[HTML][HTML] Persistent mitochondrial hyperfusion promotes G2/M accumulation and caspase-dependent cell death

LM Westrate, AD Sayfie, DM Burgenske… - PloS one, 2014 - journals.plos.org
LM Westrate, AD Sayfie, DM Burgenske, JP MacKeigan
PloS one, 2014journals.plos.org
Cancer cells have several hallmarks that define their neoplastic behavior. One is their
unabated replicative potential that allows cells to continually proliferate, and thereby
contribute to increasing tumor burden. The progression of a cell through the cell cycle is
regulated by a series of checkpoints that ensures successful transmission of genetic
information, as well as various cellular components, including organelles and protein
complexes to the two resulting daughter cells. The mitochondrial reticulum undergoes …
Cancer cells have several hallmarks that define their neoplastic behavior. One is their unabated replicative potential that allows cells to continually proliferate, and thereby contribute to increasing tumor burden. The progression of a cell through the cell cycle is regulated by a series of checkpoints that ensures successful transmission of genetic information, as well as various cellular components, including organelles and protein complexes to the two resulting daughter cells. The mitochondrial reticulum undergoes coordinated changes in shape to correspond with specific stages of the cell cycle, the most dramatic being complete mitochondrial fragmentation prior to cytokinesis. To determine whether mitochondrial fission is a required step to ensure proper mitochondrial segregation into two daughter cells, we investigated the importance of mitochondrial dynamics to cell cycle progression. We found that mitochondrial hyperfusion promotes a defect in cell cycle progression characterized by an inability for cells to exit G2/M. Additionally, extended periods of persistent mitochondrial fusion led to robust caspase-dependent cell death. The cell death signals were coordinated through activation and cleavage of caspase-8, promoting a potent death response. These results demonstrate the importance of mitochondrial dynamics in cell cycle progression, and that inhibiting mitochondrial fission regulators may provide a therapeutic strategy to target the replicative potential of cancer cells.
PLOS