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:

Myocardial infarction augments sleep to limit cardiac inflammation and damage

Abstract

Sleep is integral to cardiovascular health1,2. Yet, the circuits that connect cardiovascular pathology and sleep are incompletely understood. It remains unclear whether cardiac injury influences sleep and whether sleep-mediated neural outputs contribute to heart healing and inflammation. Here we report that in humans and mice, monocytes are actively recruited to the brain after myocardial infarction (MI) to augment sleep, which suppresses sympathetic outflow to the heart, limiting inflammation and promoting healing. After MI, microglia rapidly recruit circulating monocytes to the brain’s thalamic lateral posterior nucleus (LPN) via the choroid plexus, where they are reprogrammed to generate tumour necrosis factor (TNF). In the thalamic LPN, monocytic TNF engages Tnfrsf1a-expressing glutamatergic neurons to increase slow wave sleep pressure and abundance. Disrupting sleep after MI worsens cardiac function, decreases heart rate variability and causes spontaneous ventricular tachycardia. After MI, disrupting or curtailing sleep by manipulating glutamatergic TNF signalling in the thalamic LPN increases cardiac sympathetic input which signals through the β2-adrenergic receptor of macrophages to promote a chemotactic signature that increases monocyte influx. Poor sleep in the weeks following acute coronary syndrome increases susceptibility to secondary cardiovascular events and reduces the heart’s functional recovery. In parallel, insufficient sleep in humans reprogrammes β2-adrenergic receptor-expressing monocytes towards a chemotactic phenotype, enhancing their migratory capacity. Collectively, our data uncover cardiogenic regulation of sleep after heart injury, which restricts cardiac sympathetic input, limiting inflammation and damage.

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: MI augments sleep.
Fig. 2: Monocytes are recruited to the brain after MI to augment sleep.
Fig. 3: Monocytic TNF engages Tnfrsf1a-expressing glutamatergic neurons in the thalamic LPN to augment sleep after MI.
Fig. 4: Sleep disruption worsens cardiac function and outcome after MI.
Fig. 5: Sleep limits cardiac sympathetic input to curtail macrophage chemotactic programming and recruitment after MI.

Similar content being viewed by others

Data availability

scRNA-seq data have been deposited to NCBI Gene Expression Omnibus under the following accession numbers: GSE275071 for the human PBMC sequencing from the sleep restriction study and GSE275089 for all mouse sequencing. snRNA-seq data for human brain tissue have been deposited to NCBI Gene Expression Omnibus under the accession number GSE227781. All other necessary data are contained within the manuscript. Requests for materials can be made to the corresponding author. Source data are provided with this paper.

References

  1. St-Onge, M.-P. et al. Sleep duration and quality: impact on lifestyle behaviors and cardiometabolic health: a scientific statement from the American Heart Association. Circulation 134, e367–e386 (2016).

    PubMed  PubMed Central  Google Scholar 

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

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  3. Ziegler, K. A. et al. Immune-mediated denervation of the pineal gland underlies sleep disturbance in cardiac disease. Science 381, 285–290 (2023).

    ADS  CAS  PubMed  Google Scholar 

  4. Laugsand, L. E., Vatten, L. J., Platou, C. & Janszky, I. Insomnia and the risk of acute myocardial infarction: a population study. Circulation 124, 2073–2081 (2011).

    PubMed  Google Scholar 

  5. Daghlas, I. et al. Sleep duration and myocardial infarction. J. Am. Coll. Cardiol. 74, 1304–1314 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Clark, A., Lange, T., Hallqvist, J., Jennum, P. & Rod, N. H. Sleep impairment and prognosis of acute myocardial infarction: a prospective cohort study. Sleep 37, 851–858 (2014).

    PubMed  PubMed Central  Google Scholar 

  7. McAlpine, C. S. et al. Sleep exerts lasting effects on hematopoietic stem cell function and diversity. J. Exp. Med. 219, e20220081 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Hsueh, B. et al. Cardiogenic control of affective behavioural state. Nature 615, 292–299 (2023).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  9. Critchley, H. D. & Harrison, N. A. Visceral influences on brain and behavior. Neuron 77, 624–638 (2013).

    CAS  PubMed  Google Scholar 

  10. Mohanta, S. K. et al. Neuroimmune cardiovascular interfaces control atherosclerosis. Nature 605, 152–159 (2022).

    ADS  CAS  PubMed  Google Scholar 

  11. Jin, H., Li, M., Jeong, E., Castro-Martinez, F. & Zuker, C. S. A body–brain circuit that regulates body inflammatory responses. Nature 630, 695–703 (2024).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  12. Wheeler, E. O. & White, P. D. Insomnia due to left ventricular heart failure unrecognized as such and inadequately treated. J. Am. Med. Assoc. 129, 1158–1159 (1945).

    CAS  PubMed  Google Scholar 

  13. Madsen, M. T., Huang, C., Zangger, G., Zwisler, A. D. O. & Gögenur, I. Sleep disturbances in patients with coronary heart disease: a systematic review. J. Clin. Sleep Med. 15, 489 (2019).

    PubMed  PubMed Central  Google Scholar 

  14. Richards, D. A. et al. Distinct phenotypes induced by three degrees of transverse aortic constriction in mice. Sci. Rep. 9, 5844 (2019).

    ADS  PubMed  PubMed Central  Google Scholar 

  15. deAlmeida, A. C., van Oort, R. J. & Wehrens, X. H. T. Transverse aortic constriction in mice. J. Vis. Exp. https://doi.org/10.3791/1729 (2010).

  16. Okamoto-Mizuno, K. & Mizuno, K. Effects of thermal environment on sleep and circadian rhythm. J. Physiol. Anthropol. 31, 14 (2012).

    PubMed  PubMed Central  Google Scholar 

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

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  18. Sager, H. B. et al. Targeting interleukin-1β reduces leukocyte production after acute myocardial infarction. Circulation 132, 1880–1890 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Liu, Z. et al. Fate mapping via Ms4a3-expression history traces monocyte-derived cells. Cell 178, 1509–1525.e19 (2019).

    CAS  PubMed  Google Scholar 

  20. Cathomas, F. et al. Circulating myeloid-derived MMP8 in stress susceptibility and depression. Nature 626, 1108–1115 (2024).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  21. Jacob, L. et al. Conserved meningeal lymphatic drainage circuits in mice and humans. J. Exp. Med. 219, e20220035 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Kirst, C. et al. Mapping the fine-scale organization and plasticity of the brain vasculature. Cell 180, 780–795.e25 (2020).

    CAS  PubMed  Google Scholar 

  23. Cui, J., Xu, H. & Lehtinen, M. K. Macrophages on the margin: choroid plexus immune responses. Trends Neurosci. 44, 864–875 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Liddelow, S. A. Development of the choroid plexus and blood–CSF barrier. Front. Neurosci. 9, 123479 (2015).

    Google Scholar 

  25. McAlpine, C. S. et al. Astrocytic interleukin-3 programs microglia and limits Alzheimer’s disease. Nature 595, 701–706 (2021).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  26. Kiss, M. G. et al. Interleukin-3 coordinates glial-peripheral immune crosstalk to incite multiple sclerosis. Immunity 56, 1502–1514.e8 (2023).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Irwin, M. R. & Opp, M. R. Sleep health: reciprocal regulation of sleep and innate immunity. Neuropsychopharmacology 42, 129–155 (2017).

    CAS  PubMed  Google Scholar 

  28. Rockstrom, M. D. et al. Tumor necrosis factor alpha in sleep regulation. Sleep Med. Rev. 40, 69–78 (2018).

    PubMed  Google Scholar 

  29. Krueger, J. M. et al. Sleep as a fundamental property of neuronal assemblies. Nat. Rev. Neurosci. 9, 910–919 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Gent, T. C., Bandarabadi, M., Herrera, C. G. & Adamantidis, A. R. Thalamic dual control of sleep and wakefulness. Nat. Neurosci. 21, 974–984 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Sancho-Domingo, C., Carballo, J. L., Coloma-Carmona, A. & Buysse, D. J. Brief version of the Pittsburgh sleep quality index (B-PSQI) and measurement invariance across gender and age in a population-based sample. Psychol. Assess. 33, 111–121 (2021).

    PubMed  Google Scholar 

  32. Dick, S. A. et al. Self-renewing resident cardiac macrophages limit adverse remodeling following myocardial infarction. Nat. Immunol. 20, 29–39 (2019).

    CAS  PubMed  Google Scholar 

  33. Bajpai, G. et al. Tissue resident CCR2 and CCR2+ cardiac macrophages differentially orchestrate monocyte recruitment and fate specification following myocardial injury. Circ. Res. 124, 263–278 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Berntson, G. G. et al. Heart rate variability: origins, methods, and interpretive caveats. Psychophysiology 34, 623–648 (1997).

    CAS  PubMed  Google Scholar 

  35. Manolis, A. A. et al. The role of the autonomic nervous system in cardiac arrhythmias: The neuro-cardiac axis, more foe than friend? Trends Cardiovasc. Med. 31, 290–302 (2021).

    CAS  PubMed  Google Scholar 

  36. Carmeliet, P. & Tessier-Lavigne, M. Common mechanisms of nerve and blood vessel wiring. Nature 436, 193–200 (2005).

    ADS  CAS  PubMed  Google Scholar 

  37. Gelosa, P. et al. Cerebral derailment after myocardial infarct: mechanisms and effects of the signaling from the ischemic heart to brain. J. Mol. Med. 100, 23–41 (2022).

    CAS  PubMed  Google Scholar 

  38. Hoyer, F. F. et al. Tissue-specific macrophage responses to remote injury impact the outcome of subsequent local immune challenge. Immunity 51, 899–914.e7 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Thorp, E. B. et al. CCR2+ monocytes promote white matter injury and cognitive dysfunction after myocardial infarction. Brain. Behav. Immun. 119, 818–835 (2024).

    CAS  PubMed  Google Scholar 

  40. Leistner, D. M. et al. Differential immunological signature at the culprit site distinguishes acute coronary syndrome with intact from acute coronary syndrome with ruptured fibrous cap: results from the prospective translational OPTICO-ACS study. Eur. Heart J. 41, 3549–3560 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Gerhardt, T. et al. Culprit plaque morphology determines inflammatory risk and clinical outcomes in acute coronary syndrome. Eur. Heart J. 44, 3911–3925 (2023).

    CAS  PubMed  Google Scholar 

  42. Buysse, D. J., Reynolds, C. F., Monk, T. H., Berman, S. R. & Kupfer, D. J. The Pittsburgh sleep quality index: a new instrument for psychiatric practice and research. Psychiatry Res. 28, 193–213 (1989).

    CAS  PubMed  Google Scholar 

  43. Horne, J. A. & Ostberg, O. A self assessment questionnaire to determine morningness eveningness in human circadian rhythms. Int. J. Chronobiol. 4, 97–110 (1976).

    CAS  PubMed  Google Scholar 

  44. Full, K. M. et al. Validation of a physical activity accelerometer device worn on the hip and wrist against polysomnography. Sleep Health 4, 209–216 (2018).

    PubMed  PubMed Central  Google Scholar 

  45. Doench, J. G. et al. Optimized sgRNA design to maximize activity and minimize off-target effects of CRISPR-Cas9. Nat. Biotechnol. 34, 184–191 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Sanson, K. R. et al. Optimized libraries for CRISPR–Cas9 genetic screens with multiple modalities. Nat. Commun. 9, 5416 (2018).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Hilgendorf, I. et al. Ly-6chigh monocytes depend on nr4a1 to balance both inflammatory and reparative phases in the infarcted myocardium. Circ. Res. 114, 1611–1622 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Maki, K. A. et al. Sleep fragmentation increases blood pressure and is associated with alterations in the gut microbiome and fecal metabolome in rats. Physiol. Genomics 52, 280–292 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Topchiy, I., Fink, A. M., Maki, K. A. & Calik, M. W. Validation of PiezoSleep scoring against EEG/EMG sleep scoring in rats. Nat. Sci. Sleep 14, 1877–1886 (2022).

    PubMed  PubMed Central  Google Scholar 

  51. Yoo, J., Chepurko, V., Hajjar, R. J. & Jeong, D. Conventional method of transverse aortic constriction in mice. Methods Mol. Biol. 1816, 183–193 (2018).

    CAS  PubMed  Google Scholar 

  52. Grune, J. et al. Neutrophils incite and macrophages avert electrical storm after myocardial infarction. Nat. Cardiovasc. Res. 1, 649–664 (2022).

    PubMed  PubMed Central  Google Scholar 

  53. Li, B. et al. Cumulus provides cloud-based data analysis for large-scale single-cell and single-nucleus RNA-seq. Nat. Methods 17, 793–798 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Wolf, F. A., Angerer, P. & Theis, F. J. SCANPY: large-scale single-cell gene expression data analysis. Genome Biol. 19, 15 (2018).

    PubMed  PubMed Central  Google Scholar 

  55. Korsunsky, I. et al. Fast, sensitive and accurate integration of single-cell data with Harmony. Nat. Methods 16, 1289–1296 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Aran, D. et al. Reference-based analysis of lung single-cell sequencing reveals a transitional profibrotic macrophage. Nat. Immunol. 20, 163–172 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Fang, Z., Liu, X. & Peltz, G. GSEApy: a comprehensive package for performing gene set enrichment analysis in Python. Bioinformatics 39, btac757 (2023).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors thank the Human Immune Monitoring Core at the Icahn School of Medicine at Mount Sinai for help with sequencing; the BioMedical Engineering and Imaging Institute and the small animal imaging facility at the Icahn School of Medicine at Mount Sinai for help with MRI and echo imaging and analysis; the Mount Sinai Neuropathology Brain bank for human brain tissue; the Mount Sinai Microscopy and Advanced Bioimaging Core for help with lightsheet, confocal and immunofluorescence imaging; and K. Joyes for copy editing the manuscript text. This work was funded by the National Institutes of health (NIH) R01HL158534, R00HL151750, the Cure Alzheimer’s Fund, and an ISMMS Karen Strauss Cook Research Scholar Award (to C.S.M.); NIH R01AG082185 (to C.S.M., P.R. and D. Lee); an American Heart Association postdoctoral fellowship 24POST1196847 (to P.H.); NIH 5T32HL007824-25 (supported J.D.H.); an EMBO Long Term Fellowship (ALTF 750-2022; to J.F.d.S.); a Kayden-Lambert MGH Research Scholar Award and NIH P01-HL142494 and DP2-CA281401 (to B.P.K.); NINIH R01HL128226 and R35HL155670 (to M.-P.S.-O.); NIH T32HL007343 (supported F.M.Z.); and NIH UL1TR001873 (to Columbia University).

Author information

Authors and Affiliations

Authors

Contributions

P.H. and J.D.H. conceived the project, designed and performed experiments, analysed and interpreted data, offered intellectual input and edited the manuscript. T.G. designed and performed experiments, analysed and interpreted data, conducted the sleep and ACS trial, offered intellectual input and edited the manuscript. M.G.K. aided experiments, interpreted data and offered intellectual input. F.M.Z. and M.-P.S.-O. conducted the human sleep restriction trial and offered intellectual input. O.C. aided in implementation of the sleep questionnaire for the ACS trial, interpreted data and offered intellectual input. C.W. and D. Leistner conducted the sleep and ACS trial. S.G., A.K., A.G.Y., J.D., L.L.K., W.J., L.G., M.H., A.L. and M.G. conducted and aided experiments. Z.C., V.R., T.D., J.M.-R., D.D. and S.K.-S. conducted sequencing experiments and aided in analysis. J.F.d.S., N.J.A. and B.P.K. designed and cloned the AAV vector for Tnfrsf1a knockout and offered technical advice. J.M. and M.M.T.v.L. conducted cMRI imaging and analysis. D. Lee, J.F.F. and P.R. conducted human brain snRNA-seq and analysis. S.Y. performed MI and sham surgeries. N.S. and F.K.S. aided in supervision and offered intellectual input. C.S.M. conceived the project, supervised, directed and managed the study, performed experiments, interpreted data, designed the figures and wrote the manuscript.

Corresponding author

Correspondence to Cameron S. McAlpine.

Ethics declarations

Competing interests

C.S.M. is a consultant for Granite Bio. J.F.d.S. and B.P.K. are inventors on patents or patent applications filed by Mass General Brigham (MGB) that describe genome engineering technologies. B.P.K. is a consultant for EcoR1 capital and Novartis Venture Fund, and is on the scientific advisory board of Acrigen Biosciences, Life Edit Therapeutics and Prime Medicine. B.P.K. has a financial interest in Prime Medicine, a company that is developing therapeutic CRISPR–Cas technologies for gene editing. The interests of B.P.K. were reviewed and are managed by Massachusetts General Hospital (MGH) and MGB in accordance with their conflict-of-interest policies. The other authors declare no competing interests.

Peer review

Peer review information

Nature thanks Hafid Ait-Oufella, Rachel Rowe and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer review reports are available.

Additional information

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

Extended data figures and tables

Extended Data Fig. 1 Sleep parameters in mice with cardiovascular diseases.

a, Quantification of sleep in WT mice that consumed a chow diet and atherosclerotic Apoe−/− mice what consumed a HFD for 16 weeks. n = 8 WT mice; n = 5 Apoe−/− HFD mice. b, Quantification of SWS in sham controls and TAC mice 7 days after surgery. n = 4 sham; n = 3 TAC. c, Quantification of REM sleep in MI and sham mice up to 21 days after infarct. n = 5 mice per group. d, Hypnogram of sleep and wake states in sham and MI mice. e, Quantification of locomotor activity up to 21 days after infarct (p < 0.0001, F = 64.71). n = 4 mice per group. f, Quantification of body temperature up to 21 days after infarct (p < 0.0001, F = 19.68). n = 4 mice per group. g, SWS analysis in naïve and sham mice. n = 5 sham mice; n = 4 naive mice. Data are mean ± s.e.m. Statistical analysis was done using two-way analysis of variance. Experiments were conducted in female mice. *p < 0.05, **p < 0.01, ***p < 0.001.

Extended Data Fig. 2 Analysis of microglia after myocardial infarction.

a, UMAP of cells identified in scRNAseq of mouse brain. n = 5 pooled mice per group. b, UMAP of microglia subclusters and their frequency in day 3 sham and MI mice. n = 5 pooled mice per group. c, Expression of chemokines and activation markers in non-microglial macrophages. n = 5 pooled mice per group. d, Analysis of microglia activation markers in naive mice injected with sham or day 1 MI plasma and sacrificed 4 h later. n = 4 sham plasma; n = 5 MI plasma. e, Measurement of IL-3, IL-6, and IL-1β in plasma one day after sham or MI. n = 3-8 f, Analysis of microglia and quantification of brain monocytes 1 day after stereotactic injection of IL-3, IL-6, IL-1β or PBS into the thalamus of naive mice. n = 5. g, Microglial responses to IL-3, IL-6, and IL-1β in an ex-vivo culture assay. n = 3. Data are mean ± s.e.m. Statistical analysis was done using one-way analysis of variance and two-tailed unpaired t-tests. Experiments were conducted in female mice. *p < 0.05, **p < 0.01, ***p < 0.001.

Extended Data Fig. 3 Brain immune parameters after myocardial infarction.

a, Flow cytometry quantification of brain macrophages, microglia, T cells and neutrophils in sham and MI mice 1, 3, and 7 days after infarct. Each data point represents an individual mouse. b, Histograms of tdTomato (TdT) positivity in blood monocytes and brain microglia in Ms4a3CreRosaTomato mice. c, Analysis of GFP+ cells in the heart, brain, lung, bone marrow, and liver 1 day after sham or infarct and adoptive transfer. n = 5 per group. d, Quantification of CCL2 protein in plasma and brain, and Ccl2 mRNA transcript in blood monocytes and brain tissue. n = 12 sham and n = 14 MI plasma; n = 4 per group for monocyte transcript; n = 5 sham and n = 4 MI brain CCL2 protein; n = 15 per group for brain transcript. e, Representative immunofluorescent images and quantification of CCR2+ monocytes in the cortex and hypothalamus. n = 7 per group. f, Representative image of secondary antibody only control on human liver sections. g, Analysis of FITC-dextran signal in the brain after peripheral delivery one day after MI or sham operation. scRNAseq analysis of brain endothelial and epithelial cells 3 days after sham or MI. n = 5 sham and n = 5 MI for FITC dextran; n = 5 pooled mice per group for scRNAseq. h, Microglial analysis in CCL2RFP mice one day after sham or MI. n = 4 mice per group. i, Representative images and quantification of thalamic microglia morphology by skeletal analysis. n = 3, each dot represents one cell. j, Analysis of blood monocyte and brain microglia CD123 (IL-3Ra), brain monocytes, and microglia CCL2 in Il3rafl/fl and Cx3cr1CreERT2Il3rafl/fl mice injected with tamoxifen over 5 consecutive days and subjected to MI 3 weeks later. n = 4 per group except brain monocytes n = 7 Il3rafl/fl and n = 8 Cx3cr1CreERT2Il3rafl/fl mice. Data are mean ± s.e.m. Statistical analysis was done using two-tailed unpaired t-tests. Experiments were conducted in female mice. *p < 0.05, **p < 0.01, ***p < 0.001.

Extended Data Fig. 4 Analysis of brain monocytes, sleep regulation, and Tnfrsf1a targeting vector design.

a, Enumeration of blood Ly6Chi monocytes in MI and MI + CCR2 antagonist mice one day after injury. CCR2 antagonist was delivered to the brain via the cisterna magna. n = 5 mice per group. b, Quantification of brain Ly6Chi monocytes 1 day after MI and SWS analysis in WT sham, WT MI, and Ccr2−/− MI mice. n = 4 for immune cell quantification; for sleep analysis n = 4 WT MI; n = 5 WT sham, n = 4 Ccr2−/− MI. c, WT MI mice were injected with an anti-TCRβ antibody in the cisterna magna immediately after MI. Monocytes and SWS were quantified one day later. n = 4 control and n = 5 anti-TCRβ. d, Naive mice were injected with PBS or 30,000 monocytes sorted from the blood of a GFP mouse. Flow cytometry analysis of the transferred monocytes in the brain 2 h later. n = 5 mice per group. e, UMAP of brain monocytes and analysis of their frequencies in sham and MI mice. n = 5 pooled mice per group. f, Flow cytometry analysis of brain monocyte TNF. g, qPCR analysis of blood monocyte Tnf mRNA 1 day after sham or MI, n = 4 mice per group. scRNAseq analysis of blood monocytes 3 days after MI or sham. n = 5 pooled mice per group. h, Quantification of SWS in WT sham, WT MI, and Tnf−/− MI mice. n = 3 WT sham; n = 5 WT MI; n = 4 Tnf−/− MI. i, Schematic of the AAV genome encoding Cre expressed from the CaMKII promoter, along with an SpCas9 gRNA targeted to Tnfrsf1a and expressed from the human U6 promoter. j, Schematic for in vivo tissue specific knockdown or Tnfrsf1a, where AAV viral vectors encoding pCaMKII-Cre and the Tnfrsf1a-targeted SpCas9 gRNA are delivered via bilateral stereotactic injection into the thalamic LPN. Neuron-specific Cre recombination activates SpCas9 nuclease expression, leading to complexation of the nuclease with the Tnfrsf1a gRNA to target and knockout the Tnfrsf1a gene. k, Schematic of brain regions analysed by qPCR. Whole brain tissue was used. Expression of Tnfrsf1a in whole thalamic and cortex tissue. n = 5 WT + AAV2 MI mice; n = 4 Stopfl/fl-Cas9GFP + AAV2 MI mice. l, Enumeration of Ly6Chi monocytes in the brain of WT + AAV2 MI mice and Stopfl/fl-Cas9GFP + AAV2 MI mice 3 days after infarct. n = 3 WT + AAV2 MI mice; n = 4 Stopfl/fl-Cas9GFP + AAV2 MI mice. Data are mean ± s.e.m. Statistical analysis was done using two-way analysis of variance and two-tailed unpaired t-tests. Experiments were conducted in female mice. *p < 0.05, **p < 0.01, ***p < 0.001.

Extended Data Fig. 5 Extended analysis of sleep and cardiac function in MI and MI + SF mice.

a, Analysis of wake bouts (transitions from a sleep state to a wake state) in MI and MI + SF mice. n = 4 MI mice; n = 3 MI + SF mice. b, Evaluation of stroke volume (SV) and cardiac output (CO) by echocardiography in MI and MI + SF mice on days 3, 7, and 21 after infarct. n = 8 mice on day 3; n = 9 MI mice on day 7; n = 10 MI + SF mice on day 7; n = 8 MI mice on day 21; n = 10 MI + SF mice on day 21. c, Analysis of EF in sham mice, and mice that received a ‘medium’ or ‘large’ MI and exposed to SF or habitual sleep. Analysis was completed 3 days after sham or MI. n = 5 sham; n = 8 ‘medium’ MI ± SF; n = 5 ‘large’ MI ± SF. d, mRNA expression of Col3a1 and Col4a1 in infarcts 7 days after MI. n = 13 MI and n = 14 MI + SF. Data are mean ± s.e.m. Statistical analysis was done using one-way analysis of variance and two-tailed unpaired t-tests. Experiments were conducted in female mice. *p < 0.05, **p < 0.01, ***p < 0.001.

Extended Data Fig. 6 Inflammation and haematopoiesis in MI and MI + SF mice.

a, Flow cytometry enumeration of cardiac cells in MI and MI + SF mice. n = 7 MI; n = 8 MI + SF. b, Enumeration of monocytes and neutrophils in the infarcts of mice 3 days after receiving a ‘large’ MI and sleeping habitually or exposed to SF. n = 5 mice per group. c, Flow cytometry enumeration of blood leukocytes in MI and MI + SF mice. n = 11 MI on day 3; n = 12 MI + SF on day 3; n = 7 MI on day 7; n = 8 MI + SF on day 7; n = 7 MI day 21 monocytes; n = 9 MI + SF day 21 monocytes; n = 11 MI day 21 neutrophils; n = 12 MI + SF day 21 neutrophils. d, Flow cytometry gating and enumeration of progenitor cells in the BM of MI and MI + SF mice. Each data point represents an individual mouse. e, Analysis of BrdU incorporation into BM progenitor cells of MI and MI + SF mice 3 days after infarct. n = 5 per group. Data are mean ± s.e.m. Statistical analysis was done using two-tailed unpaired t-tests. Experiments were conducted in female mice. *p < 0.05, **p < 0.01, ***p < 0.001.

Extended Data Fig. 7 Assessment of cardiac and blood leukocytes and plasma corticosterone.

a, UMAP of heart leukocytes. n = 5 pooled mice per group. b, Subclustering of cardiac macrophages and cluster frequencies in MI and MI + SF mice. n = 5 pooled mice per group. c, UMAP depicting resident and recruited heart macrophages and defining genes. n = 5 pooled mice per group. d, DEGs in recruited macrophages. n = 5 pooled mice per group. e, UMAP of blood leukocytes and monocytes along with monocyte DEGs and top pathways of genes enriched in MI + SF versus MI mice. n = 5 pooled mice per group. f, Plasma corticosterone levels. Each data point represents an individual mouse. g, Leukocyte enumeration in the infarct. n = 9 MI + SF; n = 8 MI + SF + ADRβ2 blocker. h, Analysis of ejection fraction (EF) and infarct leukocyte abundance in WT mice transplanted with WT BM or Adrb2−/− BM and exposed to MI and SF. Analysis was performed 3 days after infarct. n = 5 WTbmWT; n = 4 WTbmAdrb2−/−. Data are mean ± s.e.m. Statistical analysis was done using two-tailed unpaired t-tests. Experiments were conducted in female mice. *p < 0.05, **p < 0.01, ***p < 0.001.

Extended Data Fig. 8 Extended analysis of human sleep restriction study and schematic of hypothesis.

a, Actigraphy measured nightly total sleep time of study participants during the habitual sleep (HS) and sleep restriction (SR) phases of the randomized crossover trial. n = 4 participants per condition. b, UMAP of scRNAseq data of PBMCs. n = 4 participants per condition. c, Pathway analysis of cluster defining genes among monocyte clusters and the top 10 cluster defining genes in cluster 2. n = 4 participants per condition. d, UMAP of chemotactic genes enriched in monocyte cluster 3. n = 4 participants per condition. e, Schematic of hypothesis. (1) Myocardial infarction activates microglia via IL-1β and (2) enhances their production of myeloid chemoattractants including CCL2 and CCL5. (3) Circulating monocytes are actively recruited to the MI brain where they release TNF which signals to glutamatergic neurons in the thalamic LPN to (4) augment sleep. (5) Enhanced sleep after MI suppresses sympathetic input to the heart which limits signalling through ADRB2 and the generation of the myeloid chemoattractants CCL3 and CCL4 to (6) suppress monocyte recruitment to the infarcted heart thus limiting inflammation and promoting healing. Data are mean ± s.e.m. Statistical analysis was done using one-way analysis of variance and two-tailed paired t-tests. *p < 0.05, **p < 0.01, ***p < 0.001.

Extended Data Table 1 Characteristics of human participants and samples in the study

Supplementary information

Reporting Summary

Supplementary Video 1

Representative iDISCO imaging of CCR2+ monocytes in the brain one day after Sham or MI operation. n = 4, representative of one experiment.

Source data

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Huynh, P., Hoffmann, J.D., Gerhardt, T. et al. Myocardial infarction augments sleep to limit cardiac inflammation and damage. Nature 635, 168–177 (2024). https://doi.org/10.1038/s41586-024-08100-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue date:

  • DOI: https://doi.org/10.1038/s41586-024-08100-w

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