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:

VTA dopaminergic neurons regulate ethologically relevant sleep–wake behaviors

Abstract

Dopaminergic ventral tegmental area (VTA) neurons are critically involved in a variety of behaviors that rely on heightened arousal, but whether they directly and causally control the generation and maintenance of wakefulness is unknown. We recorded calcium activity using fiber photometry in freely behaving mice and found arousal-state-dependent alterations in VTA dopaminergic neurons. We used chemogenetic and optogenetic manipulations together with polysomnographic recordings to demonstrate that VTA dopaminergic neurons are necessary for arousal and that their inhibition suppresses wakefulness, even in the face of ethologically relevant salient stimuli. Nevertheless, before inducing sleep, inhibition of VTA dopaminergic neurons promoted goal-directed and sleep-related nesting behavior. Optogenetic stimulation, in contrast, initiated and maintained wakefulness and suppressed sleep and sleep-related nesting behavior. We further found that different projections of VTA dopaminergic neurons differentially modulate arousal. Collectively, our findings uncover a fundamental role for VTA dopaminergic circuitry in the maintenance of the awake state and ethologically relevant sleep-related behaviors.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Figure 1: Population activity of Th+ VTA neurons across sleep–wake states.
Figure 2: Chemogenetic inhibition of Th+ VTA neurons decreases wakefulness and increases sleep.
Figure 3: Activity in Th+ VTA neurons is necessary for wake maintenance even in the face of salient stimuli.
Figure 4: Chemogenetic inhibition of Th+ VTA neurons promotes nest-building behavior.
Figure 5: Optogenetic activation of Th+ VTA neurons is sufficient to initiate wakefulness.
Figure 6: Optogenetic activation of Th+ VTA neurons is sufficient to maintain wakefulness and suppress nest-building behavior.
Figure 7: Th+ VTA neuron projections differentially modulate arousal.

Similar content being viewed by others

References

  1. Robbins, T.W. & Everitt, B.J. A role for mesencephalic dopamine in activation: commentary on Berridge (2006). Psychopharmacology (Berl.) 191, 433–437 (2007).

    Article  CAS  Google Scholar 

  2. Berridge, K.C. & Robinson, T.E. What is the role of dopamine in reward: hedonic impact, reward learning, or incentive salience? Brain Res. Brain Res. Rev. 28, 309–369 (1998).

    Article  CAS  PubMed  Google Scholar 

  3. Salamone, J.D. & Correa, M. The mysterious motivational functions of mesolimbic dopamine. Neuron 76, 470–485 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Wise, R.A. Dopamine, learning and motivation. Nat. Rev. Neurosci. 5, 483–494 (2004).

    Article  CAS  PubMed  Google Scholar 

  5. Schultz, W. Multiple dopamine functions at different time courses. Annu. Rev. Neurosci. 30, 259–288 (2007).

    Article  CAS  PubMed  Google Scholar 

  6. España, R.A. & Scammell, T.E. Sleep neurobiology from a clinical perspective. Sleep 34, 845–858 (2011).

    PubMed  PubMed Central  Google Scholar 

  7. Saper, C.B., Fuller, P.M., Pedersen, N.P., Lu, J. & Scammell, T.E. Sleep state switching. Neuron 68, 1023–1042 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Steinfels, G.F., Heym, J., Strecker, R.E. & Jacobs, B.L. Behavioral correlates of dopaminergic unit activity in freely moving cats. Brain Res. 258, 217–228 (1983).

    Article  CAS  PubMed  Google Scholar 

  9. Trulson, M.E. & Preussler, D.W. Dopamine-containing ventral tegmental area neurons in freely moving cats: activity during the sleep-waking cycle and effects of stress. Exp. Neurol. 83, 367–377 (1984).

    Article  CAS  PubMed  Google Scholar 

  10. Trulson, M.E., Preussler, D.W. & Howell, G.A. Activity of substantia nigra units across the sleep-waking cycle in freely moving cats. Neurosci. Lett. 26, 183–188 (1981).

    Article  CAS  PubMed  Google Scholar 

  11. Miller, J.D., Farber, J., Gatz, P., Roffwarg, H. & German, D.C. Activity of mesencephalic dopamine and non-dopamine neurons across stages of sleep and walking in the rat. Brain Res. 273, 133–141 (1983).

    Article  CAS  PubMed  Google Scholar 

  12. Jones, B.E., Bobillier, P., Pin, C. & Jouvet, M. The effect of lesions of catecholamine-containing neurons upon monoamine content of the brain and EEG and behavioral waking in the cat. Brain Res. 58, 157–177 (1973).

    Article  CAS  PubMed  Google Scholar 

  13. Boutrel, B. & Koob, G.F. What keeps us awake: the neuropharmacology of stimulants and wakefulness-promoting medications. Sleep 27, 1181–1194 (2004).

    Article  PubMed  Google Scholar 

  14. Wisor, J.P. et al. Dopaminergic role in stimulant-induced wakefulness. J. Neurosci. 21, 1787–1794 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Qu, W.M., Huang, Z.L., Xu, X.H., Matsumoto, N. & Urade, Y. Dopaminergic D1 and D2 receptors are essential for the arousal effect of modafinil. J. Neurosci. 28, 8462–8469 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Holst, S.C. et al. Dopaminergic role in regulating neurophysiological markers of sleep homeostasis in humans. J. Neurosci. 34, 566–573 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Dzirasa, K. et al. Dopaminergic control of sleep-wake states. J. Neurosci. 26, 10577–10589 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Qu, W.M. et al. Essential role of dopamine D2 receptor in the maintenance of wakefulness, but not in homeostatic regulation of sleep, in mice. J. Neurosci. 30, 4382–4389 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Monti, J.M. & Monti, D. The involvement of dopamine in the modulation of sleep and waking. Sleep Med. Rev. 11, 113–133 (2007).

    Article  PubMed  Google Scholar 

  20. Zhou, Q.Y. & Palmiter, R.D. Dopamine-deficient mice are severely hypoactive, adipsic, and aphagic. Cell 83, 1197–1209 (1995).

    Article  CAS  PubMed  Google Scholar 

  21. Ungerstedt, U. Adipsia and aphagia after 6-hydroxydopamine induced degeneration of the nigro-striatal dopamine system. Acta Physiol. Scand. Suppl. 367, 95–122 (1971).

    Article  CAS  PubMed  Google Scholar 

  22. Léna, 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  PubMed  CAS  Google Scholar 

  23. 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 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Lerner, T.N. et al. Intact-brain analyses reveal distinct information carried by SNc dopamine subcircuits. Cell 162, 635–647 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Alexander, G.M. et al. Remote control of neuronal activity in transgenic mice expressing evolved G protein-coupled receptors. Neuron 63, 27–39 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Hediger, H. Comparative observations on sleep. Proc. R. Soc. Med. 62, 153–156 (1969).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Meddis, R. On the function of sleep. Anim. Behav. 23, 676–691 (1975).

    Article  CAS  PubMed  Google Scholar 

  29. Moruzzi, G. Sleep and instinctive behavior. Arch. Ital. Biol. 107, 175–216 (1969).

    CAS  PubMed  Google Scholar 

  30. Gaskill, B.N. et al. Heat or insulation: behavioral titration of mouse preference for warmth or access to a nest. PLoS One 7, e32799 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Curie, T. et al. Homeostatic and circadian contribution to EEG and molecular state variables of sleep regulation. Sleep 36, 311–323 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Burgess, C.R., Tse, G., Gillis, L. & Peever, J.H. Dopaminergic regulation of sleep and cataplexy in a murine model of narcolepsy. Sleep 33, 1295–1304 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Brown, R.E., Basheer, R., McKenna, J.T., Strecker, R.E. & McCarley, R.W. Control of sleep and wakefulness. Physiol. Rev. 92, 1087–1187 (2012).

    Article  CAS  PubMed  Google Scholar 

  34. Lu, J., Jhou, T.C. & Saper, C.B. Identification of wake-active dopaminergic neurons in the ventral periaqueductal gray matter. J. Neurosci. 26, 193–202 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Hall, Z.J., Healy, S.D. & Meddle, S.L. A role for nonapeptides and dopamine in nest-building behaviour. J. Neuroendocrinol. 27, 158–165 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Lazarus, M., Chen, J.F., Urade, Y. & Huang, Z.L. Role of the basal ganglia in the control of sleep and wakefulness. Curr. Opin. Neurobiol. 23, 780–785 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Qiu, M.H. et al. The role of nucleus accumbens core/shell in sleep-wake regulation and their involvement in modafinil-induced arousal. PLoS One 7, e45471 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Andretic, R., van Swinderen, B. & Greenspan, R.J. Dopaminergic modulation of arousal in Drosophila. Curr. Biol. 15, 1165–1175 (2005).

    Article  CAS  PubMed  Google Scholar 

  39. Kume, K., Kume, S., Park, S.K., Hirsh, J. & Jackson, F.R. Dopamine is a regulator of arousal in the fruit fly. J. Neurosci. 25, 7377–7384 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Pimentel, D. et al. Operation of a homeostatic sleep switch. Nature http://dx.doi.org/10.1038/nature19055 (2016).

  41. Salamone, J.D., Cousins, M.S. & Snyder, B.J. Behavioral functions of nucleus accumbens dopamine: empirical and conceptual problems with the anhedonia hypothesis. Neurosci. Biobehav. Rev. 21, 341–359 (1997).

    Article  CAS  PubMed  Google Scholar 

  42. Zhang, F. et al. Optogenetics in freely moving mammals: dopamine and reward. Cold Spring Harb. Protoc. http://dx.doi.org/10.1101/pdb.top086330 (2015).

  43. Adamantidis, A.R. et al. Optogenetic interrogation of dopaminergic modulation of the multiple phases of reward-seeking behavior. J. Neurosci. 31, 10829–10835 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Tye, K.M. et al. Dopamine neurons modulate neural encoding and expression of depression-related behaviour. Nature 493, 537–541 (2013).

    Article  CAS  PubMed  Google Scholar 

  45. Chaudhury, D. et al. Rapid regulation of depression-related behaviours by control of midbrain dopamine neurons. Nature 493, 532–536 (2013).

    Article  CAS  PubMed  Google Scholar 

  46. Danjo, T., Yoshimi, K., Funabiki, K., Yawata, S. & Nakanishi, S. Aversive behavior induced by optogenetic inactivation of ventral tegmental area dopamine neurons is mediated by dopamine D2 receptors in the nucleus accumbens. Proc. Natl. Acad. Sci. USA 111, 6455–6460 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Cousins, D.A., Butts, K. & Young, A.H. The role of dopamine in bipolar disorder. Bipolar Disord. 11, 787–806 (2009).

    Article  CAS  PubMed  Google Scholar 

  48. Wulff, K., Gatti, S., Wettstein, J.G. & Foster, R.G. Sleep and circadian rhythm disruption in psychiatric and neurodegenerative disease. Nat. Rev. Neurosci. 11, 589–599 (2010).

    Article  CAS  PubMed  Google Scholar 

  49. Parekh, P.K., Ozburn, A.R. & McClung, C.A. Circadian clock genes: effects on dopamine, reward and addiction. Alcohol 49, 341–349 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Winton-Brown, T.T., Fusar-Poli, P., Ungless, M.A. & Howes, O.D. Dopaminergic basis of salience dysregulation in psychosis. Trends Neurosci. 37, 85–94 (2014).

    Article  CAS  PubMed  Google Scholar 

  51. Deacon, R.M. Assessing nest building in mice. Nat. Protoc. 1, 1117–1119 (2006).

    Article  PubMed  Google Scholar 

  52. Palchykova, S., Winsky-Sommerer, R., Meerlo, P., Dürr, R. & Tobler, I. Sleep deprivation impairs object recognition in mice. Neurobiol. Learn. Mem. 85, 263–271 (2006).

    Article  PubMed  Google Scholar 

  53. Anaclet, C. et al. The GABAergic parafacial zone is a medullary slow wave sleep-promoting center. Nat. Neurosci. 17, 1217–1224 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Carter, M.E. et al. Tuning arousal with optogenetic modulation of locus coeruleus neurons. Nat. Neurosci. 13, 1526–1533 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Jego, S. et al. Optogenetic identification of a rapid eye movement sleep modulatory circuit in the hypothalamus. Nat. Neurosci. 16, 1637–1643 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank T. Davidson for assistance in the set-up of the fiber photometry rig, A. Whittle for assistance in core body temperature measurements and for feedback on the manuscript, A. Rolls and P. Bonnavion for early training in sleep–wake recording and viral manipulations and discussion and J. Garner for discussion. This work was supported by National Institutes of Health (NIH) grants RO1-MH087592, RO1-MH102638 and RO1-AG04767 (L.d.L.), a Brain and Behavior Research Foundation (NARSAD) grant (L.d.L.), the US Israel Binational Science Foundation and the Klarman Family Foundation (L.d.L.), Edmond and Lily Safra Center of Brain Science (ELSC) postdoctoral fellowships (A.E.-R.) and NIH F32 AA022832 (W.J.G.). We thank the Stanford Neuroscience Microscopy Service, supported by NIH NS069375.

Author information

Authors and Affiliations

Authors

Contributions

A.E.-R. and L.d.L. conceived and designed the study. A.E.-R. performed all experiments and analyzed data. G.R. wrote the Matlab analysis code and analyzed the fiber photometry data. W.J.G. performed histology, immunostaining and confocal imaging, and W.J.G. and A.E.-R. analyzed the immunohistochemical co-expression data. J.R.J. set up the fiber photometry rig. A.E.-R. wrote the manuscript with feedback from all the authors. L.d.L. supervised the study.

Corresponding author

Correspondence to Luis de Lecea.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Th+ VTA neuron population activity across sleep–wake states.

(a-g) Transient detection. (a) Representative fluorescence trace. (b) Filtered ΔF/F signals. Pink line, signal low-pass filtered at 0 – 4 Hz. Green line, signal low-pass filtered at 0 – 40 Hz. (c) The derivative of the squared difference between the filtered signals. Pink line depicts the thresholding at the mean + 2 s.d. (d) Prospective transients (pink circles). (e) Prospective transients occurring during high ΔF/F periods. Blue line depicts the mean + 2 s.d. threshold. (f,g) Transient detected (red circles). The exact peak time of each transient was identified by taking the maximal value of the thresholded signal within each transient. (h-k) Paired comparison of (h) fluorescence, (i) transient rate, (j) transient amplitude and (k) transient rate * transient amplitude, across the different arousal states. n = 4 animals, 8 trials per animal. Different colors depict different mice. The P values were obtained from paired t-tests. (l) Durations of arousal states following a state transition. Note most of wake episodes following NREM sleep were shorter than 15 s; the animals transitioned back to NREM sleep following these brief arousals. In contrast, NREM sleep and REM sleep episodes tended to be longer. (m-o) We stereotaxically injected the control virus AAV-DJ-EF1α-DIO-GFP into the VTA of a Th-Cre mouse and implanted a fiber optic (400 µm) and EEG/EMG electrodes. (n) Representative fluorescence trace, EEG and EMG trace across spontaneous sleep-wake states. (o,p) Mean (± s.e.m.) (o) fluorescence and (p) calcium transient rate during wake, NREM sleep and REM sleep. n = 5 trials, one-way ANOVA.

Supplementary Figure 2 Spontaneous sleep–wake states of Th-Cre mice transduced with the different viral constructs.

(a,b) The percentage (mean ± s.e.m.) of time spent awake, in NREM sleep and REM sleep during 24 h of baseline sleep recording of Th-Cre mice transduced with mCherry (gray), hM4Di (red), eYFP (green) and ChR2 (blue) virus (n = 4 per group). (a) Summary of the entire 24 h period. No significant differences were found between the different groups (P > 0.24), one-way ANOVA tests. (b) Percentage per hour spent awake (left), in NREM sleep (middle) and REM sleep (right). No significant differences were found between the different groups (P > 0.86), two-way ANOVA between compound injected and time. These findings demonstrate that the viral manipulations themselves did not disrupt the normal sleep-wake cycle.

Supplementary Figure 3 Sleep–wake architecture following chemogenetic inhibition of Th+ VTA neurons.

(a,b) The percentage of time (mean ± s.e.m.) spent awake (left), in NREM sleep (middle) and REM sleep (right) during the 24 h starting with the saline or CNO (1 mg kg-1) administration to (a) mCherry and (b) hM4Di mice. n = 7 per group. * P < 0.05, *** P < 0.001, **** P < 0.0001, two-way RM ANOVA between compound injected and time, followed by Sidak’s post hoc tests. hM4Di: P(interaction) = 0.0008(wake), 0.0007(NREM), 0.058(REM). mCherry: P(interaction) > 0.2 for wake, NREM sleep and REM sleep. (c) Delta power as function of time during NREM sleep episodes following saline (left) and CNO (right) injections in hM4Di mice. n = 6 mice per group; summary of all episodes from all mice. P and R were obtained from a Pearson correlation.

Supplementary Figure 4 EEG power spectrum and arousal states in the presence of salient stimuli.

(a) EEG power spectrum of NREM sleep episodes, in CNO-treated hM4Di mice, during high-fat chow (left), a female mouse (middle), and a component of fox odor (right) presentations. (b) EEG power spectrum of wake episodes during high-fat chow (left), a female mouse (middle), and a component of fox odor (right) presentations. (c) The percentage of time (mean ± s.e.m.) spent awake, in NREM sleep and REM sleep in the presence of high fat chow (left), female mouse (middle) and a component of fox odor (right). We implanted wild-type mice with EEG/EMG electrodes and presented them with salient stimuli (one stimulus at a time, at least 4 days apart) either the light or dark phases of the day. EEG/EMG activity was monitored during 2 h, in the presence of the salient stimuli. *** P < 0.001, **** P < 0.0001, paired t-tests. These findings demonstrate that a naturalistic increase drive for sleep reduces the capacity to maintain arousal in face of some saline stimuli.

Supplementary Figure 5 Chemogenetic inhibition of Th+ VTA neurons.

(a) Representative hypnogram (left) and FFT-derived delta power (right) of a CNO-treated hM4Di mouse during the 1 h test period in a novel environment containing new nesting material. (b) Core body temperature (mean ± s.e.m.) in the course of 4 h following CNO administration in mCherry and hM4Di mice.

Supplementary Figure 6 Optogenetic activation of Th+ VTA cell bodies.

(a) Co-localization of Fos within TH cells. We delivered optical stimuli (25 Hz, during 5 s min-1, for 30 min) to the VTA of eYFP and ChR2 mice at the beginning of the light phase (ZT 2) and sacrificed the mice 90 min later. TH-positive neurons in midline cell groups from hypothalamus to hindbrain were scored for the presence of Fos. **** P < 0.0001, two-way RM ANOVA between distance and viral injection, followed by Bonferroni post hoc tests. The data demonstrate that optical stimulation produced a significant increase in Fos expression in TH+ cells throughout the anterior-posterior length of the VTA, but not in other TH+ neuronal populations. (b) Mean latency (± s.e.m.) to wake from NREM sleep following 5 s stimulation at 25 Hz, 3 days before and following sleep deprivation (SD; 4 h of gentle handling), at the same circadian time, of eYFP (left) and ChR2 (right) mice (n = 6 per group, 1 stim per mouse). P > 0.05, two-tailed paired sample t-tests. (c) EEG power spectrum during NREM sleep from eYFP (left; n = 7) and ChR2 (right; n = 8) mice, 4 s before the onset of stimulation (black) and 4 s into stimulation (gray). Quantification based on the average of 6 stimulations per mouse. (d) EEG power spectrum during wake episodes in eYFP (left) and ChR2 (right) mice during the 6-h stimulation period (ZT 0- ZT 6; light blue), the following 6 h of the light phase (ZT 6 - ZT 12; black), and the subsequent dark phase (ZT 12 – ZT 0; red). Insert represents EEG power spectrum during wake episodes across the 6 h of photostimulation in ChR2 animals. (e) EEG power spectrum of NREM sleep episodes following semi-chronic photostimulation. NREM sleep episodes EEG power spectrum during the 6 h following the termination of photostimulation in eYFP (left) and ChR2 (right) mice. Two-way RM ANOVA between frequency and time, followed by Tukey post-hoc tests. ChR2, P(time) < 0.0001, P(interaction) < 0.0001. eYFP, P(time) > 0.3. The EEG power spectrum during the 1st h following photostimulation differed significantly from the remaining 5 h of the light phase (P < 0.05 in all comparisons; Tukey post-hoc tests). (f) The percentage of time spent awake, in NREM sleep and REM sleep in mice pre-treated (45 min before) with saline / D1,D2 antagonists and undergoing a semi-chronic photostimulation (2 s per min, at 25 Hz, during 2 h; n = 4 per group). ** P < 0.01, **** P < 0.0001, two-way RM ANOVA between compound injected and viral transduction, followed by Sidak’s post hoc tests.

Supplementary Figure 7 Optogenetic interrogation of Th+ VTA efferent projections in modulation of sleep-to-wake transitions and nest-building behavior.

(a-h) Cumulative distribution of sleep-to-wake transitions following optogenetic stimulation at the NAc, mPFC, CeA and DLS at 1 and 25 Hz in eYFP and ChR2 mice during (a-d) NREM sleep and (e-h) REM sleep. Note that 25-Hz stimulation during NREM sleep in the NAc induced an almost immediate sleep-to-wake transition in about 70% of the stimulation trials. (i-l) Nesting behavior following semi-chronic optogenetic stimulation at the (i) NAc, (j) mPFC, (g) CeA and (l) DLS (2 s per min, for 3 h, at 25 Hz).The nesting score (mean ± s.e.m.) represents the amount of nesting material used and shape of the nest after a 3 h period (1, poor; 5, good). n = 4 mice per group. Wilcoxon matched-pairs signed rank test. No significant differences were found between the different groups (P > 0.5).

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–7 (PDF 2099 kb)

Supplementary Methods Checklist (PDF 585 kb)

hM4Di–saline, inaccessible palatable food.

hM4Di mouse injected with saline 45 min prior to the placement of inaccessible high fat palatable food in his home cage. (MP4 13207 kb)

hM4Di–CNO, inaccessible palatable food.

hM4Di mouse injected with CNO 45 min prior to the placement of inaccessible high fat palatable food in his home cage. (MP4 10223 kb)

hM4Di–saline, accessible female mouse.

hM4Di mouse injected with saline 45 min prior to the placement of an adult female mouse in his home cage. (MP4 11650 kb)

hM4Di–CNO, accessible female mouse.

hM4Di mouse injected with CNO 45 min prior to the placement of an adult female mouse in his home cage. (MP4 10973 kb)

hM4Di–saline, inaccessible female mouse.

hM4Di mouse injected with saline 45 min prior to the placement of an adult female mouse in his home cage. The female was restricted to a mesh cage. (MP4 18141 kb)

hM4Di–CNO, inaccessible female mouse.

hM4Di mouse injected with CNO 45 min prior to the placement of an adult female mouse in his home cage. The female was restricted to a mesh cage. (MP4 14454 kb)

hM4Di–CNO, new cage.

hM4Di mouse injected with CNO 45 min prior to his placement in a new cage containing ad-libitum food and water and a 3 g Nestlet. (MP4 18073 kb)

hM4Di–saline, new cage.

hM4Di mouse injected with saline 45 min prior to his placement in a new cage containing ad-libitum food and water and a 3 g Nestlet. (MP4 22172 kb)

hM4Di–saline, new cage + old nest.

hM4Di mouse injected with saline 45 min prior to his placement in a new cage together with his old nest, containing new nesting material. (MP4 9524 kb)

hM4Di–CNO, new cage + old nest.

hM4Di mouse injected with CNO 45 min prior to his placement in a new cage together with his old nest, containing new nesting material. (MP4 7370 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Eban-Rothschild, A., Rothschild, G., Giardino, W. et al. VTA dopaminergic neurons regulate ethologically relevant sleep–wake behaviors. Nat Neurosci 19, 1356–1366 (2016). https://doi.org/10.1038/nn.4377

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nn.4377

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