Cyclovirobuxine D pretreatment ameliorates septic heart injury through mitigation of ferroptosis

Exp Ther Med. 2023 Jul 10;26(2):407. doi: 10.3892/etm.2023.12106. eCollection 2023 Aug.

Abstract

Myocardial dysfunction is a frequent complication in patients with severe sepsis. However, effective drugs for the prevention of myocardial dysfunction and the molecular mechanisms of the disease remain elusive. The present study demonstrated that Cyclovirobuxine D (CVB-D) could improve cardiac dysfunction in a cecal ligation and puncture (CLP) model in rodents and in a lipopolysaccharide (LPS) model in vitro. Echocardiography and histopathological examination were used to detect changes in cardiac structure and function. Kits were used to detect indicators of cardiac injury, transmission electron microscopy to detect structural changes in mitochondria and reverse transcription-quantitative PCR to detect prostaglandin-endoperoxide synthase 2 and hamp expression levels. L-Glutathione and malondialdehyde levels and superoxide dismutase activity were measured using kits. Cell viability was measured with the Cell Counting Kit-8. Iron metabolism-related proteins, inflammatory factor levels and related pathway proteins were detected using western blot analysis. Changes in L-type calcium currents were detected by membrane clamp, and contractility of cardiomyocytes was measured by Ion Optix. CVB-D attenuated CLP-induced cardiac malfunction in septic rats, with changes observed in myocardial pathological structure, creatine kinase isoenzyme (CK-MB), lactate dehydrogenase (LDH) and cardiac troponin I (cTnI). CVB-D attenuated sepsis-induced lipid peroxidation and iron overload. In addition, CVB-D decreased the expression of CK-MB, LDH and cTnI, suppressed oxidative stress index levels and reduced the production of reactive oxygen species. CVB-D decreased LPS-induced cytoplasmic iron overload by increasing upregulation of iron uptake molecules. Conversely, CVB-D significantly increased the upregulation of ferroportin 1. CVB-D pretreatment significantly reduced the levels of hamp mRNA compared with the LPS-treated group. CVB-D pretreatment significantly reduced inflammatory factor levels and the ratio of phosphorylated vs. total signal transducer and activator of transcription 3. The expression of SLC7A11 and GPX4 was upregulated in septic cells pretreated with CVB-D, however treatment with ML385 largely decreased this upregulation. Of note, CVB-D inhibited the inward flow of calcium ions through the LTCC. In conclusion, these findings suggest that CVB-D alleviated sepsis-induced cardiac iron toxicity by alleviating iron metabolism.

Keywords: L-type Ca2+ channels; cyclovirobuxine D; ferroptosis; iron; nuclear factor erythroid 2-related factor 2; septic cardiomyopathy.

Grants and funding

Funding: This project was supported by the Science Foundation of Hebei Normal University (grant no. L2021B47).