[HTML][HTML] K+ regulates Ca2+ to drive inflammasome signaling: dynamic visualization of ion flux in live cells

JR Yaron, S Gangaraju, MY Rao, X Kong, L Zhang… - Cell death & …, 2015 - nature.com
JR Yaron, S Gangaraju, MY Rao, X Kong, L Zhang, F Su, Y Tian, HL Glenn, DR Meldrum
Cell death & disease, 2015nature.com
P2X 7 purinergic receptor engagement with extracellular ATP induces transmembrane
potassium and calcium flux resulting in assembly of the NLRP3 inflammasome in LPS-
primed macrophages. The role of potassium and calcium in inflammasome regulation is not
well understood, largely due to limitations in existing methods for interrogating potassium in
real time. The use of KS6, a novel sensor for selective and sensitive dynamic visualization of
intracellular potassium flux in live cells, multiplexed with the intracellular calcium sensor …
Abstract
P2X 7 purinergic receptor engagement with extracellular ATP induces transmembrane potassium and calcium flux resulting in assembly of the NLRP3 inflammasome in LPS-primed macrophages. The role of potassium and calcium in inflammasome regulation is not well understood, largely due to limitations in existing methods for interrogating potassium in real time. The use of KS6, a novel sensor for selective and sensitive dynamic visualization of intracellular potassium flux in live cells, multiplexed with the intracellular calcium sensor Fluo-4, revealed a coordinated relationship between potassium and calcium. Interestingly, the mitochondrial potassium pool was mobilized in a P2X 7 signaling, and ATP dose-dependent manner, suggesting a role for mitochondrial sensing of cytosolic ion perturbation. Through treatment with extracellular potassium we found that potassium efflux was necessary to permit sustained calcium entry, but not transient calcium flux from intracellular stores. Further, intracellular calcium chelation with BAPTA-AM indicated that P2X 7-induced potassium depletion was independent of calcium mobilization. This evidence suggests that both potassium efflux and calcium influx are necessary for mitochondrial reactive oxygen generation upstream of NLRP3 inflammasome assembly and pyroptotic cell death. We propose a model wherein potassium efflux is necessary for calcium influx, resulting in mitochondrial reactive oxygen generation to trigger the NLRP3 inflammasome.
nature.com