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.

  • Review Article
  • Published:

The nexus of chromatin regulation and intermediary metabolism

This article has been updated

Abstract

Living organisms and individual cells continuously adapt to changes in their environment. Those changes are particularly sensitive to fluctuations in the availability of energy substrates. The cellular transcriptional machinery and its chromatin-associated proteins integrate environmental inputs to mediate homeostatic responses through gene regulation. Numerous connections between products of intermediary metabolism and chromatin proteins have recently been identified. Chromatin modifications that occur in response to metabolic signals are dynamic or stable and might even be inherited transgenerationally. These emerging concepts have biological relevance to tissue homeostasis, disease and ageing.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Figure 1: DNA methylation and post-translational modifications of histones link metabolites and transcription.
Figure 2: Metabolic pathways of intermediary metabolism signal to chromatin.
Figure 3: Distinct modes of chromatin-mediated transcriptional control by intermediary metabolism products.
Figure 4: Metabolite influencers of complex biological systems.
Figure 5: Epigenetic drift and transgenerational inheritance of disease risks.

Similar content being viewed by others

Change history

  • 10 December 2013

    The TCA cycle in Figure 2 has been corrected in the online version of this Review.

References

  1. Feinberg, A. P. Phenotypic plasticity and the epigenetics of human disease. Nature 447, 433–440 (2007).

    CAS  ADS  PubMed  Google Scholar 

  2. Li, B., Carey, M. & Workman, J. L. The role of chromatin during transcription. Cell 128, 707–719 (2007).

    CAS  PubMed  Google Scholar 

  3. Strahl, B. D. & Allis, C. D. The language of covalent histone modifications. Nature 403, 41–45 (2000). This landmark review proposes for the first time the existence of a complex histone code that encodes information as the underlying logic of epigenetic transcriptional control.

    CAS  ADS  PubMed  Google Scholar 

  4. Berger, S. L. The complex language of chromatin regulation during transcription. Nature 447, 407–412 (2007).

    CAS  ADS  PubMed  Google Scholar 

  5. Jenuwein, T. & Allis, C. D. Translating the histone code. Science 293, 1074–1080 (2001).

    CAS  PubMed  Google Scholar 

  6. Kornberg, R. D. Structure of chromatin. Annu. Rev. Biochem. 46, 931–954 (1977).

    CAS  PubMed  Google Scholar 

  7. Bell, O., Tiwari, V. K., Thomä, N. H. & Schübeler, D. Determinants and dynamics of genome accessibility. Nature Rev. Genet. 12, 554–564 (2011).

    CAS  PubMed  Google Scholar 

  8. Musselman, C. A., Lalonde, M.-E., Cote, J. & Kutateladze, T. G. Perceiving the epigenetic landscape through histone readers. Nature Struct. Mol. Biol. 19, 1218–1227 (2012).

    CAS  Google Scholar 

  9. Kouzarides, T. Chromatin modifications and their function. Cell 128, 693–705 (2007).

    CAS  PubMed  Google Scholar 

  10. Kaelin, W. G. & McKnight, S. L. Influence of metabolism on epigenetics and disease. Cell 153, 56–69 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Lu, C. & Thompson, C. B. Metabolic regulation of epigenetics. Cell Metab. 16, 9–17 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Wellen, K. E. & Thompson, C. B. A two-way street: reciprocal regulation of metabolism and signalling. Nature Rev. Mol. Cell Biol. 13, 270–276 (2012).

    CAS  Google Scholar 

  13. Katada, S., Imhof, A. & Sassone-Corsi, P. Connecting threads: epigenetics and metabolism. Cell 148, 24–28 (2012).

    CAS  PubMed  Google Scholar 

  14. Teperino, R., Schoonjans, K. & Auwerx, J. Histone methyl transferases and demethylases; can they link metabolism and transcription? Cell Metab. 12, 321–327 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Bird, A. Perceptions of epigenetics. Nature 447, 396–398 (2007).

    CAS  ADS  PubMed  Google Scholar 

  16. Sasaki, H. & Matsui, Y. Epigenetic events in mammalian germ-cell development: reprogramming and beyond. Nature Rev. Genet. 9, 129–140 (2008).

    CAS  PubMed  Google Scholar 

  17. Reik, W. Stability and flexibility of epigenetic gene regulation in mammalian development. Nature 447, 425–432 (2007).

    CAS  ADS  PubMed  Google Scholar 

  18. Hirschey, M. D. et al. SIRT3 regulates mitochondrial fatty-acid oxidation by reversible enzyme deacetylation. Nature 464, 121–125 (2010).

    CAS  ADS  PubMed  PubMed Central  Google Scholar 

  19. Newman, J. C., He, W. & Verdin, E. Mitochondrial protein acylation and intermediary metabolism: regulation by sirtuins and implications for metabolic disease. J. Biol. Chem. 287, 42436–42443 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. He, W., Newman, J. C., Wang, M. Z., Ho, L. & Verdin, E. Mitochondrial sirtuins: regulators of protein acylation and metabolism. Trends Endocrinol. Metab. 23, 467–476 (2012).

    CAS  PubMed  Google Scholar 

  21. Barski, A. et al. High-resolution profiling of histone methylations in the human genome. Cell 129, 823–837 (2007).

    CAS  PubMed  Google Scholar 

  22. Forneris, F., Binda, C., Vanoni, M. A., Mattevi, A. & Battaglioli, E. Histone demethylation catalysed by LSD1 is a flavin-dependent oxidative process. FEBS Lett. 579, 2203–2207 (2005).

    CAS  PubMed  Google Scholar 

  23. Loenarz, C. & Schofield, C. J. Expanding chemical biology of 2-oxoglutarate oxygenases. Nature Chem. Biol. 4, 152–156 (2008).

    CAS  Google Scholar 

  24. Sakabe, K., Wang, Z. & Hart, G. W. β-N-acetylglucosamine (O-GlcNAc) is part of the histone code. Proc. Natl Acad. Sci. USA 107, 19915–19920 (2010).

    CAS  ADS  PubMed  PubMed Central  Google Scholar 

  25. Hart, G. W. G., Housley, M. P. M. & Slawson, C. C. Cycling of O-linked β-N-acetylglucosamine on nucleocytoplasmic proteins. Nature 446, 1017–1022 (2007).

    CAS  ADS  PubMed  Google Scholar 

  26. Hanover, J. A. J., Krause, M. W. M. & Love, D. C. D. Bittersweet memories: linking metabolism to epigenetics through O-GlcNAcylation. Nature Rev. Mol. Cell Biol. 13, 312–321 (2012).

    CAS  Google Scholar 

  27. Fong, J. J. et al. β-N-Acetylglucosamine (O-GlcNAc) is a novel regulator of mitosis-specific phosphorylations on histone H3. J. Biol. Chem. 287, 12195–12203 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Rodgers, J. T. et al. Nutrient control of glucose homeostasis through a complex of PGC-1α and SIRT1. Nature 434, 113–118 (2005).

    CAS  ADS  PubMed  Google Scholar 

  29. Bird, A. P. Functions for DNA methylation in vertebrates. Cold Spring Harb. Symp. Quant. Biol. 58, 281–285 (1993).

    CAS  PubMed  Google Scholar 

  30. Kohli, R. M. & Zhang, Y. TET enzymes, TDG and the dynamics of DNA demethylation. Nature 502, 472–479 (2013).

    CAS  ADS  PubMed  PubMed Central  Google Scholar 

  31. Portela, A. & Esteller, M. Epigenetic modifications and human disease. Nature Biotechnol. 28, 1057–1068 (2010).

    CAS  Google Scholar 

  32. Kirchner, H., Osler, M. E., Krook, A. & Zierath, J. R. Epigenetic flexibility in metabolic regulation: disease cause and prevention? Trends Cell Biol. 23, 203–209 (2013).

    CAS  PubMed  Google Scholar 

  33. Barrès, R. et al. Non-CpG methylation of the PGC-1α promoter through DNMT3B controls mitochondrial density. Cell Metab. 10, 189–198 (2009).

    PubMed  Google Scholar 

  34. Barrès, R. et al. Weight loss after gastric bypass surgery in human obesity remodels promoter methylation. Cell Rep. 3, 1020–1027 (2013).

    PubMed  Google Scholar 

  35. Anderson, O. S., Sant, K. E. & Dolinoy, D. C. Nutrition and epigenetics: an interplay of dietary methyl donors, one-carbon metabolism and DNA methylation. J. Nutr. Biochem. 23, 853–859 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Xiao, M. et al. Inhibition of α-KG-dependent histone and DNA demethylases by fumarate and succinate that are accumulated in mutations of FH and SDH tumor suppressors. Genes Dev. 26, 1326–1338 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Ladurner, A. G. Rheostat control of gene expression by metabolites. Mol. Cell 24, 1–11 (2006).

    CAS  PubMed  Google Scholar 

  38. Shi, Y. & Shi, Y. Metabolic enzymes and coenzymes in transcription — a direct link between metabolism and transcription? Trends Genet. 20, 445–452 (2004).

    CAS  PubMed  Google Scholar 

  39. Dang, L. et al. Cancer-associated IDH1 mutations produce 2-hydroxyglutarate. Nature 462, 739–744 (2009). This breakthrough article describes the neomorphic enzyme activity of cancer-associated mutant IDH1 leading to tissue accumulation of the oncometabolite 2-hydroxyglutarate.

    CAS  ADS  PubMed  PubMed Central  Google Scholar 

  40. Albaugh, B. N., Arnold, K. M. & Denu, J. M. KAT(ching) metabolism by the tail: insight into the links between lysine acetyltransferases and metabolism. ChemBioChem 12, 290–298 (2011).

    CAS  PubMed  Google Scholar 

  41. Rardin, M. J. et al. Label-free quantitative proteomics of the lysine acetylome in mitochondria identifies substrates of SIRT3 in metabolic pathways. Proc. Natl Acad. Sci. USA 110, 6601–6606 (2013).

    CAS  ADS  PubMed  PubMed Central  Google Scholar 

  42. Hebert, A. S. et al. Calorie restriction and SIRT3 trigger global reprogramming of the mitochondrial protein acetylome. Mol. Cell 49, 186–199 (2012).

    PubMed  PubMed Central  Google Scholar 

  43. Tu, B. P. & McKnight, S. L. Metabolic cycles as an underlying basis of biological oscillations. Nature Rev. Mol. Cell Biol. 7, 696–701 (2006).

    CAS  Google Scholar 

  44. Tu, B. P. et al. Cyclic changes in metabolic state during the life of a yeast cell. Proc. Natl Acad. Sci. USA 104, 16886–16891 (2007).

    CAS  ADS  PubMed  PubMed Central  Google Scholar 

  45. Berndsen, C. E. & Denu, J. M. Catalysis and substrate selection by histone/protein lysine acetyltransferases. Curr. Opin. Struct. Biol. 18, 682–689 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Cai, L., Sutter, B. M., Li, B. & Tu, B. P. Acetyl-CoA induces cell growth and proliferation by promoting the acetylation of histones at growth genes. Mol. Cell 42, 426–437 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Takahashi, H., McCaffery, J. M., Irizarry, R. A. & Boeke, J. D. Nucleocytosolic acetyl-coenzyme a synthetase is required for histone acetylation and global transcription. Mol. Cell 23, 207–217 (2006).

    CAS  PubMed  Google Scholar 

  48. Wellen, K. E. et al. ATP-citrate lyase links cellular metabolism to histone acetylation. Science 324, 1076–1080 (2009). References 46 to 48 show that acetyl-CoA increases histone acetylation in yeast and mammalian cells when its abundance is enriched in the nucleocytosolic compartment of a cell.

    CAS  ADS  PubMed  PubMed Central  Google Scholar 

  49. Haigis, M. C. & Guarente, L. P. Mammalian sirtuins — emerging roles in physiology, aging, and calorie restriction. Genes Dev. 20, 2913–2921 (2006).

    CAS  PubMed  Google Scholar 

  50. Houtkooper, R. H., Pirinen, E. & Auwerx, J. Sirtuins as regulators of metabolism and healthspan. Nature Rev. Mol. Cell Biol. 13, 225–238 (2012).

    CAS  Google Scholar 

  51. Sassone-Corsi, P. NAD+, a circadian metabolite with an epigenetic twist. Endocrinology 153, 1–5 (2012).

    CAS  PubMed  Google Scholar 

  52. Nakahata, Y., Sahar, S., Astarita, G., Kaluzova, M. & Sassone-Corsi, P. Circadian control of the NAD+ salvage pathway by CLOCK–SIRT1. Science 324, 654–657 (2009).

    CAS  ADS  PubMed  PubMed Central  Google Scholar 

  53. Nakahata, Y. et al. The NAD+-dependent deacetylase SIRT1 modulates CLOCK-mediated chromatin remodeling and circadian control. Cell 134, 329–340 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Murayama, A. et al. Epigenetic control of rDNA loci in response to intracellular energy status. Cell 133, 627–639 (2008).

    CAS  PubMed  Google Scholar 

  55. Michishita, E. et al. SIRT6 is a histone H3 lysine 9 deacetylase that modulates telomeric chromatin. Nature 452, 492–496 (2008).

    CAS  ADS  PubMed  PubMed Central  Google Scholar 

  56. Kawahara, T. L. A. et al. SIRT6 links histone H3 lysine 9 deacetylation to NF-κB-dependent gene expression and organismal life span. Cell 136, 62–74 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Mostoslavsky, R. et al. Genomic instability and aging-like phenotype in the absence of mammalian SIRT6. Cell 124, 315–329 (2006).

    CAS  PubMed  Google Scholar 

  58. Kanfi, Y. et al. The sirtuin SIRT6 regulates lifespan in male mice. Nature 483, 218–221 (2012).

    CAS  ADS  PubMed  Google Scholar 

  59. Veech, R. L. R., Eggleston, L. V. L. & Krebs, H. A. H. The redox state of free nicotinamide-adenine dinucleotide phosphate in the cytoplasm of rat liver. Biochem. J. 115, 609–619 (1969).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Gerhart-Hines, Z. et al. The cAMP/PKA pathway rapidly activates SIRT1 to promote fatty acid oxidation independently of changes in NAD. Mol. Cell 44, 851–863 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Cantó, C. et al. The NAD+ precursor nicotinamide riboside enhances oxidative metabolism and protects against high-fat diet-induced obesity. Cell Metab. 15, 838–847 (2012).

    PubMed  PubMed Central  Google Scholar 

  62. Hubbard, B. P. et al. Evidence for a common mechanism of SIRT1 regulation by allosteric activators. Science 339, 1216–1219 (2013).

    CAS  ADS  PubMed  PubMed Central  Google Scholar 

  63. Gut, P. & Verdin, E. Rejuvenating SIRT1 activators. Cell Metab. 17, 635–637 (2013).

    CAS  PubMed  Google Scholar 

  64. Hardie, D. G. D., Ross, F. A. F. & Hawley, S. A. S. AMPK: a nutrient and energy sensor that maintains energy homeostasis. Nature Rev. Mol. Cell Biol. 13, 251–262 (2012).

    CAS  Google Scholar 

  65. Bungard, D. et al. Signaling kinase AMPK activates stress-promoted transcription via histone H2B phosphorylation. Science 329, 1201–1205 (2010).

    CAS  ADS  PubMed  PubMed Central  Google Scholar 

  66. Takahashi, K. & Yamanaka, S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126, 663–676 (2006).

    CAS  PubMed  Google Scholar 

  67. Hanna, J. H., Saha, K. & Jaenisch, R. Pluripotency and cellular reprogramming: facts, hypotheses, unresolved issues. Cell 143, 508–525 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Apostolou, E. & Hochedlinger, K. Chromatin dynamics during cellular reprogramming. Nature 502, 462–471 (2013).

    CAS  ADS  PubMed  PubMed Central  Google Scholar 

  69. Wang, J. et al. Dependence of mouse embryonic stem cells on threonine catabolism. Science 325, 435–439 (2009).

    CAS  ADS  PubMed  PubMed Central  Google Scholar 

  70. Shyh-Chang, N. et al. Influence of threonine metabolism on S-adenosylmethionine and histone methylation. Science 339, 222–226 (2013). References 69 and 70 describe the dependence of pluripotency on the generation of S -adenosylmethionine by threonine catabolism in embryonic stem cells.

    ADS  PubMed  Google Scholar 

  71. Baylin, S. B. & Jones, P. A. A decade of exploring the cancer epigenome — biological and translational implications. Nature Rev. Cancer 11, 726–734 (2011).

    CAS  Google Scholar 

  72. Vander Heiden, M. G., Cantley, L. C. & Thompson, C. B. Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science 324, 1029–1033 (2009).

    CAS  ADS  PubMed  PubMed Central  Google Scholar 

  73. Vander Heiden, M. G. et al. Evidence for an alternative glycolytic pathway in rapidly proliferating cells. Science 329, 1492–1499 (2010).

    CAS  ADS  PubMed  Google Scholar 

  74. Sturm, D. et al. Hotspot mutations in H3F3A and IDH1 define distinct epigenetic and biological subgroups of glioblastoma. Cancer Cell 22, 425–437 (2012).

    CAS  PubMed  Google Scholar 

  75. Turcan, S. et al. IDH1 mutation is sufficient to establish the glioma hypermethylator phenotype. Nature 483, 479–483 (2012).

    CAS  ADS  PubMed  PubMed Central  Google Scholar 

  76. Dang, L., Jin, S. & Su, S. M. IDH mutations in glioma and acute myeloid leukemia. Trends Mol. Med. 16, 387–397 (2010).

    CAS  PubMed  Google Scholar 

  77. Waldecker, M., Kautenburger, T., Daumann, H., Busch, C. & Schrenk, D. Inhibition of histone-deacetylase activity by short-chain fatty acids and some polyphenol metabolites formed in the colon. J. Nutr. Biochem. 19, 587–593 (2008).

    CAS  PubMed  Google Scholar 

  78. Roediger, W. E. Utilization of nutrients by isolated epithelial cells of the rat colon. Gastroenterology 83, 424–429 (1982).

    CAS  PubMed  Google Scholar 

  79. Kim, Y. S. & Milner, J. A. Dietary modulation of colon cancer risk. J. Nutr. 137, 2576S–2579S (2007).

    PubMed  Google Scholar 

  80. Donohoe, D. R. et al. The Warburg effect dictates the mechanism of butyrate-mediated histone acetylation and cell proliferation. Mol. Cell 48, 612–626 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  81. Comalada, M. et al. The effects of short-chain fatty acids on colon epithelial proliferation and survival depend on the cellular phenotype. J. Cancer Res. Clin. Oncol. 132, 487–497 (2006).

    CAS  PubMed  Google Scholar 

  82. Cahill, G. F. Fuel metabolism in starvation. Annu. Rev. Nutr. 26, 1–22 (2006).

    CAS  PubMed  Google Scholar 

  83. Kashiwaya, Y. et al. D-β-hydroxybutyrate protects neurons in models of Alzheimer's and Parkinson's disease. Proc. Natl Acad. Sci. USA 97, 5440–5444 (2000).

    CAS  ADS  PubMed  PubMed Central  Google Scholar 

  84. Maalouf, M., Rho, J. M. & Mattson, M. P. The neuroprotective properties of calorie restriction, the ketogenic diet, and ketone bodies. Brain Res. Rev. 59, 293–315 (2009).

    CAS  PubMed  Google Scholar 

  85. Shimazu, T. et al. Suppression of oxidative stress by β-hydroxybutyrate, an endogenous histone deacetylase inhibitor. Science 339, 211–214 (2013). This study identifies the ketone body β-hydroxybutyrate as an endogenous inhibitor of HDACs and protective agent against oxidative damage.

    CAS  ADS  PubMed  Google Scholar 

  86. Kucharski, R., Maleszka, J., Foret, S. & Maleszka, R. Nutritional control of reproductive status in honeybees via DNA methylation. Science 319, 1827–1830 (2008).

    CAS  ADS  PubMed  Google Scholar 

  87. Fraga, M. F. M. et al. Epigenetic differences arise during the lifetime of monozygotic twins. Proc. Natl Acad. Sci. USA 102, 10604–10609 (2005). This study in humans shows that epigenetic marks increasingly differ during adult life of monozygotic twins.

    CAS  ADS  PubMed  PubMed Central  Google Scholar 

  88. Feil, R. & Fraga, M. F. Epigenetics and the environment: emerging patterns and implications. Nature Rev. Genet. 13, 97–109 (2011).

    Google Scholar 

  89. Nathan, D. M. et al. Intensive diabetes treatment and cardiovascular disease in patients with type 1 diabetes. N. Engl. J. Med. 353, 2643–2653 (2005).

    PubMed  Google Scholar 

  90. Patel, A. et al. Intensive blood glucose control and vascular outcomes in patients with type 2 diabetes. N. Engl. J. Med. 358, 2560–2572 (2008).

    CAS  PubMed  Google Scholar 

  91. El-Osta, A. Glycemic memory. Curr. Opin. Lipidol. 23, 24–29 (2012).

    CAS  PubMed  Google Scholar 

  92. Pirola, L. et al. Genome-wide analysis distinguishes hyperglycemia regulated epigenetic signatures of primary vascular cells. Genome Res. 21, 1601–1615 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  93. Villeneuve, L. M. et al. Epigenetic histone H3 lysine 9 methylation in metabolic memory and inflammatory phenotype of vascular smooth muscle cells in diabetes. Proc. Natl Acad. Sci. USA 105, 9047–9052 (2008).

    CAS  ADS  PubMed  PubMed Central  Google Scholar 

  94. Brasacchio, D. et al. Hyperglycemia induces a dynamic cooperativity of histone methylase and demethylase enzymes associated with gene-activating epigenetic marks that coexist on the lysine tail. Diabetes 58, 1229–1236 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  95. Patti, M.-E. Intergenerational programming of metabolic disease: evidence from human populations and experimental animal models. Cell. Mol. Life Sci. 70, 1597–1608 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  96. Stein, A. D. et al. Anthropometric measures in middle age after exposure to famine during gestation: evidence from the Dutch famine. Am. J. Clin. Nutr. 85, 869–876 (2007).

    CAS  PubMed  Google Scholar 

  97. Kaati, G., Bygren, L. O., Pembrey, M. & Sjöström, M. Transgenerational response to nutrition, early life circumstances and longevity. Eur. J. Hum. Genet. 15, 784–790 (2007). References 96 and 97 are classic epidemiological studies that show a relationship between nutritional status of mothers and cardiovascular and metabolic disease risk in the following generations.

    CAS  PubMed  Google Scholar 

  98. Anderson, L. M. et al. Preconceptional fasting of fathers alters serum glucose in offspring of mice. Nutrition 22, 327–331 (2006).

    CAS  PubMed  Google Scholar 

  99. Carone, B. R. et al. Paternally induced transgenerational environmental reprogramming of metabolic gene expression in mammals. Cell 143, 1084–1096 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  100. Kersten, S. et al. Peroxisome proliferator-activated receptor α mediates the adaptive response to fasting. J. Clin. Invest. 103, 1489–1498 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  101. Ng, S.-F. et al. Chronic high-fat diet in fathers programs β-cell dysfunction in female rat offspring. Nature 467, 963–966 (2010).

    CAS  ADS  PubMed  Google Scholar 

  102. ENCODE Project Consortium. An integrated encyclopedia of DNA elements in the human genome. Nature 489, 57–74 (2012).

  103. Rando, T. A. & Chang, H. Y. Aging, rejuvenation, and epigenetic reprogramming: resetting the aging clock. Cell 148, 46–57 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  104. Olshansky, S. J. et al. A potential decline in life expectancy in the United States in the 21st century. N. Engl. J. Med. 352, 1138–1145 (2005).

    CAS  PubMed  Google Scholar 

  105. Arrowsmith, C. H., Bountra, C., Fish, P. V., Lee, K. & Schapira, M. Epigenetic protein families: a new frontier for drug discovery. Nature Rev. Drug Discov. 11, 384–400 (2012).

    CAS  Google Scholar 

  106. Helin, K. & Dhanak, D. Chromatin proteins and modifications as drug targets. Nature 502, 480–488 (2013).

    CAS  ADS  PubMed  Google Scholar 

  107. Zhang, Q., Piston, D. W. & Goodman, R. H. Regulation of corepressor function by nuclear NADH. Science 295, 1895–1897 (2002).

    CAS  ADS  PubMed  Google Scholar 

  108. Paige, J. S., Nguyen-Duc, T., Song, W. & Jaffrey, S. R. Fluorescence imaging of cellular metabolites with RNA. Science 335, 1194 (2012).

    CAS  ADS  PubMed  PubMed Central  Google Scholar 

  109. Ellis, J. M. & Wolfgang, M. J. A genetically encoded metabolite sensor for malonyl-CoA. Chem. Biol. 19, 1333–1339 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  110. Tsou, P., Bin, B., Zheng, Hsu, C.-H., Sasaki, A. T. & Cantley, L. C. A fluorescent reporter of AMPK activity and cellular energy stress. Cell Metab. 13, 476–486 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  111. Niethammer, P., Grabher, C., Look, A. T. & Mitchison, T. J. A tissue-scale gradient of hydrogen peroxide mediates rapid wound detection in zebrafish. Nature 459, 996–999 (2009).

    CAS  ADS  PubMed  PubMed Central  Google Scholar 

  112. Kenyon, C. J. The genetics of ageing. Nature 464, 504–512 (2010).

    CAS  ADS  PubMed  Google Scholar 

  113. Han, S. & Brunet, A. Histone methylation makes its mark on longevity. Trends Cell Biol. 22, 42–49 (2012).

    PubMed  Google Scholar 

  114. Greer, E. L. et al. Transgenerational epigenetic inheritance of longevity in Caenorhabditis elegans. Nature 479, 365–371 (2011).

    CAS  ADS  PubMed  PubMed Central  Google Scholar 

  115. Fontana, L., Partridge, L. & Longo, V. D. Extending healthy life span — from yeast to humans. Science 328, 321–326 (2010).

    CAS  ADS  PubMed  PubMed Central  Google Scholar 

  116. Bordone, L. & Guarente, L. Calorie restriction, SIRT1 and metabolism: understanding longevity. Nature Rev. Mol. Cell Biol. 6, 298–305 (2005).

    CAS  Google Scholar 

Download references

Acknowledgements

We thank J. Carroll for graphics, G. Howard and A.-L. Lucido for editorial assistance, V. Fonseca and P. Cruz for administrative assistance. The authors are supported by funds from the Gladstone Institutes.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Eric Verdin.

Ethics declarations

Competing interests

E.V. is a member of the scientific advisory board of SIRTRIS/GSK

Additional information

Reprints and permissions information is available at www.nature.com/reprints.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Gut, P., Verdin, E. The nexus of chromatin regulation and intermediary metabolism. Nature 502, 489–498 (2013). https://doi.org/10.1038/nature12752

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature12752

This article is cited by

Comments

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

Search

Quick links

Nature Briefing

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

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