Skip to main content
Top
Published in: The Journal of Headache and Pain 1/2019

Open Access 01-12-2019 | Capsaicin | Research article

The influence of rapid eye movement sleep deprivation on nociceptive transmission and the duration of facial allodynia in rats: a behavioral and Fos immunohistochemical study

Authors: Seong Hoon Kim, Ju Yeon Park, Hae Eun Shin, Si baek Lee, Dong Woo Ryu, Tae Won Kim, Jeong Wook Park

Published in: The Journal of Headache and Pain | Issue 1/2019

Login to get access

Abstract

Background

Disrupted sleep is associated with a reciprocal influence on headaches and is one of the contributing factors in the process of chronicity. The goal of the present study was to investigate the influence of sleep on headaches using animal rapid eye movement (REM) sleep deprivation and supradural capsaicin infusion models.

Method

Sprague-Dawley rats underwent REM sleep deprivation (REMSD) for 96 h. The sensory threshold to mechanical stimuli, assessed by the von Frey monofilament test, was measured during the REMSD period. Additionally, the Fos protein expression level was measured in the trigeminocervical complex, periaqueductal gray, and hypothalamus. Following supradural infusion of capsaicin, we evaluated the duration of facial allodynia for 28 days after REMSD.

Results

After REMSD, the sensory threshold to mechanical stimuli was significantly decreased (p < 0.01) and Fos-positivity in the posterior (p = 0.010) and dorsomedial hypothalamus (p = 0.024), ventrolateral periaqueductal gray (p = 0.016), and superficial layer of the trigeminocervical complex (p = 0.019) were significantly increased. The duration of facial allodynia induced by supradural capsaicin infusion was significantly longer in the REM sleep deprivation and capsaicin infusion group (Day 10 PSD vs. Day 25 PSD).

Conclusion

The present study demonstrates that REM sleep deprivation increased nociceptive transmission from trigeminal nerve endings. Furthermore, it suggests that sleep deprivation may contribute to the chronicity of facial allodynia.
Literature
1.
go back to reference Orzel-Gryglewska J (2010) Consequences of sleep deprivation. Int J Occup Med Environ Health 23:95–114CrossRef Orzel-Gryglewska J (2010) Consequences of sleep deprivation. Int J Occup Med Environ Health 23:95–114CrossRef
2.
go back to reference Zhu B, Dong Y, Xu Z, Gompf HS, Ward SA, Xue Z et al (2012) Sleep disturbance induces neuroinflammation and impairment of learning and memory. Neurobiol Dis 48:348–355CrossRef Zhu B, Dong Y, Xu Z, Gompf HS, Ward SA, Xue Z et al (2012) Sleep disturbance induces neuroinflammation and impairment of learning and memory. Neurobiol Dis 48:348–355CrossRef
3.
go back to reference Alkadhi K, Zagaar M, Alhaider I, Salim S, Aleisa A (2013) Neurobiological consequences of sleep deprivation. Curr Neuropharmacol 11:231–249CrossRef Alkadhi K, Zagaar M, Alhaider I, Salim S, Aleisa A (2013) Neurobiological consequences of sleep deprivation. Curr Neuropharmacol 11:231–249CrossRef
4.
go back to reference Drewes AM, Svendsen L, Taagholt SJ, Bjerregard K, Nielsen KD, Hansen B (1998) Sleep in rheumatoid arthritis: a comparison with healthy subjects and studies of sleep/wake interactions. Br J Rheumatol 37:71–81CrossRef Drewes AM, Svendsen L, Taagholt SJ, Bjerregard K, Nielsen KD, Hansen B (1998) Sleep in rheumatoid arthritis: a comparison with healthy subjects and studies of sleep/wake interactions. Br J Rheumatol 37:71–81CrossRef
5.
go back to reference Agargun MY, Tekeoglu I, Gunes A, Adak B, Kara H, Ercan M (1999) Sleep quality and pain threshold in patients with fibromyalgia. Compr Psychiatry 40:226–228CrossRef Agargun MY, Tekeoglu I, Gunes A, Adak B, Kara H, Ercan M (1999) Sleep quality and pain threshold in patients with fibromyalgia. Compr Psychiatry 40:226–228CrossRef
6.
go back to reference Raymond I, Nielsen TA, Lavigne G, Manzini C, Choiniere M (2001) Quality of sleep and its daily relationship to pain intensity in hospitalized adult burn patients. Pain 92:381–388CrossRef Raymond I, Nielsen TA, Lavigne G, Manzini C, Choiniere M (2001) Quality of sleep and its daily relationship to pain intensity in hospitalized adult burn patients. Pain 92:381–388CrossRef
7.
go back to reference Finan PH, Goodin BR, Smith MT (2013) The association of sleep and pain: an update and a path forward. J Pain 14:1539–1552CrossRef Finan PH, Goodin BR, Smith MT (2013) The association of sleep and pain: an update and a path forward. J Pain 14:1539–1552CrossRef
8.
go back to reference Vanini G (2016) Sleep deprivation and recovery sleep prior to a noxious inflammatory insult influence characteristics and duration of pain. Sleep 39:133–142CrossRef Vanini G (2016) Sleep deprivation and recovery sleep prior to a noxious inflammatory insult influence characteristics and duration of pain. Sleep 39:133–142CrossRef
9.
go back to reference Bardin L (2011) The complex role of serotonin and 5-HT receptors in chronic pain. Behav Pharmacol 22:390–404CrossRef Bardin L (2011) The complex role of serotonin and 5-HT receptors in chronic pain. Behav Pharmacol 22:390–404CrossRef
10.
go back to reference Ukponmwan OE, Rupreht J, Dzoljic MR (1984) REM sleep deprivation decreases the antinociceptive property of enkephalinase-inhibition, morphine and cold-water-swim. Gen Pharmacol 15:255–258CrossRef Ukponmwan OE, Rupreht J, Dzoljic MR (1984) REM sleep deprivation decreases the antinociceptive property of enkephalinase-inhibition, morphine and cold-water-swim. Gen Pharmacol 15:255–258CrossRef
11.
go back to reference Ukponmwan OE, Rupreht J, Dzoljic M (1986) An analgesic effect of enkephalinase inhibition is modulated by monoamine oxidase-B and REM sleep deprivations. Naunyn Schmiedeberg's Arch Pharmacol 332:376–379CrossRef Ukponmwan OE, Rupreht J, Dzoljic M (1986) An analgesic effect of enkephalinase inhibition is modulated by monoamine oxidase-B and REM sleep deprivations. Naunyn Schmiedeberg's Arch Pharmacol 332:376–379CrossRef
12.
go back to reference Bartsch T, Knight YE, Goadsby PJ (2004) Activation of 5-HT(1B/1D) receptor in the periaqueductal gray inhibits nociception. Ann Neurol 56:371–381CrossRef Bartsch T, Knight YE, Goadsby PJ (2004) Activation of 5-HT(1B/1D) receptor in the periaqueductal gray inhibits nociception. Ann Neurol 56:371–381CrossRef
13.
go back to reference Bartsch T, Levy MJ, Knight YE, Goadsby PJ (2004) Differential modulation of nociceptive dural input to [hypocretin] orexin a and B receptor activation in the posterior hypothalamic area. Pain 109:367–378CrossRef Bartsch T, Levy MJ, Knight YE, Goadsby PJ (2004) Differential modulation of nociceptive dural input to [hypocretin] orexin a and B receptor activation in the posterior hypothalamic area. Pain 109:367–378CrossRef
14.
go back to reference Tomim DH, Pontarolla FM, Bertolini JF, Arase M, Tobaldini G, Lima MMS et al (2016) The Pronociceptive effect of paradoxical sleep deprivation in rats: evidence for a role of descending pain modulation mechanisms. Mol Neurobiol 53:1706–1717CrossRef Tomim DH, Pontarolla FM, Bertolini JF, Arase M, Tobaldini G, Lima MMS et al (2016) The Pronociceptive effect of paradoxical sleep deprivation in rats: evidence for a role of descending pain modulation mechanisms. Mol Neurobiol 53:1706–1717CrossRef
15.
16.
go back to reference Zebenholzer K, Andree C, Lechner A, Broessner G, Lampl C, Luthringshausen G et al (2015) Prevalence, management and burden of episodic and chronic headaches--a cross-sectional multicentre study in eight Austrian headache centres. J Headache Pain 16:531CrossRef Zebenholzer K, Andree C, Lechner A, Broessner G, Lampl C, Luthringshausen G et al (2015) Prevalence, management and burden of episodic and chronic headaches--a cross-sectional multicentre study in eight Austrian headache centres. J Headache Pain 16:531CrossRef
17.
go back to reference Ong JC, Park M (2012) Chronic headaches and insomnia: working toward a biobehavioral model. Cephalalgia 32:1059–1070CrossRef Ong JC, Park M (2012) Chronic headaches and insomnia: working toward a biobehavioral model. Cephalalgia 32:1059–1070CrossRef
18.
go back to reference Goadsby PJ (2005) Migraine, allodynia. sensitisation and all of that Eur Neurol 53(Suppl 1):10–16CrossRef Goadsby PJ (2005) Migraine, allodynia. sensitisation and all of that Eur Neurol 53(Suppl 1):10–16CrossRef
19.
go back to reference Potrebic S, Ahn AH, Skinner K, Fields HL, Basbaum AI (2003) Peptidergic nociceptors of both trigeminal and dorsal root ganglia express serotonin 1D receptors: implications for the selective antimigraine action of triptans. J Neurosci 23:10988–10997CrossRef Potrebic S, Ahn AH, Skinner K, Fields HL, Basbaum AI (2003) Peptidergic nociceptors of both trigeminal and dorsal root ganglia express serotonin 1D receptors: implications for the selective antimigraine action of triptans. J Neurosci 23:10988–10997CrossRef
20.
go back to reference Mitsikostas DD, Sanchez del Rio M (2001) Receptor systems mediating c-fos expression within trigeminal nucleus caudalis in animal models of migraine. Brain Res Brain Res Rev 35:20–35CrossRef Mitsikostas DD, Sanchez del Rio M (2001) Receptor systems mediating c-fos expression within trigeminal nucleus caudalis in animal models of migraine. Brain Res Brain Res Rev 35:20–35CrossRef
21.
go back to reference Aguggia M (2012) Allodynia and migraine. Neurol Sci 33:Suppl 1:S9–11 Aguggia M (2012) Allodynia and migraine. Neurol Sci 33:Suppl 1:S9–11
22.
go back to reference Wieseler J, Ellis A, Sprunger D, Brown K, McFadden A, Mahoney J et al (2010) A novel method for modeling facial allodynia associated with migraine in awake and freely moving rats. J Neurosci Methods 185:236–245CrossRef Wieseler J, Ellis A, Sprunger D, Brown K, McFadden A, Mahoney J et al (2010) A novel method for modeling facial allodynia associated with migraine in awake and freely moving rats. J Neurosci Methods 185:236–245CrossRef
23.
go back to reference Oshinsky ML, Gomonchareonsiri S (2007) Episodic dural stimulation in awake rats: a model for recurrent headache. Headache 47:1026–1036CrossRef Oshinsky ML, Gomonchareonsiri S (2007) Episodic dural stimulation in awake rats: a model for recurrent headache. Headache 47:1026–1036CrossRef
24.
go back to reference Cohen HB, Dement WC (1965) Sleep: changes in threshold to electroconvulsive shock in rats after deprivation of "paradoxical" phase. Science 150:1318–1319CrossRef Cohen HB, Dement WC (1965) Sleep: changes in threshold to electroconvulsive shock in rats after deprivation of "paradoxical" phase. Science 150:1318–1319CrossRef
25.
go back to reference Chaplan SR, Bach FW, Pogrel JW, Chung JM, Yaksh TL (1994) Quantitative assessment of tactile allodynia in the rat paw. J Neurosci Methods 53:55–63CrossRef Chaplan SR, Bach FW, Pogrel JW, Chung JM, Yaksh TL (1994) Quantitative assessment of tactile allodynia in the rat paw. J Neurosci Methods 53:55–63CrossRef
26.
go back to reference Vos BP, Strassman AM, Maciewicz RJ (1994) Behavioral evidence of trigeminal neuropathic pain following chronic constriction injury to the rat's infraorbital nerve. J Neurosci 14:2708–2723CrossRef Vos BP, Strassman AM, Maciewicz RJ (1994) Behavioral evidence of trigeminal neuropathic pain following chronic constriction injury to the rat's infraorbital nerve. J Neurosci 14:2708–2723CrossRef
27.
go back to reference Paxinos G, KBJ F (2003) The rat brain in stereotaxic coordinates, 2nd edn. Academic Press, London Paxinos G, KBJ F (2003) The rat brain in stereotaxic coordinates, 2nd edn. Academic Press, London
28.
go back to reference Onen SH, Alloui A, Eschalier A, Dubray C (2000) Vocalization thresholds related to noxious paw pressure are decreased by paradoxical sleep deprivation and increased after sleep recovery in rat. Neurosci Lett 291:25–28CrossRef Onen SH, Alloui A, Eschalier A, Dubray C (2000) Vocalization thresholds related to noxious paw pressure are decreased by paradoxical sleep deprivation and increased after sleep recovery in rat. Neurosci Lett 291:25–28CrossRef
29.
go back to reference Hicks RA, Coleman DD, Ferrante F, Sahatjian M, Hawkins J (1979) Pain thresholds in rats during recovery from REM sleep deprivation. Percept Mot Skills 48:687–690CrossRef Hicks RA, Coleman DD, Ferrante F, Sahatjian M, Hawkins J (1979) Pain thresholds in rats during recovery from REM sleep deprivation. Percept Mot Skills 48:687–690CrossRef
30.
go back to reference Hicks RA, Moore JD, Findley P, Hirshfield C, Humphrey V (1978) REM sleep deprivation and pain thresholds in rats. Percept Mot Skills 47:848–850CrossRef Hicks RA, Moore JD, Findley P, Hirshfield C, Humphrey V (1978) REM sleep deprivation and pain thresholds in rats. Percept Mot Skills 47:848–850CrossRef
31.
go back to reference Lovati C, D'Amico D, Raimondi E, Mariani C, Bertora P (2010) Sleep and headache: a bidirectional relationship. Expert Rev Neurother 10:105–117CrossRef Lovati C, D'Amico D, Raimondi E, Mariani C, Bertora P (2010) Sleep and headache: a bidirectional relationship. Expert Rev Neurother 10:105–117CrossRef
32.
go back to reference Cortelli P, Pierangeli G (2007) Hypothalamus and headaches. Neurol Sci 28(Suppl 2):S198–S202CrossRef Cortelli P, Pierangeli G (2007) Hypothalamus and headaches. Neurol Sci 28(Suppl 2):S198–S202CrossRef
33.
go back to reference Fuller PM, Gooley JJ, Saper CB (2006) Neurobiology of the sleep-wake cycle: sleep architecture, circadian regulation, and regulatory feedback. J Biol Rhythm 21:482–493CrossRef Fuller PM, Gooley JJ, Saper CB (2006) Neurobiology of the sleep-wake cycle: sleep architecture, circadian regulation, and regulatory feedback. J Biol Rhythm 21:482–493CrossRef
34.
go back to reference Estabrooke IV, McCarthy MT, Ko E, Chou TC, Chemelli RM, Yanagisawa M et al (2001) Fos expression in orexin neurons varies with behavioral state. J Neurosci 21:1656–1662CrossRef Estabrooke IV, McCarthy MT, Ko E, Chou TC, Chemelli RM, Yanagisawa M et al (2001) Fos expression in orexin neurons varies with behavioral state. J Neurosci 21:1656–1662CrossRef
35.
go back to reference Weiller C, May A, Limmroth V, Juptner M, Kaube H, Schayck RV et al (1995) Brain stem activation in spontaneous human migraine attacks. Nat Med 1:658–660CrossRef Weiller C, May A, Limmroth V, Juptner M, Kaube H, Schayck RV et al (1995) Brain stem activation in spontaneous human migraine attacks. Nat Med 1:658–660CrossRef
36.
go back to reference Hoffmann J, Supronsinchai W, Akerman S, Andreou AP, Winrow CJ, Renger J et al (2015) Evidence for orexinergic mechanisms in migraine. Neurobiol Dis 74:137–143CrossRef Hoffmann J, Supronsinchai W, Akerman S, Andreou AP, Winrow CJ, Renger J et al (2015) Evidence for orexinergic mechanisms in migraine. Neurobiol Dis 74:137–143CrossRef
37.
go back to reference Akerman S, Holland PR, Lasalandra MP, Goadsby PJ (2013) Endocannabinoids in the brainstem modulate dural trigeminovascular nociceptive traffic via CB1 and "triptan" receptors: implications in migraine. J Neurosci 33:14869–14877CrossRef Akerman S, Holland PR, Lasalandra MP, Goadsby PJ (2013) Endocannabinoids in the brainstem modulate dural trigeminovascular nociceptive traffic via CB1 and "triptan" receptors: implications in migraine. J Neurosci 33:14869–14877CrossRef
38.
go back to reference Benjamin L, Levy MJ, Lasalandra MP, Knight YE, Akerman S, Classey JD et al (2004) Hypothalamic activation after stimulation of the superior sagittal sinus in the cat: a Fos study. Neurobiol Dis 16:500–505CrossRef Benjamin L, Levy MJ, Lasalandra MP, Knight YE, Akerman S, Classey JD et al (2004) Hypothalamic activation after stimulation of the superior sagittal sinus in the cat: a Fos study. Neurobiol Dis 16:500–505CrossRef
39.
go back to reference Leone M, Proietti Cecchini A (2016) Deep brain stimulation in headache. Cephalalgia 36:1143–1148CrossRef Leone M, Proietti Cecchini A (2016) Deep brain stimulation in headache. Cephalalgia 36:1143–1148CrossRef
40.
go back to reference Veloso F, Kumar K, Toth C (1998) Headache secondary to deep brain implantation. Headache 38:507–515CrossRef Veloso F, Kumar K, Toth C (1998) Headache secondary to deep brain implantation. Headache 38:507–515CrossRef
41.
go back to reference Knight YE, Goadsby PJ (2001) The periaqueductal grey matter modulates trigeminovascular input: a role in migraine? Neuroscience 106:793–800CrossRef Knight YE, Goadsby PJ (2001) The periaqueductal grey matter modulates trigeminovascular input: a role in migraine? Neuroscience 106:793–800CrossRef
42.
go back to reference Hoskin KL, Bulmer DC, Lasalandra M, Jonkman A, Goadsby PJ (2001) Fos expression in the midbrain periaqueductal grey after trigeminovascular stimulation. J Anat 198:29–35CrossRef Hoskin KL, Bulmer DC, Lasalandra M, Jonkman A, Goadsby PJ (2001) Fos expression in the midbrain periaqueductal grey after trigeminovascular stimulation. J Anat 198:29–35CrossRef
43.
go back to reference Clement O, Sapin E, Libourel PA, Arthaud S, Brischoux F, Fort P et al (2012) The lateral hypothalamic area controls paradoxical (REM) sleep by means of descending projections to brainstem GABAergic neurons. J Neurosci 32:16763–16774CrossRef Clement O, Sapin E, Libourel PA, Arthaud S, Brischoux F, Fort P et al (2012) The lateral hypothalamic area controls paradoxical (REM) sleep by means of descending projections to brainstem GABAergic neurons. J Neurosci 32:16763–16774CrossRef
44.
go back to reference Petitjean F, Sakai K, Blondaux C, Jouvet M (1975) Hypersomnia by isthmic lesion in cat. II Neurophysiological and pharmacological study. Brain Res 88:439–453CrossRef Petitjean F, Sakai K, Blondaux C, Jouvet M (1975) Hypersomnia by isthmic lesion in cat. II Neurophysiological and pharmacological study. Brain Res 88:439–453CrossRef
45.
go back to reference Sastre JP, Buda C, Kitahama K, Jouvet M (1996) Importance of the ventrolateral region of the periaqueductal gray and adjacent tegmentum in the control of paradoxical sleep as studied by muscimol microinjections in the cat. Neuroscience 74:415–426CrossRef Sastre JP, Buda C, Kitahama K, Jouvet M (1996) Importance of the ventrolateral region of the periaqueductal gray and adjacent tegmentum in the control of paradoxical sleep as studied by muscimol microinjections in the cat. Neuroscience 74:415–426CrossRef
46.
go back to reference Akerman S, Holland PR, Goadsby PJ (2011) Diencephalic and brainstem mechanisms in migraine. Nat Rev Neurosci 12:570–584CrossRef Akerman S, Holland PR, Goadsby PJ (2011) Diencephalic and brainstem mechanisms in migraine. Nat Rev Neurosci 12:570–584CrossRef
47.
go back to reference Melo-Carrillo A, Lopez-Avila A (2013) A chronic animal model of migraine, induced by repeated meningeal nociception, characterized by a behavioral and pharmacological approach. Cephalalgia 33:1096–1105CrossRef Melo-Carrillo A, Lopez-Avila A (2013) A chronic animal model of migraine, induced by repeated meningeal nociception, characterized by a behavioral and pharmacological approach. Cephalalgia 33:1096–1105CrossRef
48.
go back to reference Palchykova S, Winsky-Sommerer R, Meerlo P, Durr R, Tobler I (2006) Sleep deprivation impairs object recognition in mice. Neurobiol Learn Mem 85:263–271CrossRef Palchykova S, Winsky-Sommerer R, Meerlo P, Durr R, Tobler I (2006) Sleep deprivation impairs object recognition in mice. Neurobiol Learn Mem 85:263–271CrossRef
49.
go back to reference Abel T, Havekes R, Saletin JM, Walker MP (2013) Sleep, plasticity and memory from molecules to whole-brain networks. Curr Biol 23:R774–R788CrossRef Abel T, Havekes R, Saletin JM, Walker MP (2013) Sleep, plasticity and memory from molecules to whole-brain networks. Curr Biol 23:R774–R788CrossRef
50.
go back to reference Graves LA, Heller EA, Pack AI, Abel T (2003) Sleep deprivation selectively impairs memory consolidation for contextual fear conditioning. Learn Mem 10:168–176CrossRef Graves LA, Heller EA, Pack AI, Abel T (2003) Sleep deprivation selectively impairs memory consolidation for contextual fear conditioning. Learn Mem 10:168–176CrossRef
51.
go back to reference Machado RB, Hipolide DC, Benedito-Silva AA, Tufik S (2004) Sleep deprivation induced by the modified multiple platform technique: quantification of sleep loss and recovery. Brain Res 1004:45–51CrossRef Machado RB, Hipolide DC, Benedito-Silva AA, Tufik S (2004) Sleep deprivation induced by the modified multiple platform technique: quantification of sleep loss and recovery. Brain Res 1004:45–51CrossRef
52.
go back to reference Grahnstedt S, Ursin R (1985) Platform sleep deprivation affects deep slow wave sleep in addition to REM sleep. Behav Brain Res 18:233–239CrossRef Grahnstedt S, Ursin R (1985) Platform sleep deprivation affects deep slow wave sleep in addition to REM sleep. Behav Brain Res 18:233–239CrossRef
Metadata
Title
The influence of rapid eye movement sleep deprivation on nociceptive transmission and the duration of facial allodynia in rats: a behavioral and Fos immunohistochemical study
Authors
Seong Hoon Kim
Ju Yeon Park
Hae Eun Shin
Si baek Lee
Dong Woo Ryu
Tae Won Kim
Jeong Wook Park
Publication date
01-12-2019
Publisher
Springer Milan
Published in
The Journal of Headache and Pain / Issue 1/2019
Print ISSN: 1129-2369
Electronic ISSN: 1129-2377
DOI
https://doi.org/10.1186/s10194-019-0977-0

Other articles of this Issue 1/2019

The Journal of Headache and Pain 1/2019 Go to the issue