MCU gain- and loss-of-function models define the duality of mitochondrial calcium uptake in heart failure

Background Mitochondrial calcium (mCa2+) uptake through the mitochondrial calcium uniporter channel (mtCU) stimulates metabolism to meet acute increases in cardiac energy demand. However, excessive mCa2+ uptake during stress, as in ischemia-reperfusion, initiates permeability transition and cell death. Despite these often-reported acute physiological and pathological effects, a major unresolved controversy is whether mtCU-dependent mCa2+ uptake and long-term elevation of cardiomyocyte mCa2+ contributes to the heart’s adaptation during sustained increases in workload. Objective We tested the hypothesis that mtCU-dependent mCa2+ uptake contributes to cardiac adaptation and ventricular remodeling during sustained catecholaminergic stress. Methods Mice with tamoxifen-inducible, cardiomyocyte-specific gain (αMHC-MCM x flox-stop-MCU; MCU-Tg) or loss (αMHC-MCM x Mcufl/fl; Mcu-cKO) of mtCU function received 2-wk catecholamine infusion. Results Cardiac contractility increased after 2d of isoproterenol in control, but not Mcu-cKO mice. Contractility declined and cardiac hypertrophy increased after 1-2-wk of isoproterenol in MCU-Tg mice. MCU-Tg cardiomyocytes displayed increased sensitivity to Ca2+- and isoproterenol-induced necrosis. However, loss of the mitochondrial permeability transition pore (mPTP) regulator cyclophilin D failed to attenuate contractile dysfunction and hypertrophic remodeling, and increased isoproterenol-induced cardiomyocyte death in MCU-Tg mice. Conclusions mtCU mCa2+ uptake is required for early contractile responses to adrenergic signaling, even those occurring over several days. Under sustained adrenergic load excessive MCU-dependent mCa2+ uptake drives cardiomyocyte dropout, perhaps independent of classical mitochondrial permeability transition pore opening, and compromises contractile function. These findings suggest divergent consequences for acute versus sustained mCa2+ loading, and support distinct functional roles for the mPTP in settings of acute mCa2+ overload versus persistent mCa2+ stress.

[1]  W. Koch,et al.  Enhanced NCLX-dependent mitochondrial Ca2+ efflux attenuates pathological remodeling in heart failure. , 2022, Journal of molecular and cellular cardiology.

[2]  Shangcheng Xu,et al.  Elevated MCU Expression by CaMKIIδB Limits Pathological Cardiac Remodeling , 2022, Circulation.

[3]  J. Elrod,et al.  Mitochondrial calcium exchange in physiology and disease. , 2021, Physiological reviews.

[4]  J. Elrod,et al.  Is the Failing Heart Starved of Mitochondrial Calcium? , 2021, Circulation research.

[5]  B. O’Rourke,et al.  MCU Overexpression Rescues Inotropy and Reverses Heart Failure by Reducing SR Ca2+ Leak. , 2021, Circulation research.

[6]  T. Luongo,et al.  The debate continues - What is the role of MCU and mitochondrial calcium uptake in the heart? , 2020, Journal of molecular and cellular cardiology.

[7]  E. Gao,et al.  MCUB Regulates the Molecular Composition of the Mitochondrial Calcium Uniporter Channel to Limit Mitochondrial Calcium Overload During Stress. , 2019, Circulation.

[8]  J. Kao,et al.  Voltage-energized Calcium-sensitive ATP Production by Mitochondria , 2019, Nature Metabolism.

[9]  X. Bai,et al.  Structural Mechanism of EMRE-Dependent Gating of the Human Mitochondrial Calcium Uniporter , 2019, Cell.

[10]  M. Salih,et al.  Cardiac-Specific Cre Induces Age-Dependent Dilated Cardiomyopathy (DCM) in Mice , 2019, Molecules.

[11]  J. Ge,et al.  Mitochondrial calcium uniporter inhibition provides cardioprotection in pressure overload-induced heart failure through autophagy enhancement. , 2018, International journal of cardiology.

[12]  D. Catalucci,et al.  Content of mitochondrial calcium uniporter (MCU) in cardiomyocytes is regulated by microRNA-1 in physiologic and pathologic hypertrophy , 2017, Proceedings of the National Academy of Sciences.

[13]  S. Houser,et al.  The mitochondrial Na+/Ca2+ exchanger is essential for Ca2+ homeostasis and viability , 2017, Nature.

[14]  Julia C. Liu,et al.  MICU1 Serves as a Molecular Gatekeeper to Prevent In Vivo Mitochondrial Calcium Overload. , 2016, Cell reports.

[15]  S. Houser,et al.  MCUR1 Is a Scaffold Factor for the MCU Complex Function and Promotes Mitochondrial Bioenergetics. , 2016, Cell reports.

[16]  D. Bers,et al.  Individual Cardiac Mitochondria Undergo Rare Transient Permeability Transition Pore Openings. , 2016, Circulation research.

[17]  Qun Chen,et al.  Activation of mitochondrial calpain and increased cardiac injury: beyond AIF release. , 2016, American journal of physiology. Heart and circulatory physiology.

[18]  Ken-ichi Yoshida,et al.  Mitochondrial m-calpain opens the mitochondrial permeability transition pore in ischemia-reperfusion. , 2015, International journal of cardiology.

[19]  Julia C. Liu,et al.  Assessment of cardiac function in mice lacking the mitochondrial calcium uniporter. , 2015, Journal of molecular and cellular cardiology.

[20]  Jianyi(Jay) Zhang,et al.  The Mitochondrial Calcium Uniporter Selectively Matches Metabolic Output to Acute Contractile Stress in the Heart. , 2015, Cell reports.

[21]  S. Houser,et al.  The Mitochondrial Calcium Uniporter Matches Energetic Supply with Cardiac Workload during Stress and Modulates Permeability Transition. , 2015, Cell reports.

[22]  D. Hall,et al.  The mitochondrial uniporter controls fight or flight heart rate increases , 2015, Nature Communications.

[23]  T. Abraham,et al.  Inhibiting Mitochondrial Na+/Ca2+ Exchange Prevents Sudden Death in a Guinea Pig Model of Heart Failure , 2014, Circulation research.

[24]  R. Rizzuto,et al.  MICU1 and MICU2 Finely Tune the Mitochondrial Ca2+ Uniporter by Exerting Opposite Effects on MCU Activity , 2014, Molecular cell.

[25]  J. Cheung,et al.  MICU1 motifs define mitochondrial calcium uniporter binding and activity. , 2013, Cell reports.

[26]  S. Carr,et al.  EMRE Is an Essential Component of the Mitochondrial Calcium Uniporter Complex , 2013, Science.

[27]  S. Moro,et al.  The mitochondrial calcium uniporter is a multimer that can include a dominant‐negative pore‐forming subunit , 2013, The EMBO journal.

[28]  Sangita Choudhury,et al.  Moderate and high amounts of tamoxifen in αMHC-MerCreMer mice induce a DNA damage response, leading to heart failure and death , 2013, Disease Models & Mechanisms.

[29]  Steven P Jones,et al.  Standardized bioenergetic profiling of adult mouse cardiomyocytes. , 2012, Physiological genomics.

[30]  J. Kolesar,et al.  MCUR1 is an essential component of mitochondrial Ca2+ uptake that regulates cellular metabolism , 2012, Nature Cell Biology.

[31]  M. Birnbaum,et al.  MICU1 Is an Essential Gatekeeper for MCU-Mediated Mitochondrial Ca2+ Uptake that Regulates Cell Survival , 2012, Cell.

[32]  Klitos Konstantinidis,et al.  Mechanisms of cell death in heart disease. , 2012, Arteriosclerosis, thrombosis, and vascular biology.

[33]  V. Mootha,et al.  Integrative genomics identifies MCU as an essential component of the mitochondrial calcium uniporter , 2011, Nature.

[34]  J. Farber,et al.  Cyclophilin D controls mitochondrial pore-dependent Ca(2+) exchange, metabolic flexibility, and propensity for heart failure in mice. , 2010, The Journal of clinical investigation.

[35]  Y. Pinto,et al.  Avoidance of Transient Cardiomyopathy in Cardiomyocyte-Targeted Tamoxifen-Induced MerCreMer Gene Deletion Models , 2009, Circulation research.

[36]  B. O’Rourke,et al.  Enhancing Mitochondrial Ca2+ Uptake in Myocytes From Failing Hearts Restores Energy Supply and Demand Matching , 2008, Circulation research.

[37]  D. Michele,et al.  Blebbistatin extends culture life of adult mouse cardiac myocytes and allows efficient and stable transgene expression. , 2008, American journal of physiology. Heart and circulatory physiology.

[38]  L. Scorrano,et al.  Organelle isolation: functional mitochondria from mouse liver, muscle and cultured filroblasts , 2007, Nature Protocols.

[39]  R. Schnellmann,et al.  Calpain 10: a mitochondrial calpain and its role in calcium-induced mitochondrial dysfunction. , 2006, American journal of physiology. Cell physiology.

[40]  R. Schwartz,et al.  Cardiac-Specific Deletion of Gata4 Reveals Its Requirement for Hypertrophy, Compensation, and Myocyte Viability , 2006, Circulation research.

[41]  S. Korsmeyer,et al.  Cyclophilin D is a component of mitochondrial permeability transition and mediates neuronal cell death after focal cerebral ischemia. , 2005, Proceedings of the National Academy of Sciences of the United States of America.

[42]  Jeffrey Robbins,et al.  Loss of cyclophilin D reveals a critical role for mitochondrial permeability transition in cell death , 2005, Nature.

[43]  中川 崇 Cyclophilin D-dependent mitochondrial permeability transition regulates some necrotic but not apoptotic cell death. , 2005 .

[44]  K. Mani,et al.  Death begets failure in the heart. , 2005, The Journal of clinical investigation.

[45]  M. Grounds,et al.  Evans Blue Dye as an in vivo marker of myofibre damage: optimising parameters for detecting initial myofibre membrane permeability , 2002, Journal of anatomy.

[46]  J. Molkentin,et al.  Physiologic functions of cyclophilin D and the mitochondrial permeability transition pore. , 2013, Circulation journal : official journal of the Japanese Circulation Society.