: 05/22/2024] [Accepted: 06/04/2024] [Indexed: 06/14/2024] Open
Abstract
Therapy-induced senescent tumor cells have emerged as significant drivers of tumor recurrence and disease relapse. Interestingly, reactive oxygen species (ROS) production and its associated redox signaling networks are intertwined with initiation and establishment of therapy-induced senescence. Therapy-induced senescent cells influence neighboring cells and the tumor microenvironment via their bioactive secretome known as the senescence-associated secretory phenotype (SASP). The intracellular effects of ROS are dose and context-dependent. Under normal physiological conditions, ROS is involved in various signalling pathways and cellular processes important for maintenance of cellular homeostasis, such as redox balance, stress response, inflammatory signalling, cell proliferation and cell death among others. However excess ROS accompanied by a pro-oxidant microenvironment can engender oxidative DNA damage, triggering cellular senescence. In this review, we discuss the role of ROS and the redox state dynamics in fine-tuning homeostatic processes that drive therapy-induced cell fate towards senescence establishment, as well as their influence in stimulating inflammatory signalling and SASP production. We also offer insights into interventional strategies, specifically senotherapeutics, that could potentially leverage on modulation of redox and antioxidant pathways. Lastly, we evaluate possible implications of redox rewiring during escape from therapy-induced senescence, an emerging area of research. We envision that examining therapy-induced senescence through the redox lens, integrated with time-resolved single-cell RNA sequencing combined with spatiotemporal multi-omics, could further enhance our understanding of its functional heterogeneity. This could aid identification of targetable signalling nodes to reduce disease relapse, as well as inform strategies for development of broad-spectrum senotherapeutics. Overall, our review aims to delineate redox-driven mechanisms which contribute to the biology of therapy-induced senescence and beyond, while highlighting implications for tumor initiation and recurrence.
Collapse
Affiliation(s)
- Matius Robert
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Centre for Healthy Longevity, National University Health System, Singapore; Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Brian K Kennedy
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Centre for Healthy Longevity, National University Health System, Singapore; Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| | - Karen C Crasta
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Centre for Healthy Longevity, National University Health System, Singapore; Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| |
Collapse
|
33
|
Li X, Chen T, Fu Y, Yang B, Lin X, Hou J, Yang X. Mechanism and functional verification of genes by virulence factors of P. gingivalis in ferroptosis. Arch Oral Biol 2024; 163:105965. [PMID: 38593562 DOI: 10.1016/j.archoralbio.2024.105965] [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: 02/21/2024] [Revised: 04/03/2024] [Accepted: 04/06/2024] [Indexed: 04/11/2024]
Abstract
OBJECTIVE Porphyromonas gingivalis (P. gingivalis) is a key etiological agent in periodontitis and functions as a facultative intracellular microorganism and involves many virulence factors. These virulence factors participate in multiple intracellular processes, like ferroptosis, the mechanistic underpinnings remain to be elucidated. Aim of this study was to investigate the effects of virulence factors on the host cells. DESIGN Human umbilical vein endothelial cells (HUVECs) were treated with 4% paraformaldehyde-fixed P. gingivalis, and subsequent alterations in gene expression were profiled via RNA-seq. Further, the molecules associated with ferroptosis were quantitatively analyzed using qRT-PCR and Western blot. RESULTS A total of 1125 differentially expressed genes (DEGs) were identified, encompassing 225 upregulated and 900 downregulated. Ferroptosis was conspicuously represented in the kyoto encyclopedia of genes and genomes (KEGG) enrichment analysis, with notable upregulation of Heme oxygenase 1 (HMOX1), Ferritin light chain (FTL), and Solute carrier family 3 member 2 (SLC3A2) and downregulation of Scavenger receptor class A member 5 (SCARA5) and glutaminase (GLS). Random selection of DEGs for validation through qRT-PCR corroborated the RNA-Seq data (R2 = 0.93). Kelch like ECH associated protein 1 (Keap1) protein expression decreased after 4 and 8 h, while NFE2 like bZIP transcription factor 2 (Nrf2) and HMOX1 were elevated, with significant nuclear translocation of Nrf2. CONCLUSIONS The virulence factors of P. gingivalis may potentially instigating ferroptosis through activation of the Keap1-Nrf2-HMOX1 signaling cascade, in conjunction with modulating the expression of other ferroptosis-associated elements. Further research is necessary to achieve a thorough comprehension of these complex molecular interactions.
Collapse
Affiliation(s)
- Xinzhu Li
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Ting Chen
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yinyu Fu
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Bo Yang
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xiaoyu Lin
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jin Hou
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| | - Xiaojun Yang
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
34
|
Hou Y, Wang H, Wu J, Guo H, Chen X. Dissecting the pleiotropic roles of reactive oxygen species (ROS) in lung cancer: From carcinogenesis toward therapy. Med Res Rev 2024; 44:1566-1595. [PMID: 38284170 DOI: 10.1002/med.22018] [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: 08/23/2022] [Revised: 12/14/2023] [Accepted: 01/10/2024] [Indexed: 01/30/2024]
Abstract
Lung cancer is a major cause of morbidity and mortality. The specific pulmonary structure to directly connect with ambient air makes it more susceptible to damage from airborne toxins. External oxidative stimuli and endogenous reactive oxygen species (ROS) play a crucial role in promoting lung carcinogenesis and development. The biological properties of higher ROS levels in tumor cells than in normal cells make them more sensitive and vulnerable to ROS injury. Therefore, the strategy of targeting ROS has been proposed for cancer therapy for decades. However, it is embarrassing that countless attempts at ROS-based therapies have had very limited success, and no FDA approval in the anticancer list was mechanistically based on ROS manipulation. Even compared with the untargetable proteins, such as transcription factors, ROS are more difficult to be targeted due to their chemical properties. Thus, the pleiotropic roles of ROS provide therapeutic potential for anticancer drug discovery, while a better dissection of the mechanistic action and signaling pathways is a prerequisite for future breakthroughs. This review discusses the critical roles of ROS in cancer carcinogenesis, ROS-inspired signaling pathways, and ROS-based treatment, exemplified by lung cancer. In particular, an eight considerations rule is proposed for ROS-targeting strategies and drug design and development.
Collapse
Affiliation(s)
- Ying Hou
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao, China
| | - Heng Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao, China
| | - Jiarui Wu
- Department of Clinical Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, China
| | - Hongwei Guo
- Key Laboratory of Longevity and Aging-Related Diseases of Chinese Ministry of Education, Guangxi Key Laboratory of Research and Evaluation of Bioactive Molecules & College of Pharmacy, Guangxi Medical University, Nanning, China
| | - Xiuping Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao, China
- Department of Pharmaceutical Sciences, University of Macau, Taipa, Macao, China
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macao, China
| |
Collapse
|
35
|
Cheng P, Xia R, Wang X. Ferroptosis: a promising target for fumarate hydratase-deficient tumor therapeutics literature review. Transl Cancer Res 2024; 13:3126-3141. [PMID: 38988939 PMCID: PMC11231789 DOI: 10.21037/tcr-24-21] [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: 01/04/2024] [Accepted: 04/24/2024] [Indexed: 07/12/2024]
Abstract
Background and Objective This review aims to investigate the ferroptosis mechanism of fumarate hydratase (FH)-related tumors for the purpose of possible treatment of tumors. Ferroptosis is an iron (Fe)-dependent form of regulated cell death caused by lipid peroxidation on the cell membrane. Studies have implicated FH in tumorigenesis. As mutations in the FH gene alter cellular metabolism and increase tumorigenesis risk, particularly in the kidneys. As most tumor cells require higher amounts of ferrous ions (Fe2+) than normal cells, they are more susceptible to ferroptosis. Recent studies have indicated that ferroptosis is inhibited the pathogenesis and progression of FH-deficient tumors by regulating lipid and iron metabolism, glutathione-glutathione peroxidase 4 (GSH-GPX4), nuclear factor-erythroid 2-related factor 2 (NRF2)/heme oxygenase-1 (HO-1) pathways. While the Fe2+ content is significantly lower in FH-deficient tumor cells, than that in normal cells. It is promising to promote ferroptosis by increasing the concentration of Fe2+ in cells to achieve the purpose of tumor treatment. Methods In this study, we searched for relevant articles on ferroptosis and FH-deficient tumors using PubMed database. Key Content and Findings FH is a tumor suppressor. A number of basic studies have shown that the loss of FH plays an important role in hereditary leiomyomas and tumors such as renal cell carcinoma, ovarian cancer, and other tumors. This type of tumor cells can through induce ferroptosis, inhibit proliferation, migration and invasion of tumor cells, increase the sensitivity of tumor cells to chemotherapy, and reverse the drug resistance through various molecular mechanisms. At present, the research on ferroptosis in FH-related tumors is still in the basic experimental stage. Conclusions This article reviews the anti-tumor effects and mechanisms of FH and ferroptosis, in order to further explore the medical value of ferroptosis in FH-related tumor therapy.
Collapse
Affiliation(s)
- Ping Cheng
- Department of Biochemistry and Molecular Biology, Health Science Center, Yangtze University, Jingzhou, China
| | - Ruohan Xia
- Department of Biochemistry and Molecular Biology, Health Science Center, Yangtze University, Jingzhou, China
| | - Xianwang Wang
- Department of Biochemistry and Molecular Biology, Health Science Center, Yangtze University, Jingzhou, China
- Shannan Maternal and Child Health Hospital, Shannan, China
| |
Collapse
|
36
|
Hu Y, Huang Y, Zong L, Lin J, Liu X, Ning S. Emerging roles of ferroptosis in pulmonary fibrosis: current perspectives, opportunities and challenges. Cell Death Discov 2024; 10:301. [PMID: 38914560 PMCID: PMC11196712 DOI: 10.1038/s41420-024-02078-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 06/13/2024] [Accepted: 06/18/2024] [Indexed: 06/26/2024] Open
Abstract
Pulmonary fibrosis (PF) is a chronic interstitial lung disorder characterized by abnormal myofibroblast activation, accumulation of extracellular matrix (ECM), and thickening of fibrotic alveolar walls, resulting in deteriorated lung function. PF is initiated by dysregulated wound healing processes triggered by factors such as excessive inflammation, oxidative stress, and coronavirus disease (COVID-19). Despite advancements in understanding the disease's pathogenesis, effective preventive and therapeutic interventions are currently lacking. Ferroptosis, an iron-dependent regulated cell death (RCD) mechanism involving lipid peroxidation and glutathione (GSH) depletion, exhibits unique features distinct from other RCD forms (e.g., apoptosis, necrosis, and pyroptosis). Imbalance between reactive oxygen species (ROS) production and detoxification leads to ferroptosis, causing cellular dysfunction through lipid peroxidation, protein modifications, and DNA damage. Emerging evidence points to the crucial role of ferroptosis in PF progression, driving macrophage polarization, fibroblast proliferation, and ECM deposition, ultimately contributing to alveolar cell death and lung tissue scarring. This review provides a comprehensive overview of the latest findings on the involvement and signaling mechanisms of ferroptosis in PF pathogenesis, emphasizing potential novel anti-fibrotic therapeutic approaches targeting ferroptosis for PF management.
Collapse
Affiliation(s)
- Yixiang Hu
- Department of Clinical Pharmacy, The Affiliated Xiangtan Center Hospital of Hunan University, Xiangtan, 411100, China
| | - Ying Huang
- Zhongshan Hospital of Traditional Chinese Medicine Afflilated to Guangzhou University of Chinese Medicine, Zhongshan, 528400, China
| | - Lijuan Zong
- Department of Rehabilitation Medicine, Zhongda Hospital of Southeast University, Nanjing, 210096, China
| | - Jiaxin Lin
- Department of Breast Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530000, China
| | - Xiang Liu
- Department of Clinical Pharmacy, The Affiliated Xiangtan Center Hospital of Hunan University, Xiangtan, 411100, China.
| | - Shipeng Ning
- Department of Breast Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530000, China.
| |
Collapse
|
37
|
Dhandapani S, Samad A, Liu Y, Wang R, Balusamy SR, Perumalsamy H, Kim YJ. Coprisin/Compound K Conjugated Gold Nanoparticles Induced Cell Death through Apoptosis and Ferroptosis Pathway in Adenocarcinoma Gastric Cells. ACS OMEGA 2024; 9:25932-25944. [PMID: 38911731 PMCID: PMC11190908 DOI: 10.1021/acsomega.4c00554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 03/03/2024] [Accepted: 04/26/2024] [Indexed: 06/25/2024]
Abstract
Ferroptosis and apoptosis are programmed cell death pathways with distinct characteristics. Sometimes, cancer cells are aided by the induction of a different pathway, such as ferroptosis, when they develop chemoresistance and avoid apoptosis. Identifying the nanomedicine that targets dual pathways is considered as one of the best strategies for diverse cancer types. In our previous work, we synthesized gold nanoparticles (GNP) utilizing Gluconacetobacter liquefaciens in conjunction with compound K (CK) and coprisin (CopA3), yielding GNP-CK-CopA3. Here, we assessed the inhibitory effect of GNP-CK-CopA3 on AGS cells and the induction of apoptosis using Hoechst and PI, Annexin V-FITC/PI, and qRT-PCR. Subsequently, we conducted downstream proteomic analysis and molecular dynamic stimulation to identify the underlying molecular mechanisms. Our investigation of cultured AGS cells treated with varying concentrations of GNP-CK-CopA3 demonstrated the anticancer properties of these nanoparticles. Penetration of GNP-CK-CopA3 into AGS cells was visualized using an enhanced dark field microscope. Apoptosis induction was initially confirmed by treating AGS cells with GNP-CK-CopA3, as evidenced by staining with dyes such as Hoechst and PI. Additionally, mitochondrial disruption and cellular localization induced by GNP-CK-CopA3 were validated through Mito-tracker staining and transmission electron microscopy images. Annexin V-FITC/PI staining was used to distinguish early and late-stage apoptosis or necrosis based on fluorescence patterns. The gene expression of apoptotic markers indicated the initiation of cellular apoptosis. Further, proteomic analysis suggested that the treatment of GNP-CK-CopA3 to AGS cells led to the suppression of 439 proteins and the stimulation of 832 proteins. Among these, ferroptosis emerged as a significant interconnected pathway where glutathione peroxidase 4 (GPX4) and glutathione synthetase (GSS) were significant interacting proteins. Molecular docking and dynamic simulation studies confirmed the binding affinity and stability between CopA3 and CK with GSS and GPX4 proteins, suggesting the role of GNP-CK-CopA3 in ferroptosis induction. Overall, our study showed GNP-CK-CopA3 could play a dual role by inducing apoptosis and ferroptosis to induce AGS cell death.
Collapse
Affiliation(s)
- Sanjeevram Dhandapani
- Graduate
School of Biotechnology, and College of Life Science, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea
| | - Abdus Samad
- Graduate
School of Biotechnology, and College of Life Science, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea
| | - Ying Liu
- Graduate
School of Biotechnology, and College of Life Science, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea
| | - Rongbo Wang
- Graduate
School of Biotechnology, and College of Life Science, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea
| | - Sri Renukadevi Balusamy
- Department
of Food Science and Biotechnology, Sejong
University, Gwangjin-gu, Seoul 05006, Republic of Korea
| | - Haribalan Perumalsamy
- Center
for Creative Convergence Education, Hanyang
University, Seoul 04763, Republic of Korea
- Research
Institute for Convergence of Basic Science, Hanyang University, Seoul 04763, South Korea
| | - Yeon-Ju Kim
- Graduate
School of Biotechnology, and College of Life Science, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea
| |
Collapse
|
38
|
Zou W, Gao F, Meng Z, Cai X, Chen W, Zheng Y, Ying T, Wang L, Wu J. Lactic acid responsive sequential production of hydrogen peroxide and consumption of glutathione for enhanced ferroptosis tumor therapy. J Colloid Interface Sci 2024; 663:787-800. [PMID: 38442520 DOI: 10.1016/j.jcis.2024.03.001] [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: 12/16/2023] [Revised: 02/29/2024] [Accepted: 03/01/2024] [Indexed: 03/07/2024]
Abstract
Ferroptosis is characterized by the lethal accumulation of lipid reactive oxygen species (ROS), which has great potential for tumor therapy. However, developing new ferroptosis-inducing strategies by combining nanomaterials with small molecule inducers is important. In this study, an enzyme-gated biodegradable natural-product delivery system based on lactate oxidase (LOD)-gated biodegradable iridium (Ir)-doped hollow mesoporous organosilica nanoparticles (HMONs) loaded with honokiol (HNK) (HNK@Ir-HMONs-LOD, HIHL) is designed to enhance ferroptosis in colon tumor therapy. After reaching the tumor microenvironment, the outer LOD dissociates and releases the HNK to induce ferroptosis. Moreover, the released dopant Ir4+ and disulfide-bridged organosilica frameworks deplete intracellular glutathione (GSH), which is followed by GSH-mediated Ir(IV)/Ir(III) conversion. This leads to the repression of glutathione peroxidase 4 (GPX4) activity and decomposition of intratumoral hydrogen peroxide (H2O2) into hydroxyl radicals (•OH) by Ir3+-mediated Fenton-like reactions. Moreover, LOD efficiently depletes lactic acid to facilitate the generation of H2O2 and boost the Fenton reaction, which in turn enhances ROS generation. With the synergistic effects of these cascade reactions and the release of HNK, notable ferroptosis efficacy was observed both in vitro and in vivo. This combination of natural product-induced and lactic acid-responsive sequential production of H2O2 as well as the consumption of glutathione may provide a new paradigm for achieving effective ferroptosis-based cancer therapy.
Collapse
Affiliation(s)
- Weijuan Zou
- Department of Ultrasound in Medicine, Shanghai Institute of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China
| | - Feng Gao
- Department of Ultrasonic Imaging, the First Hospital of Shanxi Medical University, Taiyuan, 030001, PR China
| | - Zheying Meng
- Department of Ultrasound in Medicine, Shanghai Institute of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China
| | - Xiaojun Cai
- Department of Ultrasound in Medicine, Shanghai Institute of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China
| | - Wu Chen
- Department of Ultrasonic Imaging, the First Hospital of Shanxi Medical University, Taiyuan, 030001, PR China
| | - Yuanyi Zheng
- Department of Ultrasound in Medicine, Shanghai Institute of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China
| | - Tao Ying
- Department of Ultrasound in Medicine, Shanghai Institute of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China.
| | - Longchen Wang
- Department of Ultrasound in Medicine, Shanghai Institute of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China.
| | - Jianrong Wu
- Department of Ultrasound in Medicine, Shanghai Institute of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China.
| |
Collapse
|
39
|
Xue P, Zhuang H, Bai T, Zeng X, Deng J, Shao S, Yan S. Iron (II)-based metal-organic framework nanozyme for boosting tumor ferroptosis through inhibiting DNA damage repair and system Xc . J Nanobiotechnology 2024; 22:228. [PMID: 38715049 PMCID: PMC11077818 DOI: 10.1186/s12951-024-02508-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 04/29/2024] [Indexed: 05/12/2024] Open
Abstract
Development of ferroptosis-inducible nanoplatforms with high efficiency and specificity is highly needed and challenging in tumor ferrotherapy. Here, we demonstrate highly effective tumor ferrotherapy using iron (II)-based metal-organic framework (FessMOF) nanoparticles, assembled from disulfide bonds and ferrous ions. The as-prepared FessMOF nanoparticles exhibit peroxidase-like activity and pH/glutathione-dependent degradability, which enables tumor-responsive catalytic therapy and glutathione depletion by the thiol/disulfide exchange to suppress glutathione peroxidase 4, respectively. Upon PEGylation and Actinomycin D (ActD) loading, the resulting FessMOF/ActD-PEG nanoplatform induces marked DNA damage and lipid peroxidation. Concurrently, we found that ActD can inhibit Xc- system and elicit ferritinophagy, which further boosts the ferrotherapeutic efficacy of the FessMOF/ActD-PEG. In vivo experiments demonstrate that our fabricated nanoplatform presents excellent biocompatibility and a high tumor inhibition rate of 91.89%.
Collapse
Affiliation(s)
- Panpan Xue
- The Straits Institute of Flexible Electronics (SIFE, Future Technologies), The Straits Laboratory of Flexible Electronics (SLoFE), Fujian Normal University, Fuzhou, Fujian, 350117, China
| | - Huilan Zhuang
- The Straits Institute of Flexible Electronics (SIFE, Future Technologies), The Straits Laboratory of Flexible Electronics (SLoFE), Fujian Normal University, Fuzhou, Fujian, 350117, China
| | - Tingjie Bai
- The Straits Institute of Flexible Electronics (SIFE, Future Technologies), The Straits Laboratory of Flexible Electronics (SLoFE), Fujian Normal University, Fuzhou, Fujian, 350117, China
| | - Xuemei Zeng
- Key Laboratory of Innate Immune Biology of Fujian Province, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University, 1 Keji Road, Fuzhou, 350117, PR China.
| | - Jinpeng Deng
- The Straits Institute of Flexible Electronics (SIFE, Future Technologies), The Straits Laboratory of Flexible Electronics (SLoFE), Fujian Normal University, Fuzhou, Fujian, 350117, China
| | - Sijie Shao
- The Straits Institute of Flexible Electronics (SIFE, Future Technologies), The Straits Laboratory of Flexible Electronics (SLoFE), Fujian Normal University, Fuzhou, Fujian, 350117, China
| | - Shuangqian Yan
- The Straits Institute of Flexible Electronics (SIFE, Future Technologies), The Straits Laboratory of Flexible Electronics (SLoFE), Fujian Normal University, Fuzhou, Fujian, 350117, China.
| |
Collapse
|
40
|
Wang H, Jiao D, Feng D, Liu Q, Huang Y, Hou J, Ding D, Zhang W. Transformable Supramolecular Self-Assembled Peptides for Cascade Self-Enhanced Ferroptosis Primed Cancer Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2311733. [PMID: 38339920 DOI: 10.1002/adma.202311733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 02/02/2024] [Indexed: 02/12/2024]
Abstract
Immunotherapy has received widespread attention for its effective and long-term tumor-eliminating ability. However, for immunogenic "cold" tumors, such as prostate cancer (PCa), the low immunogenicity of the tumor itself is a serious obstacle to efficacy. Here, this work reports a strategy to enhance PCa immunogenicity by triggering cascade self-enhanced ferroptosis in tumor cells, turning the tumor from "cold" to "hot". This work develops a transformable self-assembled peptide TEP-FFG-CRApY with alkaline phosphatase (ALP) responsiveness and glutathione peroxidase 4 (GPX4) protein targeting. TEP-FFG-CRApY self-assembles into nanoparticles under aqueous conditions and transforms into nanofibers in response to ALP during endosome/lysosome uptake into tumor cells, promoting lysosomal membrane permeabilization (LMP). On the one hand, the released TEP-FFG-CRAY nanofibers target GPX4 and selectively degrade the GPX4 protein under the light irradiation, inducing ferroptosis; on the other hand, the large amount of leaked Fe2+ further cascade to amplify the ferroptosis through the Fenton reaction. TEP-FFG-CRApY-induced immunogenic ferroptosis improves tumor cell immunogenicity by promoting the maturation of dendritic cells (DCs) and increasing intratumor T-cell infiltration. More importantly, recovered T cells further enhance ferroptosis by secreting large amounts of interferon-gamma (IFN-γ). This work provides a novel strategy for the molecular design of synergistic molecularly targeted therapy for immunogenic "cold" tumors.
Collapse
Affiliation(s)
- He Wang
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Di Jiao
- Frontiers Science Center for Cell Responses, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education and College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Dexiang Feng
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Qian Liu
- Department of Urology, Tianjin First Central Hospital, Tianjin, 300192, China
| | - Yuhua Huang
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Jianquan Hou
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Dan Ding
- Frontiers Science Center for Cell Responses, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education and College of Life Sciences, Nankai University, Tianjin, 300071, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Weijie Zhang
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| |
Collapse
|
41
|
Cao J, Zhu C, Cao Z, Ke X. CPPs-modified chitosan as permeability-enhancing chemotherapeutic combined with gene therapy nanosystem by thermosensitive hydrogel for the treatment of osteosarcoma. Int J Biol Macromol 2024; 267:130915. [PMID: 38561118 DOI: 10.1016/j.ijbiomac.2024.130915] [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: 12/19/2023] [Revised: 03/11/2024] [Accepted: 03/13/2024] [Indexed: 04/04/2024]
Abstract
BACKGROUND Chemotherapy resistance of osteosarcoma (OS) is still the crux of poor clinical curative effect.E3 ubiquitin-protein ligase Rad18 (Rad18) contributed to doxorubicin resistance in OS, which ultimately mediated DNA damage tolerance and led to a poor prognosis and chemotherapy response in patients. METHODS In this study, doxorubicin was loaded in the process of Fe2+ and siRad18 forming nanoparticles(FSD) through coordination, chitosan modified with cell penetrating peptide (H6R6) was synthesized and coated on the surface of the NPs(FSD-CHR). FSD-CHR was then dispersed in thermosensitive hydrogel(PPP) for peritumoral injection of osteosarcoma in situ. Subsequently, the physicochemical properties and molecular biological characteristics of the drug delivery system were characterized. Finally, an osteosarcoma model was established to study the anti-tumor effects of multifunctional nanoparticles and the immunotherapy effect combined with αPD-L1. RESULTS FSD-CHR has enhanced tumor tissue permeability, siRad18 can significantly reduce Dox-mediated DNA damage tolerance and enhance anti-tumor effects, and iron-based NPs show enhanced ROS upregulation. FSD-CHR@PPP showed significant inhibition of osteosarcoma growth in vivo and a reduced incidence of lung metastasis. In addition, siRad18 was unexpectedly found to enhance Dox-mediated immunogenic cell death (ICD).FSD-CHR@PPP combined with PD-L1 blocking significantly enhanced anti-tumor effects due to decreased PD-L1 enrichment. CONCLUSION Hydrogel encapsulation of permeable nanoparticles provides an effective strategy for doxorubicin-resistant OS, showing that gene therapy blocking DNA damage tolerance can enhance treatment response to chemotherapy and appears to enhance the effect of ICD inducers to activate the immune system.
Collapse
Affiliation(s)
- Jie Cao
- Department of Pharmaceutics, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, Jiangsu province, China
| | - Chenghong Zhu
- Department of Pharmaceutics, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, Jiangsu province, China
| | - Ziqi Cao
- Department of Pharmaceutics, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, Jiangsu province, China
| | - Xue Ke
- Department of Pharmaceutics, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, Jiangsu province, China.
| |
Collapse
|
42
|
Kontoghiorghes GJ. The Importance and Essentiality of Natural and Synthetic Chelators in Medicine: Increased Prospects for the Effective Treatment of Iron Overload and Iron Deficiency. Int J Mol Sci 2024; 25:4654. [PMID: 38731873 PMCID: PMC11083551 DOI: 10.3390/ijms25094654] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 04/19/2024] [Accepted: 04/22/2024] [Indexed: 05/13/2024] Open
Abstract
The supply and control of iron is essential for all cells and vital for many physiological processes. All functions and activities of iron are expressed in conjunction with iron-binding molecules. For example, natural chelators such as transferrin and chelator-iron complexes such as haem play major roles in iron metabolism and human physiology. Similarly, the mainstay treatments of the most common diseases of iron metabolism, namely iron deficiency anaemia and iron overload, involve many iron-chelator complexes and the iron-chelating drugs deferiprone (L1), deferoxamine (DF) and deferasirox. Endogenous chelators such as citric acid and glutathione and exogenous chelators such as ascorbic acid also play important roles in iron metabolism and iron homeostasis. Recent advances in the treatment of iron deficiency anaemia with effective iron complexes such as the ferric iron tri-maltol complex (feraccru or accrufer) and the effective treatment of transfusional iron overload using L1 and L1/DF combinations have decreased associated mortality and morbidity and also improved the quality of life of millions of patients. Many other chelating drugs such as ciclopirox, dexrazoxane and EDTA are used daily by millions of patients in other diseases. Similarly, many other drugs or their metabolites with iron-chelation capacity such as hydroxyurea, tetracyclines, anthracyclines and aspirin, as well as dietary molecules such as gallic acid, caffeic acid, quercetin, ellagic acid, maltol and many other phytochelators, are known to interact with iron and affect iron metabolism and related diseases. Different interactions are also observed in the presence of essential, xenobiotic, diagnostic and theranostic metal ions competing with iron. Clinical trials using L1 in Parkinson's, Alzheimer's and other neurodegenerative diseases, as well as HIV and other infections, cancer, diabetic nephropathy and anaemia of inflammation, highlight the importance of chelation therapy in many other clinical conditions. The proposed use of iron chelators for modulating ferroptosis signifies a new era in the design of new therapeutic chelation strategies in many other diseases. The introduction of artificial intelligence guidance for optimal chelation therapeutic outcomes in personalised medicine is expected to increase further the impact of chelation in medicine, as well as the survival and quality of life of millions of patients with iron metabolic disorders and also other diseases.
Collapse
Affiliation(s)
- George J Kontoghiorghes
- Postgraduate Research Institute of Science, Technology, Environment and Medicine, Limassol 3021, Cyprus
| |
Collapse
|
43
|
Chen X, Poetsch A. The Role of Cdo1 in Ferroptosis and Apoptosis in Cancer. Biomedicines 2024; 12:918. [PMID: 38672271 PMCID: PMC11047957 DOI: 10.3390/biomedicines12040918] [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: 03/19/2024] [Revised: 04/08/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
Cysteine dioxygenase type 1 (Cdo1) is a tumor suppressor gene. It regulates the metabolism of cysteine, thereby influencing the cellular antioxidative capacity. This function puts Cdo1 in a prominent position to promote ferroptosis and apoptosis. Cdo1 promotes ferroptosis mainly by decreasing the amounts of antioxidants, leading to autoperoxidation of the cell membrane through Fenton reaction. Cdo1 promotes apoptosis mainly through the product of cysteine metabolism, taurine, and low level of antioxidants. Many cancers exhibit altered function of Cdo1, underscoring its crucial role in cancer cell survival. Genetic and epigenetic alterations have been found, with methylation of Cdo1 promoter as the most common mutation. The fact that no cancer was found to be caused by altered Cdo1 function alone indicates that the tumor suppressor role of Cdo1 is mild. By compiling the current knowledge about apoptosis, ferroptosis, and the role of Cdo1, this review suggests possibilities for how the mild anticancer role of Cdo1 could be harnessed in new cancer therapies. Here, developing drugs targeting Cdo1 is considered meaningful in neoadjuvant therapies, for example, helping against the development of anti-cancer drug resistance in tumor cells.
Collapse
Affiliation(s)
| | - Ansgar Poetsch
- Queen Mary School, Nanchang University, Nanchang 330047, China;
| |
Collapse
|
44
|
Meinert M, Jessen C, Hufnagel A, Kreß JKC, Burnworth M, Däubler T, Gallasch T, Xavier da Silva TN, Dos Santos AF, Ade CP, Schmitz W, Kneitz S, Friedmann Angeli JP, Meierjohann S. Thiol starvation triggers melanoma state switching in an ATF4 and NRF2-dependent manner. Redox Biol 2024; 70:103011. [PMID: 38219574 PMCID: PMC10825660 DOI: 10.1016/j.redox.2023.103011] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 12/20/2023] [Accepted: 12/21/2023] [Indexed: 01/16/2024] Open
Abstract
The cystine/glutamate antiporter xCT is an important source of cysteine for cancer cells. Once taken up, cystine is reduced to cysteine and serves as a building block for the synthesis of glutathione, which efficiently protects cells from oxidative damage and prevents ferroptosis. As melanomas are particularly exposed to several sources of oxidative stress, we investigated the biological role of cysteine and glutathione supply by xCT in melanoma. xCT activity was abolished by genetic depletion in the Tyr::CreER; BrafCA; Ptenlox/+ melanoma model and by acute cystine withdrawal in melanoma cell lines. Both interventions profoundly impacted melanoma glutathione levels, but they were surprisingly well tolerated by murine melanomas in vivo and by most human melanoma cell lines in vitro. RNA sequencing of human melanoma cells revealed a strong adaptive upregulation of NRF2 and ATF4 pathways, which orchestrated the compensatory upregulation of genes involved in antioxidant defence and de novo cysteine biosynthesis. In addition, the joint activation of ATF4 and NRF2 triggered a phenotypic switch characterized by a reduction of differentiation genes and induction of pro-invasive features, which was also observed after erastin treatment or the inhibition of glutathione synthesis. NRF2 alone was capable of inducing the phenotypic switch in a transient manner. Together, our data show that cystine or glutathione levels regulate the phenotypic plasticity of melanoma cells by elevating ATF4 and NRF2.
Collapse
Affiliation(s)
- Madlen Meinert
- Department of Physiological Chemistry, University of Würzburg, Würzburg, Germany
| | - Christina Jessen
- Institute of Pathology, University of Würzburg, Würzburg, Germany
| | - Anita Hufnagel
- Institute of Pathology, University of Würzburg, Würzburg, Germany
| | | | - Mychal Burnworth
- Institute of Pathology, University of Würzburg, Würzburg, Germany
| | - Theo Däubler
- Institute of Pathology, University of Würzburg, Würzburg, Germany
| | - Till Gallasch
- Institute of Pathology, University of Würzburg, Würzburg, Germany
| | | | - Ancély Ferreira Dos Santos
- Rudolf-Virchow Center for Integrative and Translational Bioimaging, University of Würzburg, Würzburg, Germany
| | - Carsten Patrick Ade
- Department of Biochemistry and Molecular Biology, University of Würzburg, Würzburg, Germany
| | - Werner Schmitz
- Department of Biochemistry and Molecular Biology, University of Würzburg, Würzburg, Germany
| | - Susanne Kneitz
- Department of Biochemistry and Cell Biology, University of Würzburg, Würzburg, Germany
| | - José Pedro Friedmann Angeli
- Rudolf-Virchow Center for Integrative and Translational Bioimaging, University of Würzburg, Würzburg, Germany
| | - Svenja Meierjohann
- Department of Physiological Chemistry, University of Würzburg, Würzburg, Germany; Institute of Pathology, University of Würzburg, Würzburg, Germany; Comprehensive Cancer Center Mainfranken, University Hospital Würzburg, Würzburg, Germany.
| |
Collapse
|
45
|
Zhou Y, Chen Z, Yang M, Chen F, Yin J, Zhang Y, Zhou X, Sun X, Ni Z, Chen L, Lv Q, Zhu F, Liu S. FERREG: ferroptosis-based regulation of disease occurrence, progression and therapeutic response. Brief Bioinform 2024; 25:bbae223. [PMID: 38742521 PMCID: PMC11091744 DOI: 10.1093/bib/bbae223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 03/25/2024] [Accepted: 04/21/2024] [Indexed: 05/16/2024] Open
Abstract
Ferroptosis is a non-apoptotic, iron-dependent regulatory form of cell death characterized by the accumulation of intracellular reactive oxygen species. In recent years, a large and growing body of literature has investigated ferroptosis. Since ferroptosis is associated with various physiological activities and regulated by a variety of cellular metabolism and mitochondrial activity, ferroptosis has been closely related to the occurrence and development of many diseases, including cancer, aging, neurodegenerative diseases, ischemia-reperfusion injury and other pathological cell death. The regulation of ferroptosis mainly focuses on three pathways: system Xc-/GPX4 axis, lipid peroxidation and iron metabolism. The genes involved in these processes were divided into driver, suppressor and marker. Importantly, small molecules or drugs that mediate the expression of these genes are often good treatments in the clinic. Herein, a newly developed database, named 'FERREG', is documented to (i) providing the data of ferroptosis-related regulation of diseases occurrence, progression and drug response; (ii) explicitly describing the molecular mechanisms underlying each regulation; and (iii) fully referencing the collected data by cross-linking them to available databases. Collectively, FERREG contains 51 targets, 718 regulators, 445 ferroptosis-related drugs and 158 ferroptosis-related disease responses. FERREG can be accessed at https://idrblab.org/ferreg/.
Collapse
Affiliation(s)
- Yuan Zhou
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, School of Pharmacy, and Department of Respiratory Medicine of Affiliated Hospital, Hangzhou Normal University, Hangzhou, 311121, China
| | - Zhen Chen
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Mengjie Yang
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, School of Pharmacy, and Department of Respiratory Medicine of Affiliated Hospital, Hangzhou Normal University, Hangzhou, 311121, China
| | - Fengyun Chen
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, School of Pharmacy, and Department of Respiratory Medicine of Affiliated Hospital, Hangzhou Normal University, Hangzhou, 311121, China
| | - Jiayi Yin
- Department of Clinical Pharmacy, The First Affiliated Hospital, Zhejiang University School of Medicine
| | - Yintao Zhang
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Xuheng Zhou
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Xiuna Sun
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Ziheng Ni
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, School of Pharmacy, and Department of Respiratory Medicine of Affiliated Hospital, Hangzhou Normal University, Hangzhou, 311121, China
| | - Lu Chen
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, School of Pharmacy, and Department of Respiratory Medicine of Affiliated Hospital, Hangzhou Normal University, Hangzhou, 311121, China
| | - Qun Lv
- Department of Respiratory, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, 311121, China
| | - Feng Zhu
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou, 330110, China
| | - Shuiping Liu
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, School of Pharmacy, and Department of Respiratory Medicine of Affiliated Hospital, Hangzhou Normal University, Hangzhou, 311121, China
| |
Collapse
|
46
|
Duché G, Sanderson JM. The Chemical Reactivity of Membrane Lipids. Chem Rev 2024; 124:3284-3330. [PMID: 38498932 PMCID: PMC10979411 DOI: 10.1021/acs.chemrev.3c00608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 02/27/2024] [Accepted: 02/28/2024] [Indexed: 03/20/2024]
Abstract
It is well-known that aqueous dispersions of phospholipids spontaneously assemble into bilayer structures. These structures have numerous applications across chemistry and materials science and form the fundamental structural unit of the biological membrane. The particular environment of the lipid bilayer, with a water-poor low dielectric core surrounded by a more polar and better hydrated interfacial region, gives the membrane particular biophysical and physicochemical properties and presents a unique environment for chemical reactions to occur. Many different types of molecule spanning a range of sizes, from dissolved gases through small organics to proteins, are able to interact with membranes and promote chemical changes to lipids that subsequently affect the physicochemical properties of the bilayer. This Review describes the chemical reactivity exhibited by lipids in their membrane form, with an emphasis on conditions where the lipids are well hydrated in the form of bilayers. Key topics include the following: lytic reactions of glyceryl esters, including hydrolysis, aminolysis, and transesterification; oxidation reactions of alkenes in unsaturated fatty acids and sterols, including autoxidation and oxidation by singlet oxygen; reactivity of headgroups, particularly with reactive carbonyl species; and E/Z isomerization of alkenes. The consequences of reactivity for biological activity and biophysical properties are also discussed.
Collapse
Affiliation(s)
- Genevieve Duché
- Génie
Enzimatique et Cellulaire, Université
Technologique de Compiègne, Compiègne 60200, France
| | - John M Sanderson
- Chemistry
Department, Durham University, Durham DH1 3LE, United Kingdom
| |
Collapse
|
47
|
Jiang X, Peng Q, Peng M, Oyang L, Wang H, Liu Q, Xu X, Wu N, Tan S, Yang W, Han Y, Lin J, Xia L, Tang Y, Luo X, Dai J, Zhou Y, Liao Q. Cellular metabolism: A key player in cancer ferroptosis. Cancer Commun (Lond) 2024; 44:185-204. [PMID: 38217522 PMCID: PMC10876208 DOI: 10.1002/cac2.12519] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 12/25/2023] [Accepted: 01/02/2024] [Indexed: 01/15/2024] Open
Abstract
Cellular metabolism is the fundamental process by which cells maintain growth and self-renewal. It produces energy, furnishes raw materials, and intermediates for biomolecule synthesis, and modulates enzyme activity to sustain normal cellular functions. Cellular metabolism is the foundation of cellular life processes and plays a regulatory role in various biological functions, including programmed cell death. Ferroptosis is a recently discovered form of iron-dependent programmed cell death. The inhibition of ferroptosis plays a crucial role in tumorigenesis and tumor progression. However, the role of cellular metabolism, particularly glucose and amino acid metabolism, in cancer ferroptosis is not well understood. Here, we reviewed glucose, lipid, amino acid, iron and selenium metabolism involvement in cancer cell ferroptosis to elucidate the impact of different metabolic pathways on this process. Additionally, we provided a detailed overview of agents used to induce cancer ferroptosis. We explained that the metabolism of tumor cells plays a crucial role in maintaining intracellular redox homeostasis and that disrupting the normal metabolic processes in these cells renders them more susceptible to iron-induced cell death, resulting in enhanced tumor cell killing. The combination of ferroptosis inducers and cellular metabolism inhibitors may be a novel approach to future cancer therapy and an important strategy to advance the development of treatments.
Collapse
Affiliation(s)
- Xianjie Jiang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, P. R. China
- Public Service Platform of Tumor Organoids Technology, Changsha, Hunan, P. R. China
| | - Qiu Peng
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, P. R. China
- Public Service Platform of Tumor Organoids Technology, Changsha, Hunan, P. R. China
| | - Mingjing Peng
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, P. R. China
- Public Service Platform of Tumor Organoids Technology, Changsha, Hunan, P. R. China
| | - Linda Oyang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, P. R. China
- Public Service Platform of Tumor Organoids Technology, Changsha, Hunan, P. R. China
| | - Honghan Wang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, P. R. China
- Department of Head and Neck Surgery, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, P. R. China
| | - Qiang Liu
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, P. R. China
- Public Service Platform of Tumor Organoids Technology, Changsha, Hunan, P. R. China
| | - Xuemeng Xu
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, P. R. China
- Hengyang Medical School, University of South China, Hengyang, Hunan, P. R. China
| | - Nayiyuan Wu
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, P. R. China
- Public Service Platform of Tumor Organoids Technology, Changsha, Hunan, P. R. China
| | - Shiming Tan
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, P. R. China
| | - Wenjuan Yang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, P. R. China
| | - Yaqian Han
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, P. R. China
- Public Service Platform of Tumor Organoids Technology, Changsha, Hunan, P. R. China
| | - Jinguan Lin
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, P. R. China
| | - Longzheng Xia
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, P. R. China
| | - Yanyan Tang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, P. R. China
- Public Service Platform of Tumor Organoids Technology, Changsha, Hunan, P. R. China
| | - Xia Luo
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, P. R. China
- Public Service Platform of Tumor Organoids Technology, Changsha, Hunan, P. R. China
| | - Jie Dai
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, P. R. China
- Department of Head and Neck Surgery, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, P. R. China
| | - Yujuan Zhou
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, P. R. China
- Public Service Platform of Tumor Organoids Technology, Changsha, Hunan, P. R. China
| | - Qianjin Liao
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, P. R. China
- Public Service Platform of Tumor Organoids Technology, Changsha, Hunan, P. R. China
| |
Collapse
|
48
|
Tang X, Niu Y, Jian J, Guo Y, Wang Y, Zhu Y, Liu B. Potential applications of ferroptosis inducers and regulatory molecules in hematological malignancy therapy. Crit Rev Oncol Hematol 2024; 193:104203. [PMID: 37979734 DOI: 10.1016/j.critrevonc.2023.104203] [Citation(s) in RCA: 2] [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: 10/31/2023] [Accepted: 11/13/2023] [Indexed: 11/20/2023] Open
Abstract
Ferroptosis, a novel form of iron-dependent cell death, has emerged as a potential avenue for promoting tumor cell death by causing cell membrane rupture and the accumulation of lipid peroxides (LPO) in the cell. Since its discovery in 2012, extensive research has been conducted to explore the mechanism of ferroptosis inducers, including erastin, sulfasalazine, and sorafenib. These compounds inhibit system XC-, while Ras-selective lethal small molecule 3 (RSL3) and FION2 specifically target GPX4 to promote ferroptosis. Therefore, targeting ferroptosis presents a promising therapeutic approach for malignant tumors. While the study of ferroptosis in solid tumors has made significant progress, there is limited information available on its role in hematological tumors. This review aims to summarize the molecular mechanisms of ferroptosis inducers and discuss their clinical applications in hematological malignancies. Furthermore, the identification of non-coding RNAs (ncRNAs) and genes that regulate key molecules in the ferroptosis pathway could provide new targets and establish a molecular theoretical foundation for exploring novel ferroptosis inducers in hematological malignancies.
Collapse
Affiliation(s)
- Xiao Tang
- The First Clinical Medical College, Lanzhou University, Lanzhou 730099, China
| | - Yujie Niu
- The First Clinical Medical College, Lanzhou University, Lanzhou 730099, China
| | - Jinli Jian
- The First Clinical Medical College, Lanzhou University, Lanzhou 730099, China
| | - Yuancheng Guo
- The First Clinical Medical College, Lanzhou University, Lanzhou 730099, China
| | - Yin Wang
- The First Clinical Medical College, Lanzhou University, Lanzhou 730099, China
| | - Yu Zhu
- The First Clinical Medical College, Lanzhou University, Lanzhou 730099, China
| | - Bei Liu
- The First Clinical Medical College, Lanzhou University, Lanzhou 730099, China; Department of Hematology, The First Affiliated Hospital, Lanzhou University, Lanzhou 730099, China.
| |
Collapse
|
49
|
Zhang Y, Jiang C, Meng N. Targeting Ferroptosis: A Novel Strategy for the Treatment of Atherosclerosis. Mini Rev Med Chem 2024; 24:1262-1276. [PMID: 38284727 DOI: 10.2174/0113895575273164231130070920] [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: 08/09/2023] [Revised: 10/04/2023] [Accepted: 10/11/2023] [Indexed: 01/30/2024]
Abstract
Since ferroptosis was reported in 2012, its application prospects in various diseases have been widely considered, initially as a treatment direction for tumors. Recent studies have shown that ferroptosis is closely related to the occurrence and development of atherosclerosis. The primary mechanism is to affect the occurrence and development of atherosclerosis through intracellular iron homeostasis, ROS and lipid peroxide production and metabolism, and a variety of intracellular signaling pathways. Inhibition of ferroptosis is effective in inhibiting the development of atherosclerosis, and it can bring a new direction for treating atherosclerosis. In this review, we discuss the mechanism of ferroptosis and focus on the relationship between ferroptosis and atherosclerosis, summarize the different types of ferroptosis inhibitors that have been widely studied, and discuss some issues worthy of attention in the treatment of atherosclerosis by targeting ferroptosis.
Collapse
Affiliation(s)
- Yifan Zhang
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Chengshi Jiang
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Ning Meng
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| |
Collapse
|
50
|
Yadav VK, Choudhary N, Gacem A, Verma RK, Abul Hasan M, Tarique Imam M, Almalki ZS, Yadav KK, Park HK, Ghosh T, Kumar P, Patel A, Kalasariya H, Jeon BH, Ali AlMubarak H. Deeper insight into ferroptosis: association with Alzheimer's, Parkinson's disease, and brain tumors and their possible treatment by nanomaterials induced ferroptosis. Redox Rep 2023; 28:2269331. [PMID: 38010378 PMCID: PMC11001282 DOI: 10.1080/13510002.2023.2269331] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2023] Open
Abstract
Ferroptosis is an emerging and novel type of iron-dependent programmed cell death which is mainly caused by the excessive deposition of free intracellular iron in the brain cells. This deposited free iron exerts a ferroptosis pathway, resulting in lipid peroxidation (LiPr). There are mainly three ferroptosis pathways viz. iron metabolism-mediated cysteine/glutamate, and LiPr-mediated. Iron is required by the brain as a redox metal for several physiological activities. Due to the iron homeostasis balance disruption, the brain gets adversely affected which further causes neurodegenerative diseases (NDDs) like Parkinson's and Alzheimer's disease, strokes, and brain tumors like glioblastoma (GBS), and glioma. Nanotechnology has played an important role in the prevention and treatment of these NDDs. A synergistic effect of nanomaterials and ferroptosis could prove to be an effective and efficient approach in the field of nanomedicine. In the current review, the authors have highlighted all the latest research in the field of ferroptosis, specifically emphasizing on the role of major molecular key players and various mechanisms involved in the ferroptosis pathway. Moreover, here the authors have also addressed the correlation of ferroptosis with the pathophysiology of NDDs and theragnostic effect of ferroptosis and nanomaterials for the prevention and treatment of NDDs.
Collapse
Affiliation(s)
- Virendra Kumar Yadav
- Department of Life Sciences, Hemchandracharya North Gujarat University, Patan, India
| | - Nisha Choudhary
- Department of Life Sciences, Hemchandracharya North Gujarat University, Patan, India
| | - Amel Gacem
- Department of Physics, Faculty of Sciences, University 20 Août 1955, Skikda, Algeria
| | - Rakesh Kumar Verma
- Department of Biosciences, School of Liberal Arts & Sciences, Mody University of Science and Technology, Sikar, India
| | - Mohd Abul Hasan
- Civil Engineering Department, College of Engineering, King Khalid University, Abha, Kingdom of Saudi Arabia (KSA)
| | - Mohammad Tarique Imam
- Department of Clinical Pharmacy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj, Saudi Arabia
| | - Ziyad Saeed Almalki
- Department of Clinical Pharmacy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj, Saudi Arabia
| | - Krishna Kumar Yadav
- Faculty of Science and Technology, Madhyanchal Professional University, Bhopal, India
- Environmental and Atmospheric Sciences Research Group, Scientific Research Center, Al-Ayen University, Nasiriyah, Iraq
| | - Hyun-Kyung Park
- Department of Pediatrics, Hanyang University College of Medicine, Seoul, Republic of Korea
| | - Tathagata Ghosh
- Department of Arts, School of Liberal Arts & Sciences, Mody University of Science and Technology, Sikar, India
| | - Pankaj Kumar
- Department of Environmental Science, Parul Institute of Applied Sciences, Parul University, Vadodara, India
| | - Ashish Patel
- Department of Life Sciences, Hemchandracharya North Gujarat University, Patan, India
| | - Haresh Kalasariya
- Centre for Natural Products Discovery, School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| | - Byong-Hun Jeon
- Department of Earth Resources and Environmental Engineering, Hanyang University, Seoul, Republic of Korea
| | - Hassan Ali AlMubarak
- Division of Radiology, Department of Medicine, College of Medicine and Surgery, King Khalid University (KKU), Abha, Kingdom of Saudi Arabia
| |
Collapse
|