Your browser doesn't support javascript.
loading
Targeting KCa3.1 channels to overcome erlotinib resistance in non-small cell lung cancer cells.
Todesca, Luca Matteo; Gerke, Matthias; Bulk, Emma Etmar; Bachmann, Magdalena; Rudersdorf, Alisa; Antonuzzo, Lorenzo; Pillozzi, Serena; Düfer, Martina; Szabo, Ildiko; Schwab, Albrecht.
Afiliación
  • Todesca LM; Institute of Physiology II, University of Münster, Münster, Germany. l.m.todesca@gmail.com.
  • Gerke M; Institute of Physiology II, University of Münster, Münster, Germany.
  • Bulk EE; Institute of Physiology II, University of Münster, Münster, Germany.
  • Bachmann M; Department of Biology, University of Padova, Padua, Italy.
  • Rudersdorf A; Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, Münster, Germany.
  • Antonuzzo L; Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.
  • Pillozzi S; Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.
  • Düfer M; Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, Münster, Germany.
  • Szabo I; Department of Biology, University of Padova, Padua, Italy.
  • Schwab A; Institute of Physiology II, University of Münster, Münster, Germany.
Cell Death Discov ; 10(1): 2, 2024 Jan 04.
Article en En | MEDLINE | ID: mdl-38177097
ABSTRACT
Almost all non-small cell lung cancer (NSCLC) patients initially responding to EGFR tyrosine kinase inhibitors (TKIs) develop acquired resistance. Since KCa3.1 channels, expressed in mitochondria and plasma membrane, regulate similar behavioral traits of NSCLC cells as EGFR, we hypothesized that their blockade contributes to overcoming EGFR-TKI resistance. Meta-analysis of microarray data revealed that KCa3.1 channel expression in erlotinib-resistant NSCLC cells correlates with that of genes of integrin and apoptosis pathways. Using erlotinib-sensitive and -resistant NSCLC cells we monitored the role of mitochondrial KCa3.1 channels in integrin signaling by studying cell-matrix adhesion with single-cell force spectroscopy. Apoptosis was quantified with fluorescence-based assays. The function of mitochondrial KCa3.1 channels in these processes was assessed by measuring the mitochondrial membrane potential and by quantifying ROS production. Functional assays were supplemented by biochemical analyses. We show that KCa3.1 channel inhibition with senicapoc in erlotinib-resistant NSCLC cells increases cell adhesion by increasing ß1-integrin expression, that in turn depends on mitochondrial ROS release. Increased adhesion impairs migration of NSCLC cells in a 3D matrix. At the same time, the senicapoc-dependent ROS production induces cytochrome C release and triggers apoptosis of erlotinib-resistant NSCLC cells. Thus, KCa3.1 channel blockade overcomes EGFR-TKI resistance by inhibiting NSCLC motility and inducing apoptosis.