Elsevier

Neuropharmacology

Volume 139, 1 September 2018, Pages 52-60
Neuropharmacology

Chronic sleep restriction increases pain sensitivity over time in a periaqueductal gray and nucleus accumbens dependent manner

https://doi.org/10.1016/j.neuropharm.2018.06.022Get rights and content

Highlights

  • Sleep restriction for 6 h daily induces a pronociceptive effect.

  • The effect increases progressively from day 3 to day 12 remaining stable thereafter.

  • Two consecutive days of free sleep were not enough to reverse the effect.

  • Chronic sleep restriction increases pain sensitivity in a NAc and PAG dependent manner.

  • c-Fos protein expression within the NAc and PAG correlates with the pronociceptive effect.

Abstract

Painful conditions and sleep disturbances are major public health problems worldwide and one directly affects the other. Sleep loss increases pain prevalence and severity; while pain disturbs sleep. However, the underlying mechanisms are largely unknown. Here we asked whether chronic sleep restriction for 6 h daily progressively increases pain sensitivity and if this increase is reversed after two days of free sleep. Also, whether the pronociceptive effect of chronic sleep restriction depends on the periaqueductal grey and on the nucleus accumbens, two key regions involved in the modulation of pain and sleep-wake cycle. We showed that sleep restriction induces a pronociceptive effect characterized by a significant decrease in the mechanical paw withdrawal threshold in rats. Such effect increases progressively from day 3 to day 12 remaining stable thereafter until day 26. Two consecutive days of free sleep were not enough to reverse the effect, not even to attenuate it. This pronociceptive effect depends on the periaqueductal grey and on the nucleus accumbens, since it was prevented by their excitotoxic lesion. Complementarily, chronic sleep restriction significantly increased c-Fos protein expression within the periaqueductal grey and the nucleus accumbens and this correlates with the intensity of the pronociceptive effect, suggesting that the greater the neural activity in this regions, the greater the effect. These findings may contribute not only to understand why painful conditions are more prevalent and severe among people who sleep poorly, but also to develop therapeutic strategies to prevent this, increasing the effectiveness of pain management in this population.

Introduction

Pain conditions and sleep disorders are major public health problems worldwide (Appleton et al., 2018; Murphy et al., 2017) and there is a clear bidirectional relationship between them. There is no doubt that pain impairs sleep (Artner et al., 2013; Karaman et al., 2014) and different types of sleep impairment increase pain sensitivity (Okifuji and Hare, 2011). However, the underlying mechanisms are largely unknown.

The reason why sleep disorders are great predictors of pain development (Mork and Nilsen, 2012; Okifuji and Hare, 2011) may rely on the ability of sleep loss to disrupt endogenous pain modulation, as suggested by clinical findings (Paul-Savoie et al., 2012; Tiede et al., 2010). In fact, several brain regions that play a key role in pain modulation, such as the ventrolateral periaqueductal gray (vlPAG) (Fields, 2004), and the Nucleus Accumbens (NAc), (Gear et al., 1999), also contribute to control sleep-wake cycle (Lu et al., 2006; Oishi et al., 2017; Weber et al., 2018). Since most sleep disorders are characterized by impairment mainly in rapid eye movement (REM) sleep (Brown et al., 2012; Naiman, 2017), we have performed REM sleep deprivation (REM-SD) in rats to provide a mechanistic basis for these clinical observations. According to our previous data, REM-SD increases nociceptive responses by disrupting the PAG-RVM (periaqueductal gray – rostral ventral medulla) descending pain modulation system (Tomim et al., 2016) as well as by increasing NAc adenosinergic activity and by decreasing NAc dopaminergic activity (Sardi et al., 2018).

However, insomnia (Calhoun et al., 2014) and restriction of sleep time due to occupational or recreational reasons have become increasingly frequent in the modern society (Owens et al., 2014). In order to mimic this decrease in sleep duration in laboratory animals, some types of gentle stimulation have been used to keep them awake for long periods of time. Like selective REM-SD (Damasceno et al., 2009; Nascimento et al., 2007; Sardi et al., 2018; Tomim et al., 2016; Wei et al., 2013), sleep restriction (SR) has been associated with increased pain sensitivity in both humans (Okifuji and Hare, 2011; Tiede et al., 2010) and animals (Alexandre et al., 2017). However, the underlying mechanisms are largely unknown and many unanswered questions remain. For example: Does the pronociceptive effect of sleep restriction increase over time? If yes, when does it reach the maximum intensity? Are two days of normal sleep (mimicking free sleep on weekends) enough to normalize pain sensitivity? Is the pronociceptive effect of chronic sleep restriction also dependent on the PAG and on the NAc? If yes, the neuronal activity within these regions increases with chronic sleep restriction? This study aimed to answer these questions.

Section snippets

Animals

The experiments were performed in male Wistar rats (270–300 g). The animals were housed five per cage in a room with controlled 12:12-h light/dark cycle and temperature (23 °C ± 2), with free access to food and water. All animal experimental procedures and protocols were approved by the Committee on Animal Research of the Federal University of Parana, Brazil, and followed the guidelines of the Ethics Standards of the International Association for the Study of Pain in animals (Zimmermann, 1983).

Stereotaxic surgery and NMDA lesion

The pronociceptive effect of chronic sleep restriction and its temporal evolution

Chronic sleep restriction (CSR) for six hours daily progressively increased nociceptive response, as demonstrated by the decrease in mechanical nociceptive paw-withdrawal threshold (Fig. 1, repeated-measures (time) ANOVA – sleep condition (CSR or control procedure): F(1,14) = 248.15, p < 0.001; sleep condition x time: F(11,154) = 16.208, p < 0.001. Post hoc analysis using Tukey's test indicated that CSR decreased mechanical nociceptive threshold during the overall experiment, p < 0.003). Only

Discussion

This study demonstrated that sleep restriction for 6 h daily induces a pronociceptive effect that increases progressively from day 3 to day 12 remaining stable thereafter. Repeated two-day-periods of free sleep were neither enough to normalize pain sensitivity, nor to even attenuate the increased nociceptive response. The pronociceptive effect of CSR depends on both the NAc and the PAG, since it was prevented by the excitotoxic lesion of any one of them. Complementarily, CSR significantly

Conflicts of interest

The authors have no conflicts of interest to declare.

Acknowledgments

This study was supported by National Council for Scientific and Technological Development, Coordination for the Improvement of Higher Education Personnel and Funding Authority for Studies and Projects (FINEP, Brazil). N.F.S. and G.T. are recipient of PhD fellowships from CAPES. L.F is recipient of a research fellowship from CNPq. M.M.S.L. is the recipient of a CNPq fellowship, Research Grant no. 305986/2016-3. The authors declare that there is no conflict of interests.

References (54)

  • Q. Meng et al.

    Peri-adolescence isolation rearing alters social behavior and nociception in rats

    Neurosci. Lett.

    (2010)
  • M.J. Millan

    Descending control of pain

    Prog. Neurobiol.

    (2002)
  • J. Miranda et al.

    Effect of pain chronification and chronic pain on an endogenous pain modulation circuit in rats

    Neuroscience

    (2015)
  • D.C. Nascimento et al.

    Pain hypersensitivity induced by paradoxical sleep deprivation is not due to altered binding to brain mu-opioid receptors

    Behav. Brain Res.

    (2007)
  • Y. Oishi et al.

    The control of sleep and wakefulness by mesolimbic dopamine systems

    Neurosci. Res.

    (2017)
  • S. Schuh-Hofer et al.

    One night of total sleep deprivation promotes a state of generalized hyperalgesia: a surrogate pain model to study the relationship of insomnia and pain

    Pain

    (2013)
  • W. Tiede et al.

    Sleep restriction attenuates amplitudes and attentional modulation of pain-related evoked potentials, but augments pain ratings in healthy volunteers

    Pain

    (2010)
  • G. Tobaldini et al.

    Ascending nociceptive control contributes to the antinociceptive effect of acupuncture in a rat model of acute pain

    J. Pain

    (2014)
  • J. Townhill et al.

    Using Actiwatch to monitor circadian rhythm disturbance in Huntington' disease: a cautionary note

    J. Neurosci. Meth.

    (2016)
  • H. Wei et al.

    Spinal D-amino acid oxidase contributes to mechanical pain hypersensitivity induced by sleep deprivation in the rat

    Pharmacol. Biochem. Behav.

    (2013)
  • J.P. Zhang et al.

    Projections of nucleus accumbens adenosine A2A receptor neurons in the mouse brain and their implications in mediating sleep-wake regulation

    Front. Neuroanat.

    (2013)
  • M. Zimmermann

    Ethical guidelines for investigations of experimental pain in conscious animals

    Pain

    (1983)
  • C. Alexandre et al.

    Decreased alertness due to sleep loss increases pain sensitivity in mice

    Nat. Med.

    (2017)
  • J. Artner et al.

    Prevalence of sleep deprivation in patients with chronic neck and back pain: a retrospective evaluation of 1016 patients

    J. Pain Res.

    (2013)
  • M.N. Baliki et al.

    Corticostriatal functional connectivity predicts transition to chronic back pain

    Nat. Neurosci.

    (2012)
  • A.I. Basbaum et al.

    The origin of descending pathways in the dorsolateral funiculus of the spinal cord of the cat and rat: further studies on the anatomy of pain modulation

    J. Comp. Neurol.

    (1979)
  • R.E. Brown et al.

    Control of sleep and wakefulness

    Physiol. Rev.

    (2012)
  • Cited by (24)

    • Comorbidities of migraine: Sleep disorders

      2024, Handbook of Clinical Neurology
    • The differential effects of sleep deprivation on pain perception in individuals with or without chronic pain: A systematic review and meta-analysis

      2022, Sleep Medicine Reviews
      Citation Excerpt :

      For example, Simpson et al. [13] found that significant increases in spontaneous pain intensity and temporal summation of pain occurred in the second and third week rather than the first week of partial SD in healthy adults. An animal study that involved experimentally induced partial SD in rats showed that pain sensitivity became significant from the third day onward and continued to increase till the 12th day [59]. Further, different pain measurement protocols (e.g., types of painful stimuli or sites of stimuli applications) may influence the measured sleep-deprived pain perception.

    • Association of depressive symptoms with marital status among the middle-aged and elderly in Rural China–Serial mediating effects of sleep time, pain and life satisfaction

      2022, Journal of Affective Disorders
      Citation Excerpt :

      So emotion dysregulation could possibly mediate the relationship between sleep time and pain. Natalia F Sardi et al. (Sardi et al., 2018) found in animal research that chronic sleep restriction significantly increased the expression of c-Fos protein within the periaqueductal gray and the nucleus accumbens, which correlated with the intensity of the pronociceptive effect. Our findings confirmed the correlation between sleep time and pain.

    • At the intersection of sleep deficiency and opioid use: mechanisms and therapeutic opportunities

      2021, Translational Research
      Citation Excerpt :

      Again, when describing individual studies, we will use study-specific language. Across studies of individuals with varying substance use disorders (SUDs), it has been reported or experimentally demonstrated that sleep deficiency is related to increased pain sensitivity,20-24 emotion dysregulation,25-27 and impaired frontal-executive control and decision making.28-33 Furthermore, sleep deficiency is associated with increased propensity for lapse/relapse to substances34 including alcohol,35-38 tobacco/nicotine,26,39-41 cannabis,42-44 and methamphetamine,45 and has been hypothesized to impair extinction of drug-environment conditioning.46

    View all citing articles on Scopus
    View full text