Abstract

Mitochondrial-nucleus cross talks and mitochondrial retrograde regulation can play a significant role in cellular properties. Transmitochondrial cybrid systems (cybrids) are an excellent tool to study specific effects of altered mitochondria under a defined nuclear background. The majority of the studies using the cybrid model focused on the significance of specific mitochondrial DNA variations in mitochondrial function or tumor properties. However, most of these variants are benign polymorphisms without known functional significance. From an objective of rectifying mitochondrial defects in cancer cells and to establish mitochondria as a potential anticancer drug target, understanding the role of functional mitochondria in reversing oncogenic properties under a cancer nuclear background is very important. Here we analyzed the potential reversal of oncogenic properties of a highly metastatic cell line with the introduction of non-cancerous mitochondria. Cybrids were established by fusing the mitochondria DNA depleted 143B TK- ρ0 cells from an aggressive osteosarcoma cell line with mitochondria from benign breast epithelial cell line MCF10A, moderately metastatic breast cancer cell line MDA-MB-468 and 143B cells. In spite of the uniform cancerous nuclear background, as observed with the mitochondria donor cells, cybrids with benign mitochondria showed high mitochondrial functional properties including increased ATP synthesis, oxygen consumption and respiratory chain activities compared to cybrids with cancerous mitochondria. Interestingly, benign mitochondria could reverse different oncogenic characteristics of 143B TK- cell including cell proliferation, viability under hypoxic condition, anti-apoptotic properties, resistance to anti-cancer drug, invasion, and colony formation in soft agar, and in vivo tumor growth in nude mice. Microarray analysis suggested that several oncogenic pathways observed in cybrids with cancer mitochondria are inhibited in cybrids with non-cancerous mitochondria. These results suggest the critical oncogenic regulation by mitochondrial-nuclear cross talk and highlights rectifying mitochondrial functional properties as a promising target in cancer therapy.

Highlights

  • Cancer cells adapt to hypoxic conditions during progressive tumor cell growth by shifting the burden of energy metabolism from oxidative phosphorylation to glycolysis, referred to as the Warburg effect [1]

  • The cybrid technology described here allows the investigation of the effect mitochondria derived from cancer or non-cancer cells on mitochondrial function and oncogenic properties in a common nuclear background

  • Since the leakage of Reactive Oxygen Species (ROS) is an indication of defective mitochondrial function, ROS production was analyzed in all parental cell lines using cellpermeable probe, DCFH-DA

Read more

Summary

Introduction

Cancer cells adapt to hypoxic conditions during progressive tumor cell growth by shifting the burden of energy metabolism from oxidative phosphorylation to glycolysis, referred to as the Warburg effect [1]. The cybrid technology described here allows the investigation of the effect mitochondria derived from cancer or non-cancer cells on mitochondrial function and oncogenic properties in a common nuclear background. Non-cancerous Cells and their Cybrids Exhibit Better Mitochondrial Functional Properties Compared to Cancer Mitochondria

Results
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