Skip to main content

Lund University Publications

LUND UNIVERSITY LIBRARIES

A Unifying Mechanism for Cancer Cell Death through Ion Channel Activation by HAMLET.

Storm, Petter LU orcid ; Kjaer Klausen, Thomas ; Trulsson, Maria LU ; Ho Cs, James ; Dosnon, Marion ; Westergren, Tomas LU ; Chao, Yinxia ; Rydström, Anna LU ; Yang, Henry and Pedersen, Stine Falsig , et al. (2013) In PLoS ONE 8(3).
Abstract
Ion channels and ion fluxes control many aspects of tissue homeostasis. During oncogenic transformation, critical ion channel functions may be perturbed but conserved tumor specific ion fluxes remain to be defined. Here we used the tumoricidal protein-lipid complex HAMLET as a probe to identify ion fluxes involved in tumor cell death. We show that HAMLET activates a non-selective cation current, which reached a magnitude of 2.74±0.88 nA within 1.43±0.13 min from HAMLET application. Rapid ion fluxes were essential for HAMLET-induced carcinoma cell death as inhibitors (amiloride, BaCl2), preventing the changes in free cellular Na(+) and K(+) concentrations also prevented essential steps accompanying carcinoma cell death, including changes in... (More)
Ion channels and ion fluxes control many aspects of tissue homeostasis. During oncogenic transformation, critical ion channel functions may be perturbed but conserved tumor specific ion fluxes remain to be defined. Here we used the tumoricidal protein-lipid complex HAMLET as a probe to identify ion fluxes involved in tumor cell death. We show that HAMLET activates a non-selective cation current, which reached a magnitude of 2.74±0.88 nA within 1.43±0.13 min from HAMLET application. Rapid ion fluxes were essential for HAMLET-induced carcinoma cell death as inhibitors (amiloride, BaCl2), preventing the changes in free cellular Na(+) and K(+) concentrations also prevented essential steps accompanying carcinoma cell death, including changes in morphology, uptake, global transcription, and MAP kinase activation. Through global transcriptional analysis and phosphorylation arrays, a strong ion flux dependent p38 MAPK response was detected and inhibition of p38 signaling delayed HAMLET-induced death. Healthy, differentiated cells were resistant to HAMLET challenge, which was accompanied by innate immunity rather than p38-activation. The results suggest, for the first time, a unifying mechanism for the initiation of HAMLET's broad and rapid lethal effect on tumor cells. These findings are particularly significant in view of HAMLET's documented therapeutic efficacy in human studies and animal models. The results also suggest that HAMLET offers a two-tiered therapeutic approach, killing cancer cells while stimulating an innate immune response in surrounding healthy tissues. (Less)
Please use this url to cite or link to this publication:
author
; ; ; ; ; ; ; ; and , et al. (More)
; ; ; ; ; ; ; ; ; and (Less)
organization
publishing date
type
Contribution to journal
publication status
published
subject
in
PLoS ONE
volume
8
issue
3
article number
e58578
publisher
Public Library of Science (PLoS)
external identifiers
  • wos:000318334500102
  • pmid:23505537
  • scopus:84874736041
  • pmid:23505537
ISSN
1932-6203
DOI
10.1371/journal.pone.0058578
language
English
LU publication?
yes
id
509869db-6370-4b20-917d-19841598e5ed (old id 3628041)
alternative location
http://www.ncbi.nlm.nih.gov/pubmed/23505537?dopt=Abstract
date added to LUP
2016-04-01 13:13:52
date last changed
2024-03-13 00:50:46
@article{509869db-6370-4b20-917d-19841598e5ed,
  abstract     = {{Ion channels and ion fluxes control many aspects of tissue homeostasis. During oncogenic transformation, critical ion channel functions may be perturbed but conserved tumor specific ion fluxes remain to be defined. Here we used the tumoricidal protein-lipid complex HAMLET as a probe to identify ion fluxes involved in tumor cell death. We show that HAMLET activates a non-selective cation current, which reached a magnitude of 2.74±0.88 nA within 1.43±0.13 min from HAMLET application. Rapid ion fluxes were essential for HAMLET-induced carcinoma cell death as inhibitors (amiloride, BaCl2), preventing the changes in free cellular Na(+) and K(+) concentrations also prevented essential steps accompanying carcinoma cell death, including changes in morphology, uptake, global transcription, and MAP kinase activation. Through global transcriptional analysis and phosphorylation arrays, a strong ion flux dependent p38 MAPK response was detected and inhibition of p38 signaling delayed HAMLET-induced death. Healthy, differentiated cells were resistant to HAMLET challenge, which was accompanied by innate immunity rather than p38-activation. The results suggest, for the first time, a unifying mechanism for the initiation of HAMLET's broad and rapid lethal effect on tumor cells. These findings are particularly significant in view of HAMLET's documented therapeutic efficacy in human studies and animal models. The results also suggest that HAMLET offers a two-tiered therapeutic approach, killing cancer cells while stimulating an innate immune response in surrounding healthy tissues.}},
  author       = {{Storm, Petter and Kjaer Klausen, Thomas and Trulsson, Maria and Ho Cs, James and Dosnon, Marion and Westergren, Tomas and Chao, Yinxia and Rydström, Anna and Yang, Henry and Pedersen, Stine Falsig and Svanborg, Catharina}},
  issn         = {{1932-6203}},
  language     = {{eng}},
  number       = {{3}},
  publisher    = {{Public Library of Science (PLoS)}},
  series       = {{PLoS ONE}},
  title        = {{A Unifying Mechanism for Cancer Cell Death through Ion Channel Activation by HAMLET.}},
  url          = {{https://lup.lub.lu.se/search/files/3245580/3810988.pdf}},
  doi          = {{10.1371/journal.pone.0058578}},
  volume       = {{8}},
  year         = {{2013}},
}