Monobenzyltin Complex C1 Induces Apoptosis in MCF-7 Breast Cancer Cells through the Intrinsic Signaling Pathway and through the Targeting of MCF-7-Derived Breast Cancer Stem Cells via the Wnt/β-Catenin Signaling Pathway

PLoS One. 2016 Aug 16;11(8):e0160836. doi: 10.1371/journal.pone.0160836. eCollection 2016.

Abstract

Monobenzyltin Schiff base complex, [N-(3,5-dichloro-2-oxidobenzylidene)-4-chlorobenzyhydrazidato](o-methylbenzyl)aquatin(IV) chloride, C1, is an organotin non-platinum metal-based agent. The present study was conducted to investigate its effects on MCF-7 cells with respect to the induction of apoptosis and its inhibitory effect against MCF-7 breast cancer stem cells. As determined in a previous study, compound C1 revealed strong antiproliferative activity on MCF-7 cells with an IC50 value of 2.5 μg/mL. Annexin V/propidium iodide staining coupled with flow cytometry indicated the induction of apoptosis in treated cells. Compound C1 induced apoptosis in MCF-7 cells and was mediated through the intrinsic pathway with a reduction in mitochondrial membrane potential and mitochondrial cytochrome c release to cytosol. Complex C1 activated caspase 9 as a result of cytochrome c release. Subsequently, western blot and real time PCR revealed a significant increase in Bax and Bad expression and a significant decrease in the expression levels of Bcl2 and HSP70. Furthermore, a flow cytometric analysis showed that treatment with compound C1 caused a significant arrest of MCF-7 cells in G0/G1 phase. The inhibitory analysis of compound C1 against derived MCF-7 stem cells showed a significant reduction in the aldehyde dehydrogenase-positive cell population and a significant reduction in the population of MCF-7 cancer stem cells in primary, secondary, and tertiary mammospheres. Moreover, treatment with C1 down-regulated the Wnt/β-catenin self-renewal pathway. These findings indicate that complex C1 is a suppressive agent of MCF-7 cells that functions through the induction of apoptosis, cell cycle arrest, and the targeting of MCF-7-derived cancer stem cells. This work may lead to a better treatment strategy for the reduction of breast cancer recurrence.

MeSH terms

  • Aldehyde Dehydrogenase / metabolism
  • Antineoplastic Agents / chemistry
  • Antineoplastic Agents / pharmacology
  • Apoptosis / drug effects*
  • Biological Transport / drug effects
  • Breast Neoplasms / pathology*
  • Caspase 7 / metabolism
  • Caspase 9 / metabolism
  • Cell Self Renewal / drug effects
  • Cytochromes c / metabolism
  • G1 Phase Cell Cycle Checkpoints / drug effects
  • Gene Expression Regulation, Neoplastic / drug effects
  • Humans
  • MCF-7 Cells
  • Membrane Potential, Mitochondrial / drug effects
  • Molecular Targeted Therapy
  • Neoplastic Stem Cells / drug effects*
  • Neoplastic Stem Cells / pathology
  • Organometallic Compounds / chemistry*
  • Organometallic Compounds / pharmacology*
  • Phosphatidylserines / metabolism
  • Resting Phase, Cell Cycle / drug effects
  • Tin / chemistry*
  • Wnt Signaling Pathway / drug effects*

Substances

  • Antineoplastic Agents
  • Organometallic Compounds
  • Phosphatidylserines
  • Tin
  • Cytochromes c
  • Aldehyde Dehydrogenase
  • Caspase 7
  • Caspase 9

Grants and funding

The authors would like to express their greatest appreciation to the University of Malaya Research Grant (RG084-13BIO), IPPP grant (PG082-2013B), and UM.0000104/HIR.C1 for providing partial financial support to conduct this study.