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.

Exercise reduces inflammatory cell production and cardiovascular inflammation via instruction of hematopoietic progenitor cells

Abstract

A sedentary lifestyle, chronic inflammation and leukocytosis increase atherosclerosis; however, it remains unclear whether regular physical activity influences leukocyte production. Here we show that voluntary running decreases hematopoietic activity in mice. Exercise protects mice and humans with atherosclerosis from chronic leukocytosis but does not compromise emergency hematopoiesis in mice. Mechanistically, exercise diminishes leptin production in adipose tissue, augmenting quiescence-promoting hematopoietic niche factors in leptin-receptor-positive stromal bone marrow cells. Induced deletion of the leptin receptor in Prrx1-creERT2; Leprfl/fl mice reveals that leptin’s effect on bone marrow niche cells regulates hematopoietic stem and progenitor cell (HSPC) proliferation and leukocyte production, as well as cardiovascular inflammation and outcomes. Whereas running wheel withdrawal quickly reverses leptin levels, the impact of exercise on leukocyte production and on the HSPC epigenome and transcriptome persists for several weeks. Together, these data show that physical activity alters HSPCs via modulation of their niche, reducing hematopoietic output of inflammatory leukocytes.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Exercise increases HSPC quiescence.
Fig. 2: Exercise dampens hematopoiesis by reducing adipose tissue leptin production.
Fig. 3: Voluntary running reduces LSK chromatin accessibility.
Fig. 4: Exercise augments emergency hematopoiesis and improves survival in sepsis.
Fig. 5: Disrupting leptin signaling reduces hematopoiesis and inflammation in acute MI.
Fig. 6: Sedentary lifestyle accelerates leukocyte supply in mice and humans with atherosclerosis.

Similar content being viewed by others

Data availability

All data are included in this published article and its supplementary information files. Raw sequencing data are available from Gene Expression Omnibus under accession numbers GSE110639 and GSE124799. Raw data other than sequencing data that support the findings of this study are available from the corresponding author upon reasonable request. Source data are available for Figs. 1–6.

References

  1. Ridker, P. M. Residual inflammatory risk: addressing the obverse side of the atherosclerosis prevention coin. Eur. Heart J. 37, 1720–1722 (2016).

    PubMed  Google Scholar 

  2. Ridker, P. M. et al. Anti-inflammatory therapy with canakinumab for atherosclerotic disease. N. Engl. J. Med. 377, 1119–1131 (2017).

    CAS  PubMed  Google Scholar 

  3. Madjid, M., Awan, I., Willerson, J. T. & Casscells, S. W. Leukocyte count and coronary heart disease: implications for risk assessment. J. Am. Coll. Cardiol. 44, 1945–1956 (2004).

    PubMed  Google Scholar 

  4. Nahrendorf, M. & Swirski, F. K. Lifestyle effects on hematopoiesis and atherosclerosis. Circ. Res. 116, 884–894 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Tall, A. R. & Yvan-Charvet, L. Cholesterol, inflammation and innate immunity. Nat. Rev. Immunol. 15, 104–116 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Heidt, T. et al. Chronic variable stress activates hematopoietic stem cells. Nat. Med. 20, 754–758 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. McAlpine, C. S. et al. Sleep modulates haematopoiesis and protects against atherosclerosis. Nature 566, 383–387 (2019).

    PubMed  PubMed Central  Google Scholar 

  8. Nagareddy, P. R. et al. Adipose tissue macrophages promote myelopoiesis and monocytosis in obesity. Cell Metab. 19, 821–835 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Morrison, S. J. & Scadden, D. T. The bone marrow niche for haematopoietic stem cells. Nature 505, 327–334 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Lavie, C. J., Ozemek, C., Carbone, S., Katzmarzyk, P. T. & Blair, S. N. Sedentary behavior, exercise, and cardiovascular health. Circ. Res. 124, 799–815 (2019).

    CAS  PubMed  Google Scholar 

  11. Lee, D. C. et al. Leisure-time running reduces all-cause and cardiovascular mortality risk. J. Am. Coll. Cardiol. 64, 472–481 (2014).

    PubMed  PubMed Central  Google Scholar 

  12. Duggal, N. A., Niemiro, G., Harridge, S. D. R., Simpson, R. J. & Lord, J. M. Can physical activity ameliorate immunosenescence and thereby reduce age-related multi-morbidity. Nat. Rev. Immunol. 19, 563–572 (2019).

    CAS  PubMed  Google Scholar 

  13. Meissner, M. et al. Voluntary wheel running increases bile acid as well as cholesterol excretion and decreases atherosclerosis in hypercholesterolemic mice. Atherosclerosis 218, 323–329 (2011).

    CAS  PubMed  Google Scholar 

  14. Fukao, K. et al. Voluntary exercise ameliorates the progression of atherosclerotic lesion formation via anti-inflammatory effects in apolipoprotein E-deficient mice. J. Atheroscler. Thromb. 17, 1226–1236 (2010).

    CAS  PubMed  Google Scholar 

  15. Pellegrin, M. et al. Long-term exercise stabilizes atherosclerotic plaque in ApoE knockout mice. Med. Sci. Sports Exerc. 41, 2128–2135 (2009).

    CAS  PubMed  Google Scholar 

  16. de Visser, L., van den Bos, R. & Spruijt, B. M. Automated home cage observations as a tool to measure the effects of wheel running on cage floor locomotion. Behav. Brain Res. 160, 382–388 (2005).

    PubMed  Google Scholar 

  17. Asada, N., Takeishi, S. & Frenette, P. S. Complexity of bone marrow hematopoietic stem cell niche. Int. J. Hematol. 106, 45–54 (2017).

    PubMed  PubMed Central  Google Scholar 

  18. Abella, V. et al. Leptin in the interplay of inflammation, metabolism and immune system disorders. Nat. Rev. Rheumatol. 13, 100–109 (2017).

    CAS  PubMed  Google Scholar 

  19. Bennett, B. D. et al. A role for leptin and its cognate receptor in hematopoiesis. Curr. Biol. 6, 1170–1180 (1996).

    CAS  PubMed  Google Scholar 

  20. Hirose, H. et al. Serum leptin level: possible association with haematopoiesis in adolescents, independent of body mass index and serum insulin. Clin. Sci. 94, 633–636 (1998).

    CAS  Google Scholar 

  21. Rostas, I. et al. In middle-aged and old obese patients, training intervention reduces leptin level: a meta-analysis. PLoS ONE 12, e0182801 (2017).

    PubMed  PubMed Central  Google Scholar 

  22. Zhou, B. O., Yue, R., Murphy, M. M., Peyer, J. G. & Morrison, S. J. Leptin-receptor-expressing mesenchymal stromal cells represent the main source of bone formed by adult bone marrow. Cell Stem Cell 15, 154–168 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Ding, L., Saunders, T. L., Enikolopov, G. & Morrison, S. J. Endothelial and perivascular cells maintain haematopoietic stem cells. Nature 481, 457–462 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Yue, R., Zhou, B. O., Shimada, I. S., Zhao, Z. & Morrison, S. J. Leptin receptor promotes adipogenesis and reduces osteogenesis by regulating mesenchymal stromal cells in adult bone marrow. Cell Stem Cell 18, 782–96 (2016).

    CAS  PubMed  Google Scholar 

  25. Mercier, F. E., Sykes, D. B. & Scadden, D. T. Single targeted exon mutation creates a true congenic mouse for competitive hematopoietic stem cell transplantation: the C57BL/6-CD45.1STEM mouse. Stem Cell Rep. 6, 985–992 (2016).

    CAS  Google Scholar 

  26. Cao, J. et al. Joint profiling of chromatin accessibility and gene expression in thousands of single cells. Science 361, 1380–1385 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Lara-Astiaso, D. et al. Chromatin state dynamics during blood formation. Science 345, 943–949 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Yu, V. W. C. et al. Epigenetic memory underlies cell-autonomous heterogeneous behavior of hematopoietic stem cells. Cell 167, 1310–1322 (2016).

    CAS  PubMed  Google Scholar 

  30. Golan, K., Kollet, O. & Lapidot, T. Dynamic cross talk between S1P and CXCL12 regulates hematopoietic stem cells migration, development and bone remodeling. Pharmaceuticals 6, 1145–1169 (2013).

    PubMed  PubMed Central  Google Scholar 

  31. Ito, K. & Suda, T. Metabolic requirements for the maintenance of self-renewing stem cells. Nat. Rev. Mol. Cell Biol. 15, 243–256 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Tanaka, Y. et al. Exhaustive exercise reduces tumor necrosis factor-alpha production in response to lipopolysaccharide in mice. Neuroimmunomodulation 17, 279–286 (2010).

    CAS  PubMed  Google Scholar 

  33. Mackinnon, L. T., Chick, T. W., van As, A. & Tomasi, T. B. The effect of exercise on secretory and natural immunity. Adv. Exp. Med. Biol. 216A, 869–876 (1987).

    CAS  PubMed  Google Scholar 

  34. Shirato, K. et al. Regular voluntary exercise potentiates interleukin-1β and interleukin-18 secretion by increasing caspase-1 expression in murine macrophages. Mediators Inflamm. 2017, 9290416 (2017).

    PubMed  PubMed Central  Google Scholar 

  35. Wallerstedt, S. M., Eriksson, A. L., Niklason, A., Ohlsson, C. & Hedner, T. Serum leptin and myocardial infarction in hypertension. Blood Press. 13, 243–246 (2004).

    CAS  PubMed  Google Scholar 

  36. Rajendran, K., Devarajan, N., Ganesan, M. & Ragunathan, M. Obesity, inflammation and acute myocardial infarction—expression of leptin, IL-6 and high sensitivity-CRP in a Chennai-based population. Thromb. J. 10, 13 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Engström, G., Melander, O. & Hedblad, B. Leukocyte count and incidence of hospitalizations due to heart failure. Circ. Heart Fail. 2, 217–222 (2009).

    PubMed  Google Scholar 

  38. Tsujioka, H. et al. Impact of heterogeneity of human peripheral blood monocyte subsets on myocardial salvage in patients with primary acute myocardial infarction. J. Am. Coll. Cardiol. 54, 130–138 (2009).

    PubMed  Google Scholar 

  39. Swirski, F. K. & Nahrendorf, M. Leukocyte behavior in atherosclerosis, myocardial infarction, and heart failure. Science 339, 161–166 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Verhoeven, B. A. et al. Athero-Express: differential atherosclerotic plaque expression of mRNA and protein in relation to cardiovascular events and patient characteristics. Rationale and design. Eur. J. Epidemiol. 19, 1127–1133 (2004).

    CAS  PubMed  Google Scholar 

  41. Kelesidis, T., Kelesidis, I., Chou, S. & Mantzoros, C. S. Narrative review: the role of leptin in human physiology: emerging clinical applications. Ann. Intern. Med. 152, 93–100 (2010).

    PubMed  PubMed Central  Google Scholar 

  42. De Lisio, M. & Parise, G. Characterization of the effects of exercise training on hematopoietic stem cell quantity and function. J. Appl. Physiol. 113, 1576–1584 (2012).

    PubMed  PubMed Central  Google Scholar 

  43. Baker, J. M., De Lisio, M. & Parise, G. Endurance exercise training promotes medullary hematopoiesis. FASEB J. 25, 4348–4357 (2011).

    CAS  PubMed  Google Scholar 

  44. Agha, N. H. et al. Vigorous exercise mobilizes CD34+ hematopoietic stem cells to peripheral blood via the β2-adrenergic receptor. Brain Behav. Immun. 68, 66–75 (2018).

    CAS  PubMed  Google Scholar 

  45. Soppi, E., Varjo, P., Eskola, J. & Laitinen, L. A. Effect of strenuous physical stress on circulating lymphocyte number and function before and after training. J. Clin. Lab. Immunol. 8, 43–46 (1982).

    CAS  PubMed  Google Scholar 

  46. Johannsen, N. M. et al. Effect of different doses of aerobic exercise on total white blood cell (WBC) and WBC subfraction number in postmenopausal women: results from DREW. PLoS ONE 7, e31319 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Mabuchi, T. et al. Association between serum leptin concentration and white blood cell count in middle-aged Japanese men and women. Diabetes Metab. Res. Rev. 21, 441–447 (2005).

    CAS  PubMed  Google Scholar 

  48. Arnett, D. K. et al. 2019 ACC/AHA guideline on the primary prevention of cardiovascular disease: executive summary: a report of the American College of Cardiology/American Heart Association Task Force on clinical practice guidelines. J. Am. Coll. Cardiol. 74, 1376–1414 (2019).

    PubMed  PubMed Central  Google Scholar 

  49. Ionita, M. G. et al. High neutrophil numbers in human carotid atherosclerotic plaques are associated with characteristics of rupture-prone lesions. Arterioscler. Thromb. Vasc. Biol. 30, 1842–1848 (2010).

    CAS  PubMed  Google Scholar 

  50. Halliday, A. et al. Prevention of disabling and fatal strokes by successful carotid endarterectomy in patients without recent neurological symptoms: randomised controlled trial. Lancet 363, 1491–1502 (2004).

    CAS  PubMed  Google Scholar 

  51. ten Berg, M. J. et al. Linking laboratory and medication data: new opportunities for pharmacoepidemiological research. Clin. Chem. Lab. Med. 45, 13–19 (2007).

    PubMed  Google Scholar 

  52. Lam, S. W., Leenen, L. P. H., Solinge, W. W., Hietbrink, F. & Huisman, A. Evaluation of hematological parameters on admission for the prediction of 7-day in-hospital mortality in a large trauma cohort. Clin. Chem. Lab. Med. 49, 493–499 (2011).

    CAS  PubMed  Google Scholar 

  53. Groeneveld, K. M., Heeres, M., Leenen, L. P. H., Huisman, A. & Koenderman, L. Immunophenotyping of posttraumatic neutrophils on a routine haematology analyser. Mediators Inflamm. 2012, 509513 (2012).

    PubMed  PubMed Central  Google Scholar 

  54. Mignone, J. L., Kukekov, V., Chiang, A. S., Steindler, D. & Enikolopov, G. Neural stem and progenitor cells in nestin-GFP transgenic mice. J. Comp. Neurol. 469, 311–324 (2004).

    CAS  PubMed  Google Scholar 

  55. Méndez-Ferrer, S. et al. Mesenchymal and haematopoietic stem cells form a unique bone marrow niche. Nature 466, 829–834 (2010).

    PubMed  PubMed Central  Google Scholar 

  56. DeFalco, J. et al. Virus-assisted mapping of neural inputs to a feeding center in the hypothalamus. Science 291, 2608–2613 (2001).

    CAS  PubMed  Google Scholar 

  57. Bilic-Curcic, I. et al. Visualizing levels of osteoblast differentiation by a two-color promoter-GFP strategy: type I collagen-GFPcyan and osteocalcin-GFPtpz. Genesis 43, 87–98 (2005).

    CAS  PubMed  Google Scholar 

  58. Bjorklund, M. M. et al. Induction of atherosclerosis in mice and hamsters without germline genetic engineering. Circ. Res. 114, 1684–1689 (2014).

    CAS  PubMed  Google Scholar 

  59. Rittirsch, D., Huber-Lang, M. S., Flierl, M. A. & Ward, P. A. Immunodesign of experimental sepsis by cecal ligation and puncture. Nat. Protoc. 4, 31–36 (2008).

    Google Scholar 

  60. Konstantinides, S., Schäfer, K., Neels, J. G., Dellas, C. & Loskutoff, D. J. Inhibition of endogenous leptin protects mice from arterial and venous thrombosis. Arterioscler. Thromb. Vasc. Biol. 24, 2196–2201 (2004).

    CAS  PubMed  Google Scholar 

  61. Surwit, R. S., Edwards, C. L., Murthy, S. & Petro, A. E. Transient effects of long-term leptin supplementation in the prevention of diet-induced obesity in mice. Diabetes 49, 1203–1208 (2000).

    CAS  PubMed  Google Scholar 

  62. Kunisaki, Y. et al. Arteriolar niches maintain haematopoietic stem cell quiescence. Nature 502, 637–643 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Orr, J. S., Kennedy, A. J. & Hasty, A. H. Isolation of adipose tissue immune cells. J. Vis. Exp. 22, e50707 (2013).

    Google Scholar 

  64. Buenrostro, J. D., Wu, B., Chang, H. Y. & Greenleaf, W. J. ATAC-seq: a method for assaying chromatin accessibility genome-wide. Curr. Protoc. Mol. Biol. 109, 1–9 (2015).

    Google Scholar 

  65. Mehta, S. et al. Maintenance of macrophage transcriptional programs and intestinal homeostasis by epigenetic reader SP140. Sci. Immunol. 2, eaag3160 (2017).

    PubMed  PubMed Central  Google Scholar 

  66. Li, H. Exploring single-sample SNP and INDEL calling with whole-genome de novo assembly. Bioinformatics 28, 1838–1844 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  67. John, S. et al. Chromatin accessibility pre-determines glucocorticoid receptor binding patterns. Nat. Genet. 43, 264–268 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Robinson, M. D., McCarthy, D. J. & Smyth, G. K. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26, 139–140 (2010).

    CAS  PubMed  Google Scholar 

  69. Patro, R., Duggal, G., Love, M. I., Irizarry, R. A. & Kingsford, C. Salmon provides fast and bias-aware quantification of transcript expression. Nat. Methods 14, 417–419 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Soneson, C., Love, M. I. & Robinson, M. D. Differential analyses for RNA-seq: transcript-level estimates improve gene-level inferences. F1000Res 4, 1521 (2015).

    PubMed  Google Scholar 

  71. McCarthy, D. J., Chen, Y. & Smyth, G. K. Differential expression analysis of multifactor RNA-Seq experiments with respect to biological variation. Nucleic Acids Res. 40, 4288–4297 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Yu, G., Wang, L. G., Han, Y. & He, Q. Y. clusterProfiler: an R package for comparing biological themes among gene clusters. OMICS 16, 284–287 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Bouxsein, M. L. et al. Guidelines for assessment of bone microstructure in rodents using micro-computed tomography. J. Bone Miner. Res. 25, 1468–1486 (2010).

    PubMed  Google Scholar 

  74. Scheller, E. L. et al. Use of osmium tetroxide staining with microcomputerized tomography to visualize and quantify bone marrow adipose tissue in vivo. Methods Enzymol. 537, 123–139 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank M. Handley, E. Surette and A. Galvin of the HSCI-CRM Flow Cytometry Core Facility, Massachusetts General Hospital, for assistance with cell sorting, the Center for Skeletal Research Imaging and Biomechanical Testing Core (National Institutes of Health P30 AR066261), Massachusetts General Hospital, for bone histology and µCT imaging, the Bioanalytics Core at the Diabetes and Obesity Center, Christina Lee Brown Envirome Institute, University of Louisville, for mass spectrometry analysis, the BPF Next-Gen Sequencing Core Facility at Harvard Medical School for their support for RNA-sequencing and K. Joyes for editing the manuscript. This work was funded in part by federal funds from the National Institutes of Health (HL142494, HL139598, HL131478, HL128264, AI07087, DK040561 and T32HL076136), the European Union’s Horizon 2020 research and innovation program (grant agreement no. 667837), the Deutsche Forschungsgemeinschaft (CR 603/1-1, HO 5279/1-2 and RO 5071/1-1) and a fellowship from the Netherlands Organisation for Scientific Research (Rubicon Grant: 835.15.014). We acknowledge Servier Medical Art (https://smart.servier.com) for providing images of mice and components of the cartoon.

Author information

Authors and Affiliations

Authors

Contributions

V.F., D.R. and M.N. designed experiments. V.F., D.R., G.C., N.S., M.J.S., H.A., S.C., F.F.H., F.J., I.D.v.K., F.H., L.H., C.S.M., G.S.M., S.Z., J.G., Y.I., S.P.S., G.R.W., I.-H.L. and K.G. performed experiments and collected data. V.F., D.R., G.C., N.S., F.J., I.D.v.K., G.P., S.C.A.d.J., R.I.S., I.-H.L., J.M. and K.N. analyzed data. V.F., M.J.S., D.R., S.C., F.F.H. and G.S.M. performed surgeries. V.F., D.R., G.C., N.S., H.A., P.L., G.P., P.R., D.T.S., K.N., K.L.J., F.S. and M.N. discussed results and strategy. V.F., D.R. and M.N. wrote the manuscript, which was edited by all co-authors. M.N. supervised, directed and managed the study.

Corresponding author

Correspondence to Matthias Nahrendorf.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Michael Basson was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

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

Extended data

Extended Data Fig. 1 Effects of 6 weeks of running.

(a) Mean distance run per hour (n = 24 animals). (b) Mean daily distance over the course of 6 weeks (n = 16 animals). (c) changes in sedentary (n = 12) and exercising mice (n = 17, **p = 0.0076, two-tailed U test) compared to initial six weeks prior. (d) Heart weight adjusted for tibia length (n = 9 animals per group). (e) Food consumption during the last week of exercise (***p = 0.0002, n = 9 animals per group, two-tailed Student’s t-test). (f) Flow cytometry gating strategy for leukocytes in skeletal muscle. (g) Total leukocytes, neutrophils, monocytes and macrophages per mg muscle tissue by flow cytometry (n = 7 animals for sedentary, n = 12 for exercise). (h) Representative microCT images of the proximal metaphysis and mid-diaphysis tibia of exercising and sedentary mice. (i) Parameters of bone microstructure, including trabecular and cortical thickness, bone mineral density and polar moment of inertia by µCT (n = 6 animals per group). (j) Representative Runx2 staining of tibial proximal metaphysis. Osteoblast surface per bone surface (Ob.S/BS, n = 6 animals per group). (k) Bone formation rate as observed by incorporation of calcein (20 mg/kg, 7 days prior) and alizarin red (30 mg/kg, 2 days prior to sacrifice) during bone mineralization at the diaphysis of femurs. Distance of fluorescent label indicated by the arrow demarcates the mineralization front at different times of administration. ‘#’ denotes medullary cavity and ‘##’ trabecular bone (n = 4 animals). Data are mean ± s.e.m. We acknowledge Servier Medical Art (https://smart.servier.com) for providing images of mice and components of the cartoon.

Extended Data Fig. 2 Increased stem and progenitor cell quiescence after 6 weeks of exercise.

Mice were given 5-bromo-2- deoxyuridine (BrdU) intraperitoneally (1 mg). BrdU incorporation in (a) long-term hematopoietic stem cells (LT-HSC), short-term HSC (ST-HSC), (b) common myeloid progenitors (CMP, ***p = 0.00026), megakaryocyte erythroid progenitors (MEP, ***p = 1.028 × 10−5), granulocyte macrophage progenitors (GMP, ***p = 4.17 × 10−4, all n = 14 animals for sedentary, n = 15 for exercise), macrophage and dendritic cell progenitors (MDP, **p = 0.0070, n = 7 animals per group) and B cell progenitors (B cell prog, **p = 0.0065, n = 6 animals per group) were analyzed 22 hours later (two-tailed U test for CMP, MDP and B cell prog; two-tailed Student’s for MEPs and GMPs). (c) Flow cytometry gating for hematopoietic progenitors and representative flow cytometry plots of BrdU gating. (d) Cell cycle analysis in LSK assessed by Ki-67/ DAPI staining. Representative flow cytometry dot plots of + LSK (*p = 0.038, n = 7 animals for sedentary, n = 12 for exercise, 2 independent experiments, two-tailed U test). (e) Experimental outline for BrdU pulse-chase experiment. Mice received BrdU in drinking water for 3 weeks (baseline, n = 3 animals) prior to 3 weeks of exercise. (f) BrdU incorporation into LT-HSC, ST-HSC, multipotent progenitors (MPP, **p = 0.0074), CMP (p = 0.14), MEP (*p = 0.034) and GMP (p = 0.07, n = 9 animals for sedentary, n = 4 for exercise, two-tailed U test comparing sedentary and exercise). (g) Representative images of granulocyte macrophage colonies from sedentary and running mice. (h) Bone marrow unit assay (CFU) of bone marrow mononuclear cells (BMNCs) for complete colonies (*p = 0.036, n = 6 animals per group, 2 independent experiments, two-tailed U-test). (i) Number of HSPC per femur in sedentary and exercising mice (n = 15 animals per group). (j) Number of marrow leukocytes at Zeitgeber 13: B cells (**p = 0.0089), CD4 T cells, CD8 T cells (*p = 0.048), neutrophils (*p = 0.044), monocytes (*p = 0.041), eosinophils (*p = 0.019, n = 5 animals for sedentary, n = 6 for exercise) and NK cells (**p = 0.00099, n = 3 per group, two-tailed U test). (k) Numbers of platelets (*p = 0.016, two-tailed Student’s), red blood cells (RBC), hemoglobin (HGB) and hematocrit (HCT, n = 12 animals for sedentary and n = 11 for exercise, 4 independent experiments). Data are mean ± s.e.m.We acknowledge Servier Medical Art (https://smart.servier.com) for providing images of mice and components of the cartoon.

Extended Data Fig. 3 Neutral running effects on bone marrow neurotransmitters, corticosterone and selected hematopoietic niche cells.

(a) Mass spectrometry of norepinephrine and acetylcholine in the bone marrow after 6 weeks of exercise (n = 5 animals per group). (b) Choline acetyltransferase (ChAT) expression by bone marrow CD45+ leukocytes (n = 3 animals per group). (c) Experimental outline, administration of a competitive antagonist of the muscarinic acetylcholine receptors (atropine) during 3 weeks exercise. Leukocytes in circulation (n = 5 animals for Sed-Saline, n = 3 for Ex-Saline, n = 2 for Ex-Atropine). (d) Plasma corticosterone at Zeitgeber time (ZG 1 (n = 11 animals for sedentary, n = 8 for exercise), ZG 7 (n = 8 sedentary, n = 11 exercise), ZG 13 (n = 6 sedentary, n = 9 exercise) after exercise for 6 weeks. (e) Nestin+ stromal cells (n = 6 animals for sedentary, n = 8 for exercise, 3 independent experiments), (f) OCN+ osteoblasts, (n = 9 animals per group, 4 independent experiments) (g) endothelial cells (n = 8 animals for sedentary, n = 10 for exercise, 6 independent experiments) and (h) bone marrow macrophages (n = 5 animals per group, 2 independent experiments) were isolated by fluorescence-activated cell sorting. GFP stromal reporter mice either had access to exercise wheels for 6 weeks or remained sedentary. Representative dot plots are shown. Expression of Cxcl12, Vcam1, Kitl and Angpt1 was assessed by qPCR, ND: not detectable. (i) Numbers of stromal niche cells in sedentary and exercising mice (n = 9 and n = 8 for LepR+, n = 6 and n = 8 for Nestin+, n = 9 and n = 9 for OCN+, n = 8 and n = 10 for CD31high, n = 5 and n = 5 animals for sedentary and exercise, respectively). (j) Gene expression of several niche factors in total bone marrow by qPCR (n = 8 animals for sedentary for Ccl2 and Pf4, n = 12 for sedentary for Tgfb, Csf1, n = 16 for sedentary for Il7, Csf2, Csf3, n = 13 for exercise for Ccl2, Tgfb, n = 19 for exercise for Il7, Csf1, Csf2, Csf3, 4 independent experiments). (k) Markers for osteolineage cells (Sp7,Bglap, Runx2) and adipocytes (Lpl, Fabp4) by qPCR in total bone marrow (n = 8 animals per group, 2 independent experiments). All mRNA levels were normalized to Actb Ct values. Data are mean ± s.e.m., where appropriate. We acknowledge Servier Medical Art (https://smart.servier.com) for providing images of mice and components of the cartoon.

Extended Data Fig. 4 Exercise reduces visceral adipose tissue macrophages.

(a) Visceral adipose tissue (VAT) per mouse adjusted for (BW; ***p = 0.00069, n = 9 animals for sedentary, n = 7 for exercise, 2 independent experiments, two-tailed U test). (b) Cytokine production by visceral adipose tissue by qPCR (*p = 0.034, **p = 0.0087, n = 6 animals per group, 2 independent experiments, two-tailed U test). (c) Macrophages per mg VAT. Representative dot plots are shown (*p = 0.016, n = 5 animals for sedentary, n = 4 for exercise, two-tailed U test). (d) Experimental outline for e. Mice received BrdU in drinking water for 3 weeks (baseline, n = 4 animals) prior to 3 weeks of exercise. (e) BrdU incorporation into VAT macrophages (*p = 0.045, n = 9 animals for sedentary, n = 7 for exercise, 2 independent experiments, two-tailed U test comparing sedentary and exercise). Representative dot plots are shown. (f) Longitudinal sections of tibias were stained by perilipin (red) and counterstained by DAPI (blue). (g) Quantification of adipocyte numbers and size in the proximal metaphysis of tibias (**p = 0.0055, n = 8 animals for sedentary, n = 9 for exercise, 3 independent experiments, two-tailed U-test). (h) In vitro adipocyte differentiation assay of bone marrow stromal cells from all long bones and pelvic bones. Representative images with 100x magnification are shown (n = 4 animals per group). (i,j) Visualization of marrow adipose tissue in tibias by osmium stain by μCT and marrow adipose tissue (MAT) per marrow volume (MV) (n = 3 animals per group). (k) Leptin expression by qPCR in visceral adipose tissue (VAT; **p = 0.0022, n = 6 animals per group, two-tailed U test) and bone marrow (BM; n = 3 animals for sedentary, n = 6 for exercise). mRNA levels were normalized to Actb Ct values. Data are mean ± s.e.m. (l) Lack of correlation between tibial adipocyte size and leptin concentration (R2 = 0.0002, P = 0.96, n = 17 animals, linear regression analysis). We acknowledge Servier Medical Art (https://smart.servier.com) for providing images of mice and components of the cartoon.

Extended Data Fig. 5 Leptin supplementation and antibody neutralization.

(a) Leptin levels in blood (*p = 0.19 for both sed-leptin vs ex-saline and ex-saline vs ex-leptin, one-way analysis of variance with Sidak’s post hoc test) and bone marrow (*p = 0.013 sed-leptin vs ex-saline, *p = 0.042 sed-saline vs ex-saline, **p = 0.0038 ex-saline versus ex-leptin, n = 12 animals for sed-saline and ex-saline, n = 10 for sed-leptin, n = 13 for ex-leptin, 5 independent experiments, with Dunn’s post hoc test) as measured by ELISA. (b) LSK proliferation 22 h after intraperitoneal injection of BrdU (*p = 0.047 sed-saline versus ex-sal, P = 0.05 ex-saline versus ex-leptin, ***p = 0.00031 sed-leptin versus ex-sal, n = 12 animals for sed-saline and ex-saline, n = 10 for sed-leptin, n = 13 for ex-leptin, 5 independent experiments, one-way analysis of variance with Sidak’s post hoc test), LSK numbers and (c) expression of hematopoietic factors in bone marrow of exercising and sedentary mice implanted with osmotic minipumps as described in Fig. 2h (*p = 0.016 and **p = 0.0026 for Cxcl12, *p = 0.025 sed-saline versus ex-saline and *p = 0.046 ex-saline versus ex-leptin for Vcam1, ***p = 0.00074 ex-saline versus ex-leptin and p = 0.09 sed-saline versus ex-saline for Angpt1, n = 12 animals for sed-saline and ex-saline, n = 10 for sed-leptin, n = 13 for Ex-Leptin). (d) Running distance with either saline or leptin and access to exercise wheels for 6 weeks. Mean distance run per hour (n = 4 animals per group). (e) Injection of antibody or leptin into sedentary mice. Circulating leukocytes levels at Zeitgeber time 7 (*p = 0.010 IgG versus αLep, **p = 0.0072 αLep versus leptin) and LSK proliferation 22 h after intraperitoneal injection (**p = 0.0038 IgG versus αLep, ***p = 1.52 × 10−5, n = 6 animals for IgG, n = 7 for α-Lep, n = 4 for Leptin, 2 independent experiments, one-way analysis of variance with Sidak’s post hoc test). Data are mean ± s.e.m. We acknowledge Servier Medical Art (https://smart.servier.com) for providing images of mice and components of the cartoon.

Extended Data Fig. 6 Leptin receptor expression in the bone marrow.

(a) Representative flow cytometry dot plots of leptin receptor (LepR) expression in B cells, myeloid cells, T cells, (b) bone marrow long-term hematopoietic stem cells (LT-HSC), short-term HSC (ST-HSC), multipotent progenitors (MPP), common myeloid progenitors (CMP), megakaryocyte erythroid progenitors (MEP), granulocyte macrophage progenitors (GMP) and (c) stromal bone marrow cells (n = 3 independent experiments with similar results). (d) Bone marrow unit assay (CFU) for complete colonies (n = 4 donor animals). Bone marrow mononuclear cells (BMNCs) were plated with increasing concentrations of leptin. (e) Experimental outline for bone marrow transplantation; data shown in panel f-i. Total bone marrow was isolated from db/db donor mice and transplanted into wild type recipients. After an 8-week recovery period, mice exercised for 6 weeks or remained sedentary. (f) Leptin levels in serum by ELISA (**p = 0.0059, n = 11 animals for sedentary, n = 10 for exercise, 2 independent experiments, two-tailed Student’s t-test). (g) Circulating leukocyte levels at Zeitgeber time 7 (**p = 0.0042, n = 11 for animals sedentary, n = 10 for exercise, 2 independent experiments, two-tailed Student’s t-test). (h) BrdU incorporation into LSK 22 h after intraperitoneal injection (*p = 0.045, n = 10 animals sedentary, n = 9 for exercise, 2 independent experiments, two-tailed Student’s t-test). (i) Gene expression by qPCR in total bone marrow of Cxcl12 (*p = 0.042), Vcam1 (*p = 0.03), Kitl (*p = 0.014) and Angpt1 (* p = 0.0168, n = 11 animals for sedentary, n = 10 for exercise, 2 independent experiments, two-tailed U test for Cxcl12 and two-tailed Student’s t-test for Vcam1, Kitl, Angpt1). mRNA levels were normalized to Actb Ct values. (j) Leptin levels in blood of Leprfl/f and Prrx1-creERT2:Leprfl/fl mice measured by ELISA (n = 8 animals for Leprfl/fl and n = 11 for Prrx1-creERT2:Leprfl/fl). (k) Representative microCT images of the proximal metaphysis and mid-diaphysis tibia of Prrx1-creERT2:Leprfl/fl mice and their Leprfl/fl littermates. (b) Parameters of bone microstructure, including trabecular and cortical thickness, bone mineral density and polar moment of inertia by µCT (n = 3 animals per group). Data are mean ± s.e.m. We acknowledge Servier Medical Art (https://smart.servier.com) for providing images of mice and components of the cartoon.

Extended Data Fig. 7 Exercise effects wane after 6 sedentary weeks.

(a) Experimental outline for b-c. (b) Blood (*p = 0.012, ***p = 8.89 × 10−5) and tibial (**p = 0.0053, ***p = 0.00079) leptin concentrations measured by ELISA (n = 9 animals for sedentary and exercise, n = 13 animals for post-exercise-sedentary, 3 independent experiments, with Dunn’s post hoc test). (c) Gene expression of niche factors Cxcl12 (*p = 0.015), Vcam1 (*p = 0.038), Kitl (**p = 0.0037 sedentary versus exercise, **p = 0.0035 exercise versus post-exercise-sedentary) and Angpt1 (*p = 0.027) in whole bone marrow by qPCR (n = 9 animals for sedentary and exercise, n = 13 animals for post-exercise-sedentary, 3 independent experiments, one-way analysis of variance with Sidak’s post hoc test). mRNA levels were normalized to Actb Ct values. (d) Experimental outline for e. The post-exercise-sedentary group had access to exercise wheels for 6 weeks after which the wheels were removed for the following 6 weeks. Sedentary controls had no access, while the exercise group had access to wheels during the last 6 weeks before sacrifice. (e) Circulating leukocyte levels at Zeitgeber time 7 (*p = 0.028 sedentary versus exercise, *p = 0.045 exercise versus post-exercise sedentary) and BrdU incorporation into LSK 22 h after intraperitoneal injection (*p = 0.045 sedentary versus exercise, *p = 0.014 exercise versus post-exercise-sedentary, n = 6 animals per group, with Dunn’s post hoc test). (f) Outline of the competitive bone marrow transplantation experiments. LSK were isolated from CD45.2 donors that either exercised for 6 weeks or were sedentary. These were transplanted in a 1:1 ratio into CD45.1 recipients together with LSK isolated from Ubc-GFP mice that exercised for 6 weeks and had a 3-week post-exercise-sedentary period. Blood chimerism 8 weeks after transplantation (n = 4 animals per group, p = 0.12 for exercise versus post-exercise-sedentary donor chimerism, Wilcoxon matched-pairs signed rank test). Data are mean ± s.e.m. We acknowledge Servier Medical Art (https://smart.servier.com) for providing images of mice and components of the cartoon.

Extended Data Fig. 8 Background ATAC-seq signals are similar, while peaks are higher in LSK of sedentary mice.

(a) Average profiles of ATAC-seq tag density among randomly shuffled regions of the same size as the actual ATAC-seq peaks. These profiles are similar among different conditions, suggesting the absence of background shift between ATAC-seq signals. (b) Tracks of normalized ATAC-seq tag density for the loci of additional genes in the top ten significant genes in the cell cycle category as determined by DAVID in Fig. 3n. (c) Scatter plot of normalized tag density at ATAC-seq peaks shows comparison between LSK from sedentary versus post-exercise-sedentary cohorts. Peaks with significantly lower and higher tag density in post-running mice are highlighted in orange and black, respectively (FDR < 0.01). The top ten significant genes in the cell cycle pathway determined by DAVID (refer to d) and Mki67 are indicated; see Supplementary Table 1 for all genes. (d) Functional categories enriched among genes with differential chromatin accessibility in LSK from sedentary versus post-exercise-sedentary mice as determined by DAVID. (e) Tracks of normalized ATAC-seq tag density for the loci of the top ten significant genes in the cell cycle category as determined by DAVID in d.

Extended Data Fig. 9 Leptin in acute MI.

(a) Experimental outline for b-e. (b) Leptin blood levels on day 6 after MI (n = 5 animals for Sed-Saline and Ex-Saline, n = 6 for Ex-Leptin, 3 independent experiments). (c) Infarct CD45+ leukocyte levels on day 6 after MI (*p = 0.025, n = 5 animals for Sed-Saline and Ex-Saline, n = 6 for Ex-Leptin, 3 independent experiments, with Dunn’s post hoc test). (d) Flow cytometry gating and quantification of neutrophils (*p = 0.039 Sed-Saline versus Ex-Leptin, *p = 0.021 Ex-Saline versus Ex-Leptin), monocytes (*p = 0.018 Sed-Saline versus Ex-Leptin, *p = 0.015 Ex-Saline versus Ex-Leptin), macrophages and lymphocytes in the infarct in respective cohorts (n = 5 animals for Sed-Saline and Ex-Saline, n = 6 for Ex-Leptin, 3 independent experiments, with Dunn’s post hoc test). (e) Cardiac magnetic resonance imaging on day 21 after MI. Ejection fraction (EF), enddiastolic volume (EDV), endsystolic volume (ESV) and left ventricular (LV) mass were determined (n = 7 animals for Sed-Saline, n = 5 for Ex-Saline, n = 8 for Ex-Leptin, 3 independent experiments). (f) Experimental outline for panels g-j. (g) Circulating leukocytes at Zeitgeber 7 (**p = 0.0017, n = 8 animals for IgG and n = 10 for αLep, 3 independent experiments, two-tailed Student’s). (h) BrdU incorporation into granulocyte macrophage progenitors (GMP) 3 days after MI (p = 0.05, n = 8 animals for IgG and n = 10 for αLep, 3 independent experiments, two-tailed Student’s t-test). (i) Bone marrow unit assay (CFU) of bone marrow mononuclear cells (BMNCs) for complete colonies (***p = 0.00041, n = 7 animals for IgG and n = 10 for αLep, 3 independent experiments, two-tailed U test). (j) Neutrophils and monocytes per mg infarct tissue (*p = 0.03, n = 5 animals for IgG, n = 6 for αLep, two-tailed U test). (k) Experimental outline for l. (l) Representative immunohistochemical stainings and quantification of myeloid cells (CD11b), collagen deposition (Collagen I), and myofibroblasts (alpha smooth muscle actin) in the infarct border zone (*p = 0.026 for CD11b and Collagen I, n = 6 animals per group, two-tailed U test). Data are mean ± s.e.m. We acknowledge Servier Medical Art (https://smart.servier.com) for providing images of mice and components of the cartoon.

Extended Data Fig. 10 Stromal leptin receptor deletion attenuates atherosclerosis, inflammation and hematopoiesis.

(a) Experimental outline for b-h. Leprfl/fl mice and Prrx1-creERT2:Leprfl/fl littermates were injected with tamoxifen and received a single IV injection of AAV-PCSK9 followed by a high fat diet for 12 weeks. (b) Representative cross sections of aortic roots stained with Oil red O and assessment of lesion size (*p = 0.042, n = 8 animals per group, two-tailed Student’s t-test). (c) Flow cytometry enumeration of myeloid cells in aortas of Leprfl/fl and Prrx1-creERT2:Leprfl/fl mice (*p = 0.023, n = 8 animals per group, two-tailed Student’s t-test). (d) CD68 histological staining of aortic root lesions. Percentage of positive staining per plaque (*p = 0.029, n = 6 animals for Leprfl/fl, n = 8 for Prrx1-creERT2:Leprfl/fl, two-tailed U test). (e) Representative flow plots and statistical analysis of long-term hematopoietic stem cells (LT-HSC) in femur bone marrow (*p = 0.046, n = 9 animals per group, two-tailed Student’s t-test). (f) Bone marrow unit assay for complete colonies (CFU-C) of bone marrow mononuclear cells (BMNCs) (*p = 0.036, n = 9 animals per group, two-tailed Student’s t-test). (g) BrdU incorporation assay 22 hours after intraperitoneal injection for LT-HSC and progenitors (GMP) proliferation (*p = 0.027 for LT-HSC, *p = 0.017 for GMP, n = 9 animals per group, two-tailed Student’s t-test). (h) Circulating myeloid cells at Zeitgeber time 7 (*p = 0.014 for neutrophils, *p = 0.046 for monocytes, n = 10 animals for Leprfl/fl, n = 9 for Prrx1-creERT2:Leprfl/fl, two-tailed Student’s t-test). Data are mean ± s.e.m. (i) Athero-express cohort. The illustrates inclusion criteria for patients and separation into sedentary lifestyle and exercise groups. We acknowledge Servier Medical Art (https://smart.servier.com) for providing images of mice and components of the cartoon.

Supplementary information

Supplementary Tables

Supplementary Tables 1–5

Reporting Summary

Source data

Source Data Fig. 1

Statistical source information

Source Data Fig. 2

Statistical source information

Source Data Fig. 3

Statistical source information

Source Data Fig. 4

Statistical source information

Source Data Fig. 5

Statistical source information

Source Data Fig. 6

Statistical source information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Frodermann, V., Rohde, D., Courties, G. et al. Exercise reduces inflammatory cell production and cardiovascular inflammation via instruction of hematopoietic progenitor cells. Nat Med 25, 1761–1771 (2019). https://doi.org/10.1038/s41591-019-0633-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41591-019-0633-x

This article is cited by

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