Abstract
The aging of hematopoietic stem cells (HSCs) substantially alters their characteristics. Mitochondria, essential for cellular metabolism, play a crucial role, and their dysfunction is a hallmark of aging-induced changes. The impact of mitochondrial mass on aged HSCs remains incompletely understood. Here we demonstrate that HSCs with high mitochondrial mass during aging are not merely cells that have accumulated damaged mitochondria and become exhausted. In addition, these HSCs retain a high regenerative capacity and remain in the aging bone marrow. Furthermore, we identified GPR183 as a distinct marker characterizing aged HSCs through single-cell analysis. HSCs marked by GPR183 were also enriched in aged HSCs with high mitochondrial mass, possessing a high capacity of self-renewal. These insights deepen understanding of HSC aging and provide additional perspectives on the assessment of aged HSCs, underscoring the importance of mitochondrial dynamics in aging.
This is a preview of subscription content, access via your institution
Access options
Access through your institution
Change institution
Buy or subscribe
Access Nature and 54 other Nature Portfolio journals
Get Nature+, our best-value online-access subscription
$29.99 / 30 days
cancel any time
Learn more
Subscribe to this journal
Receive 12 digital issues and online access to articles
$119.00 per year
only $9.92 per issue
Learn more
Buy this article
Purchase on SpringerLink
Instant access to full article PDF
Buy now
Prices may be subject to local taxes which are calculated during checkout
Additional access options:
Log in
Learn about institutional subscriptions
Read our FAQs
Contact customer support
Fig. 1: Aged HSCs with a high mitochondrial mass retain robust stemness characteristics.
Fig. 2: Aged mito-Dendra2 High HSCs demonstrate a higher self-renewal capacity and a lower tendency toward differentiation than aged mito-Dendra2 Low HSCs.
Fig. 3: Single-cell multiome analysis revealed that aged mito-Dendra2 High HSCs were characterized with HSC signature and OXPHOS genes.
Fig. 4: Aged mito-Dendra2 High HSCs were characterized with higher ATP production with no increase of cellular and mitochondrial ROS.
Fig. 5: Gpr183 was identified as a marker gene specific to a subset of aged mito-Dendra2 High HSCs.
Fig. 6: Gpr183 was identified as a marker gene specific to a subset of aged mito-Dendra2 High HSCs.
Fig. 7: GPR183high aged HSCs maintain high autophagy capacity and preserve their properties well during BMT and long-term ex vivo culture.
Data availability
The raw data for the multiome and bulk RNA-seq analyses in this study have been deposited in the Gene Expression Omnibus database under accession numbers GSE262784 and GSE262396. Any other data reported in this paper are available from the lead contact upon reasonable request.
References
de Haan, G. & Lazare, S. S. Aging of hematopoietic stem cells. Blood 131, 479–487 (2018).
ArticlePubMedGoogle Scholar
Mejia-Ramirez, E. & Florian, M. C. Understanding intrinsic hematopoietic stem cell aging. Haematologica 105, 22–37 (2020).
ArticleCASPubMedPubMed CentralGoogle Scholar
Dykstra, B., Olthof, S., Schreuder, J., Ritsema, M. & de Haan, G. Clonal analysis reveals multiple functional defects of aged murine hematopoietic stem cells. J. Exp. Med. 208, 2691–2703 (2011).
ArticleCASPubMedPubMed CentralGoogle Scholar
Cho, R. H., Sieburg, H. B. & Muller-Sieburg, C. E. A new mechanism for the aging of hematopoietic stem cells: aging changes the clonal composition of the stem cell compartment but not individual stem cells. Blood 111, 5553–5561 (2008).
ArticleCASPubMedPubMed CentralGoogle Scholar
Bowman, R. L., Busque, L. & Levine, R. L. Clonal hematopoiesis and evolution to hematopoietic malignancies. Cell Stem Cell 22, 157–170 (2018).
ArticleCASPubMedPubMed CentralGoogle Scholar
Challen, G. A. & Goodell, M. A. Clonal hematopoiesis: mechanisms driving dominance of stem cell clones. Blood 136, 1590–1598 (2020).
CASPubMedPubMed CentralGoogle Scholar
Rossi, D. J. et al. Deficiencies in DNA damage repair limit the function of haematopoietic stem cells with age. Nature 447, 725–729 (2007).
ArticleCASPubMedGoogle Scholar
Kirschner, K. et al. Proliferation drives aging-related functional decline in a subpopulation of the hematopoietic stem cell compartment. Cell Rep. 19, 1503–1511 (2017).
ArticleCASPubMedPubMed CentralGoogle Scholar
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).
ArticleCASPubMedPubMed CentralGoogle Scholar
Chen, C., Liu, Y., Liu, Y. & Zheng, P. mTOR regulation and therapeutic rejuvenation of aging hematopoietic stem cells. Sci. Signal. 2, ra75 (2009).
ArticlePubMedPubMed CentralGoogle Scholar
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).
ArticleCASPubMedPubMed CentralGoogle Scholar
Tang, B. et al. Aging-disturbed FUS phase transition impairs hematopoietic stem cells by altering chromatin structure. Blood 143, 124–138 (2024).
ArticleCASPubMedGoogle Scholar
Florian, M. C. et al. Cdc42 activity regulates hematopoietic stem cell aging and rejuvenation. Cell Stem Cell 10, 520–530 (2012).
ArticleCASPubMedPubMed CentralGoogle Scholar
Suda, T., Arai, F. & Hirao, A. Hematopoietic stem cells and their niche. Trends Immunol. 26, 426–433 (2005).
ArticleCASPubMedGoogle Scholar
Nakamura-Ishizu, A., Ito, K. & Suda, T. Hematopoietic stem cell metabolism during development and aging. Dev. Cell 54, 239–255 (2020).
ArticleCASPubMedPubMed CentralGoogle Scholar
Xu, Y., Chiang, Y. H., Ho, P. C. & Vannini, N. Mitochondria dictate function and fate of HSCs and T cells. Cancer Immunol. Res. 11, 1303–1313 (2023).
ArticleCASPubMedGoogle Scholar
Morganti, C. & Ito, K. Mitochondrial contributions to hematopoietic stem cell aging. Int. J. Mol. Sci. 22, 11117 (2021).
ArticleCASPubMedPubMed CentralGoogle Scholar
Filippi, M. D. & Ghaffari, S. Mitochondria in the maintenance of hematopoietic stem cells: new perspectives and opportunities. Blood 133, 1943–1952 (2019).
ArticleCASPubMedPubMed CentralGoogle Scholar
Umemoto, T., Hashimoto, M., Matsumura, T., Nakamura-Ishizu, A. & Suda, T. Ca2+–mitochondria axis drives cell division in hematopoietic stem cells. J. Exp. Med. 215, 2097–2113 (2018).
ArticleCASPubMedPubMed CentralGoogle Scholar
Harada, K. et al. Mitochondrial electron transport chain complex II dysfunction causes premature aging of hematopoietic stem cells. Stem Cells 41, 39–49 (2023).
ArticlePubMedGoogle Scholar
Mohrin, M. et al. Stem cell aging. A mitochondrial UPR-mediated metabolic checkpoint regulates hematopoietic stem cell aging. Science 347, 1374–1377 (2015).
ArticleCASPubMedPubMed CentralGoogle Scholar
Sun, X. et al. Nicotinamide riboside attenuates age-associated metabolic and functional changes in hematopoietic stem cells. Nat. Commun. 12, 2665 (2021).
ArticleCASPubMedPubMed CentralGoogle Scholar
Girotra, M. et al. Induction of mitochondrial recycling reverts age-associated decline of the hematopoietic and immune systems. Nat. Aging 3, 1057–1066 (2023).
ArticleCASPubMedGoogle Scholar
de Almeida, M. J., Luchsinger, L. L., Corrigan, D. J., Williams, L. J. & Snoeck, H. W. Dye-independent methods reveal elevated mitochondrial mass in hematopoietic stem cells. Cell Stem Cell 21, 725–729 (2017).
ArticlePubMedPubMed CentralGoogle Scholar
Pham, A. H., McCaffery, J. M. & Chan, D. C. Mouse lines with photo-activatable mitochondria to study mitochondrial dynamics. Genesis 50, 833–843 (2012).
ArticleCASPubMedPubMed CentralGoogle Scholar
Takihara, Y. et al. High mitochondrial mass is associated with reconstitution capacity and quiescence of hematopoietic stem cells. Blood Adv. 3, 2323–2327 (2019).
ArticleCASPubMedPubMed CentralGoogle Scholar
Hinge, A. et al. Asymmetrically segregated mitochondria provide cellular memory of hematopoietic stem cell replicative history and drive HSC attrition. Cell Stem Cell 26, 420–430 (2020).
ArticleCASPubMedPubMed CentralGoogle Scholar
Ho, T. T. et al. Autophagy maintains the metabolism and function of young and old stem cells. Nature 543, 205–210 (2017).
ArticleCASPubMedPubMed CentralGoogle Scholar
Balazs, A. B., Fabian, A. J., Esmon, C. T. & Mulligan, R. C. Endothelial protein C receptor (CD201) explicitly identifies hematopoietic stem cells in murine bone marrow. Blood 107, 2317–2321 (2006).
ArticleCASPubMedPubMed CentralGoogle Scholar
Beerman, I. et al. Functionally distinct hematopoietic stem cells modulate hematopoietic lineage potential during aging by a mechanism of clonal expansion. Proc. Natl Acad. Sci. USA 107, 5465–5470 (2010).
ArticleCASPubMedPubMed CentralGoogle Scholar
Montecino-Rodriguez, E. et al. Lymphoid-biased hematopoietic stem cells are maintained with age and efficiently generate lymphoid progeny. Stem Cell Rep. 12, 584–596 (2019).
ArticleCASGoogle Scholar
Wilkinson, A. C. et al. Long-term ex vivo haematopoietic-stem-cell expansion allows nonconditioned transplantation. Nature 571, 117–121 (2019).
ArticleCASPubMedPubMed CentralGoogle Scholar
Kowalczyk, M. S. et al. Single-cell RNA-seq reveals changes in cell cycle and differentiation programs upon aging of hematopoietic stem cells. Genome Res. 25, 1860–1872 (2015).
ArticleCASPubMedPubMed CentralGoogle Scholar
Li, J. et al. STAT1 is essential for HSC function and maintains MHCIIhi stem cells that resist myeloablation and neoplastic expansion. Blood 140, 1592–1606 (2022).
ArticleCASPubMedPubMed CentralGoogle Scholar
Lauridsen, F. K. B. et al. Differences in cell cycle status underlie transcriptional heterogeneity in the HSC compartment. Cell Rep. 24, 766–780 (2018).
ArticleCASPubMedGoogle Scholar
Wilson, N. K. et al. Combined single-cell functional and gene expression analysis resolves heterogeneity within stem cell populations. Cell Stem Cell 16, 712–724 (2015).
ArticleCASPubMedPubMed CentralGoogle Scholar
Sommerkamp, P. et al. Mouse multipotent progenitor 5 cells are located at the interphase between hematopoietic stem and progenitor cells. Blood 137, 3218–3224 (2021).
ArticleCASPubMedPubMed CentralGoogle Scholar
Chitteti, B. R. et al. CD166 regulates human and murine hematopoietic stem cells and the hematopoietic niche. Blood 124, 519–529 (2014).
ArticleCASPubMedPubMed CentralGoogle Scholar
Hu, M. et al. CD63 acts as a functional marker in maintaining hematopoietic stem cell quiescence through supporting TGFβ signaling in mice. Cell Death Differ. 29, 178–191 (2022).
ArticlePubMedGoogle Scholar
Sawai, C. M. et al. Hematopoietic stem cells are the major source of multilineage hematopoiesis in adult animals. Immunity 45, 597–609 (2016).
ArticleCASPubMedPubMed CentralGoogle Scholar
Liberzon, A. et al. The Molecular Signatures Database (MSigDB) hallmark gene set collection. Cell Syst. 1, 417–425 (2015).
ArticleCASPubMedPubMed CentralGoogle Scholar
Rath, S. et al. MitoCarta3.0: an updated mitochondrial proteome now with sub-organelle localization and pathway annotations. Nucleic Acids Res. 49, D1541–D1547 (2021).
ArticleCASPubMedGoogle Scholar
Mishra, P. & Chan, D. C. Metabolic regulation of mitochondrial dynamics. J. Cell Biol. 212, 379–387 (2016).
ArticleCASPubMedPubMed CentralGoogle Scholar
Flohr Svendsen, A. et al. A comprehensive transcriptome signature of murine hematopoietic stem cell aging. Blood 138, 439–451 (2021).
ArticleCASPubMedGoogle Scholar
Bartlett, S. et al. GPR183 regulates interferons, autophagy, and bacterial growth during Mycobacterium tuberculosis infection and is associated with TB disease severity. Front. Immunol. 11, 601534 (2020).
ArticleCASPubMedPubMed CentralGoogle Scholar
Lima, T., Li, T. Y., Mottis, A. & Auwerx, J. Pleiotropic effects of mitochondria in aging. Nat. Aging 2, 199–213 (2022).
ArticlePubMedGoogle Scholar
Silva Ramos, E. et al. Mitochondrial fusion is required for regulation of mitochondrial DNA replication. PLoS Genet. 15, e1008085 (2019).
ArticlePubMedPubMed CentralGoogle Scholar
Yamamoto, R. et al. Large-scale clonal analysis resolves aging of the mouse hematopoietic stem cell compartment. Cell Stem Cell 22, 600–607 (2018).
ArticleCASPubMedPubMed CentralGoogle Scholar
Tanaka, T. et al. Identification of differentiation roots of hematopoietic stem cells by a paired-daughter assay combined with multiple barcoding. Exp. Hematol. 111, S146 (2022).
ArticleGoogle Scholar
Yien, Y. Y. & Perfetto, M. Regulation of heme synthesis by mitochondrial homeostasis proteins. Front. Cell Dev. Biol. 10, 895521 (2022).
ArticlePubMedPubMed CentralGoogle Scholar
Liang, R. et al. Restraining lysosomal activity preserves hematopoietic stem cell quiescence and potency. Cell Stem Cell 26, 359–376 (2020).
ArticleCASPubMedPubMed CentralGoogle Scholar
Watanuki, S. et al. SDHAF1 confers metabolic resilience to aging hematopoietic stem cells by promoting mitochondrial ATP production. Cell Stem Cell 31, 1145–1161 (2024).
ArticleCASPubMedGoogle Scholar
Bonora, M. et al. A mitochondrial NADPH-cholesterol axis regulates extracellular vesicle biogenesis to support hematopoietic stem cell fate. Cell Stem Cell 31, 359–377 (2024).
ArticleCASPubMedGoogle Scholar
Arosio, P., Elia, L. & Poli, M. Ferritin, cellular iron storage and regulation. IUBMB Life 69, 414–422 (2017).
ArticleCASPubMedGoogle Scholar
Yi, W. et al. Ferritin-mediated mitochondrial iron homeostasis is essential for the survival of hematopoietic stem cells and leukemic stem cells. Leukemia 5, 1003–1018 (2024).
Jiang, X., Stockwell, B. R. & Conrad, M. Ferroptosis: mechanisms, biology and role in disease. Nat. Rev. Mol. Cell Biol. 22, 266–282 (2021).
ArticlePubMedPubMed CentralGoogle Scholar
Freitas, F. P. et al. 7-Dehydrocholesterol is an endogenous suppressor of ferroptosis. Nature 626, 401–410 (2024).
ArticleCASPubMedGoogle Scholar
Hannedouche, S. et al. Oxysterols direct immune cell migration via EBI2. Nature 475, 524–527 (2011).
ArticleCASPubMedPubMed CentralGoogle Scholar
Liu, C. et al. Oxysterols direct B-cell migration through EBI2. Nature 475, 519–523 (2011).
ArticleCASPubMedGoogle Scholar
Zhang, P. et al. G protein-coupled receptor 183 facilitates endothelial-to-hematopoietic transition via Notch1 inhibition. Cell Res. 25, 1093–1107 (2015).
ArticleCASPubMedPubMed CentralGoogle Scholar
Nevius, E. et al. Oxysterols and EBI2 promote osteoclast precursor migration to bone surfaces and regulate bone mass homeostasis. J. Exp. Med. 212, 1931–1946 (2015).
ArticleCASPubMedPubMed CentralGoogle Scholar
Arcangeli, M. L. et al. JAM-B regulates maintenance of hematopoietic stem cells in the bone marrow. Blood 118, 4609–4619 (2011).
ArticleCASPubMedGoogle Scholar
Oedekoven, C. A. et al. Hematopoietic stem cells retain functional potential and molecular identity in hibernation cultures. Stem Cell Rep. 16, 1614–1628 (2021).
ArticleCASGoogle Scholar
Wang, T. et al. Loss of Nupr1 promotes engraftment by tuning the quiescence threshold of hematopoietic stem cell repository via regulating p53-checkpoint pathway. Haematologica 107, 154–166 (2022).
ArticleCASPubMedGoogle Scholar
Liu, J. et al. NUPR1 is a critical repressor of ferroptosis. Nat. Commun. 12, 647 (2021).
ArticleCASPubMedPubMed CentralGoogle Scholar
Matsumura, T. et al. Hematopoietic stem cells acquire survival advantage by loss of RUNX1 methylation identified in familial leukemia. Blood 136, 1919–1932 (2020).
ArticlePubMedGoogle Scholar
Matsumura, T. et al. A Myb enhancer-guided analysis of basophil and mast cell differentiation. Nat. Commun. 13, 7064 (2022).
ArticleCASPubMedPubMed CentralGoogle Scholar
Hao, Y. et al. Integrated analysis of multimodal single-cell data. Cell 184, 3573–3587 (2021).
ArticleCASPubMedPubMed CentralGoogle Scholar
Stuart, T., Srivastava, A., Madad, S., Lareau, C. A. & Satija, R. Single-cell chromatin state analysis with Signac. Nat. Methods 18, 1333–1341 (2021).
ArticleCASPubMedPubMed CentralGoogle Scholar
Hänzelmann, S., Castelo, R. & Guinney, J. GSVA: gene set variation analysis for microarray and RNA-seq data. BMC Bioinformatics 14, 7 (2013).
ArticlePubMedPubMed CentralGoogle Scholar
Kanehisa, M. & Goto, S. KEGG: Kyoto Encyclopedia of Genes and Genomes. Nucleic Acids Res. 28, 27–30 (2000).
ArticleCASPubMedPubMed CentralGoogle Scholar
Ritchie, M. E. et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43, e47 (2015).
ArticlePubMedPubMed CentralGoogle Scholar
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).
ArticleCASPubMedPubMed CentralGoogle Scholar
Liao, Y., Smyth, G. K. & Shi, W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics 30, 923–930 (2014).
ArticleCASPubMedGoogle Scholar
Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).
ArticlePubMedPubMed CentralGoogle Scholar
Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA 102, 15545–15550 (2005).
ArticleCASPubMedPubMed CentralGoogle Scholar
Argüello, R. J. et al. SCENITH: a flow cytometry-based method to functionally profile energy metabolism with single-cell resolution. Cell Metab. 32, 1063–1075 (2020).
ArticlePubMedPubMed CentralGoogle Scholar
Watanabe, A. et al. Noncanonical TCA cycle fosters canonical TCA cycle and mitochondrial integrity in acute myeloid leukemia. Cancer Sci. 116, 152–163 (2025).
ArticleCASPubMedGoogle Scholar
Download references
Acknowledgements
The authors thank the CSI FACS facility for technical assistance and the NUS MD2 Vivarium for mouse husbandry. This research is supported by the Singapore Translational Research Investigator Award from the National Medical Research Council in Singapore (NMRC/STaR 18 may-0004 to T. Suda); JSPS Grant-in-Aid for Scientific Research (S) (18H05284 and 26221309 to T. Suda); the Chinese Academy of Medical Sciences Innovation Fund for Medical Sciences (CIFMS) (2024-12M-3-017 to T. Suda); the National Natural Science Foundation of China (W2441024 to T. Suda); JSPS Grant-in-Aid for Challenging Research (Exploratory) (JP21K19514 to T.M.); JSPS Grant-in-Aid for Scientific Research (C) (24K11298 to T.M.); the Daiwa Securities Health Foundation (T.M.); the Novartis Foundation (Japan) for the Promotion of Science (T.M.); the SENSHIN Medical Research Foundation (T.M.); the Takeda Science Foundation (T.M.); the Naito Foundation (T.M.); and the Open Fund-Young Individual Research Grant (OF-YIRG) from the National Medical Research Council in Singapore (OFYIRG21nov-0020 to H.T.).
Author information
Author notes
These authors contributed equally: Haruhito Totani, Takayoshi Matsumura.
These authors jointly supervised this work: Takayoshi Matsumura, Toshio Suda.
Authors and Affiliations
Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
Haruhito Totani, Takayoshi Matsumura, Rui Yokomori, Yuji Takihara, Chong Yang, Lee Hui Chua, Atsushi Watanabe, Takaomi Sanda & Toshio Suda
Division of Cardiovascular and Genetic Research, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
Takayoshi Matsumura
International Research Center of Medical Sciences, Kumamoto University, Kumamoto, Japan
Terumasa Umemoto & Toshio Suda
State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
Chong Yang & Toshio Suda
Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
Takaomi Sanda
Authors
Haruhito Totani
View author publications
You can also search for this author in PubMedGoogle Scholar
2. Takayoshi Matsumura
View author publications
You can also search for this author in PubMedGoogle Scholar
3. Rui Yokomori
View author publications
You can also search for this author in PubMedGoogle Scholar
4. Terumasa Umemoto
View author publications
You can also search for this author in PubMedGoogle Scholar
5. Yuji Takihara
View author publications
You can also search for this author in PubMedGoogle Scholar
6. Chong Yang
View author publications
You can also search for this author in PubMedGoogle Scholar
7. Lee Hui Chua
View author publications
You can also search for this author in PubMedGoogle Scholar
8. Atsushi Watanabe
View author publications
You can also search for this author in PubMedGoogle Scholar
9. Takaomi Sanda
View author publications
You can also search for this author in PubMedGoogle Scholar
10. Toshio Suda
View author publications
You can also search for this author in PubMedGoogle Scholar
Contributions
H.T., T.M. and T. Suda conceived and designed the experiments. H.T., T.M., T.U., Y.T., C.Y., L.H.C., A.W. and T. Sanda performed the experiments. T.M. and R.Y. analyzed bioinformatics data. H.T. and T.M. wrote the manuscript. T.M. and T. Suda supervised the research. All authors reviewed and approved the manuscript. H.T. and T.M. contributed equally to this work.
Corresponding authors
Correspondence to Takayoshi Matsumura or Toshio Suda.
Ethics declarations
Competing interests
The authors declare no financial conflicts of interest.
Peer review
Peer review information
Nature Aging thanks Simón Méndez-Ferrer, Danica Chen 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 Flow cytometry gating strategy for HSCs.
(a) Gating Strategy for HSCs in flow cytometry. FSC, forward scatter; FSC-A, FSC-aria; FSC-H, FSC-height; FSC-W, FSC-width; SSC, side scatter; SSC-A, SSC-aria; SSC-H, SSC-height; SSC-W, SSC-width; PI, propidium iodide; Live cells, PI− cells; Lin− cells, lineage− (CD4−CD8−B220−Gr1−CD11b−Ter119−NK1.1−CD127−) cells; LS−K, Lin−Sca1−c-Kit+; LSK, Lin−Sca1+c-Kit+; MPP, CD150−CD48+ LSK; ST-HSC, CD150−CD48− LSK; SLAM (LT-HSC), CD150+CD48− LSK; ESLAM, EPCR+ SLAM-HSC. (b) Gating Strategy for classical HSCs in flow cytometry. LT-HSC, CD34−CD135− LSK; ST-HSC, CD34+CD135− LSK; MPP, CD34+CD135+ LSK; CMP, CD34+CD16/32− LS−K; GMP, CD34+CD16/32+ LS−K; MEP, CD34−CD16/32− LS−K.
Extended Data Fig. 2 Expression of mito-Dendra2 in HSCs and analysis of their phenotype during bone marrow transplantation.
(a) Mitochondrial mass in young and aged HSPCs. Representative histogram of mito-Dendra2 for ST-HSC, MPP, LS−K, and Lin−. (b) Mitochondrial DNA copy number in mito-Dendra2 Low and High HSCs (P = 0.0118, n = 6). (c) EPCR expression in aged ESLAM-HSCs by mitochondrial mass. Relative MFI of EPCR (P = 0.0001, n = 6). (d) Complete blood count post-BMT (n = 6, 8 for Low and High groups). P-values by week: white blood cell - 4 (0.0015), 8 (0.4187), 12 ( < 0.0001), 16 (0.0529); red blood cell - 4 (0.8886), 8 (0.8816), 12 (0.0300), 16 (0.8269); hemoglobin - 4 (0.4995), 8 (0.3988), 12 (0.0037), 16 (0.8101); hematocrit - 4 (0.9067), 8 (0.4799), 12 (0.0092), 16 (0.4611); platelet - 4 (0.0672), 8 (0.8458), 12 (0.5581), 16 (0.6478). (**e**-**g**) Proportion of mito-Dendra2+ cell in LSK and LS−K fractions at 16 weeks post-BMT (Low: n = 6, High: n = 8). (e) HSPCs (Extended Data Fig. 1a). P-values: ST-HSC (0.1639), MPP (0.5754), LSK (0.1102), LS−K (0.5728), Lin− (0.5853). (f) HSPCs (Extended Data Fig. 1b). P-values: ST-HSC (CD34+CD135−) (0.2781), MPP (CD34+CD135+) (0.4255), CMP (0.6850), GMP (0.5686), MEP (0.5608). (g) MPP2 (CD150+CD48− ESLAM-HSCs). P = 0.1084. (h) mito-Dendra2 expression in donor-derived cells in the HSC at 16 weeks post-BMT. Due to the lower number of donor-positive cells in the HSC fraction originating from mito-Dendra2 Low cells, the sample size is smaller compared to that of mito-Dendra2 High cells (n = 3, 7 for mito-Dendra2 Low and High). P = 0.1510. (i) Proportion of donor-derived cells post-BMT in young mice (ref. 26; n = 5, 4 for Young-Low and Young-High). P-values: SLAM (0.0374), ST-HSC (0.1513), MPP (0.0975). Note: All charts show mean ± SD. Statistical analysis: two-tailed Student’s t-test (*P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001; ns = not significant).
Source data
Extended Data Fig. 3 Single cell multiome analysis classified HSCs into four clusters.
(a) A heatmap of multiome analysis of young and aged mito-Dendra2 HSCs, showing the top differentially expressed genes in each cluster. The x-axis represents individual cells and clusters, and the y-axis represents differentially expressed genes. (b) Uniform manifold approximation and projection (UMAP) visualization of HSCs from young and aged mito-Dendra2 mice. scRNA-seq and scATAC-seq data were integrated with Weighted Nearest Neighbor (WNN) analysis. UMAP based on WNN analysis (top), scRNA-seq data only (middle), and scATAC-seq data only (bottom) were shown. In the right panels, young and aged mito-Dendra2 Low and High HSCs were depicted separately. The top leftmost panel is identical to Fig. 3b.
Extended Data Fig. 4 Single cell multiome analysis revealed that aged mito-Dendra2 High HSCs were characterized with HSC signature and oxidative phosphorylation genes.
(a) Violin plots showing differences in Surface Marker Overlap (SuMo) scores (upper) and HSC scores (lower) of young and aged mito-Dendra2 Low and High HSCs in each cluster. In the upper panel, P = 0, 2.99×10−209, 1.04×10−70, and 7.10×10−25 for Clusters 1, 2,3 and 4 (one-way Welch’s ANOVA). *P = 0, 0, 3.26×10−13, and 1.00×10−11 between aged mito-Dendra2 Low and High HSCs for Clusters 1, 2,3 and 4 (Games-Howell post hoc test). In the lower panel, P = 0, 1.90×10−172, 3.83×10−77, and 1.39×10−19 for Clusters 1, 2,3 and 4 (one-way Welch’s ANOVA). *P = 0, 3.77×10−14, 2.42×10−13, and 3.93×10−8 between aged mito-Dendra2 Low and High HSCs for Clusters 1, 2,3 and 4 (Games-Howell post hoc test). (b) Violin plot showing myeloid-biased (left) and lymphoid-biased (right) score. P = 0 and 0 (one-way Welch’s ANOVA). *P = 2.05×10−12 and 1.69×10−12 (Games-Howell post hoc test). (c) Violin plots showing differences in mRNA expression levels of ribosomal genes between aged mito-Dendra2 Low and High HSCs in clusters 1 (left) and 2 (right). *P = 1.13×10−261, 9.30×10−125, 1.93×10−264, 6.27×10−115, 9.12×10−227, 1.31×10−85, 1.62×10−273 and 2.65×10−150 from left to right, calculated by the Wilcoxon Rank Sum test, and adjusted based on Bonferroni correction with all genes using the Seurat package in R in a two-sided manner. (d) Gene set variation analysis (GSVA) using MitoCarta gene sets between aged mito-Dendra2 Low and High HSCs. Each dot represents one gene set. Two-sided adjusted P-values were calculated using the limma package. (e) Violin plots showing differences in mRNA expression levels of selected iron homeostasis genes between aged mito-Dendra2 Low and High HSCs. *P = 5.18×10−84 and 2.48×10−81 from left to right, calculated by the Wilcoxon Rank Sum test, and adjusted based on Bonferroni correction with all genes using the Seurat package in R in a two-sided manner.
Source data
Extended Data Fig. 5 scRNA-seq and bulk RNA-seq revealed genes specific to aged HSCs.
(a) Gene set enrichment analysis (GSEA) comparing young and aged LT-HSCs using 50 hallmark gene sets. The top 4 gene sets enriched in aged LT-HSCs are shown. Each solid bar represents one gene within the gene set. NES, normalized enrichment score; FDR, false discovery rate. (b) Violin plots showing mRNA expression levels of Kcnb2 and Clu in each cluster of young mito-Dendra2 Low (cyan), young mito-Dendra2 High (pink), aged mito-Dendra2 Low (blue), and aged mito-Dendra2 High HSCs (red). P = 0, 1.84×10−146, 9.94×10−67, and 2.00×10−38 (Kcnb2), and 5.76×10−100, 7.11×10−52, 8.86×10−25, and 5.37×10−11 (Clu) between aged mito-Dendra2 High HSCs and others from left to right, calculated by the Wilcoxon Rank Sum test and adjusted based on Bonferroni correction with all genes using the Seurat package in R. (c) Box plots of bulk RNA-seq showing mRNA expression levels of selected aged LT-HSC-specific genes in young (pink) and aged (cyan) LT-HSCs, ST-HSCs, and MPPs (n = 4). P = 4.30×10−3, 7.07×10−12, 1.78×10−100, 6.46×10−116, 3.07×10−86, and 0, calculated by the likelihood ratio test and adjusted by the Benjamini and Hochberg method using the DESeq2 package in R. The boxes show median values, 1st and 3rd quartiles. The whiskers extend to the most extreme data point which is no more than 1.5 times the interquartile range from the box.
Source data
Extended Data Fig. 6 The relationship between mitochondria and GPR183.
(a) Relative GPR183 expression following SR-18292 treatment, with normalized MFI value (P = 0.0029, n = 6). (b) Relative mito-Dendra2 expression following NIBR-189 and/or 7α,25-DHC treatment, with normalized MFI values (n = 3). Adjusted P-value: NIBR-189−/7α,25-DHC− vs NIBR-189−/7α,25-DHC+ (0.2551), NIBR-189+/7α,25-DHC− vs NIBR-189+/7α,25-DHC+ (0.1046), NIBR-189−/7α,25-DHC− vs NIBR-189+/7α,25-DHC− (0.2310), NIBR-189−/7α,25-DHC+ vs NIBR-189+/7α,25-DHC+ (0.7146). Note: All charts show mean ± SD. Statistical analysis: (a) two-tailed Student’s t-test and (b) one-way ANOVA (*P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001; ns = not significant).
Source data
Extended Data Fig. 7 BMT for GPR183neg and GPR183high HSCs.
(a) Complete blood count post-BMT (n = 6, 7 for GPR183neg and GPR183high groups). P-values by week: white blood cell - 4 (0.0817), 8 (0.0987), 12 (0.6245), 16 (0.0755); red blood cell - 4 (0.5566), 8 (0.0567), 12 (0.3505), 16 (0.3763); hemoglobin - 4 (0.4078), 8 (0.1669), 12 (0.8170), 16 (0.6456); hematocrit - 4 (0.4519), 8 (0.0957), 12 (0.4468), 16 (0.3169); platelet - 4 (0.0269), 8 (0.6668), 12 (0.9417), 16 (0.9568). (b) Proportion of CD45.1+ cells in PB. GPR183low (n = 6, 7 for GPR183neg and GPR183high groups). P-values by week: total cells - 4 (0.0195), 8 (0.0153), 12 (0.0219), 16 (0.0290); granulocytes - 4 (0.0049), 8 (0.0139), 12 (0.0056), 16 (0.0086); B-cells - 4 (0.0659), 8 (0.0829), 12 (0.0937), 16 (0.0933); T-cells - 4 (0.8150), 8 (0.1236), 12 (0.1091), 16 (0.2046). (c-d) Proportion of CD45.1+ cell in LSK and LS−K fractions at 16 weeks post-BMT (n = 6, 7 for GPR183neg and GPR183high groups). (c) HSPCs (Extended Data Fig. 1a). P-values: ST-HSC (0.0365), MPP (0.0268), LSK (0.0229), LS−K (0.0301), Lin− (0.0374). (d) HSPCs (Extended Data Fig. 1b). P-values: ST-HSC (CD34+CD135−) (0.0256), MPP (CD34+CD135+) (0.0370), CMP (0.0250), GMP (0.0205), MEP (0.0385). Note: All charts show mean ± SD. Statistical analysis: two-tailed Student’s t-test (*P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001; ns = not significant).
Source data
Supplementary information
Supplementary Figure
Reporting Summary
Supplementary Table 1
Supplementary Table
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. 2
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.
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
Totani, H., Matsumura, T., Yokomori, R. et al. Mitochondria-enriched hematopoietic stem cells exhibit elevated self-renewal capabilities, thriving within the context of aged bone marrow. Nat Aging (2025). https://doi.org/10.1038/s43587-025-00828-y
Download citation
Received:19 March 2024
Accepted:30 January 2025
Published:06 March 2025
DOI:https://doi.org/10.1038/s43587-025-00828-y
Share this article
Anyone you share the following link with will be able to read this content:
Get shareable link
Sorry, a shareable link is not currently available for this article.
Copy to clipboard
Provided by the Springer Nature SharedIt content-sharing initiative