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:

The SESAME complex regulates cell senescence through the generation of acetyl-CoA

Abstract

Acetyl-CoA is a central node in carbon metabolism and plays critical roles in regulatory and biosynthetic processes. The acetyl-CoA synthetase Acs2, which catalyses acetyl-CoA production from acetate, is an integral subunit of the serine-responsive SAM-containing metabolic enzyme (SESAME) complex, but the precise function of Acs2 within the SESAME complex remains unclear. Here, using budding yeast, we show that Acs2 within the SESAME complex is required for the regulation of telomere silencing and cellular senescence. Mechanistically, the SESAME complex interacts with the histone acetyltransferase SAS protein complex to promote histone H4K16 acetylation (H4K16ac) enrichment and the occupancy of bromodomain-containing protein, Bdf1, at subtelomeric regions. This interaction maintains telomere silencing by antagonizing the spreading of Sir2 along the telomeres, which is enhanced by acetate. Consequently, dissociation of Sir2 from telomeres by acetate leads to compromised telomere silencing and accelerated chronological ageing. In human endothelial cells, ACSS2, the ortholog of yeast Acs2, also interacts with H4K16 acetyltransferase hMOF and are required for acetate to increase H4K16ac, reduce telomere silencing and induce cell senescence. Altogether, our results reveal a conserved mechanism to connect cell metabolism with telomere silencing and cellular senescence.

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: SESAME complex is required to maintain global H4K16ac and H4K16ac localization at subtelomeric regions.
Fig. 2: The SESAME complex interacts with the SAS complex to promote H4K16ac.
Fig. 3: The SESAME complex and Sas2 co-regulate gene transcription and telomere silencing.
Fig. 4: The SESAME complex and SAS complex are required for acetate to induce H4K16ac.
Fig. 5: Acetate enhances the binding of Bdf1 at subtelomeric regions.
Fig. 6: The SESAME and SAS complexes are required for acetate to antagonize Sir2-mediated telomere silencing.
Fig. 7: Acetate accelerates chronological ageing in yeast and induce senescence in HUVEC cells.
Fig. 8: Acetate accelerates cell senescence in HUVECs dependent on ACSS2 and hMOF.

Similar content being viewed by others

Data availability

The accession numbers for the Acs2 and H4K16ac ChIP–seq datasets reported in this paper are GSE146143 and PRJNA695438. The accession number for the Pyk1, Sam1 ChIP–seq dataset is GSE72972. The accession numbers for the H4K16ac ChIP–seq dataset are GSE52339 and GSE108217. The accession numbers for the raw RNA-seq dataset reported in this paper are PRJNA612967, PRJNA674443, PRJNA673282 and GSE147765. Uncropped blots and source data are provided with this paper. All other data are available from the authors upon reasonable request.

References

  1. Sivanand, S., Viney, I. & Wellen, K. E. Spatiotemporal control of acetyl-CoA metabolism in chromatin regulation. Trends Biochem. Sci. 43, 61–74 (2018).

    Article  CAS  PubMed  Google Scholar 

  2. Mews, P. et al. Acetyl-CoA synthetase regulates histone acetylation and hippocampal memory. Nature 546, 381–386 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Marino, G. et al. Regulation of autophagy by cytosolic acetyl-coenzyme A. Mol. Cell 53, 710–725 (2014).

    Article  CAS  PubMed  Google Scholar 

  5. Yu, X., Ma, R., Wu, Y., Zhai, Y. & Li, S. Reciprocal regulation of metabolic reprogramming and epigenetic modifications in cancer. Front. Genet. 9, 394 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  6. Yu, X. & Li, S. Non-metabolic functions of glycolytic enzymes in tumorigenesis. Oncogene 36, 2629–2636 (2017).

    Article  CAS  PubMed  Google Scholar 

  7. Li, X. et al. Nucleus-translocated ACSS2 promotes gene transcription for lysosomal biogenesis and autophagy. Mol. Cell 66, 684–697 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Wu, Y. et al. Glycolysis regulates gene expression by promoting the cross-talk between H3K4me3 and H3K14ac in Saccharomyces cerevisiae. J. Genet. Genomics 46, 561–574 (2019).

    Article  PubMed  Google Scholar 

  9. Schug, Z. T. et al. Acetyl-CoA synthetase 2 promotes acetate utilization and maintains cancer cell growth under metabolic stress. Cancer Cell 27, 57–71 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. 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).

    Article  CAS  PubMed  Google Scholar 

  11. Klein, H. P. & Jahnke, L. Effects of aeration on formation and localization of the acetyl coenzyme A synthetases of Saccharomyces cerevisiae. J. Bacteriol. 137, 179–184 (1979).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Falcon, A. A., Chen, S., Wood, M. S. & Aris, J. P. Acetyl-coenzyme A synthetase 2 is a nuclear protein required for replicative longevity in Saccharomyces cerevisiae. Mol. Cell. Biochem. 333, 99–108 (2010).

    Article  CAS  PubMed  Google Scholar 

  13. Li, S. et al. Serine and SAM responsive complex SESAME regulates histone modification cross-talk by sensing cellular metabolism. Mol. Cell 60, 408–421 (2015).

    Article  CAS  PubMed  Google Scholar 

  14. Jaworski, D. M., Namboodiri, A. M. & Moffett, J. R. Acetate as a metabolic and epigenetic modifier of cancer therapy. J. Cell. Biochem. 117, 574–588 (2016).

    Article  CAS  PubMed  Google Scholar 

  15. Folmes, C. D. et al. Somatic oxidative bioenergetics transitions into pluripotency dependent glycolysis to facilitate nuclear reprogramming. Cell Metab. 14, 264–271 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Moussaieff, A. et al. Glycolysis-mediated changes in acetyl-CoA and histone acetylation control the early differentiation of embryonic stem cells. Cell Metab. 21, 392–402 (2015).

    Article  CAS  PubMed  Google Scholar 

  17. Murakami, C. J., Wall, V., Basisty, N. & Kaeberlein, M. Composition and acidification of the culture medium influences chronological aging similarly in vineyard and laboratory yeast. PLoS ONE 6, e24530 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Burtner, C. R., Murakami, C. J., Kennedy, B. K. & Kaeberlein, M. A molecular mechanism of chronological aging in yeast. Cell Cycle 8, 1256–1270 (2009).

    Article  CAS  PubMed  Google Scholar 

  19. Fabrizio, P. et al. Superoxide is a mediator of an altruistic aging program in Saccharomyces cerevisiae. J. Cell Biol. 166, 1055–1067 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Longo, V. D., Shadel, G. S., Kaeberlein, M. & Kennedy, B. Replicative and chronological aging in Saccharomyces cerevisiae. Cell Metab. 16, 18–31 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Dang, W. et al. Histone H4 lysine 16 acetylation regulates cellular lifespan. Nature 459, 802–807 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Millar, C. B., Kurdistani, S. K. & Grunstein, M. Acetylation of yeast histone H4 lysine 16: a switch for protein interactions in heterochromatin and euchromatin. Cold Spring Harb. Symp. Quant. Biol. 69, 193–200 (2004).

    Article  CAS  PubMed  Google Scholar 

  23. Kimura, A., Umehara, T. & Horikoshi, M. Chromosomal gradient of histone acetylation established by Sas2p and Sir2p functions as a shield against gene silencing. Nat. Genet. 32, 370–377 (2002).

    Article  PubMed  Google Scholar 

  24. Gruning, N. M. et al. Pyruvate kinase triggers a metabolic feedback loop that controls redox metabolism in respiring cells. Cell Metab. 14, 415–427 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Fleck, C. B. & Brock, M. Re-characterisation of Saccharomyces cerevisiae Ach1p: fungal CoA-transferases are involved in acetic acid detoxification. Fungal Genet. Biol. 46, 473–485 (2009).

    Article  CAS  PubMed  Google Scholar 

  26. van den Berg, M. A. et al. The two acetyl-coenzyme A synthetases of Saccharomyces cerevisiae differ with respect to kinetic properties and transcriptional regulation. J. Biol. Chem. 271, 28953–28959 (1996).

    Article  PubMed  Google Scholar 

  27. Shia, W. J., Li, B. & Workman, J. L. SAS-mediated acetylation of histone H4 Lys 16 is required for H2A.Z incorporation at subtelomeric regions in Saccharomyces cerevisiae. Genes Dev. 20, 2507–2512 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Vassylyeva, M. N. et al. Efficient, ultra-high-affinity chromatography in a one-step purification of complex proteins. Proc. Natl Acad. Sci. USA 114, E5138–E5147 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Sutton, A. et al. Sas4 and Sas5 are required for the histone acetyltransferase activity of Sas2 in the SAS complex. J. Biol. Chem. 278, 16887–16892 (2003).

    Article  CAS  PubMed  Google Scholar 

  30. McBrian, M. A. et al. Histone acetylation regulates intracellular pH. Mol. Cell 49, 310–321 (2013).

    Article  CAS  PubMed  Google Scholar 

  31. Gao, X. et al. Acetate functions as an epigenetic metabolite to promote lipid synthesis under hypoxia. Nat. Commun. 7, 11960 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Orlandi, I., Coppola, D. P. & Vai, M. Rewiring yeast acetate metabolism through MPC1 loss of function leads to mitochondrial damage and decreases chronological lifespan. Microb. Cell 1, 393–405 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Maestre, O., Garcia-Martinez, T., Peinado, R. A. & Mauricio, J. C. Effects of ADH2 overexpression in Saccharomyces bayanus during alcoholic fermentation. Appl. Environ. Microbiol. 74, 702–707 (2008).

    Article  CAS  PubMed  Google Scholar 

  34. Simpson-Lavy, K. & Kupiec, M. Carbon catabolite repression: not only for glucose. Curr. Genet. 65, 1321–1323 (2019).

  35. Saint-Prix, F., Bonquist, L. & Dequin, S. Functional analysis of the ALD gene family of Saccharomyces cerevisiae during anaerobic growth on glucose: the NADP+-dependent Ald6p and Ald5p isoforms play a major role in acetate formation. Microbiology 150, 2209–2220 (2004).

    Article  CAS  PubMed  Google Scholar 

  36. Ladurner, A. G., Inouye, C., Jain, R. & Tjian, R. Bromodomains mediate an acetyl-histone encoded antisilencing function at heterochromatin boundaries. Mol. Cell 11, 365–376 (2003).

    Article  CAS  PubMed  Google Scholar 

  37. Matangkasombut, O. & Buratowski, S. Different sensitivities of bromodomain factors 1 and 2 to histone H4 acetylation. Mol. Cell 11, 353–363 (2003).

    Article  CAS  PubMed  Google Scholar 

  38. Casatta, N., Porro, A., Orlandi, I., Brambilla, L. & Vai, M. Lack of Sir2 increases acetate consumption and decreases extracellular pro-aging factors. Biochim. Biophys. Acta 1833, 593–601 (2013).

    Article  CAS  PubMed  Google Scholar 

  39. Eisenberg, T. et al. Nucleocytosolic depletion of the energy metabolite acetyl-coenzyme a stimulates autophagy and prolongs lifespan. Cell Metab. 19, 431–444 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Cummings, J. H., Pomare, E. W., Branch, W. J., Naylor, C. P. & Macfarlane, G. T. Short-chain fatty acids in human large intestine, portal, hepatic and venous blood. Gut 28, 1221–1227 (1987).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Zhao, M., Chen, L. & Qu, H. CSGene: a literature-based database for cell senescence genes and its application to identify critical cell aging pathways and associated diseases. Cell Death Dis. 7, e2053 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Robin, J. D. et al. Telomere position effect: regulation of gene expression with progressive telomere shortening over long distances. Genes Dev. 28, 2464–2476 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  43. Wang, J. & Chen, J. SIRT1 regulates autoacetylation and histone acetyltransferase activity of TIP60. J. Biol. Chem. 285, 11458–11464 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Liao, K. et al. A feedback circuitry between polycomb signaling and fructose-1, 6-bisphosphatase enables hepatic and renal tumorigenesis. Cancer Res. 80, 675–688 (2020).

    Article  CAS  PubMed  Google Scholar 

  45. Jung, P. P. et al. Natural variation of chronological aging in the Saccharomyces cerevisiae species reveals diet-dependent mechanisms of lifespan control. NPJ Aging Mech. Dis. 4, 3 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  46. Fabrizio, P. et al. Sir2 blocks extreme lifespan extension. Cell 123, 655–667 (2005).

    Article  CAS  PubMed  Google Scholar 

  47. Vall-Llaura, N., Mir, N., Garrido, L., Vived, C. & Cabiscol, E. Redox control of yeast Sir2 activity is involved in acetic acid resistance and longevity. Redox Biol. 24, 101229 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Reverter-Branchat, G. et al. Chronological and replicative life-span extension in Saccharomyces cerevisiae by increased dosage of alcohol dehydrogenase 1. Microbiology 153, 3667–3676 (2007).

    Article  CAS  PubMed  Google Scholar 

  49. Longtine, M. S. et al. Additional modules for versatile and economical PCR-based gene deletion and modification in Saccharomyces cerevisiae. Yeast 14, 953–961 (1998).

    Article  CAS  PubMed  Google Scholar 

  50. Mei, Q. et al. Set1-catalyzed H3K4 trimethylation antagonizes the HIR/Asf1/Rtt106 repressor complex to promote histone gene expression and chronological lifespan. Nucleic Acids Res. 47, 3434–3449 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Vasilishina, A., Kropotov, A., Spivak, I. & Bernadotte, A. Relative human telomere length quantification by real-time PCR. Methods Mol. Biol. 1896, 39–44 (2019).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We sincerely thank J. D. Boeke and J. Thorner for yeast strains and antibodies. We thank Igenebook Bioinformatics Institute (Wuhan, China) for technical support. This work was supported by funding from the National Natural Science Foundation of China (31970578 to S.L. and 31872812 to X.Y.), the Natural Science Foundation of Hubei Province (2019CFA077 to X.Y. and 2017CFA066 to S.L.), National Institutes of Health (1R35GM118068 to J.L.W.) and the Stowers Institute.

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization: S.L. and X.Y.; experiments with western blots, RNA-seq, ChIP–qPCR, ChIP–seq and Co-IP were performed by W.C., X.Y., J.T. and X.L.; experiments with cell culture were performed by Y.W., X.L. and Z.Z.; animal experiments were performed by Y.W. and B.H.; CL7 affinity purification was done by L.M.; ChIP–seq and RNA-seq data were analysed by Q.Y. and W.C.; statistical analysis was performed by X.Y., W.C. and J.C.; writing, review and editing: X.Y., S.L. and J. L.W.

Corresponding author

Correspondence to Shanshan Li.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Metabolism thanks Brian Kennedy and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. 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 The SESAME complex is required to maintain the normal levels of H4K16ac.

a Analysis of relative intracellular acetyl-CoA levels in WT, sam1∆ and ser33∆ mutants. The acetyl-CoA levels were normalized to intracellular protein concentrations. b and c Quantification of western blots data in Fig. 1d. d Western blot analysis of H4K16ac and Acs2 in WT, TEFpr-ACS2 and CYCpr-ACS2 mutants. e Western blot analysis of H4K16ac in WT, acs1Δ and ach1Δ mutants. f ChIP-qPCR analysis of H4K16ac in TEFpr-ACS2 and CYCpr-ACS2 at regions with different distance to TEL VI-R. g ChIP-qPCR analysis of H4K16ac in WT and acs2-ts mutant at subtelomere regions of chromosome V (TEL V XC and TEL V XR), and chromosome VII (TEL VII XC and TEL VII XR). h ChIP-qPCR analysis of Acs2 occupancy at subtelomere regions. KRE1 and YJR011C were used as negative control regions. The untagged BY4741 and IgG were used as negative controls. i ChIP analysis of the occupancy of Sam1 (Sam1-FLAG) at regions with different distance to telomere VI-R. The untagged BY4741 was used as a negative control. The IP signals were normalized to input signals. j Venn diagram showing the overlap of genes occupied by Acs2, Sam1 and Pyk1 as determined by ChIP-seq. Acs2-bound genes were co-occupied by Pyk1 and Sam1 with P = 1.26 × 10−372 and P = 6.15 × 10−356, respectively using hypergeometric test. k Heatmap shown the binding of Acs2 at regions located within 30 kb of all 32 telomeres as determined by ChIP-seq. For Extended Data Fig. 1a–i, data represent means ± SE; n = 3 biological independent experiments. Two-sided t-tests were used for statistical analysis.

Source data

Extended Data Fig. 2 The SESAME complex interacts with the SAS complex to promote H4K16ac.

a Analysis of the genetic interaction between ACS2 and SAS2, ACS2 and SAS3, ACS2 and SAS4, ACS2 and SAS5. WT, acs2-ts, sas2Δ, acs2-ts sas2Δ, sas3Δ, acs2-ts sas3Δ, sas4Δ, acs2-ts sas4Δ, sas5Δ, and acs2-ts sas5Δ mutants were grown on YPD plates at 26 °C and 37 °C for 2–4 days. b The SESAME complex interacts with Sas2. The endogenous Acs2-FLAG was immunoprecipitated from cell lysates of Sas2-13Myc and Sas2-13Myc/Acs2-FLAG strains with anti-FLAG antibody. The Sas2-13Myc strain was used as a negative control. The co-IPed proteins were detected with anti-Myc (Sas2-13Myc), anti-Sam1, anti-Pyk1 and anti-Shm2 antibodies. c Effect of Acs2 on Sas2 expression levels as determined by western blot analysis. d Acs2 is required to maintain the integrity of SAS complex as determined by in vivo Co-IP assay. The endogenously expressed Sas5-FLAG was immunoprecipitated with anti-FLAG antibody from Sas5-FLAG and Sas5-FLAG acs2-ts cells when grown at 37 °C for 2 hr. The co-IPed Sas2 were detected by anti-Sas2 antibody. e Inactivation of Acs2 has no effect on acetylation of SAS complex. Sas5 was immunoprecipitated with anti-Myc antibody. The acetylation of SAS complex was detected with anti-acetyl lysine antibody. Ada3-FLAG was immunoprecipitated with anti-FLAG antibody and detected with anti-acetyl lysine antibody as a positive control (lane 3). For Extended Data Fig. 2a,b,d,e, shown is the typical example of 3 biological independent replicates. For Fig. 2c, data represent mean ± SE; n = 3 biological independent experiments. Two-sided t-tests were used for statistical analysis.

Source data

Extended Data Fig. 3 The SESAME complex and SAS complex co-regulate gene expression and telomere silencing.

a KEGG analysis of genes co-occupied by Acs2, Sam1 and H4K16ac. See also Fig. 3a. b Boxplots showing the overall transcription changes in WT, acs2-ts, sam1Δ, sas2Δ, and H4K16R mutants. Centre lines denote medians; box limits denote 25th and 75th percentiles; whiskers denote maxima and minima. Two-sided t-tests were used for statistical analysis. c Left panel: Venn diagrams showing the overlap of genes co-regulated by Acs2 and Sam1. Genes with fold changes in acs2-ts (≥3-fold; ≤0.5-fold), sam1Δ (≥1.5-fold; ≤0.75-fold) and P < 0.05 were considered as differentially expressed. The significance for overlapped genes between acs2-ts and sam1Δ RNA-seq datasets was indicated by the P-value, which was calculated by hypergeometric test. Right panel: KEGG analysis of genes co-regulated by Acs2 and Sam1. d-g Histogram showing the fractions of genes down-regulated in sas2Δ (d), H4K16R (e), sam1Δ (f) and acs2-ts (g) when plotted as a function of their distance to the nearest telomeres. Genes were categorized at 10-kb intervals for up to 60 kb from telomeres. One-sided χ2 tests were used. *, χ2 > 3.841, P < 0.05; ***, χ2 > 10.828, P < 0.001. h RT–qPCR analysis of the transcription of telomere-proximal genes in TEFpr-ACS2 and CYCpr-ACS2. Data represent means ± SE; n = 3 biological independent experiments. Two-sided t-tests were used for statistical analysis.

Source data

Extended Data Fig. 4 Acetate induces the global levels of H4K16ac as well as H4K16ac at subtelomere regions.

a and b Western blot analysis of the effect of acetate on histone modifications in yeast cells. WT (BY4741) cells were treated with 0–50 mM KAc for 4 hr. c Effect of KCl on H4K16ac in WT (BY4741) cells as determined by western blots. KAc treatment was used as a positive control. d Effect of KAc and KCl treatment on the medium pH after yeast cells were grown for 24 hr. e-g Western blot analysis of the effect of NaCl and NaAc on histone modifications in WT (BY4741) cells. The asterisk (*) indicates non-specific bands. h and i ChIP-seq tracks of H4K16ac at indicated genes when cells were treated with KCl and KAc. j ChIP-qPCR analysis of the effect of acetate on H4K16ac at subtelomere regions (TEL VIR, TEL V XC, TEL V XR, TEL VII XC and TEL VII XR) and telomere-proximal genes (COS8, IRC7, YCR106W, PHO11, SOR1). k Effect of glucose on H4K16ac as determined by western blots. Log phased WT (BY4741) cells were treated with YP + 0.05% glucose, YP + 1% glucose and YP + 4% glucose for 2 hr. For Extended Data Fig. 4a–g,j,k, data represent means ± SE; n = 3 biological independent experiments. Two-sided t-tests were used for statistical analysis.

Source data

Extended Data Fig. 5 Acs2 and Sas2 are required for acetate to induce H4K16ac.

a Acetate has no significant effect on Acs2 protein levels as determined by western blots. WT (BY4741) cells were treated with 0–50 mM KAc for 4 hr. KCl was added to keep the concentration of K+ at 50 mM. b Effect of acetate on H4K16ac in WT TetO7 and TetO7-ACS2 mutants as determined by Western blots. WT TetO7 and TetO7-ACS2 mutant were treated with KAc and doxycycline (dox) for 0–5 hr. c Effect of acetate on H4K16ac in WT and sam1Δ mutant. d and e Effect of acetate on H4K16ac in acs1Δ (d) and ach1Δ (e) mutants. f Acetate has no significant effect on Sas2 occupancy at subtelomere regions as determined by ChIP-qPCR. g Acetate treatment had no effect on the interaction between Sas2 and Sas5 as determined by Co-IP assay. The Sas5-13Myc/Sas2-FLAG and Sas5-13Myc/Sas2-FLAG/acs2-ts cells were grown at 37 °C and treated with or without KAc for 4 hr. Sas5 (Sas5-13Myc) was immunoprecipitated with anti-Myc agarose from cell lysates. The co-IPed Sas2 was detected by anti-FLAG (Sas2-FLAG). For Extended Data Fig. 5a–f, data represent means ± SE; n = 3 biological independent experiments. Two-sided t-tests were used for statistical analysis. For Extended Data Fig. 5g, shown is the typical example of 2 biological independent replicates.

Source data

Extended Data Fig. 6 Acetate regulates telomere silencing in budding yeast.

a Histogram showing the fraction of genes down-regulated in bdf1Δ (≤0.75-fold, P < 0.05) when plotted as a function of their distance to the nearest telomere. Genes were categorized at 10-kb intervals for up to 60 kb from telomeres. One-sided χ2 tests were used. *, χ2 > 3.841, P < 0.05; ***, χ2 > 10.828, P < 0.001. b Effect of Acs2 on Bdf1 expression as determined by western blots. c Effect of Bdf1 on the global levels of H4K16ac as determined by western blots. d ChIP analysis of H4K16ac at TEL V and TEL VII regions in WT and bdf1Δ mutant. e Effect of acetate on Bdf1 occupancy at subtelomere regions (TEL V XC, TEL V XR, TEL VII XC and TEL VII XR) as determined by ChIP-qPCR analysis. f ChIP-qPCR analysis of Sir2 occupancy at regions with different distance to TEL VI-R in TEFpr-ACS2 and CYCpr-ACS2. g Effect of Acs2 on Sir2 protein levels as determined by western blots. WT and acs2-ts mutant were grown at 37 °C for 2 hr. h ChIP-qPCR analysis of the effect of acetate on Sir2 binding at telomere-proximal genes in acs2-ts mutant. KCl was added as a control. i and j RT–qPCR analysis of the effect of KAc and sodium acetate (NaAc) on the transcription of telomere-proximal genes. KCl and NaCl were used as controls. k and l qRT-PCR analysis of the effect of acetate on the transcription of telomere-proximal genes in sam1Δ (k) and acs2-ts (l) mutants. m qRT-PCR analysis of the transcription of telomere-proximal genes in WT cells treated with YP + 2% glucose, 0.5% glucose, 0.05% glucose, 2% glycerol, 2% ethanol. For Extended Data Fig. 6b–m, data represent means ± SE; n = 3 biological independent experiments, two-sided t-tests were used for statistical analysis.

Source data

Extended Data Fig. 7 Acetate accelerates chronological aging in budding yeast.

a Boxplots showing the overall transcription changes by KAc treatment. Centre lines denote medians; box limits denote 25th and 75th percentiles; whiskers denote maxima and minima. Two-sided t-tests were used for statistical analysis. b Histogram showing the number of genes up-regulated by KAc treatment but down-regulated in sas2Δ (KAc up/sas2Δ down), H4K16R (KAc up/H4K16R down), bdf1Δ (KAc up/bdf1Δ down) when plotted as a function of their distance to the nearest telomere. Genes were categorized at 10-kb intervals for up to 60 kb from telomeres. One-sided χ2 tests were used. ***, χ2 > 10.828, P < 0.001. c-d Quantification of western blots data in Fig. 7e,f. e Analysis of the chronological life span of TEFpr-ACS2, CYCpr-ACS2, TEFpr-ACS2 sir2Δ, and CYCpr-ACS2 sir2Δ. f Analysis of the effect of acetate on the chronological life span of Sir2 overexpression cells (Sir2 OE, BY4741 genetic background). Cells (BY4741) transformed with the empty vector (EV) were used as a control. The survival percentage of cells on day 11 and day 25 was presented on the left panel. g Effect of acetate on autophagy in cells expressing the endogenous ATG8 promoter-driven GFP-ATG8. The autophagy activity was determined by a GFP (green fluorescent protein) liberation assay, which detects free GFP that is cleaved upon the delivery of endogenous promoter-driven Atg8 with an N-terminal GFP tag (GFP-Atg8) to the vacuole and subsequent proteolysis of Atg8. For Extended Data Fig. 7c–g, data represent means ± SE; n = 3 biological independent experiments, two-sided t-tests were used for statistical analysis.

Source data

Extended Data Fig. 8 Acetate accelerates cell senescence in HUVEC.

a Effect of acetate on HUVEC cell growth. b Effect of acetate on HUVEC cell cycle profiling. c-e Quantification of western blots data in Fig. 8a,b,e. f Effect of glucose (Gluc) starvation on H4K16ac in control (siControl) and ACSS2 knockdown (siACSS2) HUVEC cells. Cells were depleted of glucose for 6 hr. g Effect of glucose (Gluc) starvation on senescence of control (siControl) and ACSS2 knockdown (siACSS2) HUVEC cells as determined by SA-β-gal staining. Scale bar, 25 μm. Right panel: Quantification of the percentage of SA-β-gal positive cells. h Effect of glucose starvation on transcription of IL1A, IL1B, p16 and p21 in control (siControl) and ACSS2 knockdown (siACSS2) HUVEC cells. For Extended Data Fig. 8a–h, the quantitative data represent the mean ± SE; n = 3 biological independent experiments; two-sided t-tests were used for statistical analysis.

Source data

Extended Data Fig. 9 Acetate accelerates cell senescence dependent on ACSS2 and hMOF.

a Effect of acetate on H4K16ac in control (siControl) and MATIIα knockdown (siMATIIα) HUVEC cells. b and c Effect of acetate on senescence of control (siControl) and MATIIα knockdown (siMATIIα) cells as determined by SA-β-gal staining (b) and SAHF detection (DAPI staining) (c). d Quantification of western blots data in Fig. 8g. e Effect of acetate on H4K16ac in control and SIRT1 overexpression (SIRT1 OE) HUVEC cells. f Effect of acetate on senescence of control and SIRT1 overexpression (SIRT1 OE) HUVEC cells as determined by SA-β-gal staining. g KEGG analysis of 2533 genes differentially regulated by KAc. h Top panel: Venn diagrams showing the overlap between genes down-regulated by siACSS2 but up-regulated by KAc treatment. Bottom panel: Venn diagrams showing the overlap between genes up-regulated by siACSS2 but down-regulated by KAc treatment. i Heatmap showing genes differentially regulated by ACSS2 and acetate. j Effect of acetate and ACSS2 on telomere length in young (P10) and senescent (P22) HUVEC cells. For Extended Data Fig. 9a–f,j, the quantitative data represent the mean ± SE; n = 3 biological independent experiments; two-sided t-tests were used for statistical analysis.

Source data

Extended Data Fig. 10 A working model for how acetate regulates telomere silencing and aging in budding yeast.

a Under normal conditions, Acs2 within the SESAME complex synthesizes acetyl-CoA from glucose-derived acetate. SESAME specifically maintains normal levels of H4K16ac at subtelomere regions via its interaction with SAS complex. By promoting the binding of Bdf1, H4K16ac functions as the telomeric heterochromatin-euchromatin boundary to prevent the spreading of SIR complex. b When cells were treated with acetate, the interaction between the SESAME complex and the SAS complex is enhanced, which then increases Sas2-catalyzed H4K16ac and Bdf1 binding across the subtelomere regions. As a consequence, Sir2 is dissociated from the telomeres, leading to compromised telomere silencing and accelerated aging in yeast.

Supplementary information

Supplementary Information

Supplementary Figs. 1 and 2, unprocessed western blots and/or gels of Supplementary Figs. 1 and 2 and statistical source data of Supplementary Fig. 1

Reporting Summary

Supplementary Tables 1–14

Source data

Source Data Fig. 1

Unprocessed western blots and/or gels.

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Unprocessed western blots and/or gels.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Unprocessed western blots and/or gels.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Unprocessed western blots and/or gels.

Source Data Fig. 6

Statistical source data.

Source Data Fig. 7

Unprocessed western blots and/or gels.

Source Data Fig. 7

Statistical source data of Fig. 7.

Source Data Fig. 8

Unprocessed western blots and/or gels.

Source Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 1

Unprocessed western blots and/or gels.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Unprocessed western blots and/or gels.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Unprocessed western blots and/or gels.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Unprocessed western blots and/or gels.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Unprocessed western blots and/or gels.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Unprocessed western blots and/or gels.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 8

Unprocessed western blots and/or gels.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 9

Unprocessed western blots and/or gels.

Source Data Extended Data Fig. 9

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chen, W., Yu, X., Wu, Y. et al. The SESAME complex regulates cell senescence through the generation of acetyl-CoA. Nat Metab 3, 983–1000 (2021). https://doi.org/10.1038/s42255-021-00412-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-021-00412-9

This article is cited by

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