Volume 41 Issue 9
Sep.  2025
Turn off MathJax
Article Contents
Zhang ZJ,Li DW,Liu LW,et al.Roles and mechanism of mitophagy in myocardial injury in mice following delayed resusafter severe burns[J].Chin J Burns Wounds,2025,41(9):847-856.DOI: 10.3760/cma.j.cn501225-20250514-00226.
Citation: Zhang ZJ,Li DW,Liu LW,et al.Roles and mechanism of mitophagy in myocardial injury in mice following delayed resusafter severe burns[J].Chin J Burns Wounds,2025,41(9):847-856.DOI: 10.3760/cma.j.cn501225-20250514-00226.

Roles and mechanism of mitophagy in myocardial injury in mice following delayed resuscitation after severe burns

doi: 10.3760/cma.j.cn501225-20250514-00226
Funds:

National Key Research and Development Program 2024YFC3016604

More Information
  •   Objective  To explore the roles and mechanism of mitophagy in myocardial injury in mice following delayed resuscitation after severe burns.  Methods  This study was an experimental study. Forty-five 6-8-week-old male C57BL/6J mice were divided into sham injury group, 0 h post-injury rehydration group, 3 h post-injury rehydration group, 6 h post-injury rehydration group, and no post-injury rehydration group, with 9 mice in each group, according to the random number table (the same grouping method below). The mice in sham injury group were subjected to a sham injury, while the mice in the remaining 4 groups were subjected to full-thickness scald (hereinafter referred to as burn) of 30% total body surface area. The mice in 0 h post-injury rehydration group, 3 h post-injury rehydration group, and 6 h post-injury rehydration group received rehydration starting from 0 (immediately), 3, and 6 h post-injury, respectively. The mice in sham injury group and no post-injury rehydration group did not receive rehydration after injury. At 12 h post-injury, the serum levels of lactate dehydrogenase (LDH) and creatine kinase isoenzyme (CK-MB) were detected using an automated biochemical analyzer. Myocardial histopathological scoring was performed using Kishimoto scores after hematoxylin-eosin staining. The protein expressions of mitophagy-related proteins, including translocase of outer mitochondrial membrane 20 (TOM20), microtubule-associated protein 1 light chain 3B (LC3B), P62, and Beclin-1 in myocardial tissue were detected by Western blotting, and the ratio of LC3B-Ⅱ/LC3B-Ⅰ was calculated. Twenty-seven 6-8-week-old male C57BL/6J mice were divided into 6 h post-injury rehydration alone group, 6 h post-injury rehydration+3-methyladenine (3-MA) group, and 6 h post-injury rehydration+rapamycin group, with 9 mice in each group. One week before burn modeling, mice in 6 h post-injury rehydration+3-MA group and 6 h post-injury rehydration+rapamycin group were injected intraperitoneally with the mitophagy inhibitor 3-MA or mitophagy activator rapamycin. All the mice in 3 groups received rehydration starting from 6 h post-injury following severe burns. At 12 h post-injury, the serum levels of LDH and CK-MB were detected, myocardial histopathological scoring was performed, and the protein expressions of mitochondrial autophagy-related proteins were detected as before. The mitochondrial ultrastructure changes in myocardial tissue were observed under a transmission electron microscope after uranium-lead double staining, and Flameng scoring was performed.  Results  At 12 h post-injury, compared with those in 0 h post-injury rehydration group the serum levels of LDH and CK-MB in mice in 6 h post-injury rehydration group and no post-injury rehydration group were significantly increased (P<0.05). At 12 h post-injury, compared with that in 0 h post-injury rehydration group, the myocardial histopathological score in mice in sham injury group was significantly decreased (P<0.05), and the myocardial histopathological scores in mice in 3 h post-injury rehydration group, 6 h post-injury rehydration group, and no post-injury rehydration group were significantly increased (P<0.05). At 12 h post-injury, compared with those in 0 h post-injury rehydration group, the protein expression of Beclin-1 was significantly decreased (P<0.05), while the protein expressions of TOM20 and P62 were significantly increased (P<0.05) in myocardial tissue of mice in sham injury group; the protein expression of Beclin-1 was significantly increased (P<0.05), while the protein expressions of TOM20 and P62 were significantly decreased (P<0.05) in myocardial tissue of mice in 3 h post-injury rehydration group; the protein expressions of Beclin-1 and ratios of LC3B-Ⅱ/LC3B-Ⅰwere significantly increased (P<0.05), while the protein expressions of TOM20 and P62 were significantly decreased (P<0.05) in myocardial tissue of mice in 6 h post-injury rehydration group and no post-injury rehydration group. At 12 h post-injury, compared with those in 6 h post-injury rehydration alone group, the serum levels of LDH and CK-MB in mice in 6 h post-injury rehydration+3-MA group were significantly decreased (P<0.05), while the serum levels of LDH and CK-MB in mice in 6 h post-injury rehydration+rapamycin group were significantly increased (P<0.05). At 12 h post-injury, compared with that in 6 h post-injury rehydration alone group, the myocardial histopathological score in mice in 6 h post-injury rehydration+3-MA group was significantly decreased (P<0.05), while the myocardial histopathological score in mice in 6 h post-injury rehydration+rapamycin group was significantly increased (P<0.05). At 12 h post-injury, compared with those in 6 h post-injury rehydration alone group, the protein expression of Beclin-1 and ratio of LC3B-Ⅱ/LC3B-Ⅰ were significantly decreased (P<0.05), while the protein expressions of TOM20 and P62 were significantly increased (P<0.05) in myocardial tissue of mice in 6 h post-injury rehydration+3-MA group; the protein expression of Beclin-1 and ratio of LC3B-Ⅱ/LC3B-Ⅰwere significantly increased (P<0.05), while the protein expressions of TOM20 and P62 were significantly decreased (P<0.05) in myocardial tissue of mice in 6 h post-injury rehydration+rapamycin group. At 12 h post-injury, compared with 2.67±0.11 in 6 h post-injury rehydration alone group, the mitochondrial Flameng score (2.07±0.11) in myocardial tissue of mice in 6 h post-injury rehydration+3-MA group was significantly decreased (P<0.05), while the mitochondrial Flameng score (3.60±0.20) in myocardial tissue of mice in 6 h post-injury rehydration+rapamycin group was significantly increased (P<0.05).  Conclusions  Delayed rehydration leads to myocardial injury and excessive activation of mitophagy in mice with severe burns. The inhibition of mitophagy can mitigate myocardial injury caused by delayed rehydration after severe burns.

     

  • loading
  • [1]
    GreenhalghDG. Management of burns[J]. N Engl J Med, 2019, 380(24): 2349-2359. DOI: 10.1056/NEJMra1807442.
    [2]
    Radzikowska-BüchnerE, ŁopuszyńskaI, FliegerW, et al. An overview of recent developments in the management of burn injuries[J]. Int J Mol Sci, 2023, 24(22): 16357. DOI: 10.3390/ijms242216357.
    [3]
    BurgessM, ValderaF, VaronD, et al. The immune and regenerative response to burn injury[J]. Cells, 2022, 11(19): 3073. DOI: 10.3390/cells11193073.
    [4]
    FoldiV,CsontosC,BogarL,et al.Effects of fluid resuscitation methods on burn trauma-induced oxidative stress[J].J Burn Care Res,2009,30(6):957-966.DOI: 10.1097/BCR.0b013e3181bfb75e.
    [5]
    EndorfFW,DriesDJ.Burn resuscitation[J].Scand J Trauma Resusc Emerg Med,2011,19:69.DOI: 10.1186/1757-7241-19-69.
    [6]
    LiA,GaoM,LiuB,et al.Mitochondrial autophagy: molecular mechanisms and implications for cardiovascular disease[J].Cell Death Dis,2022,13(5):444.DOI: 10.1038/s41419-022-04906-6.
    [7]
    SzczesnyB, BrunyánszkiA, AhmadA, et al. Time-dependent and organ-specific changes in mitochondrial function, mitochondrial DNA integrity, oxidative stress and mononuclear cell infiltration in a mouse model of burn injury[J]. PLoS One, 2015, 10(12): e0143730. DOI: 10.1371/journal.pone.0143730.
    [8]
    Marín-GarcíaJ,GoldenthalMJ.Understanding the impact of mitochondrial defects in cardiovascular disease: a review[J].J Card Fail,2002,8(5):347-361.DOI: 10.1054/jcaf.2002.127774.
    [9]
    PicklesS, VigiéP, YouleRJ. Mitophagy and quality control mechanisms in mitochondrial maintenance[J]. Curr Biol, 2018, 28(4): R170-R185. DOI: 10.1016/j.cub.2018.01.004.
    [10]
    WangM, ScottSR, KoniarisLG, et al. Pathological responses of cardiac mitochondria to burn trauma[J]. Int J Mol Sci, 2020, 21(18): 6655. DOI: 10.3390/ijms21186655.
    [11]
    LiuC,LiuY,ChenH,et al.Myocardial injury: where inflammation and autophagy meet[J/OL].Burns Trauma,2023,11:tkac062[2025-05-14].https://pubmed.ncbi.nlm.nih.gov/36873283/.DOI: 10.1093/burnst/tkac062.
    [12]
    LiYZ,WuXD,LiuXH,et al.Mitophagy imbalance in cardiomyocyte ischaemia/reperfusion injury[J].Acta Physiol (Oxf),2019,225(4):e13228.DOI: 10.1111/apha.13228.
    [13]
    ChenX,ZhuY,WeiY,et al.Glutamine alleviates intestinal injury in a murine burn sepsis model by maintaining intestinal intraepithelial lymphocyte homeostasis[J].Eur J Pharmacol,2023,940:175480.DOI: 10.1016/j.ejphar.2022.175480.
    [14]
    KishimotoC,KawamataH,SakaiS,et al.Enhanced production of macrophage inflammatory protein 2 (MIP-2) by in vitro and in vivo infections with encephalomyocarditis virus and modulation of myocarditis with an antibody against MIP-2[J].J Virol,2001,75(3):1294-1300.DOI: 10.1128/JVI.75.3.1294-1300.2001.
    [15]
    DiS, WangZ, HuW, et al. The protective effects of melatonin against LPS-induced septic myocardial injury: a potential role of AMPK-mediated autophagy[J]. Front Endocrinol (Lausanne), 2020, 11: 162. DOI: 10.3389/fendo.2020.00162.
    [16]
    FlamengW,BorgersM,DaenenW,et al.Ultrastructural and cytochemical correlates of myocardial protection by cardiac hypothermia in man[J].J Thorac Cardiovasc Surg,1980,79(3):413-424.
    [17]
    GuilloryAN, ClaytonRP, HerndonDN, et al. Cardiovascular dysfunction following burn injury: what we have learned from rat and mouse models[J]. Int J Mol Sci, 2016, 17(1): 53. DOI: 10.3390/ijms17010053.
    [18]
    陈诺, 席毛毛, 阮琼芳, 等.大面积烧伤患者发生早期心肌损伤的危险因素及早期心肌损伤对预后的影响[J].中华烧伤与创面修复杂志,2023,39(5):417-423.DOI: 10.3760/cma.j.cn501225-20230308-00074.
    [19]
    黄跃生.严重烧伤早期心肌损害机制及临床意义的再认识[J].中华烧伤杂志,2016,32(5):257-259.DOI: 10.3760/cma.j.issn.1009-2587.2016.05.001.
    [20]
    FOZZARDHA.Myocardial injury in burn shock[J].Ann Surg,1961,154(1):113-119.DOI: 10.1097/00000658-196107000-00017.
    [21]
    YuT,LiuH,GaoM,et al.Dexmedetomidine regulates exosomal miR-29b-3p from macrophages and alleviates septic myocardial injury by promoting autophagy in cardiomyocytes via targeting glycogen synthase kinase 3β[J/OL].Burns Trauma,2024,12:tkae042[2025-05-14]. https://pubmed.ncbi.nlm.nih.gov/39502342/.DOI: 10.1093/burnst/tkae042.
    [22]
    LiF,HuH,WuL,et al.Ablation of mitophagy receptor FUNDC1 accentuates septic cardiomyopathy through ACSL4-dependent regulation of ferroptosis and mitochondrial integrity[J].Free Radic Biol Med,2024,225:75-86.DOI: 10.1016/j.freeradbiomed.2024.09.039.
    [23]
    HuangB, XieL, KeM, et al. Programmed release METTL3-14 inhibitor microneedle protects myocardial function by reducing Drp1 m6A modification-mediated mitochondrial fission[J]. ACS Appl Mater Interfaces, 2023, 15(40): 46583-46597. DOI: 10.1021/acsami.3c06318.
    [24]
    WenJJ,MobliK,RontoyanniVG,et al.Nuclear factor erythroid 2-related factor 2 activation and burn-induced cardiac dysfunction[J].J Am Coll Surg,2022,234(4):660-671.DOI: 10.1097/XCS.0000000000000119.
    [25]
    HuangY,WangS,ZhouJ,et al.IRF1-mediated downregulation of PGC1α contributes to cardiorenal syndrome type 4[J].Nat Commun,2020,11(1):4664.DOI: 10.1038/s41467-020-18519-0.
    [26]
    WangB,NieJ,WuL,et al.AMPKα2 protects against the development of heart failure by enhancing mitophagy via PINK1 phosphorylation[J].Circ Res,2018,122(5):712-729.DOI: 10.1161/CIRCRESAHA.117.312317.
    [27]
    ChouchaniET,PellVR,GaudeE,et al.Ischaemic accumulation of succinate controls reperfusion injury through mitochondrial ROS[J].Nature,2014,515(7527):431-435.DOI: 10.1038/nature13909.
    [28]
    BeaulantA,DiaM,PillotB,et al.Endoplasmic reticulum-mitochondria miscommunication is an early and causal trigger of hepatic insulin resistance and steatosis[J].J Hepatol,2022,77(3):710-722.DOI: 10.1016/j.jhep.2022.03.017.
    [29]
    LiL,FuW,GongX,et al.The role of G protein-coupled receptor kinase 4 in cardiomyocyte injury after myocardial infarction[J].Eur Heart J,2021,42(14):1415-1430.DOI: 10.1093/eurheartj/ehaa878.
    [30]
    XiaoR,TengM,ZhangQ,et al.Myocardial autophagy after severe burn in rats[J].PLoS One,2012,7(6):e39488.DOI: 10.1371/journal.pone.0039488.
    [31]
    AjoolabadyA,AghanejadA,BiY,et al.Enzyme-based autophagy in anti-neoplastic management: from molecular mechanisms to clinical therapeutics[J].Biochim Biophys Acta Rev Cancer,2020,1874(1):188366.DOI: 10.1016/j.bbcan.2020.188366.
    [32]
    MoralesPE,Arias-DuránC,Ávalos-GuajardoY,et al.Emerging role of mitophagy in cardiovascular physiology and pathology[J].Mol Aspects Med,2020,71:100822.DOI: 10.1016/j.mam.2019.09.006.
    [33]
    WenJJ, CumminsCB, RadhakrishnanRS. Burn-induced cardiac mitochondrial dysfunction via interruption of the PDE5A-cGMP-PKG pathway[J]. Int J Mol Sci, 2020, 21(7): 2350. DOI: 10.3390/ijms21072350.
    [34]
    XueY, FuW, YuP, et al. Ginsenoside Rc alleviates myocardial ischemia-reperfusion injury by reducing mitochondrial oxidative stress and apoptosis: role of SIRT1 activation[J]. J Agric Food Chem, 2023, 71(3): 1547-1561. DOI: 10.1021/acs.jafc.2c06926.
    [35]
    KubliDA,ZhangX,LeeY,et al.Parkin protein deficiency exacerbates cardiac injury and reduces survival following myocardial infarction[J].J Biol Chem,2013,288(2):915-926.DOI: 10.1074/jbc.M112.411363.
    [36]
    MaC,YangZ,WangJ,et al.Interleukin-1β-stimulated macrophage-derived exosomes improve myocardial injury in sepsis via regulation of mitochondrial homeostasis: experimental research[J].Int J Surg,2025,111(1):283-301.DOI: 10.1097/JS9.0000000000001915.
    [37]
    LiX,LuoW,TangY,et al.Semaglutide attenuates doxorubicin-induced cardiotoxicity by ameliorating BNIP3-Mediated mitochondrial dysfunction[J].Redox Biol,2024,72:103129.DOI: 10.1016/j.redox.2024.103129.
    [38]
    YaoX,WiggintonJG,MaassDL,et al.Estrogen-provided cardiac protection following burn trauma is mediated through a reduction in mitochondria-derived DAMPs[J].Am J Physiol Heart Circ Physiol,2014,306(6):H882-894.DOI: 10.1152/ajpheart.00475.2013.
    [39]
    ZepedaR,KuzmicicJ,ParraV,et al.Drp1 loss-of-function reduces cardiomyocyte oxygen dependence protecting the heart from ischemia-reperfusion injury[J].J Cardiovasc Pharmacol,2014,63(6):477-487.DOI: 10.1097/FJC.0000000000000071.
    [40]
    HoshinoA,MitaY,OkawaY,et al.Cytosolic p53 inhibits Parkin-mediated mitophagy and promotes mitochondrial dysfunction in the mouse heart[J].Nat Commun,2013,4:2308.DOI: 10.1038/ncomms3308.
    [41]
    WangSH,ZhuXL,WangF,et al.LncRNA H19 governs mitophagy and restores mitochondrial respiration in the heart through Pink1/Parkin signaling during obesity[J].Cell Death Dis,2021,12(6):557.DOI: 10.1038/s41419-021-03821-6.
    [42]
    da Silva RosaSC,MartensMD,FieldJT,et al.BNIP3L/Nix-induced mitochondrial fission, mitophagy, and impaired myocyte glucose uptake are abrogated by PRKA/PKA phosphorylation[J].Autophagy,2021,17(9):2257-2272.DOI: 10.1080/15548627.2020.1821548.
  • 加载中

Catalog

    通讯作者: 陈斌, bchen63@163.com
    • 1. 

      沈阳化工大学材料科学与工程学院 沈阳 110142

    1. 本站搜索
    2. 百度学术搜索
    3. 万方数据库搜索
    4. CNKI搜索

    Figures(5)  / Tables(2)

    Article Metrics

    Article views (2150) PDF downloads(9) Cited by()
    Proportional views
    Related

    /

    DownLoad:  Full-Size Img  PowerPoint
    Return
    Return