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:

Transcriptional and epigenetic dysregulation impairs generation of proliferative neural stem and progenitor cells during brain aging

Abstract

The decline in stem cell function during aging may affect the regenerative capacity of mammalian organisms; however, the gene regulatory mechanism underlying this decline remains unclear. Here we show that the aging of neural stem and progenitor cells (NSPCs) in the male mouse brain is characterized by a decrease in the generation efficacy of proliferative NSPCs rather than the changes in lineage specificity of NSPCs. We reveal that the downregulation of age-dependent genes in NSPCs drives cell aging by decreasing the population of actively proliferating NSPCs while increasing the expression of quiescence markers. We found that epigenetic deregulation of the MLL complex at promoters leads to transcriptional inactivation of age-dependent genes, highlighting the importance of the dynamic interaction between histone modifiers and gene regulatory elements in regulating transcriptional program of aging cells. Our study sheds light on the key intrinsic mechanisms driving stem cell aging through epigenetic regulators and identifies potential rejuvenation targets that could restore the function of aging stem cells.

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: Generation efficacy of proliferative NSPCs decreases in the aging brain.
Fig. 2: Downregulation of age-dependent genes drives NSPC aging.
Fig. 3: Age-dependent genes are potential rejuvenation targets for aging NSPCs.
Fig. 4: Changes in histone modifications at gene regulatory elements are related to transcriptional dysregulation of NSPC aging.
Fig. 5: Identification of the MLL complex as a key epigenetic regulator in the functional decline of aging NSPCs.
Fig. 6: Loss of function of the MLL complex results in transcriptional and epigenetic inactivation of age-dependent genes.

Similar content being viewed by others

Data availability

All the sequencing data have been submitted to the Gene Expression Omnibus and are publicly available as of the date of publication. The accession number for the sequencing data is GSE212725. The UniProtKB Mus musculus database was used for mass spectrometry data analysis (database link: https://www.uniprot.org/taxonomy/10090). All other data supporting the findings of this paper will be available from the corresponding author upon reasonable request. Source data are provided with this paper

References

  1. Oh, J., Lee, Y. D. & Wagers, A. J. Stem cell aging: mechanisms, regulators and therapeutic opportunities. Nat. Med. 20, 870–880 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Doetsch, F., Caille, I., Lim, D. A., Garcia-Verdugo, J. M. & Alvarez-Buylla, A. Subventricular zone astrocytes are neural stem cells in the adult mammalian brain. Cell 97, 703–716 (1999).

    Article  CAS  PubMed  Google Scholar 

  3. Lim, D. A. & Alvarez-Buylla, A. The adult ventricular-subventricular zone (V-SVZ) and olfactory bulb (OB) neurogenesis. Cold Spring Harb. Perspect. Biol. 8, a018820 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  4. Bondolfi, L., Ermini, F., Long, J. M., Ingram, D. K. & Jucker, M. Impact of age and caloric restriction on neurogenesis in the dentate gyrus of C57BL/6 mice. Neurobiol. Aging 25, 333–340 (2004).

    Article  CAS  PubMed  Google Scholar 

  5. Maslov, A. Y., Barone, T. A., Plunkett, R. J. & Pruitt, S. C. Neural stem cell detection, characterization, and age-related changes in the subventricular zone of mice. J. Neurosci. 24, 1726–1733 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Molofsky, A. V. et al. Increasing p16INK4a expression decreases forebrain progenitors and neurogenesis during ageing. Nature 443, 448–452 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Tobin, M. K. et al. Human hippocampal neurogenesis persists in aged adults and Alzheimer’s disease patients. Cell Stem Cell 24, 974–982 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Liu, P. et al. Histone deacetylation promotes mouse neural induction by restricting nodal-dependent mesendoderm fate. Nat. Commun. 6, 6830 (2015).

    Article  CAS  PubMed  Google Scholar 

  9. Huang, C. et al. Cbx3 maintains lineage specificity during neural differentiation. Genes Dev. 31, 241–246 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Qiao, Y., Yang, X. & Jing, N. Epigenetic regulation of early neural fate commitment. Cell Mol. Life Sci. 73, 1399–1411 (2016).

    Article  CAS  PubMed  Google Scholar 

  11. Pearson, C. A. et al. Foxp1 regulates neural stem cell self-renewal and bias toward deep layer cortical fates. Cell Rep. 30, 1964–1981 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Zhang, K. et al. Distinct functions of BMP4 during different stages of mouse ES cell neural commitment. Development 137, 2095–2105 (2010).

    Article  CAS  PubMed  Google Scholar 

  13. Hirabayashi, Y. & Gotoh, Y. Epigenetic control of neural precursor cell fate during development. Nat. Rev. Neurosci. 11, 377–388 (2010).

    Article  CAS  PubMed  Google Scholar 

  14. Kalamakis, G. et al. Quiescence modulates stem cell maintenance and regenerative capacity in the aging brain. Cell 176, 1407–1419 (2019).

    Article  CAS  PubMed  Google Scholar 

  15. Lupo, G. et al. Molecular profiling of aged neural progenitors identifies Dbx2 as a candidate regulator of age-associated neurogenic decline. Aging Cell 17, e12745 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  16. Yang, J. H. et al. Loss of epigenetic information as a cause of mammalian aging. Cell 186, 305–326 (2023).

    Article  CAS  PubMed  Google Scholar 

  17. Ellis, P. et al. SOX2, a persistent marker for multipotential neural stem cells derived from embryonic stem cells, the embryo or the adult. Dev. Neurosci. 26, 148–165 (2004).

    Article  CAS  PubMed  Google Scholar 

  18. Ferri, A. L. et al. Sox2 deficiency causes neurodegeneration and impaired neurogenesis in the adult mouse brain. Development 131, 3805–3819 (2004).

    Article  CAS  PubMed  Google Scholar 

  19. Suh, H. et al. In vivo fate analysis reveals the multipotent and self-renewal capacities of Sox2+ neural stem cells in the adult hippocampus. Cell Stem Cell 1, 515–528 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Brazel, C. Y. et al. Sox2 expression defines a heterogeneous population of neurosphere-forming cells in the adult murine brain. Aging Cell 4, 197–207 (2005).

    Article  CAS  PubMed  Google Scholar 

  21. Stuart, T. et al. Comprehensive integration of single-cell data. Cell 177, 1888–1902 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Mich, J. K. et al. Prospective identification of functionally distinct stem cells and neurosphere-initiating cells in adult mouse forebrain. eLife 3, e02669 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  23. Giachino, C. et al. Molecular diversity subdivides the adult forebrain neural stem cell population. Stem Cells 32, 70–84 (2014).

    Article  CAS  PubMed  Google Scholar 

  24. Kim, E. J., Leung, C. T., Reed, R. R. & Johnson, J. E. In vivo analysis of Ascl1 defined progenitors reveals distinct developmental dynamics during adult neurogenesis and gliogenesis. J. Neurosci. 27, 12764–12774 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Ponti, G. et al. Cell cycle and lineage progression of neural progenitors in the ventricular-subventricular zones of adult mice. Proc. Natl Acad. Sci. USA 110, E1045–E1054 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Cao, J. et al. The single-cell transcriptional landscape of mammalian organogenesis. Nature 566, 496–502 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Shin, J. et al. Single-cell RNA-seq with waterfall reveals molecular cascades underlying adult neurogenesis. Cell Stem Cell 17, 360–372 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Basak, O. et al. Troy+ brain stem cells cycle through quiescence and regulate their number by sensing niche occupancy. Proc. Natl Acad. Sci. USA 115, E610–E619 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Adusumilli, V. S. et al. ROS dynamics delineate functional states of hippocampal neural stem cells and link to their activity-dependent exit from quiescence. Cell Stem Cell 28, 300–314 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Naeve, G. S. et al. Neuritin: a gene induced by neural activity and neurotrophins that promotes neuritogenesis. Proc. Natl Acad. Sci. USA 94, 2648–2653 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Erickson, R. I., Paucar, A. A., Jackson, R. L., Visnyei, K. & Kornblum, H. Roles of insulin and transferrin in neural progenitor survival and proliferation. J. Neurosci. Res. 86, 1884–1894 (2008).

    Article  CAS  PubMed  Google Scholar 

  32. Boschian, C. et al. Impaired neuronal differentiation of neural stem cells lacking the Engrailed-2 gene. Neuroscience 386, 137–149 (2018).

    Article  CAS  PubMed  Google Scholar 

  33. Khodosevich, K. et al. Connexin45 modulates the proliferation of transit-amplifying precursor cells in the mouse subventricular zone. Proc. Natl Acad. Sci. USA 109, 20107–20112 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Young, F. I. et al. The doublesex-related Dmrta2 safeguards neural progenitor maintenance involving transcriptional regulation of Hes1. Proc. Natl Acad. Sci. USA 114, E5599–E5607 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Lim, D. A. et al. Chromatin remodelling factor Mll1 is essential for neurogenesis from postnatal neural stem cells. Nature 458, 529–533 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Apple, D. M., Mahesula, S., Fonseca, R. S., Zhu, C. & Kokovay, E. Calorie restriction protects neural stem cells from age-related deficits in the subventricular zone. Aging (Albany NY) 11, 115–126 (2019).

    Article  CAS  PubMed  Google Scholar 

  37. Parish, E. V., Mason, J. O. & Price, D. J. Expression of Barhl2 and its relationship with Pax6 expression in the forebrain of the mouse embryo. BMC Neurosci. 17, 76 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  38. Bates, M. D., Dunagan, D. T., Welch, L. C., Kaul, A. & Harvey, R. P. The Hlx homeobox transcription factor is required early in enteric nervous system development. BMC Dev. Biol. 6, 33 (2006).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Li, Z. et al. An HMGA2-IGF2BP2 axis regulates myoblast proliferation and myogenesis. Dev. Cell 23, 1176–1188 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Mori, H. et al. Expression of mouse igf2 mRNA-binding protein 3 and its implications for the developing central nervous system. J. Neurosci. Res. 64, 132–143 (2001).

    Article  CAS  PubMed  Google Scholar 

  41. Leitner, D. F. & Connor, J. R. Functional roles of transferrin in the brain. Biochim. Biophys. Acta 1820, 393–402 (2012).

    Article  CAS  PubMed  Google Scholar 

  42. Fan, X. et al. TWIST1 and chromatin regulatory proteins interact to guide neural crest cell differentiation. eLife 10, e62873 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. McKey, J., Martire, D., de Santa Barbara, P. & Faure, S. LIX1 regulates YAP1 activity and controls the proliferation and differentiation of stomach mesenchymal progenitors. BMC Biol. 14, 34 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Chua, H. C., Wulf, M., Weidling, C., Rasmussen, L. P. & Pless, S. A. The NALCN channel complex is voltage sensitive and directly modulated by extracellular calcium. Sci. Adv. 6, eaaz3154 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Preitner, N., Quan, J., Li, X., Nielsen, F. C. & Flanagan, J. G. IMP2 axonal localization, RNA interactome, and function in the development of axon trajectories. Development 143, 2753–2759 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Pastrana, E., Cheng, L. C. & Doetsch, F. Simultaneous prospective purification of adult subventricular zone neural stem cells and their progeny. Proc. Natl Acad. Sci. USA 106, 6387–6392 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Shah, P. T. et al. Single-cell transcriptomics and fate mapping of ependymal cells reveals an absence of neural stem cell function. Cell 173, 1045–1057 (2018).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  49. Huang, C. & Wu, J. C. Epigenetic modulations of induced pluripotent stem cells: novel therapies and disease models. Drug Discov. Today Dis. Models 9, e153–e160 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  50. Sun, F. et al. Promoter–enhancer communication occurs primarily within insulated neighborhoods. Mol. Cell 73, 250–263 (2019).

    Article  CAS  PubMed  Google Scholar 

  51. Chronis, C. et al. Cooperative binding of transcription factors orchestrates reprogramming. Cell 168, 442–459 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Park, J., Lee, K., Kim, K. & Yi, S. J. The role of histone modifications: from neurodevelopment to neurodiseases. Signal Transduct. Target. Ther. 7, 217 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. 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 

  54. Cho, Y. W. et al. PTIP associates with MLL3- and MLL4-containing histone H3 lysine 4 methyltransferase complex. J. Biol. Chem. 282, 20395–20406 (2007).

    Article  CAS  PubMed  Google Scholar 

  55. Dhar, S. S. et al. Trans-tail regulation of MLL4-catalyzed H3K4 methylation by H4R3 symmetric dimethylation is mediated by a tandem PHD of MLL4. Genes Dev. 26, 2749–2762 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Huisman, C. et al. The histone H3-lysine 4-methyltransferase Mll4 regulates the development of growth hormone-releasing hormone-producing neurons in the mouse hypothalamus. Nat. Commun. 12, 256 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Sridharan, R. et al. Proteomic and genomic approaches reveal critical functions of H3K9 methylation and heterochromatin protein-1γ in reprogramming to pluripotency. Nat. Cell Biol. 15, 872–882 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Miranda, C. J. et al. Aging brain microenvironment decreases hippocampal neurogenesis through Wnt-mediated survivin signaling. Aging Cell 11, 542–552 (2012).

    Article  PubMed  Google Scholar 

  59. Sun, D. et al. Epigenomic profiling of young and aged HSCs reveals concerted changes during aging that reinforce self-renewal. Cell Stem Cell 14, 673–688 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Liu, L. et al. Chromatin modifications as determinants of muscle stem cell quiescence and chronological aging. Cell Rep. 4, 189–204 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Maybury-Lewis, S. Y. et al. Changing and stable chromatin accessibility supports transcriptional overhaul during neural stem cell activation and is altered with age. Aging Cell 20, e13499 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Sun, Y. et al. Neurogenin promotes neurogenesis and inhibits glial differentiation by independent mechanisms. Cell 104, 365–376 (2001).

    Article  CAS  PubMed  Google Scholar 

  63. Sun, H. et al. Quantitative integration of epigenomic variation and transcription factor binding using MAmotif toolkit identifies an important role of IRF2 as transcription activator at gene promoters. Cell Discov. 4, 38 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  64. Liu, Z., Scannell, D. R., Eisen, M. B. & Tjian, R. Control of embryonic stem cell lineage commitment by core promoter factor, TAF3. Cell 146, 720–731 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Corenblum, M. J. et al. Reduced Nrf2 expression mediates the decline in neural stem cell function during a critical middle-age period. Aging Cell 15, 725–736 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Anandhan, A., Kirwan, K. R., Corenblum, M. J. & Madhavan, L. Enhanced NRF2 expression mitigates the decline in neural stem cell function during aging. Aging Cell 20, e13385 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Obernier, K. & Alvarez-Buylla, A. Neural stem cells: origin, heterogeneity and regulation in the adult mammalian brain. Development 146, dev156059 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Kaise, T. et al. Functional rejuvenation of aged neural stem cells by Plagl2 and anti-Dyrk1a activity. Genes Dev. 36, 23–37 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Shan, Q., Hu, Y., Chen, S. & Tian, Y. Nucleus accumbens dichotomically controls social dominance in male mice. Neuropsychopharmacology 47, 776–787 (2022).

    Article  CAS  PubMed  Google Scholar 

  70. Guo, W., Patzlaff, N. E., Jobe, E. M. & Zhao, X. Isolation of multipotent neural stem or progenitor cells from both the dentate gyrus and subventricular zone of a single adult mouse. Nat. Protoc. 7, 2005–2012 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Livak, K. J. & Schmittgen, T. D. Analysis of relative gene expression data using real-time quantitative PCR and the \(2^{-\Delta\Delta C_{\rm T}}\) method. Methods 25, 402–408 (2001).

    Article  CAS  PubMed  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank D. Qin, Q. Shan and Z. Cai (Shantou University Medical College) for their technical support of stereotactic injection of mice. Thanks to B. Wang for technical support of animal studies. Thanks to X. Hu for technical support of NSPC isolation from the SVZ. Thanks to T. Su (University of California, Los Angeles) for bioinformatic support. Thanks to C. Peng and Y. Yin of the Mass Spectrometry System at the National Facility for Protein Science in Shanghai, Shanghai Advanced Research Institute, Chinese Academy of Sciences, for data collection and analysis. This work was supported by the National Natural Science Foundation of China (grant no. 81671396, to C.H.), the Natural Science Foundation of Guangdong Province (grant no. 2017A030313780, to C.H.) and a National Institutes of Health R01 grant (grant no. GM074701, to M.C.).

Author information

Authors and Affiliations

Authors

Contributions

M.L. did most experiments and statistical analyses. H.G. performed the animal experiments. C.H. analyzed the bioinformatic data. C.H. and M.C. supervised the study.

Corresponding authors

Correspondence to Michael Carey or Chengyang Huang.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Aging thanks Orly Lazarov 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 Aged NSPCs exhibit reduced potential for differentiating into neurons compared to young NSPCs.

a, Confocal microscopy images showing immunofluorescence co-labeling of DCX and BrdU in the coronal SVZ sections from young and aged mice. Scale bar, 40 μm. The quantification of DCX and BrdU double-positive cells was performed in both the lateral and dorsolateral SVZ for each section. A total of four sections per mouse, spaced at 120 μm intervals along the rostrocaudal axis of the SVZ, were used for cell counting. Seven mice per young group. Six mice per aged group. The average number of DCX and BrdU double-positive cells per group was plotted in the chart on the right panel. P = 0.0009. **** P < 0.001. b, Immunofluorescence labeling of SOX2 in NSPCs isolated from the young and aged SVZ, respectively. Primary NSPC-derived neurospheres were dissociated into single cells for staining. Nuclei were stained with DAPI. Scale bar, 200 μm. Quantification data were plotted, showing the percentage of SOX2-positive cells among DAPI-positive cells counted from 10 microscopic fields per age group. ns: not significant.c, Immunofluorescence labeling of NESTIN in NSPCs isolated from the young and aged SVZ, respectively. The staining and quantification methods used are identical to those described in (b). d, The size of primary NSPC-derived neurospheres in young and aged groups. Neurospheres were imaged after 6 days of culture, and their diameters were measured through ImageJ software. n = 80. P = 3.05E-15. **** P < 0.001. e, Luminescent cell viability analysis of young and aged NSPCs cultured from Day 1 to Day 4. n = 3 independent experiments. Day1: P = 0.2436, Day2: P = 0.0012, Day3: P = 0.0013, Day4: P = 0.0003. *** P < 0.005. **** P < 0.001. ns: not significant. f, Cell death analysis by propidium iodide (PI) staining. Scale bar, 200 μm. Quantification data were plotted, showing the percentage of PI-positive cells among Hoechst-positive cells counted from 10 microscopic fields per age group. g, Neural differentiation starting from NSPC-derived neurosphere as neurite outgrows. After 2 days of differentiation, cells were fixed and subjected to immunofluorescence labeling of neuron marker TUJ1. The number of TUJ1+ differentiated cells with neurites that migrated away from the neurosphere was counted. Scale bar, 200 μm. Data were plotted with the percentage of TUJ1 versus DAPI-positive cells with neurites in each neurosphere differentiation. n = 10. P = 6.71E-05. h, Immunofluorescence labeling of neuron marker MAP2 four days post-differentiation. The number of MAP2+ differentiated cells that migrated away from the neurosphere was counted. Scale bar, 200 μm. n = 8 in young group. n = 13 in aged group. P = 4.83E-06. i, Immunofluorescence labeling of astrocyte marker GFAP four days post-differentiation. The number of GFAP+ differentiated cells that migrated away from the neurosphere was counted. Scale bar, 200 μm. Data were plotted with the percentage of GFAP-positive cells with astrocytic extensions versus DAPI-positive cells in each neurosphere differentiation. n = 13 per group. Data analysis for g-i: P = 3.99E-07. *** P < 0.005. **** P < 0.001. Error bars are mean ± SD in all graphs. Two-sided Student’s t-test for statistical analysis in all graphs.

Source data

Extended Data Fig. 2 Identification of NSPC activation states and lineage specificity through single-cell transcriptome.

a, Heatmap of top 10 differentially expressed marker genes in 4 clusters of NSPCs. Analysis was performed using merged scRNA-seq datasets of young and aged NSPCs. b, Ridgeline plots of merged scRNA-seq datasets of young and aged NSPCs, showing expression changes of activation markers from early type B1a cells to late type C cells. c, Violin plots showing the similar expression levels of NSPC lineage markers among 4 clusters between young and aged NSPCs. d, UMAP plots of NSPC lineage markers in young and aged NSPCs. e, Single-cell pseudotime analysis using Monocle3 shows dynamic transition of NSPC activation states along the trajectory. NSPCs with larger pseudotime values exhibit higher activity in proliferation. f, Gene ontology analysis of the DE markers of early type B1a cells. The gene number of each GO category is showed on the right side of the bar chart.

Source data

Extended Data Fig. 3 Transcriptional reversal of age-dependent genes is associated with the functional improvement of NSPCs in mice subjected to calorie restriction.

a, Percentage of BrdU and SOX2 double-positive versus SOX2-positive cells in the brain SVZs of young and aged mice subjected to AL and CR treatments. Seven mice per young group; eight mice per aged group. Twelve sections per mouse. Error bars are mean ± SD. Unpaired two-sided Student’s t-test. Young: P = 0.0146, Aged: P = 0.3789. * P < 0.05. ns: not significant. b, Quantification of DCX and BrdU double-positive cells in both the lateral and dorsolateral SVZ of young and aged mice subjected to AL and CR treatments. A total of four sections per mouse, spaced at 120 μm intervals along the rostrocaudal axis of the SVZ, were used for cell counting. Seven mice per young group. Six mice per aged group. Error bars are mean ± SD. Unpaired two-sided Student’s t-test. Young: P = 0.0024, Aged: P = 0.28. *** P < 0.005. ns: not significant. c, Representative images of neurospheres formed by primary NSPCs isolated from the SVZ of mice subjected to ad libitum (AL) feeding or calorie restriction (CR) within each age group. Scale bar, 1000 μm. The SVZ tissues were pooled from five mice per group. d, Number of neurospheres was quantified across four groups of mice as in (a). n = 3 independent cultures. Six microscopic fields were imaged from each culture replicate for cell counting. Error bars are mean ± SD. Unpaired two-sided Student’s t-test. Young_AL vs. Young_CR: P = 0.0032, Young_AL vs. Aged_AL: P = 0.0009, Aged_AL vs. Aged_CR: P = 0.5534. *** P < 0.005. **** P < 0.001. ns: not significant. e, f, Heatmap showing the fold change in transcriptional levels of age-dependent genes (Aged vs. Young, FC > 1.5) and their transcriptional reversal in young (c) or aged (d) mice subjected to calorie restriction (CR vs. AL, FC > 1.5).

Source data

Extended Data Fig. 4 Downregulation of age-dependent gene upregulates NSC quiescence markers.

a, Heatmap showing scRNA-seq differential expression analysis of the age-dependent genes identified in bulk RNA-seq in each NSPC cluster. Data are the average log2 fold change of expression. b, Heatmap showing bulk RNA-seq differential expression analysis of DE genes detected in early type B1a cells as in Fig. 1j. Fold change of RNA level (Aged_FPKM vs. Young_FPKM) and p-value were plotted. c, d, Quantitative real-time PCR analysis of relative expression (siRNA group vs. siControl group) of Aldoc and Atp1a2 in young NSPCs 48 hours after siRNA transfection. n = 3 independent assays. Error bars are mean ± SD. Unpaired two-sided Student’s t-test.

Source data

Extended Data Fig. 5 The rejuvenating effects of age-dependent genes on aged NSPCs.

a, Quantification of SOX2-positive cells in the SVZ. The average percentage of SOX2+ /DAPI+ cells was plotted based on data from three aged wild-type mice. A total of eight sections per mouse, spaced at 120 μm intervals along the rostrocaudal axis of the SVZ, were used for cell counting. Cells were quantified within the lateral and dorsolateral SVZ in each section. b–d, Quantification of BrdU+SOX2+ NSPCs in the left (Igf2bp2, Igf2bp3, and Barhl2 overexpression) and right (wild-type) sides of the SVZ. The adenovirus encoding these genes were injected in the left side of SVZ of aged mice, respectively. BrdU labeling was performed two weeks after the gene transduction. BrdU+SOX2+ NSPCs were quantified at both sides of the lateral and dorsolateral SVZ in each section. Seven sections per mouse. Three mice per group. Unpaired two-sided Student’s t-test. (b): P = 0.0396, (c): P = 5.05E-09, (d): P = 1.39E-08. * P < 0.05. **** P < 0.001. e, f, Overexpression of Igf2bp2 and Igf2bp3 increases the number of DCX-positive cells in the SVZ of aged mice. Control mice were injected with control adenovirus without expressing these genes. Confocal microscopy images showing immunofluorescence co-labeling of DCX and DAPI in the SVZ. Scale bar, 20 μm. DCX-positive cells were quantified at the injection site within the lateral and dorsolateral SVZ in each section. A total of six sections per mouse, spaced at 120 μm intervals along the rostrocaudal axis of the SVZ, were used for cell counting. Three mice per group. The average number of DCX-positive cells per group was plotted blow the images. Unpaired two-sided Student’s t-test. (e): P = 0.0355, (f): P = 0.0216. * P < 0.05. g–i, Quantitative real-time PCR analysis of relative expression of Igf2bp2, Igf2bp3 and Barhl2 40 hours after the adenovirus-mediated transduction of the respective genes in primary aged NSPCs. The control group was transduced with control adenovirus without expressing these genes. n = 3 independent assays. Unpaired two-sided Student’s t-test. j, Cell death analysis by propidium iodide (PI) staining in primary aged NSPCs transduced with the genes of Igf2bp2, Igf2bp3 and Barhl2. NSPC-formed neurospheres were dissociated into single cells for staining 48 hours following adenovirus-mediated transduction of the respective genes. Scale bar, 200 μm. k, Quantification data of PI staining shows the percentage of PI-positive cells among Hoechst-positive cells, which were counted from 10 microscopic fields per group. Error bars are mean ± SD for b-k.

Source data

Extended Data Fig. 6 Changes in average profiles of histone marks and ATAC-seq signals at promoters and enhancers in aging NSPCs.

a, Average profile of H3K4me3 and H3K27me3 at the promoters of all genes spanning TSSs in young and aged NSPCs (Two-sided Wilcoxon rank sum test, H3K4me3 data p-value = 0.2209, H3K27me3 data p-value = 6.69E-15). b, Average profile of H3K27ac signals spanning the center of enhancers of all genes in young and aged NSPCs (Two-sided Wilcoxon rank sum test, H3K27ac data p-value = 0.2684). c, Pearson correlation analysis of fold changes of RNA levels and promoter H3K4me3 signals of age-dependent genes. 95% confidence interval. d, Pearson correlation analysis of fold changes of RNA levels and enhancer H3K27ac signals of age-dependent genes. 95% confidence interval. e, f, Average profiles of ATAC- seq at promoters and enhancers of all genes show no genome-wide changes of chromatin accessibility across the regulatory elements during NSPC aging (Two-sided Wilcoxon rank sum test, promoter profile p-value = 0.9261, enhancer profile p-value = 0.6969).

Source data

Extended Data Fig. 7 Histone modifications are associated with chromatin accessibility at the promoters of age-dependent genes in aging NSPCs.

a, Heatmap shows fold change (Aged vs. Young) of RNA levels of age-dependent genes and the average signals of ATAC-seq at their regulatory elements. b, Venn diagrams show the overlap of age-dependent genes that have decreased levels of chromatin accessibility and H3K4me3 at the promoters during NSPC aging. Two-sided Hypergeometric test for significant overlap analysis, * p-value = 5.602e-26. c, Venn diagrams show the overlap of age- dependent genes that have decreased levels of chromatin accessibility and H3K27ac signals at the enhancers during NSPC aging. Two-sided Hypergeometric test, * p-value = 3.618e-16. d, Browser tracks of ATAC-seq and ChIP-seq show a similar level of signals at the promoters of NSPC lineage markers and housekeeping genes between the young and aged groups. e, Browser tracks of 3 age-dependent down-regulated genes (Igf2bp3, Twist1 and Otx2) and 2 up-regulated genes (Gpr37l1 and Ifi27) that have changes of ATAC-seq signals and histone modifications at the promoters during NSPC aging.

Source data

Extended Data Fig. 8 Age-related changes in histone modifications and transcription in aging EGFR+ NSPCs.

a, FACS plots of the isolation of EGFR-positive and 7-AAD negative NSPCs from the SVZ of young and aged mice. b, Average profile of H3K4me3 at the promoters of all genes spanning TSSs in young and aged EGFR+ NSPCs (Two-sided Wilcoxon rank sum test, H3K4me3 data p-value = 0.5428). c, Browser tracks of CUT&Tag signals of H3K4me3 at the promoters of NSPC lineage markers in young and aged EGFR+ NSPCs. d, Heatmap showing the fold change of average CUT&Tag signals of H3K4me3 at each promoter of age-dependent genes in aged NSPC-derived neurospheres or EGFR+ NSPCs compared to their young counterparts (Aged vs. Young, FC > 1.5). e, Browser tracks of CUT&Tag signals at the promoters of Barhl2, Igf2bp2, and Igf2bp3. f, Real-time qPCR analysis of relative expression of genes in aged versus young EGFR+ NSPCs. Error bars are mean ± SD. n = 3 independent assays. Two-sided Student’s t-tests. Otx2: P = 0.0021, Rimbp2: P = 0.0003, Twist1: P = 0.0099, Igf2bp3: P = 7.77E-06, Barhl2: P = 0.0001, Fam155a: P = 0.0382, Epb41l13: P = 0.0109, Fn3k: P = 0.0174, Igf2bp2: P = 7.57E-05, Angpt2: P = 0.0035. * P < 0.05. ** P < 0.01. *** P < 0.005. **** P < 0.001.

Source data

Extended Data Fig. 9 Cell death analysis by propidium iodide (PI) staining in NSPCs following knockdown of MLL complex subunits.

a, Representative immunofluorescence images showing PI staining in siRNA-transfected young NSPCs. NSPC-derived neurospheres were dissociated into single cells for PI staining 48 hours post-transfection of siRNAs. Hoechst (blue) staining visualizes the nuclei. Scale bar, 200 μm. b, Quantification of PI staining in siRNA-transfected young NSPCs, displaying the percentage of PI-positive cells among Hoechst-positive cells. The quantification was performed by counting from 10 microscopic fields per group. Error bars are mean ± SD.

Source data

Extended Data Fig. 10 Interaction between MLL complex and Cbx3 in aging NSPCs.

a, Mass spectrometry peptide counts (number of unique peptides) of MLL complex components identified in KMT2D immunoprecipitation product from young NSPC nuclear extract. b, Mass spectrometry peptide counts (number of unique peptides) of proteins identified in CBX3 immunoprecipitation product from young NSPC nuclear extract. c, Western blot analysis of endogenous MLL complex components in CBX3 and IgG immunoprecipitation products from the young and aged NSPC nuclear extracts. Bar charts show the relative levels of the indicated proteins in aged versus young IP groups analyzed by Image J software. Error bars are mean ± SD. n = 2 independent experiments, Unpaired two-sided Student’s t-tests. KMT2D: P = 0.0222, RBBP5: P = 0.0015, WDR5: P = 0.6499, CBX3: P = 0.4363. * P < 0.05. *** P < 0.005. ns: not significant.

Source data

Supplementary information

Supplementary Information

Supplementary Figure and Tables 1–3.

Reporting Summary

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

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

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

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.

Source Data Extended Data Fig. 9

Statistical source data.

Source Data Extended Data Fig. 10

Statistical source data.

Source Data Fig. 2

Unprocessed western blots.

Source Data Fig. 5

Unprocessed western blots.

Source Data Extended Data Fig. 10

Unprocessed western blots.

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

Li, M., Guo, H., Carey, M. et al. Transcriptional and epigenetic dysregulation impairs generation of proliferative neural stem and progenitor cells during brain aging. Nat Aging 4, 62–79 (2024). https://doi.org/10.1038/s43587-023-00549-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43587-023-00549-0

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