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:

Disease-associated astrocyte epigenetic memory promotes CNS pathology

Abstract

Disease-associated astrocyte subsets contribute to the pathology of neurologic diseases, including multiple sclerosis and experimental autoimmune encephalomyelitis1,2,3,4,5,6,7,8 (EAE), an experimental model for multiple sclerosis. However, little is known about the stability of these astrocyte subsets and their ability to integrate past stimulation events. Here we report the identification of an epigenetically controlled memory astrocyte subset that exhibits exacerbated pro-inflammatory responses upon rechallenge. Specifically, using a combination of single-cell RNA sequencing, assay for transposase-accessible chromatin with sequencing, chromatin immunoprecipitation with sequencing, focused interrogation of cells by nucleic acid detection and sequencing, and cell-specific in vivo CRISPR–Cas9-based genetic perturbation studies we established that astrocyte memory is controlled by the metabolic enzyme ATP-citrate lyase (ACLY), which produces acetyl coenzyme A (acetyl-CoA) that is used by histone acetyltransferase p300 to control chromatin accessibility. The number of ACLY+p300+ memory astrocytes is increased in acute and chronic EAE models, and their genetic inactivation ameliorated EAE. We also detected the pro-inflammatory memory phenotype in human astrocytes in vitro; single-cell RNA sequencing and immunohistochemistry studies detected increased numbers of ACLY+p300+ astrocytes in chronic multiple sclerosis lesions. In summary, these studies define an epigenetically controlled memory astrocyte subset that promotes CNS pathology in EAE and, potentially, multiple sclerosis. These findings may guide novel therapeutic approaches for multiple sclerosis and other neurologic diseases.

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: Pro-inflammatory stimuli induce astrocyte epigenetic memory.
Fig. 2: p300 promotes astrocyte epigenetic memory in EAE.
Fig. 3: ACLY controls p300-derived astrocyte epigenetic memory in EAE.
Fig. 4: Analysis of Acly+Ep300+ astrocytes with FIND-seq.
Fig. 5: Identification of astrocyte epigenetic memory in multiple sclerosis.

Similar content being viewed by others

Data availability

Sequencing data were deposited into GEO under the SuperSeries accession numbers GSE252551, GSE237558 and GSE252498. Source data are provided with this paper.

References

  1. Rothhammer, V. et al. Type I interferons and microbial metabolites of tryptophan modulate astrocyte activity and central nervous system inflammation via the aryl hydrocarbon receptor. Nat. Med. 22, 586–597 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Wheeler, M. A. et al. MAFG-driven astrocytes promote CNS inflammation. Nature 578, 593–599 (2020).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  3. Sanmarco, L. M. et al. Gut-licensed IFNγ+ NK cells drive LAMP1+TRAIL+ anti-inflammatory astrocytes. Nature 590, 473–479 (2021).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  4. Wheeler, M. A. et al. Environmental control of astrocyte pathogenic activities in CNS inflammation. Cell 176, 581–596.e518 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Wheeler, M. A. et al. Droplet-based forward genetic screening of astrocyte-microglia cross-talk. Science 379, 1023–1030 (2023).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  6. Clark, I. C. et al. Barcoded viral tracing of single-cell interactions in central nervous system inflammation. Science 372, eabf1230 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Rothhammer, V. et al. Microglial control of astrocytes in response to microbial metabolites. Nature 557, 724–728 (2018).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  8. Chao, C. C. et al. Metabolic control of astrocyte pathogenic activity via cPLA2-MAVS. Cell 179, 1483–1498.e1422 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Allen, N. J. & Lyons, D. A. Glia as architects of central nervous system formation and function. Science 362, 181–185 (2018).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  10. Lee, H. G., Wheeler, M. A. & Quintana, F. J. Function and therapeutic value of astrocytes in neurological diseases. Nat. Rev. Drug Discov. 21, 339–358 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Sofroniew, M. V. Astrocyte barriers to neurotoxic inflammation. Nat. Rev. Neurosci. 16, 249–263 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Yu, X. & Khakh, B. S. SnapShot: astrocyte interactions. Cell 185, 220–220.e221 (2022).

    Article  CAS  PubMed  Google Scholar 

  13. Lee, H. G., Lee, J. H., Flausino, L. E. & Quintana, F. J. Neuroinflammation: an astrocyte perspective. Sci. Transl. Med. 15, eadi7828 (2023).

    Article  CAS  PubMed  Google Scholar 

  14. Liddelow, S. A. et al. Neurotoxic reactive astrocytes are induced by activated microglia. Nature 541, 481–487 (2017).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  15. Endo, F. et al. Molecular basis of astrocyte diversity and morphology across the CNS in health and disease. Science 378, eadc9020 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Habib, N. et al. Disease-associated astrocytes in Alzheimer’s disease and aging. Nat. Nerosci. 23, 701–706 (2020).

    Article  CAS  Google Scholar 

  17. Zinkernagel, R. M. et al. On immunological memory. Annu. Rev. Immunol. 14, 333–367 (1996).

    Article  CAS  PubMed  Google Scholar 

  18. Wendeln, A. C. et al. Innate immune memory in the brain shapes neurological disease hallmarks. Nature 556, 332–338 (2018).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  19. Saeed, S. et al. Epigenetic programming of monocyte-to-macrophage differentiation and trained innate immunity. Science 345, 1251086 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Sun, J. C., Beilke, J. N. & Lanier, L. L. Adaptive immune features of natural killer cells. Nature 457, 557–561 (2009).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  21. Serafini, N. et al. Trained ILC3 responses promote intestinal defense. Science 375, 859–863 (2022).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  22. Netea, M. G. et al. Defining trained immunity and its role in health and disease. Nat. Rev. Immunol. 20, 375–388 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Perkins, N. D. et al. Regulation of NF-κB by cyclin-dependent kinases associated with the p300 coactivator. Science 275, 523–527 (1997).

    Article  CAS  PubMed  Google Scholar 

  24. Zhao, J., Li, X., Guo, M., Yu, J. & Yan, C. The common stress responsive transcription factor ATF3 binds genomic sites enriched with p300 and H3K27ac for transcriptional regulation. BMC Genomics 17, 335 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Magistretti, P. J. & Allaman, I. Lactate in the brain: from metabolic end-product to signalling molecule. Nat. Rev. Neurosci. 19, 235–249 (2018).

    Article  CAS  PubMed  Google Scholar 

  26. Pietrocola, F., Galluzzi, L., Bravo-San Pedro, J. M., Madeo, F. & Kroemer, G. Acetyl coenzyme A: a central metabolite and second messenger. Cell Metab. 21, 805–821 (2015).

    Article  CAS  PubMed  Google Scholar 

  27. Wellen, K. E. et al. ATP-citrate lyase links cellular metabolism to histone acetylation. Science 324, 1076–1080 (2009).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  28. Mayo, L. et al. Regulation of astrocyte activation by glycolipids drives chronic CNS inflammation. Nat. Med. 20, 1147–1156 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Clark, I. C. et al. Identification of astrocyte regulators by nucleic acid cytometry. Nature 614, 326–333 (2023).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  30. Absinta, M. et al. A lymphocyte-microglia-astrocyte axis in chronic active multiple sclerosis. Nature 597, 709–714 (2021).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  31. Schirmer, L. et al. Neuronal vulnerability and multilineage diversity in multiple sclerosis. Nature 573, 75–82 (2019).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  32. Cheong, J. G. et al. Epigenetic memory of coronavirus infection in innate immune cells and their progenitors. Cell 186, 3882–3902.e3824 (2023).

    Article  CAS  PubMed  Google Scholar 

  33. Yao, Y. et al. Induction of autonomous memory alveolar macrophages requires T cell help and is critical to trained immunity. Cell 175, 1634–1650.e1617 (2018).

    Article  CAS  PubMed  Google Scholar 

  34. McCarthy, G. F. & Leblond, C. P. Radioautographic evidence for slow astrocyte turnover and modest oligodendrocyte production in the corpus callosum of adult mice infused with 3H-thymidine. J. Comp. Neurol. 271, 589–603 (1988).

    Article  CAS  PubMed  Google Scholar 

  35. Chierzi, S. et al. Astrocytes transplanted during early postnatal development integrate, mature, and survive long term in mouse cortex. J. Neurosci. 43, 1509–1529 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Ogryzko, V. V., Schiltz, R. L., Russanova, V., Howard, B. H. & Nakatani, Y. The transcriptional coactivators p300 and CBP are histone acetyltransferases. Cell 87, 953–959 (1996).

    Article  CAS  PubMed  Google Scholar 

  37. Li, Q., Xiao, H. & Isobe, K. Histone acetyltransferase activities of cAMP-regulated enhancer-binding protein and p300 in tissues of fetal, young, and old mice. J. Gerontol. A 57, B93–B98 (2002).

    Article  Google Scholar 

  38. Farrelly, L. A. et al. Histone serotonylation is a permissive modification that enhances TFIID binding to H3K4me3. Nature 567, 535–539 (2019).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  39. Lepack, A. E. et al. Dopaminylation of histone H3 in ventral tegmental area regulates cocaine seeking. Science 368, 197–201 (2020).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  41. Sen, P. et al. Histone acetyltransferase p300 induces de novo super-enhancers to drive cellular senescence. Mol. Cell 73, 684–698.e688 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Furman, D. et al. Chronic inflammation in the etiology of disease across the life span. Nat. Med. 25, 1822–1832 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Li, T. Y. et al. The transcriptional coactivator CBP/p300 is an evolutionarily conserved node that promotes longevity in response to mitochondrial stress. Nat Aging 1, 165–178 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Burda, J. E. et al. Divergent transcriptional regulation of astrocyte reactivity across disorders. Nature 606, 557–564 (2022).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  45. Chen, L., Fischle, W., Verdin, E. & Greene, W. C. Duration of nuclear NF-κB action regulated by reversible acetylation. Science 293, 1653–1657 (2001).

    Article  ADS  CAS  Google Scholar 

  46. Long, L. et al. CRISPR screens unveil signal hubs for nutrient licensing of T cell immunity. Nature 600, 308–313 (2021).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  47. Beigneux, A. P. et al. ATP-citrate lyase deficiency in the mouse. J. Biol. Chem. 279, 9557–9564 (2004).

    Article  CAS  PubMed  Google Scholar 

  48. Hochrein, S. M. et al. The glucose transporter GLUT3 controls T helper 17 cell responses through glycolytic-epigenetic reprogramming. Cell Metab. 34, 516–532.e511 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Balmer, M. L. et al. Memory CD8+ T cells require increased concentrations of acetate induced by stress for optimal function. Immunity 44, 1312–1324 (2016).

    Article  CAS  PubMed  Google Scholar 

  50. Lauterbach, M. A. et al. Toll-like receptor signaling rewires macrophage metabolism and promotes histone acetylation via ATP-citrate lyase. Immunity 51, 997–1011.e1017 (2019).

    Article  CAS  PubMed  Google Scholar 

  51. Sardar, D. et al. Induction of astrocytic Slc22a3 regulates sensory processing through histone serotonylation. Science 380, eade0027 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Krausgruber, T. et al. Structural cells are key regulators of organ-specific immune responses. Nature 583, 296–302 (2020).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  53. Hatzivassiliou, G. et al. ATP citrate lyase inhibition can suppress tumor cell growth. Cancer Cell 8, 311–321 (2005).

    Article  CAS  PubMed  Google Scholar 

  54. Verschueren, K. H. G. et al. Structure of ATP citrate lyase and the origin of citrate synthase in the Krebs cycle. Nature 568, 571–575 (2019).

    Article  ADS  CAS  PubMed  Google Scholar 

  55. Wei, J. et al. An allosteric mechanism for potent inhibition of human ATP-citrate lyase. Nature 568, 566–570 (2019).

    Article  ADS  CAS  PubMed  Google Scholar 

  56. Garcia, A. D., Doan, N. B., Imura, T., Bush, T. G. & Sofroniew, M. V. GFAP-expressing progenitors are the principal source of constitutive neurogenesis in adult mouse forebrain. Nat. Neurosci. 7, 1233–1241 (2004).

    Article  CAS  PubMed  Google Scholar 

  57. Madisen, L. et al. A robust and high-throughput Cre reporting and characterization system for the whole mouse brain. Nat. Neurosci. 13, 133–140 (2010).

    Article  CAS  PubMed  Google Scholar 

  58. Everhart, M. B. et al. Duration and intensity of NF-κB activity determine the severity of endotoxin-induced acute lung injury. J. Immunol. 176, 4995–5005 (2006).

    Article  CAS  PubMed  Google Scholar 

  59. Gutierrez-Vazquez, C. & Quintana, F. J. Protocol for in vitro analysis of pro-inflammatory and metabolic functions of cultured primary murine astrocytes. STAR Protoc. 3, 101033 (2022).

    Article  CAS  PubMed  Google Scholar 

  60. Thompson, A. J. et al. Diagnosis of multiple sclerosis: 2017 revisions of the McDonald criteria. Lancet Neurol. 17, 162–173 (2018).

    Article  PubMed  Google Scholar 

  61. Dhaeze, T. et al. CD70 defines a subset of proinflammatory and CNS-pathogenic TH1/TH17 lymphocytes and is overexpressed in multiple sclerosis. Cell Mol. Immunol. 16, 652–665 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Broux, B. et al. Interleukin-26, preferentially produced by TH17 lymphocytes, regulates CNS barrier function. Neurol. Neuroimmunol. Neuroinflamm. 7, e870 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  63. Illouz, T., Madar, R., Hirsh, T., Biragyn, A. & Okun, E. Induction of an effective anti-amyloid-β humoral response in aged mice. Vaccine 39, 4817–4829 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Illouz, T. et al. Maternal antibodies facilitate amyloid-β clearance by activating Fc-receptor–Syk-mediated phagocytosis. Commun. Biol. 4, 329 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Motulsky, H. J. & Brown, R. E. Detecting outliers when fitting data with nonlinear regression—a new method based on robust nonlinear regression and the false discovery rate. BMC Bioinformatics 7, 123 (2006).

    Article  PubMed  PubMed Central  Google Scholar 

  66. Sanjana, N. E., Shalem, O. & Zhang, F. Improved vectors and genome-wide libraries for CRISPR screening. Nat. Methods 11, 783–784 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Chen, S. et al. Genome-wide CRISPR screen in a mouse model of tumor growth and metastasis. Cell 160, 1246–1260 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Lee, Y., Messing, A., Su, M. & Brenner, M. GFAP promoter elements required for region-specific and astrocyte-specific expression. Glia 56, 481–493 (2008).

    Article  PubMed  Google Scholar 

  69. Wang, T. et al. Gene essentiality profiling reveals gene networks and synthetic lethal interactions with oncogenic Ras. Cell 168, 890–903.e815 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Foo, L. C. Purification of rat and mouse astrocytes by immunopanning. Cold Spring Harb. Protoc. 2013, 421–432 (2013).

    Article  PubMed  Google Scholar 

  71. Lemaitre, F. et al. Capturing T lymphocytes’ dynamic interactions with human neural cells using time-lapse microscopy. Front. Immunol. 12, 668483 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Durafourt, B. A., Moore, C. S., Blain, M. & Antel, J. P. Isolating, culturing, and polarizing primary human adult and fetal microglia. Methods Mol. Biol. 1041, 199–211 (2013).

    Article  CAS  PubMed  Google Scholar 

  73. Kieran, N. W. et al. MicroRNA-210 regulates the metabolic and inflammatory status of primary human astrocytes. J. Neuroinflammation 19, 10 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. van Galen, P. et al. A multiplexed system for quantitative comparisons of chromatin landscapes. Mol. Cell 61, 170–180 (2016).

    Article  ADS  PubMed  Google Scholar 

  75. Untergasser, A. et al. Primer3—new capabilities and interfaces. Nucleic Acids Res. 40, e115 (2012).

  76. Trombetta, J. J. et al. Preparation of single-cell RNA-seq libraries for next generation sequencing. Curr. Protoc. Mol. Biol. 107, 4.22.1–4.22.17 (2014).

    Article  PubMed  Google Scholar 

  77. Langmead, B. & Salzberg, S. L. Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Tarasov, A., Vilella, A. J., Cuppen, E., Nijman, I. J. & Prins, P. Sambamba: fast processing of NGS alignment formats. Bioinformatics 31, 2032–2034 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Li, H. et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics 25, 2078–2079 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  80. Ramirez, F., Dundar, F., Diehl, S., Gruning, B. A. & Manke, T. deepTools: a flexible platform for exploring deep-sequencing data. Nucleic Acids Res. 42, W187–191 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Zhang, Y. et al. Model-based analysis of ChIP-seq (MACS). Genome Biol. 9, R137 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  82. Buenrostro, J. D., Giresi, P. G., Zaba, L. C., Chang, H. Y. & Greenleaf, W. J. Transposition of native chromatin for fast and sensitive epigenomic profiling of open chromatin, DNA-binding proteins and nucleosome position. Nat. Methods 10, 1213–1218 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Buenrostro, J. D., Wu, B., Chang, H. Y. & Greenleaf, W. J. ATAC-seq: a method for assaying chromatin accessibility genome-wide. Curr. Protoc. Mol. Biol. 109, 21.29.21–21.29.29 (2015).

    Article  Google Scholar 

  84. Clark, I. C., Thakur, R. & Abate, A. R. Concentric electrodes improve microfluidic droplet sorting. Lab Chip 18, 710–713 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA 102, 15545–15550 (2005).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  86. Mootha, V. K. et al. PGC-1α-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes. Nat. Genet. 34, 267–273 (2003).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported by grants NS102807, ES02530, ES029136, AI126880 from the NIH; RG4111A1 and JF2161-A-5 from the NMSS; RSG-14-198-01-LIB from the American Cancer Society; and PA-1604-08459 from the International Progressive MS Alliance. H.-G.L. was supported by a Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education (2021R1A6A3A14039088). J.-H.L. was supported by the Basic Science Research Program funded by the National Research Foundation of Korea (NRF)/Ministry of Education (2022R1A6A3A03071157) and a long-term postdoctoral fellowship funded by the Human Frontier Science Program (LT0015/2023-L). T.I. was supported by the EMBO postdoctoral fellowship (ALTF: 1009-2021). G.P. is a trainee in the Medical Scientist Training Program funded by NIH T32 GM007356. The content of this manuscript is solely the responsibility of the authors and does not necessarily represent the official views of the National Institute of General Medical Science or NIH. M.A.W. was supported by NINDS, NIMH and NCI (R01MH132632, R01MH130458, R00NS114111, T32CA207201). A.P. holds the T1 Canada Research Chair in MS and is funded by the Canada Institute of Health Research, the NMSS and the Canadian Foundation for Innovation. I.C.C. was supported by K22AI152644 and DP2AI154435 from the NIH. The authors thank L. Li for technical assistance; all members of the Quintana laboratory for helpful advice and discussions; R. Krishnan for technical assistance with flow cytometry; and BDRL members K. A. Aldinger, D. Doherty, I. G. Phelps, J. C. Dempsey, K. J. Lee and L. A. Cort.

Author information

Authors and Affiliations

Authors

Contributions

H.-G.L., V.R., M.A.W. and F.J.Q. designed the research. H-G.L., J.M.R., C.F.A., J.-H.L., L.E.F., F.P., C.-C.C., L.S., T.I., F.G., M.C., L.M.S., J.E.K., G.P., S.E.J.Z. and V.R. performed experiments. H.-G.L., Z.L., C.F.A., K.L.K. and F.J.Q. performed bioinformatic analyses. S.W.S. performed FIND-seq. J.P.A., A.P. and I.C.C. provided unique reagents and discussed and/or interpreted findings. H.-G.L. and F.J.Q. wrote the paper with input from coauthors. F.J.Q. directed and supervised the study.

Corresponding author

Correspondence to Francisco J. Quintana.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature thanks the anonymous reviewer(s) for their contribution to the peer review of this work.

Additional information

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

Extended data figures and tables

Extended Data Fig. 1 Analysis of astrocyte epigenetic memory in vivo and in vitro.

(a) Fluorescence-activated cell sorting (FACS) sorting schematic for astrocytes, microglia, and monocytes. (b) Volcano plot of differential gene expression increased (red) or decreased (blue) analyzed by RNA-seq of sorted astrocytes. (c) IPA predicted upstream regulators in isolated astrocytes. Up-regulated (red) or down-regulated (blue) in astrocytes stimulated twice (2X IL-1β + TNF) versus once (1X IL-1β + TNF) are shown. (d) GO pathway analysis of ATAC-seq accessible peaks of isolated astrocytes comparing 2X IL-1β + TNF versus 1X IL-1β + TNF. (e) Genome browser snapshots showing the ATAC-seq sequencing tracks at the Tlr2 locus. Only regions showing a significant increase (p-value < 0.05) in accessibility in astrocytes from mice stimulated twice (2X) versus once (1X) are highlighted by yellow boxes. (f) Homer DNA-motif enrichment analyses of differentially accessible peaks. (g) Experimental design for (h) to (j). Primary astrocytes received IL-1β/TNF stimulation once (1X) or twice (2X). (h) qPCR of astrocytes after 30 min activation with IL-1β/TNF on day 7 (n = 5 per group). Unpaired two-sided t-test. (i) Neuronal viability assay (n = 4 control; n = 4 1X; n = 3 2X). Unpaired two-sided t-test. (j) Effect of IL-1β/TNF stimulation on lactate release (n = 6 per group). Unpaired two-sided t-test. (k) qPCR analysis of astrocyte response after the first IL-1β/TNF stimulation (n = 5 per group). Unpaired two-sided t-test. (l) FACS analysis of EGFP expression in IL-1β/TNF stimulated primary astrocytes isolated from p65EGFP reporter mice (0 min; n = 5; Other time point; n = 6 per group). ***P < 0.0001. Unpaired two-sided t-test. (m) FACS sorting schematic of EGFP positive/negative astrocytes after PBS or IL-1β/TNF stimulation for 18-24 h. (n) Primary astrocytes isolated from p65EGFP reporter mice received IL-1β/TNF stimulation. After 18–24 h, EGFP positive astrocytes were sorted and analyzed by FACS of EGFP expression (n = 3-5 per group). Unpaired two-sided t-test. (o) Primary astrocytes isolated from p65EGFP reporter mice received PBS or IL-1β/TNF stimulation. After 18-24 h, EGFP positive/negative astrocytes were sorted and cultured for 6 days to perform qPCR (n = 4 per group). Unpaired two-sided t-test. (p) p65 activation in EGFP positive/negative astrocytes 1 h after IL-1β/TNF stimulation on day 7 (n = 5 per group). Unpaired two-sided t-test. Data shown as mean ± s.e.m.

Source Data

Extended Data Fig. 2 HAT enzyme expression and regulation of astrocyte epigenetic memory.

(a) EP300, Tip60 (KAT5), and PCAF (KAT2B) signaling network in isolated brain astrocytes that received ICV administration of IL-1β/TNF twice (2X) compared to once (1X). (b) qPCR analysis of Ep300, Kat5, and Kat2b expression in primary astrocytes stimulated after 30 min IL-1β/TNF stimulation on day 7 (n = 4 per group). Unpaired two-sided t-test. (c) Primary astrocytes were stimulated with IL-1β/TNF once (1X), twice (2X), or three times (3X). qPCR analysis of astrocytes after 30 min activation with IL-1β/TNF on day 14 (n = 4 per group). Unpaired two-sided t-test. (d) Experimental design for (e). (e) qPCR of primary astrocytes in the presence with/without C646 (p300/CBP inhibitor), MB-3 (Gcn5/PCAF inhibitor), and MG149 (Tip60 inhibitor) after 30 min stimulation with IL-1β/TNF on day 7 (n = 5 per group). Unpaired two-sided t-test. (f) Experimental design for Fig. 1j. (g) Gene expression 30 min after IL-1β/TNF stimulation on day 7 of EGFP positive/negative astrocytes treated with C646 or vehicle (n = 8 per group). Unpaired two-sided t-test. Data shown as mean ± s.e.m.

Source Data

Extended Data Fig. 3 Astrocyte epigenetic memory and Ep300 signaling in EAE.

(a) Generation of astrocyte epigenetic memory signature filtered based on adjusted p-value and fold change. Up-signature (red) or down-signature (blue) in astrocytes stimulated twice (2X) than once (1X) are shown. (b) Astrocyte epigenetic memory signature score applied to naive and EAE scRNA-seq astrocyte dataset (Priming, Peak, and Remission)10. (c) Experimental design for (d,e). (d) EAE score for (e) (n = 8 per group). Data shown as mean ± s.e.m. Naïve and EAE induced C57BL/6 mice received ICV administration of IL-1β/TNF (EAE peak, Day 22), and 18-24 h later sorted brain astrocytes were analyzed. (e) qPCR of IL-1β/TNF response of astrocytes (Naïve; n = 12; EAE; n = 8 per group). Representative data of two independent experiments. Unpaired two-sided t-test. (f) Immunostaining (left) and quantification (right) of H3K27ac+ and p300+ astrocytes from sgScrmbl- and sgEp300- transduced mice at 23 days after EAE induction (n = 6 spinal cord sections; n = 3 mice per group). Unpaired two-sided t-test. (g) FluoroMyelin dye staining and percentage of myelin loss in spinal cord from sgScrmbl- and sgEp300-treated mice (n = 9 spinal cord sections; n = 3 mice per group). Lesions indicated by arrowheads. Unpaired two-sided t-test. (h) NF-kB signaling network comparing sgEp300-transduced versus sgScrmbl-transduced astrocytes. (i) Quantification of CNS-resident cells from sgScrmbl- and sgEp300-transduced mice (n = 9 per group). Unpaired two-sided t-test. (j,k) Analysis of CNS T cells (up) and splenic T cells (bottom) from sgScrmbl- or sgEp300-transduced mice (n = 5 per group). Unpaired two-sided t-test. (l) Genome browser snapshots showing the Relb locus. Only regions showing a significant decrease (p-value < 0.05) in accessibility in sgScrmbl-transduced versus sgEp300-transduced astrocytes are highlighted by yellow boxes. (m) Chip-qPCR analysis of p300 recruitment to promoters in primary astrocytes 30 min after IL-1β/TNF stimulation on day 7 (n = 3 per group). Unpaired two-sided t-test. Data shown as mean ± s.e.m.

Source Data

Extended Data Fig. 4 ACLY and ACSS2 signaling in EAE astrocytes.

(a) qPCR analysis of Acly and Acss2 expression in primary astrocytes stimulated for 30 min with IL-1β/TNF on day 7 (n = 4 per group). Unpaired two-sided t-test. (b) Primary astrocytes received IL-1β/TNF stimulation once (1X), twice (2X), or three times (3X). qPCR analysis of astrocytes after 30 min activation with IL-1β/TNF on day 14 (n = 4 per group). Unpaired two-sided t-test. (c) Immunostaining (left) and quantification (right) of ACLY+ astrocytes in mice with/without EAE (n = 8 spinal cord sections (naïve); n = 9 spinal cord sections (EAE); n = 3 mice per group). Unpaired two-sided t-test. (d) Immunostaining (left) and quantification (right) of ACSS2+ astrocytes in mice with/without EAE (n = 6 spinal cord sections (naïve); n = 9 spinal cord sections (EAE); n = 3 mice per group). Unpaired two-sided t-test. Data shown as mean ± s.e.m.

Source Data

Extended Data Fig. 5 Astrocyte Acly signaling and ACLY + p300+ astrocytes in EAE.

(a) Immunostaining (left) and quantification (right) of H3K27ac+ and ACLY+ astrocytes from sgScrmbl- and sgAcly- transduced mice at 21days after EAE induction (n = 6 spinal cord sections; n = 3 mice per group). Unpaired two-sided t-test. (b) Staining with FluoroMyelin dye and percentage of myelin loss in spinal cord of sgScrmbl- and sgAcly-treated mice (n = 9 spinal cord sections; n = 3 mice per group). Lesions indicated by arrowheads. Unpaired two-sided t-test. (c) Quantification of CNS-resident cells from sgScrmbl- and sgAcly-transduced mice (n = 5 per group). Unpaired two-sided t-test. (d,e) Analysis of CNS T cells (up) and splenic T cells (bottom) from sgScrmbl- or sgAcly-transduced mice (n = 5 per group). Unpaired two-sided t-test. Data shown as mean ± s.e.m. (f) Immunostaining (left) and quantification (right) of ACLY+SOX9+, p300+SOX9+, and ACLY+p300+SOX9+ astrocytes in EAE and control mice (n = 6 spinal cord sections; n = 3 mice per group). Unpaired two-sided t-test. Data shown as mean ± s.e.m.

Source Data

Extended Data Fig. 6 Astrocyte epigenetic memory in NOD EAE.

(a) Experimental design for (c). (b) NOD EAE score for (c) (n = 3 per group). (c) Naïve and EAE induced NOD mice received ICV administration of IL-1β/TNF (EAE progressive, Day 124). After 18-24 h, sorted brain astrocytes were analyzed by qPCR (n = 3 per group). Unpaired two-sided t-test. (d) Immunostaining (left) and quantification (right) of H3K27ac+ and p300+ astrocytes in mice with/without NOD EAE (n = 9 spinal cord sections; n = 3 mice per group). Astrocyte H3K27ac levels were calculated as the mean signal intensity (arbitrary units) per GFAP+ cells using automated unbiased quantification. Unpaired two-sided t-test. (e) Immunostaining (left) and quantification (right) of ACLY+, p300+, and ACLY+p300+ astrocytes in mice with/without NOD EAE (n = 9 spinal cord sections; n = 3 mice per group). Unpaired two-sided t-test. (f) NOD EAE curves (sgScrmbl; n = 7; sgEp300; n = 8; sgAcly; n = 7). Lentivirus were injected at day 40. Representative data of two independent experiments. Regression slope two-sided t-test compared with sgScrmbl. (g) Volcano plot of differential gene expression determined by RNA-seq in astrocytes isolated from sgScrmbl-, sgEp300-, and sgAcly-transduced mice 64 days after NOD EAE induction (n = 3 sgScrmbl, n = 3 sgEp300, n = 2 sgAcly). (h) GSEA analysis comparing sgScrmbl-, sgEp300-, and sgAcly-transduced astrocytes. (i) Staining with FluoroMyelin dye and percentage of myelin loss from sgScrmbl-, sgEp300-, and sgAcly- transduced mice spinal cord (n = 6 spinal cord sections (sgEp300); n = 9 spinal cord sections (sgScrmbl, sgAcly); n = 3 mice per group. Lesions indicated by arrowheads. Unpaired two-sided t-test. Data shown as mean ± s.e.m.

Source Data

Extended Data Fig. 7 ACLY and p300 signaling in NOD EAE astrocytes.

(a) Immunostaining (left) and quantification (right) of H3K27ac+ and p300+ astrocytes from sgScrmbl- and sgEp300- transduced NOD mice 64 days after EAE induction (n = 6 spinal cord sections; n = 3 mice per group). Unpaired two-sided t-test. (b) Immunostaining (left) and quantification (right) of H3K27ac+ and ACLY+ astrocytes from sgScrmbl- and sgAcly- transduced NOD mice 64 days after EAE induction (n = 6 spinal cord sections; n = 3 mice per group). Unpaired two-sided t-test. (c) Quantification of CNS-resident cells from sgScrmbl-, sgEp300, and sgAcly-transduced mice (n = 7 per group). Unpaired two-sided t-test. (d,e) Analysis of CNS T cells (up) and splenic T cells (bottom) from sgScrmbl-, sgEp300, and sgAcly-transduced mice (n = 4 per group). Unpaired two-sided t-test. Data shown as mean ± s.e.m.

Source Data

Extended Data Fig. 8 Analysis of Acly+Ep300+ astrocytes by FIND-seq.

(a) FACS sorting schematic for tdTomatoGfap astrocytes. (b-d) Schematic illustration of microfluidic devices utilized in FIND-seq. (b) The bubble-triggered device has with five inlets: i) cell inlet, ii) Oligo dT primer-conjugated agarose inlet, iii) lysis buffer inlet, iv) oil inlet, and v) pressurized air inlet. (c) The re-injector device has three inlets: i) agarose bead inlet, ii) TaqMan PCR master mix inlet, and iii) oil inlet. (d) The droplet sorter device has seven inlets: i) emulsion inlet, ii) spacing oil inlet, iii) additional spacing oil inlet, iv) bias oil inlet, v) saltwater inlet (for the electrode), vi) saltwater inlet (for the moat), and vii) pressurized air inlet. (e) Astrocytes are encapsulated in an agarose bead along with the lysis buffer. The genome entrapped in the agarose bead is stained with SYBR Green I and visualized using a fluorescence microscope. (f) cDNA, produced on the agarose bead, is amplified via WTA and validated using a Bioanalyzer. (g) Principal component analysis (PCA) plot of AclyEp300, Acly+Ep300, AclyEp300+, Acly+Ep300+ EAE astrocytes. (h) Violin plot depicting Acly+Ep300+ EAE astrocyte signature expression in EAE astrocytes. (i) IPA pathway analysis up-regulated (red) in cluster 1 astrocytes. (j) EP300-NF-κB signaling network of cluster 1 astrocytes. (k) IPA pathway analysis up-regulated (red) in cluster 10 astrocytes. (l) EP300-NF-κB signaling network of cluster 10 astrocytes.

Source Data

Extended Data Fig. 9 Analysis of human astrocyte epigenetic memory and MS astrocyte scRNA-seq.

(a) Primary human fetal astrocytes received IL-1β/TNF stimulation once (1X) or twice (2X). qPCR of astrocytes in the presence with/without C646 (p300/CBP inhibitor) after 2 h stimulation with IL-1β/TNF on day 7 (n = 4 per group). Unpaired two-sided t-test. (b) Gene scatterplots of astrocyte markers. (c) Unsupervised clustering UMAP plot of astrocytes from patients with MS and control individuals from Schirmer et al.31 and Absinta et al.30. (n = 16,276 cells). WM, white matter; CI, chronic inactive; CA, chronic active. (d) Significantly enriched genes by astrocyte cell type cluster. (e) Cluster distribution of CNS cells. (f) Violin plot depicting Acly+Ep300+ EAE astrocyte signature expression in MS astrocytes. (g) Astrocyte epigenetic memory signature score in astrocyte clusters of control and MS patients.

Source Data

Extended Data Fig. 10 ACLY + p300+ astrocytes in MS patients.

(a) Immunostaining and quantification of ACLY+SOX9+, p300+SOX9+, ACLY+p300+SOX9+ astrocytes in CNS samples from MS patients with MS (n = 9 sections (Lesion); n = 6 sections (NAWM); n = 3 per patient) and controls (n = 3 sections; n = 3 per patient). WM, white matter; NAWM, normally appearing white matter. Unpaired two-sided t-test. Data shown as mean ± s.e.m.

Source Data

Supplementary information

Reporting Summary

Supplementary Table 1

Differential gene expression of 2× IL-1β+TNF versus 1× IL-1β+TNF astrocytes.

Supplementary Table 2

Differentially accessible gene of 2× IL-1β+TNF versus 1× IL-1β+TNF astrocytes.

Supplementary Table 3

Transcriptional signature of astrocyte epigenetic memory.

Supplementary Table 4

Differential gene expression of sgScrmbl- versus sgEp300-targeted astrocytes during EAE.

Supplementary Table 5

Peaks of H3K27ac ChIP–seq data of sgScrmbl- versus sgEp300-targeted astrocytes during EAE.

Supplementary Table 6

Differential gene expression of sgScrmbl- versus sgAcly-targeted astrocytes during EAE.

Supplementary Table 7

EP300–NF-κB signalling network of sgScrmbl- versus sgAcly-targeted astrocytes during EAE.

Supplementary Table 8

Differential gene expression of sgScrmbl- versus sgEp300- or sgAcly-targeted astrocytes during NOD EAE.

Supplementary Table 9

Transcriptional signature of Acly+Ep300+ EAE astrocytes.

Supplementary Table 10

Differential gene expression by cluster of astrocytes during EAE in TdTomatoGfap mice by scRNA-seq from Wheeler et al.

Supplementary Table 11

ACLY–EP300–NF-κB network of cluster 3 EAE astrocytes.

Supplementary Table 12

Differential gene expression by cluster of astrocytes in multiple sclerosis versus control samples from Schirmer et al. and Absinta et al.

Supplementary Table 13

Clinical information of samples analysed.

Supplementary Table 14

ACLY–EP300–NF-κB network of cluster 2 multiple sclerosis astrocytes.

Source data

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

Lee, HG., Rone, J.M., Li, Z. et al. Disease-associated astrocyte epigenetic memory promotes CNS pathology. Nature 627, 865–872 (2024). https://doi.org/10.1038/s41586-024-07187-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-024-07187-5

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