Vancomycin Induced Ferroptosis in Renal Injury Through the Inactivation of Recombinant Glutathione Peroxidase 4 and the Accumulation of Peroxides

Background Vancomycin (VCM) has long been used clinically to fight against Gram-positive bacterial infections. In recent decades, an increased number of kidney injury cases caused by VCM overdose have been reported. In this study, we further investigated the mechanism of VCM-overdose-induced kidney injury. Methods Immunohistochemistry (IHC) staining, RT-qPCR and Western blot assays were used to determine ki67, DDX5, PTGS2, GPX4 and SLC7A11 expressions in the kidney tissues of mice. CCK-8 and flow cytometry assays were used to determine HK2 cell viability and apoptosis. In addition, RT-qPCR and Western blot assays was applied to evaluate the expressions of ACSL4, PTGS2, GPX4, SLC7A11, DDX5 and Ki67 in HK2 cells. Results We found that VCM induced ferroptosis in vitro and in vivo. Ferrostatin-1 (Fer-1) is a potent inhibitor of ferroptosis, Fer-1 rescued cell viability and renal function renal morphology in VCM-treated cells and mice, respectively. Further, GPX4, which plays an essential role in reducing lipid hydroperoxides and preventing ferroptosis, was observed to be downregulated by VCM treatment. Interestingly, we found that GPX4-knockdown HK-2 cells exhibited a similar phenotype and gene expression level of ACSL4, PTGS2, DDX5 and Ki67 compared with VCM-treated cells, which suggested that VCM could induce ferroptosis in HK2 cells by down-regulating GPX4. Conclusion In conclusion, VCM induced renal injury in the kidney tissues of mice. In addition, VCM induced ferroptosis cell death in HK-2 cells and in the kidney tissues of mice by down-regulating GPX4 and causing the accumulation of peroxides. These data suggested that VCM could induce renal injury in vitro and in vivo via triggering ferroptosis. This study further elucidates the mechanism of VCM-induced renal injury and provides additional references for clinical use of VCM.

[1]  Yinglong Hou,et al.  ATF4 protects against sorafenib-induced cardiotoxicity by suppressing ferroptosis. , 2022, Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie.

[2]  Y. Ruan,et al.  Ferrostatin-1 alleviates oxalate-induced renal tubular epithelial cell injury, fibrosis and calcium oxalate stone formation by inhibiting ferroptosis , 2022, Molecular medicine reports.

[3]  Hui Li,et al.  Silencing of LncRNA KCNQ1OT1 confers an inhibitory effect on renal fibrosis through repressing miR-124-3p activity , 2022, Bioengineered.

[4]  Luyao Li,et al.  MicroRNA-103a-3p enhances sepsis-induced acute kidney injury via targeting CXCL12 , 2022, Bioengineered.

[5]  En-Sheng Ji,et al.  Liraglutide attenuates hepatic iron levels and ferroptosis in db/db mice , 2022, Bioengineered.

[6]  Zeqing Huang,et al.  Bupivacaine modulates the apoptosis and ferroptosis in bladder cancer via phosphatidylinositol 3-kinase (PI3K)/AKT pathway , 2022, Bioengineered.

[7]  Yuwei Zhang,et al.  Chronic intermittent hypoxia induces the pyroptosis of renal tubular epithelial cells by activating the NLRP3 inflammasome , 2022, Bioengineered.

[8]  Jinzhong Huang,et al.  Platycodin D regulates high glucose-induced ferroptosis of HK-2 cells through glutathione peroxidase 4 (GPX4) , 2022, Bioengineered.

[9]  Dan Xu,et al.  Lapatinib induces mitochondrial dysfunction to enhance oxidative stress and ferroptosis in doxorubicin-induced cardiomyocytes via inhibition of PI3K/AKT signaling pathway , 2021, Bioengineered.

[10]  S. Nakagawa,et al.  Antibiotic-induced microbiome depletion alters renal glucose metabolism and exacerbates renal injury after ischemia-reperfusion injury in mice. , 2021, American journal of physiology. Renal physiology.

[11]  E. Mao,et al.  Vitamin C reduces vancomycin-related nephrotoxicity through the inhibition of oxidative stress, apoptosis, and inflammation in mice , 2021, Annals of translational medicine.

[12]  M. D. de Jong,et al.  Impact of a vancomycin loading dose on the achievement of target vancomycin exposure in the first 24 h and on the accompanying risk of nephrotoxicity in critically ill patients , 2021, The Journal of antimicrobial chemotherapy.

[13]  J. Kellum,et al.  Acute kidney injury , 2021, Nature Reviews Disease Primers.

[14]  Bo Yang,et al.  Natural products: potential treatments for cisplatin-induced nephrotoxicity , 2021, Acta Pharmacologica Sinica.

[15]  Jiapeng Wang,et al.  Chrysophanol inhibits the osteoglycin/mTOR and activats NF2 signaling pathways to reduce viability and proliferation of malignant meningioma cells , 2021, Bioengineered.

[16]  Xuelian Bai,et al.  Lipid Peroxidation, GSH Depletion, and SLC7A11 Inhibition are Common Causes of EMT and Ferroptosis in A549 Cells, but Different in Specific Mechanisms. , 2020, DNA and cell biology.

[17]  Zhihong Yang,et al.  Periplaneta americana Extracts Accelerate Liver Regeneration via a Complex Network of Pathways , 2020, Frontiers in Pharmacology.

[18]  Xiaoqun Duan,et al.  Targeting SLC7A11 specifically suppresses the progression of colorectal cancer stem cells via inducing ferroptosis. , 2020, European journal of pharmaceutical sciences : official journal of the European Federation for Pharmaceutical Sciences.

[19]  Rongqian Wu,et al.  Involvement of GPX4 in irisin's protection against ischemia reperfusion‐induced acute kidney injury , 2020, Journal of cellular physiology.

[20]  Shi-kun Yang,et al.  Emerging Role of Ferroptosis in Acute Kidney Injury , 2019, Oxidative medicine and cellular longevity.

[21]  Eunhee Park,et al.  ROS-mediated autophagy increases intracellular iron levels and ferroptosis by ferritin and transferrin receptor regulation , 2019, Cell Death & Disease.

[22]  Hongliang Dong,et al.  Lipid Peroxidation and GPX4 Inhibition Are Common Causes for Myofibroblast Differentiation and Ferroptosis. , 2019, DNA and cell biology.

[23]  J. Farber,et al.  The Nephrotoxicity of Vancomycin , 2017, Clinical pharmacology and therapeutics.

[24]  M. Conrad,et al.  Iron and ferroptosis: A still ill‐defined liaison , 2017, IUBMB life.

[25]  L. Schöckel,et al.  Mitochondrial complex I inhibition triggers a mitophagy-dependent ROS increase leading to necroptosis and ferroptosis in melanoma cells , 2017, Cell Death and Disease.

[26]  D. Tang,et al.  Identification of ACSL4 as a biomarker and contributor of ferroptosis. , 2016, Biochemical and biophysical research communications.

[27]  Yongqiang Chen,et al.  Ferroptosis is induced following siramesine and lapatinib treatment of breast cancer cells , 2016, Cell Death and Disease.

[28]  S. Dixon,et al.  Mechanisms of ferroptosis , 2016, Cellular and Molecular Life Sciences.

[29]  K. Mergenhagen,et al.  Vancomycin Nephrotoxicity , 2014, Journal of pharmacy practice.

[30]  Matthew E. Welsch,et al.  Regulation of Ferroptotic Cancer Cell Death by GPX4 , 2014, Cell.

[31]  M. R. Lamprecht,et al.  Ferroptosis: An Iron-Dependent Form of Nonapoptotic Cell Death , 2012, Cell.

[32]  Kun Wang,et al.  Damage of oxidative stress on mitochondria during microspores development in Honglian CMS line of rice , 2007, Plant Cell Reports.

[33]  F. Őktem,et al.  In vivo evidences suggesting the role of oxidative stress in pathogenesis of vancomycin-induced nephrotoxicity: protection by erdosteine. , 2005, Toxicology.

[34]  G. Matzke,et al.  Clinical Pharmacokinetics of Vancomycin , 1986, Clinical pharmacokinetics.

[35]  A. Walch,et al.  ACSL4 dictates ferroptosis sensitivity by shaping cellular lipid composition. , 2017, Nature chemical biology.

[36]  P. Guest,et al.  Multiplex Analyses Using Real-Time Quantitative PCR. , 2017, Methods in molecular biology.

[37]  R. Brigelius-Flohé,et al.  Glutathione peroxidases. , 2013, Biochimica et biophysica acta.

[38]  R. Griffith Vancomycin use--an historical review. , 1984, The Journal of antimicrobial chemotherapy.