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:

Adiponectin receptors sustain haematopoietic stem cells throughout adulthood by protecting them from inflammation

Abstract

How are haematopoietic stem cells (HSCs) protected from inflammation, which increases with age and can deplete HSCs? Adiponectin, an anti-inflammatory factor that is not required for HSC function or haematopoiesis, promotes stem/progenitor cell proliferation after bacterial infection and myeloablation. Adiponectin binds two receptors, AdipoR1 and AdipoR2, which have ceramidase activity that increases upon adiponectin binding. Here we found that adiponectin receptors are non-cell-autonomously required in haematopoietic cells to promote HSC quiescence and self-renewal. Adiponectin receptor signalling suppresses inflammatory cytokine expression by myeloid cells and T cells, including interferon-γ and tumour necrosis factor. Without adiponectin receptors, the levels of these factors increase, chronically activating HSCs, reducing their self-renewal potential and depleting them during ageing. Pathogen infection accelerates this loss of HSC self-renewal potential. Blocking interferon-γ or tumour necrosis factor signalling partially rescues these effects. Adiponectin receptors are thus required in immune cells to sustain HSC quiescence and to prevent premature HSC depletion by reducing inflammation.

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: Adiponectin receptors promote HSC quiescence.
Fig. 2: Adiponectin receptors are required to sustain HSC function.
Fig. 3: Adiponectin receptors suppress the expression of inflammatory cytokines in bone marrow lymphocytes.
Fig. 4: Adiponectin receptors promote HSC function by reducing IFNγ signalling.
Fig. 5: Adiponectin receptors promote HSC function by reducing TNF signalling.
Fig. 6: Adiponectin receptors are required to sustain HSCs during ageing.

Similar content being viewed by others

Data availability

RNA-seq data associated with this paper are accessible in the NCBI Sequence Read Archive, BioProjects PRJNA699097 (associated with Fig. 1a–c), PRJNA729963 (associated with Fig. 3a) and PRJNA765672 (associated with Extended Data Fig. 4f). All other data supporting the findings of this study are available from the corresponding author on reasonable request. Source data are provided with this paper.

References

  1. Berg, A. H., Combs, T. P., Du, X., Brownlee, M. & Scherer, P. E. The adipocyte-secreted protein Acrp30 enhances hepatic insulin action. Nat. Med. 7, 947–953 (2001).

    Article  CAS  PubMed  Google Scholar 

  2. Yamauchi, T. et al. The fat-derived hormone adiponectin reverses insulin resistance associated with both lipoatrophy and obesity. Nat. Med. 7, 941–946 (2001).

    Article  CAS  PubMed  Google Scholar 

  3. Fruebis, J. et al. Proteolytic cleavage product of 30-kDa adipocyte complement-related protein increases fatty acid oxidation in muscle and causes weight loss in mice. Proc. Natl Acad. Sci. USA 98, 2005–2010 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Scherer, P. E., Williams, S., Fogliano, M., Baldini, G. & Lodish, H. F. A novel serum protein similar to C1q, produced exclusively in adipocytes. J. Biol. Chem. 270, 26746–26749 (1995).

    Article  CAS  PubMed  Google Scholar 

  5. Maeda, K. et al. cDNA cloning and expression of a novel adipose specific collagen-like factor, apM1 (AdiPose Most abundant Gene transcript 1). Biochem. Biophys. Res. Commun. 221, 286–289 (1996).

    Article  CAS  PubMed  Google Scholar 

  6. Nakano, Y., Tobe, T., Choi-Miura, N. H., Mazda, T. & Tomita, M. Isolation and characterization of GBP28, a novel gelatin-binding protein purified from human plasma. J. Biochem. 120, 803–812 (1996).

    Article  CAS  PubMed  Google Scholar 

  7. Hu, E., Liang, P. & Spiegelman, B. M. AdipoQ is a novel adipose-specific gene dysregulated in obesity. J. Biol. Chem. 271, 10697–10703 (1996).

    Article  CAS  PubMed  Google Scholar 

  8. Masamoto, Y. et al. Adiponectin enhances quiescence exit of murine hematopoietic stem cells and hematopoietic recovery through mTORC1 potentiation. Stem Cells 35, 1835–1848 (2017).

    Article  CAS  PubMed  Google Scholar 

  9. DiMascio, L. et al. Identification of adiponectin as a novel hemopoietic stem cell growth factor. J. Immunol. 178, 3511–3520 (2007).

    Article  CAS  PubMed  Google Scholar 

  10. Tikhonova, A. N. et al. The bone marrow microenvironment at single-cell resolution. Nature 569, 222–228 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Cawthorn, W. P. et al. Bone marrow adipose tissue is an endocrine organ that contributes to increased circulating adiponectin during caloric restriction. Cell Metab. 20, 368–375 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Ohashi, K. et al. Adiponectin promotes macrophage polarization toward an anti-inflammatory phenotype. J. Biol. Chem. 285, 6153–6160 (2010).

    Article  CAS  PubMed  Google Scholar 

  13. Yamaguchi, N. et al. Adiponectin inhibits Toll-like receptor family-induced signaling. FEBS Lett. 579, 6821–6826 (2005).

    Article  CAS  PubMed  Google Scholar 

  14. Wilk, S. et al. Adiponectin modulates NK-cell function. Eur. J. Immunol. 43, 1024–1033 (2013).

    Article  CAS  PubMed  Google Scholar 

  15. Surendar, J. et al. Adiponectin limits IFN-γ and IL-17 producing CD4 T cells in obesity by restraining cell intrinsic glycolysis. Front Immunol. 10, 2555 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Maeda, N. et al. Diet-induced insulin resistance in mice lacking adiponectin/ACRP30. Nat. Med. 8, 731–737 (2002).

    Article  CAS  PubMed  Google Scholar 

  17. Masamoto, Y. et al. Adiponectin enhances antibacterial activity of hematopoietic cells by suppressing bone marrow inflammation. Immunity 44, 1422–1433 (2016).

    Article  CAS  PubMed  Google Scholar 

  18. Holland, W. L. et al. Receptor-mediated activation of ceramidase activity initiates the pleiotropic actions of adiponectin. Nat. Med. 17, 55–63 (2011).

    Article  CAS  PubMed  Google Scholar 

  19. Vasiliauskaite-Brooks, I. et al. Structural insights into adiponectin receptors suggest ceramidase activity. Nature 544, 120–123 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Yamauchi, T. et al. Targeted disruption of AdipoR1 and AdipoR2 causes abrogation of adiponectin binding and metabolic actions. Nat. Med. 13, 332–339 (2007).

    Article  CAS  PubMed  Google Scholar 

  21. Hug, C. et al. T-cadherin is a receptor for hexameric and high-molecular-weight forms of Acrp30/adiponectin. Proc. Natl Acad. Sci. USA 101, 10308–10313 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Hannun, Y. A. & Obeid, L. M. Sphingolipids and their metabolism in physiology and disease. Nat. Rev. Mol. Cell Biol. 19, 175–191 (2018).

    Article  CAS  PubMed  Google Scholar 

  23. Xie, S. Z. et al. Sphingolipid modulation activates proteostasis programs to govern human hematopoietic stem cell self-renewal. Cell Stem Cell 25, 639–653 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Golan, K. et al. S1P promotes murine progenitor cell egress and mobilization via S1P1-mediated ROS signaling and SDF-1 release. Blood 119, 2478–2488 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Juarez, J. G. et al. Sphingosine-1-phosphate facilitates trafficking of hematopoietic stem cells and their mobilization by CXCR4 antagonists in mice. Blood 119, 707–716 (2012).

    Article  CAS  PubMed  Google Scholar 

  26. Massberg, S. et al. Immunosurveillance by hematopoietic progenitor cells trafficking through blood, lymph, and peripheral tissues. Cell 131, 994–1008 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Ergen, A. V., Boles, N. C. & Goodell, M. A. Rantes/Ccl5 influences hematopoietic stem cell subtypes and causes myeloid skewing. Blood 119, 2500–2509 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Yamashita, M. & Passegue, E. TNF-α coordinates hematopoietic stem cell survival and myeloid regeneration. Cell Stem Cell 25, 357–372 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Chambers, S. M. et al. Aging hematopoietic stem cells decline in function and exhibit epigenetic dysregulation. PLoS Biol. 5, e201 (2007).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  30. Valletta, S. et al. Micro-environmental sensing by bone marrow stroma identifies IL-6 and TGFβ1 as regulators of hematopoietic ageing. Nat. Commun. 11, 4075 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Young, K. et al. Decline in IGF1 in the bone marrow microenvironment initiates hematopoietic stem cell aging. Cell Stem Cell 28, 1473–1482 (2021).

    Article  CAS  PubMed  Google Scholar 

  32. Henry, C. J. et al. Aging-associated inflammation promotes selection for adaptive oncogenic events in B cell progenitors. J. Clin. Invest. 125, 4666–4680 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Baldridge, M. T., King, K. Y., Boles, N. C., Weksberg, D. C. & Goodell, M. A. Quiescent haematopoietic stem cells are activated by IFN-γ in response to chronic infection. Nature 465, 793–797 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Essers, M. A. et al. IFNα activates dormant haematopoietic stem cells in vivo. Nature 458, 904–908 (2009).

    Article  CAS  PubMed  Google Scholar 

  35. Matatall, K. A. et al. Chronic infection depletes hematopoietic stem cells through stress-induced terminal differentiation. Cell Rep. 17, 2584–2595 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Pietras, E. M. et al. Chronic interleukin-1 exposure drives haematopoietic stem cells towards precocious myeloid differentiation at the expense of self-renewal. Nat. Cell Biol. 18, 607–618 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Pietras, E. M., Warr, M. R. & Passegue, E. Cell cycle regulation in hematopoietic stem cells. J. Cell Biol. 195, 709–720 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. 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 

  39. Fujisaki, J. et al. In vivo imaging of Treg cells providing immune privilege to the haematopoietic stem-cell niche. Nature 474, 216–219 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. 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 

  41. Luo, Y. & Liu, M. Adiponectin: a versatile player of innate immunity. J. Mol. Cell. Biol. 8, 120–128 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Wu, X. et al. Intrinsic immunity shapes viral resistance of stem cells. Cell 172, 423–438 (2018).

    Article  CAS  PubMed  Google Scholar 

  43. Pearl-Yafe, M. et al. Tumor necrosis factor receptors support murine hematopoietic progenitor function in the early stages of engraftment. Stem Cells 28, 1270–1280 (2010).

    Article  CAS  PubMed  Google Scholar 

  44. Pronk, C. J., Veiby, O. P., Bryder, D. & Jacobsen, S. E. Tumor necrosis factor restricts hematopoietic stem cell activity in mice: involvement of two distinct receptors. J. Exp. Med. 208, 1563–1570 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Ishida, T. et al. Pre-transplantation blockade of TNF-α-mediated oxygen species accumulation protects hematopoietic stem cells. Stem Cells 35, 989–1002 (2017).

    Article  CAS  PubMed  Google Scholar 

  46. Donnelly, R. P., Freeman, S. L. & Hayes, M. P. Inhibition of IL-10 expression by IFN-γ up-regulates transcription of TNF-α in human monocytes. J. Immunol. 155, 1420–1427 (1995).

    CAS  PubMed  Google Scholar 

  47. Vila-del Sol, V., Punzon, C. & Fresno, M. IFN-γ-induced TNF-α expression is regulated by interferon regulatory factors 1 and 8 in mouse macrophages. J. Immunol. 181, 4461–4470 (2008).

    Article  CAS  PubMed  Google Scholar 

  48. Palucka, A. K., Blanck, J. P., Bennett, L., Pascual, V. & Banchereau, J. Cross-regulation of TNF and IFN-α in autoimmune diseases. Proc. Natl Acad. Sci. USA 102, 3372–3377 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Raju, R. et al. A new cecal slurry preparation protocol with improved long-term reproducibility for animal models of sepsis. PLoS ONE 9, e115705 (2014).

    Article  CAS  Google Scholar 

  50. Kuhn, R., Schwenk, F., Aguet, M. & Rajewsky, K. Inducible gene targeting in mice. Science 269, 1427–1429 (1995).

    Article  CAS  PubMed  Google Scholar 

  51. de Boer, J. et al. Transgenic mice with hematopoietic and lymphoid specific expression of Cre. Eur. J. Immunol. 33, 314–325 (2003).

    Article  PubMed  Google Scholar 

  52. Lee, S. H. et al. Identifying the initiating events of anti-Listeria responses using mice with conditional loss of IFN-γ receptor subunit 1 (IFNGR1). J. Immunol. 191, 4223–4234 (2013).

    Article  CAS  PubMed  Google Scholar 

  53. Kajimura, D. et al. Adiponectin regulates bone mass via opposite central and peripheral mechanisms through FoxO1. Cell Metab. 17, 901–915 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Yue, R., Shen, B. & Morrison, S. J. Clec11a/osteolectin is an osteogenic growth factor that promotes the maintenance of the adult skeleton. eLife 5, e18782 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  55. Trapnell, C. et al. Differential gene and transcript expression analysis of RNA-seq experiments with TopHat and Cufflinks. Nat. Protoc. 7, 562–578 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Pertea, M., Kim, D., Pertea, G. M., Leek, J. T. & Salzberg, S. L. Transcript-level expression analysis of RNA-seq experiments with HISAT, StringTie and Ballgown. Nat. Protoc. 11, 1650–1667 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. 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  CAS  PubMed  PubMed Central  Google Scholar 

  58. Mootha, V. K. et al. PGC-1α-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes. Nat. Genet. 34, 267–273 (2003).

    Article  CAS  PubMed  Google Scholar 

  59. Noguchi, K., Gel, Y. R., Brunner, E. & Konietschke, F. nparLD: an RSoftware package for the nonparametric analysis of longitudinal data in factorial experiments. J. Stat. Softw. 50, 1–23 (2012).

    Article  Google Scholar 

Download references

Acknowledgements

S.J.M. is a Howard Hughes Medical Institute (HHMI) Investigator, the Mary McDermott Cook Chair in Pediatric Genetics, the Kathryn and Gene Bishop Distinguished Chair in Pediatric Research, the director of the Hamon Laboratory for Stem Cells and Cancer and a Cancer Prevention and Research Institute of Texas Scholar. This work was supported by the National Institutes of Health (DK118745), the Moody Medical Research Institute, the Josephine Hughes Sterling Foundation and the Robert J. Kleberg, Jr. and Helen C. Kleberg Foundation (all to S.J.M.). C.E.M. was supported by a Postdoctoral Fellowship from the American Cancer Society (PF-13-245-01-LIB). E.C.J. was supported by a Postdoctoral Fellowship from the Damon Runyon Cancer Research Foundation (2278-16). R.J.B. was supported by Ruth L. Kirschstein National Research Service Award Postdoctoral Fellowship from the National Heart, Lung, and Blood Institute (F32HL122095-01). Funding for the UT Dallas/UT Southwestern Green Fellows Program (C.D.S.) was supported, in part, by the Cecil and Ida Green Foundation. We thank G. Karsenty for providing the Adipor1fl/fl and Adipor2fl/fl mice, N. Loof, C. Cantu, T. Shih, G. Wilson and the Moody Foundation Flow Cytometry Facility, M. Mulkey for mouse colony management, and the BioHPC high-performance computing cloud at the University of Texas Southwestern Medical Center for providing computational resources.

Author information

Authors and Affiliations

Authors

Contributions

C.E.M. and S.J.M. conceived the project, and designed and interpreted experiments. C.E.M. performed most of the experiments. E.C.J. and R.J.B. discussed experiments and helped interpret data. E.C.J. performed the stromal cell analysis. A.M.S. processed samples for flow cytometric analysis in Fig. 2. E.M.C. processed samples for flow cytometric analysis in Fig. 4. C.D.S. and M.A.A. performed ELISAs in Fig. 3 and processed samples for flow cytometric analysis in Fig. 6. A.M.S., E.M.C. and M.A.A. analysed blood samples from transplanted mice. G.M.C. performed histopathological analysis of spleen sections. Z.Z. performed bioinformatic and statistical analyses. C.E.M. and S.J.M. wrote the manuscript.

Corresponding author

Correspondence to Sean J. Morrison.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Cell Biology thanks Maria Carolina Florian, 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

Extended Data Fig. 1 Adiponectin receptor deficient mice are born in normal numbers and are normal in size (related to Fig. 1).

a-c. Vav1-Cre; Adipor1fl/fl; Adipor2fl/fl mice were born at the expected mendelian frequencies (a) and were grossly normal in size (b) and appearance (c) (n = 4 female and n = 8 male Vav1-Cre; Adipor1fl/fl; Adipor2fl/fl mice and n = 10 female and n = 3 male control mice in one experiment (b)). d. The number of bone marrow cells in one femur and one tibia or in the spleen of 8-10-week-old Vav1-Cre and control mice (five mice per genotype in one experiment). e. White blood cell, red blood cell, and platelet counts in the blood of 8–10-week-old Vav1-Cre and control mice (five mice per genotype in one experiment). f-i. The frequencies of HSCs (f), MPPs (g), HPCs (h), CMPs, MEPs, and GMPs (i) in bone marrow from one femur and one tibia of Vav1-Cre and control mice (five mice per genotype in one experiment). j-m. The frequencies of HSCs (j), MPPs (k), HPCs (l), CMPs, MEPs, and GMPs (m) in splenocytes from Vav1-Cre and control mice (five mice per genotype in one experiment). All data represent mean ± standard deviation and each dot reflects a different mouse. Statistical significance was assessed using Mann-Whitney tests followed by Holm-Sidak’s multiple comparisons adjustments (b and d-e) or Student’s t-tests followed by Holm-Sidak’s multiple comparisons adjustments (f-m). All statistical tests were two-sided. Source numerical data are available in the source data files.

Source data

Extended Data Fig. 2 Flow cytometry gating strategy for the isolation of hematopoietic stem cells, progenitor cells, and differentiated cells (related to Figs. 16).

a-c. Representative flow cytometry gates used to identify hematopoietic stem and progenitor cells (a-b), NK cells, CD4 + T cells, and CD8 + T cells (c) in the bone marrow. The markers used to identify each of the cell populations characterized in this study are listed in Supplementary Table 1. d. Hematoxylin and Eosin stained sections from the spleens of Vav1-Cre; Adipor1fl/fl; Adipor2fl/fl and control mice. In Vav1-Cre; Adipor1fl/fl; Adipor2fl/fl mice the spleens were enlarged (Fig. 1d) and the red pulp was expanded throughout the spleen with increased numbers of myeloid, erythroid, and megakaryocytic cells. In control spleens there was limited extramedullary hematopoiesis confined to the subcapsular region. Representative images from three mice per genotype in one experiment are shown.

Extended Data Fig. 3 Adiponectin receptors are required to sustain HSC function (related to Fig. 2).

a-b. The percentages of HSCs that formed colonies (n = 6 mice per genotype in two independent experiments) (a) and colony size (n = 3 mice per genotype in one experiment) (b) from Vav1-Cre; Adipor1fl/fl; Adipor2fl/fl and control mice. c. The percentage of whole bone marrow cells that formed CFU-GEMM, CFU-GM, CFU-G, CFU-M, and BFU-E colonies (n = 3 mice per genotype in one experiment). d. Donor CD45 + cells, T, B, and myeloid cells from the blood of mice competitively transplanted with donor bone marrow cells from Mx1-Cre; Adipor1fl/fl; Adipor2fl/fl (n = 27 recipients) or control (n = 25 recipients) mice (five donors per genotype in five independent experiments). e. Secondary recipients of bone marrow cells from the mice in (d) (n = 25 recipients from six Mx1-Cre; Adipor1fl/fl; Adipor2fl/fl donors and n = 24 recipients from five control donors in five independent experiments). All data represent mean ± standard deviation and each dot reflects a different mouse. Statistical significance was assessed using Mann-Whitney tests (d-e), a Student’s t-test (a), a multinomial logistic regression (b), and Student’s t-tests followed by Holm-Sidak’s multiple comparisons adjustments (c), or Mann-Whitney tests (d-e). All statistical tests were two-sided. Source numerical data are available in the source data files.

Source data

Extended Data Fig. 4 Adiponectin receptors suppress the production of inflammatory factors (related to Fig. 3).

a. ELISA analysis of inflammatory cytokines in spleen lysates from Vav1-Cre; Adipor1fl/fl; Adipor2fl/fl and control mice (spleen lysates from 16 (IFNγ, TNF, IL6, and Il1β) or 13 (IFNα and IFNβ) Vav1-Cre; Adipor1fl/fl; Adipor2fl/fl mice and 19 (IFNγ, TNF, IL6, and Il1β) or 14 (IFNα and IFNβ) control mice in two independent ELISAs per cytokine). b. qRT-PCR analysis of Tnf transcript levels in sorted bone marrow cells. Data are normalized to transcript levels in wildtype whole bone marrow (WBM) cells (cells sorted from six (WBM), three (HSC, HPC, CMP, MEP, GMP, T-cells, B-cells, and erythroid progenitors), or five (myeloid cells) Vav1-Cre; Adipor1fl/fl; Adipor2fl/fl mice and from seven (WBM), three (HSC, HPC, CMP, MEP, GMP, and erythroid progenitors), or four (T-cells, B-cells, and myeloid cells) control mice in one experiment). c. qRT-PCR analysis of Tnf transcripts in sorted CD4 + T cells, CD8 + T cells, or NK cells from the bone marrow (cells sorted from three mice per genotype in one experiment). d. qRT-PCR analysis of Tnf transcripts in sorted macrophages, inflammatory monocytes, and neutrophils from the bone marrow. Data are normalized to transcript levels in wildtype WBM (cells sorted from seven mice per genotype in two independent experiments). e. The frequency of LepR+ stromal cells in bone marrow from 8-10 week old Vav1-Cre; Adipor1fl/fl; Adipor2fl/fl and control mice (n = 7 mice per genotype in two independent experiments). f. RNAseq analysis of transcripts for Ifng and Tnf in LepR+ cells from 8-10 week old Vav1-Cre; Adipor1fl/fl; Adipor2fl/fl and control mice (n = 3 mice per genotype one experiment). g. The frequencies of donor and competitor HSCs in the spleens of recipient mice co-transplanted with Vav1-Cre; Adipor1fl/fl; Adipor2fl/fl or control donor cells and wildtype competitor cells 16 weeks after transplantation (n = 19 recipients from Vav1-Cre; Adipor1fl/fl; Adipor2fl/fl donors and n = 20 recipients from control donors in four independent experiments). All data represent mean ± standard deviation and each dot reflects a different mouse. Statistical significance was assessed using Student’s t-tests followed by Holm-Sidak’s multiple comparisons adjustments (a-b and d), a matched samples two-way ANOVA followed by Sidak’s multiple comparisons adjustment (c), Student’s t-tests (e), or Mann-Whitney tests followed by Holm-Sidak’s multiple comparisons adjustments (a and g). All statistical tests were two-sided. Source numerical data are available in the source data files.

Source data

Extended Data Fig. 5 Adiponectin receptors promote HSC function by reducing IFNγ receptor signaling (related to Fig. 4).

a. Bone marrow (one femur and one tibia) and spleen cellularity in Vav1-Cre; Ifngr1fl/fl and control mice (n = 7 Vav1-Cre; Ifngrfl/fl mice and n = 6 control mice in three independent experiments). b-d. The frequencies of HSCs, MPPs, HPCs (b), CMPs, MEPs, GMPs (c), and differentiated T, B, myeloid, and erythroid cells (d) in the bone marrow of Vav1-Cre; Ifngr1fl/fl and control mice (n = 7 Vav1-Cre; Ifngrfl/fl mice and n = 6 control mice in three independent experiments). e−g. The frequencies of HSCs, MPPs, HPCs (e), CMPs, MEPs, GMPs (f), and differentiated cells (g) in the spleens of Vav1-Cre; Ifngr1fl/fl and control mice (n = 7 Vav1-Cre; Ifngrfl/fl mice and n = 6 control mice in three independent experiments). h. The percentage of HSCs that incorporated a 72 hour pulse of BrdU (n = 3 Vav1-Cre; Ifngrfl/fl mice and n = 4 control mice one experiment). i-o. The frequencies of HSCs (i), MPPs (j), HPCs (k), CMPs (l), MEPs (m), GMPs (n) and differentiated cells (o) in the bone marrow of control, Vav1-Cre; Adipor1fl/fl; Adipor2fl/fl, and Vav1-Cre; Adipor1fl/fl; Adipor2fl/fl; Ifngrfl/fl mice (n = 15 Vav1-Cre; Adipor1fl/fl; Adipor2fl/fl mice, n = 13 Vav1-Cre; Adipor1fl/fl; Adipor2fl/fl; Ifngrfl/f mice and n = 21 control mice in five independent experiments). All data represent mean ± standard deviation and each dot reflects a different mouse. Statistical significance was assessed using a matched samples two-way ANOVA followed by Sidak’s multiple comparisons adjustment (a), Mann-Whitney tests followed by Holm-Sidak’s multiple comparisons adjustments (b-g), a Student’s t-test (h), or oneway ANOVAs followed by Tukey’s multiple comparisons adjustments (i-o). All statistical tests were two-sided. Source numerical data are available in the source data files.

Source data

Extended Data Fig. 6 Adiponectin receptors promote HSC function by reducing TNF levels (related to Fig. 5).

a. Bone marrow (one femur and one tibia) and spleen cellularity in Tnf deficient and control mice (n = 5 Tnf-/- mice and n = 7 control mice in four independent experiments). b-d. The frequencies of HSCs, MPPs, HPCs (b), CMPs, MEPs, GMPs (c) and differentiated T, B, myeloid, and erythroid cells (d), in the bone marrow of Tnf deficient and control mice (n = 5 Tnf-/- mice and n = 7 control mice in four independent experiments). e-g. The frequencies of HSCs, MPPs, HPCs (e), CMPs, MEPs, GMPs (f) and differentiated cells (g), in the spleens of Tnf deficient and control mice (n = 5 (e and g) or n = 4 (f) Tnf-/- mice and n = 7 control mice in four independent experiments). h. The percentage of HSCs that incorporated a 72 hour pulse of BrdU (n = 4 Tnf-/- mice and n = 9 control mice in two independent experiments) i-o. The frequencies of HSCs (i), MPPs (j), HPCs (k), CMPs (l), MEPs (m), GMPs (n) and differentiated cells (o) in the bone marrow of control, Vav1-Cre; Adipor1fl/fl; Adipor2fl/fl, and Vav1-Cre; Adipor1fl/fl; Adipor2fl/fl; Tnf-/- mice (n = 12 Vav1-Cre; Adipor1fl/fl; Adipor2fl/fl mice, n = 11 Vav1-Cre; Adipor1fl/fl; Adipor2fl/fl; Tnf-/- mice and n = 12 control mice in five independent experiments). p-q. ELISA of TNF (p) or INFg (q) in blood plasma from Vav1-Cre; Adipor1fl/fl; Adipor2fl/fl; Ifngrfl/fl (p), Vav1-Cre; Adipor1fl/fl; Adipor2fl/fl; Tnf-/- (q), Vav1-Cre; Adipor1fl/fl; Adipor2fl/fl and control mice (n = 6 Vav1-Cre; Adipor1fl/fl; Adipor2fl/fl mice, n = 10 Vav1-Cre; Adipor1fl/fl; Adipor2fl/fl; Ifngrfl/f mice and n = 17 control mice run in one ELISA per cytokine (p) and n = 13 Vav1-Cre; Adipor1fl/fl; Adipor2fl/fl mice, n = 6 Vav1-Cre; Adipor1fl/fl; Adipor2fl/fl; Tnf-/- mice and n = 12 control mice in one ELISA per cytokine (q)). All data represent mean ± standard deviation and each dot reflects a different mouse. Statistical significance was assessed using a matched samples two-way ANOVA followed by Sidak’s multiple comparisons adjustment (a), Student’s t-tests (b-h) followed by Holm-Sidak’s multiple comparisons adjustments (b-g), one-way ANOVAs followed by Tukey’s multiple comparisons adjustments (i-o and q), a Welch’s one-way ANOVA followed by Dunnett’s T3 multiple comparisons adjustment (o), or a Kruskal-Wallis test followed by Dunn’s multiple comparisons adjustment (o and p). All statistical tests were two-sided. Source numerical data are available in the source data files.

Source data

Extended Data Fig. 7 Adiponectin receptor function reduces the frequencies of splenic myeloid cells in 5-6 and 19-24 month old mice (related to Fig. 6).

a-d. The frequencies of CMPs, MEPs, GMPs (a, c), differentiated T, B, myeloid, and erythroid cells (b, d) in the bone marrow from 5-6 month old (a-b) or 19-24 month old (c-d) Vav1-Cre; Adipor1fl/fl; Adipor2fl/fl or control mice (n = 7 (a-b) or n = 5 (c-d) Vav1-Cre; Adipor1fl/fl; Adipor2fl/fl mice and n = 10 (a-b) or n = 6 (c-d) control mice in two independent experiments per age group). e-n. The frequencies of HSCs (e, j), MPPs (f, k), HPCs (g, l), CMPs, MEPs, GMPs (h, m), and differentiated T, B, myeloid, and erythroid cells (i, n) in the spleens of 5–6-month-old (e-i) and 19-24 month-old (j-n) Vav1-Cre; Adipor1fl/fl; Adipor2fl/fl or control mice (n = 7 (e-i) or n = 5 (j-n) Vav1-Cre; Adipor1fl/fl; Adipor2fl/fl mice and n = 10 (e-i) or n = 6 (j-n) control mice in two independent experiments per age group). All data represent mean ± standard deviation and each dot reflects a different mouse. Statistical significance was assessed using Mann-Whitney tests followed by Holm-Sidak’s multiple comparisons adjustments (a-n). All statistical tests were two-sided. Source numerical data are available in the source data files.

Source data

Supplementary information

Reporting Summary

Peer Review File

Supplementary Table 1

Cell populations analysed by flow cytometry in this study.

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Meacham, C.E., Jeffery, E.C., Burgess, R.J. et al. Adiponectin receptors sustain haematopoietic stem cells throughout adulthood by protecting them from inflammation. Nat Cell Biol 24, 697–707 (2022). https://doi.org/10.1038/s41556-022-00909-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-022-00909-9

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