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:

Acetylation profiling by Iseq-Kac reveals insights into HSC aging and lineage decision

A Publisher Correction to this article was published on 04 June 2025

This article has been updated

Abstract

Profiling post-translational modifications face challenges with low-input samples. We developed Iseq-Kac (internal standard-assisted enrichment-free approach for high-throughput quantitative analysis of lysine acetylation) to profile the acetylome in as few as 103–104 cells. By using a hyperacetylated internal standard, Iseq-Kac can be used in mass spectrometry (MS) to enhance MS1 signals and facilitate MS2 fragmentation of acetylated peptides. Using Iseq-Kac, we quantified 675–1,471 acetylated peptides per analysis from 104 hematopoietic stem cells (HSCs) or multipotent progenitors. Validation by targeted MS, site-specific antibodies and functional assays linked aging-related proteome and acetylome changes to HSC lineage decision. A pronounced decrease in acetylation at H4 lysine 77 (H4K77ac) was observed in aged HSCs, linked to histone deacetylase 3 (HDAC3) activity. HDAC3 inhibition or knockdown in HSCs significantly promoted lymphocyte differentiation. Mimicking H4K77ac through H4K77Q expression enhanced B cell differentiation while repressing myeloid differentiation. Overall, Iseq-Kac enables robust low-input acetylome profiling and reveals epigenetic mechanisms underlying lineage skewing in aged HSCs.

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: Concept and evaluation of the Iseq-Kac strategy.
Fig. 2: Evaluation of the HSPC proteome data generated by Iseq-Kac.
Fig. 3: Acetylome analysis reveals aging-associated changes in HSPCs.
Fig. 4: Histone modification H4K77ac is associated with lymphocyte differentiation in HSCs.
Fig. 5: RGFP966 restores aging-related H4K77 hypoacetylation and enhances lymphocyte differentiation in aged HSCs.
Fig. 6: H4K77ac promotes in vivo lymphocyte differentiation of HSCs.

Similar content being viewed by others

Data availability

Raw MS data were deposited to the ProteomeXchange Consortium through the iProX partner repository with dataset identifier PXD021651. The data for CUT&Tag and ATAC-seq were deposited to the National Center for Biotechnology information under BioProject PRJNA1236923. All data are contained within the article and Supplementary Information. Source data are provided with this paper.

Code availability

Code specifically developed for this study is available from GitHub (https://github.com/MonnaHu/Iseq-Kac/tree/master).

Change history

References

  1. Zhong, Q. et al. Protein posttranslational modifications in health and diseases: functions, regulatory mechanisms, and therapeutic implications. MedComm (2020) 4, e261 (2023).

    Article  CAS  PubMed  Google Scholar 

  2. Hansen, B. K. et al. Analysis of human acetylation stoichiometry defines mechanistic constraints on protein regulation. Nat. Commun. 10, 1055 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  3. Olsen, J. V. & Mann, M. Status of large-scale analysis of post-translational modifications by mass spectrometry. Mol. Cell. Proteom. 12, 3444–3452 (2013).

    Article  CAS  Google Scholar 

  4. Li, X. et al. Inflammation and aging: signaling pathways and intervention therapies. Signal Transduct. Target. Ther. 8, 239 (2023).

    Article  PubMed  PubMed Central  Google Scholar 

  5. Ross, J. B. et al. Depleting myeloid-biased haematopoietic stem cells rejuvenates aged immunity. Nature 628, 162–170 (2024).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Kanayama, M. et al. Skewing of the population balance of lymphoid and myeloid cells by secreted and intracellular osteopontin. Nat. Immunol. 18, 973–984 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Kasbekar, M., Mitchell, C. A., Proven, M. A. & Passegué, E. Hematopoietic stem cells through the ages: a lifetime of adaptation to organismal demands. Cell Stem Cell 30, 1403–1420 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Signer, R. A. J., Magee, J. A., Salic, A. & Morrison, S. J. Haematopoietic stem cells require a highly regulated protein synthesis rate. Nature 509, 49–54 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Zaro, B. W. et al. Proteomic analysis of young and old mouse hematopoietic stem cells and their progenitors reveals post-transcriptional regulation in stem cells. eLife 9, e62210 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Cabezas-Wallscheid, N. et al. Identification of regulatory networks in HSCs and their immediate progeny via integrated proteome, transcriptome, and DNA methylome analysis. Cell Stem Cell 15, 507–522 (2014).

    Article  CAS  PubMed  Google Scholar 

  11. Amon, S. et al. Sensitive quantitative proteomics of human hematopoietic stem and progenitor cells by data-independent acquisition mass spectrometry. Mol. Cell. Proteom. 18, 1454–1467 (2019).

    Article  CAS  Google Scholar 

  12. Jassinskaja, M. et al. Comprehensive proteomic characterization of ontogenic changes in hematopoietic stem and progenitor cells. Cell Rep. 21, 3285–3297 (2017).

    Article  CAS  PubMed  Google Scholar 

  13. Chen, Z. et al. Cohesin-mediated NF-κB signaling limits hematopoietic stem cell self-renewal in aging and inflammation. J. Exp. Med. 216, 152–175 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Budnik, B., Levy, E., Harmange, G. & Slavov, N. SCoPE-MS: mass spectrometry of single mammalian cells quantifies proteome heterogeneity during cell differentiation. Genome Biol. 19, 018–1547 (2018).

    Article  Google Scholar 

  15. Wühr, M. et al. Accurate multiplexed proteomics at the MS2 level using the complement reporter ion cluster. Anal. Chem. 84, 9214–9221 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  16. Virreira Winter, S. et al. EASI-tag enables accurate multiplexed and interference-free MS2-based proteome quantification. Nat. Methods 15, 527–530 (2018).

    Article  CAS  PubMed  Google Scholar 

  17. Schölz, C. et al. Acetylation site specificities of lysine deacetylase inhibitors in human cells. Nat. Biotechnol. 33, 415–423 (2015).

    Article  PubMed  Google Scholar 

  18. Pessoa Rodrigues, C. & Akhtar, A. Differential H4K16ac levels ensure a balance between quiescence and activation in hematopoietic stem cells. Sci. Adv. 7, eabi5987 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Wang, H. et al. SIRT6 controls hematopoietic stem cell homeostasis through epigenetic regulation of Wnt signaling. Cell Stem Cell 18, 495–507 (2016).

    Article  CAS  PubMed  Google Scholar 

  20. Wilson, A. et al. Dormant and self-renewing hematopoietic stem cells and their niches. Ann. N. Y. Acad. Sci. 1106, 64–75 (2007).

    Article  CAS  PubMed  Google Scholar 

  21. 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 

  22. Xing, Z. et al. Increased hematopoietic stem cell mobilization in aged mice. Blood 108, 2190–2197 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Grover, A. et al. Single-cell RNA sequencing reveals molecular and functional platelet bias of aged haematopoietic stem cells. Nat. Commun. 7, 11075 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Lu, Z. et al. Phosphatase, Mg2+/Mn2+ dependent 1B regulates the hematopoietic stem cells homeostasis via the Wnt/β-catenin signaling. Haematologica 109, 2144–2156 (2024).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Chan, G. et al. Essential role for Ptpn11 in survival of hematopoietic stem and progenitor cells. Blood 117, 4253–4261 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Georgopoulos, K. et al. The Ikaros gene is required for the development of all lymphoid lineages. Cell 79, 143–156 (1994).

    Article  CAS  PubMed  Google Scholar 

  27. Ng, S. Y., Yoshida, T., Zhang, J. & Georgopoulos, K. Genome-wide lineage-specific transcriptional networks underscore Ikaros-dependent lymphoid priming in hematopoietic stem cells. Immunity 30, 493–507 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Kirstetter, P., Thomas, M., Dierich, A., Kastner, P. & Chan, S. Ikaros is critical for B cell differentiation and function. Eur. J. Immunol. 32, 720–730 (2002).

    Article  CAS  PubMed  Google Scholar 

  29. Feng, X. et al. Foxp1 is an essential transcriptional regulator for the generation of quiescent naive T cells during thymocyte development. Blood 115, 510–518 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Hu, H. et al. Foxp1 is an essential transcriptional regulator of B cell development. Nat. Immunol. 7, 819–826 (2006).

    Article  CAS  PubMed  Google Scholar 

  31. Milner, J. J. et al. Runx3 programs CD8+ T cell residency in non-lymphoid tissues and tumours. Nature 552, 253–257 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Cronk, J. C. et al. Methyl-CpG binding protein 2 regulates microglia and macrophage gene expression in response to inflammatory stimuli. Immunity 42, 679–691 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Lu, H. C. et al. STAT3 signaling in myeloid cells promotes pathogenic myelin-specific T cell differentiation and autoimmune demyelination. Proc. Natl Acad. Sci. USA 117, 5430–5441 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Feng, M. et al. YBX1 is required for maintaining myeloid leukemia cell survival by regulating BCL2 stability in an m6A-dependent manner. Blood 138, 71–85 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Nakamura-Ishizu, A., Ito, K. & Suda, T. Hematopoietic stem cell metabolism during development and aging. Dev. Cell 54, 239–255 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Krug, K. et al. Proteogenomic landscape of breast cancer tumorigenesis and targeted therapy. Cell 183, 1436–1456 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Dou, Y. et al. Proteogenomic insights suggest druggable pathways in endometrial carcinoma. Cancer Cell 41, 1586–1605 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Fang, F. et al. A distinct isoform of ZNF207 controls self-renewal and pluripotency of human embryonic stem cells. Nat. Commun. 9, 4384 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Wang, M. et al. Genotoxic aldehyde stress prematurely ages hematopoietic stem cells in a p53-driven manner. Mol. Cell 83, 2417–2433 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Wang, Z. et al. Quantitative phosphoproteomic analysis of the molecular substrates of sleep need. Nature 558, 435–439 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Zeng, X. et al. A metabolic atlas of blood cells in young and aged mice identifies uridine as a metabolite to rejuvenate aged hematopoietic stem cells. Nat. Aging 4, 1477–1492 (2024).

    Article  CAS  PubMed  Google Scholar 

  42. Ermolaeva, M., Neri, F., Ori, A. & Rudolph, K. L. Cellular and epigenetic drivers of stem cell ageing. Nat. Rev. Mol. Cell Biol. 19, 594–610 (2018).

    Article  CAS  PubMed  Google Scholar 

  43. Cheung, P. et al. Single-cell chromatin modification profiling reveals increased epigenetic variations with aging. Cell 173, 1385–1397 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Adelman, E. R. et al. Aging human hematopoietic stem cells manifest profound epigenetic reprogramming of enhancers that may predispose to leukemia. Cancer Discov. 9, 1080–1101 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  45. Sen, P. et al. H3K36 methylation promotes longevity by enhancing transcriptional fidelity. Genes Dev. 29, 1362–1376 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Manlove, L. & Farrar, M. The role of NCOR co-repressors in lymphocyte development and function (P1471). J. Immunol. 190, 174.18 (2013).

    Article  Google Scholar 

  47. Fazi, F. et al. A minicircuitry comprised of microRNA-223 and transcription factors NFI-A and C/EBPα regulates human granulopoiesis. Cell 123, 819–831 (2005).

    Article  CAS  PubMed  Google Scholar 

  48. Jia, Y. et al. Hyperactive PI3Kδ predisposes naive T cells to activation via aerobic glycolysis programs. Cell Mol. Immunol. 18, 1783–1797 (2021).

    Article  CAS  PubMed  Google Scholar 

  49. Harder, K. W. et al. Perturbed myelo/erythropoiesis in Lyn-deficient mice is similar to that in mice lacking the inhibitory phosphatases SHP-1 and SHIP-1. Blood 104, 3901–3910 (2004).

    Article  CAS  PubMed  Google Scholar 

  50. Glatigny, S., Wagner, C. A. & Bettelli, E. Cutting edge: integrin α4 is required for regulatory B cell control of experimental autoimmune encephalomyelitis. J. Immunol. 196, 3542–3546 (2016).

    Article  CAS  PubMed  Google Scholar 

  51. Salehin, N., Santucci, N., Osteil, P. & Tam, P. P. L. Exploring chromatin accessibility in mouse epiblast stem cells with ATAC-Seq. Methods Mol. Biol. 2490, 93–100 (2022).

    Article  CAS  PubMed  Google Scholar 

  52. Summers, A. R. et al. HDAC3 is essential for DNA replication in hematopoietic progenitor cells. J. Clin. Invest. 123, 3112–3123 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Nguyen, H. C. B., Adlanmerini, M., Hauck, A. K. & Lazar, M. A. Dichotomous engagement of HDAC3 activity governs inflammatory responses. Nature 584, 286–290 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Stengel, K. R. et al. Histone deacetylase 3 controls a transcriptional network required for B cell maturation. Nucleic Acids Res. 47, 10612–10627 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Philips, R. L. et al. HDAC3 restrains CD8-lineage genes to maintain a bi-potential state in CD4+CD8+ thymocytes for CD4-lineage commitment. eLife 8, e43821 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  56. Yi, L. et al. Boosting to amplify signal with isobaric labeling (BASIL) strategy for comprehensive quantitative phosphoproteomic characterization of small populations of cells. Anal. Chem. 91, 5794–5801 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Hahne, H. et al. DMSO enhances electrospray response, boosting sensitivity of proteomic experiments. Nat. Methods 10, 989–991 (2013).

    Article  CAS  PubMed  Google Scholar 

  58. Akbar, A. N. & Gilroy, D. W. Aging immunity may exacerbate COVID-19. Science 369, 256–257 (2020).

    Article  CAS  PubMed  Google Scholar 

  59. Sonnett, M., Yeung, E. & Wühr, M. Accurate, sensitive, and precise multiplexed proteomics using the complement reporter ion cluster. Anal. Chem. 90, 5032–5039 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Zhang, H. et al. Genetic rescue of lineage-balanced blood cell production reveals a crucial role for STAT3 antiinflammatory activity in hematopoiesis. Proc. Natl Acad. Sci. USA 115, E2311–E2319 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank M. Wühr at Princeton University for generously sharing the TMTc+ algorithm. This work was supported by the National Natural Science Foundation of China (82025002 and 32230036 to H.H.; 82394411 and 82170113 to H. Zhang; 81570060, 82073221 and 31870826 to L.D.; 32201216 to Y.G.), National Key R&D Program of China (2022YFA1303200 to L.D.; 2019YFA0110203 to H. Zhang; 2019YFA0110201 to H.H.), National Clinical Research Center for Geriatrics at West China Hospital (Z2024JC002 to L.D.) and West China Hospital 135 project (ZYYC23013 to L.D.; ZYGD22005 to H.H.).

Author information

Authors and Affiliations

Authors

Contributions

L.D., H. Zhang and H.H. designed the entire project and wrote the paper. Y.G. and Y.L. performed the MS analysis. H. Zhan, N.W., M.L., Y.X., X.L. and Xiaodong Wang performed the FACS sorting, immunofluorescence staining, confocal microscopy, flow cytometry, immunoblotting, in vitro differentiation, HSC transplantation and CUT&Tag and ATAC-seq experiments. P.G., Y.G. and Y.D. analyzed the sequencing, proteome and acetylome data. Q.P. performed the metabolic analysis of HSCs. Y.G. and S.H. prepared the site-specific polyclonal antibodies. S.H., M.H., W.L. and K.Z. manually checked the MS2 spectra. X.J., Xiuxuan Wang and Z.X. synthesized the chemicals for this study. R.Z., L.G., Xinyuan Wang and Y.L. provided experimental help. P.W., Y.P., P.L., H.X., Y.S. and J.Q. provided helpful suggestions for this project.

Corresponding authors

Correspondence to Hongbo Hu, Huiyuan Zhang or Lunzhi Dai.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Chemical Biology thanks Hiroaki Honda, Feng-Chun Yang, Hui Ye and Balyn Zaro 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 Optimization and evaluation of the Iseq-Kac strategy.

a, Line chart showing the number of total acetylated peptides identified in 50 μg of protein from HCT116 cells labeled with different amounts of Ac-NHS. b, Fan chart depicting the number of mono- and multi-acetylated peptides identified in HCT116 samples labeled with different amounts of Ac-NHS. c, Fan chart showing the number of doubly and other charged peptides generated under different LC-MS/MS conditions. d, Fan chart showing the number of mono- and multi-acetylated doubly charged peptides under varied LC-MS/MS conditions. The C8 condition was used as a standard condition for Iseq-Kac. e, Fan chart showing the number of identified peptides at different mixing ratios, with TMTc cutoff ranging from 0.01 to 0.1. f, False discovery rate (FDR) of acetylated peptide identification at different TMTc cutoffs using a 2:1:1 pre-mixed sample. TMTc ion clusters and MS2 fragments were manually examined. g, Signal-to-noise (S/N) in the low-mass region for 169 identified acetylated peptides, calculated by Proteome Discoverer 2.3. h, Quantification of 2:1:1 pre-mixed digested peptides from HCT116 cells using reporter ions or TMTc. i,j, Quantification of HCT116 peptides pre-mixed at different ratios using reporter ions (i) or TMTc (j). k-n, Peptides from trichostatin A (TSA)-treated HCT116 cells labeled with TMT129 and TMT131 were analyzed by Iseq-Kac. The internal standard (IS) was derived from either different batches of TSA-treated HCT116 cells (k,l), or untreated HCT116 cells and colorectal cancer tissue (m,n). Representative Data from n = 2 (k,m) or n = 3 (g-j) independent experiments. Data are presented as the mean ± s.e.m.

Source data

Extended Data Fig. 2 Evaluating Iseq-Kac in low-input samples.

a-h, Peptides from trichostatin A (TSA)-treated HCT116 cells labeled with TMT129 and TMT131 were analyzed by Iseq-Kac. a,b, Venn diagrams showing the overlap of identified (a) and quantified (b) acetylated peptides between two replicates. c, Two-sided Pearson correlation analysis of acetylated peptide ratios (TSA/NC) between replicates. d, Comparison of quantitative results for histone acetylated peptides from Iseq-Kac, IP-MS, and immunoblotting. The immunoblotting data were quantified with ImageJ. e, Histone acetylation sites detected by immunoblotting. Representative western blots from n = 2 or 3 independent experiments. f, Bar graph showing the average number of identified (blue) and quantified (red) acetylated peptides from 100 to 10,000 (per channel) TSA-treated or untreated HCT116 cells. Data are presented as the mean ± s.e.m. of n = 3 independently performed experiments. g, Venn plots showing the overlap of quantified acetylated peptides across three replicates in experiments with 100 to 10,000 cells per channel. h, CV of overlapping acetylated peptides across three replicates using varying numbers of cells. i, Chord diagram showing the classes of proteins identified in different numbers of HCT116 cells. j, Bubble diagram comparing the quantitative results of histone acetylated peptides in different numbers of HCT116 cells. NBT-2015 represents data from Scholz et al.’s work, where acetylome data were obtained using an enrichment-based strategy with 20 mg of protein input.

Source data

Extended Data Fig. 3 Evaluation of proteome and acetylome data of HSCs and MPPs.

a, Gating strategy for FACS sorting or analysis of Lin- (CD3e-Ter119-B220-Gr1-CD11b-), HSC (Lin-CD127-Sca1+c-Kit+CD150+CD48-CD34-), CD150hi HSC, CD150lo HSC, MPP1 (Lin-CD127-Sca1+c-Kit+CD150+CD48+), MPP2 (Lin-CD127-Sca1+c-Kit+CD150-CD48+CD135-) and MPP3 (Lin-CD127-Sca1+c-Kit+CD150-CD48+CD135+), GMP (Lin-CD127-Sca1-c-Kit+CD34+FcγRIII/II+), CMP (Lin-CD127-Sca1-c-Kit+CD34+FcγRIII/II-), CLP (Lin-CD127+) and MEP (Lin-CD127-Sca1-c-Kit+CD34-FcγRIII/II-). b-c, Representative data of one of twelve to eighteen independent experiments, for young (b) and aged (c) HSCs and MPPs. d, Flowchart of generating cell-specific Kac libraries for HSCs and MPPs. e, Venn diagrams depicting the overlap of quantified proteins across four replicates in HSCs or MPPs. f, Correlation of quantified proteins among four replicates in HSCs or MPPs. g, Venn diagrams showing the overlap of quantified acetylated peptides among four replicates in HSCs or MPPs. h, Correlation of quantified acetylated peptides among four replicates in HSCs or MPPs.

Extended Data Fig. 4 Proteomes of young and aged HSCs and MPPs.

a, Kinases and phosphatases altered in HSCs or MPPs with age (fold change < 0.85 or > 1.176), ranked by average protein intensity. Red, upregulated in aged cells; blue, downregulated. b, Proteins involved in BCAA, TCA cycle and fatty acid metabolism pathways significantly changed in aged HSCs or MPPs. Red, upregulated in aged cells; blue, downregulated; undetectable proteins labeled as “NA”. c, Relative mRNA expression levels of Foxp1 (top) and Ikzf1 (bottom) in young (blue) and aged (orange) HSCs and MPPs, as determined by qPCR. The cumulative mean values are presented as the fold change normalized to young (n = 3-6 wells, representative data for three independent biological replicates). Data are presented as the mean ± s.e.m. Statistical significance was assessed using two-side t-test (c).

Source data

Extended Data Fig. 5 Acetylomes of young and aged HSCs and MPPs.

a, Chord diagram showing the classes of acetylated proteins identified in HSCs and MPPs. b-e, Proteins were categorized into five intervals (R1 to R5) based on average protein abundance ranking, and the proportion of acetylated proteins in each interval was calculated and compared. b, Comparison of protein classes in lowest-abundance proteins intervals (R5), based on our proteome data and Zaro et al.’s proteome data. c, Distribution of acetylated peptides identified by Iseq-Kac across five intervals (R1 to R5), based on protein intensity ranking from our study and Zaro et al.’s study. d,e, Distribution of acetylated peptides identified by classic affinity enrichment-based method across five intervals based on protein intensity ranking. Proteome and acetylome data were obtained from Krug et al.’s study (d) and Yongchao et al.’s study (e). f, Dot blotting assays to confirm the specificities of ALDH2-K430ac and ZNF207-K398ac antibodies using peptides containing acetyllysine (Modified) or unmodified lysine (Unmodified). Each dot contained 10 ng, 100 ng, and 1 µg of peptides, respectively. Representative blots from n = 2 independent experiments.

Source data

Extended Data Fig. 6 Age- and metabolism-related acetylome of hematopoietic progenitors.

a, Global ∆As analysis of acetylated proteins in HSCs and MPPs. The acetylated change (∆As) for each protein was calculated as the sum of log2(FC(A/Y)) values of all acetylated peptides of that protein. Dotted lines represent ∆As = ±1. b, Hyper- or hypo-acetylated proteins in aged HSCs or MPPs, with pathway enrichment analysis performed using Metascape (one-sided hypergeometric test). Bubble color indicates the corresponding ∆As values. c, Changes in glycolysis and TCA cycle-related metabolites in HSCs from young and aged mice, determined by mass spectrometry (MS) analysis. Data are presented as the median ± 25% of n = 12 independently performed experiments. Statistical significance was assessed using two-sided two-side t-test. d, Overrepresented NADP metabolic pathways and cellular stress responses with hyper- or hypo-acetylated proteins in aged HSCs or MPPs. Colors represent the ∆As values. Red, hyperacetylated in aged cells; blue, hypoacetylated. NA, acetylation is not detected.

Source data

Extended Data Fig. 7 The levels of H4K77ac in young and aged MPPs and LSKs.

a, MS2 spectrum of the peptide containing H4K77ac, with enlarged regions showing the low-mass reporter ion and TMTc ion clusters. b,c, Anti-H4K77ac antibody specificity verified by dot blotting (b) and immunoblotting in TSA-treated and untreated K562 cells (c). Representative blots from n = 2 independent experiments (b,c). d, Cumulative mean values showing the relative mean fluorescence intensities (MFI) of H4K77ac in HSCs and hematopoietic progenitor subsets, as measured by flow cytometry and normalized to young (n = 7-8 mice). e,f, Immunofluorescence of H4K77ac in FACS-sorted young and aged MPPs and LSKs labeled with AF647-conjugated anti-rabbit IgG antibodies. Representative confocal microscopy images of young and aged MPPs and LSKs expressing H4K77ac (red) (e, scale bar, 2μm), with fluorescence quantification of H4K77ac (f) by ImageJ (n = 106-185 cells). The fluorescence intensities were calculated per square pixel and normalized to DAPI. g, Cumulative mean value showing the MFI of H4K77ac in HSPC subpopulations as measured by flow cytometry and normalized to young (n = 6 mice). Data are presented as the mean ± s.e.m. All data are derived from three (d,f) or four (g) independent biological replicates. Statistical significance was assessed via two-way ANOVA followed (d) or two-side t-test (f).

Source data

Extended Data Fig. 8 Identification of H4K77ac-related pathways and binding proteins.

a, Fraction of reads falling within peak regions (FRiP, SEACR FDR ≤ 0.05) of H4K77ac CUT&Tag data in young and aged HSCs (n = 20000 HSCs/experiment). b,c, Overlap between genes differentially expressed (downregulated/upregulated) in aged HSCs and genes showing differentially H4K77ac enrichment (TSS ± 3 kb) at their promoters (n = 20000 HSCs/experiment). b, Venn diagrams of overlapping genes. c, Pathway enrichment from Over-Representation Analysis (ORA) of overlapping genes (Fisher’s precision probability tests. p value < 0.01). Blue, downregulated in aged HSCs; Orange, upregulated in aged HSCs. d, Relative mRNA expression levels of signature genes in young (blue) and aged (orange) HSCs, as determined by transcriptome. e, Distribution of ATAC-seq reads (OCRs) at transcription start sites (TSS ± 3 kb) of genes annotated by common peaks (Fig. 4g) associated with lymphocyte differentiation (GO:0030217 for T cell differentiation, GO:0030183 for B cell differentiation) or myeloid cell development (GO:0030099 for myeloid cell differentiation) between young and aged HSCs promoters (n = 20000 HSCs/experiment). f, Distribution of H4K77ac CUT&Tag reads at the transcription start site (TSS ± 3 kb) of genes associated with lymphocytes (GO:0030217 and GO:0030183 gene sets for T and B cell differentiation, respectively) or myeloid cell development (GO:0030099 gene sets for myeloid cell differentiation) in young (left) or aged HSCs (right) promoters (n = 20000 HSCs/experiment). g, Comparative analysis of proteins pulled down by acetylated H4K77 peptide or its unmodified form. h, Immunoblotting showing H4K77ac levels in HCT116 cells treated with HDAC3 inhibitor (RGFP966) or SIRT2 inhibitor (Thiomyristoyl) i, Representative data showing the frequencies of T lymphocytes (CD4-CD8-, CD4+CD8+, CD4+ and CD8+) derived from aged HSCs, determined at 2 weeks post-RGFP966 or DMSO treatment. j, ShRNA-Hdac3-mediated gene knockdown efficiency and H4K77ac levels in K562 cells, assessed by immunoblotting. Data are presented as the median ± 25% (a,e,f) or mean ± s.e.m. (d). All data are derived from three independently biological replicates (a,d,e,f). Representative blots from three independent experiments (h,j). Statistical significance was assessed using the two-side Wilcoxon rank sum test (d) or two-side Kruskal-Wallis rank sum test (e,f).

Source data

Extended Data Fig. 9 H4K77ac specifically regulated by the deacetylase HDAC3.

a-j, In vitro differentiation assay of aged HSCs infected with shRNA-Hdac3 (shRNA#1) or vector control. Representative data (a,c,f,h) and cumulative mean values (b,d,e,g,i,j) of T lymphocyte (CD4-CD8-, CD4+CD8+), B lymphocyte (CD19+), myeloid cell (CD11b+) frequencies and H4K77ac abundance in CD45+mCherry+ cells derived from shRNA-Hdac3-infected HSCs or vector control infected HSCs. The cumulative mean values are presented as the fold change normalized to control (n = 14-20 wells). k-m, Representative data (k,m) and cumulative mean values (l,n) of H4K77ac, H3K27ac and H3K9/K14ac in RGFP966-treated (1/5/10 µM) or DMSO-treated HSCs (n = 3 mice). o, Pre-ranked GSEA of genes enriched by H4K77ac in RGFP966-treated versus DMSO-treated HSCs from young mice. Differential peaks (p value < 0.01) were identified using DiffBind in R. p-r, Comparisons between genes co-regulated by H4K77ac and H3K27ac. p, Venn diagrams showing the numbers of overlapping genes. q, Volcano plot displaying the genes differentially expressed between aged and young HSCs based on the 304 overlapping genes. Dotted lines indicate Log2FC = ± log2(1.5), p value adjusted ≤ 0.05. Blue indicates downregulated genes in aged HSCs, orange indicates upregulated genes in aged HSCs, and gray indicates no significant difference. r, Bubble plot showing the pathway enrichment results of the 304 genes, based on GSEA. NES, normalized enrichment score. s,t, Representative immunoblotting showing the expression of H4 (s) or H3 (t) in Lin- bone marrow cells infected with H4K77 (s) or H3K27 (t) mutant virus or vector control. u,v, Cumulative mean value showing the MFI of GFP in GFP+CD45+ cells derived from HSCs ectopically expressing H4K77R/Q (u) or H3K27R/Q (v) (n = 5-8 wells for H4K77 mutations; n = 4-6 wells for H3K27 mutations). All data are derived from three independently biological replicates (a-n and u-v). Representative blots from n = 2 or 3 independent experiments (s,t). Data are presented as the mean ± s.e.m. Statistical significance was assessed using two-side t-test (b,d,e,g,i,j), two-way ANOVA followed by Tukey’s multiple comparisons adjustments (l,n), two-side Wilcoxon Rank Sum Tests (q) or two-side Permutation test (o,r).

Source data

Supplementary information

Supplementary Information

Supplementary Figs. 1–3 and legends for Supplementary Tables 1–6.

Reporting Summary

Supplementary Table 1

Optimization and evaluation of the Iseq-Kac strategy.

Supplementary Table 2

Acetylated peptides or proteins identified and quantified in the aging-associated proteome study of HSC and MPPs.

Supplementary Table 3

The parameters used in MRM for targeted validation of acetylated peptides.

Supplementary Table 4

List of H4K77ac-enriched genes in young and aged HSCs, identified by CUT&Tag assay.

Supplementary Table 5

Acetylated peptides quantified in RGFP966-treated and untreated HSCs.

Supplementary Table 6

Genes enriched with H4K77ac peaks in RGFP966-treated and DMSO-treated HSCs, identified by CUT&Tag assay.

Supplementary Data 1

The expression of H4 in young and aged MPPs and LSKs.

Supplementary Data 2

H4K77ac but not H3K27ac regulates differentiation potential of HSCs.

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

Unprocessed blots.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Unprocessed blots.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Unprocessed blots.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 8

Unprocessed blots.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 9

Unprocessed blots.

Source Data Extended Data Fig. 9

Statistical 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

Gong, Y., Zhan, H., Wei, N. et al. Acetylation profiling by Iseq-Kac reveals insights into HSC aging and lineage decision. Nat Chem Biol (2025). https://doi.org/10.1038/s41589-025-01916-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41589-025-01916-1

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