1
|
Yang Y, Shen W, Zhang Z, Dai Y, Zhang Z, Liu T, Yu J, Huang S, Ding Y, You R, Wang Z, Wu Y, Bian T. FSP1 Acts in Parallel with GPX4 to Inhibit Ferroptosis in Chronic Obstructive Pulmonary Disease. Am J Respir Cell Mol Biol 2025; 72:551-562. [PMID: 39453438 DOI: 10.1165/rcmb.2023-0467oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 10/25/2024] [Indexed: 10/26/2024] Open
Abstract
GPX4 (glutathione peroxidase 4) has recently been reported to play an important role in the pathogenesis of chronic obstructive pulmonary disease (COPD). FSP1 (ferroptosis suppressor protein-1) is a protein that defends against ferroptosis in parallel with GPX4, but its role in the pathogenesis of COPD remains unexplored, and further research is needed. Normal and COPD lung tissues were obtained from lobectomy and lung transplant specimens, respectively. FSP1-overexpressing mice were established by monthly transfection with adenoassociated virus 9-FSP1 through modified intranasal administration. The expression of FSP1, GPX4, and PTGS2 (prostaglandin-endoperoxide synthase 2) was measured by Western blotting, immunohistochemistry and other methods. The correlation between FSP1 and ferroptosis and the role of FSP1 in COPD were explored by screening the expression of ferroptosis-related genes in a COPD cell model after the inhibition and overexpression of FSP1. We then explored the underlying mechanism of low FSP1 expression in patients with COPD in vitro by methylated RNA immunoprecipitation quantitative qPCR. We found that cigarette smoke exposure can lead to an increase in lipid peroxide production and ultimately ferroptosis, which is negatively regulated by FSP1 activity. FSP1 overexpression can prevent ferroptosis and alleviate emphysema. Next, we found that decreased FSP1 expression was caused by increased N6-methyladenosine modification of FSP1 mRNA. Moreover, the level of FSP1 decreased in a YTHDF2-dependent manner. These results indicate that METTL3-induced FSP1 mRNA methylation leading to low FSP1 expression is a potential therapeutic target for COPD.
Collapse
Affiliation(s)
- Yue Yang
- Department of Respiratory Medicine, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, People's Republic of China
| | - Weiyu Shen
- Department of Respiratory Medicine, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, People's Republic of China
| | - Zheming Zhang
- Department of Respiratory Medicine, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, People's Republic of China
| | - Youai Dai
- Department of Respiratory Medicine, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, People's Republic of China
| | - Zixiao Zhang
- Department of Respiratory Medicine, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, People's Republic of China
| | - Tingting Liu
- Department of Respiratory Medicine, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, People's Republic of China
| | - Jinyan Yu
- Department of Respiratory Medicine, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, People's Republic of China
| | - Shulun Huang
- Department of Respiratory Medicine, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, People's Republic of China
| | - Yu Ding
- Department of Respiratory Medicine, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, People's Republic of China
| | - Rong You
- Department of Respiratory Medicine, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, People's Republic of China
| | - Ziteng Wang
- Department of Respiratory Medicine, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, People's Republic of China
| | - Yan Wu
- Department of Respiratory Medicine, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, People's Republic of China
| | - Tao Bian
- Department of Respiratory Medicine, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, People's Republic of China
| |
Collapse
|
2
|
Yang X, Liu Y, Cao J, Wu C, Tang L, Bian W, Chen Y, Yu L, Wu Y, Li S, Shen Y, Xia J, Du J. Targeting epigenetic and post-translational modifications of NRF2: key regulatory factors in disease treatment. Cell Death Discov 2025; 11:189. [PMID: 40258841 PMCID: PMC12012105 DOI: 10.1038/s41420-025-02491-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 04/07/2025] [Accepted: 04/10/2025] [Indexed: 04/23/2025] Open
Abstract
Nuclear factor erythroid 2-related factor 2 (NRF2) is a key transcription factor involved in regulating cellular antioxidant defense and detoxification mechanisms. It mitigates oxidative stress and xenobiotic-induced damage by inducing the expression of cytoprotective enzymes, including HO-1 and NQO1. NRF2 also modulates inflammatory responses by inhibiting pro-inflammatory genes and mediates cell death pathways, including apoptosis and ferroptosis. Targeting NRF2 offers potential therapeutic avenues for treating various diseases. NRF2 is regulated through two principal mechanisms: post-translational modifications (PTMs) and epigenetic alterations. PTMs, including phosphorylation, ubiquitination, and acetylation, play a pivotal role in modulating NRF2's stability, activity, and subcellular localization, thereby precisely controlling its function in the antioxidant response. For instance, ubiquitination can lead to NRF2 degradation and reduced antioxidant activity, while deubiquitination enhances its stability and function. Epigenetic modifications, such as DNA methylation, histone modifications, and interactions with non-coding RNAs (e.g., MALAT1, PVT1, MIR4435-2HG, and TUG1), are essential for regulating NRF2 expression by modulating chromatin architecture and gene accessibility. This paper systematically summarizes the molecular mechanisms by which PTMs and epigenetic alterations regulate NRF2, and elucidates its critical role in cellular defense and disease. By analyzing the impact of PTMs, such as phosphorylation, ubiquitination, and acetylation, as well as DNA methylation, histone modifications, and non-coding RNA interactions on NRF2 stability, activity, and expression, the study reveals the complex cellular protection network mediated by NRF2. Furthermore, the paper explores how these regulatory mechanisms affect NRF2's roles in oxidative stress, inflammation, and cell death, identifying novel therapeutic targets and strategies. This provides new insights into the treatment of NRF2-related diseases, such as cancer, neurodegenerative disorders, and metabolic syndrome. This research deepens our understanding of NRF2's role in cellular homeostasis and lays the foundation for the development of NRF2-targeted therapies.
Collapse
Affiliation(s)
- Xinyi Yang
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Yingchao Liu
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Jinghao Cao
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Cuiyun Wu
- Cancer Center, Department of Radiology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Lusheng Tang
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Wenxia Bian
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Yuhan Chen
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Lingyan Yu
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Yunyi Wu
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Sainan Li
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Yuhuan Shen
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China.
| | - Jun Xia
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China.
| | - Jing Du
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China.
| |
Collapse
|
3
|
Yang Y, Deng X, Li W, Leng Y, Xiong Y, Wang B, Gong S, Wang Y, Yang B, Li W. Targeting the epigenetic regulation of ferroptosis: a potential therapeutic approach for sepsis-associated acute kidney injury. Clin Epigenetics 2025; 17:57. [PMID: 40189571 PMCID: PMC11974148 DOI: 10.1186/s13148-025-01861-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 03/13/2025] [Indexed: 04/09/2025] Open
Abstract
Sepsis is a syndrome of organ dysfunction caused by the invasion of pathogenic microorganisms. In clinical practice, patients with sepsis are prone to concurrent acute kidney injury, which has high morbidity and mortality rates. Thus, understanding the pathogenesis of sepsis-associated acute kidney injury is of significant clinical importance. Ferroptosis is an iron-dependent programmed cell death pathway, which is proved to play a critical role in the process of sepsis-associated acute kidney injury through various mechanisms. Epigenetic regulation modulates the content and function of nucleic acids and proteins within cells through various modifications. Its impact on ferroptosis has garnered increasing attention; however, the role of epigenetic regulation targeting ferroptosis in sepsis-associated acute kidney injury has not been fully elucidated. Growing evidence suggests that epigenetic regulation can modulate ferroptosis through complex pathway networks, thereby affecting the development and prognosis of sepsis-associated acute kidney injury. This paper summarizes the impact of ferroptosis on sepsis-associated acute kidney injury and the regulatory mechanisms of epigenetic regulation on ferroptosis, providing new insights for the targeted therapy of sepsis-associated acute kidney injury.
Collapse
Affiliation(s)
- Yuhang Yang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Xinqi Deng
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Wenyuan Li
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Yan Leng
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Yonghong Xiong
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Bihan Wang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Siyuan Gong
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Yunhao Wang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Baichuan Yang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Wei Li
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
| |
Collapse
|
4
|
Ragusa R, Bufano P, Tognetti A, Laurino M, Caselli C. Recent Evidences of Epigenetic Alterations in Chronic Obstructive Pulmonary Disease (COPD): A Systematic Review. Int J Mol Sci 2025; 26:2571. [PMID: 40141213 PMCID: PMC11942187 DOI: 10.3390/ijms26062571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 03/04/2025] [Accepted: 03/09/2025] [Indexed: 03/28/2025] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a heterogeneous inflammatory condition characterized by progressive airflow limitation, which may be caused by genetic and environmental factors. Furthermore, epigenetic mechanisms could provide valuable insights into the complex interactions between environment and genes and subsequent development of the disease. The aim of this study is to provide a systematic review of the latest knowledge on epigenetic modifications that characterize COPD, summarizing epigenetic factors that could serve as potential novel biomarkers and therapeutic targets for the treatment of COPD patients. We queried the PubMed and Scopus electronic databases with specific keywords, in May 2024, according to the PRISMA guidelines, and articles were included if they met all the inclusion criteria and survived a quality assessment. We identified 5414 publications in our systematic search. Among them, only 51 articles met the criteria of COPD-associated epigenetic modifications in human patients compared to the control group. Eight studies described DNA methylation, one study histone modifications, and forty-two studies non-coding RNAs. Apoptosis and inflammatory pathways have been found to be the main mechanisms regulated by epigenetic elements in COPD patients. In addition, non-coding RNAs may be useful as biomarkers or therapeutic targets of pulmonary disease. Future studies will be needed to confirm the role of epigenetic elements associated with COPD.
Collapse
Affiliation(s)
- Rosetta Ragusa
- Institute of Clinical Physiology, CNR, 56124 Pisa, Italy; (P.B.); (M.L.)
| | - Pasquale Bufano
- Institute of Clinical Physiology, CNR, 56124 Pisa, Italy; (P.B.); (M.L.)
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, 56124 Pisa, Italy
| | | | - Marco Laurino
- Institute of Clinical Physiology, CNR, 56124 Pisa, Italy; (P.B.); (M.L.)
| | - Chiara Caselli
- Institute of Clinical Physiology, CNR, 56124 Pisa, Italy; (P.B.); (M.L.)
- Fondazione Toscana Gabriele Monasterio, 56124 Pisa, Italy
| |
Collapse
|
5
|
Khawas S, Sharma N. Cell death crosstalk in respiratory diseases: unveiling the relationship between pyroptosis and ferroptosis in asthma and COPD. Mol Cell Biochem 2025; 480:1305-1326. [PMID: 39112808 DOI: 10.1007/s11010-024-05062-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 06/29/2024] [Indexed: 02/21/2025]
Abstract
Asthma and chronic obstructive pulmonary disease (COPD) are heterogeneous obstructive diseases characterized by airflow limitations and are recognized as significant contributors to fatality all over the globe. Asthma accounts for about 4, 55,000 deaths, and COPD is the 3rd leading contributor of mortality worldwide. The pathogenesis of these two obstructive disorders is complex and involves numerous mechanistic pathways, including inflammation-mediated and non-inflammation-mediated pathways. Among all the pathological categorizations, programmed cell deaths (PCDs) play a dominating role in the progression of these obstructive diseases. The two major PCDs that are involved in structural and functional remodeling in the progression of asthma and COPD are Pyroptosis and Ferroptosis. Pyroptosis is a PCD mechanism mediated by the activation of the Nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3 (NLRP3) inflammasome, leading to the maturation and release of Interleukin-1β and Interleukin-18, whereas ferroptosis is a lipid peroxidation-associated cell death. In this review, the major molecular pathways contributing to these multifaceted cell deaths have been discussed, and crosstalk among them regarding the pathogenesis of asthma and COPD has been highlighted. Further, the possible therapeutic approaches that can be utilized to mitigate both cell deaths at once have also been illustrated.
Collapse
Affiliation(s)
- Sayak Khawas
- Department of Pharmaceutical Science & Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, India
| | - Neelima Sharma
- Department of Pharmaceutical Science & Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, India.
| |
Collapse
|
6
|
Faruqui N, Orell S, Dondi C, Leni Z, Kalbermatter DM, Gefors L, Rissler J, Vasilatou K, Mudway IS, Kåredal M, Shaw M, Larsson-Callerfelt AK. Differential Cytotoxicity and Inflammatory Responses to Particulate Matter Components in Airway Structural Cells. Int J Mol Sci 2025; 26:830. [PMID: 39859544 PMCID: PMC11765832 DOI: 10.3390/ijms26020830] [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: 12/09/2024] [Revised: 01/09/2025] [Accepted: 01/12/2025] [Indexed: 01/27/2025] Open
Abstract
Particulate matter (PM) is a major component of ambient air pollution. PM exposure is linked to numerous adverse health effects, including chronic lung diseases. Air quality guidelines designed to regulate levels of ambient PM are currently based on the mass concentration of different particle sizes, independent of their origin and chemical composition. The objective of this study was to assess the relative hazardous effects of carbonaceous particles (soot), ammonium nitrate, ammonium sulfate, and copper oxide (CuO), which are standard components of ambient air, reflecting contributions from primary combustion, secondary inorganic constituents, and non-exhaust emissions (NEE) from vehicular traffic. Human epithelial cells representing bronchial (BEAS-2B) and alveolar locations (H441 and A549) in the airways, human lung fibroblasts (HFL-1), and rat precision-cut lung slices (PCLS) were exposed in submerged cultures to different concentrations of particles for 5-72 h. Following exposure, cell viability, metabolic activity, reactive oxygen species (ROS) formation, and inflammatory responses were analyzed. CuO and, to a lesser extent, soot reduced cell viability in a dose-dependent manner, increased ROS formation, and induced inflammatory responses. Ammonium nitrate and ammonium sulfate did not elicit any significant cytotoxic responses but induced immunomodulatory alterations at very high concentrations. Our findings demonstrate that secondary inorganic components of PM have a lower hazard cytotoxicity compared with combustion-derived and indicative NEE components, and alveolar epithelial cells are more sensitive to PM exposure. This information should help to inform which sources of PM to target and feed into improved, targeted air quality guidelines.
Collapse
Affiliation(s)
- Nilofar Faruqui
- Department of Chemical & Biological Services, National Physical Laboratory, Teddington TW11 0LW, UK
| | - Sofie Orell
- Lung Biology, Department of Experimental Medical Sciences, Lund University, 221 84 Lund, Sweden
| | - Camilla Dondi
- Department of Chemical & Biological Services, National Physical Laboratory, Teddington TW11 0LW, UK
| | - Zaira Leni
- Institute of Anatomy, University of Bern, 3012 Bern, Switzerland
| | | | - Lina Gefors
- Lund University Bioimaging Centre (LBIC), Lund University, 221 84 Lund, Sweden
| | - Jenny Rissler
- Ergonomics and Aerosol Technology, Department of Design Sciences, Faculty of Engineering (LTH), Lund University, 223 62 Lund, Sweden
| | | | - Ian S. Mudway
- MRC Centre for Environment and Health, Imperial College London, London W2 1PG, UK
- National Institute of Health Protection Research Unit in Environmental Exposures and Health, London W2 1NY, UK
| | - Monica Kåredal
- Division of Occupational and Environmental Medicine, Department of Laboratory Medicine, Lund University, 223 63 Lund, Sweden
- Department of Occupational and Environmental Medicine, Region Skåne, 223 63 Lund, Sweden
| | - Michael Shaw
- Department of Chemical & Biological Services, National Physical Laboratory, Teddington TW11 0LW, UK
- Department of Computer Science, University College London, London WC1E 6BT, UK
| | | |
Collapse
|
7
|
Xu A, Xu Y, Chen H, Xiang L, Zhao X. Ginkgo biloba extract alleviates ferroptosis in lung epithelial cells induced by cigarette smoke extract through miR-3,619-5p/GPX4 axis. Toxicol Res (Camb) 2025; 14:tfae225. [PMID: 39759181 PMCID: PMC11694667 DOI: 10.1093/toxres/tfae225] [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: 05/15/2024] [Revised: 10/31/2024] [Accepted: 12/17/2024] [Indexed: 01/07/2025] Open
Abstract
Ginkgo biloba extract (GBE), a therapeutic drug, has anti-inflammatory and antioxidant effects that protect cells from harmful substances. Although GBE has been extensively studied in the prevention and treatment of lung diseases, its mechanism of action in chronic obstructive pulmonary disease (COPD) is unclear. In the present study, cigarette smoke extract (CSE) and cigarette smoke (CS) were used to induce COPD in cell and animal models. The expression of related genes and proteins was detected, and cell damage and lung tissue damage were evaluated via CCK-8 assays, flow cytometry analyses, ELISA, and HE staining. In HBE cells, the expression of miR-3,619-5p was upregulated after CSE induction. However, GBE treatment alleviated the impact of CSE on HBE cell damage and alleviated COPD in vivo. In addition, GBE treatment increased the expression of GPX4 by inhibiting the expression of miR-3,619-5p, and it reduced the release of the IL-6, IL-8, and TNF-α inflammatory factors. Moreover, GBE treatment decreased the production of ROS and MDA, as well as decreased the expression of the ferroptosis-related protein ACSL4, and it promoted the production of GSH and the expression of FTH1. Further, GBE treatment improved cell viability, inhibited ferroptosis, and ultimately alleviated COPD. The present findings suggest that GBE alleviates the progression of COPD through the inhibitory effect of the miR-3,619-5p/GPX4 axis on the ferroptosis process and that GBE may be an effective treatment option for COPD.
Collapse
Affiliation(s)
- Anhui Xu
- Department of Clinical Laboratory, The People’s Hospital of Mengzi, No. 89 Tianma Road, Mengzi, Yunnan Province 661100, China
| | - Yanmei Xu
- Department of Clinical Laboratory, The People’s Hospital of Mengzi, No. 89 Tianma Road, Mengzi, Yunnan Province 661100, China
| | - Hongbo Chen
- Department of Respiratory and Critical Care Medicine, Anning First People's Hospital Affiliated with Kunming University of Science and Technology, No. 2, Ganghe South Road, Anning, Yunnan Province 650302, China
| | - Linhua Xiang
- Department of Clinical Laboratory, The People’s Hospital of Mengzi, No. 89 Tianma Road, Mengzi, Yunnan Province 661100, China
| | - Xiao Zhao
- Department of Respiratory and Critical Care Medicine, The People’s Hospital of Mengzi, No. 89 Tianma Road, Mengzi, Yunnan Province 661100, China
| |
Collapse
|
8
|
Wang A, Liu J, Li Z, Qian Z, Yang S, Luo S, Lin J, Wu J. CC16 alleviates PM2.5-induced lung epithelial cell injury and airway inflammation in asthmatic mice by inhibiting ferroptosis. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 289:117417. [PMID: 39644566 DOI: 10.1016/j.ecoenv.2024.117417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 11/02/2024] [Accepted: 11/25/2024] [Indexed: 12/09/2024]
Abstract
BACKGROUND Exposure to PM2.5 represents a significant public health challenge, closely associated with the worsening of asthma, a condition that still lacks effective preventive measures. Club Cell 16 kDa protein (CC16), recognized for its anti-inflammatory and antioxidant properties, may serve a protective function in asthma exacerbated by PM2.5; however, the underlying mechanisms, particularly those related to ferroptosis, remain poorly understood. METHODS The impact of CC16 on inflammation and ferroptosis was assessed using a TC-1 lung epithelial cell model exposed to PM2.5, as well as an ovalbumin (OVA)-induced asthmatic mouse model also subjected to PM2.5 exposure. RESULTS CC16 significantly modulated key regulators of ferroptosis (NRF2, GPX4, SLC7A11, HO-1) and attenuated pro-inflammatory cytokines (IL-13, IL-5, IL-6, IL-1β, IL-17A) in PM2.5-exposed lung epithelial cells. Furthermore, it enhanced pulmonary function while reducing airway inflammation and mucus secretion and inhibited ferroptosis in PM2.5-induced asthmatic mice. CONCLUSION CC16 demonstrates promise as a therapeutic agent for PM2.5-induced asthma by modulating ferroptosis and alleviating airway inflammation, thereby providing a novel strategy for asthma management.
Collapse
Affiliation(s)
- Aili Wang
- Second Department of Elderly Respiratory, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangdong Provincial Geriatric Institute, Guangzhou, Guangdong 510080, China; Department of Respiratory and Critical Care Medicine, Wuhan No.1 Hospital, Wuhan, Hubei 430022, China
| | - Jianling Liu
- Second Department of Elderly Respiratory, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangdong Provincial Geriatric Institute, Guangzhou, Guangdong 510080, China; School of Medicine, South China University of Technology, Guangzhou, Guangdong 510080, China
| | - Zhangwen Li
- Second Department of Elderly Respiratory, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangdong Provincial Geriatric Institute, Guangzhou, Guangdong 510080, China; School of Medicine, South China University of Technology, Guangzhou, Guangdong 510080, China
| | - Ze Qian
- Second Department of Elderly Respiratory, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangdong Provincial Geriatric Institute, Guangzhou, Guangdong 510080, China
| | - Shuo Yang
- Department of Respiratory and Critical Care Medicine, Wuhan No.1 Hospital, Wuhan, Hubei 430022, China
| | - Shaohua Luo
- Second Department of Elderly Respiratory, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangdong Provincial Geriatric Institute, Guangzhou, Guangdong 510080, China
| | - Jinle Lin
- Department of Emergency Medicine, Affiliated Baoan Hospital of Shenzhen, The Second School of Clinical Medicine, Southern Medical University, Shenzhen, Guangdong 518101, China
| | - Jian Wu
- Second Department of Elderly Respiratory, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangdong Provincial Geriatric Institute, Guangzhou, Guangdong 510080, China.
| |
Collapse
|
9
|
Wu T, Ji M, Li T, Luo L. The molecular and metabolic landscape of ferroptosis in respiratory diseases: Pharmacological aspects. J Pharm Anal 2025; 15:101050. [PMID: 40034685 PMCID: PMC11873008 DOI: 10.1016/j.jpha.2024.101050] [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: 02/29/2024] [Revised: 07/16/2024] [Accepted: 07/19/2024] [Indexed: 03/05/2025] Open
Abstract
Ferroptosis is a form of cell death that occurs when there is an excess of reactive oxygen species (ROS), lipid peroxidation, and iron accumulation. The precise regulation of metabolic pathways, including iron, lipid, and amino acid metabolism, is crucial for cell survival. This type of cell death, which is associated with oxidative stress, is controlled by a complex network of signaling molecules and pathways. It is also implicated in various respiratory diseases such as asthma, chronic obstructive pulmonary disease (COPD), acute lung injury (ALI), lung cancer, pulmonary fibrosis (PF), and the coronavirus disease 2019 (COVID-19). To combat drug resistance, it is important to identify appropriate biological markers and treatment targets, as well as intervene in respiratory disorders to either induce or prevent ferroptosis. The focus is on the role of ferroptosis in the development of respiratory diseases and the potential of targeting ferroptosis for prevention and treatment. The review also explores the interaction between immune cell ferroptosis and inflammatory mediators in respiratory diseases, aiming to provide more effective strategies for managing cellular ferroptosis and respiratory disorders.
Collapse
Affiliation(s)
- Tong Wu
- The First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong, 524023, China
| | - Miaorong Ji
- The First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong, 524023, China
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Lianxiang Luo
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, Guangdong, 524023, China
| |
Collapse
|
10
|
Xie Y, Xie J, Li L. The Role of Methylation in Ferroptosis. J Cardiovasc Transl Res 2024; 17:1219-1228. [PMID: 39075241 DOI: 10.1007/s12265-024-10539-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 06/21/2024] [Indexed: 07/31/2024]
Abstract
Methylation modification is a crucial epigenetic alteration encompassing RNA methylation, DNA methylation, and histone methylation. Ferroptosis represents a newly discovered form of programmed cell death (PCD) in 2012, which is characterized by iron-dependent lipid peroxidation. The comprehensive investigation of ferroptosis is therefore imperative for a more profound comprehension of the pathological and pathophysiological mechanisms implicated in a wide array of diseases. Researches show that methylation modifications can exert either promotive or inhibitory effects on cell ferroptosis. Consequently, this review offers a comprehensive overview of the pivotal role played by methylation in ferroptosis, elucidating its associated factors and underlying mechanisms.
Collapse
Affiliation(s)
- Yushu Xie
- Class of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Jie Xie
- Class of Excellent Doctor, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Liang Li
- Department of Physiology, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| |
Collapse
|
11
|
Duan X, Hu T, Xu L, Li Z, Jing J, Xu D, Ding J, Li F, Jiang M, Wang J. The Correlation Analysis Between m6A Methylation Modification and Ferroptosis Induced by Cigarette Smoke in Human Bronchial Epithelium. Immun Inflamm Dis 2024; 12:e70104. [PMID: 39688470 PMCID: PMC11650891 DOI: 10.1002/iid3.70104] [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: 10/07/2023] [Revised: 11/11/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD), a prevalent respiratory condition, is characterized by long-term airway inflammation, which can lead to airway remodeling and persistent airflow restriction. Exposure to cigarette smoke is known as a major contributor to COPD development. Research has confirmed that ferroptosis and m6A modification are closely related to various inflammatory-related diseases. However, the correlation between m6A methylation and ferroptosis in COPD has not been confirmed. In this study, combined with bioinformatics analysis and molecular biology methods we investigated how m6A methylation was correlated to ferroptosis-associated genes (SLC7A11 and NQO-1) in cigarette smoke induced 16HBES cells. METHODS Two microarray datasets (GSE30063 and GSE64614) were combined to identify differentially expressed genes (DEGs) through the application of bioinformatics techniques. A cigarette smoke (CS)-induced 16HBE cells model was established. The ROS, GSH, MDA, and total iron content were detected by relevant detection kits. The expression levels associated with ferroptosis and m6A methylation modification-related genes were determined via reverse transcription-quantitative polymerase chain reaction and western blot. RESULTS Overall, 529 DEGs were identified in the above two databases. For COPD patients, significant changes were observed in FAGs (GCLC, NQO-1, SLC7A11) and m6A methylation-related genes (FTO). A negative correlation was also noted between the expression level of genes linked to ferroptosis (SLC7A11 and NQO-1) and that of the m6A methylation gene (FTO). The in vitro experiments results indicate that SLC7A11 and NQO-1 were significantly downregulated, and FTO were significantly upregulated. In addition, cigarette smoke stimulation increased the levels of MDA, LPO, and ROS, while reducing the content of GSH and total iron content in 16HBE cells. CONCLUSION Our findings explored the relationship between ferroptosis and m6A methylation in COPD, and screened out SLC7A11, NQO-1 and FTO may be critical in the pathogenesis of COPD.
Collapse
Affiliation(s)
- Xiaomei Duan
- Department of Xinjiang Laboratory of Respiratory Disease ResearchTraditional Chinese Medicine Hospital Affiliated to Xinjiang Medical UniversityUrumqiXinjiangChina
- Department of Xinjiang Clinical Research Center for Respiratory DiseasesTraditional Chinese Medicine Hospital Affiliated to Xinjiang Medical UniversityUrumqiXinjiangChina
| | - Tingting Hu
- Department of Xinjiang Laboratory of Respiratory Disease ResearchTraditional Chinese Medicine Hospital Affiliated to Xinjiang Medical UniversityUrumqiXinjiangChina
- Department of Xinjiang Clinical Research Center for Respiratory DiseasesTraditional Chinese Medicine Hospital Affiliated to Xinjiang Medical UniversityUrumqiXinjiangChina
| | - Lijuan Xu
- Department of EndocrineTraditional Chinese Medicine Hospital Affiliated to Xinjiang Medical UniversityUrumqiXinjiangChina
| | - Zheng Li
- Department of Xinjiang Laboratory of Respiratory Disease ResearchTraditional Chinese Medicine Hospital Affiliated to Xinjiang Medical UniversityUrumqiXinjiangChina
- Department of Xinjiang Clinical Research Center for Respiratory DiseasesTraditional Chinese Medicine Hospital Affiliated to Xinjiang Medical UniversityUrumqiXinjiangChina
| | - Jing Jing
- Department of Xinjiang Laboratory of Respiratory Disease ResearchTraditional Chinese Medicine Hospital Affiliated to Xinjiang Medical UniversityUrumqiXinjiangChina
- Department of Xinjiang Clinical Research Center for Respiratory DiseasesTraditional Chinese Medicine Hospital Affiliated to Xinjiang Medical UniversityUrumqiXinjiangChina
| | - Dan Xu
- Department of Xinjiang Laboratory of Respiratory Disease ResearchTraditional Chinese Medicine Hospital Affiliated to Xinjiang Medical UniversityUrumqiXinjiangChina
- Department of Xinjiang Clinical Research Center for Respiratory DiseasesTraditional Chinese Medicine Hospital Affiliated to Xinjiang Medical UniversityUrumqiXinjiangChina
| | - Jianbing Ding
- Department of Immunology, College of Basic MedicineXinjiang Medical UniversityUrumqiXinjiangChina
| | - Fengsen Li
- Department of Xinjiang Laboratory of Respiratory Disease ResearchTraditional Chinese Medicine Hospital Affiliated to Xinjiang Medical UniversityUrumqiXinjiangChina
- Department of Xinjiang Clinical Research Center for Respiratory DiseasesTraditional Chinese Medicine Hospital Affiliated to Xinjiang Medical UniversityUrumqiXinjiangChina
| | - Min Jiang
- Department of Xinjiang Laboratory of Respiratory Disease ResearchTraditional Chinese Medicine Hospital Affiliated to Xinjiang Medical UniversityUrumqiXinjiangChina
- Department of Xinjiang Clinical Research Center for Respiratory DiseasesTraditional Chinese Medicine Hospital Affiliated to Xinjiang Medical UniversityUrumqiXinjiangChina
| | - Jing Wang
- Department of Xinjiang Laboratory of Respiratory Disease ResearchTraditional Chinese Medicine Hospital Affiliated to Xinjiang Medical UniversityUrumqiXinjiangChina
- Department of Xinjiang Clinical Research Center for Respiratory DiseasesTraditional Chinese Medicine Hospital Affiliated to Xinjiang Medical UniversityUrumqiXinjiangChina
| |
Collapse
|
12
|
Du L, Zhu X, Jiang Z, Wang W, Liu P, Zhu L, Zhang F. Resveratrol inhibits ferroptosis in the lung tissues of heat stroke-induced rats via the Nrf2 pathway. BMC Pharmacol Toxicol 2024; 25:88. [PMID: 39563478 DOI: 10.1186/s40360-024-00810-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 10/30/2024] [Indexed: 11/21/2024] Open
Abstract
BACKGROUND Heat stroke (HS) can lead to the development of pulmonary ferroptosis. The inhibition of pulmonary ferroptosis during HS improves patient prognosis. The aim of this study was to investigate the effects of resveratrol (RES) on heat stress at an ambient temperature of 42 °C. METHODS Heat stress was induced in Beas-2B cells and lung injury was induced in HS rats at an ambient temperature of 42 °C. The anti-oxidative stress and anti-ferroptotic effects of RES were confirmed through tail vein injection of nuclear factor-2 associated factor (Nrf2) shRNA recombinant adeno-associated virus 6 (AAV6-shNrf2). RESULTS RES treatment attenuated the upregulation of reactive oxygen species (ROS) and malondialdehyde (MDA) levels and alleviated glutathione inhibition in HS. In addition, RES treatment reduced the accumulation of Fe2+ in heat-stressed Beas-2B cells and increased the ferroptosis resistance-related proteins FTH1, GPX4, and SLC7A11 as well as the anti-oxidative stress pathway proteins Nrf2, NQO1, and HO-1. The antioxidant and anti-ferroptotic effects of RES in heat-stressed Beas-2B cells were effectively reversed upon treatment with Nrf2-IN-1, an Nrf2 pathway inhibitor. In the HS rat model, the antioxidant and anti-ferroptotic effects of RES were reversed by an ambient temperature of 42 °C and relative humidity of 60 ± 5%. CONCLUSIONS RES effectively protected HS rats from lung injury, inhibited the accumulation of Fe2+, ROS, and MDA in the lung, and upregulated FTH1, GPX4, SLC7A11, Nrf2, NQO1, and HO-1.
Collapse
Affiliation(s)
- Liwen Du
- Department of Emergency, Ningbo No.2 Hospital. No. 41, Northwest Street, Haishu District, Ningbo, 315010, China
| | - Xueqi Zhu
- Department of Emergency, Ningbo No.2 Hospital. No. 41, Northwest Street, Haishu District, Ningbo, 315010, China
| | - Zhenluo Jiang
- Department of Emergency, Ningbo No.2 Hospital. No. 41, Northwest Street, Haishu District, Ningbo, 315010, China
| | - Weidong Wang
- Department of Emergency, Ningbo No.2 Hospital. No. 41, Northwest Street, Haishu District, Ningbo, 315010, China
| | - Peng Liu
- Department of Emergency, Ningbo No.2 Hospital. No. 41, Northwest Street, Haishu District, Ningbo, 315010, China
| | - Leilei Zhu
- Department of Emergency, Ningbo No.2 Hospital. No. 41, Northwest Street, Haishu District, Ningbo, 315010, China.
| | - Fangqi Zhang
- Department of Pulmonary and Critical Care Medicine, The 987th Hospital of Joint Logistics Support Force of Chinese PLA, Baoji, 721000, China
| |
Collapse
|
13
|
Chen Y, Jiang Z, Li X. New insights into crosstalk between Nrf2 pathway and ferroptosis in lung disease. Cell Death Dis 2024; 15:841. [PMID: 39557840 PMCID: PMC11574213 DOI: 10.1038/s41419-024-07224-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 10/30/2024] [Accepted: 11/05/2024] [Indexed: 11/20/2024]
Abstract
Ferroptosis is a distinctive process of cellular demise that is linked to amino acid metabolism, lipid oxidation, and iron oxidation. The ferroptosis cascade genes, which are closely associated with the onset of lung diseases, are among the regulatory targets of nuclear factor erythroid 2-related factor 2 (Nrf2). Although the regulation of ferroptosis is mostly mediated by Nrf2, the precise roles and underlying regulatory mechanisms of ferroptosis and Nrf2 in lung illness remain unclear. This review provides new insights from recent discoveries involving the modulation of Nrf2 and ferroptosis in a range of lung diseases. It also systematically describes regulatory mechanisms involving lipid peroxidation, intracellular antioxidant levels, ubiquitination of Nrf2, and expression of FSP1 and GPX4. Finally, it summarises active ingredients and drugs with potential for the treatment of lung diseases. With the overarching aim of expediting improvements in treatment, this review provides a reference for novel therapeutic mechanisms and offers suggestions for the development of new medications for a variety of lung disorders.
Collapse
Affiliation(s)
- Yonghu Chen
- College of Pharmacy, Yanbian University Hospital, Yanbian University, Yanji, 133002, P. R. China
| | - Zhe Jiang
- College of Pharmacy, Yanbian University Hospital, Yanbian University, Yanji, 133002, P. R. China.
| | - Xuezheng Li
- College of Pharmacy, Yanbian University Hospital, Yanbian University, Yanji, 133002, P. R. China.
| |
Collapse
|
14
|
Powers SK, Lategan-Potgieter R, Goldstein E. Exercise-induced Nrf2 activation increases antioxidant defenses in skeletal muscles. Free Radic Biol Med 2024; 224:470-478. [PMID: 39181477 DOI: 10.1016/j.freeradbiomed.2024.07.041] [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: 05/27/2024] [Accepted: 07/30/2024] [Indexed: 08/27/2024]
Abstract
Following the discovery that exercise increases the production of reactive oxygen species in contracting skeletal muscles, evidence quickly emerged that endurance exercise training increases the abundance of key antioxidant enzymes in the trained muscles. Since these early observations, knowledge about the impact that regular exercise has on skeletal muscle antioxidant capacity has increased significantly. Importantly, in recent years, our understanding of the cell signaling pathways responsible for this exercise-induced increase in antioxidant enzymes has expanded exponentially. Therefore, the goals of this review are: 1) summarize our knowledge about the influence that exercise training has on the abundance of key antioxidant enzymes in skeletal muscles; and 2) to provide a state-of-the-art review of the nuclear factor erythroid 2-related factor (Nrf2) signaling pathway that is responsible for many of the exercise-induced changes in muscle antioxidant capacity. We begin with a discussion of the sources of reactive oxygen species in contracting muscles and then examine the exercise-induced changes in the antioxidant enzymes that eliminate both superoxide radicals and hydrogen peroxide in muscle fibers. We conclude with a discussion of the advances in our understanding of the exercise-induced control of the Nrf2 signaling pathway that is responsible for the expression of numerous antioxidant proteins. In hopes of stimulating future research, we also identify gaps in our knowledge about the signaling pathways responsible for the exercise-induced increases in muscle antioxidant enzymes.
Collapse
Affiliation(s)
- Scott K Powers
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA.
| | | | - Erica Goldstein
- Department of Health Sciences, Stetson University, Deland, FL, USA
| |
Collapse
|
15
|
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] [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
|
16
|
Ouyang S, Zeng Z, He J, Luo L. Epigenetic regulation of targeted ferroptosis: A new strategy for drug development. J Pharm Anal 2024; 14:101012. [PMID: 39850234 PMCID: PMC11755343 DOI: 10.1016/j.jpha.2024.101012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 05/20/2024] [Accepted: 05/23/2024] [Indexed: 01/25/2025] Open
Abstract
Ferroptosis is a newly discovered form of cell death that is influenced by iron levels and is triggered by cellular metabolism and excessive lipid peroxidation. Epigenetic regulation plays a crucial role in the development and progression of diseases, making it essential to understand these mechanisms in order to identify potential targets for drug development and clinical treatment. The intersection of ferroptosis and epigenetics has opened up new avenues for research in drug development, offering innovative strategies for combating diseases. Recent studies have shown that epigenetic modifications can impact pathways related to ferroptosis, potentially leading to organ dysfunction. Despite the increasing focus on this relationship, the role of epigenetic regulation in drug development remains largely unexplored. This article explores current research on the interplay between epigenetic regulation and ferroptosis, delving into their regulatory mechanisms and discussing the effects of existing epigenetic modification regulators on diseases. Additionally, we highlight ongoing research on epigenetic factors involved in targeting ferroptosis in cancer, providing new insights for the development of cancer treatments.
Collapse
Affiliation(s)
- Shengli Ouyang
- The First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong, 524023, China
| | - Zeyao Zeng
- The First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong, 524023, China
| | - Jieyi He
- The First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong, 524023, China
| | - Lianxiang Luo
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, Guangdong, 524023, China
| |
Collapse
|
17
|
Shang J, Yan J, Lou H, Shou R, Zhan Y, Lu X, Fan X. Genome-wide DNA methylation sequencing reveals the involvement of ferroptosis in hepatotoxicity induced by dietary exposure to food-grade titanium dioxide. Part Fibre Toxicol 2024; 21:37. [PMID: 39294687 PMCID: PMC11409784 DOI: 10.1186/s12989-024-00598-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 09/06/2024] [Indexed: 09/21/2024] Open
Abstract
BACKGROUND Following the announcement by the European Food Safety Authority that the food additive titanium dioxide (E 171) is unsafe for human consumption, and the subsequent ban by the European Commission, concerns have intensified over the potential risks E 171 poses to human vital organs. The liver is the main organ for food-grade nanoparticle metabolism. It is increasingly being found that epigenetic changes may play an important role in nanomaterial-induced hepatotoxicity. However, the profound effects of E 171 on the liver, especially at the epigenetic level, remain largely unknown. METHODS Mice were exposed orally to human-relevant doses of two types of E 171 mixed in diet for 28 and/or 84 days. Conventional toxicology and global DNA methylation analyses were performed to assess E 171-induced hepatotoxicity and epigenetic changes. Whole genome bisulfite sequencing and further ferroptosis protein detection were used to reveal E 171-induced changes in liver methylation profiles and toxic mechanisms. RESULTS Exposed to E 171 for 28 and/or 84 days resulted in reduced global DNA methylation and hydroxymethylation in the liver of mice. E 171 exposure for 84 days elicited inflammation and damage in the mouse liver, whereas 28-day exposure did not. Whole-genome DNA methylation sequencing disclosed substantial methylation alterations at the CG and non-CG sites of the liver DNA in mice exposed to E 171 for 84 days. Mechanistic analysis of the DNA methylation alterations indicated that ferroptosis contributed to the liver toxicity induced by E 171. E 171-induced DNA methylation changes triggered NCOA4-mediated ferritinophagy, attenuated the protein levels of GPX4, FTH1, and FTL in the liver, and thereby caused ferroptosis. CONCLUSIONS Long-term oral exposure to E 171 triggers hepatotoxicity and induces methylation changes in both CG and non-CG sites of liver DNA. These epigenetic alterations activate ferroptosis in the liver through NCOA4-mediated ferritinophagy, highlighting the role of DNA methylation and ferroptosis in the potential toxicity caused by E 171 in vivo.
Collapse
Affiliation(s)
- Jiaxin Shang
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Jun Yan
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - He Lou
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Rongshang Shou
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yingqi Zhan
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Xiaoyan Lu
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- State Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314102, China.
- Jinhua Institute of Zhejiang University, Jinhua, 321299, China.
- Department of Infectious Diseases, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China.
| | - Xiaohui Fan
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- State Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314102, China.
- Jinhua Institute of Zhejiang University, Jinhua, 321299, China.
- The Joint-Laboratory of Clinical Multi-Omics Research between Zhejiang University and Ningbo Municipal Hospital of TCM, Ningbo Municipal Hospital of TCM, Ningbo, 315010, China.
| |
Collapse
|
18
|
Jiang J, Zheng Z, Chen S, Liu J, Jia J, Huang Y, Liu Q, Cheung CY, Sin DD, Yang T, Wang C. Hypoxia inducible factor (HIF) 3α prevents COPD by inhibiting alveolar epithelial cell ferroptosis via the HIF-3α-GPx4 axis. Theranostics 2024; 14:5512-5527. [PMID: 39310101 PMCID: PMC11413794 DOI: 10.7150/thno.99237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 08/10/2024] [Indexed: 09/25/2024] Open
Abstract
Rationale: COPD patients are largely asymptomatic until the late stages when prognosis is generally poor. In this study, we shifted the focus to pre-COPD and smoking stages, and found enrichment of hypoxia inducible factor (HIF)-3α is in pre-COPD samples. Smoking induced regional tissue hypoxia and emphysema have been found in COPD patients. However, the mechanisms underlying hypoxia especially HIF-3α and COPD have not been investigated. Methods: We performed bulk-RNA sequencing on 36 peripheral lung tissue specimens from non-smokers, smokers, pre-COPD and COPD patients, and using "Mfuzz" algorithm to analysis the dataset dynamically. GSE171541 and EpCAM co-localization analyses were used to explore HIF-3α localization. Further, SftpcCreert2/+R26LSL-Hif3a knock-in mice and small molecular inhibitors in vitro were used to explore the involvement of HIF-3α in the pathophysiology of COPD. Results: Reactive oxygen species (ROS) and hypoxia were enriched in pre-COPD samples, and HIF-3α was downregulated in alveolar epithelial cells in COPD. In vitro experiments using lentivirus transfection, bulk-RNA seq, and RSL3 showed that the activation of the HIF-3α-GPx4 axis inhibited alveolar epithelial cell ferroptosis when treated with cigarettes smoking extracts (CSE). Further results from SftpcCreert2/+R26LSL-Hif3a knock-in mice demonstrated overexpression of HIF-3α inhibited alveolar epithelial cells ferroptosis and prevented the decline of lung function. Conclusion: Hypoxia and oxidation-related damage begins years before the onset of COPD symptoms, suggesting the imbalance and impairment of intracellular homeostatic system. The activation of the HIF-3α-GPx4 axis is a promising treatment target. By leveraging this comprehensive analysis method, more potential targets could be found and enhancing our understanding of the pathogenesis.
Collapse
Affiliation(s)
- Junchao Jiang
- China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, CN
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity, Beijing, CN
- The University of British Columbia, Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, CA
| | - Zhoude Zheng
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity, Beijing, CN
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, CN
| | - Shengsong Chen
- First Affiliated Hospital of Nanchang University, Department of Pulmonary and Critical Care Medicine, Nanchang, Jiangxi, CN
| | - Jixiang Liu
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity, Beijing, CN
| | - Ju Jia
- Department of Infectious Disease, Beijing Friendship Hospital, Capital Medical University, Beijing, CN
| | - Yuhang Huang
- China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, CN
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity, Beijing, CN
| | - Qing Liu
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity, Beijing, CN
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, CN
| | - Chung Y Cheung
- The University of British Columbia, Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, CA
| | - Don D Sin
- The University of British Columbia, Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, CA
- The University of British Columbia, Division of Respiratory Medicine, Department of Medicine, Vancouver, BC, CA
| | - Ting Yang
- China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, CN
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity, Beijing, CN
| | - Chen Wang
- China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, CN
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity, Beijing, CN
| |
Collapse
|
19
|
Xu M, Zhang D, Yan J. Targeting ferroptosis using Chinese herbal compounds to treat respiratory diseases. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 130:155738. [PMID: 38824825 DOI: 10.1016/j.phymed.2024.155738] [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: 02/01/2024] [Revised: 04/27/2024] [Accepted: 05/14/2024] [Indexed: 06/04/2024]
Abstract
BACKGROUND Respiratory diseases pose a grave threat to human life. Therefore, understanding their pathogenesis and therapeutic strategy is important. Ferroptosis is a novel type of iron-dependent programmed cell death, distinct from apoptosis, necroptosis, and autophagy, characterised by iron, reactive oxygen species, and lipid peroxide accumulation, as well as glutathione (GSH) depletion and GSH peroxidase 4 (GPX4) inactivation. A close association between ferroptosis and the onset and progression of respiratory diseases, including chronic obstructive pulmonary disease, acute lung injury, bronchial asthma, pulmonary fibrosis, and lung cancer, has been reported. Recent studies have shown that traditional Chinese medicine (TCM) compounds exhibit unique advantages in the treatment of respiratory diseases owing to their natural properties and potential efficacy. These compounds can effectively regulate ferroptosis by modulating several key signalling pathways such as system Xc- -GSH-GPX4, NCOA4-mediated ferritinophagy, Nrf2-GPX4, and Nrf2/HO-1, thus playing a positive role in improving respiratory diseases. PURPOSE This comprehensive review systematically outlines the regulatory role of ferroptosis in the onset and progression of respiratory diseases and provides evidence for treating respiratory diseases by targeting ferroptosis with TCM compounds. These insights aim to offer potential remedies for the clinical prevention and treatment of respiratory diseases. STUDY DESIGN AND METHODS We searched scientific databases PubMed, Web of Science, Scopus, and CNKI using keywords such as "ferroptosis","respiratory diseases","chronic obstructive pulmonary disease","bronchial asthma","acute lung injury","pulmonary fibrosis","lung cancer","traditional Chinese medicine","traditional Chinese medicine compound","monomer", and "natural product" to retrieve studies on the therapeutic potential of TCM compounds in ameliorating respiratory diseases by targeting ferroptosis. The retrieved data followed PRISMA criteria (preferred reporting items for systematic review). RESULTS TCM compounds possess unique advantages in treating respiratory diseases, stemming from their natural origins and proven clinical effectiveness. TCM compounds can exert therapeutic effects on respiratory diseases by regulating ferroptosis, which mainly involves modulation of pathways such as system Xc- -GSH-GPX4,NCOA4-mediated ferritinophagy, Nrf2-GPX4, and Nrf2/HO-1. CONCLUSION TCM compounds have demonstrated promising potential in improving respiratory diseases through the regulation of ferroptosis. The identification of specific TCM-related inducers and inhibitors of ferroptosis holds great significance in developing more effective strategies. However, current research remains confined to animal and cellular studies, emphasizing the imperative for further verifications through high-quality clinical data.
Collapse
Affiliation(s)
- Mengjiao Xu
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing 100102, China
| | - Di Zhang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Jun Yan
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China.
| |
Collapse
|
20
|
Yan M, Xu S, Wang H, Dong S, Mo C. Ferroptosis in chronic obstructive pulmonary disease: From cellular mechanisms to therapeutic applications. Chin Med J (Engl) 2024; 137:1237-1239. [PMID: 38595135 PMCID: PMC11101239 DOI: 10.1097/cm9.0000000000003079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Indexed: 04/11/2024] Open
Affiliation(s)
- Mengli Yan
- The Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Department of Nephrology, Henan Provincial Clinical Research Center for Kidney Disease, Henan Provincial Key Laboratory of Kidney Disease and Immunology, Henan Provincial People’s Hospital and People’s Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Shiyu Xu
- The Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Han Wang
- Department of Biochemistry, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
- Center for RNA Science and Therapeutics, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Shoujin Dong
- Department of Pulmonary and Critical Care Medicine, Chengdu First People’s Hospital, Chengdu, Sichuan 610095, China
| | - Chunheng Mo
- The Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
21
|
Xia W, Zhang M, Liu C, Wang S, Xu A, Xia Z, Pang L, Cai Y. Exploring the therapeutic potential of tetrahydrobiopterin for heart failure with preserved ejection fraction: A path forward. Life Sci 2024; 345:122594. [PMID: 38537900 DOI: 10.1016/j.lfs.2024.122594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 03/10/2024] [Accepted: 03/24/2024] [Indexed: 04/02/2024]
Abstract
A large number of patients are affected by classical heart failure (HF) symptomatology with preserved ejection fraction (HFpEF) and multiorgan syndrome. Due to high morbidity and mortality rate, hospitalization and mortality remain serious socioeconomic problems, while the lack of effective pharmacological or device treatment means that HFpEF presents a major unmet medical need. Evidence from clinical and basic studies demonstrates that systemic inflammation, increased oxidative stress, and impaired mitochondrial function are the common pathological mechanisms in HFpEF. Tetrahydrobiopterin (BH4), beyond being an endogenous co-factor for catalyzing the conversion of some essential biomolecules, has the capacity to prevent systemic inflammation, enhance antioxidant resistance, and modulate mitochondrial energy production. Therefore, BH4 has emerged in the last decade as a promising agent to prevent or reverse the progression of disorders such as cardiovascular disease. In this review, we cover the clinical progress and limitations of using downstream targets of nitric oxide (NO) through NO donors, soluble guanylate cyclase activators, phosphodiesterase inhibitors, and sodium-glucose co-transporter 2 inhibitors in treating cardiovascular diseases, including HFpEF. We discuss the use of BH4 in association with HFpEF, providing new evidence for its potential use as a pharmacological option for treating HFpEF.
Collapse
Affiliation(s)
- Weiyi Xia
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Miao Zhang
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China; Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Guangdong, China
| | - Chang Liu
- Department of Anesthesiology, The First Hospital of Jilin University, Jilin, China
| | - Sheng Wang
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China; Department of Medicine, The University of Hong Kong, Hong Kong SAR, China; Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| | - Zhengyuan Xia
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Guangdong, China
| | - Lei Pang
- Department of Anesthesiology, The First Hospital of Jilin University, Jilin, China.
| | - Yin Cai
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China; Research Center for Chinese Medicine Innovation, The Hong Kong Polytechnic University, Hong Kong SAR, China; Research Institute for Future Food, The Hong Kong Polytechnic University, Hong Kong SAR, China.
| |
Collapse
|
22
|
Barrio E, Lerma-Puertas D, Jaulín-Pueyo JJ, Labarta JI, Gascón-Catalán A. Epigenetic modifications in the ferroptosis pathway in cord blood cells from newborns of smoking mothers and their influence on fetal growth. Reprod Toxicol 2024; 125:108581. [PMID: 38552991 DOI: 10.1016/j.reprotox.2024.108581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 03/12/2024] [Accepted: 03/22/2024] [Indexed: 04/01/2024]
Abstract
Maternal smoking during pregnancy increases oxidative stress and decreases antioxidant capacity in newborns. Uncontrolled oxidative stress plays a role in fetal development disorders and in adverse perinatal outcomes. In order to identify molecular pathways involved in low fetal growth, epigenetic modifications in newborns of smoking and non-smoking mothers were examined. Low birth weight newborns of mothers who smoked more than 10 cigarettes per day during the first trimester of pregnancy and normal birth weight newborns of mothers who did not smoke during pregnancy were included in the study. DNA was extracted from umbilical cord blood of term newborns. 125 differentially methylated regions were identified by MeDIP-Seq. Functional analysis revealed several pathways, such as ferroptosis, that were enriched in differentially methylated genes after prenatal smoke exposure. GPX4 and PCBP1 were found to be hypermethylated and associated with low fetal growth. These epigenetic modifications in ferroptosis pathway genes in newborns of smoking mothers can potentially contribute to intrauterine growth restriction through the induction of cell death via lipid peroxidation of cell membranes. The identification of epigenetic modifications in the ferroptosis pathway sheds light on the potential mechanisms underlying the pathophysiology of low birth weight in infants born to smoking mothers.
Collapse
Affiliation(s)
- Eva Barrio
- Facultad de Medicina, Universidad de Zaragoza, Spain
| | - Diego Lerma-Puertas
- Facultad de Medicina, Universidad de Zaragoza, Spain; Servicio de Obstetricia y Ginecología, Hospital Universitario Clínico Lozano Blesa, Zaragoza, Spain
| | - José Javier Jaulín-Pueyo
- Facultad de Medicina, Universidad de Zaragoza, Spain; Servicio de Pediatría. Hospital Universitario Miguel Servet, Zaragoza, Spain
| | - José Ignacio Labarta
- Facultad de Medicina, Universidad de Zaragoza, Spain; Servicio de Pediatría. Hospital Universitario Miguel Servet, Zaragoza, Spain
| | | |
Collapse
|
23
|
Guo C, Yue Y, Wang B, Chen S, Li D, Zhen F, Liu L, Zhu H, Xie M. Anemoside B4 alleviates arthritis pain via suppressing ferroptosis-mediated inflammation. J Cell Mol Med 2024; 28:e18136. [PMID: 38334255 PMCID: PMC10853948 DOI: 10.1111/jcmm.18136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 11/02/2023] [Accepted: 12/20/2023] [Indexed: 02/10/2024] Open
Abstract
Chronic pain is the key manifestations of rheumatoid arthritis. Neuroinflammation in the spinal cord drives central sensitization and chronic pain. Ferroptosis has potentially important roles in the occurrence of neuroinflammation and chronic pain. In the current study, mouse model of collagen-induced arthritis was established by intradermal injection of type II collagen in complete Freund's adjuvant (CFA) solution. CFA inducement resulted in swollen paw and ankle, mechanical and spontaneous pain, and impaired motor coordination. The spinal inflammation was triggered, astrocytes were activated, and increased NLRP3-mediated inflammatory signal was found in CFA spinal cord. Oxidative stress and ferroptosis in the spinal cord were manifested. Meanwhile, enhancive spinal GSK-3β activity and abnormal phosphorylated Drp1 were observed. To investigate the potential therapeutic options for arthritic pain, mice were intraperitoneally injected with AB4 for three consecutive days. AB4 treatment reduced pain sensitivity and increased the motor coordination. In the spinal cord, AB4 treatment inhibited NLRP3 inflammasome-mediated inflammatory response, increased antioxidation, decreased mitochondrial reactive oxygen species and ferroptosis. Furthermore, AB4 decreased GSK-3β activity by binding with GSK-3β through five electrovalent bonds. Our findings indicated that AB treatment relieves arthritis pain by inhibiting GSK-3β activation, increasing antioxidant capability, reducing Drp1-mediated mitochondrial dysfunction and suppressing neuroinflammation.
Collapse
Affiliation(s)
- Chenlu Guo
- School of PharmacyHubei University of Science and TechnologyXianningChina
| | - Yuanfen Yue
- Department of ObstetricsXianning Central Hospital, First Affiliated Hospital of Hubei University of Science and TechnologyXianningChina
| | - Bojun Wang
- Hubei Key Laboratory of Diabetes and Angiopathy, School of Basic Medical Sciences, Xianning Medical CollegeHubei University of Science and TechnologyXianningChina
| | - Shaohui Chen
- Hubei Key Laboratory of Diabetes and Angiopathy, School of Basic Medical Sciences, Xianning Medical CollegeHubei University of Science and TechnologyXianningChina
| | - Dai Li
- Hubei Key Laboratory of Diabetes and Angiopathy, School of Basic Medical Sciences, Xianning Medical CollegeHubei University of Science and TechnologyXianningChina
| | - Fangshou Zhen
- Department of PharmacyMatang Hospital of Traditional Chinese MedicineXianningChina
| | - Ling Liu
- Hubei Key Laboratory of Diabetes and Angiopathy, School of Basic Medical Sciences, Xianning Medical CollegeHubei University of Science and TechnologyXianningChina
| | - Haili Zhu
- Hubei Key Laboratory of Diabetes and Angiopathy, School of Basic Medical Sciences, Xianning Medical CollegeHubei University of Science and TechnologyXianningChina
| | - Min Xie
- Hubei Key Laboratory of Diabetes and Angiopathy, School of Basic Medical Sciences, Xianning Medical CollegeHubei University of Science and TechnologyXianningChina
| |
Collapse
|
24
|
Wang Y, Hu J, Wu S, Fleishman JS, Li Y, Xu Y, Zou W, Wang J, Feng Y, Chen J, Wang H. Targeting epigenetic and posttranslational modifications regulating ferroptosis for the treatment of diseases. Signal Transduct Target Ther 2023; 8:449. [PMID: 38072908 PMCID: PMC10711040 DOI: 10.1038/s41392-023-01720-0] [Citation(s) in RCA: 74] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 09/16/2023] [Accepted: 11/18/2023] [Indexed: 12/18/2023] Open
Abstract
Ferroptosis, a unique modality of cell death with mechanistic and morphological differences from other cell death modes, plays a pivotal role in regulating tumorigenesis and offers a new opportunity for modulating anticancer drug resistance. Aberrant epigenetic modifications and posttranslational modifications (PTMs) promote anticancer drug resistance, cancer progression, and metastasis. Accumulating studies indicate that epigenetic modifications can transcriptionally and translationally determine cancer cell vulnerability to ferroptosis and that ferroptosis functions as a driver in nervous system diseases (NSDs), cardiovascular diseases (CVDs), liver diseases, lung diseases, and kidney diseases. In this review, we first summarize the core molecular mechanisms of ferroptosis. Then, the roles of epigenetic processes, including histone PTMs, DNA methylation, and noncoding RNA regulation and PTMs, such as phosphorylation, ubiquitination, SUMOylation, acetylation, methylation, and ADP-ribosylation, are concisely discussed. The roles of epigenetic modifications and PTMs in ferroptosis regulation in the genesis of diseases, including cancers, NSD, CVDs, liver diseases, lung diseases, and kidney diseases, as well as the application of epigenetic and PTM modulators in the therapy of these diseases, are then discussed in detail. Elucidating the mechanisms of ferroptosis regulation mediated by epigenetic modifications and PTMs in cancer and other diseases will facilitate the development of promising combination therapeutic regimens containing epigenetic or PTM-targeting agents and ferroptosis inducers that can be used to overcome chemotherapeutic resistance in cancer and could be used to prevent other diseases. In addition, these mechanisms highlight potential therapeutic approaches to overcome chemoresistance in cancer or halt the genesis of other diseases.
Collapse
Affiliation(s)
- Yumin Wang
- Department of Respiratory and Critical Care Medicine, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, PR China
| | - Jing Hu
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300060, PR China
| | - Shuang Wu
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, 430000, PR China
| | - Joshua S Fleishman
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Yulin Li
- Department of Respiratory and Critical Care Medicine, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, PR China
| | - Yinshi Xu
- Department of Outpatient, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, PR China
| | - Wailong Zou
- Department of Respiratory and Critical Care Medicine, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, PR China
| | - Jinhua Wang
- Beijing Key Laboratory of Drug Target and Screening Research, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, PR China.
| | - Yukuan Feng
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, PR China.
| | - Jichao Chen
- Department of Respiratory and Critical Care Medicine, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, PR China.
| | - Hongquan Wang
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, PR China.
| |
Collapse
|
25
|
Zeng Z, Li T, Liu X, Ma Y, Luo L, Wang Z, Zhao Z, Li H, He X, Zeng H, Tao Y, Chen Y. DNA dioxygenases TET2 deficiency promotes cigarette smoke induced chronic obstructive pulmonary disease by inducing ferroptosis of lung epithelial cell. Redox Biol 2023; 67:102916. [PMID: 37812881 PMCID: PMC10579541 DOI: 10.1016/j.redox.2023.102916] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 09/21/2023] [Accepted: 10/02/2023] [Indexed: 10/11/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a significant global cause of morbidity and mortality currently. Long-term exposure of cigarette smoke (CS) inducing persistent inflammation, small airway remodeling and emphysematous lung are the distinguishing features of COPD. Ferroptosis, occurred in lung epithelial cells has recently been reported to be associated with COPD pathogenesis. DNA dioxygenase ten-eleven translocation 2 (TET2) is an important demethylase and its genetic mutation is associated with low forced expiratory volume in 1 s (FEV1) of lung function. However, its role in COPD remains elusive. Here, we found that TET2 regulates CS induced lipid peroxidation through demethylating glutathione peroxidase 4 (GPx4), thus alleviating airway epithelial cell ferroptosis in COPD. TET2 protein levels were mainly reduced in the airway epithelia of COPD patients, mouse models, and CS extract-treated bronchial epithelial cells. The deletion of TET2 triggered ferroptosis and further exaggerated CS-induced airway remodeling, inflammation, and emphysema in vivo. Moreover, we demonstrated that TET2 silencing intensified ferroptosis, while TET2 overexpression inhibited ferroptosis in airway epithelial cell treated with CSE. Mechanically, TET2 protected airway epithelial cells from CS-induced lipid peroxidation and ferroptosis through demethylating the promoter of glutathione peroxidase 4 (GPx4). Finally, co-administration of methylation inhibitor 5'-aza-2'-deoxycytidine (5-AZA) and the antioxidant N-acetyl-cysteine (NAC) have more protective effects on CS-induced COPD than either administration alone. Overall, our study reveals that TET2 is an essential modulator in the lipid peroxidation and ferroptosis of airway epithelial cell, and could act as a potential therapeutic target for CS-induced COPD.
Collapse
Affiliation(s)
- Zihang Zeng
- Department of Respiratory and Critical Care Medicine, The Second Xiangya Hospital of Central South University, Changsha, China; Research Unit of Respiratory Disease, Central South University, Changsha, China; Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, China
| | - Tiao Li
- Department of Respiratory and Critical Care Medicine, The Second Xiangya Hospital of Central South University, Changsha, China; Research Unit of Respiratory Disease, Central South University, Changsha, China; Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, China; Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiangming Liu
- Department of Respiratory and Critical Care Medicine, The Second Xiangya Hospital of Central South University, Changsha, China; Research Unit of Respiratory Disease, Central South University, Changsha, China; Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, China
| | - Yiming Ma
- Department of Respiratory and Critical Care Medicine, The Second Xiangya Hospital of Central South University, Changsha, China; Research Unit of Respiratory Disease, Central South University, Changsha, China; Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, China
| | - Lijuan Luo
- Department of Respiratory and Critical Care Medicine, The Second Xiangya Hospital of Central South University, Changsha, China; Research Unit of Respiratory Disease, Central South University, Changsha, China; Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, China
| | - ZuLi Wang
- Center for Tissue Engineering and Stem Cell Research, Guizhou Medical University, Changsha, China
| | - Zhiqi Zhao
- Department of Respiratory and Critical Care Medicine, The Second Xiangya Hospital of Central South University, Changsha, China; Research Unit of Respiratory Disease, Central South University, Changsha, China; Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, China
| | - Herui Li
- Department of Respiratory and Critical Care Medicine, The Second Xiangya Hospital of Central South University, Changsha, China; Research Unit of Respiratory Disease, Central South University, Changsha, China; Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, China
| | - Xue He
- Department of Respiratory and Critical Care Medicine, The Second Xiangya Hospital of Central South University, Changsha, China; Research Unit of Respiratory Disease, Central South University, Changsha, China; Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, China
| | - Huihui Zeng
- Department of Respiratory and Critical Care Medicine, The Second Xiangya Hospital of Central South University, Changsha, China; Research Unit of Respiratory Disease, Central South University, Changsha, China; Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, China
| | - Yongguang Tao
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, Xiangya Hospital, Central South University, Hunan, 410078, China; NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, China
| | - Yan Chen
- Department of Respiratory and Critical Care Medicine, The Second Xiangya Hospital of Central South University, Changsha, China; Research Unit of Respiratory Disease, Central South University, Changsha, China; Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, China.
| |
Collapse
|
26
|
Xie J, Liu M, Gao Y, Liu C, Wu F, Tong J, Li Z, Zhu J. Integration of metabolomics and network pharmacology to reveal the protective mechanism underlying Qibai Pingfei capsule on chronic obstructive pulmonary disease. Front Pharmacol 2023; 14:1258138. [PMID: 37920214 PMCID: PMC10618342 DOI: 10.3389/fphar.2023.1258138] [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: 07/13/2023] [Accepted: 10/09/2023] [Indexed: 11/04/2023] Open
Abstract
In this study, we have employed metabolomics technology in combination with network pharmacology to ascertain the key metabolites and hub genes. The objective was to explore the pathway of Qibai Pingfei Capsule (QBPF) in treating COPD through metabolomics. We identified 96 differential metabolites in the lung tissues of rats belonging to control and model groups, out of which 47 were observed to be critical (VIP >2, p < 0.05). Furthermore, 16 important differential metabolites were reversed after QBPF treatment. Using network pharmacology, we identified 176 core targets of 81 drug-active ingredients. Our comprehensive analysis of network pharmacology and metabolomics enabled us to identify a core target, prostaglandin-endoperoxide synthase 2 (PTGS2), and a core metabolic pathway for glutathione metabolism. Finally, the result of molecular docking showed that PTGS2 had strong binding activity to 18 compounds including Fumarine and Kaempferol, etc.. PTGS2 is a marker of ferroptosis, so we wanted to explore whether QBPF could inhibit ferroptosis in COPD. The results showed that ferroptosis was involved in the pathogenesis of COPD, and QBPF could inhibit the occurrence of ferroptosis. In conclusion, the mechanism of QBPF for treating COPD may be related to PTGS2 expression, glutathione metabolism and ferroptosis.
Collapse
Affiliation(s)
- Jinghui Xie
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Mengxiang Liu
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Yating Gao
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine, Institute of Health and Medicine, Anhui University of Chinese Medicine, Hefei, China
- The First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, Anhui, China
| | - Changan Liu
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Fan Wu
- College of Traditional Chinese Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Jiabing Tong
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine, Institute of Health and Medicine, Anhui University of Chinese Medicine, Hefei, China
- The First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, Anhui, China
- College of Traditional Chinese Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Zegeng Li
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine, Institute of Health and Medicine, Anhui University of Chinese Medicine, Hefei, China
- The First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, Anhui, China
| | - Jie Zhu
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine, Institute of Health and Medicine, Anhui University of Chinese Medicine, Hefei, China
- Institutes of Integrative Medicine, Fudan University, Shanghai, China
| |
Collapse
|
27
|
Qi Q, Yang S, Li J, Li P, Du L. Regulation of Redox Homeostasis Through DNA/RNA Methylation and Post-Translational Modifications in Cancer Progression. Antioxid Redox Signal 2023; 39:531-550. [PMID: 37462995 DOI: 10.1089/ars.2023.0371] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/25/2023]
Abstract
Significance: Aberrant redox homeostasis, characterized by the enhancement of intracellular reactive oxygen species (ROS) and antioxidant defenses, is among the well-known cancer hallmarks. Understanding the regulatory mechanisms of redox homeostasis in cancer cells has become the focus of many studies. Epigenetic and post-translational modifications (PTMs), as pivotal regulators of multiple biological processes, play critical roles in tumorigenesis and development. Recent Advances: DNA and RNA methylation are important forms of epigenetic modifications. Recent evidence suggests that DNA/RNA methylation and PTMs can modulate redox homeostasis in multiple manners including affecting key molecules in ROS production, elimination, and redox-related signaling, thereby participating in tumor progression. Critical Issues: The regulatory effects of DNA/RNA methylation and PTMs on ROS are of crucial importance for tumor progression. In this review, we introduce the dual role of ROS in cancer, and then focus on the mechanistic role of DNA/RNA methylation and PTMs, especially ubiquitination and acetylation, in regulating redox homeostasis to involve in cancer progression. Future Directions: A complete understanding of how epigenetics and PTMs function in the regulation of redox homeostasis in cancer progression might expand a new direction for the progression mechanisms and therapeutic targets of cancer. Antioxid. Redox Signal. 39, 531-550.
Collapse
Affiliation(s)
- Qiuchen Qi
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, China
- Shandong Provincial Key Laboratory of Laboratory Medicine Innovation Technology, Jinan, China
| | - Suli Yang
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, China
| | - Juan Li
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, China
- Shandong Provincial Key Laboratory of Laboratory Medicine Innovation Technology, Jinan, China
- Shandong Engineering and Technology Research Center for Tumor Marker Detection, Jinan, China
| | - Peilong Li
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, China
- Shandong Provincial Key Laboratory of Laboratory Medicine Innovation Technology, Jinan, China
- Shandong Engineering and Technology Research Center for Tumor Marker Detection, Jinan, China
| | - Lutao Du
- Shandong Provincial Key Laboratory of Laboratory Medicine Innovation Technology, Jinan, China
- Shandong Engineering and Technology Research Center for Tumor Marker Detection, Jinan, China
- Department of Clinical Laboratory, Qilu Hospital, Shandong University, Jinan, China
| |
Collapse
|
28
|
Raby KL, Michaeloudes C, Tonkin J, Chung KF, Bhavsar PK. Mechanisms of airway epithelial injury and abnormal repair in asthma and COPD. Front Immunol 2023; 14:1201658. [PMID: 37520564 PMCID: PMC10374037 DOI: 10.3389/fimmu.2023.1201658] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 06/19/2023] [Indexed: 08/01/2023] Open
Abstract
The airway epithelium comprises of different cell types and acts as a physical barrier preventing pathogens, including inhaled particles and microbes, from entering the lungs. Goblet cells and submucosal glands produce mucus that traps pathogens, which are expelled from the respiratory tract by ciliated cells. Basal cells act as progenitor cells, differentiating into different epithelial cell types, to maintain homeostasis following injury. Adherens and tight junctions between cells maintain the epithelial barrier function and regulate the movement of molecules across it. In this review we discuss how abnormal epithelial structure and function, caused by chronic injury and abnormal repair, drives airway disease and specifically asthma and chronic obstructive pulmonary disease (COPD). In both diseases, inhaled allergens, pollutants and microbes disrupt junctional complexes and promote cell death, impairing the barrier function and leading to increased penetration of pathogens and a constant airway immune response. In asthma, the inflammatory response precipitates the epithelial injury and drives abnormal basal cell differentiation. This leads to reduced ciliated cells, goblet cell hyperplasia and increased epithelial mesenchymal transition, which contribute to impaired mucociliary clearance and airway remodelling. In COPD, chronic oxidative stress and inflammation trigger premature epithelial cell senescence, which contributes to loss of epithelial integrity and airway inflammation and remodelling. Increased numbers of basal cells showing deregulated differentiation, contributes to ciliary dysfunction and mucous hyperproduction in COPD airways. Defective antioxidant, antiviral and damage repair mechanisms, possibly due to genetic or epigenetic factors, may confer susceptibility to airway epithelial dysfunction in these diseases. The current evidence suggests that a constant cycle of injury and abnormal repair of the epithelium drives chronic airway inflammation and remodelling in asthma and COPD. Mechanistic understanding of injury susceptibility and damage response may lead to improved therapies for these diseases.
Collapse
Affiliation(s)
- Katie Louise Raby
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | | | - James Tonkin
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Department of Respiratory Medicine, Royal Brompton and Harefield Hospital, London, United Kingdom
| | - Kian Fan Chung
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Department of Respiratory Medicine, Royal Brompton and Harefield Hospital, London, United Kingdom
| | - Pankaj Kumar Bhavsar
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Department of Respiratory Medicine, Royal Brompton and Harefield Hospital, London, United Kingdom
| |
Collapse
|
29
|
Wang X, Kong X, Feng X, Jiang DS. Effects of DNA, RNA, and Protein Methylation on the Regulation of Ferroptosis. Int J Biol Sci 2023; 19:3558-3575. [PMID: 37497000 PMCID: PMC10367552 DOI: 10.7150/ijbs.85454] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 06/26/2023] [Indexed: 07/28/2023] Open
Abstract
Ferroptosis is a form of programmed cell death characterized by elevated intracellular ferrous ion levels and increased lipid peroxidation. Since its discovery and characterization in 2012, considerable progress has been made in understanding the regulatory mechanisms and pathophysiological functions of ferroptosis. Recent findings suggest that numerous organ injuries (e.g., ischemia/reperfusion injury) and degenerative pathologies (e.g., aortic dissection and neurodegenerative disease) are driven by ferroptosis. Conversely, insufficient ferroptosis has been linked to tumorigenesis. Furthermore, a recent study revealed the effect of ferroptosis on hematopoietic stem cells under physiological conditions. The regulatory mechanisms of ferroptosis identified to date include mainly iron metabolism, such as iron transport and ferritinophagy, and redox systems, such as glutathione peroxidase 4 (GPX4)-glutathione (GSH), ferroptosis-suppressor-protein 1 (FSP1)-CoQ10, FSP1-vitamin K (VK), dihydroorotate dehydrogenase (DHODH)-CoQ, and GTP cyclohydrolase 1 (GCH1)-tetrahydrobiopterin (BH4). Recently, an increasing number of studies have demonstrated the important regulatory role played by epigenetic mechanisms, especially DNA, RNA, and protein methylation, in ferroptosis. In this review, we provide a critical analysis of the molecular mechanisms and regulatory networks of ferroptosis identified to date, with a focus on the regulatory role of DNA, RNA, and protein methylation. Furthermore, we discuss some debated findings and unanswered questions that should be the foci of future research in this field.
Collapse
Affiliation(s)
- Xiancan Wang
- Department of Cardiovascular Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430014, Hubei, China
| | - Xianghai Kong
- Department of Intervention & Vascular Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and echnology, Wuhan, 430014, Hubei, China
| | - Xin Feng
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ding-Sheng Jiang
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, Hubei, China
| |
Collapse
|
30
|
Liu C, Lu J, Yuan T, Xie L, Zhang L. EPC-exosomal miR-26a-5p improves airway remodeling in COPD by inhibiting ferroptosis of bronchial epithelial cells via PTGS2/PGE2 signaling pathway. Sci Rep 2023; 13:6126. [PMID: 37059741 PMCID: PMC10104834 DOI: 10.1038/s41598-023-33151-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 04/07/2023] [Indexed: 04/16/2023] Open
Abstract
We aimed to investigate whether exosomes (Exo) affected chronic obstructive pulmonary disease (COPD) by influencing ferroptosis of bronchial epithelial cells (BECs) and the mechanisms involved. Here we took the peripheral blood samples of normal subjects and COPD patients, extracted and identified endothelial progenitor cells (EPCs) and EPC-Exo. An animal model of COPD was established. Then human BECs were taken and treated with cigarette smoke extract (CSE) for 24 h to construct a COPD cell model. Next, we screened differentially expressed ferroptosis-related genes in COPD patients by bioinformatics. Bioinformatics predicted the miRNA targeting PTGS2. Then, the mechanism of action of miR-26a-5p and Exo-miR-26a-5p was investigated in vitro. We successfully isolated and identified EPC and Exo. In vitro, EPC alleviated CSE-induced ferroptosis in BECs by transporting Exo. In vivo, Exo alleviated cigarette smoke-induced ferroptosis and airway remodeling in mice. Through further validation, we found that CSE-induced ferroptosis promoted the epithelial-mesenchymal transition (EMT) of BECs. Bioinformatics analysis and validation showed that PTGS2/PGE2 pathway affected CSE-induced ferroptosis in BECs. Meanwhile, miR-26a-5p targeting PTGS2 affected CSE-induced ferroptosis in BECs. Additionally, we found that miR-26a-5p affected CSE-induced BECs EMT. Exo-miR-26a-5p alleviated CSE-induced ferroptosis and EMT. In conclusion, EPC-exosomal miR-26a-5p improved airway remodeling in COPD by inhibiting ferroptosis of BECs via the PTGS2/PGE2 pathway.
Collapse
Affiliation(s)
- Caihong Liu
- Department of Pulmonary and Critical Care Medicine, The Third Xiangya Hospital of Central South University, 138 Tongzipo Road, Yuelu District, Changsha, 410013, Hunan, China
| | - Junjuan Lu
- Department of Pulmonary and Critical Care Medicine, The Third Xiangya Hospital of Central South University, 138 Tongzipo Road, Yuelu District, Changsha, 410013, Hunan, China
| | - Ting Yuan
- Department of Nutriology, Second Xiangya Hospital, Central South University, Changsha, 410001, Hunan, China
| | - Lihua Xie
- Department of Pulmonary and Critical Care Medicine, The Third Xiangya Hospital of Central South University, 138 Tongzipo Road, Yuelu District, Changsha, 410013, Hunan, China
| | - Li Zhang
- Department of Pulmonary and Critical Care Medicine, The Third Xiangya Hospital of Central South University, 138 Tongzipo Road, Yuelu District, Changsha, 410013, Hunan, China.
| |
Collapse
|
31
|
Wang Y, Xia S. Relationship Between ACSL4-Mediated Ferroptosis and Chronic Obstructive Pulmonary Disease. Int J Chron Obstruct Pulmon Dis 2023; 18:99-111. [PMID: 36817367 PMCID: PMC9930680 DOI: 10.2147/copd.s391129] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Accepted: 01/11/2023] [Indexed: 02/12/2023] Open
Abstract
Purpose Although cigarette smoke exposure is the major risk factor for chronic obstructive pulmonary disease (COPD), the mechanism is not completely understood. The aim of the present study was to investigate whether ACSL4-mediated ferroptosis in lung epithelial cells plays a part in the COPD development process and its association. Patients and Methods In this study, animal and cell models of COPD were modelled using cigarette smoke extracts (CSEs), and cell viability, lipid ROS, iron ion deposition, and ferroptosis-related markers were measured in lung tissue and lung epithelial cells following CSE exposure. Morphological changes in mitochondria were observed in lung tissue and epithelial cells of the lung by transmission electron microscope. The expression levels of ACSL4 mRNA and protein in lung tissue and epithelial cells were measured by real-time PCR and Western blotting. In addition, animal-interfering lentivirus and cell-interfering RNA against ACSL4 were constructed in this study, ferroptosis in lung tissue and lung epithelial cells after ACSL4 interference was detected, and ACSL4 mRNA and protein expression levels were detected. Results CSE induced ferroptosis in lung tissues and lung epithelial cells, and the expression levels of ACSL4 were elevated in CSE-treated lung tissues and lung epithelial cells. After ACSL4 interference, the expression of ACSL4 decreased, mitochondrial morphology was restored, and ferroptosis in lung tissues and lung epithelial cells was alleviated. Both respiratory frequency and enhanced pause of COPD mice models decreased after ACSL4 interference. Conclusion ACSL4-mediated ferroptosis in lung epithelial cells is associated with COPD and positively correlated with ferroptosis in epithelial cells.
Collapse
Affiliation(s)
- Yingxi Wang
- Graduate School, Dalian Medical University, Dalian, Liaoning, People’s Republic of China
| | - Shuyue Xia
- Department of Respiratory and Critical Care Medicine, Central Hospital Affiliated to Shenyang Medical College, Shenyang, Liaoning, People’s Republic of China,Correspondence: Shuyue Xia, Department of Respiratory and Critical Care Medicine, Central Hospital Affiliated to Shenyang Medical College, Shenyang, Liaoning, 110075, People’s Republic of China, Tel/Fax +86-24-85715588, Email
| |
Collapse
|
32
|
McCord JM, Gao B, Hybertson BM. The Complex Genetic and Epigenetic Regulation of the Nrf2 Pathways: A Review. Antioxidants (Basel) 2023; 12:antiox12020366. [PMID: 36829925 PMCID: PMC9952775 DOI: 10.3390/antiox12020366] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/30/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Nrf2 is a major transcription factor that significantly regulates-directly or indirectly-more than 2000 genes. While many of these genes are involved in maintaining redox balance, others are involved in maintaining balance among metabolic pathways that are seemingly unrelated to oxidative stress. In the past 25 years, the number of factors involved in the activation, nuclear translocation, and deactivation of Nrf2 has continued to expand. The purpose of this review is to provide an overview of the remarkable complexity of the tortuous sequence of stop-and-go signals that not only regulate expression or repression, but may also modify transcriptional intensity as well as the specificity of promoter recognition, allowing fluidity of its gene expression profile depending on the various structural modifications the transcription factor encounters on its journey to the DNA. At present, more than 45 control points have been identified, many of which represent sites of action of the so-called Nrf2 activators. The complexity of the pathway and the synergistic interplay among combinations of control points help to explain the potential advantages seen with phytochemical compositions that simultaneously target multiple control points, compared to the traditional pharmaceutical paradigm of "one-drug, one-target".
Collapse
Affiliation(s)
- Joe M. McCord
- Pathways Bioscience, Aurora, CO 80045, USA
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Correspondence:
| | - Bifeng Gao
- Pathways Bioscience, Aurora, CO 80045, USA
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Brooks M. Hybertson
- Pathways Bioscience, Aurora, CO 80045, USA
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
33
|
Meng D, Zhu C, Jia R, Li Z, Wang W, Song S. The molecular mechanism of ferroptosis and its role in COPD. Front Med (Lausanne) 2023; 9:1052540. [PMID: 36687445 PMCID: PMC9852995 DOI: 10.3389/fmed.2022.1052540] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Accepted: 12/08/2022] [Indexed: 01/07/2023] Open
Abstract
Ferroptosis, a new type of cell death, is mainly characterized by intracellular iron accumulation and lipid peroxidation. The complex regulatory network of iron metabolism, lipid metabolism, amino acid metabolism, p53-related signaling, and Nrf2-related signaling factors is involved in the entire process of ferroptosis. It has been reported that ferroptosis is involved in the pathogenesis of neurological diseases, cancer, and ischemia-reperfusion injury. Recent studies found that ferroptosis is closely related to the pathogenesis of COPD, which, to some extent, indicates that ferroptosis is a potential therapeutic target for COPD. This article mainly discusses the related mechanisms of ferroptosis, including metabolic regulation and signaling pathway regulation, with special attention to its role in the pathogenesis of COPD, aiming to provide safe and effective therapeutic targets for chronic airway inflammatory diseases.
Collapse
Affiliation(s)
- Dandan Meng
- Department of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chengfeng Zhu
- Department of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ruixue Jia
- Department of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zongxin Li
- Department of Second Department of Haematology, Jinan Haematology Hospital, Jinan, China
| | - Wantao Wang
- Department of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China,*Correspondence: Wantao Wang ✉
| | - Suhua Song
- Department of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China,Suhua Song ✉
| |
Collapse
|
34
|
Alharbi KS, Alshehri SM, Alenezi SK. Epigenetic Optimization in Chronic Obstructive Pulmonary Disease (COPD). TARGETING EPIGENETICS IN INFLAMMATORY LUNG DISEASES 2023:99-110. [DOI: 10.1007/978-981-99-4780-5_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
35
|
Zhu X, Chen X, Qiu L, Zhu J, Wang J. Norcantharidin induces ferroptosis via the suppression of NRF2/HO-1 signaling in ovarian cancer cells. Oncol Lett 2022; 24:359. [PMID: 36168316 PMCID: PMC9478624 DOI: 10.3892/ol.2022.13479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 05/19/2022] [Indexed: 11/05/2022] Open
Abstract
Increasing evidence has indicated a crucial role of ferroptosis in ovarian cancer (OC). Norcantharidin (NCTD), a normethyl compound of cantharidin, is extensively used in clinical practice as an optional anticancer drug. However, whether NCTD leads to ferroptosis in OC has not been previously explored, at least to the best of our knowledge. In the present study, the effect of NCTD on SKOV3 and OVCAR-3 cells was evaluated. The experimental data of the present study revealed that NCTD significantly suppressed SKOV3 and OVCAR-3 cell viability in a concentration- and time-dependent manner. The results of Cell Counting Kit-8 assay revealed that NCTD treatment decreased SKOV3 and OVCAR-3 cell viability. In comparison, pre-incubation with ferrostatin-1 (Fer-1) significantly reversed the NCTD-induced reduction in SKOV3 and OVCAR-3 cell viability; however, no changes in cell viability were observed when the SKOV3 and OVCAR-3 cells were treated with NCTD, in combination with the apoptosis inhibitor, Z-VAD-FMK, the ferroptosis inhibitor, necrostatin-1, and the autophagy inhibitor, 3-methyladenine. Additionally, it was observed that NCTD markedly enhanced reactive oxygen species production and malondialdehyde and ferrous ion levels in the SKOV3 and OVCAR-3 cells; however, pre-incubation with Fer-1 abolished these effects. Flow cytometry also demonstrated a significant increase in cell death following treatment of the SKOV3 and OVCAR-3 cells with NCTD; however, pre-incubation with Fer-1 also reversed these effects. In vivo experiments demonstrated that NCTD significantly reduced tumor volume and weight. More importantly, it was revealed that nuclear factor erythroid 2-related factor 2 (NRF2), heme oxygenase 1 (HO-1), glutathione peroxidase 4 (GPX4) and solute carrier family 7 member 11 (xCT) expression levels were significantly decreased following NCTD treatment. Collectively, NCTD may represent a potent anticancer agent in OC cells, and NCTD-induced ferroptotic cell death may be achieved by inhibiting the NRF2/HO-1/GPX4/xCT axis.
Collapse
Affiliation(s)
- Xiaoyan Zhu
- Department of Obstetrics and Gynecology, Jianhu Hospital Affiliated to Nantong University, Yancheng, Jiangsu 224700, P.R. China
| | - Xiaohong Chen
- Department of Gynecology, People's Hospital of Gansu Province, Lanzhou, Gansu 730000, P.R. China
| | - Longshan Qiu
- Department of Obstetrics and Gynecology, Jianhu Hospital Affiliated to Nantong University, Yancheng, Jiangsu 224700, P.R. China
| | - Jianhua Zhu
- Department of Obstetrics and Gynecology, Jianhu Hospital Affiliated to Nantong University, Yancheng, Jiangsu 224700, P.R. China
| | - Jiancai Wang
- Department of Obstetrics and Gynecology, Jianhu Hospital Affiliated to Nantong University, Yancheng, Jiangsu 224700, P.R. China
| |
Collapse
|
36
|
Wang S, Huo J, Wei Y, Huan M, Luo Z, Li M, Wen M, Zhong X, He Z, Ma N, Qiu J, Tang X. Effect of erythromycin on the ultrastructure of human macrophages exposed to cigarette smoke extract in vitro. Ultrastruct Pathol 2022; 46:303-312. [PMID: 35686365 DOI: 10.1080/01913123.2022.2060395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 03/25/2022] [Accepted: 03/28/2022] [Indexed: 10/18/2022]
Abstract
Macrophages serve an active role in the pathophysiology of chronic obstructive pulmonary disease (COPD). Erythromycin (EM) has been verified as an effective treatment for COPD. However, there are few studies on the effect of EM on the ultrastructure of macrophages exposed to cigarette smoke extract (CSE). In the present study, human macrophages were randomly divided into three groups: The control, CSE and the CSE+EM group, using electron microscopy, the effect of EM was evaluated by comparing the ultrastructural changes between these groups. The macrophages were additionally divided into a further four groups: The control, CSE, CSE+EM 24 h and CSE+EM 48 h groups. The generation of reactive oxygen species (ROS) in each group was evaluated by detecting fluorescence intensity. It was observed that the cellular ultrastructure of the CSE group exhibited abnormal changes, though this effect was reversed back to the level of the control in the CSE+EM group. Compared with the control group, the ROS expression level was significantly increased in the CSE group (P < .05); however, compared with the CSE group, the ROS concentration was decreased in the CSE+EM 24 h (P < .05) and CSE+EM 48 h groups (P < .05), though this was more apparent in the EM 48 h group. It was concluded that EM protects human macrophages against CSE. Moreover, it was hypothesized that EM may reduce the symptoms of patients with COPD by protecting the macrophage ultrastructure from the effects of CSE, resulting in the decreased generation of ROS, inhibiting autophagy and reducing endoplasmic reticulum stress.
Collapse
Affiliation(s)
- Shaoshuang Wang
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, GX, China
| | - Jianjun Huo
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, GX, China
| | - Yanlin Wei
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, GX, China
| | - Mei Huan
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, GX, China
| | - Zhouling Luo
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, GX, China
| | - Meihua Li
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, GX, China
| | - Mingzhi Wen
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, GX, China
| | - Xiaoning Zhong
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, GX, China
| | - Zhiyi He
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, GX, China
| | - Nan Ma
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, GX, China
| | - Jufeng Qiu
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, GX, China
| | - Xiaojuan Tang
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, GX, China
| |
Collapse
|
37
|
Yang YC, Zhang MY, Liu JY, Jiang YY, Ji XL, Qu YQ. Identification of Ferroptosis-Related Hub Genes and Their Association with Immune Infiltration in Chronic Obstructive Pulmonary Disease by Bioinformatics Analysis. Int J Chron Obstruct Pulmon Dis 2022; 17:1219-1236. [PMID: 35637927 PMCID: PMC9148178 DOI: 10.2147/copd.s348569] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 04/30/2022] [Indexed: 12/15/2022] Open
Abstract
Purpose Ferroptosis and immune infiltration are involved in the pathogenesis of chronic obstructive pulmonary disease (COPD). We aim to identify ferroptosis-related hub genes and analyze their association with immune infiltration in COPD through bioinformatics methods. Materials and Methods The mRNA microarray data of GSE38974 were downloaded from Gene Expression Omnibus to obtain differentially expressed genes (DEGs). The DEGs were intersected with ferroptosis-related genes (FRGs) from FerrDb to obtain differentially expressed FRGs. GO and KEGG enrichment and protein–protein interaction (PPI) analyses of differentially expressed FRGs were conducted in R software and STRING database. The key module and hub genes were screened by Cytoscape software. MiRNAs, transcription factors and signal molecules were predicted in miRNet and NetworkAnalyst. The disease correlation in the Comparative Toxicomics Database (CTD) and the receiver operating characteristic (ROC) curves of hub genes were analyzed. Immune infiltration was evaluated by CIBERSORT algorithm. Spearman correlation analyses were conducted between hub genes and differentially infiltrated immune cells. Results Fifteen differentially expressed FRGs were identified, which were enriched in some terms involving airway inflammatory responses and structural remodeling. Five hub genes were selected including HIF1A, IL6, PTGS2, CDKN1A and ATM. Inference scores in CTD indicated their association with COPD. Two miRNAs, five transcription factors and one signal molecule were predicted. The combination of hub genes could be a fine diagnostic indicator of COPD (AUC: 0.981, CI: 0.940-1.000). Immune infiltration evaluation showed that monocytes and M0 macrophages were upregulated in COPD lung tissues, while CD8 T cells, activated NK cells, M2 macrophages, resting dendritic cells and resting mast cells were downregulated. The hub genes were significantly associated with differentially infiltrated immune cells. Conclusion We identified five ferroptosis-related hub genes (HIF1A, IL6, PTGS2, CDKN1A and ATM) in COPD, and found that they may influence the pathogenesis of COPD by regulating ferroptosis and thus affecting infiltrating immune cells.
Collapse
Affiliation(s)
- Yi-Can Yang
- Department of Pulmonary and Critical Care Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University; Shandong Key Laboratory of Infectious Respiratory Diseases, Jinan, People's Republic of China
| | - Meng-Yu Zhang
- Department of Pulmonary and Critical Care Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University; Shandong Key Laboratory of Infectious Respiratory Diseases, Jinan, People's Republic of China
| | - Jian-Yu Liu
- Department of Pulmonary and Critical Care Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University; Shandong Key Laboratory of Infectious Respiratory Diseases, Jinan, People's Republic of China
| | - Yuan-Yuan Jiang
- Department of Pulmonary and Critical Care Medicine, Qilu Hospital of Shandong University; Shandong Key Laboratory of Infectious Respiratory Diseases, Jinan, People's Republic of China
| | - Xiu-Li Ji
- Department of Pulmonary Disease, Jinan Traditional Chinese Medicine Hospital, Jinan, People's Republic of China
| | - Yi-Qing Qu
- Department of Pulmonary and Critical Care Medicine, Qilu Hospital of Shandong University; Shandong Key Laboratory of Infectious Respiratory Diseases, Jinan, People's Republic of China
| |
Collapse
|