Skip to main content
Log in

Brain histone beta-hydroxybutyrylation couples metabolism with gene expression

  • Original Article
  • Published:
Cellular and Molecular Life Sciences Aims and scope Submit manuscript

Abstract

Little is known about the impact of metabolic stimuli on brain tissue at a molecular level. The ketone body beta-hydroxybutyrate (BHB) can be a signaling molecule regulating gene transcription. Thus, we assessed lysine beta-hydroxybutyrylation (K-bhb) levels in proteins extracted from the cerebral cortex of mice undergoing a ketogenic metabolic challenge (48 h fasting). We found that fasting enhanced K-bhb in a variety of proteins including histone H3. ChIP-seq experiments showed that K9 beta-hydroxybutyrylation of H3 (H3K9-bhb) was significantly enriched by fasting on more than 8000 DNA loci. Transcriptomic analysis showed that H3K9-bhb on enhancers and promoters correlated with active gene expression. One of the most enriched functional annotations both at the epigenetic and transcriptional level was “circadian rhythms''. Indeed, we found that the diurnal oscillation of specific transcripts was modulated by fasting at distinct zeitgeber times both in the cortex and suprachiasmatic nucleus. Moreover, specific changes in locomotor activity daily features were observed during re-feeding after 48-h fasting. Thus, our results suggest that fasting remarkably impinges on the cerebral cortex transcriptional and epigenetic landscape, and BHB acts as a powerful epigenetic molecule in the brain through direct and specific histone marks remodeling in neural tissue cells.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7

Similar content being viewed by others

Data availability

RNA-seq and ChIP-seq datasets generated and analyzed during this study are available on the GEO database: accession number GSE168725.

References

  1. Gómez-Pinilla F (2008) Brain foods: the effects of nutrients on brain function. Nat Rev Neurosci 9:568–578

    Article  Google Scholar 

  2. Mattson MP, Moehl K, Ghena N et al (2018) Intermittent metabolic switching, neuroplasticity and brain health. Nat Rev Neurosci 19:80–80

    Article  Google Scholar 

  3. Tognini P, Samad M, Kinouchi K et al (2020) Reshaping circadian metabolism in the suprachiasmatic nucleus and prefrontal cortex by nutritional challenge. Proc Natl Acad Sci USA 117:29904–29913

    Article  CAS  Google Scholar 

  4. Padamsey Z, Katsanevaki D, Dupuy N, Rochefort NL (2022) Neocortex saves energy by reducing coding precision during food scarcity. Neuron 110:280-296.e10

    Article  CAS  Google Scholar 

  5. Dixon JB, Browne JL, Lambert GW et al (2013) Severely obese people with diabetes experience impaired emotional well-being associated with socioeconomic disadvantage: results from diabetes MILES—Australia. Diabetes Res Clin Pract 101:131–140

    Article  Google Scholar 

  6. Dutheil S, Ota KT, Wohleb ES et al (2016) High-fat diet induced anxiety and anhedonia: impact on brain homeostasis and inflammation. Neuropsychopharmacology 41:1874–1887

    Article  CAS  Google Scholar 

  7. Walsh ME, Shi Y, Van Remmen H (2014) The effects of dietary restriction on oxidative stress in rodents. Free Radical Biol Med 66:88–99

    Article  CAS  Google Scholar 

  8. Luo H, Chiang H-H, Louw M et al (2017) Nutrient sensing and the oxidative stress response. Trends Endocrinol Metab 28:449–460

    Article  CAS  Google Scholar 

  9. Lutas A, Yellen G (2013) The ketogenic diet: metabolic influences on brain excitability and epilepsy. Trends Neurosci 36:32–40

    Article  CAS  Google Scholar 

  10. Evangeliou A, Vlachonikolis I, Mihailidou H et al (2003) Application of a ketogenic diet in children with autistic behavior: pilot study. J Child Neurol 18:113–118

    Article  Google Scholar 

  11. Li Q, Liang J, Fu N et al (2021) A ketogenic diet and the treatment of autism spectrum disorder. Front Pediatr 9:650624

    Article  Google Scholar 

  12. van der Louw E, van der Louw E, van den Hurk D et al (2016) Ketogenic diet guidelines for infants with refractory epilepsy. Eur J Paediatr Neurol 20:798–809

    Article  Google Scholar 

  13. Paoli A, Rubini A, Volek JS, Grimaldi KA (2013) Beyond weight loss: a review of the therapeutic uses of very-low-carbohydrate (ketogenic) diets. Eur J Clin Nutr 67:789–796

    Article  CAS  Google Scholar 

  14. Murakami M, Tognini P (2022) Molecular mechanisms underlying the bioactive properties of a ketogenic diet. Nutrients. https://doi.org/10.3390/nu14040782

    Article  Google Scholar 

  15. Lee J, Duan W, Mattson MP (2002) Evidence that brain-derived neurotrophic factor is required for basal neurogenesis and mediates, in part, the enhancement of neurogenesis by dietary restriction in the hippocampus of adult mice. J Neurochem 82:1367–1375

    Article  CAS  Google Scholar 

  16. Vivar C, Potter MC, Choi J et al (2012) Monosynaptic inputs to new neurons in the dentate gyrus. Nat Commun 3:1107

    Article  Google Scholar 

  17. Landry T, Huang H (2021) Mini review: The relationship between energy status and adult hippocampal neurogenesis. Neurosci Lett 765:136261

    Article  CAS  Google Scholar 

  18. Pizzorusso T, Tognini P (2020) Interplay between metabolism, nutrition and epigenetics in shaping brain DNA methylation, neural function and behavior. Genes. https://doi.org/10.3390/genes11070742

    Article  Google Scholar 

  19. Newman JC, Verdin E (2014) Ketone bodies as signaling metabolites. Trends Endocrinol Metab 25:42–52

    Article  CAS  Google Scholar 

  20. Kimura I, Inoue D, Maeda T et al (2011) Short-chain fatty acids and ketones directly regulate sympathetic nervous system via G protein-coupled receptor 41 (GPR41). Proc Natl Acad Sci 108:8030–8035

    Article  CAS  Google Scholar 

  21. Taggart AKP, Kero J, Gan X et al (2005) (d)-β-Hydroxybutyrate inhibits adipocyte lipolysis via the nicotinic acid receptor PUMA-G. J Biol Chem 280:26649–26652

    Article  CAS  Google Scholar 

  22. Won Y-J, Lu VB, Puhl HL 3rd, Ikeda SR (2013) β-Hydroxybutyrate modulates N-type calcium channels in rat sympathetic neurons by acting as an agonist for the G-protein-coupled receptor FFA3. J Neurosci 33:19314–19325

    Article  CAS  Google Scholar 

  23. Shimazu T, Hirschey MD, Newman J et al (2013) Suppression of oxidative stress by β-hydroxybutyrate, an endogenous histone deacetylase inhibitor. Science 339:211–214

    Article  CAS  Google Scholar 

  24. Tognini P, Murakami M, Liu Y et al (2017) Distinct circadian signatures in liver and gut clocks revealed by ketogenic diet. Cell Metab 26:523-538.e5

    Article  CAS  Google Scholar 

  25. Xie Z, Zhang D, Chung D et al (2016) Metabolic regulation of gene expression by histone lysine β-hydroxybutyrylation. Mol Cell 62:194–206

    Article  CAS  Google Scholar 

  26. Hashimoto T, Cook WS, Qi C et al (2000) Defect in peroxisome proliferator-activated receptor alpha-inducible fatty acid oxidation determines the severity of hepatic steatosis in response to fasting. J Biol Chem 275:28918–28928

    Article  CAS  Google Scholar 

  27. Koubi HE, Robin JP, Dewasmes G et al (1991) Fasting-induced rise in locomotor activity in rats coincides with increased protein utilization. Physiol Behav 50:337–343

    Article  CAS  Google Scholar 

  28. Koronowski KB, Greco CM, Huang H et al (2021) Ketogenesis impact on liver metabolism revealed by proteomics of lysine β-hydroxybutyrylation. Cell Rep 36:109487

    Article  CAS  Google Scholar 

  29. Kobayashi Y, Ye Z, Hensch TK (2015) Clock genes control cortical critical period timing. Neuron 86:264–275

    Article  CAS  Google Scholar 

  30. Pizzorusso T, Medini P, Berardi N et al (2002) Reactivation of ocular dominance plasticity in the adult visual cortex. Science 298:1248–1251

    Article  CAS  Google Scholar 

  31. Wu D, Smyth GK (2012) Camera: a competitive gene set test accounting for inter-gene correlation. Nucleic Acids Res 40:e133–e133

    Article  CAS  Google Scholar 

  32. Guo JU, Ma DK, Mo H et al (2011) Neuronal activity modifies the DNA methylation landscape in the adult brain. Nat Neurosci 14:1345–1351

    Article  CAS  Google Scholar 

  33. Choi M, Wang SE, Ko SY et al (2014) Overexpression of human GATA-1 and GATA-2 interferes with spine formation and produces depressive behavior in rats. PLoS ONE 9:e109253

    Article  Google Scholar 

  34. Li Z, Cogswell M, Hixson K et al (2018) Nuclear Respiratory Factor 1 (NRF-1) controls the activity dependent transcription of the GABA-A receptor beta 1 subunit gene in neurons. Front Mol Neurosci 11:285

    Article  Google Scholar 

  35. Pereira JD, Sansom SN, Smith J et al (2010) Ezh2, the histone methyltransferase of PRC2, regulates the balance between self-renewal and differentiation in the cerebral cortex. Proc Natl Acad Sci U S A 107:15957–15962

    Article  CAS  Google Scholar 

  36. Swiss VA, Casaccia P (2010) Cell-context specific role of the E2F/Rb pathway in development and disease. Glia 58:377–390

    Google Scholar 

  37. Veyrac A, Besnard A, Caboche J et al (2014) The transcription factor Zif268/Egr1, brain plasticity, and memory. Prog Mol Biol Transl Sci 122:89–129

    Article  CAS  Google Scholar 

  38. Martin KC, Barad M, Kandel ER (2000) Local protein synthesis and its role in synapse-specific plasticity. Curr Opin Neurobiol 10:587–592

    Article  CAS  Google Scholar 

  39. Hartsock MJ, Spencer RL (2020) Memory and the circadian system: identifying candidate mechanisms by which local clocks in the brain may regulate synaptic plasticity. Neurosci Biobehav Rev 118:134–162

    Article  Google Scholar 

  40. Kinouchi K, Magnan C, Ceglia N et al (2018) Fasting imparts a switch to alternative daily pathways in liver and muscle. Cell Rep 25:3299-3314.e6

    Article  CAS  Google Scholar 

  41. Vollmers C, Gill S, DiTacchio L et al (2009) Time of feeding and the intrinsic circadian clock drive rhythms in hepatic gene expression. Proc Natl Acad Sci USA 106:21453–21458

    Article  CAS  Google Scholar 

  42. Mujica-Parodi LR, Amgalan A, Sultan SF et al (2020) Diet modulates brain network stability, a biomarker for brain aging, in young adults. Proc Natl Acad Sci USA 117:6170–6177

    Article  CAS  Google Scholar 

  43. Yang H, Shan W, Zhu F et al (2019) Ketone bodies in neurological diseases: focus on neuroprotection and underlying mechanisms. Front Neurol 10:585

    Article  Google Scholar 

  44. Simeone TA, Simeone KA, Rho JM (2017) Ketone bodies as anti-seizure agents. Neurochem Res 42:2011–2018

    Article  CAS  Google Scholar 

  45. Stephan JS, Sleiman SF (2021) Exercise factors released by the liver, muscle, and bones have promising therapeutic potential for stroke. Front Neurol 12:600365

    Article  Google Scholar 

  46. Sleiman SF, Henry J, Al-Haddad R et al (2016) Exercise promotes the expression of brain derived neurotrophic factor (BDNF) through the action of the ketone body β-hydroxybutyrate. Elife. https://doi.org/10.7554/eLife.15092

    Article  Google Scholar 

  47. Chen L, Miao Z, Xu X (2017) β-hydroxybutyrate alleviates depressive behaviors in mice possibly by increasing the histone3-lysine9-β-hydroxybutyrylation. Biochem Biophys Res Commun 490:117–122

    Article  CAS  Google Scholar 

  48. Rahman M, Muhammad S, Khan MA, et al (2014) The β-hydroxybutyrate receptor HCA2 activates a neuroprotective subset of macrophages. Nat Commun 5:3944

    Article  CAS  Google Scholar 

  49. Yin J, Han P, Tang Z et al (2015) Sirtuin 3 mediates neuroprotection of ketones against ischemic stroke. J Cereb Blood Flow Metab 35:1783–1789

    Article  CAS  Google Scholar 

  50. Telese F, Gamliel A, Skowronska-Krawczyk D et al (2013) “Seq-ing” insights into the epigenetics of neuronal gene regulation. Neuron 77:606–623

    Article  CAS  Google Scholar 

  51. Zarrinpar A, Chaix A, Panda S (2016) Daily eating patterns and their impact on health and disease. Trends Endocrinol Metab 27:69–83

    Article  CAS  Google Scholar 

  52. Tognini P, Murakami M, Sassone-Corsi P (2018) Interplay between microbes and the circadian clock. Cold Spring Harb Perspect Biol. https://doi.org/10.1101/cshperspect.a028365

    Article  Google Scholar 

  53. Eckel-Mahan K, Sassone-Corsi P (2013) Epigenetic regulation of the molecular clockwork. Prog Mol Biol Transl Sci 119:29–50

    Article  Google Scholar 

  54. Cajigas IJ, Will T, Schuman EM (2010) Protein homeostasis and synaptic plasticity. EMBO J 29:2746–2752

    Article  CAS  Google Scholar 

  55. Lupori L, Cornuti S, Mazziotti R, et al (2022) The gut microbiota of environmentally enriched mice regulates visual cortical plasticity. Cell Rep 38:110212

    Article  CAS  Google Scholar 

  56. Mottolese R, Redouté J, Costes N et al (2014) Switching brain serotonin with oxytocin. Proc Natl Acad Sci U S A 111:8637–8642

    Article  CAS  Google Scholar 

  57. Yamasue H, Domes G (2018) Oxytocin and autism spectrum disorders. Curr Top Behav Neurosci 35:449–465

    Article  CAS  Google Scholar 

  58. Borroto-Escuela DO, Ambrogini P, Chruścicka B et al (2021) The role of central serotonin neurons and 5-HT heteroreceptor complexes in the pathophysiology of depression: a historical perspective and future prospects. Int J Mol Sci. https://doi.org/10.3390/ijms22041927

    Article  Google Scholar 

  59. Weiss O, Dorfman A, Ram T et al (2017) Rats do not eat alone in public: Food-deprived rats socialize rather than competing for baits. PLoS ONE 12:e0173302

    Article  Google Scholar 

  60. Zhang X, Cao R, Niu J et al (2019) Molecular basis for hierarchical histone de-β-hydroxybutyrylation by SIRT3. Cell Discov 5:35

    Article  Google Scholar 

  61. Verdin E, Hirschey MD, Finley LWS, Haigis MC (2010) Sirtuin regulation of mitochondria: energy production, apoptosis, and signaling. Trends Biochem Sci 35:669–675

    Article  CAS  Google Scholar 

  62. Bao J, Lu Z, Joseph JJ et al (2010) Characterization of the murine SIRT3 mitochondrial localization sequence and comparison of mitochondrial enrichment and deacetylase activity of long and short SIRT3 isoforms. J Cell Biochem 110:238–247

    CAS  Google Scholar 

  63. Cooper HM, Spelbrink JN (2008) The human SIRT3 protein deacetylase is exclusively mitochondrial. Biochem J 411:279–285

    Article  CAS  Google Scholar 

  64. Hallows WC, Albaugh BN, Denu JM (2008) Where in the cell is SIRT3?—functional localization of an NAD-dependent protein deacetylase. Biochem J 411:e11–e13

    Article  CAS  Google Scholar 

  65. Huang H, Zhang D, Weng Y et al (2021) The regulatory enzymes and protein substrates for the lysine β-hydroxybutyrylation pathway. Sci Adv. https://doi.org/10.1126/sciadv.abe2771

    Article  Google Scholar 

  66. Chirichella M, Lisi S, Fantini M et al (2017) Post-translational selective intracellular silencing of acetylated proteins with de novo selected intrabodies. Nat Methods 14:279–282

    Article  CAS  Google Scholar 

  67. Patterson RE, Sears DD (2017) Metabolic effects of intermittent fasting. Annu Rev Nutr 37:371–393

    Article  CAS  Google Scholar 

  68. Sa-Nguanmoo P, Tanajak P, Kerdphoo S et al (2016) FGF21 improves cognition by restored synaptic plasticity, dendritic spine density, brain mitochondrial function and cell apoptosis in obese-insulin resistant male rats. Horm Behav 85:86–95

    Article  CAS  Google Scholar 

  69. Ferrario CR, Reagan LP (2018) Insulin-mediated synaptic plasticity in the CNS: anatomical, functional and temporal contexts. Neuropharmacology 136:182–191

    Article  CAS  Google Scholar 

  70. Silva B, Mantha OL, Schor J et al (2022) Glia fuel neurons with locally synthesized ketone bodies to sustain memory under starvation. Nat Metab 4:213–224

    Article  CAS  Google Scholar 

  71. Baldi P, Long AD (2001) A Bayesian framework for the analysis of microarray expression data: regularized t -test and statistical inferences of gene changes. Bioinformatics 17:509–519

    Article  CAS  Google Scholar 

  72. Kayala MA, Baldi P (2012) Cyber-T web server: differential analysis of high-throughput data. Nucleic Acids Res 40:W553–W559

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank Manuel Tongiani, Andrea Tognozzi, Matteo Alberti and Maria Grazia Giuliano for their help with the experiments. Special thanks to Vania Liverani and Antonella Calvello (Scuola Normale Superiore) for technical assistance in the lab. We thank Prof. Concetta Morrone and Prof. Paola Binda (University of Pisa) for their insightful comments.

Funding

This research was supported by H2020-MSCA-IF-2016 749697 GaMePLAY, University of Pisa PRA-2020, Italian Ministry of University and Research PNRR Tuscany Health Ecosystem Milestone 8.9.1 and PNRR young MSCA_0000081 iNsPIReD to PT, and funded in part by EFSD-Lilly 2019 to AG and PT. The work of SCh, MS, CM, and PB was in part supported by NIH Grant GM123558 to PB. RM was supported by Fondazione Umberto Veronesi.

Author information

Authors and Affiliations

Authors

Contributions

SC performed the experiments, analyzed the data, and prepared the figures. SCh performed the ChIP-seq and RNA-seq analysis. LL performed the daily activity analysis. FF and SR performed the Beta-hydroxybutyrilome analysis. FC, SF and AG performed the LC–MS-QTOF experiments and analysis. MS performed the RNA seq analysis. MC and FD helped with tissue harvesting and performed qPCR. FR helped with GO analysis. RM helped with correlations between ChIP-seq and RNA-seq. CM helped with ChIP-seq data alignment and analysis. PB supervised the RNA-seq and ChiP-seq data analysis. PT conceived and supervised the project, performed the experiments, and wrote the manuscript.

Corresponding author

Correspondence to Paola Tognini.

Ethics declarations

Conflict of interest

The authors have no relevant financial or non-financial interests to disclose.

Ethics approval

All experiments were carried out in accordance with the European Directives (2010/63/EU) and were approved by the Italian Ministry of Health (authorization number 354/2020-PR).

Additional information

Publisher's Note

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

Supplementary Information

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

Cornuti, S., Chen, S., Lupori, L. et al. Brain histone beta-hydroxybutyrylation couples metabolism with gene expression. Cell. Mol. Life Sci. 80, 28 (2023). https://doi.org/10.1007/s00018-022-04673-9

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1007/s00018-022-04673-9

Keywords

Navigation