Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2019 Sep 25;103(6):1044-1055.e7.
doi: 10.1016/j.neuron.2019.07.026. Epub 2019 Aug 28.

A Rare Mutation of β1-Adrenergic Receptor Affects Sleep/Wake Behaviors

Affiliations

A Rare Mutation of β1-Adrenergic Receptor Affects Sleep/Wake Behaviors

Guangsen Shi et al. Neuron. .

Abstract

Sleep is crucial for our survival, and many diseases are linked to long-term poor sleep quality. Before we can use sleep to enhance our health and performance and alleviate diseases associated with poor sleep, a greater understanding of sleep regulation is necessary. We have identified a mutation in the β1-adrenergic receptor gene in humans who require fewer hours of sleep than most. In vitro, this mutation leads to decreased protein stability and dampened signaling in response to agonist treatment. In vivo, the mice carrying the same mutation demonstrated short sleep behavior. We found that this receptor is highly expressed in the dorsal pons and that these ADRB1+ neurons are active during rapid eye movement (REM) sleep and wakefulness. Activating these neurons can lead to wakefulness, and the activity of these neurons is affected by the mutation. These results highlight the important role of β1-adrenergic receptors in sleep/wake regulation.

Keywords: ADRB1; dorsal pons; familial natural short sleep; sleep duration.

PubMed Disclaimer

Conflict of interest statement

DECLARATION OF INTERESTS

The authors declare no competing interests.

Figures

Figure 1.
Figure 1.. An ADRB1 mutation was identified in a natural short sleep family
(A) Pedigree of a family (K50025) carrying the ADRB1 mutation (A187V). See Table S1 for detailed sleep schedules. (B) The β1AR A187V mutation is located in transmembrane domain 4 and is highly conserved among invertebrate and mammalian species.
Figure 2.
Figure 2.. The β1AR A187V mutation alters protein stability and cAMP production
(A) Degradation assay of β1AR in transfected HEK293 cells. Twenty-four hours after transfection, cells were treated with 100 μg/ml CHX and harvested at indicated time points. Bands inside the two red boxes indicate β1AR protein of different sizes in the SDS gel. Quantified results are shown on the right. β1AR protein levels at the starting point (t=0 hours) were normalized to 1. (B) The β1AR A187V mutation confers altered downstream signaling output in response to isoproterenol in cultured cells. Heterozygous expression of β1AR A187V and WT leads to a reduction in cAMP production. (C and D) Western blotting results of endogenous β1AR protein from the heart (C) and brain (D) lysates of Adrb1+/+, and Adrb1+/m animals. N=4 mice per group. NS, non-specific band. Quantified results are shown on the right. (E and F) q-RTPCR results of Adrb1 mRNA normalized to Actin mRNA from the heart (E) and brain (F) tissues of Adrb1+/+ and +/m animals. N=4 mice per group. * P<0.05, **P<0.01, n.s.= not significant. Two way RM ANOVA, post-hoc Sidak’s multiple comparisons test for (A) and (B). Two-tailed Student’s t-test for (C)-(F). Error bars represent ± SEM.
Figure 3.
Figure 3.. The Adrb1-A187V mutation alters sleep/wake related behaviors in the FNSS mouse model
(A-C) Total mobile time by ANY-maze within 24 hours (A), dark phase (B) and light phase (C) were calculated in Adrb1 +/+ (N=20) and +/m (N=19) mice. (D-F) Total sleep time by EEG/EMG within 24 hours (D), dark phase (E) and light phase (F) were calculated in Adrb1 +/+ (N=9) and +/m (N=13) mice. (G-I) Total NREM sleep time within 24 hours (G), dark phase (H) and light phase (I) were calculated in Adrb1 +/+ (N=9) and +/m (N=13) mice. (J-L) Total REM sleep time during a 24 hour day (J), dark phase (K) and light phase (L) were calculated in Adrb1 +/+ (N=9) and +/m (N=13) mice. (M) Time course of EEG delta power (1.0–4.0 Hz) during NREM sleep across the light phase for Adrb1 +/+ (N=9) and +/m (N=9) mice. * P<0.05, **P<0.01, *** P<0.001, **** P<0.0001, n.s.=not significant. Two-tailed Student’s t-test for (A)-(L). Two way RM ANOVA, post-hoc Sidak’s multiple comparisons test for (M). Error bars represent ± SEM. See Figures S1 for NREM and REM sleep bouts and duration calculation.
Figure 4.
Figure 4.. Population activity of ADRB1+ neurons in DP is correlated with sleep/wake states
(A) Tg (ADRB1-Cre) mice generated using BAC technology. (B and C) Representative brain sections from ADRB1-Cre;ROSAmT/mG (B) and ADRB1-Cre;loxP-flanked-ChR2–eYFP (C) mice show high CRE activity in the DP area. Scale bar, 300 μm. See also Figures S2, S3 and S4 for further characterization. (D) Representative EEG power spectrogram, EEG, EMG and fluorescence trace across spontaneous sleep–wake states. See Figures S5A–S5C for the fiber photometry setup and Figure S5D for fluorescence during state transitions. (E) Quantified fluorescence signal across different sleep–wake states from N=6 mice. **P<0.01, two-tailed paired Student’s t-test.
Figure 5.
Figure 5.. ADRB1+ neurons in DP are wake-promoting
(A) Schematic of simultaneous optogenetic stimulation/EEG /EMG set up for recording of the sleep/wake states with stimulation of DP ADRB1+ neurons. (B) Representative slice showing viral expression and the placement of the fiber tip above the DP. Scale bar, 300μm. (C and D) Representative EEG/EMG recordings of optogenetic trials (red box, 10 seconds) initiating from NREM, REM and wake states for ChR2-eYFP(C) and eYFP (D) mice. EEG power spectrograms are shown in the upper right. Red scales and boxes indicate 10 seconds light stimulation. (E-G) Quantified results of NREM (E), REM (F) and wake (G) states for (C and D) from ChR2-eYFP (N=5) and eYFP (N=5) mice. *P<0.05, **P<0.01, ***P<0.01, **** P<0.0001. Two way RM ANOVA, post-hoc Sidak’s multiple comparisons test for (E-G). Error bars represent ± SEM.
Figure 6.
Figure 6.. Adrb1-A187V mutation alters DP neural activity
(A) Representative photometry fluorescence traces at ZT1–4 and ZT13–16 from ADRB1-Cre; Adrb1+/+ (N=8) and ADRB1-Cre; Adrb1+/m (N=7) mice. (B) Quantified ratio for (A). (C) Schematic of calcium imaging set up for recording the activity of DP ADRB1+ neurons in brain slices (upper panel). One example of calcium imaging of DP in the slice (lower panel). (D) Percentage of ADRB1+cells that respond differentially to dobutamine treatment in both Adrb1+/+ (N=7) and Adrb1+/m (N=5) brain slices. The bottom table shows the original cell numbers in different categories. See Figure S6 for the representative fluorescence traces of the cells in different categories. **P<0.01, *** P<0.001. Two-tailed Student’s t-test for (B). Chi-square test for (D). Error bars represent ± SEM.
Figure 7.
Figure 7.. The Adrb1-A187V mutation changes the electrophysiological properties of DP ADRB1+ neurons
(A) Representative trace of a single action potential recorded from a DP Adrb1 +/+ neuron in response to a 250ms, 20pA (1X) pulse from the resting membrane potential (RMP). The same neuron fired three action potentials in response to a 40pA (2X) pulse (inset). (B) Representative trace of two action potentials recorded from a DP Adrb1 +/m neuron in response to a 10 pA (1X) pulse from RMP. A 20pA (2X) pulse in the same neuron produced four action potentials (inset). (C and D) Rheobase currents (C) and action potential frequency (D) were calculated for Adrb1 +/+ (n=6) and +/m (n=6) neurons. (E and F) Representative traces of spontaneous EPSCs recorded under voltage clamp conditions (at −60 mV) with different treatments for Adrb1 +/+ (E) and Adrb1 +/m (F) neurons. (G and H) Frequency (G) and amplitude (H) before and after dobutamine (10μM) treatment were calculated for Adrb1 +/+ (n=6) and +/m (n=7) neurons. (I) Representative traces of mEPSCs recorded in one Adrb1 +/m neuron with different treatments. (J) mEPSCs frequency were calculated for Adrb1 +/m (n=6) neurons before and after dobutamine (10μM) treatment in the presence of TTX. **P<0.01, *** P<0.001, n.s. =not significant. Two-tailed Student’s t-test for (C) and (D). Two way RM ANOVA, post-hoc Sidak’s (genotype) and Tukey’s (dobutamine treatment) multiple comparisons test for (G) and (H). One way RM ANOVA, post-hoc Tukey’s multiple comparisons test for (J). Error bars represent ± SEM.

Similar articles

Cited by

References

    1. Anaclet C, Ferrari L, Arrigoni E, Bass CE, Saper CB, Lu J, and Fuller PM (2014). The GABAergic parafacial zone is a medullary slow wave sleep-promoting center. Nat. Neurosci. 17, 1217–1224. - PMC - PubMed
    1. Ardelt AA, Bhattacharyya BJ, Belmadani A, Ren D, and Miller RJ (2013). Stromal derived growth factor-1 (CXCL12) modulates synaptic transmission to immature neurons during post-ischemic cerebral repair. Exp. Neurol - PMC - PubMed
    1. Artiushin G, and Sehgal A (2017). The Drosophila circuitry of sleep–wake regulation. Curr. Opin. Neurobiol 44, 243–250. - PMC - PubMed
    1. Berridge CW (2008). Noradrenergic modulation of arousal. Brain Res. Rev 58, 1–17. - PMC - PubMed
    1. Berridge CW, Schmeichel BE, and España RA (2012). Noradrenergic modulation of wakefulness/arousal. Sleep Med. Rev 16, 187–197. - PMC - PubMed

Publication types

Substances