[HTML][HTML] Reactive oxygen species are required for driving efficient and sustained aerobic glycolysis during CD4+ T cell activation

DM Previte, EC O'Connor, EA Novak, CP Martins… - PLoS …, 2017 - journals.plos.org
DM Previte, EC O'Connor, EA Novak, CP Martins, KP Mollen, JD Piganelli
PLoS One, 2017journals.plos.org
The immune system is necessary for protecting against various pathogens. However, under
certain circumstances, self-reactive immune cells can drive autoimmunity, like that exhibited
in type 1 diabetes (T1D). CD4+ T cells are major contributors to the immunopathology in
T1D, and in order to drive optimal T cell activation, third signal reactive oxygen species
(ROS) must be present. However, the role ROS play in mediating this process remains to be
further understood. Recently, cellular metabolic programs have been shown to dictate the …
The immune system is necessary for protecting against various pathogens. However, under certain circumstances, self-reactive immune cells can drive autoimmunity, like that exhibited in type 1 diabetes (T1D). CD4+ T cells are major contributors to the immunopathology in T1D, and in order to drive optimal T cell activation, third signal reactive oxygen species (ROS) must be present. However, the role ROS play in mediating this process remains to be further understood. Recently, cellular metabolic programs have been shown to dictate the function and fate of immune cells, including CD4+ T cells. During activation, CD4+ T cells must transition metabolically from oxidative phosphorylation to aerobic glycolysis to support proliferation and effector function. As ROS are capable of modulating cellular metabolism in other models, we sought to understand if blocking ROS also regulates CD4+ T cell activation and effector function by modulating T cell metabolism. To do so, we utilized an ROS scavenging and potent antioxidant manganese metalloporphyrin (MnP). Our results demonstrate that redox modulation during activation regulates the mTOR/AMPK axis by maintaining AMPK activation, resulting in diminished mTOR activation and reduced transition to aerobic glycolysis in diabetogenic splenocytes. These results correlated with decreased Myc and Glut1 upregulation, reduced glucose uptake, and diminished lactate production. In an adoptive transfer model of T1D, animals treated with MnP demonstrated delayed diabetes progression, concurrent with reduced CD4+ T cell activation. Our results demonstrate that ROS are required for driving and sustaining T cell activation-induced metabolic reprogramming, and further support ROS as a target to minimize aberrant immune responses in autoimmunity.
PLOS