1
|
Huang X, Zhang Y, Jiang Y, Li T, Yang S, Wang Y, Yu B, Zhou M, Zhang G, Zhao X, Sun J, Sun X. Contribution of ferroptosis and SLC7A11 to light-induced photoreceptor degeneration. Neural Regen Res 2026; 21:406-416. [PMID: 39104162 PMCID: PMC12094538 DOI: 10.4103/nrr.nrr-d-23-01741] [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: 10/23/2023] [Revised: 04/27/2024] [Accepted: 07/03/2024] [Indexed: 08/07/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202601000-00043/figure1/v/2025-06-09T151831Z/r/image-tiff Progressive photoreceptor cell death is one of the main pathological features of age-related macular degeneration and eventually leads to vision loss. Ferroptosis has been demonstrated to be associated with retinal degenerative diseases. However, the molecular mechanisms underlying ferroptosis and photoreceptor cell death in age-related macular degeneration remain largely unexplored. Bioinformatics and biochemical analyses in this study revealed xC - , solute carrier family 7 member 11-regulated ferroptosis as the predominant pathological process of photoreceptor cell degeneration in a light-induced dry age-related macular degeneration mouse model. This process involves the nuclear factor-erythroid factor 2-related factor 2-solute carrier family 7 member 11-glutathione peroxidase 4 signaling pathway, through which cystine depletion, iron ion accumulation, and enhanced lipid peroxidation ultimately lead to photoreceptor cell death and subsequent visual function impairment. We demonstrated that solute carrier family 7 member 11 overexpression blocked this process by inhibiting oxidative stress in vitro and in vivo . Conversely, solute carrier family 7 member 11 knockdown or the solute carrier family 7 member 11 inhibitor sulfasalazine and ferroptosis-inducing agent erastin aggravated H 2 O 2 -induced ferroptosis of 661W cells. These findings indicate solute carrier family 7 member 11 may be a potential therapeutic target for patients with retinal degenerative diseases including age-related macular degeneration.
Collapse
Affiliation(s)
- Xiaoxu Huang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People’s Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Diseases; Shanghai Engineering Center for Visual Science and Photomedicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Yumeng Zhang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People’s Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Diseases; Shanghai Engineering Center for Visual Science and Photomedicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Yuxin Jiang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People’s Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Diseases; Shanghai Engineering Center for Visual Science and Photomedicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Tong Li
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People’s Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Diseases; Shanghai Engineering Center for Visual Science and Photomedicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Shiqi Yang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People’s Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Diseases; Shanghai Engineering Center for Visual Science and Photomedicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Yimin Wang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People’s Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Diseases; Shanghai Engineering Center for Visual Science and Photomedicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Bo Yu
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People’s Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Diseases; Shanghai Engineering Center for Visual Science and Photomedicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Minwen Zhou
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People’s Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Diseases; Shanghai Engineering Center for Visual Science and Photomedicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Guanran Zhang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People’s Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Diseases; Shanghai Engineering Center for Visual Science and Photomedicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Xiaohuan Zhao
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People’s Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Diseases; Shanghai Engineering Center for Visual Science and Photomedicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Junran Sun
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People’s Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Diseases; Shanghai Engineering Center for Visual Science and Photomedicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Xiaodong Sun
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People’s Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Diseases; Shanghai Engineering Center for Visual Science and Photomedicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| |
Collapse
|
2
|
Sun X, Li J, Dong P, Li L, Gu Z, Yuan J. Antioxidant and anti-apoptotic properties of heme oxygenase-1 in red swamp crayfish Procambarus clarkii. FISH & SHELLFISH IMMUNOLOGY 2025; 162:110348. [PMID: 40254088 DOI: 10.1016/j.fsi.2025.110348] [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: 01/21/2025] [Revised: 03/30/2025] [Accepted: 04/15/2025] [Indexed: 04/22/2025]
Abstract
Heme oxygenase-1 (HO-1), which is significantly induced in response to oxidative stress, plays a crucial role in mitigating oxidative damage. However, the function of HO-1 in crayfish remains unknown. In this study, HO-1 in Procambarus clarkii (PcHO-1) was identified, and its functional domain was conserved across different species based on sequence alignment and structural prediction. Through RT-qPCR analysis, PcHO-1 showed the highest expression level in the hepatopancreas. Under the stimulation of Aeromonas hydrophila or glufosinate ammonium (GLA), the crayfish showed oxidative stress damage, whereas the expression levels of PcHO-1 were increased. Knocking down of PcHO-1 with RNA interference significantly reduced the antioxidant capacity of crayfish compared to the control group under A. hydrophila or GLA stimulation. Furthermore, the expression level of PcHO-1 increased after induction with CoPPIX, which increased the antioxidant level of crayfish and reduced the apoptosis. These findings indicated that PcHO-1 manifests the antioxidant and anti-apoptotic capacity, thereby aiding in the repairing of damage caused by A. hydrophila or GLA in P. clarkii.
Collapse
Affiliation(s)
- Xingye Sun
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, China; National Aquatic Animal Diseases Para-reference Laboratory (HZAU), Wuhan, China
| | - Jiahao Li
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, China; Hubei Engineering Research Centre for Aquatic Animal Diseases Control and Prevention, Wuhan, China
| | - Peixiang Dong
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, China
| | - Lijuan Li
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, China; National Aquatic Animal Diseases Para-reference Laboratory (HZAU), Wuhan, China; Hubei Engineering Research Centre for Aquatic Animal Diseases Control and Prevention, Wuhan, China
| | - Zemao Gu
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, China; National Aquatic Animal Diseases Para-reference Laboratory (HZAU), Wuhan, China; Hubei Engineering Research Centre for Aquatic Animal Diseases Control and Prevention, Wuhan, China
| | - Junfa Yuan
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, China; National Aquatic Animal Diseases Para-reference Laboratory (HZAU), Wuhan, China; Hubei Engineering Research Centre for Aquatic Animal Diseases Control and Prevention, Wuhan, China.
| |
Collapse
|
3
|
Wang S, Du R, Liu J, Zhong W, Zhang C, Jiang X, Wang X, Wu Q, Tong G, Luo L. Multi-approach analysis reveals the mechanism by which Shugan Xiaozhi decoction protects against metabolic dysfunction-associated steatohepatitis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156712. [PMID: 40220418 DOI: 10.1016/j.phymed.2025.156712] [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: 12/07/2024] [Revised: 03/08/2025] [Accepted: 03/30/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND Metabolic dysfunction-associated steatohepatitis (MASH) is a human health-threatening hepatic disease with limited treatment strategies. As a clinical Traditional Chinese Medicine compound for MASH, Shugan Xiaozhi (SGXZ) decoction has a definite effect, but its mechanism in treating MASH is still not very clear. PURPOSE Exploring the potential mechanism of SGXZ decoction in treating MASH through multiomics and animal experimental validation. METHODS UPLC-ESI-MS method was used to identify the main components of SGXZ decoction. Periodic acid-schiff (PAS), picrosirius red (PSR), and oil red o staining were used to assess the effect of SGXZ decoction on MCD-induced MASH mouse model. The mechanism of SGXZ decoction on MASH was analyzed using multiomics techniques. TUNEL staining, western blot (WB), immunohistochemistry (IHC), kits, transmission electron microscopy (TEM), and immunofluorescence (IF) were used to validate the mechanism of SGXZ decoction on MASH. Finally, molecular docking and molecular dynamics simulation were used to verify the targeting between key components of SGXZ decoction and important targets for intervention. RESULTS Through UPLC-ESI-MS analysis, 30 main active ingredients were obtained from SGXZ decoction. SGXZ decoction improved MASH, as evidenced by the improvement in histopathology, hepatic function indexes, lipid and fibrosis indicators. Both proteomic and transcriptomic results suggested an important role for ferroptosis in SGXZ decoction intervention in MASH, ferroptosis-related pathways were the main significant pathways obtained from these analyses. In addition, SGXZ decoction treatment reduced cell death, inflammation, and oxidative stress levels and restored impaired mitochondrial morphology in MCD-induced MASH mice. Furthermore, Mechanism experiments proved that SGXZ decoction treatment improved iron metabolism and lipid peroxidation imbalance and activated the Xc- system in MASH mice. CONCLUSION SGXZ decoction does have a therapeutic effect on MASH, and its mechanism may be related to its regulation of p53/ SLC7A11/GPX4 pathway to reduce ferroptosis.
Collapse
Affiliation(s)
- Shuai Wang
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau 999078, PR China; Department of Hepatology, the Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, No. 15, Yingchun Road, Luohu District, Guangdong 518033, PR China; Shenzhen Key Laboratory of Liver Diseases of Chinese Medicine, No. 15, Yingchun Road, Luohu District, Guangdong, 518033, PR China
| | - Ruili Du
- The First Clinical Medical College of Henan University of Chinese Medicine, No. 19, Renmin Road, Jinshui District, Henan, 450003, PR China
| | - Jiahui Liu
- Department of Nephrology, The Fourth Clinical Medical College of Guangzhou University of Traditional Chinese Medicine, No.15, Yingchun Road, Luohu District, Guangdong 518033, PR China
| | - Weichao Zhong
- Department of Hepatology, the Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, No. 15, Yingchun Road, Luohu District, Guangdong 518033, PR China; Shenzhen Key Laboratory of Liver Diseases of Chinese Medicine, No. 15, Yingchun Road, Luohu District, Guangdong, 518033, PR China
| | - Chunmei Zhang
- School of Basic Medical Science of Luoyang Polytechnic, No. 6 Keji Avenue, Yibin District, Henan, 471099, PR China
| | - Xia Jiang
- Key Laboratory of Epigenetics and Oncology, The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, 646000, PR China
| | - Xiaohui Wang
- Department of Hepatology, the Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, No. 15, Yingchun Road, Luohu District, Guangdong 518033, PR China; Shenzhen Key Laboratory of Liver Diseases of Chinese Medicine, No. 15, Yingchun Road, Luohu District, Guangdong, 518033, PR China
| | - Qibiao Wu
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau 999078, PR China; Chinese Medicine Guangdong Laboratory (Hengqin Laboratory), Guangdong-Macao ln-Depth Cooperation Zone in Hengqin, 519000, PR China.
| | - Guangdong Tong
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau 999078, PR China; Department of Hepatology, the Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, No. 15, Yingchun Road, Luohu District, Guangdong 518033, PR China; Shenzhen Key Laboratory of Liver Diseases of Chinese Medicine, No. 15, Yingchun Road, Luohu District, Guangdong, 518033, PR China.
| | - Lidan Luo
- Department of Hepatology, the Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, No. 15, Yingchun Road, Luohu District, Guangdong 518033, PR China; Shenzhen Key Laboratory of Liver Diseases of Chinese Medicine, No. 15, Yingchun Road, Luohu District, Guangdong, 518033, PR China.
| |
Collapse
|
4
|
Alarcón-Veleiro C, López-Calvo I, Berjawi L, Lucio-Gallego S, Mato-Basalo R, Quindos-Varela M, Lesta-Mellid R, Santamarina-Caínzos I, Varela-Rodríguez S, Fraga M, Quintela M, Vizoso-Vázquez A, Arufe MC, Fafián-Labora J. Ferroptosis: An emerging strategy for managing epithelial ovarian cancer. Biomed Pharmacother 2025; 187:118065. [PMID: 40306179 DOI: 10.1016/j.biopha.2025.118065] [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: 01/09/2025] [Revised: 03/30/2025] [Accepted: 04/17/2025] [Indexed: 05/02/2025] Open
Abstract
Ferroptosis is a regulated form of cell death characterised by iron-dependent lipid peroxidation, a process intricately linked to cellular redox homeostasis. This form of cell death is induced by the accumulation of intracellular iron and the subsequent generation of reactive oxygen species (ROS), which leads to lipid peroxidation and ultimately cell death. Ferroptosis is distinct from traditional forms of cell death, such as apoptosis, and holds significant therapeutic potential, particularly in cancers harboring rat sarcoma virus (RAS) mutations, such as epithelial ovarian cancer (EOC). EOC is notoriously resistant to conventional therapies and is associated with a poor prognosis. In this review, we examine recent progress in the understanding of ferroptosis, with a particular focus on its redox biology and the complex regulatory networks involved. We also propose a novel classification system for ferroptosis modulators, grouping them into six categories (I, II, III, IV, V and VI) based on their mechanisms of action and their roles in modulating cellular redox status. By refining these categories, we aim to provide deeper insights into the role of ferroptosis in cancer biology, especially in EOC, and to identify potential therapeutic avenues. We propose that further investigation of ferroptosis in the context of redox biology could reveal novel biomarkers and therapeutic targets, offering promising strategies to overcome resistance mechanisms and improve clinical outcomes for patients with EOC and other treatment-resistant cancers.
Collapse
Affiliation(s)
- C Alarcón-Veleiro
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, INIBIC-Complejo Hospitalario Universitario A Coruña (CHUAC), Centro Interdisciplinar de Química y Biología (CICA), Universidade da Coruña (UDC), A Coruña 15008, Spain
| | - I López-Calvo
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, INIBIC-Complejo Hospitalario Universitario A Coruña (CHUAC), Centro Interdisciplinar de Química y Biología (CICA), Universidade da Coruña (UDC), A Coruña 15008, Spain; Grupo EXPRELA, Departamento de Bioloxía, Facultade de Ciencias, Rúa da Fraga, A Coruña 15071, Spain; Centro Interdisciplinar de Química de Química y Biología (CICA), Universidade da Coruña (UDC), A Coruña 15008, Spain; Instituto de Investigación Biomédica de A Coruña (INIBIC), Rúa as Xubias 84, A Coruña 15006, Spain
| | - L Berjawi
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, INIBIC-Complejo Hospitalario Universitario A Coruña (CHUAC), Centro Interdisciplinar de Química y Biología (CICA), Universidade da Coruña (UDC), A Coruña 15008, Spain
| | - S Lucio-Gallego
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, INIBIC-Complejo Hospitalario Universitario A Coruña (CHUAC), Centro Interdisciplinar de Química y Biología (CICA), Universidade da Coruña (UDC), A Coruña 15008, Spain
| | - R Mato-Basalo
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, INIBIC-Complejo Hospitalario Universitario A Coruña (CHUAC), Centro Interdisciplinar de Química y Biología (CICA), Universidade da Coruña (UDC), A Coruña 15008, Spain
| | - M Quindos-Varela
- Translational Cancer Research Group, A Coruña Biomedical Research Institute (INIBIC), Carretera del Pasaje s/n, A Coruña 15006, UK; Complexo Hospitalario Universitario de A Coruña (CHUAC), Spain
| | - R Lesta-Mellid
- Translational Cancer Research Group, A Coruña Biomedical Research Institute (INIBIC), Carretera del Pasaje s/n, A Coruña 15006, UK; Complexo Hospitalario Universitario de A Coruña (CHUAC), Spain
| | - I Santamarina-Caínzos
- Translational Cancer Research Group, A Coruña Biomedical Research Institute (INIBIC), Carretera del Pasaje s/n, A Coruña 15006, UK; Complexo Hospitalario Universitario de A Coruña (CHUAC), Spain
| | - S Varela-Rodríguez
- Translational Cancer Research Group, A Coruña Biomedical Research Institute (INIBIC), Carretera del Pasaje s/n, A Coruña 15006, UK; Complexo Hospitalario Universitario de A Coruña (CHUAC), Spain
| | - M Fraga
- Department of Anatomical Pathology, University Hospital Complex A Coruña, As Xubias 84, A Coruña 15006, Spain
| | - M Quintela
- European Cancer Stem Cell Research Institute, Cardiff University, Cardiff CF24 4HQ, UK
| | - A Vizoso-Vázquez
- Grupo EXPRELA, Departamento de Bioloxía, Facultade de Ciencias, Rúa da Fraga, A Coruña 15071, Spain
| | - M C Arufe
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, INIBIC-Complejo Hospitalario Universitario A Coruña (CHUAC), Centro Interdisciplinar de Química y Biología (CICA), Universidade da Coruña (UDC), A Coruña 15008, Spain.
| | - J Fafián-Labora
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, INIBIC-Complejo Hospitalario Universitario A Coruña (CHUAC), Centro Interdisciplinar de Química y Biología (CICA), Universidade da Coruña (UDC), A Coruña 15008, Spain.
| |
Collapse
|
5
|
Spiegel M. On the Photosensitizing Properties of Aloe-Emodin in Photodynamic Therapy: Insights from the Molecular Modeling. J Phys Chem B 2025. [PMID: 40448645 DOI: 10.1021/acs.jpcb.5c01117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2025]
Abstract
The photosensitizing properties of aloe-emodin were investigated under physiological conditions using computational chemistry tools. The neutral and monoanionic species were found to coexist in a 98:2 ratio, with dissociation causing a redshift in the absorption spectrum. Aloe-emodin exhibits high two-photon absorption cross-section values within the therapeutic window and significant transition probabilities, making it an efficient two-photon photosensitizer. Excited-state dynamics analysis revealed a triplet state quantum yield of 0.51 for the neutral species and around 0.88-0.89 for the anionic species, with triplet lifetimes of 26.0 s and 0.66 s, respectively. Both species exhibit similar Type I photoreactivity, but the neutral form more effectively oxidizes biomolecules during Type III photoreactivity. Additionally, the neutral species intercalates into DNA, particularly at the AT-TA site, inducing absorption changes and structural nucleotide rearrangements. The computational results align closely with available experimental data, further confirming their reliability.
Collapse
Affiliation(s)
- Maciej Spiegel
- Department of Organic Chemistry and Pharmaceutical Technology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| |
Collapse
|
6
|
Ji C, Wang Y, Ju Y, Liu S, Chen X, Wang J, Sun N, Tang Z, Gu P, Ji J. The role of HMOX1-mediated ferroptosis in blue light-induced damage to retinal pigment epithelium. Sci Rep 2025; 15:18949. [PMID: 40442370 PMCID: PMC12122798 DOI: 10.1038/s41598-025-03757-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 05/22/2025] [Indexed: 06/02/2025] Open
Abstract
Currently, blue light irradiation is frequently encountered in daily life and is widely considered a high-risk factor for retinal damage. In particular, blue light-induced dysfunction and death of the retinal pigment epithelium (RPE) may ultimately contribute to irreversible vision impairment and even blindness. However, the underlying pathogenic mechanism and pathogenically targeted protection against blue light-induced RPE degeneration remain unclear. In this study, through sophisticated biochemical evaluation and high-throughput sequencing, the predominant pathological process during blue light-induced RPE degeneration was confirmed to be HMOX1-mediated RPE ferroptosis, which may be involved in the Nrf2-SLC7A11-HMOX1 hierarchy. Upon further knockdown of HMOX1 with si-HMOX1 or the HMOX1 inhibitor zinc protoporphyrin (ZnPP), specific inhibition of HMOX1 overexpression significantly suppressed RPE ferroptosis. In mice, treatment with ZnPP effectively rescued RPE degeneration and visual function. These results highlighted that HMOX1-mediated ferroptosis might be a potential target for protection against blue light-induced damage to RPE cells.
Collapse
Affiliation(s)
- Chunyi Ji
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P.R. China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, P.R. China
| | - Yiqi Wang
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P.R. China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, P.R. China
| | - Yahan Ju
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P.R. China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, P.R. China
| | - Siwei Liu
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P.R. China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, P.R. China
| | - Xirui Chen
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P.R. China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, P.R. China
| | - Jiajing Wang
- Department of Ophthalmology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Na Sun
- Department of Ophthalmology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Zhimin Tang
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P.R. China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, P.R. China.
| | - Ping Gu
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P.R. China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, P.R. China.
| | - Jing Ji
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P.R. China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, P.R. China.
| |
Collapse
|
7
|
Chen Z, Zhu X, Lu MM, Ou Q, Wang X, Zhao Z, Shen Q, Wang Q, Wang Z, Xu JY, Jin C, Gao F, Wang J, Zhang J, Zhang J, Jin X, Bi Y, Lu L, Xu GT, Tian H. PHOSPHO1 Suppresses Ferroptosis in Retinal Pigment Epithelial Cells by Reducing the Levels of Phosphatidylethanolamine Molecular Species. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2505359. [PMID: 40396905 DOI: 10.1002/advs.202505359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Revised: 04/14/2025] [Indexed: 05/22/2025]
Abstract
Iron-induced lipid peroxidation of phosphatidylethanolamine (PE) species is a key driver of ferroptosis in retinal pigment epithelial (RPE) cells, a process closely associated with age-related macular degeneration (AMD). The previous studies have demonstrated that induced retinal pigment epithelial (iRPE) cells generated by transcription factor-mediated reprogramming exhibit superior therapeutic efficacy in treating AMD. In this study, it is found that these iRPE cells are resistant to ferroptosis and further identified phosphoethanolamine/phosphocholine phosphatase 1 (PHOSPHO1) as a critical regulator underlying ferroptosis resistance. Mechanistically, PHOSPHO1 inhibits ferroptosis through two distinct mechanisms. First, it reduces PE levels in the endoplasmic reticulum, thereby limiting PE-derived lipid peroxidation. Second, it suppresses autophagy and ferritinophagy, leading to a reduction in intracellular free iron accumulation. Experiments using an in vivo rat model confirm that PHOSPHO1 effectively protects RPE cells from ferroptotic damage. These findings highlight PHOSPHO1 as a potential therapeutic target for AMD, providing insights into novel ferroptosis-based intervention strategies.
Collapse
Affiliation(s)
- Zhiyang Chen
- Department of Ophthalmology of Tongji Hospital and Laboratory of Clinical and Visual Sciences of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Xiaoman Zhu
- Department of Ophthalmology of Tongji Hospital and Laboratory of Clinical and Visual Sciences of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Michael Mingze Lu
- Department of Ophthalmology of Tongji Hospital and Laboratory of Clinical and Visual Sciences of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Qingjian Ou
- Department of Ophthalmology of Tongji Hospital and Laboratory of Clinical and Visual Sciences of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, 200065, China
- Department of Physiology and Pharmacology, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Xueying Wang
- Department of Ophthalmology of Tongji Hospital and Laboratory of Clinical and Visual Sciences of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Zhenzhen Zhao
- Department of Ophthalmology of Tongji Hospital and Laboratory of Clinical and Visual Sciences of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Qi Shen
- Department of Ophthalmology of Tongji Hospital and Laboratory of Clinical and Visual Sciences of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Qian Wang
- Department of Ophthalmology of Tongji Hospital and Laboratory of Clinical and Visual Sciences of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Zhe Wang
- Department of Physiology, College of Basic Medical Sciences, Naval Medical University, Shanghai, 200433, China
| | - Jing-Ying Xu
- Department of Ophthalmology of Tongji Hospital and Laboratory of Clinical and Visual Sciences of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Caixia Jin
- Department of Ophthalmology of Tongji Hospital and Laboratory of Clinical and Visual Sciences of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Furong Gao
- Department of Ophthalmology of Tongji Hospital and Laboratory of Clinical and Visual Sciences of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Juan Wang
- Department of Ophthalmology of Tongji Hospital and Laboratory of Clinical and Visual Sciences of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Jingfa Zhang
- The International Eye Research Institute of the Chinese University of Hong Kong (Shenzhen), Shenzhen, 518000, China
| | - Jieping Zhang
- Department of Ophthalmology of Tongji Hospital and Laboratory of Clinical and Visual Sciences of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, 200065, China
- Department of Physiology and Pharmacology, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Xiaoliang Jin
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yanlong Bi
- Department of Ophthalmology of Tongji Hospital and Laboratory of Clinical and Visual Sciences of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Lixia Lu
- Department of Ophthalmology of Tongji Hospital and Laboratory of Clinical and Visual Sciences of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Guo-Tong Xu
- Department of Ophthalmology of Tongji Hospital and Laboratory of Clinical and Visual Sciences of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Haibin Tian
- Department of Ophthalmology of Tongji Hospital and Laboratory of Clinical and Visual Sciences of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, 200065, China
- Department of Physiology and Pharmacology, School of Medicine, Tongji University, Shanghai, 200092, China
| |
Collapse
|
8
|
El-Naby SMA, Khedr NF, El-Ashmawy NE, Ibrahim AO. Proanthocyanidin and mitoglitazone suppress lipogenesis by targeting ferroptosis in metabolic dysfunction-associated steatohepatitis. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04271-z. [PMID: 40387928 DOI: 10.1007/s00210-025-04271-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 05/05/2025] [Indexed: 05/20/2025]
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) can progress to liver cirrhosis, increasing mortality risk. The study investigates the role of ferroptosis-an inflammatory cell death mechanism-in MASH and evaluates the therapeutic effects of mitoglitazone and proanthocyanidin in targeting ferroptosis to mitigate MASH progression. Forty male albino mice were divided into five groups (n = 8): normal control (NC) fed a standard chow diet and given 2% DMSO; MASH group was maintained on MASH protocol (high fructose-high fat diet); mitoglitazone (Mito) group was kept on MASH protocol and given Mito (10 mg/kg/day); proanthocyanidin (Pro) group was kept on MASH protocol and given Pro (150 mg/kg/day); Mito + Pro co-treated group was given Mito and Pro parallel with MASH protocol, all treatments for 12 weeks. MASH induction significantly (p < 0.001) increased liver weight, liver index, serum liver enzymes (ALT & AST), serum glucose, insulin, insulin resistance (HOMA-IR), lipid profile (total cholesterol, triglycerides, LDL-C), ferroptosis biomarkers (total iron, soluble transferrin receptor-1 (sTfR1), and expression of liver acyl-CoA synthetase long-chain family member 4 (ACSL4) with diffused macrovesicular severe steatosis, and inflammatory cells infiltration in liver tissues compared to NC. However, HDL-cholesterol, ferroptosis biomarkers (liver glutathione peroxidase X4 (GPX4), and total glutathione peroxidase (GPX) activities and glutathione (GSH) content) were reduced significantly (p < 0.001) in MASH group compared to NC. On the other hand, Mito, Pro, and their combination significantly improved ferroptotic biomarkers (GSH, GPX4, sTFR1, and total iron and ACSL-4 gene expression), glucose homeostasis, lipid profile, liver enzymes, and histology compared to MASH group. Combining the insulin-sensitizing properties with targeting of ferroptosis, by the co-treatment with mitoglitazone (MSDC-0160) and proanthocyanidin, could be beneficial in inhibition of lipogenesis with retardation of MASH development in mice.
Collapse
Affiliation(s)
- Sohair M Abd El-Naby
- Biochemistry Department, Faculty of Pharmacy, Medical Campus, Tanta University, Tanta, Postal Code: 31527, Egypt
| | - Naglaa F Khedr
- Biochemistry Department, Faculty of Pharmacy, Medical Campus, Tanta University, Tanta, Postal Code: 31527, Egypt.
| | - Nahla E El-Ashmawy
- Biochemistry Department, Faculty of Pharmacy, Medical Campus, Tanta University, Tanta, Postal Code: 31527, Egypt
- Department of Pharmacology and Biochemistry, Faculty of Pharmacy, The British University in Egypt, El Sherouk, Postal Code: 11837, Egypt
| | - Amera O Ibrahim
- Biochemistry Department, Faculty of Pharmacy, Medical Campus, Tanta University, Tanta, Postal Code: 31527, Egypt.
| |
Collapse
|
9
|
Zeng B, Zhang C, Liang Y, Huang J, Li D, Liu Z, Liao H, Yang T, Liu M, Zou C, Liu D, Qin B. Single-cell RNA sequencing highlights a significant retinal Müller glial population in dry age-related macular degeneration. iScience 2025; 28:112464. [PMID: 40343286 PMCID: PMC12059717 DOI: 10.1016/j.isci.2025.112464] [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: 08/05/2024] [Revised: 12/21/2024] [Accepted: 04/14/2025] [Indexed: 05/11/2025] Open
Abstract
The main challenge in dissecting the cells and pathways involved in the pathogenesis of age-related macular degeneration (AMD) is the highly heterogeneous and dynamic nature of the retinal microenvironment. This study aimed to describe the comprehensive landscape of the dry AMD (dAMD) model and identify the key cell cluster contributing to dAMD. We identified a subset of Müller cells that express high levels of Sox2, which play crucial roles in homeostasis and neuroprotection in both mouse models of AMD and patients with dAMD. Additionally, the number of Sox2+ Müller cells decreased significantly during the progression of AMD, indicating these cells were damaged and underwent cell death. Interestingly, ferroptosis and apoptosis were identified as contributors to the damage of Sox2+ Müller cells. Our findings are potentially valuable not only for advancing the current understanding of dAMD progression but also for the development of treatment strategies through the protection of Müller cells.
Collapse
Affiliation(s)
- Bing Zeng
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China
- Shenzhen Aier Ophthalmic Technology Institute, Shenzhen, China
- Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Chuanhe Zhang
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China
- Shenzhen Aier Ophthalmic Technology Institute, Shenzhen, China
| | - Yifan Liang
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China
- Shenzhen Aier Ophthalmic Technology Institute, Shenzhen, China
| | - Jianguo Huang
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China
- Shenzhen Aier Ophthalmic Technology Institute, Shenzhen, China
| | - Deshuang Li
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China
- Shenzhen Aier Ophthalmic Technology Institute, Shenzhen, China
| | - Ziling Liu
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China
- Shenzhen Aier Ophthalmic Technology Institute, Shenzhen, China
| | - Hongxia Liao
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China
- Shenzhen Aier Ophthalmic Technology Institute, Shenzhen, China
| | - Tedu Yang
- Shenzhen Shendong Aier Eye Hospital, Shenzhen, China
| | - Muyun Liu
- National Engineering Research Center of Foundational Technologies for CGT Industry, Shenzhen Kenuo Medical Laboratory, Shenzhen, Guangdong, China
| | - Chang Zou
- Department of Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, the First Affiliated Hospital of Southern University of Science and Technology, Shenzhen People’s Hospital, Shenzhen, Guangdong, P.R. China
| | - Dongcheng Liu
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China
- Shenzhen Aier Ophthalmic Technology Institute, Shenzhen, China
- Aier School of Ophthalmology, Central South University, Changsha, China
| | - Bo Qin
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China
- Shenzhen Aier Ophthalmic Technology Institute, Shenzhen, China
- Aier School of Ophthalmology, Central South University, Changsha, China
| |
Collapse
|
10
|
Arthur P, Kandoi S, Kalvala A, Boirie B, Nathani A, Aare M, Bhattacharya S, Kulkarni T, Sun L, Lamba DA, Li Y, Singh M. Cannabidiol-Loaded Retinal Organoid-Derived Extracellular Vesicles Protect Oxidatively Stressed ARPE-19 Cells. Biomedicines 2025; 13:1167. [PMID: 40426994 PMCID: PMC12108686 DOI: 10.3390/biomedicines13051167] [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: 04/01/2025] [Revised: 05/03/2025] [Accepted: 05/06/2025] [Indexed: 05/29/2025] Open
Abstract
Background/Objectives: Age-related macular degeneration (AMD) is the third leading cause of irreversible blindness in elderly individuals aged over 50 years old. Oxidative stress plays a crucial role in the etiopathogenesis of multifactorial AMD disease. The phospholipid bilayer EVs derived from the culture-conditioned medium of human induced pluripotent stem cell (hiPSC) differentiated retinal organoids aid in cell-to-cell communication, signaling, and extracellular matrix remodeling. The goal of the current study is to establish and evaluate the encapsulation of a hydrophobic compound, cannabidiol (CBD), into retinal organoid-derived extracellular vesicles (EVs) for potential therapeutic use in AMD. Methods: hiPSC-derived retinal organoid EVs were encapsulated with CBD via sonication (CBD-EVs), and structural features were elucidated using atomic force microscopy, nanoparticle tracking analysis, and small/microRNA (miRNA) sequencing. ARPE-19 cells and oxidative-stressed (H2O2) ARPE-19 cells treated with CBD-EVs were assessed for cytotoxicity, apoptosis (MTT assay), reactive oxygen species (DCFDA), and antioxidant proteins (immunohistochemistry and Western blot). Results: Distinct miRNA cargo were identified in early and late retinal organoid-derived EVs, implicating their roles in retinal development, differentiation, and functionality. The therapeutic effects of CBD-loaded EVs on oxidative-stressed ARPE-19 cells showed greater viability, decreased ROS production, downregulated expression of inflammation- and apoptosis-related proteins, and upregulated expression of antioxidants by Western blot and immunocytochemistry. Conclusions: miRNAs are both prognostic and predictive biomarkers and can be a target for developing therapy since they regulate RPE physiology and diseases. Our findings indicate that CBD-EVs could potentially alleviate the course of AMD by activating the targeted proteins linked to the adenosine monophosphate kinase (AMPK) pathway. Implicating the use of CBD-EVs represents a novel frontline to promote long-term abstinence from drugs and pharmacotherapy development in treating AMD.
Collapse
Affiliation(s)
- Peggy Arthur
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA; (P.A.); (A.K.); (B.B.); (A.N.); (M.A.)
| | - Sangeetha Kandoi
- Department of Ophthalmology, University of California San Francisco, San Francisco, CA 94143, USA; (S.K.); (D.A.L.)
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA 94143, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Anil Kalvala
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA; (P.A.); (A.K.); (B.B.); (A.N.); (M.A.)
| | - Breana Boirie
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA; (P.A.); (A.K.); (B.B.); (A.N.); (M.A.)
| | - Aakash Nathani
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA; (P.A.); (A.K.); (B.B.); (A.N.); (M.A.)
| | - Mounika Aare
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA; (P.A.); (A.K.); (B.B.); (A.N.); (M.A.)
| | - Santanu Bhattacharya
- Department of Biochemistry and Molecular Biology, Mayo College of Medicine and Science, Jacksonville, FL 32224, USA; (S.B.); (T.K.)
- Department of Physiology and Biomedical Engineering, Mayo College of Medicine and Science, Jacksonville, FL 32224, USA
| | - Tanmay Kulkarni
- Department of Biochemistry and Molecular Biology, Mayo College of Medicine and Science, Jacksonville, FL 32224, USA; (S.B.); (T.K.)
| | - Li Sun
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Tallahassee, FL 32310, USA;
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL 32306, USA
| | - Deepak A. Lamba
- Department of Ophthalmology, University of California San Francisco, San Francisco, CA 94143, USA; (S.K.); (D.A.L.)
- Immunology and Regenerative Medicine, Genentech, South San Francisco, CA 94080, USA
| | - Yan Li
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Tallahassee, FL 32310, USA;
| | - Mandip Singh
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA; (P.A.); (A.K.); (B.B.); (A.N.); (M.A.)
| |
Collapse
|
11
|
Zhao X, Chen X, Xin X. MiR-6837-3p protected retinal epithelial cells from oxidative stress by targeting E2F6. Int Ophthalmol 2025; 45:183. [PMID: 40343605 DOI: 10.1007/s10792-025-03540-3] [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: 06/03/2024] [Accepted: 04/05/2025] [Indexed: 05/11/2025]
Abstract
AIM The mechanism of age-related macular degeneration (AMD) is a complex illness that is not fully understood. Therefore, the aim of this study was to investigate the expression patterns of miR-6837-3p in retinal epithelial cells. METHODS H2O2 was used to treat ARPE-19 cells for 2, 4 and 6 h to mimic the in vivo environment of AMD. MiR inhibitors and mimics were used to inhibit or overexpress miR-6837-3p in H2O2-treated ARPE-19 cells, respectively. Then, CCK8 assay, flow cytometry, and wound healing assays were conducted to assess the effects of miR-6837-3p on the behaviors of ARPE-19 cells, including cell growth, apoptosis, cycle progression, and migration. Finally, microRNA database prediction and luciferase reporter assays were used to demonstrate that miR-6837-3p targets the downstream gene E2F6. RESULTS H2O2 induced a decrease in cell viability and an increase in ROS levels in a time-dependent manner. Additionally, overexpression of miR-6837-3p increased cell viability and suppressed apoptosis in ARPE-19 cells treated with H2O2. Meanwhile, increased miR-6837-3p promoted cell cycle progression and cell migration of ARPE-19 cells. Finally, miR-6837-3p exerted anti-apoptosis and anti-oxidative stress effects by inhibiting the expression of E2F6 in ARPE-19 cells. CONCLUSIONS The MiR-6837-3p/E2F6 axis might be a target for the treatment of AMD to improve ARPE-19 cell function.
Collapse
Affiliation(s)
- Xin Zhao
- Department of Ophthalmology, Baogang Hospital of Inner Mongolia, Baotou, 014010, Inner Mongolia, China
| | - Xinru Chen
- Department of Ophthalmology, Baogang Hospital of Inner Mongolia, Baotou, 014010, Inner Mongolia, China
| | - Xiangyang Xin
- Department of Ophthalmology, Baotou Central Hospital, Baotou, 014040, Inner Mongolia, China.
| |
Collapse
|
12
|
Gao Q, Su Z, Pang X, Chen J, Luo R, Li X, Zhang C, Zhao Y. Overexpression of Heme Oxygenase 1 Enhances the Neuroprotective Effects of Exosomes in Subarachnoid Hemorrhage by Suppressing Oxidative Stress and Endoplasmic Reticulum Stress. Mol Neurobiol 2025; 62:6088-6101. [PMID: 39710823 DOI: 10.1007/s12035-024-04651-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 11/28/2024] [Indexed: 12/24/2024]
Abstract
AIMS This study aims to elucidate the therapeutic effects and underlying mechanisms of exosomes derived from Heme oxygenase 1 (HO-1)-overexpressing human umbilical cord mesenchymal stem cells (ExoHO-1) in a subarachnoid hemorrhage (SAH) mouse model. METHODS In this study, exosomes were identified using Western blotting, particle analysis, and transmission electron microscopy. The effect of ExoHO-1 and ExoCtrl on the neurological function of SAH mice was assessed using the Garcia scoring system, Beam balance, Rotarod test, and Morris water maze test. Neuronal apoptosis and survival were evaluated through TUNEL and Nissl staining. Levels of oxidative and endoplasmic reticulum stress were measured via immunofluorescence, Western blotting, DHE staining, enzyme-linked immunosorbent assay, and commercial kits. RESULTS HO-1-overexpressing human umbilical cord mesenchymal stem cells encapsulated HO-1 into their exosomes. ExoHO-1 significantly enhanced both short-term and long-term neurological function protection. By reducing the activation of the PERK/CHOP/Caspase12 pathway and decreasing oxidative stress levels, ExoHO-1 effectively inhibited neuronal apoptosis in the ipsilateral temporal cortex. CONCLUSION ExoHO-1 enhances the therapeutic efficacy of exosomes in SAH mice by countering neuronal apoptosis, primarily through the suppression of oxidative and endoplasmic reticulum stress.
Collapse
Affiliation(s)
- Qiongqiong Gao
- Department of neurology, The Third Affiliated Hospital, Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China
- Central Laboratory, The Third Affiliated Hospital, Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China
| | - Zhumin Su
- Department of neurology, The Third Affiliated Hospital, Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China
| | - Xiangxiong Pang
- Translational Medicine Research Center, Zhujiang Hospital of Southern Medical University, Guangzhou, 510282, China
| | - Jinshuo Chen
- Central Laboratory, The Third Affiliated Hospital, Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China
| | - Ruixiang Luo
- Central Laboratory, The Third Affiliated Hospital, Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China
| | - Xiaoyang Li
- Central Laboratory, The Third Affiliated Hospital, Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China
| | - Chi Zhang
- Central Laboratory, The Third Affiliated Hospital, Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China.
| | - Yun Zhao
- Department of neurology, The Third Affiliated Hospital, Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China.
- Translational Medicine Research Center, Zhujiang Hospital of Southern Medical University, Guangzhou, 510282, China.
| |
Collapse
|
13
|
Kwun MS, Lee DG. Ferroptosis-Like Death Induction in Saccharomyces cerevisiae by Gold Nanoparticles. J Microbiol Biotechnol 2025; 35:e2501029. [PMID: 40295204 PMCID: PMC12089944 DOI: 10.4014/jmb.2501.01029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Revised: 02/11/2025] [Accepted: 02/24/2025] [Indexed: 04/30/2025]
Abstract
Ferroptosis, a novel form of regulated cell death (RCD), has emerged as a promising therapeutic strategy for cancer treatment. While gold nanoparticles (AuNPs) are known to induce cell death and ferroptosis in combination with certain antibiotics, the mechanisms underlying ferroptosis in microorganisms remain poorly understood. This study aimed to investigate whether AuNPs induce ferroptosis-like cell death in the eukaryotic microbe Saccharomyces cerevisiae. Our findings revealed that AuNPs significantly reduced cell viability in S. cerevisiae, suggesting their ability to trigger cell death. Ferroptosis-related precursors, including intracellular iron overload and depletion of glutathione (GSH), were observed, leading to the inactivation of glutathione peroxidase (GPx). These changes were associated with the accumulation of reactive oxygen species (ROS) and lipid peroxidation, which amplified oxidative stress within the cells. Elevated ROS levels and lipid peroxidation further resulted in membrane rupture and the formation of 8-hydroxydeoxyguanosine, indicating DNA damage. Mitochondrial dysfunction, a hallmark of ferroptosis, was also evident. AuNP treatment caused mitochondrial membrane potential hyperpolarization and a reduction in mitochondrial membrane density. Unlike previously characterized forms of RCD, ferroptosis-like death in S. cerevisiae did not involve chromatin condensation, DNA fragmentation, or metacaspase activation. Finally, ferroptosis-like characteristics were confirmed using Liperfluo, a lipid ROS-specific probe. In conclusion, this study demonstrated that AuNPs can induce ferroptosis-like cell death in S. cerevisiae. These findings highlight the potential of AuNPs as antifungal agents and contribute to the broader understanding of ferroptosis in eukaryotic microbes.
Collapse
Affiliation(s)
- Min Seok Kwun
- School of Life Science, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Dong Gun Lee
- School of Life Science, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea
- Institute of Life Science and Biotechnology, Kyungpook National University, Daegu 41566, Republic of Korea
| |
Collapse
|
14
|
Yapici FI, Seidel E, Dahlhaus A, Weber J, Schmidt C, de Britto Chaves Filho A, Yang M, Nenchova M, Güngör E, Stroh J, Kotouza I, Beck J, Abdallah AT, Lackmann JW, Bebber CM, Androulidaki A, Kreuzaler P, Schulze A, Frezza C, von Karstedt S. An atlas of ferroptosis-induced secretomes. Cell Death Differ 2025:10.1038/s41418-025-01517-4. [PMID: 40281125 DOI: 10.1038/s41418-025-01517-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 04/06/2025] [Accepted: 04/09/2025] [Indexed: 04/29/2025] Open
Abstract
Cells undergoing regulated necrosis systemically communicate with the immune system via the release of protein and non-protein secretomes. Ferroptosis is a recently described iron-dependent type of regulated necrosis driven by massive lipid peroxidation. While membrane rupture occurs during ferroptosis, a comprehensive appraisal of ferroptotic secretomes and their potential biological activity has been lacking. Here, we apply a multi-omics approach to provide an atlas of ferroptosis-induced secretomes and reveal a novel function in macrophage priming. Proteins with assigned DAMP and innate immune system function, such as MIF, heat shock proteins (HSPs), and chaperones, were released from ferroptotic cells. Non-protein secretomes with assigned inflammatory function contained oxylipins as well as TCA- and methionine-cycle metabolites. Interestingly, incubation of bone marrow-derived macrophages (BMDMs) with ferroptotic supernatants induced transcriptional reprogramming consistent with priming. Indeed, exposure to ferroptotic supernatants enhanced LPS-induced cytokine production. These results define a catalog of ferroptosis-induced secretomes and identify a biological activity in macrophage priming with important implications for the fine-tuning of inflammatory processes.
Collapse
Affiliation(s)
- F Isil Yapici
- Department of Translational Genomics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Faculty of Medicine and University Hospital Cologne, CECAD Cluster of Excellence, University of Cologne, Cologne, Germany
| | - Eric Seidel
- Department of Translational Genomics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Faculty of Medicine and University Hospital Cologne, CECAD Cluster of Excellence, University of Cologne, Cologne, Germany
| | - Alina Dahlhaus
- Department of Translational Genomics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Faculty of Medicine and University Hospital Cologne, CECAD Cluster of Excellence, University of Cologne, Cologne, Germany
| | - Josephine Weber
- Department of Translational Genomics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Faculty of Medicine and University Hospital Cologne, CECAD Cluster of Excellence, University of Cologne, Cologne, Germany
| | - Christina Schmidt
- Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases (CECAD), Faculty of Medicine and University Hospital Cologne, Institute for Metabolomics in Ageing, University of Cologne, Cologne, Germany
- University of Cologne, Faculty of Mathematics and Natural Sciences, Institute for Genetics, Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany
- Institute for Computational Biomedicine, Faculty of Medicine, and Heidelberg University Hospital, Heidelberg University, Heidelberg, Germany
| | - Adriano de Britto Chaves Filho
- Division of Tumor Metabolism and Microenvironment, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Ming Yang
- Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases (CECAD), Faculty of Medicine and University Hospital Cologne, Institute for Metabolomics in Ageing, University of Cologne, Cologne, Germany
- University of Cologne, Faculty of Mathematics and Natural Sciences, Institute for Genetics, Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany
| | - Maria Nenchova
- Department of Translational Genomics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Faculty of Medicine and University Hospital Cologne, CECAD Cluster of Excellence, University of Cologne, Cologne, Germany
| | - Emre Güngör
- Department of Translational Genomics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Faculty of Medicine and University Hospital Cologne, CECAD Cluster of Excellence, University of Cologne, Cologne, Germany
| | - Jenny Stroh
- Department of Translational Genomics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Faculty of Medicine and University Hospital Cologne, CECAD Cluster of Excellence, University of Cologne, Cologne, Germany
| | - Ioanna Kotouza
- Department of Translational Genomics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Faculty of Medicine and University Hospital Cologne, CECAD Cluster of Excellence, University of Cologne, Cologne, Germany
| | - Julia Beck
- Department of Translational Genomics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Faculty of Medicine and University Hospital Cologne, CECAD Cluster of Excellence, University of Cologne, Cologne, Germany
| | - Ali T Abdallah
- Faculty of Medicine and University Hospital Cologne, CECAD Cluster of Excellence, University of Cologne, Cologne, Germany
- Institute of Medical Statistics and Computational Biology, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Jan-Wilm Lackmann
- University of Cologne, Faculty of Mathematics and Natural Sciences, Institute for Genetics, Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany
| | - Christina M Bebber
- Department of Translational Genomics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Faculty of Medicine and University Hospital Cologne, CECAD Cluster of Excellence, University of Cologne, Cologne, Germany
| | - Ariadne Androulidaki
- Department of Translational Genomics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Faculty of Medicine and University Hospital Cologne, CECAD Cluster of Excellence, University of Cologne, Cologne, Germany
| | - Peter Kreuzaler
- Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases (CECAD), Faculty of Medicine and University Hospital Cologne, Institute for Metabolomics in Ageing, University of Cologne, Cologne, Germany
| | - Almut Schulze
- Division of Tumor Metabolism and Microenvironment, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Christian Frezza
- Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases (CECAD), Faculty of Medicine and University Hospital Cologne, Institute for Metabolomics in Ageing, University of Cologne, Cologne, Germany
- University of Cologne, Faculty of Mathematics and Natural Sciences, Institute for Genetics, Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany
| | - Silvia von Karstedt
- Department of Translational Genomics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.
- Faculty of Medicine and University Hospital Cologne, CECAD Cluster of Excellence, University of Cologne, Cologne, Germany.
- Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.
| |
Collapse
|
15
|
Kolligundla LP, Sullivan KM, Mukhi D, Andrade-Silva M, Liu H, Guan Y, Gu X, Wu J, Doke T, Hirohama D, Guarnieri P, Hill J, Pullen SS, Kuo J, Inamoto M, Susztak K. Glutathione-specific gamma-glutamylcyclotransferase 1 ( CHAC1) increases kidney disease risk by modulating ferroptosis. Sci Transl Med 2025; 17:eadn3079. [PMID: 40267214 DOI: 10.1126/scitranslmed.adn3079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 08/20/2024] [Accepted: 04/03/2025] [Indexed: 04/25/2025]
Abstract
Genome-wide association studies (GWASs) have identified more than 1000 loci where genetic variants correlate with kidney function. However, the specific genes, cell types, and mechanisms influenced by these genetic variants remain largely uncharted. Here, we identified glutathione-specific gamma-glutamylcyclotransferase 1 (CHAC1) on chromosome 15 as affected by GWAS variants by analyzing human kidney gene expression and methylation information. Both CHAC1 RNA and protein were expressed in the loop of Henle region in mouse and human kidneys, and CHAC1 expression was higher in patients carrying disease risk variants. Using CRISPR technology, we created mice with a single functional copy of the Chac1 gene (Chac1+/-) that displayed no baseline phenotypic alterations in kidney structure or function. These mice demonstrated resilience to kidney disease in multiple models, including folic acid-induced nephropathy, adenine-induced chronic kidney disease, and uninephrectomy-streptozotocin-induced diabetic nephropathy. We further showed that CHAC1 plays a critical role in degrading the cellular antioxidant glutathione. Tubule cells isolated from Chac1+/- mice showed increased glutathione, decreased lipid peroxidation, improved cell viability, and protection against ferroptosis. Expression of ferroptosis-associated genes was also lower in mice with only one copy of Chac1. Higher CHAC1 protein also correlated with ferroptosis-related protein abundance in kidney biopsies from patients with kidney disease. This study positions CHAC1 as an important mediator of kidney disease that influences glutathione concentrations and ferroptosis, suggesting potential avenues to explore for the treatment of kidney diseases.
Collapse
Affiliation(s)
- Lakshmi P Kolligundla
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| | - Katie M Sullivan
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Department of Pediatrics, Medical College of Wisconsin Pediatric Nephrology, Milwaukee, WI 53226, USA
| | - Dhanunjay Mukhi
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| | - Magaiver Andrade-Silva
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| | - Hongbo Liu
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| | - Yuting Guan
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| | - Xiangchen Gu
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| | - Junnan Wu
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| | - Tomohito Doke
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| | - Daigoro Hirohama
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| | - Paolo Guarnieri
- Department of Cardiometabolic Diseases Research, Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT 06877, USA
| | - Jon Hill
- Department of Cardiometabolic Diseases Research, Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT 06877, USA
| | - Steven S Pullen
- Department of Cardiometabolic Diseases Research, Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT 06877, USA
| | - Jay Kuo
- Department of Cardiometabolic Diseases Research, Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT 06877, USA
| | | | - Katalin Susztak
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Department of Genetics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| |
Collapse
|
16
|
Saeed BI, Uthirapathy S, Kubaev A, Ganesan S, Shankhyan A, Gupta S, Joshi KK, Kariem M, Jasim AS, Ahmed JK. Ferroptosis as a key player in the pathogenesis and intervention therapy in liver injury: focusing on drug-induced hepatotoxicity. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04115-w. [PMID: 40244448 DOI: 10.1007/s00210-025-04115-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Accepted: 03/27/2025] [Indexed: 04/18/2025]
Abstract
Globally, drug-induced hepatotoxicity or drug-induced liver injury (DILI) is a serious clinical concern. Knowing the processes and patterns of cell death is essential for finding new therapeutic targets since there are not many alternatives to therapy for severe liver lesions. Excessive lipid peroxidation is a hallmark of ferroptosis, an iron-reliant non-apoptotic cell death linked to various liver pathologies. When iron is pathogenic, concomitant inflammation may exacerbate iron-mediated liver injury, and the hepatocyte necrosis that results is a key element in the fibrogenic response. The idea that dysregulated metabolic pathways and compromised iron homeostasis contribute to the development of liver injury by ferroptosis is being supported by new data. Various ferroptosis-linked genes and pathways have been linked to liver injury, although the molecular processes behind ferroptosis's pathogenicity are not well known. Here, we delve into the features of ferroptosis, the processes governing ferroptosis, and our current knowledge of iron metabolism. We also provide an overview of ferroptosis's involvement in the pathophysiology of liver injury, particularly DILI. Lastly, the therapeutic possibilities of ferroptosis targeting for liver injury management have been provided. Natural products, nanoparticles (NPs), mesenchymal stem cell (MSC), and their exosomes have attracted increasing attention among such therapeutics.
Collapse
Affiliation(s)
- Bahaa Ibrahim Saeed
- Medical Laboratory Techniques Department, College of Health and Medical Technology, University of Al-Maarif, Anbar, Iraq
| | - Subasini Uthirapathy
- Pharmacy Department, Tishk International University, Erbil, Kurdistan Region, Iraq
| | - Aziz Kubaev
- Department of Maxillofacial Surgery, Samarkand State Medical University, 18 Amir Temur Street, 140100, Samarkand, Uzbekistan.
| | - Subbulakshmi Ganesan
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Aman Shankhyan
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Sofia Gupta
- Department of Chemistry, Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, Mohali, 140307, Punjab, India
| | - Kamal Kant Joshi
- Department of Allied Science, Graphic Era Hill University, Dehradun, India
- Graphic Era Deemed to be University, Dehradun, Uttarakhand, India
| | - Muthena Kariem
- Department of Medical Analysis, Medical Laboratory Technique College, the Islamic University, Najaf, Iraq
| | - Ahmed Salman Jasim
- Radiology Techniques Department College of Health and Medical Techniques, Al-Mustaqbal University, 51001, Babylon, Iraq
| | - Jawad Kadhim Ahmed
- Department of Medical Laboratories Technology, AL-Nisour University College, Baghdad, Iraq
| |
Collapse
|
17
|
Zhao N, Li S, Wu H, Wei D, Pu N, Wang K, Liu Y, Tao Y, Song Z. Ferroptosis: An Energetic Villain of Age-Related Macular Degeneration. Biomedicines 2025; 13:986. [PMID: 40299661 PMCID: PMC12024642 DOI: 10.3390/biomedicines13040986] [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: 02/16/2025] [Revised: 04/01/2025] [Accepted: 04/14/2025] [Indexed: 05/01/2025] Open
Abstract
Iron homeostasis plays an important role in maintaining cellular homeostasis; however, excessive iron can promote the production of reactive oxygen species (ROS). Ferroptosis is iron-dependent programmed cell death that is characterized by excessive iron accumulation, elevated lipid peroxides, and the overproduction of ROS. The maintenance of iron homeostasis is contingent upon the activity of the transferrin receptor (TfR), ferritin (Ft), and ferroportin (FPn). In the retina, iron accumulation and lipid peroxidation can contribute to the development of age-related macular degeneration (AMD). This phenomenon can be explained by the occurrence of the Fenton reaction, in which the interaction between divalent iron and hydrogen peroxide leads to the generation of highly reactive hydroxyl radicals. The hydroxyl radicals exhibit a propensity to attack proteins, lipids, nucleic acids, and carbohydrates, thereby instigating oxidative damage and promoting lipid peroxidation. Ultimately, these processes culminate in cell death and retinal degeneration. In this context, a comprehensive understanding of the exact mechanisms underlying ferroptosis may hold significant importance for developing therapeutic interventions. This review summarizes recent findings on iron metabolism, cellular ferroptosis, and lipid metabolism in the aging retina. We also introduce developments in the therapeutic strategies using iron chelating agents. Further refinements of these knowledges would deepen our comprehension of the pathophysiology of AMD and advance the clinical management of degenerative retinopathy. A comprehensive search strategy was employed to identify relevant studies on the role of ferroptosis in AMD. We performed systematic searches of the PubMed and Web of Science electronic databases from inception to the current date. The keywords used in the search included "ferroptosis", "AMD", "age-related macular degeneration", "iron metabolism", "oxidative stress", and "ferroptosis pathways". Peer-reviewed articles, including original research, reviews, meta-analyses, and clinical studies, were included in this paper, with a focus on the molecular mechanisms of ferroptosis in AMDs. Studies not directly related to ferroptosis, iron metabolism, or oxidative stress in the context of AMD were excluded. Furthermore, articles that lacked sufficient data or were not peer-reviewed (e.g., conference abstracts, editorials, or opinion pieces) were not considered.
Collapse
Affiliation(s)
- Na Zhao
- Henan Eye Institute, Henan Eye Hospital, People’s Hospital of Zhengzhou University, Henan University School of Medicine, Henan Provincial People’s Hospital, Zhengzhou 450003, China; (N.Z.); (K.W.); (Y.L.)
- College of Medicine, Zhengzhou University, Zhengzhou 450001, China; (S.L.); (H.W.); (D.W.); (N.P.)
| | - Siyu Li
- College of Medicine, Zhengzhou University, Zhengzhou 450001, China; (S.L.); (H.W.); (D.W.); (N.P.)
| | - Hao Wu
- College of Medicine, Zhengzhou University, Zhengzhou 450001, China; (S.L.); (H.W.); (D.W.); (N.P.)
| | - Dong Wei
- College of Medicine, Zhengzhou University, Zhengzhou 450001, China; (S.L.); (H.W.); (D.W.); (N.P.)
| | - Ning Pu
- College of Medicine, Zhengzhou University, Zhengzhou 450001, China; (S.L.); (H.W.); (D.W.); (N.P.)
| | - Kexin Wang
- Henan Eye Institute, Henan Eye Hospital, People’s Hospital of Zhengzhou University, Henan University School of Medicine, Henan Provincial People’s Hospital, Zhengzhou 450003, China; (N.Z.); (K.W.); (Y.L.)
| | - Yashuang Liu
- Henan Eye Institute, Henan Eye Hospital, People’s Hospital of Zhengzhou University, Henan University School of Medicine, Henan Provincial People’s Hospital, Zhengzhou 450003, China; (N.Z.); (K.W.); (Y.L.)
| | - Ye Tao
- Henan Eye Institute, Henan Eye Hospital, People’s Hospital of Zhengzhou University, Henan University School of Medicine, Henan Provincial People’s Hospital, Zhengzhou 450003, China; (N.Z.); (K.W.); (Y.L.)
| | - Zongming Song
- Henan Eye Institute, Henan Eye Hospital, People’s Hospital of Zhengzhou University, Henan University School of Medicine, Henan Provincial People’s Hospital, Zhengzhou 450003, China; (N.Z.); (K.W.); (Y.L.)
| |
Collapse
|
18
|
Hansman DS, Lim K, Thomas D, Casson RJ, Peet DJ. Distinct metabolome and flux responses in the retinal pigment epithelium to cytokines associated with age-related macular degeneration: comparison of ARPE-19 cells and eyecups. Sci Rep 2025; 15:13012. [PMID: 40234500 PMCID: PMC12000464 DOI: 10.1038/s41598-025-93882-w] [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: 12/05/2024] [Accepted: 03/10/2025] [Indexed: 04/17/2025] Open
Abstract
Age-related macular degeneration (AMD) is associated with chronic inflammation of the retinal pigment epithelium (RPE) and elevated cytokines including TNFα, TGF-β, IL-6, and IL-1β. As a metabolic intermediary supporting aerobic glycolysis in the adjacent photoreceptors, the RPE's metabolic responses to inflammation and the optimal methods to study cytokine-driven metabolic programming remain unclear. We performed a rigorous comparison of ARPE-19 cells and rat eyecup metabolomes, revealing key distinctions. Rat eyecups exhibit higher levels of lactate and palmitate but depleted glutathione and high-energy nucleotides. Conversely, ARPE-19 cells are enriched with high-energy currency metabolites and the membrane phospholipid precursors phosphocholine and inositol. Both models showed contrasting responses to individual cytokines: ARPE-19 cells were more sensitive to TNFα, while eyecups responded more strongly to TGF-β2. Notably, a combined cytokine cocktail elicited stronger metabolic effects on ARPE-19 cells, more potently impacting both metabolite abundance (41 vs. 29) and glucose carbon flux (29 vs. 5), and influencing key RPE metabolites such as alanine, glycine, aspartate, proline, citrate, α-ketoglutarate, and palmitate. Overall, these findings position ARPE-19 cells as a more responsive platform for studying inflammatory cytokine effects on RPE metabolism and reveal critical RPE metabolites which may be linked with AMD pathogenesis.
Collapse
Affiliation(s)
- David S Hansman
- School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Kelly Lim
- South Australian Health and Medical Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Daniel Thomas
- South Australian Health and Medical Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Robert J Casson
- Discipline of Ophthalmology and Visual Science, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Daniel J Peet
- School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia.
| |
Collapse
|
19
|
Djordjevic NZ, Novakovic TR, Dolicanin ZC, Jovic ND, Babic GM. Maternal Thrombophilia Disrupts Fetal Redox Homeostasis. Reprod Sci 2025:10.1007/s43032-025-01863-1. [PMID: 40234328 DOI: 10.1007/s43032-025-01863-1] [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: 07/02/2024] [Accepted: 04/06/2025] [Indexed: 04/17/2025]
Abstract
Considering that the fetal redox homeostasis is a key factor for normal prenatal development, this study researched the effects of maternal thrombophilia on fetal redox homeostasis by assessing the redox profile of amniotic fluid cells and amniotic fluid during the second trimester of pregnancy. Concentration of redox biomarkers (superoxide anion, O2.-; hydrogen peroxide, H2O2; nitric oxide, NO; peroxynitrite, ONOO-; lipid peroxides, LPO, micronuclei, reduced glutathione, GSH; oxidized glutathione, GSSG) were assayed in the amniotic fluid cells and amniotic fluid of healthy pregnant women and pregnant women with thrombophilia gestational age from 16 to 18 weeks. Results of this study indicate that pregnant women with thrombophilia have significantly higher concentrations of O2.-, NO, ONOO-, and LPO but lower concentrations of H2O2, GSH, and GSSG in the amniotic fluid cells, as well as a higher concentration of GSSG in the amniotic fluid. No difference is shown in concentration of O2.-, H2O2, NO, ONOO-, LPO, and GSH in the amniotic fluid, as well as in frequency of micronuclei in the amniotic fluid cells among investigated groups of pregnant women. The present study provides the first evidence that babies born to mothers with thrombophilia in the second trimester of intrauterine life experience intense oxidative stress characterized by overproduction of O2.-, NO, ONOO-, and LPO, as well as GSH depletion.
Collapse
Affiliation(s)
- Natasa Z Djordjevic
- Department of Natural and Mathematical Sciences, State University of Novi Pazar, Vuka Karadzica 9, Novi Pazar, 36300, Serbia.
| | - Tanja R Novakovic
- Department for Cytogenetic Diagnostics, University Clinical Center Kragujevac, Zmaj Jovina 30, Kragujevac, 34000, Serbia
| | - Zana C Dolicanin
- Department of Biomedical Sciences, State University of Novi Pazar, Vuka Karadzica 9, Novi Pazar, 36300, Serbia
| | - Nikola D Jovic
- Clinic for Gynecology and Obstetrics, University Clinical Center Kragujevac, Zmaj Jovina 30, Kragujevac, 34000, Serbia
- Department of Gynecology and Obstetrics, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, Kragujevac, 34000, Serbia
| | - Goran M Babic
- Clinic for Gynecology and Obstetrics, University Clinical Center Kragujevac, Zmaj Jovina 30, Kragujevac, 34000, Serbia
- Department of Gynecology and Obstetrics, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, Kragujevac, 34000, Serbia
| |
Collapse
|
20
|
González-Llorente L, Andrés-Gasco M, Gil Aranda MA, Rabadán-Ros R, Zapata-Pérez R, Núñez-Delicado E, Menéndez-Coto N, García-González C, Baena-Huerta FJ, Coto-Montes A, Caso-Peláez E. The Hormetic Adaptative Capacity and Resilience to Oxidative Stress Is Strengthened by Exposome Enrichment with Air Cold Atmospheric Plasma: A Metabolome Targeted Follow-Up Approach. Biomedicines 2025; 13:949. [PMID: 40299663 PMCID: PMC12025095 DOI: 10.3390/biomedicines13040949] [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: 03/12/2025] [Revised: 04/08/2025] [Accepted: 04/09/2025] [Indexed: 05/01/2025] Open
Abstract
Background/Objectives: The exposome, encompassing all environmental influences on health, plays a pivotal role in oxidative stress-related diseases. Negative air ions (NAIs), generated via cold atmospheric plasma (CAP), have been proposed as potential modulators of oxidative resilience. This study aims to investigate the metabolic adaptations induced by prolonged exposure to an NAI-enriched environment in mice, focusing on its effects in oxidative stress markers and energy metabolism in liver and blood. Methods: Twenty male C57BL/6J mice were divided into four groups: two experimental groups exposed to NAI-enriched air generated by an Air Cold Atmospheric Plasma-Nanoparticle Removal (aCAP-NR) device for either 18 days (short-term, ST) or 28 days (long-term, LT), and two control groups without exposure. Targeted metabolomics was performed in whole blood and liver using ultra-high-performance liquid chromatography-mass spectrometry (UHPLC-MS). Statistical and pathway analyses were conducted to assess metabolic alterations. Results: Metabolic profiling revealed significant shifts in oxidative stress-related pathways, including enhanced glutathione metabolism, reduced lipid peroxidation, and modulation of purine metabolism. Short-term exposure led to increased mitochondrial efficiency and energy homeostasis, while long-term exposure induced adaptive metabolic reprogramming, with higher inosine levels suggesting enhanced antioxidant and anti-inflammatory responses. No adverse effects on systemic or hepatic health markers were observed. Conclusions: NAI exposure via aCAP-NR elicits a hormetic response, enhancing metabolic efficiency and resilience to oxidative stress. These findings suggest that controlled environmental enrichment with NAIs may serve as a novel non-invasive strategy for mitigating oxidative damage and improving metabolic health, as hormetic adaptative capacity and resilience to oxidative stress, warranting further translational research.
Collapse
Affiliation(s)
- Lucía González-Llorente
- UCAM HiTech Sport & Health Innovation Hub, Universidad Católica de Murcia, Guadalupe de Maciascoque, 30107 Murcia, Spain
- System and Precision Medicine Unit, Hospital Ribera Covadonga, 33204 Gijón, Asturias, Spain
| | - Miguel Andrés-Gasco
- UCAM HiTech Sport & Health Innovation Hub, Universidad Católica de Murcia, Guadalupe de Maciascoque, 30107 Murcia, Spain
- Health Sciences PhD Program, Universidad Católica de Murcia UCAM, Campus de los Jerónimos nº135, Guadalupe de Maciascoque, 30107 Murcia, Spain
| | - Macarena Alba Gil Aranda
- UCAM HiTech Sport & Health Innovation Hub, Universidad Católica de Murcia, Guadalupe de Maciascoque, 30107 Murcia, Spain
- Health Sciences PhD Program, Universidad Católica de Murcia UCAM, Campus de los Jerónimos nº135, Guadalupe de Maciascoque, 30107 Murcia, Spain
- Research Group of Metabolism and Gene Regulation, UCAM HiTech Sport & Health Innovation Hub, Universidad Católica de Murcia, Guadalupe de Maciascoque, 30107 Murcia, Spain
| | - Rubén Rabadán-Ros
- UCAM HiTech Sport & Health Innovation Hub, Universidad Católica de Murcia, Guadalupe de Maciascoque, 30107 Murcia, Spain
- Research Group of Metabolism and Gene Regulation, UCAM HiTech Sport & Health Innovation Hub, Universidad Católica de Murcia, Guadalupe de Maciascoque, 30107 Murcia, Spain
| | - Rubén Zapata-Pérez
- UCAM HiTech Sport & Health Innovation Hub, Universidad Católica de Murcia, Guadalupe de Maciascoque, 30107 Murcia, Spain
- Research Group of Metabolism and Gene Regulation, UCAM HiTech Sport & Health Innovation Hub, Universidad Católica de Murcia, Guadalupe de Maciascoque, 30107 Murcia, Spain
| | - Estrella Núñez-Delicado
- UCAM HiTech Sport & Health Innovation Hub, Universidad Católica de Murcia, Guadalupe de Maciascoque, 30107 Murcia, Spain
- Research Group of Molecular Recognition and Encapsulation (REM), Health Sciences Department, Universidad Católica de Murcia (UCAM), Campus de los Jerónimos 135, 30107 Guadalupe, Spain
| | - Nerea Menéndez-Coto
- Department of Morphology and Cell Biology, University of Oviedo, 33006 Oviedo, Asturias, Spain
- Research Group Oxidative Stress Knowledge and Advanced Research (OSKAR), Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Asturias, Spain
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), 33006 Oviedo, Asturias, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Asturias, Spain
| | - Claudia García-González
- Department of Morphology and Cell Biology, University of Oviedo, 33006 Oviedo, Asturias, Spain
- Research Group Oxidative Stress Knowledge and Advanced Research (OSKAR), Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Asturias, Spain
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), 33006 Oviedo, Asturias, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Asturias, Spain
| | - Francisco Javier Baena-Huerta
- Department of Morphology and Cell Biology, University of Oviedo, 33006 Oviedo, Asturias, Spain
- Research Group Oxidative Stress Knowledge and Advanced Research (OSKAR), Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Asturias, Spain
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), 33006 Oviedo, Asturias, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Asturias, Spain
| | - Ana Coto-Montes
- Department of Morphology and Cell Biology, University of Oviedo, 33006 Oviedo, Asturias, Spain
- Research Group Oxidative Stress Knowledge and Advanced Research (OSKAR), Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Asturias, Spain
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), 33006 Oviedo, Asturias, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Asturias, Spain
| | - Enrique Caso-Peláez
- UCAM HiTech Sport & Health Innovation Hub, Universidad Católica de Murcia, Guadalupe de Maciascoque, 30107 Murcia, Spain
- System and Precision Medicine Unit, Hospital Ribera Covadonga, 33204 Gijón, Asturias, Spain
| |
Collapse
|
21
|
Chen Z, Niu K, Li M, Deng Y, Zhang J, Wei D, Wang J, Zhao Y. GCLC desuccinylation regulated by oxidative stress protects human cancer cells from ferroptosis. Cell Death Differ 2025:10.1038/s41418-025-01505-8. [PMID: 40188196 DOI: 10.1038/s41418-025-01505-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 03/12/2025] [Accepted: 03/26/2025] [Indexed: 04/07/2025] Open
Abstract
Tumor cells evolve strong antioxidant capacities to counteract the abnormal high level of reactive oxygen species (ROS) in the tumor microenvironment. Glutamate-cysteine ligase catalyzing subunit (GCLC) for synthesis of antioxidant glutathione (GSH) represents the key enzyme to maintain redox homeostasis of tumor cells, however, whether its activity is regulated by posttranslational modifications, such as succinylation, remains to be clarified. Here, we demonstrate the existence of succinylation modification on GCLC by in vitro and in vivo assays. NAD-dependent deacetylase Sirtuin-2 (SIRT2) serves as the desuccinylase and catalyzes GCLC desuccinylation at sites of K38, K126, and K326. Specifically, GCLC directly interacts with SIRT2, which can be substantially enhanced upon ROS treatment. This strengthened association results in GCLC desuccinylation and activation, consequently promoting GSH synthesis and rendering cancer cells resistant to ferroptosis induction. Depletion of SIRT2 decreases total GSH level and meanwhile increases the cellular susceptibility to ferroptosis, which can mostly be rescued by introducing wild-type GCLC, but not its 3K-E mutant. We further demonstrated that histone acetyltransferase P300 serves as the succinyltransferase of GCLC, and their association is remarkably decreased after ROS treatment. Thus, SIRT2-regulated GCLC succinylation represents an essential signaling axis for cancer cells to maintain their redox balance in coping with oxidative stress-induced ferroptosis.
Collapse
Affiliation(s)
- Zixiang Chen
- China National Center for Bioinformation, Beijing, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Kaifeng Niu
- China National Center for Bioinformation, Beijing, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Mengge Li
- China National Center for Bioinformation, Beijing, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yuchun Deng
- China National Center for Bioinformation, Beijing, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Ji Zhang
- China National Center for Bioinformation, Beijing, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Di Wei
- China National Center for Bioinformation, Beijing, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Jiaqi Wang
- China National Center for Bioinformation, Beijing, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yongliang Zhao
- China National Center for Bioinformation, Beijing, China.
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
22
|
Hayashi Y, Saeki A, Yoshimoto S, Yano E, Yasukochi A, Kimura S, Utsunomiya T, Minami K, Aso Y, Hatakeyama Y, Lo YC, Hirata M, Jimi E, Kawakubo-Yasukochi T. 4-Octyl Itaconate Attenuates Cell Proliferation by Cellular Senescence via Glutathione Metabolism Disorders and Mitochondrial Dysfunction in Melanoma. Antioxid Redox Signal 2025; 42:547-565. [PMID: 39931827 DOI: 10.1089/ars.2024.0629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/16/2025]
Abstract
Aims: Itaconate (IA) is synthesized in the citric acid cycle via cis-aconitate decarboxylase (ACOD1); however, its biological significance in cancer remains incompletely understood. In previous studies, 4-octyl itaconate (OI) was used as a membrane-permeable form of IA, but little detailed verification of the difference in biological activities between IA and OI exists. Here, we investigated the direct effects of IA and OI on melanoma. Results: The proliferation of melanoma cells treated with OI was significantly suppressed in vitro, and our transcriptomic analysis revealed drastic changes in the expression of glutathione metabolism-related genes in OI-treated cells. Indeed, OI treatment decreased intracellular glutathione levels, followed by increased production of reactive oxygen species and expression of γH2AX, a marker of DNA damage, and β-galactosidase, a marker of cellular senescence. We further showed that the mitochondrial respiratory capacity in B16 cells was significantly decreased by OI treatment. OI administration also suppressed the growth of B16 tumor transplants in vivo, and the expression of γH2AX was increased in tumor tissues of OI-treated mice. In addition, minimal effects of OI treatment were observed in melanocytes and normal tissues. We also proved that not only exogenous IA, which enters intracellularly, but also endogenous IA has little effect on melanoma proliferation activity, via an investigation using Acod1-overexpressing transfectants and Acod1-deficient mice. Conclusion: This work revealed that OI disrupts the antioxidant system via the collapse of glutathione metabolism and inhibits cancer cell proliferation. Antioxid. Redox Signal. 42, 547-565.
Collapse
Affiliation(s)
- Yoshikazu Hayashi
- OBT Research Center, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
- Division of Functional Structure, Department of Morphological Biology, Fukuoka Dental College, Fukuoka, Japan
- Oral Medicine Research Center, Fukuoka Dental College, Fukuoka, Japan
| | - Ayaka Saeki
- OBT Research Center, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Shohei Yoshimoto
- Oral Medicine Research Center, Fukuoka Dental College, Fukuoka, Japan
- Division of Biomedical Sciences, Department of Morphological Biology, Section of Pathology, Fukuoka Dental College, Fukuoka, Japan
| | - Ena Yano
- OBT Research Center, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Atsushi Yasukochi
- Division of Maxillofacial Diagnostic and Surgical Sciences, Section of Oral and Maxillofacial Oncology, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Soi Kimura
- OBT Research Center, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Tomoe Utsunomiya
- OBT Research Center, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
- Division of Functional Structure, Department of Morphological Biology, Fukuoka Dental College, Fukuoka, Japan
| | - Kento Minami
- OBT Research Center, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Yuji Aso
- Department of Biobased Materials Science, Kyoto Institute of Technology, Kyoto, Japan
| | - Yuji Hatakeyama
- Division of Functional Structure, Department of Morphological Biology, Fukuoka Dental College, Fukuoka, Japan
| | - Yi-Chen Lo
- Institute of Food Sciences and Technology, National Taiwan University, Taipei, Taiwan
| | - Masato Hirata
- Oral Medicine Research Center, Fukuoka Dental College, Fukuoka, Japan
| | - Eijiro Jimi
- OBT Research Center, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
- Laboratory of Molecular and Cellular Biochemistry, Division of Oral Biological Sciences, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | | |
Collapse
|
23
|
Kozieł S, Wojtala D, Szmitka M, Lesiów M, Ziółkowska A, Sawka J, Del Carpio E, Crans DC, Komarnicka UK. Half-Sandwich Organometallic Ir(III) and Ru(II) Compounds and their Interactions with Biomolecules. Chempluschem 2025; 90:e202400621. [PMID: 39878055 DOI: 10.1002/cplu.202400621] [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: 09/24/2024] [Revised: 12/19/2024] [Indexed: 01/31/2025]
Abstract
This review highlights how a Ir(III) and Ru(II) coordination complexes can change theirs cytotoxic activity by interacting with a biomolecules such as deoxyribonucleic acid (DNA), human albumins (HSA), nicotinamide adenine dinucleotide (NADH), and glutathione (GSH). We have selected biomolecules (DNA, NADH, GSH, and HSA) based on their significant biological roles and importance in cellular processes. Moreover, this review may provide useful information for the development of new half-sandwich Ir(III) and Ru(II) complexes with desired properties and relevant biological activities. Additionally, the examples discussed here may help us better understand what happens to a metal-based drug once it enters the body.
Collapse
Affiliation(s)
- Sandra Kozieł
- Faculty of Chemistry, University of Wrocław, ul. F. Joliot-Curie 14, 50-383, Wrocław, Poland
- Department of Chemistry, Colorado State University, 80523, Fort Collins, CO, USA
| | - Daria Wojtala
- Faculty of Chemistry, University of Wrocław, ul. F. Joliot-Curie 14, 50-383, Wrocław, Poland
| | - Magdalena Szmitka
- Faculty of Chemistry, University of Wrocław, ul. F. Joliot-Curie 14, 50-383, Wrocław, Poland
| | - Monika Lesiów
- Faculty of Chemistry, University of Wrocław, ul. F. Joliot-Curie 14, 50-383, Wrocław, Poland
| | - Aleksandra Ziółkowska
- Faculty of Chemistry, University of Wrocław, ul. F. Joliot-Curie 14, 50-383, Wrocław, Poland
- Faculty of Chemistry, Wroclaw University of Science and Technology, Wybrzeże Wyspiańskiego 27, 50-370, Wrocław, Poland
| | - Jacek Sawka
- Faculty of Chemistry, University of Wrocław, ul. F. Joliot-Curie 14, 50-383, Wrocław, Poland
| | - Edgar Del Carpio
- Department of Chemistry, Colorado State University, 80523, Fort Collins, CO, USA
- Facultad de Farmacia, Escuela "Dr. Jesús María Bianco", Universidad Central de Venezuela (UCV), Paseo Los Ilustres, Los Chaguaramos, 1050, Caracas, Venezuela
| | - Debbie C Crans
- Department of Chemistry, Colorado State University, 80523, Fort Collins, CO, USA
- Cell and Molecular Biology Program, Colorado State University, 80523, Fort Collins, CO, USA
| | - Urszula K Komarnicka
- Faculty of Chemistry, University of Wrocław, ul. F. Joliot-Curie 14, 50-383, Wrocław, Poland
- Department of Chemistry, Colorado State University, 80523, Fort Collins, CO, USA
| |
Collapse
|
24
|
Fu HJ, Zhou XY, Qin DL, Qiao Q, Wang QZ, Li SY, Zhu YF, Li YP, Zhou JM, Cai H, Huang FH, Yu L, Wang L, Wu AG, Wu JM, Zhou XG. Inhibition of Ferroptosis Delays Aging and Extends Healthspan Across Multiple Species. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2416559. [PMID: 40162524 DOI: 10.1002/advs.202416559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 03/11/2025] [Indexed: 04/02/2025]
Abstract
Ferroptosis, a form of iron-dependent cell death, plays a pivotal role in age-related diseases; yet, its impact on cellular senescence and healthspan in mammals remains largely unexplored. This study identifies ferroptosis as a key regulator of cellular senescence, showing that its inhibition can significantly delay aging and extend healthspan across multiple species. During cellular senescence, ferroptosis is progressively exacerbated, marked by increased lipid peroxidation, oxidative stress, and diminished glutathione peroxidase 4 (GPX4) levels. Ferroptosis inducers such as Erastin and RSL3 accelerate senescence; while, inhibitors such as liproxstatin-1 (Lip-1) and ferrostatin-1 (Fer-1) effectively mitigate both chemically and replicatively induced senescence. In vivo, Fer-1 extends lifespan and healthspan in Caenorhabditis elegans, enhances motor function, preserves tissue integrity, and mitigates cognitive decline in both prematurely and naturally aged mice. These effects are attributed to Fer-1's upregulation of GPX4 and inhibition of ferroptosis. Notably, long-term Fer-1 treatment (over 6 months) does not adversely affect body weight or induce aging-related tissue damage but rejuvenates hematological parameters. These findings establish ferroptosis as a critical player in aging dynamics and highlight its inhibition as a promising strategy to extend healthspan and lifespan, providing valuable insights for translational approaches to combat aging and age-related decline.
Collapse
Affiliation(s)
- Hai-Jun Fu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Xing-Yue Zhou
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Da-Lian Qin
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Qan Qiao
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Qiao-Zhi Wang
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Shi-Ying Li
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Yun-Fei Zhu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Ya-Ping Li
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Jiang-Min Zhou
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Hui Cai
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Fei-Hong Huang
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Lu Yu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Long Wang
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - An-Guo Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Jian-Ming Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Xiao-Gang Zhou
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
- Central Nervous System Drug Key Laboratory of Sichuan Province, Luzhou, Sichuan, 646000, China
| |
Collapse
|
25
|
El-Sehrawy AAMA, Rashid TA, Ullah MI, Uthirapathy S, Ganesan S, Singh A, Devi A, Joshi KK, Jasim AS, Kadhim AJ. Cutting edge: ferroptosis in metabolic dysfunction-associated steatotic liver disease (MASLD) pathogenesis and therapy. Funct Integr Genomics 2025; 25:71. [PMID: 40131513 DOI: 10.1007/s10142-025-01579-0] [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: 01/06/2025] [Revised: 02/27/2025] [Accepted: 03/11/2025] [Indexed: 03/27/2025]
Abstract
Ferroptosis denotes a distinct form of controlled cell death marked by substantial iron buildup and significant lipid peroxidation, playing a crucial role in several disease processes linked to cell death. Given the liver's essential functions in iron and lipid metabolism and its vulnerability to oxidative damage, more research has investigated the correlation between ferroptosis and numerous hepatic diseases, including metabolic dysfunction-associated steatotic liver disease (MASLD), formerly known as non-alcoholic fatty liver disease (NAFLD). NAFLD has arisen as a worldwide public health concern due to elevated morbidity and high death rates. The pathogenesis of MASLD remains incompletely elucidated. Recent data suggests that ferroptosis is crucial in the pathophysiology of MASLD; nevertheless, the specific processes by which ferroptosis influences MASLD remain unclear. The present review summarizes the molecular processes of ferroptosis and its intricate regulatory networks, outlines the differing impacts of ferroptosis at different stages of MASLD, and examines possible approaches targeting ferroptosis for the therapy of MASLD, suggesting a novel approach for its management.
Collapse
Affiliation(s)
| | - Teeba Ammar Rashid
- Medical Laboratory Techniques Department, College of Health and Medical Technology, University of Al-Maarif, Anbar, Iraq.
| | - Muhammad Ikram Ullah
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, 72388, Aljouf, Saudi Arabia
| | - Subasini Uthirapathy
- Pharmacy Department, Tishk International University, Erbil, Kurdistan Region, Iraq
| | - Subbulakshmi Ganesan
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Abhayveer Singh
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India
| | - Anita Devi
- Department of Chemistry, Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, Mohali, 140307, Punjab, India
| | - Kamal Kant Joshi
- Department of Allied Science, Graphic Era Hill University, Dehradun, 248002, Uttarakhand, India
- Graphic Era Deemed to Be University, Dehradun, Uttarakhand, India
| | - Ahmed Salman Jasim
- Radiology Techniques Department, College of Health and Medical Techniques, Al-Mustaqbal University, 5100, Babylon, Iraq
| | - Abed J Kadhim
- Department of Medical Engineering, Al-Nisour University College, Baghdad, Iraq
| |
Collapse
|
26
|
Li X, Li W, Xie X, Fang T, Yang J, Shen Y, Wang Y, Wang H, Tao L, Zhang H. ROS Regulate Rotenone-induced SH-SY5Y Dopamine Neuron Death Through Ferroptosis-mediated Autophagy and Apoptosis. Mol Neurobiol 2025:10.1007/s12035-025-04824-6. [PMID: 40097764 DOI: 10.1007/s12035-025-04824-6] [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: 09/04/2024] [Accepted: 03/06/2025] [Indexed: 03/19/2025]
Abstract
Rotenone, a plant-derived natural insecticide, is widely used to induce Parkinson's disease (PD) models. However, the mechanisms of rotenone-induced cell death remain unclear. Here, we found that rotenone (0.01, 0.1, or 1 μmol/L) suppressed SH-SY5Y dopamine neuron viability and led to PD-like pathological changes, such as reduced tyrosine hydroxylase (TH) but increased α-synuclein. Rotenone increased the levels of intracellular reactive oxygen species (ROS) and mitochondrial ROS, as well as the levels of the antioxidants nuclear factor E2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1), ultimately resulting in oxidative stress. Moreover, rotenone significantly downregulated the expression of GPX4 and xCT but upregulated the expression of COX2 and NCOA4, which are markers of ferroptosis. Furthermore, rotenone decreased phosphorylated mTOR level but increased Beclin-1, ATG5, LC3 and p62 expression, suggesting that rotenone enhances autophagy and reduces autophagy flux. Additionally, rotenone reduced Bcl-2 levels and the mitochondrial membrane potential (MMP) while promoting BAX and Caspase-3 expression, thus initiating cell apoptosis. N-acetylcysteine (NAC), a ROS scavenger, and ferrostatin-1 (Fer-1) and deferoxamine (DFO), two ferroptosis inhibitors, significantly eliminated rotenone-induced autophagy and apoptosis. Moreover, ML385, a specific inhibitor of Nrf2, suppressed rotenone-induced ferroptosis. Our results demonstrated that ROS might mediate rotenone-induced PD-like pathological changes by regulating iron death, autophagy, and apoptosis. Inhibiting ferroptosis blocked the rotenone-induced increase in autophagy and apoptosis. Thus, the ability of ROS to regulate rotenone-induced death through autophagy and apoptosis is dependent on ferroptosis. The findings require validation in multiple neuronal cell lines and in vivo.
Collapse
Affiliation(s)
- Xinying Li
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China
| | - Weiran Li
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China
- Department of Clinical Medicine, School of Medicine, Qinghai University, Xining, China
| | - Xinying Xie
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China
| | - Ting Fang
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China
| | - Jingwen Yang
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China
| | - Yue Shen
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China
| | - Yicheng Wang
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China
| | - Hongyan Wang
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China
| | - Liqing Tao
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China
| | - Heng Zhang
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China.
| |
Collapse
|
27
|
Liang LF, Zhao JQ, Wu YF, Chen HJ, Huang T, Lu XH. SMAD specific E3 ubiquitin protein ligase 1 accelerates diabetic macular edema progression by WNT inhibitory factor 1. World J Diabetes 2025; 16:101328. [PMID: 40093288 PMCID: PMC11885972 DOI: 10.4239/wjd.v16.i3.101328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 12/06/2024] [Accepted: 01/02/2025] [Indexed: 01/21/2025] Open
Abstract
BACKGROUND Diabetic macular edema (DME) is the most common cause of vision loss in people with diabetes. Tight junction disruption of the retinal pigment epithelium (RPE) cells has been reported to induce DME development. SMAD-specific E3 ubiquitin protein ligase (SMURF) 1 was associated with the tight junctions of cells. However, the mechanism of SMURF1 in the DME process remains unclear. AIM To investigate the role of SMURF1 in RPE cell tight junction during DME. METHODS ARPE-19 cells treated with high glucose (HG) and desferrioxamine mesylate (DFX) for establishment of the DME cell model. DME mice models were constructed by streptozotocin induction. The trans-epithelial electrical resistance and permeability of RPE cells were analyzed. The expressions of tight junction-related and autophagy-related proteins were determined. The interaction between insulin like growth factor 2 mRNA binding protein 2 (IGF2BP2) and SMURF1 mRNA was verified by RNA immunoprecipitation (RIP). SMURF1 N6-methyladenosine (m6A) level was detected by methylated RIP. RESULTS SMURF1 and vascular endothelial growth factor (VEGF) were upregulated in DME. SMURF1 knockdown reduced HG/DFX-induced autophagy, which protected RPE cell tight junctions and ameliorated retinal damage in DME mice. SMURF1 activated the Wnt/β-catenin-VEGF signaling pathway by promoting WNT inhibitory factor (WIF) 1 ubiquitination and degradation. IGF2BP2 upregulated SMURF1 expression in an m6A modification-dependent manner. CONCLUSION M6A-modified SMURF1 promoted WIF1 ubiquitination and degradation, which activated autophagy to inhibit RPE cell tight junctions, ultimately promoting DME progression.
Collapse
Affiliation(s)
- Li-Fang Liang
- Department of Ophthalmology, Zhujiang Hospital, Southern Medical University, Guangzhou 515282, Guangdong Province, China
| | - Jia-Qi Zhao
- Department of Ophthalmology, Zhujiang Hospital, Southern Medical University, Guangzhou 515282, Guangdong Province, China
| | - Yi-Fei Wu
- Department of Ophthalmology, Zhujiang Hospital, Southern Medical University, Guangzhou 515282, Guangdong Province, China
| | - Hui-Jie Chen
- Department of Ophthalmology, Zhujiang Hospital, Southern Medical University, Guangzhou 515282, Guangdong Province, China
| | - Tian Huang
- Department of Ophthalmology, Zhujiang Hospital, Southern Medical University, Guangzhou 515282, Guangdong Province, China
| | - Xiao-He Lu
- Department of Ophthalmology, Zhujiang Hospital, Southern Medical University, Guangzhou 515282, Guangdong Province, China
| |
Collapse
|
28
|
Sun L, Niu Y, Liao B, Liu L, Peng Y, Li K, Chen X, Chen Q, Bai D. CUR-PDT induces ferroptosis of RA-FLS via the Nrf2/xCT/GPX4 pathway to inhibit proliferation in rheumatoid arthritis. Inflamm Res 2025; 74:53. [PMID: 40085199 DOI: 10.1007/s00011-025-02019-2] [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: 10/11/2024] [Revised: 02/23/2025] [Accepted: 02/25/2025] [Indexed: 03/16/2025] Open
Abstract
OBJECTIVE Ferroptosis is a non-apoptotic cell death mechanism driven by reactive oxygen species (ROS) and iron. Its significance in inflammatory arthritis is well-established, but its role in rheumatoid arthritis (RA) remains uncertain. This study aimed to clarify the mechanisms through which curcumin-mediated photodynamic therapy (CUR-PDT) triggers ferroptosis in RA fibroblast-like synoviocytes (FLSs). METHODS In vivo studies using a collagen-induced arthritis (CIA) rat model evaluated CUR-PDT effects on joint edema, synovial inflammation, and fibrosis through paw volume measurements and H&E and Masson's trichrome staining. The expression of Nrf2, xCT, and GPX4 in FLSs was assessed via ELISA and immunohistochemistry. In vitro, MH7A cells treated with TNF-α were analyzed for viability, proliferation, invasion, and migration through various assays. Mitochondrial potential and morphology were examined using JC-1 staining and transmission electron microscopy (TEM). Ferroptosis biomarkers, including ROS, malondialdehyde (MDA), glutathione (GSH), superoxide dismutase (SOD), and Fe2+ levels, were measured. Nrf2, xCT, and GPX4 levels were quantified with RT-qPCR, Western blot, and immunofluorescence. Small interfering RNA (siRNA) was employed to knock down Nrf2 to validate the effect of CUR-PDT on ferroptosis in RA-FLS. RESULTS The CUR-PDT therapy markedly reduced joint inflammation and collagen deposition in the synovial tissue of CIA rats. It effectively alleviated both inflammation and hyperplasia. Moreover, this therapy facilitated ferroptosis within the synovial tissue. In vitro analyses indicated that CUR-PDT diminished the proliferation and viability of FLSs, resulting in increased ROS levels in the cells. This cascade initiated ferroptosis, as evidenced by decreased glutathione, heightened iron concentrations, mitochondrial shrinkage, and reduced mitochondrial membrane potential. Crucially, the expression of xCT and GPX4 was significantly lowered. Interestingly, knocking down the Nrf2 gene amplified this effect, leading to an even greater reduction in xCT and GPX4 expression. In this context, RA-FLSs exhibited more pronounced ferroptotic traits, including diminished proliferation, invasion, and migration. CONCLUSIONS This study elucidated a mechanism by which CUR-PDT triggers ferroptosis in FLSs through the downregulation of the Nrf2-xCT-GPX4 signaling cascade, thereby effectively hindering the progression of RA and emphasizing the importance of targeting Nrf2 in disease advancement.
Collapse
Affiliation(s)
- Lihua Sun
- Department of Rehabilitation Medicine, Key Laboratory of Physical Medicine and Precision Rehabilitation of Chongqing Municipal Health Commission, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Yajuan Niu
- Department of Rehabilitation Medicine, Key Laboratory of Physical Medicine and Precision Rehabilitation of Chongqing Municipal Health Commission, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Bo Liao
- Department of Rehabilitation Medicine, Key Laboratory of Physical Medicine and Precision Rehabilitation of Chongqing Municipal Health Commission, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Linlin Liu
- Department of Rehabilitation Medicine, Key Laboratory of Physical Medicine and Precision Rehabilitation of Chongqing Municipal Health Commission, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Yi Peng
- Department of Rehabilitation Medicine, Key Laboratory of Physical Medicine and Precision Rehabilitation of Chongqing Municipal Health Commission, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Kaiting Li
- Department of Rehabilitation Medicine, Key Laboratory of Physical Medicine and Precision Rehabilitation of Chongqing Municipal Health Commission, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Xinhua Chen
- Department of Rehabilitation Medicine, Key Laboratory of Physical Medicine and Precision Rehabilitation of Chongqing Municipal Health Commission, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Qing Chen
- Department of Rehabilitation Medicine, Key Laboratory of Physical Medicine and Precision Rehabilitation of Chongqing Municipal Health Commission, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China.
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China.
| | - Dingqun Bai
- Department of Rehabilitation Medicine, Key Laboratory of Physical Medicine and Precision Rehabilitation of Chongqing Municipal Health Commission, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China.
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China.
| |
Collapse
|
29
|
Chen Y, Jiang F, Zeng Y, Zhang M. The role of retinal pigment epithelial senescence and the potential of senotherapeutics in age-related macular degeneration. Surv Ophthalmol 2025:S0039-6257(25)00053-0. [PMID: 40089029 DOI: 10.1016/j.survophthal.2025.03.004] [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: 10/14/2024] [Revised: 03/03/2025] [Accepted: 03/10/2025] [Indexed: 03/17/2025]
Abstract
Age-related macular degeneration (AMD) is a leading cause of visual impairment in the aging population. Evidence showing the presence of cellular senescence in retinal pigment epithelium (RPE) of patients with AMD is growing. Senescent RPE play a pivotal role in its pathogenesis. The senescent RPE suffers from structural and functional alterations and disruption of the surrounding microenvironment due to the development of the senescence-associated secretory phenotype, which contributes to metabolic dysfunctions and inflammatory responses in the retina. Senotherapeutics, including senolytics, senomorphics and others, are novel treatments targeting senescent cells and are promising treatments for AMD. As senotherapeutic targets are being developed, it is promising that the burden of AMD could be decreased.
Collapse
Affiliation(s)
- Yingying Chen
- Department of Ophthalmology and Laboratory of Macular Disease, West China Hospital, Sichuan University, China.
| | - Feipeng Jiang
- Department of Ophthalmology and Laboratory of Macular Disease, West China Hospital, Sichuan University, China.
| | - Yue Zeng
- Department of Ophthalmology and Laboratory of Macular Disease, West China Hospital, Sichuan University, China.
| | - Meixia Zhang
- Department of Ophthalmology and Laboratory of Macular Disease, West China Hospital, Sichuan University, China.
| |
Collapse
|
30
|
Ye Y, Abulizi A, Zhang Y, Lu F, An Y, Ren C, Zhang H, Wang Y, Lin D, Lu D, Li M, Yang B. Ganoderic Acid Ameliorates Ulcerative Colitis by Improving Intestinal Barrier Function via Gut Microbiota Modulation. Int J Mol Sci 2025; 26:2466. [PMID: 40141109 PMCID: PMC11942431 DOI: 10.3390/ijms26062466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 03/04/2025] [Accepted: 03/08/2025] [Indexed: 03/28/2025] Open
Abstract
Ulcerative colitis (UC) is a chronic and recurrent gastrointestinal disease that affects millions of humans worldwide and imposes a huge social and economic burden. It is necessary to find safe and efficient drugs for preventing and treating UC. The aim of this study was to determine whether ganoderic acid (GA), the main bioactive components of Ganoderma lucidum, has preventive and therapeutic effect on UC in a dextran sulfate sodium (DSS)-induced UC mouse model. Our experimental results showed that GA significantly ameliorated the body weight loss and disease activity index (DAI) of UC mice. GA significantly restored 11% of the colon length and 69% of the spleen index compared to UC mice. GA significantly decreased the intestinal inflammatory response and improved the barrier function of the intestine by upregulating the tight junction proteins Zonula occludens-1 (ZO-1), occludin and claudin-1. A co-housing experiment showed that gut microbiota accounted for the therapeutic activity of GA on UC, which was confirmed by fecal microbiota transplantation from GA-treated mice to the UC mice. Furthermore, 16S rDNA high-throughput sequencing of fecal bacteria showed that GA significantly enriched the abundance of Lactobacillus, Oscillospira, Odoribacter and Ruminococcus, which were positively correlated with colon length. Furthermore, this study found the functional metabolites, including Indole-3-acetaldehyde (IAAld), Glutamine (Gln) and Glutathione (GSH), reduced barrier damage in the Caco-2 cell model. In conclusion, this study suggests that GA could ameliorate UC by improving intestinal barrier function via modulating gut microbiota and associated metabolites.
Collapse
Affiliation(s)
- Yuwei Ye
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; (Y.Y.); (A.A.); (Y.Z.); (F.L.); (Y.A.); (C.R.); (H.Z.); (Y.W.)
| | - Abudumijiti Abulizi
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; (Y.Y.); (A.A.); (Y.Z.); (F.L.); (Y.A.); (C.R.); (H.Z.); (Y.W.)
| | - Yukun Zhang
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; (Y.Y.); (A.A.); (Y.Z.); (F.L.); (Y.A.); (C.R.); (H.Z.); (Y.W.)
| | - Feng Lu
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; (Y.Y.); (A.A.); (Y.Z.); (F.L.); (Y.A.); (C.R.); (H.Z.); (Y.W.)
| | - Yongpan An
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; (Y.Y.); (A.A.); (Y.Z.); (F.L.); (Y.A.); (C.R.); (H.Z.); (Y.W.)
| | - Chaoqun Ren
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; (Y.Y.); (A.A.); (Y.Z.); (F.L.); (Y.A.); (C.R.); (H.Z.); (Y.W.)
| | - Hang Zhang
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; (Y.Y.); (A.A.); (Y.Z.); (F.L.); (Y.A.); (C.R.); (H.Z.); (Y.W.)
| | - Yiming Wang
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; (Y.Y.); (A.A.); (Y.Z.); (F.L.); (Y.A.); (C.R.); (H.Z.); (Y.W.)
| | - Dongmei Lin
- JUNCAO Technology Research Institute, Fujian Agriculture and Forestry University, Fuzhou 350002, China;
| | - Dan Lu
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking University, Beijing 100191, China;
| | - Min Li
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; (Y.Y.); (A.A.); (Y.Z.); (F.L.); (Y.A.); (C.R.); (H.Z.); (Y.W.)
| | - Baoxue Yang
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; (Y.Y.); (A.A.); (Y.Z.); (F.L.); (Y.A.); (C.R.); (H.Z.); (Y.W.)
| |
Collapse
|
31
|
He Y, Lin Y, Song J, Song M, Nie X, Sun H, Xu C, Han Z, Cai J. From mechanisms to medicine: Ferroptosis as a Therapeutic target in liver disorders. Cell Commun Signal 2025; 23:125. [PMID: 40055721 PMCID: PMC11889974 DOI: 10.1186/s12964-025-02121-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 02/22/2025] [Indexed: 05/13/2025] Open
Abstract
In recent 10 years, ferroptosis has become a hot research direction in the scientific research community as a new way of cell death. Iron toxicity accumulation and lipotoxicity are unique features. Several studies have found that ferroptosis is involved in the regulation of the hepatic microenvironment and various hepatic metabolisms, thereby mediating the progression of related liver diseases. For example, NRF2 and FSP1, as important regulatory proteins of ferroptosis, are involved in the development of liver tumors and liver failure. In this manuscript, we present the mechanisms involved in ferroptosis, the concern of ferroptosis with the liver microenvironment and the progression of ferroptosis in various liver diseases. In addition, we summarize recent clinical advances in targeted ferroptosis therapy for related diseases. We expect that this manuscript can provide a new perspective for clinical treatment of related diseases.
Collapse
Affiliation(s)
- Yuqi He
- Department of Transfusion, The Lu'an Hospital Affiliated to Anhui Medical University, The Lu'an People's Hospital, Lu'an, Anhui Province, China
| | - Yumeng Lin
- Health Management Center, Nanjing Tongren Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Jinfeng Song
- Department of Transfusion, The Lu'an Hospital Affiliated to Anhui Medical University, The Lu'an People's Hospital, Lu'an, Anhui Province, China
| | - Mingzhu Song
- Department of Transfusion, The Lu'an Hospital Affiliated to Anhui Medical University, The Lu'an People's Hospital, Lu'an, Anhui Province, China
| | - Xiaoxia Nie
- Department of Transfusion, The Lu'an Hospital Affiliated to Anhui Medical University, The Lu'an People's Hospital, Lu'an, Anhui Province, China
| | - Hong Sun
- Department of Transfusion, The Lu'an Hospital Affiliated to Anhui Medical University, The Lu'an People's Hospital, Lu'an, Anhui Province, China
| | - Changyun Xu
- Department of Transfusion, The Lu'an Hospital Affiliated to Anhui Medical University, The Lu'an People's Hospital, Lu'an, Anhui Province, China
| | - Zhongyu Han
- Department of Transfusion, The Lu'an Hospital Affiliated to Anhui Medical University, The Lu'an People's Hospital, Lu'an, Anhui Province, China.
| | - Juan Cai
- Department of Transfusion, The Lu'an Hospital Affiliated to Anhui Medical University, The Lu'an People's Hospital, Lu'an, Anhui Province, China.
| |
Collapse
|
32
|
Yan Z, Bai Y, Zhang S, Kong L, Wang Y, Sun H, Li Y, Qiu L, Zhang R, Jiang P, Zhao D, Chen Z, Li Y, Pang H, Wang J. Quasi Fe MIL-53 nanozyme inducing ferroptosis and immunogenic cell death for cancer immunotherapy. Nat Commun 2025; 16:2290. [PMID: 40055308 PMCID: PMC11889140 DOI: 10.1038/s41467-025-57542-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 02/26/2025] [Indexed: 05/13/2025] Open
Abstract
Nanozymes offer diverse therapeutic potentials for cancer treatment which is dependent on the development of nanomaterials. Quasi-metal-organic framework is a class of metal-organic framework-derived nanomaterials with a transition state from metal-organic frameworks towards metal oxide featuring porous structure and high activity. Herein an iron-based quasi-metal-organic framework nanozyme Q-MIL-53(Fe) is reported via a controlled deligandation strategy, exhibiting enhanced peroxidase-/catalase-mimic activity and glutathione depletion capacity, whose underlying mechanisms are studied via density functional theory calculations. Q-MIL-53(Fe) demonstrates biocompatibility and superior antitumor efficacy compared to pristine MIL-53(Fe). It can activate antitumor immune response by inducing ferroptosis and immunogenic cell death, promoting dendritic cell maturation and T lymphocytes infiltration. Furthermore, a combination of Q-MIL-53(Fe) and programmed cell death protein 1 antibody amplifies cancer immunotherapy. This study validates the antitumor activity of quasi-metal-organic frameworks and its immunotherapy induction potential. It would broaden the application of quasi-metal-organic frameworks and open avenues for developing antitumor nanozymes.
Collapse
Affiliation(s)
- Zihui Yan
- School of Pharmacy, Changzhou University, Changzhou, 213164, P. R. China
| | - Yang Bai
- School of Pharmacy, Changzhou University, Changzhou, 213164, P. R. China.
| | - Songtao Zhang
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou, 225002, P. R. China
| | - Lingyi Kong
- Jiangsu Collaborative Innovation Centre of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, P. R. China
| | - Yu Wang
- Jiangsu Collaborative Innovation Centre of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, P. R. China
| | - Huilin Sun
- School of Pharmacy, Changzhou University, Changzhou, 213164, P. R. China
| | - Yi Li
- School of Pharmacy, Changzhou University, Changzhou, 213164, P. R. China
| | - Lin Qiu
- School of Pharmacy, Changzhou University, Changzhou, 213164, P. R. China
| | - Ruijie Zhang
- School of Pharmacy, Changzhou University, Changzhou, 213164, P. R. China
| | - Pengju Jiang
- School of Pharmacy, Changzhou University, Changzhou, 213164, P. R. China
| | - Donghui Zhao
- School of Pharmacy, Changzhou University, Changzhou, 213164, P. R. China
| | - Zhongyan Chen
- College of Chemistry and Materials Engineering, Wenzhou University, Wenzhou, 325035, P. R. China
| | - Yafei Li
- School of Pharmacy, Changzhou University, Changzhou, 213164, P. R. China.
- Jiangsu Collaborative Innovation Centre of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, P. R. China.
| | - Huan Pang
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou, 225002, P. R. China.
| | - Jianhao Wang
- School of Pharmacy, Changzhou University, Changzhou, 213164, P. R. China.
- School of Medical and Health Engineering, Changzhou University, Changzhou, 213164, P. R. China.
| |
Collapse
|
33
|
Wang L, Wang X, Wu H, Fan S, Lu Z. Integration of metagenomic analysis and metabolic modeling reveals microbial interactions in activated sludge systems in response to nanoplastics and plasticizers. WATER RESEARCH 2025; 271:122863. [PMID: 39644836 DOI: 10.1016/j.watres.2024.122863] [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: 06/21/2024] [Revised: 11/19/2024] [Accepted: 11/26/2024] [Indexed: 12/09/2024]
Abstract
Nanoplastics and plasticizers are prevalent in activated sludge and pose a potential threat to microbial communities in wastewater treatment systems. However, studies on the effects of nanoplastics and plasticizers on the interaction mechanisms and metabolic functions of microbial communities in activated sludge systems are still scarce. In this study, the responses of microbial interactions and metabolic functions to PVC nanoplastics (PVCNPs) and bis(2-ethylhexyl) phthalate (DEHP) in activated sludge were investigated via a combination of amplicon sequencing, metagenome sequencing, and metabolic modeling. The results revealed that DEHP had a significant effect on the microbial community under short-term exposure. DEHP contamination may increase vitamin B12 producers to enhance species collaboration in communities. Furthermore, community metabolic modeling revealed that DEHP-degrading bacteria could promote positive interactions among community members. The increased metabolic exchange flux of siderophores and glutathione in microbial communities under PVCNPs and DEHP contamination implied that microbial communities may maintain iron homeostasis in response to PVCNPs and DEHP contamination through interspecies collaboration. However, more data are needed to further validate these results. This study provides vital insights into the response mechanisms of microbial interactions to nanoplastic and plasticizer contamination in activated sludge systems.
Collapse
Affiliation(s)
- Lvjing Wang
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou 310058, China; Cancer Center, Zhejiang University, Hangzhou 310058, China
| | - Xiaoyu Wang
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou 310058, China; Cancer Center, Zhejiang University, Hangzhou 310058, China
| | - Hao Wu
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou 310058, China; Cancer Center, Zhejiang University, Hangzhou 310058, China
| | - Siqing Fan
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou 310058, China; Cancer Center, Zhejiang University, Hangzhou 310058, China
| | - Zhenmei Lu
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou 310058, China; Cancer Center, Zhejiang University, Hangzhou 310058, China; Zhejiang University-University of Edinburgh Joint Research Centre for Engineering Biology, International Campus, Zhejiang University, Haining 314400, China.
| |
Collapse
|
34
|
Sahoo SS, Manna D. Nanomaterial-Triggered Ferroptosis and Cuproptosis in Cancer Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2412462. [PMID: 40018870 DOI: 10.1002/smll.202412462] [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: 12/20/2024] [Revised: 02/04/2025] [Indexed: 03/01/2025]
Abstract
Cancer remains one of the leading causes of the death of individuals globally. Conventional treatment techniques like chemotherapy and radiation often suffer various drawbacks like toxicity and drug resistance. The study of cell death has been predominantly focused on classical forms like apoptosis, but the role of metal ions in governing controlled cell death is a fascinating and less explored area. Metal-mediated controlled cell death is a process where metal triggers cell death via a unique mechanism. Nanomaterial-based strategies have gained attention for their ability to deliver precise therapeutic agents while also triggering Regulated Cell Death (RCD) mechanisms in cancer cells. The recently discovered metal-mediated controlled cell death techniques like cuproptosis and ferroptosis can be used in cancer treatment as they can be used selectively for the treatment of drug-resistant cancer. Nano material-based delivery system can also be used for the precise delivery of the drug to the targeted sites. In this review, we have given some idea about the mechanism of metal-mediated controlled cell death techniques (ferroptosis and cuproptosis) and how we can initiate controlled cell deaths using nanomaterials for cancer treatment.
Collapse
Affiliation(s)
- Suman Sekhar Sahoo
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal, Bhopal Bypass Road, Bhopal, Madhya Pradesh, 462066, India
| | - Debasish Manna
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal, Bhopal Bypass Road, Bhopal, Madhya Pradesh, 462066, India
| |
Collapse
|
35
|
Wang SW, Li P, Liu SY, Huang DL, Zhang SJ, Zeng XX, Lan T, Mao KL, Gao Y, Cheng YF, Shen Q, Ruan YP, Mao ZJ. Astragaloside IV inhibits retinal pigment epithelial cell senescence and reduces IL-1β mRNA stability by targeting FTO-mediated m 6A methylation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 138:156408. [PMID: 39848020 DOI: 10.1016/j.phymed.2025.156408] [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: 10/20/2024] [Revised: 12/16/2024] [Accepted: 01/16/2025] [Indexed: 01/25/2025]
Abstract
BACKGROUND Resistance to senescence in retinal pigment epithelial (RPE) cells can delay the progression of age-related macular degeneration (AMD). However, the mechanisms underlying RPE cell senescence remain inadequately understood, and effective therapeutic strategies are lacking. While astragaloside IV (Ast) has demonstrated anti-aging properties, its specific effects on RPE cell senescence and potential mechanisms are not yet fully clarified. PURPOSE This study aimed to explore the impacts of Ast on RPE cell senescence and to uncover the molecular mechanisms involved. METHODS The therapeutic efficacy of Ast was assessed using sodium iodate (NaIO3)-induced adult retinal pigment epithelial cell line 19 (ARPE-19) cell models and an AMD mouse model. To investigate the mechanisms by which Ast mitigated RPE cell senescence, RNA sequencing (RNA-seq), drug affinity responsive target stability-mass spectrometry (DARTS-MS), cellular thermal shift assay (CETSA), reverse transcription quantitative PCR (RT-qPCR), as well as western blotting were conducted. RESULTS Ast significantly inhibited NaIO3-treated ARPE-19 cell senescence and protected against NaIO3-induced AMD in mice. RNA-seq analysis revealed that Ast significantly attenuated inflammation-related signaling pathways and reduced the mRNA levels of interleukin-1 beta (IL-1β). Specifically, Ast decreased the stability of IL-1β mRNA while enhancing its N6-methyladenosine (m6A) methylation. Furthermore, Ast directly interacted with fat mass and obesity-associated protein (FTO). Knockdown or pharmacological inhibition of FTO mitigated the senescence and IL-1β expression in NaIO3-treated ARPE-19 cells. FTO was essential for Ast to inhibit cellular senescence and IL-1β expression. Additionally, inhibition or knockdown of FTO conferred also provided resistance to AMD in the murine model. CONCLUSION Our results indicated that Ast significantly attenuated RPE cell senescence and showed anti-AMD properties. FTO was demonstrated to be a promising therapeutic target for AMD treatment. These findings may provide a deeper understanding of the molecular mechanisms underlying RPE cell senescence in AMD and offer potential strategies for its prevention and management.
Collapse
Affiliation(s)
- Si-Wei Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; Panvascular Diseases Research Center, the Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou 324000, China.
| | - Ping Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Shi-Yu Liu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - De-Lian Huang
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Si-Jia Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Xi-Xi Zeng
- Panvascular Diseases Research Center, the Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou 324000, China
| | - Tian Lan
- Panvascular Diseases Research Center, the Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou 324000, China
| | - Kai-Li Mao
- Panvascular Diseases Research Center, the Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou 324000, China
| | - Yuan Gao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yi-Fan Cheng
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Qing Shen
- Panvascular Diseases Research Center, the Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou 324000, China
| | - Ye-Ping Ruan
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Zhu-Jun Mao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; Department of Ophthalmology, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou 324000, China.
| |
Collapse
|
36
|
Xu H, Brown JL, Bhaskaran S, Van Remmen H. Reactive oxygen species in the pathogenesis of sarcopenia. Free Radic Biol Med 2025; 227:446-458. [PMID: 39613046 PMCID: PMC11816180 DOI: 10.1016/j.freeradbiomed.2024.11.046] [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: 03/22/2024] [Revised: 11/06/2024] [Accepted: 11/21/2024] [Indexed: 12/01/2024]
Abstract
One of the most critical factors impacting healthspan in the elderly is the loss of muscle mass and function, clinically referred to as sarcopenia. Muscle atrophy and weakness lead to loss of mobility, increased risk of injury, metabolic changes and loss of independence. Thus, defining the underlying mechanisms of sarcopenia is imperative to enable the development of effective interventions to preserve muscle function and quality in the elderly and improve healthspan. Over the past few decades, understanding the roles of mitochondrial dysfunction and oxidative stress has been a major focus of studies seeking to reveal critical molecular pathways impacted during aging. In this review, we will highlight how oxidative stress might contribute to sarcopenia by discussing the impact of oxidative stress on the loss of innervation and alteration in the neuromuscular junction (NMJ), on muscle mitochondrial function and atrophy pathways, and finally on muscle contractile function.
Collapse
Affiliation(s)
- Hongyang Xu
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, United States
| | - Jacob L Brown
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, United States; Oklahoma City VA Medical Center, Oklahoma City, OK, 73104, United States
| | - Shylesh Bhaskaran
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, United States
| | - Holly Van Remmen
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, United States; Oklahoma City VA Medical Center, Oklahoma City, OK, 73104, United States.
| |
Collapse
|
37
|
Li S, Su D, Hu S, Hu Q, Sun D. Epigallocatechin gallate ameliorates retinal pigment epithelial cell damage via the CYFIP2 /AKT pathway. Toxicol Appl Pharmacol 2025; 495:117124. [PMID: 39667565 DOI: 10.1016/j.taap.2024.117124] [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: 06/25/2024] [Revised: 09/27/2024] [Accepted: 10/07/2024] [Indexed: 12/14/2024]
Abstract
Age-related macular degeneration (AMD) is a representative age-related ophthalmic disease, and the pathogenesis of AMD remains unclear. This research intended to determine whether epigallocatechin gallate (EGCG) could alleviate the progression of AMD and the possible mechanism. We constructed three groups of mice (young, aged, and EGCG), and HE and TUNEL staining of retinal tissues was performed to observe the structural changes in the retinal pigment epithelial (RPE) layer and the level of apoptosis, respectively. Through RNA-Sequencing analysis of retinal tissues and by RT-qPCR, GO, KEGG, and literature analyses, we identified cytoplasmic fragile X mental retardation 1-interacting protein 2 (CYFIP2) as a possible effector gene for EGCG action and validated its role by immunofluorescent and western blotting experiments. The CCK-8 and Hoechst 33342 apoptosis assays, and western blotting and qRT-PCR assays showed that EGCG reduced hydrogen peroxide (H2O2)-induced apoptosis in adult human RPE (ARPE-19) cells, and the expression of Cyfip2 was changed accordingly. RNA interference analysis indicated that Cyfip2 knockdown alleviated H2O2-induced ARPE apoptosis, while its overexpression weakened EGCG's protective effect. Western blot analysis showed that Cyfip2 mediated the anti-apoptotic effect of EGCG by modulating the level of protein kinase B (Akt) phosphorylation in ARPE cells, and the activation level of phosphorylated AKT (p-AKT Ser473) in retinal tissue of the EGCG-fed group was higher than that of the aged group. Taken together, this study suggests that EGCG plays a protective role in the development of AMD and the apoptosis of ARPE cells through the Cyfip2/AKT pathway.
Collapse
Affiliation(s)
- Sijia Li
- Harbin Medical University, Harbin 150086, China; Hongqi Hospital of Mudanjiang Medical University, Mudanjiang 157011, Heilongjiang, China
| | - Dongmei Su
- Department of Genetics, NHC Key Laboratory of Reproductive Health Engineering Technology Research, National Research Institute for Family Planning, Health Department, Beijing 100081, China; Graduate School, Peking Union Medical College, Beijing 100081, China
| | - Shanshan Hu
- Hongqi Hospital of Mudanjiang Medical University, Mudanjiang 157011, Heilongjiang, China
| | - Qiang Hu
- Harbin Medical University, Harbin 150086, China
| | - Dawei Sun
- Department of Ophthalmology, the second affiliated hospital of Harbin Medical University, Harbin 150086, China.
| |
Collapse
|
38
|
Luo G, Yang W, Geng Z, Cheng Y, Xu Y, Xiao Y, Liu J. Molecular mechanism of GSH metabolism and autophagy in NAC-promoted recombinant human serum albumin and follicle stimulating hormone beta fusion protein secretion in Pichia pastoris. J Biotechnol 2025; 398:146-157. [PMID: 39710118 DOI: 10.1016/j.jbiotec.2024.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 12/11/2024] [Accepted: 12/12/2024] [Indexed: 12/24/2024]
Abstract
The Pichia pastoris expression system is a favorable platform for production of pharmaceutical proteins. Treatment of strains with N-acetyl-L-cysteine (NAC) has been shown to enhance the yield of recombinant proteins, thereby contributing to a reduction in production costs. However, the specific mechanism of action of NAC remains unclear. Previous research has indicated that glutathione (GSH) and autophagy are involved in the increased production of human serum albumin and porcine follicle-stimulating hormone β (HSA-pFSHβ) by NAC. This study investigated the potential interaction between GSH and autophagy in the production of HSA-pFSHβ. The findings indicated that sulfhydryl-free antioxidants such as melatonin, vitamin C, or vitamin E did not exhibit similar effects to NAC in enhancing HSA-pFSHβ yield. Moreover, NAC was found to enhance HSA-pFSHβ production by modulating GSH metabolism to reduce GSH consumption, increase total GSH levels, as well as glutathione peroxidase (GSH-Px) and glutathione reductase (GR) activities. Additionally, inhibition of autophagy through disruption of autophagy scaffolding proteins Atg1 or Atg11 led to an increase in recombinant HSA-pFSHβ production. Furthermore, NAC significantly decreased the phosphorylation of Slt2, and the absence of the SLT2 gene influenced the effect of NAC on HSA-pFSHβ secretion by modulating mitophagy and GSH metabolism. In conclusion, these results suggest a complex interplay between GSH metabolism and autophagy in the regulation of NAC-induced HSA-pFSHβ secretion.
Collapse
Affiliation(s)
- Gang Luo
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Wen Yang
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Zijian Geng
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Yiyi Cheng
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Yingqing Xu
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Yimeng Xiao
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Jiying Liu
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China.
| |
Collapse
|
39
|
Zuo J, Tian YX, An Q, Wu BY, Yang JR, Fan YC. Potential Biomarkers and Therapeutic Targets in Hepatitis B Virus-related Acute Liver Failure: Interplay of the Ferroptosis, Autophagy and Immune Responses. Int J Med Sci 2025; 22:806-818. [PMID: 39991755 PMCID: PMC11843133 DOI: 10.7150/ijms.106360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 01/11/2025] [Indexed: 02/12/2025] Open
Abstract
Hepatitis B virus-related acute liver failure (HBV-ALF) is characterized by a high fatality rate, its pathogenesis remains unclear and the therapeutic efficacy is limited. Ferroptosis which closely related to autophagy may be an underlying mechanism of HBV-ALF. The aim of this study was to identify key ferroptosis- and autophagy-related genes and pathways and provide insight into potential therapeutic approaches for HBV-ALF. We accessed the GSE14668 and GSE96851 datasets from the Gene Expression Omnibus (GEO) database and focused on differentially expressed genes (DEGs), ferroptosis-related DEGs (FRGs) and autophagy-related DEGs (ARGs). Hub genes were subsequently analyzed for enrichment, protein‒protein interactions (PPIs), and different immunological microenvironments, and potential hub gene were identified using MCC method and LASSO. Gene-targeted drugs were from the DGIdb and DrugBank databases.A total of 1462 DEGs were identified (726 upregulated and 736 downregulated). Enriched pathways included amino acid metabolism and immune and inflammatory responses, potentially serving as biomarkers for ALF pathogenesis. After integration with the FerrDb and HADb databases, 55 FRGs and 45 ARGs were identified. Thirteen hub genes (SLC7A11, HMOX1, G6PD, RRM2, KIF20A, HELLS, GPT2, GLS2, SPP1, CCR2, DCN, IRS1, and IGF1) were identified which closely associated with the immune microenvironment. Interplay among these genes occurred primarily through HMOX1. Moreover, we identified several hub gene-targeted drugs that may be effective in HBV-ALF treatment, such as riluzole, acetylcysteine, NADH and Vitamin E.Thirteen hub genes may play crucial roles in HBV-ALF progression, particularly, the HMOX1. Furthermore, drug target exploration offered promising avenues for therapeutic intervention in patients with HBV-ALF.
Collapse
Affiliation(s)
- Jing Zuo
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, China
- Institute of Hepatology, Shandong University, Jinan, China
| | - Yu-Xin Tian
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, China
- Institute of Hepatology, Shandong University, Jinan, China
| | - Qi An
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, China
- Institute of Hepatology, Shandong University, Jinan, China
| | - Bai-Yun Wu
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, China
- Institute of Hepatology, Shandong University, Jinan, China
| | - Jie-Ru Yang
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, China
- Institute of Hepatology, Shandong University, Jinan, China
| | - Yu-Chen Fan
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, China
- Institute of Hepatology, Shandong University, Jinan, China
| |
Collapse
|
40
|
Desterke C, Fu Y, Bonifacio-Mundaca J, Monge C, Pineau P, Mata-Garrido J, Francés R. Ferroptosis Transcriptional Regulation and Prognostic Impact in Medulloblastoma Subtypes Revealed by RNA-Seq. Antioxidants (Basel) 2025; 14:96. [PMID: 39857430 PMCID: PMC11761645 DOI: 10.3390/antiox14010096] [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: 12/11/2024] [Revised: 01/13/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Medulloblastoma (MB) is the most common malignant brain tumor in children, typically arising during infancy and childhood. Despite multimodal therapies achieving a response rate of 70% in children older than 3 years, treatment remains challenging. Ferroptosis, a form of regulated cell death, can be induced in medulloblastoma cells in vitro using erastin or RSL3. Using two independent medulloblastoma RNA-sequencing cohorts (MB-PBTA and MTAB-10767), we investigated the expression of ferroptosis-related molecules through multiple approaches, including Weighted Gene Co-Expression Network Analysis (WGCNA), molecular subtype stratification, protein-protein interaction (PPI) networks, and univariable and multivariable overall survival analyses. A prognostic expression score was computed based on a cross-validated ferroptosis signature. In training and validation cohorts, the regulation of the ferroptosis transcriptional program distinguished the four molecular subtypes of medulloblastoma. WGCNA identified nine gene modules in the MB tumor transcriptome; five correlated with molecular subtypes, implicating pathways related to oxidative stress, hypoxia, and trans-synaptic signaling. One module, associated with disease recurrence, included epigenetic regulators and nucleosome organizers. Univariable survival analyses identified a 45-gene ferroptosis prognostic signature associated with nutrient sensing, cysteine and methionine metabolism, and trans-sulfuration within a one-carbon metabolism. The top ten unfavorable ferroptosis genes included CCT3, SNX5, SQOR, G3BP1, CARS1, SLC39A14, FAM98A, FXR1, TFAP2C, and ATF4. Patients with a high ferroptosis score showed a worse prognosis, particularly in the G3 and SHH subtypes. The PPI network highlighted IL6 and CBS as unfavorable hub genes. In a multivariable overall survival model, which included gender, age, and the molecular subtype classification, the ferroptosis expression score was validated as an independent adverse prognostic marker (hazard ratio: 5.8; p-value = 1.04 × 10-9). This study demonstrates that the regulation of the ferroptosis transcriptional program is linked to medulloblastoma molecular subtypes and patient prognosis. A cross-validated ferroptosis signature was identified in two independent RNA-sequencing cohorts, and the ferroptosis score was confirmed as an independent and adverse prognostic factor in medulloblastoma.
Collapse
Affiliation(s)
- Christophe Desterke
- INSERM UMRS-1310, Faculté de Médecine du Kremlin Bicêtre, Université Paris-Saclay, F-94270 Le Kremlin-Bicêtre, France;
| | - Yuanji Fu
- INSERM, CNRS, Institut Necker Enfants Malades, Université Paris Cité, F-75015 Paris, France;
| | - Jenny Bonifacio-Mundaca
- National Tumor Bank, Department of Pathology, National Institute of Neoplastic Diseases, Surquillo 15038, Peru;
| | - Claudia Monge
- Unité Organisation Nucléaire et Oncogenèse, Institut Pasteur, Université Paris Cité, INSERM U993, F-75015 Paris, France; (C.M.); (P.P.)
| | - Pascal Pineau
- Unité Organisation Nucléaire et Oncogenèse, Institut Pasteur, Université Paris Cité, INSERM U993, F-75015 Paris, France; (C.M.); (P.P.)
| | - Jorge Mata-Garrido
- Unité Organisation Nucléaire et Oncogenèse, Institut Pasteur, Université Paris Cité, INSERM U993, F-75015 Paris, France; (C.M.); (P.P.)
| | - Raquel Francés
- Energy & Memory, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, F-75006 Paris, France
| |
Collapse
|
41
|
Yu FF, Zuo J, Wang M, Yu SY, Luo KT, Sha TT, Li Q, Dong ZC, Zhou GY, Zhang F, Guo X, Ba Y, Wang YJ. Selenomethionine alleviates T-2 toxin-induced articular chondrocyte ferroptosis via the system Xc -/GSH/GPX4 axis. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 290:117569. [PMID: 39700767 DOI: 10.1016/j.ecoenv.2024.117569] [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: 09/20/2024] [Revised: 12/15/2024] [Accepted: 12/16/2024] [Indexed: 12/21/2024]
Abstract
T-2 toxin can induce bone and cartilage development disorder, and oxidative stress plays an important role in it. It is well known that selenomethionine (Se-Met) has antioxidative stress properties and promotes the repair of cartilage lesion, but it remains unclear whether Se-Met can relieve damaged cartilage exposure to T-2 toxin. Here, the oxidative stress and ferroptosis of chondrocytes exposure to T-2 toxin were observed. Mechanistically, T-2 toxin increased ROS, lipid ROS, MDA and Fe2+ contents in chondrocytes, decreased GSH and GPX4 activity, and inhibited the system Xc-/GSH/GPX4 antioxidant axis. In addition, the mitochondria of chondrocytes shrunk and the mitochondrial crest decreased or disappeared. However, Fer-1 (Ferrostatin-1) inhibited ferroptosis induced by T-2 toxin in chondrocytes. The Se-Met alleviated lipid peroxidation, oxidative stress, and damaged mitochondrial in T-2 toxin-infected chondrocytes, enhanced antioxidant enzyme activity, and activated the system Xc-/GSH/GPX4 axis, thereby antagonizing ferroptosis of chondrocytes and alleviating articular cartilage damage. In conclusion, our findings highlight the essentiality of ferroptosis in chondrocyte caused by T-2 toxin, elucidate how Se-Met offers protection against this injury and provide research evidence for the drug treatment target of Kashin-Beck disease.
Collapse
Affiliation(s)
- Fang-Fang Yu
- Department of Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, PR China.
| | - Juan Zuo
- Department of Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, PR China; Department of Medical Technology, Zhengzhou Shuqing Medical College, Zhengzhou, Henan 450064, PR China.
| | - Miao Wang
- Department of Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, PR China.
| | - Shui-Yuan Yu
- Department of Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, PR China.
| | - Kang-Ting Luo
- Department of Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, PR China.
| | - Tong-Tong Sha
- Department of Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, PR China.
| | - Qian Li
- Department of Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, PR China.
| | - Zai-Chao Dong
- Department of Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, PR China.
| | - Guo-Yu Zhou
- Department of Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, PR China.
| | - Feng Zhang
- Institute of Endemic Diseases, School of Public Health of Health Science Center, Xi'an, Jiaotong University, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, Xi'an 710061, PR China.
| | - Xiong Guo
- Institute of Endemic Diseases, School of Public Health of Health Science Center, Xi'an, Jiaotong University, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, Xi'an 710061, PR China.
| | - Yue Ba
- Department of Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, PR China.
| | - Yan-Jie Wang
- Department of Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, PR China.
| |
Collapse
|
42
|
Zhang Y, Huang R, Liu X, Cai M, Su M, Cheng Y, Jiang J, Wang X, Peng D. Taohong siwu decoction ameliorates abnormal uterine bleeding via inhibiting ACSL4-mediated ferroptosis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 339:119130. [PMID: 39566864 DOI: 10.1016/j.jep.2024.119130] [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: 08/30/2024] [Revised: 11/06/2024] [Accepted: 11/17/2024] [Indexed: 11/22/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Taohong Siwu Decoction (TSD) is a well-known traditional Chinese medicine (TCM) prescription. It consists of six crude herbs, including Rehmannia glutinosa Libosch, Paeonia lactiflora Pall, Angelica sinensis (Oliv.) Diels, Ligusticum chuanxiong Hort., Prunus persica (L.) Batsch, Cauthamus tinctorius L. It has been used to treat blood stasis syndrome in Chinese clinics for thousands of years. According to recent research, TSD may be useful in the management of abnormal uterine bleeding (AUB). The aim of the present study is to investigate the possible mechanism of TSD on AUB after drug-induced incomplete abortion. AIM OF THE STUDY To investigate whether TSD could be effective in ameliorating AUB through inhibiting acyl-CoA synthetase long-chain family member 4 (ACSL4)-mediated ferroptosis. MATERIALS AND METHODS An incomplete medical aborting model was established and Ishikawa cell lines were utilized in vitro. The quantity of uterine bleeding was measured by alkaline hemoglobin photometry. Pathological results were observed by hematoxylin-eosin staining (HE). Mitochondrial morphology and function were measured by transmission electron microscopy. The related protein and mRNA were detected by western blot, the real-time reverse transcriptase-polymerase chain reaction (RT-qPCR). We used knockdown and overexpression of ACSL4 to investigate the influence of ferroptosis in Ishikawa cells and the impact of TSD on ferroptosis. RESULTS TSD dramatically reduced the amount and duration of bleeding as well as the endometrial inflammation of AUB. TSD improved mitochondrial characteristics, decreased ACSL4 protein and mRNA levels. The ferroptosis marker glutathione (GSH) levels were increased, on the contrary, reactive oxygen species (ROS) and iron levels decreased when TSD intervened. TSD decreased levels of the inflammatory factors and the oxidative products. CONCLUSION TSD alleviated endometrial inflammation by inhibiting ACSL4-mediated ferroptosis and exerts a protective effect of AUB.
Collapse
Affiliation(s)
- Yanyan Zhang
- Department of Pharmacology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230031, PR China
| | - Rong Huang
- Department of Pharmacology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230031, PR China; Department of Pharmacology, Anhui University of Chinese Medicine, Hefei, 230012, PR China
| | - Xiaochuang Liu
- Department of Pharmacology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230031, PR China.
| | - Ming Cai
- Department of Pharmacy, The Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230001, PR China
| | - Mengyu Su
- Department of Pharmacology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230031, PR China; Department of Pharmacology, Anhui University of Chinese Medicine, Hefei, 230012, PR China
| | - Yao Cheng
- Department of Pharmacology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230031, PR China; Department of Pharmacology, Anhui University of Chinese Medicine, Hefei, 230012, PR China
| | - Juanjuan Jiang
- Department of Pharmacology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230031, PR China; Department of Pharmacology, Anhui University of Chinese Medicine, Hefei, 230012, PR China
| | - Xuekai Wang
- Department of Pharmacology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230031, PR China; Department of Pharmacology, Anhui University of Chinese Medicine, Hefei, 230012, PR China
| | - Daiyin Peng
- Department of Pharmacology, Anhui University of Chinese Medicine, Hefei, 230012, PR China
| |
Collapse
|
43
|
Casirati E, Valenti L. SLC7A11 transporter maintains critical nonessential amino acids levels to hamper ferroptosis during MASLD progression. Sci Bull (Beijing) 2025:S2095-9273(25)00038-6. [PMID: 39837720 DOI: 10.1016/j.scib.2025.01.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2025]
Affiliation(s)
- Elia Casirati
- Precision Medicine and Biological Resource Center, Fondazione IRCCS Ca' Granda Ospedale Policlinico Milano, Milan 20122, Italy
| | - Luca Valenti
- Precision Medicine and Biological Resource Center, Fondazione IRCCS Ca' Granda Ospedale Policlinico Milano, Milan 20122, Italy; Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan 20122, Italy.
| |
Collapse
|
44
|
Wu W, Xu B, Huang H, Mao Y, Gao Y, Bu W. The role of ferroptosis in liver injury after cold ischemia-reperfusion in rats with autologous orthotopic liver transplantation. J Artif Organs 2025:10.1007/s10047-024-01488-2. [PMID: 39760970 DOI: 10.1007/s10047-024-01488-2] [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: 08/24/2024] [Accepted: 12/16/2024] [Indexed: 01/07/2025]
Abstract
Using autologous orthotopic liver transplantation (AOLT) model in rats, the effect of lipid reactive oxygen species (L-ROS) inhibitor Ferrostain-1 on ferroptosis signal pathway was observed to determine whether ferroptosis occurred in rat liver injury after cold ischemia-reperfusion (I/R). Thirty-two healthy adult SPF male SD rats, 8 ~ 10 weeks old, weight 240 ~ 260 g, were divided into four groups by the method of random number table (n = 8): sham group, I/R group, I/R + Fer-1 group, I/R + DFO group. In the I/R + Fer-1 group, ferristatin-1(5 mg /kg) was intraperitoneally injected 30 min before surgery; in the I/R + DFO group, DFO 100 mg/kg was injected intraperitoneally 1 h before operation and 12 h after operation. Blood samples were taken from the inferior hepatic vena cava 24 h after reperfusion. After anesthesia, the rats were killed and part of their liver tissue was removed. The pathological changes of liver tissue sections were observed under a high-power microscope, and the liver injury was evaluated. Serum malondialdehyde (MDA) and serum levels of ALT, AST and IL-6 were determined by the ELISA method, Reduced glutathione (GSH), glutathione peroxidase 4 (GPX4), MDA, Fe2 + and superoxide dismutase (SOD) were determined in the liver tissue. Compared with the sham group, the serum levels of the IL-6,MDA, AST and ALT in I/R group were obviously higher (P < 0.05); The levels of MDA and Fe2+ in liver tissue were significantly increased (P < 0.05).The levels of SOD, GSH and GPX4 in liver tissue were decreased. The levels of serum MDA, IL-6, AST, and ALT in the I/R + Fer-1 and I/R + DFO groups were significantly lower than those in the I/R group at 24 h after reperfusion. In the I/R + Fer-1 group, the level of MDA in liver tissue decreased significantly, while the level of SOD, GSH and GPX4 in intestinal tissue increased (P < 0.05). In The I/R + DFO group, the levels of MDA and Fe2+ in liver tissue decreased significantly, while the level of SOD in intestinal tissue increased (P < 0.05). Ferroptosis is involved in pathophysiological process of liver injury after cold ischemia-reperfusion in AOLT rats.
Collapse
Affiliation(s)
- Wei Wu
- Department of Anesthesiology, CR & WISCO General Hospital, Affiliated to Wuhan University of Science and Technology, Wuhan, 430080, China.
| | - Bei Xu
- Department of Anesthesiology, CR & WISCO General Hospital, Affiliated to Wuhan University of Science and Technology, Wuhan, 430080, China
| | - Haibin Huang
- Department of Anesthesiology, CR & WISCO General Hospital, Affiliated to Wuhan University of Science and Technology, Wuhan, 430080, China
| | - Ying Mao
- Department of Anesthesiology, CR & WISCO General Hospital, Affiliated to Wuhan University of Science and Technology, Wuhan, 430080, China
| | - Yuan Gao
- Department of Anesthesiology, CR & WISCO General Hospital, Affiliated to Wuhan University of Science and Technology, Wuhan, 430080, China
| | - Wenhao Bu
- Department of Anesthesiology, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430070, China.
| |
Collapse
|
45
|
Wang X, Wang X, Zhao Z, Wang Q, Zhu X, Ou Q, Xu JY, Lu L, Gao F, Wang J, Bi Y, Xu GT, Jin C, Tian H. DNA-Dependent Protein Kinase Catalytic Subunit Prevents Ferroptosis in Retinal Pigment Epithelial Cells. Invest Ophthalmol Vis Sci 2025; 66:50. [PMID: 39841110 PMCID: PMC11756607 DOI: 10.1167/iovs.66.1.50] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 12/28/2024] [Indexed: 01/30/2025] Open
Abstract
Purpose The purpose of this study was to investigate the activated core kinases involved in the DNA damage responses (DDR) during ferroptosis of retinal pigment epithelial (RPE) cells in vitro and their regulatory effects on ferroptosis. Methods Ferroptosis was induced by erastin in induced RPE (iRPE) cells derived from human umbilical cord mesenchymal stem cells (hUCMSCs), hUCMSCs, and induced pluripotent stem cell-derived RPE (iPSC-RPE) cells. CCK8 was employed to measure the cell viability. Calcein/PI staining was used to detect the ferroptotic cells. The γ-H2AX, 8-oxoG, and phosphorylated DNA-dependent protein kinase catalytic subunit (DNA-PKcs) were determined through immunostaining. The phosphorylation of DNA-PKcs and ERK1/2 was determined by Western blotting. Lipid peroxides were detected by BODIPY581/591-C11 staining. Results The iRPE cells exhibited a stronger ability to resist ferroptosis compared to hUCMSCs. Ferroptosis induced DNA damage in cells, and DNA-PKcs was rapidly phosphorylated in iRPE cells on the treatment of erastin. In addition, inhibition of DNA-PKcs phosphorylation promoted ferroptosis in iRPE cells, suggesting that DNA-PKcs prevents ferroptosis. Meanwhile, DNA-PKcs inhibited ERK1/2 phosphorylation only at the early stage of ferroptosis induction, whereas ERK1/2 phosphorylation played a protective role in iRPE cells. Furthermore, erastin inducing DNA-PKcs phosphorylation and inhibition of its phosphorylation promoting ferroptosis were also verified in iPSC-RPE cells. Conclusions The present study elucidates that the key DDR kinase DNA-PKcs is activated and plays protective role during ferroptosis in RPE cells in vitro, which will provide new research targets and strategies for inhibiting ferroptosis in RPE cells.
Collapse
Affiliation(s)
- Xueying Wang
- Department of Ophthalmology of Tongji Hospital and Laboratory of Clinical and Visual Sciences of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, China
| | - Xi Wang
- Department of Ophthalmology of Tongji Hospital and Laboratory of Clinical and Visual Sciences of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, China
| | - Zhenzhen Zhao
- Department of Ophthalmology of Tongji Hospital and Laboratory of Clinical and Visual Sciences of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, China
| | - Qian Wang
- Department of Ophthalmology of Tongji Hospital and Laboratory of Clinical and Visual Sciences of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, China
| | - Xiaoman Zhu
- Department of Ophthalmology of Tongji Hospital and Laboratory of Clinical and Visual Sciences of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, China
| | - Qingjian Ou
- Department of Ophthalmology of Tongji Hospital and Laboratory of Clinical and Visual Sciences of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, China
- Department of Pharmacology, School of Medicine, Tongji University, Shanghai, China
| | - Jing-Ying Xu
- Department of Ophthalmology of Tongji Hospital and Laboratory of Clinical and Visual Sciences of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, China
| | - Lixia Lu
- Department of Ophthalmology of Tongji Hospital and Laboratory of Clinical and Visual Sciences of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, China
| | - Furong Gao
- Department of Ophthalmology of Tongji Hospital and Laboratory of Clinical and Visual Sciences of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, China
| | - Juan Wang
- Department of Ophthalmology of Tongji Hospital and Laboratory of Clinical and Visual Sciences of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, China
| | - Yanlong Bi
- Department of Ophthalmology of Tongji Hospital and Laboratory of Clinical and Visual Sciences of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, China
| | - Guo-Tong Xu
- Department of Ophthalmology of Tongji Hospital and Laboratory of Clinical and Visual Sciences of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, China
| | - Caixia Jin
- Department of Ophthalmology of Tongji Hospital and Laboratory of Clinical and Visual Sciences of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, China
| | - Haibin Tian
- Department of Ophthalmology of Tongji Hospital and Laboratory of Clinical and Visual Sciences of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, China
- Department of Pharmacology, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
46
|
Wei S, Li J, Zhang Y, Li Y, Wang Y. Ferroptosis in eye diseases: a systematic review. Eye (Lond) 2025; 39:18-27. [PMID: 39379520 PMCID: PMC11733247 DOI: 10.1038/s41433-024-03371-z] [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: 01/23/2024] [Revised: 08/09/2024] [Accepted: 09/20/2024] [Indexed: 10/10/2024] Open
Abstract
Ferroptosis is a type of iron-dependent cell death that differs from apoptosis, necroptosis, autophagy, and other forms of cell death. It is mainly characterized by the accumulation of intracellular lipid peroxides, redox imbalance, and reduced levels of glutathione and glutathione peroxidase 4. Studies have demonstrated that ferroptosis plays an important regulatory role in the occurrence and development of neurodegenerative diseases, stroke, traumatic brain injury, and ischemia-reperfusion injuries. Multiple mechanisms, such as iron metabolism, ferritinophagy, p53, and p62/Keap1/Nrf2, as well as the combination of FSP1/CoQ/NADPH and hepcidin/FPN-1 can alter the vulnerability to ferroptosis. Nevertheless, there has been limited research on the development and management of ferroptosis in the realm of eye disorders, with most studies focusing on retinal conditions such as age-related macular degeneration and retinitis pigmentosa. This review offers a thorough examination of the disruption of iron homeostasis in eye disorders, investigating the underlying mechanisms. We anticipate that the occurrence of ferroptotic cell death will not only establish a fresh field of study in eye diseases, but also present a promising therapeutic target for treating these diseases.
Collapse
Affiliation(s)
- Shengsheng Wei
- Shaanxi Eye Hospital, Xi'an People's Hospital (Xi'an Fourth Hospital), Affiliated People's Hospital of Northwest University, Xi'an, China
| | - Jing Li
- Shaanxi Eye Hospital, Xi'an People's Hospital (Xi'an Fourth Hospital), Affiliated People's Hospital of Northwest University, Xi'an, China
| | - Yaohua Zhang
- Shaanxi Eye Hospital, Xi'an People's Hospital (Xi'an Fourth Hospital), Affiliated People's Hospital of Northwest University, Xi'an, China
| | - Yong Li
- Shaanxi Eye Hospital, Xi'an People's Hospital (Xi'an Fourth Hospital), Affiliated People's Hospital of Northwest University, Xi'an, China
| | - Yan Wang
- Tianjin Eye Institute, Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Hospital, Tianjin, China.
- Nankai University Eye Institute, Nankai University, Tianjin, China.
| |
Collapse
|
47
|
Hansman DS, Du J, Casson RJ, Peet DJ. Eye on the horizon: The metabolic landscape of the RPE in aging and disease. Prog Retin Eye Res 2025; 104:101306. [PMID: 39433211 PMCID: PMC11833275 DOI: 10.1016/j.preteyeres.2024.101306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/10/2024] [Accepted: 10/10/2024] [Indexed: 10/23/2024]
Abstract
To meet the prodigious bioenergetic demands of the photoreceptors, glucose and other nutrients must traverse the retinal pigment epithelium (RPE), a polarised monolayer of cells that lie at the interface between the outer retina and the choroid, the principal vascular layer of the eye. Recent investigations have revealed a metabolic ecosystem in the outer retina where the photoreceptors and RPE engage in a complex exchange of sugars, amino acids, and other metabolites. Perturbation of this delicate metabolic balance has been identified in the aging retina, as well as in age-related macular degeneration (AMD), the leading cause of blindness in the Western world. Also common in the aging and diseased retina are elevated levels of cytokines, oxidative stress, advanced glycation end-products, increased growth factor signalling, and biomechanical stress - all of which have been associated with metabolic dysregulation in non-retinal cell types and tissues. Herein, we outline the role of these factors in retinal homeostasis, aging, and disease. We discuss their effects on glucose, mitochondrial, lipid, and amino acid metabolism in tissues and cell types outside the retina, highlighting the signalling pathways through which they induce these changes. Lastly, we discuss promising avenues for future research investigating the roles of these pathological conditions on retinal metabolism, potentially offering novel therapeutic approaches to combat age-related retinal disease.
Collapse
Affiliation(s)
- David S Hansman
- School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia.
| | - Jianhai Du
- Department of Ophthalmology and Visual Sciences, Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, WV 26506, USA
| | - Robert J Casson
- Discipline of Ophthalmology and Visual Science, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Daniel J Peet
- School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
48
|
Huang Y, Li X, Xu S, Zu D, Liu H, He H, Bao Q, He Y, Liang C, Shi Y, Cheng X, Teng Y, Ye Z. Polyvinyl chloride nanoplastics suppress homology-directed repair and promote oxidative stress to induce esophageal epithelial cellular senescence and cGAS-STING-mediated inflammation. Free Radic Biol Med 2025; 226:288-301. [PMID: 39515594 DOI: 10.1016/j.freeradbiomed.2024.11.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 11/04/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Nanoplastics (NPs), which are characterized by plastic particles smaller than 1 μm, have emerged as pervasive environmental pollutants, raising concerns about their potential toxicity to living organisms. Numerous investigations have highlighted the tendency of NPs to accumulate in organs, resulting in toxic effects. Despite polyvinyl chloride (PVC) being one of the most prevalent NPs, its impact on the esophagus and the associated underlying mechanisms remain largely unknown. In this study, we investigated the impact of PVC NPs on the esophagus and found that PVC NPs exposure induces oxidative stress and elicits DNA damage responses. Further analysis revealed that PVC NPs inhibit the homology-directed repair (HDR) pathway by suppressing the expression of breast cancer susceptibility gene 2 (BRCA2) and growth factor receptor-bound protein 2 (GRB2), resulting in genomic instability. Additionally, the release of free DNA activates cGAS-STING and the downstream NF-κB signaling, elevating inflammatory factors and chemokines, which further leads to cellular senescence. In vivo experiments corroborated these findings, showing that PVC NPs induced oxidative stress, inflammation, and cellular senescence, subsequently impacting mouse behavior. This study contributes novel insights into the health risks associated with PVC NPs exposure and identifies potential therapeutic targets.
Collapse
Affiliation(s)
- Yixing Huang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China; Zhejiang University School of Medicine, Hangzhou, 310058, China; Department of Otorhinolaryngology, Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Xiao Li
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China; The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Shengfeng Xu
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Dan Zu
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China; School of Life Sciences, Tianjin University, Tianjin, 300100, China
| | - Haidong Liu
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, 310022, China; Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, 310022, China
| | - Hanyi He
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, 310022, China; Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, 310022, China
| | - Qimei Bao
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China
| | - Yanhua He
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, 310022, China; Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, 310022, China
| | - Chen Liang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China
| | - Yin Shi
- Department of Biochemistry, and Department of Pulmonology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Xiangdong Cheng
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, 310022, China; Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, 310022, China.
| | - Yaoshu Teng
- Department of Otorhinolaryngology, Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.
| | - Zu Ye
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, 310022, China; Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, 310022, China; Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Nanning, 530021, China; Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, 530021, China.
| |
Collapse
|
49
|
Liu F, Yang Z, Li J, Wu T, Li X, Zhao L, Wang W, Yu W, Zhang G, Xu Y. Targeting programmed cell death in diabetic kidney disease: from molecular mechanisms to pharmacotherapy. Mol Med 2024; 30:265. [PMID: 39707216 DOI: 10.1186/s10020-024-01020-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 11/29/2024] [Indexed: 12/23/2024] Open
Abstract
Diabetic kidney disease (DKD), one of the most prevalent microvascular complications of diabetes, arises from dysregulated glucose and lipid metabolism induced by hyperglycemia, resulting in the deterioration of renal cells such as podocytes and tubular epithelial cells. Programmed cell death (PCD), comprising apoptosis, autophagy, ferroptosis, pyroptosis, and necroptosis, represents a spectrum of cell demise processes intricately governed by genetic mechanisms in vivo. Under physiological conditions, PCD facilitates the turnover of cellular populations and serves as a protective mechanism to eliminate impaired podocytes or tubular epithelial cells, thereby preserving renal tissue homeostasis amidst hyperglycemic stress. However, existing research predominantly elucidates individual modes of cell death, neglecting the intricate interplay and mutual modulation observed among various forms of PCD. In this comprehensive review, we delineate the diverse regulatory mechanisms governing PCD and elucidate the intricate crosstalk dynamics among distinct PCD pathways. Furthermore, we review recent advancements in understanding the pathogenesis of PCD and explore their implications in DKD. Additionally, we explore the potential of natural products derived primarily from botanical sources as therapeutic agents, highlighting their multifaceted effects on modulating PCD crosstalk, thereby proposing novel strategies for DKD treatment.
Collapse
Affiliation(s)
- Fengzhao Liu
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Zhenyu Yang
- Graduate School of Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Jixin Li
- Xi Yuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Tao Wu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xiangyu Li
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, 100102, China
| | - Lijuan Zhao
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Wenru Wang
- Xi Yuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Wenfei Yu
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Guangheng Zhang
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Yunsheng Xu
- Department of Endocrinology, Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250001, China.
| |
Collapse
|
50
|
Zeng T, Lei GL, Yu ML, Zhang TY, Wang ZB, Wang SZ. The role and mechanism of various trace elements in atherosclerosis. Int Immunopharmacol 2024; 142:113188. [PMID: 39326296 DOI: 10.1016/j.intimp.2024.113188] [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: 06/28/2024] [Revised: 09/13/2024] [Accepted: 09/13/2024] [Indexed: 09/28/2024]
Abstract
Atherosclerosis is a slow and complex disease that involves various factors, including lipid metabolism disorders, oxygen-free radical production, inflammatory cell infiltration, platelet adhesion and aggregation, and local thrombosis. Trace elements play a crucial role in human health. Many trace elements, especially metallic ones, not only maintain the normal functions of organs but also participate in basic metabolic processes. The latest studies have revealed a close correlation between trace elements and the occurrence and progression of atherosclerosis. The imbalance of these trace elements can induce atherosclerosis or accelerate its progression through various mechanisms, which poses a significant threat to human health. Therefore, exploring the specific mechanism of trace elements on atherosclerosis is highly significant. In this review, we summarized the roles and mechanisms of iron, copper, zinc, magnesium, and selenium homeostasis and imbalance in atherosclerosis development, in order to identify novel targets and therapeutic strategies for treating atherosclerosis.
Collapse
Affiliation(s)
- Tao Zeng
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Sciences, Hengyang Medical School, University of South China, Hengyang 421001, China; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, China
| | - Guan-Lan Lei
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Sciences, Hengyang Medical School, University of South China, Hengyang 421001, China; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, China
| | - Mei-Ling Yu
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Sciences, Hengyang Medical School, University of South China, Hengyang 421001, China; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, China
| | - Ting-Yu Zhang
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Sciences, Hengyang Medical School, University of South China, Hengyang 421001, China; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, China
| | - Zong-Bao Wang
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Sciences, Hengyang Medical School, University of South China, Hengyang 421001, China; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, China.
| | - Shu-Zhi Wang
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Sciences, Hengyang Medical School, University of South China, Hengyang 421001, China; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, China.
| |
Collapse
|