Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2016 Mar 1.
Published in final edited form as: Ageing Res Rev. 2014 Nov 22;20:63–73. doi: 10.1016/j.arr.2014.11.001

Sleep disorders, obesity, and aging: the role of orexin

Joshua P Nixon a,d,e, Vijayakumar Mavanji a, Tammy A Butterick a,e, Charles J Billington a,c,d,e, Catherine M Kotz a,b,d,e, Jennifer A Teske a,d,e,f
PMCID: PMC4467809  NIHMSID: NIHMS644546  PMID: 25462194

Abstract

The hypothalamic neuropeptides orexin A and B (hypocretin 1 and 2) are important homeostatic mediators of central control of energy metabolism and maintenance of sleep/wake states. Dysregulation or loss of orexin signaling has been linked to narcolepsy, obesity, and age-related disorders. In this review, we present an overview of our current understanding of orexin function, focusing on sleep disorders, energy balance, and aging, in both rodents and humans. We first discuss animal models used in studies of obesity and sleep, including loss of function using transgenic or viral-mediated approaches, gain of function models using exogenous delivery of orexin receptor agonist, and naturally-occurring models in which orexin responsiveness varies by individual. We next explore rodent models of orexin in aging, presenting evidence that orexin loss contributes to age-related changes in sleep and energy balance. In the next section, we focus on clinical importance of orexin in human obesity, sleep, and aging. We include discussion of orexin loss in narcolepsy and potential importance of orexin in insomnia, correlations between animal and human studies of age-related decline, and evidence for orexin involvement in age-related changes in cognitive performance. Finally, we present a summary of recent studies of orexin in neurodegenerative disease. We conclude that orexin acts as an integrative homeostatic signal influencing numerous brain regions, and that this pivotal role results in potential dysregulation of multiple physiological processes when orexin signaling is disrupted or lost.

Keywords: orexin, hypocretin, obesity, sleep, aging, energy balance

1. INTRODUCTION

Identified by two independent groups, the endogenous neuropeptides, orexin A and B (also known as hypocretin 1 and 2), and their associated G-protein coupled orexin type 1 and 2 receptors (OX1R and OX2R, respectively, also known as hypocretin receptor type 1 and 2), constitute the multi-functional central orexin system (de Lecea et al., 1998; Sakurai et al., 1998). Orexin synthesis is relatively confined to neurons in the lateral-posterior-perifornical hypothalamus, while orexin receptors are widely distributed in a brain site-specific manner (Marcus et al., 2001; Trivedi et al., 1998). Unlike orexin synthesizing neurons, orexin fibers are ubiquitous, extensively innervating peripheral and central targets (Date et al., 1999; España et al., 2005; Nixon and Smale, 2007; Peyron et al., 1998). Due to the extensive terminal field, central orexin signaling is well positioned to integrate and orchestrate multiple physiological processes such as arousal, whole-body energy metabolism, reward seeking, autonomic function, and ventilatory control (Burdakov et al., 2013; de Lecea and Huerta, 2014; Karnani and Burdakov, 2011; Kotz et al., 2012; Mahler et al., 2012). Aberrant orexin function has been associated with several pathophysiologies, such as obesity, narcolepsy and other sleep disorders, as well as the occurrence and severity of age-related disorders (Fadel et al., 2013). Here we briefly review the literature documenting the role of orexin in sleep disorders, energy balance, and aging. We discuss animal models and clinical studies, highlighting how alterations in central orexin signaling affects body weight, food intake, sleep patterns, and progression of age-related pathologies. We conclude that central orexin signaling is a promising target for pharmacological therapies to alleviate a myriad of disorders.

2. Animal Models

2.1. Rodent models for studying the role of orexin in obesity

Initial behavioral studies suggested that orexin was important in mediating central control of ingestive behavior and energy metabolism (Bray, 2000; Lubkin and Stricker-Krongrad, 1998; Sakurai et al., 1998). These studies showed orexin A had opposite effects on energy balance since exogenous orexin A stimulated hyperphagia and energy expenditure. This is unusual in that most peptides known to stimulate ingestion also inhibit sympathetic activity and thermogenesis, reducing energy expenditure (reviewed in Bray, 2000). Subsequent studies have indicated orexin influences individual propensity for weight gain, and have shown that orexin receptor stimulation results in a net negative energy balance. Rodent models for studying the role of orexin in obesity have been developed and tested, and include genetic gain and loss-of-function models, as well as pharmacologic and outbred models of individual variability.

2.1.1. Loss-of-function

Mice lacking orexin function either through genetic knockout (KO) of the gene encoding orexin or through postnatal ablation using an ataxin toxin develop late onset obesity (Chemelli et al., 1999; Hara et al., 2001; Hara et al., 2005). In both models, food intake and energy expenditure are affected by the absence of orexin function. These animals are hypophagic (eat less), and have substantially reduced energy expenditure, which appears to be primarily due to reductions in physical activity (Hara et al., 2001). Hara et al (Hara et al., 2005) showed important phenotypic differences in deleting orexin function by these two methods: losing the orexin gene vs. losing the entire orexin neuron. With the latter approach, co-localized neurotransmitters, including dynorphin, cocaine and amphetamine-related transcript, glutamate, neuronal activity-regulated pentraxin, and others are lost, and thus the impact that these neurotransmitters and their projections have on energy balance are also affected. Studies have shown that in the orexin/ataxin-3 model and in orexin gene KO mice, the obesity phenotype depends upon the mouse genetic background, level of knockdown and environment (Fujiki et al., 2006; Hara et al., 2005). While both the KO mice with a mixed or C57Bl/6J genetic background and the orexin/ataxin-3 mice with a C57Bl/6J background are heavier than wild type mice, body weight is similar between mixed background orexin/ataxin-3 mice and wild type mice. Body weights of orexin/ataxin-3 mice are greater than orexin KO mice and the body weight of heterozygote mice on a mixed background is intermediate between that of homozygous knockouts and wild type mice. The latter suggests that severity of obesity increases as orexin function declines. Female mice exhibit a higher level of obesity, potentially indicating greater sensitivity to orexin loss in females (Fujiki et al., 2006). Together, these data suggest that orexin (e.g. vs. other co-localized factors) and genetic background are critical for the obese phenotype on orexin-manipulated animal models (Fujiki et al., 2006; Hara et al., 2005). Injection studies demonstrate that a single intraperitoneal administration of a selective orexin 1 receptor antagonist (SB-334867-A) reduces food intake in both male and female rats (Haynes et al., 2000). Further, the same antagonist delivered chronically into cerebral ventricles of leptin deficient (ob/ob) mice over 14 days reduced body weight gain by reducing food intake (Haynes et al., 2002), although leptin deficient mice have baseline differences in energy regulation, limiting interpretation.

2.1.2. Gain of function

Studies by Yanagisawa and colleagues have demonstrated that orexin overexpression promotes energy expenditure while also reducing food intake and that central administration of an orexin receptor 2 agonist reduced diet-induced obesity (Funato et al., 2009). Reduction in energy expenditure is in agreement with other studies, but reduction in food intake by an orexin agonist is surprising. It is unclear why this discrepancy exists. Nonetheless, transgenic overexpression of orexin and the orexin receptor 2 affords protection from obesity when mice are placed on a high fat diet. Novak et al have shown that daily injection of orexin A into the hypothalamic paraventricular nucleus results in weight loss in rats (Novak and Levine, 2009), and recently, Perez-Leighton et al showed that daily orexin A injections into the rostral lateral hypothalamus reduces fat mass gain in rats on a high fat diet (Perez-Leighton et al., 2012). These studies demonstrate that enhanced orexin signaling, via increase in peptide or in receptor activation, can protect against weight gain.

2.1.3. Individual variability in orexin

Studies by Kotz et al have demonstrated that outbred rats resistant to obesity induced either by diet or age have high gene and protein expression of orexin receptors (Mavanji et al., 2010; Novak et al., 2006; Teske et al., 2006). Studies by Perez-Leighton et al show that individual sensitivity to obesity depends upon the level of gene expression for prepro-orexin: outbred rats with higher levels of the orexin prepro gene are resistant to fat mass gain, whereas those with low levels are sensitive to fat mass gain (Perez-Leighton et al., 2013). In all of these studies, the obesity resistant phenotype aligned with high orexin tone, high levels of physical activity and sleep quality, and high energy expenditure, whereas the obesity prone phenotype aligned with low orexin tone, low physical activity and sleep quality, and low energy expenditure.

Collectively, animal models show that orexin signaling influences propensity for weight gain. While some studies have shown decreased food intake after orexin receptor antagonism, overall, orexin signaling appears to promote a net increase in energy expenditure. Orexin loss of function models commonly exhibit reduced energy expenditure and increased weight gain leading to obesity (Hara et al., 2005), and enhanced orexin signaling or sensitivity to orexin has been shown in multiple selectively-bred and outbred models of obesity resistance in rodents (reviewed in Butterick et al., 2013). Together these findings suggest that orexin tone and responsivity is central for mediating energy expenditure, which has a profound impact on obesity susceptibility.

2.2. Rodent models for studying the role of orexin in sleep disorders

The ability of orexin to promote wakefulness, and to maintain and stabilize behavioral states, underscores the crucial role of the orexin system in regulation of sleep and wakefulness (Brisbare-Roch et al., 2007; Nishino et al., 2000; Willie et al., 2003). Sleep disruption is a common symptom of several central nervous system disorders, and is associated with abnormal orexin function (Dauvilliers et al., 2003). While the strongest evidence supporting the role of orexin in sleep are data showing that the sleep disorder narcolepsy is caused by disrupted orexin signaling (Chemelli et al., 1999; Chen et al., 2009; Nishino et al., 2000), it is unclear how orexin contributes to other disorders of sleep and wakefulness. Mechanistic studies in rodent models can elucidate the association between altered orexin function and sleep disorders.

Narcolepsy is a rare sleep disorder characterized by excessive daytime sleepiness, short sleep latency, and sleep onset REM periods (American Academy of Sleep Medicine, 2014). Narcolepsy may present with or without cataplexy, or a brief loss of muscle tone with retained consciousness, and associated features may include disturbed nocturnal sleep, sleep paralysis, and or hypnagogic hallucinations (American Academy of Sleep Medicine, 2014; Dauvilliers et al., 2007). A deficit in the orexin system is the primary pathophysiology of this disease (Fronczek et al., 2009). Rodent models of narcolepsy that either lack the orexin gene, orexin neurons, or orexin receptors have been studied to elucidate the orexin role in this debilitating sleep disorder (reviewed by de Lecea et al., 2002; Zhang et al., 2006).

2.2.1. Genetic Models

The orexin KO mouse, produced by targeted replacement of the first orexin gene exon, was the first rodent model of orexin-deficient narcolepsy characterized (Chemelli et al., 1999). These orexin null mice exhibit behavioral arrests in the dark (active) phase that are strikingly similar to catalepsy in human narcolepsy (Chemelli et al., 1999). Chemelli et al referred to these arrests as “narcoleptic attacks” instead of cataplexy, as they were unable to determine if consciousness was preserved, a critical feature of cataplexy. Orexin null mice all displayed sleep onset REM periods, fragmented sleep, and greater REM sleep during the dark (active) phase. Development of the orexin/ataxin-3 mouse and rat, rodent models with a selective and postnatal ablation of orexin neurons, soon followed (Beuckmann et al., 2004; Hara et al., 2001; Zhang et al., 2007a). These models more closely mimic the postnatal loss of orexin neurons in human narcolepsy (Peyron et al., 2000). In these animals, orexin-containing neurons were undetectable in 15-week-old orexin/ataxin-3 mice (Hara et al., 2001), reduced by 75% as early as 4 weeks in the orexin/ataxin-3 rats (Zhang et al., 2007a), and absent by 17 weeks (Beuckmann et al., 2004). Similar to sleep observed in the orexin null mice, orexin/ataxin-3 mice and rats have fragmented sleep and increased wake to REM sleep transitions (Beuckmann et al., 2004; Hara et al., 2001; Zhang et al., 2007a; Zhang et al., 2007b). However, unlike orexin null mice, orexin/ataxin-3 mice and rats spend less time in REM sleep in the light phase (Beuckmann et al., 2004; Hara et al., 2001). While narcolepsy in humans is associated with the absence of hypothalamic orexin, the significance of orexin receptors was highlighted by dog studies of narcolepsy, caused by a mutation in the canine OX2R gene (Lin et al., 1999). Shortly thereafter, abnormal sleep patterns in single or double orexin receptor KO mice were reported (Willie et al., 2003; Willie et al., 2001). Mice lacking OX2R display several abnormalities similar to human narcolepsy (Willie et al., 2001), yet the behavioral phenotype of OX2R KO mice appears less severe than orexin null mice, as OX2R KO mice display less frequent sleep onset REM periods, less sleep/wake fragmentation, and spend less total time in REM sleep (Chemelli et al., 2000; Chemelli et al., 1999; Willie et al., 2003; Willie et al., 2001). A comparison of mice lacking OX1R or OX2R revealed differential regulation of orexin receptor subtypes on REM sleep and NREM sleep suppressed by orexin A (Mieda et al., 2011). Stimulation of OX2R was more efficacious at reducing REM sleep compared to OX1R, while both orexin receptor subtypes were equally efficacious at suppressing NREM sleep. Double orexin receptor KO mice display sleep/wake disturbances most similar to human narcolepsy (Willie et al., 2001), while OX1R KO mice have no overt behavioral abnormalities except for increased sleep/wake fragmentation (Kisanuki et al., 2000). Blocking orexin signaling at both receptors may thus be necessary to mimic symptomology in human narcolepsy. Overall, the sleep patterns of rodent models with insufficient orexin signaling resemble human narcolepsy, indicating that future genetic and neurochemical studies with these models may provide important clues to the etiology and treatment of many debilitating human sleep disorders.

2.2.2. Non-genetic models

In addition to the genetic models described above, in vivo non-genetic approaches have been used to study the role of orexin in sleep/wake regulation. Lateral hypothalamic administration of orexin B conjugated to the neurotoxin saporin eliminated up to 90% of orexin neurons (Gerashchenko et al., 2003b) inducing narcolepsy-like behavior (Gerashchenko et al., 2003a; Gerashchenko et al., 2001; Gerashchenko et al., 2003b), and mimicking the orexin neuron loss and sleep disturbances of narcolepsy. In another study, microdialysis perfusion of OX2R antisense into the pontine reticular formation for three days increased REM sleep and cataplexy (Thakkar et al., 1999). Likewise, Chen et al showed that perifornical hypothalamic infusion of short interfering RNA (siRNA) specific to prepro-orexin mRNA significantly (59%) suppressed prepro-orexin mRNA, decreased the number of orexin-positive neurons, induced cataplexy-like episodes, and increased REM sleep during the dark phase (Chen et al., 2006). The latter two studies provide evidence for the diurnal gating of REM sleep by orexins, suggesting that targeting central orexin pathways for treating sleep disorders is promising, as the effects of OX2R antisense and prepro-orexin RNA interference were reversed 4–6 days post injection. Unlike previous studies interrogating orexin function alone, stimulation of orexin neuron function by optogenetics (de Lecea and Huerta, 2014) reduced latency to wakefulness and increased sleep/wake transitions (Adamantidis et al., 2007), the latter requiring noradrenergic signaling in the locus coeruleus (Carter et al., 2012). Thus, promising non-genetic models are available to further probe the role of orexin in the regulation of sleep and sleep disorders. In summary, the discovery of orexin marks a milestone in sleep research. A thorough understanding of the orexin system and its effect on specific sleep regulatory brain sites aids development of orexin analogues and small molecule orexin receptor antagonists, as treatments for hypersomnias and insomnias, respectively.

2.3. Rodent models for studying orexin and aging

Orexin moderates physiological processes that undergo age-related change, implying that aberrant orexin signaling contributes to altered sleep/wake and metabolism during aging. Age-related decline in the orexin system has been widely documented in animals, and parallels physiological alterations in body weight regulation and sleep during aging (Downs et al., 2007; Kappeler et al., 2003; Kessler et al., 2011; Kotz et al., 2005; Porkka-Heiskanen et al., 2004a; Porkka-Heiskanen et al., 2004b; Sawai et al., 2010; Terao et al., 2002; Zhang et al., 2002, 2005a). A significant decline in orexin immunopositive neurons has been observed as early as eight months in rats (Sawai et al., 2010), which is near the age at which a time-dependent change was also observed in mice (Brownell and Conti, 2010). Aging also influences the diurnal expression of orexin A in cerebrospinal fluid (CSF). While the overall diurnal pattern is consistent between young and aged mice (Yoshida et al., 2001; Zeitzer et al., 2003), others showed that orexin A in CSF is lower in aged 21-month old Fischer rats during the light and dark cycle (Desarnaud et al., 2004). Age-related morphological change in orexin neurons has been noted in aged cats (Zhang et al., 2002) and rats (Zhang et al., 2007a) that had ‘spot-like’ structures absent in younger animals. These orexin immunopositive ‘spot-like’ structures were identified as enlarged axon terminals by electron microscopy (Zhang et al., 2005b). Zhang et al concluded that aging associated morphological changes alters orexin signaling and may underlie “aged-related function of the orexin system” (Zhang et al., 2005b). Age-dependent effects on the orexin system appear to be modified by gender and species. In contrast to other species, distribution of orexin cell bodies, number of orexin neurons, and serum orexin B concentrations are similar between aged and young rhesus macaques (Downs et al., 2007), and there is no age-dependent effect on preproorexin (Terao et al., 2002) or orexin A (Lin et al., 2002) in C57BL/6J mice. In mice, while orexin immunopositive neurons decline with increasing age, males retain more neurons compared to age-matched female C57Bl/6J mice from 6–24 months of age (Brownell and Conti, 2010). In light of studies suggesting functional heterogeneity of orexin neurons, the pattern of loss, rather than the total number of neurons lost, may also be important in determining the behavioral effect of orexin neuron loss in aging. Several studies suggest that a medial-lateral division exists in orexin neurons, with medially- and dorsomedially-located neurons projecting to arousal regions, while those located laterally play a role in food intake (España et al., 2005; Fadel et al., 2002). Dysfunctional orexin signaling related to age-associated declines in orexin neuron number, gene expression, orexin peptide levels or receptors or the pattern of loss may underlie age-related change in metabolic and sleep-related behavior.

Age-dependent neuroanatomical and morphological changes in the orexin system parallel behavioral and physiological processes including sleep (Morairty et al., 2011; Porkka-Heiskanen et al., 2004a), metabolism (Martone et al., 2013; Poehlman and Horton, 1990), insulin sensitivity (Tsuneki et al., 2008), hemodynamics (Hirota et al., 2003), and cognition (Stanley and Fadel, 2012). The functional significance of senescence on the hypothalamic orexin system is exemplified in a rodent model of healthy aging. Lou C/JaLL rats have higher hypothalamic preproorexin mRNA and lower body weight than Wistar rats, and normal food intake at 24 months of age (Kappeler et al., 2004). Likewise, in BRASTO brain-specific Sirt1-overexpressing transgenic mice, there is a youthful metabolic and behavioral phenotype coinciding with enhanced OX2R promoter activity. Aged BRASTO mice have higher physical activity, body temperature, oxygen consumption, sleep quality, and skeletal muscle mitochondrial function relative to aged-matched controls (Satoh et al., 2010; Satoh et al., 2013). Moreover, aged-BRASTO mice have greater OX2R mRNA in dorsomedial hypothalamus (Satoh et al., 2010; Satoh et al., 2013), and knockdown of Sirt1 or OX2R abrogates BRASTO heightened physical activity and body temperature. Consistent with Sirt1 enhancement of OX2R promoter activity in cultured cells (Satoh et al., 2010), Satoh and colleagues further demonstrated that a combination of Sirt1 and Nk2 homeobox 1 enhances OX2R promoter activity, suggesting that enhanced Sirt1-mediated OX2R underlies the prolonged lifespan and youthful behavior of aged-BRASTO mice.

Studies suggest a dysfunctional orexin system potentiates adiposity gain during aging. Senescent animals display reduced food intake or age-related anorexia, which has been defined as declines in caloric intake or appetitive drive with increasing age (Akimoto and Miyasaka, 2010; Kmiec, 2006, 2011; Martone et al., 2013). In addition to reduced ad libitum food intake, aged animals display diminished hyperphagic response to central orexin A infusion (Akimoto and Miyasaka, 2010; Kotz et al., 2005; Takano et al., 2004) with a concomitant decline in orexin neuron activity (Kotz et al., 2005). While reduced appetite seems discordant with body weight gain, orexin dysfunction during aging has a more profound effect on energy expenditure. Reductions in age-related orexin drive would elicit a more robust reduction in energy expenditure than energy intake, promoting body weight gain despite reduced consumption.

Synonymous with reduced food intake, enhanced satiety in aged animals may be a response to lower energy expenditure, reduced physical activity, and thermoregulatory dysfunction during aging (Blatteis, 2012; Holowatz and Kenney, 2010). It is widely appreciated that reduced physical activity parallels advancing age in humans and other species (Kmiec et al., 2013; Manini, 2010), which would be expected to contribute to reduce daily energy expenditure. We and others have demonstrated the robust contribution of orexin signaling to physical activity and energy expenditure in young rodents (reviewed in Kotz et al., 2012; Lubkin and Stricker-Krongrad, 1998; Novak and Levine, 2007). Based on the promotion of physical activity, temperature and energy expenditure by orexin, it is plausible that during aging, reduced orexin-stimulated physical activity would reduce daily physical activity, which in turn would be expected to reduce physical activity-associated energy expenditure. Reductions in resting metabolism likely contributes to lower total energy expenditure with advancing age (Elia et al., 2000). Reductions in resting metabolism related to sarcopenia due to low physical activity would also contribute to reductions in total energy expenditure. Thermoregulation may be augmented as a consequence of sarcopenia (Kenney and Buskirk, 1995). It is clear that orexin participates in diminished thermogenic capacity in rodents. Brown adipocytes within interscapular brown adipose tissue from aged mice have fewer multilocular cells and thus are morphologically more similar to white adipocytes. This morphological change parallels deficient thermoregulation and a lower ability to mobilize intracellular fuel reserves from brown adipocytes (Sellayah and Sikder, 2014). That orexin A treatment reverses the morphological and functional consequences of aged interscapular brown adipose tissue, indicated by greater uncoupling protein one mRNA, increased cold tolerance, and weight loss, supports the hypothesis that orexin dysfunction with advancing age augments energy expenditure to promote obesity during aging (Sellayah and Sikder, 2014). Moreover, orexin influences autonomic function including sympathetic outflow, implicating that age-related orexin neurodegeneration may contribute to reduced cutaneous thermoregulatory blood flow during advancing age. Together these age-related alterations in physical activity, energy expenditure (resting and physical activity-related), and thermoregulation would be expected to promote the reductions in total daily energy expenditure and increase adiposity gain observed in aged humans.

3. Clinical implications

The contribution of central orexin signaling to human pathophysiology is well recognized. Abnormalities in orexin signaling pathways underlie the pathophysiology of sleep disorders (Baumann and Bassetti, 2005a, b; Cao and Guilleminault, 2011; Dyken and Yamada, 2005; Malhotra and Kushida, 2013; Mignot, 2004; Overeem et al., 2001; Ritchie et al., 2010; Tafti et al., 2005; Taheri et al., 2002; Wisor and Kilduff, 2005; Zeitzer, 2013) such as narcolepsy (Nishino et al., 2000; Peyron et al., 2000; Thannickal et al., 2000) and may contribute to posttraumatic hypersomnia or excessive daytime sleepiness due to traumatic brain injury (Baumann, 2012; Baumann et al., 2009), post traumatic stress disorder (Strawn et al., 2010), or obstructive sleep apnea (Ahmed et al., 2012; Wang et al., 2013). Insufficient central orexin signaling has also been associated with other medical conditions (Mignot et al., 2002; Vankova et al., 2003) such as obesity (Van Cauter and Knutson, 2008), age-related anorexia (Kmiec et al., 2013), multiple system atrophy (Benarroch et al., 2007), neurological disorders (Fronczek et al., 2009), Parkinson’s disease (Fronczek et al., 2007; Thannickal et al., 2007; Wienecke et al., 2012), and Alzheimer’s disease (Slats et al., 2013). Since symptomology may not parallel CSF orexin levels (Dauvilliers et al., 2003; Martinez-Rodriguez et al., 2003; Nishino et al., 2003; Ripley et al., 2001) and low orexin levels and aberrant sleep have been reported in other medical conditions, it is difficult to verify causality (reviewed by Mignot, 2004; Mignot et al., 2002; Taheri et al., 2002). Hence, determining the contribution of abnormal orexin signaling to human pathophysiology may lead to improved therapeutic avenues.

3.1. Orexin and obesity in humans

The earliest descriptions of narcolepsy highlighted elevated BMI, body weight, and central obesity (Dahmen et al., 2001; Kok et al., 2003; Nishino et al., 2001; Schuld et al., 2000) and underscored the critical link between orexin dysfunction, sleep, and obesity. Whereas narcolepsy is caused by dramatic loss of orexin signaling, evidence suggests that milder perturbations of the orexin system may disrupt the normal orexinergic gating of sleep/wake state transitions and thus contribute to poor sleep quality. In addition, orexin deficient narcoleptic patients are more obese compared to narcoleptic patients with normal CSF orexin levels (Nishino et al., 2001). Weight reduction has been shown to increase plasma orexin in adolescents (Bronsky et al., 2007), suggesting that increased adiposity might result in reduced orexin signaling. The relationship between orexin and obesity is complex, and in many cases it is difficult to determine whether factors such as physical activity level, sleep quality, or circulating orexin levels are a consequence of or a causal factor for development of obesity.

3.2. Orexin and sleep in humans

The sleep disorder narcolepsy best exemplifies the clinical manifestations of dysfunctional central orexin signaling. Common symptoms of narcolepsy include excessive daytime sleepiness and abnormal manifestations of REM sleep noted during polysomnography, refreshing naps, mean sleep latency less than or equal to eight minutes, and two or more sleep onset REM periods on a mean sleep latency test. Associated features may include sleep paralysis, hypnagogic hallucinations, or autonomic behavior. Narcolepsy, also referred to as hypocretin deficiency syndrome and narcolepsy with or without cataplexy, has recently been subcategorized to narcolepsy type I and II in the third edition of the International Classification of Sleep Disorders (American Academy of Sleep Medicine, 2014). Absent or low levels (≤ 110 pg/ml or < 1/3 of mean values obtained in normal subjects with the same standardized assay (American Academy of Sleep Medicine, 2014)) of orexin in CSF is the hallmark feature of narcolepsy type I. When cataplexy is absent yet CSF orexin is normal or unknown, individuals are diagnosed with narcolepsy type II (American Academy of Sleep Medicine, 2014).

Narcolepsy type I is caused by a deficiency in orexin signaling due to loss of orexin neurons (reviewed in Kornum et al., 2011; Mignot, 2004; Overeem et al., 2001; Siegel, 1999; Siegel and Boehmer, 2006; Taheri et al., 2002). Strong association between narcolepsy and the human leukotype antigen DRD2 or DQB1*0602 implies autoimmune destruction of orexin neurons (reviewed by De la Herran-Arita and Garcia-Garcia, 2014; Fontana et al., 2010; Kornum et al., 2011; Lin et al., 2001; Mignot, 2004; Singh et al., 2013). Homozygosity for DQB1*0602 or heterozygosity for DQB1*0602/0301 is highly associated with narcolepsy (Hong et al., 2002; Hor et al., 2010; Kornum et al., 2011; Mignot et al., 1997; Mignot et al., 2001). However, some patients with narcolepsy present with normal orexin levels or are negative for human leukotype antigen DR2 or DQB1*0602, the percentage of the population positive for the DQB1*602 far exceeds the narcolepsy prevalence (Taheri et al., 2002) and there is a low rate of concordance for narcolepsy in monozygotic twins (Mignot, 1998), indicating that, as with other auto-immune disorders, gene-environment interactions (Kroeger and de Lecea, 2009; Taheri et al., 2002) and non-genetic factors contribute to narcolepsy pathogenesis (Ritchie et al., 2010). In rare cases, mutations or polymorphisms in the preproorexin gene or the orexin two receptor parallel a positive diagnosis (Dong et al., 2013; Peyron et al., 2000).

Experience with deficient orexin in narcolepsy led to the question of whether too much orexin activity might be responsible for insomnia. One formulation of this concept labels the problem of insomnia as hyperarousal (Bonnet and Arand, 2010; Riemann et al., 2010), driven by excess orexin. These ideas formed the basis for development of orexin antagonist medications for treatment of insomnia, and the first of these medications has been approved by the FDA. Suvorexant is a dual receptor antagonist, thereby blocking both orexin receptors. In clinical trials, suvorexant has improved sleep efficiency in short term and long term treatment (Herring et al., 2012; Michelson et al., 2014). There have been concerns that orexin antagonism might lead to narcoleptic symptoms, but no significant symptoms were observed in trials thus far (Herring et al., 2012; Michelson et al., 2014). Orexin antagonists therefore appear to be a promising avenue for treatment of an otherwise resistant condition, insomnia.

3.3. Orexin and aging in humans

Reduced energy expenditure, reduced food intake, and weight loss occur in elderly humans (Kmiec et al., 2013; Manini, 2010). This ‘anorexia of aging’ has been attributed to reduction in number or in sensitivity to a number of appetite-regulating peptides, including orexin (Kmiec et al., 2013). As summarized above, animal models show that orexin loss occurs during aging, and this loss is specific to orexin neurons rather than due to general age-related neurodegeneration (Kessler et al., 2011), although at least one study in primates has shown no correlation between age and orexin neuron number (Downs et al., 2007). Overall, the strength of the animal models suggest a similar loss of orexin would occur in aged humans. Outside of disease models, change in orexin neuron number with age has only recently been directly examined in humans (Hunt et al., 2014). This study showed a 23% decrease in orexin neuron number from infancy to late adulthood, with a 10% decline occurring between early and late adulthood (Hunt et al., 2014). Aside from this study, and those utilizing brain tissue analysis from persons with narcolepsy, studies in humans have relied mainly on plasma or CSF levels of orexin. Somewhat paradoxically, human plasma orexin levels have been shown to increase with aging, although the relationship between plasma and brain orexin is undefined, and plasma orexin likely relates more to peripheral than central orexin action. In humans, plasma orexin level correlates with age, with lower levels in subjects under 40, and higher levels among those aged 60 or more (Matsumura et al., 2002). In women, plasma orexin increases significantly during menopause (El-Sedeek et al., 2010). Among menopausal women, those receiving hormone replacement therapy have lower levels of plasma orexin than those receiving placebo (El-Sedeek et al., 2010). Plasma orexin is known to increase after weight loss in obese children and in lean and obese adults (Bronsky et al., 2007; Heinonen et al., 2005; Komaki et al., 2001), suggesting that increase in plasma orexin is not inconsistent with orexin involvement in age-related weight loss. However, increased plasma orexin is difficult to explain if age-related loss of orexin neurons occurs in humans. Studies in rodents suggest orexin receptors are also lost during aging (Porkka-Heiskanen et al., 2004a; Terao et al., 2002). If the same is true in humans, reduction in orexin receptors may result in greater production of peptide to overcome reduced sensitivity, or to increased unbound peptide, ultimately leading to higher levels of orexin in plasma. Plasma orexin level may also inaccurately reflect actual number of orexin neurons. In both rodents and humans, substantial loss of orexin neurons (50–70%) is required before significant decrease in CSF levels of orexin are evident (Fronczek et al., 2007; Gerashchenko et al., 2003b). Plasma orexin may be even more variable, as animal studies have shown orexin is produced in the enteric nervous system of the gut as well as in brain (Kirchgessner, 2002; Kirchgessner and Liu, 1999). However, a recent study failed to observe orexin-producing neurons in human gut tissue (Baumann et al., 2008), and no studies have examined whether orexin-producing neurons outside of the central nervous system decline with aging.

Aging is also associated with a number of changes affecting sleep, energy homeostasis, and cognition. While few studies have specifically investigated aging and orexin in humans, data from these and from animal studies support that a decline in either orexin neurons or orexin sensitivity in older individuals contributes to at least some aspects of these age-related changes. First, the most salient recognized function of orexins is promotion of arousal and stabilization of the sleep/wake cycle. Age-related decrease in orexin would thus be expected to negatively impact sleep duration and sleep quality, and incidence of sleep disturbance, sleep disorders, and insomnia do increase with age in humans (Drake et al., 2003; Wimmer et al., 2013). Second, orexin has been linked to promotion of energy expenditure in rodents (Kotz et al., 2008; Kotz et al., 2002; Kotz et al., 2006; Lubkin and Stricker-Krongrad, 1998; Teske et al., 2006; Thorpe and Kotz, 2005). Average daily energy expenditure in humans is known to increase throughout childhood, peaking at adolescence, and decreasing around 100–160 kCal per day each decade in women and men, respectively, throughout the remainder of life (Manini, 2010). This decline in daily energy expenditure with aging parallels reductions in physical activity observed in humans and animal models with orexin deficiencies (Hara et al., 2001; Nishino et al., 2001). Finally, orexin is known to affect performance in cognitive tests of spatial and working memory (Akbari et al., 2007; Deadwyler et al., 2007). Age-related decline in cognitive abilities are common in older adults, with incidence of diagnosis increasing from 4 to 36% between the ages of 65 and 85 (Black and Rush, 2002). In addition to normal decline in orexin during aging, orexin loss is a component of several progressive neurodegenerative diseases, including Alzheimer’s, Parkinson’s, and Huntington’s diseases (Fronczek et al., 2007; Fronczek et al., 2012; Petersen et al., 2005; Thannickal et al., 2007). Sleep disturbances and cognitive deficits are known comorbidities of these diseases. While the underlying cause of orexin loss in these disorders is unknown, it is possible that some aspect of these neurodegenerative disease processes greatly accelerate the normal age-related reduction in orexin neurons. It is increasingly evident that sleep, activity and cognition are interconnected (Horne, 2013), as age-related disturbances in one component also affect the others. It is plausible that orexin loss contributes to cognitive decline in both normal aging and in neurodegenerative disease, whether through direct involvement of orexin in cognitive tasks, or indirectly through effects on sleep and activity.

Many prior studies have shown that orexin neurons may be important mediators of cognitive performance. Orexin neurons appear to synapse directly on basal forebrain cholinergic neurons important in cognition (Castillo-Ruiz et al., 2010; Fadel et al., 2005; Frederick-Duus et al., 2007), and orexin appears to mediate long-term potentiation in the dentate gyrus of the hippocampus (Akbari et al., 2011). Prior work utilizing operant tasks shows that supplementing orexin might affect cognitive processes (Choi et al., 2010; Sharf et al., 2010; Thorpe et al., 2005). Orexin treatment improves performance in progressive ratio, fixed ratio, and delayed matching to sample tasks in rodents and primates (Choi et al., 2010; Deadwyler et al., 2007; Thorpe et al., 2005). Further, several lines of evidence support that blocking orexin action can impair cognition. The selective orexin 1 receptor antagonist SB-334867 impairs performance in progressive and variable ratio operant testing (Sharf et al., 2010), decreases performance in an attentional task in rats (Boschen et al., 2009), and impairs performance in passive avoidance and spatial memory tests (Akbari et al., 2008; Akbari et al., 2006, 2007). The wake-inducing drug modafinil, which activates orexin neurons, improves attention in rats (Morgan et al., 2007; Scammell et al., 2000), and the stimulant nicotine appears to act in part through activation of orexin neurons projecting to the basal forebrain and thalamus (Pasumarthi and Fadel, 2008). Interestingly, modafinil, nicotine, and acetylcholine all affect or are affected by orexin signaling in cognition, and modafinil, nicotine and acetylcholinesterase inhibitors are routinely used as cognitive enhancers in humans (Husain and Mehta, 2011).

While some degree of decline is to be expected with age, the relative severity of impairments caused by physical and cognitive impairments can often lead to reduced independence and quality of life by interfering with the ability to perform daily tasks (Black and Rush, 2002; Huh et al., 2011; Wang et al., 2006). If orexin loss during aging does contribute to age-related decline in sleep, energy expenditure, and cognition, therapies aimed at increasing orexin signaling may prove beneficial for the elderly by positively impacting all of these factors. There appears to be a synergistic relationship between decline in physical activity and cognitive performance in older adults, in that changes in one measure predict future changes in the other. Studies examining measures of physical ability and cognitive function have shown both that early identification of reduced cognitive function correlates with future decline in physical ability (Black and Rush, 2002; Huh et al., 2011), and that physical impairments are also indicators of future cognitive decline (Black and Rush, 2002; Wang et al., 2006). There is ample evidence that interventions that increase physical activity might improve cognitive function in all age groups (Colcombe et al., 2004; Davis et al., 2011; Etnier et al., 2006). In one study of older adults, aerobic exercise intervention improved cognitive performance in an executive attentional task and in functional MRI measures of cortical plasticity (Colcombe et al., 2004). A meta-regression of 37 studies on cognition and physical activity also showed a positive correlation between cognitive ability and physical activity (Etnier et al., 2006). Interestingly, this study found no correlation between aerobic fitness and cognitive function, suggesting that the increase in overall physical activity rather than in vigorous aerobic exercise is important in maintaining cognitive ability. If true, this implies that that loss of spontaneous physical activity inducing agents, such as orexin, might contribute to cognitive decline, and that treatments that increase spontaneous physical activity during aging might also exert a protective effect against cognitive decline. Increased spontaneous physical activity could encourage better health through physical movement, potentially counteracting normal age-related increases in sedentary behavior (Manini, 2010), and through the cognitive feedback stimulated by ambulation, especially if performed in an engaging environment (Horne, 2013).

3.4. Orexin in neurodegenerative disease

As discussed above, loss of orexin neurons and sleep disturbances are known comorbidities of several neurodegenerative diseases, including Parkinson’s (PD) and Alzheimer’s disease (AD) (Fronczek et al., 2007; Fronczek et al., 2012; Petersen et al., 2005; Thannickal et al., 2007). While many studies have focused on the importance of orexin loss in disease-related sleep disturbances, several recent reports suggest orexin may also play a role in some aspects of neurodegenerative disease pathogenesis. Orexin receptor 2 polymorphisms have been identified as a potential risk factor for development of AD (Gallone et al., 2014), however in another study loss of orexin neurons in narcoleptics failed to alter AD risk (Scammell et al., 2012). With respect to PD, the incidence of prior narcolepsy diagnosis was five times higher than expected in PD patients (Christine et al., 2012), suggesting that narcolepsy or narcolepsy treatments might influence development of PD. However, drugs aimed at treating PD might selectively damage orexin neurons (Katsuki and Michinaga, 2012), making interpretation of narcolepsy-PD links difficult. Evidence strongly links development of AD and PD with oxidative stress and mitochondrial dysfunction in the brain (Agostinho et al., 2010; Ferreiro et al., 2012; Niranjan, 2013). In recent in vitro and in vivo studies, we and others have shown that orexin is neuroprotective, reducing neuronal damage caused by ischemia or oxidative insult in hypothalamic, hippocampal, and cortical tissue (Butterick et al., 2012; Sokolowska et al., 2012; Yuan et al., 2011). While the mechanism is not fully defined, orexin appears to increase resistance to oxidative stress by upregulation of hypoxia-inducible factor 1 alpha (HIF-1α) (Butterick et al., 2013; Feng et al., 2014; Sikder and Kodadek, 2007; Yuan et al., 2011). HIF-1α is a transcription factor that alters mitochondrial activity by increasing ATP production through oxidative phosphorylation, and affects expression of transferrin, a gene important in regulating iron metabolism in the brain (Semenza, 2001; Weinreb et al., 2013). Increased oxidative stress and dysfunction in brain iron metabolism are associated with etiology of both AD and PD (Loeffler et al., 1995; Nestrasil et al., 2010; Niranjan, 2013; Weinreb et al., 2013). With respect to PD, HIF-1α activation has been proposed as a potential therapeutic treatment (Weinreb et al., 2013), and orexin has recently been shown to protect against the Parkinsonian neurotoxin 1-methyl-4-phenylpyridinium (MPP+)-induced toxicity through induction of HIF-1α in a dopamine-producing neuronal cell line (Feng et al., 2014). Taken as a whole, these results suggest that orexin might protect against development of neurodegenerative disease, and that orexin loss in AD and PD might exacerbate disease progression by increasing susceptibility to oxidative damage. While promising, the overall significance of these findings are currently unclear, and other studies have shown contradictory conclusions. For example, orexin has been positively linked to increased amyloid-β (Aβ) accumulation in a mouse model of AD (Kang et al., 2009). Sleep deprivation in this study caused an increase in amyloid plaque formation, while a dual orexin receptor antagonist decreased Aβ accumulation. These results suggest increased orexin signaling could exacerbate rather than protect against development of AD. A more recent study has shown that while Aβ accumulation is correlated with sleep disturbances in mice, prevention of amyloid plaque formation also normalizes sleep/wake cycles (Roh et al., 2012). If orexin loss contributes to disease progression of AD, sleep disruptions due to decreased orexin signaling would be expected even if plaque formation were reduced. The importance of orexin signaling in neurodegenerative disease is potentially promising but unclear at present.

4. CONCLUSION

While our focus here is on the role of orexin in energy metabolism and sleep, it is clear that this multifaceted peptide also influences other physiological processes. Orexin is likely to play an integrative role, coordinating central modulation of sleep and physical activity in the context of energy balance. While uniquely positioned to tie many disparate systems together, this connectivity has a down side. When dysfunction of the orexin system occurs, a great number of regulator and behavioral systems are affected. Because of this confound, it can be difficult to disentangle the effect of orexin on sleep, obesity, or cognition alone, for example, as orexin has been shown to have direct or indirect effects on all three. Despite these difficulties, the influence of orexin on multiple systems also presents the possibility of simplifying therapeutic treatments, as orexin-based therapies might positively impact multiple morbidities. For example, promotion of healthier sleep patterns through enhanced orexin signaling could also lead to reduction in weight and improved cognitive performance, while orexin-based therapies in the elderly designed to increase physical activity may also stabilize sleep patterns. Although orexin clearly represents only one component in the complex regulatory mechanisms underlying aging, obesity, and sleep disorders, there is clear potential benefit of developing orexin-based therapies to alleviate symptoms of these health conditions in humans.

Highlights.

  • Central orexin signaling declines with age

  • Dysregulation of orexin function is associated with obesity and sleep disorders

  • Reduced orexin impacts body weight, sleep, and age-related pathologies

  • Orexin effects on sleep and activity may also impact cognitive performance

  • Orexin may be a therapeutic target for treatment of multiple age-related disorders

Acknowledgments

Funding for this research and publication was supported by the Department of Veterans Affairs (F7212W to JAT, 5I01RX000441-04 to CMK and CJB, and 1IK2BX001686-01A1 to TAB), the National Institutes of Health-NIDDK (1R01DK100281-01A1 to CMK and CJB), and the United States Department of Agriculture (ARZT-1360220-H23-150 and ARZT-1372540-R23-131 to JAT).

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Contributor Information

Joshua P. Nixon, Email: nixon049@umn.edu.

Vijayakumar Mavanji, Email: mavan001@umn.edu.

Tammy A. Butterick, Email: butte017@umn.edu.

Charles J. Billington, Email: billi005@umn.edu.

Catherine M. Kotz, Email: kotzx004@umn.edu.

Jennifer A. Teske, Email: teskeja@email.arizona.edu.

References

  1. Adamantidis AR, Zhang F, Aravanis AM, Deisseroth K, de Lecea L. Neural substrates of awakening probed with optogenetic control of hypocretin neurons. Nature. 2007;450:420–424. doi: 10.1038/nature06310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Agostinho P, Cunha RA, Oliveira C. Neuroinflammation, oxidative stress and the pathogenesis of Alzheimer’s disease. Curr Pharm Des. 2010;16:2766–2778. doi: 10.2174/138161210793176572. [DOI] [PubMed] [Google Scholar]
  3. Ahmed WA, Tsutsumi M, Nakata S, Mori T, Nishimura Y, Fujisawa T, Kato I, Nakashima M, Kurahashi H, Suzuki K. A functional variation in the hypocretin neuropeptide precursor gene may be associated with obstructive sleep apnea syndrome in Japan. Laryngoscope. 2012;122:925–929. doi: 10.1002/lary.23179. [DOI] [PubMed] [Google Scholar]
  4. Akbari E, Motamedi F, Davoodi FG, Noorbakhshnia M, Ghanbarian E. Orexin-1 receptor mediates long-term potentiation in the dentate gyrus area of freely moving rats. Behav Brain Res. 2011;216:375–380. doi: 10.1016/j.bbr.2010.08.017. [DOI] [PubMed] [Google Scholar]
  5. 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. 2008;187:172–177. doi: 10.1016/j.bbr.2007.09.019. [DOI] [PubMed] [Google Scholar]
  6. Akbari E, Naghdi N, Motamedi F. Functional inactivation of orexin 1 receptors in CA1 region impairs acquisition, consolidation and retrieval in Morris water maze task. Behav Brain Res. 2006;173:47–52. doi: 10.1016/j.bbr.2006.05.028. [DOI] [PubMed] [Google Scholar]
  7. 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. 2007;28:650–656. doi: 10.1016/j.peptides.2006.11.002. [DOI] [PubMed] [Google Scholar]
  8. Akimoto S, Miyasaka K. Age-associated changes of appetite-regulating peptides. Geriatr Gerontol Int. 2010;10(Suppl 1):S107–119. doi: 10.1111/j.1447-0594.2010.00587.x. [DOI] [PubMed] [Google Scholar]
  9. American Academy of Sleep Medicine. International classification of sleep disorders. 3. American Academy of Sleep Medicine; Darien, IL: 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Baumann CR. Traumatic brain injury and disturbed sleep and wakefulness. Neuromolecular Med. 2012;14:205–212. doi: 10.1007/s12017-012-8178-x. [DOI] [PubMed] [Google Scholar]
  11. Baumann CR, Bassetti CL. Hypocretins (orexins) and sleep-wake disorders. Lancet Neurol. 2005a;4:673–682. doi: 10.1016/S1474-4422(05)70196-4. [DOI] [PubMed] [Google Scholar]
  12. Baumann CR, Bassetti CL. Hypocretins (orexins): clinical impact of the discovery of a neurotransmitter. Sleep Med Rev. 2005b;9:253–268. doi: 10.1016/j.smrv.2005.01.005. [DOI] [PubMed] [Google Scholar]
  13. Baumann CR, Bassetti CL, Valko PO, Haybaeck J, Keller M, Clark E, Stocker R, Tolnay M, Scammell TE. Loss of hypocretin (orexin) neurons with traumatic brain injury. Ann Neurol. 2009;66:555–559. doi: 10.1002/ana.21836. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Baumann CR, Clark EL, Pedersen NP, Hecht JL, Scammell TE. Do enteric neurons make hypocretin? Regul Pept. 2008;147:1–3. doi: 10.1016/j.regpep.2007.11.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Benarroch EE, Schmeichel AM, Sandroni P, Low PA, Parisi JE. Involvement of hypocretin neurons in multiple system atrophy. Acta Neuropathol. 2007;113:75–80. doi: 10.1007/s00401-006-0150-0. [DOI] [PubMed] [Google Scholar]
  16. Beuckmann CT, Sinton CM, Williams SC, Richardson JA, Hammer RE, Sakurai T, Yanagisawa M. Expression of a poly-glutamine-ataxin-3 transgene in orexin neurons induces narcolepsy-cataplexy in the rat. J Neurosci. 2004;24:4469–4477. doi: 10.1523/JNEUROSCI.5560-03.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Black SA, Rush RD. Cognitive and functional decline in adults aged 75 and older. J Am Geriatr Soc. 2002;50:1978–1986. doi: 10.1046/j.1532-5415.2002.50609.x. [DOI] [PubMed] [Google Scholar]
  18. Blatteis CM. Age-dependent changes in temperature regulation - a mini review. Gerontology. 2012;58:289–295. doi: 10.1159/000333148. [DOI] [PubMed] [Google Scholar]
  19. Bonnet MH, Arand DL. Hyperarousal and insomnia: state of the science. Sleep Med Rev. 2010;14:9–15. doi: 10.1016/j.smrv.2009.05.002. [DOI] [PubMed] [Google Scholar]
  20. Boschen KE, Fadel JR, Burk JA. Systemic and intrabasalis administration of the orexin-1 receptor antagonist, SB-334867, disrupts attentional performance in rats. Psychopharmacology (Berl) 2009;206:205–213. doi: 10.1007/s00213-009-1596-2. [DOI] [PubMed] [Google Scholar]
  21. Bray GA. Reciprocal relation of food intake and sympathetic activity: experimental observations and clinical implications. Int J Obes Relat Metab Disord. 2000;24(Suppl 2):S8–17. doi: 10.1038/sj.ijo.0801269. [DOI] [PubMed] [Google Scholar]
  22. Brisbare-Roch C, Dingemanse J, Koberstein R, Hoever P, Aissaoui H, Flores S, Mueller C, Nayler O, van Gerven J, de Haas SL, Hess P, Qiu C, Buchmann S, Scherz M, Weller T, Fischli W, Clozel M, Jenck F. Promotion of sleep by targeting the orexin system in rats, dogs and humans. Nat Med. 2007;13:150–155. doi: 10.1038/nm1544. [DOI] [PubMed] [Google Scholar]
  23. Bronsky J, Nedvidkova J, Zamrazilova H, Pechova M, Chada M, Kotaska K, Nevoral J, Prusa R. Dynamic changes of orexin A and leptin in obese children during body weight reduction. Physiol Res. 2007;56:89–96. doi: 10.33549/physiolres.930860. [DOI] [PubMed] [Google Scholar]
  24. Brownell SE, Conti B. Age- and gender-specific changes of hypocretin immunopositive neurons in C57Bl/6 mice. Neurosci Lett. 2010;472:29–32. doi: 10.1016/j.neulet.2010.01.048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Burdakov D, Karnani MM, Gonzalez A. Lateral hypothalamus as a sensor-regulator in respiratory and metabolic control. Physiol Behav. 2013;121:117–124. doi: 10.1016/j.physbeh.2013.03.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Butterick TA, Billington CJ, Kotz CM, Nixon JP. Orexin: pathways to obesity resistance? Rev Endocr Metab Disord. 2013;14:357–364. doi: 10.1007/s11154-013-9259-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Butterick TA, Nixon JP, Billington CJ, Kotz CM. Orexin A decreases lipid peroxidation and apoptosis in a novel hypothalamic cell model. Neurosci Lett. 2012;524:30–34. doi: 10.1016/j.neulet.2012.07.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Cao M, Guilleminault C. Hypocretin and its emerging role as a target for treatment of sleep disorders. Curr Neurol Neurosci Rep. 2011;11:227–234. doi: 10.1007/s11910-010-0172-9. [DOI] [PubMed] [Google Scholar]
  29. Carter ME, Brill J, Bonnavion P, Huguenard JR, Huerta R, de Lecea L. Mechanism for Hypocretin-mediated sleep-to-wake transitions. Proc Natl Acad Sci U S A. 2012;109:E2635–2644. doi: 10.1073/pnas.1202526109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Castillo-Ruiz A, Nixon JP, Smale L, Nunez AA. Neural activation in arousal and reward areas of the brain in day-active and night-active grass rats. Neuroscience. 2010;165:337–349. doi: 10.1016/j.neuroscience.2009.10.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Chemelli RM, Sinton CM, Yanagisawa M. Polysomnographic Characterization of Orexin-2 Receptor Knockout Mice. Sleep Med Rev. 2000;23:A255. [Google Scholar]
  32. 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. 1999;98:437–451. doi: 10.1016/s0092-8674(00)81973-x. [DOI] [PubMed] [Google Scholar]
  33. Chen L, Brown RE, McKenna JT, McCarley RW. Animal models of narcolepsy. CNS Neurol Disord Drug Targets. 2009;8:296–308. doi: 10.2174/187152709788921717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. 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. 2006;24:2039–2048. doi: 10.1111/j.1460-9568.2006.05058.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Choi DL, Davis JF, Fitzgerald ME, Benoit SC. The role of orexin-A in food motivation, reward-based feeding behavior and food-induced neuronal activation in rats. Neuroscience. 2010;167:11–20. doi: 10.1016/j.neuroscience.2010.02.002. [DOI] [PubMed] [Google Scholar]
  36. Christine CW, Marks WJ, Jr, Ostrem JL. Development of Parkinson’s disease in patients with Narcolepsy. J Neural Transm. 2012;119:697–699. doi: 10.1007/s00702-011-0761-z. [DOI] [PubMed] [Google Scholar]
  37. Colcombe SJ, Kramer AF, Erickson KI, Scalf P, McAuley E, Cohen NJ, Webb A, Jerome GJ, Marquez DX, Elavsky S. Cardiovascular fitness, cortical plasticity, and aging. Proc Natl Acad Sci U S A. 2004;101:3316–3321. doi: 10.1073/pnas.0400266101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Dahmen N, Bierbrauer J, Kasten M. Increased prevalence of obesity in narcoleptic patients and relatives. Eur Arch Psychiatry Clin Neurosci. 2001;251:85–89. doi: 10.1007/s004060170057. [DOI] [PubMed] [Google Scholar]
  39. 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. 1999;96:748–753. doi: 10.1073/pnas.96.2.748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Dauvilliers Y, Arnulf I, Mignot E. Narcolepsy with cataplexy. Lancet. 2007;369:499–511. doi: 10.1016/S0140-6736(07)60237-2. [DOI] [PubMed] [Google Scholar]
  41. Dauvilliers Y, Baumann CR, Carlander B, Bischof M, Blatter T, Lecendreux M, Maly F, Besset A, Touchon J, Billiard M, Tafti M, Bassetti CL. CSF hypocretin-1 levels in narcolepsy, Kleine-Levin syndrome, and other hypersomnias and neurological conditions. J Neurol Neurosurg Psychiatry. 2003;74:1667–1673. doi: 10.1136/jnnp.74.12.1667. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Davis CL, Tomporowski PD, McDowell JE, Austin BP, Miller PH, Yanasak NE, Allison JD, Naglieri JA. Exercise improves executive function and achievement and alters brain activation in overweight children: a randomized, controlled trial. Health Psychol. 2011;30:91–98. doi: 10.1037/a0021766. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. De la Herran-Arita AK, Garcia-Garcia F. Narcolepsy as an Immune-Mediated Disease. Sleep Disord. 2014;2014:792687. doi: 10.1155/2014/792687. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. de Lecea L, Huerta R. Hypocretin (orexin) regulation of sleep-to-wake transitions. Front Pharmacol. 2014;5:16. doi: 10.3389/fphar.2014.00016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. de Lecea L, Kilduff TS, Peyron C, Gao X, Foye PE, Danielson PE, Fukuhara C, Battenberg EL, Gautvik VT, Bartlett FS, 2nd, 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. 1998;95:322–327. doi: 10.1073/pnas.95.1.322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. de Lecea L, Sutcliffe JG, Fabre V. Hypocretins/orexins as integrators of physiological information: lessons from mutant animals. Neuropeptides. 2002;36:85–95. doi: 10.1054/npep.2002.0892. [DOI] [PubMed] [Google Scholar]
  47. 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. 2007;27:14239–14247. doi: 10.1523/JNEUROSCI.3878-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. 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. 2004;27:851–856. doi: 10.1093/sleep/27.5.851. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Dong XS, Ma SF, Cao CW, Li J, An P, Zhao L, Liu NY, Yan H, Hu QT, Mignot E, Strohl KP, Gao ZC, Zeng C, Han F. Hypocretin (orexin) neuropeptide precursor gene, HCRT, polymorphisms in early-onset narcolepsy with cataplexy. Sleep Med. 2013;14:482–487. doi: 10.1016/j.sleep.2013.01.016. [DOI] [PubMed] [Google Scholar]
  50. Downs JL, Dunn MR, Borok E, Shanabrough M, Horvath TL, Kohama SG, Urbanski HF. Orexin neuronal changes in the locus coeruleus of the aging rhesus macaque. Neurobiol Aging. 2007;28:1286–1295. doi: 10.1016/j.neurobiolaging.2006.05.025. [DOI] [PubMed] [Google Scholar]
  51. Drake CL, Roehrs T, Roth T. Insomnia causes, consequences, and therapeutics: an overview. Depress Anxiety. 2003;18:163–176. doi: 10.1002/da.10151. [DOI] [PubMed] [Google Scholar]
  52. Dyken ME, Yamada T. Narcolepsy and disorders of excessive somnolence. Prim Care. 2005;32:389–413. doi: 10.1016/j.pop.2005.02.012. [DOI] [PubMed] [Google Scholar]
  53. El-Sedeek M, Korish AA, Deef MM. Plasma orexin-A levels in postmenopausal women: possible interaction with estrogen and correlation with cardiovascular risk status. BJOG. 2010;117:488–492. doi: 10.1111/j.1471-0528.2009.02474.x. [DOI] [PubMed] [Google Scholar]
  54. Elia M, Ritz P, Stubbs RJ. Total energy expenditure in the elderly. Eur J Clin Nutr. 2000;54(Suppl 3):S92–103. doi: 10.1038/sj.ejcn.1601030. [DOI] [PubMed] [Google Scholar]
  55. España RA, Reis KM, Valentino RJ, Berridge CW. Organization of hypocretin/orexin efferents to locus coeruleus and basal forebrain arousal-related structures. J Comp Neurol. 2005;481:160–178. doi: 10.1002/cne.20369. [DOI] [PubMed] [Google Scholar]
  56. Etnier JL, Nowell PM, Landers DM, Sibley BA. A meta-regression to examine the relationship between aerobic fitness and cognitive performance. Brain Res Rev. 2006;52:119–130. doi: 10.1016/j.brainresrev.2006.01.002. [DOI] [PubMed] [Google Scholar]
  57. Fadel J, Bubser M, Deutch AY. Differential activation of orexin neurons by antipsychotic drugs associated with weight gain. J Neurosci. 2002;22:6742–6746. doi: 10.1523/JNEUROSCI.22-15-06742.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Fadel J, Pasumarthi R, Reznikov LR. Stimulation of cortical acetylcholine release by orexin A. Neuroscience. 2005;130:541–547. doi: 10.1016/j.neuroscience.2004.09.050. [DOI] [PubMed] [Google Scholar]
  59. Fadel JR, Jolivalt CG, Reagan LP. Food for thought: the role of appetitive peptides in age-related cognitive decline. Ageing Res Rev. 2013;12:764–776. doi: 10.1016/j.arr.2013.01.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Feng Y, Liu T, Li XQ, Liu Y, Zhu XY, Jankovic J, Pan TH, Wu YC. Neuroprotection by Orexin-A via HIF-1alpha induction in a cellular model of Parkinson’s disease. Neurosci Lett. 2014;579C:35–40. doi: 10.1016/j.neulet.2014.07.014. [DOI] [PubMed] [Google Scholar]
  61. Ferreiro E, Baldeiras I, Ferreira IL, Costa RO, Rego AC, Pereira CF, Oliveira CR. Mitochondrial- and endoplasmic reticulum-associated oxidative stress in Alzheimer’s disease: from pathogenesis to biomarkers. Int J Cell Biol. 2012;2012:735206. doi: 10.1155/2012/735206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Fontana A, Gast H, Reith W, Recher M, Birchler T, Bassetti CL. Narcolepsy: autoimmunity, effector T cell activation due to infection, or T cell independent, major histocompatibility complex class II induced neuronal loss? Brain. 2010;133:1300–1311. doi: 10.1093/brain/awq086. [DOI] [PubMed] [Google Scholar]
  63. Frederick-Duus D, Guyton MF, Fadel J. Food-elicited increases in cortical acetylcholine release require orexin transmission. Neuroscience. 2007;149:499–507. doi: 10.1016/j.neuroscience.2007.07.061. [DOI] [PubMed] [Google Scholar]
  64. Fronczek R, Baumann CR, Lammers GJ, Bassetti CL, Overeem S. Hypocretin/orexin disturbances in neurological disorders. Sleep Med Rev. 2009;13:9–22. doi: 10.1016/j.smrv.2008.05.002. [DOI] [PubMed] [Google Scholar]
  65. 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. 2007;130:1577–1585. doi: 10.1093/brain/awm090. [DOI] [PubMed] [Google Scholar]
  66. Fronczek R, van Geest S, Frolich M, Overeem S, Roelandse FW, Lammers GJ, Swaab DF. Hypocretin (orexin) loss in Alzheimer’s disease. Neurobiol Aging. 2012;33:1642–1650. doi: 10.1016/j.neurobiolaging.2011.03.014. [DOI] [PubMed] [Google Scholar]
  67. Fujiki N, Yoshida Y, Zhang S, Sakurai T, Yanagisawa M, Nishino S. Sex difference in body weight gain and leptin signaling in hypocretin/orexin deficient mouse models. Peptides. 2006;27:2326–2331. doi: 10.1016/j.peptides.2006.03.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Funato H, Tsai AL, Willie JT, Kisanuki Y, Williams SC, Sakurai T, Yanagisawa M. Enhanced orexin receptor-2 signaling prevents diet-induced obesity and improves leptin sensitivity. Cell Metab. 2009;9:64–76. doi: 10.1016/j.cmet.2008.10.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Gallone S, Boschi S, Rubino E, De Martino P, Scarpini E, Galimberti D, Fenoglio C, Acutis PL, Maniaci MG, Pinessi L, Rainero I. Is HCRTR2 a Genetic Risk Factor for Alzheimer’s Disease? Dement Geriatr Cogn Disord. 2014;38:245–253. doi: 10.1159/000359964. [DOI] [PubMed] [Google Scholar]
  70. 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. 2003a;116:223–235. doi: 10.1016/s0306-4522(02)00575-4. [DOI] [PubMed] [Google Scholar]
  71. Gerashchenko D, Kohls MD, Greco M, Waleh NS, Salin-Pascual R, Kilduff TS, Lappi DA, Shiromani PJ. Hypocretin-2-saporin lesions of the lateral hypothalamus produce narcoleptic-like sleep behavior in the rat. J Neurosci. 2001;21:7273–7283. doi: 10.1523/JNEUROSCI.21-18-07273.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. 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. 2003b;184:1010–1016. doi: 10.1016/S0014-4886(03)00388-1. [DOI] [PubMed] [Google Scholar]
  73. 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. 2001;30:345–354. doi: 10.1016/s0896-6273(01)00293-8. [DOI] [PubMed] [Google Scholar]
  74. Hara J, Yanagisawa M, Sakurai T. Difference in obesity phenotype between orexin-knockout mice and orexin neuron-deficient mice with same genetic background and environmental conditions. Neurosci Lett. 2005;380:239–242. doi: 10.1016/j.neulet.2005.01.046. [DOI] [PubMed] [Google Scholar]
  75. Haynes AC, Chapman H, Taylor C, Moore GB, Cawthorne MA, Tadayyon M, Clapham JC, Arch JR. Anorectic, thermogenic and anti-obesity activity of a selective orexin-1 receptor antagonist in ob/ob mice. Regul Pept. 2002;104:153–159. doi: 10.1016/s0167-0115(01)00358-5. [DOI] [PubMed] [Google Scholar]
  76. Haynes AC, Jackson B, Chapman H, Tadayyon M, Johns A, Porter RA, Arch JR. A selective orexin-1 receptor antagonist reduces food consumption in male and female rats. Regul Pept. 2000;96:45–51. doi: 10.1016/s0167-0115(00)00199-3. [DOI] [PubMed] [Google Scholar]
  77. Heinonen MV, Purhonen AK, Miettinen P, Paakkonen M, Pirinen E, Alhava E, Akerman K, Herzig KH. Apelin, orexin-A and leptin plasma levels in morbid obesity and effect of gastric banding. Regul Pept. 2005;130:7–13. doi: 10.1016/j.regpep.2005.05.003. [DOI] [PubMed] [Google Scholar]
  78. Herring WJ, Snyder E, Budd K, Hutzelmann J, Snavely D, Liu K, Lines C, Roth T, Michelson D. Orexin receptor antagonism for treatment of insomnia: a randomized clinical trial of suvorexant. Neurology. 2012;79:2265–2274. doi: 10.1212/WNL.0b013e31827688ee. [DOI] [PubMed] [Google Scholar]
  79. Hirota K, Kushikata T, Kudo M, Kudo T, Smart D, Matsuki A. Effects of central hypocretin-1 administration on hemodynamic responses in young-adult and middle-aged rats. Brain Res. 2003;981:143–150. doi: 10.1016/s0006-8993(03)03002-6. [DOI] [PubMed] [Google Scholar]
  80. Holowatz LA, Kenney WL. Peripheral mechanisms of thermoregulatory control of skin blood flow in aged humans. J Appl Physiol (1985) 2010;109:1538–1544. doi: 10.1152/japplphysiol.00338.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Hong SC, Leen K, Park SA, Han JH, Lee SP, Lin L, Okun M, Nishino S, Mignot E. HLA and hypocretin studies in Korean patients with narcolepsy. Sleep. 2002;25:440–444. [PubMed] [Google Scholar]
  82. Hor H, Kutalik Z, Dauvilliers Y, Valsesia A, Lammers GJ, Donjacour CE, Iranzo A, Santamaria J, Peraita Adrados R, Vicario JL, Overeem S, Arnulf I, Theodorou I, Jennum P, Knudsen S, Bassetti C, Mathis J, Lecendreux M, Mayer G, Geisler P, Beneto A, Petit B, Pfister C, Burki JV, Didelot G, Billiard M, Ercilla G, Verduijn W, Claas FH, Vollenweider P, Waeber G, Waterworth DM, Mooser V, Heinzer R, Beckmann JS, Bergmann S, Tafti M. Genome-wide association study identifies new HLA class II haplotypes strongly protective against narcolepsy. Nat Genet. 2010;42:786–789. doi: 10.1038/ng.647. [DOI] [PubMed] [Google Scholar]
  83. Horne J. Exercise benefits for the aging brain depend on the accompanying cognitive load: insights from sleep electroencephalogram. Sleep Med. 2013;14:1208–1213. doi: 10.1016/j.sleep.2013.05.019. [DOI] [PubMed] [Google Scholar]
  84. Huh Y, Yang EJ, Lee SA, Lim JY, Kim KW, Paik NJ. Association between executive function and physical performance in older Korean adults: findings from the Korean Longitudinal Study on Health and Aging (KLoSHA) Arch Gerontol Geriatr. 2011;52:e156–161. doi: 10.1016/j.archger.2010.10.018. [DOI] [PubMed] [Google Scholar]
  85. Hunt NJ, Rodriguez ML, Waters KA, Machaalani R. Changes in orexin (hypocretin) neuronal expression with normal aging in the human hypothalamus. Neurobiol Aging. 2014 doi: 10.1016/j.neurobiolaging.2014.08.010. [DOI] [PubMed] [Google Scholar]
  86. Husain M, Mehta MA. Cognitive enhancement by drugs in health and disease. Trends Cogn Sci. 2011;15:28–36. doi: 10.1016/j.tics.2010.11.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. 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. 2009;326:1005–1007. doi: 10.1126/science.1180962. [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Kappeler L, Gourdji D, Zizzari P, Bluet-Pajot MT, Epelbaum J. Age-associated changes in hypothalamic and pituitary neuroendocrine gene expression in the rat. J Neuroendocrinol. 2003;15:592–601. doi: 10.1046/j.1365-2826.2003.01039.x. [DOI] [PubMed] [Google Scholar]
  89. Kappeler L, Zizzari P, Alliot J, Epelbaum J, Bluet-Pajot MT. Delayed age-associated decrease in growth hormone pulsatile secretion and increased orexigenic peptide expression in the Lou C/JaLL rat. Neuroendocrinology. 2004;80:273–283. doi: 10.1159/000083610. [DOI] [PubMed] [Google Scholar]
  90. Karnani M, Burdakov D. Multiple hypothalamic circuits sense and regulate glucose levels. Am J Physiol Regul Integr Comp Physiol. 2011;300:R47–55. doi: 10.1152/ajpregu.00527.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Katsuki H, Michinaga S. Anti-Parkinson drugs and orexin neurons. Vitam Horm. 2012;89:279–290. doi: 10.1016/B978-0-12-394623-2.00015-9. [DOI] [PubMed] [Google Scholar]
  92. Kenney WL, Buskirk ER. Functional consequences of sarcopenia: effects on thermoregulation. J Gerontol A Biol Sci Med Sci. 1995;50(Spec No):78–85. doi: 10.1093/gerona/50a.special_issue.78. [DOI] [PubMed] [Google Scholar]
  93. Kessler BA, Stanley EM, Frederick-Duus D, Fadel J. Age-related loss of orexin/hypocretin neurons. Neuroscience. 2011;178:82–88. doi: 10.1016/j.neuroscience.2011.01.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Kirchgessner AL. Orexins in the brain-gut axis. Endocr Rev. 2002;23:1–15. doi: 10.1210/edrv.23.1.0454. [DOI] [PubMed] [Google Scholar]
  95. Kirchgessner AL, Liu M. Orexin synthesis and response in the gut. Neuron. 1999;24:941–951. doi: 10.1016/s0896-6273(00)81041-7. [DOI] [PubMed] [Google Scholar]
  96. Kisanuki YY, Chemelli RM, Sinton CM, Richardson JA, Hammer RE, Yanagisawa M. The Role of Orexin Receptor Type-1 (OX1R) in the Regulation of Sleep. Sleep. 2000;23:A91. [Google Scholar]
  97. Kmiec Z. Central regulation of food intake in ageing. J Physiol Pharmacol. 2006;57(Suppl 6):7–16. [PubMed] [Google Scholar]
  98. Kmiec Z. Aging and peptide control of food intake. Curr Protein Pept Sci. 2011;12:271–279. doi: 10.2174/138920311795906718. [DOI] [PubMed] [Google Scholar]
  99. Kmiec Z, Petervari E, Balasko M, Szekely M. Anorexia of aging. Vitam Horm. 2013;92:319–355. doi: 10.1016/B978-0-12-410473-0.00013-1. [DOI] [PubMed] [Google Scholar]
  100. Kok SW, Overeem S, Visscher TL, Lammers GJ, Seidell JC, Pijl H, Meinders AE. Hypocretin deficiency in narcoleptic humans is associated with abdominal obesity. Obes Res. 2003;11:1147–1154. doi: 10.1038/oby.2003.156. [DOI] [PubMed] [Google Scholar]
  101. Komaki G, Matsumoto Y, Nishikata H, Kawai K, Nozaki T, Takii M, Sogawa H, Kubo C. Orexin-A and leptin change inversely in fasting non-obese subjects. Eur J Endocrinol. 2001;144:645–651. doi: 10.1530/eje.0.1440645. [DOI] [PubMed] [Google Scholar]
  102. Kornum BR, Faraco J, Mignot E. Narcolepsy with hypocretin/orexin deficiency, infections and autoimmunity of the brain. Curr Opin Neurobiol. 2011;21:897–903. doi: 10.1016/j.conb.2011.09.003. [DOI] [PubMed] [Google Scholar]
  103. Kotz C, Nixon J, Butterick T, Perez-Leighton C, Teske J, Billington C. Brain orexin promotes obesity resistance. Ann N Y Acad Sci. 2012;1264:72–86. doi: 10.1111/j.1749-6632.2012.06585.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Kotz CM, Mullett MA, Wang C. Diminished feeding responsiveness to orexin A (hypocretin 1) in aged rats is accompanied by decreased neuronal activation. Am J Physiol Regul Integr Comp Physiol. 2005;289:R359–R366. doi: 10.1152/ajpregu.00717.2004. [DOI] [PubMed] [Google Scholar]
  105. Kotz CM, Teske JA, Billington CJ. Neuroregulation of nonexercise activity thermogenesis and obesity resistance. Am J Physiol Regul Integr Comp Physiol. 2008;294:R699–710. doi: 10.1152/ajpregu.00095.2007. [DOI] [PubMed] [Google Scholar]
  106. Kotz CM, Teske JA, Levine JA, Wang C. Feeding and activity induced by orexin A in the lateral hypothalamus in rats. Regul Pept. 2002;104:27–32. doi: 10.1016/s0167-0115(01)00346-9. [DOI] [PubMed] [Google Scholar]
  107. Kotz CM, Wang C, Teske JA, Thorpe AJ, Novak CM, Kiwaki K, Levine JA. Orexin A mediation of time spent moving in rats: neural mechanisms. Neuroscience. 2006;142:29–36. doi: 10.1016/j.neuroscience.2006.05.028. [DOI] [PubMed] [Google Scholar]
  108. Kroeger D, de Lecea L. The hypocretins and their role in narcolepsy. CNS Neurol Disord Drug Targets. 2009;8:271–280. doi: 10.2174/187152709788921645. [DOI] [PubMed] [Google Scholar]
  109. 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. 1999;98:365–376. doi: 10.1016/s0092-8674(00)81965-0. [DOI] [PubMed] [Google Scholar]
  110. Lin L, Hungs M, Mignot E. Narcolepsy and the HLA region. J Neuroimmunol. 2001;117:9–20. doi: 10.1016/s0165-5728(01)00333-2. [DOI] [PubMed] [Google Scholar]
  111. Lin L, Wisor J, Shiba T, Taheri S, Yanai K, Wurts S, Lin X, Vitaterna M, Takahashi J, Lovenberg TW, Koehl M, Uhl G, Nishino S, Mignot E. Measurement of hypocretin/orexin content in the mouse brain using an enzyme immunoassay: the effect of circadian time, age and genetic background. Peptides. 2002;23:2203–2211. doi: 10.1016/s0196-9781(02)00251-6. [DOI] [PubMed] [Google Scholar]
  112. Loeffler DA, Connor JR, Juneau PL, Snyder BS, Kanaley L, DeMaggio AJ, Nguyen H, Brickman CM, LeWitt PA. Transferrin and iron in normal, Alzheimer’s disease, and Parkinson’s disease brain regions. J Neurochem. 1995;65:710–724. doi: 10.1046/j.1471-4159.1995.65020710.x. [DOI] [PubMed] [Google Scholar]
  113. Lubkin M, Stricker-Krongrad A. Independent feeding and metabolic actions of orexins in mice. Biochem Biophys Res Commun. 1998;253:241–245. doi: 10.1006/bbrc.1998.9750. [DOI] [PubMed] [Google Scholar]
  114. Mahler SV, Smith RJ, Moorman DE, Sartor GC, Aston-Jones G. Multiple roles for orexin/hypocretin in addiction. Prog Brain Res. 2012;198:79–121. doi: 10.1016/B978-0-444-59489-1.00007-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Malhotra S, Kushida CA. Primary hypersomnias of central origin. Continuum (Minneap Minn) 2013;19:67–85. doi: 10.1212/01.CON.0000427212.05930.c4. [DOI] [PubMed] [Google Scholar]
  116. Manini TM. Energy expenditure and aging. Ageing Res Rev. 2010;9:1–11. doi: 10.1016/j.arr.2009.08.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  117. 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. 2001;435:6–25. doi: 10.1002/cne.1190. [DOI] [PubMed] [Google Scholar]
  118. Martinez-Rodriguez JE, Lin L, Iranzo A, Genis D, Marti MJ, Santamaria J, Mignot E. Decreased hypocretin-1 (Orexin-A) levels in the cerebrospinal fluid of patients with myotonic dystrophy and excessive daytime sleepiness. Sleep. 2003;26:287–290. doi: 10.1093/sleep/26.3.287. [DOI] [PubMed] [Google Scholar]
  119. Martone AM, Onder G, Vetrano DL, Ortolani E, Tosato M, Marzetti E, Landi F. Anorexia of aging: a modifiable risk factor for frailty. Nutrients. 2013;5:4126–4133. doi: 10.3390/nu5104126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  120. Matsumura T, Nakayama M, Nomura A, Naito A, Kamahara K, Kadono K, Inoue M, Homma T, Sekizawa K. Age-related changes in plasma orexin-A concentrations. Exp Gerontol. 2002;37:1127–1130. doi: 10.1016/s0531-5565(02)00092-x. [DOI] [PubMed] [Google Scholar]
  121. Mavanji V, Teske JA, Billington CJ, Kotz CM. Elevated sleep quality and orexin receptor mRNA in obesity-resistant rats. Int J Obes (Lond) 2010;34:1576–1588. doi: 10.1038/ijo.2010.93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  122. Michelson D, Snyder E, Paradis E, Chengan-Liu M, Snavely DB, Hutzelmann J, Walsh JK, Krystal AD, Benca RM, Cohn M, Lines C, Roth T, Herring WJ. Safety and efficacy of suvorexant during 1-year treatment of insomnia with subsequent abrupt treatment discontinuation: a phase 3 randomised, double-blind, placebo-controlled trial. Lancet Neurol. 2014;13:461–471. doi: 10.1016/S1474-4422(14)70053-5. [DOI] [PubMed] [Google Scholar]
  123. Mieda M, Hasegawa E, Kisanuki YY, Sinton CM, Yanagisawa M, Sakurai T. Differential roles of orexin receptor-1 and -2 in the regulation of non-REM and REM sleep. J Neurosci. 2011;31:6518–6526. doi: 10.1523/JNEUROSCI.6506-10.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. Mignot E. Genetic and familial aspects of narcolepsy. Neurology. 1998;50:S16–22. doi: 10.1212/wnl.50.2_suppl_1.s16. [DOI] [PubMed] [Google Scholar]
  125. Mignot E. Sleep, sleep disorders and hypocretin (orexin) Sleep Med. 2004;5(Suppl 1):S2–8. doi: 10.1016/s1389-9457(04)90001-9. [DOI] [PubMed] [Google Scholar]
  126. Mignot E, Hayduk R, Black J, Grumet FC, Guilleminault C. HLA DQB1*0602 is associated with cataplexy in 509 narcoleptic patients. Sleep. 1997;20:1012–1020. [PubMed] [Google Scholar]
  127. Mignot E, Lammers GJ, Ripley B, Okun M, Nevsimalova S, Overeem S, Vankova J, Black J, Harsh J, Bassetti C, Schrader H, Nishino S. The role of cerebrospinal fluid hypocretin measurement in the diagnosis of narcolepsy and other hypersomnias. Arch Neurol. 2002;59:1553–1562. doi: 10.1001/archneur.59.10.1553. [DOI] [PubMed] [Google Scholar]
  128. Mignot E, Lin L, Rogers W, Honda Y, Qiu X, Lin X, Okun M, Hohjoh H, Miki T, Hsu S, Leffell M, Grumet F, Fernandez-Vina M, Honda M, Risch N. Complex HLA-DR and -DQ interactions confer risk of narcolepsy-cataplexy in three ethnic groups. Am J Hum Genet. 2001;68:686–699. doi: 10.1086/318799. [DOI] [PMC free article] [PubMed] [Google Scholar]
  129. Morairty SR, Wisor J, Silveira K, Sinko W, Kilduff TS. The wake-promoting effects of hypocretin-1 are attenuated in old rats. Neurobiol Aging. 2011;32:1514–1527. doi: 10.1016/j.neurobiolaging.2009.07.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  130. Morgan RE, Crowley JM, Smith RH, LaRoche RB, Dopheide MM. Modafinil improves attention, inhibitory control, and reaction time in healthy, middle-aged rats. Pharmacol Biochem Behav. 2007;86:531–541. doi: 10.1016/j.pbb.2007.01.015. [DOI] [PubMed] [Google Scholar]
  131. Nestrasil I, Michaeli S, Liimatainen T, Rydeen CE, Kotz CM, Nixon JP, Hanson T, Tuite PJ. T1rho and T2rho MRI in the evaluation of Parkinson’s disease. J Neurol. 2010;257:964–968. doi: 10.1007/s00415-009-5446-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. Niranjan R. The Role of Inflammatory and Oxidative Stress Mechanisms in the Pathogenesis of Parkinson’s Disease: Focus on Astrocytes. Mol Neurobiol. 2013 doi: 10.1007/s12035-013-8483-x. [DOI] [PubMed] [Google Scholar]
  133. Nishino S, Kanbayashi T, Fujiki N, Uchino M, Ripley B, Watanabe M, Lammers GJ, Ishiguro H, Shoji S, Nishida Y, Overeem S, Toyoshima I, Yoshida Y, Shimizu T, Taheri S, Mignot E. CSF hypocretin levels in Guillain-Barre syndrome and other inflammatory neuropathies. Neurology. 2003;61:823–825. doi: 10.1212/01.wnl.0000081049.14098.50. [DOI] [PubMed] [Google Scholar]
  134. Nishino S, Ripley B, Overeem S, Lammers GJ, Mignot E. Hypocretin (orexin) deficiency in human narcolepsy. Lancet. 2000;355:39–40. doi: 10.1016/S0140-6736(99)05582-8. [DOI] [PubMed] [Google Scholar]
  135. Nishino S, Ripley B, Overeem S, Nevsimalova S, Lammers GJ, Vankova J, Okun M, Rogers W, Brooks S, Mignot E. Low cerebrospinal fluid hypocretin (Orexin) and altered energy homeostasis in human narcolepsy. Ann Neurol. 2001;50:381–388. doi: 10.1002/ana.1130. [DOI] [PubMed] [Google Scholar]
  136. 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. 2007;3:28. doi: 10.1186/1744-9081-3-28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  137. Novak CM, Kotz CM, Levine JA. Central orexin sensitivity, physical activity, and obesity in diet-induced obese and diet-resistant rats. Am J Physiol Endocrinol Metab. 2006;290:E396–403. doi: 10.1152/ajpendo.00293.2005. [DOI] [PubMed] [Google Scholar]
  138. Novak CM, Levine JA. Central neural and endocrine mechanisms of non-exercise activity thermogenesis and their potential impact on obesity. J Neuroendocrinol. 2007;19:923–940. doi: 10.1111/j.1365-2826.2007.01606.x. [DOI] [PubMed] [Google Scholar]
  139. Novak CM, Levine JA. Daily intraparaventricular orexin-A treatment induces weight loss in rats. Obesity (Silver Spring) 2009;17:1493–1498. doi: 10.1038/oby.2009.91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  140. Overeem S, Mignot E, van Dijk JG, Lammers GJ. Narcolepsy: clinical features, new pathophysiologic insights, and future perspectives. J Clin Neurophysiol. 2001;18:78–105. doi: 10.1097/00004691-200103000-00002. [DOI] [PubMed] [Google Scholar]
  141. Pasumarthi RK, Fadel J. Activation of orexin/hypocretin projections to basal forebrain and paraventricular thalamus by acute nicotine. Brain Res Bull. 2008;77:367–373. doi: 10.1016/j.brainresbull.2008.09.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  142. Perez-Leighton CE, Boland K, Billington CJ, Kotz CM. High and low activity rats: elevated intrinsic physical activity drives resistance to diet-induced obesity in non-bred rats. Obesity (Silver Spring) 2013;21:353–360. doi: 10.1002/oby.20045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  143. Perez-Leighton CE, Boland K, Teske JA, Billington C, Kotz CM. Behavioral responses to orexin, orexin receptor gene expression, and spontaneous physical activity contribute to individual sensitivity to obesity. Am J Physiol Endocrinol Metab. 2012;303:E865–874. doi: 10.1152/ajpendo.00119.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  144. Petersen A, Gil J, Maat-Schieman ML, Bjorkqvist M, Tanila H, Araujo IM, Smith R, Popovic N, Wierup N, Norlen P, Li JY, Roos RA, Sundler F, Mulder H, Brundin P. Orexin loss in Huntington’s disease. Hum Mol Genet. 2005;14:39–47. doi: 10.1093/hmg/ddi004. [DOI] [PubMed] [Google Scholar]
  145. 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. 2000;6:991–997. doi: 10.1038/79690. [DOI] [PubMed] [Google Scholar]
  146. Peyron C, Tighe DK, van den Pol AN, de Lecea L, Heller HC, Sutcliffe JG, Kilduff TS. Neurons containing hypocretin (orexin) project to multiple neuronal systems. J Neurosci. 1998;18:9996–10015. doi: 10.1523/JNEUROSCI.18-23-09996.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  147. Poehlman ET, Horton ES. Regulation of energy expenditure in aging humans. Annu Rev Nutr. 1990;10:255–275. doi: 10.1146/annurev.nu.10.070190.001351. [DOI] [PubMed] [Google Scholar]
  148. Porkka-Heiskanen T, Alanko L, Kalinchuk A, Heiskanen S, Stenberg D. The effect of age on prepro-orexin gene expression and contents of orexin A and B in the rat brain. Neurobiol Aging. 2004a;25:231–238. doi: 10.1016/s0197-4580(03)00043-5. [DOI] [PubMed] [Google Scholar]
  149. Porkka-Heiskanen T, Kalinchuk A, Alanko L, Huhtaniemi I, Stenberg D. Orexin A and B levels in the hypothalamus of female rats: the effects of the estrous cycle and age. Eur J Endocrinol. 2004b;150:737–742. doi: 10.1530/eje.0.1500737. [DOI] [PubMed] [Google Scholar]
  150. Riemann D, Spiegelhalder K, Feige B, Voderholzer U, Berger M, Perlis M, Nissen C. The hyperarousal model of insomnia: a review of the concept and its evidence. Sleep Med Rev. 2010;14:19–31. doi: 10.1016/j.smrv.2009.04.002. [DOI] [PubMed] [Google Scholar]
  151. 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. 2001;57:2253–2258. doi: 10.1212/wnl.57.12.2253. [DOI] [PubMed] [Google Scholar]
  152. Ritchie C, Okuro M, Kanbayashi T, Nishino S. Hypocretin ligand deficiency in narcolepsy: recent basic and clinical insights. Curr Neurol Neurosci Rep. 2010;10:180–189. doi: 10.1007/s11910-010-0100-z. [DOI] [PubMed] [Google Scholar]
  153. Roh JH, Huang Y, Bero AW, Kasten T, Stewart FR, Bateman RJ, Holtzman DM. Disruption of the sleep-wake cycle and diurnal fluctuation of beta-amyloid in mice with Alzheimer’s disease pathology. Sci Transl Med. 2012;4:150ra122. doi: 10.1126/scitranslmed.3004291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  154. 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. 1998;92:573–585. doi: 10.1016/s0092-8674(00)80949-6. [DOI] [PubMed] [Google Scholar]
  155. Satoh A, Brace CS, Ben-Josef G, West T, Wozniak DF, Holtzman DM, Herzog ED, Imai S. SIRT1 promotes the central adaptive response to diet restriction through activation of the dorsomedial and lateral nuclei of the hypothalamus. J Neurosci. 2010;30:10220–10232. doi: 10.1523/JNEUROSCI.1385-10.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  156. Satoh A, Brace CS, Rensing N, Cliften P, Wozniak DF, Herzog ED, Yamada KA, Imai S. Sirt1 extends life span and delays aging in mice through the regulation of Nk2 homeobox 1 in the DMH and LH. Cell Metab. 2013;18:416–430. doi: 10.1016/j.cmet.2013.07.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  157. Sawai N, Ueta Y, Nakazato M, Ozawa H. Developmental and aging change of orexin-A and -B immunoreactive neurons in the male rat hypothalamus. Neurosci Lett. 2010;468:51–55. doi: 10.1016/j.neulet.2009.10.061. [DOI] [PubMed] [Google Scholar]
  158. Scammell TE, Estabrooke IV, McCarthy MT, Chemelli RM, Yanagisawa M, Miller MS, Saper CB. Hypothalamic arousal regions are activated during modafinil-induced wakefulness. J Neurosci. 2000;20:8620–8628. doi: 10.1523/JNEUROSCI.20-22-08620.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  159. Scammell TE, Matheson JK, Honda M, Thannickal TC, Siegel JM. Coexistence of narcolepsy and Alzheimer’s disease. Neurobiol Aging. 2012;33:1318–1319. doi: 10.1016/j.neurobiolaging.2010.12.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  160. Schuld A, Hebebrand J, Geller F, Pollmacher T. Increased body-mass index in patients with narcolepsy. Lancet. 2000;355:1274–1275. doi: 10.1016/S0140-6736(05)74704-8. [DOI] [PubMed] [Google Scholar]
  161. Sellayah D, Sikder D. Orexin restores aging-related brown adipose tissue dysfunction in male mice. Endocrinology. 2014;155:485–501. doi: 10.1210/en.2013-1629. [DOI] [PubMed] [Google Scholar]
  162. Semenza GL. Hypoxia-inducible factor 1: oxygen homeostasis and disease pathophysiology. Trends Mol Med. 2001;7:345–350. doi: 10.1016/s1471-4914(01)02090-1. [DOI] [PubMed] [Google Scholar]
  163. Sharf R, Sarhan M, Brayton CE, Guarnieri DJ, Taylor JR, DiLeone RJ. Orexin signaling via the orexin 1 receptor mediates operant responding for food reinforcement. Biol Psychiatry. 2010;67:753–760. doi: 10.1016/j.biopsych.2009.12.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  164. Siegel JM. Narcolepsy: a key role for hypocretins (orexins) Cell. 1999;98:409–412. doi: 10.1016/s0092-8674(00)81969-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  165. Siegel JM, Boehmer LN. Narcolepsy and the hypocretin system--where motion meets emotion. Nat Clin Pract Neurol. 2006;2:548–556. doi: 10.1038/ncpneuro0300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  166. Sikder D, Kodadek T. The neurohormone orexin stimulates hypoxia-inducible factor-1 activity. Genes Dev. 2007;21:2995–3005. doi: 10.1101/gad.1584307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  167. Singh AK, Mahlios J, Mignot E. Genetic association, seasonal infections and autoimmune basis of narcolepsy. J Autoimmun. 2013;43:26–31. doi: 10.1016/j.jaut.2013.02.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  168. Slats D, Claassen JA, Verbeek MM, Overeem S. Reciprocal interactions between sleep, circadian rhythms and Alzheimer’s disease: focus on the role of hypocretin and melatonin. Ageing Res Rev. 2013;12:188–200. doi: 10.1016/j.arr.2012.04.003. [DOI] [PubMed] [Google Scholar]
  169. Sokolowska P, Urbanska A, Namiecinska M, Bieganska K, Zawilska JB. Orexins promote survival of rat cortical neurons. Neurosci Lett. 2012;506:303–306. doi: 10.1016/j.neulet.2011.11.028. [DOI] [PubMed] [Google Scholar]
  170. Stanley EM, Fadel J. Aging-related deficits in orexin/hypocretin modulation of the septohippocampal cholinergic system. Synapse. 2012;66:445–452. doi: 10.1002/syn.21533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  171. Strawn JR, Pyne-Geithman GJ, Ekhator NN, Horn PS, Uhde TW, Shutter LA, Baker DG, Geracioti TD., Jr Low cerebrospinal fluid and plasma orexin-A (hypocretin-1) concentrations in combat-related posttraumatic stress disorder. Psychoneuroendocrinology. 2010;35:1001–1007. doi: 10.1016/j.psyneuen.2010.01.001. [DOI] [PubMed] [Google Scholar]
  172. Tafti M, Maret S, Dauvilliers Y. Genes for normal sleep and sleep disorders. Ann Med. 2005;37:580–589. doi: 10.1080/07853890500372047. [DOI] [PubMed] [Google Scholar]
  173. Taheri S, Zeitzer JM, Mignot E. The role of hypocretins (orexins) in sleep regulation and narcolepsy. Annu Rev Neurosci. 2002;25:283–313. doi: 10.1146/annurev.neuro.25.112701.142826. [DOI] [PubMed] [Google Scholar]
  174. Takano S, Kanai S, Hosoya H, Ohta M, Uematsu H, Miyasaka K. Orexin-A does not stimulate food intake in old rats. Am J Physiol Gastrointest Liver Physiol. 2004;287:G1182–1187. doi: 10.1152/ajpgi.00218.2004. [DOI] [PubMed] [Google Scholar]
  175. Terao A, Apte-Deshpande A, Morairty S, Freund YR, Kilduff TS. Age-related decline in hypocretin (orexin) receptor 2 messenger RNA levels in the mouse brain. Neurosci Lett. 2002;332:190–194. doi: 10.1016/s0304-3940(02)00953-9. [DOI] [PubMed] [Google Scholar]
  176. Teske JA, Levine AS, Kuskowski M, Levine JA, Kotz CM. Elevated hypothalamic orexin signaling, sensitivity to orexin A, and spontaneous physical activity in obesity-resistant rats. Am J Physiol Regul Integr Comp Physiol. 2006;291:R889–899. doi: 10.1152/ajpregu.00536.2005. [DOI] [PubMed] [Google Scholar]
  177. Thakkar MM, Ramesh V, Cape EG, Winston S, Strecker RE, McCarley RW. REM sleep enhancement and behavioral cataplexy following orexin (hypocretin)-II receptor antisense perfusion in the pontine reticular formation. Sleep Res Online. 1999;2:112–120. [PubMed] [Google Scholar]
  178. Thannickal TC, Lai YY, Siegel JM. Hypocretin (orexin) cell loss in Parkinson’s disease. Brain. 2007;130:1586–1595. doi: 10.1093/brain/awm097. [DOI] [PMC free article] [PubMed] [Google Scholar]
  179. 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. 2000;27:469–474. doi: 10.1016/s0896-6273(00)00058-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  180. Thorpe AJ, Cleary JP, Levine AS, Kotz CM. Centrally administered orexin A increases motivation for sweet pellets in rats. Psychopharmacology (Berl) 2005;182:75–83. doi: 10.1007/s00213-005-0040-5. [DOI] [PubMed] [Google Scholar]
  181. Thorpe AJ, Kotz CM. Orexin A in the nucleus accumbens stimulates feeding and locomotor activity. Brain Res. 2005;1050:156–162. doi: 10.1016/j.brainres.2005.05.045. [DOI] [PubMed] [Google Scholar]
  182. Trivedi P, Yu H, MacNeil DJ, Van der Ploeg LH, Guan XM. Distribution of orexin receptor mRNA in the rat brain. FEBS Lett. 1998;438:71–75. doi: 10.1016/s0014-5793(98)01266-6. [DOI] [PubMed] [Google Scholar]
  183. Tsuneki H, Murata S, Anzawa Y, Soeda Y, Tokai E, Wada T, Kimura I, Yanagisawa M, Sakurai T, Sasaoka T. Age-related insulin resistance in hypothalamus and peripheral tissues of orexin knockout mice. Diabetologia. 2008;51:657–667. doi: 10.1007/s00125-008-0929-8. [DOI] [PubMed] [Google Scholar]
  184. Van Cauter E, Knutson KL. Sleep and the epidemic of obesity in children and adults. Eur J Endocrinol. 2008;159(Suppl 1):S59–66. doi: 10.1530/EJE-08-0298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  185. Vankova J, Stepanova I, Jech R, Elleder M, Ling L, Mignot E, Nishino S, Nevsimalova S. Sleep disturbances and hypocretin deficiency in Niemann-Pick disease type C. Sleep. 2003;26:427–430. doi: 10.1093/sleep/26.4.427. [DOI] [PubMed] [Google Scholar]
  186. Wang L, Larson EB, Bowen JD, van Belle G. Performance-based physical function and future dementia in older people. Arch Intern Med. 2006;166:1115–1120. doi: 10.1001/archinte.166.10.1115. [DOI] [PubMed] [Google Scholar]
  187. Wang W, Pan Y, Li Q, Wang L. Orexin: a potential role in the process of obstructive sleep apnea. Peptides. 2013;42:48–54. doi: 10.1016/j.peptides.2013.01.001. [DOI] [PubMed] [Google Scholar]
  188. Weinreb O, Mandel S, Youdim MB, Amit T. Targeting dysregulation of brain iron homeostasis in Parkinson’s disease by iron chelators. Free Radic Biol Med. 2013;62:52–64. doi: 10.1016/j.freeradbiomed.2013.01.017. [DOI] [PubMed] [Google Scholar]
  189. 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. 2012;21:710–717. doi: 10.1111/j.1365-2869.2012.01027.x. [DOI] [PubMed] [Google Scholar]
  190. 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. 2003;38:715–730. doi: 10.1016/s0896-6273(03)00330-1. [DOI] [PubMed] [Google Scholar]
  191. Willie JT, Chemelli RM, Sinton CM, Yanagisawa M. To eat or to sleep? Orexin in the regulation of feeding and wakefulness. Annu Rev Neurosci. 2001;24:429–458. doi: 10.1146/annurev.neuro.24.1.429. [DOI] [PubMed] [Google Scholar]
  192. Wimmer ME, Rising J, Galante RJ, Wyner A, Pack AI, Abel T. Aging in mice reduces the ability to sustain sleep/wake states. PLoS ONE. 2013;8:e81880. doi: 10.1371/journal.pone.0081880. [DOI] [PMC free article] [PubMed] [Google Scholar]
  193. Wisor JP, Kilduff TS. Molecular genetic advances in sleep research and their relevance to sleep medicine. Sleep. 2005;28:357–367. [PubMed] [Google Scholar]
  194. 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. 2001;14:1075–1081. doi: 10.1046/j.0953-816x.2001.01725.x. [DOI] [PubMed] [Google Scholar]
  195. Yuan LB, Dong HL, Zhang HP, Zhao RN, Gong G, Chen XM, Zhang LN, Xiong L. Neuroprotective effect of orexin-A is mediated by an increase of hypoxia-inducible factor-1 activity in rat. Anesthesiology. 2011;114:340–354. doi: 10.1097/ALN.0b013e318206ff6f. [DOI] [PubMed] [Google Scholar]
  196. Zeitzer JM. Control of sleep and wakefulness in health and disease. Prog Mol Biol Transl Sci. 2013;119:137–154. doi: 10.1016/B978-0-12-396971-2.00006-3. [DOI] [PubMed] [Google Scholar]
  197. Zeitzer JM, Buckmaster CL, Parker KJ, Hauck CM, Lyons DM, Mignot E. Circadian and homeostatic regulation of hypocretin in a primate model: implications for the consolidation of wakefulness. J Neurosci. 2003;23:3555–3560. doi: 10.1523/JNEUROSCI.23-08-03555.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  198. Zhang JH, Sampogna S, Morales FR, Chase MH. Age-related changes in hypocretin (orexin) immunoreactivity in the cat brainstem. Brain Res. 2002;930:206–211. doi: 10.1016/s0006-8993(02)02240-0. [DOI] [PubMed] [Google Scholar]
  199. Zhang JH, Sampogna S, Morales FR, Chase MH. Age-related changes of hypocretin in basal forebrain of guinea pig. Peptides. 2005a;26:2590–2596. doi: 10.1016/j.peptides.2005.05.003. [DOI] [PubMed] [Google Scholar]
  200. Zhang JH, Sampogna S, Morales FR, Chase MH. Age-related ultrastructural changes in hypocretinergic terminals in the brainstem and spinal cord of cats. Neurosci Lett. 2005b;373:171–174. doi: 10.1016/j.neulet.2003.08.085. [DOI] [PubMed] [Google Scholar]
  201. Zhang S, Lin L, Kaur S, Thankachan S, Blanco-Centurion C, Yanagisawa M, Mignot E, Shiromani PJ. The development of hypocretin (orexin) deficiency in hypocretin/ataxin-3 transgenic rats. Neuroscience. 2007a;148:34–43. doi: 10.1016/j.neuroscience.2007.05.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  202. Zhang S, Zeitzer JM, Sakurai T, Nishino S, Mignot E. Sleep/wake fragmentation disrupts metabolism in a mouse model of narcolepsy. J Physiol. 2007b;581:649–663. doi: 10.1113/jphysiol.2007.129510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  203. 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. 2006;126–127:139–145. doi: 10.1016/j.autneu.2006.02.021. [DOI] [PubMed] [Google Scholar]

RESOURCES