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:

Adolescent sleep shapes social novelty preference in mice

Abstract

Sleep disturbances frequently occur in neurodevelopmental disorders such as autism, but the developmental role of sleep is largely unexplored, and a causal relationship between developmental sleep defects and behavioral consequences in adulthood remains elusive. Here, we show that in mice, sleep disruption (SD) in adolescence, but not in adulthood, causes long-lasting impairment in social novelty preference. Furthermore, adolescent SD alters the activation and release patterns of dopaminergic neurons in the ventral tegmental area (VTA) in response to social novelty. This developmental sleep function is mediated by balanced VTA activity during adolescence; chemogenetic excitation mimics, whereas silencing rescues, the social deficits of adolescent SD. Finally, we show that in Shank3-mutant mice, improving sleep or rectifying VTA activity during adolescence ameliorates adult social deficits. Together, our results identify a critical role of sleep and dopaminergic activity in the development of social interaction behavior.

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: Adolescent SD induced loss of social novelty preference in adult social interactions.
Fig. 2: Adolescent SD attenuated the novelty-dependent response pattern of VTADA neurons in social interactions.
Fig. 3: Adolescent SD altered dopamine release in the NAc in response to social stimuli.
Fig. 4: Adolescent SD alters the projection profile of VTADA axons in the NAc and mPFc.
Fig. 5: Activity level of VTADA neurons during adolescence is critical for social novelty preference.
Fig. 6: Adolescent restoration of NREM sleep rescued the social interaction deficit in Shank3 InsG3680+/+ mice.

Similar content being viewed by others

Data availability

The data that support the findings of this study are available within this paper and its Supplementary Information. Source data are provided with this paper.

Code availability

All custom code used in this study is available from the corresponding authors upon reasonable request.

References

  1. Frank, M. G. & Heller, H. C. The function(s) of sleep. Handb. Exp. Pharmacol. 253, 3–34 (2019).

    Article  CAS  PubMed  Google Scholar 

  2. Anafi, R. C., Kayser, M. S. & Raizen, D. M. Exploring phylogeny to find the function of sleep. Nat. Rev. Neurosci. 20, 109–116 (2019).

    Article  CAS  PubMed  Google Scholar 

  3. Tononi, G. & Cirelli, C. Sleep and the price of plasticity: from synaptic and cellular homeostasis to memory consolidation and integration. Neuron 81, 12–34 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Huber, R. & Born, J. Sleep, synaptic connectivity, and hippocampal memory during early development. Trends Cogn. Sci. 18, 141–152 (2014).

    Article  PubMed  Google Scholar 

  5. Campbell, I. G. & Feinberg, I. Longitudinal trajectories of non-rapid eye movement delta and theta EEG as indicators of adolescent brain maturation. Proc. Natl Acad. Sci. USA 106, 5177–5180 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. de Vivo, L. et al. Developmental patterns of sleep slow wave activity and synaptic density in adolescent mice. Sleep 37, 689–700 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  7. Timofeev, I. et al. Spatio-temporal properties of sleep slow waves and implications for development. Curr. Opin. Physiol. 15, 172–182 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  8. Frank, M. G., Issa, N. P. & Stryker, M. P. Sleep enhances plasticity in the developing visual cortex. Neuron 30, 275–287 (2001).

    Article  CAS  PubMed  Google Scholar 

  9. Dumoulin Bridi, M. C. et al. Rapid eye movement sleep promotes cortical plasticity in the developing brain. Sci. Adv. 1, e1500105 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  10. Shaffery, J. P., Lopez, J., Bissette, G. & Roffwarg, H. P. Rapid eye movement sleep deprivation revives a form of developmentally regulated synaptic plasticity in the visual cortex of post-critical period rats. Neurosci. Lett. 391, 96–101 (2006).

    Article  CAS  PubMed  Google Scholar 

  11. Shaffery, J. P., Sinton, C. M., Bissette, G., Roffwarg, H. P. & Marks, G. A. Rapid eye movement sleep deprivation modifies expression of long-term potentiation in visual cortex of immature rats. Neuroscience 110, 431–443 (2002).

    Article  CAS  PubMed  Google Scholar 

  12. Maret, S., Faraguna, U., Nelson, A. B., Cirelli, C. & Tononi, G. Sleep and waking modulate spine turnover in the adolescent mouse cortex. Nat. Neurosci. 14, 1418–1420 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Yang, G. et al. Sleep promotes branch-specific formation of dendritic spines after learning. Science 344, 1173–1178 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Li, W., Ma, L., Yang, G. & Gan, W. B. REM sleep selectively prunes and maintains new synapses in development and learning. Nat. Neurosci. 20, 427–437 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. de Vivo, L. et al. Ultrastructural evidence for synaptic scaling across the wake/sleep cycle. Science 355, 507–510 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Kaskie, R. E., Graziano, B. & Ferrarelli, F. Schizophrenia and sleep disorders: links, risks, and management challenges. Nat. Sci. Sleep. 9, 227–239 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  17. Veatch, O. J. et al. Shorter sleep duration is associated with social impairment and comorbidities in ASD. Autism Res. 10, 1221–1238 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Mattai, A. A. et al. Sleep disturbances in childhood-onset schizophrenia. Schizophr. Res. 86, 123–129 (2006).

    Article  PubMed  Google Scholar 

  19. Robinson-Shelton, A. & Malow, B. A. Sleep disturbances in neurodevelopmental disorders. Curr. Psychiatry Rep. 18, 6 (2016).

    Article  PubMed  Google Scholar 

  20. Carmassi, C. et al. Systematic review of sleep disturbances and circadian sleep desynchronization in autism spectrum disorder: toward an integrative model of a self-reinforcing loop. Front. Psychiatry 10, 366 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Esbensen, A. J. & Schwichtenberg, A. J. Sleep in neurodevelopmental disorders. Int. Rev. Res. Dev. Disabil. 51, 153–191 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Bissonette, G. B. & Roesch, M. R. Development and function of the midbrain dopamine system: what we know and what we need to. Genes Brain Behav. 15, 62–73 (2016).

    Article  CAS  PubMed  Google Scholar 

  23. Marotta, R. et al. The neurochemistry of autism. Brain Sci. 10, 163 (2020).

  24. Chevallier, C., Kohls, G., Troiani, V., Brodkin, E. S. & Schultz, R. T. The social motivation theory of autism. Trends Cogn. Sci. 16, 231–239 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Paval, D. A dopamine hypothesis of autism spectrum disorder. Dev. Neurosci. 39, 355–360 (2017).

    Article  CAS  PubMed  Google Scholar 

  26. Ernst, M., Zametkin, A. J., Matochik, J. A., Pascualvaca, D. & Cohen, R. M. Low medial prefrontal dopaminergic activity in autistic children. Lancet 350, 638 (1997).

    Article  CAS  PubMed  Google Scholar 

  27. Eban-Rothschild, A., Rothschild, G., Giardino, W. J., Jones, J. R. & de Lecea, L. VTA dopaminergic neurons regulate ethologically relevant sleep-wake behaviors. Nat. Neurosci. 19, 1356–1366 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Lena, I. et al. Variations in extracellular levels of dopamine, noradrenaline, glutamate, and aspartate across the sleep–wake cycle in the medial prefrontal cortex and nucleus accumbens of freely moving rats. J. Neurosci. Res. 81, 891–899 (2005).

    Article  CAS  PubMed  Google Scholar 

  29. Dahan, L. et al. Prominent burst firing of dopaminergic neurons in the ventral tegmental area during paradoxical sleep. Neuropsychopharmacology 32, 1232–1241 (2007).

    Article  CAS  PubMed  Google Scholar 

  30. Yu, X. et al. GABA and glutamate neurons in the VTA regulate sleep and wakefulness. Nat. Neurosci. 22, 106–119 (2019).

    Article  CAS  PubMed  Google Scholar 

  31. Oishi, Y. et al. Activation of ventral tegmental area dopamine neurons produces wakefulness through dopamine D2-like receptors in mice. Brain Struct. Funct. 222, 2907–2915 (2017).

    Article  CAS  PubMed  Google Scholar 

  32. Bariselli, S. et al. Role of VTA dopamine neurons and neuroligin 3 in sociability traits related to nonfamiliar conspecific interaction. Nat. Commun. 9, 3173 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  33. Gunaydin, L. A. et al. Natural neural projection dynamics underlying social behavior. Cell 157, 1535–1551 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Zimprich, A. et al. A robust and reliable non-invasive test for stress responsivity in mice. Front. Behav. Neurosci. 8, 125 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  35. Nakajima, M., Gorlich, A. & Heintz, N. Oxytocin modulates female sociosexual behavior through a specific class of prefrontal cortical interneurons. Cell 159, 295–305 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Li, K., Nakajima, M., Ibanez-Tallon, I. & Heintz, N. A cortical circuit for sexually dimorphic oxytocin-dependent anxiety behaviors. Cell 167, 60–72 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Cao, W. et al. Gamma oscillation dysfunction in mPFC leads to social deficits in neuroligin 3 R451C knockin mice. Neuron 97, 1253–1260 (2018).

    Article  CAS  PubMed  Google Scholar 

  38. Liu, L. et al. Cell type-differential modulation of prefrontal cortical GABAergic interneurons on low gamma rhythm and social interaction. Sci. Adv. 6, eaay4073 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Huang, W. C., Zucca, A., Levy, J. & Page, D. T. Social behavior is modulated by valence-encoding mPFC–amygdala sub-circuitry. Cell Rep. 32, 107899 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Yamamuro, K. et al. A prefrontal–paraventricular thalamus circuit requires juvenile social experience to regulate adult sociability in mice. Nat. Neurosci. 23, 1240–1252 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  41. Dolen, G., Darvishzadeh, A., Huang, K. W. & Malenka, R. C. Social reward requires coordinated activity of nucleus accumbens oxytocin and serotonin. Nature 501, 179–184 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  42. Sun, F. et al. Next-generation GRAB sensors for monitoring dopaminergic activity in vivo. Nat. Methods 17, 1156–1166 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Liu, C., Goel, P. & Kaeser, P. S. Spatial and temporal scales of dopamine transmission. Nat. Rev. Neurosci. 22, 345–358 (2021).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  44. Wiedenmann, B. & Franke, W. W. Identification and localization of synaptophysin, an integral membrane glycoprotein of Mr 38,000 characteristic of presynaptic vesicles. Cell 41, 1017–1028 (1985).

    Article  CAS  PubMed  Google Scholar 

  45. Zhou, Y. et al. Mice with Shank3 mutations associated with ASD and schizophrenia display both shared and distinct defects. Neuron 89, 147–162 (2016).

    Article  CAS  PubMed  Google Scholar 

  46. Szelenyi, I. Flupirtine, a re-discovered drug, revisited. Inflamm. Res. 62, 251–258 (2013).

    Article  CAS  PubMed  Google Scholar 

  47. Li, S. B. et al. Hyperexcitable arousal circuits drive sleep instability during aging. Science 375, eabh3021 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Li, S. B., Nevarez, N., Giardino, W. J. & de Lecea, L. Optical probing of orexin/hypocretin receptor antagonists. Sleep 41, zsy141 (2018).

  49. Adamantidis, A. R., Zhang, F., Aravanis, A. M., Deisseroth, K. & de Lecea, L. Neural substrates of awakening probed with optogenetic control of hypocretin neurons. Nature 450, 420–424 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Li, S. B., Giardino, W. J. & de Lecea, L. in Current Topics in Behavioral Neurosciences, Vol. 33 (eds Lawrence, A. & de Lecea, L.) 93–104 (Springer, 2016).

  51. Schoch, S. F. et al. Across-night dynamics in traveling sleep slow waves throughout childhood. Sleep 41, zsy165 (2018).

  52. Wisor, J. P., Rempe, M. J., Schmidt, M. A., Moore, M. E. & Clegern, W. C. Sleep slow-wave activity regulates cerebral glycolytic metabolism. Cereb. Cortex 23, 1978–1987 (2013).

    Article  PubMed  Google Scholar 

  53. Bhatt, D. H., Zhang, S. & Gan, W. B. Dendritic spine dynamics. Annu. Rev. Physiol. 71, 261–282 (2009).

    Article  CAS  PubMed  Google Scholar 

  54. Bian, W. J., Miao, W. Y., He, S. J., Qiu, Z. & Yu, X. Coordinated spine pruning and maturation mediated by inter-spine competition for cadherin/catenin complexes. Cell 162, 808–822 (2015).

    Article  CAS  PubMed  Google Scholar 

  55. Moyer, C. E. & Zuo, Y. Cortical dendritic spine development and plasticity: insights from in vivo imaging. Curr. Opin. Neurobiol. 53, 76–82 (2018).

    Article  CAS  PubMed  Google Scholar 

  56. Laviola, G., Macri, S., Morley-Fletcher, S. & Adriani, W. Risk-taking behavior in adolescent mice: psychobiological determinants and early epigenetic influence. Neurosci. Biobehav. Rev. 27, 19–31 (2003).

    Article  PubMed  Google Scholar 

  57. Kurth, S. et al. Traveling slow oscillations during sleep: a marker of brain connectivity in childhood. Sleep 40, zsx121 (2017).

  58. Buchmann, A. et al. EEG sleep slow-wave activity as a mirror of cortical maturation. Cereb. Cortex 21, 607–615 (2011).

    Article  PubMed  Google Scholar 

  59. Kim, H. J. et al. Systematic analysis of expression signatures of neuronal subpopulations in the VTA. Mol. Brain 12, 110 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Krishnan, V. et al. Autism gene Ube3a and seizures impair sociability by repressing VTA Cbln1. Nature 543, 507–512 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Woolley, C. S. & McEwen, B. S. Roles of estradiol and progesterone in regulation of hippocampal dendritic spine density during the estrous cycle in the rat. J. Comp. Neurol. 336, 293–306 (1993).

    Article  CAS  PubMed  Google Scholar 

  62. Makinodan, M., Rosen, K. M., Ito, S. & Corfas, G. A critical period for social experience-dependent oligodendrocyte maturation and myelination. Science 337, 1357–1360 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Li, S. B. et al. Hypothalamic circuitry underlying stress-induced insomnia and peripheral immunosuppression. Sci. Adv. 6, eabc2590 (2020).

  64. Beier, K. T. et al. Circuit architecture of VTA dopamine neurons revealed by systematic input–output mapping. Cell 162, 622–634 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Yu, H. et al. Social touch-like tactile stimulation activates a tachykinin 1–oxytocin pathway to promote social interactions. Neuron 110, 1051–1067 (2022).

    Article  CAS  PubMed  Google Scholar 

  66. Nguyen, A. W. & Daugherty, P. S. Evolutionary optimization of fluorescent proteins for intracellular FRET. Nat. Biotechnol. 23, 355–360 (2005).

    Article  CAS  PubMed  Google Scholar 

  67. Zacharias, D. A., Violin, J. D., Newton, A. C. & Tsien, R. Y. Partitioning of lipid-modified monomeric GFPs into membrane microdomains of live cells. Science 296, 913–916 (2002).

    Article  CAS  PubMed  Google Scholar 

  68. Chan, K. Y. et al. Engineered AAVs for efficient noninvasive gene delivery to the central and peripheral nervous systems. Nat. Neurosci. 20, 1172–1179 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Gomez, J. L. et al. Chemogenetics revealed: DREADD occupancy and activation via converted clozapine. Science 357, 503–507 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Kaidanovich-Beilin, O., Lipina, T., Vukobradovic, I., Roder, J. & Woodgett, J. R. Assessment of social interaction behaviors. J. Vis. Exp. https://doi.org/10.3791/2473 (2011).

  71. Winslow, J. T. Mouse social recognition and preference. Curr. Protoc. Neurosci. Chapter 8, Unit 8.16 (2003).

    PubMed  Google Scholar 

  72. Rolls, A. et al. Optogenetic disruption of sleep continuity impairs memory consolidation. Proc. Natl Acad. Sci. USA 108, 13305–13310 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Walf, A. A. & Frye, C. A. The use of the elevated plus maze as an assay of anxiety-related behavior in rodents. Nat. Protoc. 2, 322–328 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Giardino, W. J. et al. Parallel circuits from the bed nuclei of stria terminalis to the lateral hypothalamus drive opposing emotional states. Nat. Neurosci. 21, 1084–1095 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Ungless, M. A. & Grace, A. A. Are you or aren’t you? Challenges associated with physiologically identifying dopamine neurons. Trends Neurosci. 35, 422–430 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Olivier Friard, M. G. BORIS: a free, versatile open-source event-logging software for video/audio coding and live observations. Methods Ecol. Evolution 7, 1325–1330 (2016).

    Article  Google Scholar 

  77. Scheffzuk, C. et al. Selective coupling between theta phase and neocortical fast gamma oscillations during REM-sleep in mice. PLoS ONE 6, e28489 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  78. Hung, L. W. et al. Gating of social reward by oxytocin in the ventral tegmental area. Science 357, 1406–1411 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Sgritta, M. et al. Mechanisms underlying microbial-mediated changes in social behavior in mouse models of autism spectrum disorder. Neuron 101, 246–259 (2019).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank G. Feng and J. Ding for transgenic mice and X. Yu for constructs. We thank A. Khan, K.J. Jennings, K.R. Murphy and Y. Sun for assistance. We thank A. Olson and G. Wang and the Stanford Wu Tsai Neuroscience Microscopy Service (NIH NS069375) for technical support. We thank Merck for providing the DORA12 compound. We acknowledge all lab members of the L.d.L. lab for critical feedback. This work was supported by Human Frontier Science Program fellowship LT000338/2017-L (W.-J.B.), Brain & Behavior Research Foundation NARSAD Young Investigator grant 29952 (W.-J.B.) and National Institutes of Health grants R01 MH102638 (L.d.L.), R01 MH087592 (L.d.L.), R01 MH116470 (L.d.L.), R01 DA011289 (J.A.K.), F32 NS123008 (C.L.B) and 5T32DA035165-08 (C.L.B).

Author information

Authors and Affiliations

Authors

Contributions

W.-J.B. and L.d.L. designed the project. W.-J.B. performed all experiments and data analyses except for the slice electrophysiology experiments, which were designed by J.A.K. and C.L.B and performed by C.L.B. W.-J.B. wrote the manuscript with contributions from C.L.B, J.A.K. and L.d.L.

Corresponding authors

Correspondence to Wen-Jie Bian or Luis de Lecea.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Neuroscience thanks Shlomo Wagner, William Wisden, and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

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

Extended data

Extended Data Fig. 1 Developmental shaping of social preferences and adolescent sleep disruption (SD).

a, Schematics of the three-chamber social interaction assay. b, c, Naïve WT C57BL/6J mice showed age-dependent increases of sociability (b) and social novelty preference (c) in three-chamber social interactions over the developmental course (n = 10 each). Preference indices were calculated as indicated in each graph. One-way ANOVA, b, F (3, 36) = 5.33, P = 0.004; c, F (3, 36) = 4.23, P = 0.01, Dunnett’s post-hoc comparisons to P28, b, P56, ** P = 0.007, P84, ** P = 0.005; c, P56, ** P = 0.004. df, Hourly percentage of Wake (d), NREM (e) and REM (f) sleep in adolescent mice (P35–42) over a 24-hr light-dark cycle before (Baseline, black), during (red), and after (blue) the 5 days with daily SD sessions (ZT 2–6, indicated by the grey stripe). The 3 groups of data were from the same mice (n = 4). RM 2-way ANOVA followed by Bonferroni’s post-tests, Wake, Time × Treatment F (46, 207) = 3.18, P = 9.5 × 10−9, post-test During SD vs. Baseline, ZT2-3 ** P = 0.008, ZT3-4 ** P = 0.001, ZT4-5 ** P = 0.005, ZT5-6 * P = 0.02; NREM, Time × Treatment F (46, 207) = 2.90, P = 1.4 × 10−7, post-test During SD vs. Baseline, ZT2-3 * P = 0.01, ZT3-4 *** P = 0.0007, ZT4-5 ** P = 0.007, ZT5-6 * P = 0.03; After SD vs. Baseline, ZT15-16 * P = 0.02; REM, Time × Treatment F (46, 207) = 3.23, P = 5.9 × 10−9, post-test During SD vs. Baseline, ZT2-3 *** P = 0.0008, ZT3-4 ** P = 0.002, ZT4-5 *** P = 0.0004, ZT5-6 ** P = 0.02. gk, Total amount of time (g), bout number (h) and average bout length (i) of Wake, NREM and REM states between ZT 2–6, total amount of 3 states during the following 6 hrs (ZT 6–12, j) and over the 24-hour light-dark cycle (k) (n = 9). One-way ANOVA within each state, g, Wake, F (1.48, 11.83) = 230.2, P = 0.000000001; NREM, F (1.28, 10.24) = 167.7, P = 0.00000005; REM, F (1.78, 14.21) = 79.99, P = 0.00000003; h, Wake, F (1.72, 13.75) = 54.78, P = 0.0000005; NREM, F (1.87, 14.94) = 70.18, P = P = 0.00000003; REM, F (1.42, 11.38) = 58.42, P = 0.000003; i, Wake, F (1.01, 8.07) = 24.47, P = 0.001; NREM, F (1.40, 11.18) = 67.54, P = P=0.000002; j, Wake, F (1.60, 12.81) = 11.30, P = 0.002; NREM, F (1.49, 11.94) = 11.41, P = 0.003; REM, F (1.48, 11.87) = 5.01, P = 0.03; k, Wake, F (1.93, 15.40) = 10.27, P = 0.002; NREM, F (1.80, 14.40) = 7.72, P = 0.006; REM, F (1.93, 15.43) = 7.59, P = 0.005. Dunnett’s multiple comparisons to Baseline, * P < 0.05; ** P < 0.01; *** P < 0.001. Unpaired t-test for REM in i, P = 0.31. l, m, EEG power spectrum of NREM (l) and REM (m) sleep was not altered after 5 days of SD compared to Baseline (n = 9). n, Plasma corticosterone level of adolescent mice (P35–42) immediately after the SD session on 1st day and 5th day of SD compared to naïve mice of same age receiving no shake (n = 10 each). One-way ANOVA, F (2, 27) = 2.30, P = 0.12, followed by Tukey’s multiple comparisons test, all n.s. Data are shown as mean ± s.e.m. All tests were two-sided. n.s., not significant.

Source data

Extended Data Fig. 2 Comparisons of same-sex social interaction behavior between sexes.

a, Male (left) and female (right) test mice received Ctrl or SD between P35–42 and the three-chamber assay were performed using gender-matched stimulus mice, respectively, at P56. be, Interaction time with the empty cup (E, b), stranger 1 (S1) in Trial 1(c), S1 in Trial 2 (d), and stranger 2 (S2) in Trial 2 (e) during the three-chamber test was binned every 2 min and presented for male and female test mice separately. n = 10 mice each. RM 3-way ANOVA. P value of each variable (Sex, Time and Treatment) was indicated in the graph. fj, Total interaction time (f), number of interaction bouts (g), average length of interaction bouts (h), total time spent in each chamber (i), and number of entries of each chamber (j) were presented separately for male-male interactions and female-female interactions and compared directly considering Sex, Stimulus and Treatment as 3 independent variables using RM 3-way ANOVA. n = 10 mice each. ANOVA P values are indicated in the graphs. In all parameters, Sex does not show a significant contribution to total variation (P > 0.05). Additional 2-way ANOVA was performed for total interaction time (f) and number of interaction bouts (g) in Trial 2, S1 and S2 categories (box insets) followed by Bonferroni’s post-tests comparing sexes within treatment. ANOVA P values were indicated in the box insets. All post-tests, male vs. female, P > 0.05. Data are shown as mean ± s.e.m. All tests used were two-tailed. Data of male and female subjects in this Extended Data Figure were combined and presented in Fig. 1ci. Data are shown as mean ± s.e.m. All tests were two-sided.

Source data

Extended Data Fig. 3 Behavioral probing of adolescent SD mice.

a, Male mice received Ctrl or SD between P42–49 and were tested at P52–56 using the three-chamber assay with male stimulus mice (n = 10 each). Interaction time, RM 2-way ANOVA, Stimulus × Treatment F (3, 54) = 1.20, P = 0.32, Tukey’s post-test, Trial 1 Ctrl * P = 0.03, SD ** P = 0.004, Trial 2 Ctrl ** P = 0.009, SD P = 0.11. Preference indices, Welch’s t-test, ** P = 0.007. b, Male test mice received Ctrl or SD between P35–42 and the three-chamber assay using female stimulus mice at P56 (n = 10 each). Interaction time, RM 2-way ANOVA, Stimulus × Treatment F (3, 54) = 0.28, P = 0.84, Tukey’s post-test, Trial 1 Ctrl ** P = 0.002, SD *** P = 0.0003, Trial 2 Ctrl * P = 0.046, SD *** P = 0.0002; Preference indices, Welch’s t-test, both P > 0.05. c, d, Sociability preference (c) and social novelty preference (d) were not significantly different between mice receiving Ctrl protocol during adolescence and naïve mice with normal, undisturbed sleep. Only male mice were included in comparisons. Ctrl data were the same as that in Extended Data Fig. 2 (n = 10 each). Welch’s t-test. e, Ctrl and SD mice spent equal time in the two side chambers during the habituation phase of three-chamber assay (n = 7 each). Paired t-test. f, Comparison of social interaction performance between male mice sleep-deprived using the gentle-touch method with those deprived by automated SD in 3 cohorts that were wean at approximately same time. Samples were combined and included in the SD group in Fig. 1g. g, h, Mice receiving previous Ctrl or SD (n = 13 in Ctrl; 12 in SD) at P35–42 showed similar locomotion dynamics in the three-chamber apparatus (g) and comparable total distance traveled during the whole 20-min session of three-chamber assay (h). g, RM 2-way ANOVA followed by Bonferroni’s post-test, all n.s. h, Welch’s t-test. i, Time spent in self-grooming during the three-chamber assay was similar between groups (n = 15 in Ctrl; 14 in SD). Welch’s t-test P = 0.42. j, Mice receiving SD at P35–42 showed similar latency to their first entry to the open arm when placed in an elevated plus maze at P70–84 (n = 9 in Ctrl; 8 in SD). Welch’s t-test P = 0.83. The two mice showing latency of 300 sec did not enter the open arm at all during the whole 5-min session. k, The novel object recognition test with reduced memory requirement. The Test trial was performed immediately after 10 min of Training session. m, Interaction time with each object during the Test trial was plotted to the left Y-axis, and the ratio of time spent with the novel object over total interaction time was plotted to the right Y-axis. n = 10 in Ctrl; 9 in SD. Left, paired t-test, Ctrl, ** P = 0.008, SD, * P = 0.03. Right, Welch’s t-test, * P = 0.04. l, The social memory tests. Upper, the 1 stimulus paradigm, where the same stimulus mouse S1 was re-presented to the test mouse in Trial 2 after a 30-min interval; Lower, the 2 stimuli paradigm in which Trial 2 contains a novel stimulus mouse S2 in addition to S1. n, Interaction time with the single stimulus mouse in each trial of the 1 stimulus test. n = 10 each. Paired t-test, both * P = 0.02. o, Interaction time with the 2 stimulus mice during the 2 stimulus test. n = 10 each. RM one-way ANOVA, Ctrl, F (1.96, 17.66) = 18.94, P < 0.0001; SD, F (1.68, 15.16) = 4.44, P = 0.04, Tukey’s post-test, Ctrl, S1, Trial 1 vs trial 2, *** P = 0.0007; Trial 2, S1 vs. S2, * P = 0.03, SD, S1, Trial 1 vs. Trial 2, * P = 0.03. p, Comparisons of ratio of S1 interaction (Trial 2/Trial 1) between paradigms. n = 10 each. Welch’s t-test, Ctrl, ** P = 0.009; SD, P = 0.68. Data are shown as mean ± s.e.m. All tests were two-sided.

Source data

Extended Data Fig. 4 Fiber photometry recording of VTADA activity during social interactions.

a, Social performance of DATCre mice in the fiber photometry setup (NS, normal sleep. n = 14 in Ctrl/NS; 9 in SD, including mice with poor GCaMP signals). RM 2-way ANOVA, Stimulus × Treatment F (3, 63) = 4.49, P = 0.006, followed by Tukey’s post-test, Trial 1, *** P < 0.0001, ** P = 0.004; Trial 2, ** P = 0.002. b, The mean amplitude of Ca2+ transients detected during the 20 min session (Trials 1 + 2) were similar between Ctrl and SD groups (n = 6 mice, transient peak Z-score were first averaged within each animal). Welch’s t-test, P = 0.41. c, d, Representative traces of GCaMP signals in Ctrl and SD mice during Trial 1 (c) and Trial 2 (d) of the social interaction assay. Colored stripes indicate interaction bouts. Red circles on trace indicate Ca2+ transients detected. e, Example GCaMP traces of the first, second, third and last interaction bout of each category. Time 0 s indicates bout onset, and yellow stripes indicate bout duration. f, g, Ctrl and SD mice in their home-cages were given a food pellet, and the VTA GCaMP6f signals were recorded and aligned to the time point when they first contacted the food pellets. GCaMP6f traces of 10–12 trials from n = 4 mice (2–3 trials each animal). across all trials were averaged and shown for Ctrl (f) and SD (g) animals. Shaded area indicates s.e.m. Insets show area under curve (AUC) of GCaMP6f signals within 0–10 s upon contact with the food pellet or a neutral object of similar size (for example, a Q-tip head), n = 4 mice, paired t-test, Ctrl, * P = 0.04; SD, ** P = 0.002. h, Relative changes in AUC, n = 4 mice, Welch’s t-test, P = 0.56. Data are shown as mean ± s.e.m. All tests were two-sided.

Source data

Extended Data Fig. 5 Adolescent SD increased the membrane resistance but did not affect the intrinsic excitability of or synaptic inputs to VTADA neurons.

a, Representative image of a horizontal acute brain slice from a DATCre::Ai14 mouse. VTADA neurons were filled with neurobiotin during whole-cell recording. The slices were subsequently fixed in 4% PFA and stained with Streptavidin (Alexa FluorTM 488 conjugate, Invitrogen) and antibodies against TH. Scale bar, 100 µm. b, Representative traces of spontaneous firing measured in a cell from a Ctrl (top, black) and SD animal (bottom, red; scale 50 mV and 5 s). c, Evoked firing in response to depolarizing current (90 pA, upper square pulses in gray) in a Ctrl (top black trace) and SD animal (red top trace; scale bars 50 mV and 0.1 s). The bottom two traces and current steps in Ctrl and SD conditions illustrate the hyperpolarizing response to a -20 pA step used for measurements of membrane resistance (indicated by the connected gray dashed [baseline] and vertical solid lines [deflection amplitude]). d, There were no observed differences in spontaneous firing frequency (n = 23 Ctrl and 27 SD neurons from 8 mice in each group, t(14) = 0.6394, P = 0.5329; data transformed with a square root, nested t-test) between the control (gray) and sleep-deprived animals (red). e, f, There were no effects of sleep deprivation (red) on the number of action potentials (APs) fired (n = 23 Ctrl and 26 SD neurons, F(16, 752) = 0.1899, P = 0.9998; two-way repeated measure ANOVA) or the firing frequency (n = 23 Ctrl and 26 SD neurons, F(16, 752) = 0.4933, P = 0.9511; two-way repeated measure ANOVA) in response to current pulse injections after adolescent sleep deprivation. g, h, There were no observed differences in hyperpolarization-activated (Ih) currents (n = 22 neurons from 7 Ctrl mice and 27 neurons from 8 SD mice, t(13) = 0.6605, P = 0.5204; nested t-test) or capacitance (n = 23 Ctrl and 27 SD neurons from 8 mice in each group, t(14) = 1.364, P = 0.1941; nested t-test) between Ctrl (left, gray) and SD animals (right, red). i, Membrane resistance was significantly increased in SD animals (M = 769.7 ± 54.32 MΩ; red) relative to Ctrl (M = 574.8 ± 36.01 MΩ; gray; n = 23 Ctrl and 27 SD neurons from 8 mice in each group, t(14) = 2.396, *P = 0.0311; nested t-test). j, k, SD did not have a significant effect on the coefficient of variation (CV) of rostrally evoked excitatory (n = 13 neurons from 9 mice in each group, t(24) = 0.9020, P = 0.3760; data transformed with square root, nested t-test) or inhibitory synaptic inputs (n = 10 neurons from 8 SD mice and 13 neurons from 10 Ctrl mice, t(21) = 1.795, P = 0.0870; nested t-test) to VTADA neurons. l, m, No observed differences between Ctrl (gray) and SD animals (red) in the paired pulse ratio (PPR) of rostrally evoked excitatory (n = 13 neurons from 9 mice in each group, t(24) = 0.0684, P = 0.9460; nested t-test) or inhibitory inputs (n = 10 neurons from 8 SD mice and 13 neurons from 10 Ctrl mice, t(16) = 1.927, P = 0.0720; data transformed with square root, nested t-test) to VTADA neurons. n, o, SD had no effect on the frequency (n = 10 neurons from 9 Ctrl mice and 9 neurons from 8 SD mice, t(15) = 0.0400, P = 0.9686; nested t-test) or amplitude (n = 10 neurons from 9 Ctrl mice and 9 neurons from 8 SD mice, t(17) = 1.544, P = 0.1410; nested t-test) of miniature excitatory postsynaptic currents (mEPSCs) in VTADA neurons. p, q, No significant differences in the amplitude (n = 10 neurons from 8 mice in each group, t(14) = 0.4431, P = 0.6645; nested t-test) or frequency (n = 10 neurons from 8 mice in each group, t(14) = 0.9301, P = 0.3681; nested t-test) of miniature inhibitory postsynaptic currents (mIPSCs) recorded in VTADA neurons from Ctrl (gray) or SD (red) animals. For all bar graphs, each bar represents 1 animal while the individual data points are from neurons recorded from that animal. Data are shown as mean ± s.e.m. All tests were two-sided.

Source data

Extended Data Fig. 6 Axonal projections of VTADA neurons in NAc and mPFC.

a, Numbers of infected VTADA neurons in DATCre mice (n = 5 in Ctrl; 2 in SD). b, Representative image showing layer 1–3 of mPFC (anterior cingulate area). Only very few SYP-mRuby puncta were present in layer 1 compared to deeper layers. Scale bar, 50 µm. c, Quantification of areal density of SYP-mRuby puncta in NAc as well as layer 2/3 and layer 5/6 of mPFC. 17–24 fields were imaged in each region from 3 animals, and SYP-mRuby puncta were counted. F (2, 55) = 19.79, P < 0.0001 by one-way ANOVA, followed by Tukey’s post-test, *** P < 0.0001. d-f, A single plane of Z-stack confocal image in separate and merged channel views showing the soma of a mPFC pyramidal neuron (same neuron in Fig. 4f, Ctrl). Arrows indicate SYP-mRuby puncta and corresponding ‘pits’ on the cytoplasmic membrane of the target neuron. Scale bar, 5 µm. g, k, n, Representative examples of MSN dendrites in NAc (g) and apical oblique dendrites (k) or basal dendrites (n) from mPFC pyramidal neurons in Ctrl and SD animals. Scale bar, 5 µm. h-j, Total spine density of NAc MSNs (h, n = 48 neurons from 5 mice in Ctrl; 39 neurons from 4 mice in SD), percentage of dendritic spines in contact with SYP-mRuby puncta (i, n = 48 neurons from 5 mice in Ctrl; 39 neurons from 4 mice in SD) and normalized integrated intensity of SYP-mRuby puncta that colocalized with labeled spines (j, n = 25 neurons from 4 mice in Ctrl; 21 neurons from 3 mice in SD) were not changed by adolescent SD. l, m, Total spine density (l) and percentage of SYP-mRuby-contacting spines (m) on apical oblique dendrites of mPFC pyramidal neurons (n = 37 neurons from 5 mice in Ctrl; 33 neurons from 4 mice in SD). o, p, Total spine density (o) and percentage of SYP-mRuby-contacting spines (p) on basal dendrites of mPFC pyramidal neurons (n = 40 neurons from 6 mice in Ctrl; 31 neurons from 4 mice in SD). h–p, all P > 0.05 by Welch’s t-test. Data are shown as mean ± s.e.m. All tests were two-sided.

Source data

Extended Data Fig. 7 VTADA neurons were overexcited by SD during adolescence.

DATCre mice received AAV-DJ-EF1α-DIO-GCaMP6f injections in VTA at P21 and implantations for fiber photometry and EEG/EMG recording at P30. Simultaneous fiber photometry and EEG/EMG recordings were performed at P37–38. a–d, Representative GCaMP (top, black), EEG (middle, red) and EMG (bottom, blue) signals during spontaneous NREM sleep (a), wake (b), REM sleep (c) and SD sessions (d). Red circles on GCaMP traces indicate Ca2+ transients detected. e, f, Quantification of frequency (e) and amplitude (peak Z-score, f) of Ca2+ transients detected in each state. n = 4 mice. e, RM one-way ANOVA, F (3, 9) = 5.68, P = 0.02, followed by Dunnett’s multiple comparisons to NREM, ** P = 0.007; NREM vs. SD, # P = 0.01 by paired t-test. f, RM one-way ANOVA, F (3, 9) = 2.57, P = 0.12; NREM vs. Wake, # P = 0.02, NREM vs SD, # P = 0.02, by paired t-test. Data are shown as mean ± s.e.m. All tests were two-sided.

Source data

Extended Data Fig. 8 CNO administration in adolescent hM3DqDAT and hM4DiDAT mice.

a, Cre- and hM3DqDAT mice were placed in an open-field arena 30 min following the i.p. injection of CNO (2 mg/kg). Total distance traveled in the arena during an 8-min session was measured (n = 5). * P = 0.01 by Welch’s t-test. b, c, The 2-bottle free-choice drinking test. Cre- and hM3DqDAT mice were kept in a cage where they have free access to the bottles containing H2O and CNO solution (50 mg/L), respectively, on opposite sides of the cage. The volume of H2O and CNO solution consumed over 24 hours were recorded. Compared to Ctrl mice, SD mice drank more CNO solution than H2O (b), and the preference for CNO (c), measured by the portion of CNO volume consumed over the total liquid consumed (H2O + CNO), was increased in the SD group (n = 5). b, * P = 0.01 by paired t-test; c, ** P = 0.003 by Welch’s t-test. d, Social performance of Cre- and mCherryDAT animals in the three-chamber assay at P56. eg, DATCre mice were injected with AAV-DJ-EF1α–DIO-hM3Dq in VTA at P21 and EEG recording was performed at P35–42. CNO (2mg/kg, i.p.) or saline was administered to the animal at ZT2. Hourly percentage of Wake (e), NREM (f) and REM (g) states during the 12-hr light phase are shown. Control group includes one mCherryDAT mouse with CNO injection and three hM3DqDAT mice with saline injections (total n = 4); hM3DqDAT + CNO, n = 5. RM 2-way ANOVA followed by Bonferroni’s post-test, e, f, both ** P = 0.006. hj, DATCre mice were injected with AAV-DIO-hM4Di in VTA at P21 and EEG recording was performed at P35–42. Mice were subjected to SD protocol during ZT 2–6, and CNO (1mg/kg, i.p.) was administered to the animal at ZT2 and ZT4. Compared to the baseline recording (1 day before SD + CNO treatment) where the animals did not receive SD or CNO, the SD protocol can still effectively deprive both NREM (i) and REM (j) sleep even with the presence of CNO (n = 4). RM 2-way ANOVA followed by Bonferroni’s post-test, h, ZT 2–3 *** P = 0.0005, ZT 3–4 *** P < 0.0001, ZT4–5 ** P = 0.009, ZT5-6 * P =0.04; i, ZT 2–3 *** P = 0.0002, ZT 3–4 *** P < 0.0001, ZT 4–5 * P = 0.01; j, ZT 3–4 ** P = 0.001. k, l, Quantification of each state during the 4 hours after the CNO injection in hM3DqDAT mice (k) and in hM4DiDAT mice with concurrent SD (l) as well as respective controls. k, n = 4 in control; 5 in hM3DqDAT + CNO, Wake, * P = 0.03; NREM, * P = 0.02 by Welch’s t-test. l, n = 4 mice each, Wake, ** P = 0.003; NREM, ** P = 0.005 by Welch’s t-test. REM, * P = 0.03 by Mann-Whitney test. m, No significant difference was found in the social memory test between Cre– and hM3DqDAT mice after adolescent CNO injections (n = 5). Left, * P = 0.02, ** P = 0.005 by paired t-test. Right, P = 0.67 by Welch’s t-test. n, The DATCre mice that received AAV-DJ-EF1α-DIO-hM4Di-mCherry (hM4DiDAT) or mCherry control virus (mCherryDAT) on water regulation were given a single injection of CNO (2 mg/kg) and subsequently allowed access to both H2O and sucrose solution (1%, w/v) for 4 hours. The mCherryDAT mice showed strong preference to the sucrose, whereas the hM4DiDAT mice, although still consuming more sucrose solution than water, showed a significantly lowered sucrose preference, which was calculated as the volume of sucrose solution consumed divided by the total liquid volume consumed (H2O + sucrose, n = 5). Left, mCherryDAT, ** P = 0.001, hM4DiDAT, ** P = 0.005 by paired t-test. Right, ** P = 0.006 by Welch’s t-test. o, Novel object recognition test with reduced memory requirement was performed on adult mCherryDAT mice and hM4DiDAT mice (P56–60) with prior adolescent SD and CNO injections (P37–41). n = 7 in mCherryDAT; 9 in hM4DiDAT. Left, Wilcoxon matched-pairs signed rank test, mCherryDAT, * P = 0.02; hM4DiDAT, P = 0.10. Right, Welch’s t-test, P = 0.81. Data are shown as mean ± s.e.m. All tests were two-sided. n.s., not significant.

Source data

Extended Data Fig. 9 Adolescent sleep defects and adult in Shank3 InsG3680 mice.

a–c, Three-chamber social test was performed in InsG3680+/+ mutants (Homo, n = 8) and WT littermates (n = 9) at P56. Absolute interaction time and percentage of each interaction category are shown in a and b, respectively. Preference indices are shown in c. a, RM 2-way ANOVA, Stimulus × Genotype F (3, 45) = 2.18, P = 0.10 followed by Tukey’s post-test, P values from the post-test are indicated on the graph; b, RM 2-way ANOVA, Stimulus × Genotype F (3, 45) = 1.97, P = 0.13, Tukey’s post-test, *** P < 0.0001, ** P = 0.004, * P = 0.01; c, Sociability, Mann-Whitney test, P = 0.13, Social Novelty, Welch’s t-test, * P = 0.04. d–f, Hourly percentage of Wake (d), NREM (e) and REM (f) states over the 24-hr light-dark cycle at P35–42 in adolescent InsG3680 mice (n = 5 in WT; 8 in Homo). g, h, Amount of Wake, NREM and REM states in InsG3680 mice at P35–42 (n = 5 in WT; 8 in Homo) in the light phase (g) and the dark phase (h). Welch’s t-test, g, Wake, * P = 0.02 (t = 3.06, df = 8.24), NREM, * P = 0.04 (t = 2.32, df = 9.26), REM, * P = 0.03 (t = 2.46, df = 10.86). h, all P > 0.05. i–l, EEG Power spectrum of NREM (i) and REM(k) sleep at P35–42 (n = 5 in WT; 8 in Homo). Relative EEG powers of each frequency band are shown in j for NREM and l for REM. Welch’s t-test, j, Delta, P = 0.10, Theta, P = 0.63, Alpha, * P = 0.02, Beta, P = 0.43; l, Delta, P = 0.08, Theta, * P = 0.03, Alpha, P = 0.15, Beta, P = 0.90. m, The pAAV-mTH-Cre construct. Cre expression is under control of mouse tyrosine hydroxylase (TH) promoter. n–q, Representative images of a InsG3680 mouse which received injection of AAV-DJ-mTH-Cre + AAV-DJ-EF1α-DIO-mCherry in VTA and after two weeks of viral expression. Red, mCherry fluorescence; Green, TH immunostaining; Blue, DAPI. Scale bars, 200 µm. r, Dual labeling strategy in InsG3680 mice. s, Quantification showing the efficiency (% of TH/mCherry double positive neurons in total TH+ neurons) and specificity (% of double positive neurons in total mCherry+ neurons) of dopaminergic neuron labeling within the VTA area using co-injection of AAV-DJ-mTH-Cre and AAV-DJ-EF1α-DIO-mCherry (or -hM4Di-mCherry). n = 3 animals. t, The VTA axonal terminals in NAc were examined in WT (n = 40 neurons from 4 animals) and Homo (n = 38 neurons from 4 animals) InsG3680 mice at P70, using the dual labeling strategy with AAV-mTH-Cre. Welch’s t-test, P = 0.0000003. Data are shown as mean ± s.e.m. All tests were two-sided. n.s., not significant.

Source data

Extended Data Fig. 10 Effects of Flupirtine and DORA12 treatments on adolescent sleep and adult social interactions in Shank3 InsG3680 mice.

a–d, Hourly percentage of Wake (a) and REM (b) state over a 24-hr light-dark cycle and the quantification of each state during the light (c) or dark (d) phase in adolescent homozygous InsG3680 mice with Flupirtine treatment (n = 7 in Veh; 8 in Flup). RM 2-way ANOVA, a, Wake, Time x Treatment, F (23, 299) = 1.74, P = 0.02; b, REM, Time x Treatment, F (23, 299) = 0.90, P = 0.61, Bonferroni’s post-test, Wake ** P = 0.002, * P = 0.04. c, d, Welch’s t-test, NREM in Light Phase, * P = 0.048. e–h, Hourly percentage of Wake (f), NREM (g) and REM (h) state over a 24-hr light-dark cycle in adolescent homozygous InsG3680 mice with DORA treatment. Arrows indicate injections of DORA12 or vehicle (50% PEG400 in saline) at ZT2. n = 5 each. RM 2-way ANOVA with Bonferroni’s post-test. i–k, Quantification of each state during the light (i) or dark (j) phase and 4 hours following the injection of DORA12 or vehicle (k). n = 5 each. Welch’s t-test, k, NREM, * P = 0.04. l–n, Homozygous InsG3680 mice received daily DORA12 (20 mg/kg, i.p.) or vehicle (50% PEG400 in saline) for 5 consecutive days during P37–41, and three-chamber social interaction assay was performed at P56. n = 12 in Veh; 11 in DORA. Interaction time (l), RM 2-way ANOVA, Stimulus × Treatment F (3, 63) = 1.43, P = 0.24, Tukey’s post-test, Trial 1, Veh, ** P = 0.004, DORA, ** P = 0.003; Trial 2, Veh, P = 0.45, DORA, * P = 0.03. Preference indices of sociability (m) and social novelty (n), Welch’s t-test, m, t = 0.93, df = 19.22, P = 0.36, n, t = 3.01, df = 16.29, ** P = 0.008. Data are shown as mean ± s.e.m. All tests were two-sided. n.s., not significant.

Source data

Supplementary information

Supplementary Information

Supplementary Table 1. Statistical details of tests used in Figs. 1–6.

Reporting Summary

Source data

Source Data Fig. 1

Statistical source data

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 9

Statistical source data.

Source Data Extended Data Fig. 10

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Bian, WJ., Brewer, C.L., Kauer, J.A. et al. Adolescent sleep shapes social novelty preference in mice. Nat Neurosci 25, 912–923 (2022). https://doi.org/10.1038/s41593-022-01076-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41593-022-01076-8

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