Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2015 Jul 7;12(1):15-22.
doi: 10.1016/j.celrep.2015.06.002. Epub 2015 Jun 25.

The Mitochondrial Calcium Uniporter Selectively Matches Metabolic Output to Acute Contractile Stress in the Heart

Affiliations

The Mitochondrial Calcium Uniporter Selectively Matches Metabolic Output to Acute Contractile Stress in the Heart

Jennifer Q Kwong et al. Cell Rep. .

Abstract

In the heart, augmented Ca(2+) fluxing drives contractility and ATP generation through mitochondrial Ca(2+) loading. Pathologic mitochondrial Ca(2+) overload with ischemic injury triggers mitochondrial permeability transition pore (MPTP) opening and cardiomyocyte death. Mitochondrial Ca(2+) uptake is primarily mediated by the mitochondrial Ca(2+) uniporter (MCU). Here, we generated mice with adult and cardiomyocyte-specific deletion of Mcu, which produced mitochondria refractory to acute Ca(2+) uptake, with impaired ATP production, and inhibited MPTP opening upon acute Ca(2+) challenge. Mice lacking Mcu in the adult heart were also protected from acute ischemia-reperfusion injury. However, resting/basal mitochondrial Ca(2+) levels were normal in hearts of Mcu-deleted mice, and mitochondria lacking MCU eventually loaded with Ca(2+) after stress stimulation. Indeed, Mcu-deleted mice were unable to immediately sprint on a treadmill unless warmed up for 30 min. Hence, MCU is a dedicated regulator of short-term mitochondrial Ca(2+) loading underlying a "fight-or-flight" response that acutely matches cardiac workload with ATP production.

PubMed Disclaimer

Figures

Figure 1
Figure 1. Cardiomyocyte-specific deletion of Mcu impairs mitochondrial Ca2+uptake
(A) Targeting strategy for the Mcu locus to generate the Mcufl/fl mice where exons 5 and 6 were flanked with LoxP sites (triangles). Mcufl/fl mice were crossed to α-MHC MerCreMer (MCM) mice to generate the Mcufl/fl-MCM animals. (B) Tamoxifen dosing to induce MerCreMer activity was given to 8 week-old animals for 4 weeks, followed by examination at 18 and 52 weeks of age. (C) Western blots of MCU and mNCX expression in cardiac mitochondria. The COXI subunit of mitochondrial Complex IV was used as a protein loading control. (D) Quantification of Ca2+ content from isolated cardiac mitochondria from the indicated genotypes of mice. (E) Quantification of baseline mitochondrial Ca2+ content in permeabilized myocytes from the indicated genotypes of mice. (F) The effect of Ru360 (1 μM) on mitochondrial Ca2+ uptake as measured by calcium-green 5N fluorescence in the solution. Mitochondria were challenged with 100 μM CaCl2 additions (arrows). (G) Mitochondrial Ca2+ uptake in mitochondria from hearts of Mcufl/fl vs. Mcufll/fl-MCM mice. Mitochondria were challenged with 200 μM CaCl2 additions (arrows). (H) Measurement of mitochondrial Ca2+ uptake in permeabilized myocytes as assessed by Rhod-2 fluorescence in the indicated groups of mice, with or without Ru360. (I) Quantification of Rhod-2 signal 14 min after Ca2+ addition as shown in H. *P<0.05 vs Mcufl/fl. All values reported as mean ± SEM. (J) Measurement of mitochondrial Ca2+ efflux as mediated by mNCX and leak, assessed by Rhod-2 fluorescence in adult cardiomyocytes. (K) Quantification of rates of mNCX Ca2+ efflux as shown in J. All values reported as mean ± SEM. *P<0.05 versus Mcufl/fl See also Figure S1
Figure 2
Figure 2. Loss of MCU from the adult heart does not lead to pathology at baseline or with pathologic stress stimulation
(A) Time-course for the analyses of cardiac function in response to aging following Mcu deletion. (B) Transverse H&E heart sections at 200X magnification. (C) Representative electron micrographs from heart sections. Scale bar is 500 nm. (D and F) Heart-weight normalized to body-weight ratios (HW/BW) at 18 and 52 weeks of age. (E and G) Echocardiographic measurement of fractional shortening (FS%) at 18 and 52 weeks of age. (H) Time course for generation of mice and analyses of cardiac function following TAC surgery. (I) H&E-stained transverse heart sections 8 weeks after TAC surgery, at 200X magnification. (J-L) HW/BW, (K) cardiomyocyte cross-sectional area and (L) FS% in the indicated groups of mice 8 weeks following TAC. All values reported as mean ± SEM. *P<0.05 versus Mcufl/fl sham
Figure 3
Figure 3. MCU is required for acute mitochondrial Ca2+ stress signaling
(A) Time-course for the cardiac ischemia-reperfusion experiment. (B) Representative images of transverse heart sections stained with 2,3,5-triphenyltetrazolium chloride following ischemia-reperfusion injury from the indicated groups. Ischemic area is outlined in yellow. (C) Quantification of the ischemic area versus area at risk. *P< 0.05 versus all other groups. All values presented as mean ± SEM. (D) Cardiomyocyte viability in response to ionomycin treatment (625 nM, 24h). *P<0.05 versus control. All values presented as mean ± SEM. (E) Mitochondrial swelling in response to Ca2+ challenge (200 μM CaCl2). Controls were 5 μM cyclosporine A (CsA) and 2 μM Ru360. The red arrows show the critical experimental group where swelling is inhibited with Mcu deletion alone. (F) Quantification of MPTP opening frequency in permeabilized cardiomyocytes. MPTP opening is measured by loss of mitochondrial membrane potential (TMRM signal; inset). Myocytes were challenged with 100 nM free Ca2+ and 1 μM CsA was used as a control. *P< 0.005 versus control. All values presented as mean ± SEM. (G) Western blots of CypD, VDAC, and ANT protein expression in purified cardiac mitochondria from the indicated genotypes of mice.
Figure 4
Figure 4. Acute versus chronic regulation of mitochondrial Ca2+ and metabolism due to MCU activity
(A) State 3 mitochondrial oxygen consumption in purified cardiac mitochondria from the indicated mice stimulated with 100 μM Ca2+ with or without 2 μM Ru360. *P<0.05 versus ADP baseline; #P<0.05 versus Mcufl/fl Ca2+. All values presented as mean ± SEM. (B) Mitochondrial ATP synthesis in isolated mitochondria at baseline and stimulated with 400 μM CaCl2. 5 μM Ru360 was used as a control. *P<0.05 versus ADP baseline; #P<0.05 versus Mcufl/fl Ca2+. All values presented as mean ± SEM. (C) Relative oxygen consumption rates (OCR) of Mcufl/fl control adult cardiomyocytes with or without Ru360 (5 μM) in response to 3.125 nM isoproterenol. (D) OCR in Mcufl/fl-MCM vs. Mcufl/fl adult cardiomyocytes in response to 3.125 nM isoproterenol. *P<0.05 versus control. All values presented as mean ± SEM. (E) Maximal rates of cardiac contraction as measured in a closed chest mouse in response to increasing doses of dobutamine. Dobutamine was increased from 0 to 32 ng/g/min and then maintained at 32 ng/g/min for an additional hour. *P<0.05 versus control. All values presented as mean ± SEM. (F) Total mitochondrial Ca2+ content measured from hearts taken at the indicated time-points from indicated groups of mice following dobutamine administration of the experiment shown in E. *P<0.05 versus control. All values presented as mean ± SEM. (G) Treadmill performance as quantified by maximum sprint time in the Mcufl/fl-MCM vs. Mcufl/fl controls. Animals were subjected to two different protocols where they were allowed either a 2 minute warm-up or a 30 minute warm-up before reaching maximum sprint speed. *P<0.05 versus control. Number of mice used is shown in the bars. All values presented as mean ± SEM. See also Figures S2 and S3.

Similar articles

Cited by

References

    1. Abel ED, Doenst T. Mitochondrial adaptations to physiological vs. pathological cardiac hypertrophy. Cardiovasc Res. 2011;90:234–242. - PMC - PubMed
    1. Arany Z, Novikov M, Chin S, Ma Y, Rosenzweig A, Spiegelman BM. Transverse aortic constriction leads to accelerated heart failure in mice lacking PPAR-gamma coactivator 1alpha. Proc Natl Acad Sci U S A. 2006;103:10086–10091. - PMC - PubMed
    1. Baughman JM, Perocchi F, Girgis HS, Plovanich M, Belcher-Timme CA, Sancak Y, Bao XR, Strittmatter L, Goldberger O, Bogorad RL, et al. Integrative genomics identifies MCU as an essential component of the mitochondrial calcium uniporter. Nature. 2011;476:341–345. - PMC - PubMed
    1. Beutner G, Sharma VK, Giovannucci DR, Yule DI, Sheu SS. Identification of a ryanodine receptor in rat heart mitochondria. J Biol Chem. 2001;276:21482–21488. - PubMed
    1. Bondarenko AI, Jean-Quartier C, Malli R, Graier WF. Characterization of distinct single-channel properties of Ca(2)(+) inward currents in mitochondria. Pflugers Arch. 2013;465:997–1010. - PMC - PubMed

Publication types