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:

Depleting myeloid-biased haematopoietic stem cells rejuvenates aged immunity

Abstract

Ageing of the immune system is characterized by decreased lymphopoiesis and adaptive immunity, and increased inflammation and myeloid pathologies1,2. Age-related changes in populations of self-renewing haematopoietic stem cells (HSCs) are thought to underlie these phenomena3. During youth, HSCs with balanced output of lymphoid and myeloid cells (bal-HSCs) predominate over HSCs with myeloid-biased output (my-HSCs), thereby promoting the lymphopoiesis required for initiating adaptive immune responses, while limiting the production of myeloid cells, which can be pro-inflammatory4. Ageing is associated with increased proportions of my-HSCs, resulting in decreased lymphopoiesis and increased myelopoiesis3,5,6. Transfer of bal-HSCs results in abundant lymphoid and myeloid cells, a stable phenotype that is retained after secondary transfer; my-HSCs also retain their patterns of production after secondary transfer5. The origin and potential interconversion of these two subsets is still unclear. If they are separate subsets postnatally, it might be possible to reverse the ageing phenotype by eliminating my-HSCs in aged mice. Here we demonstrate that antibody-mediated depletion of my-HSCs in aged mice restores characteristic features of a more youthful immune system, including increasing common lymphocyte progenitors, naive T cells and B cells, while decreasing age-related markers of immune decline. Depletion of my-HSCs in aged mice improves primary and secondary adaptive immune responses to viral infection. These findings may have relevance to the understanding and intervention of diseases exacerbated or caused by dominance of the haematopoietic system by my-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: Identification of cell-surface antigens on my-HSCs.
Fig. 2: Antibody-mediated depletion of my-HSCs in vivo.
Fig. 3: Depletion of my-HSCs in aged mice restores youthful immune features.
Fig. 4: Antibody conditioning enhances functional immunity in aged mice.
Fig. 5: Mouse my-HSC markers are enriched in aged human HSCs.

Similar content being viewed by others

Data availability

Data for all graphical representations are provided as source data. RNA-seq data have been deposited at the GEO under accession code GSE252062 and the Sequence Read Archive (SRA) under BioProject PRJNA1054066. The following publicly available datasets were used: GSE43729 (ref. 16), GSE39553 (ref. 24), GSE48893 (ref. 25), GSE109546 (ref. 26), GSE27686 (ref. 27), GSE44923 (ref. 28), GSE128050 (ref. 29), GSE47819 (ref. 30), GSE130504 (ref. 19), GSE112769 (ref. 22), E-MEXP-3935 (ref. 21), GSE32719 (ref. 3), GSE104406 (ref. 53), GSE69408 (ref. 54), GSE115348 (ref. 55), GSE107594 (ref. 104), GSE111410 (ref. 56), GSE55689 (ref. 57), GSE74246 (ref. 58), GSE132040 (ref. 108), GSE87633 (ref. 109) and GSE100428 (ref. 18). Source data are provided with this paper.

References

  1. Morrison, S. J., Wandycz, A. M., Akashi, K., Globerson, A. & Weissman, I. L. The aging of hematopoietic stem cells. Nat. Med. 2, 1011–1016 (1996).

    Article  CAS  PubMed  Google Scholar 

  2. Rossi, D. J., Jamieson, C. H. & Weissman, I. L. Stems cells and the pathways to aging and cancer. Cell 132, 681–696 (2008).

    Article  CAS  PubMed  Google Scholar 

  3. Pang, W. W. et al. Human bone marrow hematopoietic stem cells are increased in frequency and myeloid-biased with age. Proc. Natl Acad. Sci. USA 108, 20012–20017 (2011).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  4. Yamamoto, R. & Nakauchi, H. In vivo clonal analysis of aging hematopoietic stem cells. Mech. Ageing Dev. 192, 111378 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. 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).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  6. Rossi, D. J. et al. Cell intrinsic alterations underlie hematopoietic stem cell aging. Proc. Natl Acad. Sci. USA 102, 9194–9199 (2005).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  7. Muller-Sieburg, C. E., Cho, R. H., Karlsson, L., Huang, J. F. & Sieburg, H. B. Myeloid-biased hematopoietic stem cells have extensive self-renewal capacity but generate diminished lymphoid progeny with impaired IL-7 responsiveness. Blood 103, 4111–4118 (2004).

    Article  CAS  PubMed  Google Scholar 

  8. Sudo, K., Ema, H., Morita, Y. & Nakauchi, H. Age-associated characteristics of murine hematopoietic stem cells. J. Exp. Med. 192, 1273–1280 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Sieburg, H. B. et al. The hematopoietic stem compartment consists of a limited number of discrete stem cell subsets. Blood 107, 2311–2316 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Dykstra, B. et al. Long-term propagation of distinct hematopoietic differentiation programs in vivo. Cell Stem Cell 1, 218–229 (2007).

    Article  CAS  PubMed  Google Scholar 

  11. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Min, H., Montecino-Rodriguez, E. & Dorshkind, K. Effects of aging on the common lymphoid progenitor to pro-B cell transition. J. Immunol. 176, 1007–1012 (2006).

    Article  CAS  PubMed  Google Scholar 

  13. Montecino-Rodriguez, E., Berent-Maoz, B. & Dorshkind, K. Causes, consequences, and reversal of immune system aging. J. Clin. Invest. 123, 958–965 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Yang, D. & de Haan, G. Inflammation and aging of hematopoietic stem cells in their niche. Cells 10, 1849 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Chen, J. Y. et al. Hoxb5 marks long-term haematopoietic stem cells and reveals a homogenous perivascular niche. Nature 530, 223–227 (2016).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  16. Beerman, I. et al. Proliferation-dependent alterations of the DNA methylation landscape underlie hematopoietic stem cell aging. Cell Stem Cell 12, 413–425 (2013).

    Article  CAS  PubMed  Google Scholar 

  17. Gekas, C. & Graf, T. CD41 expression marks myeloid-biased adult hematopoietic stem cells and increases with age. Blood 121, 4463–4472 (2013).

    Article  CAS  PubMed  Google Scholar 

  18. Mann, M. et al. Heterogeneous responses of hematopoietic stem cells to inflammatory stimuli are altered with age. Cell Rep. 25, 2992–3005 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Gulati, G. S. et al. Neogenin-1 distinguishes between myeloid-biased and balanced. Proc. Natl Acad. Sci. USA 116, 25115–25125 (2019).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  20. Flohr Svendsen, A. et al. A comprehensive transcriptome signature of murine hematopoietic stem cell aging. Blood 138, 439–451 (2021).

    Article  CAS  PubMed  Google Scholar 

  21. Sanjuan-Pla, A. et al. Platelet-biased stem cells reside at the apex of the haematopoietic stem-cell hierarchy. Nature 502, 232–236 (2013).

    Article  ADS  CAS  PubMed  Google Scholar 

  22. 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).

    Article  CAS  Google Scholar 

  23. 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 

  24. Bersenev, A. et al. Lnk deficiency partially mitigates hematopoietic stem cell aging. Aging Cell 11, 949–959 (2012).

    Article  CAS  PubMed  Google Scholar 

  25. Flach, J. et al. Replication stress is a potent driver of functional decline in ageing haematopoietic stem cells. Nature 512, 198–202 (2014).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  26. Maryanovich, M. et al. Adrenergic nerve degeneration in bone marrow drives aging of the hematopoietic stem cell niche. Nat. Med. 24, 782–791 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Norddahl, G. L. et al. Accumulating mitochondrial DNA mutations drive premature hematopoietic aging phenotypes distinct from physiological stem cell aging. Cell Stem Cell 8, 499–510 (2011).

    Article  CAS  PubMed  Google Scholar 

  28. Wahlestedt, M. et al. An epigenetic component of hematopoietic stem cell aging amenable to reprogramming into a young state. Blood 121, 4257–4264 (2013).

    Article  CAS  PubMed  Google Scholar 

  29. Renders, S. et al. Niche derived netrin-1 regulates hematopoietic stem cell dormancy via its receptor neogenin-1. Nat. Commun. 12, 608 (2021).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  30. 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 

  31. Seita, J. et al. Gene Expression Commons: an open platform for absolute gene expression profiling. PLoS ONE 7, e40321 (2012).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  32. Akashi, K., Traver, D., Miyamoto, T. & Weissman, I. L. A clonogenic common myeloid progenitor that gives rise to all myeloid lineages. Nature 404, 193–197 (2000).

    Article  ADS  CAS  PubMed  Google Scholar 

  33. Kondo, M., Weissman, I. L. & Akashi, K. Identification of clonogenic common lymphoid progenitors in mouse bone marrow. Cell 91, 661–672 (1997).

    Article  CAS  PubMed  Google Scholar 

  34. George, B. M. et al. Antibody conditioning enables MHC-mismatched hematopoietic stem cell transplants and organ graft tolerance. Cell Stem Cell 25, 185–192 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Czechowicz, A., Kraft, D., Weissman, I. L. & Bhattacharya, D. Efficient transplantation via antibody-based clearance of hematopoietic stem cell niches. Science 318, 1296–1299 (2007).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  36. Jaiswal, S. et al. CD47 is upregulated on circulating hematopoietic stem cells and leukemia cells to avoid phagocytosis. Cell 138, 271–285 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Kuribayashi, W. et al. Limited rejuvenation of aged hematopoietic stem cells in young bone marrow niche. J. Exp. Med. 218, e20192283 (2021).

    Article  CAS  PubMed  Google Scholar 

  38. Morrison, S. J. & Weissman, I. L. The long-term repopulating subset of hematopoietic stem cells is deterministic and isolatable by phenotype. Immunity 1, 661–673 (1994).

    Article  CAS  PubMed  Google Scholar 

  39. Akashi, K., Kondo, M. & Weissman, I. L. Two distinct pathways of positive selection for thymocytes. Proc. Natl Acad. Sci. USA 95, 2486–2491 (1998).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  40. Gattinoni, L. et al. A human memory T cell subset with stem cell-like properties. Nat. Med. 17, 1290–1297 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Elyahu, Y. et al. Aging promotes reorganization of the CD4 T cell landscape toward extreme regulatory and effector phenotypes. Sci. Adv. 5, eaaw8330 (2019).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  42. Hao, Y., O’Neill, P., Naradikian, M. S., Scholz, J. L. & Cancro, M. P. A B-cell subset uniquely responsive to innate stimuli accumulates in aged mice. Blood 118, 1294–1304 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Pioli, P. D., Casero, D., Montecino-Rodriguez, E., Morrison, S. L. & Dorshkind, K. Plasma cells are obligate effectors of enhanced myelopoiesis in aging bone marrow. Immunity 51, 351–366 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Kovtonyuk, L. V. et al. IL-1 mediates microbiome-induced inflammaging of hematopoietic stem cells in mice. Blood 139, 44–58 (2022).

    Article  CAS  PubMed  Google Scholar 

  45. Collier, D. A. et al. Age-related immune response heterogeneity to SARS-CoV-2 vaccine BNT162b2. Nature 596, 417–422 (2021).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  46. Myers, L. & Hasenkrug, K. J. Retroviral immunology: lessons from a mouse model. Immunol. Res. 43, 160–166 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Dittmer, U. et al. Friend retrovirus studies reveal complex interactions between intrinsic, innate and adaptive immunity. FEMS Microbiol. Rev. 43, 435–456 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Dittmer, U., Brooks, D. M. & Hasenkrug, K. J. Requirement for multiple lymphocyte subsets in protection by a live attenuated vaccine against retroviral infection. Nat. Med. 5, 189–193 (1999).

    Article  CAS  PubMed  Google Scholar 

  49. Dittmer, U., Brooks, D. M. & Hasenkrug, K. J. Characterization of a live-attenuated retroviral vaccine demonstrates protection via immune mechanisms. J. Virol. 72, 6554–6558 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Dittmer, U., Brooks, D. M. & Hasenkrug, K. J. Protection against establishment of retroviral persistence by vaccination with a live attenuated virus. J. Virol. 73, 3753–3757 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Hasenkrug, K. J. & Dittmer, U. The role of CD4 and CD8 T cells in recovery and protection from retroviral infection: lessons from the Friend virus model. Virology 272, 244–249 (2000).

    Article  CAS  PubMed  Google Scholar 

  52. Larochelle, A. et al. Human and rhesus macaque hematopoietic stem cells cannot be purified based only on SLAM family markers. Blood 117, 1550–1554 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. 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 

  54. Rundberg Nilsson, A., Soneji, S., Adolfsson, S., Bryder, D. & Pronk, C. J. Human and murine hematopoietic stem cell aging is associated with functional impairments and intrinsic megakaryocytic/erythroid bias. PLoS ONE 11, e0158369 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  55. Hennrich, M. L. et al. Cell-specific proteome analyses of human bone marrow reveal molecular features of age-dependent functional decline. Nat. Commun. 9, 4004 (2018).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  56. Tong, J. et al. Hematopoietic stem cell heterogeneity is linked to the initiation and therapeutic response of myeloproliferative neoplasms. Cell Stem Cell 28, 502–513 (2021).

    Article  CAS  PubMed  Google Scholar 

  57. Woll, P. S. et al. Myelodysplastic syndromes are propagated by rare and distinct human cancer stem cells in vivo. Cancer Cell 25, 794–808 (2014).

    Article  CAS  PubMed  Google Scholar 

  58. Corces, M. R. et al. Lineage-specific and single-cell chromatin accessibility charts human hematopoiesis and leukemia evolution. Nat. Genet. 48, 1193–1203 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Park, C. Y., Majeti, R. & Weissman, I. L. In vivo evaluation of human hematopoiesis through xenotransplantation of purified hematopoietic stem cells from umbilical cord blood. Nat. Protoc. 3, 1932–1940 (2008).

    Article  CAS  PubMed  Google Scholar 

  60. Bhattacharya, D. et al. Transcriptional profiling of antigen-dependent murine B cell differentiation and memory formation. J. Immunol. 179, 6808–6819 (2007).

    Article  CAS  PubMed  Google Scholar 

  61. Luckey, C. J. et al. Memory T and memory B cells share a transcriptional program of self-renewal with long-term hematopoietic stem cells. Proc. Natl Acad. Sci. USA 103, 3304–3309 (2006).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  62. Saggau, C. et al. The pre-exposure SARS-CoV-2-specific T cell repertoire determines the quality of the immune response to vaccination. Immunity. 55, 1924–1939 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Merad, M., Blish, C. A., Sallusto, F. & Iwasaki, A. The immunology and immunopathology of COVID-19. Science 375, 1122–1127 (2022).

    Article  ADS  CAS  PubMed  Google Scholar 

  64. Jaiswal, S. & Weissman, I. L. Hematopoietic stem and progenitor cells and the inflammatory response. Ann. N. Y. Acad. Sci. 1174, 118–121 (2009).

    Article  ADS  CAS  PubMed  Google Scholar 

  65. Hirata, Y. et al. CD150high bone marrow Tregs maintain hematopoietic stem cell quiescence and immune privilege via adenosine. Cell Stem Cell 22, 445–453 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Jamieson, C. H. M. & Weissman, I. L. Stem-cell aging and pathways to precancer evolution. N. Engl. J. Med. 389, 1310–1319 (2023).

    Article  CAS  PubMed  Google Scholar 

  67. Busque, L. et al. Recurrent somatic TET2 mutations in normal elderly individuals with clonal hematopoiesis. Nat. Genet. 44, 1179–1181 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Jan, M. et al. Clonal evolution of preleukemic hematopoietic stem cells precedes human acute myeloid leukemia. Sci. Transl. Med. 4, 149ra118 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  69. Jaiswal, S. & Ebert, B. L. Clonal hematopoiesis in human aging and disease. Science 366, eaan4673 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Jaiswal, S. et al. Clonal hematopoiesis and risk of atherosclerotic cardiovascular disease. N. Engl. J. Med. 377, 111–121 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  71. Majeti, R. et al. Clonal expansion of stem/progenitor cells in cancer, fibrotic diseases, and atherosclerosis, and CD47 protection of pathogenic cells. Annu. Rev. Med. 73, 307–320 (2022).

    Article  CAS  PubMed  Google Scholar 

  72. Spangrude, G. J., Heimfeld, S. & Weissman, I. L. Purification and characterization of mouse hematopoietic stem cells. Science 241, 58–62 (1988).

    Article  ADS  CAS  PubMed  Google Scholar 

  73. Osawa, M., Hanada, K., Hamada, H. & Nakauchi, H. Long-term lymphohematopoietic reconstitution by a single CD34-low/negative hematopoietic stem cell. Science 273, 242–245 (1996).

    Article  ADS  CAS  PubMed  Google Scholar 

  74. Smith, L. G., Weissman, I. L. & Heimfeld, S. Clonal analysis of hematopoietic stem-cell differentiation in vivo. Proc. Natl Acad. Sci. USA 88, 2788–2792 (1991).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  75. Yamamoto, R. et al. Large-scale clonal analysis resolves aging of the mouse hematopoietic stem cell compartment. Cell Stem Cell 22, 600–607 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Myers, L. M. et al. A functional subset of CD8+ T cells during chronic exhaustion is defined by SIRPα expression. Nat. Commun. 10, 794 (2019).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  77. Chesebro, B. et al. Characterization of mouse monoclonal antibodies specific for Friend murine leukemia virus-induced erythroleukemia cells: friend-specific and FMR-specific antigens. Virology 112, 131–144 (1981).

    Article  CAS  PubMed  Google Scholar 

  78. Marsh-Wakefield, F. M. et al. Making the most of high-dimensional cytometry data. Immunol. Cell Biol. 99, 680–696 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  79. Liechti, T. et al. An updated guide for the perplexed: cytometry in the high-dimensional era. Nat. Immunol. 22, 1190–1197 (2021).

    Article  CAS  PubMed  Google Scholar 

  80. Ashhurst, T. M. et al. Integration, exploration, and analysis of high-dimensional single-cell cytometry data using Spectre. Cytometry A 101, 237–253 (2022).

    Article  CAS  PubMed  Google Scholar 

  81. Levine, J. H. et al. Data-driven phenotypic dissection of AML reveals progenitor-like cells that correlate with prognosis. Cell 162, 184–197 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. McInnes, L., Healy, J. & Melville, J. UMAP: uniform manifold approximation and projection for dimension reduction. Preprint at arxiv.org/abs/1802.03426 (2018).

  83. Baum, C. M., Weissman, I. L., Tsukamoto, A. S., Buckle, A. M. & Peault, B. Isolation of a candidate human hematopoietic stem-cell population. Proc. Natl Acad. Sci. USA 89, 2804–2808 (1992).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  84. Yiu, Y. Y. et al. CD47 blockade leads to chemokine-dependent monocyte infiltration and loss of B cells from the splenic marginal zone. J. Immunol. 208, 1371–1377 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Brignani, S. et al. Remotely produced and axon-derived Netrin-1 instructs GABAergic neuron migration and dopaminergic substantia nigra development. Neuron 107, 684–702 (2020).

    Article  CAS  PubMed  Google Scholar 

  86. Hadi, T. et al. Macrophage-derived netrin-1 promotes abdominal aortic aneurysm formation by activating MMP3 in vascular smooth muscle cells. Nat. Commun. 9, 5022 (2018).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  87. König, K. et al. The axonal guidance receptor neogenin promotes acute inflammation. PLoS ONE 7, e32145 (2012).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  88. Li, N. et al. Upregulation of neogenin-1 by a CREB1-BAF47 complex in vascular endothelial cells is implicated in atherogenesis. Front. Cell Dev. Biol. 10, 803029 (2022).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  89. Robinson, R. A. et al. Simultaneous binding of guidance cues NET1 and RGM blocks extracellular NEO1 signaling. Cell 184, 2103–2120 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Schlegel, M. et al. Inhibition of neogenin dampens hepatic ischemia-reperfusion injury. Crit. Care Med. 42, e610–e619 (2014).

    Article  CAS  PubMed  Google Scholar 

  91. Schlegel, M. et al. Inhibition of neogenin fosters resolution of inflammation and tissue regeneration. J. Clin. Invest. 128, 4711–4726 (2019).

    Article  Google Scholar 

  92. van den Heuvel, D. M., Hellemons, A. J. & Pasterkamp, R. J. Spatiotemporal expression of repulsive guidance molecules (RGMs) and their receptor neogenin in the mouse brain. PLoS ONE 8, e55828 (2013).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  93. Keren, Z. et al. B-cell depletion reactivates B lymphopoiesis in the BM and rejuvenates the B lineage in aging. Blood 117, 3104–3112 (2011).

    Article  CAS  PubMed  Google Scholar 

  94. Säwén, P. et al. Mitotic history reveals distinct stem cell populations and their contributions to hematopoiesis. Cell Rep. 14, 2809–2818 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  95. Boivin, G. et al. Durable and controlled depletion of neutrophils in mice. Nat. Commun. 11, 2762 (2020).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  96. Chhabra, A. et al. Hematopoietic stem cell transplantation in immunocompetent hosts without radiation or chemotherapy. Sci. Transl. Med. 8, 351ra105 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  97. Iglewicz, B. & Hoaglin, D. C. How to Detect and Handle Outliers (Asq Press, 1993).

  98. Guide for the Care and Use of Laboratory Animals (National Research Council, 2010).

  99. Robertson, S. J. et al. Suppression of acute anti-friend virus CD8+ T-cell responses by coinfection with lactate dehydrogenase-elevating virus. J. Virol. 82, 408–418 (2008).

    Article  CAS  PubMed  Google Scholar 

  100. Chesebro, B., Wehrly, K. & Stimpfling, J. Host genetic control of recovery from Friend leukemia virus-induced splenomegaly: mapping of a gene within the major histocompatability complex. J. Exp. Med. 140, 1457–1467 (1974).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Lander, M. R. & Chattopadhyay, S. K. A Mus dunni cell line that lacks sequences closely related to endogenous murine leukemia viruses and can be infected by ectropic, amphotropic, xenotropic, and mink cell focus-forming viruses. J. Virol. 52, 695–698 (1984).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Robertson, M. N. et al. Production of monoclonal antibodies reactive with a denatured form of the Friend murine leukemia virus gp70 envelope protein: use in a focal infectivity assay, immunohistochemical studies, electron microscopy and western blotting. J. Virol. Methods 34, 255–271 (1991).

    Article  CAS  PubMed  Google Scholar 

  103. Horton, H. et al. Optimization and validation of an 8-color intracellular cytokine staining (ICS) assay to quantify antigen-specific T cells induced by vaccination. J. Immunol. Methods 323, 39–54 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Kumar, P. et al. HMGA2 promotes long-term engraftment and myeloerythroid differentiation of human hematopoietic stem and progenitor cells. Blood Adv. 3, 681–691 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Mahi, N. A., Najafabadi, M. F., Pilarczyk, M., Kouril, M. & Medvedovic, M. GREIN: an interactive web platform for re-analyzing GEO RNA-seq data. Sci. Rep. 9, 7580 (2019).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  106. Barrett, T. et al. NCBI GEO: archive for functional genomics data sets-update. Nucleic Acids Res. 41, D991–D995 (2013).

    Article  CAS  PubMed  Google Scholar 

  107. Edgar, R., Domrachev, M. & Lash, A. E. Gene Expression Omnibus: NCBI gene expression and hybridization array data repository. Nucleic Acids Res. 30, 207–210 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. The Tabula Muris Consortium. Single-cell transcriptomics of 20 mouse organs creates a Tabula Muris. Nature 562, 367–372 (2018).

    Article  ADS  CAS  Google Scholar 

  109. Kadoki, M. et al. Organism-level analysis of vaccination reveals networks of protection across tissues. Cell 171, 398–413 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Kleverov, M. et al. Phantasus: web-application for visual and interactive gene expression analysis. Preprint at bioRxiv https://doi.org/10.1101/2022.12.10.519861 (2022).

  111. 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).

    Article  PubMed  PubMed Central  Google Scholar 

  112. Ritchie, M. E. et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43, e47 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  113. 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).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  114. Liao, Y., Wang, J., Jaehnig, E. J., Shi, Z. & Zhang, B. WebGestalt 2019: gene set analysis toolkit with revamped UIs and APIs. Nucleic Acids Res. 47, W199–W205 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Seita, J. & Weissman, I. L. Hematopoietic stem cell: self-renewal versus differentiation. Wiley Interdiscip. Rev. Syst. Biol. Med. 2, 640–653 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Helbling, P. M. et al. Global transcriptomic profiling of the bone marrow stromal microenvironment during postnatal development, aging, and inflammation. Cell Rep. 29, 3313–3330 (2019).

    Article  CAS  PubMed  Google Scholar 

  117. Akashi, K. & Weissman, I. L. The c-kit+ maturation pathway in mouse thymic T cell development: lineages and selection. Immunity 5, 147–161 (1996).

    Article  CAS  PubMed  Google Scholar 

  118. Loder, F. et al. B cell development in the spleen takes place in discrete steps and is determined by the quality of B cell receptor-derived signals. J. Exp. Med. 190, 75–89 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Leins, H. et al. Aged murine hematopoietic stem cells drive aging-associated immune remodeling. Blood 132, 565–576 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Goardon, N. et al. Coexistence of LMPP-like and GMP-like leukemia stem cells in acute myeloid leukemia. Cancer Cell 19, 138–152 (2011).

    Article  CAS  PubMed  Google Scholar 

  121. Manz, M. G., Miyamoto, T., Akashi, K. & Weissman, I. L. Prospective isolation of human clonogenic common myeloid progenitors. Proc. Natl Acad. Sci. USA 99, 11872–11877 (2002).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank the members of the Weissman and Hasenkrug laboratories for advice and discussions; A. McCarty, T. Naik, L. Quinn and T. Raveh for technical and logistical support; A. Banuelos, G. Blacker, B. George, G. Gulati, J. Liu, R. Sinha, M. Tal, N. Womack and Y. Yiu for general help, advice and experimental support; C. Carswell-Crumpton, C. Pan, J. Pasillas and the staff at the Stanford Institute for Stem Cell Biology and Regenerative Medicine FACS Core for flow cytometry assistance; H. Maecker and I. Herschmann of the Stanford Human Immune Monitoring Center (HIMC) for assistance with immunoassays; and the members of the Rocky Mountain Veterinary Branch (RMVB), especially T. Wiediger, for excellent care of the aged mice. This work was partially funded by the Intramural Research Program of the National Institute of Allergy and Infectious Diseases, National Institutes of Health, USA; the NIH/NCI Outstanding Investigator Award (R35CA220434 to I.L.W.); the NIH NIDDK (R01DK115600 to I.L.W.); the NIH NIAID (R01AI143889 to I.L.W.); and the Virginia and D.K. Ludwig Fund for Cancer Research (to I.L.W.). J.B.R. was supported by the Stanford Radiation Oncology Kaplan Research Fellowship, the RSNA Resident/Fellow Research Grant, and the Stanford Cancer Institute Fellowship Award and the Ellie Guardino Research Fund. This work was supported by the Stanford Cancer Institute, an NCI-designated Comprehensive Cancer Center. J.J.N. was supported by Stanford University Medical Scientist Training Program grant T32-GM007365 and T32-GM145402. E.D. was supported by grants from NIH NIDDK (5T32DK098132-09 and 1TL1DK139565-01). The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript. Some illustrations were created using BioRender.

Author information

Authors and Affiliations

Authors

Contributions

J.B.R. and L.M.M. contributed equally to this work and they both have the right to be listed first in bibliographic documents. J.B.R. and L.M.M. conceived and performed experiments, analysed and interpreted all the data, and wrote the paper. J.J.N., M.M.C., A.B.C., R.J.M. and E.D. performed experiments and analysed data. L.M.M., M.M.C., A.B.C. and R.J.M. performed the Friend virus experiments. J.B.R. and E.D. designed and performed the RNA-seq and transplant experiments. I.L.W. and K.J.H. conceived experiments, supervised the research, interpreted results and wrote the paper. All of the authors reviewed, edited and approved the manuscript.

Corresponding authors

Correspondence to Kim J. Hasenkrug or Irving L. Weissman.

Ethics declarations

Competing interests

I.L.W. is listed as an inventor on patents related to CD47 licensed to Gilead Sciences, but has no financial interests in Gilead; he is also a co-founder and equity holder of Bitterroot Bio, PHeast and 48 Bio; he is on the scientific advisory board of Appia. J.B.R. is a co-founder and equity holder of 48 Bio. I.L.W., K.J.H., J.B.R., L.M.M. and J.N.N. are listed as co-inventors on a pending patent application related to this work. The other authors declare no competing interests.

Peer review

Peer review information

Nature thanks Jennifer Trowbridge and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data figures and tables

Extended Data Fig. 1 Expression of my-HSC markers in HSPCs, mature cells, and tissues.

a-l, Expression of my-HSC candidate markers, Slamf1 (CD150) (a), Neo1 (NEO1) (b), Itga2b (CD41) (c), Selp (CD62p) (d), Cd38 (CD38) (e), Itgb3 (CD61) (f), Itgav (CD51) (g), Procr (CD201) (h), Tie2 (i), Esam (j), Eng (CD105) (k), Cd9 (CD9) (l), in HSC and HSPCs in normal mouse BM (top panels), and in young versus old bone marrow (bottom panels). Data and images from al generated and obtained directly from Gene Expression Commons31; scale bars represent log2 Signal Intensity (top) and Gene Expression Activity (bottom) as defined by Gene Expression Commons31. m, Heatmap of relative RNA expression for CD150 (Slamf1), NEO1 (Neo1), CD62p (Selp), CD41 (Itga2b), CD38 (Cd38), CD51 (Itgav), and CD61 (Itgb3) in HSCs, MPPs, Progenitors, Myeloid, and Lymphoid cells. Processed data for 23 cell types were obtained directly from Gulati19 Supplementary Table 1. Fold-enrichment = [(average percentile of HSCs)/(average percentile of all other cell types)+100], as described in this publication. no, RNA expression of CD150 (Slamf1), NEO1 (Neo1), CD62p (Selp), and CD41 (Itga2b) in bulk mouse tissues from two independent datasets: Tabula Muris108 (n) or Kadoki109 (o). For no, Values are z-score normalized for each gene across all tissues.

Source Data

Extended Data Fig. 2 Gating strategy for total HSCs, my-HSCs, bal-HSCs, and HPCs.

a, Schematic to identify and validate my-HSC cell-surface antigens. The diagram was created using BioRender. b, Representative flow-cytometry gating of mouse BM to identify total HSC (LincKIT+Sca1+FLT3CD34CD150+), my-HSC (LincKIT+Sca1+FLT3CD34CD150High), bal-HSC (LincKIT+Sca1+FLT3CD34CD150Low), MPPs115 [MPPa (LincKIT+Sca1+FLT3CD34+CD150+), MPPb (LincKIT+Sca1+FLT3CD34+CD150), MPPc (LincKIT+Sca1+FLT3+CD34+CD150)], OPP (LincKIT+Sca1), CMP&GMP (LincKIT+Sca1CD34+CD41), MkP (LincKIT+Sca1CD34+CD41+), MEP (LincKIT+Sca1CD34CD41+), CLP (LincKITLoSca1LoIL7Ra+FLT3+). Panels are after excluding dead cells, doublets, and lineage-positive (CD3+, or Ly-6G+/C+, or CD11b+, or CD45R+, or Ter-119+) cells. Used for Fig. 1b–k, Fig. 2a–f, Fig. 3b–d, Fig. 4a, Extended Data Fig. 2c–k, Extended Data Fig. 3a–l, Extended Data Fig. 4a–s, Extended Data Fig. 5c–s, Extended Data Fig. 6a Extended Data Fig. 8c–h. Illustration of Hematopoietic Stem and Progenitor Cell (HSPC) Tree Analysis. CMP is combined CMP&GMP. Gate to define my-HSC vs. bal-HSC was set as described previously5. cf, Relative expression of CD41 (c), CD38 (d), CD51 (e), CD61 (f), on HSC and HSPCs. MFI values for each marker were obtained for each population and normalized from 0–1 based on the lowest to highest expression. gh, Relative cell-surface levels (g) and percent-positive cells (h) for CD150, NEO1, CD62p, CD41, CD38, CD51, and CD61, on lineage-positive high and low cells, total HSCs, and HPCs in the BM. For cell-surface levels (g), MFI values for each marker were obtained for each population and normalized from 0–100 based on the lowest to highest expression. i, Percentage of total HSCs that are CD41+ (y-axis) vs. mouse age in weeks (x-axis); n = 21 mice. j, Mouse age (x-axis) vs. the frequency of total HSCs (my-HSC+bal-HSC) as a percentage of live cells in the (i) total BM (left y-axis, red) or (ii) cKIT-enriched BM (right y-axis, blue) in untreated mice; n = 13 mice. k, Percent-positive of my-HSCs vs. bal-HSCs for CD47 (k, top) using independent anti-CD47 clones (MIAP301, left; MIAP410, right), and for cKIT (k, bottom) using independent anti-cKIT clones (ACK2, left; 2B8, right). Mouse ages: 4–6 months (bi, k), 3–23 months (j). For ak, BM was cKIT-enriched prior to analysis. For j, total BM (non cKIT-enriched) was also examined. p-values and R values calculated with one-tailed Pearson correlation coefficient (ij). n represents independent mice.

Source Data

Extended Data Fig. 3 Anti-CD150 non-masking antibodies and FACS gating to isolate HSCs.

a, Schematic to identify anti-CD150 antibodies not masked by anti-CD150 antibody clone 1 (TC15), used in panels bd; BM cells were incubated with saturating concentrations of unlabelled anti-CD150 antibody clone 1 (TC15) and then stained with PE anti-CD150 clones 2, 3, 4 (Q38, 9D1, mShad150). bd, Saturating concentrations of unlabelled anti-CD150 antibody clone TC15 blocks staining with PE anti-CD150 clone 4 (mShad150) (d), but does not block staining of PE anti-CD150 clones 2, 3 (Q38, 9D1) (bc). e, Schematic to determine if anti-CD150 clones 2, 3, 4 (Q38, 9D1, mShad150) identify the same population as anti-CD150 clone 1 (TC15); used in panels fh; BM cells were incubated with PECy-7 anti-CD150 antibody clone 1 (TC15) and with PE anti-CD150 clones 2, 3, 4 (Q38, 9D1, mShad150). fh, Co-staining with anti-CD150 clones 2, 3 (Q38, 9D1) identifies the same population as anti-CD150 antibody clone TC15 (fg). Co-staining with anti-CD150 clone 4 (mShad150) and anti-CD150 antibody clone 1 (TC15) is mutually blocked (h). i, Schematic to identify anti-CD150 antibodies that are not blocked by anti-CD150 clone mShad150, used in panel j; BM cells are incubated with saturating concentrations of unlabelled anti-CD150 antibody clone mShad150 and then stained with PE anti-CD150 clone 2 (Q38). j, Saturating concentrations of unlabelled anti-CD150 antibody clone mShad150 does not block staining of PE anti-CD150 clone 2 (Q38) (j). k, Schematic to determine if co-staining with anti-CD150 clone 2 (Q38) identifies the same population as anti-CD150 antibody clone mShad150; used in panel l; BM cells are incubated with PECy-7 anti-CD150 antibody clone mShad150 and with PE anti-CD150 clone 2 (Q38). l, Co-staining with anti-CD150 clones 2 (Q38) identifies the same population as anti-CD150 antibody clone mShad150 (l). mn, Representative FACS gating of mouse BM to sort total HSC (LincKIT+Sca1+FLT3CD34CD150+) from aged (11 months) control mice (Aged, A) or aged (11 months) mice with my-HSC depletion (Aged+Conditioning, A + C), used to sort HSCs for RNA-sequencing experiment presented in Fig. 2g, h, Extended Data Fig. 4x, and used to sort HSCs for transplant experiments presented in Fig. 2i–k, Extended Data Fig. 4t–v. My-HSC depletion was performed with anti-NEO1+anti-CD62p+anti-cKIT+CD47 and cells were collected at day 9 post-treatment; n = 3 mice (A), n = 3 mice (A + C). The diagrams (a, e, i, k) were created using BioRender. Mouse ages approximately: 3 months (ah), 5–8 months (il), 11 months (mn). For (an), BM was cKIT-enriched prior to FACS analysis or sorting. n represents independent mice.

Extended Data Fig. 4 Antibody-mediated depletion of my-HSCs in vivo.

ad, Frequency (% live) (a, c) or absolute number (b, d) of my-HSCs and bal-HSCs (ab) or of NEO1+ HSCs and NEO1 HSCs (cd) after anti-CD150 conditioning (anti-CD150, anti-CD150+anti-CD47, or anti-CD150+anti-CD47+anti-cKIT); n = 3 mice per group. e, Total HSCs (my-HSCs+bal-HSCs) as percent-live in mice receiving anti-CD47; n = 5 mice per group; P = 0.3637. f, Percentage of total HSCs that are my-HSCs in mice receiving anti-CD47; n = 5 mice per group; P = 0.6628. g, Total HSCs (my-HSCs+bal-HSCs) as a percent-live, in mice receiving anti-CD47+anti-cKIT; n = 4 mice per group; P = 0.0973. h, Percentage of total HSCs that are my-HSCs in mice receiving anti-CD47+anti-cKIT; n = 4 mice per group; P = 0.2805. i, Percentage of total HSCs that are NEO1+ HSCs in mice receiving anti-CD150 (IgG2a)+anti-CD47+anti-cKIT (e.g., protocol CD150v2); n = 4 mice per group; *P = 0.0353. jn, Frequency (% live) of CLPs (j), IL7Ra+ cells (k), CMPs&GMPs (l), MkPs (m), and MEPs (n), after anti-CD150, anti-CD62p, or anti-NEO1 conditioning protocols. Values relative to mean of untreated control mice and log2-transformed; n = 3 mice per group (NEO1v1, NEO1v2, CD150v1); n = 4 mice per group (CD62p, CD150v2). o, Ratio of frequency (% live) for Lymphoid to Myeloid Progenitors (CLP)/(CMP&GMP), after anti-CD150, anti-CD62p, or anti-NEO1 antibody protocols. Values relative to mean of untreated control mice and log2-transformed; n = 3 mice per group (NEO1v1, NEO1v2, CD150v1); n = 4 mice per group (CD62p, CD150v2). p, Frequency (% live) of my-HSCs, bal-HSCs, NEO1+ HSCs, and NEO1- HSCs after treatment with anti-CD150, anti-CD62p, or anti-NEO1 protocols; n = 3 mice per group (anti-CD150v1); n = 4 mice per group (anti-CD62p); n = 3 control mice and n = 6 treated mice (anti-NEO1, combined 90 ug & 200 ug protocols). Values relative to mean of control mice and log2-transformed. For jp, values for treated mice (filled circles); values for control mice (unfilled circles). q, Correlation of fraction of live cells for my-HSCs vs. NEO1+ HSCs (left), and bal-HSCs vs. NEO1- HSCs (right), of control mice and mice receiving anti-CD150, anti-CD62p, or anti-NEO1 protocols, in cKIT-enriched BM; n = 3 mice (anti-CD150v1); n = 4 mice (anti-CD62p); n = 3 control mice and n = 6 treated mice (anti-NEO1, combined 90 ug & 200 ug protocols). r, Comparison of the absolute number of cells in total (non-cKIT-enriched) BM (y-axis) vs. frequency of cells as a percent-live (in cKIT-enriched) BM (x-axis), for my-HSCs, bal-HSCs, CLP, CMP&GMP, and MkP in control mice and mice receiving anti-CD150, anti-CD150+anti-CD47, or anti-CD150+anti-CD47+anti-cKIT; n = 3 mice per condition. s, Comparison of the frequency of cells as a fraction of total (non-cKIT-enriched) BM (x-axis) vs. the frequency of cells as a fraction of cKIT-enriched BM (y-axis), for cell populations in control mice and mice receiving anti-CD150, anti-CD150+anti-CD47, or anti-CD150+anti-CD47+anti-cKIT; n = 3 mice per condition. Data for qs log10-transformed. tv, HSC lineage potential 8-weeks after transplantation of 100 total HSCs FACS-sorted from CD45.2 aged mice without (A) or with my-HSC depletion (A + C) into CD45.1 recipients. Donor Myeloid to Lymphoid Ratio (t, *P = 0.0275) and Percent-Chimerism Ratio (u, *P = 0.0340). For each recipient (y-axis), % donor cells (bottom x-axis) that are Myeloid (red bars) or Lymphoid (blue bars) and the Myeloid/Lymphoid log2-ratio (red circles) (top x-axis) (v); n = 13 recipient mice (A), n = 14 recipient mice (A + C). w, Donor Myeloid/Lymphoid Ratio 16-weeks after transplantation of 100 total HSCs FACS-sorted from CD45.2 aged mice without (A) or with my-HSC depletion (A + C) into CD45.1 recipients; n = 12 recipient mice (A), n = 14 recipient mice (A + C); *P = 0.0232. x, Top 200 differentially expressed genes ranked by p-value for RNA-seq comparison between (A) vs. (A + C), based on Fragments Per Kilobase of transcript per Million mapped reads (FPKM); log2-transformed. Heatmap generated using Phantasus110 (v1.21.5) with FPKM values as input and Limma112 to define differentially expressed genes; n = 3 mice (A), n = 3 mice (A + C). Mouse ages approximately: 6-7 months (ad, rs), 7–9 months (gi), 5–9 months (jq), 11 months (donors, tw), 2 months (recipients, v). For (a, c, gs), BM was cKIT-enriched prior to analysis. For (b, d, ef, rs), total BM (non cKIT-enriched) was examined. P-values obtained by ordinary one-way ANOVA followed by one-tailed Dunnett’s multiple comparisons test with non-treated as control (ad), unpaired parametric one-tailed t-test (i-k, p, tu), unpaired non-parametric one-tailed t-test (w), or unpaired parametric two-tailed t-test (eh, lo). p-values and R values calculated with one-tailed Pearson correlation coefficient (qs). CD150v1 is rat IgG2b anti-CD150 protocol; CD150v2 is rat IgG2a anti-CD150 protocol; NEO1v2 includes mouse IgG2a anti-goat; α, anti-; ns, not significant. Bars indicate mean +/− s.e.m. n represents independent mice; *P < 0.05, **P < 0.005, ***P < 0.0005; Exact P-values provided as source data.

Source Data

Extended Data Fig. 5 Optimization of NEO1 depletion protocol in vitro and in vivo.

ab, Strategy to restore balanced lineage output by depleting my-HSCs. ch, Anti-NEO1 antibody saturation curve (c) determined from in vitro antibody dilution series (dh). i, Schematic of in vivo saturation experiments with anti-NEO1 antibody; used in panels jm. jm, Dose-dependent relationship between anti-NEO1 antibody dose (0 ug, 30 ug, 90 ug, 200 ug), when combined with anti-CD47 and anti-cKIT, on the relative depletion of NEO1+ HSCs (j), and increase in CLPs (k). Optimal concentration in yellow; n = 3 mice per group. l, The increase in NEO1 HSCs after anti-NEO1 dose escalation (0 ug, 30 ug, 90 ug, 200 ug) is correlated with the increase in CLPs. m, Impact on the ratio of Bal-HSCs/My-HSCs (black) and NEO1 HSCs/NEO1+ HSCs (blue) as a percentage of live cells after anti-NEO1 antibody dose-escalation (0 ug, 30 ug, 90 ug, 200 ug), when combined with anti-CD47 and anti-cKIT; n = 3 per group. Values relative to mean value of control (0 ug) mice and log2-transformed. n, Schematic of double-antibody strategy to target NEO1, whereby mouse IgG2a or IgG2b monoclonal anti-goat antibodies are administered 24 h after goat anti-NEO193,94. op, Schematic (o) to demonstrate that saturating concentrations of mouse IgG2a or IgG2b anti-goat do not reduce ability of donkey anti-goat AF488 detect goat anti-NEO1 antibody (p). qs, Schematic (q) of experiment demonstrating that mouse anti-goat IgG2a A555 (r) and IgG2b PE (s) antibodies identify the same population as donkey anti-goat AF488 by flow-cytometry. The diagrams (a, b, i, n, o, q) were created using BioRender. Mouse ages approximately: 5–7 months (ch, ns), 6–9 months (im). For (cs), BM was cKIT-enriched prior to analysis. For correlation p-values, one-tailed Pearson correlation coefficient (Rp), and one-tailed Spearman correlation coefficient (Rs) were calculated (l). p-values obtained by ordinary one-way ANOVA followed by one-tailed Dunnett’s multiple comparisons test with 0ug condition as control (m). MaG, mouse-anti goat; DaG, donkey anti-goat. Bars indicate mean +/− s.e.m. n represents independent mice; *P < 0.05, **P < 0.005, ***P < 0.0005; Exact P-values provided as source data.

Source Data

Extended Data Fig. 6 My-HSC depletion restores features of a youthful immune system.

a, Ratio of frequency of my-HSCs to bal-HSCs (% live) in aged (A), or aged+conditioning mice (A + C) after 8-weeks; n = 4 (A), n = 9 (A + C); P = 0.0346. bd, Statistical significance (y-axis, -log10p) vs. fold-change (x-axis, log2-FC) of plasma protein levels at week-8 for (A)/(Y) (b), (A)/(A + C) (c), or (A)/(Y & A + C) (d) comparisons. Grey bar (y = 1–1.3); values above grey bar p < 0.05; n = 6 (Y, A), n = 5 (A + C). e, Overlap of top 17% of plasma proteins, ranked by statistical significance, increased in (A)/(Y) and decreased in (A + C)/(A). f, Estimated plasma concentration of CXCL5116 at week-8; n = 6 (Y, A), n = 5 (A + C). g, Relative plasma abundance of inflammatory proteins at week-8; n = 6 (Y, A), n = 5 (A + C). Values relative to the mean for (Y) and log2-transformed. hi, Frequency of CD11b+Ly6G/C+ (h) and CD11b+SIRPa+ (i) mature myeloid cells in the blood approximately 1-week after antibody-conditioning; n = 3 (A), n = 4 (A, A + C). jk, Frequency of mature B cells (B220+CD19+CD43CD93IgM+IgD+) (j), and progenitor B cells (B220+CD19 B cells) (k), in the BM approximately 1-week after antibody-conditioning; n = 5 (A), n = 6 (A, A + C). Thymus weight (l) and frequency of thymic subsets 1–8 as defined by Akashi & Weissman117, as % of total CD45+ cells in the thymus (n), 8-weeks after antibody-conditioning; representative FACS (m); n = 3 (Y, A), n = 9 (A + C). Populations enriched for transitional intermediate cells (areas 3&4)117 highlighted in box (n). Mouse ages: Y (3–6 months); A & A + C (18–24 months). For (a), BM was cKIT-enriched prior to analysis. p-values obtained by unpaired parametric one-tailed t-test (a), ordinary one-way ANOVA followed by one-tailed Dunnett’s multiple comparisons test using Aged as control (f), one-way ANOVA followed by Holm multiple comparisons test (bd, g), ordinary one-way ANOVA followed by two-tailed Dunnett’s multiple comparisons test using Aged as control (hi, l, n), or Brown-Forsythe and Welch ANOVA tests followed by Dunnett’s T3 multiple comparisons test using Aged as control (jk). Bars indicate mean +/− s.e.m. n represents independent mice; *P < 0.05, **P < 0.005, ***P < 0.0005; Exact P-values provided as source data.

Source Data

Extended Data Fig. 7 My-HSC depletion increases naïve T cells and B cells in aged mice.

ac, Absolute numbers of (a) naïve (CD44CD62L+); *P = 0.0430, (b) central memory (CM: CD44+CD62L+); *P = 0.0177, or (c) effector memory (EM: CD44+CD62L); P = 0.576, T cells (CD4 & CD8), per mL of blood, approximately 8-weeks post-treatment. Values log2-transformed; n = 9 (A), n = 14 (A + C), mice pooled from 2 independent experiments. d, Absolute numbers of mature B cells (IgM+IgD+) per mL of blood, approximately 8-weeks post-treatment; *P = 0.0498. Values log2-transformed; n = 6 (A), n = 5 (A + C). e, Absolute numbers of CD45+ cells per mL of blood, approximately 8-weeks post-treatment. Values log2-transformed; n = 3 (A), n = 9 (A + C); P = 0.069. fg, Percentage of CM (f) and EM (g) subsets per total T cells (CD4 & CD8), 8-weeks post-treatment; n = 6 (Y, A), n = 5 (A + C). hj, Frequency relative to Aged mice of T cell (CD4 & CD8) subsets 8-weeks after antibody treatment (h). Naïve, CM, and EM subsets were defined by 12-marker cluster-based analysis (ij); n = 6 (Y, A), n = 5 (A + C). Values relative to mean for Aged control mice and log2-transformed. Mouse ages: Y (3–6 months); A & A + C (18–24 months). p-values obtained by unpaired parametric one-tailed t-test (a, d), unpaired parametric two-tailed t-test (bc, e), or ordinary one-way ANOVA followed by two-tailed (fg) or one-tailed (h) Dunnett’s multiple comparisons test using Aged as control. Bars indicate mean +/− s.e.m. n represents independent mice; *P < 0.05, **P < 0.005, ***P < 0.0005; Exact P-values provided as source data.

Source Data

Extended Data Fig. 8 Flow-cytometry gating strategy for T cells, B cells, and myeloid cells.

ac, Gating strategy to identify: (b) naïve (CD44CD62L+), central memory (CD44+CD62L+), and effector memory (CD44+CD62L) T cells (combined CD4 & CD8), or (c) CD4 T cells that are PD1+CD62L or PD1CD62L+41 in the blood, used for Fig. 3e,g, Extended Data Fig. 7a–c, Extended Data Fig. 7f–g. df, Gating strategy to identify: (e) mature B cells (CD19+B220+IgM+IgD+)118, or (f) Aged B Cells ABCs119 (CD19+IgM+CD93CD43 CD21/CD35CD23)42 in the blood, used for Fig. 3f,h, Extended Data Fig. 7d. gi, Gating strategy to identify (h) progenitor B cells (B220+CD19), or (i) mature B cells (B220+CD19+CD43CD93IgM+IgD+) in the bone-marrow, used for Extended Data Fig. 6j,k. jl, Gating strategy to identify (k) CD11b+Ly6G/C+ myeloid cells, or (l) CD11b+SIRPa+ myeloid cells in the blood, used for Extended Data Fig. 6h,i.

Extended Data Fig. 9 Antibody-conditioning enhances functional immunity to infection.

a, Schematic of model to determine the impact of antibody-conditioning on functional immunity. The diagram was created using BioRender. b, Gating to identify Ter119+ cells (Ter119+CD19CD3CD45+/lo) and antigen-infected cells (Ag34+Ter119+) in mouse spleens, used for Extended Data Fig. 9i,j. ch, My-HSC and NEO1+ HSC absolute numbers in total BM (c, e), or as percentage of total HSCs (d, f), with correlations (h), 10-weeks after anti-NEO1v2 conditioning. Representative FACS on total HSCs (g). A and A + C mice received vaccination at Week 8; n = 2 (Y), n = 7 (A), n = 10 (A + C). i, Total number of Ter119+ cells per mouse spleen in mice that were Naïve, Infected, or Vaccinated & Infected with FV. Representative FACS histograms for Ter119 expression, gated on all single cells. Each row represents an independent mouse. n = 9 (Y, naïve), n = 13 (Y, FV infected), n = 13 (Y, vaccinated & FV infected), n = 6 (A, naïve), n = 10 (A, FV infected), n = 8 (A, vaccinated & FV infected), n = 5 (A + C, naïve), n = 9 (A + C, vaccinated & FV infected). Data log10-transformed. j, Total number of antigen-infected (Ag34+Ter119+) cells per spleen in mice that were: Infected, or Vaccinated & Infected with FV. Representative FACS histograms for Ag34 expression, gated on Ter119+ cells. Each row represents an independent mouse; n = 13 (Y, FV infected), n = 13 (Y, vaccinated & FV infected), n = 10 (A, FV infected), n = 8 (A, vaccinated & FV infected), n = 9 (A + C, vaccinated & FV infected). Data log10(x + 1)-transformed. k, Correlation of Infectious virus levels (Infectious Centres; x-axis) vs. Dextramer+CD44+ cells (Percent of splenic CD8 + T cells; y-axis) in Vaccinated & FV-infected mice; n = 8 (A, vaccinated & FV-infected), n = 9 (A + C, vaccinated & FV-infected). Data log10-transformed. For ik, data from experiments using the anti-NEO1v1 protocol (open circles) or the anti-NEO1v2 protocol (closed circles) were combined. Mouse ages: Y (3–6 months); A & A + C (21-22 months). For (cf), BM was cKIT-enriched prior to analysis. For (c, e, h), total BM (non-cKIT-enriched) was also analysed to calculate total numbers of cells. p-values obtained by one-tailed unpaired parametric t-test (cf), two-tailed Pearson correlation coefficient (h, k), or ordinary one-way ANOVA followed by Tukey’s multiple comparisons test (ij). NEO1v2 protocol is NEO1v1 protocol (anti-NEO1+ anti-CD47+anti-cKIT)+mouse (IgG2a) anti-goat. Inf., FV infected without vaccination; Vacc. & Inf., FV infected with vaccination, Vacc. & Inf., FV infected with vaccination; LOD, limit of detection; n.s., not significant. n represents independent mice. Bars indicate mean +/− s.e.m. (cf) or bars depict median (ij). *P < 0.05, **P < 0.005, ***P < 0.0005; Exact P-values provided as source data.

Source Data

Extended Data Fig. 10 Mouse my-HSC markers are enriched in aged human HSCs.

a, Relative mRNA expression of CD62p (Selp, P = 0.0243), CD41 (Itga2b, P = 0.0001), CD61 (Itgb3, P = 0.0135), CD150 (Slamf1, P = 0.179), and NEO1 (Neo1, P = 0.433) in human HSCs isolated from young (age 20–26) and old (age >70) donors; data obtained from Nilsson54, GSE69408. b, Relative mRNA expression of CD62p (Selp, P = 0.0001), CD41 (Itga2b, P = 0.0027), CD61 (Itgb3, P = 0.0116), and NEO1 (Neo1, P = 0.274) in human HSCs isolated from young (age 18–30) or old (age 65–75) donors; data obtained from Adelman53 (GSE104406). ce, Correlation of relative mRNA expression of CD62p (c), CD41 (d), and CD61 (e) in human HSCs as compared to donor age; data obtained from Adelman53 (GSE104406). For ae, values relative to mean of young samples. f, Heatmap depicting expression of candidate markers in independent datasets of human: HMGA2+ vs. HMGA2 CD34+ cells (e104), MPN (f56) or MDS (g57) vs. normal HSCs, and Pre-AML vs. normal HSCs (h58). gh, Representative FACS to identify human HSCs (LinCD34+CD38CD45RACD90+)59, MPPs (LinCD34+CD38CD45RACD90)59, LMPPs (LinCD34+CD38CD45RA+)120, CMPs & MEPs (LinCD34+CD38+CD45RA)121, and GMPs (LinCD34+CD38+CD45RA+)121 in normal human BM59, used for Fig. 5f–i, Extended Data Fig. 10i–s. Samples are post CD34+-enrichment. im, FACS of human HSCs depicting fluorescence-minus-one (FMO) control (i), anti-CD62p clone AK4 (j), anti-CD62p clone Psel.KO2.3 (k), anti-CD62p clone AC1.2 (l), and anti-CD150 (m); representative for n = 3 donors (j); n = 2 donors (km). n, Illustration of human Hematopoietic Stem and Progenitor Cell (HSPC) Tree Analysis (n). os, Relative expression of CD62p (o), CD150 (p), ESAM (q), and CD166 (r), on human HSPCs, with summary (s), showing percent positive HSCs, and normalized MFI for each marker on HSPCs. For os, FACS median fluorescent intensity (MFI) values for each marker were obtained for each population, divided by the MFI for the FMO control, and then normalized from 0–100 based on the lowest to highest expression. Red colour scale corresponds to normalized MFI values. Blue, purple, maroon scale corresponds to bins for HSC positivity (20–50%, 51–70%, and 71–100). t, Model to rejuvenate aged immune systems by depleting myeloid-biased hematopoietic stem cells. The diagram was created using BioRender. p-values obtained by unpaired parametric one-tailed t-test (ab), or p-values and R values calculated with one-tailed Pearson correlation coefficient (ce). MDS, myelodysplastic syndrome; MPN, myeloproliferative neoplasms. Bars indicate mean +/− s.e.m. *P < 0.05, **P < 0.005, ***P < 0.0005; Exact P-values provided as source data.

Source Data

Supplementary information

Reporting Summary

Peer Review File

Supplementary Table 1

Genes differentially expressed between HSCs isolated from control versus antibody-treated mice.

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

Ross, J.B., Myers, L.M., Noh, J.J. et al. Depleting myeloid-biased haematopoietic stem cells rejuvenates aged immunity. Nature 628, 162–170 (2024). https://doi.org/10.1038/s41586-024-07238-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-024-07238-x

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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