14
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      METTL3‐mediated N6‐methyladenosine exacerbates ferroptosis via m6A‐IGF2BP2‐dependent mitochondrial metabolic reprogramming in sepsis‐induced acute lung injury

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Neutrophil extracellular traps (NETs), released by polymorphonuclear neutrophils (PMNs), exert a robust antimicrobial function in infectious diseases such as sepsis. NETs also contribute to the pathogenesis and exacerbation of sepsis. Although the lung is highly vulnerable to infections, few studies have explored the role of NETs in sepsis‐induced acute lung injury (SI‐ALI). We demonstrate that NETs induce SI‐ALI via enhanced ferroptosis in alveolar epithelial cells. Our findings reveal that the excessive release of NETs in patients and mice with SI‐ALI is accompanied by upregulation of ferroptosis depending on METTL3‐induced m6A modification of hypoxia‐inducible factor‐1α (HIF‐1α) and subsequent mitochondrial metabolic reprogramming. In addition to conducting METTL3 overexpression and knockdown experiments in vitro, we also investigated the impact of ferroptosis on SI‐ALI caused by NETs in a caecum ligation and puncture (CLP)‐induced SI‐ALI model using METTL3 condition knockout (CKO) mice and wild‐type mice. Our results indicate the crucial role of NETs in the progression of SI‐ALI via NET‐activated METTL3 m6A‐IGF2BP2‐dependent m6A modification of HIF‐1α, which further contributes to metabolic reprogramming and ferroptosis in alveolar epithelial cells.

          Abstract

          Highlights

          • NETs induce ferroptosis in alveolar epithelial cells via activation of METTL3‐mediated m6A modification.

          • METTL3 induces HIF‐1α upregulation via m6A‐IGF2BP2‐dependent mechanism to exacerbate ferroptosis.

          • METTL3 induces enhanced glycolysis and decreased oxidative phosphorylation in alveolar epithelial cells.

          • Inhibiting NET formation via peptidyl arginine deiminase 4 knockout attenuates ferroptosis and sepsis‐associated lung damage in mice.

          Related collections

          Most cited references48

          • Record: found
          • Abstract: found
          • Article: not found

          Ferroptosis: mechanisms, biology and role in disease

          The research field of ferroptosis has seen exponential growth over the past few years, since the term was coined in 2012. This unique modality of cell death, driven by iron-dependent phospholipid peroxidation, is regulated by multiple cellular metabolic pathways, including redox homeostasis, iron handling, mitochondrial activity and metabolism of amino acids, lipids and sugars, in addition to various signalling pathways relevant to disease. Numerous organ injuries and degenerative pathologies are driven by ferroptosis. Intriguingly, therapy-resistant cancer cells, particularly those in the mesenchymal state and prone to metastasis, are exquisitely vulnerable to ferroptosis. As such, pharmacological modulation of ferroptosis, via both its induction and its inhibition, holds great potential for the treatment of drug-resistant cancers, ischaemic organ injuries and other degenerative diseases linked to extensive lipid peroxidation. In this Review, we provide a critical analysis of the current molecular mechanisms and regulatory networks of ferroptosis, the potential physiological functions of ferroptosis in tumour suppression and immune surveillance, and its pathological roles, together with a potential for therapeutic targeting. Importantly, as in all rapidly evolving research areas, challenges exist due to misconceptions and inappropriate experimental methods. This Review also aims to address these issues and to provide practical guidelines for enhancing reproducibility and reliability in studies of ferroptosis. Finally, we discuss important concepts and pressing questions that should be the focus of future ferroptosis research.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            COVID-19 and its implications for thrombosis and anticoagulation

            Severe acute respiratory syndrome coronavirus 2, coronavirus disease 2019 (COVID-19)-induced infection can be associated with a coagulopathy, findings consistent with infection-induced inflammatory changes as observed in patients with disseminated intravascular coagulopathy (DIC). The lack of prior immunity to COVID-19 has resulted in large numbers of infected patients across the globe and uncertainty regarding management of the complications that arise in the course of this viral illness. The lungs are the target organ for COVID-19; patients develop acute lung injury that can progress to respiratory failure, although multiorgan failure can also occur. The initial coagulopathy of COVID-19 presents with prominent elevation of D-dimer and fibrin/fibrinogen-degradation products, whereas abnormalities in prothrombin time, partial thromboplastin time, and platelet counts are relatively uncommon in initial presentations. Coagulation test screening, including the measurement of D-dimer and fibrinogen levels, is suggested. COVID-19–associated coagulopathy should be managed as it would be for any critically ill patient, following the established practice of using thromboembolic prophylaxis for critically ill hospitalized patients, and standard supportive care measures for those with sepsis-induced coagulopathy or DIC. Although D-dimer, sepsis physiology, and consumptive coagulopathy are indicators of mortality, current data do not suggest the use of full-intensity anticoagulation doses unless otherwise clinically indicated. Even though there is an associated coagulopathy with COVID-19, bleeding manifestations, even in those with DIC, have not been reported. If bleeding does occur, standard guidelines for the management of DIC and bleeding should be followed.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Acute respiratory distress syndrome

              The acute respiratory distress syndrome (ARDS) is a common cause of respiratory failure in critically ill patients and is defined by the acute onset of noncardiogenic pulmonary oedema, hypoxaemia and the need for mechanical ventilation. ARDS occurs most often in the setting of pneumonia, sepsis, aspiration of gastric contents or severe trauma and is present in ~10% of all patients in intensive care units worldwide. Despite some improvements, mortality remains high at 30–40% in most studies. Pathological specimens from patients with ARDS frequently reveal diffuse alveolar damage, and laboratory studies have demonstrated both alveolar epithelial and lung endothelial injury, resulting in accumulation of protein-rich inflammatory oedematous fluid in the alveolar space. Diagnosis is based on consensus syndromic criteria, with modifications for under-resourced settings and in paediatric patients. Treatment focuses on lung-protective ventilation; no specific pharmacotherapies have been identified. Long-term outcomes of patients with ARDS are increasingly recognized as important research targets, as many patients survive ARDS only to have ongoing functional and/or psychological sequelae. Future directions include efforts to facilitate earlier recognition of ARDS, identifying responsive subsets of patients and ongoing efforts to understand fundamental mechanisms of lung injury to design specific treatments.
                Bookmark

                Author and article information

                Contributors
                eliteromes@126.com
                zhpsqxt@126.com
                jcata@mdanderson.org
                miaochangh@163.com
                Journal
                Clin Transl Med
                Clin Transl Med
                10.1002/(ISSN)2001-1326
                CTM2
                Clinical and Translational Medicine
                John Wiley and Sons Inc. (Hoboken )
                2001-1326
                15 September 2023
                September 2023
                : 13
                : 9 ( doiID: 10.1002/ctm2.v13.9 )
                : e1389
                Affiliations
                [ 1 ] Department of Anesthesiology Zhongshan Hospital Fudan University Shanghai China
                [ 2 ] Shanghai Key Laboratory of Perioperative Stress and Protection Shanghai China
                [ 3 ] Department of Anesthesiology Shanghai Medical College Fudan University Shanghai China
                [ 4 ] Department of Cardiac Surgery Ohio State University Columbus Ohio USA
                [ 5 ] Department of Biomedical Engineering Ohio State University Columbus Ohio USA
                [ 6 ] Department of Anesthesiology Affiliated Hospital of Xuzhou Medical University Xuzhou China
                [ 7 ] Department of Anesthesiology and Perioperative Medicine University of Texas‐MD Anderson Cancer Center Houston Texas USA
                [ 8 ] Anesthesiology and Surgical Oncology Research Group Houston Texas USA
                Author notes
                [*] [* ] Correspondence

                Changhong Miao and Hao Zhang, Department of Anesthesiology, Zhongshan Hospital, Fudan University, 180# Feng‐Lin Road, Shanghai, China.

                Email: miaochangh@ 123456163.com and eliteromes@ 123456126.com

                Zhiping Wang, Department of Anesthesiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.

                Email: zhpsqxt@ 123456126.com

                Juan P. Cata, Department of Anesthesiology and Perioperative Medicine, University of Texas‐MD Anderson Cancer Center, Houston, TX, USA.

                Email: jcata@ 123456mdanderson.org

                Author information
                https://orcid.org/0000-0003-3516-8308
                Article
                CTM21389
                10.1002/ctm2.1389
                10504453
                37715457
                24bb3536-d210-4794-b94f-03e6b3e7cab9
                © 2023 The Authors. Clinical and Translational Medicine published by John Wiley & Sons Australia, Ltd on behalf of Shanghai Institute of Clinical Bioinformatics.

                This is an open access article under the terms of the http://creativecommons.org/licenses/by/4.0/ License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.

                History
                : 13 August 2023
                : 10 March 2023
                : 18 August 2023
                Page count
                Figures: 9, Tables: 0, Pages: 20, Words: 9361
                Funding
                Funded by: National Natural Science Foundation of China , doi 10.13039/501100001809;
                Award ID: 82102253
                Funded by: Natural Science Foundation of Shanghai , doi 10.13039/100007219;
                Award ID: 21ZR1413400
                Funded by: Shanghai Sailing Program
                Award ID: 21YF1406800
                Funded by: Shanghai Municipal 2021 “Science and Technology Innovation Action Plan”
                Award ID: 21JC1401400
                Categories
                Research Article
                Research Articles
                Custom metadata
                2.0
                September 2023
                Converter:WILEY_ML3GV2_TO_JATSPMC version:6.3.3 mode:remove_FC converted:16.09.2023

                Medicine
                ferroptosis,metabolic reprogramming,n6‐methylation,neutrophil extracellular traps,sepsis‐induced acute lung injury

                Comments

                Comment on this article