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:

Kinase signalling in excitatory neurons regulates sleep quantity and depth

Abstract

Progress has been made in the elucidation of sleep and wakefulness regulation at the neurocircuit level1,2. However, the intracellular signalling pathways that regulate sleep and the neuron groups in which these intracellular mechanisms work remain largely unknown. Here, using a forward genetics approach in mice, we identify histone deacetylase 4 (HDAC4) as a sleep-regulating molecule. Haploinsufficiency of Hdac4, a substrate of salt-inducible kinase 3 (SIK3)3, increased sleep. By contrast, mice that lacked SIK3 or its upstream kinase LKB1 in neurons or with a Hdac4S245A mutation that confers resistance to phosphorylation by SIK3 showed decreased sleep. These findings indicate that LKB1–SIK3–HDAC4 constitute a signalling cascade that regulates sleep and wakefulness. We also performed targeted manipulation of SIK3 and HDAC4 in specific neurons and brain regions. This showed that SIK3 signalling in excitatory neurons located in the cerebral cortex and the hypothalamus positively regulates EEG delta power during non-rapid eye movement sleep (NREMS) and NREMS amount, respectively. A subset of transcripts biased towards synaptic functions was commonly regulated in cortical glutamatergic neurons through the expression of a gain-of-function allele of Sik3 and through sleep deprivation. These findings suggest that NREMS quantity and depth are regulated by distinct groups of excitatory neurons through common intracellular signals. This study provides a basis for linking intracellular events and circuit-level mechanisms that control NREMS.

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: Loss-of-function mutations of Hdac4 and Hdac5 increase NREMS.
Fig. 2: SIK3–HDAC4 interactions on sleep and wakefulness.
Fig. 3: Sleep–wakefulness of neural LKB1-deficient and SIK3 kinase activity-modified mice.
Fig. 4: Loss of Hdac4 or Sik3 in excitatory neurons decreases sleep.
Fig. 5: Sik3Slp in excitatory neurons increases sleep.
Fig. 6: Single-nucleus transcriptome changes and the molecular pathway associated with sleep need.

Similar content being viewed by others

Data availability

Raw and processed snRNA-seq data have been deposited into the NCBI Gene Expression Omnibus database under accession number GSE214337. All detailed statistics for sleep and behaviour studies are included in the Supplementary Information. All other data supporting this study are available from the corresponding authors without restriction. Source data are provided with this paper.

Code availability

Code used in this study is available from the corresponding authors without restriction.

References

  1. Adamantidis, A. R., Gutierrez Herrera, C. & Gent, T. C. Oscillating circuitries in the sleeping brain. Nat. Rev. Neurosci. 20, 746–762 (2019).

    Article  CAS  Google Scholar 

  2. Liu, D. & Dan, Y. A motor theory of sleep–wake control: arousal–action circuit. Annu. Rev. Neurosci. 42, 27–46 (2019).

    Article  CAS  Google Scholar 

  3. Funato, H. et al. Forward-genetics analysis of sleep in randomly mutagenized mice. Nature 539, 378–383 (2016).

    Article  ADS  CAS  Google Scholar 

  4. Borbély, A. The two-process model of sleep regulation: beginnings and outlook. J. Sleep Res. 31, e13598 (2022).

    Article  Google Scholar 

  5. Reichert, S., Pavón Arocas, O. & Rihel, J. The neuropeptide galanin is required for homeostatic rebound sleep following increased neuronal activity. Neuron 104, 370–384.e5 (2019).

    Article  CAS  Google Scholar 

  6. Oikonomou, G. et al. The serotonergic Raphe promote sleep in zebrafish and mice. Neuron 103, 686–701.e8 (2019).

    Article  CAS  Google Scholar 

  7. Ma, Y. et al. Galanin neurons unite sleep homeostasis and α2-adrenergic sedation. Curr. Biol. 29, 3315–3322.e3 (2019).

    Article  CAS  Google Scholar 

  8. Honda, T. et al. A single phosphorylation site of SIK3 regulates daily sleep amounts and sleep need in mice. Proc. Natl Acad. Sci. USA 115, 10458–10463 (2018).

    Article  ADS  CAS  Google Scholar 

  9. Iwasaki, K. et al. Induction of mutant Sik3Sleepy allele in neurons in late infancy increases sleep need. J. Neurosci. 41, 2733–2746 (2021).

    Article  CAS  Google Scholar 

  10. Vega, R. B. et al. Histone deacetylase 4 controls chondrocyte hypertrophy during skeletogenesis. Cell 119, 555–566 (2004).

    Article  CAS  Google Scholar 

  11. Sando, R. 3rd et al. HDAC4 governs a transcriptional program essential for synaptic plasticity and memory. Cell 151, 821–834 (2012).

    Article  CAS  Google Scholar 

  12. Zhu, Y. et al. Class IIa HDACs regulate learning and memory through dynamic experience-dependent repression of transcription. Nat. Commun. 10, 3469 (2019).

    Article  ADS  Google Scholar 

  13. Yang, X.-J. & Seto, E. The Rpd3/Hda1 family of lysine deacetylases: from bacteria and yeast to mice and men. Nat. Rev. Mol. Cell Biol. 9, 206–218 (2008).

    Article  CAS  Google Scholar 

  14. Park, S.-Y. & Kim, J.-S. A short guide to histone deacetylases including recent progress on class II enzymes. Exp. Mol. Med. 52, 204–212 (2020).

    Article  CAS  Google Scholar 

  15. Berdeaux, R. et al. SIK1 is a class II HDAC kinase that promotes survival of skeletal myocytes. Nat. Med. 13, 597–603 (2007).

    Article  CAS  Google Scholar 

  16. Henriksson, E. et al. SIK2 regulates CRTCs, HDAC4 and glucose uptake in adipocytes. J. Cell Sci. 128, 472–486 (2015).

    CAS  Google Scholar 

  17. Wang, Z. et al. Quantitative phosphoproteomic analysis of the molecular substrates of sleep need. Nature 558, 435–439 (2018).

    Article  ADS  CAS  Google Scholar 

  18. van der Linden, A. M., Nolan, K. M. & Sengupta, P. KIN-29 SIK regulates chemoreceptor gene expression via an MEF2 transcription factor and a class II HDAC. EMBO J. 26, 358–370 (2007).

    Article  Google Scholar 

  19. Lanjuin, A. & Sengupta, P. Regulation of chemosensory receptor expression and sensory signaling by the KIN-29 Ser/Thr kinase. Neuron 33, 369–381 (2002).

    Article  CAS  Google Scholar 

  20. Grubbs, J. J., Lopes, L. E., van der Linden, A. M. & Raizen, D. M. A salt-induced kinase is required for the metabolic regulation of sleep. PLoS Biol. 18, e3000220 (2020).

    Article  CAS  Google Scholar 

  21. Darling, N. J., Toth, R., Arthur, J. S. C. & Clark, K. Inhibition of SIK2 and SIK3 during differentiation enhances the anti-inflammatory phenotype of macrophages. Biochem. J. 474, 521–537 (2017).

    Article  CAS  Google Scholar 

  22. Lizcano, J. M. et al. LKB1 is a master kinase that activates 13 kinases of the AMPK subfamily, including MARK/PAR-1. EMBO J. 23, 833–843 (2004).

    Article  CAS  Google Scholar 

  23. Uebi, T. et al. Involvement of SIK3 in glucose and lipid homeostasis in mice. PLoS ONE 7, e37803 (2012).

    Article  ADS  CAS  Google Scholar 

  24. Franken, P., Chollet, D. & Tafti, M. The homeostatic regulation of sleep need is under genetic control. J. Neurosci. 21, 2610–2621 (2001).

    Article  CAS  Google Scholar 

  25. Vassalli, A. & Franken, P. Hypocretin (orexin) is critical in sustaining theta/gamma-rich waking behaviors that drive sleep need. Proc. Natl Acad. Sci. USA 114, E5464–E5473 (2017).

    Article  ADS  CAS  Google Scholar 

  26. Cirelli, C. The genetic and molecular regulation of sleep: from fruit flies to humans. Nat. Rev. Neurosci. 10, 549–560 (2009).

    Article  CAS  Google Scholar 

  27. Zhou, R. et al. A signalling pathway for transcriptional regulation of sleep amount in mice. Nature https://doi.org/10.1038/s41586-022-05510-6 (2022).

  28. Williams, S. R. et al. Haploinsufficiency of HDAC4 causes brachydactyly mental retardation syndrome, with brachydactyly type E, developmental delays, and behavioral problems. Am. J. Hum. Genet. 87, 219–228 (2010).

    Article  CAS  Google Scholar 

  29. Hobara, T. et al. Altered gene expression of histone deacetylases in mood disorder patients. J. Psychiatr. Res. 44, 263–270 (2010).

    Article  Google Scholar 

  30. Vyazovskiy, V. V. et al. Local sleep in awake rats. Nature 472, 443–447 (2011).

    Article  ADS  CAS  Google Scholar 

  31. Krone, L. B. et al. A role for the cortex in sleep–wake regulation. Nat. Neurosci. 24, 1210–1215 (2021).

    Article  CAS  Google Scholar 

  32. Brüning, F. et al. Sleep–wake cycles drive daily dynamics of synaptic phosphorylation. Science 366, eaav3617 (2019).

    Article  Google Scholar 

  33. Chung, S. et al. Identification of preoptic sleep neurons using retrograde labelling and gene profiling. Nature 545, 477–481 (2017).

    Article  ADS  CAS  Google Scholar 

  34. Potthoff, M. J. et al. Histone deacetylase degradation and MEF2 activation promote the formation of slow-twitch myofibers. J. Clin. Invest. 117, 2459–2467 (2007).

    Article  CAS  Google Scholar 

  35. Miyoshi, C. et al. Methodology and theoretical basis of forward genetic screening for sleep/wakefulness in mice. Proc. Natl Acad. Sci. USA 116, 16062–16067 (2019).

    Article  ADS  CAS  Google Scholar 

  36. Mizuno, S. et al. Simple generation of albino C57BL/6J mice with G291T mutation in the tyrosinase gene by the CRISPR/Cas9 system. Mamm. Genome 25, 327–334 (2014).

    Article  CAS  Google Scholar 

  37. Sato, Y. et al. A mutation in transcription factor MAFB causes focal segmental glomerulosclerosis with Duane retraction syndrome. Kidney Int. 94, 396–407 (2018).

    Article  CAS  Google Scholar 

  38. Iwasaki, K., Hotta-Hirashima, N., Funato, H. & Yanagisawa, M. Protocol for sleep analysis in the brain of genetically modified adult mice. STAR Protoc. 2, 100982 (2021).

    Article  CAS  Google Scholar 

  39. Suzuki-Abe, H. et al. Metabolomic and pharmacologic analyses of brain substances associated with sleep pressure in mice. Neurosci. Res. 177, 16–24 (2022).

    Article  CAS  Google Scholar 

  40. Park, M. et al. Loss of the conserved PKA sites of SIK1 and SIK2 increases sleep need. Sci. Rep. 10, 8676 (2020).

    Article  ADS  CAS  Google Scholar 

  41. Misztal, K. et al. TCF7L2 mediates the cellular and behavioral response to chronic lithium treatment in animal models. Neuropharmacology 113, 490–501 (2017).

    Article  CAS  Google Scholar 

  42. Guo, P. et al. Rapid and simplified purification of recombinant adeno-associated virus. J. Virol. Methods 183, 139–146 (2012).

    Article  CAS  Google Scholar 

  43. Honda, T. et al. Ablation of ventral midbrain/pons GABA neurons induces mania-like behaviors with altered sleep homeostasis and dopamine D2R-mediated sleep reduction. iScience 23, 101240 (2020).

    Article  ADS  CAS  Google Scholar 

  44. Takase, K., Tsuneoka, Y., Oda, S., Kuroda, M. & Funato, H. High-fat diet feeding alters olfactory-, social-, and reward-related behaviors of mice independent of obesity. Obesity 24, 886–894 (2016).

    Article  CAS  Google Scholar 

  45. Kumar, D. et al. Sparse activity of hippocampal adult-born neurons during REM sleep is necessary for memory consolidation. Neuron 107, 552–565.e10 (2020).

    Article  CAS  Google Scholar 

  46. Li, C. H. & Lee, C. K. Minimum cross entropy thresholding. Pattern Recognit26, 617–625 (1993).

    Article  ADS  Google Scholar 

  47. Fujiyama, T. et al. Forebrain Ptf1a is required for sexual differentiation of the brain. Cell Rep. 24, 79–94 (2018).

    Article  CAS  Google Scholar 

  48. Funato, H., Saito-Nakazato, Y. & Takahashi, H. Axonal growth from the habenular nucleus along the neuromere boundary region of the diencephalon is regulated by semaphorin 3F and netrin-1. Mol. Cell. Neurosci. 16, 206–220 (2000).

    Article  CAS  Google Scholar 

  49. Henriksson, E. et al. The AMPK-related kinase SIK2 is regulated by cAMP via phosphorylation at Ser 358 in adipocytes. Biochem. J. 444, 503–514 (2012).

    Article  CAS  Google Scholar 

  50. Hossain, M. S. et al. Identification of mutations through dominant screening for obesity using C57BL/6 substrains. Sci. Rep. 6, 32453 (2016).

    Article  ADS  CAS  Google Scholar 

  51. Barbosa, A. C. et al. MEF2C, a transcription factor that facilitates learning and memory by negative regulation of synapse numbers and function. Proc. Natl Acad. Sci. USA 105, 9391–9396 (2008).

    Article  ADS  CAS  Google Scholar 

  52. Stuart, T. et al. Comprehensive integration of single-cell data. Cell 177, 1888–1902.e21 (2019).

    Article  CAS  Google Scholar 

  53. Hao, Y. et al. Integrated analysis of multimodal single-cell data. Cell 184, 3573–3587.e29 (2021).

    Article  CAS  Google Scholar 

  54. Bragina, L. et al. Analysis of synaptotagmin, SV2, and Rab3 expression in cortical glutamatergic and GABAergic axon terminals. Front. Cell. Neurosci. 5, 32 (2011).

    CAS  Google Scholar 

  55. Ashburner, M. et al. Gene ontology: tool for the unification of biology. The Gene Ontology Consortium. Nat. Genet. 25, 25–29 (2000).

    Article  CAS  Google Scholar 

  56. Gene Ontology Consortium. The Gene Ontology resource: enriching a GOld mine. Nucleic Acids Res. 49, D325–D334 (2021).

    Article  Google Scholar 

  57. Raizen, D. M. et al. Lethargus is a Caenorhabditis elegans sleep-like state. Nature 451, 569–572 (2008).

    Article  ADS  CAS  Google Scholar 

  58. Singh, K. et al. C. elegans Notch signaling regulates adult chemosensory response and larval molting quiescence. Curr. Biol. 21, 825–834 (2011).

    Article  CAS  Google Scholar 

  59. Yu, Z. et al. Beyond t test and ANOVA: applications of mixed-effects models for more rigorous statistical analysis in neuroscience research. Neuron 110, 21–35 (2021).

    Article  Google Scholar 

Download references

Acknowledgements

We thank all members of Yanagisawa/ Funato Lab and the International Institute for Integrative Sleep Medicine, especially K. Ebihara, M. Taniguchi, M. Sato for technical support, K. Iida, A. Kuno, M. Lazarus, E. Hasegawa, K. Roy, S. Wakana, T. Suzuki and I. Miura for technical advice and discussion, D. Kawaguchi for sharing Foxg1cre mice, and E. Olson for sharing Hdac4flox mice. This work was supported by the World Premier International Research Center Initiative from MEXT to M.Y., JSPS KAKENHI (17H06095, 22H04918 to M.Y. and H.F.; 17H04023, 17H05583, 20H00567 to H.F.; 26507003, 18968064 to C.M. and H.F.; 15J00393, 18J21517 to F.A.; 18K14811 to T.F.; and 20J12137 to K.I.), JST CREST (JPMJCR1655 to M.Y.), AMED (JP21zf0127005 to M.Y.), JSPS DC2 (17J07957 to S.J.K.), the University of Tsukuba Basic Research Support Program Type A (to S.J.K.), and the Funding Program for World-Leading Innovative R&D on Science and Technology (FIRST Program) from JSPS (to M.Y.).

Author information

Authors and Affiliations

Authors

Contributions

S.J.K., N.H.-H., F.A., T.K., K.I., S.N., H.K. and N.A. contributed equally to this work. S.J.K. carried out experiments for all Hdac-modified mice. N.H.-H. performed sleep analyses of the Sik3Slp-flox mice. F.A. produced and performed sleep analysis of Sik3flox mice. T.K. performed sleep analysis of Lkb1-modified mice. K.I. performed snRNA-seq experiments and analysis. S.N. produced and performed sleep analysis of Sik3T221E mice. H.K. produced and performed sleep analysis of Sik3T221A mice. N.A. produced and performed sleep analysis of Hdac4S245A mice. T.M. performed cell culture experiments. A.I. performed western blotting. M.K. and Y.T. performed histology. D.K. performed memory experiments. S.K., J.C., T.T. and A.E. performed sleep analyses. C.M. performed sleep analyses and pharmacology experiments. S. Miyazaki and Y.H. contributed to C.elegans data generation. T.F., S. Mizuno, F.S. and S.T. contributed to gene-modified mouse generation. M.M. and Q.L. contributed to data interpretation. S.J.K., M.Y. and H.F. wrote the manuscript. M.Y. and H.F. conceived and supervised the entire project.

Corresponding authors

Correspondence to Masashi Yanagisawa or Hiromasa Funato.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature thanks the anonymous reviewers for their contribution to the peer review of this work. Peer reviewer reports are available.

Additional information

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

Extended data figures and tables

Extended Data Fig. 1 Splice mutations of Hdac4 and Hdac5 genes increased NREMS.

a. Haplotype analysis of the Slp2 pedigree for wake time based on C57B6J/C57B6N SNPs (N2, n = 31). b. Exome sequencing identified single nucleotide changes within the LOD score peak (upper) and total wake time (lower) of each mouse. Blue and red bars indicate normal and short wake time, respectively. c. RT-PCR showing short Hdac4 transcripts specific to Hdac4Slp2/+ mice. Hrt, heart; Ctx, cerebral cortex; Hyp, hypothalamus. Representative images from three biological replicates are shown. d. Immunoblot of brain homogenates of wild-type and Hdac4Slp2/+ mice using anti-HDAC4 antibody. Anti-β-actin antibody was used as loading control. Quantitation of HDAC4 band intensity against β-actin (Right). Representative blots from three biological replicates are shown. e. Structures of HDAC4,5,7 and 9 with phosphorylation sites and deacetylase (DAC) domain. Numbers on the DAC domain indicate the region encoded by the skipped exon. f. Genomic DNA and transcripts of class IIa Hdacs. Red triangles indicate the splice acceptor site mutations (SA) of Hdac4, 5, 7, and 9 genes. Black triangles indicate stop codon produced due to the frameshift. g, h. RT-PCR of Hdac4SA (g) and Hdac5SA (h) mouse cortical tissues (3 mice per group). Shorter bands (asterisk) corresponding to exon skipping variants. i–k. Total wake time (i), NREMS episode duration (j), and total time spent in REMS (k) of Hdac4 mice (+/+, n = 26; SA/+, n = 26). l. NREMS delta power under the basal condition and after 6-hour sleep deprivation (SD) of Hdac4SA mice. m. Change in NREMS delta power after sleep deprivation relative to each ZT under the basal condition of Hdac4 mice (+/+, n = 10; SA/+, n = 13). n, o. Relative NREMS delta power to the basal condition (n) and time spent in NREMS (o) for 3 h from ZT13. HDAC4 inhibitor, LMK235, was intracerebroventricularly administered at ZT11.5. n = 12. p–r. Total time spent in wake (p), NREMS episode duration (q), and total time spent in REMS (r) of Hdac5 mice (+/+, n = 17; SA/+, n = 21; SA/SA, n = 7). s, t. NREMS delta power under the basal condition and after 6-hour sleep deprivation (s) and change in NREMS delta power after sleep deprivation relative to each ZT under the basal condition (t) of Hdac5 mice (+/+, n = 14; SA/+, n = 21; SA/SA, n = 7). u, v. Diurnal NREMS delta density (u) and EEG power spectra during NREMS (v) of Hdac4SA; Hdac5SA mice. Hdac4+/+; Hdac5SA/+ mice (n = 16) and Hdac4SA/+; Hdac5SA/+ mice (n = 10). Data shown as mean ± s.e.m.; two-sided P values. Unpaired t-test (f, v); two-way ANOVA with Tukey’s test (ik, pr); mixed-effects model (m, t, u), with Dunnett’s test (n, o); two-way repeated measures ANOVA with Sidak’s (l) or Tukey’s test (s).

Source data

Extended Data Fig. 2 Retina and behavioral characterization of HDAC4SA mice.

a. Representative immunostaining from at least 3 biological replicates using anti-rhodopsin antibody of wild-type and Hdac4SA retinas. Sections were also stained with hematoxylin. Scale bar, 150 μm. b, c. Body temperature of Hdac4SA mice (+/+, n = 8; SA/+, n = 7). Data points are averaged every 10 min. d. Daily food intake over 4 days of chow diet of Hdac4SA mice (+/+, n = 14; SA/+, n = 13). e, f. Total distance (e) and time spent in the center area (f) of Hdac4SA mice (+/+, n = 12; SA/+, n = 11). g. Time spent in the open area on the elevated plus maze of Hdac4SA mice (+/+, n = 11; SA/+, n = 14). h. The time of immobility during the last 4 min of the tail suspension test of Hdac4SA mice (+/+, n = 13; SA/+, n = 13). i. The time of immobility during the last 4 min of the forced swim test of Hdac4SA mice (+/+, n = 13; SA/+, n = 14). j–l. Fear conditioning test. Shock reactivity during conditioning (j) was movements for 2 s immediately before shock (Pre-shock) and for 2 s during the first shock (Shock). Freezing in the tone fear conditioning task (k) and in contextual fear conditioning task (l). Mice were placed in the non-conditioned context, and the tone was played during the last 3 min of the session Hdac4SA mice (+/+, n = 14; SA/+, n = 15). Data shown as mean ± s.e.m.; two-sided P values. Two-way repeated measures ANOVA (b); unpaired t-test (cf, hl). Mann-Whitney U-test (g).

Source data

Extended Data Fig. 3 Sleep/wakefulness of Hdac7SA and Hdac9SA mice.

a. RT-PCR of Hdac7SA and Hdac9SA mouse brains. Shorter bands (asterisk) corresponding to exon skipping variants. Representative images from three biological replicates. b–f. Total wake time (b), total NREMS time (c), NREMS episode duration (d), diurnal NREMS delta density (e), and EEG power spectra during NREMS (f) of Hdac7SA mice (+/+, n = 12; SA/+, n = 12). g–k. Total wake time (g), total NREMS time (h), NREMS episode duration (i), diurnal NREMS delta density (j), and EEG power spectra during NREMS (k) of Hdac9SA mice (+/+, n = 15; SA/+, n = 15; SA/SA, n = 13). l. Nuclear HDAC4 intensity at ZT6 and ZT12.5 of the primary motor cortex in constant darkness (ZT6, n = 10; ZT12.5, n = 10). m, n. Representative result of western immunostaining (m) of nucleo-cytoplasmic fractionated cerebral cortex. Densitometric analysis of HDAC4 over β-actin of the cytoplasmic fractions. Six biological replicates (n). o. In vitro kinase assay of SIK3 using HDAC4 peptide. p. Immunoblots of PC12 cell homogenates using HDAC4 antibodies after the addition of HG9-91-01. Two technical replicates. q, r. NREMS episode duration (q) and total REMS time (r) of Hdac4 mice (+/+, n = 35; S245A/+, n = 20). s. NREMS delta power after 6 hour-sleep deprivation of Hdac4 mice (+/+, n = 29; S245A/+, n = 12). Data shown as mean ± s.e.m.; two-sided P values. Two-way ANOVA (bd, gi, q, r) with Tukey’s test (q); mixed-effects model (e, j); unpaired t-test (f, l, n); one-way ANOVA (k), two-way repeated measures ANOVA with Sidak’s test (s).

Source data

Extended Data Fig. 4 Lethargus of C. elegans had-4 and kin-29 mutants.

a. During lethargus, hda-4(oy59) (n = 15) and kin-29(oy39) hda-4(oy59) (n = 14) mutant worms exhibited increased quiescence compared to wild-type worms (n = 42), whereas kin-29(oy39) mutant worms (n = 12) exhibited decreased quiescence. b. Increased quiescence during lethargus in the hda-4(oy59) mutant was rescued by transfection with genomic hda-4 (n = 9), but not with an injection marker alone (n = 10). c, d. Comparison of the duration of bout of motion (c) and quiescence (d) during lethargus revealed that the quiescent bouts were selectively affected. e, f. The quiescent bouts durations were significantly decreased in genomic hda-4 transfected worms. Data shown as mean ± s.e.m.; two-sided P values. Steel-Dwass multiple comparison tests (a, c, d); Mann-Whitney U-tests (b, e, f).

Source data

Extended Data Fig. 5 Sleep/wakefulness of Lkb1 and Sik3 gene-modified mice.

a. In vitro kinase assay for SIK3, SIK3 (T221A) and SIK3 (T221E). b–d. Total wake time (b), NREMS episode duration (c), total REMS time (d) of Syn1creERT; Lkb1flox mice (+/+, n = 9; flox/flox, n = 14). e. NREMS delta power for 2 h after 6-hour sleep deprivation of Syn1creERT; Lkb1flox mice (+/+, n = 8; flox/flox, n = 10). f–i. Total wake time (f), NREMS episode duration (g), total REMS time (h), and NREMS delta power under the basal condition and after 6-hour sleep deprivation (i) of Sik3 mice (+/+, n = 11; T221A/+, n = 12). j–l. Total wake time (j), NREMS episode duration (k), and total REMS time (l) of Syn1creERT; Lkb1flox; Sik3 mice (+/+, n = 14; T221E/T221E, n = 15). m–o. Total wake time (m), NREMS episode duration (n), and total REMS time (o) of Sik3 mice (Slp/+, n = 7; Slp-T221A/+, n = 10). p. NREMS delta power under the basal condition and after 6-hour sleep deprivation of Sik3 mice (Slp/+, n = 6; Slp-T221A/+, n = 10). Data shown as mean ± s.e.m.; two-sided P values. Two-way ANOVA (c, f, g, h, k, o) with Tukey’s test (b, d, j, ln); two-way repeated measures with Sidak’s test (e, i, p).

Source data

Extended Data Fig. 6 Sleep/wakefulness of neuron-type-specific Hdac4-deficient mice.

a. HDAC4 immunoreactivity was abolished in nestincre; Hdac4flox/flox mice (right), compared to nestincre; Hdac4+/+ mice (left). Scale bars, 1 mm. b. EEG power spectra during NREMS of Vglut2cre; Hdac4flox mice (+/+, n = 21; flox/+, n = 13; flox/flox, n = 12). c. NREMS delta power after 6 hour-sleep deprivation of Vglut2cre; Hdac4flox mice (+/+, n = 13; flox/+, n = 13, flox/flox, n = 11). d–g. Total wake time (d), total NREMS time (e), diurnal NREMS delta density (f), and EEG power spectra during NREMS (g) of Camk2acre; Hdac4flox mice (+/+, n = 13; flox/+, n = 11; flox/flox, n = 11). h–k. Total wake time (h), total NREMS time (i), diurnal NREMS delta density (j) and EEG power spectra during NREMS (k) of Vgatcre; Hdac4flox mice (+/+, n = 12; flox/+, n = 9; flox/flox, n = 11). Data shown as mean ± s.e.m.; two-sided P values. Two-way ANOVA (d, e) with Tukey’s test (h, i); mixed-effects model (f, j); one-way ANOVA (b, g, k); two-way repeated measures ANOVA with Tukey’s test (c).

Source data

Extended Data Fig. 7 Histological analysis of HDAC4, SIK3 and Cre drivers.

a–f. X-gal staining of Vglut2cre; ROSA-LacZ (a), Vglut1cre; ROSA-LacZ (b), Camk2acre; ROSA-LacZ (c), Vgatcre; ROSA-LacZ mice (d), Foxg1cre; ROSA-LacZ (e), and Foxd1cre; ROSA-LacZ (f). Scale bars, 1 mm. g. Visualization of SIK3 of the cerebral cortex (left) and the ventromedial nucleus of the hypothalamus (VMH, right) using anti-HA antibody of HA-Sik3 knock-in mouse brains. Scale bars, 40 μm. h, i. Representative result of Sik3 mRNA expression in the brain of nestin+/+; Sik3flox mouse (h) and nestincre; Sik3flox mouse (i). Three biological replicates. High magnification images of visual cortex. Scale bars, 500 μm, 250 μm. j. Representative result of western blotting using anti-SIK3 antibody showing SIK3 expression in nestin+/+; Sik3flox mouse brain, but not in nestincre; Sik3 flox mouse brain. Three biological replicates. k–n. Representative results of three Sik3 mRNA expression in the brains of Sik3flox (k), nestincre; Sik3flox (l), Vglut2cre; Sik3flox (m), and Vgatcre; Sik3flox mouse (n). Vglut2cre; Sik3flox mice showed reduced SIK3 expression in the thalamus (TH) whereas Vgatcre; Sik3flox mice showed reduced SIK3 expression in the thalamic reticular nucleus (TRH). Nestincre; Sik3flox mice showed reduced SIK3 expression in both TH and TRH. Sections were lightly counterstained with Nissl. Scale bars, 400 μm.

Extended Data Fig. 8 Sleep/wakefulness of neuron-type-specific Sik3-deficient mice.

a–c. NREMS episode duration (a), total wake time (b), total REMS time (c) of Vglut2cre; Sik3 flox mice (+/+, n = 16; flox/flox, n = 17). d,e. NREMS delta power under basal condition and after 6-h sleep deprivation (d) and relative NREMS delta power after sleep deprivation to each ZT under the basal condition (e) of Vglut2cre; Sik3 flox mice (+/+, n = 10; flox/flox, n = 17). f—h. NREMS episode duration (f), total wake time (g), total REMS time (h) of Vglut1cre; Sik3 flox mice (+/+, n = 12; flox/flox, n = 12). i–j. NREMS delta power under basal condition and after 6-h sleep deprivation (i) and relative NREMS delta power after sleep deprivation to each ZT under the basal condition (j) of Vglut1cre; Sik3 flox mice (+/+, n = 10; flox/flox, n = 12). k—p. Total NREMS time (k), total wake time (l), total REMS time (m), NREMS episode duration (n), diurnal NREMS delta density (o), and EEG power during NREMS (p) of Camk2acre; Sik3 flox mice (+/+, n = 11; flox/flox, n = 11). q. NREMS delta power under basal condition and after 6-h sleep deprivation of Camk2acre; Sik3 flox mice (+/+, n = 9; flox/flox, n = 11). r–w. Total NREMS time (r), total wake time (s), total REMS time (t), NREMS episode duration (u), diurnal NREMS delta density (v), and EEG power during NREMS (w) of Vgatcre; Sik3 flox mice (+/+, n = 14; flox/flox, n = 8). x. NREMS delta power under basal condition and after 6-h sleep deprivation of Vgatcre; Sik3 flox mice (+/+, n = 7; flox/flox, n = 6). y–aa. NREMS episode duration (y), total wake time (z), total REMS time (aa) of Foxg1cre; Sik3 flox mice (+/+, n = 8; flox/flox, n = 11). ab. NREMS delta power under basal condition and after 6-h sleep deprivation of Foxg1cre; Sik3 flox mice (+/+, n = 8; flox/flox, n = 11). ac–ae. NREMS episode duration (ac), total wake time (ad), total REMS time (ae) of Foxd1cre; Sik3 flox mice (+/+, n = 10; flox/flox, n = 14). af. NREMS delta power under basal condition and after 6-h sleep deprivation of Foxd1cre; Sik3 flox mice (+/+, n = 10; flox/flox, n = 13). Data shown as mean ± s.e.m.; two-sided P values. Two-way ANOVA with Tukey’s test (ac, fh, k-n, ru, yaa, acae); two-way repeated measures ANOVA with Sidak’s test (d, i, q, x, ab, af); mixed-effects model (e, j, o, v); unpaired t-test (p, w).

Source data

Extended Data Fig. 9 Sik3Slp in excitatory neurons increased NREMS.

a–f. NREMS episode duration of Vglut2cre; Sik3Slp-flox mice (a)(+/+, n = 19; Slp-flox/+, n = 17; Slp-flox/Slp-flox, n = 20), Vglut1cre; Sik3Slp-flox mice (b)(+/+, n = 11; Slp-flox/+ n = 11; Slp-flox/Slp-flox, n = 10), Camk2acre; Sik3Slp-flox mice (c)(+/+, n = 11; Slp-flox/+, n = 13; Slp-flox/Slp-flox, n = 10), Vgatcre; Sik3Slp-flox mice (d)(+/+, n = 10; Slp-flox/+, n = 13; Slp-flox/Slp-flox, n = 12), Foxg1cre; Sik3Slp-flox mice (e)(+/+, n = 17; Slp-flox/+, n = 12; Slp-flox/Slp-flox, n = 14), and Foxd1cre; Sik3Slp-flox mice (f)(+/+, n = 27; Slp-flox/+, n = 16; Slp-flox/Slp-flox, n = 21). g–l. Total wake time of Vglut2cre; Sik3Slp-flox mice (g), Vglut1cre; Sik3Slp-flox mice (h), Camk2acre; Sik3Slp-flox mice (i), Vgatcre; Sik3Slp-flox mice (j), Foxg1cre; Sik3Slp-flox mice (k), and Foxd1cre; Sik3Slp-flox mice (l). Colours are as for af, respectively. m-r. Total REMS time of Vglut2cre; Sik3Slp-flox mice (m), Vglut1cre; Sik3Slp-flox mice (n), Camk2acre; Sik3Slp-flox mice (o), Vgatcre; Sik3Slp-flox mice (p), Foxg1cre; Sik3Slp-flox mice (q), and Foxd1cre; Sik3Slp-flox mice (r). Colours are as for af, respectively. s, t. Relative NREMS delta power changes after sleep deprivation to each ZT under the basal condition of Vglu2cre; Sik3Slp-flox mice (s)(+/+, n = 13; Slp-flox/Slp-flox, n = 16) and Foxd1cre; Sik3Slp-flox mice (t)(+/+, n = 10; Slp-flox/Slp-flox, n = 11). u–z. Process S simulation for sleep need dynamics. Td/Ti ratio of Hdac4SA mice (u)(+/+, n = 10; SA/+, n = 13), Hdac5SA mice (v)(+/+, n = 9; SA/+, n = 13; SA/SA, n = 7), Vglut2cre; Sik3flox mice (w)(+/+, n = 10; flox/flox, n = 17), Vglut1cre; Sik3flox mice (x)(+/+, n = 10; flox/flox, n = 12), Vglut2cre; Sik3Slp-flox mice (y)(+/+, n = 16; Slp-flox/Slp-flox, n = 13), and Foxd1cre; Sik3Slp-flox mice (z)(+/+, n = 11; Slp-flox/Slp-flox, n = 10). Data shown as mean ± s.e.m.; two-sided P values. Two-way ANOVA with Tukey’s test when applicable (ar); mixed-effects model (s, t); unpaired t-test (u, wz); one-way ANOVA with Tukey’s test (v).

Source data

Extended Data Fig. 10 Single-nucleus RNA-seq analysis of the cerebral cortex transduced with AAV-Sik3Slp and of sleep-deprived mice.

a. The number of detected reads in nuclei of WPRE-positive cells of each cluster from the cerebral cortex transduced with AAV-Sik3Slp or AAV-Sik3. b. The number of detected reads in nuclei of each cluster from the cerebral cortex of sleep-deprived or ad libitum sleep mice. c, d. The number of genes detected in nuclei of each cluster from the cerebral cortex transduced with AAV-Sik3Slp or AAV-Sik3 (c), and sleep-deprived or ad libitum sleep mice (d). e. WPRE-positive nuclei were displayed on UMAP of nuclei from the cerebral cortex transduced with AAV-Sik3Slp or AAV-Sik3 (6 mice per group). f. Volcano plot of genes in the GABAergic cluster of nuclei from the cerebral cortex transduced with AAV-Sik3Slp or AAV-Sik3. The vertical axis indicates -log10(adjusted P value) of Wilcoxon rank-sum test with Bonferroni correction. The horizontal axis indicates log2(fold change) calculated from AAV-Sik3Slp compared with AAV-Sik3. Each point represents a gene detected in at least 10% of nuclei in any conditions. Black circles, P ≥ 0.05 or |log2(fold change)|≤0.14. Red circles, P < 0.05 and log2(fold change) > 0.14. Blue circles, P < 0.05 and log2(fold change) < −0.14. g–j. Volcano plots of genes in the GABAergic cluster (g), astrocyte cluster (h), oligodendrocyte cluster (i), and microglia cluster (j) of nuclei from the cerebral cortex of sleep-deprived (n = 4) or ad libitum sleep mice (n = 6). k, l. Expression level of Bdnf in nuclei from the cerebral cortex transduced with AAV-Sik3Slp or AAV-Sik3 (k) and the cerebral cortex of sleep-deprived or ad libitum sleep mice (l).

Supplementary information

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

Kim, S.J., Hotta-Hirashima, N., Asano, F. et al. Kinase signalling in excitatory neurons regulates sleep quantity and depth. Nature 612, 512–518 (2022). https://doi.org/10.1038/s41586-022-05450-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-022-05450-1

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