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:

PCYT2-regulated lipid biosynthesis is critical to muscle health and ageing

An Author Correction to this article was published on 06 April 2023

This article has been updated

Abstract

Muscle degeneration is the most prevalent cause for frailty and dependency in inherited diseases and ageing. Elucidation of pathophysiological mechanisms, as well as effective treatments for muscle diseases, represents an important goal in improving human health. Here, we show that the lipid synthesis enzyme phosphatidylethanolamine cytidyltransferase (PCYT2/ECT) is critical to muscle health. Human deficiency in PCYT2 causes a severe disease with failure to thrive and progressive weakness. pcyt2-mutant zebrafish and muscle-specific Pcyt2-knockout mice recapitulate the participant phenotypes, with failure to thrive, progressive muscle weakness and accelerated ageing. Mechanistically, muscle Pcyt2 deficiency affects cellular bioenergetics and membrane lipid bilayer structure and stability. PCYT2 activity declines in ageing muscles of mice and humans, and adeno-associated virus-based delivery of PCYT2 ameliorates muscle weakness in Pcyt2-knockout and old mice, offering a therapy for individuals with a rare disease and muscle ageing. Thus, PCYT2 plays a fundamental and conserved role in vertebrate muscle health, linking PCYT2 and PCYT2-synthesized lipids to severe muscle dystrophy and ageing.

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: Phenotypes of human PCYT2 rare disease mutations and pcyt2-mutant zebrafish.
Fig. 2: Pcyt2 deficiency affects muscle stem cell fusion and muscle hypertrophic growth.
Fig. 3: Inactivation of Pcyt2 in mice leads to progressive weakness, muscle atrophy, inflammation and accelerated ageing.
Fig. 4: Pcyt2 deficiency severely affects muscle mitochondrial homeostasis as opposed to brown fat mitochondria.
Fig. 5: Loss of Pcyt2 results in altered muscle membrane architectures.
Fig. 6: Pcyt2 is essential for muscle membrane integrity and strain tolerance.
Fig. 7: Adenovirus-based Pcyt2 gene therapy in mice is efficient for treatment of Pcyt2 deficiency-induced muscle pathology.
Fig. 8: Pcyt2 activity is reduced in aged muscles from humans and mice and Pcyt2 gene delivery ameliorates age-related atrophy in sarcopenic mice.

Similar content being viewed by others

Data availability

The original datasets used in the RNA sequencing analysis can be accessed at NCBI archived under Gene Expression Omnibus accession codes GSM6785661, GSM6785662, GSM6785663, GSM6785664, GSM6785665 and GSM6785666. Mass spectrometry lipidomics data for Figs. 1, 4 and 8 and Extended Data Fig. 7 are provided in the Supplementary Information. Further information and requests for resources and reagents should be directed to and will be fulfilled by the corresponding authors. Source data are provided with this paper.

Change history

References

  1. Davies, K. E. & Nowak, K. J. Molecular mechanisms of muscular dystrophies: old and new players. Nat. Rev. Mol. Cell Biol. 7, 762–773 (2006).

    Article  CAS  PubMed  Google Scholar 

  2. Wilkinson, D. J., Piasecki, M. & Atherton, P. J. The age-related loss of skeletal muscle mass and function: measurement and physiology of muscle fibre atrophy and muscle fibre loss in humans. Ageing Res. Rev. 47, 123–132 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Shevchenko, A. & Simons, K. Lipidomics: coming to grips with lipid diversity. Nat. Rev. Mol. Cell Biol. 11, 593–598 (2010).

    Article  CAS  PubMed  Google Scholar 

  4. Harayama, T. & Riezman, H. Understanding the diversity of membrane lipid composition. Nat. Rev. Mol. Cell Biol. 19, 281–296 (2018).

    Article  CAS  PubMed  Google Scholar 

  5. Gibellini, F. & Smith, T. K. The Kennedy pathway—de novo synthesis of phosphatidylethanolamine and phosphatidylcholine. IUBMB Life 62, 414–428 (2010).

    Article  CAS  PubMed  Google Scholar 

  6. Vaz, F. M. et al. Mutations in PCYT2 disrupt etherlipid biosynthesis and cause a complex hereditary spastic paraplegia. Brain 142, 3382–3397 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  7. Ahmed, M. Y. et al. A mutation of EPT1 (SELENOI) underlies a new disorder of Kennedy pathway phospholipid biosynthesis. Brain 140, aww318–aww554 (2017).

    Article  Google Scholar 

  8. Horibata, Y. et al. EPT1 (selenoprotein I) is critical for the neural development and maintenance of plasmalogen in humans. J. Lipid Res. 59, 1015–1026 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Horibata, Y. & Hirabayashi, Y. Identification and characterization of human ethanolaminephosphotransferase. J. Lipid Res. 48, 503–508 (2007).

    Article  CAS  PubMed  Google Scholar 

  10. Uhlén, M. et al. Tissue-based map of the human proteome. Science 347, 1260419 (2015).

    Article  PubMed  Google Scholar 

  11. Naef, V. et al. Swimming in deep water: zebrafish modeling of complicated forms of hereditary spastic paraplegia and spastic ataxia. Front. Neurosci. 13, 1311 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  12. Fassier, C., Hazan, J. & Melki, J. Mouse models of autosomal dominant spastic paraplegia. in Movement Disorders: Genetics and Models 2nd edn, 1073–1086 (ed Mark S. LeDoux) (Elsevier, 2015). https://doi.org/10.1016/B978-0-12-405195-9.00070-6

  13. Blackstone, C. Murine models of autosomal recessive hereditary spastic paraplegia. in Movement Disorders: Genetics and Models 2nd edn, 1087–1093 (ed Mark S. LeDoux) (Elsevier, 2015). https://doi.org/10.1016/B978-0-12-405195-9.00071-8

  14. Rapisarda, R., Muntoni, F., Gobbi, P. & Dubowitz, V. Duchenne muscular dystrophy presenting with failure to thrive. Arch. Dis. Child. 72, 437–438 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Fullerton, M. D., Hakimuddin, F. & Bakovic, M. Developmental and metabolic effects of disruption of the mouse CTP:phosphoethanolamine cytidylyltransferase gene (Pcyt2). Mol. Cell. Biol. 27, 3327–3336 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Francetic, T. & Li, Q. Skeletal myogenesis and Myf5 activation. Transcription 2, 109–114 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  17. White, R. B., Biérinx, A. S., Gnocchi, V. F. & Zammit, P. S. Dynamics of muscle fibre growth during postnatal mouse development. BMC Dev. Biol. 10, 21 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Demonbreun, A. R., Biersmith, B. H. & McNally, E. M. Membrane fusion in muscle development and repair. Semin. Cell Dev. Biol. 45, 48–56 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Emoto, K. & Umeda, M. An essential role for a membrane lipid in cytokinesis. J. Cell Biol. 149, 1215–1224 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Mccarthy, J. J. et al. Effective fiber hypertrophy in satellite cell-depleted skeletal muscle. Development 138, 3657–3666 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Archer, J. E., Gardner, A. C., Roper, H. P., Chikermane, A. A. & Tatman, A. J. Duchenne muscular dystrophy: the management of scoliosis. J. Spine Surg. 2, 185–194 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Seale, P. et al. PRDM16 controls a brown fat/skeletal muscle switch. Nature 454, 961–967 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Eguchi, J. et al. Transcriptional control of adipose lipid handling by IRF4. Cell Metab. 13, 249–259 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Weiland, D. et al. Imbalance of mitochondrial respiratory chain complexes in the epidermis induces severe skin inflammation. J. Invest. Dermatol. 138, 132–140 (2018).

    Article  CAS  PubMed  Google Scholar 

  25. Shin, J. M. et al. Targeted deletion of Crif1 in mouse epidermis impairs skin homeostasis and hair morphogenesis. Sci. Rep. 7, 44828 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Nguyen, T. T. et al. Loss of Miro1-directed mitochondrial movement results in a novel murine model for neuron disease. Proc. Natl Acad. Sci. USA 111, 3631–3640 (2014).

    Article  Google Scholar 

  27. Choi, M. J. et al. An adipocyte-specific defect in oxidative phosphorylation increases systemic energy expenditure and protects against diet-induced obesity in mouse models. Diabetologia 63, 837–852 (2020).

    Article  CAS  PubMed  Google Scholar 

  28. Vernochet, C. et al. Adipose tissue mitochondrial dysfunction triggers a lipodystrophic syndrome with insulin resistance, hepatosteatosis and cardiovascular complications. FASEB J. 28, 4408–4419 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Sustarsic, E. G. et al. Cardiolipin synthesis in brown and beige fat mitochondria is essential for systemic energy homeostasis. Cell Metab. 28, 159–174 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Srivillibhuthur, M. et al. TFAM is required for maturation of the fetal and adult intestinal epithelium. Dev. Biol. 439, 92–101 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Selathurai, A. et al. The CDP-ethanolamine pathway regulates skeletal muscle diacylglycerol content and mitochondrial biogenesis without altering insulin sensitivity. Cell Metab. 21, 718–730 (2015).

    Article  CAS  PubMed  Google Scholar 

  32. Beedle, A. M. et al. Mouse fukutin deletion impairs dystroglycan processing and recapitulates muscular dystrophy. J. Clin. Invest. 122, 3330–3342 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Potthoff, M. J. et al. Regulation of skeletal muscle sarcomere integrity and postnatal muscle function by Mef2c. Mol. Cell. Biol. 27, 8143–8151 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Brüning, J. C. et al. A muscle-specific insulin receptor knockout exhibits features of the metabolic syndrome of NIDDM without altering glucose tolerance. Mol. Cell 2, 559–569 (1998).

    Article  PubMed  Google Scholar 

  35. Chamberlain, J. S., Jaynes, J. B. & Hauschka, S. D. Regulation of creatine kinase induction in differentiating mouse myoblasts. Mol. Cell. Biol. 5, 484–492 (1985).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Schlame, M., Xu, Y., Erdjument-Bromage, H., Neubert, T. A. & Ren, M. Lipidome-wide 13C flux analysis: a novel tool to estimate the turnover of lipids in organisms and cultures. J. Lipid Res. 61, 95–104 (2020).

    Article  CAS  PubMed  Google Scholar 

  37. Van Meer, G., Voelker, D. R. & Feigenson, G. W. Membrane lipids: where they are and how they behave. Nat. Rev. Mol. Cell Biol. 9, 112–124 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  38. Nair, U. et al. A role for Atg8-PE deconjugation in autophagosome biogenesis. Autophagy 8, 780–793 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Martinez-Lopez, N. et al. Autophagy in Myf5+ progenitors regulates energy and glucose homeostasis through control of brown fat and skeletal muscle development. EMBO Rep. 14, 795–803 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Roses, A. D. & Appel, S. H. Inherited membrane disorders of muscle: Duchenne muscular dystrophy and myotonic muscular dystrophy. in Physiology of Membrane Disorders (eds T. E. Andreoli et al.) 801–815 (Springer, 1978).

  41. Steinkühler, J., Sezgin, E., Urbančič, I., Eggeling, C. & Dimova, R. Mechanical properties of plasma membrane vesicles correlate with lipid order, viscosity and cell density. Commun. Biol. 2, 337 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  42. Elsayad, K. et al. Mapping the subcellular mechanical properties of live cells in tissues with fluorescence emission-Brillouin imaging. Sci. Signal. 9, rs5 (2016).

    Article  PubMed  Google Scholar 

  43. Demonbreun, A. R. & McNally, E. M. Plasma membrane repair in health and disease. Curr. Top. Membr. 77, 67–96 (2016).

    Article  CAS  PubMed  Google Scholar 

  44. 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  PubMed  PubMed Central  Google Scholar 

  45. Bansal, D. et al. Defective membrane repair in dysferlin-deficient muscular dystrophy. Nature 423, 168–172 (2003).

    Article  CAS  PubMed  Google Scholar 

  46. Hody, S., Croisier, J. L., Bury, T., Rogister, B. & Leprince, P. Eccentric muscle contractions: risks and benefits. Front. Physiol. 10, 536 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Ramos, J. N. et al. Development of novel micro-dystrophins with enhanced functionality. Mol. Ther. 27, 623–635 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Crudele, J. M. & Chamberlain, J. S. AAV-based gene therapies for the muscular dystrophies. Hum. Mol. Genet. 28, R102–R107 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Gault, M. L. & Willems, M. E. T. Aging, functional capacity and eccentric exercise training. Aging Dis. 4, 351–363 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  50. Manfredi, T. G. et al. Plasma creatine kinase activity and exercise-induced muscle damage in older men. Med. Sci. Sports Exerc. 23, 1028–1034 (1991).

    Article  CAS  PubMed  Google Scholar 

  51. Roth, S. M. et al. High-volume, heavy-resistance strength training and muscle damage in young and older women. J. Appl. Physiol. 88, 1112–1118 (2000).

    Article  CAS  PubMed  Google Scholar 

  52. McGreevy, J. W., Hakim, C. H., McIntosh, M. A. & Duan, D. Animal models of Duchenne muscular dystrophy: from basic mechanisms to gene therapy. Dis. Model. Mech. 8, 195–213 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Vance, J. E. & Tasseva, G. Formation and function of phosphatidylserine and phosphatidylethanolamine in mammalian cells. Biochim. Biophys. Acta 1831, 543–554 (2013).

    Article  CAS  PubMed  Google Scholar 

  54. Kuter, K. et al. Adaptation within mitochondrial oxidative phosphorylation supercomplexes and membrane viscosity during degeneration of dopaminergic neurons in an animal model of early Parkinson’s disease. Biochim. Biophys. Acta 1862, 741–753 (2016).

    Article  CAS  PubMed  Google Scholar 

  55. Möller, M. N., Lancaster, J. R. & Denicola, A. The interaction of reactive oxygen and nitrogen species with membranes. Curr. Top. Membr. https://doi.org/10.1016/S1063-5823(08)00202-0 (2008).

  56. Yusupov, M. et al. Effect of head group and lipid tail oxidation in the cell membrane revealed through integrated simulations and experiments. Sci. Rep. 7, 5761 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Ylikallio, E. & Suomalainen, A. Mechanisms of mitochondrial diseases. Ann. Med. 44, 41–59 (2012).

    Article  CAS  PubMed  Google Scholar 

  58. Kaiyrzhanov, R. et al. Defective phosphatidylethanolamine biosynthesis leads to a broad ataxia-spasticity spectrum. Brain 144, e30 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  59. Felsenthal, N. & Zelzer, E. Mechanical regulation of musculoskeletal system development. Development 144, 4271–4283 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Zick, M., Stroupe, C., Orr, A., Douville, D. & Wickner, W. T. Membranes linked by trans-SNARE complexes require lipids prone to non-bilayer structure for progression to fusion. Elife 3, e01879 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  61. Budai, Z. et al. Impaired skeletal muscle development and regeneration in transglutaminase 2 knockout mice. Cells 10, 3089 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Quach, N. L., Biressi, S., Reichardt, L. F., Keller, C. & Rando, T. A. Focal adhesion kinase signaling regulates the expression of caveolin 3 and β1 integrin, genes essential for normal myoblast fusion. Mol. Biol. Cell 20, 3422–3435 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Horsley, V. et al. Regulation of the growth of multinucleated muscle cells by an NFATC2-dependent pathway. J. Cell Biol. 153, 329–338 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Tran, T. H., Shi, X., Zaia, J. & Ai, X. Heparan sulfate 6-O-endosulfatases (Sulfs) coordinate the Wnt signaling pathways to regulate myoblast fusion during skeletal muscle regeneration. J. Biol. Chem. 287, 32651–32664 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Meyers, R. M. et al. Computational correction of copy number effect improves specificity of CRISPR–Cas9 essentiality screens in cancer cells. Nat. Genet. 49, 1779–1784 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Dawaliby, R. et al. Phosphatidylethanolamine is a key regulator of membrane fluidity in eukaryotic cells. J. Biol. Chem. 291, 3658–3667 (2016).

    Article  CAS  PubMed  Google Scholar 

  67. Mitsuhashi, S. et al. A congenital muscular dystrophy with mitochondrial structural abnormalities caused by defective de novo phosphatidylcholine biosynthesis. Am. J. Hum. Genet. 88, 845–851 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Sher, R. B. et al. A rostrocaudal muscular dystrophy caused by a defect in choline kinase beta, the first enzyme in phosphatidylcholine biosynthesis. J. Biol. Chem. 281, 4938–4948 (2006).

    Article  CAS  PubMed  Google Scholar 

  69. De Winter, J. et al. PCYT2 mutations disrupting etherlipid biosynthesis: phenotypes converging on the CDP-ethanolamine pathway. Brain 144, e17 (2021).

    Article  PubMed  Google Scholar 

  70. Shavlakadze, T. et al. Age-related gene expression signature in rats demonstrate early, late, and linear transcriptional changes from multiple tissues. Cell Rep. 28, 3263–3273 (2019).

    Article  CAS  PubMed  Google Scholar 

  71. Cree, M. G. et al. Intramuscular and liver triglycerides are increased in the elderly. J. Clin. Endocrinol. Metab. 89, 3864–3871 (2004).

    Article  CAS  PubMed  Google Scholar 

  72. Pavlovic, Z. & Bakovic, M. Regulation of phosphatidylethanolamine homeostasis-the critical role of CTP:phosphoethanolamine cytidylyltransferase (Pcyt2). Int. J. Mol. Sci. 14, 2529–2550 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Leonardi, R., Frank, M. W., Jackson, P. D., Rock, C. O. & Jackowski, S. Elimination of the CDP-ethanolamine pathway disrupts hepatic lipid homeostasis. J. Biol. Chem. 284, 27077–27089 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Guyenet, S. J. et al. A simple composite phenotype scoring system for evaluating mouse models of cerebellar ataxia. J. Vis. Exp. https://doi.org/10.3791/1787 (2010).

  75. Halbert, C. L., Allen, J. M. & Chamberlain, J. S. AAV6 vector production and purification for muscle gene therapy. Methods Mol. Biol. 1687, 257–266 (2018).

    Article  CAS  PubMed  Google Scholar 

  76. Sezgin, E. et al. Elucidating membrane structure and protein behavior using giant plasma membrane vesicles. Nat. Protoc. 7, 1042–1051 (2012).

    Article  CAS  PubMed  Google Scholar 

  77. Elsayad, K. et al. Mechanical properties of cellulose fibers measured by Brillouin spectroscopy. Cellulose 27, 4209–4220 (2020).

    Article  CAS  Google Scholar 

  78. Xiao, S., Weiner, A. M. & Lin, C. A dispersion law for virtually imaged phased-array spectral dispersers based on paraxial wave theory. IEEE J. Quantum Electron. https://doi.org/10.1109/JQE.2004.825210 (2004).

  79. Lefebvre, R., Pouvreau, S., Collet, C., Allard, B. & Jacquemond, V. Whole-cell voltage clamp on skeletal muscle fibers with the silicone-clamp technique. Methods Mol. Biol. 1183, 159–170 (2014).

    Article  PubMed  Google Scholar 

  80. Kutchukian, C. et al. Phosphatidylinositol 3-kinase inhibition restores Ca2+ release defects and prolongs survival in myotubularin-deficient mice. Proc. Natl Acad. Sci. USA 113, 14432–14437 (2016).

Download references

Acknowledgements

The authors thank the participants and their families for participating in the study. We thank all members of our laboratories for helpful discussions. We are grateful to Vienna BioCenter Core Facilities: Mouse Phenotyping Unit, Histopathology Unit, Bioinformatics Unit, BioOptics Unit, Electron Microscopy Unit and Comparative Medicine Unit. We are grateful to the Lipidomics Facility, and K. Klavins and T. Hannich at the CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences for assistance with lipidomics analysis. We also thank T. Huan and A. Hui (UBC Vancouver) for mouse tissue and mitochondria lipidomics analysis. We thank A. Klymchenko (Laboratoire de Bioimagerie et Pathologies Université de Strasbourg, Strasbourg, France) for providing the NR12S probe. We are thankful to the Sen. Paul D. Wellstone Muscular Dystrophy Cooperative Specialized Research Center Viral Vector Core Facility for AAV6 production. We also thank K. P. Campbell and M. E. Anderson (University of Iowa, Carver College of Medicine) for advice on muscle tissue handling. We thank A. Al-Qassabi from the Sultan Qaboos University for the clinical assessment of the participants. D.C. and J.M.P. are supported by the Austrian Federal Ministry of Education, Science and Research, the Austrian Academy of Sciences, and the City of Vienna, and grants from the Austrian Science Fund (FWF) Wittgenstein award (Z 271-B19), the T. von Zastrow Foundation, and a Canada 150 Research Chairs Program (F18-01336). J.S.C. is supported by grants RO1AR44533 and P50AR065139 from the US National Institutes of Health. C.K. is supported by a grant from the Agence Nationale de la Recherche (ANR-18-CE14-0007-01). A.V.K. is supported by European Union’s Horizon 2020 research and innovation programme under the Marie Skłodowska-Curie grant agreement no. 67544, and an Austrian Science Fund (FWF; no P-33799). A.W. is supported by Austrian Research Promotion Agency (FFG) project no 867674. E.S. is supported by a SciLifeLab fellowship and Karolinska Institutet Foundation Grants. Work in the laboratory of G.S.-F. is supported by the Austrian Academy of Sciences, the European Research Council (ERC AdG 695214 GameofGates) and the Innovative Medicines Initiative 2 Joint Undertaking (grant agreement no. 777372, ReSOLUTE). S.B., M.L. and R.Y. acknowledge the support of the Spastic Paraplegia Foundation.

Author information

Authors and Affiliations

Authors

Contributions

D.C. and J.M.P. designed and supervised the mouse study and wrote the manuscript with input from the co-authors. All experiments were performed and established by D.C. with the following exceptions: K.E. performed BLSM measurements. E.S. performed GPMV isolation and image analysis with assistance from D.C. E.K. performed synergic ablation assay and analysis under supervision form Z.R. F.T. performed treadmill experiment with assistance from D.C. and under supervision from Z.R. R.Y. collected and analysed zebrafish models under supervision from M.L. L.X.H. and V.S. performed lipidomics analysis under supervision of G.S.-F. A.T. and S.G. performed Pcyt2 enzyme activity analysis under supervision of M.B. A.A. performed in vivo brown fat activity and Ucp1 RT–PCR analysis under supervision from C. Knauf. A.W. performed respiration analysis under supervision from A. Kozlov. M.O. performed and analysed mouse calorimetry experiments. C. Kutchukian. and C.S. performed myofibre calcium kinetics experiment, with analysis and supervision under V.J. S.J.F.C. assisted with western blot experiments. M.N. performed bioinformatic analysis of RNA sequencing and of efficiency of Pcyt2 deletion in mice. A. Kavirayani performed myositis scoring. N.D.M. performed atomic force microscopy measurements and data analysis with assistance from D.C. T.S. and B.H. assisted in histological analysis. L.H. performed AAV6 intravenous injections. A.H. assisted in tissue sampling for western blot experiments. S.J. provided Pcyt2-floxed mice. E.R. and T.G. collected human muscle biopsy samples. J.S.C. generated the AAV6 vector and provided guidance with all AAV experiments. J.M., F.A.-M. and S.B. identified PCYT2 human mutant carriers, collected growth data and generated growth curves.

Corresponding authors

Correspondence to Domagoj Cikes, Kareem Elsayad or Josef M. Penninger.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Metabolism thanks the anonymous reviewers for their contribution to the peer review of this work. Primary Handling Editor: Isabella Samuelson, in collaboration with the Nature Metabolism team

Additional information

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

Extended data

Extended Data Fig. 1 PE synthesis pathways and EPT1 rare disease mutation carriers.

(A) Schematic diagram of phosphatidylcholines (PC), phosphatidylethanolamines (PE) and phosphatidylserine (PS) phospholipids synthesis. EK-Ethanolamine kinase; PCYT2- CTP:phosphoethanolamine cytidylyltransferase; EPT1- ethanolaminephosphotransferase 1; PSS2- Phosphatidylserine Synthase 2; PSD- Phosphatidylserine decarboxylase; CK- Choline kinase; PCYT1- Choline-phosphate cytidylyltransferase; CEPT1- Choline/ethanolaminephosphotransferase 1; PSS1- Phosphatidylserine Synthase 1. (B) Height and weight gains of three patients (#1 male, #2 female, #3 male) carrying the homozygous missense variant c.335 G > C (p.Arg112Pro) in the EPT1 gene. Controls indicate WHO standards of median weights and heights at the respective ages +/− 2 standard deviations (SD).

Source data

Extended Data Fig. 2 Analysis of Pcyt2 deletion in mice.

(A) Schematic diagram of exon 2 deletion in Myf5Cre-Pcyt2 male mice and confirmation by RNA sequencing. Exon and introns structures as well as LoxP sites targeted to exon 2 and loss of exon 2 upon Cre-mediated recombination are shown for the murine Pcyt2 locus. n = 3 animals per group.

Extended Data Fig. 3 Characterization of Myf5Cre-Pcyt2 mice.

(A) Body weights of control and Myf5Cre-Pcyt2 male mice at P1 and P4. (B) Body length gains of control and Myf5Cre-Pcyt2 male mice. n = 6 per group for body length analysis. (C) Body weights of 2 months old control and Myf5Cre-Pcyt2 female mice. (D) Body lengths of 2 months old control and Myf5Cre-Pcyt2 female mice. (E) Skeletal muscle and tissue weight isolated from (E) 10 day old control (n = 6) and Myf5Cre-Pcyt2 (n = 8) and (F) 2 months old (P56) control (n = 8) and Myf5Cre-Pcyt2 (n = 7) littermate male mice. QA, quadriceps; GC, gastrocnemius; TA, tibialis anterior muscles. Liver and spleen weights are shown as controls. Scale bars 1 cm. (G, H) Gross skeletal muscle appearance of 56 days old control and Myf5Cre-Pcyt2 male littermates. (I) mRNA TPM levels of enzymes from the PE and PC branch of the phospholipid synthesis Kennedy pathway. n = 3 mice per group. Data are shown as means ± SEM. Data are shown as means ± SEM. Each dot represents data point from individual mice unless stated otherwise. *p < 0.05, **p < 0.01, ***p < 0.001, and ****p < 0.0001, n.s. not significant (unpaired Student t-test).

Source data

Extended Data Fig. 4 Myoblast proliferation assessment in Myf5Cre-Pcyt2 mice.

(A) Representative images and quantification of BrdU labelled quadriceps from 2 days old control and Myf5Cre-Pcyt2 male mice. Images were taken under 5x magnification, and ≥2000nuclei were counted and analysed. N = 4 animals per group. Scale bar 60 µm. (B) Representative images and quantification of EdU labelled primary myoblasts in cell culture isolated from control and Myf5Cre-Pcyt2 male mice. 18 biological replicate cultures from 3 independent isolations were analysed and images were taken under 5x magnification. ≥100 nuclei counted per each culture. Each dot represents the number of EdU positive cells per each culture. Scale bar 50 µm. (C) Number of Pax7 positive nuclei in quadriceps from 6 months old male control (n = 5) and Myf5Cre-Pcyt2 mice (n = 4). ≥100 nuclei per each individual section from each mouse were counted. Scale bar 50 µm. Data are shown as means ± SEM. Unpaired Student t-test with Welch correction was used for statistical analysis. Each dot represents the number of EdU positive cells per each culture. Scale bar 50 µm. (C) Number of Pax7 positive nuclei in quadriceps from 6 months old control (n = 5) and Myf5Cre-Pcyt2 mice (n = 4). ≥100 nuclei per each individual section from each mouse were counted. Scale bar 50 µm. Data are shown as means ± SEM. Unpaired Student t-test with Welch correction was used for statistical analysis.

Source data

Extended Data Fig. 5 Myofiber type distribution in skeletal muscle of Myf5Cre-Pcyt2 mice.

(A) Western blot analysis of critical regulators of protein synthesis and translation S6K1 and 4E-BP1 in overloaded M. plantaris from Control and Myf5Cre-Pcyt2 male mice. Each lane represents individual mice. Two-Way ANOVA with multiple comparison followed by Bonferroni correction was used for statistical analysis. (B) Representative images and quantification of MyHC!, MyhCIIA and MyHCIIB fibres in skeletal muscle (quadriceps) from 6 months old control and Myf5Cre-Pcyt2 male mice. Images were taken under 5x magnification, and ≥100 myofibers were counted at 3 different matching histological areas. N = 4 animals per group. Scale bar 500 µm. (C) Representative images and quantification of oxidative and glycolytic fibres in skeletal muscle (quadriceps) from 6 months old control and Myf5Cre-Pcyt2 male mice. Images were taken under 10x magnification, and ≥1000 myofibers were counted at matching histological areas. N = 5 animals per group. Scale bar 500 µm. (D) Total number of fibres in skeletal muscle (quadriceps) from 6 months old control and Myf5Cre-Pcyt2 male mice. Images were taken under 2.5x magnification. N = 5 animals per group. Scale bar 500 µm. Data are shown as means ± SEM. Unless otherwise stated, unpaired Student t-test with Welch correction was used for statistical analysis.

Source data

Extended Data Fig. 6 Muscle inflammation and metabolic assessment of Myf5Cre-Pcyt2 mice.

(A) Grip strength of 6 months old control and Myf5Cre-Pcyt2 females. Each dot represents one mouse, values are average of three measurements per mouse. (B) Representative electron microscopy images of quadriceps of 15 months old control and Myf5Cre-Pcyt2 male mice. Note accumulation of tubular aggregates in the mutant animals (red arrows). Representative images of 3 animals per group are shown. Scale bar 2 µm. (C) Characterization of muscle inflammation in 12 months old Myf5Cre-Pcyt2 male mice. Helper T cells (CD4+) and cytotoxic T cells (CD8+) are shown. Scale bar 100 µm for H&E stained and 50 µm for immune cell staining. Representative staining of 3 animals per group are shown. (D) Inflammatory cytokine levels in the quadriceps of 12 months old Myf5Cre-Pcyt2 male mice. (E) Fed blood glucose levels on normal chow diet of 8 months old control and Myf5Cre-Pcyt2 male mice. (F) Food consumption analysis of 6 month and 8 months old control and Myf5Cre-Pcyt2 mice (G) Cage activity of 6 months old male control and Myf5Cre-Pcyt2 mice (n = 12 per group). Multiple ANOVA was used to analyse the data. (H) Energy expenditure of 6 months old male control and Myf5Cre-Pcyt2 mice during the resting (light) and active (dark) phases. (I) Cage activity under thermoneutrality of 6 months old male control and Myf5Cre-Pcyt2 mice (n = 6 per group). Multiple ANOVA was used to analyse the data. (J) Energy expenditure of 6 months old male control and Myf5Cre-Pcyt2 mice during the resting (light) and active (dark) phases under thermoneutrality. (K) Grip strength assessment of 6 months old male control and Myf5Cre-Pcyt2 mice under thermoneutrality. Data are shown as means ± SEM. Each dot represents data point from individual mice unless stated otherwise. *p < 0.05, **p < 0.01, ***p < 0.001, and ****p < 0.0001, n.s. not significant. Unpaired Student t-test with Welch correction was used for statistical analysis unless stated otherwise.

Source data

Extended Data Fig. 7 Characterization of the brown adipose tissue from Myf5Cre-Pcyt2 mice.

(A, B) Lipidomics analyses from brown fat isolated from 10-day old Myf5Cre -Pcyt2 and control male mice. n = 4 per group. (C) Brown fat differentiation in lipid free conditions from 2-day old primary pre-adipocytes isolated from control and Myf5Cre -Pcyt2 male mice. Scale bar 50 µm. (D, E) Brown fat activity as addressed by exposure of 6-month-old control and Myf5Cre-Pcyt2 male mice to cold (4C) or during fasting. (F) Ucp1 mRNA levels in brown fat of 6-month-old control and Myf5Cre-Pcyt2 male mice. (G) Mitochondrial content in brown adipose tissue. (H) BAT mitochondrial structure of 6-month-old Myf5Cre-Pcyt2 male mice. Representative images of 3 animals per group are shown. Scale bar 1 µm. (I, J) Complex I and II activities of brown fat mitochondria. Paired Student t-test was used to analyse the data. Data are shown as means ± SEM. Each dot represents data point from individual mice unless stated otherwise. *p < 0.05, **p < 0.01, ***p < 0.001, and ****p < 0.0001, n.s. not significant (unpaired Student t-test, unless otherwise stated).

Source data

Extended Data Fig. 8 Specific inactivation of Pcyt2 in multiple mouse tissues.

(A) Schematic diagram to generate adipose tissue specific Pcyt2 deficient male mice (AdipoQCre-Pcyt2). Credit: IMP-IMBA Graphics Department. (B) Body weights and appearances of 6 months old control and AdipoQCre-Pcyt2 male mice. (C) Fasting blood glucose of 6 months old control and AdipoQCre-Pcyt2 male littermates fed a chow diet. (D) Schematic diagram of motor neuron specific Pcyt2 deficient male mice (Mnx1Cre-Pcyt2). Credit: IMP-IMBA Graphics Department. (E) Body weights of 8 months old control and Mnx1Cre-Pcyt2 male mice. (F) Absence of any overt clasping behaviour and appearance in 8 months old Mnx1Cre-Pcyt2 male mice. (G) Schematic diagram of intestine epithelium specific Pcyt2 deficient male mice (VilinCre-Pcyt2). Credit: IMP-IMBA Graphics Department. (H) Body weights of 6 months old control and VilinCre-Pcyt2 littermates. (I) Histological sections of intestine isolated from 12 months old control and VilinCre-Pcyt2 male mice. Scale bar 100 μm. (J) Schematic diagram of skin epithelium Pcyt2 deficient male mice (K14Cre-Pcyt2). Representative images of 3 animals per group are shown. Credit: IMP-IMBA Graphics Department. (K) Body weights and appearances of 6 months old control and K14Cre-Pcyt2 male mice. (L) Histological sections of skin isolated from 12 months old control and K14Cre-Pcyt2 male littermates. Representative images of 3 animals per group are shown. Scale bar 100 μm. (M) Schematic diagram of mature muscle specific Pcyt2 deficient male mice (MCKCre-Pcyt2). Credit: IMP-IMBA Graphics Department. (N) Grip strength of 18 months old control and muscle specific MckCre-Pcyt2 male mice. Data are shown as means ± SEM. Each dot represents individual mice, each mouse was tested in triplicates. Mean values ± SEM are displayed. *p < 0.05, **p < 0.01, ***p < 0.001, and ****p < 0.0001, n.s. not significant (unpaired Student t-test).

Source data

Extended Data Fig. 9 Assessment of mitochondrial homeostasis and SS-31 treatment.

(A, B) Muscle mitochondrial function assessed by measurements of complex II linked activity on isolated mitochondria from 2 months and from (C) 6 months old control and Myf5Cre-Pcyt2 male mice respectively. Paired Student t-test was used for statistical analysis. (D, E) Ultrastructure and total numbers of muscle mitochondria from 8 months -old control (n = 6) and Myf5Cre-Pcyt2 male mice (n = 6). Scale bar 200 nm. Unpaired Student t-test with Welch correction was used for statistical analysis (F) Function of muscle mitochondria under increasing concentrations of phosphoethanolamine, as assessed by measurements of complex I linked activity on isolated mitochondria from 2 months old control and Myf5Cre-Pcyt2 male mice. N = 3 mice per group. Two-Way ANOVA with multiple comparison followed by Bonferroni correction was used for statistical analysis. (G, H) Grip strength and organ weight measurements of 6 months old control (vehicle) and Myf5Cre-Pcyt2 male mice that have been treated with either vehicle or ss-31 compound for two months. Credit: IMP-IMBA Graphics Department. Data are shown as means ± SEM. Multiple comparison One-Way ANOVA with Dunnett correction was used for statistical analysis.

Source data

Extended Data Fig. 10 Assessment of calcium handling, and autophagy markers in skeletal muscle.

(A) Representative images of SR ultrastructure in skeletal muscles from 6 months old male control and Myf5Cre-Pcyt2 mice. Scale bar 200 nm. (B) Voltage-dependence of the peak rate of sarcoplasmic reticulum (SR) Ca2+ release (d[CaTot]/dt) measured from rhod-2 Ca2+ transients in fibres from male control (n = 6 mice and 23 myofibres) and Myf5Cre-Pcyt2 (n = 5 mice and 21 myofibres). (C) Decline of voltage-activated fluo-4FF Ca2+ transients in muscle fibers from control (n = 2 mice and 6 myofibers) and Myf5Cre-Pcyt2 (n = 3 mice and 8 myofibers) in response to an exhausting voltage stimulation protocol. (D) LC3 I/II and p62 levels in quadriceps from 8 months old control and Myf5Cre-Pcyt2 male mice under fed and fasting (24 h) conditions. N = 3 mice per group. (E) LC3 I/II and p62 levels in diaphragm from 8 months old control and Myf5Cre-Pcyt2 male mice under fed and fasting (24 h) conditions. N = 3 mice per group. (F, G) Quantification of p62 levels under fed and fasting conditions from quadriceps and diaphragm muscle respectively. Each dot represents individual mice. Data are shown as means ± SEM. Unpaired Student t-test with Welch correction was used for statistical analysis.

Source data

Supplementary information

Supplementary Information

Supplementary Methods and Supplementary Figs. 1–8

Reporting Summary

Laser-induced myofiber damage assessment on myofibres isolated 6-month-old control mice.

Laser-induced myofibre damage assessment on myofibres isolated 6-month-old control mice.

Laser-induced myofibre damage assessment on myofibres isolated 6-month-old Myf5Cre Pcyt2 mice.

Laser-induced myofibre damage assessment on myofibres isolated 6-month-old Myf5Cre Pcyt2 mice.

Video of the last bout of eccentric exercise (day 13) of 6-month-old control and Myf5Cre-Pcyt2 mice. First, mice were adjusted to a lower speed (9 m min−1; t = 0–10 s), followed by a higher speed bout (20 m min−1; t = 10–40 s).

Supplementary Data 1

Statistical source data for Supplementary Fig. 4.

Supplementary Data 2

Statistical source data for Supplementary Fig. 5.

Supplementary Data 3

Statistical source data for Supplementary Fig. 7.

Supplementary Data 4

Statistical source data for Supplementary Fig. 8.

Supplementary Data 5

Untargeted lipidome analysis of skeletal muscle at P10.

Supplementary Data 6

Untargeted lipidome analysis of BAT at P10.

Supplementary Data 7

Untargeted lipidome analysis of skeletal muscle mitochondria.

Supplementary Data 8

Untargeted lipidome analysis of young and old skeletal muscle.

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 4

Unprocessed western blots.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Fig. 7

Statistical source data.

Source Data Fig. 7

Unprocessed western blots.

Source Data Fig. 8

Statistical source data.

Source Data Fig. 8

Unprocessed western blots.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 5

Unprocessed western blots.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 9

Statistical source data.

Source Data Extended Data Fig. 10

Statistical source data.

Source Data Extended Data Fig. 10

Unprocessed western blots.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Cikes, D., Elsayad, K., Sezgin, E. et al. PCYT2-regulated lipid biosynthesis is critical to muscle health and ageing. Nat Metab 5, 495–515 (2023). https://doi.org/10.1038/s42255-023-00766-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-023-00766-2

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