1
|
Song R, Yin S, Wu J, Yan J. Neuronal regulated cell death in aging-related neurodegenerative diseases: key pathways and therapeutic potentials. Neural Regen Res 2025; 20:2245-2263. [PMID: 39104166 PMCID: PMC11759035 DOI: 10.4103/nrr.nrr-d-24-00025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 05/13/2024] [Accepted: 06/18/2024] [Indexed: 08/07/2024] Open
Abstract
Regulated cell death (such as apoptosis, necroptosis, pyroptosis, autophagy, cuproptosis, ferroptosis, disulfidptosis) involves complex signaling pathways and molecular effectors, and has been proven to be an important regulatory mechanism for regulating neuronal aging and death. However, excessive activation of regulated cell death may lead to the progression of aging-related diseases. This review summarizes recent advances in the understanding of seven forms of regulated cell death in age-related diseases. Notably, the newly identified ferroptosis and cuproptosis have been implicated in the risk of cognitive impairment and neurodegenerative diseases. These forms of cell death exacerbate disease progression by promoting inflammation, oxidative stress, and pathological protein aggregation. The review also provides an overview of key signaling pathways and crosstalk mechanisms among these regulated cell death forms, with a focus on ferroptosis, cuproptosis, and disulfidptosis. For instance, FDX1 directly induces cuproptosis by regulating copper ion valency and dihydrolipoamide S-acetyltransferase aggregation, while copper mediates glutathione peroxidase 4 degradation, enhancing ferroptosis sensitivity. Additionally, inhibiting the Xc- transport system to prevent ferroptosis can increase disulfide formation and shift the NADP + /NADPH ratio, transitioning ferroptosis to disulfidptosis. These insights help to uncover the potential connections among these novel regulated cell death forms and differentiate them from traditional regulated cell death mechanisms. In conclusion, identifying key targets and their crosstalk points among various regulated cell death pathways may aid in developing specific biomarkers to reverse the aging clock and treat age-related neurodegenerative conditions.
Collapse
Affiliation(s)
- Run Song
- Department of Neurology, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan Province, China
- Neuromolecular Biology Laboratory, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan Province, China
| | - Shiyi Yin
- Department of Neurology, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan Province, China
- Neuromolecular Biology Laboratory, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan Province, China
| | - Jiannan Wu
- Neuromolecular Biology Laboratory, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan Province, China
| | - Junqiang Yan
- Department of Neurology, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan Province, China
- Neuromolecular Biology Laboratory, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan Province, China
| |
Collapse
|
2
|
Tang K, Ye T, He Y, Ba X, Xia D, Peng E, Chen Z, Ye Z, Yang X. Ferroptosis, necroptosis, and pyroptosis in calcium oxalate crystal-induced kidney injury. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167791. [PMID: 40086520 DOI: 10.1016/j.bbadis.2025.167791] [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: 11/30/2024] [Revised: 01/24/2025] [Accepted: 03/04/2025] [Indexed: 03/16/2025]
Abstract
Kidney stones represent a highly prevalent urological disorder worldwide, with high incidence and recurrence rates. Calcium oxalate (CaOx) crystal-induced kidney injury serves as the foundational mechanism for the formation and progression of CaOx stones. Regulated cell death (RCD) such as ferroptosis, necroptosis, and pyroptosis are essential in the pathophysiological process of kidney injury. Ferroptosis, a newly discovered RCD, is characterized by its reliance on iron-mediated lipid peroxidation. Necroptosis, a widely studied programmed necrosis, initiates with a necrotic phenotype that resembles apoptosis in appearance. Pyroptosis, a type of RCD that involves the gasdermin protein, is accompanied by inflammation and immune response. In recent years, increasing amounts of evidence has demonstrated that ferroptosis, necroptosis, and pyroptosis are significant pathophysiological processes involved in CaOx crystal-induced kidney injury. Herein, we summed up the roles of ferroptosis, necroptosis, and pyroptosis in CaOx crystal-induced kidney injury. Furthermore, we delved into the curative potential of ferroptosis, necroptosis, and pyroptosis in CaOx crystal-induced kidney injury.
Collapse
Affiliation(s)
- Kun Tang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Ye
- Department of Geriatric Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu He
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaozhuo Ba
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ding Xia
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ejun Peng
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhiqiang Chen
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhangqun Ye
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoqi Yang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
3
|
Guilbaud E, Naulin F, Meziani L, Deutsch E, Galluzzi L. Impact of radiation therapy on the immunological tumor microenvironment. Cell Chem Biol 2025; 32:678-693. [PMID: 40280118 DOI: 10.1016/j.chembiol.2025.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 02/22/2025] [Accepted: 04/03/2025] [Indexed: 04/29/2025]
Abstract
External beam radiation therapy (RT) is a cornerstone of modern cancer management, being utilized in both curative and palliative settings due to its safety, efficacy, and widespread availability. A primary biological effect of RT is DNA damage, which leads to significant cytostatic and cytotoxic effects. Importantly, malignant cells possess a limited capacity for DNA repair compared to normal cells, and when combined with irradiation techniques that minimize damage to healthy tissues, this creates an advantageous therapeutic window. However, the clinical effectiveness of RT also appears to involve both direct and indirect interactions between RT and non-transformed components of the tumoral ecosystem, particularly immune cells. In this review, we describe the molecular and cellular mechanisms by which irradiated cancer cells modify the immunological tumor microenvironment and how such changes ultimately impact tumor growth.
Collapse
Affiliation(s)
- Emma Guilbaud
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Flavie Naulin
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA; Department of Radiotherapy, Gustave Roussy Cancer Campus, Villejuif, France; INSERM RAMO-IT U1030, Villejuif, France; Faculty of Medicine, University of Paris-Saclay, Le Kremlin, Bicêtre, France
| | - Lydia Meziani
- Department of Radiotherapy, Gustave Roussy Cancer Campus, Villejuif, France; INSERM RAMO-IT U1030, Villejuif, France; Faculty of Medicine, University of Paris-Saclay, Le Kremlin, Bicêtre, France
| | - Eric Deutsch
- Department of Radiotherapy, Gustave Roussy Cancer Campus, Villejuif, France; INSERM RAMO-IT U1030, Villejuif, France; Faculty of Medicine, University of Paris-Saclay, Le Kremlin, Bicêtre, France.
| | - Lorenzo Galluzzi
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA.
| |
Collapse
|
4
|
Wu Q, Qi S, Kang Z, Bai X, Li Z, Cheng J, Dong X. PANoptosis in Sepsis: A Central Role and Emerging Therapeutic Target. J Inflamm Res 2025; 18:6245-6261. [PMID: 40386177 PMCID: PMC12085136 DOI: 10.2147/jir.s513367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 05/04/2025] [Indexed: 05/20/2025] Open
Abstract
The pathogenesis of sepsis is intricately linked to regulated cell death. As a novel form of regulated cell death, PANoptosis plays a critical role in driving the inflammatory response, impairing immune cell function, and contributing to multi-organ dysfunction in sepsis. This review explores the molecular mechanisms underlying PANoptosis and its involvement in sepsis. By activating multiple pathways, PANoptosis promotes the release of inflammatory cytokines, triggering a cytokine storm that disrupts immune cell homeostasis and exacerbates organ damage. Emerging therapeutic strategies targeting PANoptosis, including chemotherapeutic agents and herbal remedies, are showing potential for clinical application. The concept of targeting PANoptosis offers a promising avenue for developing innovative treatments for sepsis.
Collapse
Affiliation(s)
- Qiqi Wu
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Siyuan Qi
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Zhaofeng Kang
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Xiangjun Bai
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Zhanfei Li
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Jing Cheng
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Xijie Dong
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| |
Collapse
|
5
|
Engelen Y, Demuynck R, Ramon J, Breckpot K, De Smedt S, Lajoinie GPR, Braeckmans K, Krysko DV, Lentacker I. Immunogenic cell death as interplay between physical anticancer modalities and immunotherapy. J Control Release 2025:113721. [PMID: 40368187 DOI: 10.1016/j.jconrel.2025.113721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 04/07/2025] [Accepted: 04/09/2025] [Indexed: 05/16/2025]
Abstract
Current cancer treatment strategies in practice nowadays often face limitations in effectiveness due to factors such as resistance, recurrence, or suboptimal outcomes. Traditional approaches like chemotherapy often come with severe systemic side effects due to their non-specific action, prompting the development of more targeted therapies. Among these, physical ablation techniques such as radiotherapy (RT) and focused ultrasound (FUS) have gained attention for their ability to precisely target malignant tissues, reduce physical and mental stress for the patients, and minimize recovery time. These therapies also aim to stimulate the immune system through a process referred to as immunogenic cell death (ICD), enhancing the body's ability to fight cancer, explaining abscopal effects. RT has been the most established of the abovementioned techniques for decades, and will not be included in the review. While initially focused on complete tumor ablation, these techniques are now shifting towards milder, more controlled applications that induce ICD without extensive tissue damage. This review explores how physical ablation therapies can harness ICD to boost anticancer immunity, emphasizing their potential to complement immunotherapies and improve outcomes for cancer patients.
Collapse
Affiliation(s)
- Y Engelen
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; Ghent Research Group on Nanomedicines, Faculty of Pharmaceutical Sciences, Ghent University, 9000 Ghent, Belgium.
| | - R Demuynck
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; Cell Death Investigation and Therapy (CDIT) Laboratory, Anatomy and Embryology Unit, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - J Ramon
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; Biophotonics Research Group, Ghent University, 9000 Ghent, Belgium
| | - K Breckpot
- Laboratory for Molecular and Cellular Therapy, Translational Oncology Research Center, Department of Biomedical Sciences, Faculty of Pharmacy and Medicine, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - S De Smedt
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; Ghent Research Group on Nanomedicines, Faculty of Pharmaceutical Sciences, Ghent University, 9000 Ghent, Belgium
| | - G P R Lajoinie
- Physics of Fluids Group, Technical Medical (TechMed) Center, and Max Planck Center for Complex Fluid Dynamics, University of Twente, Enschede, the Netherlands
| | - K Braeckmans
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; Biophotonics Research Group, Ghent University, 9000 Ghent, Belgium
| | - D V Krysko
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; Cell Death Investigation and Therapy (CDIT) Laboratory, Anatomy and Embryology Unit, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - I Lentacker
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; Ghent Research Group on Nanomedicines, Faculty of Pharmaceutical Sciences, Ghent University, 9000 Ghent, Belgium
| |
Collapse
|
6
|
Kurmangaliyeva S, Baktikulova K, Tkachenko V, Seitkhanova B, Shapambayev N, Rakhimzhanova F, Almagambetova A, Kurmangaliyev K. An Overview of Hexavalent Chromium-Induced Necroptosis, Pyroptosis, and Ferroptosis. Biol Trace Elem Res 2025; 203:2619-2635. [PMID: 39287767 DOI: 10.1007/s12011-024-04376-1] [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: 08/16/2024] [Accepted: 09/10/2024] [Indexed: 09/19/2024]
Abstract
Heavy metals are common environmental industrial pollutants. Due to anthropogenic activity, chromium, especially its hexavalent form [Cr(VI)], is a widespread environmental contaminant that poses a threat to human health. In this review paper, we summarize the currently reported molecular mechanisms involved in chromium toxicity with a focus on the induction of pro-inflammatory non-apoptotic cell death pathways such as necroptosis, pyroptosis, and ferroptosis. The review highlights the ability of chromium to induce necroptosis, pyroptosis, and ferroptosis revealing the signaling pathways involved. Cr(VI) can induce RIPK1/RIPK3-dependent necroptosis both in vitro and in vivo. Chromium toxicity is associated with pyroptotic NLRP3 inflammasome/caspase-1/gasdermin D-dependent secretion of IL-1β and IL-18. Furthermore, this review emphasizes the role of redox imbalance and intracellular iron accumulation in Cr(VI)-induced ferroptosis. Of note, the crosstalk between the investigated lethal subroutines in chromium-induced toxicity is primarily mediated by reactive oxygen species (ROS), which are suggested to act as a rheostat determining the cell death pathway in cells exposed to chromium. The current study provides novel insights into the pro-inflammatory effects of chromium, since necroptosis, pyroptosis, and ferroptosis affect inflammation owing to their immunogenic properties linked primarily with damage-associated molecular patterns. Inhibition of these non-apoptotic lethal subroutines can be considered a therapeutic strategy to reduce the toxicity of heavy metals, including chromium.
Collapse
Affiliation(s)
- Saulesh Kurmangaliyeva
- Department of Microbiology, Virology and Immunology, West Kazakhstan Marat Ospanov Medical University, 68 Maresyev St, Aktobe, Republic of Kazakhstan
| | - Kristina Baktikulova
- Department of Microbiology, Virology and Immunology, West Kazakhstan Marat Ospanov Medical University, 68 Maresyev St, Aktobe, Republic of Kazakhstan.
| | - Viktoriya Tkachenko
- State Institution "Republican Scientific and Practical Centre of Sports, " 8 Narochanskaya St, Minsk, Republic of Belarus
| | - Bibigul Seitkhanova
- Department of Microbiology, Virology and Immunology, South Kazakhstan Medical Academy, Al-Farabi Sq, Shymkent, Republic of Kazakhstan
| | - Nasriddin Shapambayev
- Department of General Practitioner - 1, Khoja Akhmet Yasawi International Kazakh-Turkish University, 7/7 Baitursynov St, Shymkent, Republic of Kazakhstan
| | - Farida Rakhimzhanova
- Department of Microbiology, NCJSC "Semey Medical University, " 103 Abay St, Semey, Republic of Kazakhstan
| | - Altyn Almagambetova
- Department of Phthisiology and Dermatovenerology, West Kazakhstan Marat Ospanov Medical University, 68 Maresyev St, Aktobe, Republic of Kazakhstan
| | - Kairat Kurmangaliyev
- Department of Microbiology, Virology and Immunology, West Kazakhstan Marat Ospanov Medical University, 68 Maresyev St, Aktobe, Republic of Kazakhstan
| |
Collapse
|
7
|
Belaidi AA, Bush AI, Ayton S. Apolipoprotein E in Alzheimer's disease: molecular insights and therapeutic opportunities. Mol Neurodegener 2025; 20:47. [PMID: 40275327 PMCID: PMC12023563 DOI: 10.1186/s13024-025-00843-y] [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: 01/30/2025] [Accepted: 04/14/2025] [Indexed: 04/26/2025] Open
Abstract
Apolipoprotein E (APOE- gene; apoE- protein) is the strongest genetic modulator of late-onset Alzheimer's disease (AD), with its three major isoforms conferring risk for disease ε2 < ε3 < ε4. Emerging protective gene variants, such as APOE Christchurch and the COLBOS variant of REELIN, an alternative target of certain apoE receptors, offer novel insights into resilience against AD. In recent years, the role of apoE has been shown to extend beyond its primary function in lipid transport, influencing multiple biological processes, including amyloid-β (Aβ) aggregation, tau pathology, neuroinflammation, autophagy, cerebrovascular integrity and protection from lipid peroxidation and the resulting ferroptotic cell death. While the detrimental influence of apoE ε4 on these and other processes has been well described, the molecular mechanisms underpinning this disadvantage require further enunciation, particularly to realize therapeutic opportunities related to apoE. This review explores the multifaceted roles of apoE in AD pathogenesis, emphasizing recent discoveries and translational approaches to target apoE-mediated pathways. These findings underscore the potential for apoE-based therapeutic strategies to prevent or mitigate AD in genetically at-risk populations.
Collapse
Affiliation(s)
- Abdel Ali Belaidi
- The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, 3052, Australia.
- The Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, 3052, Australia.
| | - Ashley I Bush
- The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, 3052, Australia
- The Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Scott Ayton
- The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, 3052, Australia
- The Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, 3052, Australia
| |
Collapse
|
8
|
Ge T, Chen Y, Pang L, Shao J, Chen Z. Roles of PANoptosis and related genes in acute liver failure: neoteric insight from bioinformatics analysis and animal experiment verification. J Zhejiang Univ Sci B 2025; 26:353-370. [PMID: 40274384 PMCID: PMC12021540 DOI: 10.1631/jzus.b2300678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 04/23/2024] [Indexed: 04/26/2025]
Abstract
BACKGROUND: PANoptosis has the features of pyroptosis, apoptosis, and necroptosis. Numerous studies have confirmed the diverse roles of various types of cell death in acute liver failure (ALF), but limited attention has been given to the crosstalk among them. In this study, we aimed to explore the role of PANoptosis in ALF and uncover new targets for its prevention or treatment. METHODS: Three ALF-related datasets (GSE14668, GSE62029, and GSE74000) were downloaded from the Gene Expression Omnibus (GEO) database to identify differentially expressed genes (DEGs). Hub genes were identified through intersecting DEGs, genes obtained from weighted gene co-expression network analysis (WGCNA), and genes related to PANoptosis. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), protein‒protein interaction (PPI) analyses and gene set enrichment analysis (GSEA) were performed to determine functional roles. Verification was performed using an ALF mouse model. RESULTS: Our results showed that expression of seven hub genes (B-cell lymphoma-2-modifying factor (BMF), B-cell lymphoma-2-interacting protein 3-like (BNIP3L), Caspase-1 (CASP1), receptor-interacting protein kinase 3 (RIPK3), uveal autoantigen with coiled-coil domains and ankyrin repeats protein (UACA), uncoordinated-5 homolog B receptor (UNC5B), and Z-DNA-binding protein 1 (ZBP1)) was up-regulated in liver samples of patients. However, in the ALF mouse model, the expression of BNIP3L, RIPK3, phosphorylated RIPK3 (P-RIPK3), UACA, and cleaved caspase-1 was up-regulated, while the expression of CASP1 and UNC5B was down-regulated. The expression of ZBP1 and BMF increased only during the development of ALF, and there was no significant change in the end stage. Immunofluorescence of mouse liver tissue showed that macrophages expressed all seven markers. Western blot results showed that pyroptosis, apoptosis, and necroptosis were always involved in lipopolysaccharide (LPS)/ d-galactosamine (d-gal)-induced ALF mice. The ALF cell model showed that bone marrow-derived macrophages (BMDMs) form PANoptosomes after LPS stimulation. CONCLUSIONS: Our results suggest that PANoptosis of macrophages promotes the development of ALF. The seven new ALF biomarkers identified and validated in this study may contribute to further investigation of diagnostic markers or novel therapeutic targets of ALF.
Collapse
Affiliation(s)
- Tiantian Ge
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Yao Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Lantian Pang
- Department of Infectious Diseases, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Junwei Shao
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The Second Affiliated Hospital, Zhejiang University, School of Medicine, Hangzhou 310009, China.
| | - Zhi Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China.
| |
Collapse
|
9
|
McNally MA, Lau LA, Granak S, Hike D, Liu X, Yu X, Donahue RA, Chibnik LB, Ortiz JV, Che A, Chavez-Valdez R, Northington FJ, Staley KJ. Ongoing loss of viable neurons for weeks after mild hypoxia-ischaemia. Brain Commun 2025; 7:fcaf153. [PMID: 40297712 PMCID: PMC12034461 DOI: 10.1093/braincomms/fcaf153] [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: 12/17/2024] [Revised: 03/25/2025] [Accepted: 04/16/2025] [Indexed: 04/30/2025] Open
Abstract
Mild hypoxic-ischaemic encephalopathy is common in neonates, and there are no evidence-based therapies. By school age, 30-40% of those patients experience adverse neurodevelopmental outcomes. The nature and progression of mild injury is poorly understood. We studied the evolution of mild perinatal brain injury using longitudinal two-photon imaging of transgenic fluorescent calcium-sensitive and insensitive proteins to provide a novel readout of neuronal viability and activity at cellular resolution in vitro and in vivo. In vitro, perinatal organotypic hippocampal cultures underwent 15-20 min of oxygen-glucose deprivation. In vivo, mild hypoxia-ischaemia was completed at post-natal day 10 with carotid ligation and 15 min of hypoxia (FiO2, 0.08). Consistent with a mild injury, minimal immediate neuronal death was seen in vitro or in vivo, and there was no volumetric evidence of injury by ex vivo MRI 2.5 weeks after injury (n = 3 pups/group). However, in both the hippocampus and neocortex, these mild injuries resulted in delayed and progressive neuronal loss by the second week after injury compared to controls; measured by fluorophore quenching (n = 6 slices/group in vitro, P < 0.001; n = 8 pups/group in vivo, P < 0.01). Mild hypoxia-ischaemia transiently suppressed cortical network calcium activity in vivo for over 2 h after injury (versus sham, n = 13 pups/group; P < 0.01). No post-injury seizures were seen. By 24 h, network activity fully recovered, and there was no disruption in the development of normal cortical activity for 11 days (n = 8 pups/group). The participation in network activity of individual neurons destined to die in vivo was indistinguishable from those that survived up to 4 days post-injury (n = 8 pups/group). Despite a lack of significant immediate neuronal death and only transient disruptions of network activity, mild perinatal brain injury resulted in a delayed and progressive increase of neuronal death in the hippocampus and neocortex. Neurons that died late were functioning normally for days after injury, suggesting a new pathophysiology of neuronal death after mild injury. Critically, the neurons destined to die late demonstrated multiple biomarkers of viability long after mild injury, suggesting their later death may be modified with neuroprotective interventions.
Collapse
Affiliation(s)
- Melanie A McNally
- Department of Neurology, Harvard Medical School and Massachusetts General Hospital, Boston, MA 02114, USA
| | - Lauren A Lau
- Department of Neurology, Harvard Medical School and Massachusetts General Hospital, Boston, MA 02114, USA
| | - Simon Granak
- Department of Neurology, Harvard Medical School and Massachusetts General Hospital, Boston, MA 02114, USA
| | - David Hike
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Harvard Medical School and Massachusetts General Hospital, Boston, MA 02129, USA
| | - Xiaochen Liu
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Harvard Medical School and Massachusetts General Hospital, Boston, MA 02129, USA
| | - Xin Yu
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Harvard Medical School and Massachusetts General Hospital, Boston, MA 02129, USA
| | - Rachel A Donahue
- Department of Medicine, Biostatics, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Lori B Chibnik
- Department of Neurology, Harvard Medical School and Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA 02115, USA
| | - John V Ortiz
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Alicia Che
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Raul Chavez-Valdez
- Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Frances J Northington
- Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Kevin J Staley
- Department of Neurology, Harvard Medical School and Massachusetts General Hospital, Boston, MA 02114, USA
| |
Collapse
|
10
|
Saadh MJ, Saeed TN, Alfarttoosi KH, Sanghvi G, Roopashree R, Thakur V, Lakshmi L, Kubaev A, Taher WM, Alwan M, Jawad MJ, Al-Nuaimi AMA. Exosomes and MicroRNAs: key modulators of macrophage polarization in sepsis pathophysiology. Eur J Med Res 2025; 30:298. [PMID: 40247413 PMCID: PMC12007276 DOI: 10.1186/s40001-025-02561-z] [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: 01/26/2025] [Accepted: 04/06/2025] [Indexed: 04/19/2025] Open
Abstract
Sepsis is a highly dangerous and complex condition that can result in death. It is characterized by a strong reaction to an infection, causing dysfunction in multiple bodily systems and a high risk of mortality. The transformation of macrophages is a vital stage in the procedure as they possess the capability to interchange between two separate types: M1, which promotes inflammation, and M2, which inhibits inflammation. The choice greatly affects the immune response of the host. This analysis underscores the rapidly expanding roles of exosomes and microRNAs (miRNAs) in regulating the trajectory of macrophage polarization during episodes of sepsis. Exosomes, extremely small extracellular vesicles, facilitate cellular communication by transferring biologically active compounds, including miRNAs, proteins, and lipids. We investigate the impact of changes in exosome production and composition caused by sepsis on macrophage polarization and function. Unique microRNAs present in exosomes play a significant role in controlling crucial signaling pathways that govern the phenotype of macrophages. Through thorough examination of recent progress in this area, we clarify the ways in which miRNAs derived from exosomes can either aggravate or alleviate the inflammatory reactions that occur during sepsis. This revelation not only deepens our comprehension of the underlying mechanisms of sepsis, but it also reveals potential new biomarkers and targets for treatment. This assessment aims to amalgamate diverse research investigations and propose potential avenues for future investigations on the influence that exosomes and miRNAs have on macrophage polarization and the body's response to sepsis. These entities are essential for controlling the host's reaction to sepsis and hold important functions in this mechanism.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan
| | - Tamara Nazar Saeed
- Department of Medical Laboratory Technics, College of Health and Medical Technology, Alnoor University, Mosul, Iraq.
| | | | - Gaurav Sanghvi
- Department of Microbiology, Faculty of Science, Marwadi University Research Center, Marwadi University, Rajkot, Gujarat, 360003, India
| | - R Roopashree
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Vishal Thakur
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India
| | - L Lakshmi
- Department of Nursing, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, India
| | - Aziz Kubaev
- Department of Maxillofacial Surgery, Samarkand State Medical University, 18 Amir Temur Street, 140100, Samarkand, Uzbekistan
| | - Waam Mohammed Taher
- College of Nursing, National University of Science and Technology, Dhi Qar, Iraq
| | - Mariem Alwan
- Pharmacy College, Al-Farahidi University, Baghdad, Iraq
| | | | | |
Collapse
|
11
|
Li Y, Huang H, Gao J, Lu J, Kang G, Ge Y, Jiang W, Cai X, Zhang G, Liu L. A TLK2-mediated calcium-driven cell death pathway links neuronal degeneration to nuclear envelope disruption. Nat Commun 2025; 16:3419. [PMID: 40210858 PMCID: PMC11986041 DOI: 10.1038/s41467-025-58737-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 04/01/2025] [Indexed: 04/12/2025] Open
Abstract
Calcium overload drives neuronal cell death, but its mechanisms remain unclear. Previous studies in Drosophila implicated tousled-like kinase (TLK) in this process. Here, we investigated TLK2, the mammalian homolog, in calcium overload-induced neuronal death. We found that calcium overload enhances TLK2 expression, multimerization, and phosphorylation, increasing its kinase activity. Inhibiting TLK2 via RNA interference or a small-molecule inhibitor reduced neuronal death, while TLK2 overexpression triggered nuclear envelope (NE) rupture, nuclear enlargement, multinucleation, and cell cycle reentry markers. A protein complex involving TLK2, dynein light chain LC8, and myosin IIA was linked to NE disruption. In mouse models of glaucoma, TLK2 contributed to retinal ganglion cell degeneration, connecting calcium overload to neurodegeneration. We propose "CaToptosis" (Calcium-induced Tousled-like kinase-mediated cell death) as a distinct neuronal death pathway.
Collapse
Affiliation(s)
- Yajie Li
- Department of Biochemistry and Molecular Biology School of Basic Medicine, Capital Medical University, Youanmen, Beijing, 100069, China
- Oujiang Laboratory, School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Huaiyuan Huang
- Department of Biochemistry and Molecular Biology School of Basic Medicine, Capital Medical University, Youanmen, Beijing, 100069, China
| | - Jingwen Gao
- Department of Biochemistry and Molecular Biology School of Basic Medicine, Capital Medical University, Youanmen, Beijing, 100069, China
| | - Jinhong Lu
- Department of Biochemistry and Molecular Biology School of Basic Medicine, Capital Medical University, Youanmen, Beijing, 100069, China
| | - Guifeng Kang
- School of Pharmaceutical Sciences, Capital Medical University, Youanmen, Beijing, 100069, China
| | - Yipeng Ge
- Department of Cardiovascular Surgery, Beijing Anzhen Hospital, Capital Medical University, Anzhen Road 2#, Chaoyang District, Beijing, 10029, China
| | - Wencan Jiang
- Department of Clinical Diagnosis, Laboratory of Beijing Tiantan Hospital and Capital Medical University, Beijing, 100070, China
- Laboratory diagnosis platform for nervous system infectious diseases, Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100070, China
| | - Xiang Cai
- Oujiang Laboratory, School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Guojun Zhang
- Department of Clinical Diagnosis, Laboratory of Beijing Tiantan Hospital and Capital Medical University, Beijing, 100070, China.
- Laboratory diagnosis platform for nervous system infectious diseases, Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100070, China.
| | - Lei Liu
- Department of Biochemistry and Molecular Biology School of Basic Medicine, Capital Medical University, Youanmen, Beijing, 100069, China.
- Laboratory diagnosis platform for nervous system infectious diseases, Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100070, China.
| |
Collapse
|
12
|
Prabhune NM, Ameen B, Prabhu S. Therapeutic potential of synthetic and natural iron chelators against ferroptosis. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:3527-3555. [PMID: 39601820 DOI: 10.1007/s00210-024-03640-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 11/15/2024] [Indexed: 11/29/2024]
Abstract
Ferroptosis, a regulated form of cell death, is characterized by iron accumulation that results in the production of reactive oxygen species. This further causes lipid peroxidation and damage to the cellular components, eventually culminating into oxidative stress. Recent studies have highlighted the pivotal role of ferroptosis in the pathophysiological development and progression of various diseases such as β-thalassemia, hemochromatosis, and neurodegenerative disorders like AD and PD. Extensive efforts are in progress to understand the molecular mechanisms governing the role of ferroptosis in these conditions, and chelation therapy stands out as a potential approach to mitigate ferroptosis and its related implications in their development. There are currently both synthetic and natural iron chelators that are being researched for their potential as ferroptosis inhibitors. While synthetic chelators are relatively well-established and studied, their short plasma half-life and toxic side effects necessitate the exploration and identification of natural products that can act as efficient and safe iron chelators. In this review, we comprehensively discuss both synthetic and natural iron chelators as potential therapeutic strategies against ferroptosis-induced pathologies.
Collapse
Affiliation(s)
- Nupura Manish Prabhune
- Department of Cellular and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Bilal Ameen
- Department of Cellular and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Sudharshan Prabhu
- Department of Cellular and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| |
Collapse
|
13
|
Su W, Wang H, Pan J, Zhou Q. Advances in Sonodynamic Therapy: Focus on Ferroptosis. J Med Chem 2025; 68:5976-5992. [PMID: 40063557 DOI: 10.1021/acs.jmedchem.4c02603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2025]
Abstract
Ferroptosis is a nonapoptotic form of cell death discovered in 2012. Noninvasive treatments regulating ferroptosis are important for a wide range of diseases. Among the noninvasive treatments, sonodynamic therapy (SDT) has become promising due to its strong tissue penetration and few side effects. In recent years, targeted drug delivery platforms constructed on the basis of SDT have provided an efficient delivery mode for the regulation of ferroptosis. Based on the latest research reports, this Perspective introduces the basic mechanism of SDT and the influencing factors of therapeutic effects, elucidates the significance of ferroptosis-targeted SDT, and summarizes the recent studies on ferroptosis-targeted SDT through different pathways. We also present innovative studies of composite ultrasound-responsive drug delivery platforms. Finally, a brief summary and outlook based on current ferroptosis-targeted SDT are presented.
Collapse
Affiliation(s)
- Wendi Su
- Echo Lab, Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Hao Wang
- Echo Lab, Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Juhong Pan
- Echo Lab, Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Qing Zhou
- Echo Lab, Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan 430060, China
| |
Collapse
|
14
|
Zhou L, Fan S, Zhang W, Gong Z, Wang D, Tang D. The battle within: cell death by phagocytosis in cancer. Clin Transl Oncol 2025; 27:871-886. [PMID: 39167272 DOI: 10.1007/s12094-024-03650-x] [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: 06/06/2024] [Accepted: 07/25/2024] [Indexed: 08/23/2024]
Abstract
The process by which living cells are phagocytosed and digested to death is called cell death by phagocytosis, a term that has just recently been generalized and redefined. It is characterized by the phagocytosis of living cells and the cessation of cell death by phagocytosis. Phagocytosis of dead cells is a widely discussed issue in cancer, cell death by phagocytosis can stimulate phagocytosis and stimulate adaptive immunity in tumors, and at the same time, do not-eat-me signaling is an important site for cancer cells to evade recognition by phagocytes. Therefore, we discuss in this review cell death by phagocytosis occurring in cancer tissues and emphasize the difference between this new concept and the phagocytosis of dead tumor cells. Immediately thereafter, we describe the mechanisms by which cell death by phagocytosis occurs and how tumors escape phagocytosis. Finally, we summarize the potential clinical uses of cell death by phagocytosis in tumor therapy and strive to provide ideas for tumor therapy.
Collapse
Affiliation(s)
- Lujia Zhou
- Department of Clinical Medicine, Medical College, Yangzhou University, Yangzhou, 225001, Jiangsu, China
| | - Shiying Fan
- Department of Clinical Medicine, Medical College, Yangzhou University, Yangzhou, 225001, Jiangsu, China
| | - Wenjie Zhang
- School of Medicine, Chongqing University, Chongqing, 400030, China
| | - Zhiyuan Gong
- Department of Clinical Medicine, Medical College, Yangzhou University, Yangzhou, 225001, Jiangsu, China
| | - Daorong Wang
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, 225000, China
| | - Dong Tang
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, 225000, China.
| |
Collapse
|
15
|
Jhetam Z, Martins-Furness C, Slabber C, Munro OQ, Nel M, Harmse L. Copper complexes induce haem oxygenase-1 (HMOX1) and cause apoptotic cell death in pancreatic cancer cells. J Inorg Biochem 2025; 264:112815. [PMID: 39740375 DOI: 10.1016/j.jinorgbio.2024.112815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 12/14/2024] [Accepted: 12/15/2024] [Indexed: 01/02/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC), the most common pancreatic malignancy, has a dismal 5-year survival rate, making palliative chemotherapy the only treatment option. Targeted therapy has limited efficacy in PDAC, underscoring the need for novel therapeutic approaches. The inducible stress-response protein, haem oxygenase-1 (HMOX1), has been implicated in treatment failure in PDAC. Copper coordination complexes have shown promise as anticancer agents against various cancers, and are associated with apoptotic cell death. The different ligands to which copper is complexed, determine the specificity and efficacy of each complex. Three different classes of copper complexes were evaluated for anti-cancer activity against AsPC-1 and MIA PaCa-2 pancreatic cancer cell lines. A copper-phenanthroline-theophylline complex (CuPhTh2), a copper-8-aminoquinoline-naphthyl complex (Cu8AqN), and two copper-aromatic-isoindoline complexes (CuAIsI) were effective inhibitors of cell proliferation with clinically relevant IC50 values below 5 μM. The copper complexes caused reactive oxygen species (ROS) formation, promoted annexin-V binding, disrupted the mitochondrial membrane potential (MMP) and activated caspase-9 and caspase-3/7, confirming apoptotic cell death. Expression of nuclear HMOX1 was increased in both cell lines, with the CuPhTh2 complex being the most active. Inhibition of HMOX1 activity significantly decreased the IC50 values of these copper complexes suggesting that HMOX1 inhibition may alter treatment outcomes in PDAC.
Collapse
Affiliation(s)
- Zakeeya Jhetam
- Division of Pharmacology, Department of Pharmacy and Pharmacology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown 2193, South Africa
| | - Carla Martins-Furness
- Division of Pharmacology, Department of Pharmacy and Pharmacology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown 2193, South Africa
| | - Cathy Slabber
- Molecular Sciences Institute, School of Chemistry, University of the Witwatersrand, 1 Jan Smut Ave, Braamfontein, Johannesburg 2017, South Africa
| | - Orde Q Munro
- Molecular Sciences Institute, School of Chemistry, University of the Witwatersrand, 1 Jan Smut Ave, Braamfontein, Johannesburg 2017, South Africa; School of Chemistry, University of Leeds, Woodhouse Lane, Leeds LS2 9JT, UK
| | - Marietha Nel
- Dept of Surgery, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown 2193, South Africa
| | - Leonie Harmse
- Division of Pharmacology, Department of Pharmacy and Pharmacology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown 2193, South Africa.
| |
Collapse
|
16
|
Sant’Angelo D, Descamps G, Lecomte V, Stanicki D, Penninckx S, Dragan T, Van Gestel D, Laurent S, Journe F. Therapeutic Approaches with Iron Oxide Nanoparticles to Induce Ferroptosis and Overcome Radioresistance in Cancers. Pharmaceuticals (Basel) 2025; 18:325. [PMID: 40143107 PMCID: PMC11945075 DOI: 10.3390/ph18030325] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 02/15/2025] [Accepted: 02/18/2025] [Indexed: 03/28/2025] Open
Abstract
The emergence of nanotechnology in medicine, particularly using iron oxide nanoparticles (IONPs), may impact cancer treatment strategies. IONPs exhibit unique properties, such as superparamagnetism, biocompatibility, and ease of surface modification, making them ideal candidates for imaging, and therapeutic interventions. Their application in targeted drug delivery, especially with traditional chemotherapeutic agents like cisplatin, has shown potential in overcoming limitations such as low bioavailability and systemic toxicity of chemotherapies. Moreover, IONPs, by releasing iron ions, can induce ferroptosis, a form of iron-dependent cell death, which offers a promising pathway to reverse radio- and chemoresistance in cancer therapy. In particular, IONPs demonstrate significant potential as radiosensitisers, enhancing the effects of radiotherapy by promoting reactive oxygen species (ROS) generation, lipid peroxidation, and modulating the tumour microenvironment to stimulate antitumour immune responses. This review explores the multifunctional roles of IONPs in radiosensitisation through ferroptosis induction, highlighting their promise in advancing treatment for head and neck cancers. Additional research is crucial to fully addressing their potential in clinical settings, offering a novel approach to personalised cancer treatment.
Collapse
Affiliation(s)
- Dorianne Sant’Angelo
- Department of Human Biology and Toxicology (Cancer Research Unit), Faculty of Medicine, Research Institute for Health Sciences and Technology, University of Mons (UMONS), 7000 Mons, Belgium
- Laboratory of Clinical and Experimental Oncology (LOCE), Institute Jules Bordet, HUB, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| | - Géraldine Descamps
- Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons (UMONS), 7000 Mons, Belgium; (G.D.); (V.L.); (D.S.); (S.L.)
| | - Valentin Lecomte
- Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons (UMONS), 7000 Mons, Belgium; (G.D.); (V.L.); (D.S.); (S.L.)
| | - Dimitri Stanicki
- Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons (UMONS), 7000 Mons, Belgium; (G.D.); (V.L.); (D.S.); (S.L.)
| | - Sébastien Penninckx
- Department of Medical Physics, Institut Jules Bordet, HUB, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium;
- Department of Radiotherapy, Institute Jules Bordet, Hopital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium; (T.D.); (D.V.G.)
| | - Tatiana Dragan
- Department of Radiotherapy, Institute Jules Bordet, Hopital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium; (T.D.); (D.V.G.)
| | - Dirk Van Gestel
- Department of Radiotherapy, Institute Jules Bordet, Hopital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium; (T.D.); (D.V.G.)
| | - Sophie Laurent
- Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons (UMONS), 7000 Mons, Belgium; (G.D.); (V.L.); (D.S.); (S.L.)
| | - Fabrice Journe
- Department of Human Biology and Toxicology (Cancer Research Unit), Faculty of Medicine, Research Institute for Health Sciences and Technology, University of Mons (UMONS), 7000 Mons, Belgium
- Laboratory of Clinical and Experimental Oncology (LOCE), Institute Jules Bordet, HUB, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| |
Collapse
|
17
|
Deng RM, Huang G, Wang T, Zhou J. Regulated programmed cell death in sepsis associated acute lung injury: From pathogenesis to therapy. Int Immunopharmacol 2025; 148:114111. [PMID: 39832461 DOI: 10.1016/j.intimp.2025.114111] [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: 09/09/2024] [Revised: 11/28/2024] [Accepted: 01/15/2025] [Indexed: 01/22/2025]
Abstract
Sepsis associated acute lung injury (SALI) is a common complication in patients with severe sepsis and a disease with high morbidity and mortality in ICU patients. The main mechanism of SALI is pulmonary hypoperfusion due to hypotension and shock caused by sepsis, which leads to ischemic necrosis of alveolar endothelial cells and eventually lung failure. At present, SALI therapy mainly includes antibiotic therapy, fluid resuscitation, transfusion products and vasoactive drugs, but these strategies are not satisfactory. Therefore, focusing on the role of different cell death patterns in SALI may help in the search for effective treatments. Understanding the molecular mechanisms of SALI and identifying pathways that inhibit lung cell death are critical to developing effective drug therapies to prevent the progression of SALI. Cell death is controlled by programmed cell death (PCD) pathways, including apoptosis, necroptosis, ferroptosis, pyroptosis and autophagy. There is growing evidence that PCD plays an important role in the pathogenesis of SALI, and inhibitors of various types of PCD represent a promising therapeutic strategy. Therefore, understanding the role and mechanism of PCD in SALI is conducive to our understanding of its pathological mechanism, and is of great significance for the treatment of SALI. In this article, we discuss recent advances in the role of PCD in SALI, show how different signaling pathways (such as NF-κB, PI3K/Akt, mTOR, and Nrf2) regulate PCD to regulate SALI development, and discuss the associations between various types of PCD. The aim is to explore the molecular mechanism behind SALI and to find new targets for SALI therapy.
Collapse
Affiliation(s)
- Rui-Ming Deng
- Department of Anesthesiology, Ganzhou People's Hospital, 16 Meiguan Avenue, Ganzhou, Jiangxi Province 341000, PR China.
| | - Guiming Huang
- Department of Anesthesiology, Ganzhou People's Hospital, 16 Meiguan Avenue, Ganzhou, Jiangxi Province 341000, PR China
| | - Tingting Wang
- Department of Anaesthesia, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, 200137, PR China
| | - Juan Zhou
- Department of Thyroid and Breast Surgery, Ganzhou People's Hospital, 16 Meiguan Avenue, Ganzhou, Jiangxi Province 341000, PR China.
| |
Collapse
|
18
|
Bekeschus S, Roessler K, Kepp O, Freund E. Gas Plasma Technology and Immunogenic Cell Death: Implications for Chordoma Treatment. Cancers (Basel) 2025; 17:681. [PMID: 40002275 PMCID: PMC11852646 DOI: 10.3390/cancers17040681] [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: 12/23/2024] [Revised: 02/12/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025] Open
Abstract
Cancer is the second-leading cause of death in developed societies. Specifically, cancers of the spine and brain come with significant therapeutic challenges. Chordomas are semi-malignant tumors that develop from embryonic residuals at the skull base (clival) or coccyx (sacral). Small tumor fragments can remain in the operation cavities during surgical resection, forming new tumor sites. This requires repeated surgeries or the application of proton-beam radiation and chemotherapy, which often do not lead to complete remission of the tumors. Hence, there is a need for novel therapeutic avenues that are not limited to killing visible tumors but can be applied after surgery to decrease chordoma recurrences. Reactive oxygen species (ROS) generated locally via novel medical gas plasma technologies are one potential approach to address this clinical problem. Previously, broad-spectrum free radicals generated by these cold physical plasmas operated at about body temperature were shown to oxidize cancer cells to the disadvantage of their growth and induce immunogenic cancer cell death (ICD), ultimately promoting anticancer immunity. This review outlines the clinical challenges of chordoma therapy, how medical gas plasma technology could serve as an adjuvant treatment modality, and potential immune-related mechanisms of action that could extend the longevity of gas plasma therapy beyond its acute local tissue effects.
Collapse
Affiliation(s)
- Sander Bekeschus
- ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP), 17489 Greifswald, Germany
- Department of Dermatology and Venerology, Rostock University Medical Center, 18057 Rostock, Germany
| | - Karl Roessler
- Department of Neurosurgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Oliver Kepp
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, 75005 Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, 94800 Villejuif, France
- Faculté de Médecine, Université de Paris Saclay, 94270 Kremlin Bicêtre, France
| | - Eric Freund
- Department of Neurosurgery, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
19
|
Lv Y, Han S, Sun F, Zhang Y, Qu X, Li H, Gu W, Xu Q, Yao S, Chen X, Jin Y. Decreased miR-128-3p in serum exosomes from polycystic ovary syndrome induces ferroptosis in granulosa cells via the p38/JNK/SLC7A11 axis through targeting CSF1. Cell Death Discov 2025; 11:64. [PMID: 39966422 PMCID: PMC11836375 DOI: 10.1038/s41420-025-02331-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 01/10/2025] [Accepted: 01/28/2025] [Indexed: 02/20/2025] Open
Abstract
Increasing evidence suggests that non-coding small RNAs (miRNAs) carried by exosomes (EXOs) play important roles in the development and treatment of polycystic ovary syndrome (PCOS). In this study, we demonstrate that PCOS mouse serum-derived EXOs promote granulosa cells (GCs) ferroptosis, and induce the occurrence of a PCOS-like phenotype in vivo. Notably, EXO miRNA sequencing combined with in vitro gain- and loss-of-function assays revealed that miR-128-3p, which is absent in the serum-derived EXOs of mice with PCOS, regulates lipid peroxidation and GC sensitivity to ferroptosis inducers. Mechanistically, overexpression of CSF1, a direct target of miR-128-3p, reversed the anti-ferroptotic effect of miR-128-3p. Conversely, ferroptosis induction was mitigated in CSF1-downregulated GCs. Furthermore, we demonstrated that miR-128-3p inhibition activates the p38/JNK pathway via CSF1, leading to NRF2-mediated down-regulation of SLC7A11 transcription, which triggers GC iron overload. Moreover, intrathecal miR-128-3p AgomiR injection into mouse ovaries ameliorated PCOS-like characteristics and restored fertility in letrozole-induced mice. The study reveals the pathological mechanisms of PCOS based on circulating EXOs and provides the first evidence of the roles of miR-128-3p and CSF1 in ovarian GCs. This discovery is expected to provide promising therapeutic targets for the treatment of PCOS.
Collapse
Affiliation(s)
- Yanqiu Lv
- Department of Animal Science, College of Agriculture, Yanbian University, Jilin, China
| | - Shengzhong Han
- Department of Animal Science, College of Agriculture, Yanbian University, Jilin, China
- Jilin Provincial Key Laboratory of Transgenic Animal and Embryo Engineering, Yanbian University, Yanji, China
| | - Fuliang Sun
- Department of Animal Science, College of Agriculture, Yanbian University, Jilin, China
| | - Yuyang Zhang
- Department of Animal Science, College of Agriculture, Yanbian University, Jilin, China
| | - Xinglin Qu
- Department of Animal Science, College of Agriculture, Yanbian University, Jilin, China
| | - Hao Li
- Department of Animal Science, College of Agriculture, Yanbian University, Jilin, China
| | - Weiyu Gu
- Department of Animal Science, College of Agriculture, Yanbian University, Jilin, China
| | - Qinglong Xu
- Department of Animal Science, College of Agriculture, Yanbian University, Jilin, China
| | - Shunfa Yao
- Department of Animal Science, College of Agriculture, Yanbian University, Jilin, China
| | - Xuan Chen
- Department of Animal Science, College of Agriculture, Yanbian University, Jilin, China.
| | - Yi Jin
- Department of Animal Science, College of Agriculture, Yanbian University, Jilin, China.
- Jilin Provincial Key Laboratory of Transgenic Animal and Embryo Engineering, Yanbian University, Yanji, China.
| |
Collapse
|
20
|
Erdogan MK, Ozer G. Synergistic Anticancer Effects of Bleomycin and Hesperidin Combination on A549 Non-Small Cell Lung Cancer Cells: Antiproliferative, Apoptotic, Anti-Angiogenic, and Autophagic Insights. Pharmaceuticals (Basel) 2025; 18:254. [PMID: 40006067 PMCID: PMC11859711 DOI: 10.3390/ph18020254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 01/21/2025] [Accepted: 01/26/2025] [Indexed: 02/27/2025] Open
Abstract
Background: This study investigated the combined effects of hesperidin (Hesp), a natural flavonoid, with bleomycin (BL), a commonly used chemotherapy agent, on A549 human lung cancer cells. Methods: Key parameters assessed included cell viability, colony formation, and cell migration, alongside the expression of apoptotic and autophagic markers (p53, p21, Bax, cleaved PARP, and Beclin-1), VEGF levels, and caspase-3 activity. Results: The findings revealed that the Hesp + BL combination significantly amplified antiproliferative, apoptotic, anti-angiogenic, and autophagic effects compared to either treatment alone. The combination therapy effectively inhibited colony formation and cell migration while markedly reducing VEGF levels, indicating strong anti-angiogenic properties. Apoptotic markers such as p53, p21, Bax, and cPARP were significantly upregulated, with caspase-3 activity confirming robust apoptosis induction. Furthermore, autophagy was notably enhanced, as reflected by increased Beclin-1 expression. Conclusions: Synergistic interactions between Hesp and BL, validated through combination index analysis, underscore the therapeutic potential of this combination. These findings underscore the therapeutic potential of the Hesp + BL combination as a promising strategy for lung cancer treatment, meriting further exploration in diverse lung cancer cell lines to validate and expand its applicability in developing novel therapeutic approaches.
Collapse
Affiliation(s)
- Mehmet Kadir Erdogan
- Department of Molecular Biology and Genetics, Faculty of Arts and Sciences, Bingol University, 12000 Bingol, Türkiye
| | - Guleser Ozer
- Department of Biology, Science Institute, Bingol University, 12000 Bingol, Türkiye
| |
Collapse
|
21
|
Halici H, Un H, Celik S, Karakoy Z, Bayraktutan Z, Ozlu C, Cadirci E, Halici Z, Atila A, Mercantepe F. Low-dose Bee Venom as a Potential Therapeutic Agent Against Human Chronic Myeloid Leukaemia Cells. Protein J 2025:10.1007/s10930-025-10251-2. [PMID: 39924635 DOI: 10.1007/s10930-025-10251-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/31/2025] [Indexed: 02/11/2025]
Abstract
Bee venom is secreted by a gland in the abdominal cavity of bees. The venom, especially that of honeybees, contains certain enzymes and peptides that, when administered in high doses, are effective against various diseases. Peptides such as melittin and phospholipase A2 can target various cancer cells. In this study, we investigated the antiproliferative effects of administering low-dose bee venom in K-562 chronic myeloid leukaemia cells. Our proteomic study revealed regional variation of the content of bee venom and high levels of melittin, apamin and secapin, as well as phospholipase A2 and hyaluronidase. In addition, eight new, previously unidentified proteins were identified. The effects of bee venom on cell viability and drug-cell interaction were investigated at 24, 48 and 72 h. According to the MTT (3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide) results, the bee venom decreased K-562 cell viability dose-dependently at all time points. Cell viability decreased 48 and 72 h after bee venom administration but increased in the control group left untreated for 72 h. The inhibition percentages for the highest bee venom concentration (0.4 µM) at 24, 48 and 72 h were 55%, 80% and 92%, respectively. The cell-drug interactions indicated that the cell surfaces, which were smooth and clear before drug application, gradually deteriorated and started to shrink after the application. In conclusion, at increasing doses, bee venom was found to have a strong antiproliferative effect in K-562 chronic myeloid leukaemia cell lines.
Collapse
Affiliation(s)
- Hamza Halici
- Department of Hınıs Vocational Training School, Ataturk University, Erzurum, Turkey
| | - Harun Un
- Department of Biochemistry, Agri Ibrahim Cecen University, Faculty of Pharmacy, Ağrı, Turkey
| | - Saffet Celik
- Trakya University, Technological Research, Development and Design Application and Research Center, Edirne, Turkey
| | - Zeynep Karakoy
- Department of Pharmacology, Erzincan Binali Yıldırım University, Faculty of Pharmacy, Erzincan, Turkey
| | - Zafer Bayraktutan
- Department of Biochemistry, Faculty of Medicine, Ataturk University, Erzurum, Turkey
| | - Can Ozlu
- Department of Internal Medicine, Division of Hematology, University of Health Sciences, Kütahya, Turkey
| | - Elif Cadirci
- Department of Pharmacology, Ataturk University, Faculty of Medicine, Erzurum, Turkey.
| | - Zekai Halici
- Department of Pharmacology, Ataturk University, Faculty of Medicine, Erzurum, Turkey
| | - Alptug Atila
- Department of Analytical Chemistry, Ataturk University Faculty of Pharmacy, Erzurum, Turkey
| | - Filiz Mercantepe
- Department of Internal Medicine, Division of Endocrinology and Metabolic Diseases, Recep Tayyip Erdoğan University Training and Research Hospital, Rize, Turkey
| |
Collapse
|
22
|
Zhou D, Li P, Yu S, Cui Z, Xu T, Ouyang L. Optimizing extrusion-based 3D bioprinting of plant cells with enhanced resolution and cell viability. Biofabrication 2025; 17:025008. [PMID: 39847862 DOI: 10.1088/1758-5090/adada1] [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: 08/05/2024] [Accepted: 01/23/2025] [Indexed: 01/25/2025]
Abstract
3D bioprinting of plant cells has emerged as a promising technology for plant cell immobilization and related applications. Despite the numerous progress in mammalian cell printing, the bioprinting of plant cells is still in its infancy and needs further investigation. Here, we present a systematic study on optimizing the 3D bioprinting of plant cells, using carrots as an example, towards enhanced resolution and cell viability. We mainly investigated the effects of cell cluster forms and nozzle size on the rheological, extrusion, and printability properties of plant cell bioinks, as well as on the resultant cell viability and growth. We found that when the printing nozzle is larger than 85% of the cell clusters embedded in the bioink, smooth extrusion and good printability can be achieved together with considerable cell viability and long-term growth. Specifically, we optimized a bioink composited with suspension-cultured carrot cells, which exhibited better uniformity, smoother extrusion, and higher cell viability over 1 month culture compared to those with the regular callus or fragmented callus. This work provides a practical guideline for optimizing plant cell bioprinting from the bioink development to the printing outcome assessment. It highlights the importance of selecting a matched nozzle and cell cluster and might provide insights for a better understanding and exploitation of plant cell bioprinting.
Collapse
Affiliation(s)
- Dezhi Zhou
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, People's Republic of China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing 100084, People's Republic of China
- Biomanufacturing and Engineering Living Systems Innovation International Talents Base (111 Base), Beijing 100084, People's Republic of China
| | - Peixi Li
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, People's Republic of China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing 100084, People's Republic of China
- Biomanufacturing and Engineering Living Systems Innovation International Talents Base (111 Base), Beijing 100084, People's Republic of China
| | - Shuang Yu
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, People's Republic of China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing 100084, People's Republic of China
- Biomanufacturing and Engineering Living Systems Innovation International Talents Base (111 Base), Beijing 100084, People's Republic of China
| | - Zhenhua Cui
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, People's Republic of China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing 100084, People's Republic of China
- Biomanufacturing and Engineering Living Systems Innovation International Talents Base (111 Base), Beijing 100084, People's Republic of China
| | - Tao Xu
- Research Institute of Tsinghua University in Shenzhen, Shenzhen 518057, People's Republic of China
| | - Liliang Ouyang
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, People's Republic of China
- Tsinghua University, Beijing 100084, People's Republic of China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing 100084, People's Republic of China
- Biomanufacturing and Engineering Living Systems Innovation International Talents Base (111 Base), Beijing 100084, People's Republic of China
| |
Collapse
|
23
|
Zhu L, Liu Y, Wang K, Wang N. Regulated cell death in acute myocardial infarction: Molecular mechanisms and therapeutic implications. Ageing Res Rev 2025; 104:102629. [PMID: 39644925 DOI: 10.1016/j.arr.2024.102629] [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: 09/09/2024] [Revised: 11/22/2024] [Accepted: 12/03/2024] [Indexed: 12/09/2024]
Abstract
Acute myocardial infarction (AMI), primarily caused by coronary atherosclerosis, initiates a series of events that culminate in the obstruction of coronary arteries, resulting in severe myocardial ischemia and hypoxia. The subsequent myocardial ischemia/reperfusion (I/R) injury further aggravates cardiac damage, leading to a decline in heart function and the risk of life-threatening complications. The complex interplay of multiple regulated cell death (RCD) pathways plays a pivotal role in the pathogenesis of AMI. Each RCD pathway is orchestrated by a symphony of molecular regulatory mechanisms, highlighting the dynamic changes and critical roles of key effector molecules. Strategic disruption or inhibition of these molecular targets offers a tantalizing prospect for mitigating or even averting the onset of RCD, thereby limiting the extensive loss of cardiomyocytes and the progression of detrimental myocardial fibrosis. This review systematically summarizes the mechanisms underlying various forms of RCD, provides an in-depth exploration of the pathogenesis of AMI through the lens of RCD, and highlights a range of promising therapeutic targets that hold the potential to revolutionize the management of AMI.
Collapse
Affiliation(s)
- Lili Zhu
- Department of Pathology, Hunan Cancer Hospital, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yiyang Liu
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China; Key Laboratory of Sepsis Translational Medicine of Hunan, Central South University, Changsha, Hunan, China; National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan, China
| | - Kangkai Wang
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China; Key Laboratory of Sepsis Translational Medicine of Hunan, Central South University, Changsha, Hunan, China; National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan, China
| | - Nian Wang
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China; Key Laboratory of Sepsis Translational Medicine of Hunan, Central South University, Changsha, Hunan, China; National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan, China.
| |
Collapse
|
24
|
Zhang Y, Yi S, Luan M. Advances in non-apoptotic regulated cell death: implications for malignant tumor treatment. Front Oncol 2025; 15:1519119. [PMID: 39949740 PMCID: PMC11821507 DOI: 10.3389/fonc.2025.1519119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 01/15/2025] [Indexed: 02/16/2025] Open
Abstract
Cell death mechanisms are broadly classified into accidental cell death (ACD) and regulated cell death (RCD). ACD such as necrosis, is an uncontrolled, accidental process, while RCD is tightly regulated by specific signaling pathways and molecular mechanisms. Tumor cells are characterized by their ability to evade cell death and sustain uncontrolled proliferation. The failure of programmed cell death is a key contributor to tumor initiation, progression, and resistance to cancer therapies. Traditionally, research has focused primarily on apoptosis as the dominant form of RCD in cancer. However, emerging evidence highlights the importance of other non-apoptotic forms of RCD, such as pyroptosis, ferroptosis, necroptosis, and parthanatos, in tumorigenesis and treatment response. These pathways are gaining attention for their potential roles in overcoming therapy resistance. In this review, we will discuss the recent advances in the study of non-apoptotic cell death pathways in malignant tumors and explore their therapeutic implications, offering insights into new targets for cancer treatment strategies.
Collapse
Affiliation(s)
- Yizheng Zhang
- Department of Pathology and Neuropathology, University Hospital and Comprehensive Cancer Center Tübingen, Tübingen, Germany
| | - Shiqi Yi
- Department of Obstetrics and Gynecology, West China Second Hospital, Sichuan University, Chengdu, China
| | - Mingyuan Luan
- Department of Pathology and Neuropathology, University Hospital and Comprehensive Cancer Center Tübingen, Tübingen, Germany
| |
Collapse
|
25
|
Markitantova Y, Simirskii V. Retinal Pigment Epithelium Under Oxidative Stress: Chaperoning Autophagy and Beyond. Int J Mol Sci 2025; 26:1193. [PMID: 39940964 PMCID: PMC11818496 DOI: 10.3390/ijms26031193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/24/2025] [Accepted: 01/28/2025] [Indexed: 02/16/2025] Open
Abstract
The structural and functional integrity of the retinal pigment epithelium (RPE) plays a key role in the normal functioning of the visual system. RPE cells are characterized by an efficient system of photoreceptor outer segment phagocytosis, high metabolic activity, and risk of oxidative damage. RPE dysfunction is a common pathological feature in various retinal diseases. Dysregulation of RPE cell proteostasis and redox homeostasis is accompanied by increased reactive oxygen species generation during the impairment of phagocytosis, lysosomal and mitochondrial failure, and an accumulation of waste lipidic and protein aggregates. They are the inducers of RPE dysfunction and can trigger specific pathways of cell death. Autophagy serves as important mechanism in the endogenous defense system, controlling RPE homeostasis and survival under normal conditions and cellular responses under stress conditions through the degradation of intracellular components. Impairment of the autophagy process itself can result in cell death. In this review, we summarize the classical types of oxidative stress-induced autophagy in the RPE with an emphasis on autophagy mediated by molecular chaperones. Heat shock proteins, which represent hubs connecting the life supporting pathways of RPE cells, play a special role in these mechanisms. Regulation of oxidative stress-counteracting autophagy is an essential strategy for protecting the RPE against pathological damage when preventing retinal degenerative disease progression.
Collapse
Affiliation(s)
- Yuliya Markitantova
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 119334 Moscow, Russia;
| | | |
Collapse
|
26
|
Song K, Wu Y, Tan S. Caspases in PANoptosis. Curr Res Transl Med 2025; 73:103502. [PMID: 39985853 DOI: 10.1016/j.retram.2025.103502] [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: 06/19/2024] [Revised: 02/13/2025] [Accepted: 02/19/2025] [Indexed: 02/24/2025]
Abstract
Recent studies prove that the three well-established cell death pathways-pyroptosis, apoptosis, and necroptosis-are not isolated but rather engage in extensive crosstalk. PANoptosis, a newly identified pathway of inflammatory regulated cell death (RCD), integrates characteristics of apoptosis, pyroptosis, and necroptosis. Caspases are a family of conserved cysteine proteases that play critical roles in pyroptosis, apoptosis, and necroptosis. Similarly, caspases also play a role in PANoptosis. In this paper, we review the molecular mechanisms of these three RCDs and the crosstalk between them. We also delineate the discovery of PANoptosis and its association with disease. Furthermore, we discuss the caspase function in PANoptosis, mainly focusing on caspase-6 and caspase-8 molecules. This review describes the key molecules, especially caspases, in the context of PANoptosis research, aiming to provide a foundation for targeted interventions in PANoptosis-associated diseases.
Collapse
Affiliation(s)
- Kaiyuan Song
- Department of Pathophysiology, Xiangya School of Basic Medicine Science, Central South University, Changsha, PR China; Sepsis Translational Medicine Key Laboratory of Hunan Province, Central South University, Changsha, PR China; National Medicine Functional Experimental Teaching Center, Central South University, Changsha, PR China
| | - Yongbin Wu
- Department of Pathophysiology, Xiangya School of Basic Medicine Science, Central South University, Changsha, PR China; Sepsis Translational Medicine Key Laboratory of Hunan Province, Central South University, Changsha, PR China; National Medicine Functional Experimental Teaching Center, Central South University, Changsha, PR China
| | - Sipin Tan
- Department of Pathophysiology, Xiangya School of Basic Medicine Science, Central South University, Changsha, PR China; Sepsis Translational Medicine Key Laboratory of Hunan Province, Central South University, Changsha, PR China; National Medicine Functional Experimental Teaching Center, Central South University, Changsha, PR China.
| |
Collapse
|
27
|
Catanzaro E, Beltrán-Visiedo M, Galluzzi L, Krysko DV. Immunogenicity of cell death and cancer immunotherapy with immune checkpoint inhibitors. Cell Mol Immunol 2025; 22:24-39. [PMID: 39653769 PMCID: PMC11685666 DOI: 10.1038/s41423-024-01245-8] [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] [Received: 10/17/2024] [Accepted: 11/18/2024] [Indexed: 12/13/2024] Open
Abstract
While immunotherapy with immune checkpoint inhibitors (ICIs) has revolutionized the clinical management of various malignancies, a large fraction of patients are refractory to ICIs employed as standalone therapeutics, necessitating the development of combinatorial treatment strategies. Immunogenic cell death (ICD) inducers have attracted considerable interest as combinatorial partners for ICIs, at least in part owing to their ability to initiate a tumor-targeting adaptive immune response. However, compared with either approach alone, combinatorial regimens involving ICD inducers and ICIs have not always shown superior clinical activity. Here, we discuss accumulating evidence on the therapeutic interactions between ICD inducers and immunotherapy with ICIs in oncological settings, identify key factors that may explain discrepancies between preclinical and clinical findings, and propose strategies that address existing challenges to increase the efficacy of these combinations in patients with cancer.
Collapse
Affiliation(s)
- Elena Catanzaro
- Cell Death Investigation and Therapy (CDIT) Laboratory, Anatomy and Embryology Unit, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent, Ghent University, Ghent, Belgium
| | - Manuel Beltrán-Visiedo
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Lorenzo Galluzzi
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA.
| | - Dmitri V Krysko
- Cell Death Investigation and Therapy (CDIT) Laboratory, Anatomy and Embryology Unit, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium.
- Cancer Research Institute Ghent, Ghent University, Ghent, Belgium.
| |
Collapse
|
28
|
Madorran E, Ambrož M, Knez J, Sobočan M. An Overview of the Current State of Cell Viability Assessment Methods Using OECD Classification. Int J Mol Sci 2024; 26:220. [PMID: 39796074 PMCID: PMC11719996 DOI: 10.3390/ijms26010220] [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: 12/03/2024] [Revised: 12/27/2024] [Accepted: 12/29/2024] [Indexed: 01/13/2025] Open
Abstract
Over the past century, numerous methods for assessing cell viability have been developed, and there are many different ways to categorize these methods accordingly. We have chosen to use the Organisation for Economic Co-operation and Development (OECD) classification due to its regulatory importance. The OECD categorizes these methods into four groups: non-invasive cell structure damage, invasive cell structure damage, cell growth, and cellular metabolism. Despite the variety of cell viability methods available, they can all be categorized within these four groups, except for two novel methods based on the cell membrane potential, which we added to the list. Each method operates on different principles and has its own advantages and disadvantages, making it essential for researchers to choose the method that best fits their experimental design. This review aims to assist researchers in making this decision by describing these methods regarding their potential use and providing direct references to the cell viability assessment methods. Additionally, we use the OECD classification to facilitate potential regulatory use and to highlight the need for adding a new category to their list.
Collapse
Affiliation(s)
- Eneko Madorran
- Faculty of Medicine, Institute of Anatomy, Histology and Embryology, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia
- Faculty of Medicine, Institute of Translational and Clinical Research, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia; (M.A.); (M.S.)
| | - Miha Ambrož
- Faculty of Medicine, Institute of Translational and Clinical Research, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia; (M.A.); (M.S.)
| | - Jure Knez
- Department for Gynaecologic Oncology and Oncology of the Breast, University Division for Gynaecology and Perinatology, Ljubljanska ulica 5, 2000 Maribor, Slovenia;
| | - Monika Sobočan
- Faculty of Medicine, Institute of Translational and Clinical Research, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia; (M.A.); (M.S.)
- Division of Gynaecology and Perinatology, University Medical Centre Maribor, Ljubljanska ulica 5, 2000 Maribor, Slovenia
| |
Collapse
|
29
|
Ye C, Yang X, Zhu L, Chang G, Hu Y, Wang W. Macrophage-derived exosomal miR-2137 regulates pyroptosis in LPS-induced acute lung injury. Int Immunopharmacol 2024; 143:113549. [PMID: 39550844 DOI: 10.1016/j.intimp.2024.113549] [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: 04/18/2024] [Revised: 10/16/2024] [Accepted: 10/29/2024] [Indexed: 11/19/2024]
Abstract
BACKGROUND Alveolar macrophages (AMs) play a predominant role in acute lung injury (ALI). However, the role of macrophage-derived exosomal miRNAs in lipopolysaccharide (LPS)-induced ALI has not been determined. METHODS We previously reported that exosomes in the bronchoalveolar lavage fluid (BALF) of mice with ALI were derived predominantly from macrophages. Exosomal small RNA sequencing was conducted to identify the miRNA profiles. Exosomes derived from LPS-induced macrophages (LPS-exos) were intravenously administered to C57BL/6J mice, after which lung injury and pyroptosis were assessed. LPS-exos were cultured with alveolar epithelial cells (AECs) to further validate the results of the animal studies. RESULTS LPS-exos promoted lung inflammation and pyroptosis in vivo and in vitro. MiR-2137 was significantly upregulated in both LPS-exos and in MLE-12 cells. LPS-exos reduced cell viability, promoted the expression of LDH and inflammatory cytokines, and exacerbated vacuolization in MLE-12 cells. The administration of miR-2137 mimics and LPS-treated exosomes further strengthened these effects and enhanced pyroptosis mediated by NLRP3, Caspase1, ASC, and GSDMD. MiR-2137 mediated the effects of LPS-exos by targeting Wnt9a in AECs. In addition, the miR-2137 inhibitor markedly decreased the severity of LPS-exo-induced histological lesions, inflammation and pyroptosis in the lung. CONCLUSION Exosomal miR-2137 derived from AMs contributes to LPS-induced ALI by inducing AEC pyroptosis through the targeting of Wnt9a to activate the Wnt signaling pathway. This study revealed that AMs and AECs interact in ALI, providing novel strategies for ALI treatment.
Collapse
Affiliation(s)
- Cong Ye
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
| | - Xiaodong Yang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
| | - Lin Zhu
- Department of Geriatrics, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Guilin Chang
- Department of Geriatrics, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yu Hu
- Department of Geriatrics, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Weixi Wang
- Department of Geriatrics, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
30
|
McNally MA, Lau LA, Granak S, Hike D, Liu X, Yu X, Donahue RA, Chibnik LB, Ortiz JV, Che A, Northington F, Staley K. Ongoing loss of viable neurons for weeks after mild perinatal hypoxia-ischemia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.19.629457. [PMID: 39763962 PMCID: PMC11702593 DOI: 10.1101/2024.12.19.629457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
Mild hypoxic-ischemic encephalopathy is common in neonates with no evidence-based therapies, and 30-40% of patients experience adverse outcomes. The nature and progression of mild injury is poorly understood. Thus, we studied the evolution of mild perinatal brain injury using longitudinal two-photon imaging of transgenic fluorescent proteins as a novel readout of neuronal viability and activity at cellular resolution. In vitro, perinatal murine organotypic hippocampal cultures underwent 15-20 minutes of oxygen-glucose deprivation. In vivo, mild hypoxia-ischemia was completed in post-natal day 10 mouse pups of both sexes with carotid ligation and 15 minutes of hypoxia. Consistent with a mild injury, minimal immediate neuronal death was seen and there was no volumetric evidence of injury by ex vivo MRI 2.5 weeks after injury. In both the hippocampus and neocortex, these mild injuries resulted in a significantly delayed and progressive neuronal loss in the second week after injury, measured by fluorophore quenching. Mild hypoxia-ischemia transiently suppressed cortical network activity followed by normal maturation. No post-injury seizures were seen. The participation in network activity of individual neurons destined to die was indistinguishable from those that survived for 4 days post-injury. In conclusion, our results showed that mild perinatal brain injury resulted in a prolonged increase of neuronal death. Neurons that died late were functioning normally for days after injury, suggesting a new pathophysiology of neuronal death. Critically, the neurons destined to die late demonstrated multiple biomarkers of viability long after mild injury, suggesting their later death may be modified with neuroprotective interventions. SIGNIFICANCE STATEMENT Neonatal encephalopathy due to peripartum hypoxia-ischemia (HI) is a major cause of neonatal mortality and morbidity worldwide. Of these infants, most are categorized as having mild HI. Infants with mild HI have significant long-term disabilities. There are currently no evidence-based therapies, largely because the progression and pathophysiology of mild injury is poorly understood. We have identified, for the first time, that mild perinatal HI results in a delayed and prolonged increase in neuronal death. The cortical and hippocampal neurons that die over a week after injury participate normally in neural network activity and exhibit robust viability for many days after injury, indicating a novel pathophysiology of neuronal death. Clinically, these data suggest an extended therapeutic window for mild perinatal HI.
Collapse
|
31
|
Amantakul A, Amantakul A, Pojchamarnwiputh S, Chattipakorn N, Chattipakorn SC, Sripetchwandee J. Targeting mitochondria and programmed cell death as potential interventions for metastatic castration-resistant prostate cancer. Clin Transl Oncol 2024:10.1007/s12094-024-03784-y. [PMID: 39681803 DOI: 10.1007/s12094-024-03784-y] [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: 08/17/2024] [Accepted: 11/02/2024] [Indexed: 12/18/2024]
Abstract
Prostate cancer is one of the major causes of morbidity and mortality in men worldwide. Most patients with prostate cancer will turn into end-of-life stage when those tumor cells become metastatic castration-resistant prostate cancer (mCRPC). The mCRPC subsequently developed a resistance to androgen signaling. The current regimens for mCRPC therapy are still ineffective. Much evidence from in vitro and in vivo studies explored the roles of therapeutic interventions targeted at the mitochondria and programmed cell death for prostate cancer therapy. The present review will focus on the recent medications which targeted at mitochondria and programmed cell death in mCRPC and the significant findings from each study will be summarized and discussed. Development of therapeutic interventions, particularly at mitochondrial and cytotoxic targets for treatment of mCRPC without inducing cellular toxicity of normal tissues will be considered as the novel therapeutic strategy for mCRPC.
Collapse
Affiliation(s)
- Amonlaya Amantakul
- Department of Diagnostic Radiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Akara Amantakul
- Department of Urology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Suwalee Pojchamarnwiputh
- Department of Diagnostic Radiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Department of Physiology, Neurophysiology Unit, Chiang Mai University, Chiang Mai, 50200, Thailand
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Siriporn Chaisin Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Department of Physiology, Neurophysiology Unit, Chiang Mai University, Chiang Mai, 50200, Thailand
- Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand
| | - Jirapas Sripetchwandee
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Department of Physiology, Neurophysiology Unit, Chiang Mai University, Chiang Mai, 50200, Thailand.
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.
| |
Collapse
|
32
|
Prokopiuk V, Onishchenko A, Pazura Y, Bespalova I, Kökbaş U, Tryfonyuk L, Mateychenko P, Kot K, Kurmangaliyeva S, Kot Y, Yefimova S, Tkachenko A. Nanostructured zinc carbonate hydroxide microflakes: assessing the toxicity against erythrocytes and L929 cells in vitro. NANOTECHNOLOGY 2024; 36:085102. [PMID: 39637441 DOI: 10.1088/1361-6528/ad9aac] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 12/05/2024] [Indexed: 12/07/2024]
Abstract
Nanostructured materials have been suggested to be used as a source of dietary zinc for livestock animals. In this study, we assessed the cytotoxicity of newly synthesized nanostructured zinc carbonate hydroxide (ZnCH) Zn5(CO3)(OH)6microflakes. Cytotoxicity of the microflakes was assessed against murine L929 cell line and rat mature erythrocytes. Viability, motility, cell death pathways, implication of Ca2+, reactive oxygen species and reactive nitrogen species (RNS) signaling, caspases, and alterations of cell membranes following exposure of L929 cells to the microflakes were assessed. To assess hemocompatibility of the Zn-containing microflakes, osmotic fragility and hemolysis assays were performed, as well as multiple eryptosis parameters were evaluated. Our findings indicate a dose-response cytotoxicity of ZnCH microflakes against L929 cells with no toxicity observed for low concentrations (10 mg l-1and below). At high concentrations (25 mg l-1and above), ZnCH microflakes promoted nitrosyl stress, Ca2+- and caspase-dependent apoptosis, and altered lipid order of cell membranes in a dose-dependent manner, evidenced by up to 7-fold elevation of RNS-dependent fluorescence, 2.9-fold enhancement of Fura 2-dependent fluorescence, over 20-fold elevation of caspases-dependent fluorescence (caspase-3, caspase-8, and caspase-9), and up to 4.4-fold increase in the ratiometric index of the NR12S probe. Surprisingly, toxicity to enucleated mature erythrocytes was found to be lower compared to L929 cells. ZnCH microflakes induced eryptosis associated with oxidative stress, nitrosyl stress, Ca2+signaling and recruitment of caspases at 25-50-100 mg l-1. Eryptosis assays were found to be more sensitive than evaluation of hemolysis. Zn5(CO3)(OH)6microflakes show no cytotoxicity at low concentrations indicating their potential as a source of zinc for livestock animals.
Collapse
Affiliation(s)
- Volodymyr Prokopiuk
- Research Institute of Experimental and Clinical Medicine, Kharkiv National Medical University, 4 Nauky ave, 61022 Kharkiv, Ukraine
- Department of Cryobiochemistry, Institute for Problems of Cryobiology and Cryomedicine of the National Academy of Sciences of Ukraine, 23 Pereyaslavskaya st, 61015 Kharkiv, Ukraine
| | - Anatolii Onishchenko
- Department of Cryobiochemistry, Institute for Problems of Cryobiology and Cryomedicine of the National Academy of Sciences of Ukraine, 23 Pereyaslavskaya st, 61015 Kharkiv, Ukraine
| | - Yuliia Pazura
- Department of Nanostructured Materials, Institute for Scintillation Materials of the National Academy of Sciences of Ukraine, 60 Nauky ave, 61072 Kharkiv, Ukraine
| | - Iryna Bespalova
- Department of Nanostructured Materials, Institute for Scintillation Materials of the National Academy of Sciences of Ukraine, 60 Nauky ave, 61072 Kharkiv, Ukraine
| | - Umut Kökbaş
- Medical Biochemistry Department, Nevsehir Haci Bektas Veli University, 2000 Evler Mah. Zübeyde Hanım Cad, 50300 Nevşehir, Turkey
| | - Liliya Tryfonyuk
- Institute of Health, National University of Water and Environmental Engineering, 11 Soborna st, 33028 Rivne, Ukraine
| | - Pavlo Mateychenko
- Institute for Single Crystals, National Academy of Sciences of Ukraine, 60 Nauky ave, 61072 Kharkiv, Ukraine
| | - Kateryna Kot
- Department of Biochemistry, V. N. Karazin Kharkiv National University, 4 Svobody sq, 61022 Kharkiv, Ukraine
| | - Saulesh Kurmangaliyeva
- Department of Microbiology, Virology and Immunology, West Kazakhstan Marat Ospanov Medical University, 68 Maresyev st, 030012 Aktobe, Kazakhstan
| | - Yurii Kot
- Department of Biochemistry, V. N. Karazin Kharkiv National University, 4 Svobody sq, 61022 Kharkiv, Ukraine
| | - Svetlana Yefimova
- Department of Nanostructured Materials, Institute for Scintillation Materials of the National Academy of Sciences of Ukraine, 60 Nauky ave, 61072 Kharkiv, Ukraine
| | - Anton Tkachenko
- BIOCEV, First Faculty of Medicine, Charles University, Průmyslová 595, 25250 Vestec, Czech Republic
| |
Collapse
|
33
|
Martínez-Sanmiguel JJ, Zarate-Triviño D, García-García MP, García-Martín JM, Mayoral Á, Huttel Y, Martínez L, Cholula-Díaz JL. Antitumor activity of bimetallic silver/gold nanoparticles against MCF-7 breast cancer cells. RSC Adv 2024; 14:39102-39111. [PMID: 39664251 PMCID: PMC11629938 DOI: 10.1039/d4ra06227b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 10/27/2024] [Indexed: 12/13/2024] Open
Abstract
Breast cancer poses a global threat with rising incidence and high mortality. Conventional treatments, including chemotherapy, radiation, surgery, and immunotherapy, have side effects, such as resistance issues and adverse effects due to genetic mutations. Meanwhile, noble metal nanoparticles (NPs) synthesized using environmentally friendly methods offer alternative treatments. Bimetallic gold (Au) and silver (Ag) NPs, using natural compounds like starch as stabilizers, enhance biomedical applications, including breast cancer therapies. In this work, the optical properties, stability, and particle size of colloidal bimetallic Ag/Au NPs were analyzed using UV-visible spectroscopy and ζ-potential measurements. The structural properties of the NPs were studied by powder X-ray diffraction (PXRD), while the morphology, chemical composition and particle size were determined using scanning transmission electron microscopy (STEM). The antitumor properties of the Ag/Au NPs were analyzed on human breast cancer cells (MCF-7) using the MTT viability method, reactive oxygen species (ROS) production, and genotoxicity assays. Peripheral blood mononuclear cells (PBMCs) were used as a reference of healthy cells. UV-vis spectroscopy and EDX mapping analysis confirmed the synthesis of bimetallic Ag/Au NPs. Localized surface plasmon resonance (LSPR) absorption bands shifted from 407 nm (Ag) to 524 nm (Au) based on the chemical composition of the NPs. The Ag/Au NPs showed cytocompatibility in PBMCs and a dose-dependent anticancer effect against MCF-7 cancer cells, as well as cell death dependent on ROS production was observed, particularly in NPs with atomic compositions of 50 and 75 at% Ag. This biological activity of the bimetallic NPs was associated with genotoxic damage of 20-24% greater than that observed in the monometallic counterparts. This study demonstrated the synthesis of mono- and bimetallic Ag/Au NPs using a rapid, reproducible and environmentally friendly method, with successful biomedical application against human breast cancer MCF-7 cells.
Collapse
Affiliation(s)
- Juan J Martínez-Sanmiguel
- School of Engineering and Sciences, Tecnológico de Monterrey Av. Eugenio Garza Sada 2501 Monterrey 64849 N.L. Mexico +52 8183582000
| | - Diana Zarate-Triviño
- Laboratorio de Inmunología y Virología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León San Nicolas de los Garza Nuevo León 66455 Mexico
| | - María Paula García-García
- School of Engineering and Sciences, Tecnológico de Monterrey Av. Eugenio Garza Sada 2501 Monterrey 64849 N.L. Mexico +52 8183582000
| | - José Miguel García-Martín
- Instituto de Micro y Nanotecnología (IMN-CNM), CSIC (CEI UAM+CSIC) Isaac Newton 8 28760 Tres Cantos Spain
| | - Álvaro Mayoral
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza Pedro Cerbuna 50009 Zaragoza Spain
| | - Yves Huttel
- Instituto de Ciencia de Materiales de Madrid (ICMM), CSIC Sor Juana Inés de la Cruz 3 28049 Madrid Spain
| | - Lidia Martínez
- Instituto de Ciencia de Materiales de Madrid (ICMM), CSIC Sor Juana Inés de la Cruz 3 28049 Madrid Spain
| | - Jorge L Cholula-Díaz
- School of Engineering and Sciences, Tecnológico de Monterrey Av. Eugenio Garza Sada 2501 Monterrey 64849 N.L. Mexico +52 8183582000
| |
Collapse
|
34
|
Altahla R, Alshorman J, Tao X. Ferroptosis plays a role in osteoarthritis. ALL LIFE 2024; 17. [DOI: 10.1080/26895293.2024.2391292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 07/28/2024] [Indexed: 01/06/2025] Open
Affiliation(s)
- Ruba Altahla
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Jamal Alshorman
- Department of Orthopedics, The Second Affiliated Hospital, Hubei University of Science and Technology, Xiang Ning, People’s Republic of China
| | - Xu Tao
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| |
Collapse
|
35
|
Tkachenko A, Havranek O. Erythronecroptosis: an overview of necroptosis or programmed necrosis in red blood cells. Mol Cell Biochem 2024; 479:3273-3291. [PMID: 38427167 DOI: 10.1007/s11010-024-04948-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 01/20/2024] [Indexed: 03/02/2024]
Abstract
Necroptosis is considered a programmed necrosis that requires receptor-interacting protein kinase 1 (RIPK1), receptor-interacting protein kinase 3 (RIPK3), and pore-forming mixed lineage kinase domain-like protein (MLKL) to trigger a regulated cell membrane lysis. Membrane rupture in necroptosis has been shown to fuel innate immune response due to release of damage-associated molecular patterns (DAMPs). Recently published studies indicate that mature erythrocytes can undergo necroptosis as well. In this review, we provide an outline of multiple cell death modes occurring in erythrocytes, discuss possible immunological aspects of diverse erythrocyte cell deaths, summarize available evidence related to the ability of erythrocytes to undergo necroptosis, outline key involved molecular mechanisms, and discuss the potential implication of erythrocyte necroptosis in the physiology and pathophysiology. Furthermore, we aim to highlight the interplay between necroptosis and eryptosis signaling in erythrocytes, emphasizing specific characteristics of these pathways distinct from their counterparts in nucleated cells. Thus, our review provides a comprehensive summary of the current knowledge of necroptosis in erythrocytes. To reflect critical differences between necroptosis of nucleated cells and necroptosis of erythrocytes, we suggest a term erythronecroptosis for necroptosis of enucleated cells.
Collapse
Affiliation(s)
- Anton Tkachenko
- BIOCEV, First Faculty of Medicine, Charles University, Prumyslova 595, 25250, Vestec, Czech Republic.
| | - Ondrej Havranek
- BIOCEV, First Faculty of Medicine, Charles University, Prumyslova 595, 25250, Vestec, Czech Republic
- First Department of Internal Medicine-Hematology, General University Hospital and First Faculty of Medicine, Charles University, Prague, Czech Republic
| |
Collapse
|
36
|
Cui K, Wang K, Huang Z. Ferroptosis and the tumor microenvironment. J Exp Clin Cancer Res 2024; 43:315. [PMID: 39614322 PMCID: PMC11607824 DOI: 10.1186/s13046-024-03235-0] [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: 09/18/2024] [Accepted: 11/15/2024] [Indexed: 12/01/2024] Open
Abstract
Ferroptosis is a type of regulated cell death characterized by its non-apoptotic, iron-dependent and oxidative nature. Since its discovery in 2012, extensive research has demonstrated its pivotal roles in tumorigenesis, metastasis and cancer therapy. The tumor microenvironment (TME) is a complex ecosystem comprising cancer cells, non-cancer cells, extracellular matrix, metabolites and cytokines. Recent studies have underscored a new paradigm in which non-cancer cells in the TME, such as immune and stromal cells, also play significant roles in regulating tumor progression and therapeutic resistance typically through complicated crosstalk with cancer cells. Notably, this crosstalk in the TME were partially mediated through ferrotopsis-related mechanisms. This review provides a comprehensive and systematic summary of the current findings concerning the roles of ferroptosis in the TME and how ferroptosis-mediated TME reprogramming impacts cancer therapeutic resistance and progression. Additionally, this review outlines various ferroptosis-related therapeutic strategies aimed at targeting the TME.
Collapse
Affiliation(s)
- Kaisa Cui
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Huihe Road 200, Wuxi, Jiangsu, 214062, China
- Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Lihu Avenue 1800, Wuxi, Jiangsu, 214122, China
| | - Kang Wang
- Department of Radiology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Zhaohui Huang
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Huihe Road 200, Wuxi, Jiangsu, 214062, China.
- Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Lihu Avenue 1800, Wuxi, Jiangsu, 214122, China.
| |
Collapse
|
37
|
Zhang W, Jiang L, Tong X, He H, Zheng Y, Xia Z. Sepsis-Induced Endothelial Dysfunction: Permeability and Regulated Cell Death. J Inflamm Res 2024; 17:9953-9973. [PMID: 39628705 PMCID: PMC11612565 DOI: 10.2147/jir.s479926] [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: 05/26/2024] [Accepted: 10/15/2024] [Indexed: 12/06/2024] Open
Abstract
Sepsis is a life-threatening organ dysfunction caused by a dysregulated host response to infection. Endothelial cells (ECs) are an important cell type typically affected in sepsis, resulting in compromised barrier function and various forms of regulated cell death (RCD). However, the precise mechanisms underlying sepsis-induced EC damage remain unclear. This review summarizes the recent research progress on factors and mechanisms that may affect the permeability and RCD of ECs under septic conditions, including glycocalyx, damage-associated molecular patterns, and various forms of RCD in ECs, such as apoptosis, pyroptosis, ferroptosis, and autophagy. This review offers important insights into the underlying mechanisms of endothelial dysfunction in sepsis, aiming to contribute to developing small-molecule targeted clinical therapies.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People’s Republic of China
| | - Luofeng Jiang
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People’s Republic of China
| | - Xirui Tong
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People’s Republic of China
| | - Heng He
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People’s Republic of China
| | - Yongjun Zheng
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People’s Republic of China
| | - Zhaofan Xia
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People’s Republic of China
- Research Unit of Key Techniques for Treatment of burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, Shanghai, 200433, People’s Republic of China
| |
Collapse
|
38
|
Cao Z, Tian K, Ran Y, Zhou H, Zhou L, Ding Y, Tang X. Beclin-1: a therapeutic target at the intersection of autophagy, immunotherapy, and cancer treatment. Front Immunol 2024; 15:1506426. [PMID: 39650649 PMCID: PMC11621085 DOI: 10.3389/fimmu.2024.1506426] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 11/01/2024] [Indexed: 12/11/2024] Open
Abstract
The significant identification of Beclin-1's function in regulating autophagy flow signified a significant progression in our understanding of cellular operations. Beclin-1 acts as a scaffold for forming the PI3KC3 complex, controlling autophagy and cellular trafficking processes in a complicated way. This intricate protein has garnered considerable attention due to its substantial impact on the development of tumors. Strong evidence indicates Beclin-1 plays a critical role in controlling autophagy in various human cancer types and its intricate connection with apoptosis and ferroptosis. The potential of Beclin-1 as a viable target for cancer therapy is highlighted by its associations with key autophagy regulators such as AMPK, mTOR, and ATGs. Beclin-1 controls the growth and dissemination of tumors by autophagy. It also affects how tumors react to therapies such as chemotherapy and radiation therapy. The role of Beclin-1 in autophagy can influence apoptosis, depending on whether it supports cell survival or leads to cell death. Beclin-1 plays a crucial role in ferroptosis by increasing ATG5 levels, which in turn promotes autophagy-triggered ferroptosis. Finally, we analyzed the possible function of Beclin-1 in tumor immunology and drug sensitivity in cancers. In general, Beclin-1 has a significant impact on regulating autophagy, offering various potentials for medical intervention and altering our understanding of cancer biology.
Collapse
Affiliation(s)
- Zhumin Cao
- Department of Hepatobiliary Surgery, The Seventh People’s Hospital of Chongqing, Chongqing, China
| | - Ke Tian
- Department of Hepatobiliary Surgery, The Seventh People’s Hospital of Chongqing, Chongqing, China
| | - Yincheng Ran
- Department of Hepatobiliary Surgery, The Seventh People’s Hospital of Chongqing, Chongqing, China
| | - Haonan Zhou
- Department of Hepatobiliary Surgery, The Seventh People’s Hospital of Chongqing, Chongqing, China
| | - Lei Zhou
- Department of Hepatobiliary Surgery, The Seventh People’s Hospital of Chongqing, Chongqing, China
| | - Yana Ding
- Department of Hepatobiliary Surgery, District Traditional Chinese Medicine Hospital, Chongqing, China
| | - Xiaowei Tang
- Department of Hepatobiliary Surgery, District Traditional Chinese Medicine Hospital, Chongqing, China
| |
Collapse
|
39
|
Galassi C, Chan TA, Vitale I, Galluzzi L. The hallmarks of cancer immune evasion. Cancer Cell 2024; 42:1825-1863. [PMID: 39393356 DOI: 10.1016/j.ccell.2024.09.010] [Citation(s) in RCA: 81] [Impact Index Per Article: 81.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/27/2024] [Accepted: 09/16/2024] [Indexed: 10/13/2024]
Abstract
According to the widely accepted "three Es" model, the host immune system eliminates malignant cell precursors and contains microscopic neoplasms in a dynamic equilibrium, preventing cancer outgrowth until neoplastic cells acquire genetic or epigenetic alterations that enable immune escape. This immunoevasive phenotype originates from various mechanisms that can be classified under a novel "three Cs" conceptual framework: (1) camouflage, which hides cancer cells from immune recognition, (2) coercion, which directly or indirectly interferes with immune effector cells, and (3) cytoprotection, which shields malignant cells from immune cytotoxicity. Blocking the ability of neoplastic cells to evade the host immune system is crucial for increasing the efficacy of modern immunotherapy and conventional therapeutic strategies that ultimately activate anticancer immunosurveillance. Here, we review key hallmarks of cancer immune evasion under the "three Cs" framework and discuss promising strategies targeting such immunoevasive mechanisms.
Collapse
Affiliation(s)
- Claudia Galassi
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Timothy A Chan
- Department of Radiation Oncology, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH, USA; Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA; National Center for Regenerative Medicine, Cleveland, OH, USA; Case Comprehensive Cancer Center, Cleveland, OH, USA
| | - Ilio Vitale
- Italian Institute for Genomic Medicine, c/o IRCSS Candiolo, Torino, Italy; Candiolo Cancer Institute, FPO - IRCCS, Candiolo, Italy.
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, USA; Sandra and Edward Meyer Cancer Center, New York, NY, USA; Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA; Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA.
| |
Collapse
|
40
|
Li L, Du C. Fungal Apoptosis-Related Proteins. Microorganisms 2024; 12:2289. [PMID: 39597678 PMCID: PMC11596484 DOI: 10.3390/microorganisms12112289] [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: 10/17/2024] [Revised: 11/04/2024] [Accepted: 11/09/2024] [Indexed: 11/29/2024] Open
Abstract
Programmed cell death (PCD) plays a crucial role in the development and homeostasis maintenance of multicellular organisms. Apoptosis is a form of PCD that prevents pathological development by eliminating damaged or useless cells. Despite the complexity of fungal apoptosis mechanisms being similar to those of plants and metazoans, fungal apoptosis lacks the core regulatory elements of animal apoptosis. Apoptosis-like PCD in fungi can be triggered by a variety of internal and external factors, participating in biological processes such as growth, development, and stress response. Although the core regulatory elements are not fully understood, apoptosis-inducing factor and metacaspase have been found to be involved. This article summarizes various proteins closely related to fungal apoptosis, such as apoptosis-inducing factor, metacaspase, and inhibitors of apoptosis proteins, as well as their structures and functions. This research provides new strategies and ideas for the development of natural drugs targeting fungal apoptosis and the control of fungal diseases.
Collapse
Affiliation(s)
| | - Chunmei Du
- Engineering Research Center of Agricultural Microbiology Technology, Ministry of Education, Heilongjiang Provincial Key Laboratory of Plant Genetic Engineering and Biological Fermentation Engineering for Cold Region, Key Laboratory of Microbiology, College of Heilongjiang Province, School of Life Sciences, Heilongjiang University, Harbin 150080, China;
| |
Collapse
|
41
|
Kunst C, Tümen D, Ernst M, Tews HC, Müller M, Gülow K. Paraptosis-A Distinct Pathway to Cell Death. Int J Mol Sci 2024; 25:11478. [PMID: 39519031 PMCID: PMC11546839 DOI: 10.3390/ijms252111478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 10/22/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Cell death is a critical biological process necessary for development, tissue maintenance, and defense against diseases. To date, more than 20 forms of cell death have been identified, each defined by unique molecular pathways. Understanding these different forms of cell death is essential for investigating the pathogenesis of diseases such as cancer, neurodegenerative disorders, and autoimmune conditions and developing appropriate therapies. Paraptosis is a distinct form of regulated cell death characterized by cytoplasmic vacuolation and dilatation of cellular organelles like the mitochondria and endoplasmic reticulum (ER). It is regulated by several signaling pathways, for instance, those associated with ER stress, calcium overload, oxidative stress, and specific cascades such as insulin-like growth factor I receptor (IGF-IR) and its downstream signaling pathways comprising mitogen-activated protein kinases (MAPKs) and Jun N-terminal kinase (JNK). Paraptosis has been observed in diverse biological contexts, including development and cellular stress responses in neuronal, retinal, endothelial, and muscle cells. The induction of paraptosis is increasingly important in anticancer therapy, as it targets non-apoptotic stress responses in tumor cells, which can be utilized to induce cell death. This approach enhances treatment efficacy and addresses drug resistance, particularly in cases where cancer cells are resistant to apoptosis. Combining paraptosis-inducing agents with traditional therapies holds promise for enhancing treatment efficacy and overcoming drug resistance, suggesting a valuable strategy in anticancer therapy.
Collapse
Affiliation(s)
- Claudia Kunst
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, Rheumatology, Immunology, and Infectious Diseases, University Hospital Regensburg, 93053 Regensburg, Germany; (D.T.); (M.E.); (H.C.T.); (M.M.); (K.G.)
| | | | | | | | | | | |
Collapse
|
42
|
Chen SY, Wu J, Chen Y, Wang YE, Setayeshpour Y, Federico C, Mestre AA, Lin CC, Chi JT. NINJ1 regulates ferroptosis via xCT antiporter interaction and CoA modulation. Cell Death Dis 2024; 15:755. [PMID: 39424803 PMCID: PMC11489787 DOI: 10.1038/s41419-024-07135-1] [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: 05/13/2024] [Revised: 10/02/2024] [Accepted: 10/04/2024] [Indexed: 10/21/2024]
Abstract
Ninjurin-1 (NINJ1), initially identified as a stress-induced protein in neurons, recently emerged as a key mediator of plasma membrane rupture (PMR) during apoptosis, necrosis, and pyroptosis. However, its involvement in ferroptosis is less well elucidated. Here, we demonstrate that NINJ1 also plays a crucial role in ferroptosis, but through a distinct mechanism. NINJ1 knockdown significantly protected cancer cells against ferroptosis induced only by xCT inhibitors but no other classes of ferroptosis-inducing compounds (FINs). Glycine, known to inhibit canonical NINJ1-mediated membrane rupture in other cell deaths, had no impact on ferroptosis. A compound screen revealed that the ferroptosis protective effect caused by NINJ1 knockdown can be abolished by pantothenate kinase inhibitor (PANKi), buthionine sulfoximine (BSO), and diethylmaleate (DEM). These results suggest that this ferroptosis protection is mediated via Coenzyme A (CoA) and glutathione (GSH), both of which were found to be elevated upon NINJ1 knockdown. Furthermore, we discovered that NINJ1 interacts with the xCT antiporter, which is responsible for cystine uptake for the biosynthesis of CoA and GSH. The removal of NINJ1 increased xCT levels and stability, enhancing cystine uptake and thereby providing protection against ferroptosis. Conversely, NINJ1 overexpression reduced xCT levels and sensitized ferroptosis. These findings reveal that NINJ1 regulates ferroptosis via a non-canonical mechanism, distinct from other regulated cell deaths.
Collapse
Affiliation(s)
- Ssu-Yu Chen
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Jianli Wu
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Yubin Chen
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Ya-En Wang
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Yasaman Setayeshpour
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Chiara Federico
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Alexander A Mestre
- Department of Biochemistry, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Chao-Chieh Lin
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, 27710, USA.
| | - Jen-Tsan Chi
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, 27710, USA.
- Center for Advanced Genomic Technologies, Duke University School of Medicine, Durham, NC, 27710, USA.
| |
Collapse
|
43
|
Richardson KK, Adam GO, Ling W, Warren A, Marques-Carvalho A, Thostenson JD, Krager K, Aykin-Burns N, Byrum SD, Almeida M, Kim HN. Mitochondrial protein deacetylation by SIRT3 in osteoclasts promotes bone resorption with aging in female mice. Mol Metab 2024; 88:102012. [PMID: 39154858 PMCID: PMC11399565 DOI: 10.1016/j.molmet.2024.102012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 08/12/2024] [Accepted: 08/12/2024] [Indexed: 08/20/2024] Open
Abstract
OBJECTIVES The mitochondrial deacetylase sirtuin-3 (SIRT3) is necessary for the increased bone resorption and enhanced function of mitochondria in osteoclasts that occur with advancing age; how SIRT3 drives bone resorption remains elusive. METHODS To determine the role of SIRT3 in osteoclast mitochondria, we used mice with conditional loss of Sirt3 in osteoclast lineage and mice with germline deletion of either Sirt3 or its known target Pink1. RESULTS SIRT3 stimulates mitochondrial quality in osteoclasts in a PINK1-independent manner, promoting mitochondrial activity and osteoclast maturation and function, thereby contributing to bone loss in female but not male mice. Quantitative analyses of global proteomes and acetylomes revealed that deletion of Sirt3 dramatically increased acetylation of osteoclast mitochondrial proteins, particularly ATPase inhibitory factor 1 (ATPIF1), an essential protein for mitophagy. Inhibition of mitophagy via mdivi-1 recapitulated the effect of deletion of Sirt3 or Atpif1 in osteoclast formation and mitochondrial function. CONCLUSIONS Decreasing mitophagic flux in osteoclasts may be a promising pharmacotherapeutic approach to treat osteoporosis in older adults.
Collapse
Affiliation(s)
- Kimberly K Richardson
- Center for Musculoskeletal Disease Research, USA; Division of Endocrinology, Department of Internal Medicine, USA
| | - Gareeballah Osman Adam
- Center for Musculoskeletal Disease Research, USA; Division of Endocrinology, Department of Internal Medicine, USA
| | - Wen Ling
- Center for Musculoskeletal Disease Research, USA; Division of Endocrinology, Department of Internal Medicine, USA
| | - Aaron Warren
- Center for Musculoskeletal Disease Research, USA; Division of Endocrinology, Department of Internal Medicine, USA
| | - Adriana Marques-Carvalho
- Center for Musculoskeletal Disease Research, USA; Division of Endocrinology, Department of Internal Medicine, USA
| | - Jeff D Thostenson
- Center for Musculoskeletal Disease Research, USA; Department of Biostatistics, USA
| | - Kimberly Krager
- Division of Radiation Health, Department of Pharmaceutical Sciences, USA
| | - Nukhet Aykin-Burns
- Division of Radiation Health, Department of Pharmaceutical Sciences, USA
| | - Stephanie D Byrum
- Department of Biochemistry and Molecular Biology, USA; Arkansas Children's Research Institute, Little Rock, AR, USA
| | - Maria Almeida
- Center for Musculoskeletal Disease Research, USA; Division of Endocrinology, Department of Internal Medicine, USA; Department of Orthopedic Surgery, University of Arkansas for Medical Sciences, USA
| | - Ha-Neui Kim
- Center for Musculoskeletal Disease Research, USA; Division of Endocrinology, Department of Internal Medicine, USA.
| |
Collapse
|
44
|
Maremonti F, Tonnus W, Gavali S, Bornstein S, Shah A, Giacca M, Linkermann A. Ferroptosis-based advanced therapies as treatment approaches for metabolic and cardiovascular diseases. Cell Death Differ 2024; 31:1104-1112. [PMID: 39068204 PMCID: PMC11369293 DOI: 10.1038/s41418-024-01350-1] [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: 05/12/2024] [Revised: 07/10/2024] [Accepted: 07/18/2024] [Indexed: 07/30/2024] Open
Abstract
Ferroptosis has attracted attention throughout the last decade because of its tremendous clinical importance. Here, we review the rapidly growing body of literature on how inhibition of ferroptosis may be harnessed for the treatment of common diseases, and we focus on metabolic and cardiovascular unmet medical needs. We introduce four classes of preclinically established ferroptosis inhibitors (ferrostatins) such as iron chelators, radical trapping agents that function in the cytoplasmic compartment, lipophilic radical trapping antioxidants and ninjurin-1 (NINJ1) specific monoclonal antibodies. In contrast to ferroptosis inducers that cause serious untoward effects such as acute kidney tubular necrosis, the side effect profile of ferrostatins appears to be limited. We also consider ferroptosis as a potential side effect itself when several advanced therapies harnessing small-interfering RNA (siRNA)-based treatment approaches are tested. Importantly, clinical trial design is impeded by the lack of an appropriate biomarker for ferroptosis detection in serum samples or tissue biopsies. However, we discuss favorable clinical scenarios suited for the design of anti-ferroptosis clinical trials to test such first-in-class compounds. We conclude that targeting ferroptosis exhibits outstanding treatment options for metabolic and cardiovascular diseases, but we have only begun to translate this knowledge into clinically relevant applications.
Collapse
Affiliation(s)
- Francesca Maremonti
- Division of Nephrology, Medical Clinic III, University Hospital Dresden, Technische Universität Dresden, Dresden, Germany
- Department of Medicine V, University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany
- Department of Internal Medicine 3, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Wulf Tonnus
- Division of Nephrology, Medical Clinic III, University Hospital Dresden, Technische Universität Dresden, Dresden, Germany
- Department of Internal Medicine 3, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Shubhangi Gavali
- Division of Nephrology, Medical Clinic III, University Hospital Dresden, Technische Universität Dresden, Dresden, Germany
- Department of Medicine V, University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany
- Department of Internal Medicine 3, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Stefan Bornstein
- Department of Internal Medicine 3, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
- Diabetes and Nutritional Sciences, King's College London, London, UK
- Center for Regenerative Therapies, Technische Universität Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden of Helmholtz Centre Munich at University Clinic Carl Gustav Carus of TU Dresden Faculty of Medicine, Dresden, Germany
| | - Ajay Shah
- King's College London British Heart Foundation Centre, School of Cardiovascular & Metabolic Medicine and Sciences, London, UK
| | - Mauro Giacca
- King's College London British Heart Foundation Centre, School of Cardiovascular & Metabolic Medicine and Sciences, London, UK
| | - Andreas Linkermann
- Division of Nephrology, Medical Clinic III, University Hospital Dresden, Technische Universität Dresden, Dresden, Germany.
- Department of Medicine V, University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany.
- Department of Internal Medicine 3, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany.
- Division of Nephrology, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
45
|
Liu J, Li L, He S, Zheng X, Zhu D, Kong G, Li P. EXPLORING THE PROGNOSTIC NECROPTOSIS-RELATED GENES AND UNDERLYING MECHANISM IN SEPSIS USING BIOINFORMATICS. Shock 2024; 62:363-374. [PMID: 38920136 PMCID: PMC11460741 DOI: 10.1097/shk.0000000000002414] [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: 03/12/2024] [Revised: 04/10/2024] [Accepted: 05/28/2024] [Indexed: 06/27/2024]
Abstract
ABSTRACT Sepsis is a life-threatening disease due to a dysregulated host response to infection, with an unknown regulatory mechanism for prognostic necroptosis-related genes (NRGs). Using GEO datasets GSE65682 and GSE134347, we identified six NRG biomarkers ( ATRX , TSC1 , CD40 , BACH2 , BCL2 , and LEF1 ) with survival and diagnostic significance through Kaplan-Meier (KM) and receiver operating characteristic (ROC) analyses. Afterward, the ingenuity pathway analysis (IPA) highlighted enrichment in hepatic fibrosis pathways and BEX2 protein. Moreover, we examined their regulatory targets and functional links with necroptotic signaling molecules via miRDB, TargetScan, Network analyst, and GeneMANIA. The molecular regulatory network displayed that hsa-miR-5195-3p and hsa-miR-145-5p regulated ATRX, BACH2, and CD40, while YY1 showed strong connectivity, concurrently controlling LEF1, ATRX, BCL2, BACH2, and CD40. CD40 exhibited similar expression patterns to RIPK3 and MLKL, and LEF1 was functionally associated with MLKL. Additionally, DrugBank analysis identified paclitaxel, docetaxel, and rasagiline as potential BCL2-targeting sepsis treatments. Finally, real-time quantitative PCR confirmed ATRX, TSC1, and LEF1 downregulation in sepsis samples, contrasting CD40's increased expression in CTL samples. In conclusion, ATRX , TSC1 , CD40 , BACH2 , BCL2 , and LEF1 may be critical regulatory targets of necroptosis in sepsis, providing a basis for further necroptosis-related studies in sepsis.
Collapse
Affiliation(s)
- Jie Liu
- General Practice, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Lin Li
- National-Local Joint Engineering Research Center of Biodiagnosis & Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Shuyang He
- Queen Mary School of Nanchang University, Nanchang, Jiangxi, China
| | - Xin Zheng
- Department of Emergency, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Dan Zhu
- Department of Emergency, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Guangyao Kong
- National-Local Joint Engineering Research Center of Biodiagnosis & Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Ping Li
- General Practice, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| |
Collapse
|
46
|
Berndt C, Alborzinia H, Amen VS, Ayton S, Barayeu U, Bartelt A, Bayir H, Bebber CM, Birsoy K, Böttcher JP, Brabletz S, Brabletz T, Brown AR, Brüne B, Bulli G, Bruneau A, Chen Q, DeNicola GM, Dick TP, Distéfano A, Dixon SJ, Engler JB, Esser-von Bieren J, Fedorova M, Friedmann Angeli JP, Friese MA, Fuhrmann DC, García-Sáez AJ, Garbowicz K, Götz M, Gu W, Hammerich L, Hassannia B, Jiang X, Jeridi A, Kang YP, Kagan VE, Konrad DB, Kotschi S, Lei P, Le Tertre M, Lev S, Liang D, Linkermann A, Lohr C, Lorenz S, Luedde T, Methner A, Michalke B, Milton AV, Min J, Mishima E, Müller S, Motohashi H, Muckenthaler MU, Murakami S, Olzmann JA, Pagnussat G, Pan Z, Papagiannakopoulos T, Pedrera Puentes L, Pratt DA, Proneth B, Ramsauer L, Rodriguez R, Saito Y, Schmidt F, Schmitt C, Schulze A, Schwab A, Schwantes A, Soula M, Spitzlberger B, Stockwell BR, Thewes L, Thorn-Seshold O, Toyokuni S, Tonnus W, Trumpp A, Vandenabeele P, Vanden Berghe T, Venkataramani V, Vogel FCE, von Karstedt S, Wang F, Westermann F, Wientjens C, Wilhelm C, Wölk M, Wu K, Yang X, Yu F, Zou Y, Conrad M. Ferroptosis in health and disease. Redox Biol 2024; 75:103211. [PMID: 38908072 PMCID: PMC11253697 DOI: 10.1016/j.redox.2024.103211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 05/24/2024] [Accepted: 05/24/2024] [Indexed: 06/24/2024] Open
Abstract
Ferroptosis is a pervasive non-apoptotic form of cell death highly relevant in various degenerative diseases and malignancies. The hallmark of ferroptosis is uncontrolled and overwhelming peroxidation of polyunsaturated fatty acids contained in membrane phospholipids, which eventually leads to rupture of the plasma membrane. Ferroptosis is unique in that it is essentially a spontaneous, uncatalyzed chemical process based on perturbed iron and redox homeostasis contributing to the cell death process, but that it is nonetheless modulated by many metabolic nodes that impinge on the cells' susceptibility to ferroptosis. Among the various nodes affecting ferroptosis sensitivity, several have emerged as promising candidates for pharmacological intervention, rendering ferroptosis-related proteins attractive targets for the treatment of numerous currently incurable diseases. Herein, the current members of a Germany-wide research consortium focusing on ferroptosis research, as well as key external experts in ferroptosis who have made seminal contributions to this rapidly growing and exciting field of research, have gathered to provide a comprehensive, state-of-the-art review on ferroptosis. Specific topics include: basic mechanisms, in vivo relevance, specialized methodologies, chemical and pharmacological tools, and the potential contribution of ferroptosis to disease etiopathology and progression. We hope that this article will not only provide established scientists and newcomers to the field with an overview of the multiple facets of ferroptosis, but also encourage additional efforts to characterize further molecular pathways modulating ferroptosis, with the ultimate goal to develop novel pharmacotherapies to tackle the various diseases associated with - or caused by - ferroptosis.
Collapse
Affiliation(s)
- Carsten Berndt
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Hamed Alborzinia
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM GGmbH), Heidelberg, Germany; Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Vera Skafar Amen
- Rudolf Virchow Zentrum, Center for Integrative and Translational Bioimaging - University of Würzburg, Germany
| | - Scott Ayton
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Australia
| | - Uladzimir Barayeu
- Division of Redox Regulation, DKFZ-ZMBH Alliance, German Cancer Research Center (DKFZ) Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, 69120, Heidelberg, Germany; Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Alexander Bartelt
- Institute for Cardiovascular Prevention (IPEK), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany; Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, Neuherberg, Germany; German Center for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - Hülya Bayir
- Department of Pediatrics, Columbia University, New York City, NY, USA
| | - Christina M Bebber
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Translational Genomics, Cologne, Germany; CECAD Cluster of Excellence, University of Cologne, Cologne, Germany
| | - Kivanc Birsoy
- Laboratory of Metabolic Regulation and Genetics, Rockefeller University, New York City, NY, USA
| | - Jan P Böttcher
- Institute of Molecular Immunology, School of Medicine, Technical University of Munich (TUM), Germany
| | - Simone Brabletz
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg, Germany
| | - Thomas Brabletz
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg, Germany
| | - Ashley R Brown
- Department of Biological Sciences, Columbia University, New York City, NY, USA
| | - Bernhard Brüne
- Institute of Biochemistry1-Pathobiochemistry, Goethe-Universität, Frankfurt Am Main, Germany
| | - Giorgia Bulli
- Department of Physiological Genomics, Ludwig-Maximilians-University, Munich, Germany
| | - Alix Bruneau
- Department of Hepatology and Gastroenterology, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Berlin, Germany
| | - Quan Chen
- College of Life Sciences, Nankai University, Tianjin, China
| | - Gina M DeNicola
- Department of Metabolism and Physiology, Moffitt Cancer Center, Tampa, FL, USA
| | - Tobias P Dick
- Division of Redox Regulation, DKFZ-ZMBH Alliance, German Cancer Research Center (DKFZ) Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, 69120, Heidelberg, Germany
| | - Ayelén Distéfano
- Instituto de Investigaciones Biológicas, CONICET, National University of Mar Del Plata, Argentina
| | - Scott J Dixon
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Jan B Engler
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Germany
| | | | - Maria Fedorova
- Center of Membrane Biochemistry and Lipid Research, University Hospital Carl Gustav Carus and Faculty of Medicine of TU Dresden, Germany
| | - José Pedro Friedmann Angeli
- Rudolf Virchow Zentrum, Center for Integrative and Translational Bioimaging - University of Würzburg, Germany
| | - Manuel A Friese
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Germany
| | - Dominic C Fuhrmann
- Institute of Biochemistry1-Pathobiochemistry, Goethe-Universität, Frankfurt Am Main, Germany
| | - Ana J García-Sáez
- Institute for Genetics, CECAD, University of Cologne, Germany; Max Planck Institute of Biophysics, Frankfurt/Main, Germany
| | | | - Magdalena Götz
- Department of Physiological Genomics, Ludwig-Maximilians-University, Munich, Germany; Institute of Stem Cell Research, Helmholtz Center Munich, Germany
| | - Wei Gu
- Institute for Cancer Genetics, And Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA; Department of Pathology and Cell Biology, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Linda Hammerich
- Department of Hepatology and Gastroenterology, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Berlin, Germany
| | | | - Xuejun Jiang
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Aicha Jeridi
- Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Comprehensive Pneumology Center (CPC-M), Germany, Member of the German Center for Lung Research (DZL)
| | - Yun Pyo Kang
- College of Pharmacy and Research Institute of Pharmaceutical Science, Seoul National University, Republic of Korea
| | | | - David B Konrad
- Department of Pharmacy, Ludwig-Maximilians-University, Munich, Germany
| | - Stefan Kotschi
- Institute for Cardiovascular Prevention (IPEK), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Peng Lei
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Marlène Le Tertre
- Center for Translational Biomedical Iron Research, Heidelberg University, Germany
| | - Sima Lev
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Deguang Liang
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Andreas Linkermann
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Germany; Division of Nephrology, Department of Medicine, Albert Einstein College of Medicine, New York, NY, USA
| | - Carolin Lohr
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Svenja Lorenz
- Institute of Metabolism and Cell Death, Helmholtz Center Munich, Germany
| | - Tom Luedde
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Axel Methner
- Institute of Molecular Medicine, Johannes Gutenberg-Universität Mainz, Germany
| | - Bernhard Michalke
- Research Unit Analytical Biogeochemistry, Helmholtz Center Munich, Germany
| | - Anna V Milton
- Department of Pharmacy, Ludwig-Maximilians-University, Munich, Germany
| | - Junxia Min
- School of Medicine, Zhejiang University, Hangzhou, China
| | - Eikan Mishima
- Institute of Metabolism and Cell Death, Helmholtz Center Munich, Germany
| | | | - Hozumi Motohashi
- Department of Gene Expression Regulation, Tohoku University, Sendai, Japan
| | | | - Shohei Murakami
- Department of Gene Expression Regulation, Tohoku University, Sendai, Japan
| | - James A Olzmann
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA; Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA, USA; Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Gabriela Pagnussat
- Instituto de Investigaciones Biológicas, CONICET, National University of Mar Del Plata, Argentina
| | - Zijan Pan
- School of Life Sciences, Westlake University, Hangzhou, China
| | | | | | - Derek A Pratt
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Canada
| | - Bettina Proneth
- Institute of Metabolism and Cell Death, Helmholtz Center Munich, Germany
| | - Lukas Ramsauer
- Institute of Molecular Immunology, School of Medicine, Technical University of Munich (TUM), Germany
| | | | - Yoshiro Saito
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Felix Schmidt
- Institute of Molecular Medicine, Johannes Gutenberg-Universität Mainz, Germany
| | - Carina Schmitt
- Department of Pharmacy, Ludwig-Maximilians-University, Munich, Germany
| | - Almut Schulze
- Division of Tumour Metabolism and Microenvironment, DKFZ Heidelberg and DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Annemarie Schwab
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg, Germany
| | - Anna Schwantes
- Institute of Biochemistry1-Pathobiochemistry, Goethe-Universität, Frankfurt Am Main, Germany
| | - Mariluz Soula
- Laboratory of Metabolic Regulation and Genetics, Rockefeller University, New York City, NY, USA
| | - Benedikt Spitzlberger
- Department of Immunobiology, Université de Lausanne, Switzerland; Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, Munich, Germany
| | - Brent R Stockwell
- Department of Biological Sciences, Columbia University, New York City, NY, USA; Department of Pathology and Cell Biology, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA; Department of Chemistry, Columbia University, New York, NY, USA
| | - Leonie Thewes
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | | | - Shinya Toyokuni
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, Japan; Center for Low-temperature Plasma Sciences, Nagoya University, Nagoya, Japan; Center for Integrated Sciences of Low-temperature Plasma Core Research (iPlasma Core), Tokai National Higher Education and Research System, Nagoya, Japan
| | - Wulf Tonnus
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Germany
| | - Andreas Trumpp
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM GGmbH), Heidelberg, Germany; Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Peter Vandenabeele
- VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Tom Vanden Berghe
- Department of Biomedical Sciences, University of Antwerp, Belgium; VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Vivek Venkataramani
- Comprehensive Cancer Center Mainfranken, University Hospital Würzburg, Germany
| | - Felix C E Vogel
- Division of Tumour Metabolism and Microenvironment, DKFZ Heidelberg and DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Silvia von Karstedt
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Translational Genomics, Cologne, Germany; CECAD Cluster of Excellence, University of Cologne, Cologne, Germany; University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Molecular Medicine Cologne, Germany
| | - Fudi Wang
- School of Medicine, Zhejiang University, Hangzhou, China
| | | | - Chantal Wientjens
- Immunopathology Unit, Institute of Clinical Chemistry and Clinical Pharmacology, Medical Faculty, University Hospital Bonn, University of Bonn, Germany
| | - Christoph Wilhelm
- Immunopathology Unit, Institute of Clinical Chemistry and Clinical Pharmacology, Medical Faculty, University Hospital Bonn, University of Bonn, Germany
| | - Michele Wölk
- Center of Membrane Biochemistry and Lipid Research, University Hospital Carl Gustav Carus and Faculty of Medicine of TU Dresden, Germany
| | - Katherine Wu
- Department of Pathology, Grossman School of Medicine, New York University, NY, USA
| | - Xin Yang
- Institute for Cancer Genetics, And Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Fan Yu
- College of Life Sciences, Nankai University, Tianjin, China
| | - Yilong Zou
- School of Life Sciences, Westlake University, Hangzhou, China; Westlake Four-Dimensional Dynamic Metabolomics (Meta4D) Laboratory, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
| | - Marcus Conrad
- Institute of Metabolism and Cell Death, Helmholtz Center Munich, Germany.
| |
Collapse
|
47
|
Wang LY, Liu XJ, Li QQ, Zhu Y, Ren HL, Song JN, Zeng J, Mei J, Tian HX, Rong DC, Zhang SH. The romantic history of signaling pathway discovery in cell death: an updated review. Mol Cell Biochem 2024; 479:2255-2272. [PMID: 37851176 DOI: 10.1007/s11010-023-04873-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 10/05/2023] [Indexed: 10/19/2023]
Abstract
Cell death is a fundamental physiological process in all living organisms. Processes such as embryonic development, organ formation, tissue growth, organismal immunity, and drug response are accompanied by cell death. In recent years with the development of electron microscopy as well as biological techniques, especially the discovery of novel death modes such as ferroptosis, cuprotosis, alkaliptosis, oxeiptosis, and disulfidptosis, researchers have been promoted to have a deeper understanding of cell death modes. In this systematic review, we examined the current understanding of modes of cell death, including the recently discovered novel death modes. Our analysis highlights the common and unique pathways of these death modes, as well as their impact on surrounding cells and the organism as a whole. Our aim was to provide a comprehensive overview of the current state of research on cell death, with a focus on identifying gaps in our knowledge and opportunities for future investigation. We also presented a new insight for macroscopic intracellular survival patterns, namely that intracellular molecular homeostasis is central to the balance of different cell death modes, and this viewpoint can be well justified by the signaling crosstalk of different death modes. These concepts can facilitate the future research about cell death in clinical diagnosis, drug development, and therapeutic modalities.
Collapse
Affiliation(s)
- Lei-Yun Wang
- Department of Pharmacy, Traditional Chinese and Western Medicine Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, People's Republic of China
- Department of Pharmacy, Wuhan No.1 Hospital, Wuhan, 430022, Hubei, People's Republic of China
| | - Xing-Jian Liu
- Oujiang Laboratory, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China
| | - Qiu-Qi Li
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, People's Republic of China
| | - Ying Zhu
- Department of Pharmacy, Traditional Chinese and Western Medicine Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, People's Republic of China
- Department of Pharmacy, Wuhan No.1 Hospital, Wuhan, 430022, Hubei, People's Republic of China
| | - Hui-Li Ren
- Department of Pharmacy, Traditional Chinese and Western Medicine Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, People's Republic of China
- Department of Pharmacy, Wuhan No.1 Hospital, Wuhan, 430022, Hubei, People's Republic of China
| | - Jia-Nan Song
- Oujiang Laboratory, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China
| | - Jun Zeng
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Jie Mei
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, 410008, Hunan, People's Republic of China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China
| | - Hui-Xiang Tian
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China.
| | - Ding-Chao Rong
- Department of Orthopaedic Surgery, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510150, Guangdong, People's Republic of China.
| | - Shao-Hui Zhang
- Department of Pharmacy, Traditional Chinese and Western Medicine Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, People's Republic of China.
- Department of Pharmacy, Wuhan No.1 Hospital, Wuhan, 430022, Hubei, People's Republic of China.
| |
Collapse
|
48
|
Zhuang T, Wang S, Yu X, He X, Guo H, Ou C. Current status and future perspectives of platelet-derived extracellular vesicles in cancer diagnosis and treatment. Biomark Res 2024; 12:88. [PMID: 39183323 PMCID: PMC11346179 DOI: 10.1186/s40364-024-00639-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 08/12/2024] [Indexed: 08/27/2024] Open
Abstract
Platelets are a significant component of the cell population in the tumour microenvironment (TME). Platelets influence other immune cells and perform cross-talk with tumour cells, playing an important role in tumour development. Extracellular vesicles (EVs) are small membrane vesicles released from the cells into the TME. They can transfer biological information, including proteins, nucleic acids, and metabolites, from secretory cells to target receptor cells. This process affects the progression of various human diseases, particularly cancer. In recent years, several studies have demonstrated that platelet-derived extracellular vesicles (PEVs) can help regulate the malignant biological behaviours of tumours, including malignant proliferation, resistance to cell death, invasion and metastasis, metabolic reprogramming, immunity, and angiogenesis. Consequently, PEVs have been identified as key regulators of tumour progression. Therefore, targeting PEVs is a potential strategy for tumour treatment. Furthermore, the extensive use of nanomaterials in medical research has indicated that engineered PEVs are ideal delivery systems for therapeutic drugs. Recent studies have demonstrated that PEV engineering technologies play a pivotal role in the treatment of tumours by combining photothermal therapy, immunotherapy, and chemotherapy. In addition, aberrant changes in PEVs are closely associated with the clinicopathological features of patients with tumours, which may serve as liquid biopsy markers for early diagnosis, monitoring disease progression, and the prognostic assessment of patients with tumours. A comprehensive investigation into the role and potential mechanisms of PEVs in tumourigenesis may provide novel diagnostic biomarkers and potential therapeutic strategies for treating human tumours.
Collapse
Affiliation(s)
- Tongtao Zhuang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Shenrong Wang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Xiaoqian Yu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Xiaoyun He
- Departments of Ultrasound Imaging, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Hongbin Guo
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| | - Chunlin Ou
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
49
|
DJALDETTI MEIR. Immunomodulatory and chemopreventive effects of resveratrol on the digestive system cancers. Oncol Res 2024; 32:1389-1399. [PMID: 39220125 PMCID: PMC11361903 DOI: 10.32604/or.2024.049745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 05/23/2024] [Indexed: 09/04/2024] Open
Abstract
Resveratrol (RSV), the primary polyphenol found in grapes, has been revealed to have anti-inflammatory properties by reducing the capacity of the peripheral blood mononuclear cells to produce pro-inflammatory cytokines, including IL-1β, IL-6, IL-1ra and TNFα. Considering the close association between chronic inflammation and cancer development, RSV's immunomodulatory properties are one way by which the polyphenol may inhibit cancer initiation, proliferation, neovascularization, and migration. Resveratrol influences the generation of microtumor environment which is one of the key factors in cancer progress. In addition to immunomodulation, RSV inhibits cancer development by expressing anti-oxidant effects, causing cell cycle arrest, stimulating the function of certain enzymes, and activating cell signaling pathways. The end outcome is one of the various forms of cell death, including apoptosis, pyroptosis, necroptosis, and more, as it has been observed in vitro. RSV has been shown to act against cancer in practically every organ, while its effects on colon cancer have been documented more frequently. It is remarkable that longer-term clinical studies that may have established the potential for this natural substance to serve as a therapeutic adjuvant to traditional anti-cancer medications were not prompted by the encouraging outcomes seen with cancer cells treated with non-toxic doses of resveratrol. The current review aims to assess the recent findings about the immunological and anti-cancer characteristics of RSV, with a particular emphasis on cancers of the digestive tract, as a challenge for future clinical research that may contribute to the better prognosis of cancer.
Collapse
Affiliation(s)
- MEIR DJALDETTI
- />Laboratory for Immunology and Hematology Research, Rabin Medical Center, Hasharon Hospital, Petah-Tiqva, the Sackler School of Medicine, Tel-Aviv University, Ramat Aviv, 69978, Israel
| |
Collapse
|
50
|
Cooper KN, Potempa J, Bagaitkar J. Dying for a cause: The pathogenic manipulation of cell death and efferocytic pathways. Mol Oral Microbiol 2024; 39:165-179. [PMID: 37786286 PMCID: PMC10985052 DOI: 10.1111/omi.12436] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 08/21/2023] [Accepted: 09/21/2023] [Indexed: 10/04/2023]
Abstract
Cell death is a natural consequence of infection. However, although the induction of cell death was solely thought to benefit the pathogen, compelling data now show that the activation of cell death pathways serves as a nuanced antimicrobial strategy that couples pathogen elimination with the generation of inflammatory cytokines and the priming of innate and adaptive cellular immunity. Following cell death, the phagocytic uptake of the infected dead cell by antigen-presenting cells and the subsequent lysosomal fusion of the apoptotic body containing the pathogen serve as an important antimicrobial mechanism that furthers the development of downstream adaptive immune responses. Despite the complexity of regulated cell death pathways, pathogens are highly adept at evading them. Here, we provide an overview of the remarkable diversity of cell death and efferocytic pathways and discuss illustrative examples of virulence strategies employed by pathogens, including oral pathogens, to counter their activation and persist within the host.
Collapse
Affiliation(s)
- Kelley N Cooper
- Department of Immunology and Microbiology, University of Louisville, Louisville, KY
- Department of Oral Immunology and Infectious Diseases, University of Louisville, Louisville, KY
| | - Jan Potempa
- Department of Oral Immunology and Infectious Diseases, University of Louisville, Louisville, KY
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Juhi Bagaitkar
- Center for Microbial Pathogenesis, Nationwide Children’s Hospital, Columbus, OH
- Department of Pediatrics, The Ohio State College of Medicine, Columbus, OH
| |
Collapse
|