Mitochondrial redox signaling enables repair of injured skeletal muscle cells

A Horn, JH Van der Meulen, A Defour, M Hogarth… - Science …, 2017 - science.org
A Horn, JH Van der Meulen, A Defour, M Hogarth, SC Sreetama, A Reed, L Scheffer…
Science signaling, 2017science.org
Strain and physical trauma to mechanically active cells, such as skeletal muscle myofibers,
injures their plasma membranes, and mitochondrial function is required for their repair. We
found that mitochondrial function was also needed for plasma membrane repair in
myoblasts as well as nonmuscle cells, which depended on mitochondrial uptake of calcium
through the mitochondrial calcium uniporter (MCU). Calcium uptake transiently increased
the mitochondrial production of reactive oxygen species (ROS), which locally activated the …
Strain and physical trauma to mechanically active cells, such as skeletal muscle myofibers, injures their plasma membranes, and mitochondrial function is required for their repair. We found that mitochondrial function was also needed for plasma membrane repair in myoblasts as well as nonmuscle cells, which depended on mitochondrial uptake of calcium through the mitochondrial calcium uniporter (MCU). Calcium uptake transiently increased the mitochondrial production of reactive oxygen species (ROS), which locally activated the guanosine triphosphatase (GTPase) RhoA, triggering F-actin accumulation at the site of injury and facilitating membrane repair. Blocking mitochondrial calcium uptake or ROS production prevented injury-triggered RhoA activation, actin polymerization, and plasma membrane repair. This repair mechanism was shared between myoblasts, nonmuscle cells, and mature skeletal myofibers. Quenching mitochondrial ROS in myofibers during eccentric exercise ex vivo caused increased damage to myofibers, resulting in a greater loss of muscle force. These results suggest a physiological role for mitochondria in plasma membrane repair in injured cells, a role that highlights a beneficial effect of ROS.
AAAS