中文English
ISSN 1001-5256 (Print)
ISSN 2097-3497 (Online)
CN 22-1108/R
Volume 40 Issue 1
Jan.  2024
Turn off MathJax
Article Contents

Menaquinone-4 exerts a protective effect against carbon tetrachloride-induced acute liver injury in mice by alleviating ferroptosis

DOI: 10.12449/JCH240121
Research funding:

National Natural Science Foundation of China (82073566);

The Program of Excellent Young Talents in Universities of Anhui Province (gxyq2019014);

Clinical Pharmacy and Pharmacology (2020) ;

Anhui Public Health Clinical Center, Supported by North District Scientific Research and Cultivation Foundation of the First Affiliated Hospital of Anhui Medical University (2023YKJ14);

Anhui Public Health Clinical Center, Supported by North District Scientific Research and Cultivation Foundation of the First Affiliated Hospital of Anhui Medical University (2023YKJ06);

Anhui Public Health Clinical Center, Supported by North District Scientific Research and Cultivation Foundation of the First Affiliated Hospital of Anhui Medical University (2023YKJ11)

More Information
  • Corresponding author: WANG Jianqing, jianqingwang81@126.com (ORCID: 0000-0002-7935-9520)
  • Received Date: 2023-03-30
  • Accepted Date: 2023-05-29
  • Published Date: 2024-01-23
  •   Objective  To investigate whether menaquinone-4 (MK-4) can exert a protective effect against carbon tetrachloride (CCl4)-induced acute liver injury (ALI) in mice by alleviating ferroptosis.  Methods  After adaptive feeding, adult male ICR mice, aged 8 weeks, were divided into Control group, MK-4 group, CCl4 model group (6-hour, 12-hour, and 24-hour), and MK-4+CCl4 group (6-hour, 12-hour, and 24-hour), with 6 mice in each group. The mice in the Control group were given intraperitoneal injection of an equal dose of corn oil; the mice in the MK-4 group were given intraperitoneal injection of 40 mg/kg MK-4 solution, followed by an equal dose of corn oil after 1 hour; the mice in the MK-4+CCl4 group (6-hour, 12-hour, and 24-hour) were given intraperitoneal injection of 40 mg/kg MK-4 solution, and after 1 hour, the mice in this group and the CCl4 model group (6-hour, 12-hour, and 24-hour) were given intraperitoneal injection of 0.3 mL/kg CCl4 solution, with samples collected at 6, 12, and 24 hours. HE staining was used to observe the pathological changes of mouse liver; Prussian blue staining was used to observe iron accumulation in liver tissue; a biochemical analyzer was used to measure the serum levels of aspartate aminotransferase (AST) and alanine aminotransferase (ALT); related kits were used to measure the levels of tissue iron content and the oxidative stress indices malondialdehyde (MDA) and glutathione (GSH) in liver homogenate; RT-PCR was used to measure the expression levels of ferroptosis marker genes (acyl-CoA synthetase long-chain family member 4 [ACSL4], prostaglandin-endoperoxide synthase 2 [PTGS2], and glutathione peroxidase 4 [GPX4]) and iron metabolism-related genes (hemojuvelin [HJV], transferrin receptor 1 [TFR1], and ferroportin [FPN]), and Western blot was used to measure the protein expression level of GPX4. A one-way analysis of variance was used for comparison of continuous data between multiple groups, and the least significant difference t-test was used for further comparison between two groups.  Results  In the aging study, compared with the Control group, the CCl4 model group (6-hour, 12-hour, and 24-hour) had significant increases in liver weight coefficient and the serum levels of ALT and AST (all P<0.05), and HE staining also showed that liver injury gradually aggravated over time. Meanwhile, compared with the CCl4 model group (6-hour, 12-hour, and 24-hour), the MK-4+CCl4 (12-hour) group had significant reductions in liver weight coefficient and the serum levels of ALT and AST (all P<0.05), with a reduction in the necrotic area of liver tissue, and therefore, 12-hour mouse tissue samples were used for detection in the following study. Compared with the Control group, the CCl4 group had a significant increase in MDA and a significant reduction in GSH (both P<0.05), and compared with the CCl4 group, the MK-4+CCl4 group had a significant reduction in MDA and a significant increase in GSH (both P<0.05). Compared with the Control group, the CCl4 group had significant increases in the key ferroptosis indices ASCL4 and PTGS2 and a significant reduction in GPX4 (all P<0.05); compared with the CCl4 group, the MK-4+CCl4 group had significant reductions in the mRNA expression levels of ASCL4 and PTGS2 and a significant increase in the mRNA expression level of GPX4 (all P<0.05). Western blotting showed that compared with the Control group, the CCl4 group had a significant reduction in the protein expression level of GPX4 (P<0.05), and compared with the CCl4 group, the MK-4+CCl4 group had a significant increase in the protein expression level of GPX4 (P<0.05). Prussian blue staining showed that compared with the Control group, the CCl4 group had a significant increase in iron accumulation; after MK-4 intervention, compared with the CCl4 group, the MK-4+CCl4 group had a significant reduction in iron accumulation. As for the measurement of iron metabolism genes in mouse liver, compared with the Control group, the CCl4 group had a significant increase in iron content, significant reductions in the mRNA expression levels of FPN and HJV, and a significant increase in the mRNA expression level of TFR1 (all P<0.05); after protection with MK-4, there was a significant reduction in iron content, significant increases in the mRNA expression levels of FPN and HJV, and a significant reduction in the mRNA expression level of TFR1 (all P<0.05).  Conclusion  MK-4 intervention in advance can alleviate CCl4-induced ALI in mice, possibly by inhibiting ferroptosis and improving the expression of iron metabolism-related genes in mouse liver.

     

  • loading
  • [1]
    SHRESTHA DB, BUDHATHOKI P, SEDHAI YR, et al. N-acetyl cysteine versus standard of care for non-acetaminophen induced acute liver injury: A systematic review and meta-analysis[J]. Ann Hepatol, 2021, 24: 100340. DOI: 10.1016/j.aohep.2021.100340.
    [2]
    BHAKUNI GS, BEDI O, BARIWAL J, et al. Animal models of hepatotoxicity[J]. Inflamm Res, 2016, 65( 1): 13- 24. DOI: 10.1007/s00011-015-0883-0.
    [3]
    LIANG DG, MINIKES AM, JIANG XJ. Ferroptosis at the intersection of lipid metabolism and cellular signaling[J]. Mol Cell, 2022, 82( 12): 2215- 2227. DOI: 10.1016/j.molcel.2022.03.022.
    [4]
    JIANG XJ, STOCKWELL BR, CONRAD M. Ferroptosis: Mechanisms, biology and role in disease[J]. Nat Rev Mol Cell Biol, 2021, 22( 4): 266- 282. DOI: 10.1038/s41580-020-00324-8.
    [5]
    LIN FY, CHEN WY, ZHOU JH, et al. Mesenchymal stem cells protect against ferroptosis via exosome-mediated stabilization of SLC7A11 in acute liver injury[J]. Cell Death Dis, 2022, 13( 3): 271. DOI: 10.1038/s41419-022-04708-w.
    [6]
    VERVOORT LM, RONDEN JE, THIJSSEN HH. The potent antioxidant activity of the vitamin K cycle in microsomal lipid peroxidation[J]. Biochem Pharmacol, 1997, 54( 8): 871- 876. DOI: 10.1016/s0006-2952(97)00254-2.
    [7]
    LI JR, LIN JC, WANG H, et al. Novel role of vitamin k in preventing oxidative injury to developing oligodendrocytes and neurons[J]. J Neurosci, 2003, 23( 13): 5816- 5826. DOI: 10.1523/JNEUROSCI.23-13-05816.2003.
    [8]
    MISHIMA E, ITO J, WU ZJ, et al. A non-canonical vitamin K cycle is a potent ferroptosis suppressor[J]. Nature, 2022, 608( 7924): 778- 783. DOI: 10.1038/s41586-022-05022-3.
    [9]
    DIXON SJ, LEMBERG KM, LAMPRECHT MR, et al. Ferroptosis: An iron-dependent form of nonapoptotic cell death[J]. Cell, 2012, 149( 5): 1060- 1072. DOI: 10.1016/j.cell.2012.03.042.
    [10]
    QIAN MY, LIU MY, TAO CL, et al. Research progress of macrophages death[J]. Chin J Immun, 2023, 39( 1): 204- 210. DOI: 10.3969/j.issn.1000-484X.2023.01.034.

    钱敏雅, 刘满宇, 陶晨璐, 等. 巨噬细胞死亡方式的研究进展[J]. 中国免疫学杂志, 2023, 39( 1): 204- 210. DOI: 10.3969/j.issn.1000-484X.2023.01.034.
    [11]
    SAMRA YA, HAMED MF, EL-SHEAKH AR. Hepatoprotective effect of allicin against acetaminophen-induced liver injury: Role of inflammasome pathway, apoptosis, and liver regeneration[J]. J Biochem Mol Toxicol, 2020, 34( 5): e22470. DOI: 10.1002/jbt.22470.
    [12]
    SATO T, SCHURGERS LJ, UENISHI K. Comparison of menaquinone-4 and menaquinone-7 bioavailability in healthy women[J]. Nutr J, 2012, 11: 93. DOI: 10.1186/1475-2891-11-93.
    [13]
    YANG WS, SRIRAMARATNAM R, WELSCH ME, et al. Regulation of ferroptotic cancer cell death by GPX4[J]. Cell, 2014, 156( 1-2): 317- 331. DOI: 10.1016/j.cell.2013.12.010.
    [14]
    STOCKWELL BR, ANGELI JPF, BAYIR H, et al. Ferroptosis: A regulated cell death nexus linking metabolism, redox biology, and disease[J]. Cell, 2017, 171( 2): 273- 285. DOI: 10.1016/j.cell.2017.09.021.
    [15]
    PIETRANGELO A. Genetics, genetic testing, and management of hemochromatosis: 15 years since hepcidin[J]. Gastroenterology, 2015, 149( 5): 1240- 1251.e4. DOI: 10.1053/j.gastro.2015.06.045.
    [16]
    ZHANG L, LIAO YQ, XIA QC, et al. Ferroptosis regulatory signaling pathway and its research progress in related diseases[J]. Chin J Clin Pharmacol Ther, 2022, 27( 2): 227- 234. DOI: 10.12092/j.issn.1009-2501.2022.02.015.

    张亮, 廖勇群, 夏秦川, 等. 铁死亡调控信号通路以及在相关疾病中的研究进展[J]. 中国临床药理学与治疗学, 2022, 27( 2): 227- 234. DOI: 10.12092/j.issn.1009-2501.2022.02.015.
    [17]
    YANG WS, STOCKWELL BR. Ferroptosis: Death by lipid peroxidation[J]. Trends Cell Biol, 2016, 26( 3): 165- 176. DOI: 10.1016/j.tcb.2015.10.014.
    [18]
    FENG H, STOCKWELL BR. Unsolved mysteries: How does lipid peroxidation cause ferroptosis[J]. PLoS Biol, 2018, 16( 5): e2006203. DOI: 10.1371/journal.pbio.2006203.
  • 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(6)  / Tables(3)

    Article Metrics

    Article views (155) PDF downloads(27) Cited by()
    Proportional views
    Related

    /

    DownLoad:  Full-Size Img  PowerPoint
    Return
    Return