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.

  • Letter
  • Published:

Sleep modulates haematopoiesis and protects against atherosclerosis

Abstract

Sleep is integral to life1. Although insufficient or disrupted sleep increases the risk of multiple pathological conditions, including cardiovascular disease2, we know little about the cellular and molecular mechanisms by which sleep maintains cardiovascular health. Here we report that sleep regulates haematopoiesis and protects against atherosclerosis in mice. We show that mice subjected to sleep fragmentation produce more Ly-6Chigh monocytes, develop larger atherosclerotic lesions and produce less hypocretin—a stimulatory and wake-promoting neuropeptide—in the lateral hypothalamus. Hypocretin controls myelopoiesis by restricting the production of CSF1 by hypocretin-receptor-expressing pre-neutrophils in the bone marrow. Whereas hypocretin-null and haematopoietic hypocretin-receptor-null mice develop monocytosis and accelerated atherosclerosis, sleep-fragmented mice with either haematopoietic CSF1 deficiency or hypocretin supplementation have reduced numbers of circulating monocytes and smaller atherosclerotic lesions. Together, these results identify a neuro-immune axis that links sleep to haematopoiesis and atherosclerosis.

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: Sleep fragmentation aggravates atherosclerosis, increases haematopoiesis and decreases production of hypocretin in the hypothalamus.
Fig. 2: Hypocretin suppresses haematopoiesis and atherosclerosis.
Fig. 3: Hypocretin controls production of CSF1 by pre-neutrophils in the bone marrow.
Fig. 4: Haematopoietic CSF1 deletion or hypocretin supplementation protects against sleep fragmentation-induced haematopoiesis and atherosclerosis.

Similar content being viewed by others

Data availability

All relevant data are included in the paper and its Supplementary Information. Source Data for Figs. 14 are available in the online version of the paper.

References

  1. Hublin, C., Partinen, M., Koskenvuo, M. & Kaprio, J. Sleep and mortality: a population-based 22-year follow-up study. Sleep 30, 1245–1253 (2007).

    Article  Google Scholar 

  2. Cappuccio, F. P., Cooper, D., D’Elia, L., Strazzullo, P. & Miller, M. A. Sleep duration predicts cardiovascular outcomes: a systematic review and meta-analysis of prospective studies. Eur. Heart J. 32, 1484–1492 (2011).

    Article  Google Scholar 

  3. Hafner, M., Stepanek, M., Taylor, J., Troxel, W. M. & van Stolk, C. Why sleep matters—the economic costs of insufficient sleep: a cross-country comparative analysis. Rand Health Q. 6, 11 (2017).

    PubMed  PubMed Central  Google Scholar 

  4. Ford, E. S., Cunningham, T. J. & Croft, J. B. Trends in self-reported sleep duration among US adults from 1985 to 2012. Sleep 38, 829–832 (2015).

    Article  Google Scholar 

  5. Cappuccio, F. P. et al. Meta-analysis of short sleep duration and obesity in children and adults. Sleep 31, 619–626 (2008).

    Article  Google Scholar 

  6. Shan, Z. et al. Sleep duration and risk of type 2 diabetes: a meta-analysis of prospective studies. Diabetes Care 38, 529–537 (2015).

    Article  Google Scholar 

  7. Blask, D. E. Melatonin, sleep disturbance and cancer risk. Sleep Med. Rev. 13, 257–264 (2009).

    Article  Google Scholar 

  8. Carreras, A. et al. Chronic sleep fragmentation induces endothelial dysfunction and structural vascular changes in mice. Sleep 37, 1817–1824 (2014).

    Article  Google Scholar 

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

    Article  ADS  CAS  Google Scholar 

  10. Scheiermann, C. et al. Adrenergic nerves govern circadian leukocyte recruitment to tissues. Immunity 37, 290–301 (2012).

    Article  CAS  Google Scholar 

  11. He, W. et al. Circadian expression of migratory factors establishes lineage-specific signatures that guide the homing of leukocyte subsets to tissues. Immunity 49, 1175–1190 (2018).

    Article  CAS  Google Scholar 

  12. Lasselin, J., Rehman, J. U., Åkerstedt, T., Lekander, M. & Axelsson, J. Effect of long-term sleep restriction and subsequent recovery sleep on the diurnal rhythms of white blood cell subpopulations. Brain Behav. Immun. 47, 93–99 (2015).

    Article  Google Scholar 

  13. Geovanini, G. R. et al.; Association between obstructive sleep apnea and cardiovascular risk factors: variation by age, sex, and race. The multi-ethnic study of atherosclerosis. Ann. Am. Thorac. Soc. 15, 970–977 (2018).

    Article  Google Scholar 

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

    Article  CAS  Google Scholar 

  15. Li, X., Marchant, N. J. & Shaham, Y. Opposing roles of cotransmission of dynorphin and hypocretin on reward and motivation. Proc. Natl Acad. Sci. USA 111, 5765–5766 (2014).

    Article  ADS  CAS  Google Scholar 

  16. Fu, L. Y., Acuna-Goycolea, C. & van den Pol, A. N. Neuropeptide Y inhibits hypocretin/orexin neurons by multiple presynaptic and postsynaptic mechanisms: tonic depression of the hypothalamic arousal system. J. Neurosci. 24, 8741–8751 (2004).

    Article  CAS  Google Scholar 

  17. Scammell, T. E., Arrigoni, E. & Lipton, J. O. Neural circuitry of wakefulness and sleep. Neuron 93, 747–765 (2017).

    Article  CAS  Google Scholar 

  18. Latorre, D. et al. T cells in patients with narcolepsy target self-antigens of hypocretin neurons. Nature 562, 63–68 (2018).

    Article  ADS  CAS  Google Scholar 

  19. Hartmann, F. J. et al. High-dimensional single-cell analysis reveals the immune signature of narcolepsy. J. Exp. Med. 213, 2621–2633 (2016).

    Article  CAS  Google Scholar 

  20. Ibrahim, N. E. et al. circulating concentrations of orexin A predict left ventricular myocardial remodeling. J. Am. Coll. Cardiol. 68, 2238–2240 (2016).

    Article  CAS  Google Scholar 

  21. Perez, M. V. et al. Systems genomics identifies a key role for hypocretin/orexin receptor-2 in human heart failure. J. Am. Coll. Cardiol. 66, 2522–2533 (2015).

    Article  CAS  Google Scholar 

  22. Adam, J. A. et al. Decreased plasma orexin-A levels in obese individuals. Int. J. Obes. Relat. Metab. Disord. 26, 274–276 (2002).

    Article  CAS  Google Scholar 

  23. Ohayon, M. M. Narcolepsy is complicated by high medical and psychiatric comorbidities: a comparison with the general population. Sleep Med. 14, 488–492 (2013).

    Article  Google Scholar 

  24. Mochizuki, T. et al. Behavioral state instability in orexin knock-out mice. J. Neurosci. 24, 6291–6300 (2004).

    Article  CAS  Google Scholar 

  25. Sellayah, D., Bharaj, P. & Sikder, D. Orexin is required for brown adipose tissue development, differentiation, and function. Cell Metab. 14, 478–490 (2011).

    Article  CAS  Google Scholar 

  26. Aspelund, A. et al. A dural lymphatic vascular system that drains brain interstitial fluid and macromolecules. J. Exp. Med. 212, 991–999 (2015).

    Article  CAS  Google Scholar 

  27. Louveau, A. et al. Structural and functional features of central nervous system lymphatic vessels. Nature 523, 337–341 (2015).

    Article  ADS  Google Scholar 

  28. Evrard, M. et al. Developmental analysis of bone marrow neutrophils reveals populations specialized in expansion, trafficking, and effector functions. Immunity 48, 364–379 (2018).

    Article  CAS  Google Scholar 

  29. Li, S., Franken, P. & Vassalli, A. Bidirectional and context-dependent changes in theta and gamma oscillatory brain activity in noradrenergic cell-specific hypocretin/orexin receptor 1-KO mice. Sci. Rep. 8, 15474 (2018).

    Article  ADS  Google Scholar 

  30. Mossadegh-Keller, N. et al. M-CSF instructs myeloid lineage fate in single haematopoietic stem cells. Nature 497, 239–243 (2013).

    Article  ADS  CAS  Google Scholar 

  31. Chemelli, R. M. et al. Narcolepsy in orexin knockout mice: molecular genetics of sleep regulation. Cell 98, 437–451 (1999).

    Article  CAS  Google Scholar 

  32. Vassalli, A., Li, S. & Tafti, M. Comment on “Antibodies to influenza nucleoprotein cross-react with human hypocretin receptor 2”. Sci. Transl. Med. 7, 314le2 (2015).

    Article  Google Scholar 

  33. 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).

    Article  CAS  Google Scholar 

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

    Article  ADS  Google Scholar 

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

    Article  ADS  CAS  Google Scholar 

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

    Article  ADS  CAS  Google Scholar 

  37. 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).

    Article  CAS  Google Scholar 

  38. Swirski, F. K. et al. Identification of splenic reservoir monocytes and their deployment to inflammatory sites. Science 325, 612–616 (2009).

    Article  ADS  CAS  Google Scholar 

  39. Robbins, C. S. et al. Local proliferation dominates lesional macrophage accumulation in atherosclerosis. Nat. Med. 19, 1166–1172 (2013).

    Article  CAS  Google Scholar 

  40. Anzai, A. et al. The infarcted myocardium solicits GM-CSF for the detrimental oversupply of inflammatory leukocytes. J. Exp. Med. 214, 3293–3310 (2017).

    Article  CAS  Google Scholar 

  41. Da Mesquita, S. et al. Functional aspects of meningeal lymphatics in ageing and Alzheimer’s disease. Nature 560, 185–191 (2018).

    Article  ADS  Google Scholar 

  42. Ono, T., Kanbayashi, T., Yoshizawa, K., Nishino, S. & Shimizu, T. Measurement of cerebrospinal fluid orexin-A (hypocretin-1) by enzyme-linked immunosorbent assay: a comparison with radioimmunoassay. Psychiatry Clin. Neurosci. 72, 849–850 (2018).

    Article  Google Scholar 

  43. Refinetti, R., Lissen, G. C. & Halberg, F. Procedures for numerical analysis of circadian rhythms. Biol. Rhythm Res. 38, 275–325 (2007).

    Article  Google Scholar 

Download references

Acknowledgements

This work was supported in part by NIH grants R35 HL135752, R01 HL128264, P01 HL131478, an American Heart Association Established Investigator Award, and the Patricia and Scott Eston MGH Research Scholar (to F.K.S.); NIH grant R35 HL139598 (to M.N.); Swiss National Science Foundation grants 31003A_125323 and 31003A_144282 (to A.V.); a CIHR postdoctoral fellowship and a Banting postdoctoral fellowship (to C.S.M.); the doctoral program Cell Communication in Health and Disease (CCHD) funded by the Austrian Science Fund (to M.G.K.); a Swedish Research Council postdoctoral fellowship (to S.R.); an American Heart Association postdoctoral fellowship (to S.H.); a postdoctoral fellowship from the Fondation pour la Recherche Medicale (to C.V.); the German Research Foundation (DFG; 331536185 to F.K. and 398190272 to W.C.P.); and a Boehringer-Ingelheim-Fonds MD fellowship (to L.H.). We thank A. Lichtman for providing the PCSK9 adenovirus, D. Scadden for providing stromal cell reporter mice and K. Joyes for editing the manuscript.

Reviewer information

Nature thanks P. S. Frenette, V. Papayannopoulos, A. R. Tall and A. Yamanaka for their contribution to the peer review of this work.

Author information

Authors and Affiliations

Authors

Contributions

C.S.M. conceived the project, designed and performed experiments, analysed and interpreted data, made the figures and wrote the manuscript; M.G.K. designed and performed experiments, and analysed and interpreted data; S.R., S.H., A.V., A.A., C.V., C.T.C., J.E.M., F.K., W.C.P., V.F., A.M.F., A.F.G., L.H., Y.I. and F.F.H. performed experiments; A.V., C.J.B., P.L., M.T., T.E.S. and M.N. provided intellectual input and edited the manuscript; A.V., M.T. and T.E.S. provided materials; F.K.S. conceived the project, designed experiments, interpreted data and wrote the manuscript.

Corresponding author

Correspondence to Filip K. Swirski.

Ethics declarations

Competing interests

The authors declare no competing interests.

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 Effects of sleep fragmentation on metabolic and cellular parameters.

a, Image of a sleep fragmentation cage. b, Body weight (n = 10 per group). c, Plasma cholesterol at ZT3 (n = 5 per group). d, Plasma glucose at ZT3 (n = 5 per group). e, Glucose tolerance test (GTT) beginning at ZT3 (light period) and ZT12 (dark period) (n = 4 per group). fh, Apoe/ mice were placed in sleep fragmentation chambers where the sweep bar operated during the dark period (ZT12–0) when mice are normally awake. Control mice were maintained in sleep fragmentation chambers with a stationary sweep bar. f, Assessment of atherosclerosis and lesion area (n = 5 per group). g, Assessment of blood Ly-6Chigh monocytes and neutrophils (n = 5 per group). h, Assessment of bone marrow LSK cells and proliferation (n = 5 per group). i, Aortic macrophage proliferation in Apoe/ and Apoe/ SF mice after 16 weeks of sleep fragmentation at ZT3 and ZT14 (n = 5 Apoe/ mice; n = 4 Apoe/ SF mice). j, Quantification of B cells, CD4+ T cells and CD8+ T cells in the blood of Apoe−/− and Apoe−/− SF mice at ZT3 (n = 10 Apoe/ mice; for B and CD4 T cells, n = 6 Apoe/ SF mice and for CD8 T cells, n = 7 Apoe/ SF mice). k, Quantification of B cells, CD4+ T cells and CD8+ T cells in the spleen of Apoe−/− and Apoe−/− SF mice at ZT3 (n = 10 Apoe/ mice; n = 7 Apoe/ SF mice). l, Quantification of B cells in the bone marrow of Apoe−/− and Apoe−/− SF mice at ZT3 (n = 10 Apoe−/− mice; n = 7 Apoe/ SF mice). Data are mean ± s.e.m.

Extended Data Fig. 2 Sleep and circadian migration of leukocytes.

ad, Quantification of Ly-6Chigh monocytes and neutrophils at ZT3 and ZT14 in the spleen (a), bone marrow (b), lung (c) and liver (d) of Apoe−/− mice and Apoe−/− mice after 16 weeks of sleep fragmentation. Group sizes are indicated in the figure. Data are mean ± s.e.m., *P < 0.05, **P < 0.01, two-way ANOVA.

Extended Data Fig. 3 Sleep-mediated haematopoiesis and extramedullary haematopoiesis.

a, Gating strategy and quantification of haematopoietic progenitor cells at ZT3 in the bone marrow (n = 10 Apoe−/− mice, except for GMPs, n = 11 mice; n = 10 Apoe/ SF mice, except MPP3 and MPP4, n = 9 mice). LtHSCs, long-term haematopoietic stem cells; StHSC, short-term haematopoietic stem cells. b, Gating strategy and quantification of haematopoietic progenitor cells at ZT3 in the spleen (n = 9 Apoe−/− mice, except CMPs, n = 10 mice; n = 9 Apoe−/− SF mice). c, C57BL/6 wild-type mice that were fed a regular chow diet were subjected to sleep fragmentation for 16 weeks after which Ly-6Chigh monocytes, neutrophils and LSK cells were quantified at ZT3 (n = 8 wild-type mice, except n = 9 mice for spleen Ly-6Chigh monocytes, n = 5 mice for bone marrow neutrophils, n = 10 mice for bone marrow LSK cells; n = 4 wild-type SF mice, except n = 9 mice for bone marrow LSK cells). Data are mean ± s.e.m., *P < 0.05, **P < 0.01, ***P < 0.001, two-tailed Mann–Whitney U-tests.

Extended Data Fig. 4 Sleep fragmentation does not alter bone structure and does not depend on the microbiome.

a, b, μCT analysis of trabeculae (a) and cortical bone structure (b) of Apoe−/− mice and Apoe−/− mice after 16 weeks of sleep fragmentation. The bone volume fraction (BV/TV), bone mineral density (BMD), trabecular number (Tb.N), trabecular thickness (Tb.Th), structural model index (SMI), cortical tissue mineral density (Ct.TMD), cortical area (Ct.Ar), total area (T.Ar), cortical thickness (Ct.Th) and cortical porosity (Ct.Porosity) were analysed (n = 9 per group). c, d, Analysis of leukocytosis in SF (c) and Hcrt−/− (d) mice at ZT3 after receiving a cocktail of antibiotics in drinking water for 4 weeks (n = 3 per group). Data are mean ± s.e.m., *P < 0.05, **P < 0.01, ***P < 0.001, two-tailed Mann–Whitney U-tests.

Extended Data Fig. 5 Sleep fragmentation does not activate the peripheral sympathetic nervous system but has effects on hypothalamic gene transcription and food consumption.

a, Plasma corticosterone levels in Apoe−/− mice and Apoe−/− mice after 16 weeks of sleep fragmentation at ZT3 and ZT14 (n = 4 mice per group, except n = 5 Apoe−/− mice at ZT3). b, Systolic and diastolic blood pressure at ZT3 (n = 4 mice per group). c, Immunohistochemical analysis and quantification of tyrosine hydroxylaze (TH) staining in the bone marrow of Apoe−/− mice, Apoe−/− SF mice and Apoe−/− mice subjected to 3 weeks of chronic variable stress (n = 4 Apoe−/− mice; n = 4 Apoe−/− SF mice; n = 3 Apoe−/− stressed mice). d, Quantification at ZT3 of blood Ly-6Chigh monocytes and neutrophils, bone marrow LSK cells and proliferation in Apoe−/− mice and Apoe−/− SF mice after antagonism of the β3 receptor for 4 weeks (n = 3 Apoe−/− mice treated with β3 blocker; n = 4 Apoe−/− SF mice treated with β3 blocker). e, Quantification of time in outer zone during open field test (n = 9 Apoe−/− mice; n = 8 Apoe−/− SF mice). f, Quantification of time spent in light box during light–dark box test (n = 6 per group). g, Quantification of time in new arm during Y-maze test (n = 8 Apoe−/− mice; n = 5 Apoe−/− SF mice). h, Analysis of neuropeptide expression in the hypothalamus at ZT3 (n = 5 Apoe−/− mice, except n = 6 for Pmch, Tph2, Gad1and Npy; n = 5 Apoe−/− SF mice, except n = 4 for Npy). i, Neuropeptide receptor expression in the hypothalamus at ZT3 (n = 6 Apoe−/− mice, except Hcrtr1, n = 10 mice; n = 5 Apoe−/− SF mice, except Hcrtr2, n = 6 mice). j, Circadian gene expression in the hypothalamus at ZT3 and ZT14 (n = 3 Apoe−/− mice; n = 4 Apoe−/− SF mice). k, Mouse food consumption during the course of sleep fragmentation (n = 4 Apoe−/− mice at ZT3, except n = 6 Apoe−/− mice on HFD for 16 weeks at ZT3; n = 4 Apoe−/− SF mice at ZT3, except n = 6 Apoe−/− SF mice on HFD for 16 weeks at ZT3; n = 5 Apoe−/− mice at ZT14, except n = 4 Apoe−/− mice on HFD for 10 weeks at ZT14 and n = 6 Apoe−/− mice on HFD for 16 weeks at ZT14; n = 5 Apoe−/− SF mice at ZT14, except n = 4 Apoe−/− SF mice on HFD for 10 weeks at ZT14 and n = 6 Apoe−/− SF mice on HFD for 16 weeks at ZT14). Data are mean ± s.e.m., *P < 0.05, **P < 0.01, two-tailed Mann–Whitney U-tests. ns, not significant.

Extended Data Fig. 6 Hypothalamic expression of hypocretin and dynorphin.

a, Hypothalamic expression of hypocretin and quantification of blood Ly-6Chigh monocytes and neutrophils in Apoe−/− mice after 6, 8 and 12 weeks of sleep fragmentation (for Hcrt, n = 4 Apoe−/− mice, except n = 5 for Apoe−/− mice after 12 weeks; for Hcrt, n = 4 Apoe−/− SF mice; for blood cells at 6 weeks, n = 5 mice per group; for blood cells at 8 weeks, n = 4 mice per group; for blood cells at 12 weeks, n = 9 mice per group). b, Sections of the hypothalamus stained for dynorphin and hypocretin. c, Quantification of hypothalamic dynorphin+ cells per high-powered field of view (n = 5 Apoe−/− mice; n = 4 Apoe−/− SF mice, of two independent experiments). d, e, Dynorphin (Pdny) mRNA expression in the hypothalamus of SF mice (d; n = 6 Apoe−/− mice; n = 5 Apoe−/− SF mice) and Hcrt−/− mice (e; n = 4 wild-type mice; n = 5 Hcrt−/− mice). f, TUNEL staining of hypothalamic sections from Apoe−/− and Apoe−/− SF mice (representative of four biological replicates) along with a positive control of TUNEL-stained myocardium 1 day after myocardial infarction (MI) (n = 1). gi, Apoe−/− mice were sleep-fragmented for 16 weeks then allowed to recover and sleep normally for 10 weeks. Control mice slept normally for 26 weeks. g, Analysis of hypothalamic hypocretin expression (n = 5 Apoe−/− mice; n = 4 Apoe−/− SF mice). h, Blood Ly-6Chigh monocytes and neutrophils (n = 5 mice per group). i, Quantification of bone marrow LSK cells and proliferation of LSK cells (n = 5 mice per group). Data are mean ± s.e.m., *P < 0.05, **P < 0.01, two-tailed Mann–Whitney U-tests.

Extended Data Fig. 7 Haematopoiesis in hypocretin-deficient mice.

ac, Quantification of the number of leukocytes in wild-type and Hcrt−/− mice at ZT3 in blood (a; n = 5 mice per group), spleen (b; for Ly-6Chigh monocytes and neutrophils, n = 7 wild-type mice and n = 8 Hcrt−/− mice; for B, CD8+ T, and CD4+ T cells, n = 5 wild-type mice and n = 6 Hcrt−/− mice) and bone marrow (c; for Ly-6Chigh monocytes and neutrophils, n = 7 wild-type mice and n = 8 Hcrt−/− mice; for B and T cells, n = 5 wild-type mice and n = 6 Hcrt−/− mice; for CMPs, GMPs and MDPs, n = 7 wild-type mice and n = 8 Hcrt−/− mice; for LSK populations, n = 5 wild-type mice and n = 6 Hcrt−/− mice). Data are mean ± s.e.m., **P < 0.01, ***P < 0.001, two-tailed Mann–Whitney U-tests.

Extended Data Fig. 8 Hypocretin and hypocretin receptor-1 expression and production.

a, b, Relative Hcrt mRNA expression in tissues (a; n = 3) and sorted bone marrow cells (b; n = 4). c, Hcrt expression in the bone marrow and bone in Apoe−/− mice and in Apoe−/− mice subjected to sleep fragmentation for 16 weeks (n = 5 Apoe−/− mice; n = 4 Apoe−/− SF mice). d, Hypocretin-1 protein levels in cerebrospinal fluid (CSF), plasma and bone marrow (BM) fluid of wild-type and Hcrt−/− mice (n = 4 mice per group). e, Hypocretin-1 protein levels in the plasma and bone marrow fluid of Hcrt−/− mice 3 h after intra-cisterna magna (i.c.m.) injection of HCRT-1 or PBS. (n = 4 mice per group). f, Relative Hcrtr1 mRNA expression in tissues (n = 4 mice, except aorta and spleen, n = 3). g, Hcrtr2 expression in sorted bone marrow cells (n = 4 mice). h, Granulocyte–macrophage colony forming units (CFU-GM) from bone marrow cells of wild-type mice exposed to hypocretin-1 ex vivo in culture medium (n = 3 per group). i, Assessment of hypocretin receptor-1 protein in hypothalamus and sorted bone marrow neutrophils by western blot. Data are mean ± s.e.m.

Extended Data Fig. 9 Hypocretin, bone marrow neutrophils and HCRTR1.

a, Flow cytometry gating strategy for bone marrow pre-neutrophils, immature neutrophils and mature neutrophils. b, HCRTR1 (GFP) in bone marrow and blood neutrophils from WT;bmHcrtr1GFP/GFP mice. c, mRNA expression in cultured bone marrow pre-neutrophils exposed to LPS and/or HCRT-1 (for untreated n = 3 mice, except Mpo, n = 6 mice; for HCRT-1, n = 3 mice, expect Csf1, n = 4 mice and Mpo, n = 6 mice; for LPS, n = 3 mice except Csf1, n = 7 mice and Mpo, n = 6 mice; for LPS and HCRT-1, n = 3 mice, except Csf1, n = 11 mice, Csf2, n = 4 mice and Mpo, n = 6 mice). d, Csf1 expression in sorted bone marrow cells of wild-type and Hcrt−/− mice (n = 5 wild-type mice; n = 6 Hcrt−/− mice). e, Analysis of mRNA transcript expression in bone marrow leukocytes of Apoe−/− mice after 16 weeks of sleep fragmentation (for Apoe−/−, n = 5 mice, except Il10, Il34, Cxcl12, Csf3, n = 4 mice and Csf1, n = 9 mice; for Apoe−/− SF mice, n = 6 mice, except Il5, Il1β, Il6, Il34, Cxcl12, Csf3, n = 5, Il10, n = 5 mice and Csf1, n = 12). nd, not detected. f, Blood neutrophils in WT;bmHcrtr1GFP/GFP mice over 24 h (n = 3 per group). Data presented as mean ± s.e.m., **P < 0.01, one-way ANOVA.

Extended Data Fig. 10 Haematopoietic CSF1 deletion protects against haematopoiesis and atherosclerosis in hypocretin-deficient mice.

a, Schematic of chimeric models. b, c, Quantification of Ly-6Chigh monocytes and neutrophils in blood (b; n = 4 WT;bmWT mice; n = 6 Hcrt−/−;bmWT mice; n = 3 WT;bmCsf1−/− mice; n = 5 Hcrt−/−;bmCsf1−/− mice) and bone marrow (c; n = 4 WT;bmWT mice; n = 6 Hcrt−/−;bmWT mice; n = 3 WT;bmCsf1−/− mice; n = 5 Hcrt−/−;bmCsf1−/− mice). d, Quantification of the number of LSK cells (n = 4 WT;bmWT mice; n = 6 Hcrt−/−;bmWT mice; n = 3 WT;bmCsf1−/− mice; n = 5 Hcrt−/−;bmCsf1−/− mice) and proliferation (n = 4 WT;bmWT mice; n = 4 Hcrt−/−;bmWT mice; n = 3 WT;bmCsf1−/− mice; n = 5 Hcrt−/−;bmCsf1−/− mice) in bone marrow. e, Quantification of CMPs, GMPs and MDPs in chimeric mice (n = 4 WT;bmWT mice; n = 6 Hcrt−/−;bmWT mice; n = 3 WT;bmCsf1−/− mice; n = 5 Hcrt−/−;bmCsf1−/− mice). f, Bone marrow CSF1 levels (n = 4 WT;bmWT mice; n = 8 Hcrt−/−;bmWT mice; n = 4 WT;bmCsf1−/− mice; n = 7 Hcrt−/−;bmCsf1−/− mice). g, Schematic of chimeric models receiving Adv-PCSK9 and fed a HCD for 12 weeks. h, Plasma cholesterol levels (n = 7 WT;bmWT mice; n = 10 Hcrt−/−;bmWT mice; n = 5 WT;bmCsf1−/− mice; n = 6 Hcrt−/−;bmCsf1−/− mice). i, Images of cross-sections of aortic roots stained with oil red O and quantification of atherosclerosis in chimeric mice (n = 7 WT;bmWT mice; n = 9 Hcrt−/−;bmWT mice; n = 5 WT;bmCsf1−/− mice; n = 6 Hcrt−/−;bmCsf1−/− mice). Data are mean ± s.e.m., *P < 0.05, **P < 0.01, ***P < 0.001, one-way ANOVA.

Supplementary information

Reporting Summary

Video 1

Video demonstrating the sleep fragmentation (SF) chambers used in the study (Lafayette Instrument, Lafayette, IN). The sweep bar moves along the bottom of the cage every 2 minutes gently waking the mice.

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

McAlpine, C.S., Kiss, M.G., Rattik, S. et al. Sleep modulates haematopoiesis and protects against atherosclerosis. Nature 566, 383–387 (2019). https://doi.org/10.1038/s41586-019-0948-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-019-0948-2

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