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.

  • Article
  • Published:

Mitochondrial pyruvate carriers are required for myocardial stress adaptation

A Publisher Correction to this article was published on 18 November 2020

This article has been updated

Abstract

In addition to fatty acids, glucose and lactate are important myocardial substrates under physiologic and stress conditions. They are metabolized to pyruvate, which enters mitochondria via the mitochondrial pyruvate carrier (MPC) for citric acid cycle metabolism. In the present study, we show that MPC-mediated mitochondrial pyruvate utilization is essential for the partitioning of glucose-derived cytosolic metabolic intermediates, which modulate myocardial stress adaptation. Mice with cardiomyocyte-restricted deletion of subunit 1 of MPC (cMPC1−/−) developed age-dependent pathologic cardiac hypertrophy, transitioning to a dilated cardiomyopathy and premature death. Hypertrophied hearts accumulated lactate, pyruvate and glycogen, and displayed increased protein O-linked N-acetylglucosamine, which was prevented by increasing availability of non-glucose substrates in vivo by a ketogenic diet (KD) or a high-fat diet, which reversed the structural, metabolic and functional remodelling of non-stressed cMPC1−/− hearts. Although concurrent short-term KDs did not rescue cMPC1−/− hearts from rapid decompensation and early mortality after pressure overload, 3 weeks of a KD before transverse aortic constriction was sufficient to rescue this phenotype. Together, our results highlight the centrality of pyruvate metabolism to myocardial metabolism and function.

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

Fig. 1: Generation and phenotype of cMPC1−/− mice.
Fig. 2: Substrate metabolism and partitioning in cMPC1−/− hearts.
Fig. 3: Concentrations of metabolic intermediates in perfused cMPC1−/− hearts.
Fig. 4: 13C-labelled enrichment of metabolites in 13C-labelled, substrate-perfused cMPC1−/− hearts.
Fig. 5: KD feeding prevents cardiac dysfunction in cMPC1−/− mice.
Fig. 6: Alternative substrate feeding reverses cardiac dysfunction in cMPC1−/− mice.
Fig. 7: Metabolite profiles in KD hearts.
Fig. 8: Divergent effects of KD feeding on PO-induced cardiac remodelling in cMPC1−/− mice.

Similar content being viewed by others

Data availability

All data, apart from the western blots, that support the findings of the present study are available from the corresponding author upon reasonable request. Source data for western blots are provided with this paper.

Change history

  • 18 November 2020

    An amendment to this paper has been published and can be accessed via a link at the top of the paper.

References

  1. Wende, A. R., Brahma, M. K., McGinnis, G. R. & Young, M. E. Metabolic origins of heart failure. JACC Basic Transl. Sci. 2, 297–310 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  2. Doenst, T., Nguyen, T. D. & Abel, E. D. Cardiac metabolism in heart failure: implications beyond ATP production. Circ. Res. 113, 709–724 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Owen, O. E., Kalhan, S. C. & Hanson, R. W. The key role of anaplerosis and cataplerosis for citric acid cycle function. J. Biol. Chem. 277, 30409–30412 (2002).

    Article  CAS  PubMed  Google Scholar 

  4. Des Rosiers, C., Labarthe, F., Lloyd, S. G. & Chatham, J. C. Cardiac anaplerosis in health and disease: food for thought. Cardiovasc. Res. 90, 210–219 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Pound, K. M. et al. Substrate-enzyme competition attenuates upregulated anaplerotic flux through malic enzyme in hypertrophied rat heart and restores triacylglyceride content: attenuating upregulated anaplerosis in hypertrophy. Circ. Res. 104, 805–812 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Lommi, J. et al. Blood ketone bodies in congestive heart failure. J. Am. Coll. Cardiol. 28, 665–672 (1996).

    Article  CAS  PubMed  Google Scholar 

  7. Aubert, G. et al. The failing heart relies on ketone bodies as a fuel. Circulation 133, 698–705 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Ho, K. L. et al. Increased ketone body oxidation provides additional energy for the failing heart without improving cardiac efficiency. Cardiovasc. Res. 115, 1606–1616 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Huang, Y., Zhou, M., Sun, H. & Wang, Y. Branched-chain amino acid metabolism in heart disease: an epiphenomenon or a real culprit? Cardiovasc. Res. 90, 220–223 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Marazzi, G., Rosanio, S., Caminiti, G., Dioguardi, F. S. & Mercuro, G. The role of amino acids in the modulation of cardiac metabolism during ischemia and heart failure. Curr. Pharm. Des. 14, 2592–2604 (2008).

    Article  CAS  PubMed  Google Scholar 

  11. Drake, K. J., Sidorov, V. Y., McGuinness, O. P., Wasserman, D. H. & Wikswo, J. P. Amino acids as metabolic substrates during cardiac ischemia. Exp. Biol. Med. 237, 1369–1378 (2012).

    Article  CAS  Google Scholar 

  12. Allard, M. F., Schonekess, B. O., Henning, S. L., English, D. R. & Lopaschuk, G. D. Contribution of oxidative metabolism and glycolysis to ATP production in hypertrophied hearts. Am. J. Physiol. 267, H742–H750 (1994).

    CAS  PubMed  Google Scholar 

  13. Kagaya, Y. et al. Effects of long-term pressure overload on regional myocardial glucose and free fatty acid uptake in rats. A quantitative autoradiographic study. Circulation 81, 1353–1361 (1990).

    Article  CAS  PubMed  Google Scholar 

  14. Karwi, Q. G., Uddin, G. M., Ho, K. L. & Lopaschuk, G. D. Loss of metabolic flexibility in the failing heart. Front. Cardiovasc. Med. 5, 68 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  15. Chandramouli, C. et al. Myocardial glycogen dynamics: new perspectives on disease mechanisms. Clin. Exp. Pharm. Physiol. 42, 415–425 (2015).

    Article  CAS  Google Scholar 

  16. Leong, H. S., Brownsey, R. W., Kulpa, J. E. & Allard, M. F. Glycolysis and pyruvate oxidation in cardiac hypertrophy—why so unbalanced? Comp. Biochem Physiol. A Mol. Integr. Physiol. 135, 499–513 (2003).

    Article  CAS  PubMed  Google Scholar 

  17. Lopaschuk, G. D., Wambolt, R. B. & Barr, R. L. An imbalance between glycolysis and glucose oxidation is a possible explanation for the detrimental effects of high levels of fatty acids during aerobic reperfusion of ischemic hearts. J. Pharm. Exp. Ther. 264, 135–144 (1993).

    CAS  Google Scholar 

  18. Nascimben, L. et al. Mechanisms for increased glycolysis in the hypertrophied rat heart. Hypertension 44, 662–667 (2004).

    Article  CAS  PubMed  Google Scholar 

  19. Comte, B. et al. A 13C mass isotopomer study of anaplerotic pyruvate carboxylation in perfused rat hearts. J. Biol. Chem. 272, 26125–26131 (1997).

    Article  CAS  PubMed  Google Scholar 

  20. Peuhkurinen, K. J., Nuutinen, E. M., Pietilainen, E. P., Hiltunen, J. K. & Hassinen, I. E. Role of pyruvate carboxylation in the energy-linked regulation of pool sizes of tricarboxylic acid-cycle intermediates in the myocardium. Biochem. J. 208, 577–581 (1982).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Pisarenko, O. I., Solomatina, E. S. & Studneva, I. M. The role of amino acid catabolism in the formation of the tricarboxylic acid cycle intermediates and ammonia in anoxic rat heart. Biochim. Biophys. Acta 885, 154–161 (1986).

    Article  CAS  PubMed  Google Scholar 

  22. Gibala, M. J., Young, M. E. & Taegtmeyer, H. Anaplerosis of the citric acid cycle: role in energy metabolism of heart and skeletal muscle. Acta Physiol. Scand. 168, 657–665 (2000).

    Article  CAS  PubMed  Google Scholar 

  23. Opie, L. H. & Mansford, K. R. L. The value of lactate and pyruvate measurements in the assessment of the redox state of free nicotinamide-adenine dinucleotide in the cytoplasm of perfused rat heart. Eur. J. Clin. Invest. 1, 295–306 (1971).

    Article  CAS  PubMed  Google Scholar 

  24. Lazo, P. A. & Sols, A. Pyruvate dehydrogenase complex of ascites tumour. Activation by AMP and other properties of potential significance in metabolic regulation. Biochem. J. 190, 705–710 (1980).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Comte, B., Vincent, G., Bouchard, B. & Des Rosiers, C. Probing the origin of acetyl-CoA and oxaloacetate entering the citric acid cycle from the 13C labeling of citrate released by perfused rat hearts. J. Biol. Chem. 272, 26117–26124 (1997).

    Article  CAS  PubMed  Google Scholar 

  26. Bricker, D. K. et al. A mitochondrial pyruvate carrier required for pyruvate uptake in yeast, Drosophila, and humans. Science 337, 96–100 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Halestrap, A. P. The mitochondrial pyruvate carrier: has it been unearthed at last? Cell Metab. 16, 141–143 (2012).

    Article  CAS  PubMed  Google Scholar 

  28. Herzig, S. et al. Identification and functional expression of the mitochondrial pyruvate carrier. Science 337, 93–96 (2012).

    Article  CAS  PubMed  Google Scholar 

  29. Vanderperre, B. et al. Embryonic lethality of mitochondrial pyruvate carrier 1 deficient mouse can be rescued by a ketogenic diet. PLoS Genet. 12, e1006056 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  30. Gray, L. R. et al. Hepatic mitochondrial pyruvate carrier 1 is required for efficient regulation of gluconeogenesis and whole-body glucose homeostasis. Cell Metab. 22, 669–681 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. McCommis, K. S. et al. Loss of mitochondrial pyruvate carrier 2 in the liver leads to defects in gluconeogenesis and compensation via pyruvate–alanine cycling. Cell Metab. 22, 682–694 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Rauckhorst, A. J. et al. The mitochondrial pyruvate carrier mediates high fat diet-induced increases in hepatic TCA cycle capacity. Mol. Metab. 6, 1468–1479 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Sharma, A. et al. Impaired skeletal muscle mitochondrial pyruvate uptake rewires glucose metabolism to drive whole-body leanness. eLife 8, e45873 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Fernandez-Caggiano, M. et al. Analysis of mitochondrial proteins in the surviving myocardium after ischemia identifies mitochondrial pyruvate carrier expression as possible mediator of tissue viability. Mol. Cell Proteom. 15, 246–255 (2016).

    Article  CAS  Google Scholar 

  35. Abel, E. D. et al. Cardiac hypertrophy with preserved contractile function after selective deletion of GLUT4 from the heart. J. Clin. Invest. 104, 1703–1714 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Krebs, H. A. & Gascoyne, T. The redox state of the nicotinamide-adenine dinucleotides in rat liver homogenates. Biochem. J. 108, 513–520 (1968).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Ruiz, M., Gelinas, R., Vaillant, F., Lauzier, B. & Des Rosiers, C. Metabolic tracing using stable isotope-labeled substrates and mass spectrometry in the perfused mouse heart. Methods Enzymol. 561, 107–147 (2015).

    Article  CAS  PubMed  Google Scholar 

  38. Halestrap, A. P. & Denton, R. M. The specificity and metabolic implications of the inhibition of pyruvate transport in isolated mitochondria and intact tissue preparations by alpha-cyano-4-hydroxycinnamate and related compounds. Biochem. J. 148, 97–106 (1975).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Contreras-Baeza, Y. et al. Monocarboxylate transporter 4 (MCT4) is a high affinity transporter capable of exporting lactate in high-lactate microenvironments. J. Biol. Chem. 294, 20135–20147 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Lindblom, P. et al. Isoforms of alanine aminotransferases in human tissues and serum: differential tissue expression using novel antibodies. Arch. Biochem. Biophys. 466, 66–77 (2007).

    Article  CAS  PubMed  Google Scholar 

  41. O’Donnell, J. M., Kalichira, A., Bi, J. & Lewandowski, E. D. In vivo, cardiac-specific knockdown of a target protein, malic enzyme-1, in rat via adenoviral delivery of DNA for non-native miRNA. Curr. Gene Ther. 12, 454–462 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  42. Lahey, R. et al. Enhanced redox state and efficiency of glucose oxidation with miR based suppression of maladaptive NADPH-dependent malic enzyme 1 expression in hypertrophied hearts. Circ. Res. 122, 836–845 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Funk, A. M. et al. Effects of deuteration on transamination and oxidation of hyperpolarized 13C-pyruvate in the isolated heart. J. Magn. Reson. 301, 102–108 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Brooks, G. A. The science and translation of lactate shuttle theory. Cell Metab. 27, 757–785 (2018).

    Article  CAS  PubMed  Google Scholar 

  45. Chen, Y. J. et al. Lactate metabolism is associated with mammalian mitochondria. Nat. Chem. Biol. 12, 937–943 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Taylor, E. B. Functional properties of the mitochondrial carrier system. Trends Cell Biol. 27, 633–644 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Ferron, M., Denis, M., Persello, A., Rathagirishnan, R. & Lauzier, B. Protein O-GlcNacylation in cardiac pathologies: past, present, puture. Front. Endocrinol. 9, 819 (2018).

    Article  Google Scholar 

  48. Arad, M. et al. Glycogen storage diseases presenting as hypertrophic cardiomyopathy. N. Engl. J. Med. 352, 362–372 (2005).

    Article  CAS  PubMed  Google Scholar 

  49. Puchalska, P. & Crawford, P. A. Multi-dimensional roles of ketone bodies in fuel metabolism, signaling, and therapeutics. Cell Metab. 25, 262–284 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Ruan, H. B. & Crawford, P. A. Ketone bodies as epigenetic modifiers. Curr. Opin. Clin. Nutr. Metab. Care 21, 260–266 (2018).

    Article  CAS  PubMed  Google Scholar 

  51. Horton, J. L. et al. The failing heart utilizes 3-hydroxybutyrate as a metabolic stress defense. JCI Insight 4, e124079 (2019).

    Article  PubMed Central  Google Scholar 

  52. Fernandez-Caggiano, M. et al. Mitochondrial pyruvate carrier abundance mediates pathological cardiac hypertrophy. Nat. Metabol. https://doi.org/10.1038/s42255-020-00276-5 (2020).

  53. McCommis, K. S. et al. Nutritional modulation of heart failure in mitochondrial pyruvate carrier–deficient mice. Nat. Metabol. https://doi.org/10.1038/s42255-020-00296-1 (2020).

  54. Ye, J. et al. Primer-BLAST: a tool to design target-specific primers for polymerase chain reaction. BMC Bioinform. 13, 134 (2012).

    Article  CAS  Google Scholar 

  55. Chong, J., Wishart, D. S. & Xia, J. Using MetaboAnalyst 4.0 for comprehensive and integrative metabolomics data analysis. Curr. Protoc. Bioinformatics 68, e86 (2019).

    PubMed  Google Scholar 

  56. Riehle, C. et al. PGC-1β deficiency accelerates the transition to heart failure in pressure overload hypertrophy. Circ. Res. 109, 783–793 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Vaillant, F. et al. Ivabradine and metoprolol differentially affect cardiac glucose metabolism despite similar heart rate reduction in a mouse model of dyslipidemia. Am. J. Physiol. Heart Circ. Physiol. 311, H991–H1003 (2016).

    Article  PubMed  Google Scholar 

  58. Huang, X. et al. X13CMS: global tracking of isotopic labels in untargeted metabolomics. Anal. Chem. 86, 1632–1639 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by: American Heart Association (AHA; grant nos. 16SFRN31810000 to E.D.A. and 15POST22940024 to Y.Z.); Montreal Heart Institute Foundation (CDR); National Institutes of Health (NIH; grant nos. OD019941 to R.M.W., and R01 DK104998 and R00 AR059190 to E.B.T.); T32 (grant no. HL007638 to A.J.R.); American Diabetes Association (grant no. 1-18-PDF-060 (to A.J.R.); and NIH (grant nos. F32 DK101183 to L.R.G., U54DK110858, 1S10OD021505 and 1S10OD018210 to J.E.C., R01HL113057, R01HL132525 and R01HL049244 to E.D.L. and DK091538 to P.A.C.).

Author information

Authors and Affiliations

Authors

Contributions

Y.Z. and E.D.A. designed the research. Y.Z., P.V.T., J.D.C., I.R.-F., J.M.M., J.S., A.D.P., F.T., L.M.T., A.J.R., L.R.G., P.P., T.R.F., R.M., K.Z., W.J.K., T.C., S.H., K.L., K.M.K., J.L.S., L.H., R.M.W. and J.E.C. performed the research. E.B.T. and J.R. provided materials and methodology support. Y.Z., E.D.L., P.A.C., C.D.R. and E.D.A. analysed the data. P.A.C., E.D.L. and C.D.R. contributed to the writing. Y.Z. and E.D.A. wrote the paper.

Corresponding author

Correspondence to E. Dale Abel.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Primary Handling Editor: George Caputa.

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

Extended data

Extended Data Fig. 1 Mitochondrial Characterization of cMPC1-/- hearts.

a, b, MPC protein levels were determined by Western blots in whole heart lysates from 4-week-old cMPC1-/- mice. Images are representative of n = 8 per group. c, Palmitoyl-carnitine driven oxygen consumption by seahorse respirometry in isolated mitochondria (n = 6 both groups). d, Expression of selected electron transport chain (ETC) subunits (from Complex I-V) and VDAC by Western blot in heart lysates from control and cMPC1-/- mice. Images are representative of n = 3 per group. e, mtDNA copy number determined by qPCR analysis and normalized to the nuclear gene RPL13A in 8-week-old cMPC1-/- hearts (Control, 12; cMPC1-/-, 10). f, Representative TEM images of cMPC1-/- hearts from 4 and 8-week-old mice. Images are representative of n = 9 (4-week-old control and cMPC1-/-)/6(8-week-old control)/12(8-week-old cMPC1-/-). g, Quantification of mitochondrial number, volume density and size (4 weeks: Control, 9; cMPC1-/-, 9; 8 weeks: Control, 6; cMPC1-/-, 12). Data are presented as mean ± SEM and analyzed by two-tailed unpaired Student’s t-test.

Source data

Extended Data Fig. 2 Glycolysis enzymes and intermediates in cMPC1-/- hearts.

a, Glycolysis-derived metabolic intermediates in 8-week-old control and cMPC1-/- hearts were determined by GC-MS. Mice were random fed before sacrifice. (Control, 15; cMPC1-/-, 11). b, hexokinase I (HK I), GAPDH, O-GlcNAc transferase (OGT) and glycogen synthase (GS) blots were performed in lysates of cardiomyocytes isolated from 8-week-old control and cMPC1-/- mice. Protein quantification was normalized to total Coomassie blue staining. (n = 5 both groups). Data are presented as mean ± SEM and analyzed by two-tailed unpaired Student’s t-test.

Source data

Extended Data Fig. 3 Flux scheme of 13C-labeled substrate utilization in cMPC1-/- hearts.

Schematic depicting metabolic fate of uniformly labeled glucose ([U-13C6]-glucose) into glycolysis, the pentose phosphate pathway (PPP), serine biosynthesis pathway (SBP) and the Citric Acid Cycle (CAC). Closed and open circles represent 13C-labeled and 12C-labeled carbons respectively.

Extended Data Fig. 4 13C-MPE for pyruvate, lactate, alanine and serine from [U-13C6]-glucose perfusion.

13C-isotopomer labeling pattern of pyruvate (a), lactate (b), alanine (c) and serine (d) following [U-13C6]-glucose perfusion (Control, 7; cMPC1-/-, 9). Data are presented as mean ± SEM and P values were determined by two-tailed unpaired Student’s t-test.

Extended Data Fig. 5 13C-isotopomer labeling pattern of CAC intermediates from 13C-β-HB perfusion.

13C-labeled CAC intermediates analysis of Langendorff-perfused hearts perfused with [2,4-13C2]-β-HB and unlabeled glucose, palmitate and lactate. Fractional enrichment 13C-labeled isotopomers of citrate (a), glutamate (b), succinate (c), α-ketoglutarate (KG) (d), malate (e), aspartate (f) and β-hydroxybutyrate (HB) (g) were determined by LC-MS (Control, 3; cMPC1-/-, 5). Data are presented as mean ± SEM and P values were determined by two-tailed unpaired Student’s t-test.

Extended Data Fig. 6 Expression levels of hypertrophic markers and selected transcripts encoding metabolic genes in the cMPC1-/- hearts on 2920X and ketogenic diet.

The cMPC1-/- mice under protocol 2 were analyzed for gene expression after 8-week-feeding on Ketogenic (Keto) Diet or control diet (2920X). Sample sizes: n = 4 (Control-2920X), n = 4 (cMPC1-/–2920X), n = 5 (Control-Keto), n = 6 (cMPC1-/–Keto). Data are presented as mean ± SEM and P values were determined by two-way ANOVA followed by Tukey multiple comparison test.

Extended Data Fig. 7 Cardiac function of cMPC1-/- after switching ketogenic diet to regular chow.

10-week-old control and cMPC1-/- mice were fed a Keto Diet for 8 weeks and then 50% of mice of each genotype were switched to regular chow for 6 weeks. The feeding scheme is shown in panel (a). LV mass (b) and ejection fraction (c) were determined via echocardiography at the age of 22 weeks (4 weeks after chow switch). Heart weight and tibia length (d) was determined at the age of 24 weeks. Sample sizes: n = 10 (Control-2920X), n = 8 (cMPC1-/–2920X), n = 9 (Control-Keto), n = 8 (cMPC1-/–Keto). Data are presented as mean ± SEM and P values were determined by two-way ANOVA followed by Tukey multiple comparison test.

Extended Data Fig. 8 Effects of ketogenic diet feeding on pressure overload-induced cardiac remodeling in WT mice.

ac, 8-week-old WT C57Bl6/J mice were fed with chow and Keto Diet 1 day before TAC surgery. LV mass (a) and ejection fraction (b) were measured by echocardiography prior to surgery and 3 weeks post TAC. Heart weight normalized to tibia length (c) was determined at the time of sacrifice. (n = 5 for both groups). df, 12-week-old WT C57Bl6/J mice were fed with chow and Keto Diet 1 day before sham/TAC surgery. LV mass (d) and ejection fraction (e) were measured by echocardiography prior to surgery and 3 weeks post TAC. Heart weight normalized to tibia length (f) was determined at the time of sacrifice. (n = 10 for Keto Diet-TAC group and n = 5 for other groups). Data are presented as mean ± SEM and P value was determined by two-way ANOVA followed by Tukey multiple comparison test.

Extended Data Fig. 9 mRNA level of ME isoforms and ALT activity in cMPC1-/- hearts.

a, mRNA level of three malic enzyme isoforms were determined by qPCR in the hearts from control and cMPC1-/- mice (Control, 6; cMPC1-/-, 5). b, c, ALT activity (Cayman 700260) were determined in the hearts from 8-week-old and 18-week-old control and cMPC1-/- mice (8-week-old group: Control, 6; cMPC1-/-, 4. 18-week-old group: Control, 7; cMPC1-/-,7). Data are presented as mean ± SEM and P values were determined by two-tailed unpaired Student’s t-test.

Extended Data Fig. 10 13C-labeled CAC intermediates analysis from [U-13C5]-glutamine perfusion.

13C-labeled CAC intermediates analysis of Langendorff-perfused hearts perfused with 0.5 mM [U-13C5]-glutamine and unlabeled substrates (10 mM glucose, 0.4 mM palmitate, 0.5 mM lactate, and 0.1 mM β-HB). 13C-MPE of glutamine (a), glutamate (b), alpha ketoglutarate (a-KG) (c), citrate (d), malate (e), succinate (f) and pyruvate (g) were determined by GC-MS (Control, 6; cMPC1-/-, 6). Data are presented as mean ± SEM and P values were determined by two-tailed unpaired Student’s t-test.

Supplementary information

Reporting Summary

Supplementary Tables

Supplementary Tables 1 and 2

Source data

Source Data Fig. 1

Unprocessed western blots.

Source Data Extended Data Fig. 1

Unprocessed western blots.

Source Data Extended Data Fig. 2

Unprocessed western blots.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhang, Y., Taufalele, P.V., Cochran, J.D. et al. Mitochondrial pyruvate carriers are required for myocardial stress adaptation. Nat Metab 2, 1248–1264 (2020). https://doi.org/10.1038/s42255-020-00288-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-020-00288-1

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research