Amentoflavone mitigates doxorubicin-induced cardiotoxicity by suppressing cardiomyocyte pyroptosis and inflammation through inhibition of the STING/NLRP3 signalling pathway

Phytomedicine. 2023 Aug:117:154922. doi: 10.1016/j.phymed.2023.154922. Epub 2023 Jun 10.

Abstract

Background: Doxorubicin (DOX) is a potent anticancer chemotherapeutic agent whose clinical application is substantially constrained by its cardiotoxicity. The pathophysiology of DOX-induced cardiotoxicity manifests as cardiomyocyte pyroptosis and inflammation. Amentoflavone (AMF) is a naturally occurring biflavone possessing anti-pyroptotic and anti-inflammatory properties. However, the mechanism through which AMF alleviates DOX-induced cardiotoxicity remains undetermined.

Purpose: This study aimed at investigating the role of AMF in alleviating DOX-induced cardiotoxicity.

Study design and methods: To assess the in vivo effect of AMF, DOX was intraperitoneally administered into a mouse model to induce cardiotoxicity. To elucidate the underlying mechanisms, the activities of STING/NLRP3 were quantified using the NLRP3 agonist nigericin and the STING agonist amidobenzimidazole (ABZI). Primary cardiomyocytes isolated from neonatal Sprague-Dawley rats were treated with saline (vehicle) or DOX with or without AMF and/or ABZI. The echocardiogram, haemodynamics, cardiac injury markers, heart/body weight ratio, and pathological alterations were monitored; the STING/NLRP3 pathway-associated proteins were detected by western blot and cardiomyocyte pyroptosis was analysed by immunofluorescence staining of cleaved N-terminal GSDMD and scanning electron microscopy. Furthermore, we evaluated the potential of AMF in compromising the anticancer effects of DOX in human breast cancer cell lines.

Results: AMF substantially alleviated cardiac dysfunction and reduced heart/body weight ratio and myocardial damage in mice models of DOX-induced cardiotoxicity. AMF effectively suppressed DOX-mediated upregulation of IL-1β, IL-18, TNF-α, and pyroptosis-related proteins, including NLRP3, cleaved caspase-1, and cleaved N-terminal GSDMD. The levels of apoptosis-related proteins, namely Bax, cleaved caspase-3, and BCL-2 were not affected. In addition, AMF inhibited STING phosphorylation in DOX-affected hearts. Intriguingly, the administration of nigericin or ABZI dampened the cardioprotective effects of AMF. The in vitro anti-pyroptotic effect of AMF was demonstrated in attenuating the DOX-induced reduction in cardiomyocyte cell viability, upregulation of cleaved N-terminal GSDMD, and pyroptotic morphology alteration at the microstructural level. AMF exhibited a synergistic effect with DOX to reduce the viability of human breast cancer cells.

Conclusion: AMF alleviates DOX-induced cardiotoxicity by suppressing cardiomyocyte pyroptosis and inflammation via inhibition of the STING/NLRP3 signalling pathway, thereby validating its efficacy as a cardioprotective agent.

Keywords: Amentoflavone; Cardiomyocyte pyroptosis; Doxorubicin-induced cardiotoxicity; Inflammation; STING/NLRP3 signalling pathway.

MeSH terms

  • Animals
  • Apoptosis Regulatory Proteins / metabolism
  • Body Weight
  • Breast Neoplasms* / pathology
  • Cardiotoxicity / drug therapy
  • Cardiotoxicity / metabolism
  • Doxorubicin / pharmacology
  • Female
  • Humans
  • Inflammation / metabolism
  • Mice
  • Myocytes, Cardiac*
  • NLR Family, Pyrin Domain-Containing 3 Protein / metabolism
  • Nigericin / adverse effects
  • Nigericin / metabolism
  • Pyroptosis
  • Rats
  • Rats, Sprague-Dawley

Substances

  • NLR Family, Pyrin Domain-Containing 3 Protein
  • amentoflavone
  • Nigericin
  • Doxorubicin
  • Apoptosis Regulatory Proteins