The loss of orexinergic neurons, which release orexins, results in narcolepsy. Orexins participate in the regulation of many physiological functions, and their role as wake-promoting molecules has been widely described. Less is known about the involvement of orexins in body temperature and respiratory regulation. The aim of this study was to investigate if orexin peptides modulate respiratory regulation as a function of ambient temperature (Ta) during different sleep stages. Respiratory phenotype of male orexin knockout (KO-ORX, N=9) and wild-type (WT, N=8) mice was studied at thermoneutrality (Ta=30°C) or during mild cold exposure (Ta=20°C) inside a whole-body plethysmography chamber. The states of wakefulness (W), non-rapid eye movement sleep (NREMS) and rapid eye movement sleep (REMS) were scored non-invasively, using a previously validated technique. In both WT and KO-ORX mice, Ta strongly and significantly affected ventilatory period and minute ventilation values during NREMS and REMS; moreover, the occurrence rate of sleep apneas in NREMS was significantly reduced at Ta=20°C compared with Ta=30°C. Overall, there were no differences in respiratory regulation during sleep between WT and KO-ORX mice, except for sigh occurrence rate, which was significantly increased at Ta=20°C compared with Ta=30°C in WT mice, but not in KO-ORX mice. These results do not support a main role for orexin peptides in the temperature-dependent modulation of respiratory regulation during sleep. However, we showed that the occurrence rate of sleep apneas critically depends on Ta, without any significant effect of orexin peptides.

Orexins (orexin A and B), also known as hypocretins (hypocretin 1 and 2, respectively), are neuropeptides identified in 1998 by two independent research groups (de Lecea et al., 1998; Sakurai et al., 1998). The location of orexin-containing cell bodies is restricted to the lateral hypothalamus, the perifornical area and the dorsomedial hypothalamus. However, orexinergic projections and receptors are widely distributed in the hypothalamus, thalamus, cerebral cortex, circumventricular organs, brainstem and spinal cord. Originally described as involved in sleep/arousal and feeding regulation (de Lecea et al., 1998; Sakurai et al., 1998), orexin peptides show widespread connections with many different brain regions, and can affect multiple physiological functions (Peyron et al., 1998); indeed, their loss promotes narcoleptic phenotypes in both humans (Peyron et al., 2000) and rodents (Chemelli et al., 1999; Hara et al., 2001). Moreover, orexins have also emerged as major regulatory actors of numerous biological processes, and their critical role in orchestrating behavioral and autonomic responses to environmental challenges have been clearly demonstrated (Zoccoli et al., 2011). On these bases, we hypothesized that orexins may play a role in respiratory regulation during sleep, and that this effect may be affected by temperature of the sleep environment. At present, less is known about their involvement in other regulatory systems such as body temperature and respiratory regulation.

List of symbols and abbreviations
     
  • ATX3-ORX

    ataxin3-orexin

  •  
  • KO-ORX

    orexin knockout

  •  
  • NREMS

    non-rapid eye movement sleep

  •  
  • REMS

    rapid eye movement sleep

  •  
  • s.d.1

    short-term breath-to-breath (VP or VT) variability

  •  
  • s.d.2

    long-term breath-to-breath (VP or VT) variability

  •  
  • Ta

    ambient temperature

  •  
  • VE

    minute ventilation

  •  
  • VP

    ventilatory period

  •  
  • VT

    tidal volume

  •  
  • WBP

    whole-body plethysmography

  •  
  • WT

    wild-type

  •  
  • Δsigh

    difference between sigh occurrence rate at Ta=20°C and that at Ta=30°C

Lateral ventricles, cisterna magna or intrathecal microinjections of orexin A increase tidal volume (VT) and phrenic electromyographic burst amplitude. Orexin A exerts a dose-dependent decrease in ventilatory period (VP) when administered in the lateral ventricle (Zhang et al., 2005), but not when administered at the same doses in the cisterna magna (Zhang et al., 2005) or at higher doses in ventrolateral medulla or the spinal cord (Young et al., 2005). In orexin knockout (KO-ORX) mice, which have congenital orexin deficiency, VP, VT and minute ventilation (VE) were reported to not differ significantly from those of wild-type (WT) mice (Nakamura et al., 2007). However, it has been reported that KO-ORX mice show frequent sleep apneas, suggesting a critical role of orexins in respiratory regulation during sleep (Nakamura et al., 2007). Central and obstructive sleep apnea have been found to be associated with narcolepsy type 1 in adult patients (Chokroverty, 1986; Hoshino et al., 2019; Pataka et al., 2012; Pizza et al., 2013; Sansa et al., 2010), but these data have not been confirmed in pediatric patients with narcolepsy type 1 (Filardi et al., 2020). To date, contrasting data have also been reported on the role of orexins in body temperature regulation. Some studies indicated a possible involvement of orexins in thermogenesis (Kuwaki, 2015; Madden et al., 2012; Mohammed et al., 2016; Nakamura et al., 2003; Tupone et al., 2011), while other experiments suggested that orexins are not necessary to modulate the effects of ambient temperature (Ta) on the wake–sleep cycle or cardiovascular regulation (Lo Martire et al., 2012). In this respect, it should be noted that respiratory regulation and thermoregulation are intertwined. In particular, respiratory variables change as a function of Ta (Gordon, 1985). This modulation was verified in WT mice, in which reduced Ta was found to entail a decrease in VP values, and an increase in VT values. However, these changes were described without taking into account the wake–sleep state (Hodges et al., 2008). WT mice exposed to cold Ta exhibit an increase in thermoregulatory energy metabolic rate during wakefulness, which is an adaptive strategy (Hodges and Richerson, 2008; Hodges et al., 2008).

This study was designed primarily to investigate if orexin peptides modulate respiratory regulation as a function of Ta in different sleep stages. We addressed this question measuring VT, VP and VE in KO-ORX mice exposed to mild cold stress (Ta=20°C) compared with thermoneutrality (Ta=30°C). The secondary aim of the study was to verify if the described increase in sleep apnea occurrence rate in KO-ORX is modulated by exposure to different Ta (20 versus 30°C). Based on the available evidence, we hypothesized that in the absence of orexin peptides, the effects of Ta on breathing variables would be blunted, and that the occurrence rate of sleep apneas would be increased.

The study protocol was approved by the Bologna University ethics committee on animal experimentation and complied with the National Institutes of Health guide for the care and use of laboratory animals. All the experiments included in this study were executed non-invasively. Experiments were performed on eight male C57Bl/6J WT mice and nine congenic (≥10 generations of backcrossing) male KO-ORX mice matched for age (45.3±0.4 and 44.9±0.6 weeks, respectively). The mice were maintained at 23°C with a 12 h:12 h light:dark cycle and free access to food and water. Genotypes were assessed as previously described (Bastianini et al., 2011). The experimental protocol consisted of two recordings performed with the mouse placed inside a whole-body plethysmography (WBP) chamber (PLY4223; Buxco, Wilmington, NC, USA) flushed with air at 1.5 litres h−1 and exposed either to mild cold stress (Ta=20°C) or to thermoneutrality (Ta=30°C). Each animal underwent two recording sessions, one at Ta=20°C and the other at Ta=30°C. The order of the recording sessions was randomly chosen and balanced between experimental groups, leaving a 2–28 day interval between sessions. The groups were also matched for the interval between sessions (8.7±2.6 versus 10.4±3.2 days in KO-ORX versus WT mice, t15=−0.412, P=0.686). Each recording session lasted for 8 h during the rest (light) period, starting at lights-on at 09:00 h. The respiratory signal was derived from the differential pressure between the mouse chamber and a second reference chamber, measured with a high-precision differential pressure transducer (DP103-06+CD223 digital transducer indicator; Validyne Engineering, Northridge, CA, USA). Differential pressure and chamber humidity and temperature were continuously recorded, digitized and stored at 128, 4 and 4 Hz, respectively. The system was calibrated with a 100 μl microsyringe (Hamilton, Reno, NV, USA) at the end of each recording. Wakefulness, non-rapid eye movement sleep (NREMS) and rapid eye movement sleep (REMS) were scored based on inspection of the raw respiratory recordings, with a procedure validated against gold standard conventional electroencephalography and electromyography (Bastianini et al., 2017). As in previous studies with this technique (Bastianini et al., 2015; Silvani et al., 2014), quantitative analysis of breathing was restricted to stable sleep episodes ≥12 s because of the frequent occurrence of movement artefacts during wakefulness. Breath-to-breath values of VP, VT and VE were obtained as previously reported for each sleep state (Bastianini et al., 2017). The variability of VP and VT was analysed with a technique originally proposed for the study of heart rate variability (Brennan et al., 2001) and already applied to respiratory physiology (Bastianini et al., 2015; Silvani et al., 2014). Briefly, the short-term (breath-to-breath) and long-term variability of VP and VT were calculated based on Poincaré plots, in which the abscissa and ordinate of each point indicate the duration or amplitude of the Nth and (N+1)th successive breaths, respectively. In this analysis, the standard deviation of VP and VT values around the axis oriented with the line of identity of the Poincaré plot estimates the short-term (breath-to-breath) variability of VP and VT (s.d.1), while the standard deviation of VP and VT values around the orthogonal axis estimates long-term variability (s.d.2). The mean values of VP, VT, VE and the s.d.1 and s.d.2 of VP and VT were computed for each mouse after exclusion of the breaths with VP and/or VT that deviated more than 3 s.d. from the respective mean value in the whole recording (Bastianini et al., 2015). These computations were thus protected from the effects of breaths with extreme values of VP and/or VT. Finally, apneas and augmented breaths (sighs) were automatically detected as breaths with values of VP (apneas) or VT (sighs) more than three times the average values of VP or VT, respectively, for each mouse and sleep state, and detection accuracy was checked on raw recordings (Bastianini et al., 2015; Silvani et al., 2014). Because augmented breaths (sighs) often precede apneas during NREMS, we further categorized NREMS apneas as post-sigh apneas if they followed a sigh by ≤8 s or as spontaneous apneas if they followed a sigh by >8 s (Bastianini et al., 2019).

At the end of the experimental protocol, mice were perfused, under deep anesthesia (isoflurane 4%, inhalation route), with saline followed by 4% paraformaldehyde. Brains were cryoprotected in phosphate-buffered saline (PBS) with 20% sucrose and coronally sectioned at 30 µm using a cryostat microtome at −22.0°C. Hypothalamic sections were processed for immunoreactivity for orexin A. Briefly, free-floating sections were washed in 0.3% Triton X-100 in PBS for 30 min. After blocking for 90 min with 3% bovine serum albumin (Sigma-Aldrich, Milan, Italy) in 0.3% Triton X-100 in PBS, sections were incubated overnight at 4°C with rabbit anti-orexin A antiserum (Phoenix Pharmaceuticals, Burlingame, CA, USA) diluted 1:5000 in 0.3% Triton X-100 in PBS and 1% bovine serum albumin (BSA). Sections were then washed in 0.3% Triton X-100 in PBS for 30 min and incubated for 2 h with a Cy3-conjugated AffiniPure Donkey anti-rabbit IgG (Jackson ImmunoResearch, West Grove, PA, USA) secondary fluorescent antibody diluted 1:200 in 0.3% Triton X-100 in PBS and 1% BSA. Sections were counterstained with Hoechst 33342 (Sigma-Aldrich) in order to label cell nuclei. Immunofluorescence images were taken with a Nikon Eclipse TE 2000-S inverted microscope (Nikon Corporation, Kawasaki, Japan) equipped with a Nikon DS-Qi2 digital camera. The same lot of primary antiserum was used for staining the brains of WT and KO-ORX mice. Control and experimental animals were processed simultaneously for staining to avoid any batch-to-batch variation.

Statistical analysis was performed using SPSS software (SPSS Inc., Chicago, IL, USA) with two- or three-way mixed design ANOVAs. In all analyses, genotype was the between-subject factor (two levels: KO-ORX versus WT), and Ta was a within-subject (i.e. repeated measure) factor (two levels: 20 versus 30°C). Most analyses included the sleep state as another within-subject factor (two levels: NREMS versus REMS) as appropriate. In order to limit the complexity of the interpretation of statistical analysis, for the special case of the analysis of breathing variability we replaced the sleep state factor with a variability index factor (two levels: s.d.1 versus s.d.2), and ran two separate ANOVAs for NREMS and REMS. An independent t-test was then applied to compare the differences between the two experimental groups, whereas a dependent t-test was used to compare the effect of different conditions (i.e. sleep states or Ta) on a variable that was not affected by mouse genotype (thus we considered both experimental groups as a whole). Results are shown means±s.e.m. with significance at P<0.05.

First, we verified the validity of the KO-ORX mice used in this study as a model of orexin deficiency and of body weight and wake–sleep cycle alterations related to narcolepsy type 1. The orexin A antiserum produced no staining in the brains of KO-ORX mice, whereas it clearly labelled orexin A neurons in WT brains (Fig. 1).

The body weight of KO-ORX mice before each recording was significantly higher than that of WT mice (34.2±1.2 versus 30.0±0.7 g, t12.3=−2.962, P=0.012).

The percentage of recording time spent in wakefulness, NREMS or REMS did not differ significantly between KO-ORX and WT mice (two-way ANOVA: for wakefulness F1,15=0.109, P=0.746; for NREMS F1,15=0.226, P=0.641; for REMS F1,15=0.353, P=0.561), nor was it significantly affected by Ta (for wakefulness F1,15=0.484, P=0.497; for NREMS F1,15=0.595, P=0.452; for REMS F1,15=0.001, P=0.977) (Table 1). However, two-way ANOVA revealed significant main effects of orexin deficiency (i.e. KO-ORX versus WT), with no significant interaction between Ta and orexin deficiency, on specific features of sleep architecture. In particular, KO-ORX mice showed significant fragmentation of wakefulness (i.e. reduced episode duration) (F1,15=7.404, P=0.016) and NREMS (F1,15=5.945, P=0.028) episodes, and shorter REMS latency (F1,15=19.313, P=0.001) compared with WT mice, irrespective of Ta (for fragmentation of wakefulness episodes F1,15<0.001, P=0.991; for fragmentation of NREMS episodes F1,15=0.126, P=0.728; for REMS latency F1,15=4.534, P=0.0502). Accordingly, KO-ORX and WT mice constituted distinct clusters on a scatterplot of reduced REMS latency and reduced wakefulness episode mean duration, particularly at Ta=20°C (Fig. 2). Overall, these results are fully consistent with previous work on KO-ORX mice (Bastianini et al., 2011; Chemelli et al., 1999), confirming the validity of the KO-ORX mice used in this study as a model of orexin deficiency and narcolepsy type 1.

The analysis of respiratory variables demonstrated that in both KO-ORX and WT mice, Ta strongly and significantly affected VP and VE values (three-way ANOVA, main effect: for VP F1,15=97.273, P<0.001; for VEF1,15=17.515, P=0.001) (Fig. 3), but did not have any significant effect on VT (ANOVA, main effect: F1,15=1.133, P=0.304). In particular, the values of VP were significantly lower and those of VE were significantly higher at Ta=20°C than at Ta=30°C, irrespective of the sleep state and orexin deficiency (temperature×sleep state interaction on VP: F1,15=2.285, P=0.151; temperature×orexin deficiency interaction on VP: F1,15=0.433, P=0.520; temperature×sleep state interaction on VE: F1,15=3.668, P=0.075; temperature×orexin deficiency interaction on VE: F1,15=0.001, P=0.980).

Three-way ANOVA did not reveal any significant main effect of orexin deficiency on sleep apnea occurrence rate (F1,15=1.934, P=0.185), but revealed a significant interaction between Ta and sleep state (F1,15=5.039, P=0.040) on this variable. Dependent t-test considering both experimental groups together confirmed that sleep apnea occurrence rate increased with Ta during NREMS (t16=−4.601, P<0.001) but not during REMS (t16=0.351, P=0.730) (Fig. 4). Moreover, categorization of NREMS apneas as post-sigh or spontaneous as a function of their proximity to a preceding sigh (Table 2) indicated that the occurrence rate of both post-sigh and spontaneous sleep apneas significantly increased with Ta (dependent t-test: t16=−5.573, P<0.001 and t16=−2.348, P=0.032, respectively).

As sighs occur almost exclusively during NREMS in mice (Bastianini et al., 2019), the analysis of their occurrence rate was restricted to NREMS. We found a significant interaction between orexin deficiency and Ta on sigh occurrence rate (two-way ANOVA, F1,15=6.297, P=0.024). Sighs were significantly increased at Ta=20°C compared with Ta=30°C in WT mice (12.7±2.3 versus 8.2±1.8 episodes h−1, respectively; paired t-test, t7=4.111, P=0.005), but not in KO-ORX mice (7.9±1.2 versus 7.2±0.8 episodes h−1, respectively; paired t-test, t8=0.642, P=0.539). In order to better describe this interaction effect, for each mouse we calculated the difference between sigh occurrence rate at Ta=20°C and that at Ta=30°C (Δsigh). Δsigh was positive, and was significantly lower in KO-ORX compared with WT mice (independent t-test, t15=2.509, P=0.024) (Fig. 4).

Sleep apneas and sighs are extreme manifestations of VP and VT variability, respectively. We went on to analyse the overall variability of VP and VT with a method protected from the effects of extreme values of breathing variables (see Materials and Methods). Three-way ANOVA showed a significant main effect of Ta on VP variability (F1,15=110.909, P<0.001), whereas it did not highlight any significant effect of orexin deficiency (F1,15=0.002, P=0.968). In particular, the short-term (breath-to-breath, s.d.1) and the long-term (s.d.2) variability of VP were significantly lower at Ta=20°C than at Ta=30°C both in NREMS (dependent t-test, considering both experimental groups together, t16=−7.185, P<0.001 for s.d.1 and t16=−10.501, P<0.001 for s.d.2) and in REMS (dependent t-test, considering both experimental groups together, t16=−8.546, P<0.001 for s.d.1 and t16=−9.201, P<0.001 for s.d.2) (Fig. 5). Three-way ANOVA of VT variability failed to show significant main effects of Ta (F1,15=3.408, P=0.085) and orexin deficiency (F1,15=0.117, P=0.738), whereas it highlighted a significant Ta×VT variability interaction (F1,15=11.079, P=0.005). In particular, the long-term variability (s.d.2) of VT was significantly lower at Ta=20°C than at Ta=30°C in NREMS (dependent t-test, t16=3.602, P=0.003) (Fig. 6).

We investigated if orexin peptides modulate respiratory regulation as a function of Ta. Our findings do not support a significant role for orexin peptides in the temperature-dependent modulation of breathing in different sleep states with one notable exception. Δsigh during NREMS was significantly lower in ORX-KO mice than in WT mice (Fig. 4). The mechanisms underlying apneas and sighs are still incompletely understood. Sighs may be the results of an inspiratory-augmenting reflex elicited by activation of lung and chest wall receptors in response to reduced lung compliance, or by stimulation of peripheral chemoreceptors in response to hypoxia or hypercapnia (Nakamura et al., 2003; Qureshi et al., 2009). There is evidence that orexins contribute to central chemoreflex sensitivity to carbon dioxide levels, but this contribution appears to be restricted to wakefulness (Nakamura et al., 2007), particularly in the daily active period, which corresponds to the dark period in rats and mice (Li and Nattie, 2010). This effect is thus unlikely to explain our findings concerning sigh occurrence rate during sleep in the light (rest) period. This raises the hypothesis that orexins are also involved in the integration of afferent signals from chest wall receptors.

However, these experiments showed for the first time that the occurrence rate of sleep apneas critically depends on Ta, being exacerbated at thermoneutrality (Ta=30°C) and reduced by mild cold exposure (Ta=20°C), without any significant effect of orexin peptides.

The conclusion that effects of Ta on breathing do not significantly depend on orexin peptides is in line with previous results showing that in orexin-ataxin3 transgenic mice with genetic ablation of orexin neurons (ATX3-ORX) the effects of Ta on cardiovascular regulation during different sleep states was preserved compared with WT controls (Lo Martire et al., 2012). Nevertheless, more experiments are needed to understand whether different co-transmitters, released by orexin neurons together with orexin peptides (Bonnavion et al., 2016), play a role in the adaptive breathing responses to different values of Ta. Ablation of orexin neurons in ATX3-ORX mice or rats resulted in intolerance to cold exposure (Ta=5°C) (Mohammed et al., 2016; Takahashi et al., 2013), suggesting the importance of these neurons in driving the cold defence responses. In particular, in ATX3-ORX mice but not in ORX-KO mice, abdominal temperature fell rapidly and reached the endpoint of 30°C within 50–150 min of cold exposure (Takahashi et al., 2013). These data suggest that orexin co-transmitters, rather than orexin peptides, participate in the homeostatic response to cold exposure.

In our experiments, Ta did not modulate the percentage of recording time spent in the different states of the wake–sleep cycle (Table 1). This result is at variance with our previous results on ATX3-ORX and WT mice (Lo Martire et al., 2012). However, some differences between these experiments must be acknowledged. First, in this study we recorded non-instrumented mice with a pure C57BL/6J background, whereas we obtained our previous data (Lo Martire et al., 2012) on chronically instrumented mice with a hybrid genetic background (75% C57BL/6J and 25% DBA/2J). Moreover, during this study we recorded mice inside a WBP chamber for only 8 h during the light (resting) period, and in the previous study we recorded freely moving mice for 48 h at each of two values of Ta (Lo Martire et al., 2012). Thirdly, in this study we changed the value of Ta acutely at the beginning of each recording session inside a WBP chamber, whereas in the previous study the values of Ta were changed 24 h before each recording session in the home cage (Lo Martire et al., 2012).

We found that when KO-ORX and WT mice were exposed to mild cold stress (Ta=20°C), they coped with this challenge by increasing breathing rate and VE relative to the exposure to Ta=30°C (Fig. 3). This strategy has already been described in WT mice (Hodges and Richerson, 2008; Hodges et al., 2008), but only during wakefulness, with no available data during sleep. Moreover, no data on breathing adaptive response to Ta changes have been reported so far in narcoleptic KO-ORX mice lacking orexin peptides.

In our experiments, KO-ORX mice did not exhibit a higher sleep apnea occurrence rate compared with WT controls (Fig. 4; Table 2). Thus, our data challenge the view that the loss of orexin peptides is sufficient to increase sleep apnea rate in mice (Nakamura et al., 2007), but they are in broad agreement with recent findings on pediatric children with narcolepsy type 1 (Filardi et al., 2020). Stress related to chronic instrumentation in previous reports (Nakamura et al., 2007), compared with our own experiments using intact mice, might help to explain mouse differences, as the occurrence rate of sleep apneas increases with corticosterone levels (Ren et al., 2012).

A new and interesting finding of our study is that NREMS apneas were less frequent in conditions of mild cold exposure (Ta=20°C) than at thermoneutrality (Ta=30°C) (Fig. 4). This was an unexpected but robust finding as it was observed both in KO-ORX and WT mice and it concerned both post-sigh and spontaneous sleep apneas (Table 2). It is worth noting that this phenomenon is related to a specific interaction between thermoregulatory responses and NREMS state. Indeed, while we observed similar Ta-dependent increases in the mean value and in the variability of VP during both NREMS and REMS (Figs 3 and 5), the Ta-dependent increase in sleep apnea occurrence rate was restricted to NREMS (Fig. 4; Table 2). This finding is also of practical interest, as it highlights the critical importance of controlling for Ta when phenotyping mice for sleep apneas, similar to what has long been recommended when phenotyping mice for cardiovascular variables (Swoap et al., 2004).

Congenital loss of orexin peptides in KO-ORX mice may lead to developmental compensation. However, at least the behavioural phenotype (Chemelli et al., 1999) and the sleep-related blood pressure control (Bastianini et al., 2011) of KO-ORX mice strikingly resemble those of patients with narcolepsy type 1, who have a near-complete loss of orexin neurons (Berteotti and Silvani, 2018). Our conclusion that orexin peptides are not necessary for most aspects of breathing control as a function of Ta, with the notable exception of sigh occurrence rate, would therefore not be trivial if confirmed. However, the physiological effects of orexin peptides result from binding to two orexin receptors. Importantly, orexin peptides may exert different effects depending on the orexin receptor type (cf. Kakizaki et al., 2019; Willie et al., 2003). Thus, our conclusion is compatible with contrasting effects of activation of the two orexin receptor types on breathing as a function of Ta, and does not imply that orexins have no effect at all on breathing. To make that implication, it would be necessary to perform pharmacological studies targeting orexin action on each of the two orexin receptors.

Our study has a few limitations. We studied male mice to facilitate comparison with previous work on breathing in KO-ORX mice, which also was performed on male mice only (Nakamura et al., 2003, 2007). Nevertheless, the fact that we did not include female mice in the study is a limitation of our work. We have not measured the thermoneutral zone of the particular mice under study, and we are not aware of previous data published particularly on KO-ORX mice. For reference, the thermoneutral zone for C57BL/6J mice at approximatively 3 months of age lies between 29 and 31°C (Ganeshan and Chawla, 2017). Our sample size (N=8/9 per group) compares favourably with that of previous work on sleep apneas in KO-ORX mice (Nakamura et al., 2007; N=5 per group). Nevertheless, we did not perform a statistical power analysis to estimate our type II statistical error rate.

In conclusion, our findings do not support a role for orexin peptides in the temperature-dependent modulation of respiratory regulation in different sleep states. Orexin peptides only modulated the sigh occurrence rate during NREMS. However, the occurrence of sleep apneas critically depends on Ta, being exacerbated at thermoneutrality and reduced by mild cold exposure, without any significant effect of the lack of orexin peptides.

We thank the Centre for Applied Biomedical Research – CRBA, Sant’Orsola University Hospital, Bologna, Italy, for support in mouse genotyping.

Author contributions

Conceptualization: A.S., G.Z.; Methodology: A.S., G.Z.; Formal analysis: C.B., V.L.M., S.A., S.B., G.M.; Investigation: C.B., V.L.M., S.A., S.B., G.M.; Writing - original draft: C.B., V.L.M.; Writing - review & editing: C.B., V.L.M., S.A., S.B., A.S., G.Z.; Visualization: C.B., V.L.M.; Supervision: G.Z.; Funding acquisition: C.B., A.S., G.Z.

Funding

This work was supported by the University of Bologna (RFO funds to C.B., A.S. and G.Z.).

Bastianini
,
S.
,
Alvente
,
S.
,
Berteotti
,
C.
,
Bosi
,
M.
,
Lo Martire
,
V.
,
Silvani
,
A.
,
Valli
,
A.
and
Zoccoli
,
G.
(
2019
).
Post-sigh sleep apneas in mice: systematic review and data-driven definition
.
J. Sleep Res.
28
,
e12845
.
Bastianini
,
S.
,
Alvente
,
S.
,
Berteotti
,
C.
,
Lo Martire
,
V.
,
Silvani
,
A.
,
Swoap
,
S. J.
,
Valli
,
A.
,
Zoccoli
,
G.
and
Cohen
,
G.
(
2017
).
Accurate discrimination of the wake-sleep states of mice using non-invasive whole-body plethysmography
.
Sci. Rep.
7
,
41698
.
Bastianini
,
S.
,
Silvani
,
A.
,
Berteotti
,
C.
,
Elghozi
,
J.-L.
,
Franzini
,
C.
,
Lenzi
,
P.
,
Lo Martire
,
V. L.
and
Zoccoli
,
G.
(
2011
).
Sleep related changes in blood pressure in hypocretin-deficient narcoleptic mice
.
Sleep
34
,
213
-
218
.
Bastianini
,
S.
,
Silvani
,
A.
,
Berteotti
,
C.
,
Lo Martire
,
V.
,
Cohen
,
G.
,
Ohtsu
,
H.
,
Lin
,
J.-S.
and
Zoccoli
,
G.
(
2015
).
Histamine transmission modulates the phenotype of murine narcolepsy caused by orexin neuron deficiency
.
PLoS ONE
10
,
e0140520
.
Berteotti
,
C.
and
Silvani
,
A.
(
2018
).
The link between narcolepsy and autonomic cardiovascular dysfunction: a translational perspective
.
Clin. Auton. Res.
28
,
545
-
555
.
Bonnavion
,
P.
,
Mickelsen
,
L. E.
,
Fujita
,
A.
,
de Lecea
,
L.
and
Jackson
,
A. C.
(
2016
).
Hubs and spokes of the lateral hypothalamus: cell types, circuits and behaviour
.
J. Physiol.
594
,
6443
-
6462
.
Brennan
,
M.
,
Palaniswami
,
M.
and
Kamen
,
P.
(
2001
).
Do existing measures of Poincare plot geometry reflect nonlinear features of heart rate variability?
IEEE Trans. Biomed. Eng.
48
,
1342
-
1347
.
Chemelli
,
R. M.
,
Willie
,
J. T.
,
Sinton
,
C. M.
,
Elmquist
,
J. K.
,
Scammell
,
T.
,
Lee
,
C.
,
Richardson
,
J. A.
,
Williams
,
S. C.
,
Xiong
,
Y.
,
Kisanuki
,
Y.
, et al. 
(
1999
).
Narcolepsy in orexin knockout mice: molecular genetics of sleep regulation
.
Cell
98
,
437
-
451
.
Chokroverty
,
S.
(
1986
).
Sleep apnea in narcolepsy
.
Sleep
9
,
250
-
253
.
de Lecea
,
L.
,
Kilduff
,
T. S.
,
Peyron
,
C.
,
Gao
,
X.-B.
,
Foye
,
P. E.
,
Danielson
,
P. E.
,
Fukuhara
,
C.
,
Battenberg
,
E. L. F.
,
Gautvik
,
V. T.
,
Bartlett
,
F. S.
, et al. 
(
1998
).
The hypocretins: hypothalamus-specific peptides with neuroexcitatory activity
.
Proc. Natl. Acad. Sci. USA
95
,
322
-
327
.
Filardi
,
M.
,
Demir
,
N.
,
Pizza
,
F.
,
Vandi
,
S.
,
Antelmi
,
E.
,
Noce
,
S.
,
Bruni
,
O.
and
Plazzi
,
G.
(
2020
).
Prevalence and neurophysiological correlates of sleep disordered breathing in pediatric type 1 narcolepsy
.
Sleep Med.
65
,
8
-
12
.
Ganeshan
,
K.
and
Chawla
,
A.
(
2017
).
Warming the mouse to model human diseases
.
Nat. Rev. Endocrinol.
13
,
458
-
465
.
Gordon
,
C. J.
(
1985
).
Relationship between autonomic and behavioral thermoregulation in the mouse
.
Physiol. Behav.
34
,
687
-
690
.
Hara
,
J.
,
Beuckmann
,
C. T.
,
Nambu
,
T.
,
Willie
,
J. T.
,
Chemelli
,
R. M.
,
Sinton
,
C. M.
,
Sugiyama
,
F.
,
Yagami
,
K.-I.
,
Goto
,
K.
,
Yanagisawa
,
M.
, et al. 
(
2001
).
Genetic ablation of orexin neurons in mice results in narcolepsy, hypophagia, and obesity
.
Neuron
30
,
345
-
354
.
Hodges
,
M. R.
and
Richerson
,
G. B.
(
2008
).
Interaction between defects in ventilatory and thermoregulatory control in mice lacking 5-HT neurons
.
Respir. Physiol. Neurobiol.
164
,
350
-
357
.
Hodges
,
M. R.
,
Tattersall
,
G. J.
,
Harris
,
M. B.
,
McEvoy
,
S. D.
,
Richerson
,
D. N.
,
Deneris
,
E. S.
,
Johnson
,
R. L.
,
Chen
,
Z.-F.
and
Richerson
,
G. B.
(
2008
).
Defects in breathing and thermoregulation in mice with near-complete absence of central serotonin neurons
.
J. Neurosci.
28
,
2495
-
2505
.
Hoshino
,
T.
,
Sasanabe
,
R.
,
Mano
,
M.
,
Nomura
,
A.
,
Kato
,
C.
,
Sato
,
M.
,
Imai
,
M.
,
Murotani
,
K.
,
Guilleminault
,
C.
and
Shiomi
,
T.
(
2019
).
Prevalence of rapid eye movement-related obstructive sleep apnea in adult narcolepsy
.
Intern. Med.
58
,
2151
-
2157
.
Kakizaki
,
M.
,
Tsuneoka
,
Y.
,
Takase
,
K.
,
Kim
,
S. J.
,
Choi
,
J.
,
Ikkyu
,
A.
,
Abe
,
M.
,
Sakimura
,
K.
,
Yanagisawa
,
M.
and
Funato
,
H.
(
2019
).
Differential roles of each orexin receptor signaling in obesity
.
iScience
20
,
1
-
13
.
Kuwaki
,
T.
(
2015
).
Thermoregulation under pressure: a role for orexin neurons
.
Temperature
2
,
379
-
391
.
Li
,
A.
and
Nattie
,
E.
(
2010
).
Antagonism of rat orexin receptors by almorexant attenuates central chemoreception in wakefulness in the active period of the diurnal cycle
.
J. Physiol.
588
,
2935
-
2944
.
Lo Martire
,
V.
,
Silvani
,
A.
,
Bastianini
,
S.
,
Berteotti
,
C.
and
Zoccoli
,
G.
(
2012
).
Effects of ambient temperature on sleep and cardiovascular regulation in mice: the role of hypocretin/orexin neurons
.
PLoS ONE
7
,
e47032
.
Madden
,
C. J.
,
Tupone
,
D.
and
Morrison
,
S. F.
(
2012
).
Orexin modulates brown adipose tissue thermogenesis
.
Biomol. Concepts
3
,
381
-
386
.
Mohammed
,
M.
,
Yanagisawa
,
M.
,
Blessing
,
W.
and
Ootsuka
,
Y.
(
2016
).
Attenuated cold defense responses in orexin neuron-ablated rats
.
Temperature
3
,
465
-
475
.
Nakamura
,
A.
,
Fukuda
,
Y.
and
Kuwaki
,
T.
(
2003
).
Sleep apnea and effect of chemostimulation on breathing instability in mice
.
J. Appl. Physiol.
94
,
525
-
532
.
Nakamura
,
A.
,
Zhang
,
W.
,
Yanagisawa
,
M.
,
Fukuda
,
Y.
and
Kuwaki
,
T.
(
2007
).
Vigilance state-dependent attenuation of hypercapnic chemoreflex and exaggerated sleep apnea in orexin knockout mice
.
J. Appl. Physiol.
102
,
241
-
248
.
Pataka
,
A. D.
,
Frangulyan
,
R. R.
,
Mackay
,
T. W.
,
Douglas
,
N. J.
and
Riha
,
R. L.
(
2012
).
Narcolepsy and sleep-disordered breathing
.
Eur. J. Neurol.
19
,
696
-
702
.
Peyron
,
C.
,
Faraco
,
J.
,
Rogers
,
W.
,
Ripley
,
B.
,
Overeem
,
S.
,
Charnay
,
Y.
,
Nevsimalova
,
S.
,
Aldrich
,
M.
,
Reynolds
,
D.
,
Albin
,
R.
, et al. 
(
2000
).
A mutation in a case of early onset narcolepsy and a generalized absence of hypocretin peptides in human narcoleptic brains
.
Nat. Med.
6
,
991
-
997
.
Peyron
,
C.
,
Tighe
,
D. K.
,
van den Pol
,
A. N.
,
de Lecea
,
L.
,
Heller
,
H. C.
,
Sutcliffe
,
J. G.
and
Kilduff
,
T. S.
(
1998
).
Neurons containing hypocretin (orexin) project to multiple neuronal systems
.
J. Neurosci.
18
,
9996
-
10015
.
Pizza
,
F.
,
Tartarotti
,
S.
,
Poryazova
,
R.
,
Baumann
,
C. R.
and
Bassetti
,
C. L.
(
2013
).
Sleep-disordered breathing and periodic limb movements in narcolepsy with cataplexy: a systematic analysis of 35 consecutive patients
.
Eur. Neurol.
70
,
22
-
26
.
Qureshi
,
M.
,
Khalil
,
M.
,
Kwiatkowski
,
K.
and
Alvaro
,
R. E.
(
2009
).
Morphology of sighs and their role in the control of breathing in preterm infants, term infants and adults
.
Neonatology
96
,
43
-
49
.
Ren
,
J.
,
Ding
,
X.
,
Funk
,
G. D.
and
Greer
,
J. J.
(
2012
).
Anxiety-related mechanisms of respiratory dysfunction in a mouse model of Rett syndrome
.
J. Neurosci.
32
,
17230
-
17240
.
Sakurai
,
T.
,
Amemiya
,
A.
,
Ishii
,
M.
,
Matsuzaki
,
I.
,
Chemelli
,
R. M.
,
Tanaka
,
H.
,
Williams
,
S. C.
,
Richardson
,
J. A.
,
Kozlowski
,
G. P.
,
Wilson
,
S.
, et al. 
(
1998
).
Orexins and orexin receptors: a family of hypothalamic neuropeptides and G protein-coupled receptors that regulate feeding behavior
.
Cell
92
,
573
-
585
.
Sansa
,
G.
,
Iranzo
,
A.
and
Santamaria
,
J.
(
2010
).
Obstructive sleep apnea in narcolepsy
.
Sleep Med.
11
,
93
-
95
.
Silvani
,
A.
,
Berteotti
,
C.
,
Bastianini
,
S.
,
Cohen
,
G.
,
Lo Martire
,
V.
,
Mazza
,
R.
,
Pagotto
,
U.
,
Quarta
,
C.
and
Zoccoli
,
G.
(
2014
).
Cardiorespiratory anomalies in mice lacking CB1 cannabinoid receptors
.
PLoS ONE
9
,
e100536
.
Swoap
,
S. J.
,
Overton
,
J. M.
and
Garber
,
G.
(
2004
).
Effect of ambient temperature on cardiovascular parameters in rats and mice: a comparative approach
.
Am. J. Physiol. Integr. Comp. Physiol.
287
,
R391
-
R396
.
Takahashi
,
Y.
,
Zhang
,
W.
,
Sameshima
,
K.
,
Kuroki
,
C.
,
Matsumoto
,
A.
,
Sunanaga
,
J.
,
Kono
,
Y.
,
Sakurai
,
T.
,
Kanmura
,
Y.
and
Kuwaki
,
T.
(
2013
).
Orexin neurons are indispensable for prostaglandin E2-induced fever and defence against environmental cooling in mice
.
J. Physiol.
591
,
5623
-
5643
.
Tupone
,
D.
,
Madden
,
C. J.
,
Cano
,
G.
and
Morrison
,
S. F.
(
2011
).
An orexinergic projection from perifornical hypothalamus to raphe pallidus increases rat brown adipose tissue thermogenesis
.
J. Neurosci.
31
,
15944
-
15955
.
Willie
,
J. T.
,
Chemelli
,
R. M.
,
Sinton
,
C. M.
,
Tokita
,
S.
,
Williams
,
S. C.
,
Kisanuki
,
Y. Y.
,
Marcus
,
J. N.
,
Lee
,
C.
,
Elmquist
,
J. K.
,
Kohlmeier
,
K. A.
, et al. 
(
2003
).
Distinct narcolepsy syndromes in orexin receptor-2 and Orexin null mice: molecular genetic dissection of non-REM and REM sleep regulatory processes
.
Neuron
38
,
715
-
730
.
Young
,
J. K.
,
Wu
,
M.
,
Manaye
,
K. F.
,
Kc
,
P.
,
Allard
,
J. S.
,
Mack
,
S. O.
and
Haxhiu
,
M. A.
(
2005
).
Orexin stimulates breathing via medullary and spinal pathways
.
J. Appl. Physiol.
98
,
1387
-
1395
.
Zhang
,
W.
,
Fukuda
,
Y.
and
Kuwaki
,
T.
(
2005
).
Respiratory and cardiovascular actions of orexin-A in mice
.
Neurosci. Lett.
385
,
131
-
136
.
Zoccoli
,
G.
,
Amici
,
R.
and
Silvani
,
A.
(
2011
).
The hypothalamus and its functions
. In
Narcolepsy
(ed.
C. R.
Baumann
,
C. L.
Bassetti
and
T. E.
Scammell
), pp.
191
-
203
.
New York
:
Springer
.

Competing interests

The authors declare no competing or financial interests.