1
|
Dong W, Xiao Z, Ma H, Cao T, Gong D, Fan Z. A mitochondrial targeted near-infrared ratio fluorescent probe for ferroptosis related hydrogen polysulfides imaging in arthritis. J Colloid Interface Sci 2025; 695:137774. [PMID: 40319520 DOI: 10.1016/j.jcis.2025.137774] [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: 03/03/2025] [Revised: 04/30/2025] [Accepted: 05/01/2025] [Indexed: 05/07/2025]
Abstract
Hydrogen polysulfide (H2Sn, n > 1), as an active sulfur substance derived from hydrogen sulfide, is considered a new potential signaling molecule that occupies an irreplaceable position in physiological regulation and signal transduction processes. Ferroptosis, an iron-dependent form of programmed cell death, is caused by the iron dependent accumulation of lipid peroxides associated with reactive oxygen species. The process of ferroptosis generates abundant reactive oxygen species, indirectly promoting the elevation of intracellular hydrogen polysulfide levels. There is evidence to suggest a close relationship between ferroptosis and arthritis. This work developed a near-infrared ratiometric fluorescent probe (Mito-S4) with a large Stokes shift (∼240 nm) based on nucleophilic addition mechanism, which can quickly (18 s) and sensitively (Detection Limit = 0.165 μM) detect H2Sn in cells. The mechanism of action was verified through theoretical calculations and nuclear magnetic analysis, and the probe Mito-S4 was successfully applied to visualize H2Sn in cellular mitochondria. The cell imaging results showed that the probe successfully achieved tracking of H2Sn in mitochondria during inflammation and ferroptosis processes. In addition, pathological section experiments have confirmed that the probe has good in vivo imaging ability. It is worth mentioning that further in vivo imaging experiments have for the first time delved into the mechanism of H2Sn in the process of ferroptosis in arthritis, which can provide new research methods and therapeutic targets for the treatment of arthritis.
Collapse
Affiliation(s)
- Wenhua Dong
- Key Laboratory of Magnetic Molecules & Magnetic Information Materials Ministry of Education, The School of Chemical and Material Science, Shanxi Normal University, Taiyuan 030031, PR China
| | - Ziwen Xiao
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, PR China
| | - Hong Ma
- College of Chemistry and Materials, Taiyuan Normal University, Jinzhong 030619, PR China
| | - Ting Cao
- Key Laboratory of Magnetic Molecules & Magnetic Information Materials Ministry of Education, The School of Chemical and Material Science, Shanxi Normal University, Taiyuan 030031, PR China.
| | - Deyan Gong
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, PR China.
| | - Zhefeng Fan
- Key Laboratory of Magnetic Molecules & Magnetic Information Materials Ministry of Education, The School of Chemical and Material Science, Shanxi Normal University, Taiyuan 030031, PR China.
| |
Collapse
|
2
|
Sui X, Zhou H, Wang T. GLUT1 maintains esophageal cancer stem cell-like characteristics by inhibiting autophagy-dependent ferroptosis via EGFR. Exp Cell Res 2025; 449:114600. [PMID: 40368081 DOI: 10.1016/j.yexcr.2025.114600] [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: 02/08/2025] [Revised: 05/06/2025] [Accepted: 05/07/2025] [Indexed: 05/16/2025]
Abstract
Esophageal cancer, a highly malignant tumor with poor prognosis, is characterized by the presence of cancer stem cells (CSCs) that drive tumor initiation, metastasis, and recurrence. This study investigates the molecular mechanism by which glucose transporter 1 (GLUT1) maintains esophageal CSC-like properties through regulation of autophagy-dependent ferroptosis via epidermal growth factor receptor (EGFR). Using shRNA to knock down GLUT1 or EGFR and constructing a GLUT1 overexpression vector in KYSE520 cells, we employed western blotting, qRT-PCR, flow cytometry, sphere formation, Transwell assays, and xenograft models to assess stemness markers (NANOG, OCT4, SOX2), autophagic flux (LC3B, P62, Beclin1), and ferroptosis-related parameters (ROS, Fe2+, GSH, GPX4, COX2). Mechanistic analyses included co-immunoprecipitation to validate the GLUT1-EGFR interaction, chloroquine to inhibit autophagy, and cycloheximide/MG132 to evaluate EGFR protein stability. Results showed that GLUT1 depletion reduced CSC marker expression, increased ROS and Fe2+ levels, depleted GSH, and induced lipid peroxidation, sensitizing cells to ferroptosis while activating autophagy (elevated LC3 II/I, Beclin1; reduced P62); autophagy inhibition exacerbated cell death, indicating a protective role for autophagy in this context. GLUT1 directly bound to EGFR, stabilizing the receptor by blocking ubiquitin-proteasome-mediated degradation, whereas EGFR knockdown enhanced autophagic flux and reversed GLUT1-overexpression-induced ferroptosis resistance and stemness maintenance. In vivo, GLUT1 knockdown suppressed tumor growth and lung metastasis, and clinical samples revealed a positive correlation between GLUT1 and EGFR expression, linked to advanced TNM stages and poor survival. Collectively, these findings demonstrate that GLUT1 preserves esophageal CSC-like characteristics by stabilizing EGFR to inhibit autophagy-dependent ferroptosis, highlighting targeting GLUT1 as a potential therapeutic strategy to eliminate CSCs and combat esophageal cancer progression.
Collapse
Affiliation(s)
- Xin Sui
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China.
| | - Haining Zhou
- Department of Digestive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
| | - Tingting Wang
- Department of Endocrinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
| |
Collapse
|
3
|
Wang Y, Lu J, Lin B, Chen J, Lin F, Zheng Q, Xue X, Wei Y, Chen S, Xu N. Integrated analysis of MIOX gene in prognosis of clear-cell renal cell carcinoma. Cell Death Dis 2025; 16:368. [PMID: 40341358 PMCID: PMC12062366 DOI: 10.1038/s41419-025-07698-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 04/16/2025] [Accepted: 04/25/2025] [Indexed: 05/10/2025]
Abstract
Clear-cell renal cell carcinoma (ccRCC) is a highly aggressive malignancy that originates in the kidney. It often exhibits a limited response or can be refractory to a wide range of anti-cancer therapies, including tyrosine kinase inhibitors (TKIs) and immune checkpoint inhibitors. Ferroptosis is a form of oxidative, iron-dependent cell death characterized by lipid peroxidation. Targeting ferroptosis may offer a promising alternative therapeutic strategy for cancer cells that are resistant to existing treatments. The impact of ferroptosis-related genes on the prognosis of ccRCC patients is still not fully understood. In this study, we identified 30 differentially expressed ferroptosis-related genes in ccRCC samples compared to normal tissues using data from The Cancer Genome Atlas (TCGA). Lasso regression analyses, along with Kaplan-Meier analysis, were conducted to identify genes associated with prognosis. Based on scRNA-seq and spatial transcriptome analysis, we identified specificity of MIOX in ccRCC. Furthermore, MIOX demonstrated the highest significance, highlighting its independent prognostic value as a single gene in ccRCC. Our findings suggest that MIOX could serve as potential targets for therapeutic interventions in ccRCC.
Collapse
Affiliation(s)
- Yiqiu Wang
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Jiayi Lu
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bohan Lin
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Jiayin Chen
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Fei Lin
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Qingshui Zheng
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Xueyi Xue
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Yong Wei
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Shaohao Chen
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
| | - Ning Xu
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
4
|
Seke M, Stankovic A, Zivkovic M. Capacity of fullerenols to modulate neurodegeneration induced by ferroptosis: Focus on multiple sclerosis. Mult Scler Relat Disord 2025; 97:106378. [PMID: 40088719 DOI: 10.1016/j.msard.2025.106378] [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: 12/27/2023] [Revised: 01/10/2025] [Accepted: 03/05/2025] [Indexed: 03/17/2025]
Abstract
Multiple sclerosis is an inflammatory disease of the central nervous system (CNS), characterized by oligodendrocyte loss and demyelination of axons leading to neurodegeneration and severe neurological disability. Despite the existing drugs that have immunomodulatory effects an adequate therapy that slow down or stop neuronal death has not yet been found. Oxidative stress accompanied by excessive release of iron into the extracellular space, mitochondrial damage and lipid peroxidation are important factors in the controlled cell death named ferroptosis, latterly recognized in MS. As the fullerenols exhibit potent antioxidant activity, recent results imply that they could have protective effects by suppressing ferroptosis. Based on the current knowledge we addressed the main mechanisms of the protective effects of fullerenols in the CNS in relation to ferroptosis. Inhibition of inflammation, iron overload and lipid peroxidation through the signal transduction mechanism of Nuclear Factor Erythroid 2-Related Factor 2 (NRF2), chelation of heavy metals and free radical scavenging using fullerenols are proposed as benefitial strategy preventing MS progression. Current review connects ferroptosis molecular targets and important factors of MS progression, with biomedical properties and mechanisms of fullerenols' actions, to propose new treatment strategies that could be addaptobale in other neurodegenerative diseases.
Collapse
Affiliation(s)
- Mariana Seke
- Laboratory for Radiobiology and Molecular Genetics, ˮVinčaˮ Institute of Nuclear Sciences -National Institute of The Republic of Serbia, University of Belgrade, Mike Petrovica Alasa 12-14, Belgrade 11 000, Serbia
| | - Aleksandra Stankovic
- Laboratory for Radiobiology and Molecular Genetics, ˮVinčaˮ Institute of Nuclear Sciences -National Institute of The Republic of Serbia, University of Belgrade, Mike Petrovica Alasa 12-14, Belgrade 11 000, Serbia
| | - Maja Zivkovic
- Laboratory for Radiobiology and Molecular Genetics, ˮVinčaˮ Institute of Nuclear Sciences -National Institute of The Republic of Serbia, University of Belgrade, Mike Petrovica Alasa 12-14, Belgrade 11 000, Serbia.
| |
Collapse
|
5
|
Ma C, Hu H, Liu H, Zhong C, Wu B, Lv C, Tian Y. Lipotoxicity, lipid peroxidation and ferroptosis: a dilemma in cancer therapy. Cell Biol Toxicol 2025; 41:75. [PMID: 40285867 PMCID: PMC12033115 DOI: 10.1007/s10565-025-10025-7] [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: 11/27/2024] [Accepted: 04/13/2025] [Indexed: 04/29/2025]
Abstract
The vulnerability of tumor cells to lipid peroxidation, driven by redox imbalance and lipid overabundance within the tumor microenvironment (TME), has become a focal point for novel antitumor strategies. Ferroptosis, a form of regulated cell death predicated on lipid peroxidation, is emerging as a promising approach. Beyond their role in directly eliminating tumor cells, lipid peroxidation and its products, such as 4-hydroxynonenal (HNE), exert an additional influence by damaging DNA and shaping an environment conducive to tumor growth and metastasis. This process polarizes macrophages towards a pro-inflammatory phenotype, dampens the antigen-presenting capacity of dendritic cells (DCs), and undermines the cytotoxic functions of T and NK cells. Furthermore, it transforms neutrophils into pro-tumorigenic polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs). The lipid peroxidation of stroma cells also contributes to tumor progression. Although advanced nanotherapies have shown the ability to target tumor cells precisely, they often overlook the nuanced effects of lipid peroxidation products. In this review, we highlight a synergistic mechanism in which lipid peroxidation products and ferroptosis contribute to an immunosuppressive state that is temporally distinct from cell death. This insight broadens our understanding of ferroptosis-derived immunosuppression, encompassing all types of immune cells within the TME. This review aims to catalyze further research in this underexplored area, emphasizing the potential of lipid peroxidation products to hinder the clinical translation of ferroptosis-based therapies.
Collapse
Affiliation(s)
- Chuhan Ma
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China
| | - Huixin Hu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China
| | - Hao Liu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China
| | - Chongli Zhong
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China
| | - Baokang Wu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China
| | - Chao Lv
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China.
| | - Yu Tian
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China.
| |
Collapse
|
6
|
Wang Y, Dang H, Zhu G, Tian Y. PDP1 related ferroptosis risk signature indicates distinct immune microenvironment and prognosis of breast cancer patients. Front Pharmacol 2025; 16:1551325. [PMID: 40337509 PMCID: PMC12055530 DOI: 10.3389/fphar.2025.1551325] [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: 12/25/2024] [Accepted: 02/17/2025] [Indexed: 05/09/2025] Open
Abstract
Objective We aim to construct a RiskScore model to aid in the early prognosis of breast cancer (BC). Methods BC mRNA expression profiles were obtained from TCGA and GEO databases. Differential gene expression analysis identifies PDP1-ferroptosis-related genes. LASSO Cox regression was utilized to screen genes to build a RiskScore model, and survival analysis were performed to investigate the reliability in BC prognosis. Immune cell infiltration proportions were calculated using CIBERSORT and xCell algorithms. Single-cell data processing and analysis were conducted using "Seurat", "monocle", and "iTALK" packages. PDP1 was silenced to validate its influence on the target genes. Results Data from public databases revealed significant upregulation of PDP1 in BC samples compared to normal tissues. A RiskScore model based on PDP1-related differential ferroptosis-related genes (FRGs) ACSL1, BNIP3, and EMC2 was developed, which effectively predicted BC patient prognosis. High-risk BC samples exhibited poorer overall survival and were associated with immune microenvironment. The model remained significant in multivariate Cox regression analysis, indicating that it could independently predict the survival of BC patients. ACSL1, BNIP3, and EMC2 were downregulated after knockdown of PDP1. Conclusion RiskScore model constructed by PDP1-ferroptosis-related genes ACSL1, BNIP3, and EMC2 is able to help predict the prognosis of BC patients.
Collapse
Affiliation(s)
- Yufeng Wang
- Department of Breast Medical Oncology, Affiliated Cancer Hospital of Sun Yat-sen University, Gansu Hospital, Lanzhou, Gansu, China
| | - Huifen Dang
- Department of Breast Medical Oncology, Affiliated Cancer Hospital of Sun Yat-sen University, Gansu Hospital, Lanzhou, Gansu, China
| | - Gongjian Zhu
- Department of Science and Education Section, Affiliated Cancer Hospital of Sun Yat-sen University, Gansu Hospital, Lanzhou, Gansu, China
| | - Yingxia Tian
- Department of Breast Medical Oncology, Affiliated Cancer Hospital of Sun Yat-sen University, Gansu Hospital, Lanzhou, Gansu, China
| |
Collapse
|
7
|
He L, Ren W, Cheng W, Chen J, Lai J, Wu Y, Wu Z, Bao D, Wei Y, Piao JG. Arsenene-Vanadene nanodots co-activate Apoptosis/Ferroptosis for enhanced chemo-immunotherapy. Acta Biomater 2025; 196:453-470. [PMID: 40032219 DOI: 10.1016/j.actbio.2025.02.059] [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: 11/06/2024] [Revised: 02/21/2025] [Accepted: 02/26/2025] [Indexed: 03/05/2025]
Abstract
Triple-Negative Breast Cancer (TNBC) represents a highly aggressive subtype of breast cancer with an unfavorable prognosis, characterized by minimal immune infiltration and pronounced immune suppression, resulting in a limited response to immunotherapy. In this study, a multifunctional Arsenene-Vanadene nanodot (AsV) drug delivery system is introduced, which responds to the tumor microenvironment by releasing arsenic and vanadium. Arsenic undergoes oxidation to generate highly toxic trivalent arsenic, which induces apoptosis in tumor cells while utilizing apoptotic cell debris to transiently activate the immune system. Additionally, arsenic binds to cysteine, indirectly facilitating ferroptosis. Concurrently, vanadium's redox cycling properties are harnessed to trigger a Fenton-like reaction, promoting lipid peroxidation. Furthermore, ferroptosis is enhanced through the depletion of glutathione and inactivation of glutathione peroxidase 4 (GPX4), leading to the release of damage-associated molecular patterns and thereby amplifying the anti-tumor immune response. This study represents the first instance of integrating arsenene's apoptosis-inducing properties with vanadium's ferroptosis-enhancing effects, providing a synergistic approach to improving the immunotherapeutic response and offering a potential strategy for enhancing TNBC prognosis. STATEMENT OF SIGNIFICANCE: Triple-negative breast cancer (TNBC) exhibits resistance to immunotherapy due to its highly immunosuppressive tumor microenvironment. In this study, tumour-responsive Arsenene-Vanadene nanodots (AsV) were developed to induce a synergistic effect by triggering apoptosis and ferroptosis through microenvironment-specific mechanisms. The arsenic component generates cytotoxic trivalent arsenic, promoting apoptosis while binding to cysteine, thereby reducing GSH synthesis. Simultaneously, vanadium initiates lipid peroxidation through Fenton-like reactions and disruption of the glutathione/GPX4 axis, further amplifying ferroptotic cell death. This dual-action system transforms tumor cell debris into immune-stimulating signals while circumventing conventional immunotherapy limitations. As the first strategy integrating arsenic-induced apoptosis with vanadium-enhanced ferroptosis, this approach provides a mechanistic framework to overcome TNBC immunosuppression through coordinated cell death pathways, demonstrating potential for precision nanomedicine applications.
Collapse
Affiliation(s)
- Li He
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, PR China
| | - WeiYe Ren
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, PR China
| | - WeiYi Cheng
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, PR China
| | - JingQuan Chen
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, PR China
| | - Jianjun Lai
- Department of Oncology, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou 310013, PR China
| | - Yajun Wu
- Department of Oncology, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou 310013, PR China
| | - Zhibing Wu
- Department of Oncology, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou 310013, PR China; Department of Radiation Oncology, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou 310013, PR China.
| | - Dandan Bao
- Department of Dermatology & Cosmetology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310006, PR China.
| | - Yinghui Wei
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, PR China; Jinhua Academy of Zhejiang Chinese Medical University.
| | - Ji-Gang Piao
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, PR China.
| |
Collapse
|
8
|
Khalilzad MA, Mohammadi J, Amirsaadat S, Najafi S, Zare S, Nilforoushzadeh MA, Khalilzad M, Amirkhani MA, Peyrovan A, Khalili SFS, Farahani A, Zare S. Therapeutic potential of apoptotic vesicles in modulating inflammation, immune responses, and tissue regeneration. J Nanobiotechnology 2025; 23:260. [PMID: 40170079 PMCID: PMC11960034 DOI: 10.1186/s12951-025-03278-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 02/24/2025] [Indexed: 04/03/2025] Open
Abstract
The process of apoptosis plays a crucial role in tissue homeostasis, immune system regulation, and organ formation. Apoptotic vesicles (ApoEVs) are involved in efferocytosis, the process by which phagocytes ingest dead cells. ApoEVs also have potential therapeutic applications in cancer treatment, ischemic diseases, and their anti-inflammatory properties make them incredibly versatile for medical applications. These vesicles can induce apoptosis in cancer cells, provide tumor antigens for cancer vaccines, and even serve as effective drug delivery systems. Moreover, they can target hypoxic cells, inhibit inflammatory cell death pathways, and promote tissue regeneration. Also, their potential in addressing inflammatory disorders such as gastrointestinal ailments, osteoarthritis, and diabetes is promising. Additionally, ApoEVs can polarize anti-inflammatory immune cells and suppress inflammatory immune responses which make them a viable option for addressing the unmet need for novel anti-inflammatory medications. Despite a wealth of reviews examining the applications of ApoEVs, very few have thoroughly investigated the mechanisms underlying their anti-inflammatory effects. This distinctive approach positions the current review as timely and immensely relevant, illuminating the intriguing ways these entities function beyond their established advantages.
Collapse
Affiliation(s)
- Mohammad Amin Khalilzad
- Department of Life Sciences Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, 143951561, Iran
- Skin and Stem Cell Research Center, Tehran University of Medical Sciences, Tehran, Iran
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Javad Mohammadi
- Department of Life Sciences Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, 143951561, Iran.
| | - Soumayeh Amirsaadat
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sajad Najafi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Biotechnology and Medicinal Plants Research Center, Ilam University of Medical Sciences, Ilam, Iran
| | - Sona Zare
- Skin and Stem Cell Research Center, Tehran University of Medical Sciences, Tehran, Iran.
- Laserin Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Stem Cell and Regenerative Medicine Institute, Sharif University of Technology, Tehran, Iran.
- Department of Mechanical Engineering, Sharif University of Technology, Tehran, Iran.
| | - Mohammad Ali Nilforoushzadeh
- Skin and Stem Cell Research Center, Tehran University of Medical Sciences, Tehran, Iran.
- Skin Repair Research Center, Jordan Dermatology and Hair Transplantation Center, Tehran, Iran.
| | - Mitra Khalilzad
- Brain Mapping Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Aysan Peyrovan
- Skin and Stem Cell Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Atefeh Farahani
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Solmaz Zare
- Laserin Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
9
|
Lan J, Cai D, Gou S, Bai Y, Lei H, Li Y, Chen Y, Zhao Y, Shen J, Wu X, Li M, Chen M, Li X, Sun Y, Gu L, Li W, Wang F, Cho CH, Zhang Y, Zheng X, Xiao Z, Du F. The dynamic role of ferroptosis in cancer immunoediting: Implications for immunotherapy. Pharmacol Res 2025; 214:107674. [PMID: 40020885 DOI: 10.1016/j.phrs.2025.107674] [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: 12/19/2024] [Revised: 02/14/2025] [Accepted: 02/23/2025] [Indexed: 03/03/2025]
Abstract
Currently, cancer immunotherapy strategies are primarily formulated based on the patient's present condition, representing a "static" treatment approach. However, cancer progression is inherently "dynamic," as the immune environment is not fixed but undergoes continuous changes. This dynamism is characterized by the ongoing interactions between tumor cells and immune cells, which ultimately lead to alterations in the tumor immune microenvironment. This process can be effectively elucidated by the concept of cancer immunoediting, which divides tumor development into three phases: "elimination," "equilibrium," and "escape." Consequently, adjusting immunotherapy regimens based on these distinct phases may enhance patient survival and improve prognosis. Targeting ferroptosis is an emerging area in cancer immunotherapy, and our findings reveal that the antioxidant systems associated with ferroptosis possess dual roles, functioning differently across the three phases of cancer immunoediting. Therefore, this review delve into the dual role of the ferroptosis antioxidant system in tumor development and progression. It also propose immunotherapy strategies targeting ferroptosis at different stages, ultimately aiming to illuminate the significant implications of targeting ferroptosis at various phases for cancer immunotherapy.
Collapse
Affiliation(s)
- Jiarui Lan
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China
| | - Dan Cai
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China
| | - Shuang Gou
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China
| | - Yulin Bai
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China
| | - Huaqing Lei
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China
| | - Yan Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China
| | - Yu Chen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China
| | - Yueshui Zhao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China
| | - Jing Shen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China
| | - Xu Wu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China
| | - Mingxing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China
| | - Meijuan Chen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China
| | - Xiaobing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China
| | - Yuhong Sun
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China
| | - Li Gu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China
| | - Wanping Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China
| | - Fang Wang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China
| | - Chi Hin Cho
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Yan Zhang
- Department of Oncology, Luzhou People's Hospital, Luzhou, Sichuan 646000, China
| | - Xin Zheng
- Department of Oncology, Luzhou People's Hospital, Luzhou, Sichuan 646000, China.
| | - Zhangang Xiao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China.
| | - Fukuan Du
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China.
| |
Collapse
|
10
|
Johnson B, Guo Q, Chaludiya K, Kim S. The Proimmunomodulatory and Anti-immunomodulatory Effects of Radiotherapy in Oncologic Care. Hematol Oncol Clin North Am 2025; 39:399-411. [PMID: 39827043 PMCID: PMC11932133 DOI: 10.1016/j.hoc.2024.11.009] [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] [Indexed: 01/22/2025]
Abstract
The abscopal effect in radiotherapy (RT) refers to the phenomenon where localized radiation treatment causes regression of distant, nonirradiated tumors. Although rare, recent research shows that combining radiation with immunotherapies, such as immune checkpoint inhibitors, can enhance this effect. The interaction between radiation-induced cell death, immune responses, and the tumor microenvironment manifests in competing biologic mechanisms resulting in complex immunologic outcomes. In order to maximize the therapeutic advantages of the immunogenic effect of RT in the future, further studies are needed to fully understand its biologic underpinnings.
Collapse
Affiliation(s)
- Bryan Johnson
- Department of Radiation Oncology, Mayo Clinic Florida, 4500 San Pablo Road S, Jacksonville, FL 32224, USA
| | - Qianyu Guo
- Department of Radiation Oncology, Mayo Clinic Florida, 4500 San Pablo Road S, Jacksonville, FL 32224, USA; Department of Internal Medicine, Mayo Clinic Florida, 4500 San Pablo Road S, Jacksonville, FL 32224, USA
| | - Keyur Chaludiya
- Department of Laboratory Medicine, Mayo Clinic Minnesota, 150 3rd Street SW, Rochester, MN 55902, USA
| | - Sungjune Kim
- Department of Radiation Oncology, Mayo Clinic Florida, 4500 San Pablo Road S, Jacksonville, FL 32224, USA.
| |
Collapse
|
11
|
Ju Y, Lv Y, Liu X, Lu J, Shi Y, Guo H, Xu S, Tian J, Yang J, Zhong J. Role of long non-coding RNAs in the regulation of ferroptosis in tumors. Front Immunol 2025; 16:1568567. [PMID: 40191204 PMCID: PMC11968707 DOI: 10.3389/fimmu.2025.1568567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Accepted: 03/06/2025] [Indexed: 04/09/2025] Open
Abstract
Normal cells begin to grow indefinitely and immortalize to form tumor cells after an external stimulus resulting in a genetic mutation. Effective killing of tumor cells is the basis of various cancer therapies. Ferroptosis is a class of cell death types dependent on iron and cellular lipid peroxidation. Tumors themselves are iron-dependent, and conventional radiotherapy also sensitizes cancer cells to ferroptosis. Increasing the sensitivity of tumor cells to ferroptosis may be a potential therapeutic strategy to overcome the resistance mechanisms of conventional cancer therapy. Long noncoding RNAs (LncRNAs) are a class of transcripts more than 200 nucleotides in length that regulate gene expression at multiple levels and are involved in biological processes such as cell differentiation, cell cycle arrest, and maintenance of tumor stemness. Recent studies have found that lncRNAs regulate ferroptosis of tumor cells through multiple mechanisms and may influence or ameliorate tumor resistance to chemotherapeutic agents. With the continuous maturation of nanomaterials technology, it may provide new means for cancer treatment by regulating the levels of ferroptosis-related lncRNAs inside tumors as well as increasing the levels of Fe2+ and ROS inside tumors. In this paper, we systematically introduce the regulatory mechanism of lncRNAs in ferroptosis, the role of ferroptosis in tumor immunotherapy and the application of lncRNAs combined with ferroptosis in nanomaterials, which provides new perspectives for tumor therapy.
Collapse
Affiliation(s)
- Ying Ju
- Department of Gynecology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Yuanhao Lv
- Department of Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Xu Liu
- Department of Anesthesia and Perioperative Medicine, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Jing Lu
- Department of Gynecology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Yashen Shi
- Department of Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Huimin Guo
- Department of Gynecology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Siguang Xu
- Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Jiaqi Tian
- Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Jun Yang
- Department of Gynecology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Jiateng Zhong
- Department of Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
- Xinxiang Engineering Technology Research Center of Digestive Tumor Molecular Diagnosis, the First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
12
|
Rithvik A, Wadhavane S, Rasool M. Decoding poly (RC)-binding protein 1 (PCBP1), the underrated guard at the foothill of ferroptosis. Pathol Res Pract 2025; 266:155771. [PMID: 39700662 DOI: 10.1016/j.prp.2024.155771] [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: 10/12/2024] [Revised: 12/10/2024] [Accepted: 12/11/2024] [Indexed: 12/21/2024]
Abstract
PCBP1 is a multifunctional adaptor protein, whose function as an iron chaperone and epigenetic regulator of several chemical messengers involved in ferroptosis has garnered much attention. Herein, this review, several attempts have been made to simplify our understanding of the complex roles of PCBP1. The review begins by elucidating the relevance of PCBP1 in key events governing ferroptosis. We expeditiously shed light on some of the important mechanisms that have critical implications for the ferroptosis landscape. For instance, senescence, EMT, hypoxia, and regulation of the cell cycle and immune checkpoints, among others, have been demonstrated to influence ferroptosis sensitivity to varying degrees. Thus, this review entails a conscious attempt to carefully examine the relevance of PCBP1 in such potential mechanisms. Furthermore, we investigated the therapeutic relevance of PCBP1 in tumor biology and autoimmunity, while underscoring the contrasting perspective of ferroptosis targeting across the disease spectrum. Finally, we debate the different strategies that can be exploited to target PCBP1 in promoting or inhibiting ferroptosis.
Collapse
Affiliation(s)
- Arulkumaran Rithvik
- Immunopathology Lab, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu 632014, India
| | - Sakshi Wadhavane
- Immunopathology Lab, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu 632014, India
| | - Mahaboobkhan Rasool
- Immunopathology Lab, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu 632014, India.
| |
Collapse
|
13
|
Musella L, Afonso Castro A, Lai X, Widmann M, Vera J. ENQUIRE automatically reconstructs, expands, and drives enrichment analysis of gene and Mesh co-occurrence networks from context-specific biomedical literature. PLoS Comput Biol 2025; 21:e1012745. [PMID: 39932993 PMCID: PMC11844901 DOI: 10.1371/journal.pcbi.1012745] [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: 03/22/2024] [Revised: 02/21/2025] [Accepted: 12/20/2024] [Indexed: 02/13/2025] Open
Abstract
The accelerating growth of scientific literature overwhelms our capacity to manually distil complex phenomena like molecular networks linked to diseases. Moreover, biases in biomedical research and database annotation limit our interpretation of facts and generation of hypotheses. ENQUIRE (Expanding Networks by Querying Unexpectedly Inter-Related Entities) offers a time- and resource-efficient alternative to manual literature curation and database mining. ENQUIRE reconstructs and expands co-occurrence networks of genes and biomedical ontologies from user-selected input corpora and network-inferred PubMed queries. Its modest resource usage and the integration of text mining, automatic querying, and network-based statistics mitigating literature biases makes ENQUIRE unique in its broad-scope applications. For example, ENQUIRE can generate co-occurrence gene networks that reflect high-confidence, functional networks. When tested on case studies spanning cancer, cell differentiation, and immunity, ENQUIRE identified interlinked genes and enriched pathways unique to each topic, thereby preserving their underlying context specificity. ENQUIRE supports biomedical researchers by easing literature annotation, boosting hypothesis formulation, and facilitating the identification of molecular targets for subsequent experimentation.
Collapse
Affiliation(s)
- Luca Musella
- Laboratory of Systems Tumor Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Deutsches Zentrum Immuntherapie, BZKF, and Uniklinikum Erlangen, Erlangen, Germany
| | - Alejandro Afonso Castro
- Laboratory of Systems Tumor Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Deutsches Zentrum Immuntherapie, BZKF, and Uniklinikum Erlangen, Erlangen, Germany
| | - Xin Lai
- Laboratory of Systems Tumor Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Deutsches Zentrum Immuntherapie, BZKF, and Uniklinikum Erlangen, Erlangen, Germany
- Faculty of Medicine and Health Technology, Systems and Network Medicine Lab, Biomedicine Unit, Tampere University, Tampere, Finland
| | - Max Widmann
- Laboratory of Systems Tumor Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Deutsches Zentrum Immuntherapie, BZKF, and Uniklinikum Erlangen, Erlangen, Germany
- University of Konstanz, Konstanz, Germany
| | - Julio Vera
- Laboratory of Systems Tumor Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Deutsches Zentrum Immuntherapie, BZKF, and Uniklinikum Erlangen, Erlangen, Germany
| |
Collapse
|
14
|
Zhang Z, Xu H, He J, Hu Q, Liu Y, Xu Z, Lou W, Wu W, Zhang L, Pu N, Shi C, Xu Y, Wang W, Liu L. Inhibition of KLF5 promotes ferroptosis via the ZEB1/HMOX1 axis to enhance sensitivity to oxaliplatin in cancer cells. Cell Death Dis 2025; 16:28. [PMID: 39827156 PMCID: PMC11743205 DOI: 10.1038/s41419-025-07330-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 12/04/2024] [Accepted: 01/07/2025] [Indexed: 01/22/2025]
Abstract
As a novel form of nonapoptotic cell death, ferroptosis is developing into a promising therapeutic target of dedifferentiating and therapy-refractory cancers. However, its application in pancreatic cancer is still unknown. In the preliminary research, we found that F-box and WD repeat domain-containing 7 (FBW7) inhibited the migration and proliferation of pancreatic cancer cells through its substrate c-Myc. We further found that another key substrate of FBW7, KLF5, could inhibit ferroptosis. Inhibiting KLF5 significantly enhances the cytotoxicity of oxaliplatin rather than other chemotherapy drugs. Mechanistically, we found that KLF5 inhibited the expression of heme oxygenase 1 (HMOX1) via repressing zinc finger E-box-binding homeobox 1 (ZEB1). Inhibition of KLF5 facilitated the cytotoxic effect of oxaliplatin via promoting ferroptosis. Oxaliplatin combined with KLF5 inhibitor significantly potentiated cell death in vitro and inhibited tumor growth in vivo compared with either treatment alone. These results reveal a critical role of KLF5 in sensitized chemotherapy of pancreatic cancer, and suggest that ferroptosis combined with platinum-based chemotherapy rather than gemcitabine-based chemotherapy is expected to bring better therapeutic effects.
Collapse
Affiliation(s)
- Zheng Zhang
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Huaxiang Xu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Junyi He
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qiangsheng Hu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yuxin Liu
- Institute of liver diseases, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Zijin Xu
- Department of General Surgery, Qingpu Branch of Zhongshan Hospital Affiliated to Fudan University, Shanghai, China
| | - Wenhui Lou
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wenchuan Wu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lei Zhang
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ning Pu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chenye Shi
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yaolin Xu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wenquan Wang
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China.
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Liang Liu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China.
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
15
|
Famurewa AC, Prabhune NM, Prabhu S. Natural product mitigation of ferroptosis in platinum-based chemotherapy toxicity: targeting the underpinning oxidative signaling pathways. J Pharm Pharmacol 2025; 77:1-17. [PMID: 39485898 DOI: 10.1093/jpp/rgae132] [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: 04/21/2024] [Accepted: 09/30/2024] [Indexed: 11/03/2024]
Abstract
OBJECTIVES Platinum-based anticancer chemotherapy (PAC) represents a cornerstone in cancer treatment, retaining its status as the gold standard therapy. However, PAC's efficacy is countered by significant toxicities, such as nephrotoxicity, ototoxicity, and neurotoxicity. Recent studies have linked these toxicities to ferroptosis, characterized by iron accumulation, reactive oxygen species generation, and lipid peroxidation. This review explores the mechanisms underlying PAC-induced toxicities, focusing on the involvement of ferroptosis with three major PAC drugs-cisplatin, carboplatin, and oxaliplatin. Further, we provide a comprehensive analysis of the natural product mitigation of PAC-induced ferroptotic toxicity. KEY FINDINGS The mechanistic role of ferroptosis in cisplatin- and oxaliplatin-induced toxicities has been investigated, while studies on carboplatin-induced ferroptotic toxicities are lacking. Natural compounds targeting molecular pathways of ferroptosis have been explored to mitigate PAC-induced ferroptotic toxicity. CONCLUSION While ferroptosis in cisplatin- and oxaliplatin-induced toxicities has been investigated, there remains a notable dearth of studies examining its involvement in carboplatin-induced toxicities. Hence, further exploration is warranted to define the role of ferroptosis in carboplatin-induced toxicities, and its further mitigation. Moreover, in-depth mechanistic evaluation is necessary to establish natural products evaluated against PAC-induced ferroptosis, as PAC adjuvants.
Collapse
Affiliation(s)
- Ademola C Famurewa
- Department of Medical Biochemistry, Faculty of Basic Medical Sciences, College of Medical Sciences, Alex Ekwueme Federal University, Ikwo 482103, Ebonyi State, Nigeria
- Centre for Natural Products Discovery, School of Pharmacy and Biomolecular Sciences, Faculty of Science, Liverpool John Moores University, Byrom Street, Liverpool L3 3AF, United Kingdom
| | - Nupura Manish Prabhune
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, India
| | - Sudharshan Prabhu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, India
| |
Collapse
|
16
|
Yi J, Zhang W, Li Y, Ren H, Xiang Y, Qiao C. Recent advances in crosstalk between immune cells and cancer cells with ferroptosis. Life Sci 2025; 360:123279. [PMID: 39608446 DOI: 10.1016/j.lfs.2024.123279] [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: 08/01/2024] [Revised: 11/12/2024] [Accepted: 11/25/2024] [Indexed: 11/30/2024]
Abstract
Ferroptosis, a regulated form of cell death distinct from apoptosis and necrosis. Key hallmarks include iron-dependent lipid peroxidation, glutathione depletion, and intracellular iron accumulation, all of which are counteracted by specific cellular defenses. However, the immunosuppressive effects of ferroptosis induction in cancer immunotherapy remain unresolved. This review summarizes the recent advancements in ferroptosis research, focusing on its defensive mechanisms. It analyzes how ferroptosis affects both cancer and immune cells, highlighting its potential inhibitory effects on anti-tumor immunity and possible promotion of pro-tumor immune responses. Finally, this review briefly introduces case studies that combined ferroptosis induction with immunotherapy, offering novel perspectives for cancer treatment.
Collapse
Affiliation(s)
- Jinfeng Yi
- Department of Pathology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Wanting Zhang
- Department of Pathology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Yingpu Li
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150000, China; NHC Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province 150001, China
| | - He Ren
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150000, China
| | - Yuhang Xiang
- Department of Pathology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Cong Qiao
- Department of Pathology, Harbin Medical University, Harbin, Heilongjiang, China.
| |
Collapse
|
17
|
Yuan Z, Han X, Xiao M, Zhu T, Xu Y, Tang Q, Lian C, Wang Z, Li J, Wang B, Li C, Xiang X, Jin R, Liu Y, Yu X, Zhang K, Li S, Ray M, Li R, Gruzdev A, Shao S, Shao F, Wang H, Lian W, Tang Y, Chen D, Lei Y, Jin X, Li Q, Long W, Huang H, DeMayo FJ, Liu J. Overexpression of ELF3 in the PTEN-deficient lung epithelium promotes lung cancer development by inhibiting ferroptosis. Cell Death Dis 2024; 15:897. [PMID: 39695109 PMCID: PMC11655876 DOI: 10.1038/s41419-024-07274-5] [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: 06/19/2024] [Revised: 11/21/2024] [Accepted: 11/29/2024] [Indexed: 12/20/2024]
Abstract
Ferroptosis has been shown to play a crucial role in preventing cancer development, but the underlying mechanisms of dysregulated genes and genetic alternations driving cancer development by regulating ferroptosis remain unclear. Here, we showed that the synergistic role of ELF3 overexpression and PTEN deficiency in driving lung cancer development was highly dependent on the regulation of ferroptosis. Human ELF3 (hELF3) overexpression in murine lung epithelial cells only caused hyperplasia with increased proliferation and ferroptosis. hELF3 overexpression and Pten genetic disruption significantly induced lung tumor development with increased proliferation and inhibited ferroptosis. Mechanistically, we found it was due to the induction of SCL7A11, a typical ferroptosis inhibitor, and ELF3 directly and positively regulated SCL7A11 in the PTEN-deficient background. Erastin-mediated inhibition of SCL7A11 induced ferroptosis in cells with ELF3 overexpression and PTEN deficiency and thus inhibited cell colony formation and tumor development. Clinically, human lung tumors showed a negative correlation between ELF3 and PTEN expression and a positive correlation between ELF3 and SCL7A11 in a subset of human lung tumors with PTEN-low expression. ELF3 and SCL7A11 expression levels were negatively associated with lung cancer patients' survival rates. In summary, ferroptosis induction can effectively attenuate lung tumor development induced by ELF3 overexpression and PTEN downregulation or loss-of-function mutations.
Collapse
Affiliation(s)
- Zengzhuang Yuan
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital, and Centre for Infection Immunity and Cancer (IIC) of Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Xinyan Han
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital, and Centre for Infection Immunity and Cancer (IIC) of Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Manyu Xiao
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital, and Centre for Infection Immunity and Cancer (IIC) of Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Taoyu Zhu
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital, and Centre for Infection Immunity and Cancer (IIC) of Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Edinburgh Medical School: Biomedical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
| | - Yaping Xu
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital, and Centre for Infection Immunity and Cancer (IIC) of Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Edinburgh Medical School: Biomedical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
| | - Qian Tang
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital, and Centre for Infection Immunity and Cancer (IIC) of Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Edinburgh Medical School: Biomedical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
| | - Chen Lian
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital, and Centre for Infection Immunity and Cancer (IIC) of Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Zijin Wang
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital, and Centre for Infection Immunity and Cancer (IIC) of Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Edinburgh Medical School: Biomedical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
| | - Junming Li
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital, and Centre for Infection Immunity and Cancer (IIC) of Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Edinburgh Medical School: Biomedical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
| | - Boyu Wang
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital, and Centre for Infection Immunity and Cancer (IIC) of Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Changhui Li
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital, and Centre for Infection Immunity and Cancer (IIC) of Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Edinburgh Medical School: Biomedical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
| | - Xiaochen Xiang
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital, and Centre for Infection Immunity and Cancer (IIC) of Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Edinburgh Medical School: Biomedical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
| | - Ruobai Jin
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital, and Centre for Infection Immunity and Cancer (IIC) of Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Edinburgh Medical School: Biomedical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
| | - Yufei Liu
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital, and Centre for Infection Immunity and Cancer (IIC) of Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Edinburgh Medical School: Biomedical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
| | - Xinyu Yu
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital, and Centre for Infection Immunity and Cancer (IIC) of Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Edinburgh Medical School: Biomedical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
| | - Kehang Zhang
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital, and Centre for Infection Immunity and Cancer (IIC) of Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Edinburgh Medical School: Biomedical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
| | - Songsong Li
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital, and Centre for Infection Immunity and Cancer (IIC) of Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Madhumita Ray
- Reproductive & Developmental Biology Laboratory, National Institute of Environmental Health Sciences (NIEHS), Research Triangle Park, NC, USA
| | - Rong Li
- Department of Obstetrics, Gynecology and Women' Health, University of Missouri, Columbia, MO, USA
| | - Artiom Gruzdev
- Gene Editing and Mouse Model Core, National Institute of Environmental Health Sciences (NIEHS), Research Triangle Park, NC, USA
| | - Shiqun Shao
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China
| | - Fangwei Shao
- Zhejiang University-University of Illinois Urbana-Champaign Institute, Zhejiang University, Haining, China
- Biomedical and Heath Translational Research Center of Zhejiang Province, Haining, Zhejiang, China
- National Key Laboratory of Biobased Transportation Fuel Technology, ZJU-UIUC Institute, Zhejiang University, Hangzhou, China
| | - Hua Wang
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Wang Lian
- Department of Thoracic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Yong Tang
- Department of Thoracic Surgery, Shenzhen Nanshan People's Hospital, Shenzhen, China
| | - Di Chen
- Center for Regeneration and Cell Therapy of Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Ying Lei
- Department of Respiratory and Critical Care Medicine, The Quzhou Affiliated Hospital of Wenzhou Medical Hospital, Quzhou People's Hospital, Wenzhou, China
| | - Xuru Jin
- Department of Respiratory and Critical Care Medicine, The Quzhou Affiliated Hospital of Wenzhou Medical Hospital, Quzhou People's Hospital, Wenzhou, China
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qinglin Li
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Weiwen Long
- Department of Biochemistry and Molecular Biology, Boonshoft School of Medicine, Wright State University, Dayton, OH, USA
| | - Huaqiong Huang
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Francesco J DeMayo
- Reproductive & Developmental Biology Laboratory, National Institute of Environmental Health Sciences (NIEHS), Research Triangle Park, NC, USA
| | - Jian Liu
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital, and Centre for Infection Immunity and Cancer (IIC) of Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China.
- Edinburgh Medical School: Biomedical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK.
- Biomedical and Heath Translational Research Center of Zhejiang Province, Haining, Zhejiang, China.
- Zhejiang Key Laboratory of Medical Imaging Artificial Intelligence, Haining, Zhejiang, China.
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
18
|
Ning L, Chen D, Han J, Xie G, Sun J. Global research trends and frontiers in ferroptosis in hepatocellular carcinoma: a bibliometric and visualization study. Front Oncol 2024; 14:1474496. [PMID: 39723378 PMCID: PMC11668663 DOI: 10.3389/fonc.2024.1474496] [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: 08/01/2024] [Accepted: 11/26/2024] [Indexed: 12/28/2024] Open
Abstract
Background Since the emergence of the hot topic of "ferroptosis," numerous studies have explored its role in hepatocellular carcinoma (HCC), revealing its significance in the disease's pathogenesis, progression, and treatment. However, there remains a significant gap in the quantitative analysis of ferroptosis in HCC. Therefore, this study aims to comprehensively assess the research progress and evolution in this field through bibliometric and citation analysis. Method On June 27, 2024, the author conducted a literature search, extracting relevant publications from the Web of Science Core Collection (WOSCC) Science Citation Index Expanded (SCIE) spanning from January 2010 to December 2023. Subsequently, the compiled documents were subjected to bibliometric evaluation and analysis using visualization tools such as R package "bibliometrix", CiteSpace and VOSviewer. Result The search yielded 576 papers by 3,925 authors, encompassing contributions from 34 countries and 685 institutions, published across 250 journals, including 25,889 co-cited references from 2,600 journals. Notably, China leads with a significant publication count of 481 articles (accounting for 83.5%) and demonstrates the strongest collaboration with the United States. The multifaceted role of ferroptosis in hepatocellular carcinoma (HCC) has garnered considerable attention. In recent years, research into disease prognosis, the tumor microenvironment, and targeted therapies involving immunology has become key themes and emerging frontiers in this field. Conclusion This study meticulously compiled and analyzed the current discourse and emerging perspectives on ferroptosis in HCC. Identifying research trends and hotspots offers valuable guidance for future investigations and provides a basis for the development of novel therapeutic strategies to improve HCC prognosis and treatment outcomes.
Collapse
Affiliation(s)
- Lin Ning
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Di Chen
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Department of Hepatobiliary Medicine, Jinan, China
| | - Jie Han
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Department of Hepatobiliary Medicine, Jinan, China
| | - Guanyue Xie
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Department of Hepatobiliary Medicine, Jinan, China
| | - Jianguang Sun
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
19
|
Shi J, Cui G, Jin Y, Mi B, Liu K, Zhao L, Bao K, Lu Z, Liu J, Wang Y, He H, Guo Z. Glutathione-Depleted Photodynamic Nanoadjuvant for Triggering Nonferrous Ferroptosis to Amplify Radiotherapy of Breast Cancer. Adv Healthc Mater 2024; 13:e2402474. [PMID: 39397336 DOI: 10.1002/adhm.202402474] [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: 07/06/2024] [Revised: 09/10/2024] [Indexed: 10/15/2024]
Abstract
Radiotherapy plays a crucial role in the treatment of advanced breast cancer, but the increased antioxidant system, especially the rise in glutathione (GSH), presents a significant obstacle to its effectiveness. To address this challenge, a versatile GSH-depleted photodynamic nanoadjuvant is developed to augment the efficacy of radiotherapy for breast cancer treatment. This nanoadjuvant operates within the tumor microenvironment to effectively deplete intracellular GSH through a sequence of cascaded processes, including GSH exhaustion, biosynthetic inhibition, and photodynamic oxidation. This leads to a notable accumulation of lipid peroxides (LPO) and subsequent suppression of glutathione peroxidase 4 (GPX4) activity. Consequently, the combined GSH depletion induced by the nanoadjuvant markedly promotes nonferrous ferroptosis, thereby contributing to the augmentation of antitumor efficiency during radiotherapy in breast cancer. This work presents an innovative approach to designing and synthesizing biocompatible nanoadjuvants with the goal of improving the efficacy of radiotherapy for breast cancer in prospective clinical scenarios.
Collapse
Affiliation(s)
- Jiangnan Shi
- State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, and School of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, China
| | - Guoqing Cui
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Yaqi Jin
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Boyu Mi
- State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, and School of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, China
| | - Kenan Liu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Linqian Zhao
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Kewang Bao
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Ziyao Lu
- State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, and School of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, China
| | - Jie Liu
- School of Chemistry and Life Sciences, Suzhou University of Science and Technology, Suzhou, 215000, China
| | - Yuwei Wang
- State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, and School of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, China
| | - Hui He
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Zhengqing Guo
- State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, and School of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, China
| |
Collapse
|
20
|
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
|
21
|
De Leon-Oliva D, Boaru DL, Minaya-Bravo AM, De Castro-Martinez P, Fraile-Martinez O, Garcia-Montero C, Cobo-Prieto D, Barrena-Blázquez S, Lopez-Gonzalez L, Albillos A, Alvarez-Mon M, Saez MA, Diaz-Pedrero R, Ortega MA. Improving understanding of ferroptosis: Molecular mechanisms, connection with cellular senescence and implications for aging. Heliyon 2024; 10:e39684. [PMID: 39553553 PMCID: PMC11564042 DOI: 10.1016/j.heliyon.2024.e39684] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 10/20/2024] [Accepted: 10/21/2024] [Indexed: 11/19/2024] Open
Abstract
In the face of cell damage, cells can initiate a response ranging from survival to death, the balance being crucial for tissue homeostasis and overall health. Cell death, in both accidental and regulated forms, plays a fundamental role in maintaining tissue homeostasis. Among the regulated mechanisms of cell death, ferroptosis has garnered attention for its iron-dependent phospholipid (PL) peroxidation and its implications in aging and age-related disorders, as well as for its therapeutic relevance. In this review, we provide an overview of the mechanisms, regulation, and physiological and pathological roles of ferroptosis. We present new insights into the relationship between ferroptosis, cellular senescence and aging, emphasizing how alterations in ferroptosis pathways contribute to aging-related tissue dysfunction. In addition, we examine the therapeutic potential of ferroptosis in aging-related diseases, offering innovative insights into future interventions aimed at mitigating the effects of aging and promoting longevity.
Collapse
Affiliation(s)
- Diego De Leon-Oliva
- Department of Medicine and Medical Specialities, (CIBERehd), Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
| | - Diego Liviu Boaru
- Department of Medicine and Medical Specialities, (CIBERehd), Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
| | - Ana M. Minaya-Bravo
- Department of Medicine and Medical Specialities, (CIBERehd), Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
| | - Patricia De Castro-Martinez
- Department of Medicine and Medical Specialities, (CIBERehd), Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
| | - Oscar Fraile-Martinez
- Department of Medicine and Medical Specialities, (CIBERehd), Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
| | - Cielo Garcia-Montero
- Department of Medicine and Medical Specialities, (CIBERehd), Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
| | - David Cobo-Prieto
- Department of Medicine and Medical Specialities, (CIBERehd), Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
- Immune System Diseases-Rheumatology Service, Central University Hospital of Defence-UAH Madrid, 28801, Alcala de Henares, Spain
| | - Silvestra Barrena-Blázquez
- Department of Medicine and Medical Specialities, (CIBERehd), Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
- Department of General and Digestive Surgery, General and Digestive Surgery, Príncipe de Asturias Universitary Hospital, Alcala de Henares, Spain
| | - Laura Lopez-Gonzalez
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
| | - Agustín Albillos
- Department of Medicine and Medical Specialities, (CIBERehd), Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
- Gastroenterology and Hepatology Service, Ramón y Cajal University Hospital, University of Alcalá, IRYCIS, Network Biomedical Research Center for Liver and Digestive Diseases (CIBERehd), Carlos III Health Institute, Madrid, Spain
| | - Melchor Alvarez-Mon
- Department of Medicine and Medical Specialities, (CIBERehd), Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
- Immune System Diseases-Rheumatology, Oncology Service an Internal Medicine (CIBEREHD), University Hospital Príncipe de Asturias, 28806, Alcala de Henares, Spain
| | - Miguel A. Saez
- Department of Medicine and Medical Specialities, (CIBERehd), Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
- Pathological Anatomy Service, Central University Hospital of Defence-UAH Madrid, 28801, Alcala de Henares, Spain
| | - Raul Diaz-Pedrero
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
- Department of General and Digestive Surgery, General and Digestive Surgery, Príncipe de Asturias Universitary Hospital, Alcala de Henares, Spain
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
| | - Miguel A. Ortega
- Department of Medicine and Medical Specialities, (CIBERehd), Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
| |
Collapse
|
22
|
Guan J, Min S, Xia Y, Guo Z, Zhou X. Identifying colorectal cancer subtypes and establishing a prognostic model using metabolic plasticity and ferroptosis genes. Sci Rep 2024; 14:27277. [PMID: 39516556 PMCID: PMC11549462 DOI: 10.1038/s41598-024-78505-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
Metabolic plasticity and ferroptosis are essential for colorectal cancer (CRC) progression. The effects and prognostic value of metabolic plasticity- and ferroptosis-related genes (MPFRGs) in CRC remain unclear. We established a prognostic model for CRC patients by identifying important genes in metabolic plasticity and ferroptosis. Data of CRC patients were retrieved from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus; MPFRG data were obtained from GeneCards and FerrDb. We performed functional (to explore differences between the two metabolic subtypes) and single-sample gene set (to assess the immune environment) enrichment analyses. Immunophenotype, tumor immunological dysfunction, and exclusion scores were assessed to determine patient immune responses. A least absolute shrinkage and selection operator-Cox regression model comprising 10 significant differentially expressed genes of metabolic plasticity and ferroptosis (MPFDEGs) was constructed using TCGA training cohort and validated using the GSE17536 and GSE39582 datasets. We established a nomogram comprising metabolic plasticity- and ferroptosis-based signatures, revealing the clinical application and potential molecular mechanisms underlying the role of MPFRGs in CRC. Our model (developed based on 10 MPFDEGs) is efficient for calculating the overall survival of CRC patients. Our findings provide new strategies for the clinical management and individualized treatment of these patients.
Collapse
Affiliation(s)
- Jingwen Guan
- Department of Pathology, Suzhou Hospital of Anhui Medical University, Anhui, China.
| | - Simin Min
- Department of Science and Education Section, Suzhou Hospital of Anhui Medical University, Anhui, China
| | - Yan Xia
- Department of Pathology, Suzhou Hospital of Anhui Medical University, Anhui, China
| | - Zhiguo Guo
- Department of Gastroenterology, Suzhou Hospital of Anhui Medical University, Anhui, China
| | - Xiaolan Zhou
- Department of Gastroenterology, Suzhou Hospital of Anhui Medical University, Anhui, China
| |
Collapse
|
23
|
Liang Y, Qiu S, Zou Y, Leung ELH, Luo L. Ferroptosis-Modulating Natural Products for Targeting Inflammation-Related Diseases: Challenges and Opportunities in Manipulating Redox Signaling. Antioxid Redox Signal 2024; 41:976-991. [PMID: 39001833 DOI: 10.1089/ars.2024.0556] [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] [Indexed: 07/15/2024]
Abstract
Significance: Numerous disorders are linked to ferroptosis, a form of programmed cell death triggered by lipid peroxidation accumulation rather than apoptosis. Inflammation is the body's defensive response to stimuli and is also caused by inflammatory chemicals that can harm the body. The treatment of inflammatory diseases by focusing on the signaling pathways and mechanisms of ferroptosis has emerged as a new area worthy of extensive research. Recent Advances: Studies in cellular and animal models of inflammatory diseases have shown that ferroptosis markers are activated and lipid peroxidation levels are increased. Natural products (NPs) are gaining importance due to their ability to target ferroptosis pathways, particularly the Nuclear factor E2-related factor 2 signaling pathway, thereby suppressing inflammation and the release of pro-inflammatory cytokines. Critical Issues: This article provides an overview of ferroptosis, focusing on the signaling pathways and mechanisms connecting it to inflammation. It also explores the potential use of NPs as a treatment for inflammatory diseases and ferroptosis. Future Directions: NPs offer unique advantages, including multicomponent properties, multi-bio-targeting capabilities, and minimal side effects. Further research may facilitate the early clinical application of NPs to develop innovative treatment strategies. Antioxid. Redox Signal. 41, 976-991.
Collapse
Affiliation(s)
- Yongyi Liang
- School of Ocean and Tropical Medicine, The Marine Biomedical Research Institute of Guangdong Zhanjiang, Guangdong Medical University, Zhanjiang, China
| | - Shaojun Qiu
- School of Ocean and Tropical Medicine, The Marine Biomedical Research Institute of Guangdong Zhanjiang, Guangdong Medical University, Zhanjiang, China
| | - Youwen Zou
- School of Ocean and Tropical Medicine, The Marine Biomedical Research Institute of Guangdong Zhanjiang, Guangdong Medical University, Zhanjiang, China
| | - Elaine Lai-Han Leung
- Faculty of Health Sciences, Cancer Centre, University of Macau, Zhuhai, Macao Special Administrative Region of China
- MOE Frontiers Science Centre for Precision Oncology, University of Macau, Zhuhai, Macao Special Administrative Region of China
- State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Zhuhai, Macao Special Administrative Region of China
| | - Lianxiang Luo
- School of Ocean and Tropical Medicine, The Marine Biomedical Research Institute of Guangdong Zhanjiang, Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
24
|
Piamsiri C, Fefelova N, Pamarthi SH, Gwathmey JK, Chattipakorn SC, Chattipakorn N, Xie LH. Potential Roles of IP 3 Receptors and Calcium in Programmed Cell Death and Implications in Cardiovascular Diseases. Biomolecules 2024; 14:1334. [PMID: 39456267 PMCID: PMC11506173 DOI: 10.3390/biom14101334] [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: 09/23/2024] [Revised: 10/17/2024] [Accepted: 10/18/2024] [Indexed: 10/28/2024] Open
Abstract
Inositol 1,4,5-trisphosphate receptors (IP3Rs) play a crucial role in maintaining intracellular/cytosolic calcium ion (Ca2+i) homeostasis. The release of Ca2+ from IP3Rs serves as a second messenger and a modulatory factor influencing various intracellular and interorganelle communications during both physiological and pathological processes. Accumulating evidence from in vitro, in vivo, and clinical studies supports the notion that the overactivation of IP3Rs is linked to the pathogenesis of various cardiac conditions. The overactivation of IP3Rs results in the dysregulation of Ca2+ concentration ([Ca2+]) within cytosolic, mitochondrial, and nucleoplasmic cellular compartments. In cardiovascular pathologies, two isoforms of IP3Rs, i.e., IP3R1 and IP3R2, have been identified. Notably, IP3R1 plays a pivotal role in cardiac ischemia and diabetes-induced arrhythmias, while IP3R2 is implicated in sepsis-induced cardiomyopathy and cardiac hypertrophy. Furthermore, IP3Rs have been reported to be involved in various programmed cell death (PCD) pathways, such as apoptosis, pyroptosis, and ferroptosis underscoring their multifaceted roles in cardiac pathophysiology. Based on these findings, it is evident that exploring potential therapeutic avenues becomes crucial. Both genetic ablation and pharmacological intervention using IP3R antagonists have emerged as promising strategies against IP3R-related pathologies suggesting their potential therapeutic potency. This review summarizes the roles of IP3Rs in cardiac physiology and pathology and establishes a foundational understanding with a particular focus on their involvement in the various PCD pathways within the context of cardiovascular diseases.
Collapse
Affiliation(s)
- Chanon Piamsiri
- Department of Cell Biology and Molecular Medicine, Rutgers University-New Jersey Medical School, Newark, NJ 07103, USA; (C.P.); (N.F.)
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nadezhda Fefelova
- Department of Cell Biology and Molecular Medicine, Rutgers University-New Jersey Medical School, Newark, NJ 07103, USA; (C.P.); (N.F.)
| | - Sri Harika Pamarthi
- Department of Cell Biology and Molecular Medicine, Rutgers University-New Jersey Medical School, Newark, NJ 07103, USA; (C.P.); (N.F.)
| | - Judith K. Gwathmey
- Department of Cell Biology and Molecular Medicine, Rutgers University-New Jersey Medical School, Newark, NJ 07103, USA; (C.P.); (N.F.)
| | - Siriporn C. Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Lai-Hua Xie
- Department of Cell Biology and Molecular Medicine, Rutgers University-New Jersey Medical School, Newark, NJ 07103, USA; (C.P.); (N.F.)
| |
Collapse
|
25
|
Ouyang S, Zeng Z, He J, Luo L. Epigenetic regulation of targeted ferroptosis: A new strategy for drug development. J Pharm Anal 2024; 14:101012. [PMID: 39850234 PMCID: PMC11755343 DOI: 10.1016/j.jpha.2024.101012] [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: 01/28/2024] [Revised: 05/20/2024] [Accepted: 05/23/2024] [Indexed: 01/25/2025] Open
Abstract
Ferroptosis is a newly discovered form of cell death that is influenced by iron levels and is triggered by cellular metabolism and excessive lipid peroxidation. Epigenetic regulation plays a crucial role in the development and progression of diseases, making it essential to understand these mechanisms in order to identify potential targets for drug development and clinical treatment. The intersection of ferroptosis and epigenetics has opened up new avenues for research in drug development, offering innovative strategies for combating diseases. Recent studies have shown that epigenetic modifications can impact pathways related to ferroptosis, potentially leading to organ dysfunction. Despite the increasing focus on this relationship, the role of epigenetic regulation in drug development remains largely unexplored. This article explores current research on the interplay between epigenetic regulation and ferroptosis, delving into their regulatory mechanisms and discussing the effects of existing epigenetic modification regulators on diseases. Additionally, we highlight ongoing research on epigenetic factors involved in targeting ferroptosis in cancer, providing new insights for the development of cancer treatments.
Collapse
Affiliation(s)
- Shengli Ouyang
- The First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong, 524023, China
| | - Zeyao Zeng
- The First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong, 524023, China
| | - Jieyi He
- The First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong, 524023, 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
|
26
|
Yeon Kim S, Tang M, Lu T, Chih SY, Li W. Ferroptosis in glioma therapy: advancements in sensitizing strategies and the complex tumor-promoting roles. Brain Res 2024; 1840:149045. [PMID: 38821335 PMCID: PMC11323215 DOI: 10.1016/j.brainres.2024.149045] [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: 02/27/2024] [Revised: 05/03/2024] [Accepted: 05/28/2024] [Indexed: 06/02/2024]
Abstract
Ferroptosis, an iron-dependent form of non-apoptotic regulated cell death, is induced by the accumulation of lipid peroxides on cellular membranes. Over the past decade, ferroptosis has emerged as a crucial process implicated in various physiological and pathological systems. Positioned as an alternative modality of cell death, ferroptosis holds promise for eliminating cancer cells that have developed resistance to apoptosis induced by conventional therapeutics. This has led to a growing interest in leveraging ferroptosis for cancer therapy across diverse malignancies. Gliomas are tumors arising from glial or precursor cells, with glioblastoma (GBM) being the most common malignant primary brain tumor that is associated with a dismal prognosis. This review provides a summary of recent advancements in the exploration of ferroptosis-sensitizing methods, with a specific focus on their potential application in enhancing the treatment of gliomas. In addition to summarizing the therapeutic potential, this review also discusses the intricate interplay of ferroptosis and its potential tumor-promoting roles within gliomas. Recognizing these dual roles is essential, as they could potentially complicate the therapeutic benefits of ferroptosis. Exploring strategies aimed at circumventing these tumor-promoting roles could enhance the overall therapeutic efficacy of ferroptosis in the context of glioma treatment.
Collapse
Affiliation(s)
- Soo Yeon Kim
- Division of Hematology and Oncology, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA
| | - Miaolu Tang
- Division of Hematology and Oncology, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA
| | - Tong Lu
- Division of Hematology and Oncology, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA
| | - Stephen Y Chih
- Division of Hematology and Oncology, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA; Medical Scientist Training Program, Penn State College of Medicine, Hershey, PA, USA
| | - Wei Li
- Division of Hematology and Oncology, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA; Penn State Cancer Institute, Penn State College of Medicine, Hershey, PA, USA; Department of Biochemistry and Molecular Biology, Penn State College of Medicine, Hershey, PA, USA.
| |
Collapse
|
27
|
Biscop E, Baroen J, De Backer J, Vanden Berghe W, Smits E, Bogaerts A, Lin A. Characterization of regulated cancer cell death pathways induced by the different modalities of non-thermal plasma treatment. Cell Death Discov 2024; 10:416. [PMID: 39349444 PMCID: PMC11442809 DOI: 10.1038/s41420-024-02178-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 09/02/2024] [Accepted: 09/06/2024] [Indexed: 10/02/2024] Open
Abstract
Non-thermal plasma (NTP) has shown promising anti-cancer effects, but there is still limited knowledge about the underlying cell death mechanisms induced by NTP and inherent differences between NTP treatment modalities. This study aimed to investigate four major regulated cell death (RCD) pathways, namely apoptosis, pyroptosis, necroptosis, and ferroptosis, in melanoma cancer cells following NTP treatment, and to provide an overview of molecular mechanistic differences between direct and indirect NTP treatment modalities. To discriminate which cell death pathways were triggered after treatment, specific inhibitors of apoptosis, pyroptosis, necroptosis, and ferroptosis were evaluated. RCD-specific molecular pathways were further investigated to validate the findings with inhibitors. Both direct and indirect NTP treatment increased caspase 3/7 and annexin V expression, indicative of apoptosis, as well as lipid peroxidation, characteristic of ferroptosis. Pyroptosis, on the other hand, was only induced by direct NTP treatment, evidenced by increased caspase 1 activity, whereas necroptosis was stimulated in a cell line-dependent manner. These findings highlight the molecular differences and implications of direct and indirect NTP treatment for cancer therapy. Altogether, activation of multiple cell death pathways offers advantages in minimizing treatment resistance and enhancing therapeutic efficacy, particularly in a combination setting. Understanding the mechanisms underlying NTP-induced RCD will enable the development of strategic combination therapies targeting multiple pathways to achieve cancer lethality.
Collapse
Affiliation(s)
- Eline Biscop
- PLASMANT, Department of Chemistry, University of Antwerp, Antwerp, Belgium.
- Center for Oncological Research - Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Antwerp, Belgium.
| | - Jana Baroen
- PLASMANT, Department of Chemistry, University of Antwerp, Antwerp, Belgium
- Center for Oncological Research - Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Antwerp, Belgium
| | - Joey De Backer
- Cell Death Signaling Lab, University of Antwerp, Antwerp, Belgium
| | | | - Evelien Smits
- Center for Oncological Research - Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Antwerp, Belgium
| | - Annemie Bogaerts
- PLASMANT, Department of Chemistry, University of Antwerp, Antwerp, Belgium
| | - Abraham Lin
- PLASMANT, Department of Chemistry, University of Antwerp, Antwerp, Belgium.
- Center for Oncological Research - Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
28
|
Gawargi FI, Mishra PK. MMP9 drives ferroptosis by regulating GPX4 and iron signaling. iScience 2024; 27:110622. [PMID: 39252956 PMCID: PMC11382059 DOI: 10.1016/j.isci.2024.110622] [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: 12/12/2023] [Revised: 02/08/2024] [Accepted: 07/26/2024] [Indexed: 09/11/2024] Open
Abstract
Ferroptosis, defined by the suppression of glutathione peroxidase-4 (GPX4) and iron overload, is a distinctive form of regulated cell death. Our in-depth research identifies matrix metalloproteinase-9 (MMP9) as a critical modulator of ferroptosis through its influence on GPX4 and iron homeostasis. Employing an innovative MMP9 construct without collagenase activity, we reveal that active MMP9 interacts with GPX4 and glutathione reductase, reducing GPX4 expression and activity. Furthermore, MMP9 suppresses key transcription factors (SP1, CREB1, NRF2, FOXO3, and ATF4), alongside GPX1 and ferroptosis suppressor protein-1 (FSP1), thereby disrupting the cellular redox balance. MMP9 regulates iron metabolism by modulating iron import, storage, and export via a network of protein interactions. LC-MS/MS has identified 83 proteins that interact with MMP9 at subcellular levels, implicating them in ferroptosis regulation. Integrated pathway analysis (IPA) highlights MMP9's extensive influence on ferroptosis pathways, underscoring its potential as a therapeutic target in conditions with altered redox homeostasis and iron metabolism.
Collapse
Affiliation(s)
- Flobater I Gawargi
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Paras K Mishra
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
29
|
Zhang J, Zhang F, Zhang L, Zhang M, Liu S, Ma Y. Screening and molecular docking verification of feature genes related to phospholipid metabolism in hepatocarcinoma caused by hepatitis B. Lipids Health Dis 2024; 23:268. [PMID: 39182089 PMCID: PMC11344459 DOI: 10.1186/s12944-024-02253-3] [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: 06/25/2024] [Accepted: 08/13/2024] [Indexed: 08/27/2024] Open
Abstract
BACKGROUND The progression of tumours is related to abnormal phospholipid metabolism. This study is anticipated to present a fresh perspective for disease therapy targets of hepatocarcinoma caused by hepatitis B virus in the future by screening feature genes related to phospholipid metabolism. METHODS This study analysed GSE121248 to pinpoint differentially expressed genes (DEGs). By examining the overlap between the metabolism-related genes and DEGs, the research focused on the genes involved in phospholipid metabolism. To find feature genes, functional enrichment studies were carried out and a network diagram was proposed. These findings were validated via data base of The Cancer Genome Atlas (TCGA). Further analyses included immune infiltration studies and metabolomics. Finally, the relationships between differentially abundant metabolites and feature genes were confirmed by molecular docking, providing a thorough comprehension of the molecular mechanisms. RESULTS The seven genes with the highest degree of connection (PTGS2, IGF1, SPP1, BCHE, NR1I2, NAMPT, and FABP1) were identified as feature genes. In the TCGA database, the seven feature genes also had certain diagnostic efficiency. Immune infiltration analysis revealed that feature genes regulate the infiltration of various immune cells. Metabolomics successfully identified the different metabolites of the phospholipid metabolism pathway between patients and normal individuals. The docking study indicated that different metabolites may play essential roles in causing disease by targeting feature genes. CONCLUSIONS In this study, for the first time, it reveals the possible involvement of genes linked to phospholipid metabolism-related genes using bioinformatics analysis. Identifying genes and probable therapeutic targets could provide clues for the further treatment of disease.
Collapse
Affiliation(s)
- Jian Zhang
- Department of Clinical Laboratory, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Fengmei Zhang
- Department of Clinical Laboratory, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Artificial Cell Engineering Technology Research Center, The Third Central Hospital of Tianjin, Tianjin Institute of Hepatobiliary Disease, Tianjin, 300170, China
| | - Lei Zhang
- Department of Clinical Laboratory, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Artificial Cell Engineering Technology Research Center, The Third Central Hospital of Tianjin, Tianjin Institute of Hepatobiliary Disease, Tianjin, 300170, China.
| | - Meiling Zhang
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Shuye Liu
- Department of Clinical Laboratory, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Artificial Cell Engineering Technology Research Center, The Third Central Hospital of Tianjin, Tianjin Institute of Hepatobiliary Disease, Tianjin, 300170, China.
| | - Ying Ma
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China.
| |
Collapse
|
30
|
Su X, Ding X, Ding C, Wang G, Fu C, Liu F, Shi J, He W. The role of JMJD2A in immune evasion and malignant behavior of esophageal squamous cell carcinoma. Int Immunopharmacol 2024; 137:112401. [PMID: 38878485 DOI: 10.1016/j.intimp.2024.112401] [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: 11/26/2023] [Revised: 06/01/2024] [Accepted: 06/02/2024] [Indexed: 07/11/2024]
Abstract
OBJECTIVE This study aimed to investigate the role of JMJD2A in radiotherapy tolerance of esophageal squamous cell carcinoma (ESCC). METHODS The levels of H3K9me3 modification were analyzed in anti-PD-1 therapy non-responder or responder patients, and the expression differences of H3K9me3-related modifying enzymes were assessed in TCGA-ESCC and ICGC cohorts. Subsequently, JMJD2A was knocked down in ESCC cells using CRISPR-Cas9 or lentivirus-mediated shRNA, and changes in malignant behavior of ESCC cells were observed. RNA-seq, ATAC-seq, and ChIP-seq analyses were then conducted to investigate the genes and downstream signaling pathways regulated by JMJD2A, and functional validation experiments were performed to analyze the role of downstream regulated genes and pathways in ESCC malignant behavior and immune evasion. RESULTS JMJD2A was significantly overexpressed in ESCC and anti-PD-1 therapy non-responders. Knockdown or deletion of JMJD2A significantly promoted the malignant behavior and immune evasion of ESCC. JMJD2A facilitated the structural changes in chromatin and promoted the binding of SMARCA4 to super-enhancers, thereby inducing the expression of GPX4. This resulted in the inhibition of radiation-induced DNA damage and cell ferroptosis, ultimately promoting the malignant behavior and immune evasion of ESCC cells. CONCLUSION JMJD2A plays an indispensable role in the malignant behavior and immune evasion of ESCC. It regulates the binding of SMARCA4 to super-enhancers and affects the chromatin's epigenetic landscape, thereby promoting the expression of GPX4 and attenuating iron-mediated cell death caused by radiotherapy. Consequently, it triggers the malignant behavior and immune evasion of ESCC cells.
Collapse
Affiliation(s)
- Xiangyu Su
- Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China; School of Medicine, Southeast University, Nanjing 210009, China
| | - Xu Ding
- Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China; School of Medicine, Southeast University, Nanjing 210009, China
| | - Chenxi Ding
- Department of Internal Medicine of Traditional Chinese Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Guoqing Wang
- Department of Pathology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Chenchun Fu
- Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Fei Liu
- Department of Medical Oncology, Luhe People's Hospital of Nanjing, Nanjing 210000, China
| | - Jinjun Shi
- Department of Ultrasound, Zhongda Hospital, Medical School, Southeast University, Nanjing 210009, China.
| | - Wei He
- Department of Thoracic surgery, Zhongda Hospital, Medical School, Southeast University, Nanjing 210009, China.
| |
Collapse
|
31
|
Wang Z, Huang J, MinYang, Fu L, Liu S, Huang J, Han J, Zhao X. Identification of the ferroptosis-related prognostic gene signature in mesothelioma. Gene 2024; 919:148498. [PMID: 38670397 DOI: 10.1016/j.gene.2024.148498] [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: 12/13/2023] [Revised: 04/19/2024] [Accepted: 04/22/2024] [Indexed: 04/28/2024]
Abstract
Mesothelioma, an uncommon yet highly aggressive malignant neoplasm, presents challenges in the effectiveness of current therapeutic approaches. Ferroptosis, a non-apoptotic mechanism of cellular demise, exhibits a substantial association with the progression of diverse cancer forms. It is important to acknowledge that there exists a significant association between ferroptosis and the advancement of various forms of cancer. Nevertheless, the precise role of ferroptosis regulatory factors within the context of mesothelioma remains enigmatic. In our investigation, we initially scrutinized the prognostic significance of 24 ferroptosis regulatory factors in the realm of mesothelioma. Our observations unveiled that heightened expression levels of CARS1, CDKN1A, TFRC, FANCD2, FDFT1, HSPB1, SLC1A5, SLC7A11, coupled with reduced DPP4 expression, were indicative of an unfavorable prognosis. Built upon the nine previously discussed prognostic genes, the ferroptosis prognostic model offers a reliable means to forecast mesothelioma patients' survival with a substantial degree of precision. Furthermore, a notable correlation emerged between these prognostic ferroptosis regulators and parameters such as immune cell infiltration, tumor mutation burden, microsatellite instability, and PD-L1 expression in the context of mesothelioma. Within this cadre of nine ferroptosis regulatory factors with prognostic relevance, FANCD2 exhibited the most pronounced prognostic influence, as elucidated by our analyses. Subsequently, we executed a validation process employing clinical specimens sourced from our institution, thus confirming that heightened FANCD2 expression is a discernible harbinger of an adverse prognosis in the context of mesothelioma. In vitro experiments revealed that knocking down FANCD2 markedly suppressed the proliferation, migration, and ability of mesothelioma cells to attract immune cells. Furthermore, our findings also showed that reducing FANCD2 levels heightened the vulnerability of mesothelioma cells to inducers of ferroptosis. Furthermore, an extensive pan-cancer analysis uncovered a robust association between FANCD2 and the gene expression linked to immune checkpoints, thereby signifying an adverse prognosis across a broad spectrum of cancer types. Additional research is warranted to validate these findings.
Collapse
Affiliation(s)
- Zairui Wang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China; Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Jialin Huang
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - MinYang
- Department of Pathology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Liren Fu
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Shijie Liu
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Jianghua Huang
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
| | - Jingjing Han
- Department of Pathology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
| | - Xiaohui Zhao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China; Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China; Department of Oncology, Shenshan Medical Centre, Memorial Hospital of Sun Yat-Sen University, Shanwei, 516621.
| |
Collapse
|
32
|
Kang H, Meng F, Liu F, Xie M, Lai H, Li P, Zhang X. Nanomedicines Targeting Ferroptosis to Treat Stress-Related Diseases. Int J Nanomedicine 2024; 19:8189-8210. [PMID: 39157732 PMCID: PMC11328858 DOI: 10.2147/ijn.s476948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 08/03/2024] [Indexed: 08/20/2024] Open
Abstract
Ferroptosis, a unique form of regulated cell death driven by iron-dependent lethal lipid peroxidation, is implicated in various stress-related diseases like neurodegeneration, vasculopathy, and metabolic disturbance. Stress-related diseases encompass widespread medical disorders that are influenced or exacerbated by stress. These stressors can manifest in various organ or tissue systems and have significant implications for human overall health. Understanding ferroptosis in these diseases offers insights for therapeutic strategies targeting relevant pathways. This review explores ferroptosis mechanisms, its role in pathophysiology, its connection to stress-related diseases, and the potential of ferroptosis-targeted nanomedicines in treating conditions. This monograph also delves into the engineering of ferroptosis-targeted nanomedicines for tackling stress-related diseases, including cancer, cardia-cerebrovascular, neurodegenerative, metabolic and inflammatory diseases. Anyhow, nanotherapy targeting ferroptosis holds promise by both promoting and suppressing ferroptosis for managing stress-related diseases.
Collapse
Affiliation(s)
- Hao Kang
- Department of Medicinal Chemistry and Pharmaceutical Analysis, Anhui College of Traditional Chinese Medicine, Wuhu, People’s Republic of China
- Wuhu Modern Technology Research and Development Center of Chinese Medicine and Functional Food, Wuhu, People’s Republic of China
| | - Fansu Meng
- Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Traditional Chinese Medicine, Zhongshan, People’s Republic of China
| | - Fengjie Liu
- Department of Pharmaceutics, College of Pharmacy, Jinan University, Guangzhou, People’s Republic of China
| | - Mengjie Xie
- Department of Pharmaceutics, College of Pharmacy, Jinan University, Guangzhou, People’s Republic of China
| | - Haibiao Lai
- Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Traditional Chinese Medicine, Zhongshan, People’s Republic of China
| | - Pengfei Li
- Department of Oncology, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, People’s Republic of China
| | - Xingwang Zhang
- Department of Pharmaceutics, College of Pharmacy, Jinan University, Guangzhou, People’s Republic of China
| |
Collapse
|
33
|
Liu X, Ren B, Ren J, Gu M, You L, Zhao Y. The significant role of amino acid metabolic reprogramming in cancer. Cell Commun Signal 2024; 22:380. [PMID: 39069612 DOI: 10.1186/s12964-024-01760-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 07/21/2024] [Indexed: 07/30/2024] Open
Abstract
Amino acid metabolism plays a pivotal role in tumor microenvironment, influencing various aspects of cancer progression. The metabolic reprogramming of amino acids in tumor cells is intricately linked to protein synthesis, nucleotide synthesis, modulation of signaling pathways, regulation of tumor cell metabolism, maintenance of oxidative stress homeostasis, and epigenetic modifications. Furthermore, the dysregulation of amino acid metabolism also impacts tumor microenvironment and tumor immunity. Amino acids can act as signaling molecules that modulate immune cell function and immune tolerance within the tumor microenvironment, reshaping the anti-tumor immune response and promoting immune evasion by cancer cells. Moreover, amino acid metabolism can influence the behavior of stromal cells, such as cancer-associated fibroblasts, regulate ECM remodeling and promote angiogenesis, thereby facilitating tumor growth and metastasis. Understanding the intricate interplay between amino acid metabolism and the tumor microenvironment is of crucial significance. Expanding our knowledge of the multifaceted roles of amino acid metabolism in tumor microenvironment holds significant promise for the development of more effective cancer therapies aimed at disrupting the metabolic dependencies of cancer cells and modulating the tumor microenvironment to enhance anti-tumor immune responses and inhibit tumor progression.
Collapse
Affiliation(s)
- Xiaohong Liu
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, P.R, 100023, China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, P.R, China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, P.R, China
| | - Bo Ren
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, P.R, 100023, China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, P.R, China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, P.R, China
| | - Jie Ren
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, P.R, 100023, China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, P.R, China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, P.R, China
| | - Minzhi Gu
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, P.R, 100023, China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, P.R, China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, P.R, China
| | - Lei You
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, P.R, 100023, China.
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, P.R, China.
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, P.R, China.
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, P.R, 100023, China.
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, P.R, China.
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, P.R, China.
| |
Collapse
|
34
|
Zhao R, Sukocheva O, Tse E, Neganova M, Aleksandrova Y, Zheng Y, Gu H, Zhao D, Madhunapantula SV, Zhu X, Liu J, Fan R. Cuproptosis, the novel type of oxidation-induced cell death in thoracic cancers: can it enhance the success of immunotherapy? Cell Commun Signal 2024; 22:379. [PMID: 39068453 PMCID: PMC11282696 DOI: 10.1186/s12964-024-01743-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 07/08/2024] [Indexed: 07/30/2024] Open
Abstract
Copper is an important metal micronutrient, required for the balanced growth and normal physiological functions of human organism. Copper-related toxicity and dysbalanced metabolism were associated with the disruption of intracellular respiration and the development of various diseases, including cancer. Notably, copper-induced cell death was defined as cuproptosis which was also observed in malignant cells, representing an attractive anti-cancer instrument. Excess of intracellular copper leads to the aggregation of lipoylation proteins and toxic stress, ultimately resulting in the activation of cell death. Differential expression of cuproptosis-related genes was detected in normal and malignant tissues. Cuproptosis-related genes were also linked to the regulation of oxidative stress, immune cell responses, and composition of tumor microenvironment. Activation of cuproptosis was associated with increased expression of redox-metabolism-regulating genes, such as ferredoxin 1 (FDX1), lipoic acid synthetase (LIAS), lipoyltransferase 1 (LIPT1), dihydrolipoamide dehydrogenase (DLD), drolipoamide S-acetyltransferase (DLAT), pyruvate dehydrogenase E1 subunit alpha 1 (PDHA1), and pyruvate dehydrogenase E1 subunit beta (PDHB)). Accordingly, copper-activated network was suggested as an attractive target in cancer therapy. Mechanisms of cuproptosis and regulation of cuproptosis-related genes in different cancers and tumor microenvironment are discussed in this study. The analysis of current findings indicates that therapeutic regulation of copper signaling, and activation of cuproptosis-related targets may provide an effective tool for the improvement of immunotherapy regimens.
Collapse
Affiliation(s)
- Ruiwen Zhao
- The Department of Radiation Oncology & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Olga Sukocheva
- Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Port Rd, Adelaide, SA, 5000, Australia.
| | - Edmund Tse
- Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Port Rd, Adelaide, SA, 5000, Australia
| | - Margarita Neganova
- Nesmeyanov Institute of Organoelement Compounds, Russian Academy of Sciences, Moscow, 119991, Russia
| | - Yulia Aleksandrova
- Nesmeyanov Institute of Organoelement Compounds, Russian Academy of Sciences, Moscow, 119991, Russia
| | - Yufei Zheng
- The Department of Radiation Oncology & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Hao Gu
- The Department of Radiation Oncology & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Deyao Zhao
- The Department of Radiation Oncology & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - SabbaRao V Madhunapantula
- Special Interest Group in Cancer Biology and Cancer Stem Cells (SIG-CBCSC), Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education & Research, Mysuru, Karnataka, 570015, India
| | - Xiaorong Zhu
- The Department of Radiation Oncology & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Junqi Liu
- The Department of Radiation Oncology & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Ruitai Fan
- The Department of Radiation Oncology & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
35
|
Kapper C, Oppelt P, Arbeithuber B, Gyunesh AA, Vilusic I, Stelzl P, Rezk-Füreder M. Targeting ferroptosis in ovarian cancer: Novel strategies to overcome chemotherapy resistance. Life Sci 2024; 349:122720. [PMID: 38762066 DOI: 10.1016/j.lfs.2024.122720] [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: 03/08/2024] [Revised: 05/02/2024] [Accepted: 05/13/2024] [Indexed: 05/20/2024]
Abstract
AIMS This review investigates the role of ferroptosis in combating chemotherapy resistance in ovarian cancer, with a focus on its underlying mechanisms and therapeutic implications. MAIN METHODS A database search was conducted up to December 2023 using PubMed, Scopus, Google Scholar, Web of Science, and the Cochrane Library. The keywords "ovarian cancer," "ferroptosis," "cisplatin," and "cisplatin resistance" were employed. We included studies that offered original data on the application of ferroptosis in platinum-based chemotherapy, focusing on both in-vitro and in-vivo research models. KEY FINDINGS Our review reveals that ferroptosis significantly influences drug resistance in ovarian cancer. It investigates the existing studies to understand the role of ferroptosis in platinum resistance and explores its underlying mechanisms and assesses potential therapeutic strategies that uses ferroptosis to improve outcomes. The findings underscore the importance of ferroptosis in enhancing the effectiveness of platinum-based treatments and improving patient prognosis. SIGNIFICANCE The potential of ferroptosis induction to develop novel therapeutic strategies against ovarian cancer, especially in cisplatin-resistant cases, is promising. The preliminary nature of these findings highlights the necessity for further research to bring these insights into clinical practice. This would not only improve treatment outcomes and prognosis but also encourage ongoing studies into ferroptosis as a viable therapeutic approach.
Collapse
Affiliation(s)
- Celine Kapper
- Experimental Gynaecology, Obstetrics and Gynaecological Endocrinology, Johannes Kepler University Linz, Altenberger Strasse 69, 4040 Linz, Austria
| | - Peter Oppelt
- Experimental Gynaecology, Obstetrics and Gynaecological Endocrinology, Johannes Kepler University Linz, Altenberger Strasse 69, 4040 Linz, Austria; Department for Gynaecology, Obstetrics and Gynaecological Endocrinology, Kepler University Hospital, Johannes Kepler University Linz, Austria
| | - Barbara Arbeithuber
- Experimental Gynaecology, Obstetrics and Gynaecological Endocrinology, Johannes Kepler University Linz, Altenberger Strasse 69, 4040 Linz, Austria
| | - Ayberk Alp Gyunesh
- Experimental Gynaecology, Obstetrics and Gynaecological Endocrinology, Johannes Kepler University Linz, Altenberger Strasse 69, 4040 Linz, Austria
| | - Ivona Vilusic
- Experimental Gynaecology, Obstetrics and Gynaecological Endocrinology, Johannes Kepler University Linz, Altenberger Strasse 69, 4040 Linz, Austria
| | - Patrick Stelzl
- Department for Gynaecology, Obstetrics and Gynaecological Endocrinology, Kepler University Hospital, Johannes Kepler University Linz, Austria
| | - Marlene Rezk-Füreder
- Experimental Gynaecology, Obstetrics and Gynaecological Endocrinology, Johannes Kepler University Linz, Altenberger Strasse 69, 4040 Linz, Austria.
| |
Collapse
|
36
|
Wu R, Li J, Tian H, Song D, Zhao T, Tian Y, Zouboulis CC, Jiang J, Zhu M. Unveiling the mechanism of photothermal therapy in acne man-agement: targeting sebaceous gland ferroptosis via umbilical cord mesenchymal stem cell membrane-encapsulated Au-Ag-PDA. Front Bioeng Biotechnol 2024; 12:1426477. [PMID: 38915336 PMCID: PMC11194360 DOI: 10.3389/fbioe.2024.1426477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 05/20/2024] [Indexed: 06/26/2024] Open
Abstract
Background Branched gold and silver nanoparticles coated with polydopamine (Au-Ag-PDA) demonstrate high photothermal conversion efficiency. Utilizing umbilical cord mesenchymal stem cell membranes (MSCM) as an effective drug delivery system, our preliminary studies investigated the suppression of sebum secretion in sebaceous glands using MSCM-coated Au-Ag-PDA nano-particles (Au-Ag-PDA@MSCM) combined with 808 nm laser irradiation, showing potential for dermatological applications in acne treatment. Methods This study employs proteomic analysis, complemented by subsequent techniques such as Western blotting (WB), small interfering RNA (siRNA), and transmission electron microscopy, to further investigate the differential mechanisms by which Au-Ag-PDA and Au-Ag-PDA@MSCM-mediated photothermal therapy (PTT) suppress sebum secretion. Results Our proteomic analysis indicated mitochondrial respiratory chain damage in sebaceous gland tissues post-PTT, with further validation revealing ferroptosis in sebaceous cells and tissues. Acyl-CoA Synthetase Long-Chain Family Member 4 (Acsl4) has been identified as a critical target, with Au-Ag-PDA@MSCM demonstrating enhanced ferroptotic effects. Conclusion These findings significantly advance our understanding of how PTT mediated by Au-Ag-PDA@MSCM nanoparticles reduces sebum secretion and underscore the pivotal role of MSCM in inducing ferroptosis in sebaceous glands, thus providing a robust theoretical foundation for employing PTT via specific molecular pathways in acne treatment.
Collapse
Affiliation(s)
- Ronghui Wu
- Department of Dermatology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jing Li
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Hao Tian
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Dandan Song
- Department of Dermatology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Tianqi Zhao
- Department of Dermatology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yangyang Tian
- Department of Dermatology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Christos C. Zouboulis
- Departments of Dermatology, Venereology, Allergology and Immunology, Staedtisches Klinikum Dessau, Brandenburg Medical School Theodor Fontane and Faculty of Health Sciences Brandenburg, Dessau, Germany
| | - Jinlan Jiang
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Mingji Zhu
- Department of Dermatology, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
37
|
Li Y, Tuerxun H, Zhao Y, Liu X, Li X, Wen S, Zhao Y. The new era of lung cancer therapy: Combining immunotherapy with ferroptosis. Crit Rev Oncol Hematol 2024; 198:104359. [PMID: 38615871 DOI: 10.1016/j.critrevonc.2024.104359] [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: 10/29/2023] [Revised: 03/12/2024] [Accepted: 04/10/2024] [Indexed: 04/16/2024] Open
Abstract
Ferroptosis is an unconventional programmed cell death mode caused by phospholipid peroxidation dependent on iron. Emerging immunotherapies (especially immune checkpoint inhibitors) have the potential to enhance lung cancer patients' long-term survival. Although immunotherapy has yielded significant positive applications in some patients, there are still many mechanisms that can cause lung cancer cells to evade immunity, thus leading to the failure of targeted therapies. Immune-tolerant cancer cells are insensitive to conventional death pathways such as apoptosis and necrosis, whereas mesenchymal and metastasis-prone cancer cells are particularly vulnerable to ferroptosis, which plays a vital role in mediating immune tolerance resistance by tumors and immune cells. As a result, triggering lung cancer cell ferroptosis holds significant therapeutic potential for drug-resistant malignancies. Here, we summarize the mechanisms underlying the suppression of ferroptosis in lung cancer, highlight its function in the lung cancer immune microenvironment, and propose possible therapeutic strategies.
Collapse
Affiliation(s)
- Yawen Li
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Halahati Tuerxun
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Yixin Zhao
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Xingyu Liu
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Xi Li
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Shuhui Wen
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Yuguang Zhao
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin 130021, China.
| |
Collapse
|
38
|
Nejadi Orang F, Abdoli Shadbad M. Competing endogenous RNA networks and ferroptosis in cancer: novel therapeutic targets. Cell Death Dis 2024; 15:357. [PMID: 38778030 PMCID: PMC11111666 DOI: 10.1038/s41419-024-06732-4] [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: 12/12/2023] [Revised: 05/02/2024] [Accepted: 05/08/2024] [Indexed: 05/25/2024]
Abstract
As a newly identified regulated cell death, ferroptosis is a metabolically driven process that relies on iron and is associated with polyunsaturated fatty acyl peroxidation, elevated levels of reactive oxygen species (ROS), and mitochondrial damage. This distinct regulated cell death is dysregulated in various cancers; activating ferroptosis in malignant cells increases cancer immunotherapy and chemoradiotherapy responses across different malignancies. Over the last decade, accumulating research has provided evidence of cross-talk between non-coding RNAs (ncRNAs) and competing endogenous RNA (ceRNA) networks and highlighted their significance in developing and progressing malignancies. Aside from pharmaceutical agents to regulate ferroptosis, recent studies have shed light on the potential of restoring dysregulated ferroptosis-related ceRNA networks in cancer treatment. The present study provides a comprehensive and up-to-date review of the ferroptosis significance, ferroptosis pathways, the role of ferroptosis in cancer immunotherapy and chemoradiotherapy, ceRNA biogenesis, and ferroptosis-regulating ceRNA networks in different cancers. The provided insights can offer the authorship with state-of-the-art findings and future perspectives regarding the ferroptosis and ferroptosis-related ceRNA networks and their implication in the treatment and determining the prognosis of affected patients.
Collapse
Affiliation(s)
| | - Mahdi Abdoli Shadbad
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Immunology, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
39
|
Fan Z, Wu S, Deng H, Li G, Huang L, Liu H. Light-Triggered Nanozymes Remodel the Tumor Hypoxic and Immunosuppressive Microenvironment for Ferroptosis-Enhanced Antitumor Immunity. ACS NANO 2024; 18:12261-12275. [PMID: 38683132 DOI: 10.1021/acsnano.4c00844] [Citation(s) in RCA: 40] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/01/2024]
Abstract
Cancer immunotherapy holds significant promise for addressing diverse malignancies. Nevertheless, its efficacy remains constrained by the intricate tumor immunosuppressive microenvironment. Herein, a light-triggered nanozyme Fe-TCPP-R848-PEG (Fe-MOF-RP) was designed for remodeling the immunosuppressive microenvironment. The Fe-TCPP-MOFs were utilized not only as a core catalysis component against tumor destruction but also as a biocompatible delivery vector of an immunologic agonist, improving its long circulation and tumor enrichment. Concurrently, it catalyzes the decomposition of H2O2 within the tumor, yielding oxygen to augment photodynamic therapy. The induced ferroptosis, in synergy with photodynamic therapy, prompts the liberation of tumor-associated antigens from tumor cells inducing immunogenic cell death. Phototriggered on-demand release of R848 agonists stimulated the maturation of dendritic cells and reverted the tumor-promoting M2 phenotypes into adoptive M1 macrophages, which further reshaped the tumor immunosuppressive microenvironment. Notably, the nanozyme effectively restrains well-established tumors, such as B16F10 melanoma. Moreover, it demonstrates a distal tumor-inhibiting effect upon in situ light treatment. What is more, in a lung metastasis model, it elicits robust immune memory, conferring enduring protection against tumor rechallenge. Our study presents a straightforward and broadly applicable strategy for crafting nanozymes with the potential to effectively thwart cancer recurrence and metastasis.
Collapse
Affiliation(s)
- Zhijin Fan
- Guangdong Provincial Key Laboratory of Urology, Guangdong Engineering Research Center of Urinary Minimally Invasive Surgery Robot and Intelligent Equipment, Guangzhou Institute of Urology, Department of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510120, China
- School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Sicheng Wu
- Guangdong Provincial Key Laboratory of Urology, Guangdong Engineering Research Center of Urinary Minimally Invasive Surgery Robot and Intelligent Equipment, Guangzhou Institute of Urology, Department of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510120, China
| | - Huaping Deng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Guanlin Li
- Guangdong Provincial Key Laboratory of Urology, Guangdong Engineering Research Center of Urinary Minimally Invasive Surgery Robot and Intelligent Equipment, Guangzhou Institute of Urology, Department of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510120, China
| | - Linghong Huang
- Guangdong Provincial Key Laboratory of Urology, Guangdong Engineering Research Center of Urinary Minimally Invasive Surgery Robot and Intelligent Equipment, Guangzhou Institute of Urology, Department of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510120, China
| | - Hongxing Liu
- Guangdong Provincial Key Laboratory of Urology, Guangdong Engineering Research Center of Urinary Minimally Invasive Surgery Robot and Intelligent Equipment, Guangzhou Institute of Urology, Department of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510120, China
| |
Collapse
|
40
|
Khan A, Huo Y, Guo Y, Shi J, Hou Y. Ferroptosis is an effective strategy for cancer therapy. Med Oncol 2024; 41:124. [PMID: 38652406 DOI: 10.1007/s12032-024-02317-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 01/29/2024] [Indexed: 04/25/2024]
Abstract
Ferroptosis is a form of intracellular iron-dependent cell death that differs from necrosis, autophagy and apoptosis. Intracellular iron mediates Fenton reaction resulting in lipid peroxidation production, which in turn promotes cell death. Although cancer cell exhibit's ability to escape ferroptosis by multiple pathways such as SLC7A11, GPX4, induction of ferroptosis could inhibit cancer cell proliferation, migration and invasion. In tumor microenvironment, ferroptosis could affect immune cell (T cells, macrophages etc.) activity, which in turn regulates tumor immune escape. In addition, ferroptosis in cancer cells could activate immune cell activity by antigen processing and presentation. Therefore, ferroptosis could be an effective strategy for cancer therapy such as chemotherapy, radiotherapy, and immunotherapy. In this paper, we reviewed the role of ferroptosis on tumor progression and therapy, which may provide a strategy for cancer treatment.
Collapse
Affiliation(s)
- Afrasyab Khan
- School of Life Sciences, Jiangsu University, Zhenjiang, 212013, Jiangsu Province, People's Republic of China
| | - Yu Huo
- School of Life Sciences, Jiangsu University, Zhenjiang, 212013, Jiangsu Province, People's Republic of China
| | - Yilei Guo
- School of Life Sciences, Jiangsu University, Zhenjiang, 212013, Jiangsu Province, People's Republic of China
| | - Juanjuan Shi
- School of Life Sciences, Jiangsu University, Zhenjiang, 212013, Jiangsu Province, People's Republic of China
| | - Yongzhong Hou
- School of Life Sciences, Jiangsu University, Zhenjiang, 212013, Jiangsu Province, People's Republic of China.
- , Zhenjiang, People's Republic of China.
| |
Collapse
|
41
|
Santoro N, Salutari P, Di Ianni M, Marra A. Precision Medicine Approaches in Acute Myeloid Leukemia with Adverse Genetics. Int J Mol Sci 2024; 25:4259. [PMID: 38673842 PMCID: PMC11050344 DOI: 10.3390/ijms25084259] [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: 03/11/2024] [Revised: 04/08/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
The treatment of acute myeloid leukemia (AML) with adverse genetics remains unsatisfactory, with very low response rates to standard chemotherapy and shorter durations of remission commonly observed in these patients. The complex biology of AML with adverse genetics is continuously evolving. Herein, we discuss recent advances in the field focusing on the contribution of molecular drivers of leukemia biogenesis and evolution and on the alterations of the immune system that can be exploited with immune-based therapeutic strategies. We focus on the biological rationales for combining targeted therapy and immunotherapy, which are currently being investigated in ongoing trials, and could hopefully ameliorate the poor outcomes of patients affected by AML with adverse genetics.
Collapse
Affiliation(s)
- Nicole Santoro
- Hematology Unit, Department of Hematology and Oncology, Ospedale Civile “Santo Spirito”, 65122 Pescara, Italy; (P.S.); (M.D.I.)
| | - Prassede Salutari
- Hematology Unit, Department of Hematology and Oncology, Ospedale Civile “Santo Spirito”, 65122 Pescara, Italy; (P.S.); (M.D.I.)
| | - Mauro Di Ianni
- Hematology Unit, Department of Hematology and Oncology, Ospedale Civile “Santo Spirito”, 65122 Pescara, Italy; (P.S.); (M.D.I.)
- Department of Medicine and Science of Aging, “G.D’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Andrea Marra
- Laboratory of Molecular Medicine and Biotechnology, Department of Medicine, University Campus Bio-Medico of Rome, 00128 Rome, Italy
- Institute of Translational Pharmacology, National Research Council of Italy (CNR), 00196 Rome, Italy
| |
Collapse
|
42
|
Ashoub MH, Razavi R, Heydaryan K, Salavati-Niasari M, Amiri M. Targeting ferroptosis for leukemia therapy: exploring novel strategies from its mechanisms and role in leukemia based on nanotechnology. Eur J Med Res 2024; 29:224. [PMID: 38594732 PMCID: PMC11003188 DOI: 10.1186/s40001-024-01822-7] [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: 10/05/2023] [Accepted: 03/30/2024] [Indexed: 04/11/2024] Open
Abstract
The latest findings in iron metabolism and the newly uncovered process of ferroptosis have paved the way for new potential strategies in anti-leukemia treatments. In the current project, we reviewed and summarized the current role of nanomedicine in the treatment and diagnosis of leukemia through a comparison made between traditional approaches applied in the treatment and diagnosis of leukemia via the existing investigations about the ferroptosis molecular mechanisms involved in various anti-tumor treatments. The application of nanotechnology and other novel technologies may provide a new direction in ferroptosis-driven leukemia therapies. The article explores the potential of targeting ferroptosis, a new form of regulated cell death, as a new therapeutic strategy for leukemia. It discusses the mechanisms of ferroptosis and its role in leukemia and how nanotechnology can enhance the delivery and efficacy of ferroptosis-inducing agents. The article not only highlights the promise of ferroptosis-targeted therapies and nanotechnology in revolutionizing leukemia treatment, but also calls for further research to overcome challenges and fully realize the clinical potential of this innovative approach. Finally, it discusses the challenges and opportunities in clinical applications of ferroptosis.
Collapse
Affiliation(s)
- Muhammad Hossein Ashoub
- Department of Hematology and Medical Laboratory Sciences, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran
- Stem Cells and Regenerative Medicine Innovation Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Razieh Razavi
- Department of Chemistry, Faculty of Science, University of Jiroft, Jiroft, Iran
| | - Kamran Heydaryan
- Department of Medical Biochemical Analysis, Cihan University-Erbil, Kurdistan Region, Iraq
| | - Masoud Salavati-Niasari
- Institute of Nano Science and Nano Technology, University of Kashan, P.O. Box 87317-51167, Kashan, Iran
| | - Mahnaz Amiri
- Student Research Committee, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran.
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Science, Kerman, Iran.
| |
Collapse
|
43
|
Stoess C, Choi YK, Onyuru J, Friess H, Hoffman HM, Hartmann D, Feldstein AE. Cell Death in Liver Disease and Liver Surgery. Biomedicines 2024; 12:559. [PMID: 38540172 PMCID: PMC10968531 DOI: 10.3390/biomedicines12030559] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 02/15/2024] [Accepted: 02/17/2024] [Indexed: 01/03/2025] Open
Abstract
Cell death is crucial for maintaining tissue balance and responding to diseases. However, under pathological conditions, the surge in dying cells results in an overwhelming presence of cell debris and the release of danger signals. In the liver, this gives rise to hepatic inflammation and hepatocellular cell death, which are key factors in various liver diseases caused by viruses, toxins, metabolic issues, or autoimmune factors. Both clinical and in vivo studies strongly affirm that hepatocyte death serves as a catalyst in the progression of liver disease. This advancement is characterized by successive stages of inflammation, fibrosis, and cirrhosis, culminating in a higher risk of tumor development. In this review, we explore pivotal forms of cell death, including apoptosis, pyroptosis, and necroptosis, examining their roles in both acute and chronic liver conditions, including liver cancer. Furthermore, we discuss the significance of cell death in liver surgery and ischemia-reperfusion injury. Our objective is to illuminate the molecular mechanisms governing cell death in liver diseases, as this understanding is crucial for identifying therapeutic opportunities aimed at modulating cell death pathways.
Collapse
Affiliation(s)
- Christian Stoess
- Department of Pediatric Gastroenterology, University of California San Diego, 9500 Gilman Dr, La Jolla, CA 92093, USA; (C.S.)
- Department of Surgery, TUM School of Medicine, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| | - Yeon-Kyung Choi
- Department of Pediatric Gastroenterology, University of California San Diego, 9500 Gilman Dr, La Jolla, CA 92093, USA; (C.S.)
- Department of Internal Medicine, School of Medicine, Kyungpook National University Chilgok Hospital, Kyungpook National University, Daegu 41404, Republic of Korea
| | - Janset Onyuru
- Department of Pediatric Allergy, Immunology and Rheumatology, University of California San Diego, La Jolla, CA 92093, USA
| | - Helmut Friess
- Department of Surgery, TUM School of Medicine, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| | - Hal M. Hoffman
- Department of Pediatric Allergy, Immunology and Rheumatology, University of California San Diego, La Jolla, CA 92093, USA
| | - Daniel Hartmann
- Department of Surgery, TUM School of Medicine, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| | - Ariel E. Feldstein
- Department of Pediatric Gastroenterology, University of California San Diego, 9500 Gilman Dr, La Jolla, CA 92093, USA; (C.S.)
- Novo Nordisk, Global Drug Discovery, Ørestads Boulevard 108, 2300 Copenhagen, Denmark
| |
Collapse
|
44
|
Yuan Q, Fang Y, Guo J, Zhang Z, Liao J, Kuang J. Therapeutic potential and mechanisms of Rifaximin in ameliorating iron overload-induced ferroptosis and liver fibrosis in vivo and in vitro. Toxicol Appl Pharmacol 2024; 484:116845. [PMID: 38331104 DOI: 10.1016/j.taap.2024.116845] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 02/02/2024] [Accepted: 02/05/2024] [Indexed: 02/10/2024]
Abstract
Liver fibrosis could progress to liver cirrhosis with several contributing factors, one being iron overload which triggers ferroptosis, a form of regulated cell death. Rifaximin, a non-absorbable antibiotic, has shown promise in mitigating fibrosis, primarily by modulating gut microbiota. This study investigated the effects and mechanisms of rifaximin on iron overload-related hepatic fibrosis and ferroptosis. In an iron overload-induced liver fibrosis model in mice and in ferric ammonium citrate (FAC)-stimulated primary hepatocytes, treatment with rifaximin showed significant therapeutic effects. Specifically, it ameliorated the processes of ferroptosis triggered by iron overload, reduced liver injury, and alleviated fibrosis. This was demonstrated by decreased iron accumulation in the liver, improved liver function, and reduced fibrotic area and collagen deposition. Rifaximin also modulated key proteins related to iron homeostasis and ferroptosis, including reduced expression of TFR1, a protein facilitating cellular iron uptake, and increased expression of Fpn and FTH, proteins involved in iron export and storage. In the context of oxidative stress, rifaximin treatment led to a decrease in lipid peroxidation, evidenced by reduced levels of reactive oxygen species (ROS) and malondialdehyde (MDA), and an increase in the reduced glutathione (GSH) and decrease in oxidized glutathione (GSSG). Notably, rifaximin's potential functions were associated with the TGF-β pathway, evidenced by suppressed Tgfb1 protein levels and ratios of phosphorylated to total Smad2 and Smad3, whereas increased Smad7 phosphorylation. These findings indicate rifaximin's therapeutic potential in managing liver fibrosis by modulating the TGF-β pathway and reducing iron overload-induced damage. Further research is required to confirm these results and explore their clinical implications.
Collapse
Affiliation(s)
- Qi Yuan
- Department of Hepatopathy, The Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha 410005, China
| | - Yuan Fang
- Department of Hepatopathy, The Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha 410005, China.
| | - Jingyun Guo
- Department of Hepatopathy, The Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha 410005, China
| | - Zheng Zhang
- Department of Hepatopathy, The Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha 410005, China
| | - Jinmao Liao
- Department of Hepatopathy, The Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha 410005, China
| | - Jia Kuang
- Department of Hepatopathy, The Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha 410005, China
| |
Collapse
|
45
|
Menendez JA, Cuyàs E, Encinar JA, Vander Steen T, Verdura S, Llop‐Hernández À, López J, Serrano‐Hervás E, Osuna S, Martin‐Castillo B, Lupu R. Fatty acid synthase (FASN) signalome: A molecular guide for precision oncology. Mol Oncol 2024; 18:479-516. [PMID: 38158755 PMCID: PMC10920094 DOI: 10.1002/1878-0261.13582] [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: 10/02/2023] [Revised: 10/27/2023] [Accepted: 12/28/2023] [Indexed: 01/03/2024] Open
Abstract
The initial excitement generated more than two decades ago by the discovery of drugs targeting fatty acid synthase (FASN)-catalyzed de novo lipogenesis for cancer therapy was short-lived. However, the advent of the first clinical-grade FASN inhibitor (TVB-2640; denifanstat), which is currently being studied in various phase II trials, and the exciting advances in understanding the FASN signalome are fueling a renewed interest in FASN-targeted strategies for the treatment and prevention of cancer. Here, we provide a detailed overview of how FASN can drive phenotypic plasticity and cell fate decisions, mitochondrial regulation of cell death, immune escape and organ-specific metastatic potential. We then present a variety of FASN-targeted therapeutic approaches that address the major challenges facing FASN therapy. These include limitations of current FASN inhibitors and the lack of precision tools to maximize the therapeutic potential of FASN inhibitors in the clinic. Rethinking the role of FASN as a signal transducer in cancer pathogenesis may provide molecularly driven strategies to optimize FASN as a long-awaited target for cancer therapeutics.
Collapse
Affiliation(s)
- Javier A. Menendez
- Metabolism & Cancer Group, Program Against Cancer Therapeutic Resistance (ProCURE)Catalan Institute of OncologyGironaSpain
- Girona Biomedical Research InstituteGironaSpain
| | - Elisabet Cuyàs
- Metabolism & Cancer Group, Program Against Cancer Therapeutic Resistance (ProCURE)Catalan Institute of OncologyGironaSpain
- Girona Biomedical Research InstituteGironaSpain
| | - Jose Antonio Encinar
- Institute of Research, Development and Innovation in Biotechnology of Elche (IDiBE) and Molecular and Cell Biology Institute (IBMC)Miguel Hernández University (UMH)ElcheSpain
| | - Travis Vander Steen
- Division of Experimental Pathology, Department of Laboratory Medicine and PathologyMayo ClinicRochesterMNUSA
- Mayo Clinic Cancer CenterRochesterMNUSA
- Department of Biochemistry and Molecular Biology LaboratoryMayo Clinic LaboratoryRochesterMNUSA
| | - Sara Verdura
- Metabolism & Cancer Group, Program Against Cancer Therapeutic Resistance (ProCURE)Catalan Institute of OncologyGironaSpain
- Girona Biomedical Research InstituteGironaSpain
| | - Àngela Llop‐Hernández
- Metabolism & Cancer Group, Program Against Cancer Therapeutic Resistance (ProCURE)Catalan Institute of OncologyGironaSpain
- Girona Biomedical Research InstituteGironaSpain
| | - Júlia López
- Metabolism & Cancer Group, Program Against Cancer Therapeutic Resistance (ProCURE)Catalan Institute of OncologyGironaSpain
- Girona Biomedical Research InstituteGironaSpain
| | - Eila Serrano‐Hervás
- Metabolism & Cancer Group, Program Against Cancer Therapeutic Resistance (ProCURE)Catalan Institute of OncologyGironaSpain
- Girona Biomedical Research InstituteGironaSpain
- CompBioLab Group, Institut de Química Computacional i Catàlisi (IQCC) and Departament de QuímicaUniversitat de GironaGironaSpain
| | - Sílvia Osuna
- CompBioLab Group, Institut de Química Computacional i Catàlisi (IQCC) and Departament de QuímicaUniversitat de GironaGironaSpain
- ICREABarcelonaSpain
| | - Begoña Martin‐Castillo
- Metabolism & Cancer Group, Program Against Cancer Therapeutic Resistance (ProCURE)Catalan Institute of OncologyGironaSpain
- Girona Biomedical Research InstituteGironaSpain
- Unit of Clinical ResearchCatalan Institute of OncologyGironaSpain
| | - Ruth Lupu
- Division of Experimental Pathology, Department of Laboratory Medicine and PathologyMayo ClinicRochesterMNUSA
- Mayo Clinic Cancer CenterRochesterMNUSA
- Department of Biochemistry and Molecular Biology LaboratoryMayo Clinic LaboratoryRochesterMNUSA
| |
Collapse
|
46
|
Song G, Li M, Fan S, Qin M, Shao B, Dai W, Zhang H, Wang X, He B, Zhang Q. Boosting synergism of chemo- and immuno-therapies via switching paclitaxel-induced apoptosis to mevalonate metabolism-triggered ferroptosis by bisphosphonate coordination lipid nanogranules. Acta Pharm Sin B 2024; 14:836-853. [PMID: 38322346 PMCID: PMC10840482 DOI: 10.1016/j.apsb.2023.08.029] [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: 04/02/2023] [Revised: 07/13/2023] [Accepted: 07/25/2023] [Indexed: 02/08/2024] Open
Abstract
Conventional chemotherapy based on cytotoxic drugs is facing tough challenges recently following the advances of monoclonal antibodies and molecularly targeted drugs. It is critical to inspire new potential to remodel the value of this classical therapeutic strategy. Here, we fabricate bisphosphonate coordination lipid nanogranules (BC-LNPs) and load paclitaxel (PTX) to boost the chemo- and immuno-therapeutic synergism of cytotoxic drugs. Alendronate in BC-LNPs@PTX, a bisphosphonate to block mevalonate metabolism, works as both the structure and drug constituent in nanogranules, where alendronate coordinated with calcium ions to form the particle core. The synergy of alendronate enhances the efficacy of paclitaxel, suppresses tumor metastasis, and alters the cytotoxic mechanism. Differing from the paclitaxel-induced apoptosis, the involvement of alendronate inhibits the mevalonate metabolism, changes the mitochondrial morphology, disturbs the redox homeostasis, and causes the accumulation of mitochondrial ROS and lethal lipid peroxides (LPO). These factors finally trigger the ferroptosis of tumor cells, an immunogenic cell death mode, which remodels the suppressive tumor immune microenvironment and synergizes with immunotherapy. Therefore, by switching paclitaxel-induced apoptosis to mevalonate metabolism-triggered ferroptosis, BC-LNPs@PTX provides new insight into the development of cytotoxic drugs and highlights the potential of metabolism regulation in cancer therapy.
Collapse
Affiliation(s)
- Ge Song
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Minghui Li
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Shumin Fan
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Mengmeng Qin
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Bin Shao
- Department of Medical Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital, Beijing 100142, China
| | - Wenbing Dai
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Hua Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xueqing Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Bing He
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Qiang Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| |
Collapse
|
47
|
Consoli V, Fallica AN, Sorrenti V, Pittalà V, Vanella L. Novel Insights on Ferroptosis Modulation as Potential Strategy for Cancer Treatment: When Nature Kills. Antioxid Redox Signal 2024; 40:40-85. [PMID: 37132605 PMCID: PMC10824235 DOI: 10.1089/ars.2022.0179] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 04/19/2023] [Accepted: 04/20/2023] [Indexed: 05/04/2023]
Abstract
Significance: The multifactorial nature of the mechanisms implicated in cancer development still represents a major issue for the success of established antitumor therapies. The discovery of ferroptosis, a novel form of programmed cell death distinct from apoptosis, along with the identification of the molecular pathways activated during its execution, has led to the uncovering of novel molecules characterized by ferroptosis-inducing properties. Recent advances: As of today, the ferroptosis-inducing properties of compounds derived from natural sources have been investigated and interesting findings have been reported both in vitro and in vivo. Critical Issues: Despite the efforts made so far, only a limited number of synthetic compounds have been identified as ferroptosis inducers, and their utilization is still limited to basic research. In this review, we analyzed the most important biochemical pathways involved in ferroptosis execution, with particular attention to the newest literature findings on canonical and non-canonical hallmarks, together with mechanisms of action of natural compounds identified as novel ferroptosis inducers. Compounds have been classified based on their chemical structure, and modulation of ferroptosis-related biochemical pathways has been reported. Future Directions: The outcomes herein collected represent a fascinating starting point from which to take hints for future drug discovery studies aimed at identifying ferroptosis-inducing natural compounds for anticancer therapies. Antioxid. Redox Signal. 40, 40-85.
Collapse
Affiliation(s)
- Valeria Consoli
- Department of Drug and Health Sciences, University of Catania, Catania, Italy
| | | | - Valeria Sorrenti
- Department of Drug and Health Sciences, University of Catania, Catania, Italy
- Department of Drug and Health Sciences, CERNUT—Research Centre on Nutraceuticals and Health Products, University of Catania, Catania, Italy
| | - Valeria Pittalà
- Department of Drug and Health Sciences, University of Catania, Catania, Italy
- Department of Drug and Health Sciences, CERNUT—Research Centre on Nutraceuticals and Health Products, University of Catania, Catania, Italy
| | - Luca Vanella
- Department of Drug and Health Sciences, University of Catania, Catania, Italy
- Department of Drug and Health Sciences, CERNUT—Research Centre on Nutraceuticals and Health Products, University of Catania, Catania, Italy
| |
Collapse
|
48
|
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
|
49
|
Lei Y, Lei X, Zhu A, Xie S, Zhang T, Wang C, Song A, Wang X, Shu G, Deng X. Ethanol Extract of Rosa rugosa Ameliorates Acetaminophen-Induced Liver Injury via Upregulating Sirt1 and Subsequent Potentiation of LKB1/AMPK/Nrf2 Cascade in Hepatocytes. Molecules 2023; 28:7307. [PMID: 37959727 PMCID: PMC10649261 DOI: 10.3390/molecules28217307] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 10/21/2023] [Accepted: 10/26/2023] [Indexed: 11/15/2023] Open
Abstract
Acetaminophen (APAP)-induced liver injury is a common hepatic disease resulting from drug abuse. Few targeted treatments are available clinically nowadays. The flower bud of Rosa rugosa has a wide range of biological activities. However, it is unclear whether it alleviates liver injury caused by APAP. Here, we prepared an ethanol extract of Rosa rugosa (ERS) and analyzed its chemical profile. Furthermore, we revealed that ERS significantly ameliorated APAP-induced apoptosis and ferroptosis in AML-12 hepatocytes and dampened APAP-mediated cytotoxicity. In AML-12 cells, ERS elevated Sirt1 expression, boosted the LKB1/AMPK/Nrf2 axis, and thereby crippled APAP-induced intracellular oxidative stress. Both EX527 and NAM, which are chemically unrelated inhibitors of Sirt1, blocked ERS-induced activation of LKB1/AMPK/Nrf2 signaling. The protection of ERS against APAP-triggered toxicity in AML-12 cells was subsequently abolished. As expression of LKB1 was knocked down, ERS still upregulated Sirt1 but failed to activate AMPK/Nrf2 cascade or suppress cytotoxicity provoked by APAP. Results of in vivo experiments showed that ERS attenuated APAP-caused hepatocyte apoptosis and ferroptosis and improved liver injury and inflammation. Consistently, ERS boosted Sirt1 expression, increased phosphorylations of LKB1 and AMPK, and promoted Nrf2 nuclear translocation in the livers of APAP-intoxicated mice. Hepatic transcriptions of HO-1 and GCLC, which are downstream antioxidant genes of Nrf2, were also significantly increased in response to ERS. Our results collectively indicated that ERS effectively attenuates APAP-induced liver injury by activating LKB1/AMPK/Nrf2 cascade. Upregulated expression of Sirt1 plays a crucial role in ERS-mediated activation of LKB1.
Collapse
Affiliation(s)
- Yecheng Lei
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan 430074, China; (Y.L.)
| | - Xiao Lei
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan 430074, China; (Y.L.)
| | - Anqi Zhu
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan 430074, China; (Y.L.)
| | - Shijie Xie
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan 430074, China; (Y.L.)
| | - Tiantian Zhang
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan 430074, China; (Y.L.)
| | - Chuo Wang
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan 430074, China; (Y.L.)
| | - Anning Song
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan 430074, China; (Y.L.)
| | - Xiaoming Wang
- School of Life Sciences, Nanjing University, Nanjing 210023, China;
| | - Guangwen Shu
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan 430074, China; (Y.L.)
| | - Xukun Deng
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan 430074, China; (Y.L.)
| |
Collapse
|
50
|
Liu Y, Lu S, Wu LL, Yang L, Yang L, Wang J. The diversified role of mitochondria in ferroptosis in cancer. Cell Death Dis 2023; 14:519. [PMID: 37580393 PMCID: PMC10425449 DOI: 10.1038/s41419-023-06045-y] [Citation(s) in RCA: 146] [Impact Index Per Article: 73.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 06/23/2023] [Accepted: 08/07/2023] [Indexed: 08/16/2023]
Abstract
Ferroptosis is a form of regulated cell death induced by iron-dependent lipid peroxidation, and it has been studied extensively since its discovery in 2012. Induced by iron overload and ROS accumulation, ferroptosis is modulated by various cellular metabolic and signaling pathways. The GSH-GPX4 pathway, the FSP1-CoQ10 pathway, the GCH1-BH4 pathway, the DHODH-CoQH2 system and the sex hormones suppress ferroptosis. Mitochondrial iron metabolism regulates ferroptosis and mitochondria also undergo a morphological change during ferroptosis, these changes include increased membrane density and reduced mitochondrial cristae. Moreover, mitochondrial energy metabolism changes during ferroptosis, the increased oxidative phosphorylation and ATP production rates lead to a decrease in the glycolysis rate. In addition, excessive oxidative stress induces irreversible damage to mitochondria, diminishing organelle integrity. ROS production, mitochondrial membrane potential, mitochondrial fusion and fission, and mitophagy also function in ferroptosis. Notably, some ferroptosis inhibitors target mitochondria. Ferroptosis is a major mechanism for cell death associated with the progression of cancer. Metastasis-prone or metastatic cancer cells are more susceptible to ferroptosis. Inducing ferroptosis in tumor cells shows very promising potential for treating drug-resistant cancers. In this review, we present a brief retrospect of the discovery and the characteristics of ferroptosis, then we discuss the regulation of ferroptosis and highlight the unique role played by mitochondria in the ferroptosis of cancer cells. Furthermore, we explain how ferroptosis functions as a double-edged sword as well as novel therapies aimed at selectively manipulating cell death for cancer eradication.
Collapse
Affiliation(s)
- Yu'e Liu
- Institute of Hepatobiliary and Pancreatic Surgery, Department of Hepatobiliary and Pancreatic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Shiping Lu
- Center for Translational Research in infection and Inflammation, School of Medicine, Tulane University, New Orleans, LA, 70112, USA
| | - Lei-Lei Wu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, 200433, Shanghai, China
| | - Liang Yang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, China
| | - Lixue Yang
- Department of Biliary Tract Surgery II, Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China.
| | - Jinghan Wang
- Institute of Hepatobiliary and Pancreatic Surgery, Department of Hepatobiliary and Pancreatic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
| |
Collapse
|