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:

Mitochondrial pyruvate carrier abundance mediates pathological cardiac hypertrophy

Abstract

Cardiomyocytes rely on metabolic substrates, not only to fuel cardiac output, but also for growth and remodelling during stress. Here we show that mitochondrial pyruvate carrier (MPC) abundance mediates pathological cardiac hypertrophy. MPC abundance was reduced in failing hypertrophic human hearts, as well as in the myocardium of mice induced to fail by angiotensin II or through transverse aortic constriction. Constitutive knockout of cardiomyocyte MPC1/2 in mice resulted in cardiac hypertrophy and reduced survival, while tamoxifen-induced cardiomyocyte-specific reduction of MPC1/2 to the attenuated levels observed during pressure overload was sufficient to induce hypertrophy with impaired cardiac function. Failing hearts from cardiomyocyte-restricted knockout mice displayed increased abundance of anabolic metabolites, including amino acids and pentose phosphate pathway intermediates and reducing cofactors. These hearts showed a concomitant decrease in carbon flux into mitochondrial tricarboxylic acid cycle intermediates, as corroborated by complementary 1,2-[13C2]glucose tracer studies. In contrast, inducible cardiomyocyte overexpression of MPC1/2 resulted in increased tricarboxylic acid cycle intermediates, and sustained carrier expression during transverse aortic constriction protected against cardiac hypertrophy and failure. Collectively, our findings demonstrate that loss of the MPC1/2 causally mediates adverse cardiac remodelling.

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: MPC1 and MPC2 abundance is lower in hearts of patients with hypertrophic cardiomyopathy and mice induced to undergo pathological hypertrophy.
Fig. 2: MPC1 or MPC2 knockout from birth induces heart hypertrophy and increases mortality.
Fig. 3: Inducibly decreasing MPC expression in adult mice switches metabolism to an anabolic programme that causes hypertrophic growth.
Fig. 4: Conserved MPC expression in MPC1 overexpressers in a TAC model of cardiac hypertrophy is cardioprotective and limits aberrant growth.

Similar content being viewed by others

Data availability

The data that support the findings of this study are available from the corresponding author on request. Human Metabolome Technologies uses the Human Metabolome Database (HMDB) (https://hmdb.ca/). The metabolomic data and the HMDB link for each metabolite are available online as Supplementary Table 1 and Supplementary Table 2. Source data for Figs. 1–4 and Extended Data Figs. 1–3, 5 and 6 are available with the paper.

The Ensembl Gene entry used for the generation of MPC1 conditional knockout with tetO knock-in was ENSMUSG00000023861.

The Ensembl Gene entry used for the generation of MPC2 conditional knockout with tetO knock-in was ENSMUSG00000026568.

References

  1. Lopaschuk, G. D., Belke, D. D., Gamble, J., Itoi, T. & Schonekess, B. O. Regulation of fatty acid oxidation in the mammalian heart in health and disease. Biochim. Biophys. Acta 1213, 263–276 (1994).

    Article  CAS  Google Scholar 

  2. Kerr, P. M., Suleiman, M. S. & Halestrap, A. P. Reversal of permeability transition during recovery of hearts from ischemia and its enhancement by pyruvate. Am. J. Physiol. 276, H496–H502 (1999).

    CAS  PubMed  Google Scholar 

  3. Barger, P. M. & Kelly, D. P. Fatty acid utilization in the hypertrophied and failing heart: molecular regulatory mechanisms. Am. J. Med. Sci. 318, 36–42 (1999).

    Article  CAS  Google Scholar 

  4. Razeghi, P. et al. Metabolic gene expression in fetal and failing human heart. Circulation 104, 2923–2931 (2001).

    Article  CAS  Google Scholar 

  5. Oliver, M. F., Kurien, V. A. & Greenwood, T. W. Relation between serum-free-fatty-acids and arrhythmias and death after acute myocardial infarction. Lancet 1, 710–715 (1968).

    Article  CAS  Google Scholar 

  6. Jung, W. I. et al. 31P NMR spectroscopy detects metabolic abnormalities in asymptomatic patients with hypertrophic cardiomyopathy. Circulation 97, 2536–2542 (1998).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  8. 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  Google Scholar 

  9. Kolwicz, S. C. Jr. & Tian, R. Glucose metabolism and cardiac hypertrophy. Cardiovascular Res. 90, 194–201 (2011).

    Article  CAS  Google Scholar 

  10. 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 

  11. Sorokina, N. et al. Recruitment of compensatory pathways to sustain oxidative flux with reduced carnitine palmitoyltransferase I activity characterizes inefficiency in energy metabolism in hypertrophied hearts. Circulation 115, 2033–2041 (2007).

    Article  CAS  Google Scholar 

  12. Taegtmeyer, H. & Overturf, M. L. Effects of moderate hypertension on cardiac function and metabolism in the rabbit. Hypertension 11, 416–426 (1988).

    Article  CAS  Google Scholar 

  13. Smith, S. H., Kramer, M. F., Reis, I., Bishop, S. P. & Ingwall, J. S. Regional changes in creatine kinase and myocyte size in hypertensive and nonhypertensive cardiac hypertrophy. Circulation Res. 67, 1334–1344 (1990).

    Article  CAS  Google Scholar 

  14. 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  Google Scholar 

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

    Article  CAS  Google Scholar 

  16. Schell, J. C. et al. Control of intestinal stem cell function and proliferation by mitochondrial pyruvate metabolism. Nat. Cell Biol. 19, 1027–1036 (2017).

    Article  CAS  Google Scholar 

  17. Flores, A. et al. Lactate dehydrogenase activity drives hair follicle stem cell activation. Nat. Cell Biol. 19, 1017–1026 (2017).

    Article  CAS  Google Scholar 

  18. Zhong, Y. et al. Application of mitochondrial pyruvate carrier blocker UK5099 creates metabolic reprogram and greater stem-like properties in LnCap prostate cancer cells in vitro. Oncotarget 6, 37758–37769 (2015).

    Article  Google Scholar 

  19. Li, X. et al. Mitochondrial pyruvate carrier function determines cell stemness and metabolic reprogramming in cancer cells. Oncotarget 8, 46363–46380 (2017).

    Article  Google Scholar 

  20. Schell, J. C. et al. A role for the mitochondrial pyruvate carrier as a repressor of the Warburg effect and colon cancer cell growth. Mol. Cell 56, 400–413 (2014).

    Article  CAS  Google Scholar 

  21. Li, X. et al. MPC1 and MPC2 expressions are associated with favorable clinical outcomes in prostate cancer. BMC Cancer 16, 894 (2016).

    Article  CAS  Google Scholar 

  22. Xiao, B. et al. Downregulation of COUP-TFII inhibits glioblastoma growth via targeting MPC1. Oncol. Lett. 15, 9697–9702 (2018).

    PubMed  PubMed Central  Google Scholar 

  23. 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  Google Scholar 

  24. El Alaoui-Talibi, Z., Guendouz, A., Moravec, M. & Moravec, J. Control of oxidative metabolism in volume-overloaded rat hearts: effect of propionyl-l-carnitine. Am. J. Physiol. 272, H1615–H1624 (1997).

    CAS  PubMed  Google Scholar 

  25. Wambolt, R. B. et al. Glucose utilization and glycogen turnover are accelerated in hypertrophied rat hearts during severe low-flow ischemia. J. Mol. Cell. Cardiol. 31, 493–502 (1999).

    Article  CAS  Google Scholar 

  26. Zimmer, H. G. Regulation of and intervention into the oxidative pentose phosphate pathway and adenine nucleotide metabolism in the heart. Mol. Cell. Biochem. 160-161, 101–109 (1996).

    Article  CAS  Google Scholar 

  27. Gupte, S. A. et al. Glucose-6-phosphate dehydrogenase-derived NADPH fuels superoxide production in the failing heart. J. Mol. Cell. Cardiol. 41, 340–349 (2006).

    Article  CAS  Google Scholar 

  28. Hildyard, J. C., Ammala, C., Dukes, I. D., Thomson, S. A. & Halestrap, A. P. Identification and characterisation of a new class of highly specific and potent inhibitors of the mitochondrial pyruvate carrier. Biochim. Biophys. Acta 1707, 221–230 (2005).

    Article  CAS  Google Scholar 

  29. 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 

  30. van Bilsen, M., van Nieuwenhoven, F. A. & van der Vusse, G. J. Metabolic remodelling of the failing heart: beneficial or detrimental? Cardiovasc. Res. 81, 420–428 (2009).

    Article  Google Scholar 

  31. 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).

  32. Zhang, Y. et al. Mitochondrial pyruvate carriers are required for myocardial stress adaptation. Nat. Metabol. https://doi.org/10.1038/s42255-020-00288-1 (2020).

  33. UK Government Home Office. Guidance on the Operation of the Animals (Scientific Procedures) Act 1986 (The Stationery Office, 2014).

  34. National Research Council (US) Committee for the Update of the Guide for the Care and Use of Laboratory Animals. Guide for the Care and Use of Laboratory Animals 8th edn (National Academies Press, 2011).

  35. Mackay, G. M., Zheng, L., van den Broek, N. J. & Gottlieb, E. Analysis of cell metabolism using LC-MS and isotope tracers. Methods Enzymol. 561, 171–196 (2015).

    Article  CAS  Google Scholar 

  36. Carpenter, K. L. H. et al. 13C-labelled microdialysis studies of cerebral metabolism in TBI patients. Eur. J. Pharm. Sci. 16, 87–97 (2014).

    Article  Google Scholar 

Download references

Acknowledgements

This work was supported by the British Heart Foundation, the European Research Council (ERC Advanced award) and the Medical Research Council. P.E. is supported by The Barts Charity Cardiovascular Programme Award G00913. We also thank K. Hartmann for her technical assistance and Biobank of ‘A Coruña’ (XXIAC-Instituto de Investigación Biomédica de A Coruña) for providing healthy heart tissue samples. T.E. acknowledges support from NIHR Biomedical Research Centre at Guy’s and St Thomas’ NHS Foundation Trust and KCL; the Centre of Excellence in Medical Engineering funded by the Welcome Trust and EPSRC (WT 088641/Z/09/Z) and the KCL Comprehensive Cancer Imaging Centre funded by the Cancer Research UK (CRUK) and EPSRC in association with MRC and DoH. We acknowledge the metabolic flux analysis facility of the Barts School of Medicine and Dentistry created with the support of the Barts and the London Charity, grant MGU0401.

Author information

Authors and Affiliations

Authors

Contributions

P.E. and M.F.-C. conceived the project. M.F.-C., A.K., A.A.F. and O.P. conducted experiments. S.K. performed histology. K.B. and V.M. conducted the C13 flux metabolomics using LC–MS analysis. N.D., M.G.C.-L. and M.G.V. collected human tissue samples and clinical data. M.F.-C. and T.R.E. analysed the metabolomic data. P.E. and M.F.-C. wrote the manuscript. All authors discussed the results and reviewed the manuscript.

Corresponding authors

Correspondence to Mariana Fernandez-Caggiano or Philip Eaton.

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

a, Cardiac function of wild type mice injected with saline or angiotensin presented as ejection fraction, fractional shortening, cardiac output and stroke volume. Development of cardiac hypertrophy was indexed by the heart weight/tibia length ratio (n = 5 mice/group). b, Cardiac function of wild type mice subjected to a sham or thoracic aortic constriction operation presented as ejection fraction, fractional shortening, cardiac output and stroke volume. Development of cardiac hypertrophy was indexed by the heart weight/tibia length ratio (n = 5 mice/group). All statistical significances (P < 0.05) were calculated using unpaired, two-tailed, Student’s t-tests. Data are presented as mean ± S.E.M.

Source data

Extended Data Fig. 2 Cardiac function of partial MPC1 KO (-/ + CreMPC1), Mhy6Cre control and wild type mice was indexed using echocardiography.

Cardiac function is presented as interventricular septum thickness at end-diastole and end systole (IVSd and IVSs), left ventricular internal-diastolic (LVIDd), internal-systolic dimension (LVIDs), left ventricular posterior wall in diastole and systole (LVPWd and LVPWs), end diastolic and end systolic volume (Vol d and Vol s), stroke volume, ejection fraction, and fractional shortening (n = 9–10 mice/group). Statistical significances (P < 0.05) were calculated using one-way ANOVA adjusted using Dunnett’s test for multiple comparisons. Data are presented as mean ± S.E.M.

Source data

Extended Data Fig. 3 Western immunoblotting analysis of homozygous (TAXMPC1 KO) or heterozygous (TAXMPC1+/-) mouse hearts showed that tamoxifen injection to 8-week-old transgenic TAXMPC1 adults promoted a significant reduction in MPC1 and MPC2 protein abundance (n = 4–6 biologically independent heart samples/group).

All statistical significances (P < 0.05) were calculated using unpaired, two-tailed, Student’s t-tests. Data are presented as mean ± S.E.M.

Source data

Extended Data Fig. 4 Immunofluorescence analysis of dystrophin (green), MPC1 (red) and DAPI enabled measurement of cardiomyocyte cross-sectional area.

TAXMPC1+/-, which are characterised by partial knockout of MPC expression, were hypertrophic as evidenced by the presence of larger cells. (Scale bar represents 25 μm). Representative image for n = 3 biologically independent hearts/group.

Extended Data Fig. 5 Diagram adapted from Carpenter et al36.

to show measurement of flux towards the PPP. TAXMPC1 + /- hearts presented an increase in doubly and singly labelled 13C lactate and pyruvate (n = 5–6 biologically independent heart samples/group). Peak area was normalized by HEPES. All statistical significances (P < 0.05) were calculated using unpaired, two-tailed, Student’s t-tests. Data are presented as mean ± S.E.M.

Source data

Extended Data Fig. 6

a, Proliferation and growth of H9C2 cells was increased in response to the mitochondrial pyruvate carrier inhibitor UK5099 (10 µM or 20 µM). b, H9C2 cells treated with 20 µM UK5099 showed a significant reduction in NADP/NADPH and NAD/NADH ratios, whereas only the NAD/NADH ratio was significant in H9C2 cells treated with 10 µM UK5099 (n = 4 biologically independent cell samples/group). Statistical significances (P < 0.05) were calculated using one-way ANOVA adjusted using Dunnett’s test for multiple comparisons. Data are presented as mean ± S.E.M.

Source data

Supplementary information

Reporting Summary

Supplementary Tables 1 and 2

Supplementary Table 1: Quantitative estimation of target metabolites Control-Cre and TAXMPC1; Supplementary Table 2: Quantitative estimation of target metabolites Control-tTA and tTAMPC1 hearts.

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 1

Unprocessed western blots.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 2

Unprocessed western blots.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 3

Unprocessed western blots.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 4

Unprocessed western blots.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 3

Unprocessed western blots.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Fernandez-Caggiano, M., Kamynina, A., Francois, A.A. et al. Mitochondrial pyruvate carrier abundance mediates pathological cardiac hypertrophy. Nat Metab 2, 1223–1231 (2020). https://doi.org/10.1038/s42255-020-00276-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-020-00276-5

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