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:

Destabilizing heterochromatin by APOE mediates senescence

Abstract

Apolipoprotein E (APOE) is a component of lipoprotein particles that function in the homeostasis of cholesterol and other lipids. Although APOE is genetically associated with human longevity and Alzheimer’s disease, its mechanistic role in aging is largely unknown. Here, we used human genetic, stress-induced and physiological cellular aging models to explore APOE-driven processes in stem cell homeostasis and aging. We report that in aged human mesenchymal progenitor cells (MPCs), APOE accumulation is a driver for cellular senescence. By contrast, CRISPR–Cas9-mediated deletion of APOE endows human MPCs with resistance to cellular senescence. Mechanistically, we discovered that APOE functions as a destabilizer for heterochromatin. Specifically, increased APOE leads to the degradation of nuclear lamina proteins and a heterochromatin-associated protein KRAB-associated protein 1 via the autophagy–lysosomal pathway, thereby disrupting heterochromatin and causing senescence. Altogether, our findings uncover a role of APOE as an epigenetic mediator of senescence and provide potential targets to ameliorate aging-related 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: APOE expression increases in senescent human MPCs.
Fig. 2: Depletion of APOE attenuates cellular senescence in human MPCs.
Fig. 3: APOE promotes the degradation of nuclear envelope and heterochromatin-associated proteins.
Fig. 4: APOE interrupts nuclear envelope homeostasis and heterochromatin organization.
Fig. 5: Depletion of APOE stabilizes heterochromatin.
Fig. 6: Knockdown of APOE exhibits a geroprotective effect in senescent human MPCs.

Similar content being viewed by others

Data availability

The high-throughput sequencing data including RNA-seq, DamID-seq, ATAC-seq and whole-genome sequencing generated in this study have been deposited with the Genome Sequence Archive in the National Genomics Data Center, Beijing Institute of Genomics (China National Center for Bioinformation) of the CAS under accession no. HRA000668 and are publicly accessible at https://ngdc.cncb.ac.cn/gsa-human/browse/HRA000668. MS data have been deposited with the ProteomeXchange Consortium via the PRoteomics IDEntifications partner repository with the dataset identifier PXD024414.

Code availability

All custom analysis code and scripts for reproducing the key conclusions in this study can be found at https://github.com/QianzhaoJ/APOE_2021.

References

  1. Hauser, P. S., Narayanaswami, V. & Ryan, R. O. Apolipoprotein E: from lipid transport to neurobiology. Prog. Lipid Res. 50, 62–74 (2011).

    Article  CAS  PubMed  Google Scholar 

  2. Tao, Q.-Q. et al. Associations between apolipoprotein E genotypes and serum levels of glucose, cholesterol, and triglycerides in a cognitively normal aging Han Chinese population. Clin. Interv. Aging 9, 1063–1067 (2014).

    PubMed  PubMed Central  Google Scholar 

  3. Wagner, T., Bartelt, A., Schlein, C. & Heeren, J. Genetic dissection of tissue-specific apolipoprotein E function for hypercholesterolemia and diet-induced obesity. PLoS ONE 10, e0145102 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Huang, R. et al. Lowering circulating apolipoprotein E levels improves aged bone fracture healing. JCI Insight 4, e129144 (2019).

    Article  PubMed Central  Google Scholar 

  5. Schilling, A. F. et al. Increased bone formation in mice lacking apolipoprotein E. J. Bone Miner. Res. 20, 274–282 (2005).

    Article  CAS  PubMed  Google Scholar 

  6. Bartelt, A. et al. Apolipoprotein E-dependent inverse regulation of vertebral bone and adipose tissue mass in C57Bl/6 mice: modulation by diet-induced obesity. Bone 47, 736–745 (2010).

    Article  CAS  PubMed  Google Scholar 

  7. Milad, N. et al. Increased plasma lipid levels exacerbate muscle pathology in the mdx mouse model of Duchenne muscular dystrophy. Skelet. Muscle 7, 19 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Yin, Y. & Wang, Z. ApoE and neurodegenerative diseases in aging. Adv. Exp. Med. Biol. 1086, 77–92 (2018).

    Article  CAS  PubMed  Google Scholar 

  9. Schupf, N. et al. Apolipoprotein E and familial longevity. Neurobiol. Aging 34, 1287–1291 (2013).

    Article  CAS  PubMed  Google Scholar 

  10. Ryu, S., Atzmon, G., Barzilai, N., Raghavachari, N. & Suh, Y. Genetic landscape of APOE in human longevity revealed by high-throughput sequencing. Mech. Ageing Dev. 155, 7–9 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Terao, A. et al. Immune response gene expression increases in the aging murine hippocampus. J. Neuroimmunol. 132, 99–112 (2002).

    Article  CAS  PubMed  Google Scholar 

  12. Jung, J.-S. et al. Adipose-derived stem/stromal cells recapitulate aging biomarkers and show reduced stem cell plasticity affecting their adipogenic differentiation capacity. Cell. Reprogram. 21, 187–199 (2019).

    Article  CAS  PubMed  Google Scholar 

  13. Di Narzo, A. F. et al. High-throughput characterization of blood serum proteomics of IBD patients with respect to aging and genetic factors. PLoS Genet. 13, e1006565 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Finch, C. E. & Shams, S. Apolipoprotein E and sex bias in cerebrovascular aging of men and mice. Trends Neurosci. 39, 625–637 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Huang, L. et al. Cellular senescence: a pathogenic mechanism of pelvic organ prolapse (review). Mol. Med. Rep. 22, 2155–2162 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Carreno, G., Guiho, R. & Martinez-Barbera, J. P. Cell senescence in neuropathology: a focus on neurodegeneration and tumours. Neuropathol. Appl. Neurobiol. 47, 359–378 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  17. McHugh, D. & Gil, J. Senescence and aging: causes, consequences, and therapeutic avenues. J. Cell Biol. 217, 65–77 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Liu, H., Xia, X. & Li, B. Mesenchymal stem cell aging: mechanisms and influences on skeletal and non-skeletal tissues. Exp. Biol. Med. (Maywood) 240, 1099–1106 (2015).

    Article  CAS  Google Scholar 

  19. Schurgers, L. J. et al. Initiation and propagation of vascular calcification is regulated by a concert of platelet- and smooth muscle cell-derived extracellular vesicles. Front. Cardiovasc. Med. 5, 36 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Yan, P. et al. FOXO3-engineered human ESC-derived vascular cells promote vascular protection and regeneration. Cell Stem Cell 24, 447–461.e8 (2019).

    Article  CAS  PubMed  Google Scholar 

  21. López-Otín, C., Blasco, M. A., Partridge, L., Serrano, M. & Kroemer, G. The hallmarks of aging. Cell 153, 1194–1217 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Dodig, S., Čepelak, I. & Pavić, I. Hallmarks of senescence and aging. Biochem. Med. (Zagreb) 29, 030501 (2019).

    Article  Google Scholar 

  23. Zhang, W. et al. Aging stem cells. A Werner syndrome stem cell model unveils heterochromatin alterations as a driver of human aging. Science 348, 1160–1163 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Zhang, W., Qu, J., Liu, G.-H. & Belmonte, J. C. I. The ageing epigenome and its rejuvenation. Nat. Rev. Mol. Cell Biol. 21, 137–150 (2020).

    Article  CAS  PubMed  Google Scholar 

  25. Zhang, W., Song, M., Qu, J. & Liu, G.-H. Epigenetic modifications in cardiovascular aging and diseases. Circ. Res. 123, 773–786 (2018).

    Article  CAS  PubMed  Google Scholar 

  26. Lenain, C. et al. Massive reshaping of genome–nuclear lamina interactions during oncogene-induced senescence. Genome Res. 27, 1634–1644 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. De Cecco, M. et al. Transposable elements become active and mobile in the genomes of aging mammalian somatic tissues. Aging (Albany NY) 5, 867–883 (2013).

    Article  Google Scholar 

  28. Ucar, D. et al. The chromatin accessibility signature of human immune aging stems from CD8+ T cells. J. Exp. Med. 214, 3123–3144 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Li, X., Xiong, X. & Yi, C. Epitranscriptome sequencing technologies: decoding RNA modifications. Nat. Methods 14, 23–31 (2016).

    Article  CAS  PubMed  Google Scholar 

  30. Deng, L. et al. Stabilizing heterochromatin by DGCR8 alleviates senescence and osteoarthritis. Nat. Commun. 10, 3329 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Ren, R., Ocampo, A., Liu, G.-H. & Izpisua Belmonte, J. C. Regulation of stem cell aging by metabolism and epigenetics. Cell Metab. 26, 460–474 (2017).

    Article  CAS  PubMed  Google Scholar 

  32. Kim, W. S. et al. Analysis of apolipoprotein E nuclear localization using green fluorescent protein and biotinylation approaches. Biochem. J. 409, 701–709 (2008).

    Article  CAS  PubMed  Google Scholar 

  33. Rohn, T. T. & Moore, Z. D. Nuclear localization of apolipoprotein E4: a new trick for an old protein. Int. J. Neurol. Neurother. 4, 067 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Theendakara, V., Bredesen, D. E. & Rao, R. V. Downregulation of protein phosphatase 2A by apolipoprotein E: implications for Alzheimer’s disease. Mol. Cell. Neurosci. 83, 83–91 (2017).

    Article  CAS  PubMed  Google Scholar 

  35. Quinn, C. M. et al. Induction of fibroblast apolipoprotein E expression during apoptosis, starvation-induced growth arrest and mitosis. Biochem. J. 378, 753–761 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Fu, L. et al. Up-regulation of FOXD1 by YAP alleviates senescence and osteoarthritis. PLoS Biol. 17, e3000201 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Wang, S. et al. ATF6 safeguards organelle homeostasis and cellular aging in human mesenchymal stem cells. Cell Discov. 4, 2 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Liang, C. et al. Stabilization of heterochromatin by CLOCK promotes stem cell rejuvenation and cartilage regeneration. Cell Res. 31, 187–205 (2021).

    Article  CAS  PubMed  Google Scholar 

  39. Watanabe, S., Kawamoto, S., Ohtani, N. & Hara, E. Impact of senescence-associated secretory phenotype and its potential as a therapeutic target for senescence-associated diseases. Cancer Sci. 108, 563–569 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Johnson, L. A. et al. Apolipoprotein E-low density lipoprotein receptor interaction affects spatial memory retention and brain ApoE levels in an isoform-dependent manner. Neurobiol. Dis. 64, 150–162 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Dou, Z. et al. Autophagy mediates degradation of nuclear lamina. Nature 527, 105–109 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Park, Y.-E. et al. Autophagic degradation of nuclear components in mammalian cells. Autophagy 5, 795–804 (2009).

    Article  CAS  PubMed  Google Scholar 

  43. Lenain, C., Gusyatiner, O., Douma, S., van den Broek, B. & Peeper, D. S. Autophagy-mediated degradation of nuclear envelope proteins during oncogene-induced senescence. Carcinogenesis 36, 1263–1274 (2015).

    Article  CAS  PubMed  Google Scholar 

  44. Xie, W. et al. HIV-1 exposure triggers autophagic degradation of stathmin and hyperstabilization of microtubules to disrupt epithelial cell junctions. Signal Transduct. Target. Ther. 5, 79 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Gorbunova, V., Boeke, J. D., Helfand, S. L. & Sedivy, J. M. Human genomics. Sleeping dogs of the genome. Science 346, 1187–1188 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Simon, M. et al. LINE1 derepression in aged wild-type and SIRT6-deficient mice drives inflammation. Cell Metab. 29, 871–885.e5 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. De Cecco, M. et al. L1 drives IFN in senescent cells and promotes age-associated inflammation. Nature 566, 73–78 (2019).

    Article  CAS  PubMed  Google Scholar 

  48. Van Meter, M. et al. SIRT6 represses LINE1 retrotransposons by ribosylating KAP1 but this repression fails with stress and age. Nat. Commun. 5, 5011 (2014).

    Article  CAS  PubMed  Google Scholar 

  49. Bi, S. et al. SIRT7 antagonizes human stem cell aging as a heterochromatin stabilizer. Protein Cell 11, 483–504 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Hu, H. et al. ZKSCAN3 counteracts cellular senescence by stabilizing heterochromatin. Nucleic Acids Res. 48, 6001–6018 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Martins, F., Sousa, J., Pereira, C. D., da Cruz E Silva, O. A. B. & Rebelo, S. Nuclear envelope dysfunction and its contribution to the aging process. Aging Cell 19, e13143 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Briand, N. & Collas, P. Lamina-associated domains: peripheral matters and internal affairs. Genome Biol. 21, 85 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  53. Canat, A., Veillet, A., Bonnet, A. & Therizols, P. Genome anchoring to nuclear landmarks drives functional compartmentalization of the nuclear space. Brief. Funct. Genomics 19, 101–110 (2020).

    Article  CAS  PubMed  Google Scholar 

  54. Joung, J. et al. Genome-scale CRISPR–Cas9 knockout and transcriptional activation screening. Nat. Protoc. 12, 828–863 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Solovei, I. et al. LBR and lamin A/C sequentially tether peripheral heterochromatin and inversely regulate differentiation. Cell 152, 584–598 (2013).

    Article  CAS  PubMed  Google Scholar 

  56. Shan, H. et al. Large-scale chemical screen identifies Gallic acid as a geroprotector for human stem cells. Protein Cell https://doi.org/10.1007/s13238-021-00872-5 (2021).

  57. Liu, C.-C., Liu, C.-C., Kanekiyo, T., Xu, H. & Bu, G. Apolipoprotein E and Alzheimer disease: risk, mechanisms and therapy. Nat. Rev. Neurol. 9, 106–118 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Belloy, M. E., Napolioni, V. & Greicius, M. D. A quarter century of APOE and Alzheimer’s disease: progress to date and the path forward. Neuron 101, 820–838 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Zhang, W., Qu, J., Suzuki, K., Liu, G. H., & Izpisua Belmonte, J. C. Concealing cellular defects in pluripotent stem cells. Trends Cell Biol. 23, 587–592 (2013).

    Article  CAS  PubMed  Google Scholar 

  60. Bachmeier, C. et al. A multifaceted role for apoE in the clearance of Beta-amyloid across the blood–brain barrier. Neurodegener. Dis. 11, 13–21 (2013).

    Article  CAS  PubMed  Google Scholar 

  61. Fernandez, C. G., Hamby, M. E., McReynolds, M. L. & Ray, W. J. The role of APOE4 in disrupting the homeostatic functions of astrocytes and microglia in aging and Alzheimer’s disease. Front. Aging Neurosci. 11, 14 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Huang, Y. & Mahley, R. W. Apolipoprotein E: structure and function in lipid metabolism, neurobiology, and Alzheimer’s diseases. Neurobiol. Dis. 72, 3–12 (2014).

    Article  CAS  PubMed  Google Scholar 

  63. Mahley, R. W. Central nervous system lipoproteins: ApoE and regulation of cholesterol metabolism. Arterioscler. Thromb. Vasc. Biol. 36, 1305–1315 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Marais, A. D. Apolipoprotein E in lipoprotein metabolism, health and cardiovascular disease. Pathology 51, 165–176 (2019).

    Article  CAS  PubMed  Google Scholar 

  65. Theendakara, V. et al. Direct transcriptional effects of apolipoprotein E. J. Neurosci. 36, 685–700 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Love, J. E. et al. Nuclear uptake of an amino-terminal fragment of apolipoprotein E4 promotes cell death and localizes within microglia of the Alzheimer’s disease brain. Int. J. Physiol. Pathophysiol. Pharmacol. 9, 40–57 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Levros, L.-C. Jr., Labrie, M., Charfi, C. & Rassart, E. Binding and repressive activities of apolipoprotein E3 and E4 isoforms on the human ApoD promoter. Mol. Neurobiol. 48, 669–680 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Madireddy, A. & Gerhardt, J. Replication through repetitive DNA elements and their role in human diseases. Adv. Exp. Med. Biol. 1042, 549–581 (2017).

    Article  CAS  PubMed  Google Scholar 

  69. Guo, C. et al. Tau activates transposable elements in Alzheimer’s disease. Cell Rep. 23, 2874–2880 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Ho, Y. Y., Deckelbaum, R. J., Chen, Y., Vogel, T. & Talmage, D. A. Apolipoprotein E inhibits serum-stimulated cell proliferation and enhances serum-independent cell proliferation. J. Biol. Chem. 276, 43455–43462 (2001).

    Article  CAS  PubMed  Google Scholar 

  71. Vogel, T. et al. Apolipoprotein E: a potent inhibitor of endothelial and tumor cell proliferation. J. Cell. Biochem. 54, 299–308 (1994).

    Article  CAS  PubMed  Google Scholar 

  72. Ishigami, M., Swertfeger, D. K., Granholm, N. A. & Hui, D. Y. Apolipoprotein E inhibits platelet-derived growth factor-induced vascular smooth muscle cell migration and proliferation by suppressing signal transduction and preventing cell entry to G1 phase. J. Biol. Chem. 273, 20156–20161 (1998).

    Article  CAS  PubMed  Google Scholar 

  73. Roos, A. et al. Myopathy in Marinesco–Sjögren syndrome links endoplasmic reticulum chaperone dysfunction to nuclear envelope pathology. Acta Neuropathol. 127, 761–777 (2014).

    Article  CAS  PubMed  Google Scholar 

  74. Nava, M. M. et al. Heterochromatin-driven nuclear softening protects the genome against mechanical stress-induced damage. Cell 181, 800–817.e22 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Ren, X. et al. Maintenance of nucleolar homeostasis by CBX4 alleviates senescence and osteoarthritis. Cell Rep. 26, 3643–3656.e7 (2019).

    Article  CAS  PubMed  Google Scholar 

  76. Zhang, X. et al. Telomere-dependent and telomere-independent roles of RAP1 in regulating human stem cell homeostasis. Protein Cell 10, 649–667 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Guo, Y. et al. Visualizing intracellular organelle and cytoskeletal interactions at nanoscale resolution on millisecond timescales. Cell 175, 1430–1442.e17 (2018).

    Article  CAS  PubMed  Google Scholar 

  78. Jeong, W.-J. et al. WDR76 is a RAS binding protein that functions as a tumor suppressor via RAS degradation. Nat. Commun. 10, 295 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Konermann, S. et al. Genome-scale transcriptional activation by an engineered CRISPR–Cas9 complex. Nature 517, 583–588 (2015).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank L. Bai, R. Bai, S. Ma, J. Lu, J.Chen and Y. Yang for their administrative assistance, X. Zhang, W. Li, J. Jia and J. Jiao for their help with the animal experiments, L. Wang, J. Yang, L. Fu, W. Wang, C. Liang, Z. Liu for their technical support, J. Wang for his help with the LC–MS/MS assay, J. Jia and S. Sun for their help with the FACS experiment and C. Peng for her help with the TEM sample preparation. The processing of high-throughput sequencing data was conducted on the ‘Era’ Petascale supercomputer of the Computer Network Information Center of the CAS. This work was supported by the National Key Research and Development Program of China (no. 2018YFC2000100), the Strategic Priority Research Program of CAS (no. XDA16010000), Beijing Natural Science Foundation (nos. Z190019, JQ20031), the National Key Research and Development Program of China (nos. 2021YFF1201000, 2020YFA0112200, 2020YFA0113400, 2020YFA0804000, 2020YFA0803401 2019YFA0802202, 2019YFA0110100, 2018YFA0107203, 2018YFC2000400 2017YFA0103304 and 2017YFA0102802), the National Natural Science Foundation of China (nos. 81901433, 81921006, 81625009, 91749202, 81861168034, 91949209, 82125011, 92049304, 92049116, 81822018, 81870228, 81922027, 82071588, 82122024, 32100937, 32121001, 31970597 and 81801399), the Program of the Beijing Municipal Science and Technology Commission (no. Z191100001519005), the Key Research Program of the Chinese Academy of Sciences (no. KFZD-SW-221), the K. C. Wong Education Foundation (nos. GJTD-2019-06 and GJTD-2019-08), the 14th Five-year Network Security and Informatization Plan of CAS (no. WX145XQ07-18), the Science and Technology Service Network Initiative of CAS (no. KFJ-STS-QYZD-2021-08-001), CAS Special Research Assistant Program, Youth Innovation Promotion Association of CAS (nos. E1CAZW0401 and 2020085), CAS Project for Young Scientists in Basic Research (YSBR-012), the Non-profit Central Research Institute Fund of the Chinese Academy of Medical Sciences (no. 2020-JKCS-011), the State Key Laboratory of Stem Cell and Reproductive Biology, the State Key Laboratory of Membrane Biology and the Milky Way Research Foundation.

Author information

Authors and Affiliations

Authors

Contributions

G.H.L., J.Q., W.Z. and M.S. conceived the work and supervised the experiments. H.Z., Z. Wu and K.Y. performed the phenotypic and mechanistic analyses. Q.J. and S.W. performed bioinformatics analyses. Z. Wu, Z. Wang and J.H. performed the RNA-seq, DamID-seq and ATAC-seq library construction. Q.C. performed RNA and DNA analyses. H.H. and Y.C. performed immunostaining analysis and animal experiments. Q.W. performed the plasmid construction. D.H. and Z.J. performed the lentivirus packaging and protein stability analysis. G.H.L., J.Q., W.Z., M.S., H.Z., Q.J., Z. Wu and S.W. performed the data analysis. G.H.L., J.Q., W.Z., M.S., J.C.I.B., H.Z., Q.J., Z. Wu, S.W., J.R. and J.L. wrote the manuscript.

Corresponding authors

Correspondence to Moshi Song, Weiqi Zhang, Jing Qu or Guang-Hui Liu.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Aging thanks Karl Riabowol, Zhixun Dou and the other, 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

Extended Data Fig. 1 Generation and characterization of APOE+/+ and APOE-/- hESCs.

a, RT-qPCR showing the mRNA levels of APOE and CDKN1A (P21) in RS-hMPCs (P3), HGPS-hMPCs (P3), and WS-hMPCs (P3). GAPDH was used as the internal control. b, Western blot analysis of APOE in hMPCs (P4) transduced with lentiviruses expressing luciferase (Luc) or APOE. c, Schematic diagram of CRISPR-dCas9-mediated transcriptional activation of endogenous APOE. d, Western blot analysis of APOE in hMPCs transfected with non-targeting (negative control, sg-NC) or APOE-targeting (sg-APOE) sgRNAs using the CRISPR-dCas9 transcriptional activation system. e, Schematic diagram of APOE knockout strategy through CRISPR-Cas9-mediated non-homologous end joining (NHEJ). Sequencing results showed a 1-bp (T/A) insertion introduced by genome editing. f, Western blot analysis of the APOE protein level in APOE+/+ hESCs and APOE-/- hESCs. g, Immunofluorescence analysis of pluripotency markers OCT4, NANOG, and SOX2 in APOE+/+ hESC and APOE-/- hESC. Scale bars, 25 μm. Phase-contrast images were shown on the right. Scale bars, 250 μm. h, Immunofluorescence analysis of TuJ1, SMA, and FOXA2 to evaluate the differentiation potential of APOE+/+ hESC and APOE-/- hESC toward lineages of all three germ layers, that is, ectoderm, mesoderm, and endoderm, respectively. Scale bars, 25 μm. i, Cell cycle analysis of APOE+/+ hESCs and APOE-/- hESCs. j, Immunofluorescence analysis of Ki67 in APOE+/+ hESCs and APOE-/- hESCs. Scale bars, 25 μm. k, Detection of the DNA methylation status of the OCT4 promoter in APOE+/+ hESCs and APOE-/- hESCs. l, Karyotype analysis of APOE+/+ hESCs and APOE-/- hESCs. m, The copy number variation (CNV) analysis by whole genome sequencing of APOE+/+ hESCs and APOE-/- hESCs. a,ij, A Two-tailed Student’s t-test was used for statistical analysis. Data are presented as the means ± s.e.m. a, n = 4 biological repeats. b,d,f,g, n = 3 biological repeats. b,d,f, For western blot analysis, β-Tubulin was used as the loading control.

Source data

Extended Data Fig. 2 Characterization of APOE+/+ and APOE-/- hMPCs.

a, Absence of markers (CD34, CD43, and CD45) irrelevant to hMPCs by flow cytometry analysis in APOE+/+ (P3) hMPCs and APOE-/- (P3) hMPCs. b, Characterization of the osteogenesis capacity of APOE+/+ (P3) hMPCs and APOE-/- (P3) hMPCs by Von Kossa and Osteocalcin staining. c, Characterization of the adipogenesis capacity of APOE+/+ (P3) hMPCs and APOE-/- (P3) hMPCs by Oil Red O and FABP4 staining. d, Characterization of the chondrogenesis capacity of APOE+/+ (P3) hMPCs and APOE-/- (P3) hMPCs by Toluidine blue staining. e, CNV analysis of APOE+/+ (P3) hMPCs and APOE-/- (P3) hMPCs. f, Heatmap showing the Euclidean distance of RNA-seq replicates in APOE+/+ (P3) hMPCs and APOE-/- (P3) hMPCs. The color key from dark to light represents relatively low to high Euclidean distance, respectively. g, Gene set enrichment analysis showing that SASP-associated genes were downregulated in APOE-/- (P3) hMPCs compared to APOE+/+ (P3) hMPCs. h, Immunofluorescence analysis of γH2AX foci in APOE+/+ (P9) hMPCs and APOE-/- (P9) hMPCs. i, Western blot analysis of APOE in APOE-/- (P9) hMPCs transduced with lentiviruses expressing luciferase (Luc) or APOE. β-Tubulin was used as the loading control. bd,h, A Two-tailed Student’s t-test was used for statistical analysis. bd,hi, Data are presented as the means ± s.e.m. n = 3 biological repeats. b-d, Scale bars, 50 μm. h, Scale bars, 25 μm.

Source data

Extended Data Fig. 3 APOE interacts with nuclear envelope proteins and heterochromatin-associated proteins.

a, Networks showing the potential APOE-interacting proteins associated with ‘Chromosomal region’ and ‘Nuclear envelope’ Gene Ontology (GO) annotations. The color key represents relatively low to high coverages of APOE-interacting proteins, respectively. b, Co-IP analysis in APOE+/+ (P4) hMPCs and APOE-/- (P4) hMPCs indicated no interaction between APOE and Nucleolin or β-Tubulin, referred to Fig. 3c as negative controls. c, Immunofluorescence analysis of APOE-FLAG and Emerin in APOE-/- (P9) hMPCs transduced with lentiviruses expressing FLAG-tagged APOE (left). Fluorescence signal density plots show the distributions of Emerin (red) and APOE (green) signals in APOE-/- hMPCs, as measured between peripheries. Individual cell measurements are shown as thin lines and average curves are shown as thick lines (right). d, Western blot analysis of APOE in APOE+/+ hMPCs and APOE-/- hMPCs transduced with lentiviruses expressing FLAG-tagged APOE. e, Protein stability analysis of KAP1, LBR and Emerin in APOE+/+ (P4) hMPCs transduced with lentiviruses expressing Luc-FLAG or APOE-FLAG. Protein levels of KAP1, LBR and Emerin at indicated time points in the presence of a protein synthesis inhibitor cycloheximide (CHX) treatment were determined by western blotting. f, Immunofluorescence analysis of H3K9me3 in APOE-/- (P4) hMPCs transduced with lentiviruses expressing FLAG-tagged APOE, 3×NLS-tagged APOE and 3×NES-tagged APOE. g, Immunofluorescence analysis of Ki67 in APOE-/- (P4) hMPCs transduced with lentiviruses expressing FLAG-tagged APOE, 3×NLS-tagged APOE and 3×NES-tagged APOE. h, Co-IP verification for the interaction between LC3 and APOE-FLAG in HEK293T cells. i, Immunofluorescence analysis of Emerin and LAMP2 in APOE+/+ hMPCs transduced with lentiviruses expressing FLAG-tagged APOE and treated with 50 nM bafilomycin A1 (BFA1) for 12 h. j, Western blot analysis of heterochromatin proteins in APOE+/+ hMPCs and APOE-/- hMPCs after treatment with si-NC or si-ATG5. d,e, β-Tubulin was used as the loading control. j, GAPDH was used as the loading control. eg,j, A Two-tailed Student’s t-test was used for statistical analysis. b,d,hi, n = 3 biological repeats. c, n=13. e,g,j, n = 3 independent experiments. f, n=150 cells per group. c,i, Scale bars, 10 μm. f,g, Scale bars, 25 μm.

Source data

Extended Data Fig. 4 DamID-seq and ATAC-seq analyses of APOE+/+ and APOE-/- hMPCs.

a, Heatmap showing the Euclidean distance of DamID-seq replicates in APOE+/+ (P3) hMPCs and APOE-/- (P3) hMPCs. The color key from dark to light represents relatively low to high Euclidean distance, respectively. b, Heatmaps showing increased DamID signals at LINE1, Satellite, and Alu regions in APOE-/- (P3) hMPCs compared to APOE+/+ (P3) hMPCs. The color key from blue to red represents relatively low to high DamID signals, respectively. c, Heatmap showing the Euclidean distance of ATAC-seq replicates in APOE+/+ (P3) hMPCs and APOE-/- (P3) hMPCs. The color key from dark to light represents relatively low to high Euclidean distance, respectively. d, Left, scatter plot showing the differential ATAC-seq peaks identified by Diffbind in APOE-/- (P3) hMPCs compared to APOE+/+ (P3) hMPCs. Red and blue dots represent the opened and closed ATAC-seq peaks in APOE-/- hMPCs, respectively. Right, heatmaps showing the ATAC signals within opened and closed ATAC-seq peaks in both APOE+/+ (P3) hMPCs and APOE-/- (P3) hMPCs. e, Genomic element distribution analysis of opened and closed ATAC peaks in APOE-/- (P3) hMPCs compared to APOE+/+ (P3) hMPCs. f, Heatmap showing decreased ATAC signals ranging from 5-kb upstream to 5-kb downstream of ATAC-seq peaks at LAD-localized LINE1, Satellite, and Alu regions in APOE-/- (P3) hMPCs compared to APOE+/+ (P3) hMPCs. The color key from light to dark represents relatively low to high ATAC signals, respectively.

Extended Data Fig. 5 Knockdown of KAP1 or LBR accelerates cell senescence in APOE-/- hMPCs.

a, Western blot analysis of KAP1 in APOE-/- (P3) hMPCs transduced with lentiviral sh-GL2 or sh-KAP1. b, Clonal expansion assay in APOE-/- (P4) hMPCs transduced with lentiviral sh-GL2 or sh-KAP1. c, Immunofluorescence analysis of Ki67 in APOE-/- (P4) hMPCs transduced with lentiviral sh-GL2 or sh-KAP1. The white arrows indicate Ki67-positive cells. d, SA-β-gal staining of APOE-/- (P4) hMPCs transduced with lentiviral sh-GL2 or sh-KAP1. e, Western blot analysis of LBR in APOE-/- (P4) hMPCs transduced with lentiviral sh-GL2 or sh-LBR. f, Clonal expansion assay in APOE-/- (P4) hMPCs transduced with lentiviral sh-GL2 or sh-LBR. g, Immunofluorescence analysis of Ki67 in APOE-/- (P4) hMPCs transduced with lentiviral sh-GL2 or sh-LBR. The white arrows indicate Ki67-positive cells. h, SA-β-gal staining of APOE-/- (P4) hMPCs transduced with lentiviral sh-GL2 or sh-LBR. bd,fh, A Two-tailed Student’s t-test was used for statistical analysis. Data are presented as the means ± s,e,m. a,cd,e,gh, n = 3 independent experiments. b,f, n = 4 biological repeats. cd,gh, Scale bars, 25 μm. a,e, β-Tubulin was used as the loading control. c,g, The white arrows indicate Ki67+ cells.

Source data

Extended Data Fig. 6 Knockdown of APOE delays physiological and pathological aging in hMPCs.

a, Western blot analysis of APOE in replicativesenescent human MPCs (P9), HGPS human MPCs (P9), Werner syndrome human MPCs (P9) and primary hMPCs (P8) from an aged individual transduced with lentiviral sh-GL2 or sh-APOE. β-Tubulin was used as the loading control. b, Nuclear DNA staining with Hoechst 33342 in replicativesenescent human MPCs (P9), HGPS human MPCs (P9), Werner syndrome human MPCs (P9) and primary hMPCs (P8) from an aged individual transduced with lentiviral sh-GL2 or sh-APOE. c, Immunofluorescence analysis of Ki67 in replicativesenescent human MPCs (P9), HGPS human MPCs (P9), Werner syndrome human MPCs (P9), and primary hMPCs (P8) from an aged individual transduced with lentiviral sh-GL2 or sh-APOE. d, SA-β-gal staining of replicativesenescent human MPCs (P9), HGPS human MPCs (P9), Werner syndrome human MPCs (P9), and primary hMPCs (P8) from an aged individual transduced with lentiviral sh-GL2 or sh-APOE. bd, A Two-tailed Student’s t-test was used for statistical analysis. a,cd, Data are presented as the means ± s,e,m. n = 3 independent experiments. b, n = 300 cells per group. b, Scale bars, 7.5 μm. c,d, Scale bars, 25 μm. a, β-Tubulin was used as the loading control. c, The white arrows indicate Ki67+ cells.

Source data

Extended Data Fig. 7 Knockdown of APOE delays hMPC senescence induced by different stressors.

ad, Phenotypic analyses of sh-APOE-infected hMPCs (P2) after H2O2 treatment including immunofluorescence analysis of H3K9me3 (a), SA-β-gal staining (b), immunofluorescence analysis of Ki67 (c) and clonal expansion assay (d). eh, Phenotypic analyses of sh-APOE-infected hMPCs (P2) after UV irradiation including immunofluorescence analysis of H3K9me3 (e), SA-β-gal staining (f), immunofluorescence analysis of Ki67 (g) and clonal expansion assay (h). il, Phenotypic analyses of sh-APOE-infected hMPCs (P2) upon H-Rasv12 overexpression including immunofluorescence analysis of H3K9me3 (i), SA-β-gal staining (j), immunofluorescence analysis of Ki67 (k) and clonal expansion assay (l). al, A Two-tailed Student’s t-test was used for statistical analysis. a,e,i, n = 300 cells per group. bc,fg,jk, n = 3 independent experiments. d,h,l, n = 4 biological repeats. ac,eg,ik, Scale bars, 25 μm. c,g,k, The white arrows indicate Ki67+ cells.

Source data

Extended Data Fig. 8 Knockdown of APOE delays human skin fibroblast senescence.

ac, Phenotypic analysis of human skin fibroblasts (P10) transduced with lentiviruses expressing luciferase (Luc) or APOE. Immunofluorescence analysis of H3K9me3 (a) and Ki67 (b), clonal expansion assay (c) in human skin fibroblasts transduced with lentiviruses expressing Luc or APOE. df, Phenotypic analysis of human skin replicative-senescent fibroblasts (P14) transduced with lentiviral sh-GL2 or sh-APOE. Immunofluorescence analysis of H3K9me3 (d) and Ki67 (e), clonal expansion assay (f) in human skin fibroblasts transduced with lentiviral sh-GL2 or sh-APOE. gi, Phenotypic analyses of sh-APOE-transduced human skin fibroblasts (P2) after H2O2 treatment including immunofluorescence analysis of H3K9me3 (g), immunofluorescence analysis of Ki67 (h) and clonal expansion assay (i). jl, Phenotypic analyses of sh-APOE-transduced human skin fibroblasts (P2) upon H-Rasv12 overexpression including immunofluorescence analysis of H3K9me3 (j), immunofluorescence analysis of Ki67 (k) and clonal expansion assay (l). al, A Two-tailed Student’s t-test was used for statistical analysis. a,d,g,j, Data are presented as the means ± s.e.m. n = 150 cells per group. b,e,h,k, n = 3 independent experiments. c,f,i,l, n = 4 biological repeats. a,b,d,e,g,h, Scale bars, 25 μm. b,e,h,k, The white arrows indicate Ki67+ cells.

Source data

Supplementary information

Supplementary Information

Supplementary Methods and examples of gating strategy.

Reporting Summary

Supplementary Tables

Legends and descriptive captions of all the supplementary tables listed in the Excel sheet.

Source data

Source Data Fig. 1

Unprocessed western blots.

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Unprocessed western blots.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Unprocessed western blots.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 1

Unprocessed western blots.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Unprocessed western blots.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Unprocessed western blots.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 5

Unprocessed western blots.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Unprocessed western blots.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 8

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhao, H., Ji, Q., Wu, Z. et al. Destabilizing heterochromatin by APOE mediates senescence. Nat Aging 2, 303–316 (2022). https://doi.org/10.1038/s43587-022-00186-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43587-022-00186-z

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research