1
|
Wo B, Liu S, Liang Z, Li X. Treatment with human placental extracts inhibits allergic rhinitis by modulating AMPK/SHP1/SHP2/STING signaling. Mol Med Rep 2025; 32:183. [PMID: 40280106 PMCID: PMC12059460 DOI: 10.3892/mmr.2025.13548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 03/11/2025] [Indexed: 04/29/2025] Open
Abstract
The present study aimed to investigate the regulatory effects and mechanisms of human placental extracts (HPE) on rats and cell models of ovalbumin (OVA)‑induced allergic rhinitis (AR). IFN‑y and LPS induced AR in vitro. A total of 32 male Sprague‑Dawley (SD) rats were randomly divided into the following four groups: Sham group, model group, model + HPE group and model + HPE + AMPK inhibitor group (n=8 rats/group). With the exception of the sham group, the remaining three groups were sensitized with OVA to establish an AR model, followed by various treatments. Hematoxylin and eosin staining was utilized to observe morphological changes in the nasal mucosa, ELISA was employed to measure serum levels of IL‑1β, interferon (IFN)β, immunoglobulin (Ig)E, IgG1 and IgG2a, and western blotting was conducted to assess protein expression across the groups. The sham group exhibited intact tissue structure with no notable pathological alterations. The model group demonstrated pronounced pathological features, including extensive infiltration of inflammatory cells, tissue shedding and edema. The model + HPE group revealed a gradual restoration of tissue architecture, characterized by reduced edema and inflammatory infiltration, whereas the model + HPE + AMPK inhibitor group again exhibited significant inflammatory cell infiltration and other pathological manifestations. Compared with the sham operation group, the levels of IL‑1β, IFNβ, IgE, IgG1 and IgG2a in the serum of the model group were elevated. The levels of IL‑1β, IFNβ, IgE, IgG1 and IgG2a in the model + HPE group were lower than those in the model group. In addition, the levels of IL‑1β, IFNβ, IgE, IgG1 and IgG2a in the model + HPE + AMPK inhibitor group were higher than those in the model + HPE group. Relative to the sham group, the expression levels of phosphorylated (p)‑AMPK/total (t)‑AMPK, p‑Src homology 2‑containing phosphatase (SHP)1/t‑SHP1 and p‑SHP2/t‑SHP2 were diminished, whereas the expression levels of p‑STING/t‑STING and p‑TBK1/t‑TBK1 were heightened in the model group. In comparison to the model group, the expression levels of p‑AMPK/t‑AMPK, p‑SHP1/t‑SHP1 and p‑SHP2/t‑SHP2 were enhanced, whereas the expression levels of p‑STING/t‑STING and p‑TBK1/t‑TBK1 were reduced in the model + HPE group. Conversely, when compared with the model + HPE group, the expression levels of p‑AMPK/t‑AMPK, p‑SHP1/t‑SHP1 and p‑SHP2/t‑SHP2 were decreased, whereas those of p‑STING/t‑STING and p‑TBK1/t‑TBK1 were increased in the model + HPE + AMPK inhibitor group. In conclusion, HPE may inhibit AR by modulating the AMPK/SHP1/SHP2/STING signaling pathway.
Collapse
Affiliation(s)
- Beibei Wo
- Department of Otolaryngology Head and Neck Surgery, Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Shuang Liu
- Department of Pathology, The 980th Hospital of People's Liberation Army (PLA) Joint Logistics Support Force, Shijiazhuang, Hebei 050082, P.R. China
| | - Zihui Liang
- Department of Surgery, Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Xiaoming Li
- Department of Otolaryngology Head and Neck Surgery, Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| |
Collapse
|
2
|
Leal Y, Valenzuela-Muñoz V, Gallardo-Escárate C. Fish vaccines promote blood cell transcriptional remodeling in Atlantic salmon against pathogens. FISH & SHELLFISH IMMUNOLOGY 2025; 162:110356. [PMID: 40258434 DOI: 10.1016/j.fsi.2025.110356] [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: 11/27/2024] [Revised: 03/20/2025] [Accepted: 04/17/2025] [Indexed: 04/23/2025]
Abstract
Chilean salmon farming confronts persistent challenges, including climate change risks and pathogens, where the most prevalent diseases impacting Atlantic salmon are Caligidosis and Rickettsial Salmonid Septicemia (SRS). As a sustainable strategy, fish vaccines hold promise for preventing diseases and reducing the use of antibiotics. While most studies on Atlantic salmon responses to vaccines emphasize transcriptome profiling from tissues such as the liver, head kidney, and skin, blood cell transcriptomics to monitor immune response dynamics is emerging as a promising tool in salmon aquaculture. This study evaluated the Atlantic salmon blood cell transcriptome in response to vaccination and subsequent infection with the sea louse Caligus rogercresseyi and the intracellular bacterium Piscirickettsia salmonis. The vaccination trial included four groups: fish immunized with the recombinant IPath® vaccine and two commercial vaccines currently used in Chile for salmon production. (BlueGuard® and Alpha Ject LiVac® SRS), and the unvaccinated control group. The group vaccinated with IPath® showed a higher transcriptomic response than commercial vaccines. Additionally, all three groups significantly modulated genes associated with iron homeostasis and metabolism. Furthermore, the HIF-1 signaling pathway and ferroptosis were notably activated in the IPath® group, suggesting a potential role of IPath® in the hypoxia response and cell death. This research highlights the effectiveness of using Atlantic salmon blood cells to assess immune responses, offering valuable insights into the fish immune system without resorting to lethal sampling.
Collapse
Affiliation(s)
- Yeny Leal
- Interdisciplinary Center for Aquaculture Research (INCAR), Universidad de Concepción, P.O. Box 160-C, Concepción, 4030000, Chile
| | - Valentina Valenzuela-Muñoz
- Interdisciplinary Center for Aquaculture Research (INCAR), Universidad de Concepción, P.O. Box 160-C, Concepción, 4030000, Chile
| | - Cristian Gallardo-Escárate
- Interdisciplinary Center for Aquaculture Research (INCAR), Universidad de Concepción, P.O. Box 160-C, Concepción, 4030000, Chile.
| |
Collapse
|
3
|
Gao D, Chen T, Dong J, He Y, Ge N, Guo J, Sun S, Yang F. Ferroptosis at the crossroads: Insights and advances in non-neoplastic pancreatic diseases. Int Immunopharmacol 2025; 158:114870. [PMID: 40383100 DOI: 10.1016/j.intimp.2025.114870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 03/26/2025] [Accepted: 05/10/2025] [Indexed: 05/20/2025]
Abstract
Ferroptosis is a form of regulated cell death characterized by iron accumulation and increased lipid peroxidation, primarily counteracted by a range of antioxidant molecules, including glutathione (GSH), glutathione peroxidase 4 (GPX4), ubiquinone, tetrahydrofolate, and nuclear respiratory factor 2. Furthermore, the process of ferroptosis is intricately influenced by the opposing actions of the p53 tumor suppressor gene and activated transcription factors 3 and 4, which can either facilitate or hinder ferroptotic cell death depending on the cellular context. This form of cell death is significantly associated with various pancreatic disorders, including both acute and chronic pancreatitis, as well as diabetes mellitus. In this review, we thoroughly investigate the mechanisms underlying ferroptosis, focusing on iron overload, lipid peroxidation, and the regulatory molecules involved in ferroptosis modulation (notably the system xc-/GSH/GPX4 axis), along with the relevant signaling pathways. We also examine the role of ferroptosis in non-neoplastic pancreatic diseases such as pancreatitis and diabetes mellitus while identifying novel therapeutic agents that target ferroptosis, potentially paving the way for innovative treatment strategies for pancreatic conditions.
Collapse
Affiliation(s)
- Duolun Gao
- Department of Gastroenterology, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, People's Republic of China; Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, People's Republic of China
| | - Tingting Chen
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, People's Republic of China
| | - Jize Dong
- Department of Gastroenterology, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, People's Republic of China; Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, People's Republic of China
| | - Yanjie He
- Department of Surgery, New York University School of Medicine and NYU-Langone Medical Center, New York 10012, NY, USA
| | - Nan Ge
- Department of Gastroenterology, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, People's Republic of China; Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, People's Republic of China
| | - Jintao Guo
- Department of Gastroenterology, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, People's Republic of China; Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, People's Republic of China
| | - Siyu Sun
- Department of Gastroenterology, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, People's Republic of China; Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, People's Republic of China.
| | - Fan Yang
- Department of Gastroenterology, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, People's Republic of China; Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, People's Republic of China.
| |
Collapse
|
4
|
Liu T, Huang Y, Wang Y, Shen H. Disrupting the immune homeostasis: the emerging role of macrophage ferroptosis in autoimmune diseases. Int Immunopharmacol 2025; 157:114745. [PMID: 40319750 DOI: 10.1016/j.intimp.2025.114745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 03/18/2025] [Accepted: 04/24/2025] [Indexed: 05/07/2025]
Abstract
Autoimmune diseases are a class of chronic disorders characterized by the aberrant activation of the immune system, where macrophages play a central role in regulating immune responses during disease onset and progression. Ferroptosis, a form of iron-dependent programmed cell death, has recently attracted significant interest due to its involvement in various pathological conditions. In macrophages, ferroptosis not only compromises cell viability but also disrupts immune homeostasis by promoting pro-inflammatory responses and suppressing anti-inflammatory pathways, thereby intensifying inflammation and exacerbating disease severity. While substantial progress has been made in elucidating macrophage ferroptosis in atherosclerosis and oncology, its precise mechanistic role in autoimmune diseases remains largely unexplored. This review systematically summarizes the molecular mechanisms of macrophage ferroptosis and its regulatory effects on immune homeostasis, with particular emphasis on its role in autoimmune diseases, including rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), inflammatory bowel disease (IBD), multiple sclerosis (MS), and systemic sclerosis (SSc). Furthermore, we discuss potential therapeutic targets related to macrophage ferroptosis in these conditions. By integrating current knowledge, this review aims to provide a theoretical framework and novel perspectives for developing innovative therapeutic strategies targeting autoimmune diseases.
Collapse
Affiliation(s)
- Tianfu Liu
- Department of Hepatology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, Gansu, China
| | - Yichen Huang
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, Gansu, China
| | - Yizhe Wang
- Department of Respiratory and Critical Care Medicine, The First People Hospital of Lanzhou, Lanzhou 730050, Gansu, China
| | - Haili Shen
- Department of Rheumatology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, Gansu, China.
| |
Collapse
|
5
|
Zhou S, Zhu Y, Wu Y, Zhang X, Kong X, Zhao X, Xiang H, Shang D. New insights on metabolic reprogramming in macrophage plasticity. Int Immunopharmacol 2025; 157:114797. [PMID: 40339492 DOI: 10.1016/j.intimp.2025.114797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 04/18/2025] [Accepted: 04/30/2025] [Indexed: 05/10/2025]
Abstract
Macrophages are the first line of defense in the innate immune system. Macrophages have two subtypes: classically activated macrophages (M1) and alternatively activated macrophages (M2), with different phenotypes and functions. They play a critical role in defending against pathogens and maintaining internal homeostasis. Macrophages have great plasticity in their biological characteristics. Although the regulation of macrophage plasticity has not been fully elucidated, accumulated evidence supports that microenvironmental differences are the root cause for macrophage differentiation into different subtypes. These differences alter macrophage plasticity by modulating key metabolites, activating downstream gene transcription, and influencing phagocytosis, cytokine secretion, and immune regulation. Herein, we systematically summarize metabolic reprogramming, including glucose, lipid, amino acid, ion, vitamin, nucleotide, and butyrate metabolism, as key regulators affecting macrophage polarization, providing new insights for developing targeted drugs that modulate macrophage plasticity.
Collapse
Affiliation(s)
- Siyu Zhou
- Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China; Institute (College) of Integrative Medicine, Dalian Medical University, Dalian 116044, China
| | - Yutong Zhu
- Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China; Institute (College) of Integrative Medicine, Dalian Medical University, Dalian 116044, China
| | - Yu Wu
- Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China; Institute (College) of Integrative Medicine, Dalian Medical University, Dalian 116044, China
| | - Xiaonan Zhang
- Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China; Institute (College) of Integrative Medicine, Dalian Medical University, Dalian 116044, China
| | - Xin Kong
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China; College of Pharmacy, Dalian Medical University, Dalian 116011, China
| | - Xinya Zhao
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China; College of Pharmacy, Dalian Medical University, Dalian 116011, China
| | - Hong Xiang
- Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| | - Dong Shang
- Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China; Institute (College) of Integrative Medicine, Dalian Medical University, Dalian 116044, China; Department of General Surgery, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| |
Collapse
|
6
|
Pliouta L, Lampsas S, Kountouri A, Korakas E, Thymis J, Kassi E, Oikonomou E, Ikonomidis I, Lambadiari V. Mitochondrial Dysfunction in the Development and Progression of Cardiometabolic Diseases: A Narrative Review. J Clin Med 2025; 14:3706. [PMID: 40507468 PMCID: PMC12155522 DOI: 10.3390/jcm14113706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2025] [Revised: 05/19/2025] [Accepted: 05/23/2025] [Indexed: 06/16/2025] Open
Abstract
Mitochondria play a central role in energy metabolism and continuously adapt through dynamic processes such as fusion and fission. When the balance between these processes is disrupted, it can lead to mitochondrial dysfunction and increased oxidative stress, contributing to the development and progression of various cardiometabolic diseases (CMDs). Their role is crucial in diabetes mellitus (DM), since their dysfunction drives β-cell apoptosis, immune activation, and chronic inflammation through excessive ROS production, worsening endogenous insulin secretion. Moreover, sympathetic nervous system activation and altered dynamics, contribute to hypertension through oxidative stress, impaired mitophagy, endothelial dysfunction, and cardiomyocyte hypertrophy. Furthermore, the role of mitochondria is catalytic in endothelial dysfunction through excessive reactive oxygen species (ROS) production, disrupting the vascular tone, permeability, and apoptosis, while impairing antioxidant defense and promoting inflammatory processes. Mitochondrial oxidative stress, resulting from an imbalance between ROS/Reactive nitrogen species (RNS) imbalance, promotes atherosclerotic alterations and oxidative modification of oxidizing low-density lipoprotein (LDL). Mitochondrial DNA (mtDNA), situated in close proximity to the inner mitochondrial membrane where ROS are generated, is particularly susceptible to oxidative damage. ROS activate redox-sensitive inflammatory signaling pathways, notably the nuclear factor kappa B (NF-κB) pathway, leading to the transcriptional upregulation of proinflammatory cytokines, chemokines, and adhesion molecules. This proinflammatory milieu promotes endothelial activation and monocyte recruitment, thereby perpetuating local inflammation and enhancing atherogenesis. Additionally, mitochondrial disruptions in heart failure promote further ischemic injury and excessive oxidative stress release and impair ATP production and Ca2⁺ dysregulation, contributing to cell death, fibrosis, and decreased cardiac performance. This narrative review aims to investigate the intricate relationship between mitochondrial dysfunction and CMDs.
Collapse
Affiliation(s)
- Loukia Pliouta
- Diabetes Center, 2nd Department of Internal Medicine, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece; (L.P.); (S.L.); (A.K.); (E.K.); (V.L.)
| | - Stamatios Lampsas
- Diabetes Center, 2nd Department of Internal Medicine, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece; (L.P.); (S.L.); (A.K.); (E.K.); (V.L.)
| | - Aikaterini Kountouri
- Diabetes Center, 2nd Department of Internal Medicine, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece; (L.P.); (S.L.); (A.K.); (E.K.); (V.L.)
| | - Emmanouil Korakas
- Diabetes Center, 2nd Department of Internal Medicine, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece; (L.P.); (S.L.); (A.K.); (E.K.); (V.L.)
| | - John Thymis
- 2nd Cardiology Department, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 15772 Athens, Greece;
| | - Eva Kassi
- Endocrine Unit, 1st Department of Propaedeutic and Internal Medicine, Laiko Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Evangelos Oikonomou
- 3rd Department of Cardiology, “Sotiria” Thoracic Diseases Hospital of Athens, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Ignatios Ikonomidis
- 2nd Cardiology Department, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 15772 Athens, Greece;
| | - Vaia Lambadiari
- Diabetes Center, 2nd Department of Internal Medicine, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece; (L.P.); (S.L.); (A.K.); (E.K.); (V.L.)
| |
Collapse
|
7
|
Li AL, Guo KZ, Yu LR, Ge J, Zhou W, Zhang JP. Intercellular communication after myocardial infarction: Macrophage as the centerpiece. Ageing Res Rev 2025; 109:102757. [PMID: 40320153 DOI: 10.1016/j.arr.2025.102757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 04/16/2025] [Accepted: 04/25/2025] [Indexed: 05/16/2025]
Abstract
Post-myocardial infarction (MI) injury, repair, and remodeling are complex biological events orchestrated by the heart and immune cell populations, with immune-inflammation at the core. Macrophages, as the main immune cell population active throughout the post-MI injury to repair processes, are the core of this "drama". Recently, single-cell sequencing and other techniques have revealed the heterogeneity of macrophage origins and the complexity of macrophage subpopulation transformation and intercellular communication after MI. Defining the changes in macrophage subpopulation dynamics and macrophage-centered intercellular communication after MI may represent new targeted therapeutic strategies. It also helps to select the optimal time point for anti-inflammatory or pro-repair accurately. Therefore, in this review, we summarize the major macrophage subpopulations active at different times after MI and their functional characteristics based on gene expression profiles. Meanwhile, we summarize macrophage-centered intercellular communication, focusing on how macrophages interact with cardiomyocytes, neutrophils, fibroblasts, endothelial cells, and other cardiac cells. Together, these dominate the transition from inflammatory injury to fibrotic repair in the infarcted heart. We also focus on the regulatory potential of immune metabolism in macrophage subpopulation transformation and intercellular communication after MI, particularly providing new insights about lactylation. We conclude by emphasizing macrophage-centric targeting strategies and clinical translational potential, to provide ideas for the clinical treatment of MI.
Collapse
Affiliation(s)
- Ao-Lin Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300183, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China; Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Kang-Zheng Guo
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300183, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China; Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Le-Rong Yu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300183, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China; Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Jun Ge
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300183, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China; Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Wei Zhou
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300183, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China; Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Jun-Ping Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300183, China.
| |
Collapse
|
8
|
Zhang Z, She L, Bai M. Efficacy of exosomes in acute kidney injury treatment and the associated mechanism (Review). Mol Med Rep 2025; 31:137. [PMID: 40145555 PMCID: PMC11963750 DOI: 10.3892/mmr.2025.13503] [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: 11/23/2024] [Accepted: 01/31/2025] [Indexed: 03/28/2025] Open
Abstract
Acute kidney injury (AKI) is a syndrome characterized by rapid loss of renal function with a high morbidity and mortality. However, due to the complex pathophysiologic mechanisms of AKI, no specific treatment for this disease is currently available. Animal models have demonstrated the protective effects of exosomes on AKI; however, the underlying mechanisms require further investigation. The present review focuses on the efficacy of exosomes derived from different cell sources, including mesenchymal stem cells, endothelial progenitor cells and tubular epithelial cells, in the treatment of AKI and the associated mechanism. Furthermore, the effects of exosomal contents, including microRNAs, circular RNAs, long non‑coding RNAs, messenger RNAs and proteins, on the repair of renal tubules, protection against renal tubular epithelial cell injury, protection against fibrosis, inhibition of early endoplasmic reticulum stress and mediation of inflammation during AKI are also summarized in the present review.
Collapse
Affiliation(s)
- Zehao Zhang
- Department of Nephrology, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Lecheng She
- Department of Nephrology, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Ming Bai
- Department of Nephrology, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| |
Collapse
|
9
|
Attique I, Haider Z, Khan M, Hassan S, Soliman MM, Ibrahim WN, Anjum S. Reactive Oxygen Species: From Tumorigenesis to Therapeutic Strategies in Cancer. Cancer Med 2025; 14:e70947. [PMID: 40377005 DOI: 10.1002/cam4.70947] [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: 11/26/2024] [Revised: 04/27/2025] [Accepted: 04/29/2025] [Indexed: 05/18/2025] Open
Abstract
BACKGROUND Reactive oxygen species (ROS), a class of highly reactive molecules, are closely linked to the pathogenesis of various cancers. While ROS primarily originate from normal cellular processes, external stimuli can also contribute to their production. Cancer cells typically exhibit elevated ROS levels due to disrupted redox homeostasis, characterized by an imbalance between antioxidant and oxidant species. ROS play a dual role in cancer biology: at moderate levels, they facilitate tumor progression by regulating oncogenes and tumor suppressor genes, inducing mutations, promoting proliferation, extracellular matrix remodeling, invasion, immune modulation, and angiogenesis. However, excessive ROS levels can cause cellular damage and initiate apoptosis, necroptosis, or ferroptosis. METHODS This review explores molecular targets involved in redox homeostasis dysregulation and examines the impact of ROS on the tumor microenvironment (TME). Literature from recent in vitro and in vivo studies was analyzed to assess how ROS modulation contributes to cancer development and therapy. RESULTS Findings indicate that ROS influence cancer progression through various pathways and cellular mechanisms. Targeting ROS synthesis or enhancing ROS accumulation in tumor cells has shown promising anticancer effects. These therapeutic strategies exhibit significant potential to impair tumor growth while also interacting with elements of the TME. CONCLUSION The ROS serve as both promoters and suppressors of cancer depending on their intracellular concentration. Their complex role offers valuable opportunities for targeted cancer therapies. While challenges remain in precisely modulating ROS for therapeutic benefit, they hold promise as synergistic agents alongside conventional treatments, opening new avenues in cancer management.
Collapse
Affiliation(s)
- Iqra Attique
- Department of Biotechnology, Kinnaird College for Women University, Lahore, Pakistan
| | - Zahra Haider
- Department of Biotechnology, Kinnaird College for Women University, Lahore, Pakistan
| | - Maha Khan
- Department of Biotechnology, Lahore College for Women University, Lahore, Pakistan
| | - Samina Hassan
- Department of Botany, Kinnaird College for Women University, Lahore, Pakistan
| | - Mohamed Mohamed Soliman
- Clinical Laboratory Sciences Department, Turabah University College, Taif University, Taif, Saudi Arabia
- Biochemistry Department, Faculty of Veterinary Medicine, Benha University, Toukh, Egypt
| | - Wisam Nabeel Ibrahim
- Department of Biomedical Science, College of Health Sciences, QU Health, Qatar University, Doha, Qatar
| | - Sumaira Anjum
- Department of Biotechnology, Kinnaird College for Women University, Lahore, Pakistan
| |
Collapse
|
10
|
Wang S, Yang J, Zhen C, Wang H, Shang P. Electromagnetic fields regulate iron metabolism: From mechanisms to applications. J Adv Res 2025:S2090-1232(25)00288-7. [PMID: 40311754 DOI: 10.1016/j.jare.2025.04.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 04/06/2025] [Accepted: 04/28/2025] [Indexed: 05/03/2025] Open
Abstract
BACKGROUND Electromagnetic fields (EMFs), as a form of physical therapy, have been widely applied in biomedicine. Iron, the most abundant trace metal in living organisms, plays a critical role in various physiological processes, and imbalances in its metabolism are closely associated with the development and progression of numerous diseases. Numerous studies have demonstrated that EMF exposureinduces significant changes in both systemic and cellular iron metabolism. AIM OF REVIEW This review aims to summarize the evidence and potential biophysical mechanisms underlying the role of EMFs in regulating iron metabolism, thereby enhancing the understanding of their biological mechanisms and expanding their potential applications in biomedical fields. KEY SCIENTIFIC CONCEPTS OF REVIEW In this review, we have synthesized research findings and proposed the hypothesis that the biophysical mechanisms of EMFs regulate iron metabolism involve the special electromagnetic properties of iron-containing proteins and iron-enriched tissues, as well as the modulation of membrane structure and function, ion channels, and the generation and activity of Reactive Oxygen Species (ROS). Then, the review summarizes the latest advances in the effects of EMFs on iron metabolism and their safety, as well as their impact on immunoregulation, cardiovascular diseases, neurological diseases, orthopedic diseases, diabetes, liver injury, and cancer.
Collapse
Affiliation(s)
- Shenghang Wang
- Department of Spine Surgery, People's Hospital of Longhua, Shenzhen, China; Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen, China
| | - Jiancheng Yang
- Department of Osteoporosis, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Chenxiao Zhen
- Department of Spine Surgery, People's Hospital of Longhua, Shenzhen, China; Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen, China; School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Huiru Wang
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen, China; School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Peng Shang
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen, China.
| |
Collapse
|
11
|
俞 佳, 周 薏, 钱 春, 穆 蓝, 阙 任. [Effects of liver fibrosis induced by iron overload on M2 polarization of macrophages in mice]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2025; 45:684-691. [PMID: 40294917 PMCID: PMC12037300 DOI: 10.12122/j.issn.1673-4254.2025.04.02] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Indexed: 04/30/2025]
Abstract
OBJECTIVES To observe the evolution of intrahepatic macrophage polarization in mice with liver fibrosis induced by iron overload. METHODS Thirty-two C57BL/6 mice (6-8 weeks) were randomized into control group (n=8) and liver fibrosis model group (n=24) induced by aidly intraperitoneal injection of iron dextran. At the 3rd, 5th, and 7th weeks of modeling, 8 mice in the model group were sacrificed for observing liver fibrosis using Masson, Sirius Red and immunohistochemical staining and detecting serum levels of ALT, AST and the levels of serum iron, ferritin, liver total Fe and ferrous Fe. iNOS+/F4/80+ cells and CD206+/F4/80+ cells were detected by double immunofluorescence assay to observe the proportion and distribution of M1 and M2 macrophages. The hepatic expressions of Arg-1, iNOS, IL-6, IL-10, and TNF‑α proteins were detected using Western blotting or ELISA, and the expression of CD206 mRNA was detected using RT-PCR. RESULTS The mice in the model group showed gradual increase of fibrous tissue hyperplasia in the portal area over time, structural destruction of the hepatic lobules and formation of pseudolobules. With the passage of time during modeling, the rat models showed significantly increased hepatic expressions of α-SMA and COL-1, elevated serum levels of ALT, AST, Fe, ferritin, and increased liver total Fe and ferrous Fe levels. The expressions of M1 polarization markers IL-6, TNF‑α, and iNOS all increased with time and reached their peak levels at the 3rd week; The expressions of M2 polarization markers (IL-10 and Arg-1 proteins and CD206 mRNA) significantly increased in the 3rd week and but decreased in the 5th and 7th weeks. CONCLUSIONS Iron overload promotes M1 polarization of macrophages in mice. Liver fibrosis in the early stage promotes M2 polarization of macrophages but negatively regulate M2 polarization at later stages.
Collapse
|
12
|
Peng Y, Sheng J, Liu T, He R, Xu P. Identifying key targets and immune environment in wound healing based on iron overload-related genes. Arch Dermatol Res 2025; 317:719. [PMID: 40252113 DOI: 10.1007/s00403-025-04140-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 02/26/2025] [Accepted: 03/09/2025] [Indexed: 04/21/2025]
Abstract
Wound healing (WH) poses a significant socio-economic burden due to its high incidence and recurrence rates. Iron overload (IO) could be a factor leading to delayed WH. This study thus analyzed IO-related genes (IORGs) in WH, offering possibilities for developing new therapeutic strategies. Differential gene expression (DEGs) analysis was conducted between the WH group and intact skin (IS) group, intersected with IORGs to obtain differentially expressed IORGs (DE-IORGs). Functional enrichment analysis and potential drug screening were performed on DE-IORGs. A protein-protein interaction (PPI) network of DE-IORGs was constructed, and hub genes were identified using CytoHubba and MCODE methods. ROC curves of hub genes were plotted, and their expression levels in WH and IS groups as well as inter-gene correlations were analyzed. Additionally, immune infiltration variances in WH and IS groups, along with miRNA and TFs of hub genes, were examined. Finally, the effect of EGFR on skin wound healing was verified by scratch healing assay. 39 DE-IORGs were predominantly enriched in signaling pathways like HIF-1 signaling pathway and Th17 cell differentiation. Potential drugs for treating WH (e.g., felbamate, SA-94315, GANT-58, rucaparib) were identified. Three hub genes related to IO in WH were pinpointed (HIF1A, CDKN2A, EGFR) with diagnostic value. Immune infiltration analysis showed higher levels of immune cells like endothelial cells and macrophages in the WH group. Additionally, 55 miRNAs (e.g., hsa-mir-200a-3p, hsa-mir-218-5p) and 2 TFs (L3MBTL2, ZNF76) regulating the three hub genes were predicted. Cell experiments showed that EGFR could promote skin wound healing. The study suggested HIF1A, CDKN2A, and EGFR as potential diagnostic biomarkers for effective WH diagnosis, offering new insights into identifying potenti1al therapeutic targets for WH treatment.
Collapse
Affiliation(s)
- Yinbo Peng
- Department of Burns and Plastic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201900, China
- Department of Plastic Surgery and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201900, China
- Institute of Traumatic Medicine of Shanghai Jiao Tong University School of Medicine, 280 Mohe Road, Shanghai, 201900, China
| | - Juxiang Sheng
- Department of Burns and Plastic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201900, China
- Department of Plastic Surgery and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201900, China
| | - Tiantian Liu
- Department of Burns and Plastic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201900, China
- Department of Plastic Surgery and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201900, China
- Institute of Traumatic Medicine of Shanghai Jiao Tong University School of Medicine, 280 Mohe Road, Shanghai, 201900, China
| | - Ruizhe He
- Department of Burns and Plastic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201900, China
- Department of Plastic Surgery and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201900, China
- Institute of Traumatic Medicine of Shanghai Jiao Tong University School of Medicine, 280 Mohe Road, Shanghai, 201900, China
| | - Peng Xu
- Department of Burns and Plastic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201900, China.
- Department of Plastic Surgery and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201900, China.
- Institute of Traumatic Medicine of Shanghai Jiao Tong University School of Medicine, 280 Mohe Road, Shanghai, 201900, China.
| |
Collapse
|
13
|
Jinson S, Zhang Z, Lancaster GI, Murphy AJ, Morgan PK. Iron, lipid peroxidation, and ferroptosis play pathogenic roles in atherosclerosis. Cardiovasc Res 2025; 121:44-61. [PMID: 39739567 DOI: 10.1093/cvr/cvae270] [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: 07/30/2024] [Revised: 10/22/2024] [Accepted: 12/05/2024] [Indexed: 01/02/2025] Open
Abstract
Oxidation of lipids, excessive cell death, and iron deposition are prominent features of human atherosclerotic plaques. While extensive research has established the detrimental roles of lipid oxidation and apoptosis in atherosclerosis development, the involvement of iron in atherogenesis is not yet fully understood. With the emergence of an iron-dependent form of cell death termed ferroptosis, new attention has been brought to the complex inter-play among iron, ferroptosis, and atherosclerosis. Mechanistically, ferroptosis is caused by the lethal accumulation of iron-mediated lipid peroxides. Emerging studies have underscored ferroptosis as a contributor to worsened atherosclerosis. Herein, we review the evidence that oxidative damage and iron overload in the context of atherosclerosis may promote ferroptosis within plaques. Furthermore, we summarize recent findings of lipid peroxidation, thereby potentially ferroptosis, in various plaque cell types-such as endothelial cells, macrophages, dendritic cells, T cells, and vascular smooth muscle cells-across different stages of atherosclerosis. Understanding how these processes influence atherosclerotic plaque progression may permit targeting stage-dependent ferroptosis in each cell population and could provide a rationale for developing cell type-specific intervention strategies to mitigate atherogenic ferroptosis effectively.
Collapse
Affiliation(s)
- Swetha Jinson
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Grattan Street, Parkville, VIC 3010, Australia
| | - Ziyang Zhang
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Grattan Street, Parkville, VIC 3010, Australia
| | - Graeme I Lancaster
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Grattan Street, Parkville, VIC 3010, Australia
| | - Andrew J Murphy
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Grattan Street, Parkville, VIC 3010, Australia
| | - Pooranee K Morgan
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Grattan Street, Parkville, VIC 3010, Australia
| |
Collapse
|
14
|
Yuan Z, Chen Y, Xin Y, Zhang Y, Dong Z, Wang J, Wang X, Yang G, Li S. Key role of the CSE/transsulfuration pathway in macrophage phenotypic change under iron overload. J Trace Elem Med Biol 2025; 88:127611. [PMID: 39914135 DOI: 10.1016/j.jtemb.2025.127611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 11/22/2024] [Accepted: 01/31/2025] [Indexed: 03/24/2025]
Abstract
BACKGROUND Iron homeostasis has a significant impact on the phenotypic transformation of macrophages and is implicated in various diseases. In this study, we evaluated the effect of cystathionine-gamma-lyase (CSE)/transsulfuration pathway in iron-overload induced macrophage phenotype change. METHODS The biochemical parameters, such as qRT-PCR, western blot, fluorescence staining, were assessed both in vitro and in vivo. RESULTS Iron overload disrupts iron metabolism and alters the expression of genes involved in iron transport, resulting in the polarization of macrophages towards the M1 phenotype and an alternating activation state of M2. Meanwhile, excessive iron led to an increase in lipid peroxidation levels and disrupted cysteine metabolism. By utilizing erastin to inhibit SLC7A11 activity and block exogenous cysteine uptake, we were able to observe the exacerbation of the proinflammatory state in macrophages under conditions of cysteine deprivation. The CSE/transsulfuration pathway, serves as the primary route for endogenous cysteine synthesis. In the presence of iron overload, the expression of CSE was upregulated and further enhanced by cysteine deprivation. Deletion of CSE in CSE-knockout mice exacerbated the inflammatory transition of iron-overloaded macrophages by impacting cysteine metabolism and ferritinophagy. CONCLUSION The CSE/transsulfuration pathway regulated macrophage phenotype change under iron-overload, which may offer novel insights into potential therapeutic strategies for iron overload-related disorders.
Collapse
Affiliation(s)
- Zhaoji Yuan
- Metabolism and Disease Research Center, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250013, China; Shandong Institute of Endocrine and Metabolic Diseases, Shandong First Medical University, Jinan, Shandong 250062, China
| | - Yuxuan Chen
- Department of Cell Biology, Shandong University, Jinan, Shandong 250012, China
| | - Yijun Xin
- Metabolism and Disease Research Center, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250013, China; Shandong Institute of Endocrine and Metabolic Diseases, Shandong First Medical University, Jinan, Shandong 250062, China
| | - Yong Zhang
- Metabolism and Disease Research Center, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250013, China
| | - Zihao Dong
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Jianxu Wang
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Xiangdong Wang
- Department of Cell Biology, Shandong University, Jinan, Shandong 250012, China
| | - Guang Yang
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China.
| | - Siying Li
- Metabolism and Disease Research Center, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250013, China; Shandong Institute of Endocrine and Metabolic Diseases, Shandong First Medical University, Jinan, Shandong 250062, China.
| |
Collapse
|
15
|
Liang Y, Zhao Y, Qi Z, Li X, Zhao Y. Ferroptosis: CD8 +T cells' blade to destroy tumor cells or poison for self-destruction. Cell Death Discov 2025; 11:128. [PMID: 40169575 PMCID: PMC11962101 DOI: 10.1038/s41420-025-02415-x] [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: 11/03/2024] [Revised: 02/19/2025] [Accepted: 03/19/2025] [Indexed: 04/03/2025] Open
Abstract
Ferroptosis represents an emerging, iron-dependent form of cell death driven by lipid peroxidation. In recent years, it has garnered significant attention in the realm of cancer immunotherapy, particularly in studies involving immune checkpoint inhibitors. This form of cell death not only enhances our comprehension of the tumor microenvironment but is also considered a promising therapeutic strategy to address tumor resistance, investigate immune activation mechanisms, and facilitate the development of cancer vaccines. The combination of immunotherapy with ferroptosis provides innovative targets and fresh perspectives for advancing cancer treatment. Nevertheless, tumor cells appear to possess a wider array of ferroptosis evasion strategies compared to CD8+T cells, which have been conclusively shown to be more vulnerable to ferroptosis. Furthermore, ferroptosis in the TME can create a favorable environment for tumor survival and invasion. Under this premise, both inducing tumor cell ferroptosis and inhibiting T cell ferroptosis will impact antitumor immunity to some extent, and even make the final result run counter to our therapeutic purpose. This paper systematically elucidates the dual-edged sword role of ferroptosis in the antitumor process of T cells, briefly outlining the complexity of ferroptosis within the TME. It explores potential side effects associated with ferroptosis-inducing therapies and critically considers the combined application of ferroptosis-based therapies with ICIs. Furthermore, it highlights the current challenges faced by this combined therapeutic approach and points out future directions for development.
Collapse
Affiliation(s)
- Yuan Liang
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin, China
| | - Yixin Zhao
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin, China
| | - Zhaoyang Qi
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin, China
| | - Xinru Li
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin, China
| | - Yuguang Zhao
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin, China.
| |
Collapse
|
16
|
Veschi V, Verona F, Di Bella S, Turdo A, Gaggianesi M, Di Franco S, Mangiapane LR, Modica C, Lo Iacono M, Bianca P, Brancato OR, D'Accardo C, Porcelli G, Lentini VL, Sperduti I, Sciacca E, Fitzgerald P, Lopez-Perez D, Martine P, Brown K, Giannini G, Appella E, Stassi G, Todaro M. C1Q + TPP1 + macrophages promote colon cancer progression through SETD8-driven p53 methylation. Mol Cancer 2025; 24:102. [PMID: 40165182 PMCID: PMC11956498 DOI: 10.1186/s12943-025-02293-y] [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: 06/14/2024] [Accepted: 03/05/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND In many tumors, the tumor suppressor TP53 is not mutated, but functionally inactivated. However, mechanisms underlying p53 functional inactivation remain poorly understood. SETD8 is the sole enzyme known to mono-methylate p53 on lysine 382 (p53K382me1), resulting in the inhibition of its pro-apoptotic and growth-arresting functions. METHODS We analyzed SETD8 and p53K382me1 expression in clinical colorectal cancer (CRC) and inflammatory bowel disease (IBD) samples. Histopathological examinations, RNA sequencing, ChIP assay and preclinical in vivo CRC models, were used to assess the functional role of p53 inactivation in tumor cells and immune cell infiltration. RESULTS By integrating bulk RNAseq and scRNAseq approaches in CRC patients, SETD8-mediated p53 regulation resulted the most significantly enriched pathway. p53K382me1 expression was confined to colorectal cancer stem cells (CR-CSCs) and C1Q+ TPP1+ tumor-associated macrophages (TAMs) in CRC patient tissues, with high levels predicting decreased survival probability. TAMs promote p53 functional inactivation in CR-CSCs through IL-6 and MCP-1 secretion and increased levels of CEBPD, which directly binds SETD8 promoter thus enhancing its transcription. The direct binding of C1Q present on macrophages and C1Q receptor (C1QR) present on cancer stem cells mediates the cross-talk between the two cell compartments. As monotherapy, SETD8 genetic and pharmacological (UNC0379) inhibition affects the tumor growth and metastasis formation in CRC mouse avatars, with enhanced effects observed when combined with IL-6 receptor targeting. CONCLUSIONS These findings suggest that p53K382me1 may be an early step in tumor initiation, especially in inflammation-induced CRC, and could serve as a functional biomarker and therapeutic target in adjuvant setting for advanced CRCs.
Collapse
Affiliation(s)
- Veronica Veschi
- Department of Precision Medicine in Medical, Surgical and Critical Care, University of Palermo, Palermo, 90127, Italy.
- Department of Molecular Medicine, University of Rome La Sapienza, Rome, 00161, Italy.
| | - Francesco Verona
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, 90127, Italy
| | - Sebastiano Di Bella
- Department of Precision Medicine in Medical, Surgical and Critical Care, University of Palermo, Palermo, 90127, Italy
| | - Alice Turdo
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, 90127, Italy
| | - Miriam Gaggianesi
- Department of Precision Medicine in Medical, Surgical and Critical Care, University of Palermo, Palermo, 90127, Italy
| | - Simone Di Franco
- Department of Precision Medicine in Medical, Surgical and Critical Care, University of Palermo, Palermo, 90127, Italy
| | - Laura Rosa Mangiapane
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, 90127, Italy
| | - Chiara Modica
- Department of Precision Medicine in Medical, Surgical and Critical Care, University of Palermo, Palermo, 90127, Italy
| | - Melania Lo Iacono
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, 90127, Italy
| | - Paola Bianca
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, 90127, Italy
| | - Ornella Roberta Brancato
- Department of Precision Medicine in Medical, Surgical and Critical Care, University of Palermo, Palermo, 90127, Italy
| | - Caterina D'Accardo
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, 90127, Italy
| | - Gaetana Porcelli
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, 90127, Italy
| | | | - Isabella Sperduti
- Clinical Trial Center, Biostatistics and Bioinformatics Unit, IRCCS - Regina Elena National Cancer Institute, Rome, 00144, Italy
| | - Elisabetta Sciacca
- Centre for Experimental Medicine and Rheumatology, the London School of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, Bartsand, London, UK
| | - Peter Fitzgerald
- Genome Analysis Unit, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - David Lopez-Perez
- Chemical Immunology Section, Laboratory of Cell Biology, National Cancer Institute, Bethesda, MD, USA
| | - Pierre Martine
- Chemical Immunology Section, Laboratory of Cell Biology, National Cancer Institute, Bethesda, MD, USA
| | - Kate Brown
- Chemical Immunology Section, Laboratory of Cell Biology, National Cancer Institute, Bethesda, MD, USA
| | - Giuseppe Giannini
- Department of Molecular Medicine, University of Rome La Sapienza, Rome, 00161, Italy
- Istituto Pasteur-Fondazione Cenci Bolognetti, University of Rome La Sapienza, Rome, 00161, Italy
| | - Ettore Appella
- Chemical Immunology Section, Laboratory of Cell Biology, National Cancer Institute, Bethesda, MD, USA
| | - Giorgio Stassi
- Department of Precision Medicine in Medical, Surgical and Critical Care, University of Palermo, Palermo, 90127, Italy.
| | - Matilde Todaro
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, 90127, Italy
- Azienda Ospedaliera Universitaria Policlinico "Paolo Giaccone" (AOUP), Palermo, Italy
| |
Collapse
|
17
|
Li S, Yuan H, Yang XZ, Xu X, Yu W, Wu Y, Yao S, Xie J, He W, Guo Z, Chen Y. Synergistic Antitumor Immunotherapy via Mitochondria Regulation in Macrophages and Tumor Cells by an Iridium Photosensitizer. ACS CENTRAL SCIENCE 2025; 11:441-451. [PMID: 40161955 PMCID: PMC11950858 DOI: 10.1021/acscentsci.4c02156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 02/13/2025] [Accepted: 03/03/2025] [Indexed: 04/02/2025]
Abstract
Mitochondrial targeting has emerged as an attractive method for antitumor treatment. However, most of the mitochondria targeted drugs focused on inhibiting tumor cells, while their potential for activation of immune responses in the tumor microenvironment has rarely been described. In this study, we report a photosensitive iridium complex MitoIrL2, which enabled the simultaneous mitochondrial modulation of macrophages and tumor cells to achieve synergistic antitumor immunity. The adjustment of the mitochondrial respiratory chain, HIF-1α, and the NF-κB pathway in macrophages drove the metabolic reprogramming from oxidative phosphorylation (OXPHOS) to glycolysis, converting protumor M2 into the antitumor M1 phenotype. Downregulated expression of immunosuppressive checkpoint SIRPα has also been observed on macrophages. Meanwhile, the mitochondrial targeting MitoIrL2 enhanced the immunogenic cell death of tumor cells and reversed the immunosuppressive tumor microenvironment, which activated the systemic immune response and established long-term immune memory in vivo. This work illustrates a promising strategy to simultaneously regulate macrophages toward the antitumor phenotype and enhance immunogenic cell death in tumor cells for synergistic antitumor immunotherapy.
Collapse
Affiliation(s)
- Shumeng Li
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Chemistry and Biomedicine Innovation Center
(ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing 210023, Jiangsu, P.R. China
| | - Hao Yuan
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Chemistry and Biomedicine Innovation Center
(ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing 210023, Jiangsu, P.R. China
| | - Xiu-Zhi Yang
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Chemistry and Biomedicine Innovation Center
(ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing 210023, Jiangsu, P.R. China
| | - Xinyu Xu
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Chemistry and Biomedicine Innovation Center
(ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing 210023, Jiangsu, P.R. China
| | - Wenhao Yu
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Chemistry and Biomedicine Innovation Center
(ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing 210023, Jiangsu, P.R. China
| | - Yanping Wu
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Chemistry and Biomedicine Innovation Center
(ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing 210023, Jiangsu, P.R. China
| | - Shankun Yao
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Chemistry and Biomedicine Innovation Center
(ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing 210023, Jiangsu, P.R. China
| | - Jin Xie
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Chemistry and Biomedicine Innovation Center
(ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing 210023, Jiangsu, P.R. China
| | - Weijiang He
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Chemistry and Biomedicine Innovation Center
(ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing 210023, Jiangsu, P.R. China
| | - Zijian Guo
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Chemistry and Biomedicine Innovation Center
(ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing 210023, Jiangsu, P.R. China
| | - Yuncong Chen
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Chemistry and Biomedicine Innovation Center
(ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing 210023, Jiangsu, P.R. China
- Department
of Cardiothoracic Surgery, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing 210008, Jiangsu, P.R. China
| |
Collapse
|
18
|
Zhang S, Guan M, Ren T, Li N, Ding Q, Sun D, Zhu H. Prexasertib exerts a synergistic effect on the antitumor activity of Lenvatinib through ALOX15-mediated ferroptosis in hepatocellular carcinoma. Int Immunopharmacol 2025; 150:114278. [PMID: 39954659 DOI: 10.1016/j.intimp.2025.114278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 02/03/2025] [Accepted: 02/07/2025] [Indexed: 02/17/2025]
Abstract
Hepatocellular carcinoma (HCC) is one of the most prevalent and lethal malignancies worldwide. Lenvatinib, a potent multi-receptor tyrosine kinase inhibitor approved for the treatment of advanced HCC, demonstrates limited clinical efficacy. Therefore, there is an urgent need to investigate therapeutic strategies that combine Lenvatinib with other anticancer agents. Lenvatinib induces DNA damage in tumor cells, and the inhibition of the DNA damage response (DDR) pathway is hypothesized to enhance Lenvatinib-induced tumor cell death. In this study, we initially observed that Lenvatinib upregulated phosphorylated checkpoint kinase 1 (CHK1) protein levels, a key molecule in the DDR pathway, in HCC cells. This observation prompted us to investigate the antitumor efficacy of combining Lenvatinib with Prexasertib, a novel CHK1 inhibitor. The combination demonstrated synergistic anticancer effects in HCC cells. Mechanistically, treatment with Lenvatinib and Prexasertib resulted in cell death primarily through ferroptosis. Furthermore, we found that Lenvatinib and Prexasertib cooperatively upregulated ALOX15 expression, which culminated in the induction of ferroptosis. Taken together, our findings suggest the potential application of Prexasertib in combination with Lenvatinib as a promising therapeutic strategy for HCC treatment.
Collapse
Affiliation(s)
- Shiyu Zhang
- Department of Medical Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Mingcheng Guan
- Department of Medical Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Tianyuan Ren
- Key Laboratory of Synthetic Biology Regulatory Element, Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Suzhou Institute of Systems Medicine, Suzhou, China
| | - Na Li
- Department of Medical Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Qian Ding
- Department of Medical Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Di Sun
- Department of Medical Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hong Zhu
- Department of Medical Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
19
|
Wang J, Zhao R, Ma J, Qin J, Zhang H, Guo J, Chang X, Zhang W. Biallelic FDXR mutations induce ferroptosis in a rare mitochondrial disease with ataxia. Free Radic Biol Med 2025; 230:248-262. [PMID: 39954867 DOI: 10.1016/j.freeradbiomed.2025.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/06/2025] [Accepted: 02/09/2025] [Indexed: 02/17/2025]
Abstract
Biallelic mutations in the FDXR are known to cause rare mitochondrial diseases. However, the underlying pathogenic mechanisms remain elusive. This study investigated a patient affected by optic atrophy, ataxia, and peripheral neuropathy resulting from compound heterozygous mutations in FDXR. Structural abnormalities in mitochondria were observed in muscle and nerve tissues. Lymphoblastic cell lines (LCLs) and muscle samples from the patient exhibited signs of mitochondrial dysfunction, iron overload, oxidative stress, and lipid peroxidation. Dysregulation of the glutathione peroxidase-4 was noted in the LCLs. Furthermore, treatment with deferoxamine, N-acetyl-cysteine, and ferrostatin-1 effectively alleviated oxidative stress and cell death. Cortical neurons demonstrate that FDXR deficiency impacts the morphogenesis of neurites. Collectively, these findings suggest that ferroptosis plays a significant role in the pathogenesis of FDXR-associated diseases. Additionally, idebenone appeared to have protective effects against various cellular injuries induced by FDXR mutations, providing novel insights and therapeutic approaches for the treatment of FDXR-associated diseases.
Collapse
Affiliation(s)
- Juan Wang
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Rongjuan Zhao
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Jing Ma
- First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Jiangbo Qin
- Department of Radiology, First Hospital of Shanxi Medical University, China
| | - Huiqiu Zhang
- First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Junhong Guo
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Xueli Chang
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, China.
| | - Wei Zhang
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
20
|
Lin J, Li B, Guo X, Li G, Zhang Q, Wang W. Key Mechanisms of Oxidative Stress-Induced Ferroptosis in Heart Failure with Preserved Ejection Fraction and Potential Therapeutic Approaches. Rev Cardiovasc Med 2025; 26:26613. [PMID: 40160560 PMCID: PMC11951494 DOI: 10.31083/rcm26613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 12/02/2024] [Accepted: 12/10/2024] [Indexed: 04/02/2025] Open
Abstract
The prevalence of heart failure with preserved ejection fraction (HFpEF) is increasing annually, particularly among patients with metabolic disorders such as hypertension and diabetes. However, there is currently no treatment capable of altering the natural course of HFpEF. Recently, the interplay between oxidative stress and ferroptosis in cardiovascular diseases has drawn extensive attention; however, minimal research has been published on the mechanisms of oxidative stress and ferroptosis in HFpEF. This paper reviews the relevant mechanisms through which oxidative stress is induced and promotes ferroptosis during the development of HFpEF. The review also explores more efficacious treatment approaches for HFpEF by inhibiting oxidative stress and ferroptosis, thereby offering a theoretical foundation for verifying the feasibility of these methods for further research. As tumor-targeted therapy progresses, the survival period of tumor patients is prolonged, and cardiovascular events have gradually emerged as one of the most crucial causes of death among tumor patients. Hence, inhibiting the vascular endothelial growth factor (VEGF) pathway has become a major target in tumor treatment, significantly enhancing patient survival. Nevertheless, secondary cardiovascular complications and events, such as myocardial injury and subsequent heart failure, have severely impacted patient survival and quality of life. Therefore we have also explored the potential mechanism through which novel targeted anti-cancer drugs induce HFpEF via ferroptosis. Additionally, we reviewed the specific modes of action for preventing and treating HFpEF without influencing their anti-cancer therapeutic effect.
Collapse
Affiliation(s)
- Junling Lin
- Department of Cardiovascular Center, First Affiliated Hospital of Huzhou University, 313000 Huzhou, Zhejiang, China
| | - Bingtao Li
- Department of Cardiovascular Center, First Affiliated Hospital of Huzhou University, 313000 Huzhou, Zhejiang, China
| | - Xueqi Guo
- Department of Cardiovascular Center, First Affiliated Hospital of Huzhou University, 313000 Huzhou, Zhejiang, China
| | - Guodong Li
- Department of Cardiovascular Center, First Affiliated Hospital of Huzhou University, 313000 Huzhou, Zhejiang, China
| | - Qi Zhang
- Department of Cardiovascular Center, First Affiliated Hospital of Huzhou University, 313000 Huzhou, Zhejiang, China
| | - Wenjuan Wang
- Department of Cardiovascular Center, First Affiliated Hospital of Huzhou University, 313000 Huzhou, Zhejiang, China
| |
Collapse
|
21
|
Wang Z, Guo Z, Zhang Q, Yang C, Shi X, Wen Q, Xue Y, Zhang Z, Wang J. Relationship between iron deficiency and severity of tuberculosis: Influence on T cell subsets. iScience 2025; 28:111709. [PMID: 39898042 PMCID: PMC11783395 DOI: 10.1016/j.isci.2024.111709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/16/2024] [Accepted: 12/27/2024] [Indexed: 02/04/2025] Open
Abstract
Tuberculosis (TB) remains a leading cause of death globally, with nearly half of TB patients experiencing iron deficiency. The role of iron supplementation as an adjunct therapy remains controversial. This study examines the impact of iron deficiency on TB progression and immune function. We conducted a case-control study involving 808 pulmonary TB patients recruited from Changzhou Third People's Hospital (2018-2022) to investigate the association between serum iron levels and TB severity. Additionally, we evaluated the relationship between baseline serum iron levels and pulmonary lesion characteristics during antituberculosis treatment using a cohort study of 89 patients. We observed that low serum iron was associated with more severe lung symptoms, decreased MAIT, Vδ2+, and Treg cell percentages, and increased interleukin-1β (IL-1β) and IL-7 levels. Findings suggest that iron deficiency may exacerbate lung lesions by altering T cell subsets and cytokine profiles.
Collapse
Affiliation(s)
- Zheyue Wang
- Department of Epidemiology, Key Laboratory of Public Health Safety and Emergency Prevention and Control Technology of Higher Education Institutions in Jiangsu Province, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou 213004, China
- Department of Epidemiology, Gusu School, Nanjing Medical University, Nanjing 211166, China
- National Vaccine Innovation Platform, Nanjing Medical University, Nanjing 211166, China
| | - Zhenpeng Guo
- Department of Epidemiology, Key Laboratory of Public Health Safety and Emergency Prevention and Control Technology of Higher Education Institutions in Jiangsu Province, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou 213004, China
| | - Qiang Zhang
- Department of Epidemiology, Key Laboratory of Public Health Safety and Emergency Prevention and Control Technology of Higher Education Institutions in Jiangsu Province, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Chenchen Yang
- Department of Epidemiology, Key Laboratory of Public Health Safety and Emergency Prevention and Control Technology of Higher Education Institutions in Jiangsu Province, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Xinling Shi
- Department of Epidemiology, Key Laboratory of Public Health Safety and Emergency Prevention and Control Technology of Higher Education Institutions in Jiangsu Province, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Qin Wen
- Department of Epidemiology, Key Laboratory of Public Health Safety and Emergency Prevention and Control Technology of Higher Education Institutions in Jiangsu Province, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Yuan Xue
- Changzhou Medical Center, Nanjing Medical University, Changzhou 213004, China
| | - Zhixin Zhang
- Changzhou Medical Center, Nanjing Medical University, Changzhou 213004, China
- Department of Pulmonary Diseases, The Third People’s Hospital of Changzhou, Changzhou 213001, China
| | - Jianming Wang
- Department of Epidemiology, Key Laboratory of Public Health Safety and Emergency Prevention and Control Technology of Higher Education Institutions in Jiangsu Province, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou 213004, China
- Department of Epidemiology, Gusu School, Nanjing Medical University, Nanjing 211166, China
- National Vaccine Innovation Platform, Nanjing Medical University, Nanjing 211166, China
| |
Collapse
|
22
|
Zhao Y, Chen Z, Xie S, Xiao F, Hu Q, Ju Z. The emerging role and therapeutical implications of ferroptosis in wound healing. BURNS & TRAUMA 2025; 13:tkae082. [PMID: 39958433 PMCID: PMC11827611 DOI: 10.1093/burnst/tkae082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 11/11/2024] [Accepted: 12/03/2024] [Indexed: 02/18/2025]
Abstract
Wound healing is a complex biological process involving multiple steps, including hemostasis, inflammation, proliferation, and remodeling. A novel form of regulated cell death, ferroptosis, has garnered attention because of its involvement in these processes. Ferroptosis is characterized by the accumulation of lipid peroxides and is tightly regulated by lipid metabolism, iron metabolism, and the lipid-peroxide repair network, all of which exert a significant influence on wound healing. This review highlights the current findings and emerging concepts regarding the multifaceted roles of ferroptosis throughout the stages of normal and chronic wound healing. Additionally, the potential of targeted interventions aimed at modulating ferroptosis to improve wound-healing outcomes is discussed.
Collapse
Affiliation(s)
- Yanan Zhao
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Department of Developmental & Regenerative Medicine, College of Life Science and Technology, Jinan University, No. 601, Huangpu Avenue West, Tianhe District, Guangzhou, 510632, China
| | - Zhiyang Chen
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Department of Developmental & Regenerative Medicine, College of Life Science and Technology, Jinan University, No. 601, Huangpu Avenue West, Tianhe District, Guangzhou, 510632, China
| | - Shenghao Xie
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Department of Developmental & Regenerative Medicine, College of Life Science and Technology, Jinan University, No. 601, Huangpu Avenue West, Tianhe District, Guangzhou, 510632, China
| | - Feng Xiao
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Department of Developmental & Regenerative Medicine, College of Life Science and Technology, Jinan University, No. 601, Huangpu Avenue West, Tianhe District, Guangzhou, 510632, China
| | - Qian Hu
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Department of Developmental & Regenerative Medicine, College of Life Science and Technology, Jinan University, No. 601, Huangpu Avenue West, Tianhe District, Guangzhou, 510632, China
| | - Zhenyu Ju
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Department of Developmental & Regenerative Medicine, College of Life Science and Technology, Jinan University, No. 601, Huangpu Avenue West, Tianhe District, Guangzhou, 510632, China
| |
Collapse
|
23
|
Tian Z, Wang X, Chen S, Guo Z, Di J, Xiang C. Mitochondria-Targeted Biomaterials-Regulating Macrophage Polarization Opens New Perspectives for Disease Treatment. Int J Nanomedicine 2025; 20:1509-1528. [PMID: 39925677 PMCID: PMC11806677 DOI: 10.2147/ijn.s505591] [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: 11/09/2024] [Accepted: 01/18/2025] [Indexed: 02/11/2025] Open
Abstract
Macrophage immunotherapy is an emerging therapeutic approach designed for modulating the immune response to alleviate disease symptoms. The balance between pro-inflammatory and anti-inflammatory macrophages plays a pivotal role in the progression of inflammatory diseases. Mitochondria, often referred to as the "power plants" of the cell, are essential organelles responsible for critical functions such as energy metabolism, material synthesis, and signal transduction. The functional state of mitochondria is closely linked to macrophage polarization, prompting interest in therapeutic strategies that target mitochondria to regulate this process. To this end, biomaterials with excellent targeting capabilities and effective therapeutic properties have been developed to influence mitochondrial function and regulate macrophage polarization. However, a comprehensive summary of biomaterial-driven modulation of mitochondrial function to control macrophage phenotypes is still lacking. This review highlights the critical role of mitochondrial function in macrophage polarization and discusses therapeutic strategies mediated by biomaterials, including mitochondria-targeted biomaterials. Finally, the prospects and challenges of the use of these biomaterials in disease modulation have been explored, emphasizing their potential to be translated to the clinic. It is anticipated that this review will serve as a valuable resource for materials scientists and clinicians in the development of next-generation mitochondria-targeted biomaterials.
Collapse
Affiliation(s)
- Zui Tian
- Department of Orthopedics, Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Xudong Wang
- Department of Orthopedics, Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Shuai Chen
- Department of Orthopedics, Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Zijian Guo
- Department of Orthopedics, Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Jingkai Di
- Department of Orthopedics, Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Chuan Xiang
- Department of Orthopedics, Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| |
Collapse
|
24
|
Mao L, Ma Y, Wen X, Luo Z, Zhu H, Kong J, Liu S, Fan X, Wang J, He C, Wu YL. Iron-glucose oxidase nanogel assembly for amplified starvation-ferroptosis anti-tumor therapy. Int J Biol Macromol 2025; 289:138804. [PMID: 39689793 DOI: 10.1016/j.ijbiomac.2024.138804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 12/14/2024] [Accepted: 12/14/2024] [Indexed: 12/19/2024]
Abstract
Developing advanced and effective enzyme-drug systems for cancer treatment is of significant interest. Herein, a novel approach is reported to create a highly active and robust enzyme-drug system. Glucose oxidase nanogels (nGOx) are first synthesized by polymerization on the surface of GOx using vinylimidazole as comonomers. Fe3+ are utilized to induce self-assembly of nGOx through the imidazole-metal coordination interaction to form GOx nanogel clusters (Fe@nGOx), enhancing the permeability and retention of nGOx into tumor cells by EPR effect. nGOx can deplete glucose in the presence of oxygen and generate H2O2, which is converted to highly cytotoxic hydroxyl radical (·OH) by Fe3+ and GSH, and the proximity between Fe3+ and GOx act in tandem for enhanced tumor therapy. The FeIII/FeII redox cycle reacts with GSH and H2O2, enabling continuous generation of ·OH within tumor cells, thus facilitating the anticancer effect. Moreover, the generation of H2O2 and ·OH can further promote the repolarization of tumor-associated macrophages from an M2 phenotype towards an M1 phenotype polarization, thus enhancing immune response. The cascade reaction between GOx and Fe3+/Fe2+ endows Fe@nGOx with excellent anti-tumor efficacy in mice models, highlighting its potential as a promising anticancer drug for clinical applications. This work establishes a new platform for utilizing enzyme/protein and metal ion complexes in versatile applications, advancing the field of enzyme-based cancer therapies.
Collapse
Affiliation(s)
- Liuzhou Mao
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, PR China
| | - Yedong Ma
- Department of Materials Science and Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Republic of Singapore
| | - Xiaoqing Wen
- Drug clinical trial institution, The first affiliated hospital of Xiamen university, Xiamen, Fujian, PR China
| | - Zheng Luo
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, PR China
| | - Houjuan Zhu
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology, and Research (A∗STAR), 2 Fusionopolis Way, Innovis, Singapore 138634, Republic of Singapore
| | - Junhua Kong
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology, and Research (A∗STAR), 2 Fusionopolis Way, Innovis, Singapore 138634, Republic of Singapore
| | - Siqi Liu
- Department of Materials Science and Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Republic of Singapore
| | - Xiaotong Fan
- Institute of Sustainability for Chemicals, Energy and Environment (ISCE2), Agency for Science, Technology and Research (A*STAR), 1 Pesek Road, Jurong Island, Singapore 627833, Republic of Singapore.
| | - Jinling Wang
- Department of Emergency and Critical Care Center, The Second Affiliated Hospital of Guangdong Medical University, No. 12 Minyou Road, Xiashan, Zhanjiang, Guangdong 524003, PR China.
| | - Chaobin He
- Department of Materials Science and Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Republic of Singapore; Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology, and Research (A∗STAR), 2 Fusionopolis Way, Innovis, Singapore 138634, Republic of Singapore.
| | - Yun-Long Wu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, PR China.
| |
Collapse
|
25
|
Jang JH, Kim DH, Chun KS. Tumor microenvironment regulation by reactive oxygen species-mediated inflammasome activation. Arch Pharm Res 2025; 48:115-131. [PMID: 39888519 DOI: 10.1007/s12272-025-01532-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 01/16/2025] [Indexed: 02/01/2025]
Abstract
Tumor microenvironment (TME) is composed of diverse cell types whose interactions, both direct and indirect, significantly influence tumorigenesis and therapeutic outcomes. Within TME, reactive oxygen species (ROS) are produced by various cells and exhibit a dual role: moderate ROS levels promote tumor initiation and progression, whereas excessive levels induce cancer cell death, influencing the efficacy of anticancer therapies. Inflammasomes, cytosolic multiprotein complexes, are pivotal in multiple stages of tumorigenesis and play a crucial role in establishing the inflammatory TME. By releasing cytokines such as IL-1β and IL-18, inflammasomes contribute to immune cell recruitment and sustain a chronic inflammatory state that supports tumor growth. ROS are critical regulators of inflammasome activation, with the impact of ROS-mediated activation differing across cell types, leading to distinct influences on tumor progression and therapeutic responses. This review explores how ROS drive inflammasome activation in various TME-associated cells and the reciprocal ROS generation induced by inflammasomes, examining their multifaceted impact on tumorigenesis and therapeutic efficacy. By elucidating the complex interplay between ROS and inflammasomes in TME, we provide insights into potential therapeutic approaches that could modulate cancer progression and enhance treatment outcomes.
Collapse
Affiliation(s)
- Jeong-Hoon Jang
- College of Pharmacy, Daegu Catholic University, Gyeongsan-si, Gyeongbuk, 38430, Republic of Korea
| | - Do-Hee Kim
- Department of Chemistry, Kyonggi University, Suwon, 16227, Republic of Korea
| | - Kyung-Soo Chun
- College of Pharmacy, Keimyung University, Daegu, 42601, Republic of Korea.
| |
Collapse
|
26
|
Zhu L, Liu Y, Wang K, Wang N. Regulated cell death in acute myocardial infarction: Molecular mechanisms and therapeutic implications. Ageing Res Rev 2025; 104:102629. [PMID: 39644925 DOI: 10.1016/j.arr.2024.102629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 11/22/2024] [Accepted: 12/03/2024] [Indexed: 12/09/2024]
Abstract
Acute myocardial infarction (AMI), primarily caused by coronary atherosclerosis, initiates a series of events that culminate in the obstruction of coronary arteries, resulting in severe myocardial ischemia and hypoxia. The subsequent myocardial ischemia/reperfusion (I/R) injury further aggravates cardiac damage, leading to a decline in heart function and the risk of life-threatening complications. The complex interplay of multiple regulated cell death (RCD) pathways plays a pivotal role in the pathogenesis of AMI. Each RCD pathway is orchestrated by a symphony of molecular regulatory mechanisms, highlighting the dynamic changes and critical roles of key effector molecules. Strategic disruption or inhibition of these molecular targets offers a tantalizing prospect for mitigating or even averting the onset of RCD, thereby limiting the extensive loss of cardiomyocytes and the progression of detrimental myocardial fibrosis. This review systematically summarizes the mechanisms underlying various forms of RCD, provides an in-depth exploration of the pathogenesis of AMI through the lens of RCD, and highlights a range of promising therapeutic targets that hold the potential to revolutionize the management of AMI.
Collapse
Affiliation(s)
- Lili Zhu
- Department of Pathology, Hunan Cancer Hospital, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yiyang Liu
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China; Key Laboratory of Sepsis Translational Medicine of Hunan, Central South University, Changsha, Hunan, China; National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan, China
| | - Kangkai Wang
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China; Key Laboratory of Sepsis Translational Medicine of Hunan, Central South University, Changsha, Hunan, China; National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan, China
| | - Nian Wang
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China; Key Laboratory of Sepsis Translational Medicine of Hunan, Central South University, Changsha, Hunan, China; National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan, China.
| |
Collapse
|
27
|
Lu J, Lu F, Peng Z, Zhang Z, Jiang W, Meng X, Yi X, Chen T, Fei Z, Wang Y, Yi J, Deng X, Zhang J, Wang Z, Xiao Q. Clodronate liposome-mediated macrophage depletion ameliorates iron overload-induced dry eye disease. Exp Eye Res 2025; 251:110204. [PMID: 39662663 DOI: 10.1016/j.exer.2024.110204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 10/08/2024] [Accepted: 12/08/2024] [Indexed: 12/13/2024]
Abstract
Dry eye disease (DED) is a prevalent ophthalmic disease that affects millions of people worldwide. Iron overload and macrophage inflammation have been implicated in the development of murine DED, though the specific role of macrophages under iron overload conditions remains unclear. This study aimed to establish a novel iron overload-induced mouse model of DED and investigate macrophage involvement. The model was induced via intraperitoneal injection of D-glucoside iron. Results showed that macrophage depletion via clodronate liposomes (CL) significantly mitigated iron deposit, decreased ocular surface inflammation, improved tear production and restored the structure of ocular surface tissues. Furthermore, CL specifically targeted pro-inflammatory M1 macrophages and reduced levels of the inflammatory cytokines IL-1β, IL-6, and TNF-α, effectively alleviating symptoms of DED. In conclusion, this study characterized a novel iron overload-induced DED mouse model and demenstrated that macrophage depletion mitigated the pathological changes in ocular surface and lacrimal gland tissues caused by iron overload, suggesting potential therapeutic strategies for further investigation in the treatment of DED.
Collapse
Affiliation(s)
- Jing Lu
- The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Fangfang Lu
- The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Zhengwu Peng
- The Affiliated Chenzhou Hospital, Hengyang Medical School, University of South China, Chenzhou, 423000, Hunan, China
| | - Zihe Zhang
- The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Weijie Jiang
- The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Xia Meng
- The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Xin Yi
- The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Tuo Chen
- The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Zhigang Fei
- The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Yu Wang
- The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Jiahuan Yi
- The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Xujie Deng
- The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Jia Zhang
- The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| | - Zhi Wang
- The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| | - Qiguo Xiao
- The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| |
Collapse
|
28
|
Hanna M, Akabawy AMA, Khalifa MM, Elbaset MA, Imam RA, Seddiek H. Intracellular iron accumulation throughout the progression of sepsis influences the phenotype and function of activated macrophages in renal tissue damage. Front Physiol 2025; 16:1430946. [PMID: 39949667 PMCID: PMC11821637 DOI: 10.3389/fphys.2025.1430946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 01/06/2025] [Indexed: 02/16/2025] Open
Abstract
Sepsis, the most common cause of acute kidney injury, remains a major socioeconomic burden. A dysregulated immune response leads to progressive organ dysfunction. Although numerous inflammatory pathways were described, most are still vague and need to be studied in terms of the mechanisms to improve the therapeutic intervention. We tackled the relationship between intracellular iron overload and macrophage polarization within 6, 24, and 72 h of sepsis induction. In our study, sepsis-induced kidney injury was caused by using the cecal ligation and puncture (CLP) model. Our results indicated severe renal tissue damage with a progressive increase in serum BUN and creatinine with architectural tissue damage and positive PAS staining. There was increased expression of CD8+ CD68+ M1 macrophage markers with upregulation of iNOS and co-expression of CD163+. Alternatively, Arg1+ Fizz1+ M2 macrophage markers were downregulated with increased iNOS/Arg1 ratio. TFR1, cubilin, and DMT1, as iron transport systems, were increased compared to sham but were significant after 72 h, while ZIP8 showed no significant change. There was a correlation between iron overload and M1 macrophage polarization with CD163+ phenotype, together with fibrotic changes. The intracellular iron overload with downregulation of ferritin was strongly related to macrophage polarization that was exaggerated at 72 h. Finally, early introduced therapy to target free iron during sepsis is a proposed novel solution for protecting the renal tissue from acute injury due to macrophage activation that may end up with chronic kidney injury, if not mortality.
Collapse
Affiliation(s)
- Mira Hanna
- Department of Medical Physiology, Faculty of Medicine, Kasr Al-Ainy, Cairo University, Cairo, Egypt
| | - Ahmed M. A. Akabawy
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Mohamed Mansour Khalifa
- Department of Medical Physiology, Faculty of Medicine, Kasr Al-Ainy, Cairo University, Cairo, Egypt
- Department of Medical Physiology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Marawan Abd Elbaset
- Department of Pharmacology, Medical Research and Clinical Studies Institute, National Research Centre, Cairo, Egypt
| | - Reda Abdelnasser Imam
- Department of Anatomy and Embryology, Faculty of Medicine, Kasr Al-Ainy, Cairo University, Cairo, Egypt
| | - Hanan Seddiek
- Department of Medical Physiology, Faculty of Medicine, Kasr Al-Ainy, Cairo University, Cairo, Egypt
| |
Collapse
|
29
|
Yuan J, Liu C, Jiang C, Liu N, Yang Z, Xing H. RSL3 induces ferroptosis by activating the NF-κB signalling pathway to enhance the chemosensitivity of triple-negative breast cancer cells to paclitaxel. Sci Rep 2025; 15:1654. [PMID: 39794456 PMCID: PMC11724089 DOI: 10.1038/s41598-025-85774-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 01/06/2025] [Indexed: 01/13/2025] Open
Abstract
Chemotherapy resistance in triple-negative breast cancer (TNBC) leads to poor therapeutic effects and a poor prognosis. Given that paclitaxel-based chemotherapy is the main treatment method for TNBC, enhancing its chemosensitivity has been a research focus. Induced ferroptosis of tumour cells has been proven to increase chemosensitivity, but its ability to sensitize TNBC cells to paclitaxel (PTX) is unknown. In our experiments, measurements of viability and proliferation validated the synergistic effect of PTX combined with RSL3 on TNBC cells. The accumulation of intracellular Fe2+ and lipid reactive oxygen species, as well as the expression of malondialdehyde, illustrated that RSL3 enhanced the chemosensitivity of TNBC to PTX by inducing ferroptosis. Through transcriptome sequencing, a series of differentially expressed genes were identified, in which the expression of cytokines, such as CXCLs, was significantly increased in the treatment group, and the effect of combination therapy on TNBC was enriched mainly in the NFκB signalling pathway. In subsequent validation experiments, the use of the NF-κB inhibitor BAY11-7082 reversed the inhibitory effects of PTX and RSL3 on TNBC cell activity. In a xenograft immunodeficient mouse model, the inhibitory effects of PTX and RSL3 on TNBC in vivo were further verified. Our research validated the synergistic effects of PTX and RSL3 both in vivo and in vitro, with RSL3 inducing ferroptosis by activating the NF-κB signalling pathway, thereby increasing the chemosensitivity of TNBC to PTX. This study provides new insights for improving the therapeutic efficacy of treatment strategies.
Collapse
Affiliation(s)
- Jialin Yuan
- Department of Breast Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Cong Liu
- Department of Breast Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Chengwei Jiang
- Department of Pathology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Ning Liu
- Department of Breast Surgery, China-Japan Union Hospital of Jilin University, Changchun, China.
| | - Zhaoying Yang
- Department of Breast Surgery, China-Japan Union Hospital of Jilin University, Changchun, China.
| | - Hua Xing
- Department of Breast Surgery, China-Japan Union Hospital of Jilin University, Changchun, China.
| |
Collapse
|
30
|
Sun Z, Qin Y, Zhang X. Identification and validation of five ferroptosis-related molecular signatures in keloids based on multiple transcriptome data analysis. Front Mol Biosci 2025; 11:1490745. [PMID: 39834787 PMCID: PMC11743277 DOI: 10.3389/fmolb.2024.1490745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 11/27/2024] [Indexed: 01/22/2025] Open
Abstract
Introduction Keloids are a common skin disorder characterized by excessive fibrous tissue proliferation, which can significantly impact patients' health. Ferroptosis, a form of regulated cell death, plays a crucial role in the development of fibrosis; however, its role in the mechanisms of keloid formation remains poorly understood. Methods This study aimed to identify key genes associated with ferroptosis in keloid formation. Data from the NCBI GEO database, including GSE145725, GSE7890, and GSE44270, were analyzed, comprising a total of 24 keloid and 17 normal skin samples. Additionally, single-cell data from GSE181316, which included 8 samples with complete expression profiles, were also evaluated. Differentially expressed genes were identified, and ferroptosis-related genes were extracted from the GeneCards database. LASSO regression was used to select key genes associated with keloids. Validation was performed using qRT-PCR and Western blot (WB) analysis on tissue samples from five keloid and five normal skin biopsies. Results A total of 471 differentially expressed genes were identified in the GSE145725 dataset, including 225 upregulated and 246 downregulated genes. Five ferroptosis-related genes were selected through gene intersection and LASSO regression. Two of these genes were upregulated, while three were downregulated in keloid tissue. Further analysis through GSEA pathway enrichment, GSVA gene set variation, immune cell infiltration analysis, and single-cell sequencing revealed that these genes were primarily involved in the fibrotic process. The qRT-PCR and WB results confirmed the expression patterns of these genes. Discussion This study provides novel insights into the molecular mechanisms of ferroptosis in keloid formation. The identified ferroptosis-related genes could serve as potential biomarkers or therapeutic targets for treating keloids.
Collapse
Affiliation(s)
| | - Yonghong Qin
- Department of Plastic Surgery, Second Hospital and Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Xuanfen Zhang
- Department of Plastic Surgery, Second Hospital and Clinical Medical School, Lanzhou University, Lanzhou, China
| |
Collapse
|
31
|
Chen L, Hu P, Fang W, Wu T, Shi J. Nebulized Immunotherapy of Orthotopic Lung Cancer by Mild Magnetothermal-Based Innate Immunity Activations. Angew Chem Int Ed Engl 2025; 64:e202413127. [PMID: 39343740 DOI: 10.1002/anie.202413127] [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: 07/12/2024] [Revised: 09/08/2024] [Accepted: 09/27/2024] [Indexed: 10/01/2024]
Abstract
Advances in adaptive immunity have greatly contributed to the development of cancer immunotherapy. However, its over-low efficacy and insufficient invasion of immune cells in the tumor tissue, and safety problems caused by cytokine storm, have seriously impeded further clinical application for solid tumor immunotherapy. Notably, the immune microenvironment of the lungs is naturally enriched with alveolar macrophages (AMs). Herein, we introduce a novel nebulized magnetothermal immunotherapy strategy to treat orthotopic lung cancer by using magnetothermal nanomaterial (Zn-CoFe2O4@Zn-MnFe2O4-PEG, named ZCMP), which can release iron ions via an acid/thermal-catalytic reaction to maximize the use of lung's immune environment through the cascade activations of AMs and natural killer (NK) cells. Nebulized administration greatly enhance drug bioavailability by localized drug accumulation at the lesion site. Upon mild magnetic hyperthermia, the released iron ions catalyze endogenous H2O2 decomposition to produce reactive oxygen species (ROS), which triggers the M1 polarization of AMs, and the resultant inflammatory cytokine IFN-β, IL-1β and IL-15 releases to activate c-Jun, STAT5 and GZMB related signaling pathways, promoting NK cells proliferation and activation. This innovative strategy optimally utilizes the lung's immune environment and shows excellent immunotherapeutic outcomes against orthotopic lung cancer.
Collapse
Affiliation(s)
- Lizhu Chen
- School of Chemical Science and Engineering, Institute of Advanced Study, Shanghai Key Laboratory of Chemical Assessment and Sustainability, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, 200092, Shanghai, P.R. China
| | - Ping Hu
- School of Chemical Science and Engineering, Institute of Advanced Study, Shanghai Key Laboratory of Chemical Assessment and Sustainability, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, 200092, Shanghai, P.R. China
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, 200050, Shanghai, P.R. China
| | - Wenming Fang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, 200050, Shanghai, P.R. China
| | - Tong Wu
- School of Chemical Science and Engineering, Institute of Advanced Study, Shanghai Key Laboratory of Chemical Assessment and Sustainability, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, 200092, Shanghai, P.R. China
| | - Jianlin Shi
- School of Chemical Science and Engineering, Institute of Advanced Study, Shanghai Key Laboratory of Chemical Assessment and Sustainability, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, 200092, Shanghai, P.R. China
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, 200050, Shanghai, P.R. China
| |
Collapse
|
32
|
Li G, Wu M, Chen K, Xu Y, Zhang X, Chen Y, Zhang H, Zhang R, Huang X. ROS-mediated M1 polarization-necroptosis crosstalk involved in Di-(2-ethylhexyl) phthalate-induced chicken liver injury. Poult Sci 2025; 104:104558. [PMID: 39631278 PMCID: PMC11665341 DOI: 10.1016/j.psj.2024.104558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 11/10/2024] [Accepted: 11/21/2024] [Indexed: 12/07/2024] Open
Abstract
The widespread use of plasticizers poses a serious threat to the environment and poultry health. Di-(2-ethylhexyl) phthalate (DEHP) is a commonly used plasticizer that can cause liver damage with prolonged exposure. Oxidative stress is closely associated with DEHP toxicity. Macrophage polarization plays an important role in many physiological and pathological processes and regulates disease development. This study aims to elucidate the mechanism of chronic DEHP exposure leading to chicken liver injury through oxidative stress-induced M1 polarization-necroptosis. In this study, the DEHP exposure model of chicken liver and the single and co-culture model of LMH and HD11 cells were established. With increasing dose and time, DEHP decreased body weight, increased liver coefficient, raised activities of liver function indicators and caused pathological liver damage in chickens. Further studies revealed the increase of reactive oxygen species (ROS) level and malonaldehyde (MDA) content, and the decrease of total antioxidant capacity (T-AOC) level, total superoxide dismutase (T-SOD) and glutathione peroxidase (GSH-Px) activities, which led to excessive oxidative stress in the liver. In addition, there was increased infiltration of liver macrophages (CD68), upregulation of M1 polarization indicators (CD86, iNOS, IL-1β, TNF-α) and downregulation of M2 polarization indicators (CD163, Arg-1, IL-10, TGF-β) and appearance of necroptosis (RIPK1, RIPK3, MLKL). The vitro experiments confirmed the addition of N-acetylcysteine (NAC) inhibited M1 polarization and necroptosis. Besides, M1 polarization of HD11 cells promoted necroptosis of LMH cells in the HD11-LMH co-culture system. In brief, ROS-mediated M1 polarization-necroptosis is involved in DEHP-induced liver injury. This study provides a reference for environmental toxicant exposure in livestock and poultry farming.
Collapse
Affiliation(s)
- Guangxing Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Menglin Wu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Kaiting Chen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Yue Xu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Xiandan Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Yang Chen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Hong Zhang
- Liaoning Petmate Biotechnology Co, PR China
| | - Ruili Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Xiaodan Huang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China.
| |
Collapse
|
33
|
Mo Y, Zou Z, Chen E. [Research progress on ferroptosis regulation in tumor immunity of hepatocellular carcinoma]. Zhejiang Da Xue Xue Bao Yi Xue Ban 2024; 53:715-725. [PMID: 39694527 PMCID: PMC11726010 DOI: 10.3724/zdxbyxb-2024-0117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 06/30/2024] [Indexed: 12/20/2024]
Abstract
Ferroptosis is a form of regulated cell death, which is dependent on iron metabolism imbalance and characterized by lipid peroxidation. Ferroptosis plays a crucial role in various pathological processes. Studies have shown that the occurrence of ferroptosis is closely associated with the progression of hepatocellular carcinoma (HCC). Ferroptosis is involved in regulating the lipid metabolism, iron homeostasis, mitochondrial metabolism, and redox processes in HCC. Additionally, ferroptosis plays a key role in HCC tumor immunity by modulating the phenotype and function of various immune cells in the tumor microenvironment, affecting tumor immune escape and progression. Ferroptosis-induced lipid peroxidation and oxidative stress can promote the polarization of M1 macrophages and enhance the pro-inflammatory response in tumors, inhibiting immune suppressive cells such as myeloid-derived suppressor cells and regulatory T cells to disrupt their immune suppression function. The regulation of expression of ferroptosis-related molecules such as GPX4 and SLC7A11 not only affects the sensitivity of tumor cells to immunotherapy but also directly influences the activity and survival of effector cells such as T cells and dendritic cells, further enhancing or weakening host antitumor immune response. Targeting ferroptosis has demonstrated significant clinical potential in HCC treatment. Induction of ferroptosis by nanomedicines and molecular targeting strategies can directly kill tumor cells or enhance antitumor immune responses. The integration of multimodal therapies with immunotherapy further expands the application of ferroptosis targeting as a cancer therapy. This article reviews the relationship between ferroptosis and antitumor immune responses and the role of ferroptosis in HCC progression from the perspective of tumor immune microenvironment, to provide insights for the development of antitumor immune therapies targeting ferroptosis.
Collapse
Affiliation(s)
- Yuqian Mo
- School of Public Health, Guangdong Medical University, Dongguan 523808, Guangdong Province, China.
| | - Zhilin Zou
- School of Public Health, Guangdong Medical University, Dongguan 523808, Guangdong Province, China
| | - Erbao Chen
- Department of Hepatobiliary and Pancreatic Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, Guangdong Province, China.
| |
Collapse
|
34
|
Zhang P, Liu W, Wang S, Wang Y, Han H. Ferroptosisand Its Role in the Treatment of Sepsis-Related Organ Injury: Mechanisms and Potential Therapeutic Approaches. Infect Drug Resist 2024; 17:5715-5727. [PMID: 39720615 PMCID: PMC11668052 DOI: 10.2147/idr.s496568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 12/12/2024] [Indexed: 12/26/2024] Open
Abstract
Sepsis is a complicated clinical disease caused by a defective host response to infection, leading to elevated morbidity and fatality globally. Sepsis patients have a significant risk of life-threatening organ damage, including hearts, brains, lungs, kidneys, and livers. Nevertheless, the molecular pathways driving organ injury in sepsis are not well known. Ferroptosis, a non-apoptotic cell death, occurs due to iron metabolism disturbance and lipid peroxide buildup. Multiple studies indicate that ferroptosis has a significant role in decreasing inflammation and lipid peroxidation during sepsis. Ferroptosis inhibitors and medications, aimed at the most studied ferroptosis process, including Xc-system, Nrf2/GPX4 axis, and NCOA4-FTH1-mediated ferritinophagy, alleviating sepsis effectively. However, few clinical trials demonstrated ferroptosis-targeted drugs's effectiveness in sepsis. Our study examines ferroptosis-targeted medicinal agents and their potential benefits for treating sepsis-associated organ impairment. This review indicates that ferroptosis suppression by pharmaceutical means may be a useful therapy for sepsis-associated organ injury.
Collapse
Affiliation(s)
- Pengyu Zhang
- The Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People’s Republic of China
| | - Wendi Liu
- School of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People’s Republic of China
| | - Shu Wang
- School of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People’s Republic of China
| | - Yuan Wang
- Department of Histology and Embryology, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People’s Republic of China
| | - Han Han
- School of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People’s Republic of China
| |
Collapse
|
35
|
Lu S, Liu Z, Qi M, Wang Y, Chang L, Bai X, Jiao Y, Chen X, Zhen J. Ferroptosis and its role in osteoarthritis: mechanisms, biomarkers, and therapeutic perspectives. Front Cell Dev Biol 2024; 12:1510390. [PMID: 39744014 PMCID: PMC11688369 DOI: 10.3389/fcell.2024.1510390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 12/04/2024] [Indexed: 01/04/2025] Open
Abstract
Osteoarthritis (OA) is one of the leading causes of disability worldwide, characterized by a complex pathological process involving cartilage degradation, synovial inflammation, and subchondral bone remodeling. In recent years, ferroptosis, a form of programmed cell death driven by iron-dependent lipid peroxidation, has been recognized as playing a critical role in the onset and progression of OA. Investigating the molecular mechanisms of ferroptosis and its involvement in OA may offer novel strategies for diagnosing and treating this disease. This review first outlines the core mechanisms of ferroptosis, with a particular focus on the roles of critical molecules such as Glutathione Peroxidase 4 (GPX4), Transferrin Receptor 1 (TfR1), and Nuclear Receptor Coactivator 4 (NCOA4). Subsequently, this study examines the specific impacts of ferroptosis on the pathophysiology of OA. Building on this, the potential of ferroptosis-related biomarkers for OA diagnosis and treatment is highlighted, along with proposed therapeutic strategies targeting ferroptosis regulation. This review aims to deepen the understanding of ferroptosis mechanisms and advance the clinical application of regulatory therapies for OA.
Collapse
Affiliation(s)
- Shanyu Lu
- College of Medical Imaging, Shanxi Medical University, Taiyuan, Shanxi, China
- Department of Imaging, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory for Immunomicroecology, Taiyuan, Shanxi, China
| | - Zhenyu Liu
- College of Medical Imaging, Shanxi Medical University, Taiyuan, Shanxi, China
- Department of Imaging, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory for Immunomicroecology, Taiyuan, Shanxi, China
| | - Meiling Qi
- College of Medical Imaging, Shanxi Medical University, Taiyuan, Shanxi, China
- Department of Imaging, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory for Immunomicroecology, Taiyuan, Shanxi, China
| | - Yingchao Wang
- College of Medical Imaging, Shanxi Medical University, Taiyuan, Shanxi, China
- Department of Imaging, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Le Chang
- College of Medical Imaging, Shanxi Medical University, Taiyuan, Shanxi, China
- Department of Imaging, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiaolong Bai
- College of Medical Imaging, Shanxi Medical University, Taiyuan, Shanxi, China
- Department of Imaging, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yingguang Jiao
- College of Medical Imaging, Shanxi Medical University, Taiyuan, Shanxi, China
- Department of Imaging, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xinyao Chen
- College of Medical Imaging, Shanxi Medical University, Taiyuan, Shanxi, China
- Department of Imaging, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Junping Zhen
- College of Medical Imaging, Shanxi Medical University, Taiyuan, Shanxi, China
- Department of Imaging, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Molecular Imaging Laboratory, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
36
|
Hajra D, Rajmani RS, Chaudhary AD, Gupta SK, Chakravortty D. Salmonella-induced SIRT1 and SIRT3 are crucial for maintaining the metabolic switch in bacteria and host for successful pathogenesis. eLife 2024; 13:RP93125. [PMID: 39693143 PMCID: PMC11655064 DOI: 10.7554/elife.93125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2024] Open
Abstract
Sirtuins are the major players in host immunometabolic regulation. However, the role of sirtuins in the modulation of the immune metabolism pertaining to salmonellosis is largely unknown. Here, our investigation focussed on the role of two important sirtuins, SIRT1 and SIRT3, shedding light on their impact on intracellular Salmonella's metabolic switch and pathogenesis establishment. Our study indicated the ability of the live Salmonella Typhimurium to differentially regulate the levels of SIRT1 and SIRT3 for maintaining the high glycolytic metabolism and low fatty acid metabolism in Salmonella. Perturbing SIRT1 or SIRT3 through knockdown or inhibition resulted in a remarkable shift in the host metabolism to low fatty acid oxidation and high glycolysis. This switch led to decreased proliferation of Salmonella in the macrophages. Further, Salmonella-induced higher levels of SIRT1 and SIRT3 led to a skewed polarization state of the macrophages from a pro-inflammatory M1 state toward an immunosuppressive M2, making it more conducive for the intracellular life of Salmonella. Alongside, governing immunological functions by modulating p65 NF-κB acetylation, SIRT1, and SIRT3 also skew Salmonella-induced host metabolic switch by regulating the acetylation status of HIF-1α and PDHA1. Interestingly, though knockdown of SIRT1/3 attenuated Salmonella proliferation in macrophages, in in vivo mice model of infection, inhibition or knockdown of SIRT1/3 led to more dissemination and higher organ burden, which can be attributed to enhanced ROS and IL-6 production. Our study hence reports for the first time that Salmonella modulates SIRT1/3 levels to maintain its own metabolism for successful pathogenesis.
Collapse
Grants
- SPM-07/079(0293)/2019-EMR-I (CSIR Shyama Prasad Mukherjee Fellowship) Council of Scientific and Industrial Research, India
- DAE00195 Department of Atomic Energy, Government of India
- DBT-IISC Department of Biotechnology, Ministry of Science & Technology, India
- BT/RLF/re-entry/14/2019 Department of Biotechnology, Ministry of Science and Technology, India
- Department of Biotechnology, Ministry of Science & Technology, India
- Indian Council of Medical Research
- Department of Science & Technology, Ministry of Science and Technology, India
Collapse
Affiliation(s)
- Dipasree Hajra
- Department of Microbiology and Cell Biology, Indian Institute of ScienceBangaloreIndia
| | - Raju S Rajmani
- Centre of Infectious Disease Research, Indian Institute of ScienceBangaloreIndia
| | - Ayushi Devendrasingh Chaudhary
- Pharmacology Division, CSIR-Central Drug Research InstituteLucknowIndia
- Academy of Scientific and Innovative Research (AcSIR)GhaziabadIndia
| | - Shashi Kumar Gupta
- Pharmacology Division, CSIR-Central Drug Research InstituteLucknowIndia
- Academy of Scientific and Innovative Research (AcSIR)GhaziabadIndia
| | - Dipshikha Chakravortty
- Department of Microbiology and Cell Biology, Indian Institute of ScienceBangaloreIndia
- Adjunct Faculty, School of Biology, Indian Institute of Science Education and ResearchThiruvananthapuramIndia
| |
Collapse
|
37
|
O'Reilly A, Zhao W, Wickström S, Arnér ESJ, Kiessling R. Reactive oxygen species: Janus-faced molecules in the era of modern cancer therapy. J Immunother Cancer 2024; 12:e009409. [PMID: 39645234 PMCID: PMC11629020 DOI: 10.1136/jitc-2024-009409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 11/11/2024] [Indexed: 12/09/2024] Open
Abstract
Oxidative stress, that is, an unbalanced increase in reactive oxygen species (ROS), contributes to tumor-induced immune suppression and limits the efficacy of immunotherapy. Cancer cells have inherently increased ROS production, intracellularly through metabolic perturbations and extracellularly through activation of NADPH oxidases, which promotes cancer progression. Further increased ROS production or impaired antioxidant systems, induced, for example, by chemotherapy or radiotherapy, can preferentially kill cancer cells over healthy cells. Inflammatory cell-derived ROS mediate immunosuppressive effects of myeloid-derived suppressor cells and activated granulocytes, hampering antitumor effector cells such as T cells and natural killer (NK) cells. Cancer therapies modulating ROS levels in tumors may thus have entirely different consequences when targeting cancer cells versus immune cells. Here we discuss the possibility of developing more efficient cancer therapies based on reduction-oxidation modulation, as either monotherapies or in combination with immunotherapy. Short-term, systemic administration of antioxidants or drugs blocking ROS production can boost the immune system and act in synergy with immunotherapy. However, prolonged use of antioxidants can instead enhance tumor progression. Alternatives to systemic antioxidant administration are under development where gene-modified or activated T cells and NK cells are shielded ex vivo against the harmful effects of ROS before the infusion to patients with cancer.
Collapse
Affiliation(s)
- Aine O'Reilly
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Department of Medicine, University College Cork, Cork, Ireland
- The Christie NHS Foundation Trust, Manchester, UK
| | - Wenchao Zhao
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Stina Wickström
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Theme Cancer, Patient area Head and Neck, Lung and Skin, Karolinska University Hospital, Stockholm, Sweden
| | - Elias S J Arnér
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
- Department of Selenoprotein Research and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, Hungary
| | - Rolf Kiessling
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Theme Cancer, Patient area Head and Neck, Lung and Skin, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
38
|
Zhu Z, Cao H, Yan H, Liu H, Hong Z, Sun A, Liu T, Mao F. Prognostic iron-metabolism signature robustly stratifies single-cell characteristics of hepatocellular carcinoma. Comput Struct Biotechnol J 2024; 23:929-941. [PMID: 38375529 PMCID: PMC10875160 DOI: 10.1016/j.csbj.2024.01.022] [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: 10/26/2023] [Revised: 01/23/2024] [Accepted: 01/29/2024] [Indexed: 02/21/2024] Open
Abstract
Cancer immunotherapy has shown to be a promising method in treating hepatocellular carcinoma (HCC), but suboptimal responses in patients are attributed to cellular and molecular heterogeneity. Iron metabolism-related genes (IRGs) are important in maintaining immune system homeostasis and have the potential to help develop new strategies for HCC treatment. Herein, we constructed and validated the iron-metabolism gene prognostic index (IPX) using univariate Cox proportional hazards regression and LASSO Cox regression analysis, successfully categorizing HCC patients into two groups with distinct survival risks. Then, we performed single-sample gene set enrichment analysis, weighted correlation network analysis, gene ontology enrichment analysis, cellular lineage analysis, and SCENIC analysis to reveal the key determinants underlying the ability of this model based on bulk and single-cell transcriptomic data. We identified several driver transcription factors specifically activated in specific malignant cell sub-populations to contribute to the adverse survival outcomes in the IPX-high subgroup. Within the tumor microenvironment (TME), T cells displayed significant diversity in their cellular characteristics and experienced changes in their developmental paths within distinct clusters identified by IPX. Interestingly, the proportion of Treg cells was increased in the high-risk group compared with the low-risk group. These results suggest that iron-metabolism could be involved in reshaping the TME, thereby disrupting the cell cycle of immune cells. This study utilized IRGs to construct a novel and reliable model, which can be used to assess the prognosis of patients with HCC and further clarify the molecular mechanisms of IRGs in HCC at single-cell resolution.
Collapse
Affiliation(s)
- Zhipeng Zhu
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing 100191, China
- Cancer Center, Peking University Third Hospital, Beijing 100191, China
| | - Huang Cao
- School of Medicine, Xiamen University, Xiamen, Fujian 361100, China
| | - Hongyu Yan
- School of Medicine, Xiamen University, Xiamen, Fujian 361100, China
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Hanzhi Liu
- The Third Clinical Medical College, Gannan Medical University, Ganzhou, Jiangxi 341000, China
| | - Zaifa Hong
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, School of Medicine, Xiamen University, Xiamen, Fujian 361103, China
| | - Anran Sun
- Oncology Research Center, Foresea Life Insurance Guangzhou General Hospital, Guangzhou, Guangdong 511300, China
- Research Center for Translational Medicine, The First Affiliated Hospital, School of Medicine, Xiamen University, Xiamen, Fujian 361003, China
| | - Tong Liu
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing 100191, China
- Cancer Center, Peking University Third Hospital, Beijing 100191, China
| | - Fengbiao Mao
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing 100191, China
- Cancer Center, Peking University Third Hospital, Beijing 100191, China
| |
Collapse
|
39
|
Zhao Y, Zheng G, Yang S, Liu S, Wu Y, Miao Y, Liang Z, Hua Y, Zhang J, Shi J, Li D, Cheng Y, Zhang Y, Chen Y, Fan G, Ma C. The plant extract PNS mitigates atherosclerosis via promoting Nrf2-mediated inhibition of ferroptosis through reducing USP2-mediated Keap1 deubiquitination. Br J Pharmacol 2024; 181:4822-4844. [PMID: 39228119 DOI: 10.1111/bph.17311] [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: 12/14/2023] [Revised: 06/04/2024] [Accepted: 06/23/2024] [Indexed: 09/05/2024] Open
Abstract
BACKGROUND AND PURPOSE Atherosclerosis is the basis of cardiovascular disease. Ferroptosis is a form of programmed cell death characterized by lipid peroxidation, which contributes to atherogenesis. The plant extract PNS (Panax notoginseng saponins), containing the main active ingredients of Panax notoginseng, exhibits anti-atherogenic properties. Herein, we determined whether PNS and its major components could attenuate atherosclerosis by suppressing ferroptosis and revealed the underlying mechanism(s). EXPERIMENTAL APPROACH The anti-atherogenic effects of PNS and their association with inhibition of ferroptosis was determined in apoE-/- mice. In vitro, the anti-ferroptotic effect and mechanism(s) of PNS components were demonstrated in the presence of ferroptosis inducers. Expression of ferroptosis markers and the ubiquitination of Keap1 were evaluated in USP2-/- macrophages. Finally, the anti-atherogenic effect of USP2 knockout was determined by using USP2-/- mice treated with high-fat diet (HFD) and AAV-PCSK9. KEY RESULTS PNS inhibited ferroptosis and atherosclerosis in vivo. PNS suppressed ferroptosis and ferroptosis-aggravated foam cell formation and inflammation in vitro. Mechanistically, PNS and its components activated Nrf2 by antagonizing Keap1, which was attributed to the inhibition of USP2 expression. USP2 knockout antagonized ferroptosis and ferroptosis-aggravated foam cell formation and inflammation, thus mitigating atherosclerosis. USP2 knockout abolished inhibitory effects of PNS on foam cell formation and inflammation in vitro. CONCLUSION AND IMPLICATIONS PNS reduced USP2-mediated Keap1 de-ubiquitination and promoted Keap1 degradation, thereby activating Nrf2, improving iron metabolism and reducing lipid peroxidation, thus contributing to an anti-atherosclerotic outcome. Our study revealed the mechanism(s) underlying inhibition of ferroptosis and atherosclerosis by PNS.
Collapse
Affiliation(s)
- Yun Zhao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Guobin Zheng
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Shu Yang
- Department of Geriatrics, The First Affiliated Hospital of Southern University of Science and Technology (Shenzhen People's Hospital), Shenzhen, China
| | - Shangjing Liu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yifan Wu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yaodong Miao
- Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhen Liang
- Department of Geriatrics, The First Affiliated Hospital of Southern University of Science and Technology (Shenzhen People's Hospital), Shenzhen, China
| | - Yunqing Hua
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Jing Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Jia Shi
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Dan Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yanfei Cheng
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yunsha Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yuanli Chen
- Anhui Provincial International Science and Technology Cooperation Base for Major Metabolic Diseases and Nutritional Interventions, Key Laboratory of Major Metabolic Diseases and Nutritional Regulation of Anhui Department of Education, School of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Guanwei Fan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Chuanrui Ma
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| |
Collapse
|
40
|
Chen Y, Shen YQ. Role of reactive oxygen species in regulating epigenetic modifications. Cell Signal 2024; 125:111502. [PMID: 39521028 DOI: 10.1016/j.cellsig.2024.111502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 10/24/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024]
Abstract
Reactive oxygen species (ROS) originate from diverse sources and regulate multiple signaling pathways within the cellular environment. Their generation is intricately controlled, and disruptions in their signaling or atypical levels can precipitate pathological conditions. Epigenetics, the examination of heritable alterations in gene expression independent of changes in the genetic code, has been implicated in the pathogenesis of various diseases through aberrant epigenetic modifications. The significant contribution of epigenetic modifications to disease progression underscores their potential as crucial therapeutic targets for a wide array of medical conditions. This study begins by providing an overview of ROS and epigenetics, followed by a discussion on the mechanisms of epigenetic modifications such as DNA methylation, histone modification, and RNA modification-mediated regulation. Subsequently, a detailed examination of the interaction between ROS and epigenetic modifications is presented, offering new perspectives and avenues for exploring the mechanisms underlying specific epigenetic diseases and the development of novel therapeutics.
Collapse
Affiliation(s)
- Yutong Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Ying-Qiang Shen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, PR China.
| |
Collapse
|
41
|
Wang J, Lin Y, Liu N, Hu M, Zhang M. Differential expression of ferroptosis-related proteins in urinary exosomes: potential indicators for monitoring acute gout attack. Front Mol Biosci 2024; 11:1476631. [PMID: 39606034 PMCID: PMC11599738 DOI: 10.3389/fmolb.2024.1476631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 10/21/2024] [Indexed: 11/29/2024] Open
Abstract
Background Gout is the most prevalent form of inflammatory arthritis, characterized by significant pain during acute episodes. Current diagnostic and monitoring techniques are invasive and fail to predict the onset of acute attacks. Recent studies have implicated ferroptosis-related proteins in the pathogenesis of inflammation and gout; however, their clinical relevance in gout patients remains largely unexplored. This study aimed to evaluate the expression of these proteins in urinary exosomes from gout patients and to investigate their potential as noninvasive biomarkers. Methods Utilizing data-independent acquisition (DIA) mass spectrometry and advanced bioinformatics techniques, we assessed the expression of ferroptosis-related proteins in the urinary exosomes of three groups: acute gout patients (AD group), intermittent gout patients (ID group), and normal controls (NC group). We constructed receiver operating characteristic (ROC) curves to determine the clinical utility of these proteins in monitoring acute gout attacks. Results Our analysis of urinary exosome proteomics identified 13 ferroptosis-related proteins. Notably, in comparison to the ID group, the proteins ACSL4, VDAC2, GPX4, and GSS were significantly upregulated in the AD group. ROC curve analysis revealed that the presence of ACSL4, VDAC2, and GPX4 in urinary exosomes possesses substantial predictive value for acute gout attacks. Conclusion In patients with gout, numerous protein alterations occur within urinary exosomes. Specifically, changes in ferroptosis-related proteins such as ACSL4, VDAC2, GPX4, and GSS may serve as promising biomarkers for the monitoring of acute gout attacks.
Collapse
Affiliation(s)
- Jitu Wang
- Clinical Laboratory Medicine, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Urinary Cellular Molecular Diagnostics, Beijing, China
| | - Yubin Lin
- Clinical Laboratory Medicine, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Urinary Cellular Molecular Diagnostics, Beijing, China
| | - Na Liu
- Clinical Laboratory Medicine, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Urinary Cellular Molecular Diagnostics, Beijing, China
| | - Mei Hu
- Clinical Laboratory Medicine, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Urinary Cellular Molecular Diagnostics, Beijing, China
| | - Man Zhang
- Clinical Laboratory Medicine, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Urinary Cellular Molecular Diagnostics, Beijing, China
- Institute of Regenerative Medicine and Laboratory Technology Innovation, Qingdao University, Qingdao, China
| |
Collapse
|
42
|
Wu L, Lin H, Li S, Huang Y, Sun Y, Shu S, Luo T, Liang T, Lai W, Rao J, Hu Z, Peng H. Macrophage iron dyshomeostasis promotes aging-related renal fibrosis. Aging Cell 2024; 23:e14275. [PMID: 39016438 PMCID: PMC11561705 DOI: 10.1111/acel.14275] [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: 12/22/2023] [Revised: 05/31/2024] [Accepted: 07/01/2024] [Indexed: 07/18/2024] Open
Abstract
Renal aging, marked by the accumulation of senescent cells and chronic low-grade inflammation, leads to renal interstitial fibrosis and impaired function. In this study, we investigate the role of macrophages, a key regulator of inflammation, in renal aging by analyzing kidney single-cell RNA sequencing data of C57BL/6J mice from 8 weeks to 24 months. Our findings elucidate the dynamic changes in the proportion of kidney cell types during renal aging and reveal that increased macrophage infiltration contributes to chronic low-grade inflammation, with these macrophages exhibiting senescence and activation of ferroptosis signaling. CellChat analysis indicates enhanced communications between macrophages and tubular cells during aging. Suppressing ferroptosis alleviates macrophage-mediated tubular partial epithelial-mesenchymal transition in vitro, thereby mitigating the expression of fibrosis-related genes. Using SCENIC analysis, we infer Stat1 as a key age-related transcription factor promoting iron dyshomeostasis and ferroptosis in macrophages by regulating the expression of Pcbp1, an iron chaperone protein that inhibits ferroptosis. Furthermore, through virtual screening and molecular docking from a library of anti-aging compounds, we construct a docking model targeting Pcbp1, which indicates that the natural small molecule compound Rutin can suppress macrophage senescence and ferroptosis by preserving Pcbp1. In summary, our study underscores the crucial role of macrophage iron dyshomeostasis and ferroptosis in renal aging. Our results also suggest Pcbp1 as an intervention target in aging-related renal fibrosis and highlight Rutin as a potential therapeutic agent in mitigating age-related renal chronic low-grade inflammation and fibrosis.
Collapse
Affiliation(s)
- Lingzhi Wu
- Nephrology Division, Department of Medicine, the Third Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Hongchun Lin
- Nephrology Division, Department of Medicine, the Third Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Shaomin Li
- Nephrology Division, Department of Medicine, the Third Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Yuebo Huang
- Nephrology Division, Department of Medicine, the Third Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Yuxiang Sun
- Nephrology Division, Department of Medicine, the Third Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Shuangshuang Shu
- Nephrology Division, Department of Medicine, the Third Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Ting Luo
- Nephrology Division, Department of Medicine, the Third Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Tiantian Liang
- Nephrology Division, Department of Medicine, the Third Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Weiyan Lai
- Nephrology Division, Department of Medicine, the Third Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Jialing Rao
- Nephrology Division, Department of Medicine, the Third Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Zhaoyong Hu
- Nephrology Division, Department of MedicineBaylor College of MedicineHoustonTXUSA
| | - Hui Peng
- Nephrology Division, Department of Medicine, the Third Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhouChina
| |
Collapse
|
43
|
Fei J, Liu L, Li JF, Zhou Q, Wei Y, Zhou TD, Fu L. Associations of Vitamin D With GPX4 and Iron Parameters in Chronic Obstructive Pulmonary Disease Patients: A Case-Control Study. Can Respir J 2024; 2024:4505905. [PMID: 39502871 PMCID: PMC11535414 DOI: 10.1155/2024/4505905] [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: 10/26/2023] [Revised: 09/10/2024] [Accepted: 10/09/2024] [Indexed: 11/08/2024] Open
Abstract
Background: Vitamin D deficiency elevates the risk of chronic obstructive pulmonary disease (COPD) patients. Iron parameters elevation and glutathione peroxidase 4 (GPX4) reduction are involved in the process of COPD. The goal is to explore the associations of vitamin D with GPX4 and iron parameters in COPD patients through a case-control study. Methods: COPD patients and control subjects were enrolled. Serum samples and lung tissues were collected. Serum vitamin D and iron levels and pulmonary ferritin and GPX4 expressions were determined. In addition, human pulmonary epithelial cells (BEAS-2B) were incubated with 1,25(OH)2D3 (100 nM), the active form of vitamin D3. Then, vitamin D receptor (VDR) and nuclear factor (erythroid-derived 2)-like 2 (Nrf-2) signaling were detected. Results: In patients with COPD, serum 25-hydroxyvitamin D (25(OH)D) decreased, and iron and ferritin levels in serum and lung tissues increased. Furthermore, pulmonary expression of GPX4 was reduced. Correlative analyzes indicated that lung function was inversely correlated with iron parameters and positively correlated with GPX4. The results showed that serum 25(OH)D deficiency was associated with an elevation in serum iron parameters and a reduction in pulmonary GPX4. In addition, VDR- and Nrf-2-positive lung nuclei were decreased in COPD patients than in control subjects. In patients with COPD, the results indicated a positive relationship between VDR and Nrf-2. Further analysis revealed that Nrf-2-positive nuclei were negatively correlated with iron parameters. In vitro experiments found that 1,25(OH)2D3 treatment activated VDR signaling and elevated the expression of Nrf-2 and GPX4 in BEAS-2B cells. Conclusions: Vitamin D deficiency is positively associated with GPX4 reduction and iron parameters elevation in COPD patients. It is recommended to explore the role of vitamin D supplementation in the progression of COPD.
Collapse
Affiliation(s)
- Jun Fei
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Anhui Medical University, Hefei 230601, Anhui, China
- Institute of Respiratory Diseases, Second Affiliated Hospital of Anhui Medical University, Hefei 230601, Anhui, China
- Anhui Province Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Bengbu Medical College, Bengbu 233004, Anhui, China
| | - Ling Liu
- Department of Respiratory and Critical Care Medicine, People's Hospital of Yingshan, Fuyang 236000, Anhui, China
| | - Jia-Fei Li
- Department of Respiratory and Critical Care Medicine, First People's Hospital of Chuzhou, Chuzhou 239001, Anhui, China
| | - Qiang Zhou
- Department of Clinical Laboratory, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China
| | - Yu Wei
- Department of Clinical Laboratory, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China
| | - Ting-Dong Zhou
- Department of Clinical Laboratory, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China
| | - Lin Fu
- Anhui Province Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Bengbu Medical College, Bengbu 233004, Anhui, China
- Center for Big Data and Population Health of IHM, Anhui Medical University, Hefei, Anhui 230032, China
| |
Collapse
|
44
|
Dong W, Xu H, Wei W, Ning R, Chang Y. Advances in the study of ferroptosis and its relationship to autoimmune diseases. Int Immunopharmacol 2024; 140:112819. [PMID: 39096870 DOI: 10.1016/j.intimp.2024.112819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/10/2024] [Accepted: 07/25/2024] [Indexed: 08/05/2024]
Abstract
Ferroptosis represents a novel mode of programmed cell death characterized by the intracellular accumulation of iron and lipid peroxidation, culminating in oxidative stress and subsequent cell demise. Mounting evidence demonstrates that ferroptosis contributes significantly to the onset and progression of diverse pathological conditions and diseases, including infections, neurodegenerative disorders, tissue ischemia-reperfusion injury, and immune dysregulation. Recent investigations have underscored the pivotal role of ferroptosis in the pathogenesis of rheumatoid arthritis, ulcerative colitis, systemic lupus erythematosus, and asthma. This review provides a comprehensive overview of the current understanding of the regulatory mechanisms governing ferroptosis, particularly its interplay with iron, lipid, and amino acid metabolism. Furthermore, we explore the implications of ferroptosis in autoimmune diseases and deliberate on its potential as a promising therapeutic target for diverse autoimmune disorders.
Collapse
Affiliation(s)
- Weibo Dong
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Hepeng Xu
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Rende Ning
- The Third Affiliated Hospital of Anhui Medical University (The First People's Hospital of Hefei), 390 Huaihe Road, Hefei 230061, Anhui, China.
| | - Yan Chang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China; Laboratory Animal Center, Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
45
|
Wang T, Zheng Y, Zhang J, Wu Z. Targeting ferroptosis promotes diabetic wound healing via Nrf2 activation. Heliyon 2024; 10:e37477. [PMID: 39421383 PMCID: PMC11483302 DOI: 10.1016/j.heliyon.2024.e37477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 09/04/2024] [Accepted: 09/04/2024] [Indexed: 10/19/2024] Open
Abstract
Wound healing impairment is a frequent diabetes problem leading to amputation. Hyperglycemia induces the overproduction of reactive oxygen species (ROS), iron overload and sustained inflammation, resulting in the persistence of chronic wounds. However, the intrinsic mechanisms of impaired diabetic wound healing remain enigmatic. A new non-apoptotic regulatory cellular death called Ferroptosis, is distinguished by iron-driven lipid peroxidation products accumulation along with insufficient antioxidant enzymes. A decline in antioxidant capacity, excess accumulation of peroxidation of iron and lipid have been identified in wound sites of streptozotocin-induced diabetes mellitus (DM) rats and elevated glucose (EG)-cultured macrophages. Additionally, sustained inflammation and increased inflammatory cytokines were observed in DM rats and HG-cultured macrophages. Importantly, ferrostatin-1 (Fer-1) is a ferroptosis suppressor treatment significantly ameliorated diabetes-related ferroptosis and inflammation. This treatment also enhanced cell proliferation and neovascularization, ultimately thereby accelerating diabetic wound healing. Meanwhile, our study demonstrated that an anti-ferroptotic and anti-inflammatory effects of Fer-1 were mediated through stimulation of nuclear erythroid-associated factor 2 (Nrf2). The current study may provide a new rationale for diabetic wound healing.
Collapse
Affiliation(s)
- Tongcai Wang
- NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
- Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, Tianjin, 300134, China
- Department of Geriatric Medical Center, Inner Mongolia people's Hospital, 20 Zhaowuda Road, Hohhot, 010021, Inner Mongolia, China
| | - Yin Zheng
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
- Shandong Institute of Endocrine and Metabolic Diseases, Jinan, 250021, China
| | - Jun Zhang
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
- Shandong Institute of Endocrine and Metabolic Diseases, Jinan, 250021, China
| | - Zhongming Wu
- NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
- Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, Tianjin, 300134, China
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
- Shandong Institute of Endocrine and Metabolic Diseases, Jinan, 250021, China
| |
Collapse
|
46
|
Yin J, Xu X, Guo Y, Sun C, Yang Y, Liu H, Yu P, Wu T, Song X. Repair and regeneration: ferroptosis in the process of remodeling and fibrosis in impaired organs. Cell Death Discov 2024; 10:424. [PMID: 39358326 PMCID: PMC11447141 DOI: 10.1038/s41420-024-02181-2] [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: 04/02/2024] [Revised: 09/01/2024] [Accepted: 09/13/2024] [Indexed: 10/04/2024] Open
Abstract
As common clinical-pathological processes, wound healing and tissue remodelling following injury or stimulation are essential topics in medical research. Promoting the effective healing of prolonged wounds, improving tissue repair and regeneration, and preventing fibrosis are important and challenging issues in clinical practice. Ferroptosis, which is characterized by iron overload and lipid peroxidation, is a nontraditional form of regulated cell death. Emerging evidence indicates that dysregulated metabolic pathways and impaired iron homeostasis play important roles in various healing and regeneration processes via ferroptosis. Thus, we review the intrinsic mechanisms of tissue repair and remodeling via ferroptosis in different organs and systems under various conditions, including the inflammatory response in skin wounds, remodeling of joints and cartilage, and fibrosis in multiple organs. Additionally, we summarize the common underlying mechanisms, key molecules, and targeted drugs for ferroptosis in repair and regeneration. Finally, we discuss the potential of therapeutic agents, small molecules, and novel materials emerging for targeting ferroptosis to promote wound healing and tissue repair and attenuate fibrosis.
Collapse
Affiliation(s)
- Jiali Yin
- Department of Otolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China
| | - Xinjun Xu
- Department of Otolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China
| | - Ying Guo
- Department of Otolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China
| | - Caiyu Sun
- Department of Otolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China
| | - Yujuan Yang
- Department of Otolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China
| | - Huifang Liu
- Department of Otolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China
- Second Clinical Medicine College, Binzhou Medical University, Yantai, Shandong, 264003, China
| | - Pengyi Yu
- Department of Otolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China
| | - Tong Wu
- Qingdao Medical College, Qingdao University, Qingdao, 266071, China.
- Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266000, China.
| | - Xicheng Song
- Department of Otolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China.
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai, Shandong, China.
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China.
| |
Collapse
|
47
|
Lan W, Yang L, Tan X. Crosstalk between ferroptosis and macrophages: potential value for targeted treatment in diseases. Mol Cell Biochem 2024; 479:2523-2543. [PMID: 37880443 DOI: 10.1007/s11010-023-04871-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 10/05/2023] [Indexed: 10/27/2023]
Abstract
Ferroptosis is a newly identified form of programmed cell death that is connected to iron-dependent lipid peroxidization. It involves a variety of physiological processes involving iron metabolism, lipid metabolism, oxidative stress, and biosynthesis of nicotinamide adenine dinucleotide phosphate, glutathione, and coenzyme Q10. So far, it has been discovered to contribute to the pathological process of many diseases, such as myocardial infarction, acute kidney injury, atherosclerosis, and so on. Macrophages are innate immune system cells that regulate metabolism, phagocytize pathogens and dead cells, mediate inflammatory reactions, promote tissue repair, etc. Emerging evidence shows strong associations between macrophages and ferroptosis, which can provide us with a deeper comprehension of the pathological process of diseases and new targets for the treatments. In this review, we summarized the crosstalk between macrophages and ferroptosis and anatomized the application of this association in disease treatments, both non-neoplastic and neoplastic diseases. In addition, we have also addressed problems that remain to be investigated, in the hope of inspiring novel therapeutic strategies for diseases.
Collapse
Affiliation(s)
- Wanxin Lan
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Operative Dentistry and Endodontics West China Hospital of Stomatology, Sichuan University, 14# 3rd Section, Renmin South Road, Chengdu, 610041, Sichuan, China
| | - Lei Yang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Operative Dentistry and Endodontics West China Hospital of Stomatology, Sichuan University, 14# 3rd Section, Renmin South Road, Chengdu, 610041, Sichuan, China
| | - Xuelian Tan
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Operative Dentistry and Endodontics West China Hospital of Stomatology, Sichuan University, 14# 3rd Section, Renmin South Road, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
48
|
Wei R, Wei P, Yuan H, Yi X, Aschner M, Jiang YM, Li SJ. Inflammation in Metal-Induced Neurological Disorders and Neurodegenerative Diseases. Biol Trace Elem Res 2024; 202:4459-4481. [PMID: 38206494 DOI: 10.1007/s12011-023-04041-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 12/23/2023] [Indexed: 01/12/2024]
Abstract
Essential metals play critical roles in maintaining human health as they participate in various physiological activities. Nonetheless, both excessive accumulation and deficiency of these metals may result in neurotoxicity secondary to neuroinflammation and the activation of microglia and astrocytes. Activation of these cells can promote the release of pro-inflammatory cytokines. It is well known that neuroinflammation plays a critical role in metal-induced neurotoxicity as well as the development of neurological disorders, such as Alzheimer's disease (AD), Parkinson's disease (PD), and multiple sclerosis (MS). Initially seen as a defense mechanism, persistent inflammatory responses are now considered harmful. Astrocytes and microglia are key regulators of neuroinflammation in the central nervous system, and their excessive activation may induce sustained neuroinflammation. Therefore, in this review, we aim to emphasize the important role and molecular mechanisms underlying metal-induced neurotoxicity. Our objective is to raise the awareness on metal-induced neuroinflammation in neurological disorders. However, it is not only just neuroinflammation that different metals could induce; they can also cause harm to the nervous system through oxidative stress, apoptosis, and autophagy, to name a few. The primary pathophysiological mechanism by which these metals induce neurological disorders remains to be determined. In addition, given the various pathways through which individuals are exposed to metals, it is necessary to also consider the effects of co-exposure to multiple metals on neurological disorders.
Collapse
Affiliation(s)
- Ruokun Wei
- Toxicology Department, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, Guangxi, China
| | - Peiqi Wei
- Toxicology Department, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, Guangxi, China
| | - Haiyan Yuan
- Toxicology Department, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, Guangxi, China
| | - Xiang Yi
- Toxicology Department, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, Guangxi, China
| | - Michael Aschner
- The Department of Molecular Pharmacology at Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Yue-Ming Jiang
- Toxicology Department, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, China.
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, Guangxi, China.
| | - Shao-Jun Li
- Toxicology Department, School of Public Health, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, China.
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, 22 Shuang-yong Rd., Nanning, 530021, Guangxi, China.
| |
Collapse
|
49
|
Zhang Y, Hu K, Shang Z, Yang X, Cao L. Ferroptosis: Regulatory mechanisms and potential targets for bone metabolism: A review. Medicine (Baltimore) 2024; 103:e39158. [PMID: 39331895 PMCID: PMC11441915 DOI: 10.1097/md.0000000000039158] [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: 11/15/2023] [Accepted: 07/10/2024] [Indexed: 09/29/2024] Open
Abstract
Bone homeostasis is a homeostasis process constructed by osteoblast bone formation and osteoclast bone resorption. Bone homeostasis imbalance and dysfunction are the basis for the development of various orthopedic diseases such as osteoporosis, osteoarthritis, and steroid-induced avascular necrosis of femoral head. Previous studies have demonstrated that ferroptosis can induce lipid peroxidation through the generation of reactive oxygen species, activate a number of signaling pathways, and participate in the regulation of osteoblast bone formation and osteoclast bone resorption, resulting in bone homeostasis imbalance, which is an important factor in the pathogenesis of many orthopedic diseases, but the mechanism of ferroptosis is still unknown. In recent years, it has been found that, in addition to iron metabolism and intracellular antioxidant system imbalance, organelle dysfunction is also a key factor affecting ferroptosis. This paper takes this as the starting point, reviews the latest literature reports at home and abroad, elaborates the pathogenesis and regulatory pathways of ferroptosis and the relationship between ferroptosis and various organelles, and summarizes the mechanism by which ferroptosis mediates bone homeostasis imbalance, with the aim of providing new directions for the research related to ferroptosis and new ideas for the prevention and treatment of bone and joint diseases.
Collapse
Affiliation(s)
- Yongjie Zhang
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Kangyi Hu
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Zhengya Shang
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Xiaorui Yang
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Linzhong Cao
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
- The Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, China
| |
Collapse
|
50
|
Huangfu S, Zheng J, He J, Liao J, Jiang H, Zhou H, Pan J. Protective role of seleno-amino acid against IBD via ferroptosis inhibition in enteral nutrition therapy. iScience 2024; 27:110494. [PMID: 39290833 PMCID: PMC11407031 DOI: 10.1016/j.isci.2024.110494] [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: 02/20/2024] [Revised: 04/23/2024] [Accepted: 07/09/2024] [Indexed: 09/19/2024] Open
Abstract
The interplay between intestinal barrier degradation and trace element insufficiency worsens inflammatory bowel disease (IBD). Selenium (se) is essential for glutathione peroxidase 4 (GPX4) synthesis, which protects against intestinal epithelial cell injury in IBD. However, malnutrition and malabsorption limit the availability of dietary selenium. This study investigated the protective effects of naturally occurring seleno-amino acids on the intestinal barrier in an IBD animal model by promoting GPX4 synthesis. L-se-methylselenocystine (seMc) supplementation reversed decreased GPX4 expression levels, alleviated glutathione depletion and scavenged reactive oxygen species in vitro. In vivo, enteral nutrition combined with seMc protected the intestinal barrier and alleviated IBD-related symptoms by inhibiting ferroptosis and reversing lipid peroxidation in epithelial cells while reducing immune cell infiltration. Our findings suggest that seleno-amino acid-based nutritional formulations may provide a basis for nutritional support to alleviate complex cycles between intestinal barrier damage and malnutrition in IBD patients.
Collapse
Affiliation(s)
- Shuchen Huangfu
- Department of General Surgery, the First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| | - Jie Zheng
- Department of Food Science and Engineering, Jinan University, Guangzhou 510632, China
| | - Jiashuai He
- Department of General Surgery, the First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| | - Jin Liao
- Department of Food Science and Engineering, Jinan University, Guangzhou 510632, China
| | - Haiping Jiang
- Department of General Surgery, the First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| | - Hua Zhou
- Department of Food Science and Engineering, Jinan University, Guangzhou 510632, China
| | - Jinghua Pan
- Department of General Surgery, the First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| |
Collapse
|