Comprehensive Physiology Wiley Online Library

Orexin in Respiratory and Autonomic Regulation, Health and Diseases

Full Article on Wiley Online Library



Abstract

Orexin neurons, located in the hypothalamus, produce orexin‐A and orexin‐B neuropeptides and send widespread projections throughout the central nervous system, including many nuclei that are critically involved in sleep‐wake, cardiorespiratory, and autonomic regulation. Significant progress has been made to better understand the roles of orexins in the control of breathing and autonomic functions since the discovery of orexins in 1998. Orexin neurons are CO2/pH chemosensitive and blockade of orexin receptors with orexin receptor antagonists can significantly attenuate ventilatory response to hypercapnia or CO2 chemoreflex. Animal models with orexin abnormalities, for example, too little or too much, have all been reported to have significant alterations in breathing, central chemoreception (hypercapnic chemoreflex), blood pressure, thermoregulation, and cardiorespiratory responses to stress. More recent studies further show that abnormalities of the orexin system are linked to many neurological disorders in addition to narcolepsy, for example, sleep disorders, neurodegenerative disorders, neurogenic hypertension, and sudden infant death syndrome. These new findings have significantly advanced the knowledge in understanding the underlying mechanism of orexin‐associated health and diseases while providing a new pathway for possible treatments. In this article, we will discuss some of the progresses in basic research and in health and diseases. © 2020 American Physiological Society. Compr Physiol 10:345‐363, 2020.

Keywords: orexin; control of breathing and autonomic functions; central chemoreception; neurogenic diseases

Comprehensive Physiology offers downloadable PowerPoint presentations of figures for non-profit, educational use, provided the content is not modified and full credit is given to the author and publication.

Download a PowerPoint presentation of all images


Figure 1. Figure 1. Schematic representation showing the broad anatomical and functional connections between orexin neurons and other neuronal systems. It summarizes how the orexin system may be critically linked with the cardiorespiratory and autonomic regulatory system and the vigilance‐state dependent behavioral regulatory system. Arrows indicate the excitatory connections, while circles indicate the inhibitory connections. AMG, amygdala; BAT, brown adipose tissue; BNST, bed nucleus of the stria terminalis; DR, dorsal raphe; LC, locus coeruleus; MLR, medullary locomotor region; MR, medullary raphe; NTS, nucleus tractus solitarius; PAG, periaqueductal gray; PBC, pre‐Bötzinger complex; PBN, parabrachial nucleus; PVN, paraventricular nucleus; RTN, retrotrapezoid nucleus; RVLM, rostral ventrolateral medulla where sympathetic cardiovascular premotor neurons are located; SCN, suprachiasmatic nucleus; TMN, tuberomammillary nucleus; VLPO, ventrolateral preoptic nucleus. Adapted, with permission, from Kuwaki, 2015 78.
Figure 2. Figure 2. Transgenic orexin deficient mice have lower mean arterial blood pressure and attenuated ventilatory response to CO2 or hypercapnic chemoreflex. (A) Ventilatory response to hypercapnia is severely attenuated in Orexin‐KO than wild‐type (WT) controls only in wakefulness. (*P < 0.05 compared with WT mice. P < 0.05 compared with the data during awake). (B) Resting blood pressure is significantly lower in orexin‐KO than WT controls in both light and dark diurnal cycles. ORX‐KO, orexin knockout mice; WT, wild‐type mice; SWS, slow‐wave sleep; REM, rapid‐eye movement sleep. Adapted, with permission, from Kayaba et al. 2013 71, and Kuwaki et al. 2010 79.
Figure 3. Figure 3. Spontaneously hypertensive rats have an excessive number of orexin‐producing neurons, high mean arterial blood pressure (MABP), and exaggerated ventilatory response to hypercapnia. (A) Distribution of OX‐ir neurons in three hypothalamic zones and whole hypothalamus in SHR (dark gray bar) and WKY (white bar) at young and adult age. (B) Resting MABP with (hatched bars) and without (black bars) treatment of a dual OXR antagonist, almorexant (Amxt), in young and adult SHRs versus that in age‐matched WKY rats (gray bars) in wakefulness, NREM, and REM sleep. (C) Ventilatory response (% change of V˙E) in normoxic hypercapnia with Amxt treatment (hatched bars) and without (black bars) in young and adult SHRs versus that in normotensive young and adult WKY rats (gray bars). DMH, dorsomedial hypothalamus; f, fornix; Hyp, hypothalamus (all three zones); LHA, lateral hypothalamic area; PeF, perifornical hypothalamus 94.
Figure 4. Figure 4. Orexins are involved in regulation of central chemoreception. Schematic sagittal brain section shows the location of orexin neurons and two brainstem putative central chemoreceptor sites, the RTN and medullary raphe. (E) Adapted, with permission, from Nambu et al. 115. Orexin neurons themselves are CO2/pH chemosensitive in brain slices of the hypothalamus of transgenic mice that express GFP only in orexin neurons. Demonstrates the effect of CO2‐induced low pHe on orexin neurons (A‐a). Effect of HEPES‐buffered low pHe on orexin neurons (A‐b). (a,b) Adapted, with permission, from Williams et al. 2007 172. Blocking OXRs with a dual OXR antagonist, Amxt, via oral administration significantly lowers the ventilatory response to hypercapnia only in wakefulness in the dark period (B). Focal inhibition of OX1R in the RTN (C) or medullary raphe (D) significantly attenuated CO2 chemoreflex. (B) Adapted, with permission, from Li and Nattie 2010 91; (C) Adapted, with permission, from Dias et al. 2009 36; (D) Adapted, with permission, from Dias et al. 2010 35. aCSF, artificial cerebrospinal fluid; RTN, retrotrapezoid nucleus.
Figure 5. Figure 5. The effects of OXA on hypoglossal motor (HMNs) neuronal activity and of eliminating orexin neurons on genioglossal muscle activity (GG‐EMG). Orexin‐A concentration‐dependently increased the firing rate of HMNs in brain slices of neonatal rats (A). Bilateral lesions of orexin neurons in the hypothalamus of adult rats via orexin‐B‐SAP (400 nL per side, 0.43 mg/mL) decreased the respiratory‐related GG‐EMG (B, lower panel) compared to control‐injected rats (B, top panel). Tonic activity of GG‐EMG is not affected while respiratory‐related activity is significantly attenuated post‐lesion (C). Adapted, with permission, from Zhang et al. 2014 181.
Figure 6. Figure 6. The effects of OX1R antagonist and OXB agonist on orexin‐induced increase in mean arterial pressure (MAP) and sympathetic nerve activity (SNA) in the RVLM (A) and spinal cord (B). OX1R antagonist, SB334867, can significantly attenuate the orexin A‐induced MAP and sSNA effects in the RVLM (A). OXA dose‐dependently increased MAP and sSNA and OX1R antagonism can block such OXA‐induced effects in the spinal cord (B). ***P < 0.001, **P < 0.01, *P < 0.05, significantly different from PBS. Adapted, with permission, from Shahid et al. 2011 151 and Shahid et al. 2012 152.
Figure 7. Figure 7. OX‐KO and OX neuron‐ablated (ORX‐AB) mice differentially respond to cold stress. Mice were exposed to a cold environment (5°C) for 4 h while the abdominal temperature was continuously monitored with a telemetric system. The thin lines indicate the data from an individual animal, and the thick lines are the mean ± SEM of orexin‐knockout mice (ORX‐KO), orexin neuron‐ablated mice (ORX‐AB), and their corresponding wild‐type littermates (WTKO and WTAB). Adapted, with permission, from Kuwaki 78.
Figure 8. Figure 8. Change of number of orexin neurons in the hypothalamus and orexin processes in the pons in SIDS and non‐SIDS cases. Box and whisker plot shows that total number of OXA and OXB immunoreactivity was decreased by up to 21% within the hypothalamus in SIDS versus non‐SIDS cases (A and B). In the pons, a 40% to 50% decrease in OXA in all pontine nuclei (C), similar OXB findings in the LC, LDT, DTg, and Pn (D) in SIDS cases compared to non‐SIDS cases. *p ≤ 0.05; **p ≤ 0.01; ***p ≤ 0.001. Adapted, with permission, from Hunt et al. 2015 66 with permission.


Figure 1. Schematic representation showing the broad anatomical and functional connections between orexin neurons and other neuronal systems. It summarizes how the orexin system may be critically linked with the cardiorespiratory and autonomic regulatory system and the vigilance‐state dependent behavioral regulatory system. Arrows indicate the excitatory connections, while circles indicate the inhibitory connections. AMG, amygdala; BAT, brown adipose tissue; BNST, bed nucleus of the stria terminalis; DR, dorsal raphe; LC, locus coeruleus; MLR, medullary locomotor region; MR, medullary raphe; NTS, nucleus tractus solitarius; PAG, periaqueductal gray; PBC, pre‐Bötzinger complex; PBN, parabrachial nucleus; PVN, paraventricular nucleus; RTN, retrotrapezoid nucleus; RVLM, rostral ventrolateral medulla where sympathetic cardiovascular premotor neurons are located; SCN, suprachiasmatic nucleus; TMN, tuberomammillary nucleus; VLPO, ventrolateral preoptic nucleus. Adapted, with permission, from Kuwaki, 2015 78.


Figure 2. Transgenic orexin deficient mice have lower mean arterial blood pressure and attenuated ventilatory response to CO2 or hypercapnic chemoreflex. (A) Ventilatory response to hypercapnia is severely attenuated in Orexin‐KO than wild‐type (WT) controls only in wakefulness. (*P < 0.05 compared with WT mice. P < 0.05 compared with the data during awake). (B) Resting blood pressure is significantly lower in orexin‐KO than WT controls in both light and dark diurnal cycles. ORX‐KO, orexin knockout mice; WT, wild‐type mice; SWS, slow‐wave sleep; REM, rapid‐eye movement sleep. Adapted, with permission, from Kayaba et al. 2013 71, and Kuwaki et al. 2010 79.


Figure 3. Spontaneously hypertensive rats have an excessive number of orexin‐producing neurons, high mean arterial blood pressure (MABP), and exaggerated ventilatory response to hypercapnia. (A) Distribution of OX‐ir neurons in three hypothalamic zones and whole hypothalamus in SHR (dark gray bar) and WKY (white bar) at young and adult age. (B) Resting MABP with (hatched bars) and without (black bars) treatment of a dual OXR antagonist, almorexant (Amxt), in young and adult SHRs versus that in age‐matched WKY rats (gray bars) in wakefulness, NREM, and REM sleep. (C) Ventilatory response (% change of V˙E) in normoxic hypercapnia with Amxt treatment (hatched bars) and without (black bars) in young and adult SHRs versus that in normotensive young and adult WKY rats (gray bars). DMH, dorsomedial hypothalamus; f, fornix; Hyp, hypothalamus (all three zones); LHA, lateral hypothalamic area; PeF, perifornical hypothalamus 94.


Figure 4. Orexins are involved in regulation of central chemoreception. Schematic sagittal brain section shows the location of orexin neurons and two brainstem putative central chemoreceptor sites, the RTN and medullary raphe. (E) Adapted, with permission, from Nambu et al. 115. Orexin neurons themselves are CO2/pH chemosensitive in brain slices of the hypothalamus of transgenic mice that express GFP only in orexin neurons. Demonstrates the effect of CO2‐induced low pHe on orexin neurons (A‐a). Effect of HEPES‐buffered low pHe on orexin neurons (A‐b). (a,b) Adapted, with permission, from Williams et al. 2007 172. Blocking OXRs with a dual OXR antagonist, Amxt, via oral administration significantly lowers the ventilatory response to hypercapnia only in wakefulness in the dark period (B). Focal inhibition of OX1R in the RTN (C) or medullary raphe (D) significantly attenuated CO2 chemoreflex. (B) Adapted, with permission, from Li and Nattie 2010 91; (C) Adapted, with permission, from Dias et al. 2009 36; (D) Adapted, with permission, from Dias et al. 2010 35. aCSF, artificial cerebrospinal fluid; RTN, retrotrapezoid nucleus.


Figure 5. The effects of OXA on hypoglossal motor (HMNs) neuronal activity and of eliminating orexin neurons on genioglossal muscle activity (GG‐EMG). Orexin‐A concentration‐dependently increased the firing rate of HMNs in brain slices of neonatal rats (A). Bilateral lesions of orexin neurons in the hypothalamus of adult rats via orexin‐B‐SAP (400 nL per side, 0.43 mg/mL) decreased the respiratory‐related GG‐EMG (B, lower panel) compared to control‐injected rats (B, top panel). Tonic activity of GG‐EMG is not affected while respiratory‐related activity is significantly attenuated post‐lesion (C). Adapted, with permission, from Zhang et al. 2014 181.


Figure 6. The effects of OX1R antagonist and OXB agonist on orexin‐induced increase in mean arterial pressure (MAP) and sympathetic nerve activity (SNA) in the RVLM (A) and spinal cord (B). OX1R antagonist, SB334867, can significantly attenuate the orexin A‐induced MAP and sSNA effects in the RVLM (A). OXA dose‐dependently increased MAP and sSNA and OX1R antagonism can block such OXA‐induced effects in the spinal cord (B). ***P < 0.001, **P < 0.01, *P < 0.05, significantly different from PBS. Adapted, with permission, from Shahid et al. 2011 151 and Shahid et al. 2012 152.


Figure 7. OX‐KO and OX neuron‐ablated (ORX‐AB) mice differentially respond to cold stress. Mice were exposed to a cold environment (5°C) for 4 h while the abdominal temperature was continuously monitored with a telemetric system. The thin lines indicate the data from an individual animal, and the thick lines are the mean ± SEM of orexin‐knockout mice (ORX‐KO), orexin neuron‐ablated mice (ORX‐AB), and their corresponding wild‐type littermates (WTKO and WTAB). Adapted, with permission, from Kuwaki 78.


Figure 8. Change of number of orexin neurons in the hypothalamus and orexin processes in the pons in SIDS and non‐SIDS cases. Box and whisker plot shows that total number of OXA and OXB immunoreactivity was decreased by up to 21% within the hypothalamus in SIDS versus non‐SIDS cases (A and B). In the pons, a 40% to 50% decrease in OXA in all pontine nuclei (C), similar OXB findings in the LC, LDT, DTg, and Pn (D) in SIDS cases compared to non‐SIDS cases. *p ≤ 0.05; **p ≤ 0.01; ***p ≤ 0.001. Adapted, with permission, from Hunt et al. 2015 66 with permission.
References
 1.Abbott SB, Kanbar R, Bochorishvili G, Coates MB, Stornetta RL, Guyenet PG. C1 neurons excite locus coeruleus and A5 noradrenergic neurons along with sympathetic outflow in rats. J Physiol 590: 2897‐2915, 2012. DOI: 10.1113/jphysiol.2012.232157.
 2.Akbari E, Motamedi F, Naghdi N, Noorbakhshnia M. The effect of antagonization of orexin 1 receptors in CA1 and dentate gyrus regions on memory processing in passive avoidance task. Behav Brain Res 187: 172‐177, 2008. DOI: 10.1016/j.bbr.2007.09.019.
 3.Akbari E, Naghdi N, Motamedi F. The selective orexin 1 receptor antagonist SB‐334867‐A impairs acquisition and consolidation but not retrieval of spatial memory in Morris water maze. Peptides 28: 650‐656, 2007. DOI: 10.1016/j.peptides.2006.11.002.
 4.Aksu K, Firat Guven S, Aksu F, Ciftci B, Ulukavak Ciftci T, Aksaray S, Sipit T, Peker Y. Obstructive sleep apnoea, cigarette smoking and plasma orexin‐A in a sleep clinic cohort. J Int Med Res 37: 331‐340, 2009. DOI: 10.1177/147323000903700207.
 5.Amin R, Simakajornboon N, Szczesniak R, Inge T. Early improvement in obstructive sleep apnea and increase in orexin levels after bariatric surgery in adolescents and young adults. Surg Obes Relat Dis 13: 95‐100, 2017. DOI: 10.1016/j.soard.2016.05.023.
 6.Antunes VR, Brailoiu GC, Kwok EH, Scruggs P, Dun NJ. Orexins/hypocretins excite rat sympathetic preganglionic neurons in vivo and in vitro. Am J Physiol Regul Integr Comp Physiol 281: R1801‐R1807, 2001.
 7.Arima Y, Yokota S, Fujitani M. Lateral parabrachial neurons innervate orexin neurons projecting to brainstem arousal areas in the rat. Sci Rep 9: 2830, 2019. DOI: 10.1038/s41598‐019‐39063‐y.
 8.Arnulf I, Konofal E, Merino‐Andreu M, Houeto JL, Mesnage V, Welter ML, Lacomblez L, Golmard JL, Derenne JP, Agid Y. Parkinson's disease and sleepiness: An integral part of PD. Neurology 58: 1019‐1024, 2002.
 9.Arnulf I, Merino‐Andreu M, Bloch F, Konofal E, Vidailhet M, Cochen V, Derenne JP, Agid Y. REM sleep behavior disorder and REM sleep without atonia in patients with progressive supranuclear palsy. Sleep 28: 349‐354, 2005.
 10.Badami VM, Rice CD, Lois JH, Madrecha J, Yates BJ. Distribution of hypothalamic neurons with orexin (hypocretin) or melanin concentrating hormone (MCH) immunoreactivity and multisynaptic connections with diaphragm motoneurons. Brain Res 1323: 119‐126, 2010. DOI: 10.1016/j.brainres.2010.02.002.
 11.Bochorishvili G, Nguyen T, Coates MB, Viar KE, Stornetta RL, Guyenet PG. The orexinergic neurons receive synaptic input from C1 cells in rats. J Comp Neurol 522: 3834‐3846, 2014. DOI: 10.1002/cne.23643.
 12.Brownell SE, Conti B. Age‐ and gender‐specific changes of hypocretin immunopositive neurons in C57Bl/6 mice. Neurosci Lett 472: 29‐32, 2010. DOI: 10.1016/j.neulet.2010.01.048.
 13.Brunnstrom HR, Englund EM. Cause of death in patients with dementia disorders. Eur J Neurol 16: 488‐492, 2009. DOI: 10.1111/j.1468‐1331.2008.02503.x.
 14.Burdakov D, Karnani MM, Gonzalez A. Lateral hypothalamus as a sensor‐regulator in respiratory and metabolic control. Physiol Behav 121: 117‐124, 2013. DOI: 10.1016/j.physbeh.2013.03.023.
 15.Busquets X, Barbe F, Barcelo A, de la Pena M, Sigritz N, Mayoralas LR, Ladaria A, Agusti A. Decreased plasma levels of orexin‐A in sleep apnea. Respiration 71: 575‐579, 2004. DOI: 10.1159/000081757.
 16.Carrive P, Orexin KT. Central modulation of cardiovascular and respiratory function. Curr Top Behav Neurosci 33: 157‐196, 2017. DOI: 10.1007/7854_2016_46.
 17.Cedernaes J, Osorio RS, Varga AW, Kam K, Schioth HB, Benedict C. Candidate mechanisms underlying the association between sleep‐wake disruptions and Alzheimer's disease. Sleep Med Rev 31: 102‐111, 2017. DOI: 10.1016/j.smrv.2016.02.002.
 18.Chaudhuri KR, Healy DG, Schapira AH. Non‐motor symptoms of Parkinson's disease: Diagnosis and management. Lancet Neurol 5: 235‐245, 2006. DOI: 10.1016/S1474‐4422(06)70373‐8.
 19.Chemelli RM, Willie JT, Sinton CM, Elmquist JK, Scammell T, Lee C, Richardson JA, Williams SC, Xiong Y, Kisanuki Y, Fitch TE, Nakazato M, Hammer RE, Saper CB, Yanagisawa M. Narcolepsy in orexin knockout mice: Molecular genetics of sleep regulation. Cell 98: 437‐451, 1999.
 20.Chen CT, Hwang LL, Chang JK, Dun NJ. Pressor effects of orexins injected intracisternally and to rostral ventrolateral medulla of anesthetized rats. Am J Physiol Regul Integr Comp Physiol 278: R692‐R697, 2000.
 21.Chen L, Thakkar MM, Winston S, Bolortuya Y, Basheer R, McCarley RW. REM sleep changes in rats induced by siRNA‐mediated orexin knockdown. Eur J Neurosci 24: 2039‐2048, 2006. DOI: 10.1111/j.1460‐9568.2006.05058.x.
 22.Chen W, Ye J, Han D, Yin G, Wang B, Zhang Y. Association of prepro‐orexin polymorphism with obstructive sleep apnea/hypopnea syndrome. Am J Otolaryngol 33: 31‐36, 2012. DOI: 10.1016/j.amjoto.2010.12.005.
 23.Ciriello J, Li Z, de Oliveira CV. Cardioacceleratory responses to hypocretin‐1 injections into rostral ventromedial medulla. Brain Res 991: 84‐95, 2003.
 24.Ciriello J, McMurray JC, Babic T, de Oliveira CV. Collateral axonal projections from hypothalamic hypocretin neurons to cardiovascular sites in nucleus ambiguus and nucleus tractus solitarius. Brain Res 991: 133‐141, 2003.
 25.Clifford L, Dampney BW, Carrive P. Spontaneously hypertensive rats have more orexin neurons in their medial hypothalamus than normotensive rats. Exp Physiol 100: 388‐398, 2015. DOI: 10.1113/expphysiol.2014.084137.
 26.Date Y, Ueta Y, Yamashita H, Yamaguchi H, Matsukura S, Kangawa K, Sakurai T, Yanagisawa M, Nakazato M. Orexins, orexigenic hypothalamic peptides, interact with autonomic, neuroendocrine and neuroregulatory systems. Proc Natl Acad Sci U S A 96: 748‐753, 1999.
 27.de Lecea L. Hypocretins and the neurobiology of sleep‐wake mechanisms. Prog Brain Res 198: 15‐24, 2012. DOI: 10.1016/B978‐0‐444‐59489‐1.00003‐3.
 28.de Lecea L, Kilduff TS, Peyron C, Gao X, Foye PE, Danielson PE, Fukuhara C, Battenberg EL, Gautvik VT, Bartlett FS II, Frankel WN, van den Pol AN, Bloom FE, Gautvik KM, Sutcliffe JG. The hypocretins: Hypothalamus‐specific peptides with neuroexcitatory activity. Proc Natl Acad Sci U S A 95: 322‐327, 1998.
 29.de Oliveira CV, Ciriello J. Cardiovascular responses to hypocretin‐1 in nucleus ambiguus of the ovariectomized female rat. Brain Res 986: 148‐156, 2003.
 30.de Oliveira CV, Rosas‐Arellano MP, Solano‐Flores LP, Ciriello J. Cardiovascular effects of hypocretin‐1 in nucleus of the solitary tract. Am J Physiol Heart Circ Physiol 284: H1369‐H1377, 2003. DOI: 10.1152/ajpheart.00877.2002.
 31.Deadwyler SA, Porrino L, Siegel JM, Hampson RE. Systemic and nasal delivery of orexin‐A (Hypocretin‐1) reduces the effects of sleep deprivation on cognitive performance in nonhuman primates. J Neurosci 27: 14239‐14247, 2007. DOI: 10.1523/JNEUROSCI.3878‐07.2007.
 32.Deng BS, Nakamura A, Zhang W, Yanagisawa M, Fukuda Y, Kuwaki T. Contribution of orexin in hypercapnic chemoreflex: Evidence from genetic and pharmacological disruption and supplementation studies in mice. J Appl Physiol (1985) 103: 1772‐1779, 2007. DOI: 10.1152/japplphysiol.00075.2007.
 33.Dergacheva O, Mendelowitz D. Combined hypoxia and hypercapnia, but not hypoxia alone, suppresses neurotransmission from orexin to hypothalamic paraventricular spinally‐projecting neurons in weanling rats. Brain Res 1679: 33‐38, 2018. DOI: 10.1016/j.brainres.2017.11.015.
 34.Desarnaud F, Murillo‐Rodriguez E, Lin L, Xu M, Gerashchenko D, Shiromani SN, Nishino S, Mignot E, Shiromani PJ. The diurnal rhythm of hypocretin in young and old F344 rats. Sleep 27: 851‐856, 2004.
 35.Dias MB, Li A, Nattie E. The orexin receptor 1 (OX1R) in the rostral medullary raphe contributes to the hypercapnic chemoreflex in wakefulness, during the active period of the diurnal cycle. Respir Physiol Neurobiol 170: 96‐102, 2010. DOI: 10.1016/j.resp.2009.12.002.
 36.Dias MB, Li A, Nattie EE. Antagonism of orexin receptor‐1 in the retrotrapezoid nucleus inhibits the ventilatory response to hypercapnia predominantly in wakefulness. J Physiol 587: 2059‐2067, 2009. DOI: 10.1113/jphysiol.2008.168260.
 37.Drouot X, Moutereau S, Nguyen JP, Lefaucheur JP, Creange A, Remy P, Goldenberg F, d'Ortho MP. Low levels of ventricular CSF orexin/hypocretin in advanced PD. Neurology 61: 540‐543, 2003.
 38.Du MK, Hunt NJ, Waters KA, Machaalani R. Cumulative effects of repetitive intermittent hypercapnic hypoxia on orexin in the developing piglet hypothalamus. Int J Dev Neurosci 48: 1‐8, 2016. DOI: 10.1016/j.ijdevneu.2015.10.007.
 39.Dunn S, Moushey A, Dong R, Li A. Postnatal developmental changes of orexin in spontaneously hypertensive rats. FASEB J 31: 866.865, 2017.
 40.Dutschmann M, Kron M, Morschel M, Gestreau C. Activation of Orexin B receptors in the pontine Kolliker‐Fuse nucleus modulates pre‐inspiratory hypoglossal motor activity in rat. Respir Physiol Neurobiol 159: 232‐235, 2007. DOI: 10.1016/j.resp.2007.06.004.
 41.Emamian F, Khazaie H, Tahmasian M, Leschziner GD, Morrell MJ, Hsiung GY, Rosenzweig I, Sepehry AA. The association between obstructive sleep apnea and Alzheimer's disease: A meta‐analysis perspective. Front Aging Neurosci 8: 78, 2016. DOI: 10.3389/fnagi.2016.00078.
 42.Follwell MJ, Ferguson AV. Cellular mechanisms of orexin actions on paraventricular nucleus neurones in rat hypothalamus. J Physiol 545: 855‐867, 2002.
 43.Fronczek R, Overeem S, Lee SY, Hegeman IM, van Pelt J, van Duinen SG, Lammers GJ, Swaab DF. Hypocretin (orexin) loss in Parkinson's disease. Brain 130: 1577‐1585, 2007. DOI: 10.1093/brain/awm090.
 44.Fronczek R, Overeem S, Lee SY, Hegeman IM, van Pelt J, van Duinen SG, Lammers GJ, Swaab DF. Hypocretin (orexin) loss and sleep disturbances in Parkinson's disease. Brain 131: e88, 2008. DOI: 10.1093/brain/awm222.
 45.Fronczek R, van Geest S, Frolich M, Overeem S, Roelandse FW, Lammers GJ, Swaab DF. Hypocretin (orexin) loss in Alzheimer's disease. Neurobiol Aging 33: 1642‐1650, 2012. DOI: 10.1016/j.neurobiolaging.2011.03.014.
 46.Fujiki N, Yoshida Y, Ripley B, Honda K, Mignot E, Nishino S. Changes in CSF hypocretin‐1 (orexin A) levels in rats across 24 hours and in response to food deprivation. Neuroreport 12: 993‐997, 2001.
 47.Fung SJ, Yamuy J, Sampogna S, Morales FR, Chase MH. Hypocretin (orexin) input to trigeminal and hypoglossal motoneurons in the cat: A double‐labeling immunohistochemical study. Brain Res 903: 257‐262, 2001.
 48.Gerashchenko D, Blanco‐Centurion C, Greco MA, Shiromani PJ. Effects of lateral hypothalamic lesion with the neurotoxin hypocretin‐2‐saporin on sleep in Long‐Evans rats. Neuroscience 116: 223‐235, 2003.
 49.Gerashchenko D, Murillo‐Rodriguez E, Lin L, Xu M, Hallett L, Nishino S, Mignot E, Shiromani PJ. Relationship between CSF hypocretin levels and hypocretin neuronal loss. Exp Neurol 184: 1010‐1016, 2003. DOI: 10.1016/S0014‐4886(03)00388‐1.
 50.Gestreau C, Bevengut M, Dutschmann M. The dual role of the orexin/hypocretin system in modulating wakefulness and respiratory drive. Curr Opin Pulm Med 14: 512‐518, 2008. DOI: 10.1097/MCP.0b013e32831311d3.
 51.Gonzalez JA, Jensen LT, Doyle SE, Miranda‐Anaya M, Menaker M, Fugger L, Bayliss DA, Burdakov D. Deletion of TASK1 and TASK3 channels disrupts intrinsic excitability but does not abolish glucose or pH responses of orexin/hypocretin neurons. Eur J Neurosci 30: 57‐64, 2009. DOI: 10.1111/j.1460‐9568.2009.06789.x.
 52.Gubellini P, Kachidian P. Animal models of Parkinson's disease: An updated overview. Rev Neurol (Paris) 171: 750‐761, 2015. DOI: 10.1016/j.neurol.2015.07.011.
 53.Guerreiro R, Bras J. The age factor in Alzheimer's disease. Genome Med 7: 106, 2015. DOI: 10.1186/s13073‐015‐0232‐5.
 54.Guerreiro S, Florence C, Rousseau E, Hamadat S, Hirsch EC, Michel PP. The sleep‐modulating peptide orexin‐B protects midbrain dopamine neurons from degeneration, alone or in cooperation with nicotine. Mol Pharmacol 87: 525‐532, 2015. DOI: 10.1124/mol.114.095703.
 55.Guyenet PG, Bayliss DA. Neural control of breathing and CO2 homeostasis. Neuron 87: 946‐961, 2015. DOI: 10.1016/j.neuron.2015.08.001.
 56.Hadadianpour Z, Fatehi F, Ayoobi F, Kaeidi A, Shamsizadeh A, Fatemi I. The effect of orexin‐A on motor and cognitive functions in a rat model of Parkinson's disease. Neurol Res 39: 845‐851, 2017. DOI: 10.1080/01616412.2017.1352185.
 57.Han F. Respiratory regulation in narcolepsy. Sleep Breath 16: 241‐245, 2012. DOI: 10.1007/s11325‐011‐0489‐x.
 58.Hara J, Beuckmann CT, Nambu T, Willie JT, Chemelli RM, Sinton CM, Sugiyama F, Yagami K, Goto K, Yanagisawa M, Sakurai T. Genetic ablation of orexin neurons in mice results in narcolepsy, hypophagia, and obesity. Neuron 30: 345‐354, 2001.
 59.Harrison TA, Chen CT, Dun NJ, Chang JK. Hypothalamic orexin A‐immunoreactive neurons project to the rat dorsal medulla. Neurosci Lett 273: 17‐20, 1999.
 60.Hervieu GJ, Cluderay JE, Harrison DC, Roberts JC, Leslie RA. Gene expression and protein distribution of the orexin‐1 receptor in the rat brain and spinal cord. Neuroscience 103: 777‐797, 2001.
 61.Hinrichsen CF, Maskrey M, Mortola JP. Ventilatory and metabolic responses to cold and hypoxia in conscious rats with discrete hypothalamic lesions. Respir Physiol 111: 247‐256, 1998.
 62.Huang SC, Dai YW, Lee YH, Chiou LC, Hwang LL. Orexins depolarize rostral ventrolateral medulla neurons and increase arterial pressure and heart rate in rats mainly via orexin 2 receptors. J Pharmacol Exp Ther 334: 522‐529, 2010. DOI: 10.1124/jpet.110.167791.
 63.Hughes TM, Lockhart SN, Smagula SF. Blood pressure's role in Alzheimer disease pathology. Am J Geriatr Psychiatry 26: 23‐24, 2018. DOI: 10.1016/j.jagp.2017.09.019.
 64.Hunt NJ, Rodriguez ML, Waters KA, Machaalani R. Changes in orexin (hypocretin) neuronal expression with normal aging in the human hypothalamus. Neurobiol Aging 36: 292‐300, 2015. DOI: 10.1016/j.neurobiolaging.2014.08.010.
 65.Hunt NJ, Waters KA, Machaalani R. Promotion of the unfolding protein response in orexin/dynorphin neurons in sudden infant death syndrome (SIDS): Elevated pPERK and ATF4 expression. Mol Neurobiol 54: 7171‐7185, 2017. DOI: 10.1007/s12035‐016‐0234‐3.
 66.Hunt NJ, Waters KA, Rodriguez ML, Machaalani R. Decreased orexin (hypocretin) immunoreactivity in the hypothalamus and pontine nuclei in sudden infant death syndrome. Acta Neuropathol 130: 185‐198, 2015. DOI: 10.1007/s00401‐015‐1437‐9.
 67.Iigaya K, Horiuchi J, McDowall LM, Lam AC, Sediqi Y, Polson JW, Carrive P, Dampney RA. Blockade of orexin receptors with Almorexant reduces cardiorespiratory responses evoked from the hypothalamus but not baro‐ or chemoreceptor reflex responses. Am J Physiol Regul Integr Comp Physiol 303: R1011‐R1022, 2012. DOI: 10.1152/ajpregu.00263.2012.
 68.Jackson KL, Dampney BW, Moretti JL, Stevenson ER, Davern PJ, Carrive P, Head GA. Contribution of orexin to the neurogenic hypertension in BPH/2J mice. Hypertension 67: 959‐969, 2016. DOI: 10.1161/HYPERTENSIONAHA.115.07053.
 69.Johnson PL, Truitt W, Fitz SD, Minick PE, Dietrich A, Sanghani S, Traskman‐Bendz L, Goddard AW, Brundin L, Shekhar A. A key role for orexin in panic anxiety. Nat Med 16: 111‐115, 2010. DOI: 10.1038/nm.2075.
 70.Kang JE, Lim MM, Bateman RJ, Lee JJ, Smyth LP, Cirrito JR, Fujiki N, Nishino S, Holtzman DM. Amyloid‐beta dynamics are regulated by orexin and the sleep‐wake cycle. Science 326: 1005‐1007, 2009. DOI: 10.1126/science.1180962.
 71.Kayaba Y, Nakamura A, Kasuya Y, Ohuchi T, Yanagisawa M, Komuro I, Fukuda Y, Kuwaki T. Attenuated defense response and low basal blood pressure in orexin knockout mice. Am J Physiol Regul Integr Comp Physiol 285: R581‐R593, 2003. DOI: 10.1152/ajpregu.00671.2002.
 72.Kessler BA, Stanley EM, Frederick‐Duus D, Fadel J. Age‐related loss of orexin/hypocretin neurons. Neuroscience 178: 82‐88, 2011. DOI: 10.1016/j.neuroscience.2011.01.031.
 73.Kiyashchenko LI, Mileykovskiy BY, Maidment N, Lam HA, Wu MF, John J, Peever J, Siegel JM. Release of hypocretin (orexin) during waking and sleep states. J Neurosci 22: 5282‐5286, 2002.
 74.Kukkonen JP, Holmqvist T, Ammoun S, Akerman KE. Functions of the orexinergic/hypocretinergic system. Am J Physiol Cell Physiol 283: C1567‐C1591, 2002. DOI: 10.1152/ajpcell.00055.2002.
 75.Kuo TB, Lai CJ, Shaw FZ, Lai CW, Yang CC. Sleep‐related sympathovagal imbalance in SHR. Am J Physiol Heart Circ Physiol 286: H1170‐H1176, 2004. DOI: 10.1152/ajpheart.00418.2003.
 76.Kuo TB, Shaw FZ, Lai CJ, Lai CW, Yang CC. Changes in sleep patterns in spontaneously hypertensive rats. Sleep 27: 406‐412, 2004.
 77.Kuo TB, Yang CC. Sleep‐related changes in cardiovascular neural regulation in spontaneously hypertensive rats. Circulation 112: 849‐854, 2005. DOI: 10.1161/CIRCULATIONAHA.104.503920.
 78.Kuwaki T. Thermoregulation under pressure: A role for orexin neurons. Temperature (Austin) 2: 379‐391, 2015. DOI: 10.1080/23328940.2015.1066921.
 79.Kuwaki T, Li A, Nattie E. State‐dependent central chemoreception: A role of orexin. Respir Physiol Neurobiol 173: 223‐229, 2010. DOI: 10.1016/j.resp.2010.02.006.
 80.Kuwaki T, Zhang W. Orexin neurons as arousal‐associated modulators of central cardiorespiratory regulation. Respir Physiol Neurobiol 174: 43‐54, 2010. DOI: 10.1016/j.resp.2010.04.018.
 81.Kuwaki T, Zhang W, Nakamura A, Deng BS. Emotional and state‐dependent modification of cardiorespiratory function: Role of orexinergic neurons. Auton Neurosci 142: 11‐16, 2008. DOI: 10.1016/j.autneu.2008.03.004.
 82.Langen B, Dost R. Comparison of SHR, WKY and Wistar rats in different behavioural animal models: Effect of dopamine D1 and alpha2 agonists. Atten Defic Hyperact Disord 3: 1‐12, 2011. DOI: 10.1007/s12402‐010‐0034‐y.
 83.Lavezzi AM, Ferrero S, Roncati L, Matturri L, Pusiol T. Impaired orexin receptor expression in the Kolliker‐Fuse nucleus in sudden infant death syndrome: Possible involvement of this nucleus in arousal pathophysiology. Neurol Res 38: 706‐716, 2016. DOI: 10.1080/01616412.2016.1201632.
 84.Lazarenko RM, Stornetta RL, Bayliss DA, Guyenet PG. Orexin A activates retrotrapezoid neurons in mice. Respir Physiol Neurobiol 175: 283‐287, 2011. DOI: 10.1016/j.resp.2010.12.003.
 85.Lee MG, Hassani OK, Jones BE. Discharge of identified orexin/hypocretin neurons across the sleep‐waking cycle. J Neurosci 25: 6716‐6720, 2005. DOI: 10.1523/JNEUROSCI.1887‐05.2005.
 86.Lee YH, Dai YW, Huang SC, Li TL, Hwang LL. Blockade of central orexin 2 receptors reduces arterial pressure in spontaneously hypertensive rats. Exp Physiol 98: 1145‐1155, 2013. DOI: 10.1113/expphysiol.2013.072298.
 87.Lee YH, Tsai MC, Li TL, Dai YW, Huang SC, Hwang LL. Spontaneously hypertensive rats have more orexin neurons in the hypothalamus and enhanced orexinergic input and orexin 2 receptor‐associated nitric oxide signalling in the rostral ventrolateral medulla. Exp Physiol 100: 993‐1007, 2015. DOI: 10.1113/EP085016.
 88.Lennon MJ, Makkar SR, Crawford JD, Sachdev PS. Midlife hypertension and Alzheimer's disease: A systematic review and meta‐analysis. J Alzheimers Dis, 2019. DOI: 10.3233/JAD‐190474.
 89.Li A, Hindmarch CC, Nattie EE, Paton JF. Antagonism of orexin receptors significantly lowers blood pressure in spontaneously hypertensive rats. J Physiol 591: 4237‐4248, 2013. DOI: 10.1113/jphysiol.2013.256271.
 90.Li A, Nattie E. CO2 dialysis in one chemoreceptor site, the RTN: Stimulus intensity and sensitivity in the awake rat. Respir Physiol Neurobiol 133: 11‐22, 2002.
 91.Li A, Nattie E. 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, 2010. DOI: 10.1113/jphysiol.2010.191288.
 92.Li A, Nattie E. Orexin, cardio‐respiratory function, and hypertension. Front Neurosci 8: 22, 2014. DOI: 10.3389/fnins.2014.00022.
 93.Li A, Randall M, Nattie EE. CO(2) microdialysis in retrotrapezoid nucleus of the rat increases breathing in wakefulness but not in sleep. J Appl Physiol 87: 910‐919, 1999.
 94.Li A, Roy SH, Nattie EE. An augmented CO2 chemoreflex and overactive orexin system are linked with hypertension in young and adult spontaneously hypertensive rats. J Physiol 594: 4967‐4980, 2016. DOI: 10.1113/JP272199.
 95.Li N, Li A, Nattie E. Focal microdialysis of CO(2) in the perifornical‐hypothalamic area increases ventilation during wakefulness but not NREM sleep. Respir Physiol Neurobiol 185: 349‐355, 2013. DOI: 10.1016/j.resp.2012.09.007.
 96.Li SB, Jones JR, de Lecea L. Hypocretins, neural systems, physiology, and psychiatric disorders. Curr Psychiatry Rep 18: 7, 2016. DOI: 10.1007/s11920‐015‐0639‐0.
 97.Liguori C, Romigi A, Nuccetelli M, Zannino S, Sancesario G, Martorana A, Albanese M, Mercuri NB, Izzi F, Bernardini S, Nitti A, Sancesario GM, Sica F, Marciani MG, Placidi F. Orexinergic system dysregulation, sleep impairment, and cognitive decline in Alzheimer disease. JAMA Neurol 71: 1498‐1505, 2014. DOI: 10.1001/jamaneurol.2014.2510.
 98.Lin L, Faraco J, Li R, Kadotani H, Rogers W, Lin X, Qiu X, de Jong PJ, Nishino S, Mignot E. The sleep disorder canine narcolepsy is caused by a mutation in the hypocretin (orexin) receptor 2 gene. Cell 98: 365‐376, 1999.
 99.Lingueglia E. Acid‐sensing ion channels in sensory perception. J Biol Chem 282: 17325‐17329, 2007. DOI: 10.1074/jbc.R700011200.
 100.Liu MF, Xue Y, Liu C, Liu YH, Diao HL, Wang Y, Pan YP, Chen L. Orexin‐A exerts neuroprotective effects via OX1R in Parkinson's disease. Front Neurosci 12: 835, 2018. DOI: 10.3389/fnins.2018.00835.
 101.Llewellyn‐Smith IJ, Martin CL, Marcus JN, Yanagisawa M, Minson JB, Scammell TE. Orexin‐immunoreactive inputs to rat sympathetic preganglionic neurons. Neurosci Lett 351: 115‐119, 2003.
 102.Louis GW, Leinninger GM, Rhodes CJ, Myers MG Jr. Direct innervation and modulation of orexin neurons by lateral hypothalamic LepRb neurons. J Neurosci 30: 11278‐11287, 2010. DOI: 10.1523/JNEUROSCI.1340‐10.2010.
 103.Luo G, Ambati A, Lin L, Bonvalet M, Partinen M, Ji X, Maecker HT, Mignot EJ. Autoimmunity to hypocretin and molecular mimicry to flu in type 1 narcolepsy. Proc Natl Acad Sci U S A 115: E12323‐E12332, 2018. DOI: 10.1073/pnas.1818150116.
 104.Luong LN, Carrive P. Orexin microinjection in the medullary raphe increases heart rate and arterial pressure but does not reduce tail skin blood flow in the awake rat. Neuroscience 202: 209‐217, 2012. DOI: 10.1016/j.neuroscience.2011.11.073.
 105.Machado BH, Bonagamba LG, Dun SL, Kwok EH, Dun NJ. Pressor response to microinjection of orexin/hypocretin into rostral ventrolateral medulla of awake rats. Regul Pept 104: 75‐81, 2002. DOI: 10.1016/S0167‐0115(01)00351‐2.
 106.Maeda T, Nagata K, Kondo H, Kanbayashi T. Parkinson's disease comorbid with narcolepsy presenting low CSF hypocretin/orexin level. Sleep Med 7: 662, 2006. DOI: 10.1016/j.sleep.2006.05.017.
 107.Mahoney CE, Cogswell A, Koralnik IJ, Scammell TE. The neurobiological basis of narcolepsy. Nat Rev Neurosci 20: 83‐93, 2019. DOI: 10.1038/s41583‐018‐0097‐x.
 108.Marcus JN, Aschkenasi CJ, Lee CE, Chemelli RM, Saper CB, Yanagisawa M, Elmquist JK. Differential expression of orexin receptors 1 and 2 in the rat brain. J Comp Neurol 435: 6‐25, 2001.
 109.Maskrey M, Hinrichsen CF. Respiratory responses to combined hypoxia and hypothermia in rats after posterior hypothalamic lesions. Pflugers Arch 426: 371‐377, 1994.
 110.Matsumura K, Tsuchihashi T, Abe I. Central orexin‐A augments sympathoadrenal outflow in conscious rabbits. Hypertension 37: 1382‐1387, 2001.
 111.Matthews KA, Xu W, Gaglioti AH, Holt JB, Croft JB, Mack D, McGuire LC. Racial and ethnic estimates of Alzheimer's disease and related dementias in the United States (2015‐2060) in adults aged >/=65 years. Alzheimers Dement 15: 17‐24, 2019. DOI: 10.1016/j.jalz.2018.06.3063.
 112.Mieda M. The roles of orexins in sleep/wake regulation. Neurosci Res 118: 56‐65, 2017. DOI: 10.1016/j.neures.2017.03.015.
 113.Mileykovskiy BY, Kiyashchenko LI, Siegel JM. Behavioral correlates of activity in identified hypocretin/orexin neurons. Neuron 46: 787‐798, 2005. DOI: 10.1016/j.neuron.2005.04.035.
 114.Nakamura A, Zhang W, Yanagisawa M, Fukuda Y, Kuwaki T. Vigilance state‐dependent attenuation of hypercapnic chemoreflex and exaggerated sleep apnea in orexin knockout mice. J Appl Physiol 102: 241‐248, 2007. DOI: 10.1152/japplphysiol.00679.2006.
 115.Nambu T, Sakurai T, Mizukami K, Hosoya Y, Yanagisawa M, Goto K. Distribution of orexin neurons in the adult rat brain. Brain Res 827: 243‐260, 1999.
 116.Nattie E, Li A. Central chemoreception is a complex system function that involves multiple brain stem sites. J Appl Physiol (1985) 106: 1464‐1466, 2009. DOI: 10.1152/japplphysiol.00112.2008.
 117.Nattie E, Li A. Central chemoreception in wakefulness and sleep: Evidence for a distributed network and a role for orexin. J Appl Physiol (1985) 108: 1417‐1424, 2010. DOI: 10.1152/japplphysiol.01261.2009.
 118.Nattie E, Li A. Central chemoreceptors: Locations and functions. Compr Physiol 2: 221‐254, 2012. DOI: 10.1002/cphy.c100083.
 119.Nattie E, Li A. Respiration and autonomic regulation and orexin. Prog Brain Res 198: 25‐46, 2012. DOI: 10.1016/B978‐0‐444‐59489‐1.00004‐5.
 120.Nattie EE, Li A. CO2 dialysis in the medullary raphe of the rat increases ventilation in sleep. J Appl Physiol (1985) 90: 1247‐1257, 2001. DOI: 10.1152/jappl.2001.90.4.1247.
 121.Nattie EE, Li A. CO2 dialysis in nucleus tractus solitarius region of rat increases ventilation in sleep and wakefulness. J Appl Physiol (1985) 92: 2119‐2130, 2002. DOI: 10.1152/japplphysiol.01128.2001.
 122.Nishijima T, Sakurai S, Arihara Z, Takahashi K. Plasma orexin‐A‐like immunoreactivity in patients with sleep apnea hypopnea syndrome. Peptides 24: 407‐411, 2003.
 123.Nishino S, Ripley B, Overeem S, Lammers GJ, Mignot E. Hypocretin (orexin) deficiency in human narcolepsy. Lancet 355: 39‐40, 2000. DOI: 10.1016/S0140‐6736(99)05582‐8.
 124.Nixon JP, Mavanji V, Butterick TA, Billington CJ, Kotz CM, Teske JA. Sleep disorders, obesity, and aging: The role of orexin. Ageing Res Rev 20: 63‐73, 2015. DOI: 10.1016/j.arr.2014.11.001.
 125.Nixon JP, Smale L. A comparative analysis of the distribution of immunoreactive orexin A and B in the brains of nocturnal and diurnal rodents. Behav Brain Funct 3: 28, 2007. DOI: 10.1186/1744‐9081‐3‐28.
 126.Oliveira LM, Falquetto B, Moreira TS, Takakura AC. Orexinergic neurons are involved in the chemosensory control of breathing during the dark phase in a Parkinson's disease model. Exp Neurol 309: 107‐118, 2018. DOI: 10.1016/j.expneurol.2018.08.004.
 127.Orexin CP. Stress and central cardiovascular control. A link with hypertension? Neurosci Biobehav Rev 74: 376‐392, 2017. DOI: 10.1016/j.neubiorev.2016.06.044.
 128.Overeem S, Scammell TE, Lammers GJ. Hypocretin/orexin and sleep: Implications for the pathophysiology and diagnosis of narcolepsy. Curr Opin Neurol 15: 739‐745, 2002. DOI: 10.1097/01.wco.0000044800.53746.5a.
 129.Peever JH, Lai YY, Siegel JM. Excitatory effects of hypocretin‐1 (orexin‐A) in the trigeminal motor nucleus are reversed by NMDA antagonism. J Neurophysiol 89: 2591‐2600, 2003. DOI: 10.1152/jn.00968.2002.
 130.Peppard PE, Young T, Barnet JH, Palta M, Hagen EW, Hla KM. Increased prevalence of sleep‐disordered breathing in adults. Am J Epidemiol 177: 1006‐1014, 2013. DOI: 10.1093/aje/kws342.
 131.Peyron C, Faraco J, Rogers W, Ripley B, Overeem S, Charnay Y, Nevsimalova S, Aldrich M, Reynolds D, Albin R, Li R, Hungs M, Pedrazzoli M, Padigaru M, Kucherlapati M, Fan J, Maki R, Lammers GJ, Bouras C, Kucherlapati R, Nishino S, Mignot E. 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, 2000. DOI: 10.1038/79690.
 132.Polsek D, Gildeh N, Cash D, Winsky‐Sommerer R, Williams SCR, Turkheimer F, Leschziner GD, Morrell MJ, Rosenzweig I. Obstructive sleep apnoea and Alzheimer's disease: In search of shared pathomechanisms. Neurosci Biobehav Rev 86: 142‐149, 2018. DOI: 10.1016/j.neubiorev.2017.12.004.
 133.Puskas N, Papp RS, Gallatz K, Palkovits M. Interactions between orexin‐immunoreactive fibers and adrenaline or noradrenaline‐expressing neurons of the lower brainstem in rats and mice. Peptides 31: 1589‐1597, 2010. DOI: 10.1016/j.peptides.2010.04.020.
 134.Redgate ES, Gellhorn E. Respiratory activity and the hypothalamus. Am J Physiol 193: 189‐194, 1958. DOI: 10.1152/ajplegacy.1958.193.1.189.
 135.Ripley B, Overeem S, Fujiki N, Nevsimalova S, Uchino M, Yesavage J, Di Monte D, Dohi K, Melberg A, Lammers GJ, Nishida Y, Roelandse FW, Hungs M, Mignot E, Nishino S. CSF hypocretin/orexin levels in narcolepsy and other neurological conditions. Neurology 57: 2253‐2258, 2001.
 136.Roh JH, Jiang H, Finn MB, Stewart FR, Mahan TE, Cirrito JR, Heda A, Snider BJ, Li M, Yanagisawa M, de Lecea L, Holtzman DM. Potential role of orexin and sleep modulation in the pathogenesis of Alzheimer's disease. J Exp Med 211: 2487‐2496, 2014. DOI: 10.1084/jem.20141788.
 137.Rosin DL, Chang DA, Guyenet PG. Afferent and efferent connections of the rat retrotrapezoid nucleus. J Comp Neurol 499: 64‐89, 2006. DOI: 10.1002/cne.21105.
 138.Rye DB. Parkinson's disease and RLS: The dopaminergic bridge. Sleep Med 5: 317‐328, 2004. DOI: 10.1016/j.sleep.2004.01.016.
 139.Sagvolden T, Johansen EB. Rat models of ADHD. Curr Top Behav Neurosci 9: 301‐315, 2012. DOI: 10.1007/7854_2011_126.
 140.Saito YC, Tsujino N, Abe M, Yamazaki M, Sakimura K, Sakurai T. Serotonergic input to orexin neurons plays a role in maintaining wakefulness and REM sleep architecture. Front Neurosci 12: 892, 2018. DOI: 10.3389/fnins.2018.00892.
 141.Sakurai S, Nishijima T, Takahashi S, Yamauchi K, Arihara Z, Takahashi K. Low plasma orexin‐A levels were improved by continuous positive airway pressure treatment in patients with severe obstructive sleep apnea‐hypopnea syndrome. Chest 127: 731‐737, 2005. DOI: 10.1378/chest.127.3.731.
 142.Sakurai T. The neural circuit of orexin (hypocretin): Maintaining sleep and wakefulness. Nat Rev Neurosci 8: 171‐181, 2007. DOI: 10.1038/nrn2092.
 143.Sakurai T. Orexin deficiency and narcolepsy. Curr Opin Neurobiol 23: 760‐766, 2013. DOI: 10.1016/j.conb.2013.04.007.
 144.Sakurai T, Amemiya A, Ishii M, Matsuzaki I, Chemelli RM, Tanaka H, Williams SC, Richardson JA, Kozlowski GP, Wilson S, Arch JR, Buckingham RE, Haynes AC, Carr SA, Annan RS, McNulty DE, Liu WS, Terrett JA, Elshourbagy NA, Bergsma DJ, Yanagisawa M. Orexins and orexin receptors: A family of hypothalamic neuropeptides and G protein‐coupled receptors that regulate feeding behavior. Cell 92: 573‐585, 1998.
 145.Sakurai T, Mieda M, Tsujino N. The orexin system: Roles in sleep/wake regulation. Ann N Y Acad Sci 1200: 149‐161, 2010. DOI: 10.1111/j.1749‐6632.2010.05513.x.
 146.Sakurai T, Nagata R, Yamanaka A, Kawamura H, Tsujino N, Muraki Y, Kageyama H, Kunita S, Takahashi S, Goto K, Koyama Y, Shioda S, Yanagisawa M. Input of orexin/hypocretin neurons revealed by a genetically encoded tracer in mice. Neuron 46: 297‐308, 2005. DOI: 10.1016/j.neuron.2005.03.010.
 147.Samson WK, Gosnell B, Chang JK, Resch ZT, Murphy TC. Cardiovascular regulatory actions of the hypocretins in brain. Brain Res 831: 248‐253, 1999.
 148.Sansa G, Iranzo A, Santamaria J. Obstructive sleep apnea in narcolepsy. Sleep Med 11: 93‐95, 2010. DOI: 10.1016/j.sleep.2009.02.009.
 149.Sargin D. The role of the orexin system in stress response. Neuropharmacology, 2018. DOI: 10.1016/j.neuropharm.2018.09.034.
 150.Schwarting RK, Huston JP. The unilateral 6‐hydroxydopamine lesion model in behavioral brain research. Analysis of functional deficits, recovery and treatments. Prog Neurobiol 50: 275‐331, 1996.
 151.Shahid IZ, Rahman AA, Pilowsky PM. Intrathecal orexin A increases sympathetic outflow and respiratory drive, enhances baroreflex sensitivity and blocks the somato‐sympathetic reflex. Br J Pharmacol 162: 961‐973, 2011. DOI: 10.1111/j.1476‐5381.2010.01102.x.
 152.Shahid IZ, Rahman AA, Pilowsky PM. Orexin A in rat rostral ventrolateral medulla is pressor, sympatho‐excitatory, increases barosensitivity and attenuates the somato‐sympathetic reflex. Br J Pharmacol 165: 2292‐2303, 2012. DOI: 10.1111/j.1476‐5381.2011.01694.x.
 153.Shirasaka T, Kunitake T, Takasaki M, Kannan H. Neuronal effects of orexins: Relevant to sympathetic and cardiovascular functions. Regul Pept 104: 91‐95, 2002. DOI: 10.1016/S0167‐0115(01)00352‐4.
 154.Shirasaka T, Miyahara S, Kunitake T, Jin QH, Kato K, Takasaki M, Kannan H. Orexin depolarizes rat hypothalamic paraventricular nucleus neurons. Am J Physiol Regul Integr Comp Physiol 281: R1114‐R1118, 2001.
 155.Shirasaka T, Nakazato M, Matsukura S, Takasaki M, Kannan H. Sympathetic and cardiovascular actions of orexins in conscious rats. Am J Physiol 277: R1780‐R1785, 1999.
 156.Smith PM, Connolly BC, Ferguson AV. Microinjection of orexin into the rat nucleus tractus solitarius causes increases in blood pressure. Brain Res 950: 261‐267, 2002.
 157.Song N, Zhang G, Geng W, Liu Z, Jin W, Li L, Cao Y, Zhu D, Yu J, Shen L. Acid sensing ion channel 1 in lateral hypothalamus contributes to breathing control. PLoS One 7: e39982, 2012. DOI: 10.1371/journal.pone.0039982.
 158.Sunanaga J, Deng BS, Zhang W, Kanmura Y, Kuwaki T. CO2 activates orexin‐containing neurons in mice. Respir Physiol Neurobiol 166: 184‐186, 2009. DOI: 10.1016/j.resp.2009.03.006.
 159.Tabuchi S, Tsunematsu T, Black SW, Tominaga M, Maruyama M, Takagi K, Minokoshi Y, Sakurai T, Kilduff TS, Yamanaka A. Conditional ablation of orexin/hypocretin neurons: A new mouse model for the study of narcolepsy and orexin system function. J Neurosci 34: 6495‐6509, 2014. DOI: 10.1523/JNEUROSCI.0073‐14.2014.
 160.Takahashi Y, Zhang W, Sameshima K, Kuroki C, Matsumoto A, Sunanaga J, Kono Y, Sakurai T, Kanmura Y, Kuwaki T. Orexin neurons are indispensable for prostaglandin E2‐induced fever and defence against environmental cooling in mice. J Physiol 591: 5623‐5643, 2013. DOI: 10.1113/jphysiol.2013.261271.
 161.Tarasiuk A, Levi A, Assadi MH, Troib A, Segev Y. Orexin plays a role in growth impediment induced by obstructive sleep breathing in rats. Sleep 39: 887‐897, 2016. DOI: 10.5665/sleep.5648.
 162.Tarasiuk A, Levi A, Berdugo‐Boura N, Yahalom A, Segev Y. Role of orexin in respiratory and sleep homeostasis during upper airway obstruction in rats. Sleep 37: 987‐998, 2014. DOI: 10.5665/sleep.3676.
 163.Teran FA, Massey CA, Richerson GB. Serotonin neurons and central respiratory chemoreception: Where are we now? Prog Brain Res 209: 207‐233, 2014. DOI: 10.1016/B978‐0‐444‐63274‐6.00011‐4.
 164.Thannickal TC, Lai YY, Siegel JM. Hypocretin (orexin) cell loss in Parkinson's disease. Brain 130: 1586‐1595, 2007. DOI: 10.1093/brain/awm097.
 165.Thannickal TC, Moore RY, Nienhuis R, Ramanathan L, Gulyani S, Aldrich M, Cornford M, Siegel JM. Reduced number of hypocretin neurons in human narcolepsy. Neuron 27: 469‐474, 2000.
 166.Tupone D, Madden CJ, Cano G, Morrison SF. An orexinergic projection from perifornical hypothalamus to raphe pallidus increases rat brown adipose tissue thermogenesis. J Neurosci 31: 15944‐15955, 2011. DOI: 10.1523/JNEUROSCI.3909‐11.2011.
 167.Waldrop TG. Posterior hypothalamic modulation of the respiratory response to CO2 in cats. Pflugers Arch 418: 7‐13, 1991.
 168.Waldrop TG, Mullins DC, Millhorn DE. Control of respiration by the hypothalamus and by feedback from contracting muscles in cats. Respir Physiol 64: 317‐328, 1986.
 169.Wang W, Li Q, Pan Y, Zhu D, Wang L. Influence of hypercapnia on the synthesis of neuropeptides and their receptors in murine brain. Respirology 18: 102‐107, 2013. DOI: 10.1111/j.1440‐1843.2012.02245.x.
 170.Waters KA, Hunt NJ, Machaalani R. Neuropathology of sudden infant death syndrome: Hypothalamus. In: Duncan JR, Byard RW, editors. SIDS Sudden Infant and Early Childhood Death: The Past, the Present and the Future. Adelaide (AU): University of Adelaide Press, 2018.
 171.Wienecke M, Werth E, Poryazova R, Baumann‐Vogel H, Bassetti CL, Weller M, Waldvogel D, Storch A, Baumann CR. Progressive dopamine and hypocretin deficiencies in Parkinson's disease: Is there an impact on sleep and wakefulness? J Sleep Res 21: 710‐717, 2012. DOI: 10.1111/j.1365‐2869.2012.01027.x.
 172.Williams RH, Jensen LT, Verkhratsky A, Fugger L, Burdakov D. Control of hypothalamic orexin neurons by acid and CO2. Proc Natl Acad Sci U S A 104: 10685‐10690, 2007. DOI: 10.1073/pnas.0702676104.
 173.Willie JT, Chemelli RM, Sinton CM, Tokita S, Williams SC, Kisanuki YY, Marcus JN, Lee C, Elmquist JK, Kohlmeier KA, Leonard CS, Richardson JA, Hammer RE, Yanagisawa M. 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, 2003.
 174.Xiao F, Jiang M, Du D, Xia C, Wang J, Cao Y, Shen L, Zhu D. Orexin A regulates cardiovascular responses in stress‐induced hypertensive rats. Neuropharmacology 67: 16‐24, 2013. DOI: 10.1016/j.neuropharm.2012.10.021.
 175.Yang B, Ferguson AV. Orexin‐A depolarizes nucleus tractus solitarius neurons through effects on nonselective cationic and K+ conductances. J Neurophysiol 89: 2167‐2175, 2003. DOI: 10.1152/jn.01088.2002.
 176.Yang B, Samson WK, Ferguson AV. Excitatory effects of orexin‐A on nucleus tractus solitarius neurons are mediated by phospholipase C and protein kinase C. J Neurosci 23: 6215‐6222, 2003.
 177.Yokota S, Oka T, Asano H, Yasui Y. Orexinergic fibers are in contact with Kolliker‐Fuse nucleus neurons projecting to the respiration‐related nuclei in the medulla oblongata and spinal cord of the rat. Brain Res 1648: 512‐523, 2016. DOI: 10.1016/j.brainres.2016.08.020.
 178.Yoshida K, McCormack S, Espana RA, Crocker A, Scammell TE. Afferents to the orexin neurons of the rat brain. J Comp Neurol 494: 845‐861, 2006. DOI: 10.1002/cne.20859.
 179.Yoshida Y, Fujiki N, Nakajima T, Ripley B, Matsumura H, Yoneda H, Mignot E, Nishino S. Fluctuation of extracellular hypocretin‐1 (orexin A) levels in the rat in relation to the light‐dark cycle and sleep‐wake activities. Eur J Neurosci 14: 1075‐1081, 2001.
 180.Young JK, Wu M, Manaye KF, Kc P, Allard JS, Mack SO, Haxhiu MA. Orexin stimulates breathing via medullary and spinal pathways. J Appl Physiol 98: 1387‐1395, 2005. DOI: 10.1152/japplphysiol.00914.2004.
 181.Zhang GH, Liu ZL, Zhang BJ, Geng WY, Song NN, Zhou W, Cao YX, Li SQ, Huang ZL, Shen LL. Orexin A activates hypoglossal motoneurons and enhances genioglossus muscle activity in rats. Br J Pharmacol 171: 4233‐4246, 2014. DOI: 10.1111/bph.12784.
 182.Zhang W, Fukuda Y, Kuwaki T. Respiratory and cardiovascular actions of orexin‐A in mice. Neurosci Lett 385: 131‐136, 2005. DOI: 10.1016/j.neulet.2005.05.032.
 183.Zhang W, Sakurai T, Fukuda Y, Kuwaki T. Orexin neuron‐mediated skeletal muscle vasodilation and shift of baroreflex during defense response in mice. Am J Physiol Regul Integr Comp Physiol 290: R1654‐R1663, 2006. DOI: 10.1152/ajpregu.00704.2005.
 184.Zhang W, Shimoyama M, Fukuda Y, Kuwaki T. Multiple components of the defense response depend on orexin: Evidence from orexin knockout mice and orexin neuron‐ablated mice. Auton Neurosci 126–127: 139‐145, 2006. DOI: 10.1016/j.autneu.2006.02.021.
 185.Zhang W, Sunanaga J, Takahashi Y, Mori T, Sakurai T, Kanmura Y, Kuwaki T. Orexin neurons are indispensable for stress‐induced thermogenesis in mice. J Physiol 588: 4117‐4129, 2010. DOI: 10.1113/jphysiol.2010.195099.
 186.Zheng H, Patterson LM, Berthoud HR. Orexin‐A projections to the caudal medulla and orexin‐induced c‐Fos expression, food intake, and autonomic function. J Comp Neurol 485: 127‐142, 2005. DOI: 10.1002/cne.20515.
 187.Zhu Y, Fenik P, Zhan G, Xin R, Veasey SC. Degeneration in arousal neurons in chronic sleep disruption modeling sleep apnea. Front Neurol 6: 109, 2015. DOI: 10.3389/fneur.2015.00109.

Teaching Material

Savannah Barnett and Aihua Li. Orexin in Respiratory and Autonomic Regulation, Health and Diseases. Compr Physiol 10 : 2020, 345-363.

Didactic Synopsis

Major Teaching Points:

1. Understanding the hypothalamic neuropeptides, e.g., orexins, is necessary to master the underline mechanism of vigilance state-dependent control of cardiorespiratory and autonomic function and orexin-associated diseases.

2. Orexins play important roles in regulation cardio-respiratory and autonomic functions in different vigilance state.

    a) Many neurons in the cardio-respiratory and sleep-wake control centers are innervated by orexin neurons and express orexin receptors.
    b) Orexins excite cardio-respiratory and sympathetic activity, promote wakefulness and maintain upper airway patency.

3. Animals with orexin deficiency have hypotension, attenuated hypercapnic chemoreflex and fight-or-flight stress response, while animals with too much orexin, e.g., spontaneous hypertensive rats, have exaggerated CO2 chemoreflex, hyperactive and hypertension.

4. Dysfunctional orexin system are associated with many neurogenic disorders, including

    a) Sleep disorders, e.g., narcolepsy and obstructive sleep apnea
    b) Neurodegenerative diseases, e.g., Parkinson's disease and Alzheimer's disease
    c) Neurogenic hypertension
    d) Sudden infant death syndrome

Didactic Legends

The following legends to the figures that appear throughout the article are written to be useful for teaching.

Figure 1. Teaching point: Orexin neurons are anatomical and functional connected with many other neuronal systems e.g., cardio-respiratory and autonomic regulatory system and the vigilance-state dependent behavioral regulatory system.

Figure 2. Teaching point: Orexin deficiency leads to a severely attenuated ventilatory response to hypercapnia or hypercapnic chemoreflex during wakefulness, and lower resting blood pressure in both light and dark diurnal cycles in transgenic orexin knockout mice.

Figure 3. Teaching point: An overactive orexin system has been linked to neurogenic hypertension. Spontaneously hypertensive rats, the most commonly used animal model for human essential hypertension, have excess orexin neurons from young. Treating with a dual orexin receptor blocker can significantly lower higher blood pressure and exaggerated hypercapnic chemoreflex in these hypertensive rats.

Figure 4. Teaching point: Orexin is importantly involved in CO2 chemoreception or hypercapnic chemoreflex. Orexin neurons in the hypothalamus are pH/CO2 chemosensitive, and block orexin receptors with orexin receptor antagonist focally in two chemosensitive sites or systemically can significantly attenuate CO2 chemoreflex.

Figure 5. Teaching point: Orexin signaling is important in the control of airway patency, and decreased orexin inputs may have pathological impacts on the airway reflexes.

Figure 6. Teaching point: Administration of orexin can dose-dependently increase sympathetic activity and blood pressure, and administration of orexin receptor antagonist can block such orexin-induced excitatory effects.

Figure 7. Teaching point: The role of orexin in thermoregulation is complex, that loss of orexin producing neurons can significantly attenuate cold-stress response.

 

Figure 8. Teaching point: Dysfunctional orexin system is also linked to SIDS, and the total number of orexin neurons was significantly lower within the hypothalamus in SIDS cases (up to ~ 21%) compare to non-SIDS cases. 


Related Articles:

Teaching Material

Contact Editor

Submit a note to the editor about this article by filling in the form below.

* Required Field

How to Cite

Savannah Barnett, Aihua Li. Orexin in Respiratory and Autonomic Regulation, Health and Diseases. Compr Physiol 2020, 10: 345-363. doi: 10.1002/cphy.c190013