Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Letter
  • Published:

The mitochondrial Na+/Ca2+ exchanger is essential for Ca2+ homeostasis and viability

Abstract

Mitochondrial calcium (mCa2+) has a central role in both metabolic regulation and cell death signalling, however its role in homeostatic function and disease is controversial1. Slc8b1 encodes the mitochondrial Na+/Ca2+ exchanger (NCLX), which is proposed to be the primary mechanism for mCa2+ extrusion in excitable cells2,3. Here we show that tamoxifen-induced deletion of Slc8b1 in adult mouse hearts causes sudden death, with less than 13% of affected mice surviving after 14 days. Lethality correlated with severe myocardial dysfunction and fulminant heart failure. Mechanistically, cardiac pathology was attributed to mCa2+ overload driving increased generation of superoxide and necrotic cell death, which was rescued by genetic inhibition of mitochondrial permeability transition pore activation. Corroborating these findings, overexpression of NCLX in the mouse heart by conditional transgenesis had the beneficial effect of augmenting mCa2+ clearance, preventing permeability transition and protecting against ischaemia-induced cardiomyocyte necrosis and heart failure. These results demonstrate the essential nature of mCa2+ efflux in cellular function and suggest that augmenting mCa2+ efflux may be a viable therapeutic strategy in disease.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Deletion of Slc8b1 from adult cardiomyocytes causes left ventricular remodelling and heart failure followed by sudden death.
Figure 2: Deletion of Slc8b1 in adult mice ablates mCa2+ efflux resulting in mitochondrial-dependent necrotic cell death.
Figure 3: Cardiomyocyte overexpression of NCLX increases mCa2+ efflux capacity.
Figure 4: Overexpression of NCLX protects against myocardial ischaemic reperfusion injury and ischaemic heart failure.

Similar content being viewed by others

References

  1. Murphy, E. et al. Unresolved questions from the analysis of mice lacking MCU expression. Biochem. Biophys. Res. Commun. 449, 384–385 (2014)

    Article  CAS  Google Scholar 

  2. Palty, R. et al. NCLX is an essential component of mitochondrial Na+/Ca2+ exchange. Proc. Natl Acad. Sci. USA 107, 436–441 (2010)

    Article  CAS  ADS  Google Scholar 

  3. Sekler, I. Standing of giants shoulders the story of the mitochondrial Na+Ca2+ exchanger. Biochem. Biophys. Res. Commun. 460, 50–52 (2015)

    Article  CAS  Google Scholar 

  4. Antony, A. N. et al. MICU1 regulation of mitochondrial Ca2+ uptake dictates survival and tissue regeneration. Nat. Commun. 7, 10955 (2016)

    Article  CAS  ADS  Google Scholar 

  5. Mallilankaraman, K. et al. MICU1 is an essential gatekeeper for MCU-mediated mitochondrial Ca2+ uptake that regulates cell survival. Cell 151, 630–644 (2012)

    Article  CAS  Google Scholar 

  6. Mallilankaraman, K. et al. MCUR1 is an essential component of mitochondrial Ca2+ uptake that regulates cellular metabolism. Nat. Cell Biol. 14, 1336–1343 (2012)

    Article  CAS  Google Scholar 

  7. De Stefani, D., Raffaello, A., Teardo, E., Szabò, I. & Rizzuto, R. A forty-kilodalton protein of the inner membrane is the mitochondrial calcium uniporter. Nature 476, 336–340 (2011)

    Article  CAS  ADS  Google Scholar 

  8. Baughman, J. M. et al. Integrative genomics identifies MCU as an essential component of the mitochondrial calcium uniporter. Nature 476, 341–345 (2011)

    Article  CAS  ADS  Google Scholar 

  9. Perocchi, F. et al. MICU1 encodes a mitochondrial EF hand protein required for Ca2+ uptake. Nature 467, 291–296 (2010)

    Article  CAS  ADS  Google Scholar 

  10. Sancak, Y. et al. EMRE is an essential component of the mitochondrial calcium uniporter complex. Science 342, 1379–1382 (2013)

    Article  CAS  ADS  Google Scholar 

  11. Bers, D. M. Calcium cycling and signaling in cardiac myocytes. Annu. Rev. Physiol. 70, 23–49 (2008)

    Article  CAS  Google Scholar 

  12. Maack, C. & O’Rourke, B. Excitation–contraction coupling and mitochondrial energetics. Basic Res. Cardiol. 102, 369–392 (2007)

    Article  CAS  Google Scholar 

  13. Luongo, T. S. et al. The mitochondrial calcium uniporter matches energetic supply with cardiac workload during stress and modulates permeability transition. Cell Reports 12, 23–34 (2015)

    Article  CAS  Google Scholar 

  14. Pan, X. et al. The physiological role of mitochondrial calcium revealed by mice lacking the mitochondrial calcium uniporter. Nat. Cell Biol. 15, 1464–1472 (2013)

    Article  CAS  Google Scholar 

  15. Kwong, J. Q. et al. The mitochondrial calcium uniporter selectively matches metabolic output to acute contractile stress in the heart. Cell Reports 12, 15–22 (2015)

    Article  CAS  Google Scholar 

  16. Palty, R. & Sekler, I. The mitochondrial Na+/Ca2+ exchanger. Cell Calcium 52, 9–15 (2012)

    Article  CAS  Google Scholar 

  17. Kostic, M. et al. PKA phosphorylation of NCLX reverses mitochondrial calcium overload and depolarization, promoting survival of PINK1-deficient dopaminergic neurons. Cell Reports 13, 376–386 (2015)

    Article  CAS  Google Scholar 

  18. Skarnes, W. C. et al. A conditional knockout resource for the genome-wide study of mouse gene function. Nature 474, 337–342 (2011)

    Article  CAS  Google Scholar 

  19. Sohal, D. S. et al. Temporally regulated and tissue-specific gene manipulations in the adult and embryonic heart using a tamoxifen-inducible Cre protein. Circ. Res. 89, 20–25 (2001)

    Article  CAS  Google Scholar 

  20. Davis, J., Maillet, M., Miano, J. M. & Molkentin, J. D. Lost in transgenesis: a user’s guide for genetically manipulating the mouse in cardiac research. Circ. Res. 111, 761–777 (2012)

    Article  CAS  Google Scholar 

  21. Brookes, P. S., Yoon, Y., Robotham, J. L., Anders, M. W. & Sheu, S. S. Calcium, ATP, and ROS: a mitochondrial love-hate triangle. Am. J. Physiol. Cell Physiol. 287, C817–C833 (2004)

    CAS  Google Scholar 

  22. Baines, C. P. et al. Loss of cyclophilin D reveals a critical role for mitochondrial permeability transition in cell death. Nature 434, 658–662 (2005)

    Article  CAS  ADS  Google Scholar 

  23. Oka, T. et al. Cardiac-specific deletion of Gata4 reveals its requirement for hypertrophy, compensation, and myocyte viability. Circ. Res. 98, 837–845 (2006)

    Article  CAS  Google Scholar 

  24. Boyman, L. et al. Calcium movement in cardiac mitochondria. Biophys. J. 107, 1289–1301 (2014)

    Article  CAS  ADS  Google Scholar 

  25. Bers, D. M. Altered cardiac myocyte Ca regulation in heart failure. Physiology (Bethesda) 21, 380–387 (2006)

    CAS  Google Scholar 

  26. Kohlhaas, M. et al. Elevated cytosolic Na+ increases mitochondrial formation of reactive oxygen species in failing cardiac myocytes. Circulation 121, 1606–1613 (2010)

    Article  CAS  Google Scholar 

  27. Liu, T. & O’Rourke, B. Enhancing mitochondrial Ca2+ uptake in myocytes from failing hearts restores energy supply and demand matching. Circ. Res. 103, 279–288 (2008)

    Article  CAS  Google Scholar 

  28. Nakayama, H. et al. Ca2+- and mitochondrial-dependent cardiomyocyte necrosis as a primary mediator of heart failure. J. Clin. Invest. 117, 2431–2444 (2007)

    Article  CAS  Google Scholar 

  29. Holmström, K. M. et al. Assessment of cardiac function in mice lacking the mitochondrial calcium uniporter. J. Mol. Cell. Cardiol. 85, 178–182 (2015)

    Article  Google Scholar 

  30. Williams, G. S., Boyman, L. & Lederer, W. J. Mitochondrial calcium and the regulation of metabolism in the heart. J. Mol. Cell. Cardiol. 78, 35–45 (2015)

    Article  CAS  Google Scholar 

  31. Reynolds, E. S. The use of lead citrate at high pH as an electron-opaque stain in electron microscopy. J. Cell Biol. 17, 208–212 (1963)

    Article  CAS  Google Scholar 

  32. Zhang, X. et al. Persistent increases in Ca2+ influx through Cav1.2 shortens action potential and causes Ca2+ overload-induced afterdepolarizations and arrhythmias. Basic Res. Cardiol. 111, 4 (2016)

    Article  Google Scholar 

  33. Frezza, C., Cipolat, S. & Scorrano, L. Organelle isolation: functional mitochondria from mouse liver, muscle and cultured fibroblasts. Nat. Protocols 2, 287–295 (2007)

    Article  CAS  Google Scholar 

  34. Hamer, P. W., McGeachie, J. M., Davies, M. J. & Grounds, M. D. Evans Blue Dye as an in vivo marker of myofibre damage: optimising parameters for detecting initial myofibre membrane permeability. J. Anat. 200, 69–79 (2002)

    Article  CAS  Google Scholar 

  35. Dipla, K., Mattiello, J. A., Jeevanandam, V., Houser, S. R. & Margulies, K. B. Myocyte recovery after mechanical circulatory support in humans with end-stage heart failure. Circulation 97, 2316–2322 (1998)

    Article  CAS  Google Scholar 

  36. Gao, E. et al. A novel and efficient model of coronary artery ligation and myocardial infarction in the mouse. Circ. Res. 107, 1445–1453 (2010)

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank T. Tierney, N. Shah, and P. Kelly for technical assistance in the Elrod Laboratory. We thank S. Modla for her expertise in TEM sample processing (Delaware Biotechnology Institute). This study was supported by grants to J.W.E. from the NIH (R01 HL123966, P01 DA037830 sub-8614) and AHA (14SDG18910041) and (15PRE25080299 to T.S.L.), (16PRE31030038 to A.A.L.), and (17PRE33460423 to J.P.L.).

Author information

Authors and Affiliations

Authors

Contributions

J.W.E. and T.S.L. contributed to study design, data analysis and writing the paper; T.S.L. was involved with all assays and data collection; J.P.L. performed western blots and NADH assays; P.G. performed ACM iCa2+ transient analysis; M.N. performed myocardial infarction data acquisition; A.A.L. assisted with the ischaemia reperfusion study and mitochondrial membrane potential assays; S.S. and M.M. assisted with efflux analysis in permeabilized cells; A.C.C. performed qPCR assays; D.K. performed histology; E.G. performed ischaemia reperfusion and myocardial infarction surgeries; J.H.v.B. and J.D.M. aided mutant mouse generation; E.J.T. supplied human heart samples; X.C. performed radiotelemeter implantation and assisted with electrocardiogram analysis; J.W.E., J.H.v.B, T.S.L., S.R.H., J.P.L., A.C.C. and A.A.L. assisted with data interpretation and edited the manuscript.

Corresponding author

Correspondence to John W. Elrod.

Additional information

Reviewer Information: Nature thanks M. Murphy and the other anonymous reviewer(s) for their contribution to the peer review of this work.

Publisher's note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data figures and tables

Extended Data Figure 1 shRNA knockdown of NCLX and biophysical characterization in non-excitable cells.

a, qPCR analysis of SLC8B1 mRNA expression in shRNA stable knockdown HeLa cell lines versus scramble shRNA control cell line (scr. con). b, mCa2+ transients recorded in cells expressing the genetic mCa2+ sensor, mitycam, during histamine treatment (100 μM). Mean intensity shown as solid lines, thin dashed lines display s.e.m. c, mCa2+ peak amplitude. d, Per cent mCa2+ efflux versus scramble shRNA control. n = 42–49 cells per group. eg, HeLa cells were loaded with the Ca2+ sensor, Fura-FF, and the Δψ sensor, JC-1, permeabilized with digitonin (dg) and treated with SERCA inhibitor, thapsigargin (thaps) for simultaneous ratiometric monitoring during repetitive additions of 5 μM Ca2+ (black arrows). FCCP was used to collapse Δψ at the conclusion of each experiment. h, mCa2+ uptake capacity versus scramble shRNA control cells following 20 μM Ca2+ (fourth pulse). i, JC-1-derived Δψ before FCCP addition. n = 3 experiments per assay; *P < 0.05, ***P < 0.001 versus scramble shRNA control.

Extended Data Figure 2 Characterization of Slc8b1 conditional knockout following tamoxifen administration.

a, Images of chimeric founder mice and estimated per cent chimerism, black coat colour correlates with mutant ES cell contribution to development. b, Image of gel with PCR genotyping results for Slc8b1 loxP targeted mice. c, Left ventricular end-systolic dimension (LVESD) three days after tamoxifen administration. d, Curve fitting of mitochondrial swelling traces after addition of 500 μM Ca2+. e, Representative images of MitoSOX Red-stained ACMs (scale bar, 40 μm). f, Fold change in MitoSOX Red intensity three days after tamoxifen administration. n = 42–52 ACMs per group. g, Representative ECG recordings of Slc8b1fl/fl × MCM mice (n = 4–7) at baseline, three days and five days after tamoxifen treatment, and day of sinus arrest. h, Quantitation of PR interval. i, Quantitation of QRS interval. j, Quantitation of heart rate (HR). BPM, beats per minute. k, Western blot of proposed MPTP components from heart protein lysate: CypD and ANT, VDAC shown in Fig. 1d. l, Size distribution (perimeter) of mitochondria analysed in electron microscopy images three days after tamoxifen (minimum 200 mitochondria quantified per mouse, n = 3–4 mice per group). m, Quantification of NAD+/NADH ratio three days after tamoxifen administration (fold change versus control). n = 6–7 per group. n, Western blots of total pyruvate dehydrogenase (PDH E1α) and phosphorylated-PDH (p-PDH) from heart protein lysate. o, Fold change in the ratio of p-PDH/total PDH E1α versus control. n = 3 per group; *P < 0.05, **P < 0.01, ***P < 0.001; ns, not significant.

Source data

Extended Data Figure 3 Examination of Ca2+ flux following αMHC-Cre-mediated deletion of Slc8b1.

a, Adult heart protein lysate isolated from: Slc8b1fl/fl × αMHC-Cre mice and controls were examined by western blot for NCLX expression and various other proteins involved in mCa2+ exchange: MCU, MCUb, MICU1, EMRE, LETM1, VDAC; and OXPHOS components: ATP synthase subunit α (CV-Sα), complex III subunit core 2 (CIII-C2), complex IV subunit I (CIV-SI), and complex I subunit NDUFB8 (CI-SIV). n = 3 hearts per group. b, c, Series of mitycam mCa2+ transients recorded during pacing (0.1 Hz). d, mCa2+ time to peak amplitude. e, Per cent mCa2+ efflux 10 s after pacing. f, Tetramethylrhodamine, ethyl ester (TMRE) (Δψ) in intact ACM. n = 23–24 ACMs per group. g, h, Representative recordings of iCa2+ transients (Fluo-4) in ACMs paced at 1 Hz with or without isoproterenol (Iso, 100 nM). i, iCa2+ peak amplitude. j, iCa2+ time to peak amplitude. k, iCa2+ time to 50% decay. l, Time to 90% decay. n = 10 cells per group. m, n, Permeabilized ACM mCa2+ uptake rate (m) and per cent uptake (n) after Ca2+ addition. *P < 0.05, **P < 0.01, ***P < 0.001.

Extended Data Figure 4 Characterization of NCLX transgenic mice.

a, b, Series of mitycam mCa2+ transients recorded during pacing (0.1 Hz). c, Tetramethylrhodamine, ethyl ester (TMRE) (Δψ) in intact ACMs. n = 23–36 ACMs per group. d, e, Representative traces of iCa2+ transients (Fluo-4) in ACMs paced at 1 Hz with or without isoproterenol (100 nM). f, iCa2+ peak amplitude. g, iCa2+ time to peak amplitude. h, Time to 50% decay. i, Time to 90% decay (n = 15 cells per group). j, Recording of transients in Digitonin (dig)-permeabilized ACMs loaded with Fura-FF, treated with thapsigargin (thaps) and placed in 20 μM bath Ca2+ before treatment with the MCU inhibitor, Ru360, to quantify the rate of mCa2+ efflux independent of uptake. kq, Seahorse analysis of mitochondrial oxygen consumption rates (OCR) in ACMs in the presence of pyruvate or palmitate. m, Basal OCR. n, ATP-linked respiration after addition of ATP synthase inhibitor, oligomycin. o, Maximal respiration after addition of protonophore, FCCP. p, Spare respiratory capacity (max − basal). q, Proton leak (post-oligomycin OCR – non-mitochondrial OCR). n = 11–15 per condition. r, Quantification of NAD+/NADH ratio three days after tamoxifen (fold change versus control). n = 4–5 per group. s, Western blot of total pyruvate dehydrogenase (PDH E1α) and phosphorylated-PDH (p-PDH) from heart protein lysate. *P < 0.05, ***P < 0.001.

Source data

Extended Data Figure 5 Analysis of NCLX-Tg mice 24 h after ichaemia reperfusion and 4 weeks after myocardial infarction.

a, Quantification of TUNEL+ interstitial cells after ischaemia reperfusion. be, B-mode speckle tracking analysis of left ventricular function 4 weeks after myocardial infarction: longitudinal strain (b), longitudinal strain rate (c), radial strain (d), radial strain rate (e). n = 20 tTA and n = 18 NCLX-Tg. f, Lung oedema (wet − dry lung weight). n = 7 per sham group, n = 17 per myocardial infarction group. g, Kaplan–Meier survival curves post myocardial infarction. n = 28 tTA and n = 27 NCLX-Tg. hl, qPCR quantification of mRNA expression in hearts from sham-treated mice or 4 weeks after myocardial infarction. Nppa, atrial natriuretic peptide; Nppb, brain natriuretic peptide; Spp1, osteopontin; Postn1, periostin; Acta2, smooth muscle α-actin (sham, n = 3 group; myocardial infarction, n = 7 per group). m, Representative images of H&E-stained heart sections from the infarct border zone 4 weeks post myocardial infarction. Scale bar, 400 μm. n, o, qPCR analysis of mRNA expression for inflammatory cytokines 4 weeks post myocardial infarction. Il1b, interleukin-1β; Il6, interleukin-6. n = 3 per sham group, n = 7 per myocardial infarction group. p, Western blot of pyruvate dehydrogenase subunits and phosphorylated-PDH (p-PDH) from heart protein lysate 4 weeks post myocardial infarction. q, Fold-change in the ratio of p-PDH/total PDH E1α versus tTA control. n = 4 per group; *P < 0.05, **P < 0.01, ***P < 0.001, versus αMHC tTA post injury; #P < 0.05, ##P < 0.01 versus sham control.

Source data

Extended Data Table 1 Electrocardiography measurements of Slc8b1fl/fl × MCM mice (n = 7) before and after the delivery of tamoxifen
Extended Data Table 2 Genotype distribution of Slc8b1 × αMHC-Cre mice

Supplementary information

Supplementary Figure

This file contains Supplementary Figure 1, the uncropped blots and band density results. (PDF 1699 kb)

PowerPoint slides

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Luongo, T., Lambert, J., Gross, P. et al. The mitochondrial Na+/Ca2+ exchanger is essential for Ca2+ homeostasis and viability. Nature 545, 93–97 (2017). https://doi.org/10.1038/nature22082

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature22082

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing