Abstract
Aging of hematopoietic stem cells (HSCs) is accompanied by impaired self-renewal ability, myeloid skewing, immunodeficiencies and increased susceptibility to malignancies. Although previous studies highlighted the pivotal roles of individual metabolites in hematopoiesis, comprehensive and high-resolution metabolomic profiles of different hematopoietic cells across ages are still lacking. In this study, we created a metabolome atlas of different blood cells across ages in mice. We reveal here that purine, pyrimidine and retinol metabolism are enriched in young hematopoietic stem and progenitor cells (HSPCs), whereas glutamate and sphingolipid metabolism are concentrated in aged HSPCs. Through metabolic screening, we identified uridine as a potential regulator to rejuvenate aged HSPCs. Mechanistically, uridine treatment upregulates the FoxO signaling pathway and enhances self-renewal while suppressing inflammation in aged HSCs. Finally, we constructed an open-source platform for public easy access and metabolomic analysis in blood cells. Collectively, we provide a resource for metabolic studies in hematopoiesis that can contribute to future anti-aging metabolite screening.
This is a preview of subscription content, access via your institution
Access options
Access Nature and 54 other Nature Portfolio journals
Get Nature+, our best-value online-access subscription
$29.99 / 30 days
cancel any time
Subscribe to this journal
Receive 12 digital issues and online access to articles
$119.00 per year
only $9.92 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to full article PDF
Prices may be subject to local taxes which are calculated during checkout
Similar content being viewed by others
Data availability
The data used in this study have been placed into Figshare (https://doi.org/10.6084/m9.figshare.22708162.v1) and the Gene Expression Omnibus (GSE240731). Public online databases (KEGG (https://www.kegg.jp/) and HMDB (https://hmdb.ca/)) were applied to match metabolites based on the exact mass data (m/z). All data supporting the findings of this study are available within the article or from the corresponding authors upon reasonable request.
Code availability
The code used in this study has been placed into Figshare (https://doi.org/10.6084/m9.figshare.22708162.v1).
References
Orkin, S. H. & Zon, L. I. Hematopoiesis: an evolving paradigm for stem cell biology. Cell 132, 631–644 (2008).
Laurenti, E. & Göttgens, B. From haematopoietic stem cells to complex differentiation landscapes. Nature 553, 418–426 (2018).
Li, X. et al. Mechanisms and rejuvenation strategies for aged hematopoietic stem cells. J. Hematol. Oncol. 13, 31 (2020).
Mittelbrunn, M. & Kroemer, G. Hallmarks of T cell aging. Nat. Immunol. 22, 687–698 (2021).
de Graaf, C. A. et al. Haemopedia: an expression atlas of murine hematopoietic cells. Stem Cell Rep. 7, 571–582 (2016).
Choi, J. et al. Haemopedia RNA-seq: a database of gene expression during haematopoiesis in mice and humans. Nucleic Acids Res. 47, D780–D785 (2019).
Wendorff, A. A. et al. Epigenetic reversal of hematopoietic stem cell aging in Phf6-knockout mice. Nat. Aging 2, 1008–1023 (2022).
Wilkinson, A. C., Morita, M., Nakauchi, H. & Yamazaki, S. Branched-chain amino acid depletion conditions bone marrow for hematopoietic stem cell transplantation avoiding amino acid imbalance-associated toxicity. Exp. Hematol. 63, 12–16 (2018).
Li, C. et al. Loss of sphingosine kinase 2 promotes the expansion of hematopoietic stem cells by improving their metabolic fitness. Blood 140, 1686–1701 (2022).
Guerrero, A. et al. 3-deazaadenosine (3DA) alleviates senescence to promote cellular fitness and cell therapy efficiency in mice. Nat. Aging 2, 851–866 (2022).
Gonzalez-Menendez, P. et al. Arginine metabolism regulates human erythroid differentiation through hypusination of eIF5A. Blood 141, 2520–2536 (2023).
Rattigan, K. M., Zarou, M. M. & Helgason, G. V. Metabolism in stem cell-driven leukemia: parallels between hematopoiesis and immunity. Blood 141, 2553–2565 (2023).
Takubo, K. et al. Regulation of the HIF-1α level is essential for hematopoietic stem cells. Cell Stem Cell 7, 391–402 (2010).
Takubo, K. et al. Regulation of glycolysis by Pdk functions as a metabolic checkpoint for cell cycle quiescence in hematopoietic stem cells. Cell Stem Cell 12, 49–61 (2013).
Simsek, T. et al. The distinct metabolic profile of hematopoietic stem cells reflects their location in a hypoxic niche. Cell Stem Cell 7, 380–390 (2010).
DeVilbiss, A. W. et al. Metabolomic profiling of rare cell populations isolated by flow cytometry from tissues. eLife 10, e61980 (2021).
Agathocleous, M. et al. Ascorbate regulates haematopoietic stem cell function and leukaemogenesis. Nature 549, 476–481 (2017).
Taya, Y. et al. Depleting dietary valine permits nonmyeloablative mouse hematopoietic stem cell transplantation. Science 354, 1152–1155 (2016).
Lim, S. A., Su, W., Chapman, N. M. & Chi, H. Lipid metabolism in T cell signaling and function. Nat. Chem. Biol. 18, 470–481 (2022).
Corrado, M. et al. Dynamic cardiolipin synthesis is required for CD8+ T cell immunity. Cell Metab. 32, 981–995 (2020).
Alving, A. S., Carson, P. E., Flanagan, C. L. & Ickes, C. E. Enzymatic deficiency in primaquine-sensitive erythrocytes. Science 124, 484–485 (1956).
Vogel, M. et al. HPRT and purine salvaging are critical for hematopoietic stem cell function. Stem Cells 37, 1606–1614 (2019).
Amorim, J. A. et al. Mitochondrial and metabolic dysfunction in ageing and age-related diseases. Nat. Rev. Endocrinol. 18, 243–258 (2022).
Ding, C. et al. A Treg-specific long noncoding RNA maintains immune-metabolic homeostasis in aging liver. Nat. Aging 3, 813–828 (2023).
Fumagalli, M., Lecca, D., Abbracchio, M. P. & Ceruti, S. Pathophysiological role of purines and pyrimidines in neurodevelopment: unveiling new pharmacological approaches to congenital brain diseases. Front. Pharmacol. 8, 941 (2017).
Liu, Z. et al. Cross-species metabolomic analysis identifies uridine as a potent regeneration promoting factor. Cell Discov. 8, 6 (2022).
Zeng, X. et al. Integrated single-cell bioinformatics analysis reveals intrinsic and extrinsic biological characteristics of hematopoietic stem cell aging. Front. Genet. 12, 745786 (2021).
Wu, L. et al. LepR+ niche cell-derived AREG compromises hematopoietic stem cell maintenance under conditions of DNA repair deficiency and aging. Blood 142, 1529–1542 (2023).
Colom Díaz, P. A., Mistry, J. J. & Trowbridge, J. J. Hematopoietic stem cell aging and leukemia transformation. Blood 142, 533–542 (2023).
Cabezas-Wallscheid, N. et al. Vitamin A-retinoic acid signaling regulates hematopoietic stem cell dormancy. Cell 169, 807–823 (2017).
Chang, P. et al. Seizure control by decanoic acid through direct AMPA receptor inhibition. Brain 139, 431–443 (2016).
Yamamoto, S., Takehara, M. & Ushimaru, M. Inhibitory action of linoleamide and oleamide toward sarco/endoplasmic reticulum Ca2+-ATPase. Biochim. Biophys. Acta Gen. Subj. 1861, 3399–3405 (2017).
Tothova, Z. et al. FoxOs are critical mediators of hematopoietic stem cell resistance to physiologic oxidative stress. Cell 128, 325–339 (2007).
Tothova, Z. & Gilliland, D. G. FoxO transcription factors and stem cell homeostasis: insights from the hematopoietic system. Cell Stem Cell 1, 140–152 (2007).
Caiado, F., Pietras, E. M. & Manz, M. G. Inflammation as a regulator of hematopoietic stem cell function in disease, aging, and clonal selection. J. Exp. Med. 218, e20201541 (2021).
Zeng, X. et al. Fecal microbiota transplantation from young mice rejuvenates aged hematopoietic stem cells by suppressing inflammation. Blood 141, 1691–1707 (2023).
Nakamura-Ishizu, A., Ito, K. & Suda, T. Hematopoietic stem cell metabolism during development and aging. Dev. Cell 54, 239–255 (2020).
Wang, Y. H. et al. Cell-state-specific metabolic dependency in hematopoiesis and leukemogenesis. Cell 158, 1309–1323 (2014).
Menéndez-Gutiérrez, M. P. et al. Retinoid X receptor promotes hematopoietic stem cell fitness and quiescence and preserves hematopoietic homeostasis. Blood 141, 592–608 (2023).
Field, M. S., Lan, X., Stover, D. M. & Stover, P. J. Dietary uridine decreases tumorigenesis in the ApcMin/+ model of intestinal cancer. Curr. Dev. Nutr. 2, nzy013 (2018).
Nakada, D. et al. Oestrogen increases haematopoietic stem-cell self-renewal in females and during pregnancy. Nature 505, 555–558 (2014).
Shahidi, N. T. Androgens and erythropoiesis. N. Engl. J. Med. 289, 72–80 (1973).
Pellin, D. et al. A comprehensive single cell transcriptional landscape of human hematopoietic progenitors. Nat. Commun. 10, 2395 (2019).
Watcham, S., Kucinski, I. & Gottgens, B. New insights into hematopoietic differentiation landscapes from single-cell RNA sequencing. Blood 133, 1415–1426 (2019).
Olsson, A. et al. Single-cell analysis of mixed-lineage states leading to a binary cell fate choice. Nature 537, 698–702 (2016).
Byun, S. et al. Postprandial FGF19-induced phosphorylation by Src is critical for FXR function in bile acid homeostasis. Nat. Commun. 9, 2590 (2018).
Dunn, W. B. et al. Procedures for large-scale metabolic profiling of serum and plasma using gas chromatography and liquid chromatography coupled to mass spectrometry. Nat. Protoc. 6, 1060–1083 (2011).
Dyar, K. A. et al. Atlas of circadian metabolism reveals system-wide coordination and communication between clocks. Cell 174, 1571–1585 (2018).
R Core Team. R: a lan guage and environment for statistical computing. http://www.R-project.org/ (2013).
Krijthe, J. H. Rtsne: t-distributed stochastic neighbor embedding using Barnes–Hut implementation. https://github.com/jkrijthe/Rtsne (2015).
Pang, Z., Chong, J., Li, S. & Xia, J. MetaboAnalystR 3.0: toward an optimized workflow for global metabolomics. Metabolites 10, 186 (2020).
Livak, K. J. & Schmittgen, T. D. Analysis of relative gene expression data using real-time quantitative PCR and the \(2^{-\Delta\Delta{C}_{\mathrm{T}}}\) method. Methods 25, 402–408 (2001).
Hu, Y. & Smyth, G. K. ELDA: extreme limiting dilution analysis for comparing depleted and enriched populations in stem cell and other assays. J. Immunol. Methods 347, 70–78 (2009).
Kim, D., Paggi, J. M., Park, C., Bennett, C. & Salzberg, S. L. Graph-based genome alignment and genotyping with HISAT2 and HISAT-genotype. Nat. Biotechnol. 37, 907–915 (2019).
Wu, T. et al. clusterProfiler 4.0: a universal enrichment tool for interpreting omics data. Innovation (Camb.) 2, 100141 (2021).
Acknowledgements
This work was supported by grants from the National Key Research and Development Program of China (2022YFA1103500 to P.Q.); the National Natural Science Foundation of China (82130003 to H.H., 82270221 to J.Y., 82200255 to M.Z., 82222003 to P.Q., 92268117 to P.Q. and 82161138028 to P.Q.); the Key R&D Project of the Zhejiang Provincial Science and Technology Department (2021C03010 to H.H.); the Key R&D Program of Zhejiang Province (2024SSYS0023 to H.H. and 2024SSYS0024 to P.Q.); the Research Center of Prevention and Treatment of Senescence Syndrome, Zhejiang University School of Medicine (2022020002 to P.Q.); the Zhejiang Provincial Natural Science Foundation of China (Z24H080001 to P.Q. and LQ22H080005 to M.Z.); the Department of Science and Technology of Zhejiang Province (2023R01012 to P.Q.); and the Fundamental Research Funds for the Central Universities (226-2024-00007 to P.Q.). We are grateful for the technical assistance provided by B. Xu from Liangzhu Laboratory, Zhejiang University, and for the support and help from members of the core facility platform of Zhejiang University School of Medicine and the Laboratory Animal Center of Zhejiang University. The authors acknowledge BioRender (https://www.biorender.com/), as flow diagrams in this study were created with the BioRender platform.
Author information
Authors and Affiliations
Contributions
H.H., P.Q. and X.Z. conceived the study. X.Z., C.S., Y.H., K.H., Xiaoqing Li and C.W. curated the data. X.Z., C.S., Y.H., K.H., Xiaoqing Li and C.W. analyzed the data. H.H. and P.Q. determined the methodology. H.H., Q.P. and J.Y. acquired funding. X.Z. and C.S. wrote the original draft. C.S., C.W., L.D., J.C., S.H., K.H., Y.X., M.Z., W.S., Q.L., J.Y., Y.H., Z.Z., Xia Li, P.Q. and H.H. reviewed and edited the manuscript.
Corresponding authors
Ethics declarations
Competing interests
The authors declare no competing interests.
Peer review
Peer review information
Nature Aging thanks Toshio Suda and the other, anonymous, reviewer(s) for their contributions 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 Metabolome of different blood cell types during aging.
(a–d) Flow cytometry gating strategy. (a) cKit+ cells enriched by immunomagnetic sorting were used for further FACS. After being discriminated from debris by gating on forward scatter (FSC) and side scatter (SSC), LSK cells, CLPs, CMPs, GMPs and MEPs were collected from BMs. (b) Megakaryocytes, macrophages, dendritic cells, monocytes and granulocytes were collected from BM cells. (c) CD19− cells enriched by immunomagnetic sorting in spleen were used for further FACS. CD4 T cells, CD8 T cells and NK cells were collected by FACS. (d) Erythrocytes were collected from PB cells. (e) Bar graphs showing the count of metabolites detected in each cell type in aged mice. (f) Violin plot showing the relative abundance of detected metabolites in different cell types in aged mice. (g) Pie chart showing the superclass of the detected metabolites in aged mice. (h) T-SNE plot showing the clustering result of different cell types in aged mice.
Extended Data Fig. 2 Quantitative analysis of glycolysis-related metabolites in different cell types.
Graphs show mean ± SEM, with statistical significance determined by one-way ANOVA followed by Dunnett’s multiple comparison test (n = 4 per group, 6–8 weeks, C57BL/6J female mice).
Extended Data Fig. 3 Identification of cell-specific metabolic signatures.
(a) Wind-rose plots showing the count of cell-specific metabolic signatures in different cell types. (b-o) Representative KEGG analysis results of cell-specific metabolic signatures in CLPs, CMPs, GMPs, MEPs, B cells, CD4 T cells, CD8 T cells, NK cells, megakaryocytes, macrophages, dendritic cells, monocytes, granulocytes and erythrocytes.
Extended Data Fig. 4 Enrichment analysis of aging-related metabolites.
(a–ab) Representative KEGG analysis results of differential metabolites during aging in CLPs (a,b), CMPs (c,d), B cells (e,j), GMPs (f,g), macrophages (h,i), dendritic cells (k,l), CD4 T cells (m,n), erythrocytes (o,t), MEPs (p,q), CD8 T cells (r,s), granulocytes (u,v), megakaryocytes (w,x), monocytes (y,z) and NK cells (aa,ab).
Extended Data Fig. 5 Screening of metabolites rejuvenating aged HSPCs.
(a–c) Absolute number of LT-HSCs, ST-HSCs and MPPs at different uridine concentrations when acting on aged HSPCs (n = 3 samples). (d, e) Percentage and absolute number of LSK cells at different guanosine concentrations when acting on aged HSPCs (n = 3 samples). (f–h) Absolute number of LSK cells after different metabolite interventions when acting on aged HSPCs (n = 3 samples). (i) Number of total colonies measured for aged HSPCs treated with guanosine in methylcellulose (n = 3 samples). (j) Number of total colonies measured for uncultured young and aged HSPCs in methylcellulose (n = 4 samples). (k, l) The percentage of Dcfh-da positive aged HSPCs (n = 3 samples). (m) Line graph of donor-derived CD45.2+ reconstitution in CD45.1+ mice receiving 1000 aged LSK cells post transplantation (vehicle group, 2 male and 2 female, 6–8 weeks C57BL/6 J; guanosine group, 1 male and 2 female, 6–8 weeks C57BL/6 J). (n, o) Absolute number of HSPCs and LT-HSCs at different concentrations of uridine when acting on young HSPCs (n = 3 samples). (p) In the in vitro uridine (200 μM) administration experiment, line graph of donor-derived CD45.2+ reconstitution in CD45.1+ mice receiving 1000 young HSPCs post transplantation (n = 6 mice per group, 3 male and 3 female, 6–8 weeks C57BL/6J). a, b, c, d, e, f, g, i, n, o, Graphs show mean ± SEM, with statistical significance determined by one-way ANOVA followed by Dunnett’s multiple comparison test. h, j, k, l, m, p, Graphs show mean ± SEM, with statistical significance determined by two-tail unpaired student’s t-test.
Extended Data Fig. 6 Uridine improved cellular function of aged HSCs in vivo.
(a) Absolute number of HSPCs in aged mice administered different concentrations of uridine orally (n = 6 mice in 0 and 0.25 mg/ml group, female, 18–20 months, C57BL/6 J; n = 5 mice in 0.05, 2.5, and 10 mg/ml group, female, 18–20 months, C57BL/6 J). (b) Number of total colonies measured for HSPCs from aged mice administered different concentrations of uridine orally (n = 6 mice in 0 and 0.25 mg/ml group, female, 18–20 months, C57BL/6 J; n = 5 mice in 0.05, 2.5, and 10 mg/ml group, female, 18–20 months, C57BL/6 J). (c–e) Concentration of creatinine, aspartate aminotransferase, and alanine aminotransferase in the serum from aged mice administered different concentrations of uridine orally (n = 5 mice per group, female, 18–20 months, C57BL/6 J). (f–k) Absolute number of LT-HSCs, ST-HSCs, MPPs, CMPs, GMPs, and MEPs in aged mice orally administered uridine (0.25 mg/ml) (n = 6 mice per group, female, 18–20 months, C57BL/6 J). a–e, Graphs show mean ± SEM, with statistical significance determined by one-way ANOVA followed by Dunnett’s multiple comparison test. f–k, Graphs show mean ± SEM, with statistical significance determined by two-tail unpaired student’s t-test.
Extended Data Fig. 7 Brief introduction of the open-source platform for analyzing metabolites across different blood cells and during aging.
The functions of the website including browsing experimental design and methods, performing dimensionality reduction analysis, retrieving the metabolic markers of specific cells in hematopoietic system, assessing the correlations between metabolites, retrieving the distribution of specific metabolites among different cell types, obtaining the differential metabolites in different cell types during aging and downloading the experimental data.
Supplementary information
Supplementary Information
Supplementary Figs. 1 and 2.
Supplementary Data 1
Blood cell-type-specific metabolic signatures.
Supplementary Data 2
Aging-related metabolites in all blood cell types.
Supplementary Data 3
Differentially expressed genes in RNA sequencing.
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 Fig. 7
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.
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.
About this article
Cite this article
Zeng, X., Shi, C., Han, Y. 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 (2024). https://doi.org/10.1038/s43587-024-00669-1
Received:
Accepted:
Published:
DOI: https://doi.org/10.1038/s43587-024-00669-1