Deficiency of mitochondrial calcium uniporter abrogates iron overload-induced cardiac dysfunction by reducing ferroptosis

N Fefelova, S Wongjaikam, SH Pamarthi… - Basic research in …, 2023 - Springer
N Fefelova, S Wongjaikam, SH Pamarthi, N Siri-Angkul, T Comollo, A Kumari, V Garg…
Basic research in cardiology, 2023Springer
Iron overload associated cardiac dysfunction remains a significant clinical challenge whose
underlying mechanism (s) have yet to be defined. We aim to evaluate the involvement of the
mitochondrial Ca2+ uniporter (MCU) in cardiac dysfunction and determine its role in the
occurrence of ferroptosis. Iron overload was established in control (MCUfl/fl) and conditional
MCU knockout (MCUfl/fl-MCM) mice. LV function was reduced by chronic iron loading in
MCUfl/fl mice, but not in MCUfl/fl-MCM mice. The level of mitochondrial iron and reactive …
Abstract
Iron overload associated cardiac dysfunction remains a significant clinical challenge whose underlying mechanism(s) have yet to be defined. We aim to evaluate the involvement of the mitochondrial Ca2+ uniporter (MCU) in cardiac dysfunction and determine its role in the occurrence of ferroptosis. Iron overload was established in control (MCUfl/fl) and conditional MCU knockout (MCUfl/fl-MCM) mice. LV function was reduced by chronic iron loading in MCUfl/fl mice, but not in MCUfl/fl-MCM mice. The level of mitochondrial iron and reactive oxygen species were increased and mitochondrial membrane potential and spare respiratory capacity (SRC) were reduced in MCUfl/fl cardiomyocytes, but not in MCUfl/fl-MCM cardiomyocytes. After iron loading, lipid oxidation levels were increased in MCUfl/fl, but not in MCUfl/fl-MCM hearts. Ferrostatin-1, a selective ferroptosis inhibitor, reduced lipid peroxidation and maintained LV function in vivo after chronic iron treatment in MCUfl/fl hearts. Isolated cardiomyocytes from MCUfl/fl mice demonstrated ferroptosis after acute iron treatment. Moreover, Ca2+ transient amplitude and cell contractility were both significantly reduced in isolated cardiomyocytes from chronically Fe treated MCUfl/fl hearts. However, ferroptosis was not induced in cardiomyocytes from MCUfl/fl-MCM hearts nor was there a reduction in Ca2+ transient amplitude or cardiomyocyte contractility. We conclude that mitochondrial iron uptake is dependent on MCU, which plays an essential role in causing mitochondrial dysfunction and ferroptosis under iron overload conditions in the heart. Cardiac-specific deficiency of MCU prevents the development of ferroptosis and iron overload-induced cardiac dysfunction.
Springer