Abstract

Cardenolide glycosides are natural compounds known to inhibit the ion pumping function of the Na+/K+-ATPase in cellular systems. Interestingly, various cancer cell types are highly susceptible to cardenolide glycosides. Herein, we explore the cardenolide glycoside Acovenoside A (AcoA) with respect to its influences on human A549 non-small cell lung cancer (NSCLC) cells. We found that exposure to AcoA, digoxin and ouabain increases intracellular sodium and ATP levels indicating that the ion pumping function of the transmembrane Na+/K+-ATPase is effectively inhibited. Like digoxin and ouabain, AcoA inhibits transcription factor NF-κB activation and induces apoptotic cell death in NSCLC cells. This was confirmed by a preclinical in vivo model in which AcoA treatment of NSCLC xenografts grown on chick chorioallantoic membranes inhibited the expression of proliferation antigen Ki-67 and induced apoptotic DNA strand breaks. We aimed to elucidate the underlying mechanisms. The Na+/K+-ATPase transmembrane complex contains Src kinase and epidermal growth factor receptor (EGFR). Indeed, we found that AcoA activates Src kinase in A549 cells, but not in a cell-free assay using recombinant Src kinase. Src kinase is a downstream target of EGFR, and correlation analysis using the NCI60 database pointed to a role of EGFR in cardenolide glycoside-induced cancer cell death. Accordingly, NSCLC cells expressing hyperphosphorylated EGFRmut exhibited resistance to AcoA. To investigate the interaction between cardenolide glycosides and EGFR in detail, we performed immunoblotting studies: Whereas ligand binding and EGFR phosphorylation were not significantly affected, ubiquitinated EGFR accumulated after prolonged incubation with AcoA. To visualize EGFR trafficking we used A549 cells transfected with a fluorescent biosensor which binds to activated EGFR. Pretreatment with AcoA and digoxin induced accumulation of EGFR in endosomal compartments thus inhibiting EGF-induced EGFR degradation comparable to the Na+ ionophore monensin, a known inducer of EGFR endosomal arrest. Intracellular Na+ concentrations regulate EGFR trafficking and signaling. Na+ homeostasis is maintained by the Na+/K+-ATPase, which might account for its close interaction with the EGFR. Cardenolide glycosides inhibit the ATP-dependent Na+/K+ exchange through the Na+/K+-ATPase resulting in higher intracellular Na+ levels. Our data provide first evidence that this impedes efficient EGFR trafficking at the endosomal compartment.

Highlights

  • The term cardenolide glycoside is used for a diverse group of naturally derived substances composed of a steroid skeleton linked to a sugar moiety at the C3 position and a lactone substituent at the C17 position (Diederich et al, 2017)

  • Our findings using ELISA technique and A549 cells confirm these data, and we show that the inhibition of epidermal growth factor receptor (EGFR) degradation by monensin and cardenolide glycosides persists after continued exposure for at least 24 h (Figures 8A,B)

  • Our data confirm that Acovenoside A (AcoA) displays typical pharmacodynamic features of the cardenolide glycoside family and exhibits cytotoxic potential in non-small cell lung cancer (NSCLC) cells equivalent to digoxin and ouabain

Read more

Summary

Introduction

The term cardenolide glycoside is used for a diverse group of naturally derived substances composed of a steroid skeleton linked to a sugar moiety at the C3 position and a lactone substituent at the C17 position (Diederich et al, 2017). Cardenolide glycosides target the α-subunit of Na+/K+ATPases located on cellular membranes. The ATP-consuming ion pumping function of the Na+/K+-ATPase transporting 3 Na+ out of and 2 K+ into the cell was early understood and utilized. The molecular functions ascribed to “non-pumping” Na+/K+-ATPase reach far beyond ionic gradients and comprise regulation of protein kinase cascades, transcription factors, membrane transporters, and receptors (Liang et al, 2007). By interfering with Na+/K+-ATPase-related processes, cardenolide glycosides in the nanomolar range are affecting essential mechanisms of cell metabolism, e.g. transcription and translation, glycolysis and immune responses, and can direct cellular fate towards either proliferation or death (Sepp et al, 2014; Diederich et al, 2017; Orlov et al, 2017)

Objectives
Methods
Results
Discussion
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call