Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2024 Mar 28.
Published in final edited form as: Int Rev Neurobiol. 2023 Jan 13;168:93–175. doi: 10.1016/bs.irn.2022.11.002

Metabotropic glutamate receptor function and regulation of sleep-wake cycles

Kimberly M Holter 1, Bethany E Pierce 1, Robert W Gould 1,*
PMCID: PMC10973983  NIHMSID: NIHMS1977914  PMID: 36868636

Abstract

Metabotropic glutamate (mGlu) receptors are the most abundant family of G-protein coupled receptors and are widely expressed throughout the central nervous system (CNS). Alterations in glutamate homeostasis, including dysregulations in mGlu receptor function, have been indicated as key contributors to multiple CNS disorders. Fluctuations in mGlu receptor expression and function also occur across diurnal sleep-wake cycles. Sleep disturbances including insomnia are frequently comorbid with neuropsychiatric, neurodevelopmental, and neurodegenerative conditions. These often precede behavioral symptoms and/or correlate with symptom severity and relapse. Chronic sleep disturbances may also be a consequence of primary symptom progression and can exacerbate neurodegeneration in disorders including Alzheimer’s disease (AD). Thus, there is a bidirectional relationship between sleep disturbances and CNS disorders; disrupted sleep may serve as both a cause and a consequence of the disorder. Importantly, comorbid sleep disturbances are rarely a direct target of primary pharmacological treatments for neuropsychiatric disorders even though improving sleep can positively impact other symptom clusters. This chapter details known roles of mGlu receptor subtypes in both sleep-wake regulation and CNS disorders focusing on schizophrenia, major depressive disorder, post-traumatic stress disorder, AD, and substance use disorder (cocaine and opioid). In this chapter, preclinical electrophysiological, genetic, and pharmacological studies are described, and, when possible, human genetic, imaging, and post-mortem studies are also discussed. In addition to reviewing the important relationships between sleep, mGlu receptors, and CNS disorders, this chapter highlights the development of selective mGlu receptor ligands that hold promise for improving both primary symptoms and sleep disturbances.

1. Introduction

The word “sleep” holds different meaning and emotional weight for different individuals. While many people undergo an easy transition from waking to a peaceful state of altered consciousness, initiating and maintaining sleep is a fitful chore for others. Though there have been substantial advancements in understanding the underlying functions of homeostatic sleep, including its role in synaptic remodeling critical for memory consolidation (Manoach & Stickgold, 2019; Martin, Monroe, & Diering, 2019), many purposes remain unresolved. Although sleeping and waking states may be perceived as dichotomous processes, they are intimately intertwined. Acute sleep disruptions lead to decreased daytime arousal and cognitive impairments as well as increased fatigue or daytime somnolence. Further, chronic sleep disruptions including insomnia are often comorbid with neuropsychiatric, neurodevelopmental, and neurodegenerative disorders (Benca & Buysse, 2018; Benca, William, Thisted, & Gillin, 1992; Riemann, Krone, Wulff, & Nissen, 2020; Sprecher, Ferrarelli, & Benca, 2015; Wulff, Gatti, Wettstein, & Foster, 2010). Increasing evidence suggests a direct relationship between sleep disruptions and symptom onset and/or severity in central nervous system (CNS) disorders (Reeve, Sheaves, & Freeman, 2015; Riemann et al., 2020; Veatch et al., 2017; Wulff et al., 2010). Importantly, the Diagnostics and Statistic Manual, 5th Edition (DSM-5) delineated insomnia, defined by difficulty falling asleep and/or staying asleep, as a separate diagnosis from other mental disorders as opposed to a primary or secondary symptom, and, thus, sleep disturbances should be a primary target of pharmacotherapies (APA, 2013; Benca & Buysse, 2018).

Sleep is an intricately regulated process involving multiple circuits and neurotransmitter systems. These systems work in concert to actively inhibit arousal-related processes and shift from asynchronous electrical activity during wake to synchronous, slow oscillatory activity during sleep (Scammell, Arrigoni, & Lipton, 2017). Seminal studies employing region-specific brain lesions and genetic manipulations in murine models as well as pharmacological studies have identified roles of noradrenergic, dopaminergic, cholinergic, histaminergic, serotonergic and orexinergic systems in sleep and arousal (for review, see España & Scammell, 2011; Weber & Dan, 2016). More recent developments suggest that the glutamatergic system is also extensively involved in sleep-wake processes ( Jones, 2020; Martin et al., 2019; Saper & Fuller, 2017; Tononi & Cirelli, 2014). Although a critical regulator of several neurobiological processes including memory consolidation, the role of glutamate in sleep-specific processes is not as rigorously understood. With lines of evidence supporting a role of glutamatergic dysregulation in both CNS disorders and sleep impairments, examining the relationship between glutamate, sleep and CNS disorders is timely.

This chapter highlights research examining metabotropic glutamate (mGlu) receptor involvement in the regulation of sleep alone and in the context of CNS disorders. We focus on schizophrenia, major depressive disorder (MDD), post-traumatic stress disorder (PTSD), Alzheimer’s disease (AD), and substance use disorder (SUD). It is important to note that these disorders are selected examples and that sleep disturbances and disrupted mGlu function are associated with many other CNS disorders. In this chapter, we review preclinical electrophysiological, genetic, and pharmacological studies contributing to our understanding of how various mGlu receptors impact sleep architecture and duration. When possible, we also discuss related human genetic, imaging, and post-mortem studies. Ultimately, we work to shed light on the underappreciated contribution of the glutamate system to sleep and, thereby, comorbid sleep disruptions in CNS disorders. As remediation of sleep disruptions should be a complementary focus of drug development efforts, this chapter highlights novel pharmacotherapeutic approaches targeting mGlu receptors that may effectively address both sleep disturbances and primary symptoms of the neuropsychiatric disorder.

2. Overview of metabotropic glutamate receptors (mGlu receptors)

Glutamate, the major excitatory neurotransmitter in the CNS, regulates rapid excitatory neurotransmission via ionotropic glutamate (iGlu) receptors and slower neuromodulatory effects via mGlu receptors. iGlu receptors are ligand-gated ion channels subdivided into four functional classes: α-amino-3-hydroxy-5-methyl-iso-xazolepropionic acid (AMPA) receptors, N-methyl-d-aspartate (NMDA) receptors, kainate receptors, and GluD receptors (Hansen et al., 2021). mGlu receptors are G-protein coupled receptors (GPCRs) and are the most abundant receptor family in the CNS with 8 distinct subtypes (mGlu1-mGlu8). These class C GPCRs are widely distributed, located on both neurons and glial cells influencing glutamatergic, GABAergic, and other neuromodulatory synaptic transmission (Luessen & Conn, 2022; Maksymetz, Moran, & Conn, 2017; Niswender & Conn, 2010). mGlu receptors are subdivided into three groups (Group I: mGlu1 and mGlu5; Group II: mGlu2 and mGlu3; Group III: mGlu4, mGlu6, mGlu7, and mGlu8) based on sequence homology, G-protein coupling, and agonist selectivity (Conn & Pin, 1997; Niswender & Conn, 2010; Wang & Zhuo, 2012). Acknowledging the extensive distribution and neuromodulatory role of mGlu receptors throughout the CNS, it is not surprising that preclinical and clinical studies suggest altered mGlu receptor expression and function contributes to many neuropsychiatric, neurodevelopmental, and neurodegenerative disorders. In the following section, we provide a brief overview of the general functional and behavioral roles of the different mGlu receptor subtypes.

2.1. Group I mGlu receptor distribution and function

Group I mGlu receptors (mGlu1/5) are widely distributed throughout the CNS in brain regions particularly relevant for learning, memory and motivation. mGlu5 receptors are densely present in the cortex, striatum, and hippocampus whereas mGlu1 receptors are found primarily in the cerebellum, thalamus, and hypothalamus (Niswender & Conn, 2010; Olive, 2010; Romano et al., 1995; Shigemoto et al., 1993; Shigemoto, Nakanishi, & Mizuno, 1992). These predominantly postsynaptic receptors are Gq/11-coupled and localized on glutamatergic and GABAergic terminals (Luessen & Conn, 2022; Niswender & Conn, 2010). Activation of Gq/11-coupled receptors canonically stimulates phospholipase C (PLC) and phosphoinositide hydrolysis leading to intracellular calcium mobilization. However, mGlu1/5 receptors also activate several additional pathways including downstream effectors in the mitogen-activate protein kinase/extracellular protein kinase (MAPK/ERK) pathway (Luessen & Conn, 2022; Niswender & Conn, 2010; Page et al., 2006). Furthermore, mGlu1/5 receptors are critical regulators of activity-dependent synaptic plasticity, influencing both long-term potentiation (LTP) and long-term depression (LTD), and this is in part attributed to their tight coupling with NMDARs (Anwyl, 1999; Joffe, Centanni, Jaramillo, Winder, & Conn, 2018; Lüscher & Huber, 2010; Lutzu & Castillo, 2021; Lu et al., 1997). As these functions are outside the scope of this chapter, readers are referred to several excellent reviews (Luessen & Conn, 2022; Niswender & Conn, 2010; Olive, 2010; Shigemoto et al., 1993). Given the role of group I mGlu receptors in regulating synaptic plasticity and, thereby, learning and memory (for review, see Olive, 2010), it is not surprising that they have been implicated in neurodegenerative and neuropsychiatric disorders associated with cognitive disruptions including AD (Bruno et al., 2000; Kumar, Dhull, & Mishra, 2015), schizophrenia (Kinney et al., 2003; Maksymetz et al., 2017; Nicoletti et al., 2019), Parkinson’s disease (Morin, Grégoire, Gomez-Mancilla, Gasparini, & di Paolo, 2010), MDD (Lindemann et al., 2015), and SUD (Gould et al., 2016; McGeehan & Olive, 2003; Veeneman et al., 2011). As will be described below, dysregulation of either mGlu1 or mGlu5 receptors also affects sleep which may further contribute to primary symptoms associated with these disorders (Aguilar, Strecker, Basheer, & Mcnally, 2020; Cavas, Scesa, & Navarro, 2013b; Holter et al., 2021).

2.2. Group II mGlu receptor distribution and function

Group II mGlu receptors (mGlu2 and mGlu3) are localized on both presynaptic and postsynaptic membranes and, similar to group I, are abundantly present in regions important for learning, memory, and motivation including the prefrontal cortex (PFC), hippocampus, striatum, and amygdala (Crupi, Impellizzeri, & Cuzzocrea, 2019; Luessen & Conn, 2022; Maksymetz et al., 2017). mGlu3 receptors are also expressed on astrocytes, and this contributes to anti-inflammatory properties that may be neuroprotective (Bruno et al., 1998; Nicoletti et al., 2011). Group II mGlu receptors are Gi/o coupled and inhibit adenylyl cyclase and phosphatidyl inositol 3-kinase which reduces intracellular calcium mobilization. These receptors also inhibit MAPK/ERK pathways (Luessen & Conn, 2022; Nicoletti et al., 2011; Niswender & Conn, 2010). Importantly, the primary function of presynaptic mGlu2/3 receptors is to work as autoreceptors and maintain glutamate homeostasis via inhibition of release at glutamatergic and GABAergic synapses following periods of glutamate efflux (Conn & Jones, 2009; Luessen & Conn, 2022; Maksymetz et al., 2017; Mazzitelli, Palazzo, Maione, & Neugebauer, 2018; Moussawi & Kalivas, 2010; Wright, Arnold, Wheeler, Ornstein, & Schoepp, 2001). In general, group II mGlu receptors contribute to synaptic plasticity by reducing synaptic excitability; receptor activation leads to decreased EPSC amplitude and induction of postsynaptic hippocampal LTD (Altinbilek & Manahan-Vaughan, 2009; Grueter & Winder, 2005; Joffe et al., 2020; Walker et al., 2015; Yokoi et al., 1996). Behaviorally, mGlu2/3 receptors have been shown to influence spatial learning and working memory (de Filippis et al., 2015; Lyon et al., 2011). Findings indicate pharmacological activation of mGlu2/3 receptors can improve attention and working memory in acute pharmacological models of cognitive disruptions (e.g. NMDAR antagonists) in both humans and animals (Greco, Invernizzi, & Carli, 2005; Griebel et al., 2016; Krystal et al., 2005; Moghaddam & Adams, 1998). Activation may also produce anxiolytic- and antidepressant-like effects in rodent models (Dogra & Conn, 2021). As such, group II mGlu receptors may be promising pharmacotherapeutic targets for multiple disorders including schizophrenia (Hackler et al., 2010; Hiyoshi, Hikichi, Karasawa, & Chaki, 2014; Maksymetz et al., 2017; Moghaddam & Adams, 1998; Sokolenko, Hudson, Nithianantharajah, & Jones, 2019), AD (Lee et al., 2004; Richards et al., 2010), MDD (Chaki, 2017), pain (Davidson et al., 2017; Jones, Eberle, Peters, Monn, & Shannon, 2005; Mazzitelli et al., 2018), and SUD (Cleva & Olive, 2012; Hao, Martin-Fardon, & Weiss, 2010; Rodd et al., 2006).

2.3. Group III distribution and function

Group III mGlu receptors (mGlu4, mGlu6, mGlu7, mGlu8) are localized pre- and post-synaptically on glutamatergic and GABAergic terminals with differential expression throughout the CNS (Luessen & Conn, 2022; Niswender & Conn, 2010). mGlu4 and mGlu8 receptors are located presynaptically with regionally-restricted expression in the cerebellum and hippocampus (Luessen & Conn, 2022; Niswender & Conn, 2010a; Zhai et al., 2002). mGlu6 receptors, unlike others, have strict localization to the retina, particularly on ON bipolar cells, and, thus, are not relevant for this chapter (Crupi et al., 2019; Luessen & Conn, 2022; Niswender & Conn, 2010). Lastly, mGlu7 receptors are presynaptic and widely distributed throughout the CNS with expression in regions including the hippocampus, thalamus, hypothalamus and amygdala. mGlu7 is unique in that it has a very low affinity for glutamate and, thus, it serves to only function in times of high synaptic activity to fine-tune and prevent excessive glutamate levels (Niswender & Conn, 2010). In general, group III mGlu receptors contribute to spontaneous glutamate transmission and hippocampal-mediated long-term synaptic plasticity (Altinbilek & Manahan-Vaughan, 2007). For example, mGlu4 knockout mice had enhanced hippocampal LTP, though the PFC was unaffected (Iscru et al., 2013). Furthermore, bath application of the mGlu4 PAM foliglurax to corticostriatal slices reduced glutamatergic transmission, as shown through a reduction in spontaneous EPSC frequency (Calabrese et al., 2022). Pharmacological activation of mGlu7 induced LTD in mossy fiber inputs to stratum lucidum interneurons (SLINs) (Pelkey, Lavezzari, Racca, Roche, & McBain, 2005). In Schaffer collateral-CA1 synapses, activation of mGlu7 potentiated a submaximal level of LTP and inhibition of mGlu7 prevented the induction of LTP in these synapses (Kalinichev et al., 2013; Klar et al., 2015). Lastly, activation of mGlu8 reduced field EPSPs induced by lateral perforant path (LPP) afferents in hippocampal slices (Zhai et al., 2002). In line with their regions of expression and preclinical findings, mGlu4 receptors are being explored for their pharmacotherapeutic potential in Parkinson’s disease and schizophrenia (Battaglia et al., 2006; Calabrese et al., 2022; Charvin, 2018; Luessen & Conn, 2022; Niswender & Conn, 2010). The mGlu7 receptor has been a suggested target for disorders including AD (Crupi et al., 2019), neuropathic and inflammatory pain (Marabese et al., 2007; Palazzo, Fu, Ji, Maione, & Neugebauer, 2008), SUD (Li, Xi, & Markou, 2013), MDD and anxiety disorders (Bradley et al., 2012; O’Connor et al., 2013; Palucha & Pilc, 2007; Pałucha-Poniewiera & Pilc, 2013), and epilepsy (Girard et al., 2019; Sansig et al., 2001). Lastly, activation of mGlu8 may be a promising therapeutic approach for anxiety disorders (Duvoisin et al., 2010; Linden et al., 2002; Niswender & Conn, 2010) and additional research supports mGlu8 as a target for pain (Crupi et al., 2019; Marabese et al., 2007; Palazzo et al., 2008).

3. Sleep

Sleep is defined as an easily reversible state of reduced consciousness. However, from a neurobiological perspective, sleep is not passive. Rather, sleep is actively regulated by inhibition of arousal circuits which contrast the cortical activation and arousal that occurs during wake. The two-process model of homeostatic sleep regulation posits that sleep is driven by an interaction of sleep debt(Process S) with the circadian pacemaker (Process C). Process S increases during extended waking periods and decreases following periods of sleep, and the circadian pacemaker follows a similar oscillatory pattern, regulating sleep to specific times of day (Borbély, Daan, Wirz-Justice, & Deboer, 2016; Borbély & Wirz-Justice, 1982; Daan, Beersma, & Borbély, 1984). Circadian factors influencing sleep regulation are outside the scope of this chapter but are thoroughly detailed in both Logan and McClung (2019) and Rosenwasser and Turek (2015). There are many neuroanatomical and neurochemical systems that contribute to sleep-wake regulation, and we refer the reader to many excellent reviews (Brown, Basheer, McKenna, Strecker, & McCarley, 2012; España & Scammell, 2011; Saper, Fuller, Pedersen, Lu, & Scammell, 2010; Saper, Scammell, & Lu, 2005). The following sections provide a brief overview of human and nonhuman sleep and the neurochemical systems regulating sleep-wake cycles with a specific focus on glutamatergic involvement.

3.1. Sleep stages and characteristics

On average, healthy adults sleep 6–8h and undergo 4–5 sleep cycles per night (Hor & Tafti, 2009). Each cycle is comprised of non-rapid eye movement (NREM) and rapid eye movement (REM) sleep and lasts approximately 90min in duration (le Bon, Lanquart, Hein, & Loas, 2019). In humans, NREM sleep is divided into three stages, termed N1, N2, and N3. The American Academy of Sleep Medicine (AASM) defines N1 as the lightest stage of sleep, representing a transition between wake and sleep when external stimuli can still be processed (Iber, Ancoli-Israel, & Chesson, 2007). N2 sleep is classified as light sleep and N3 is considered deep sleep, or slow wave sleep (SWS). Overall, in one night of sleep, the percentage of time in N1, N2, and N3 is ~5%, 50% and 20% with the remaining 25% being REM sleep (Iber et al., 2007). As a result of increased sleep debt throughout the day, N3 stage sleep (and duration of bouts) is generally more prevalent in the first half of the night whereas duration of REM bouts are generally longer in the second half of the night. However, the actual duration and percentage of time in each stage changes across development and normal aging in healthy populations (Ohayon, Carskadon, Guilleminault, & Vitiello, 2004).

Sleep stages and transitions are identified using electroencephalography (EEG) to measure and determine changes in brain oscillatory activity. The transition from wake to N1 sleep is associated with a shift from high/mixed frequency, lower amplitude EEG patterns to predominately lower frequency, higher amplitude oscillations. This includes a decrease in alpha band [~8–13Hz] activity compared to waking states. N2 sleep is associated with sleep spindles (periodic synchronous burst firing in the sigma [~12–15Hz] range) and K-complexes (brief waveforms characterized by high-amplitude spike and rebound patterns). N3 sleep, or SWS, is associated with high amplitude slow wave activity (SWA, delta band, [~0–4Hz]). Lastly, REM sleep is associated with an increase in theta [~4–8Hz] activity relative to delta activity as well as an increase in beta [18–30Hz] and gamma [>30Hz] frequencies. Historically, REM sleep was termed paradoxical sleep as the EEG waveforms paradoxically resemble EEG activity during waking states (Boissard et al., 2002; España & Scammell, 2011).

Humans and nonhuman primates are monophasic sleepers whereas rodents are polyphasic sleepers, rapidly cycling through sleep-wake states over a 24-h period. Despite these differences, from a neurophysiological perspective, mechanisms regulating sleep-wake cycles are evolutionarily well-conserved. Rodents are nocturnal and spend roughly 80% of their time sleeping during the light (inactive) phase and 20% of their time sleeping during the dark (active) phase when under a typical 12 h:12h light:dark cycle. Due to the more rapid sleep cycle (~8–15min/cycle), sleep is typically classified into REM and NREM sleep. Although EEG activity in rodent sleep stages are comparable to humans, electromyography (EMG; muscle activity) is often used in rodents to accurately distinguish REM sleep, which demonstrates a distinct muscle atonia, from waking periods. Research in animals using lesions and genetic and pharmacological manipulations has provided extensive insight into the mechanisms regulating sleep-wake transitions. These studies have also elucidated roles of different sleep variables in maintaining healthy daily function and how irregularities may contribute to various CNS disorders.

3.2. Glutamate and the sleep-wake cycle

Historically, the ascending reticular activation system (ARAS) was solely implicated in regulating waking and arousal. Brainstem nuclei, that were primarily thought to be monoaminergic (dopamine, norepinephrine, serotonin, histamine) and cholinergic, innervate thalamic projections to the cortex. Additional projections to the hypothalamus and basal forebrain innervate orexin/hypocretin neurons and forebrain cholinergic neurons, respectively, which then further project to the cortex (Saper & Fuller, 2017). Early seminal studies by Moruzzi and Magoun (1949) demonstrated that inactivation of the reticular formation and ARAS produced a passive state resembling sleep. However, more recent research demonstrated that lesions to different components of the ARAS had a minimal, if any, effect on sleep-wake durations in rats, suggesting there must be additional contributors (for detailed review see Saper & Fuller, 2017). Glutamatergic projections from the brainstem parabrachial and pedunculopontine nuclei to the basal forebrain as well as projections from the hypothalamic sup-ramammillary area to the cortex have since been shown to contribute substantially to arousal and wake-promoting effects (see Saper & Fuller, 2017). Thus, multiple neurons, including glutamatergic neurons, in the brainstem, hypothalamus and basal forebrain regulate cortical arousal and waking.

Sleep promotion occurs via GABAergic inhibition of each of these wake-promoting nuclei. GABA neurons within the ventrolateral (VLPO) and median preoptic (MNPO) nuclei of the hypothalamus innervate all wake-regulating monoaminergic and cholinergic brainstem nuclei (Sherin, Elmquist, Torrealba, & Saper, 1998; Suntsova, Szymusiak, Alam, Guzman-Marin, & McGinty, 2002). Additional GABA neurons in the brainstem reticular formation inhibit glutamate neurons in the parabrachial nucleus (Anaclet et al., 2014). These GABA neurons are almost entirely inactive during waking periods, and NREM sleep is initiated when these neurons are activated to inhibit cortical activity. This shift in inhibitory balance from wake to sleep is in part driven by several metabolic and circadian influences including adenosine, prostaglandins, cytokines, and growth hormone-releasing hormone (Obal & Krueger, 2003). For example, extracellular concentrations of adenosine increase following prolonged periods of waking as active neurons hydrolyze ATP, and concentrations dissipate following sleep (Porkka-Heiskanen et al., 1997). Importantly, adenosine acts as a neuromodulator to inhibit arousal circuits and disinhibit VLPO GABA neurons thereby exerting sleep-promoting effects (Obal & Krueger, 2003).

There are multiple neuronal populations that contribute specifically to REM sleep. These include subsets of cholinergic neurons in the laterodorsal and pedunculopontine tegmentum (LTD/PPT) as well as the basal forebrain which promote cortical activation through acetylcholine release (Marrosu et al., 1995; Williams, Comisarow, Day, Fibiger, & Reiner, 1994). Additionally, neurons containing melanin-concentrating hormone and GABA neurons in the hypothalamus are active during REM sleep demonstrating inhibitory effects on wake-promoting nuclei (Verret et al., 2003). Lastly, a subset of glutamate neurons active in the sublaterodorsal (SLD) nucleus that project to the inhibitory neurons in the medulla and spinal cord are thought to contribute to muscle atonia during REM sleep (Boissard et al., 2002).

The distinct patterns of EEG activity that occur throughout sleep-wake cycles are derived from interactions between the subcortical systems described above (brainstem, basal forebrain, and hypothalamus) and the thalamus and cortex. Activity in the thalamocortical (TC) circuit also undergoes dynamic changes across sleep-wake cycles. The TC circuit is comprised of two groups of neurons. TC projection neurons are predominantly glutamatergic and relay sensory and motor information to the cortex. Thalamic relay neurons (TRNs) are predominantly GABAergic and are innervated by and can inhibit TC neurons (España & Scammell, 2011; Huguenard & McCormick, 2007). During wake and REM sleep, monoaminergic and cholinergic neurotransmitter release depolarizes thalamic neurons resulting in desynchronized fast, low-amplitude cortical oscillations and increasedsensitivity to incoming stimuli (Aston-Jones, Smith, Moorman, & Richardson, 2009; Hu, Steriade, & Deschênes, 1989). In contrast, the firing patterns shift during NREM sleep as these neurons become hyperpolarized and undergo synchronized burst firing, reducing responsivity to external stimuli (Brown et al., 2012; España & Scammell, 2011; Livingstone & Hubel, 1981; Llinás & Steriade, 2006; Mccormick & Bal, 1997; Steriade, Iosif, & Apostol, 1968). These reciprocally acting projections in part drive cortical delta waves and sleep spindles during NREM sleep (España & Scammell, 2011; Steriade, Domich, Oakson, & Deschfines, 1987). Additional inhibitory feedback loops between cortical glutamate neurons and thalamic GABA neurons further contribute to cortical oscillatory rhythms and sleep spindle generation (Steriade et al., 1987). Lastly, Yu et al. (2019) recently found that activation of glutamate neurons in the VTA projecting to both the nucleus accumbens (NAc) and the lateral hypothalamus increased time awake, whereas inhibition decreased waking bout duration and increased frequency of wake: NREM sleep transitions. In short, there are many overlapping circuits and neurotransmitter systems involved in the transition and maintenance of sleep-wake cycles and the associated oscillatory functions, and the glutamate system is a strong contributor.

Preclinical and clinical studies support a strong relationship between extracellular glutamate and the different sleep stages. EEG studies paired with simultaneous in vivo microdialysis recordings in the orbitofrontal cortex found extracellular glutamate concentrations to be highest during REM sleep, modest during waking periods and lowest during NREM sleep in rats (Lopez-Rodriguez, Medina-Ceja, Wilson, Jhung, & Morales-Villagran, 2007). Amperometric detection of glutamate in the cortex showed rapid increases in glutamate at the beginning of wake and REM sleep and rapid decreases during NREM sleep (Dash, Douglas, Vyazovskiy, Cirelli, & Tononi, 2009). Additional microdialysis studies revealed similar elevations in glutamate levels during wake in the pontine reticular formation (Watson, Lydic, & Baghdoyan, 2011), though glutamate concentrations detected in the thalamus were contrastingly highest during NREM sleep (Kékesi, Dobolyi, Salfay, Nyitrai, & Juhász, 1997). These changes in glutamate concentrations likely align with the shift from asynchronous cortical excitation during wake to synchronous TC regulation during NREM (España & Scammell, 2011). Additionally, evidence from rodent amperometry studies and human studies using proton magnetic resonance spectroscopy (1H-MRS) indicates that glutamate levels and receptor availability are not only state-dependent but also sensitive to additional factors including general sleep history and disruptions in sleep homeostasis that occur following periods of extended waking (Dash et al., 2009; Weigend et al., 2019).

Lastly, the glutamate system has a major role in regulating state-dependent neurobiological changes in synaptic plasticity. The sleep homeostasis hypothesis suggests that a key role of sleep is to maintain a balance in synaptic strength. Notably, the number of synapses undergoing LTP and LTD shift between sleep and wake (Tononi & Cirelli, 2014). During time awake, as daily learning occurs, synapses progressively strengthen and undergo LTP-like potentiation. This is followed by synaptic weakening and LTD during sleep (Tononi & Cirelli, 2014; Vyazovskiy, Cirelli, Pfister-Genskow, Faraguna, & Tononi, 2008). This synaptic weakening functions predominantly to prevent saturation of synapses, which would result in an inability to form new connections; this synaptic weakening is also an important process in memory consolidation (Diering et al., 2017; Martin et al., 2019; Tononi & Cirelli, 2014). Supporting evidence for the morphological changes that occur in sleep and wake has emerged from studies using Drosophila. During time awake, an increase in synapse number as well as in scaffold and post-synaptic proteins Bruchpilot (BRP) and Discs-large (DLG), both key regulators of glutamate release, were reported. These changes were exacerbated in periods of extended wake as a result of sleep deprivation and reduced during periods of sleep (Bushey, Tononi, & Cirelli, 2011; Gilestro, Tononi, & Cirelli, 2009). Importantly, in rodents, both iGlu (GluA1-containing AMPARs) and mGlu receptors, (primarily group I mGlu receptors, detailed below), have been found to orchestrate these synaptic changes (Diering et al., 2017; Vyazovskiy et al., 2008). Briefly, synaptic expression of cortical and hippocampal GluA1-containing AMPARs is highest during wake and lowest during NREM sleep, which corresponds with electrophysiological findings of synaptic potentiation (Vyazovskiy et al., 2008). Cognitive impairments, a primary symptom of many CNS disorders, may be a direct result of aberrant regulation of these synaptic plasticity changes.

3.3. mGlu receptors and sleep

Many studies spanning from Drosophila to humans highlight important contributions of the different mGlu receptor subtypes in sleep-wake regulation. On the most basic level, genetic knockdown of the only mGlu receptor in Drosophila (DmGluRA) produced drastic changes in sleep-wake behavior, notably increasing time spent asleep during the day and decreasing time spent asleep at night (Ly, Lee, Strus, Prober, & Naidoo, 2020). Additional support for mGlu involvement in sleep regulation is derived from EEG studies in mice with a constitutive knockout of a single mGlu receptor subtype or altered function of a downstream signaling protein (see Table 1). Recent development of pharmacological tool compounds with mGlu receptor subtype specificity has also added to this literature base (see Table 2). The following section describes literature examining genetic and pharmacological manipulations in rodents, a majority of which involves the group I and II mGlu receptor classes. Although less well characterized, some evidence supports the role of group III mGlu receptors in sleep-wake regulation. Lastly, when available, relevant human postmortem, GWAS, and neuroimaging studies are also included.

Table 1.

Effects of subtype-specific mGlu receptor genetic alterations on sleep in rodents.

Awake Sleep NREM REM
Condition Phase Duration Total duration Duration Bouts Bout duration Delta Duration Bouts Bout duration Theta
*PLCβ4−/− 1,2 Light n.s., ↑ n.s. n.s.
Dark n.s., ↑ n.s.
TC-restricted PLCβ4−/− 1 Light n.s. n.s.
Dark n.s. n.s. n.s. n.s.
*mGlu1 Mutation3 24 hr n.s.
mGlu2/34 Light
Dark n.s.
mGlu55,6,7 Light n.s., ↓ n.s. n.s.,↑ n.s. n.s.,↓ n.s. n.s., ↓
Dark n.s., ↑ n.s. n.s.,↑ n.s., ↓ n.s. n.s. n.s., ↓
24 hr n.s.
mGlu78 Light n.s. n.s. n.s. n.s. n.s. n.s.
Dark n.s. n.s. n.s.
*

PLCβ4−/− is a downstream effector in the mGlu1 pathway; mGlu1 mutation is loss of function.

n.s., not significant; —, not determined.

Table 2.

Pharmacological studies examining subtype-specific mGlu receptor compounds on sleep in rodents.

Awake NREM REM
Compound Duration Duration Latency Bouts Bout duration Duration Latency Bouts Bout duration
mGlu5 receptor NAMS (within 4–6h post-dosing)
MPEP1,2 (1–20mg/kg, IP) n.s.
MTEP1 (1–10 mg/kg, IP) n.s. n.s. n.s.
GRN-5293 (0.32–1mg/kg, SC)
Mavoglurant3 (1–3.2mg/kg, SC)
Basimglurant4 (0.03–0.3mg/kg, PO)
VU04242385 (1–30mg/kg, IP) n.s.
M-5MPEP (partial NAM)5 (18–56.6 mg/kg, IP) n.s. n.s. n.s.
mGlu5 Receptor PAMS (within 4–6h post-dosing)
LSN28146171,6 (2.5–10mg/kg, PO)
LSN24633596 (0.3–3mg/kg, PO)
ADX472731,6 (10–300 mg/kg, PO) n.s. n.s.
CDPPB6,7 (30mg/kg, IP) ↑ ; n.s. n.s.
mGlu2/3 Receptor Antagonists/NAMS (within 4–6h post-dosing)
LY341495 (antagonist)8,9 (1–10mg/kg, SC)
LY3020371 (antagonist)10 (3–10mg/kg, IP)
Ro4491533 (NAM)8 (2.5–40 mg/kg, PO)
mGlu2/3 Receptor Agonists/PAMS (within 4–6h post-dosing)
LY379268 (agonist)10,11 (10 mg/kg, PO10; 0.25–1 mg/kg SC11) n.s. (dark) ↑(light) n.s. (dark) ↓(light)
LY354740 (agonist)12 (1–10mg/kg, SC) n.s. n.s. n.s. n.s. n.s.
BINA (PAM)12 (1–40mg/kg, SC) n.s. n.s.
JNJ-42153605 (PAM)13 (3 mg/kg, PO) n.s. n.s. n.s.
JNJ-40411813 (PAM)14 (3–30mg/kg, PO)
JNJ-40068782 (PAM)15 (3–30mg/kg, PO) n.s. n.s.
THIIC (PAM; 12h post-dosing)16 (10–30mg/kg, PO)
mGlu2/3 receptor agonists/PAMS (within 4–6h post-dosing)
(S)-3,4-dicarboxyphenylglycine17 (5–20mg/kg, IP) n.s. n.s. n.s. n.s. n.s. n.s. n.s. n.s. n.s.

3.3.1. Group I mGlu receptors

Perhaps one of the most relevant contributions of mGlu1/5 receptors is to maintain homeostasis in synaptic size and strength throughout the sleep-wake cycle via dynamic changes in the mGlu1/5/Homer protein complex. Homer family proteins (Homer1, Homer2, and Homer3) are adaptor proteins that each contain several isoforms as a product of alternative splicing (Shiraishi-Yamaguchi & Furuichi, 2007). Homer protein variants are differentially recruited to the synapse during sleep and wake and, thereby, influence downstream effector systems of mGlu1/5. During wake, mGlu1/5 receptors are anchored to the excitatory post synaptic density via long-form Homer proteins, which contain both an Ena/VASP (EVP) domain and a coiled-coil domain. These long-form Homer proteins link mGlu1/5 receptors to their primary signaling effector, the inositol triphosphate (IP3) receptor, in the endoplasmic reticulum as well as Shank1-3 proteins, facilitating cell excitability and increased GluA1-containing AMPAR levels during arousal states (Diering et al., 2017; Martin et al., 2019). Following prolonged waking periods, the activity-dependent immediate early gene Homer1a, a short-form protein containing only an EVP domain, is targeted to the synapse (Martin et al., 2019; Shiraishi-Yamaguchi & Furuichi, 2007). Homer1a uncouples the mGlu1/5 receptor from the IP3 receptor, resulting in signaling through alternate pathways and reduced synaptic GluA1 (Kammermeier & Worley, 2007; Ronesi & Huber, 2008). While the specific downstream pathways of the mGlu1/5/Homer1a protein complex are not yet extensively studied, there is support for increased MAPK/ERK signaling (Diering et al., 2017; Martin et al., 2019). Importantly, regulation of synaptic levels of Homer1a has been found to be mediated in part by neuromodulators including noradrenaline and adenosine which suppress and promote Homer1a synapse targeting, respectively (Diering et al., 2017). Although Homer1a mRNA is highest during wake, cortical synaptic levels are highest during sleep and states of increased sleep drive suggesting Homer1a levels are activity-dependent and contribute to sleep-maintained synaptic homeostasis (Diering et al., 2017; Maret et al., 2007). Overall, this process, termed homeostatic scaling-down, yields restorative benefits, increasing memory consolidation and opportunities for future learning (de Vivo et al., 2017; Diering et al., 2017; Martin et al., 2019; Tononi & Cirelli, 2014).

3.3.1.1. mGlu1 receptors and sleep

In general, mGlu1 has been implicated in modulating sleep duration. Recently, two loss-of-function mutations of GRM1, the gene encoding mGlu1, were identified in two families diagnosed with familial natural short sleep (FNSS) (Shi et al., 2021). Individuals with FNSS can sleep less than 6h a day (compared to the average 8h of sleep) with no known adverse consequences (Aeschbach et al., 2003). When these mutations were introduced into mice, significant decreases in total and NREM sleep time were found with no effects on REM sleep time (Shi et al., 2021). No other studies to date have directly examined region-or circuit specific GRM1 genetic knockouts to further elucidate their involvement in sleep regulation. However, immunohistochemistry studies have shown that mGlu1 expression was higher in rodents following a 12-h sleep deprivation period (Tadavarty, Rajput, Wong, Kumar, & Sastry, 2011), suggesting that mGlu1 receptors may also serve a role in homeostatic regulation of sleep.

Additional studies examining downstream signaling pathways of mGlu1 have elucidated the important role of this receptor in sleep via expression on TC neurons (Shigemoto et al., 1992). Activation of mGlu1 leads to downstream PLC signaling, specifically the PLCβ4 isoform in TC neurons (Watanabe et al., 1998). Thus, to investigate the function of mGlu1 in sleep, studies have examined sleep in PLCβ4-deficient (PLCβ4−/−) mice. Both global PLCβ4−/− and TC-restricted PLCβ4−/− mice displayed longer total NREM sleep time as well as longer NREM and REM episodes. Delta power was also increased during NREM sleep (Hong et al., 2016). Alternately, a separate study reported no differences in NREM sleep in PLCβ4−/− mice compared to WT mice but did find increased total REM duration, abnormal wake-to-REM transitions, and altered ultradian body temperature rhythms (Ikeda et al., 2009). Altogether, though the GRM1 mutation studies and studies using PLCβ4−/−-deficient mice suggest different roles of mGlu1 in sleep, it is clear that expression of mGlu1 and the associated signaling pathways in TC circuits is likely involved in the transitions and maintenance of wake and sleep. Pharmacological studies examining receptor subtype-selective mGlu1 compounds on sleep-wake architecture are also lacking, in large part due to the unavailability of selective ligands until recently (Luessen & Conn, 2022).

3.3.1.2. mGlu5 receptors and sleep

Similar to the mGlu1 receptor, human and animal studies suggest the mGlu5 receptor serves a role in homeostatic regulation of sleep, with several lines of evidence supporting a role in sleep need/drive. Positron Emission Tomography (PET) imaging studies in healthy humans reported global increases in mGlu5 receptor availability following a sleep deprivation period (Hefti et al., 2013; Holst et al., 2017; Weigend et al., 2019). mGlu5 receptor availability was positively correlated with both subjective and objective measures of sleepiness as well as delta power during NREM sleep (Holst et al., 2017). Multiple EEG studies comparing mGlu5 knockout (KO) mice with wild type (WT) mice corroborate these findings. First, mGlu5 receptor KO mice showed less time awake and greater duration of NREM sleep during the active phase, though sleep was reportedly more fragmented (Aguilar et al., 2020). Alterations in spectral power were also found in mGlu5 KO mice including decreased delta power during NREM, sleep, theta power during REM sleep, and alpha power during waking periods. These spectral changes could have implications in sleep quality and sleep homeostasis, sleep-dependent memory consolidation, and TRN function, respectively (Aguilar et al., 2020). Interestingly, some (Ahnaou, Langlois, et al., 2015; Ahnaou, Raeymaekers, et al., 2015; Holst et al., 2017) but not all (Aguilar et al., 2020) studies reported a lack of build-up in delta power following increased time awake across the dark (active) phase and following sleep deprivation, suggesting altered homeostatic sleep drive in KO animals. Additionally, one study reported extended waking periods following sleep deprivation in KO animals in contrast to the expected rebound sleep found in WT animals (Holst et al., 2017). However, a separate study reported a more rapid sleep rebound in mGlu5 KO mice (Aguilar et al., 2020). Lastly, mGlu5 KO mice showed higher basal gamma power during wake in both the light and dark phase (Ahnaou, Langlois, et al., 2015; Ahnaou, Raeymaekers, et al., 2015) as well as during NREM and REM sleep (Aguilar et al., 2020) suggesting heightened arousal and inability to shift from asynchronous to synchronous oscillatory activity.

Pharmacological studies using subtype-selective compounds have further aided in our understanding of the contributions of the mGlu5 receptor to sleep regulation and underlying brain oscillatory activity (see Table 2 summarizing effects of mGlu selective compounds on sleep). Most highly selective compounds targeting mGlu5 receptors include negative and positive allosteric modulators (NAMs and PAMs, respectively). NAMs and PAMs bind to an allosteric site and either reduce or enhance receptor function, respectively when a ligand binds to the orthosteric binding site, (Niswender & Conn, 2010). In rodent EEG studies, traditional mGlu5 NAMs administered in the light cycle including MPEP and MTEP dose-dependently increased NREM sleep time and bout duration as well as REM latency and decreased REM duration within the 4–6h post-administration (Ahnaou, Langlois, et al., 2015; Ahnaou, Raeymaekers, et al., 2015; Cavas et al., 2013b; Harvey et al., 2013). Increased NREM duration, but not decreased REM duration, is consistent with results from mGlu5 KO studies. Novel, more selective mGlu5 compounds have also been examined. In contrast to MTEP and MPEP, basimglurant (RO4917523, RG7090; administered 2h into the dark cycle) and VU0424238 (administered 2h into the light cycle), both demonstrated initial but transient wake-promoting effects (Holter et al., 2021; Lindemann et al., 2015). However, as the wake-promoting effects of VU0424238 dissipated, selective reductions in REM sleep persisted for 8–10h following administration (Holter et al., 2021). Furthermore, all mGlu5 NAMs mentioned above modulated quantitative EEG by producing increases in NREM delta power, which is possibly reflective of sleep drive, though this finding is inconsistent with studies in mGlu5 KO mice (Harvey et al., 2013; Holter et al., 2021; Lindemann et al., 2015). However, similar to KO mice, multiple mGlu5 NAMs increased high frequency gamma power during waking periods, which may be reflective of increased arousal (Ahnaou, Langlois, et al., 2015; Ahnaou, Raeymaekers, et al., 2015; Holter et al., 2021). Interestingly, M-5MPEP, a partial mGlu5 NAM (at full receptor occupancy, there is only 50% functional inhibition), selectively decreased REM sleep without affecting quantitative EEG (Holter et al., 2021; Rodriguez et al., 2005). Lastly, in contrast, to mGlu5 NAMs, mGlu5 PAMs administered in the light phase including LSN2814617, CDPPB and ADX47273 demonstrated wake-promoting effects for up to 7h following administration (Ahnaou, Langlois, et al., 2015; Ahnaou, Raeymaekers, et al., 2015; Gilmour et al., 2013; Parmentier-Batteur et al., 2012). In the case of LSN2814617 (3mg/kg), while wake and REM sleep effects normalized at around 6h post-administration, decreases in deep sleep duration did not normalize until the onset of the dark cycle at 10h post-administration (Ahnaou, Langlois, et al., 2015; Ahnaou, Raeymaekers, et al., 2015). It is important to note tolerance developed to the wake-promoting effects of CDPPB, such that sleep-wake durations and latencies were not different from baseline following 7 days of repeated administration (Parmentier-Batteur et al., 2012).

Overall, mGlu5 receptor activity influences multiple stages of the sleep-wake cycle. Human studies support a relationship between mGlu5 receptor availability and sleep homeostasis/sleep need. In rodent models, both mGlu5 KO mice and pharmacological studies confirm altered sleep-wake patterns. Of note, differences in selectivity, potency, and pharmacokinetics of mGlu5 compounds need to be considered as these may contribute to a state-specific effect (e.g. selective REM sleep reduction) or initial wake-promotion that may artificially reduce both NREM and REM sleep; off-target effects of some compounds, especially MTEP and MPEP, may also be of influence (Montana et al., 2009).

3.3.2. Group II mGlu receptors and sleep

Similar to group I mGlu receptors, substantial preclinical literature describes the modulatory role of group II mGlu receptors on sleep. Using video analysis to determine sleep vs waking periods, Grm2/3−/− mice had significant reductions in sleep time during both the light and dark phase as well as fragmented sleep (increased number of sleep bouts and decreased bout duration) (Pritchett et al., 2015). However, two additional studies using EEG reported no marked differences between Grm2/3−/− and Grm2−/− mice in waking, NREM and REM durations compared to WT mice (Ahnaou et al., 2009; Wood et al., 2018).

Pharmacological modulation of mGlu2/3 receptors, however, has produced robust changes in sleep/wake patterns. Activation of mGlu2/3 during the light cycle via agonists including LY379268 and LY354740 selectively increased REM sleep latency and reduced REM sleep duration without impacting wake or NREM sleep for 7–12h following administration (Ahnaou et al., 2009; Feinberg et al., 2002). mGlu2/3 agonists also reduced high frequency oscillatory activity in the 10–50Hz range indicating that, in addition to selectively reducing REM sleep, these compounds also reduce cortical arousal during wake (Ahnaou et al., 2009; Feinberg et al., 2002). Importantly, these effects were absent in Grm2−/− mice suggesting that a reduction in REM sleep is specifically attributed to activation of mGlu2 and not mGlu3 receptors (Ahnaou et al., 2009; Wood et al., 2018). mGlu2/3 agonists have similar effects on sleep when compared to mGlu2/3 PAMs including BINA, JNJ-42153605, THIIC and JNJ-40411813, although the latter two compounds also increased sleep duration and bout length for 8–12h following administration (Ahnaou et al., 2009; Cid et al., 2012; Fell et al., 2011; Lavreysen etal., 2013). Effects of mGlu2/3 PAMs were also absent in Grm2−/− mice (Ahnaou et al., 2009; Wood et al., 2018). In contrast, mGlu2/3 antagonists (LY341495 and LY3020371) administered in both the light and dark phase and NAM (Ro-4491533) increased wake duration (both active and passive) and non-selectively decreased total sleep time. These wake-promoting effects were long-lasting, such that administration in the light cycle resulted in sustained changes through the beginning of the dark cycle (Ahnaou et al., 2014). Likely tied to increased arousal, increased gamma power was reported following administration, and this opposes effects to receptor activation (Ahnaou et al., 2014; Feinberg et al., 2005; Wood et al., 2018). Interestingly, reduced, yet still measurable, effects of the mGlu2/3 antagonist LY3020371 and NAM Ro-4491533 were present in mGlu2−/− rats suggesting that both mGlu2 and mGlu3 contribute to wake promotion (Wood et al., 2018). As noted above, acute pharmacological studies recapitulate some, but not all, findings from knockout mice.

3.3.3. Group III mGlu receptors and sleep

Unlike group I and II mGlu receptors, the influence of group III mGlu receptors on sleep-wake regulation are understudied. To our knowledge, no study has examined the influence of mGlu4 receptors on sleep-wake architecture in KO mice or with selective ligands despite an extensive catalogue of pharmacological compounds and widespread interest in mGlu4 receptors with regard to Parkinson’s Disease (Hopkins, Lindsley, & Niswender, 2009). Furthermore, research examining the influence mGlu7 and mGlu8 receptors on sleep regulation is scarce. A single study reported increased time awake and decreased NREM sleep duration in mice with a GRM7 deletion, the gene that encodes mGlu7, compared to WT mice. Additionally, though these Grm7−/− mice displayed no change in overall REM sleep duration, they demonstrated decreased average REM bout duration and increased bout numbers, suggesting changes in the maintenance of REM sleep (Fisher et al., 2020). In part attributed to the scarcity of selective compounds, pharmacological studies are also limited. The mGlu7 PAM AMN082 produced differential effects on sleep in healthy rats. Acute administration of lower doses (5 and 10mg/kg) during the light phase increased NREM sleep and decreased time awake whereas a high dose (20mg/kg) increased time awake and decreased both NREM and REM sleep (Cavas, Scesa, & Navarro, 2013a). However, similar effects were present when examined in Grm7−/− mice (Ahnaou, Raeyemaekers, Huysmans, & Drinkenburg, 2016) suggesting off-target effects. Thus, there is still a need to examine more selective compounds to better characterize the role for mGlu7 in sleep regulation. Lastly, a single study reports that the mGlu8 agonist (S)-3,4-DCPG did not affect any sleep parameter in healthy rats (Cavas et al., 2017).

Overall, the individual mGlu receptors have differential roles in sleep/wake regulation. In general, both pharmacological and knockout models examining group I receptors identified contributions to both NREM and REM sleep time. Further, in conjunction with human findings, several rodent studies identified changes in spectral activity that reflected contributions to homeostatic sleep drive. In contrast, group II receptors appear to have less profound contributions in EEG knockout studies, and, in pharmacological studies, many compounds appear to produce REM-selective inhibition (agonists/PAMs) or robust wake-promotion (antagonists/NAMs). Lastly, while some roles of group III receptors have been reported, the literature is still in early stages with genetic and pharmacological advancements necessary to further our understanding of their role (or lack thereof ) in regulating sleep.

3.4. mGlu receptors, neuropsychiatric disorders and sleep

Comorbid sleep disturbances including insomnia have been found in the majority of CNS disorders (Benca & Buysse, 2018; Winkelman & de Lecea, 2020; Wulff et al., 2010). Although primarily diagnosed subjectively and defined as difficulty initiating or maintaining sleep, insomnia is also associated with aberrant quantitative EEG characteristics including increased high frequency (beta/gamma) activity at sleep onset and during NREM sleep (for review, see Zhao et al., 2021). Importantly, the DSM-5 delineated insomnia as a separate diagnosis from other mental disorders as opposed to a primary or secondary symptom (APA, 2013). Thus, as a separate diagnosis, insomnia should also be a primary target of pharmacotherapies (Benca & Buysse, 2018). Furthermore, the DSM-5 emphasizes the bidirectional relationship between insomnia and neuropsychiatric illness, where both disorders can serve as a risk factor for the development of one another (Benca & Buysse, 2018; Winkelman & de Lecea, 2020). Support for insomnia as a risk factor stemmed from data collected from the National Institute of Mental Health’s Epidemiologic Catchment Area Survey. This survey collected comprehensive information on mental disorders in the United States and included questions about sleep disturbances in addition to current psychiatric symptoms. Findings from this survey suggested that insomnia increased the risk of first episode diagnosis of MDD (Ford & Kamerow, 1989). As discussed below, preexisting insomnia has also been implicated in the onset of SUD, PTSD, schizophrenia, and AD (Irwin & Vitiello, 2019; Maher, Rego, & Asnis, 2006; Manoach & Stickgold, 2009; Neylan et al., 2021; Riemann, Berger, & Voderholzer, 2001; Roehrs, Sibai, & Roth, 2021; Zhang et al., 2022).

It is important to understand how the glutamate system may contribute to the comorbid and bidirectional relationship between sleep and CNS disorders. As there are ongoing efforts to develop mGlu receptor subtype-selective treatments for these conditions, current and future research should work to determine if mGlu receptor compounds can target both sleep disturbances and the comorbid neuropsychiatric disorders. In the following sections, we present preclinical and clinical literature detailing known alterations in glutamatergic function associated with schizophrenia, MDD, PTSD, AD, and SUD as well as the comorbid sleep disruptions frequently linked with these conditions. As the field is still expanding, we build on the predominately preclinical pharmacological data above and speculate on possible mGlu receptor manipulations/strategies to improve sleep disturbances in addition to other symptoms of the CNS disorder.

3.4.1. Schizophrenia

Schizophrenia is a neuropsychiatric disorder that affects around 1% of the human population and is characterized by three primary symptoms clusters: positive, negative, and cognitive ( Jones, Watson, & Fone, 2011; Manoach & Stickgold, 2019; Winship et al., 2019). Sleep disruptions represent an additional underappreciated symptom cluster (Manoach & Stickgold, 2019). Sleep disturbances are reported in both medicated and drug-naïve patients, affecting ~80% of all patients, and are linked with symptom prevalence and severity (Chen et al., 2020; Cohrs, 2008; Korenic et al., 2020; Manoach & Stickgold, 2019; Sprecher et al., 2015). Patients commonly experience insomnia and related increases in NREM sleep latency as well as reduced sleep quality (e.g. reduced delta power in N3 sleep) and sleep efficiency (defined as the ratio of time spent asleep when in bed) (Benca & Buysse, 2018; Manoach & Stickgold, 2019; Sprecher et al., 2015). Decreased REM sleep latency has also been found during acute psychosis, though reported changes in REM sleep duration in patients with schizophrenia are inconsistent throughout literature (Pritchett et al., 2012; Sprecher et al., 2015). Importantly, sleep disturbances can precede the onset of schizophrenia and associated symptoms. Insomnia has been found to precede the onset of positive symptoms including hallucinations and paranoia (for review, see Reeve et al., 2015), and, similarly, decreased NREM sleep duration and quality has been associated with the onset of negative and cognitive symptoms (Göder et al., 2004; Göder et al., 2006; Keshavan et al., 1995). Sleep spindles, which are important for memory consolidation and cognition, also occur less frequently in medicated patients with schizophrenia relative to healthy humans (Göder et al., 2006, 2015; Manoach & Stickgold, 2019; Wamsley et al., 2012). Together, altered sleep duration and architecture are directly associated with all primary symptom clusters of schizophrenia.

Current antipsychotic medications, which exert therapeutic effects via blockade of dopamine D2 and serotonin 5-HT2A receptors, affect sleep and can contribute to results of sleep studies conducted in medicated patients. Both first- and second-generation antipsychotics increase total sleep time in healthy humans and in patients with schizophrenia in part by decreasing monoaminergic mediated arousal circuits, and these sedative effects may persist during wake (Göder et al., 2004; Manoach et al., 2010; Manoach & Stickgold, 2009). Although literature is mixed on specific changes, second generation antipsychotics have commonly been found to increase both NREM and REM sleep time in patients with schizophrenia (for review and more detailed descriptions, see Cohrs, 2008). Importantly, antipsychotic medications are used as off-label agents to treat insomnia (Benca & Buysse, 2018). Thus, although many of the current FDA-approved medications can increase sleep in patients with schizophrenia, novel, more effective pharmacotherapies that target cognitive impairments and have a lower adverse effect risk than current antipsychotics are needed.

Although antipsychotics were initially developed on the premise that excessive dopamine in the striatum contributed to the positive symptoms, growing evidence supports glutamatergic dysregulation, specifically N-methyl-d-aspartate receptor (NMDAR) hypofunction, as a key contributor to multiple symptoms of schizophrenia (Coyle, 2006; Javitt, 2007; Jones et al., 2011; Kantrowitz & Javitt, 2012). This hypothesis stemmed from early studies that reported psychotic-like behavior in healthy humans following administration of NMDAR antagonists (Davies & Beech, 1960; Luby, Cohen, Rosenbaum, Gottlieb, & Kelley, 1959). Since then, multiple studies have reported similarities in brain function, psychotic-like behaviors, and cognitive impairments between healthy humans administered NMDAR-antagonists and patients with schizophrenia, and NMDAR antagonists have been integrated into many preclinical studies to model all symptom clusters (for review, see Javitt, 2007; Jones et al., 2011). Genetic, functional imaging, and post-mortem brain tissue studies have provided substantial evidence of neurobiological alterations in the glutamate system in patients with schizophrenia (Hu, MacDonald, Elswick, & Sweet, 2015; Korenic et al., 2020). For example, altered pyramidal cell morphology was found in the cortex of post-mortem brain tissue, including decreased dendritic spine density, dendritic length, and somal size (Black et al., 2004; Glantz & Lewis, 2000; Kalus, Müller, Zuschratter, & Senitz, 2000; Pierri, Volk, Auh, Sampson, & Lewis, 2001, 2003). Changes in enzymes that have a role in glutamate synthesis and metabolism have also been reported. Protein expression of glutamine synthetase was lower and activity of phosphate-activated glutaminase (PAG) was higher in the PFC. These enzymes are responsible for converting glutamate to glutamine and vice versa suggesting impairment in glutamate metabolism in patients with schizophrenia compared to healthy controls (Burbaeva et al., 2003; Gluck, Thomas, Davis, & Haroutunian, 2002). Importantly, studies have found that alterations in glutamate neurotransmission correspond with associated sleep impairments. Using [1H]-MRS, Korenic et al., 2020 found poor sleep quality in patients with schizophrenia was associated with higher Glx (glutamate and glutamine) levels in the hippocampus and lower levels of glutamate in the anterior cingulate cortex. The glutamate levels in the anterior cingulate cortex correlated with lower total scores on the Brief Psychiatric Rating Scale (BPRS). Moreover, poor sleep quality corresponded with heightened severity of positive symptoms. Taken together, these clinical studies firmly support a relationship between glutamate dysregulation, schizophrenia, and sleep.

Alterations in mGlu receptors have also been identified. Post-mortem tissue from patients with schizophrenia revealed higher protein expression of mGlu5 and several mGlu5 trafficking proteins in the hippocampus (Matosin et al., 2015). Conversely, a PET imaging study reported lower cortical mGlu5 binding potential in patients with schizophrenia compared to healthy controls and lower binding was associated with an increase in negative symptoms and worse cognitive performance (Régio Brambilla et al., 2020). Lastly, polymorphisms of GRM5, the gene encoding the mGlu5 receptor, have also been reported in patients with schizophrenia (Matosin et al., 2018). This relationship between mGlu5 and symptoms of schizophrenia is further supported by pharmacological studies. Some mGlu5 NAMs, including fenobam, induced psychotomimetic-like effects in both humans and animals. These compounds were also associated with increased resting-state high frequency gamma power, which is commonly found in patients with schizophrenia (Ahnaou, Langlois, et al., 2015; Ahnaou, Raeymaekers, et al., 2015; Holter et al., 2021; Homayoun, Stefani, Adams, Tamagan, & Moghaddam, 2004; Jacob et al., 2009). The mGlu5 receptor is coupled to the NMDAR (Niswender & Conn, 2010; Shigemoto et al., 1993), and, thus, one hypothesized mechanism through which mGlu5 NAMs produce these psychotomimetic-like effects is via downstream NMDAR inhibition (Sengmany & Gregory, 2016). Furthermore, as described above, global knockout of mGlu5 in mice recapitulated some sleep deficits similar to those in patients with schizophrenia (Aguilar et al., 2020). Taken together, these preclinical and clinical studies support an underlying role of mGlu5 hypofunction in schizophrenia symptomology.

While overwhelming evidence suggests NMDAR dysregulation may underlie all symptom clusters of schizophrenia, directly targeting NMDARs as a therapeutic approach has been met with adverse effects including seizures. Thus, modulating mGlu receptors represents a more tolerable approach than direct NMDAR activation, and all groups of mGlu receptors have gained preclinical support as potential therapeutic targets (Dogra & Conn, 2022; Luessen & Conn, 2022). Activation/allosteric modulation of group I mGlu receptors has shown promise through increasing presynaptic glutamate release and postsynaptic NMDAR function. Support for the use of both mGlu1 and mGlu5 PAMs spans multiple preclinical models of all primary symptom clusters. For example, compounds targeting either receptor have reversed amphetamine and NMDAR antagonist-induced hyperlocomotion, deficits in paired pulse inhibition, and deficits in multiple cognitive assessments (for review, seeDogra & Conn, 2022; Maksymetz et al., 2017). Although mGlu5 PAMs increase time awake and reduce sleep in healthy rodents, the potential for these compounds to modulate sleep spindles still warrants examination of the sleep-altering effects in animal models of schizophrenia. As noted above, patients with schizophrenia and mGlu5 KO mice showed lower sleep spindle frequencies (Aguilar et al., 2020; Göder et al., 2006, 2015; Manoach & Stickgold, 2019; Wamsley et al., 2012). Sleep spindles are generated in the TRN (Bazhenov, Timofeev, Steriade, & Sejnowski, 2000; Ferrarelli & Tononi, 2017) and are in part mediated by group I mGlu receptor activation (Sun et al., 2016). Thus, both mGlu1 and mGlu5 PAMs may enhance sleep spindle activity and, thereby, memory consolidation. Patients with schizophrenia also report excessive daytime sleepiness which may be a product of insomnia, reduced sleep quality, and/or the sedative effects of current antipsychotics (Cohrs, 2008; Miller, 2004; Sharma, 2016). Increasing arousal/time awake during the day could aid with sleep consolidation at night, similar to the primary goal of the cognitive behavioral therapy approach for insomnia (Haynes, Talbert, Fox, & Close, 2018). These can also be employed as an adjunct treatment to counteract the sedative effects of many antipsychotics. mGlu5 PAMs have not yet been examined on sleep-wake activity in preclinical models of schizophrenia, though these studies are critical to determine potential of these compounds to normalize sleep disturbances. It is also important to consider time of dosing in future studies to mitigate undesired wake promoting/sleep-disrupting effects.

Group II mGlu receptors are also promising pharmacotherapeutic targets for schizophrenia. NMDAR hypofunction induced by ketamine and PCP ultimately leads to increased levels of synaptic glutamate in regions including the PFC (Lorrain, Baccei, Bristow, Anderson, & Varney, 2003; Maksymetz et al., 2017; Moghaddam & Jackson, 2003). Activation of mGlu2/3 presynaptic autoreceptors causes a reduction in synaptic glutamate concentrations (Dogra & Conn, 2022; Maksymetz et al., 2017) and, thus, may be a viable mechanism to attenuate this aberrant glutamate efflux. Similar to group I PAMs, mGlu2/3 agonists and PAMs have shown efficacy in preclinical models of all primary symptom clusters of schizophrenia (Benneyworth et al., 2007; Galicietal., 2006; Greco etal., 2005; Griebel et al., 2016; Hackler et al., 2010; Kawaura, Karasawa, & Hikichi, 2016; Maksymetz et al., 2017; Moghaddam & Adams, 1998). Following extensive support from preclinical studies, the mGlu2/3 agonist LY2140023 advanced into clinical trials for treatment of schizophrenia but had mixed results in phase II studies. However, significant improvements in positive and negative symptoms were reported in some trials supporting activation of mGlu2/3 as a mechanism for further pursuit (for review, see Maksymetz et al., 2017). Unlike mGlu5 PAMs, mGlu2/3 PAMs may influence sleep directly by reducing extracellular glutamate levels. Newer, more selective compounds increased NREM sleep time and bout length (Fell et al., 2011; Lavreysen et al., 2013) and, thus, may be of benefit to patients experiencing insomnia. Furthermore, although not directly investigated, mGlu2 receptors are also located in the TRN and regulate signaling (Crabtree, Lodge, Bashir, & Isaac, 2013) suggesting a possible, but not yet studied, influence on sleep spindle activity.

In summary, both mGlu5 and mGlu2/3 PAMs represent promising therapeutic approaches for multiple symptoms of schizophrenia. While speculative, these compounds may appropriately address associated sleep disturbances. It is important to note that preclinical studies also support targeting group III mGlu receptors, specifically with mGlu4 PAMs and mGlu7 NAMs, for all primary symptom clusters of schizophrenia (for review, see Dogra & Conn, 2022 and Luessen & Conn, 2022). However, as reiterated throughout this chapter, further research is needed to understand the potential of group III compounds to target schizophrenia-associated sleep disturbances.

3.4.2. Major depressive disorder

Major depressive disorder (MDD) is characterized by the DSM-5 as having at least five symptoms including depressed mood, loss of interest in activities, fatigue, and sleep disturbances for a 2-week period (APA, 2013). Sleep disturbances are reported in around 90% of patients with MDD, and, while the majority of these patients report insomnia, there is a smaller subset of patients (15–35%) who report hypersomnia (Armitage, 2007; Reynolds & Kupfer, 1987; Steiger & Pawlowski, 2019). Further, there appears to be a strong bidirectional relationship between depression and insomnia. First, in an epidemiological study examining an adult sample from the UK population, insomnia was linked to preexisting depression (Morphy, Dunn, Lewis, Boardman, & Croft, 2007). Multiple epidemiological studies have also found preexisting insomnia was associated with the development of depression (Fang, Tu, Sheng, & Shao, 2019; Ford & Kamerow, 1989; Morphy et al., 2007; Riemann et al., 2020). However, it is important to note that while the majority of patients with depression have comorbid insomnia, only around 20% of patients with insomnia are diagnosed with depression (Breslau, Roth, Rosenthal, & Andreski, 1996). Polysomnography studies reveal that, in general, patients with depression experience a reduction in overall sleep time and display increased NREM sleep latency alongside decreased NREM sleep duration and increased wake after sleep onset (WASO) (Benca et al., 1992; Steiger & Kimura, 2010; Wichniak, Wierzbicka, & Jernajczyk, 2013; Wichniak, Wierzbicka, Walęcka, & Jernajczyk, 2017). This is paired with reduced delta power during NREM sleep (Armitage, 1995; Kupfer, Reynolds, Ulrich, & Grochocinski, 1986; Steiger & Kimura, 2010). Interestingly, in contrast to NREM sleep, patients with MDD concurrently experience an increase in REM sleep duration and density and a reduction in REM sleep latency (Foster, Kupfer, Coble, & McPartland, 1976; Steiger & Pawlowski, 2019; Wichniak et al., 2013). Although variable, most antidepressant medications selectively decrease REM sleep and increase REM sleep latency without producing concomitant decreases in NREM sleep (Steiger & Pawlowski, 2019; Wichniak et al., 2017). Antidepressants have also been found to increase NREM sleep delta power (Kupfer, Ehlers, Pollock, Swami Nathan, & Perel, 1989; Steiger & Kimura, 2010).Importantly, the ability of medications to mitigate REM sleep abnormalities has been shown to correspond with treatment efficacy, reiterating the importance of improving sleep for overall treatment outcome (Riemann et al., 2001).

There is substantial evidence for glutamatergic contributions to the pathology of MDD and some even suggest there should be a glutamate hypothesis of depression (Sanacora, Treccani, & Popoli, 2012). Several studies in clinical populations have shown differences in glutamate levels in the CSF, plasma and platelets, and post-mortem brain tissue between individuals with MDD and healthy individuals. For example, increased glutamate levels in the plasma have been found in patients with MDD relative to a healthy control group (Altamura et al., 1993; Kim, Schmid-Burgk, Claus, & Kornhuber, 1982; Mauri et al., 1998). In contrast, decreased glutamate levels were reported in the CSF in patients with refractory affective disorder (includes both unipolar and bipolar patients) (Frye, Tsai, Huggins, Coyle, & Post, 2007), although earlier studies examining glutamine levels in the CSF found increases in patients with unipolar and bipolar depression (Levine et al., 2000). In studies examining post-mortem tissue, increased glutamate levels were found in the frontal cortex (Hashimoto, Sawa, & Iyo, 2007) and dorsolateral prefrontal cortex (Lan et al., 2009). Lastly, several studies have examined glutamate levels in patients with MDD using 1H-MRS. Collectively, a meta-analysis reported consistent decreases in concentrations of glutamate both globally and specifically in the anterior cingulate cortex (Luykx et al., 2012). In short, these clinical findings have pointed to clear dysregulations in glutamate levels in patients with MDD, and the sampling methodology, brain-region, and/or heterogeneity associated with MDD may have influenced varying findings across studies.

Targeting the glutamate system through both iGlu and mGlu receptors has been explored for depression. Early findings suggest that ketamine, an NMDAR antagonist, produced rapid-acting antidepressant effects, and NMDAR antagonism remains a promising therapeutic approach (Berman et al., 2000; Raab-Graham, Workman, Namjoshi, & Niere, 2016; Trullas & Skolnick, 1990; Zarate et al., 2006). Of relevance, ketamine administration to individuals with MDD decreased mGlu5 receptor availability in several brain regions and hippocampal mGlu5 availability correlated with Montgomery-Å sberg Depression Rating Scale (MADRS) total score (Esterlis et al., 2018). Thus, long-standing research also supports targeting mGlu receptors as a treatment approach for MDD. Functional antagonism of mGlu5 displayed therapeutic potential in preclinical models that have predictive validity for antidepressant medications including the forced swim test (FST) and tail suspension test (TST) (Felts et al., 2017; Gould et al., 2016; Hughes et al., 2013; Kato et al., 2015; Lindemann et al., 2015). Importantly, extensive preclinical work suggests mGlu5 NAMs may be beneficial for treating sleep disturbances associated with MDD. For example, the mGlu5 NAM basimglurant significantly increased REM latency and decreased REM sleep and the REM/NREM ratio in healthy rats when administered during the active phase. Furthermore, basimglurant increased delta power during NREM sleep. This profile is similar to other mGlu5 NAMs (e.g. Holter et al., 2021) and consistent with several clinically prescribed antidepressants (Steiger & Pawlowski, 2019; Wichniak et al., 2013, 2017). Furthermore, some mGlu5 NAMs have progressed into clinical trials for MDD, though historically with negative outcomes due to the adverse effects noted above (e.g. fenobam, Pecknold, McClure, Appeltauer, Wrzesinski, & Allan, 1982). However, basimglurant advanced to phase II clinical trials as an adjunctive therapy for depression. Although the primary endpoint criteria, change in MADRS total score, was not met with this compound, significant results were found in secondary endpoint criteria including other self-report measures of depressive symptoms (Quiroz et al., 2016). It is critical to note that sleep was not evaluated in this clinical trial.

Clinical evidence, although mixed, also supports a role of group II mGlu receptors in MDD. One study reported increased mGlu2/3 protein levels in the PFC of postmortem tissue (Feyissa et al., 2010) while a PET imaging study reported significantly lower binding in the anterior cingulate cortex in patients with MDD suggesting the relationship between mGlu2/3 and MDD is complex (Mcomish et al., 2016). Both agonists and antagonists of group II mGlu receptors have been pursued in preclinical and clinical studies for their antidepressant-like potential. Antagonism of the mGlu2/3 receptor has been most heavily pursued because these compounds have similar mechanisms to ketamine. Ultimately, similar to subanesthetic doses of ketamine, administration of an mGlu2/3 antagonist should lead to increased glutamate release because presynaptic mGlu2/3 receptors function as autoreceptors (Chaki, Koike, & Fukumoto, 2019). Aside from sleep, preclinical studies showed mGlu2/3 antagonists and NAMs display antidepressant-like effects in preclinical models including the FST and TST (Chaki et al., 2004; Fukumoto, Iijima, & Chaki, 2016; Witkin et al., 2016). Interestingly, both mGlu2 and mGlu3 contribute to this effect because selective compounds VU6001966 (mGlu2 NAM) and VU650786 (mGlu3 NAM) both decreased immobility time in the FST ( Joffe et al., 2020). Additionally, the mGlu2/3 NAM decoglurant advanced to clinical trials as an adjunctive treatment to SSRIs or SNRIs but was unsuccessful in meeting primary or secondary endpoint criteria. However, complexities with the clinical trial including a high placebo response made it difficult to adequately evaluate the compound (Umbricht et al., 2020). Importantly, mGlu2/3 antagonists may be relevant to target sleep disturbances in patients with depression because of their ability to reduce REM sleep. However, applicability of mGlu2/3 antagonists may be population-dependent, perhaps better suited for the sub-population presenting with hypersomnia, as mGlu2/3 antagonists also have wake-promoting effects.

mGlu2/3 agonists and PAMs may have greater potential for targeting sleep disturbances in MDD. Several of these compounds have also displayed antidepressant-like effects in preclinical screens including the FST (Fell et al., 2011). Furthermore, the mGlu2/3 agonist LY379268 demonstrated promising antidepressant-like effects when administered as an adjunctive treatment to fluoxetine and may help bridge the gap between treatment initiation and delayed onset of the therapeutic effects of classic antidepressants (Matrisciano et al., 2007). Importantly, similar to mGlu2/3 antagonists, these compounds also significantly decrease REM sleep time and increase REM latency, but, unlike mGlu2/3 antagonists, most do not promote wake (Ahnaou et al., 2009; Cid et al., 2012). Thus, this approach may be more beneficial to those who are concurrently experiencing insomnia. Because elevated extracellular glutamate concentrations were found in the orbitofrontal cortex during REM sleep in the rat (Lopez-Rodriguez et al., 2007), activation of mGlu2/3 agonists may be a more suitable approach to correct REM sleep abnormalities in MDD by reducing overall synaptic glutamate levels and selectively suppressing REM sleep (Ahnaou et al., 2009).

In summary, reducing glutamate function via mGlu5 NAMs or mGlu2/3 PAMs may be a viable approach to target primary symptoms and sleep disruptions in MDD. As evaluated with basimglurant, the ability of new compounds to normalize sleep disruptions associated with MDD should be integrated into future clinical trial design.

3.4.3. Post-traumatic stress disorder

Post-traumatic stress disorder (PTSD) affects roughly 4% of the population and is associated with fear, anxiety, avoidance, negative mood, intrusive thoughts, hyperarousal, and sleep disturbances, including insomnia, that persist following a traumatic event (Kessler, Wai, Demler, & Walters, 2005; Kilpatrick et al., 2013). While objective measures of sleep within the PTSD population are inconclusive, perhaps due to the heterogeneity of individuals and their associated trauma, many studies report increased light sleep, decreased SWS, increased REM density, and decreased REM bout length (Kobayashi, Boarts, & Delahanty, 2007; Kobayashi, Huntley, Lavela, & Mellman, 2012; Lewis et al., 2020; Woodward, Leskin, & Sheikh, 2002). Subjectively, patients with PTSD report sleep disturbances including nightmares, sleep terrors, limb movements, and dream enactment (Baird et al., 2018). Additionally, the prevalence of sleep disordered breathing (e.g. sleep apnea) and sleep movement disorders, including periodic limb movement disorder and REM movement disorders, are higher among those with PTSD (Baird et al., 2018; Maher et al., 2006; Neylan et al., 2021). Sleep disturbances have also been found to correlate with prevalence and severity of other symptoms. For example, insomnia severity correlated with intrusion and hyperarousal (Lauterbach, Behnke, & McSweeney, 2011). The frequency and severity of nightmares was associated with a higher prevalence of hopelessness and suicidal behaviors (Littlewood, Gooding, Panagioti, & Kyle, 2016). Further, in support of a bidirectional relationship, it is well-established that preexisting sleep disturbances influence the development of PTSD. Insomnia and stress reactivity prior to a motor vehicle collision predicted subsequent PTSD diagnosis (Neylan et al., 2021). Additionally, some studies evaluating self-reported sleep measures including nightmares and insomnia prior to deployment corresponded to subsequent PTSD development in combat veterans (van Liempt, van Zuiden, Westenberg, Super, & Vermetten, 2013; Wang et al., 2019). Given the important role of sleep in regulating stress, emotion, and learning and memory, it is not surprising that dysregulated sleep may exacerbate the development and progression of PTSD.

Glutamatergic function has been shown to be altered in individuals with PTSD which may directly contribute to PTSD symptoms including altered fear reactivity, hyperarousal, and dysregulated sleep. Using 13C-acetate MRS, patients with PTSD showed reduced glutamatergic synaptic strength in the PFC compared to healthy controls (Averill et al., 2022). Similarly, in rodents, the single prolonged stress model reduced glutamatergic excitation of pyramidal neurons in the infralimbic medial PFC (Nawreen, Baccei, & Herman, 2021). Another study using 1H-MRS imaging in humans reported lower GABA and increased glutamate levels in the cortex in patients with PTSD, and these findings correlated with higher subjective ratings of insomnia (Meyerhoff, Mon, Metzler, & Neylan, 2014). In further support of research that suggests sleep disturbances may exacerbate neurobiological factors in PTSD, studies conducted in healthy humans found a single night of sleep deprivation altered functional connectivity between the amygdala and PFC and was associated with aberrant amygdalar reactivity (Killgore, 2013; Yoo, Gujar, Hu, Jolesz, & Walker, 2007). These data support the prevailing theory that inadequate top-down PFC inhibitory control leads to hyperactivity in the amygdala and over-reactivity to stressful stimuli (Rauch, Shin, & Phelps, 2006). Furthermore, as many of the sleep disturbances and other symptoms derived from hyperarousal occur in part because of increased glutamate activation of the hippocampus and amygdala, putative pharmacotherapeutics should attenuate glutamate to restore excitatory:inhibitory balance.

Current treatments for PTSD include cognitive behavioral therapy and exposure therapy, which are aimed to help individuals re-process traumatic events and re-structure responses in an effort to attenuate hyperarousal and responsivity to environmental stimuli (American Psychological Association, 2017). Treatment of the emotion-related symptoms of PTSD have been found to positively impact sleep. For example, subjective and objective measures of sleep improved in patients with PTSD following successful outcomes of trauma-targeted psychotherapies (e.g. remission of emotional symptoms) (Rousseau et al., 2021). While severity of sleep disturbances may be reduced following behavioral treatment approaches, they are not fully resolved (Taylor et al., 2020). There are only two FDA-approved pharmacological treatments for PTSD, the antidepressant medications sertraline and paroxetine (Brady et al., 2000; Krystal et al., 2017; Marshall, Beebe, Oldham, & Zaninelli, 2001). However, both antidepressants, in addition to commonly used anxiolytics, target depressed mood and anxiety symptoms rather than directly targeting hyperarousal or sleep disturbances. Since NMDAR hyperactivity has been associated with this hyperarousal and PTSD formation after trauma, NMDAR antagonists including ketamine have been proposed to both target this hyperarousal and provide rapid antidepressant effects (Liriano, Hatten, & Schwartz, 2019). A single dose of ketamine decreased subjective measures of intrusion, avoidance, and hyperarousal in patients with PTSD (Feder et al., 2014). Additionally, ketamine reduced the likelihood of developing PTSD after burn injury (McGhee, Maani, Garza, Gaylord, & Black, 2008). The partial NMDAR agonist D-cycloserine has also been investigated and was administered alongside exposure therapy to enhance extinction learning (Baker, Cates, & Luthin, 2017; George et al., 2018). Lastly, while sleep aids such as benzodiazepines, zolpidem, buspirone, and gabapentin have been tested in addition to cognitive behavioral therapy for PTSD-induced sleep disturbances, results are variable (Maher et al., 2006). Thus, as reiterated throughout, novel, disorder-specific medications that target sleep are still needed.

Emerging evidence supports targeting both group I and group II mGlu receptors for symptoms of PTSD and associated sleep disturbances. Importantly, unlike clinical and preclinical studies for other CNS disorders, mGlu receptor compounds have been assessed for their ability to improve sleep disturbances in the context of animal models of traumatic stress. First, functional antagonism of the mGlu5 receptor may be effective in treating both sleep disruptions and hyperarousal associated with PTSD. In vivo PET imaging showed that patients with PTSD have higher mGlu5 receptor availability in the hippocampus relative to healthy controls, and mGlu5 receptor availability correlated with heightened avoidance symptoms and suicidal ideations (Davis et al., 2019; Esterlis, Holmes, Sharma, Krystal, & Delorenzo, 2018; Holmes et al., 2017). In preclinical rodent models, treatments targeting mGlu5 receptors also show promise in treating PTSD due to their role in fear, avoidance, and extinction learning (Riedel, Casabona, Platt, MacPhail, & Nicoletti, 2000). As previously discussed, mGlu5 NAMs inhibit arousal and have been shown to selectively reduce REM sleep, and may modulate sleep disruptions induced by single prolonged stress in rodents (Nedelcovych et al., 2015). Given that the hippocampus influences memory consolidation and REM sleep and the amygdala regulates emotional processing (Rasch & Born, 2013; Wang, Liu, Li, Qu, & Huang, 2021), it is theorized that hippocampal and amygdalar hyperactivity during REM sleep decreases extinction learning, increases emotional response to trauma, and increases PTSD symptom manifestation (Bottary et al., 2020; Cominski, Jiao, Catuzzi, Stewart, & Pang, 2014; Pace-Schott, Germain, & Milad, 2015; Straus et al., 2018). Thus, mGlu5 NAMs may be a two-fold treatment approach that facilitates extinction learning and targets the sleep disturbances associated with PTSD.

To date, most work has focused on mGlu5 as a potential treatment for PTSD and associated sleep impairments. However, activation of group II receptors also produces selective decreases in REM sleep and preclinical studies support group II receptors as a therapeutic target for PTSD. Given their-expression in regions including the amygdala, both mGlu2 and mGlu3 receptors have been implicated in fear memory and extinction learning (Kim et al., 2015; Linden et al., 2005; Potter, Zanos, & Gould, 2020; Sweeten, Adkins, Wellman, & Sanford, 2021). Of relevance, Sweeten et al., 2021 explored the therapeutic potential of mGlu2/3 agonist LY379268 for sleep disturbances associated with fear extinction and found that a microinjection of LY379268 to the basolateral amygdala normalized the increased REM response associated with context re-exposure. Thus, group II receptors remain an exciting area of future research and should be further pursued. Lastly, it is important to note group III receptors (notably mGlu7 and mGlu8) also modulate learning and memory, including aversive and extinction learning, though their effects on sleep disturbances have not yet been studied (Fendt et al., 2013; O’Connor et al., 2019).

3.4.4. Alzheimer’s disease

As the most common form of dementia, Alzheimer’s Disease (AD) is characterized by neurodegeneration, aggregation of extracellular amyloid β (Aβ) into amyloid plaques, and the aggregation of intracellular tau into neurofibrillary tangles (NFTs), which begin to accumulate approximately 15 years before cognitive decline ( Jack et al., 2010; Jack et al., 2016). Importantly, sleep disruptions are common in both clinical populations (even in prodromal stages) and preclinical models, and these have correlated with cognitive decline (Lim, Kowgier, Yu, Buchman, & Bennett, 2013; Potvin et al., 2012; Romanella et al., 2021). In clinical populations, total sleep time and SWS percentage were decreased and REM sleep latency and waking after sleep onset (WASO) were increased in patients with AD when compared to healthy controls (Bliwise et al., 1989; Zhang et al., 2022). Furthermore, a recent meta-analysis examining polysomnographic sleep changes in patients with AD found a significant positive relationship between SWS and REM sleep duration and Mini-Mental Sate Examination (MMSE) scores, such that reduced sleep duration correlated with lower MMSE scores (Zhang et al., 2022).

Electroencephalography (EEG) studies in patients with AD have reported several age-related differences, especially in spectral waveforms, that may directly contribute to cognitive dysfunction and AD pathology (Lyashenko, Poluektov, Levin, & Pchelina, 2016; Scullin & Bliwise, 2015a, 2015b; Song et al., 2015; Xie et al., 2013; Zhang et al., 2022). Patients with AD demonstrated increased power in low frequency (delta/theta) bands and decreased alpha power when awake compared to healthy aged or young cohorts (Czigler et al., 2008; Rondina et al., 2016). These waveforms each have distinct roles in cognitive processes and, thus, may correspond with AD-associated cognitive decline. Additionally, findings of reduced delta power (0.5–4Hz) during NREM sleep, as well as sleep spindle abnormalities including reduced density, amplitude, and duration, have been reported in patients with AD; these further correlated with impaired cognitive performance (Scullin & Bliwise, 2015b; Weng, Lei, & Yu, 2020).

Importantly, disrupted sleep has been implicated as both a cause and a consequence of AD pathology. Homeostatic sleep is important for Aβ clearance, and, in healthy adult participants, disrupted slow wave activity was found to increase AD pathology and Aβ levels (Ju et al., 2017; Lucey et al., 2019). Furthermore, healthy individuals with amyloid deposition demonstrated worsened sleep efficiency compared to individuals without amyloid deposition (Ju et al., 2013). Murine models of AD-like pathology also support a relationship between sleep and pathological development. For example, in transgenic mice expressing the human amyloid precursor protein (APP), Aβ accumulation was observed in brain interstitial fluid following sleep deprivation (Kang et al., 2009). Additionally, sleep disturbances associated with AD may also contribute to sundown syndrome, a phenomenon in which neuropsychiatric symptoms such as delusions, irritation, and depression are exacerbated during the late afternoon and night hours (Canevelli et al., 2016). Although the underlying causes of these are not well understood, possible contributing factors include abnormal sleep schedules (e.g. reduced nighttime sleep and increased daytime napping) as well as circadian disruptions (e.g. degeneration of the SCN and decreased melatonin release) (Canevelli et al., 2016; Srinivasan, Pandi-Perumal, Cardinali, Poeggeler, & Hardeland, 2006; Stopa et al., 1999). Taken together, identifying at risk individuals with sleep disturbances early in life may present a relevant treatment window to slow or prevent pathological progression.

Recent evidence points to a glutamate hypothesis of AD, which is supported by the increased release and decreased clearance of synaptic glutamate (Revett, Baker, Jhamandas, & Kar, 2013; Tok, Ahnaou, & Drinkenburg, 2021). These alterations in glutamate levels may contribute to network hyperexcitability and, thereby, pathological development (Tok et al., 2021). Glutamate receptors have also been targeted for the treatment of AD. Aβ-mediated neurotoxicity has been hypothesized to occur via an NMDAR-activity dependent mechanism, and, thus, memantine, a non-competitive NMDAR antagonist, has been FDA-approved as a treatment for moderate to severe AD (Kumar et al., 2015; Miguel-Hidalgo, Alvarez, Cacabelos, & Quack, 2002; Rogawski & Wenk, 2003; Shankar et al., 2007). However, memantine is limited in that it only relieves symptoms and does not improve existing pathology (Matsunaga et al., 2018).

Several studies support targeting both group I and group II mGlu receptors for AD. Because mGlu5 is coupled to the NMDAR (Niswender & Conn, 2010; Shigemoto et al., 1993), reducing mGlu5 function may exert neuroprotective effects by blocking Aβ-mediated neurotoxicity, similar to memantine, with lesser adverse effects (Kumar et al., 2015). Furthermore, mGlu5 has been shown to directly engage with Aβ oligomers (Aβo). One possible contributor to AD pathology is Aβo binding to cellular prion protein (PrPc) which was shown in neuronal cultures to ultimately induce synaptic dysfunction (Laurén, Gimbel, Nygaard, Gilbert, & Strittmatter, 2009). Interaction of the Aβo-PrPc complex with mGlu5 induces downstream calcium mobilization and increases Aβ formation, and these effects may be associated with deficits in learning and memory (Hamilton, Esseltine, Devries, Cregan, & Ferguson, 2014; Um et al., 2013). In support of this interaction, both deletion of mGlu5 and blockade of mGlu5 with NAMs including MTEP in a mouse model of AD (APPswe/PS1ΔE9 mice) reversed deficits in spatial learning and working memory and prevented Aβo-associated changes in synaptic density (Budgett, Bakker, Sergeev, Bennett, & Bradley, 2022; Hamilton et al., 2014, 2016; Um et al., 2013). Lastly, there is preclinical support for both inhibition and activation of group II mGlu receptors. Support for inhibition as a therapeutic approach stems from findings that mGlu2 receptor activation increased accumulation of Aβ42 in cortical synaptoneurosomes (Kim et al., 2010). Pharmacological activation of mGlu2 receptors also contributed to microglial neurotoxicity, whereas blockade of mGlu2 receptors reduced microglia apoptosis (Taylor, Diemel, Cuzner, & Pocock, 2002). Importantly, increased mGlu2 receptor expression was found in hippocampal neurons in post-mortem tissue from patients with AD when compared to age-matched controls suggesting a close relationship between mGlu2 and AD pathogenesis (Lee et al., 2004). Conversely, there is some support for mGlu2/3 receptor activation for the treatment of AD. Caraci et al., 2011 found activation of both mGlu2 and mGlu3 receptors with LY379268 exerted neuroprotective effects, reducing Aβ-mediated neuronal death. Further, neuroprotective effects of LY379268 did not occur in cells lacking mGlu3 receptors and selective activation of mGlu2 receptors increased Aβ-mediated neurotoxicity suggesting a role for mGlu3 receptor activation in these neuroprotective effects (Caraci et al., 2011; Caraci, Nicoletti, & Copani, 2018).

mGlu5 NAMs may also hold promise for targeting sleep disturbances associated with AD. Aforementioned effects of mGlu5 NAMs on sleep, primarily increases in NREM sleep and NREM sleep quality, could mitigate the accelerated Aβ deposition caused by sleep disruptions and, thereby, slow cognitive impairment and behavioral effects (Ahnaou, Langlois, et al., 2015; Ahnaou, Raeymaekers, et al., 2015; Cavas et al., 2013b). Furthermore, while speculative, arousal enhancing effects of some mGlu5 NAMs may be favorable to target sundowning effects if administered at the correct time of day. For example, VU042438 has initial wake-promoting effects followed by compensatory increases in NREM sleep and increased NREM sleep quality (Holter et al., 2021). This profile could normalize sleep patterns that are perceived contributors to sundowning pathophysiology, serving to prevent daytime napping and increase nighttime sleep. However, further research is needed to determine if effects of mGlu5 NAMs on sleep in healthy rodents will translate similarly to rodents in models of AD-pathology and humans with and without AD. Lastly, while both inhibition and activation of mGlu2/3 receptors has been explored for the possible benefits in targeting AD pathology, the effects of these compounds on sleep in healthy animals (both decreased REM sleep) appear to counteract the desired effects in patients with Alzheimer’s disease. Nonetheless, examining these compounds on sleep in the context of AD is still important to acknowledge and address in future studies.

3.4.5. Substance use disorders

Current research suggests a bidirectional relationship between sleep and substance use/misuse. On one hand, sleep disturbances increase the risk of licit and illicit drug use which contributes to the subsequent development of substance use disorders (SUDs). For example, reduced sleep duration or quality as well as social jetlag (reduced sleep time on weekends) during adolescence is associated with increased problematic drinking and continued illicit substance use in adulthood (Dolsen & Harvey, 2017; Hasler & Pedersen, 2020; Roehrs et al., 2021; Troxel et al., 2021). Conversely, substance use also induces both acute and long-lasting sleep disruptions (Koob & Colrain, 2020; Roehrs et al., 2021; Valentino & Volkow, 2020). Although all illicit substances, as well as alcohol, cannabis, and nicotine alter sleep, specific disturbances and severity vary depending on a number of factors including, but not limited to, substance, duration, and frequency/typography of use. Chronic drug exposure associated with SUD often leads to long-lasting sleep disruptions including insomnia that last for weeks to months into abstinence. Importantly, sleep disturbances during abstinence have been correlated with increased craving and risk of relapse (Brower, 2003; Dolsen & Harvey, 2017; Lydon-Staley et al., 2017). Thus, as with other CNS disorders, targeting the sleep disruptions associated with SUD represents a promising treatment approach to not only prevent progression from recreational use to SUD but also to reduce relapse following abstinence. While most substances affect sleep (Hser et al., 2017; Jaehne, Loessl, Bárkai, Riemann, & Hornyak, 2009; Mondino et al., 2021), the following sections focus on relevant clinical and preclinical literature describing effects of cocaine and opioids on sleep.

3.4.5.1. Cocaine use disorder (CUD)

There is growing preclinical and clinical evidence for the presence of sleep disturbances throughout various stages of cocaine use disorder (CUD). As a stimulant, acute use of cocaine is associated with increased arousal and time awake and decreased total sleep time across species (Bjorness & Greene, 2018, 2021; Post, Gillin, Wyatt, & Goodwin, 1974; Schierenbeck, Riemann, Berger, & Hornyak, 2008). However, abstinence following chronic use is associated with persistent alterations in sleep. In general, polysomnography studies show a decrease in REM sleep on the first day of abstinence (Schierenbeck et al., 2008). Sleep disturbances further into abstinence include decreased total and SWS time, decreased REM latency, and increased REM duration, which may be a REM rebound effect (Bjorness & Greene, 2022; Schierenbeck et al., 2008). During early abstinence (<1 week), objective polysomnography and subjective self-report measures of sleep concurrently indicate decreased sleep time, increased sleep latency, and decreased sleep quality. Interestingly, although most patients continue to display insomnia-like symptoms on objective measures during later abstinence periods (>2 weeks), subjective reports do not align, as patients report either unchanged or improved sleep (Morgan et al., 2006; Morgan, Perry, Cho, Krystal, & D’Souza, 2006; Morgan & Malison, 2007; Pace-Schott et al., 2005). Importantly, the DSM-5 characterizes these sleep disturbances as a subset of stimulant-induced disorders, which are symptoms that last months to years into abstinence (APA, 2013).

Surprisingly, few studies to date have examined cocaine-associated sleep alterations in animal models of CUD. One nonhuman primate study using actigraphy-based assessments reported sleep fragmentation and decreased NREM sleep during abstinence following cocaine self-administration (Cortes et al., 2016). Further, only two rodent studies have used polysomnography to examine effects of repeated cocaine exposure on sleep. Compared to a drug-naïve baseline, Chen and colleagues reported sleep fragmentation and, in contrast to human reports, both decreased NREM and REM sleep following one week of cocaine self-administration (Chen etal., 2015). These effects were only in part recapitulated in a study examining sleep following one week of repeated, non-contingent administration of cocaine in mice (Bjorness & Greene, 2022). Thus, this suggests frequency, dose, duration, and route of administration (contingent vs non-contingent) may be influential factors. Lastly, reduced REM sleep was correlated with increased operant responding to a cocaine cue (incubation paradigm; a preclinical model of drug seeking) and restoring REM sleep reduced cue-associated responding (Chen et al., 2015). Thus, this study established a direct relationship between cocaine induced sleep-disruptions and future drug-related behaviors.

Impaired glutamate homeostasis has been widely implicated in CUD. The primary reinforcing effects of cocaine are attributed to the blockade of the dopamine transporter in the mesocorticolimbic dopamine pathway (Di Chiara & Imperato, 1988), and chronic cocaine self-administration is associated with a hypodopaminergic state (Goldstein & Volkow, 2002; Gould, Porrino, & Nader, 2012; Mateo, Lack, Morgan, Roberts, & Jones, 2005; Siciliano, Ferris, & Jones, 2016). However, cocaine self-administration also induces long-lasting glutamate-mediated increases in synaptic plasticity in mesocorticolimbic regions including the VTA, NAc, and PFC (Niedzielska-Andres et al., 2021; van Huijstee & Mansvelder, 2014). During abstinence, exposure to cocaine-paired cues increases cortical glutamate concentrations in the NAc, and this excessive glutamate-mediated response drives increased DA release (Korpi et al., 2015; Shin, Gadzhanova, Roughead, Ward, & Pont, 2016), a purported mechanism contributing to craving. Excessive cue-reactivity following extended periods of abstinence is associated with an accumulation of calcium-permeable AMPA receptors (CP-AMPARs; for review, see Wolf, 2016), and this process has been shown to be regulated via mGlu1 receptor activity (McCutcheon et al., 2011). Importantly, Chen and colleagues (2015) demonstrated that altering sleep impacted CP-AMPARs and subsequent cocaine-related behaviors in rats. Specifically, cocaine-induced REM sleep disruptions correlated with the accumulation of CP-AMPARs and restoring REM sleep during abstinence reduced CP-AMPARs and cocaine cue-reactivity (Chen et al., 2015). Alterations in mGlu receptors are also present during abstinence following cocaine self-administration. Rats showed lower mGlu2/3 receptor expression (Logan et al., 2020), density (Pomierny-Chamiolo, Miszkiel, Frankowska, & Mizera, 2017), and function (Moussawi et al., 2011) in the NAc as well as lower mGlu5 receptor expression in regions including the NAc and striatum (Hao et al., 2010; Knackstedt, Trantham-Davidson, & Schwendt, 2014; Swanson, Baker, Carson, Worley, & Kalivas, 2001).

3.4.5.2. Opioid use disorder (OUD)

Understanding the direct effects of opioids on human sleep is complicated by a number of factors including the type/frequency of use (prescription use, misuse, OUD), the presence or absence of pain, and whether studies use subjective or objective measures. Studies conducted in healthy, pain-free humans reported that opioid administration increased lighter NREM sleep stages (N1 and N2) and decreased N3 and REM sleep (Dimsdale, Norman, DeJardin, & Wallace, 2007; Shaw, Lavigne, & Mayer, 2006). Prescription opioid-dependent users with chronic pain experience subjective and objective (actigraphy-based) decreases in sleep duration and sleep quality (Hartwell, Pfeifer, McCauley, Moran-Santa Maria, & Back, 2014), and, in some instances, prescription opioids have been shown to further exacerbate sleep disturbances associated with chronic pain (Robertson et al., 2016). Moreover, illicit opioid use may affect sleep differently than FDA-approved opioids for the treatment of pain and OUD based on differences in potency/efficacy, route of administration, and related pharmacokinetic factors. In studies that used the Pittsburg Sleep Quality Index (PQSI) to assess sleep, 70–84% of sampled participants in methadone maintenance therapy were poor sleepers, and PQSI scores correlated with methadone dosage (Hsu et al., 2012; Stein et al., 2004). These studies are not exhaustive but highlight the complexity of interpreting opioid-related effects on sleep and reiterate that sleep is negatively impacted across conditions.

Specifically, within the context of OUD, sleep impairments including insomnia are some of the most common complaints during acute (days to weeks) and protracted (weeks to months) withdrawal (Dunn, Huhn, Bergeria, Gipson, & Weerts, 2019). Other common symptoms reported during abstinence periods in non-treatment seeking individuals with OUD include restless sleep, fatigue, and unusual dreaming (Hartwell et al., 2014; Tripathi, Rao, Dhawan, Jain, & Sinha, 2020). For example, increased N1 and reduced N2 sleep was associated with relapse in heroin detoxification patients at a 6-month follow-up assessment (Rady, Mekky, Moulokheya, & Elsheshai, 2020). Sleep disruption can also lead to decreased pain tolerance, which may influence subsequent opioid use (Eacret, Veasey, & Blendy, 2020). Together, similar to cocaine, chronic opioid use is associated with long-lasting sleep alterations and influences the likelihood of relapse.

Animal studies provide the opportunity to directly assess the effects of opioids on sleep, controlling for a number of the aforementioned confounding variables associated with human studies. In rats, acute morphine administration reduced NREM and REM sleep, yet NREM, but not REM, sleep duration returned to normal within three consecutive days of administration (Khazan, Weeks, & Schroeder, 1967). Acute effects of opioids are largely consistent between humans and rodents, except that opioids increased delta power during NREM sleep in rodents whereas opioids decreased delta power during N3 sleep in humans (Gauthier, Guzick, Brummett, Baghdoyan, & Lydic, 2011; Shaw et al., 2006). Additionally, actigraphy-based assessments in non-human primates were used to examine acute effects of the FDA-approved treatments for OUD on sleep-like parameters including sleep latency and efficiency (Berro, Zamarripa, Talley, Freeman, & Rowlett, 2022). Interestingly, while methadone (full mu opioid receptor agonist) and buprenorphine (partial mu opioid receptor agonist) both increased and decreased sleep latency and efficiency, respectively, disruptive effects of buprenorphine were longer lasting. In contrast, naltrexone (mu opioid receptor antagonist) reduced sleep latency and increased efficiency (Berro et al., 2022). While these studies were conducted in non-opioid dependent monkeys and did not employ polysomnography, they elegantly distinguished effects of activating or inhibiting opioid receptors on sleep-like parameters.

Similar to CUD, few preclinical studies have examined effects of chronic opioid self-administration and abstinence on sleep/wake cycles. In the only study to our knowledge, rats given 6-hr access to self-administer heroin during the light cycle for 2 weeks demonstrated a complete reversal in sleep pattern compared to a saline control group. REM and NREM sleep were increased during the dark period but decreased during the light period (Coffey, Guan, Grigson, & Fang, 2016). While NREM sleep duration returned to baseline levels in both the light and dark period within three days of abstinence, REM sleep impairments in the dark period persisted. These findings suggest altered recovery duration and possible underlying neurobiological alterations following heroin self-administration. Future preclinical research is needed to understand, in a well-controlled setting, how chronic opioid self-administration affects sleep and how FDA-approved opioid treatments affect sleep during abstinence.

There are many known and posited mechanisms through which opioids affect sleep-wake regulation. Reinforcing effects (e.g. abuse liability) of opioids are attributed to activation of mu-opioid receptors (MORs). MORs are broadly distributed throughout the CNS, influencing reward, pain, stress reactivity, mood, cognition and sleep (for review, see Welsch, Bailly, Darcq, & Kieffer, 2020). Due to the broad receptor distribution, opioids likely affect sleep-wake regulation via multiple circuits including excitation of the mesolimbic dopamine system and direct activation of MORs on hypocretin-containing hypothalamic neurons (Eacret et al., 2020). Additionally, opioids influence the suprachiasmatic nucleus affecting arousal and circadian rhythm and disrupt sleep by inhibiting respiratory activity and contributing to sleep apnea (Bergum, Berezin, & Vigh, 2022; Rosen et al., 2019; Wang & Teichtahl, 2007). While these are noted mechanisms through which acute exposure may alter sleep-wake regulation, similar to CUD, neurobiological alterations spanning many of these circuits may contribute to the long-lasting sleep disturbances associated with OUD.

Extensive evidence implicates excessive glutamate associated with OUD as a contributor to opioid-associated withdrawal symptoms and risk for relapse. In fact, hyperglutamatergic function may be a critical component of withdrawal. Glutamate concentrations within the locus coeruleus (LC), a noradrenergic nucleus in the brainstem, have been shown to increase during naloxone-precipitated withdrawal in opioid-dependent rats (Feng, Zhang, Rockhold, & Ho, 1995) and acute injections of glutamate into the LC induced behavioral signs of withdrawal similar to naloxone in morphine-dependent animals (Medrano, Mendiguren, & Pineda, 2015; Sekiya, Nakagawa, Ozawa, Minami, & Satoh, 2004; Tokuyama, Wakabayashi, & Ho, 1995; Zhu, Rockhold, & Ho, 1998). Given that the LC is largely implicated in OUD and associated withdrawal, as well as a key contributor for wake-promoting circuits, reducing glutamatergic function may be a viable therapeutic approach for multiple symptoms of OUD during abstinence including sleep disturbances. Additionally, like cocaine, long-lasting glutamatergic-mediated synaptic plasticity changes likely contribute to drug craving and risk of relapse (van Huijstee & Mansvelder, 2014). While less well characterized, recent evidence shows increased CP-AMPARs in the NAc that contribute to drug seeking during abstinence following oxycodone self-administration, which is a finding similar to cocaine studies (Wong, Zimbelman, Milovanovic, Wolf, & Stefanik, 2022).

3.4.5.3. Modulating mGlu receptors as treatments for SUD

There are currently 3 FDA-approved pharmacological treatments for OUD and no pharmacological treatments for CUD. While each is successful in some regard, all have adverse risk factors and none directly target sleep disturbances. Decades of research have examined compounds targeting various mGlu receptor subtypes as treatments for SUD. For example, mGlu5 antagonists/NAMs as well as mGlu2/3 PAMs have shown promise in reducing cocaine and opioid-related behaviors in rodent and nonhuman primate models of drug taking or seeking (Amato et al., 2013; Gould et al., 2016; Gould, Felts, & Jones, 2017; Johnson & Lovinger, 2020; Keck et al., 2013; Kumaresan et al., 2009; Lee, Platt, Rowlett, Adewale, & Spealman, 2005; Lou, Chen, Liu, Ruan, & Zhou, 2014; Salling, Grassetti, Ferrera, Martinez, & Foltin, 2021; van der Kam, de Vry, & Tzschentke, 2007). Moreover, many of these compounds have demonstrated anxiolytic-like and antidepressant-like activity suggesting potential to alleviate anxiety/depressive symptoms often present during withdrawal (Gould et al., 2016). Lastly, cocaine abstinence-induced accumulation of CP-AMPARs can be restored followed administration of an mGlu1 receptor agonist (McCutcheon et al., 2011), although this has not been examined in the context of sleep, nor extended to studies involving opioids.

A causal relationship has not yet been established between sleep disruptions and altered mGlu receptor function during abstinence following cocaine or opioid self-administration. However, based on KO and pharmacological studies in drug-naïve rodents (Tables 1 and 2), we can speculate on possible treatment approaches. Results from several studies suggest targeting mGlu1 and mGlu2/3 receptors for CUD and OUD may benefit both abstinence-related behaviors and abstinence-induced sleep disturbances including insomnia and decreases in REM sleep. For example, mGlu1 agonists may alter CP-AMPARs during abstinence in part by altering sleep. CP-AMPARs have a strong role in regulating sleep and sleep-maintained synaptic homeostasis and, thus, balancing these may normalize the restorative benefits of sleep including memory consolidation (for review, see Shepherd, 2012 and Martin et al., 2019). Additionally, some novel mGlu2/3 PAMs may be able to target aberrant decreases in NREM sleep associated with abstinence. While mGlu5 NAMs are promising for reducing drug taking/seeking, these compounds may not produce the desired effects on sleep as it relates to findings in some, but not all, rodent and human studies (i.e. mGlu5 NAMs reduce NREM and REM sleep when increased NREM and REM sleep is needed). However, conflicting evidence between clinical and preclinical studies that show increased and decreased REM sleep during abstinence, respectively, indicates the appropriate way to target REM sleep still needs further investigation. Additionally, this does not discount the ability of these compounds to normalize possible sleep disturbances associated with stimulant-induced disorders including anxiety and depression. Lastly, though there is minimal literature surrounding group III mGlu receptors and sleep, it is important to note that activating mGlu7 and mGlu8 receptors was effective in attenuating self-administration, reinstatement, and withdrawal as it relates to cocaine and opioids (Hajasova, Canestrelli, Acher, Noble, & Marie, 2018; Kahvandi et al., 2021; Li et al., 2013; Luessen & Conn, 2022; Pałucha-Poniewiera & Pilc, 2013; Salling et al., 2021). Together, these studies provide compelling rationale to further examine group I-III mGlu receptor compounds in multiple preclinical models of SUD, with a particular need to examine associated impacts on sleep disturbances.

4. Discussion

As described in this chapter and summarized in Tables 3 and 4, sleep disturbances including insomnia are prevalent in many neuropsychiatric disorders, and there is a clear bidirectional relationship between the two. Sleep disturbances can precede traditional symptom onset and/or correlate with symptom severity or relapse (e.g. schizophrenia, MDD, SUD, AD). Sleep disturbances may also contribute to or exacerbate neuropathology associated with disorders including AD. Although disrupted sleep is common in both subjective and objective reports, primary pharmacological treatments for neuropsychiatric disorders have historically disregarded sleep as a modifiable symptom. Importantly, the DSM-5 distinguished CNS disorders and sleep disturbances as separate diagnoses. While common sleep medications including benzodiazepines and z-drugs are often prescribed for many neuropsychiatric disorders, they all decrease arousal by increasing inhibitory GABA activity (Benca & Buysse, 2018). Thus, depending on the disorder, these compounds may not directly affect the disrupted neurobiology/circuits implicated in disrupted sleep-wake activity in a disease-specific manner. Given that many CNS disorders have seen a dearth of novel treatments, future advancements should investigate sleep modifying pharmacotherapies in parallel with strategies to mitigate other symptoms.

Table 3.

Objective measures of sleep disruptions in neuropsychiatric disorders.

Neuropsychiatric condition NREM REM Awakenings qEEG Sleep spindles Comorbidity
Duration Latency Duration latency Density Bout Length % WASO Sleep fragmentation NREM Delta Number Density Sleep disorders
Schizophrenia
1,4–13
n.s n.s
Major Depressive Disorder
1,14–24
Post-traumatic stress disorder
25–39
↓ ; n.s ↕ ; n.s n.s n.s ↑ ; n.s
Alzheimer’s Disease
1,40–42
↓ ; n.s
Substance Use Disorders: Withdrawal and Abstinence Cocaine (Early Abstinence)
2,3,43–50
Cocaine (Later Abstinence)
2,3,43–49
↑ ; n.s -
Opioid (w/o medication assistance)
2,3,50–57

n.s., not significant; —, not determined; qEEG, quantitative EEG; REM Density, number of REMbouts per minute of sleep; % REM, duration of REM out of total sleep time; WASO, time awake after sleep onset until final awakening; Early abstinence, <1 week; Later abstinence, >2 weeks.

1Benca et al. (1992), 2Angarita, Emadi, Hodges, and Morgan (2016), 3Dolsen and Harvey (2017), 4Cohrs (2008), 5Gerstenberg et al. (2020), 6Göder et al. (2006), 7Hodges, Pittman, and Morgan (2017), 8Kaskie, Graziano, and Ferrarelli (2017), 9Keshavan et al. (1998), 10Manoach et al. (2004), 11Manoach and Stickgold (2009), 12Manoach and Stickgold (2019), 13Sprecher et al. (2015), 14Armitage, Hudson, Trivedi, and Rush (1995), 15Armitage, Hoffmann, Fitch, Morel, and Bonato (1995), 16Bovy et al. (2021), 17Coble, Foster, and Kupfer (1976), 18Steiger, Pawlowski, and Kimura (2015), 19Steiger and Kimura (2010), 20Steiger and Pawlowski (2019), 21Steiger et al. (2015), 22Tesler et al. (2016), 23Wichniak, Antczak, Wierzbicka, and Jernajczyk (2002), 24Wichniak et al. (2013), 25Baird et al. (2018), 26Khazaie and Masoudi (2016), 27Kobayashi et al. (2007), Kobayashi et al. (2012), 29Krakow, Ulibarri, Moore, and McIver (2015), 30Laniepce et al. (2020), 31Le Bon et al. (1997), 32Leeman-markowski et al. (2020), 33Lewis et al. (2020), 34Mellman, Bustamante, Fins, Pigeon, and Nolan (2002), 35Mellman, Pigeon, Nowell, and Nolan (2007), 36Mellman, Kobayashi, Lavela, Wilson, and Hall Brown (2014), 37Neylan et al. (2021), 38Swift (2020), 39Woodward et al. (2002), 40Bliwise (1993), 41Scullin and Bliwise (2015a, 2015b), 42Zhang et al. (2022), 43Bjorness and Greene (2021), 44Chen et al. (2015), 45Johanson, Roehrs, Schuh, and Warbasse (1999), 46Matuskey, Pittman, Forselius, Malison, and Morgan (2011), 47Morgan, Pace-Schott, et al. (2006), Morgan, Perry, et al., 2006, 48Schierenbeck et al. (2008), 49Valladares and Irwin (2007), 50Chen, Ting, Wu, Lin, and Gossop (2017), 51Eckert and Yaggi (2022), 52Gauthier et al. (2011), 53Kay (1975), 54Khazan and Colasanti (1972), 55Rosen et al. (2019), 56Shaw et al. (2006), 57Wang and Teichtahl (2007).

Table 4.

Subjective assessments of sleep disruptions in neuropsychiatric disorders.

Neuropsychiatric condition Sleep Nightmares Limb movement Daytime napping
Duration Latency Quality WASO
Schizophrenia
2–7
Major depressive disorder
8–11
Post-traumatic stress disorder
12–16
Alzheimer’s disease
17–22
Substance use disorders: Withdrawal and abstinence Cocaine (Early abstinence) 1,23–27
Cocaine (Later abstinence) 1,23–27
Opioid (w/o medication assistance)
1,28–30

As highlighted throughout, glutamate is a key regulator of homeostatic sleep, and glutamate dysfunction is commonly associated with many CNS disorders. Thus, it is important to investigate how glutamate dysfunction may directly contribute to sleep disturbances associated with specific CNS disorders. This chapter provided an overview of known roles of glutamatergic and, more specifically, mGlu receptor regulation of the sleep-wake cycle. Key contributions of multiple mGlu receptor subtypes in sleep regulation are discussed alone and in context of multiple CNS disorders. This chapter also highlights current knowledge gaps and speculates on plausible mGlu receptor subtypes to target that mitigate both the primary symptoms of a specific disorder and the associated sleep disturbances. We argue that, while possible to identify a singular treatment to augment sleep across multiple conditions, given the heterogeneity of CNS disorders, unique treatments based on neurobiological alterations may have greater therapeutic efficacy across multiple symptoms.

As summarized in Table 1 and 2, much of our understanding of mGlu receptor contributions to sleep regulation stem from constitutive knockout studies or studies using acute pharmacological challenges with subtype selective (or preferring) compounds. While these studies are vital to our basic understanding, methodological considerations should be discussed to assist with accurate interpretation of findings. For example, the absence of altered baseline sleep between wild type mice and mice with genetically altered receptor expression/function should not entirely rule out the possible involvement of that receptor influencing sleep regulation. Given the highly conserved circuitry regulating the sleep-wake cycle, constitutive knockout may result in compensatory functional and circuit-level changes that mask subtle differences. Moreover, baseline sleep comparisons reflect normal sleep regulation but not the mechanisms regulating sleep following a perturbation (sleep restriction/deprivation, stressor, etc.). For example, mGlu5 studies found disruptions in homeostatic sleep drive in KO animals following a sleep deprivation period (Ahnaou, Raeymaekers, et al., 2015; Holst et al., 2017). Importantly, while many studies have examined direct effects of mGlu receptor knockouts (notably mGlu2/3, mGlu5, mGlu7) or altered downstream signaling (e.g. PLCβ4, a downstream effector of mGlu1) as shown in Table 1, direct effects of mGlu1, mGlu4, and mGlu8 have not yet been examined. Furthermore, recent advances in neuroscience methods afford the exciting opportunity to examine sleep-wake activity changes at baseline and in response to perturbations. These include studies with conditional genetic alterations in mice as well as circuit/cell-specific modulation via opto/chemogenetic techniques.

Similar methodological considerations should be reviewed when designing/interpreting pharmacological studies including time of dosing and selectivity of pharmacological effects. In rodent studies, compounds administered during the dark (active) phase can provide information regarding general sleep-promoting effects (or sedative effects based on quantitative assessments of spectral power distribution) whereas compound administration during the light (quiescent) phase may better align with investigating wake-promoting/sleep-disrupting effects, NREM:REM ratios/latencies, and delta power during NREM sleep. Given the known fluctuations in both neurotransmitters and receptor cycling across diurnal periods, compounds may have divergent profiles on sleep-wake duration and architecture depending on time of dosing. Examining pharmacological effects on rodent sleep-wake behavior in both periods is even more critical when considering that recently developed compounds with increased mGlu receptor subtype selectivity predominately involve allosteric modulators (for review, see Luessen & Conn, 2022). Unlike compounds that bind to the orthosteric site, that activate/inhibit receptor function regardless of endogenous physiological conditions, allosteric modulators require the presence of an endogenous agonist (e.g. glutamate) to augment intracellular signaling. In this regard, allosteric modulation may provide unprecedented temporal selectivity, effectively modulating signaling when endogenous glutamate is present at higher concentrations (wake and REM) without altering sleep/wake state when glutamate is lower (NREM). Importantly, most pharmacological studies included in this chapter examined acute but not chronic pharmacological effects on sleep-wake cycles. Many studies have also not examined effects of these compounds on sleep in higher order, gyrencephalic species with more analogous sleep-wake patterns to humans. Lastly, it must be noted that, given the absence of literature examining mGlu receptor modulation on sleep in preclinical models of a CNS disorder, we have speculated on most potential pharmacotherapeutic applications. Future studies are thus needed to expand on findings from pharmacological assessments in otherwise healthy animals and incorporate these methodologies into animal models of disease states and/or in clinical populations to confirm/refute these hypotheses.

In summary, normalizing sleep disturbances associated with CNS disorders represents a novel treatment approach. Improving sleep may serve as a critical component in the therapeutic outcome, and restoration of homeostatic sleep may have several downstream benefits for other primary symptoms of a CNS disorder. Recent development of mGlu receptor-subtype selective ligands presents an opportunity to further interrogate the role of group I, II and III mGlu receptor regulation of the sleep-wake cycle with separate or synergistic effects on other symptoms. In general, group I mGlu receptors appear to be involved in homeostatic sleep pressure in addition to influences in sleep duration (Aguilar et al., 2020; Martin et al., 2019) whereas group II receptors appear to be more so involved in cortical arousal and selective effects on REM sleep following activation (Ahnaou et al., 2009; Pritchett et al., 2015). Fewer studies have examined group III, though their role (or lack thereof) will likely be determined in future studies with the recent development of more subtype-selective ligands (Lin et al., 2022; Luessen & Conn, 2022).

Importantly, the potential for specific mGlu receptors and the associated pharmacological effect (activation or inhibition) to ameliorate sleep disturbances and primary symptoms varies across disorders. For example, where mGlu5 agonists/PAMs may have pharmacotherapeutic relevance for sleep disturbances and symptoms in schizophrenia (Maksymetz et al., 2017), mGlu5 antagonists/NAMs may be more relevant for MDD, PTSD, AD, and SUD (Hamilton et al., 2014; Luessen & Conn, 2022). Likewise, both activation and inhibition of mGlu2/3 may be therapeutically relevant for MDD (Dogra & Conn, 2021) whereas activation may be most relevant with regard to schizophrenia (Maksymetz et al., 2017). The current, most effective therapies for both schizophrenia and depression influence sleep, predominantly through increasing sleep duration (antipsychotics; Cohrs, 2008) or selectively decreasing REM sleep (antidepressants; Steiger & Pawlowski, 2019; Wichniak et al., 2017). Thus, systematically examining sleep as a primary or secondary endpoint should be considered in the development of novel therapies. Recent preclinical studies investigating mGlu5 NAMs and mGlu2/3 PAMs have shown antidepressant-like effects, both in classic models and on sleep (Ahnaou et al., 2009; Gould et al., 2016; Holter et al., 2021; Lindemann et al., 2015). Several compounds targeting mGlu receptors have advanced to clinical trials for disorders including schizophrenia and MDD. For example, both mGlu5 NAMs (Quiroz et al., 2016) and mGlu2/3 antagonists (see Maksymetz et al., 2017 for discussion) have advanced to phase II for the treatment of MDD but were stalled due to a lack of significant effects on primary endpoints despite promising results on secondary endpoints. As multiple development programs progress forward with novel mGlu receptor subtype selective compounds for various indications, incorporating sleep measures into preclinical and clinical studies (although costly) should be considered. Lastly, while this chapter highlights the importance of mGlu receptor function on sleep in the context of schizophrenia, MDD, PTSD, SUD, and AD, there is impetus for targeting sleep disturbances as a treatment approach for numerous other neurodegenerative and neurodevelopmental disorders.

References

  1. Aeschbach D, Sher L, Postolache TT, Matthews JR, Jackson MA, & Wehr TA (2003). A longer biological night in long sleepers than in short sleepers. The Journal of Clinical Endocrinology and Metabolism, 88(1), 26–30. 10.1210/jc.2002-020827. [DOI] [PubMed] [Google Scholar]
  2. Ağargün MY, Kara H, & Solmaz M (1997). Subjective sleep quality and suicidality in patients with major depression. Journal of Psychiatric Research, 31(3), 377–381. 10.1016/S0022-3956(96)00037-4. [DOI] [PubMed] [Google Scholar]
  3. Aguilar XDD, Strecker RE, Basheer R, & Mcnally JM (2020). Alterations in sleep, sleep spindle, and EEG power in mGluR5 knockout mice. Journal of Neurophysiology, 123(1), 22–33. 10.1152/jn.00532.2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Ahnaou A, Dautzenberg FM, Geys H, Imogai H, Gibelin A, Moechars D, et al. (2009). Modulation of group II metabotropic glutamate receptor (mGlu2) elicits common changes in rat and mice sleep-wake architecture. European Journal of Pharmacology, 603(1–3), 62–72. 10.1016/j.ejphar.2008.11.018. [DOI] [PubMed] [Google Scholar]
  5. Ahnaou A, Langlois X, Steckler T, Bartolome-Nebreda JM, & Drinkenburg WHIM (2015). Negative versus positive allosteric modulation of metabotropic glutamate receptors (mGluR5): Indices for potential pro-cognitive drug properties based on EEG network oscillations and sleep-wake organization in rats. Psychopharmacology, 232(6), 1107–1122. 10.1007/s00213-014-3746-4. [DOI] [PubMed] [Google Scholar]
  6. Ahnaou A, Raeyemaekers L, Huysmans H, & Drinkenburg WHIM (2016). Off-target potential of AMN082 on sleep EEG and related physiological variables: Evidence from mGluR7 (−/−) mice. Behavioural Brain Research, 311, 287–297. 10.1016/j.bbr.2016.05.035. [DOI] [PubMed] [Google Scholar]
  7. Ahnaou A, Raeymaekers L, Steckler T, & Drinkenbrug WHIM (2015). Relevance of the metabotropic glutamate receptor (mGluR5) in the regulation of NREM-REM sleep cycle and homeostasis: Evidence from mGluR5 (−/−) mice. Behavioural Brain Research, 282, 218–226. 10.1016/j.bbr.2015.01.009. [DOI] [PubMed] [Google Scholar]
  8. Ahnaou A, ver Donck L, & Drinkenburg WHIM (2014). Blockade of the metabotropic glutamate (mGluR2) modulates arousal through vigilance states transitions: Evidence from sleep-wake EEG in rodents. Behavioural Brain Research, 270, 56–67. 10.1016/j.bbr.2014.05.003. [DOI] [PubMed] [Google Scholar]
  9. Alam A, & Chengappa KNR (2011). Obstructive sleep apnoea and schizophrenia: A primer for psychiatrists. Acta Neuropsychiatrica, 23(5), 201–209. 10.1111/j.1601-5215.2011.00589.x. [DOI] [PubMed] [Google Scholar]
  10. Altamura CA, Mauri MC, Ferrara A, Moro AR, D’Andrea G, & Zamberlan F (1993). Plasma and platelet excitatory amino acids in psychiatric disorders. The American Journal of Psychiatry, 150(11), 1731–1733. 10.1176/ajp.150.11.1731. [DOI] [PubMed] [Google Scholar]
  11. Altinbilek B, & Manahan-Vaughan D (2007). Antagonism of group III metabotropic glutamate receptors results in impairment of LTD but not LTP in the hippocampal CA1 region, and prevents long-term spatial memory. The European Journal of Neuroscience, 26(5), 1166–1172. 10.1111/j.1460-9568.2007.05742.x. [DOI] [PubMed] [Google Scholar]
  12. Altinbilek B, & Manahan-Vaughan D (2009). A specific role for group II metabotropic glutamate receptors in hippocampal long-term depression and spatial memory. Neuroscience, 158(1), 149–158. 10.1016/j.neuroscience.2008.07.045. [DOI] [PubMed] [Google Scholar]
  13. Amato RJ, Felts AS, Rodriguez AL, Venable DF, Morrison RD, Byers FW, et al. (2013). Substituted 1-phenyl-3-(pyridin-2-yl)urea negative allosteric modulators of mGlu5: Discovery of a new tool compound VU0463841 with activity in rat models of cocaine addiction. ACS Chemical Neuroscience, 4(8), 1217–1228. 10.1021/cn400070k. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. American Psychological Association. (2017). Clinical Practice Guideline for the Treatment of Posttraumatic Stress Disorder (PTSD). Vol. 139. Washington, DC: APA, Guideline Development Panel for the Treatment of Posttraumatic Stress Disorder in Adults. 10.1162/jocn. [DOI] [Google Scholar]
  15. Anaclet C, Ferrari L, Arrigoni E, Bass CE, Saper CB, Lu J, et al. (2014). The GABAergic parafacial zone is a medullary slow wave sleep-promoting center. Nature Neuroscience, 17(9), 1217–1224. 10.1038/nn.3789. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Angarita GA, Emadi N, Hodges S, & Morgan PT (2016). Sleep abnormalities associated with alcohol, cannabis, cocaine, and opiate use: A comprehensive review. Addiction Science and Clinical Practice, 11(1), 1–17. 10.1186/s13722-016-0056-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Anwyl R (1999). Metabotropic glutamate receptors: Electrophysiological properties and role in plasticity. Brain Research. Brain Research Reviews, 29(1), 83–120. 10.1016/s0165-0173(98)00050-2. [DOI] [PubMed] [Google Scholar]
  18. APA. (2013). Diagnostic and statistical manual of mental disorders (5th ed.). [Google Scholar]
  19. Armitage R (1995). The distribution of EEG frequencies in REM and NREM sleep stages in healthy young adults. Sleep, 18(5), 334–341. 10.1093/sleep/18.5.334. [DOI] [PubMed] [Google Scholar]
  20. Armitage R (2007). Sleep and circadian rhythms in mood disorders. Acta Psychiatrica Scandinavica, 433, 104–115. 10.1111/j.1600-0447.2007.00968.x. [DOI] [PubMed] [Google Scholar]
  21. Armitage R, Hoffmann R, Fitch T, Morel C, & Bonato R (1995). A comparison of period amplitude and power spectral analysis of sleep EEG in normal adults and depressed outpatients. Psychiatry Research, 56(3), 245–256. 10.1016/0165-1781(95)02615-4. [DOI] [PubMed] [Google Scholar]
  22. Armitage R, Hudson A, Trivedi M, & Rush AJ (1995). Sex differences in the distribution of EEG frequencies during sleep: Unipolar depressed outpatients. Journal of Affective Disorders, 34(2), 121–129. 10.1016/0165-0327(95)00009-C. [DOI] [PubMed] [Google Scholar]
  23. Aston-Jones G, Smith RJ, Moorman DE, & Richardson KA (2009). Role of lateral hypothalamic orexin neurons in reward processing and addiction. Neuropharmacology, 56(Suppl. 1), 112–121. 10.1016/j.neuropharm.2008.06.060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Averill LA, Jiang L, Purohit P, Coppoli A, Averill CL, Roscoe J, et al. (2022). Prefrontal glutamate neurotransmission in PTSD: A novel approach to estimate synaptic strength in vivo in humans. Chronic Stress, 6. 10.1177/24705470221092734. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Baird T, Theal R, Gleeson S, McLeay S, O’Sullivan R, Initiative PTSD, et al. (2018). Detailed polysomnography in Australian Vietnam veterans with and without posttraumatic stress disorder. Journal of Clinical Sleep Medicine, 14(9), 1577–1586. 10.5664/jcsm.7340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Baker JF, Cates ME, & Luthin DR (2017). D-cycloserine in the treatment of post-traumatic stress disorder. Mental Health Clinician, 7(2), 88–94. 10.9740/mhc.2017.03.088. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Battaglia G, Busceti CL, Molinaro G, Biagioni F, Traficante A, Nicoletti F, et al. (2006). Pharmacological activation of mGlu4 metabotropic glutamate receptors reduces nigrostriatal degeneration in mice treated with 1-methyl-4-phenyl-1,2,3,6-tetra-hydropyridine. The Journal of Neuroscience, 26(27), 7222–7229. 10.1523/JNEUROSCI.1595-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Bazhenov M, Timofeev I, Steriade M, & Sejnowski T (2000). Spiking-bursting activity in the thalamic reticular nucleus initiates sequences of spindle oscillations in thalamic networks. Journal of Neurophysiology, 84(2), 1076–1087. 10.1152/jn.2000.84.2.1076. [DOI] [PubMed] [Google Scholar]
  29. Benca RM, & Buysse DJ (2018). Reconsidering insomnia as a disorder rather than just a symptom in psychiatric practice. The Journal of Clinical Psychiatry, 79(1). 10.4088/JCP.me17008ah1c. [DOI] [PubMed] [Google Scholar]
  30. Benca RM, William H, Thisted RA, & Gillin JC (1992). Sleep and psychiatric disorders: A meta-analysis. Sleep and Psychiatric Disorders, Vol. 49, 651. Benca RM, Obermeyer WH, Thisted RA, Gillin JC. 10.1001/archpsyc.1992.01820080059010. [DOI] [PubMed] [Google Scholar]
  31. Benneyworth MA, Xiang Z, Smith RL, Garcia EE, Conn PJ, & Sanders-Bush E (2007). A selective positive allosteric modulator of metabotropic glutamate receptor subtype 2 blocks a hallucinogenic drug model of psychosis. Molecular Pharmacology, 72(2), 477–484. 10.1124/mol.107.035170. [DOI] [PubMed] [Google Scholar]
  32. Bergum N, Berezin C-T, & Vigh J (2022). A retinal contribution to opioid-induced sleep disorders? Frontiers in Neuroscience, 16, 981939. 10.3389/fnins.2022.981939. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Berman RM, Cappiello A, Anand A, Oren DA, Heninger GR, Charney DS, et al. (2000). Antidepressant effects of ketamine in depressed patients. Biological Psychiatry, 47(4), 351–354. 10.1016/S0006-3223(99)00230-9. [DOI] [PubMed] [Google Scholar]
  34. Berro LF, Zamarripa CA, Talley JT, Freeman KB, & Rowlett JK (2022). Effects of methadone, buprenorphine, and naltrexone on actigraphy-based sleep-like parameters in male rhesus monkeys. Addictive Behaviors, 135, 107433. 10.1016/j.addbeh.2022.107433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Bjorness TE, & Greene RW (2018). Dose response of acute cocaine on sleep/waking behavior in mice. Neurobiology of Sleep and Circadian Rhythms, 5, 84–93. 10.1016/j.nbscr.2018.02.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Bjorness TE, & Greene RW (2021). Interaction between cocaine use and sleep behavior: A comprehensive review of cocaine’s disrupting influence on sleep behavior and sleep disruptions influence on reward seeking. Pharmacology Biochemistry and Behavior, 206(November 2020), 173194. 10.1016/j.pbb.2021.173194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Bjorness TE, & Greene RW (2022). Arousal-mediated sleep disturbance persists during cocaine abstinence in male mice. Frontiers in Neuroscience, 16, 868049. 10.3389/fnins.2022.868049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Black JE, Kodish IM, Grossman AW, Klintsova AY, Orlovskaya D, Vostrikov V, et al. (2004). Pathology of layer V pyramidal neurons in the prefrontal cortex of patients with schizophrenia. The American Journal of Psychiatry, 161(4), 742–744. 10.1176/appi.ajp.161.4.742. [DOI] [PubMed] [Google Scholar]
  39. Bliwise DL (1993). Sleep in normal aging and dementia. Sleep, 16(1), 40–81. 10.1093/sleep/16.1.40. [DOI] [PubMed] [Google Scholar]
  40. Bliwise DL, Tinklenberg J, Yesavage JA, Davies H, Pursley AM, Petta DE, et al. (1989). REM latency in Alzheimer’s disease. Biological Psychiatry, 25(3), 320–328. 10.1016/0006-3223(89)90179-0. [DOI] [PubMed] [Google Scholar]
  41. Boissard R, Gervasoni D, Schmidt MH, Barbagli B, Fort P, & Luppi PH (2002). The rat ponto-medullary network responsible for paradoxical sleep onset and maintenance: A combined microinjection and functional neuroanatomical study. European Journal of Neuroscience, 16(10), 1959–1973. 10.1046/j.1460-9568.2002.02257.x. [DOI] [PubMed] [Google Scholar]
  42. Borbély AA, Daan S, Wirz-Justice A, & Deboer T (2016). The two-process model of sleep regulation: A reappraisal. Journal of Sleep Research, 25(2), 131–143. 10.1111/jsr.12371. [DOI] [PubMed] [Google Scholar]
  43. Borbély AA, & Wirz-Justice A (1982). Sleep, sleep deprivation and depression. A hypothesis derived from a model of sleep regulation. Human Neurobiology, 1(3), 205–210. [PubMed] [Google Scholar]
  44. Borges CR, Piovezan RD, Poyares DR, Busatto Filho G, Studart-Neto A, Coutinho AM, et al. (2021). Subjective sleep parameters in prodromal Alzheimer’s disease: A case-control study. Brazilian Journal of Psychiatry, 43(5), 510–513. 10.1590/1516-4446-2020-1503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Bottary R, Seo J, Daffre C, Gazecki S, Moore KN, Kopotiyenko K, et al. (2020). Fear extinction memory is negatively associated with REM sleep in insomnia disorder. Sleep, 43(7), 1–12. 10.1093/SLEEP/ZSAA007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Bovy L, Weber FD, Tendolkar I, Fernández G, Czisch M, Steiger A, et al. (2021). Non-REM sleep in major depressive disorder. BioRxiv. 2021.03.19.436132 https://www.biorxiv.org/content/10.1101/2021.03.19.436132v1%0Ahttps://www.biorxiv.org/content/10.1101/2021.03.19.436132v1.abstract. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Bradley SR, Uslaner JM, Flick RB, Lee A, Groover KM, & Hutson PH (2012). The mGluR7 allosteric agonist AMN082 produces antidepressant-like effects by modulating glutamatergic signaling. Pharmacology, Biochemistry, and Behavior, 101(1), 35–40. 10.1016/j.pbb.2011.11.006. [DOI] [PubMed] [Google Scholar]
  48. Brady K, Pearlstein T, Asnis GM, Baker D, Rothbaum B, Sikes CR, et al. (2000). Efficacy and safety of sertraline treatment of posttraumatic stress disorder: A randomized controlled trial. JAMA, 283(14), 1837–1844. 10.1001/jama.283.14.1837. [DOI] [PubMed] [Google Scholar]
  49. Breslau N, Roth T, Rosenthal L, & Andreski P (1996). Sleep disturbance and psychiatric disorders: A longitudinal epidemiological study of young adults. Biological Psychiatry, 39(6), 411–418. 10.1016/0006-3223(95)00188-3. [DOI] [PubMed] [Google Scholar]
  50. Brower KJ (2003). Insomnia, alcoholism and relapse. Sleep Medicine Reviews, 7(6), 523–539. 10.1016/s1087-0792(03)90005-0. [DOI] [PubMed] [Google Scholar]
  51. Brown RE, Basheer R, McKenna JT, Strecker RE, & McCarley RW (2012). Control of sleep and wakefulness. Physiological Reviews, 92(3), 1087–1187. 10.1152/physrev.00032.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Bruno V, Battaglia G, Casabona G, Copani A, Caciagli F, & Nicoletti F (1998). Neuroprotection by glial metabotropic glutamate receptors is mediated by transforming growth factor-beta. The Journal of Neuroscience: The Official Journal of the Society for Neuroscience, 18(23), 9594–9600. 10.1523/JNEUROSCI.18-23-09594.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Bruno V, Ksiazek I, Battaglia G, Lukic S, Leonhardt T, Sauer D, et al. (2000). Selective blockade of metabotropic glutamate receptor subtype 5 is neuroprotective. Neuropharmacology, 39(12), 2223–2230. 10.1016/S0028-3908(00)00079-4. [DOI] [PubMed] [Google Scholar]
  54. Budgett RF, Bakker G, Sergeev E, Bennett KA, & Bradley SJ (2022). Targeting the type 5 metabotropic glutamate receptor: A potential therapeutic strategy for neurodegenerative diseases? Frontiers in Pharmacology, 13, 1–20. 10.3389/fphar.2022.893422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Burbaeva GS, Boksha IS, Turishcheva MS, Vorobyeva EA, Savushkina OK, & Tereshkina EB (2003). Glutamine synthetase and glutamate dehydrogenase in the prefrontal cortex of patients with schizophrenia. Progress in Neuro-Psychopharmacology & Biological Psychiatry, 27(4), 675–680. 10.1016/s0278-5846(03)00078-2. [DOI] [PubMed] [Google Scholar]
  56. Bushey D, Tononi G, & Cirelli C (2011). Sleep and synaptic homeostasis: Structural evidence in Drosophila. Science, 332(6037), 1576–1581. 10.1126/science.1202839. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Calabrese V, Picconi B, Heck N, Campanelli F, Natale G, Marino G, et al. (2022). A positive allosteric modulator of mGlu4 receptors restores striatal plasticity in an animal model of l-Dopa-induced dyskinesia. Neuropharmacology, 218, 109205. 10.1016/j.neuropharm.2022.109205. [DOI] [PubMed] [Google Scholar]
  58. Canevelli M, Valletta M, Trebbastoni A, Sarli G, D’Antonio F, Tariciotti L, et al. (2016). Sundowning in dementia: Clinical relevance, pathophysiological determinants, and therapeutic approaches. Frontiers in Medicine, 3, 73. 10.3389/fmed.2016.00073. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Caraci F, Molinaro G, Battaglia G, Giuffrida ML, Riozzi B, Traficante A, et al. (2011). Targeting group II metabotropic glutamate (mGlu) receptors for the treatment of psychosis associated with Alzheimer’s disease: Selective activation of mGlu2 receptors amplifies beta-amyloid toxicity in cultured neurons, whereas dual activation of mGlu2 and mGlu3 receptors is neuroprotective. Molecular Pharmacology, 79(3), 618–626. 10.1124/mol.110.067488. [DOI] [PubMed] [Google Scholar]
  60. Caraci F, Nicoletti F, & Copani A (2018). Metabotropic glutamate receptors: The potential for therapeutic applications in Alzheimer’s disease. Current Opinion in Pharmacology, 38, 1–7. 10.1016/j.coph.2017.12.001. [DOI] [PubMed] [Google Scholar]
  61. Cavas M, Scesa G, Martín-López M, & Navarro JF (2017). Selective agonism of mGlu8 receptors by (S)-3,4-dicarboxyphenylglycine does not affect sleep stages in the rat. Pharmacological Reports, 69(1), 97–104. 10.1016/j.pharep.2016.09.019. [DOI] [PubMed] [Google Scholar]
  62. Cavas M, Scesa G, & Navarro JF (2013a). Positive allosteric modulation of mGlu7 receptors by AMN082 affects sleep and wakefulness in the rat. Pharmacology Biochemistry and Behavior, 103(4), 756–763. 10.1016/j.pbb.2012.12.008. [DOI] [PubMed] [Google Scholar]
  63. Cavas M, Scesa G, & Navarro JF (2013b). Effects of MPEP, a selective metabotropic glutamate mGlu5 ligand, on sleep and wakefulness in the rat. Progress in Neuro-Psychopharmacology and Biological Psychiatry, 40, 18–25. 10.1016/J.PNPBP.2012.09.011. [DOI] [PubMed] [Google Scholar]
  64. Chaki S (2017). mGlu2/3 receptor antagonists as novel antidepressants. Trends in Pharmacological Sciences, 38(6), 569–580. 10.1016/j.tips.2017.03.008. [DOI] [PubMed] [Google Scholar]
  65. Chaki S, Koike H, & Fukumoto K (2019). Targeting of metabotropic glutamate receptors for the development of novel antidepressants. Chronic Stress, 3. 10.1177/2470547019837712. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Chaki S, Yoshikawa R, Hirota S, Shimazaki T, Maeda M, Kawashima N, et al. (2004). MGS0039: A potent and selective group II metabotropic glutamate receptor antagonist with antidepressant-like activity. Neuropharmacology, 46(4), 457–467. 10.1016/j.neuropharm.2003.10.009. [DOI] [PubMed] [Google Scholar]
  67. Charvin D (2018). mGlu4 allosteric modulation for treating Parkinson’s disease. Neuropharmacology, 135, 308–315. 10.1016/j.neuropharm.2018.03.027. [DOI] [PubMed] [Google Scholar]
  68. Chen MH, Korenic SA, Wickwire EM, Wijtenburg SA, Hong LE, & Rowland LM (2020). Sex differences in subjective sleep quality patterns in Schizophrenia. Behavioral Sleep Medicine, 18(5), 668–679. 10.1080/15402002.2019.1660168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Chen VCH, Ting H, Wu MH, Lin TY, & Gossop M (2017). Sleep disturbance and its associations with severity of dependence, depression and quality of life among heroin-dependent patients: A cross-sectional descriptive study. Substance Abuse: Treatment, Prevention, and Policy, 12(1), 1–9. 10.1186/s13011-017-0101-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Chen B, Wang Y, Liu X, Liu Z, Dong Y, & Huang YH (2015). Sleep regulates incubation of cocaine craving. J Neurosci, 35(39), 13300–13310. 10.1523/jneurosci.1065-15.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Chiu VW, Ree M, Janca A, & Waters F (2016). Sleep in Schizophrenia: Exploring subjective experiences of sleep problems, and implications for treatment. Psychiatric Quarterly, 87(4), 633–648. 10.1007/s11126-015-9415-x. [DOI] [PubMed] [Google Scholar]
  72. Chouinard S, Poulin J, Stip E, & Godbout R (2004). Sleep in untreated patients with schizophrenia: A meta-analysis. Schizophrenia Bulletin, 30(4), 957–967. 10.1093/oxfordjournals.schbul.a007145. [DOI] [PubMed] [Google Scholar]
  73. Cid JM, Tresadern G, Vega JA, de Lucas AI, Matesanz E, Iturrino L, et al. (2012). Discovery of 3-cyclopropylmethyl-7-(4-phenylpiperidin-1-yl)-8-trifluoromethyl[1,2,4] triazolo[4,3-a]pyridine (JNJ-42153605): A positive allosteric modulator of the metabotropic glutamate 2 receptor. Journal of Medicinal Chemistry, 55(20), 8770–8789. 10.1021/jm3010724. [DOI] [PubMed] [Google Scholar]
  74. Cleva RM, & Olive MF (2012). mGlu receptors and drug addiction. Wiley Interdisciplinary Reviews. Membrane Transport and Signaling, 1(3), 281–295. 10.1002/wmts.18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Coble P, Foster FG, & Kupfer DJ (1976). Electroencephalographic sleep diagnosis of primary depression. Archives of General Psychiatry, 33(9), 1124–1127. 10.1001/archpsyc.1976.01770090114012. [DOI] [PubMed] [Google Scholar]
  76. Coffey AA, Guan Z, Grigson PS, & Fang J (2016). Reversal of the sleep–wake cycle by heroin self-administration in rats. Brain Research Bulletin, 123, 33–46. 10.1016/j.brainresbull.2015.09.008. [DOI] [PubMed] [Google Scholar]
  77. Cohrs S (2008). Sleep disturbances in patients with schizophrenia: Impact and effect of antipsychotics. CNS Drugs. 10.2165/00023210-200822110-00004. [DOI] [PubMed] [Google Scholar]
  78. Cominski TP, Jiao X, Catuzzi JE, Stewart AL, & Pang KCH (2014). The role of the hippocampus in avoidance learning and anxiety vulnerability. Frontiers in Behavioral Neuroscience, 8, 1–10. 10.3389/fnbeh.2014.00273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Conn PJ, & Jones CK (2009). Promise of mGluR2/3 activators in psychiatry. Neuropsychopharmacology, 34(1), 248–249. 10.1038/npp.2008.156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Conn PJ, & Pin JP (1997). Pharmacology and functions of metabotropic glutamate receptors. Annual Review of Pharmacology and Toxicology, 37, 205–237. 10.1146/annurev.pharmtox.37.1.205. [DOI] [PubMed] [Google Scholar]
  81. Cortes JA, Gomez G, Ehnerd C, Gurnsey K, Nicolazzo J, Bradberry CW, et al. (2016). Altered activity-based sleep measures in rhesus monkeys following cocaine self-administration and abstinence. Drug and Alcohol Dependence, 163, 202–208. 10.1016/j.drugalcdep.2016.04.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Coyle JT (2006). Glutamate and schizophrenia: Beyond the dopamine hypothesis. Cellular and Molecular Neurobiology, 26(4–6), 365–384. 10.1007/s10571-006-9062-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Crabtree JW, Lodge D, Bashir ZI, & Isaac JTR (2013). GABAA, NMDA and mGlu2 receptors tonically regulate inhibition and excitation in the thalamic reticular nucleus. European Journal of Neuroscience, 37(6), 850–859. 10.1111/ejn.12098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Crupi R, Impellizzeri D, & Cuzzocrea S (2019). Role of metabotropic glutamate receptors in neurological disorders. Frontiers in Molecular Neuroscience, 12, 1–11. 10.3389/fnmol.2019.00020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Czigler B, Csikós D, Hidasi Z, Anna Gaál Z, Csibri É, Kiss É, et al. (2008). Quantitative EEG in early Alzheimer’s disease patients–Power spectrum and complexity features. International Journal of Psychophysiology, 68(1), 75–80. 10.1016/j.ijpsycho.2007.11.002. [DOI] [PubMed] [Google Scholar]
  86. Daan S, Beersma DG, & Borbély AA (1984). Timing of human sleep: Recovery process gated by a circadian pacemaker. The American Journal of Physiology, 246(2 Pt. 2), R161–R183. 10.1152/ajpregu.1984.246.2.R161. [DOI] [PubMed] [Google Scholar]
  87. Dash MB, Douglas CL, Vyazovskiy VV, Cirelli C, & Tononi G (2009). Long-term homeostasis of extracellular glutamate in the rat cerebral cortex across sleep and waking states. Journal of Neuroscience, 29(3), 620–629. 10.1523/JNEUROSCI.5486-08.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Davidson S,Golden J,Copits B,Pradipta R, Vogt S,Valtcheva M, et al. (2017). Group II mGluRs suppress hyperexcitability in mouse and human nociceptors. Physiology & Behavior, 176(5), 139–148. 10.1097/j.pain.0000000000000621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. Davies BM, & Beech HR (1960). The effect of 1-arylcylohexylamine (sernyl) on twelve normal volunteers. The Journal of Mental Science, 106, 912–924. 10.1192/bjp.106.444.912. [DOI] [PubMed] [Google Scholar]
  90. Davis MT, Hillmer A, Holmes SE, Pietrzak RH, DellaGioia N, Nabulsi N, et al. (2019). In vivo evidence for dysregulation of mGluR5 as a biomarker of suicidal ideation. Proceedings of the National Academy of Sciences of the United States of America, 166(23), 11490–11495. 10.1073/pnas.1818871116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. de Filippis B, Lyon L, Taylor A, Lane T, Burnet PWJ, Harrison PJ, et al. (2015). The role of group II metabotropic glutamate receptors in cognition and anxiety: Comparative studies in GRM2−/−, GRM3−/− and GRM2/3−/− knockout mice. Neuropharmacology, 89, 19–32. 10.1016/j.neuropharm.2014.08.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. de Vivo L, Bellesi M, Marshall W, Bushong EA, Ellisman MH, Tononi G, et al. (2017). Ultrastructural evidence for synaptic scaling across the wake/sleep cycle. Science (New York, N.Y.), 355(6324), 507–510. 10.1126/science.aah5982. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Denis D, Bottary R, Cunningham TJ, Zeng S, Daffre C, Oliver KL, et al. (2021). Sleep power spectral density and spindles in PTSD and their relationship to symptom severity. Frontiers in Psychiatry, 12(November), 1–14. 10.3389/fpsyt.2021.766647. [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Di Chiara G, & Imperato A (1988). Drugs abused by humans preferentially increase synaptic dopamine concentrations in the mesolimbic system of freely moving rats. Proceedings of the National Academy of Sciences of the United States of America, 85(14), 5274–5278. 10.1073/pnas.85.14.5274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Diering GH, Nirujogi RS, Roth RH, Worley PF, Pandey A, & Huganir RL (2017). Homer1a drives homeostatic scaling-down of excitatory synapses during sleep. Science, 515(February), 511–515. 10.1126/science.aai8355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Dimsdale JE, Norman D, DeJardin D, & Wallace MS (2007). The effect of opioids on sleep architecture. Journal of Clinical Sleep Medicine, 3(1), 33–36. [PubMed] [Google Scholar]
  97. Dogra S, & Conn PJ (2021). Targeting metabotropic glutamate receptors for the treatment of depression and other stress-related disorders. Neuropharmacology, 196, 108687. 10.1016/j.neuropharm.2021.108687. [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Dogra S, & Conn PJ (2022). Metabotropic glutamate receptors as emerging targets for the treatment of Schizophrenia. Molecular Pharmacology, 101(5), 275–285. 10.1124/molpharm.121.000460. [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Dolsen MR, & Harvey AG (2017). Life-time history of insomnia and hypersomnia symptoms as correlates of alcohol, cocaine and heroin use and relapse among adults seeking substance use treatment in the United States from 1991 to 1994. Addiction, 112(6), 1104–1111. 10.1111/add.13772. [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Dule A, Ahmed G, Tessema W, & Soboka M (2020). Sleep quality in Schizophrenia. Journal of Mental Health & Clinical Psychology, 4(4), 57–64. 10.29245/2578-2959/2020/4.1223. [DOI] [Google Scholar]
  101. Dunn KE, Huhn AS, Bergeria CL, Gipson CD, & Weerts EM (2019). Non-opioid neurotransmitter systems that contribute to the opioid withdrawal syndrome: A review of preclinical and human evidence. Journal of Pharmacology and Experimental Therapeutics, 371(2), 422–452. 10.1124/jpet.119.258004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Duvoisin RM, Pfankuch T, Wilson JM, Grabell J, Chhajlani V, Brown DG, et al. (2010). Acute pharmacological modulation of mGluR8 reduces measures of anxiety. Behavioural Brain Research, 212(2), 130–134. 10.1037/12218-043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Eacret D, Veasey SC, & Blendy JA (2020). Bidirectional relationship between opioids and disrupted sleep: Putative mechanisms. Molecular Pharmacology, 98(4), 445–453. 10.1124/mol.119.119107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Eckert DJ, & Yaggi HK (2022). Opioid use disorder, sleep deficiency, and ventilatory control: Bidirectional mechanisms and therapeutic targets. American Journal of Respiratory and Critical Care Medicine, 206, 937–949. 10.1164/rccm.202108-2014ci. [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. España RA, & Scammell TE (2011). Sleep neurobiology from a clinical perspective. Sleep, 34(7), 845–858. 10.5665/SLEEP.1112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Esterlis I, DellaGioia N, Pietrzak RH, Matuskey D, Nabulsi N, Abdallah CG, et al. (2018). Ketamine-induced reduction in mGluR5 availability is associated with an antidepressant response: An [11C]ABP688 and PET imaging study in depression. Molecular Psychiatry, 23(4), 824–832. 10.1038/mp.2017.58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Esterlis I, Holmes SE, Sharma P, Krystal JH, & Delorenzo C (2018). mGluR5 and stress disorders: Knowledge gained from receptor imaging studies. Biological Psychiatry, 84(2), 95–105. 10.1016/j.biopsych.2017.08.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. Fang H, Tu S, Sheng J, & Shao A (2019). Depression in sleep disturbance: A review on a bidirectional relationship, mechanisms and treatment. Journal of Cellular and Molecular Medicine, 23(4), 2324–2332. 10.1111/jcmm.14170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Feder A, Parides MK, Murrough JW, Perez AM, Morgan JE, Saxena S, et al. (2014). Efficacy of intravenous ketamine for treatment of chronic posttraumatic stress disorder: A randomized clinical trial. JAMA Psychiatry, 71(6), 681–688. 10.1001/jamapsychiatry.2014.62. [DOI] [PubMed] [Google Scholar]
  110. Feinberg I, Campbell IG, Schoepp DD, & Anderson K (2002). The selective group mGlu2/3 receptor agonist LY379268 suppresses REM sleep and fast EEG in the rat. Pharmacology Biochemistry and Behavior, 73(2), 467–474. 10.1016/S0091-3057(02)00843-2. [DOI] [PubMed] [Google Scholar]
  111. Feinberg I, Schoepp DD, Hsieh KC, Darchia N, & Campbell IG (2005). The metabotropic glutamate (mGLU)2/3 receptor antagonist LY341495 [2S-2-amino-2-(1S,2S-2-carboxycyclopropyl-1-yl)-3-(xanth-9-yl)propanoic acid] stimulates waking and fast electroencephalogram power and blocks the effects of the mGLU2/3 receptor agonist LY. Journal of Pharmacology and Experimental Therapeutics, 312(2), 826–833. 10.1124/jpet.104.076547. [DOI] [PubMed] [Google Scholar]
  112. Fell MJ, Witkin JM, Falcone JF, Katner JS, Perry KW, Hart J, et al. (2011). N-(4-((2-(trifluoromethyl)-3-hydroxy-4-(isobutyryl)phenoxy)methyl) benzyl)-1-methyl-1h-imidazole-4-carboxamide (THIIC), a novel metabotropic glutamate 2 potentiator with potential anxiolytic/antidepressant properties: In vivo profiling suggests a link bet. Journal of Pharmacology and Experimental Therapeutics, 336(1), 165–177. 10.1124/jpet.110.172957. [DOI] [PubMed] [Google Scholar]
  113. Felts AS, Rodriguez AL, Blobaum AL, Morrison RD, Bates BS, Thompson Gray A, et al. (2017). Discovery of N-(5-fluoropyridin-2-yl)-6-methyl-4-(pyrimidin-5-yloxy)picolinamide (VU0424238): A novel negative allosteric modulator of metabotropic glutamate receptor subtype 5 selected for clinical evaluation. Journal of Medicinal Chemistry, 60(12), 5072–5085. 10.1021/acs.jmedchem.7b00410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Fendt M, Imobersteg S, Peterlik D, Chaperon F, Mattes C, Wittmann C, et al. (2013). Differential roles of mGlu7 and mGlu8 in amygdala-dependent behavior and physiology. Neuropharmacology, 72, 215–223. 10.1016/j.neuropharm.2013.04.052. [DOI] [PubMed] [Google Scholar]
  115. Feng YZ, Zhang T, Rockhold RW, & Ho IK (1995). Increased locus coeruleus glutamate levels are associated with naloxone-precipitated withdrawal from butorphanol in the rat. Neurochemical Research, 20(6), 745–751. 10.1007/BF01705544. [DOI] [PubMed] [Google Scholar]
  116. Ferrarelli F, & Tononi G (2017). Reduced sleep spindle activity point to a TRN-MD thalamus-PFC circuit dysfunction in schizophrenia. Schizophrenia Research, 180, 36–43. 10.1016/j.schres.2016.05.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  117. Feyissa AM, Woolverton WL, Miguel-Hidalgo JJ, Wang Z, Kyle PB, Hasler G, et al. (2010). Elevated level of metabotropic glutamate receptor 2/3 in the prefrontal cortex in major depression. Progress in Neuro-Psychopharmacology and Biological Psychiatry, 34(2), 279–283. 10.1016/j.pnpbp.2009.11.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Fisher NM, Gould RW, Gogliotti RG, McDonald AJ, Badivuku H, Chennareddy S, et al. (2020). Phenotypic profiling of mGlu(7) knockout mice reveals new implications for neurodevelopmental disorders. Genes, Brain, and Behavior, 19(7), e12654. 10.1111/gbb.12654. [DOI] [PMC free article] [PubMed] [Google Scholar]
  119. Ford DE, & Kamerow DB (1989). Epidemiologic study of sleep disturbances and psychiatric disorders. An opportunity for prevention? JAMA, 262(11), 1479–1484. 10.1001/jama.262.11.1479. [DOI] [PubMed] [Google Scholar]
  120. Foster FG, Kupfer DJ, Coble P, & McPartland RJ (1976). Rapid eye movement sleep density. An objective indicator in severe medical-depressive syndromes. Archives of General Psychiatry, 33(9), 1119–1123. 10.1001/archpsyc.1976.01770090109011. [DOI] [PubMed] [Google Scholar]
  121. Frye MA, Tsai GE, Huggins T, Coyle JT, & Post RM (2007). Low cerebrospinal fluid glutamate and glycine in refractory affective disorder. Biological Psychiatry, 61(2), 162–166. 10.1016/j.biopsych.2006.01.024. [DOI] [PubMed] [Google Scholar]
  122. Fukumoto K, Iijima M, & Chaki S (2016). The antidepressant effects of an mGlu2/3 receptor antagonist and ketamine require AMPA receptor stimulation in the mPFC and subsequent activation of the 5-HT neurons in the DRN. Neuropsychopharmacology, 41(4), 1046–1056. 10.1038/npp.2015.233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  123. Galici R, Jones CK, Hemstapat K, Nong Y, Echemendia NG, Williams LC, et al. (2006). Biphenyl-indanone A, a positive allosteric modulator of the metabotropic glutamate receptor subtype 2, has antipsychotic- and anxiolytic-like effects in mice. Journal of Pharmacology and Experimental Therapeutics, 318(1), 173–185. 10.1124/jpet.106.102046. [DOI] [PubMed] [Google Scholar]
  124. Gauthier EA, Guzick SE, Brummett CM, Baghdoyan HA, & Lydic R (2011). Buprenorphine disrupts sleep and decreases adenosine concentrations in sleep-regulating brain regions of sprague dawley rat. Anesthesiology, 115(4), 743–753. 10.1097/ALN.0b013e31822e9f85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. George SA, Rodriguez-Santiago M, Riley J, Abelson JL, Floresco SB, & Liberzon I (2018). D-Cycloserine facilitates reversal in an animal model of post-traumatic stress disorder. Behavioural Brain Research, 347, 332–338. 10.1016/j.bbr.2018.03.037. [DOI] [PubMed] [Google Scholar]
  126. Gerstenberg M, Furrer M, Tesler N, Franscini M, Walitza S, & Huber R (2020). Reduced sleep spindle density in adolescent patients with early-onset schizophrenia compared to major depressive disorder and healthy controls. Schizophrenia Research, 221, 20–28. 10.1016/j.schres.2019.11.060. [DOI] [PubMed] [Google Scholar]
  127. Gilestro GF, Tononi G, & Cirelli C (2009). Widespread changes in synaptic markers as a function of sleep and wakefulness in drosophila. Science, 324(5923), 109–112. 10.1126/science.1166673. [DOI] [PMC free article] [PubMed] [Google Scholar]
  128. Gilmour G, Broad LM, Wafford KA, Britton T, Colvin EM, Fivush A, et al. (2013). In vitro characterisation of the novel positive allosteric modulators of the mGlu5 receptor, LSN2463359 and LSN2814617, and their effects on sleep architecture and operant responding in the rat. Neuropharmacology, 64, 224–239. 10.1016/j.neuropharm.2012.07.030. [DOI] [PubMed] [Google Scholar]
  129. Girard B, Tuduri P, Moreno MP, Sakkaki S, Barboux C, Bouschet T, et al. (2019). The mGlu7 receptor provides protective effects against epileptogenesis and epileptic seizures. Neurobiology of Disease, 129, 13–28. 10.1016/j.nbd.2019.04.016. [DOI] [PubMed] [Google Scholar]
  130. Glantz LA, & Lewis DA (2000). Decreased dendritic spine density on prefrontal cortical pyramidal neurons in schizophrenia. Archives of General Psychiatry, 57(1), 65–73. 10.1001/archpsyc.57.1.65. [DOI] [PubMed] [Google Scholar]
  131. Gluck MR, Thomas RG, Davis KL, & Haroutunian V (2002). Implications for altered glutamate and GABA metabolism in the dorsolateral prefrontal cortex of aged schizophrenic patients. The American Journal of Psychiatry, 159(7), 1165–1173. 10.1176/appi.ajp.159.7.1165. [DOI] [PubMed] [Google Scholar]
  132. Göder R, Aldenhoff JB, Boigs M, Braun S, Koch J, & Fritzer G (2006). Delta power in sleep in relation to neuropsychological performance in healthy subjects and schizophrenia patients. The Journal of Neuropsychiatry and Clinical Neurosciences, 18(4), 529–535. 10.1176/jnp.2006.18.4.529. [DOI] [PubMed] [Google Scholar]
  133. Göder R, Boigs M, Braun S, Friege L, Fritzer G, Aldenhoff JB, et al. (2004). Impairment of visuospatial memory is associated with decreased slow wave sleep in schizophrenia. Journal of Psychiatric Research, 38(6), 591–599. 10.1016/j.jpsychires.2004.04.005. [DOI] [PubMed] [Google Scholar]
  134. Göder R, Graf A, Ballhausen F, Weinhold S, Baier PC, Junghanns K, et al. (2015). Impairment of sleep-related memory consolidation in schizophrenia: Relevance of sleep spindles? Sleep Medicine, 16(5), 564–569. 10.1016/j.sleep.2014.12.022. [DOI] [PubMed] [Google Scholar]
  135. Goldstein RZ, & Volkow ND (2002). Drug addiction and its underlying neurobiological basis: Neuroimaging evidence for the involvement of the frontal cortex. Am J Psychiatry, 159(10), 1642–1652. 10.1176/appi.ajp.159.10.1642. [DOI] [PMC free article] [PubMed] [Google Scholar]
  136. Gould RW, Amato RJ, Bubser M, Joffe ME, Nedelcovych MT, Thompson AD, et al. (2016). Partial mGlu 5 negative allosteric modulators attenuate cocaine-mediated behaviors and lack psychotomimetic-like effects. Neuropsychopharmacology, 41(4), 1166–1178. 10.1038/npp.2015.265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  137. Gould RW, Felts AS, & Jones CK (2017). Negative allosteric modulators of the metabotropic glutamate receptor subtype 5 for the treatment of cocaine use disorder. In The neuroscience of cocaine: Mechanisms and treatment. 10.1016/B978-0-12-803750-8.00071-3. [DOI] [Google Scholar]
  138. Gould RW, Porrino LJ, & Nader MA (2012). Nonhuman primate models of addiction and PET imaging: Dopamine system dysregulation. Current Topics in Behavioral Neurosciences, 11, 25–44. 10.1007/7854_2011_168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  139. Greco B, Invernizzi RW, & Carli M (2005). Phencyclidine-induced impairment in attention and response control depends on the background genotype of mice: Reversal by the mGLU2/3 receptor agonist LY379268. Psychopharmacology, 179(1), 68–76. 10.1007/s00213-004-2127-9. [DOI] [PubMed] [Google Scholar]
  140. Griebel G, Pichat P, Boulay D, Naimoli V, Potestio L, Featherstone R, et al. (2016). The mGluR2 positive allosteric modulator, SAR218645, improves memory and attention deficits in translational models of cognitive symptoms associated with schizophrenia. Scientific Reports, 6, 1–23. 10.1038/srep35320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  141. Grueter BA, & Winder DG (2005). Group II and III metabotropic glutamate receptors suppress excitatory synaptic transmission in the dorsolateral bed nucleus of the stria terminalis. Neuropsychopharmacology, 30(7), 1302–1311. 10.1038/sj.npp.1300672. [DOI] [PubMed] [Google Scholar]
  142. Gupta R, Ali R, & Ray R (2018). Willis-Ekbom disease/restless legs syndrome in patients with opioid withdrawal. Sleep Medicine, 45, 39–43. 10.1016/j.sleep.2017.09.028. [DOI] [PubMed] [Google Scholar]
  143. Hackler EA, Byun NE, Jones CK, Williams JM, Baheza R, Sengupta S, et al. (2010). Selective potentiation of the metabotropic glutamate receptor subtype 2 blocks phencyclidine-induced hyperlocomotion and brain activation. Neuroscience, 168(1), 209–218. 10.1016/j.neuroscience.2010.02.057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  144. Hajasova Z, Canestrelli C, Acher F, Noble F, & Marie N (2018). Role of mGlu7 receptor in morphine rewarding effects is uncovered by a novel orthosteric agonist. Neuropharmacology, 131, 424–430. 10.1016/j.neuropharm.2018.01.002. [DOI] [PubMed] [Google Scholar]
  145. Hamilton A, Esseltine JL, Devries RA, Cregan SP, & Ferguson SSG (2014). Metabotropic glutamate receptor 5 knockout reduces cognitive impairment and pathogenesis in a mouse model of Alzheimer’s disease. Molecular Brain, 7(1), 1–12. 10.1186/1756-6606-7-40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  146. Hamilton A, Vasefi M, vander Tuin C, McQuaid RJ, Anisman H, & Ferguson SSG (2016). Chronic pharmacological mGluR5 inhibition prevents cognitive impairment and reduces pathogenesis in an Alzheimer disease mouse model. Cell Reports, 15(9), 1859–1865. 10.1016/j.celrep.2016.04.077. [DOI] [PubMed] [Google Scholar]
  147. Hansen KB, Wollmuth LP, Bowie D, Furukawa H, Menniti FS, Sobolevsky AI, et al. (2021). Structure, function, and pharmacology of glutamate receptor ion channels. Pharmacological Reviews, 73(4), 298–487. 10.1124/pharmrev.120.000131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  148. Hao Y, Martin-Fardon R, & Weiss F (2010). Behavioral and functional evidence of metabotropic glutamate receptor 2/3 and metabotropic glutamate receptor 5 dysregulation in cocaine-escalated rats: Factor in the transition to dependence. Biological Psychiatry, 68(3), 240–248. 10.1016/j.biopsych.2010.02.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  149. Hartwell EE, Pfeifer JG, McCauley JL, Moran-Santa Maria M, & Back SE (2014). Sleep disturbances and pain among individuals with prescription opioid dependence. Addictive Behaviors, 39(10), 1537–1542. 10.1016/j.addbeh.2014.05.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  150. Harvey BD, Siok CJ, Kiss T, Volfson D, Grimwood S, Shaffer CL, et al. (2013). Neurophysiological signals as potential translatable biomarkers for modulation of metabotropic glutamate 5 receptors. Neuropharmacology, 75, 19–30. 10.1016/j.neuropharm.2013.06.020. [DOI] [PubMed] [Google Scholar]
  151. Hashimoto K, Sawa A, & Iyo M (2007). Increased levels of glutamate in brains from patients with mood disorders. Biological Psychiatry, 62(11), 1310–1316. 10.1016/j.biopsych.2007.03.017. [DOI] [PubMed] [Google Scholar]
  152. Hasler BP, & Pedersen SL (2020). Sleep and circadian risk factors for alcohol problems: A brief overview and proposed mechanisms. Current Opinion in Psychology, 34, 57–62. 10.1016/j.copsyc.2019.09.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  153. Haynes J, Talbert M, Fox S, & Close E (2018). Cognitive behavioral therapy in the treatment of insomnia. Southern Medical Journal, 111(2), 75–80. 10.14423/SMJ.0000000000000769. [DOI] [PubMed] [Google Scholar]
  154. Hefti K, Holst SC, Sovago J, Bachmann V, Buck A, Ametamey SM, et al. (2013). Increased metabotropic glutamate receptor subtype 5 availability in human brain after one night without sleep. Biological Psychiatry, 73(2), 161–168. 10.1016/j.biopsych.2012.07.030. [DOI] [PubMed] [Google Scholar]
  155. Hiyoshi T, Hikichi H, Karasawa JI, & Chaki S (2014). Metabotropic glutamate receptors regulate cortical gamma hyperactivities elicited by ketamine in rats. Neuroscience Letters, 567, 30–34. 10.1016/j.neulet.2014.03.025. [DOI] [PubMed] [Google Scholar]
  156. Hodges SE, Pittman B, & Morgan PT (2017). Sleep perception and misperception in chronic cocaine users during abstinence. Sleep, 40(3). 10.1093/sleep/zsw069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  157. Holmes SE, Girgenti MJ, Davis MT, Pietrzak RH, Dellagioia N, Nabulsi N, et al. (2017). Altered metabotropic glutamate receptor 5 markers in PTSD: In vivo and postmortem evidence. Proceedings of the National Academy of Sciences of the United States of America, 114(31), 8390–8395. 10.1073/pnas.1701749114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  158. Holst SC, Sousek A, Hefti K, Saberi-Moghadam S, Buck A, Ametamey SM, et al. (2017). Cerebral mGluR5 availability contributes to elevated sleep need and behavioral adjustment after sleep deprivation. ELife, 6, 1–23. 10.7554/eLife.28751.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  159. Holter KM, Lekander AD, LaValley CM, Bedingham EG, Pierce BE, Sands LP, et al. (2021). Partial mGlu5 negative allosteric modulator M-5MPEP demonstrates antidepressant-like effects on sleep without affecting cognition or quantitative EEG. Frontiers in Neuroscience, 15, 700822. 10.3389/fnins.2021.700822. [DOI] [PMC free article] [PubMed] [Google Scholar]
  160. Homayoun H, Stefani MR, Adams BW, Tamagan GD, & Moghaddam B (2004). Functional interaction between NMDA and mGlu5 receptors: Effects on working memory, instrumental learning, motor behaviors, and dopamine release. Neuropsychopharmacology, 29(7), 1259–1269. 10.1038/sj.npp.1300417. [DOI] [PubMed] [Google Scholar]
  161. Hong J, Lee J, Song K, Ha GE, Yang YR, Ma JS, et al. (2016). The thalamic mGluR1-PLCβ4 pathway is critical in sleep architecture. Molecular Brain, 9(1), 1–12. 10.1186/s13041-016-0276-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  162. Hopkins CR, Lindsley CW, & Niswender CM (2009). mGluR4-positive allosteric modulation as potential treatment for Parkinson’s disease. Future Medicinal Chemistry, 1(3), 501–513. 10.4155/fmc.09.38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  163. Hor H, & Tafti M (2009). How much sleep do we need? Science, 325(5942), 825–826. 10.1126/science.1178713. [DOI] [PubMed] [Google Scholar]
  164. Hser Y-I, Mooney LJ, Huang D, Zhu Y, Tomko RL, McClure E, et al. (2017). Reductions in cannabis use are associated with improvements in anxiety, depression, and sleep quality, but not quality of life. Journal of Substance Abuse Treatment, 81, 53–58. 10.1016/j.jsat.2017.07.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  165. Hsu WY, Chiu NY, Liu JT, Wang CH, Chang TG, Liao YC, et al. (2012). Sleep quality in heroin addicts under methadone maintenance treatment. Acta Neuropsychiatrica, 24(6), 356–360. 10.1111/j.1601-5215.2011.00628.x. [DOI] [PubMed] [Google Scholar]
  166. Hu W, MacDonald ML, Elswick DE, & Sweet RA (2015). The glutamate hypothesis of schizophrenia: Evidence from human brain tissue studies. Annals of the New York Academy of Sciences, 1338, 38–57. 10.1111/nyas.12547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  167. Hu B, Steriade M, & Deschênes M (1989). The cellular mechanism of thalamic ponto-geniculo-occipital waves. Neuroscience, 31(1), 25–35. 10.1016/0306-4522(89)90028-6. [DOI] [PubMed] [Google Scholar]
  168. Hughes ZA, Neal SJ, Smith DL, Sukoff Rizzo SJ, Pulicicchio CM, Lotarski S, et al. (2013). Negative allosteric modulation of metabotropic glutamate receptor 5 results in broad spectrum activity relevant to treatment resistant depression. Neuropharmacology, 66, 202–214. 10.1016/j.neuropharm.2012.04.007. [DOI] [PubMed] [Google Scholar]
  169. Huguenard JR, & McCormick DA (2007). Thalamic synchrony and dynamic regulation of global forebrain oscillations. Trends in Neurosciences, 30(7), 350–356. 10.1016/j.tins.2007.05.007. [DOI] [PubMed] [Google Scholar]
  170. Iber C, Ancoli-Israel S, & Chesson A (2007). The AASM manual for the scoring of sleep and associated events: Rules, terminology, and technical specification (1st ed.). American Academy of Sleep Medicine. [Google Scholar]
  171. Ikeda M, Hirono M, Sugiyama T, Moriya T, Ikeda-Sagara M, Eguchi N, et al. (2009). Phospholipase C-β4 is essential for the progression of the normal sleep sequence and ultradian body temperature rhythms in mice. PLoS One, 4(11). 10.1371/journal.pone.0007737. [DOI] [PMC free article] [PubMed] [Google Scholar]
  172. Irwin MR, & Vitiello MV (2019). Implications of sleep disturbance and inflammation for Alzheimer’s disease dementia. The Lancet. Neurology, 18(3), 296–306. 10.1016/S1474-4422(18)30450-2. [DOI] [PubMed] [Google Scholar]
  173. Iscru E, Goddyn H, Ahmed T, Callaerts-Vegh Z, D’Hooge R, & Balschun D (2013). Improved spatial learning is associated with increased hippocampal but not prefrontal long-term potentiation in mGluR4 knockout mice. Genes, Brain, and Behavior, 12(6), 615–625. 10.1111/gbb.12052. [DOI] [PubMed] [Google Scholar]
  174. Jack CR,Bennett DA,Blennow K,Carrillo MC, Feldman HH,Frisoni GB,et al. (2016). A/T/N: An unbiased descriptive classification scheme for Alzheimer disease biomarkers. Neurology, 87(5), 539–547. 10.1212/WNL.0000000000002923. [DOI] [PMC free article] [PubMed] [Google Scholar]
  175. Jack CR, Knopman DS, Jagust WJ, Shaw LM, Aisen PS, Weiner MW, et al. (2010). Hypothetical model of dynamic biomarkers of the Alzheimer’s pathological cascade. The Lancet. Neurology, 9(1), 119–128. 10.1016/S1474-4422(09)70299-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  176. Jacob W, Gravius A, Pietraszek M, Nagel J, Belozertseva I, Shekunova E, et al. (2009). The anxiolytic and analgesic properties of fenobam, a potent mGlu5 receptor antagonist, in relation to the impairment of learning. Neuropharmacology, 57(2), 97–108. 10.1016/J.NEUROPHARM.2009.04.011. [DOI] [PubMed] [Google Scholar]
  177. Jaehne A, Loessl B, Bárkai Z, Riemann D, & Hornyak M (2009). Effects of nicotine on sleep during consumption, withdrawal and replacement therapy. Sleep Medicine Reviews, 13(5), 363–377. 10.1016/j.smrv.2008.12.003. [DOI] [PubMed] [Google Scholar]
  178. Javitt DC (2007). Glutamate and Schizophrenia: Phencyclidine, N-Methyl-d-aspartate receptors, and dopamine-glutamate interactions. International Review of Neurobiology, 78, 69–108. 10.1016/S0074-7742(06)78003-5. [DOI] [PubMed] [Google Scholar]
  179. Joffe ME, Centanni SW, Jaramillo AA, Winder DG, & Conn PJ (2018). Metabotropic glutamate receptors in alcohol use disorder: Physiology, plasticity, and promising pharmacotherapies. ACS Chemical Neuroscience, 9(9), 2188–2204. 10.1021/acschemneuro.8b00200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  180. Joffe ME, Santiago CI, Oliver KH, Maksymetz J, Harris NA, Engers JL, et al. (2020). mGlu2 and mGlu3 negative allosteric modulators divergently enhance thalamocortical transmission and exert rapid antidepressant-like effects. Neuron, 105(1), 46–59.e3. 10.1016/j.neuron.2019.09.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  181. Johanson CE, Roehrs T, Schuh K, & Warbasse L (1999). The effects of cocaine on mood and sleep in cocaine-dependent males. Experimental and Clinical Psychopharmacology, 7(4), 338–346. 10.1037/1064-1297.7.4.338. [DOI] [PubMed] [Google Scholar]
  182. Johnson KA, & Lovinger DM (2020). Allosteric modulation of metabotropic glutamate receptors in alcohol use disorder: Insights from preclinical investigations. Advances in Pharmacology, 88, 193–232. 10.1016/bs.apha.2020.02.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  183. Jones BE (2020). Arousal and sleep circuits. Neuropsychopharmacology, 45(1), 6–20. 10.1038/s41386-019-0444-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  184. Jones CK, Eberle EL, Peters SC, Monn JA, & Shannon HE (2005). Analgesic effects of the selective group II (mGlu2/3) metabotropic glutamate receptor agonists LY379268 and LY389795 in persistent and inflammatory pain models after acute and repeated dosing. Neuropharmacology, 49(Suppl), 206–218. 10.1016/j.neuropharm.2005.05.008. [DOI] [PubMed] [Google Scholar]
  185. Jones C, Watson D, & Fone K (2011). Animal models of schizophrenia. British Journal of Pharmacology, 164(4), 1162–1194. 10.1111/j.1476-5381.2011.01386.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  186. Ju Y-ES, McLeland JS, Toedebusch CD, Xiong C, Fagan AM, Duntley SP, et al. (2013). Sleep quality and preclinical Alzheimer disease. JAMA Neurology, 70(5), 587–593. 10.1001/jamaneurol.2013.2334. [DOI] [PMC free article] [PubMed] [Google Scholar]
  187. Ju Y-ES, Ooms SJ, Sutphen C, Macauley SL, Zangrilli MA, Jerome G, et al. (2017). Slow wave sleep disruption increases cerebrospinal fluid amyloid-β levels. Brain: A Journal of Neurology, 140(8), 2104–2111. 10.1093/brain/awx148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  188. Kahvandi N, Ebrahimi Z, Karimi SA, Shahidi S, Salehi I, Naderishahab M, et al. (2021). The effect of the mGlu8 receptor agonist, (S)-3,4-DCPG on acquisition and expression of morphine-induced conditioned place preference in male rats. Behavioral and Brain Functions, 17(1), 1–10. 10.1186/s12993-021-00174-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  189. Kalinichev M, Rouillier M, Girard F, Royer-Urios I, Bournique B, Finn T, et al. (2013). ADX71743, a potent and selective negative allosteric modulator of metabotropic glutamate receptor 7: In vitro and in vivo characterization. The Journal of Pharmacology and Experimental Therapeutics, 344(3), 624–636. 10.1124/jpet.112.200915. [DOI] [PubMed] [Google Scholar]
  190. Kalus P, Müller TJ, Zuschratter W, & Senitz D (2000). The dendritic architecture of prefrontal pyramidal neurons in schizophrenic patients. Neuroreport, 11(16), 3621–3625. 10.1097/00001756-200011090-00044. [DOI] [PubMed] [Google Scholar]
  191. Kammermeier PJ, & Worley PF (2007). Homer 1a uncouples metabotropic glutamate receptor 5 from postsynaptic effectors. Proceedings of the National Academy of Sciences of the United States of America, 104(14), 6055–6060. 10.1073/pnas.0608991104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  192. Kang J-E, Lim MM, Bateman RJ, Lee JJ, Smyth LP, Cirrito JR, et al. (2009). Amyloid-beta dynamics are regulated by orexin and the sleep-wake cycle. Science (New York, N.Y.), 326(5955), 1005–1007. 10.1126/science.1180962. [DOI] [PMC free article] [PubMed] [Google Scholar]
  193. Kantrowitz J, & Javitt DC (2012). Glutamatergic transmission in Schizophrenia. Current Opinion in Psychiatry, 25(2), 96–102. 10.1097/YCO.0b013e32835035b2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  194. Kaskie RE, Graziano B, & Ferrarelli F (2017). Schizophrenia and sleep disorders: Links, risks, and management challenges. Nature and Science of Sleep, 9, 227–239. 10.2147/NSS.S121076. [DOI] [PMC free article] [PubMed] [Google Scholar]
  195. Kato T, Takata M, Kitaichi M, Kassai M, Inoue M, Ishikawa C, et al. (2015). DSR-98776, a novel selective mGlu5 receptor negative allosteric modulator with potent antidepressant and antimanic activity. European Journal of Pharmacology, 757, 11–20. 10.1016/j.ejphar.2015.03.024. [DOI] [PubMed] [Google Scholar]
  196. Kawaura K, Karasawa JI, & Hikichi H (2016). Stimulation of the metabotropic glutamate (mGlu) 2 receptor attenuates the MK-801-induced increase in the immobility time in the forced swimming test in rats. Pharmacological Reports, 68(1), 80–84. 10.1016/j.pharep.2015.05.027. [DOI] [PubMed] [Google Scholar]
  197. Kay DC (1975). Human sleep during chronic morphine intoxication. Psychopharmacologia, 44(2), 117–124. 10.1007/BF00420997. [DOI] [PubMed] [Google Scholar]
  198. Keck TM, Yang H-J, Bi GH, Huang Y, Zhang HY, Srivastava R, et al. (2013). Fenobam sulfate inhibits cocaine-taking and cocaine-seeking behavior in rats: Implications for addiction treatment in humans. NIH Public Access, 229(2), 253–265. 10.1007/s00213-013-3106-9.Fenobam. [DOI] [PMC free article] [PubMed] [Google Scholar]
  199. Kekesi KA, Dobolyi Á, Salfay O, Nyitrai G, & Juhász G (1997). Slow wave sleep is accompanied by release of certain amino acids in the thalamus of cats. NeuroReport, 8(5), 1183–1186. 10.1097/00001756-199703240-00025. [DOI] [PubMed] [Google Scholar]
  200. Keshavan MS, Miewald J, Haas G, Sweeney J, Ganguli R, & Reynolds CF (1995). Slow-wave sleep and symptomatology in schizophrenia and related psychotic disorders. Journal of Psychiatric Research. 10.1016/0022-3956(95)00023-X. [DOI] [PubMed] [Google Scholar]
  201. Keshavan MS, Reynolds CF, Miewald JM, Montrose DM, Sweeney JA, Vasko RC, et al. (1998). Delta sleep deficits in Schizophrenia. Archives of General Psychiatry, 55(5), 443. 10.1001/archpsyc.55.5.443. [DOI] [PubMed] [Google Scholar]
  202. Kessler RC, Wai TC, Demler O, & Walters EE (2005). Prevalence, severity, and comorbidity of 12-month DSM-IV disorders in the National Comorbidity Survey replication. Archives of General Psychiatry, 62(6), 617–627. 10.1001/arch-psyc.62.6.617. [DOI] [PMC free article] [PubMed] [Google Scholar]
  203. Khazaie H, & Masoudi M (2016). Sleep disturbances in veterans with chronic war-induced PTSD. Journal of Injury and Violence Research, 8(2), 99–107. 10.5249/jivr.v8i2.808. [DOI] [PMC free article] [PubMed] [Google Scholar]
  204. Khazan N, & Colasanti B (1972). Protracted rebound in rapid movement sleep time and electroencephalogram voltage output in morphine-dependent rats upon withdrawal. Journal of Pharmacology and Experimental Therapeutics, 183(1), 23–30. [PubMed] [Google Scholar]
  205. Khazan K, Weeks JR, & Schroeder LA (1967). Electroencephalographic, electromyo-graphic and behavioral correlates during a cycle of self-maintained morphine addiction in the rat. The Journal of Pharmacology and Experimental Therapeutics, 155(3), 521–531. [PubMed] [Google Scholar]
  206. Killgore WDS (2013). Self-reported sleep correlates with prefrontal-amygdala functional connectivity and emotional functioning. Sleep, 36(11), 1597–1608. 10.5665/sleep.3106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  207. Kilpatrick DG, Resnick HS, Milanak ME, Miller MW, Keyes KM, & Friedman MJ (2013). Prevalence Using DSM-IV and DSM-5 criteria. J Trauma Stress, 26(5), 537–547. 10.1002/jts.21848. [DOI] [PMC free article] [PubMed] [Google Scholar]
  208. Kim JH, Ahn JH, Min CY, Yoo DM, & Choi HG (2021). Association between sleep quality and subjective cognitive decline: Evidence from a community health survey. Sleep Medicine, 83, 123–131. 10.1016/j.sleep.2021.04.031. [DOI] [PubMed] [Google Scholar]
  209. Kim J, An B, Kim J, Park S, Park S, Hong I, et al. (2015). MGluR2/3 in the lateral amygdala is required for fear extinction: Cortical input synapses onto the lateral amygdala as a target site of the mGluR2/3 action. Neuropsychopharmacology, 40(13), 2916–2928. 10.1038/npp.2015.145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  210. Kim SH, Fraser PE, Westaway D, St George-Hyslop PH, Ehrlich ME, & Gandy S (2010). Group II metabotropic glutamate receptor stimulation triggers production and release of Alzheimer’s amyloid(beta)42 from isolated intact nerve terminals. The Journal of Neuroscience, 30(11), 3870–3875. 10.1523/JNEUROSCI.4717-09.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  211. Kim JS, Schmid-Burgk W, Claus D, & Kornhuber HH (1982). Increased serum glutamate in depressed patients. Archiv Für Psychiatrie Und Nervenkrankheiten , 232(4), 299–304. 10.1007/BF00345492. [DOI] [PubMed] [Google Scholar]
  212. Kinney GG, Burno M, Campbell UC, Hernandez LM, Rodriguez D, Bristow LJ, et al. (2003). Metabotropic glutamate subtype 5 receptors modulate locomotor activity and sensorimotor gating in rodents. J Pharmacol Exp Ther, 306(1), 116–123. 10.1124/jpet.103.048702. [DOI] [PubMed] [Google Scholar]
  213. Klar R, Walker AG, Ghose D, Grueter BA, Engers DW, Hopkins CR, et al. (2015). Activation of metabotropic glutamate receptor 7 is required for induction of long-term potentiation at SC-CA1 synapses in the hippocampus. The Journal of Neuroscience: The Official Journal of the Society for Neuroscience, 35(19), 7600–7615. 10.1523/JNEUROSCI.4543-14.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  214. Knackstedt LA, Trantham-Davidson HL, & Schwendt M (2014). The role of ventral and dorsal striatum mGluR5 in relapse to cocaine-seeking and extinction learning. Addiction Biology, 19(1), 87–101. 10.1111/adb.12061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  215. Kobayashi I, Boarts JM, & Delahanty DL (2007). Polysomnographically measured sleep abnormalities in PTSD: A meta-analytic review. Psychophysiology, 44(4), 660–669. 10.1111/j.1469-8986.2007.537.x. [DOI] [PubMed] [Google Scholar]
  216. Kobayashi I, Huntley E, Lavela J, & Mellman TA (2012). Subjectively and objectively measured sleep with and without posttraumatic stress disorder and trauma exposure. Sleep, 35(7), 957–965. 10.5665/sleep.1960. [DOI] [PMC free article] [PubMed] [Google Scholar]
  217. Koob GF, & Colrain IM (2020). Alcohol use disorder and sleep disturbances: A feed-forward allostatic framework. Neuropsychopharmacology. 10.1038/s41386-019-0446-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  218. Korenic SA, Klingaman EA, Wickwire EM, Gaston FE, Chen H, Wijtenburg SA, et al. (2020). Sleep quality is related to brain glutamate and symptom severity in schizophrenia. Journal of Psychiatric Research, 120(October 2019), 14–20. 10.1016/j.jpsychires.2019.10.006. [DOI] [PubMed] [Google Scholar]
  219. Korpi ER, den Hollander B, Farooq U, Vashchinkina E, Rajkumar R, Nutt DJ, et al. (2015). Mechanisms of action and persistent neuroplasticity by drugs of abuse. Pharmacological Reviews, 67(4), 872–1004. 10.1124/pr.115.010967. [DOI] [PubMed] [Google Scholar]
  220. Krakow BJ, Ulibarri VA, Moore BA, & McIver ND (2015). Posttraumatic stress disorder and sleep-disordered breathing: A review of comorbidity research. Sleep Medicine Reviews, 24, 37–45. 10.1016/j.smrv.2014.11.001. [DOI] [PubMed] [Google Scholar]
  221. Krystal JH, Abi-Saab W, Perry E, D’Souza DC, Liu N, Gueorguieva R, et al. (2005). Preliminary evidence of attenuation of the disruptive effects of the NMDA glutamate receptor antagonist, ketamine, on working memory by pretreatment with the group II metabotropic glutamate receptor agonist, LY354740, in healthy human subjects. Psychopharmacology, 179(1), 303–309. 10.1007/s00213-004-1982-8. [DOI] [PubMed] [Google Scholar]
  222. Krystal JH, Davis LL, Neylan TC, Raskind MA, Schnurr PP, Stein MB, et al. (2017). It is time to address the crisis in the pharmacotherapy of posttraumatic stress disorder: A consensus statement of the PTSD psychopharmacology working group. Biological Psychiatry, 82(7), e51–e59. 10.1016/j.biopsych.2017.03.007. [DOI] [PubMed] [Google Scholar]
  223. Kumar A, Dhull DK, & Mishra PS (2015). Therapeutic potential of mGluR5 targeting in Alzheimer’s disease. Frontiers in Neuroscience, 9, 1–9. 10.3389/fnins.2015.00215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  224. Kumaresan V, Yuan M, Yee J, Famous KR, Anderson SM, Schmidt HD, et al. (2009). Metabotropic glutamate receptor 5 (mGluR5) antagonists attenuate cocaine priming- and cue-induced reinstatement of cocaine seeking. Behavioural Brain Research. 10.1016/j.bbr.2009.03.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  225. Kuo CY, Hsiao HT, Lo IH, & Nikolai T (2021). Association between obstructive sleep apnea, its treatment, and Alzheimer’s disease: Systematic mini-review. Frontiers in Aging Neuroscience, 12, 1–6. 10.3389/fnagi.2020.591737. [DOI] [PMC free article] [PubMed] [Google Scholar]
  226. Kupfer DJ, Ehlers CL, Pollock BG, Swami Nathan R, & Perel JM (1989). Clomipramine and EEG sleep in depression. Psychiatry Research, 30(2), 165–180. 10.1016/0165-1781(89)90158-3. [DOI] [PubMed] [Google Scholar]
  227. Kupfer DJ, Reynolds CF, Ulrich RF, & Grochocinski VJ (1986).Comparison of automated REM and slow-wave sleep analysis in young and middle-aged depressed subjects. Biological Psychiatry, 21(2), 189–200. 10.1016/0006-3223(86)90146-0. [DOI] [PubMed] [Google Scholar]
  228. Lan MJ, McLoughlin GA, Griffin JL, Tsang TM, Huang JTJ, Yuan P, et al. (2009). Metabonomic analysis identifies molecular changes associated with the pathophysiology and drug treatment of bipolar disorder. Molecular Psychiatry, 14(3), 269–279. 10.1038/sj.mp.4002130. [DOI] [PubMed] [Google Scholar]
  229. Laniepce A, Cabe N, Andre C, Bertran F, Boudehent C, Lahbairi N, et al. (2020). The effect of alcohol withdrawal syndrome severity on sleep, brain and cognition. Brain Communications, 2(2). 10.1093/braincomms/fcaa123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  230. Laurén J, Gimbel DA, Nygaard HB, Gilbert JW, & Strittmatter SM (2009). Cellular prion protein mediates impairment of synaptic plasticity by amyloid-B oligomers. Nature, 457(7233), 1128–1132. 10.1038/nature07761. [DOI] [PMC free article] [PubMed] [Google Scholar]
  231. Lauterbach D, Behnke C, & McSweeney LB (2011). Sleep problems among persons with a lifetime history of posttraumatic stress disorder alone and in combination with a lifetime history of other psychiatric disorders: A replication and extension. Comprehensive Psychiatry, 52(6), 580–586. 10.1016/j.comppsych.2011.01.007. [DOI] [PubMed] [Google Scholar]
  232. Lavreysen H, Langlois X, Ahnaou A, Drinkenburg W, te Riele P, Biesmans I, et al. (2013). Pharmacological characterization of JNJ-40068782, a new potent, selective, and systemically active positive allosteric modulator of the mGlu2 receptor and its radioligand [3H]JNJ-40068782. The Journal of Pharmacology and Experimental Therapeutics, 346(3), 514–527. 10.1124/jpet.113.204990. [DOI] [PubMed] [Google Scholar]
  233. Lavreysen H, Langlois X, Donck LV, Nuñez JM, Pype S, Lütjens R, & Megens A (2015). Preclinical evaluation of the antipsychotic potential of the mGlu2-positive allosteric modulator JNJ-40411813. Pharmacology Research & Perspectives, 3(2), e00097. 10.1002/prp2.97. [DOI] [PMC free article] [PubMed] [Google Scholar]
  234. le Bon O, Lanquart J-P, Hein M, & Loas G (2019). Sleep ultradian cycling: Statistical distribution and links with other sleep variables, depression, insomnia and sleepiness—A retrospective study on 2,312 polysomnograms. Psychiatry Research, 279, 140–147. 10.1016/j.psychres.2018.12.141. [DOI] [PubMed] [Google Scholar]
  235. le Bon O, Verbanck P, Hoffmann G, Murphy JR, Staner L, de Groote D, et al. (1997). Sleep in detoxified alcoholics: Impairment of most standard sleep parameters and increased risk for sleep apnea, but not for myoclonias—A controlled study. Journal of Studies on Alcohol, 58(1), 30–36. 10.15288/jsa.1997.58.30. [DOI] [PubMed] [Google Scholar]
  236. Lee H, Ogawa O, Zhu X, O’Neill MJ, Petersen RB, Castellani RJ, et al. (2004). Aberrant expression of metabotropic glutamate receptor 2 in the vulnerable neurons of Alzheimer’s disease. Acta Neuropathologica, 107(4), 365–371. 10.1007/s00401-004-0820-8. [DOI] [PubMed] [Google Scholar]
  237. Lee B, Platt DM, Rowlett JK, Adewale AS, & Spealman RD (2005). Attenuation of behavioral effects of cocaine by the metabotropic glutamate receptor 5 antagonist 2-methyl-6-(phenylethynyl)-pyridine in squirrel monkeys: Comparison with dizocilpine. Journal of Pharmacology and Experimental Therapeutics, 312(3), 1232–1240. 10.1124/jpet.104.078733. [DOI] [PubMed] [Google Scholar]
  238. Leeman-markowski BA, Meador KJ, Moo LR, Cole AJ, Hoch DB, Garcia E, et al. (2020). U.S. Department of Veterans Affairs, 40(March 2005), 315–324. 10.1016/j.yebeh.2018.06.047.Does. [DOI] [Google Scholar]
  239. Levine J, Panchalingam K, Rapoport A, Gershon S, McClure RJ, & Pettegrew JW (2000). Increased cerebrospinal fluid glutamine levels in depressed patients. Biological Psychiatry, 47(7), 586–593. 10.1016/s0006-3223(99)00284-x. [DOI] [PubMed] [Google Scholar]
  240. Lewis C, Lewis K, Kitchiner N, Isaac S, Jones I, & Bisson JI (2020). Sleep disturbance in post-traumatic stress disorder (PTSD): A systematic review and meta-analysis of actigraphy studies. European Journal of Psychotraumatology, 11(1). 10.1080/20008198.2020.1767349. [DOI] [PMC free article] [PubMed] [Google Scholar]
  241. Li P, Gao L, Yu L, Zheng X, Ulsa MC, Yang HW, et al. (2022). Daytime napping and Alzheimer’s dementia: A potential bidirectional relationship. Alzheimer’s and Dementia, 2021, 1–11. 10.1002/alz.12636. [DOI] [PMC free article] [PubMed] [Google Scholar]
  242. Li X, Xi Z-X, & Markou A (2013). Metabotropic glutamate 7 (mGlu7) receptor: A target for medication development for the treatment of cocaine dependence. Neuropharmacology, 66, 12–23. 10.1016/j.neuropharm.2012.04.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  243. Lim ASP, Kowgier M, Yu L, Buchman AS, & Bennett DA (2013). Sleep fragmentation and the risk of incident Alzheimer’s disease and cognitive decline in older persons. Sleep, 36(7), 1027–1032. 10.5665/sleep.2802. [DOI] [PMC free article] [PubMed] [Google Scholar]
  244. Lin X, Fisher NM, Dogra S, Senter RK, Reed CW, Kalbfleisch JJ, et al. (2022). Differential activity of mGlu7 allosteric modulators provides evidence for mGlu7/8 heterodimers at hippocampal Schaffer collateral-CA1 synapses. The Journal of Biological Chemistry, 298(10), 102458. 10.1016/j.jbc.2022.102458. [DOI] [PMC free article] [PubMed] [Google Scholar]
  245. Lindemann L, Porter RH, Scharf SH, Kuennecke B, Bruns A, von Kienlin M, et al. (2015). Pharmacology of basimglurant (RO4917523, RG7090), a unique metabotropic glutamate receptor 5 negative allosteric modulator in clinical development for depression. The Journal of Pharmacology and Experimental Therapeutics, 353(1), 213–233. 10.1124/jpet.114.222463. [DOI] [PubMed] [Google Scholar]
  246. Linden AM, Johnson BG, Peters SC, Shannon HE, Tian M, Wang Y, et al. (2002). Increased anxiety-related behavior in mice deficient for metabotropic glutamate 8 (mGlu8) receptor. Neuropharmacology, 43(2), 251–259. 10.1016/S0028-3908(02)00079-5. [DOI] [PubMed] [Google Scholar]
  247. Linden AM, Shannon H, Baez M, Yu JL, Koester A, & Schoepp DD (2005). Anxiolytic-like activity of the mGLU2/3 receptor agonist LY354740 in the elevated plus maze test is disrupted in metabotropic glutamate receptor 2 and 3 knock-out mice. Psychopharmacology, 179(1), 284–291. 10.1007/s00213-004-2098-x. [DOI] [PubMed] [Google Scholar]
  248. Liriano F, Hatten C, & Schwartz TL (2019). Ketamine as treatment for post-traumatic stress disorder: A review. Drugs in Context, 8, 1–7. 10.7573/dic.212305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  249. Littlewood DL, Gooding PA, Panagioti M, & Kyle SD (2016). Nightmares and suicide in posttraumatic stress disorder: The mediating role of defeat, entrapment, and hopelessness. Journal of Clinical Sleep Medicine, 12(3), 393–399. 10.5664/jcsm.5592. [DOI] [PMC free article] [PubMed] [Google Scholar]
  250. Livingstone MS, & Hubel DH (1981). Effects of sleep and arousal on the processing of visual information in the cat. Nature, 291(5816), 554–561. 10.1038/291554a0. [DOI] [PubMed] [Google Scholar]
  251. Llinás RR, & Steriade M (2006). Bursting of thalamic neurons and states of vigilance. Journal of Neurophysiology, 95(6), 3297–3308. 10.1152/jn.00166.2006. [DOI] [PubMed] [Google Scholar]
  252. Logan CN, Bechard AR, Hamor PU, Wu L, Schwendt M, & Knackstedt LA (2020). Ceftriaxone and mGlu2/3 interactions in the nucleus accumbens core affect the reinstatement of cocaine-seeking in male and female rats. 10.1007/s00213-020-05514y/Published. [DOI] [PMC free article] [PubMed] [Google Scholar]
  253. Logan RW, & McClung CA (2019). Rhythms of life: Circadian disruption and brain disorders across the lifespan. Nature Reviews. Neuroscience, 20(1), 49–65. 10.1038/s41583-018-0088-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  254. Lopez J, Hoffmann R, Armitage R, & Are A (2010). Reduced sleep spindle activity in early-onset and elevated risk for depression running head: Sleep spindles and adolescent MDD. Journal of the American Academy of Child and Adolescent Psychiatry, 49(9), 934–943. 10.1016/j.jaac.2010.05.014.Reduced. [DOI] [PMC free article] [PubMed] [Google Scholar]
  255. Lopez-Rodriguez F, Medina-Ceja L, Wilson CL, Jhung D, & Morales-Villagran A (2007). Changes in extracellular glutamate levels in rat orbitofrontal cortex during sleep and wakefulness. Archives of Medical Research, 38(1), 52–55. 10.1016/j.arcmed.2006.07.004. [DOI] [PubMed] [Google Scholar]
  256. Lorrain DS, Baccei CS, Bristow LJ, Anderson JJ, & Varney MA (2003). Effects of ketamine and N-methyl-D-aspartate on glutamate and dopamine release in the rat prefrontal cortex: modulation by a group II selective metabotropic glutamate receptor agonist LY379268. Neuroscience, 117(3), 697–706. 10.1016/s0306-4522(02)00652-8. [DOI] [PubMed] [Google Scholar]
  257. Lou Z, Chen L, Liu H, Ruan L, & Zhou W (2014). Blockade of mGluR5 in the nucleus accumbens shell but not core attenuates heroin seeking behavior in rats. Acta Pharmacologica Sinica, 35(12), 1485–1492. 10.1038/aps.2014.93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  258. Lu YM, Jia Z, Janus C, Henderson JT, Gerlai R, Wojtowicz JM, et al. (1997). Mice lacking metabotropic glutamate receptor 5 show impaired learning and reduced CA1 long-term potentiation (LTP) but normal CA3 LTP. Journal of Neuroscience, 17(13), 5196–5205. 10.1523/jneurosci.17-13-05196.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  259. Luby ED, Cohen BD, Rosenbaum G, Gottlieb JS, & Kelley R (1959). Study of a new schizophrenomimetic drug; sernyl. AMA Archives of Neurology and Psychiatry, 81(3), 363–369. 10.1001/archneurpsyc.1959.02340150095011. [DOI] [PubMed] [Google Scholar]
  260. Lucey BP, Mccullough A, Landsness EC, Toedebusch CD, Mcleland JS, Zaza AM, et al. (2019). Reduced non-rapid eye movement sleep is associated with tau pathology in early Alzheimer’s disease. Science Translational Medicine, 11-(474). 10.1126/scitranslmed.aau6550.Reduced. [DOI] [PMC free article] [PubMed] [Google Scholar]
  261. Luessen DJ, & Conn PJ (2022). Allosteric modulators of metabotropic glutamate receptors as novel therapeutics for neuropsychiatric disease. Pharmacological Reviews, 74(3), 630–661. 10.1124/pharmrev.121.000540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  262. Luscher C, & Huber KM (2010). Group 1 mGluR-dependent synaptic long-term depression: Mechanisms and implications for circuitry and disease. Neuron, 65(4), 445–459. 10.1016/j.neuron.2010.01.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  263. Lutzu S, & Castillo PE (2021). Modulation of NMDA receptors by G-protein-coupled receptors: Role in synaptic transmission, plasticity and beyond. Neuroscience, 456, 27–42. 10.1016/j.neuroscience.2020.02.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  264. Luykx JJ, Laban KG, van den Heuvel MP, Boks MPM, Mandl RCW, Kahn RS, et al. (2012). Region and state specific glutamate downregulation in major depressive disorder: A meta-analysis of 1H-MRS findings. Neuroscience and Biobehavioral Reviews, 36(1), 198–205. 10.1016/j.neubiorev.2011.05.014. [DOI] [PubMed] [Google Scholar]
  265. Ly S, Lee DA, Strus E, Prober DA, & Naidoo N (2020). Evolutionarily conserved regulation of sleep by the protein translational regulator PERK. Current Biology : CB, 30(9), 1639–1648.e3. 10.1016/j.cub.2020.02.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  266. Lyashenko EA, Poluektov MG, Levin OS, & Pchelina PV (2016). Age-related sleep changes and its implication in neurodegenerative diseases. Current Aging Science, 9(1), 26–33. 10.2174/1874609809666151130220219. [DOI] [PubMed] [Google Scholar]
  267. Lydon-Staley DM, Harrington Cleveland H, Huhn AS, Cleveland MJ, Harris J, Stankoski D, et al. (2017). Daily sleep quality affects drug craving, partially through indirect associations with positive affect, in patients in treatment for nonmedical use of prescription drugs. Paper Knowledge Toward a Media History of Documents, 65, 275–282. 10.1016/j.addbeh.2016.08.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  268. Lyon L, Burnet PW, Kew JN, Corti C, Rawlins JNP, Lane T, et al. (2011). Fractionation of spatial memory in GRM2/3 (mGlu2/mGlu3) double knockout mice reveals a role for group II metabotropic glutamate receptors at the interface between arousal and cognition. Neuropsychopharmacology, 36(13), 2616–2628. 10.1038/npp.2011.145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  269. Maglione JE, Ancoli-Israel S, Peters KW, Paudel ML, Yaffe K, Ensrud KE, et al. (2014). Subjective and objective sleep disturbance and longitudinal risk of depression in a cohort of older women. Sleep, 37(7), 1179–1187. 10.5665/sleep.3834. [DOI] [PMC free article] [PubMed] [Google Scholar]
  270. Maher MJ, Rego SA, & Asnis GM (2006). Sleep disturbances in patients with post-traumatic stress disorder: Epidemiology, impact and approaches to management. CNS Drugs, 20(7), 567–590. 10.2165/00023210-200620070-00003. [DOI] [PubMed] [Google Scholar]
  271. Maksymetz J, Moran SP, & Conn PJ (2017). Targeting metabotropic glutamate receptors for novel treatments of schizophrenia. Molecular Brain, 10(1), 1–19. 10.1186/s13041-017-0293-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  272. Manoach DS, Cain MS, Vangel MG, Khurana A, Goff DC, & Stickgold R (2004). A failure of sleep-dependent procedural learning in chronic, medicated schizophrenia. Biological psychiatry, 56(12), 951–956. 10.1016/j.biopsych.2004.09.012. [DOI] [PubMed] [Google Scholar]
  273. Manoach DS, & Stickgold R (2009). Does abnormal sleep impair memory consolidation in Schizophrenia? Frontiers in Human Neuroscience, 3, 21. 10.3389/neuro.09.021.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  274. Manoach DS, & Stickgold R (2019). Abnormal sleep spindles, memory consolidation, and Schizophrenia. Annual Review of Clinical Psychology, 15, 451–479. 10.1146/annurev-clinpsy-050718-095754. [DOI] [PMC free article] [PubMed] [Google Scholar]
  275. Manoach DS, Thakkar KN, Stroynowski E, Ely A, McKinley SK, Wamsley E, et al. (2010). Reduced overnight consolidation of procedural learning in chronic medicated schizophrenia is related to specific sleep stages. Journal of Psychiatric Research, 44(2), 112–120. 10.1016/j.jpsychires.2009.06.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  276. Marabese I, Rossi F, Palazzo E, De Novellis V, Starowicz K, Cristino L, et al. (2007). Periaqueductal gray metabotropic glutamate receptor subtype 7 and 8 mediate opposite effects on amino acid release, rostral ventromedial medulla cell activities, and thermal nociception. Journal of Neurophysiology, 98(1), 43–53. 10.1152/jn.00356.2007. [DOI] [PubMed] [Google Scholar]
  277. Maret S, Dorsaz S, Gurcel L, Pradervand S, Petit B, Pfister C, et al. (2007). Homer1a is a core brain molecular correlate of sleep loss. Proceedings of the National Academy of Sciences of the United States of America, 104(50), 20090–20095. 10.1073/pnas.0710131104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  278. Marrosu F, Portas C, Mascia MS, Casu MA, Fà M, Giagheddu M, et al. (1995). Microdialysis measurement of cortical and hippocampal acetylcholine release during sleep-wake cycle in freely moving cats. Brain Research, 671(2), 329–332. 10.1016/0006-8993(94)01399-3. [DOI] [PubMed] [Google Scholar]
  279. Marshall RD, Beebe KL, Oldham M, & Zaninelli R (2001). Efficacy and safety of paroxetine treatment for chronic PTSD: A fixed-dose, placebo-controlled study. American Journal of Psychiatry, 158(12), 1982–1988. 10.1176/appi.ajp.158.12.1982. [DOI] [PubMed] [Google Scholar]
  280. Martin SC, Monroe SK, & Diering GH (2019). Homer1a and mGluR1/5 signaling in homeostatic sleep drive and output. Yale Journal of Biology and Medicine, 92(1), 93–101. [PMC free article] [PubMed] [Google Scholar]
  281. Mateo Y, Lack CM, Morgan D, Roberts DCS, & Jones SR (2005). Reduced dopamine terminal function and insensitivity to cocaine following cocaine binge self-administration and deprivation. Neuropsychopharmacology, 30(8), 1455–1463. 10.1038/sj.npp.1300687. [DOI] [PubMed] [Google Scholar]
  282. Matosin N, Fernandez-Enright F, Lum JS, Andrews JL, Engel M, Huang X-F, et al. (2015). Metabotropic glutamate receptor 5, and its trafficking molecules Norbin and Tamalin, are increased in the CA1 hippocampal region of subjects with schizophrenia. Schizophrenia Research, 166(1–3), 212–218. 10.1016/j.schres.2015.05.001. [DOI] [PubMed] [Google Scholar]
  283. Matosin N, Newell KA, Quidé Y, Andrews JL, Teroganova N, Green MJ, et al. (2018). Effects of common GRM5 genetic variants on cognition, hippocampal volume and mGluR5 protein levels in schizophrenia. Brain Imaging and Behavior, 12(2), 509–517. 10.1007/s11682-017-9712-0. [DOI] [PubMed] [Google Scholar]
  284. Matrisciano F, Panaccione I, Zusso M, Giusti P, Tatarelli R, Iacovelli L, et al. (2007). Group-II metabotropic glutamate receptor ligands as adjunctive drugs in the treatment of depression: A new strategy to shorten the latency of antidepressant medication? Molecular Psychiatry, 12(8), 704–706. 10.1038/sj.mp.4002005. [DOI] [PubMed] [Google Scholar]
  285. Matsunaga S, Kishi T, Nomura I, Sakuma K, Okuya M, Ikuta T, et al. (2018). The efficacy and safety of memantine for the treatment of Alzheimer’s disease. Expert Opinion on Drug Safety, 17(10), 1053–1061. 10.1080/14740338.2018.1524870. [DOI] [PubMed] [Google Scholar]
  286. Matuskey D, Pittman B, Forselius E, Malison RT, & Morgan PT (2011). A multistudy analysis of the effects of early cocaine abstinence on sleep. Drug and Alcohol Dependence, 115(1–2), 62–66. 10.1016/j.drugalcdep.2010.10.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  287. Mauri MC, Ferrara A, Boscati L, Bravin S, Zamberlan F, Alecci M, et al. (1998). Plasma and platelet amino acid concentrations in patients affected by major depression and under fluvoxamine treatment. Neuropsychobiology, 37(3), 124–129. 10.1159/000026491. [DOI] [PubMed] [Google Scholar]
  288. Mazzitelli M, Palazzo E, Maione S, & Neugebauer V (2018). Group II metabotropic glutamate receptors: Role in pain mechanisms and pain modulation. Frontiers in Molecular Neuroscience, 11, 1–11. 10.3389/fnmol.2018.00383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  289. Mccormick DA, & Bal T (1997). Sleep and arousal: Thalamocortical mechanisms. Annual Review of Neuroscience, 20, 185–215. 10.1146/annurev.neuro.20.1.185. [DOI] [PubMed] [Google Scholar]
  290. McCutcheon JE, Loweth JA, Ford KA, Marinelli M, Wolf ME, & Tseng KY (2011). Group I mGlur activation reverses cocaine-induced accumulation of calcium-permeable AMPA receptors in nucleus accumbens synapses via a protein kinase C-dependent mechanism. Journal of Neuroscience, 31(41), 14536–14541. 10.1523/JNEUROSCI.3625-11.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  291. McGeehan AJ, & Olive MF (2003). The mGluR5 antagonist MPEP reduces the conditioned rewarding effects of cocaine but not other drugs of abuse. Synapse, 47(3), 240–242. 10.1002/syn.10166. [DOI] [PubMed] [Google Scholar]
  292. McGhee LL, Maani CV, Garza TH, Gaylord KM, & Black IH (2008). The correlation between ketamine and posttraumatic stress disorder in burned service members. The Journal of Trauma, 64(2 Suppl). 10.1097/ta.0b013e318160ba1d. [DOI] [PubMed] [Google Scholar]
  293. Mcomish CE, Pavey G, Gibbons A, Hopper S, Udawela M, Scarr E, et al. (2016). Lower [3H]LY341495 binding to mGlu2/3 receptors in the anterior cingulate of subjects with major depressive disorder but not bipolar disorder or Schizophrenia. Journal of Affective Disorders, 190, 241–248. 10.1016/j.jad.2015.10.004. [DOI] [PubMed] [Google Scholar]
  294. Medrano MC, Mendiguren A, & Pineda J (2015). Effect of ceftriaxone and topiramate treatments on naltrexone-precipitated morphine withdrawal and glutamate receptor desensitization in the rat locus coeruleus. Psychopharmacology, 232(15), 2795–2809. 10.1007/s00213-015-3913-2. [DOI] [PubMed] [Google Scholar]
  295. Mellman TA, Bustamante V, Fins AI, Pigeon WR, & Nolan B (2002). REM sleep and the early development of posttraumatic stress disorder. American Journal of Psychiatry, 159(10), 1696–1701. 10.1176/appi.ajp.159.10.1696. [DOI] [PubMed] [Google Scholar]
  296. Mellman TA, Kobayashi I, Lavela J, Wilson B, & Hall Brown TS (2014). A relationship between REM sleep measures and the duration of posttraumatic stress disorder in a young adult urban minority population. Sleep, 37(8), 1321–1326. 10.5665/sleep.3922. [DOI] [PMC free article] [PubMed] [Google Scholar]
  297. Mellman TA, Pigeon WR, Nowell PD, & Nolan B (2007). Relationships between REM sleep findings and PTSD symptoms during the early aftermath of trauma. Journal of Traumatic Stress, 20(5), 893–901. 10.1002/jts.20246. [DOI] [PubMed] [Google Scholar]
  298. Meyerhoff DJ, Mon A, Metzler T, & Neylan TC (2014). Cortical gamma-aminobutyric acid and glutamate in posttraumatic stress disorder and their relationships to self-reported sleep quality. Sleep, 37(5), 893–900. 10.5665/sleep.3654. [DOI] [PMC free article] [PubMed] [Google Scholar]
  299. Miguel-Hidalgo JJ, Alvarez XA, Cacabelos R, & Quack G (2002). Neuroprotection by memantine against neurodegeneration induced by β-amyloid(1–40). Brain Research, 958(1), 210–221. 10.1016/S0006-8993(02)03731-9. [DOI] [PubMed] [Google Scholar]
  300. Miller DD (2004). Atypical antipsychotics: Sleep, sedation, and efficacy. Primary Care Companion to the Journal of Clinical Psychiatry, 6(Suppl. 2), 3–7. [PMC free article] [PubMed] [Google Scholar]
  301. Moghaddam B, & Adams BW (1998). Reversal of phencyclidine effects by a group II metabotropic glutamate receptor agonist in rats. Science, 281(5381), 1349–1352. 10.1126/science.281.5381.1349. [DOI] [PubMed] [Google Scholar]
  302. Moghaddam B, & Jackson ME (2003). Glutamatergic animal models of Schizophrenia. Annals Of The New York Academy of Sciences, 1003, 131–137. 10.1196/annals.1300.065. [DOI] [PubMed] [Google Scholar]
  303. Mondino A, Cavelli M, González J, Murillo-Rodriguez E, Torterolo P, & Falconi A (2021). Effects of Cannabis consumption on sleep. Advances in Experimental Medicine and Biology, 1297, 147–162. 10.1007/978-3-030-61663-2_11. [DOI] [PubMed] [Google Scholar]
  304. Montana MC, Cavallone LF, Stubbert KK, Stefanescu AD, Kharasch ED, & Gereau RW IV. (2009). The metabotropic glutamate receptor subtype 5 antagonist fenobam is analgesic and has improved in vivo selectivity compared with the prototypical antagonist 2-methyl-6-(phenylethynyl)-pyridine. Journal of Pharmacology and Experimental Therapeutics, 330(3), 834–843. 10.1124/jpet.109.154138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  305. Morgan PT, & Malison RT (2007). Cocaine and sleep: Early abstinence. The Scientific World Journal, 7(Suppl. 2), 223–230. 10.1100/tsw.2007.209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  306. Morgan PT, Pace-Schott EF, Sahul ZH, Coric V, Stickgold R, & Malison RT (2006). Sleep, sleep-dependent procedural learning and vigilance in chronic cocaine users: Evidence for occult insomnia. Drug and Alcohol Dependence, 82(3), 238–249. 10.1016/j.drugalcdep.2005.09.014. [DOI] [PubMed] [Google Scholar]
  307. Morgan CJA, Perry EB, Cho HS, Krystal JH, & D’Souza DC (2006). Greater vulnerability to the amnestic effects of ketamine in males. Psychopharmacology, 187(4), 405–414. 10.1007/s00213-006-0409-0. [DOI] [PubMed] [Google Scholar]
  308. Morin N, Grégoire L, Gomez-Mancilla B, Gasparini F, & di Paolo T (2010). Effect of the metabotropic glutamate receptor type 5 antagonists MPEP and MTEP in Parkinsonian monkeys. Neuropharmacology, 58(7), 981–986. 10.1016/J.NEUROPHARM.2009.12.024. [DOI] [PubMed] [Google Scholar]
  309. Morphy H, Dunn KM, Lewis M, Boardman HF, & Croft PR (2007). Epidemiology of insomnia: A longitudinal study in a UK population. Sleep, 30(3), 274–280. [PubMed] [Google Scholar]
  310. Moruzzi G, & Magoun HW (1949). Brain stem reticular formation and activation of the EEG. Electroencephalography and Clinical Neurophysiology, 1(1–4), 455–473. [PubMed] [Google Scholar]
  311. Moussawi K, & Kalivas PW (2010). Group II metabotropic glutamate receptors (mGlu2/3) in drug addiction. European Journal of Pharmacology, 639(1–3), 115–122. 10.1016/j.ejphar.2010.01.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  312. Moussawi K, Zhou W, Shen H, Reichel CM, See RE, Carr DB, et al. (2011). Reversing cocaine-induced synaptic potentiation provides enduring protection from relapse. Proceedings of the National Academy of Sciences of the United States of America, 108(1), 385–390. 10.1073/pnas.1011265108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  313. Nawreen N, Baccei ML, & Herman JP (2021). Single prolonged stress reduces intrinsic excitability and excitatory synaptic drive onto pyramidal neurons in the infralimbic prefrontal cortex of adult male rats. Frontiers in Cellular Neuroscience, 15(July), 1–11. 10.3389/fncel.2021.705660. [DOI] [PMC free article] [PubMed] [Google Scholar]
  314. Nedelcovych M, Gould R, Gong X, Felts A, Grannan M, Thompson A, et al. (2015). Selective antagonism of mGlu5 alters sleep-wake and spectral EEG and ameliorates behavioral abnormalities in a rodent model of traumatic stress. The FASEB Journal, 29(S1). 10.1096/fasebj.29.1_supplement.615.8. [DOI] [Google Scholar]
  315. Neylan TC, Kessler RC, Ressler KJ, Clifford G, Beaudoin FL, An X, et al. (2021). Prior sleep problems and adverse post-traumatic neuropsychiatric sequelae of motor vehicle collision in the AURORA study. Sleep, 44(3). 10.1093/sleep/zsaa200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  316. Nicoletti F, Bockaert J, Collingridge GL, Conn PJ, Ferraguti F, Schoepp DD, et al. (2011). Metabotropic glutamate receptors: From the workbench to the bedside. Neuropharmacology, 60(7–8), 1017–1041. 10.1016/j.neuropharm.2010.10.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  317. Nicoletti F, Orlando R, Menna LD, Cannella M, Notartomaso S, Mascio G, et al. (2019). Targeting mGlu receptors for optimization of antipsychotic activity and disease-modifying effect in Schizophrenia. Frontiers in Psychiatry, 10, 1–10. 10.3389/fpsyt.2019.00049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  318. Niedzielska-Andres E, Pomierny-Chamioło L, Andres M, Walczak M, Knackstedt LA, Filip M, et al. (2021). Cocaine use disorder: A look at metabotropic glutamate receptors and glutamate transporters. Pharmacology and Therapeutics, 221. 10.1016/j.pharmthera.2020.107797. [DOI] [PubMed] [Google Scholar]
  319. Niswender CM, & Conn PJ (2010). Metabotropic glutamate receptors: physiology, pharmacology, and disease. Annual Review of Pharmacology and Toxicology, 50, 295–322. 10.1146/annurev.pharmtox.011008.145533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  320. O’Connor RM, McCafferty CP, Bravo JA, Singewald N, Holmes A, & Cryan JF (2019). Increased amygdalar metabotropic glutamate receptor 7 mRNA in a genetic mouse model of impaired fear extinction. Psychopharmacology, 236(1), 265–272. 10.1007/s00213-018-5031-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  321. O’Connor RM, Thakker DR, Schmutz M, van der Putten H, Hoyer D, Flor PJ, et al. (2013). Adult siRNA-induced knockdown of mGlu7 receptors reduces anxiety in the mouse. Neuropharmacology, 72, 66–73. 10.1016/j.neuropharm.2013.03.028. [DOI] [PubMed] [Google Scholar]
  322. Obal F, & Krueger JM (2003). Biochemical regulation of non-rapid-eye-movement sleep. Frontiers in Bioscience, 8, d520–d550. 10.2741/1033. [DOI] [PubMed] [Google Scholar]
  323. Ohayon MM, Carskadon MA, Guilleminault C, & Vitiello MV (2004). Meta-analysis of quantitative sleep parameters from childhood to old age in healthy individuals: Developing normative sleep values across the human lifespan. Sleep, 27(7), 1255–1273. 10.1093/sleep/27.7.1255. [DOI] [PubMed] [Google Scholar]
  324. Olive MF (2010). Cognitive effects of Group I metabotropic glutamate receptor ligands in the context of drug addiction. European Journal of Pharmacology, 639(1–3), 47–58. 10.1016/j.ejphar.2010.01.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  325. Pace-Schott EF, Germain A, & Milad MR (2015). Sleep and REM sleep disturbance in the pathophysiology of PTSD: The role of extinction memory. Biology of Mood and Anxiety Disorders, 5(1), 1–19. 10.1186/s13587-015-0018-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  326. Pace-Schott EF, Stickgold R, Muzur A, Wigren PE, Ward AS, Hart CL, et al. (2005). Sleep quality deteriorates over a binge-abstinence cycle in chronic smoked cocaine users. Psychopharmacology, 179(4), 873–883. 10.1007/s00213-004-2088-z. [DOI] [PubMed] [Google Scholar]
  327. Page G, Khidir FAL, Pain S, Barrier L, Fauconneau B, Guillard O, et al. (2006). Group I metabotropic glutamate receptors activate the p70S6 kinase via both mammalian target of rapamycin (mTOR) and extracellular signal-regulated kinase (ERK 1/2) signaling pathways in rat striatal and hippocampal synaptoneurosomes. Neurochemistry International, 49(4), 413–421. 10.1016/j.neuint.2006.01.020. [DOI] [PubMed] [Google Scholar]
  328. Palazzo E, Fu Y, Ji G, Maione S, & Neugebauer V (2008). Group III mGluR7 and mGluR8 in the amygdala differentially modulate nocifensive and affective pain behaviors. Neuropharmacology, 55(4), 537–545. 10.1016/j.neuropharm.2008.05.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  329. Palucha A, & Pilc A (2007). Metabotropic glutamate receptor ligands as possible anxiolytic and antidepressant drugs. Pharmacology & Therapeutics, 115(1), 116–147. 10.1016/j.pharmthera.2007.04.007. [DOI] [PubMed] [Google Scholar]
  330. Pałucha-Poniewiera A, & Pilc A (2013). A selective mGlu7 receptor antagonist MMPIP reversed antidepressant-like effects of AMN082 in rats. Behavioural Brain Research, 238, 109–112. 10.1016/j.bbr.2012.10.004. [DOI] [PubMed] [Google Scholar]
  331. Parmentier-Batteur S, Obrien JA, Doran S, Nguyen SJ, Flick RB, Uslaner JM, et al. (2012). Differential effects of the mGluR5 positive allosteric modulator CDPPB in the cortex and striatum following repeated administration. Neuropharmacology, 62(3), 1453–1460. 10.1016/j.neuropharm.2010.11.013. [DOI] [PubMed] [Google Scholar]
  332. Pecknold JC, McClure DJ, Appeltauer L, Wrzesinski L, & Allan T (1982). Treatment of anxiety using fenobam (a nonbenzodiazepine) in a double-blind standard (diazepam) placebo-controlled study. Journal of Clinical Psychopharmacology, 2(2), 129–133. [PubMed] [Google Scholar]
  333. Pelkey KA, Lavezzari G, Racca C, Roche KW, & McBain CJ (2005). mGluR7 is a metaplastic switch controlling bidirectional plasticity of feedforward inhibition. Neuron, 46(1), 89–102. 10.1016/j.neuron.2005.02.011. [DOI] [PubMed] [Google Scholar]
  334. Pierri JN, Volk CL, Auh S, Sampson A, & Lewis DA (2001). Decreased somal size of deep layer 3 pyramidal neurons in the prefrontal cortex of subjects with schizophrenia. Archives of General Psychiatry, 58(5), 466–473. 10.1001/archpsyc.58.5.466. [DOI] [PubMed] [Google Scholar]
  335. Pierri JN, Volk CLE, Auh S, Sampson A, & Lewis DA (2003). Somal size of prefrontal cortical pyramidal neurons in schizophrenia: Differential effects across neuronal subpopulations. Biological Psychiatry, 54(2), 111–120. 10.1016/s0006-3223(03)00294-4. [DOI] [PubMed] [Google Scholar]
  336. Pomierny-Chamiolo, Miszkiel J, Frankowska M, & Mizera J (2017). Neuroadaptive changes in metabotropic glutamate mGlu2/3R expression during different phases of cocaine addiction in rats. Pharmacological Reports, 69(5), 1073–1081. [DOI] [PubMed] [Google Scholar]
  337. Porkka-Heiskanen T, Strecker RE, Thakkar M, Bjorkum AA, Greene RW, & McCarley RW (1997). Adenosine: A mediator of the sleep-inducing effects of prolonged wakefulness. Science, 276(5316), 1265–1268. 10.1126/science.276.5316.1265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  338. Post RM, Gillin JC, Wyatt RJ, & Goodwin FK (1974). The effect of orally administered cocaine on sleep of depressed patients. Psychopharmacologia, 37(1), 59–66. 10.1007/BF00426683. [DOI] [PubMed] [Google Scholar]
  339. Potter LE, Zanos P, & Gould TD (2020). Antidepressant effects and mechanisms of group II mGlu receptor-specific negative allosteric modulators. Neuron, 105(1), 139–148. 10.1016/j.neuron.2019.12.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  340. Potvin O, Lorrain D, Forget H, Dube M, Grenier S, Preville M, et al. (2012). Sleep quality and 1-year incident cognitive impairment in community-dwelling older adults. Sleep, 35(4), 491–499. 10.5665/sleep.1732. [DOI] [PMC free article] [PubMed] [Google Scholar]
  341. Pritchett D, Jagannath A, Brown LA, Tam SKE, Hasan S, Gatti S, et al. (2015). Deletion of metabotropic glutamate receptors 2 and 3 (mGlu2 & mGlu3) in mice disrupts sleep and wheel-running activity, and increases the sensitivity of the circadian system to light. PLoS One, 10(5), 1–21. 10.1371/journal.pone.0125523. [DOI] [PMC free article] [PubMed] [Google Scholar]
  342. Pritchett D, Wulff K, Oliver PL, Bannerman DM, Davies KE, Harrison PJ, et al. (2012). Evaluating the links between schizophrenia and sleep and circadian rhythm disruption. Journal of Neural Transmission, 119(10), 1061–1075. 10.1007/s00702-012-0817-8. [DOI] [PubMed] [Google Scholar]
  343. Quiroz JA, Tamburri P, Deptula D, Banken L, Beyer U, Rabbia M, et al. (2016). Efficacy and safety of basimglurant as adjunctive therapy for major depression: A randomized clinical trial. JAMA Psychiatry, 73(7), 675–684. 10.1001/jamapsychiatry.2016.0838. [DOI] [PubMed] [Google Scholar]
  344. Raab-Graham KF, Workman ER, Namjoshi S, & Niere F (2016). Pushing the threshold: How NMDAR antagonists induce homeostasis through protein synthesis to remedy depression. Brain Research, 1647, 94–104. 10.1016/j.brainres.2016.04.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  345. Rady A, Mekky J, Moulokheya T, & Elsheshai A (2020). Polysomnographic correlates for the risk of relapse in detoxified opiate-misuse patients. Neuropsychiatric Disease and Treatment, 16, 3187–3196. 10.2147/NDT.S284337. [DOI] [PMC free article] [PubMed] [Google Scholar]
  346. Rasch B, & Born J (2013). About sleep’s role in memory. Physiological Reviews, 93(2), 681–766. 10.1152/physrev.00032.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  347. Rauch SL, Shin LM, & Phelps EA (2006). Neurocircuitry models of posttraumatic stress disorder and extinction: Human neuroimaging research-past, present, and future. Biological Psychiatry, 60(4), 376–382. 10.1016/j.biopsych.2006.06.004. [DOI] [PubMed] [Google Scholar]
  348. Reeve S, Sheaves B, & Freeman D (2015). The role of sleep dysfunction in the occurrence of delusions and hallucinations: A systematic review. Clinical Psychology Review, 42, 96–115. 10.1016/j.cpr.2015.09.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  349. Régio Brambilla C, Veselinović T, Rajkumar R, Mauler J, Orth L, Ruch A, et al. (2020). mGluR5 receptor availability is associated with lower levels of negative symptoms and better cognition in male patients with chronic schizophrenia. Human Brain Mapping, 41(10), 2762–2781. 10.1002/hbm.24976. [DOI] [PMC free article] [PubMed] [Google Scholar]
  350. Revett TJ, Baker GB, Jhamandas J, & Kar S (2013). Glutamate system, amyloid β peptides and tau protein: Functional interrelationships and relevance to Alzheimer disease pathology. Journal of Psychiatry and Neuroscience, 38(1), 6–23. 10.1503/jpn.110190. [DOI] [PMC free article] [PubMed] [Google Scholar]
  351. Reynolds CF, & Kupfer DJ (1987). Sleep research in affective illness: State of the art circa 1987. Sleep, 10(3), 199–215. 10.1093/sleep/10.3.199. [DOI] [PubMed] [Google Scholar]
  352. Richards G, Messer J, Faull RLM, Stadler H, Wichmann J, Huguenin P, et al. (2010). Altered distribution of mGlu2 receptors in β-amyloid-affected brain regions of Alzheimer cases and aged PS2APP mice. Brain Research, 1363, 180–190. 10.1016/j.brainres.2010.09.072. [DOI] [PubMed] [Google Scholar]
  353. Riedel G, Casabona G, Platt B, MacPhail EM, & Nicoletti F (2000). Fear conditioning-induced time- and subregion-specific increase in expression of mGlu5 receptor protein in rat hippocampus. Neuropharmacology, 39(11), 1943–1951. 10.1016/S0028-3908(00)00037-X. [DOI] [PubMed] [Google Scholar]
  354. Riemann D, Berger M, & Voderholzer U (2001). Sleep and depression—Results from psychobiological studies: An overview. Biological Psychology, 57(1–3), 67–103. 10.1016/S0301-0511(01)00090-4. [DOI] [PubMed] [Google Scholar]
  355. Riemann D, Krone LB, Wulff K, & Nissen C (2020). Sleep, insomnia, and depression. Neuropsychopharmacology, 45(1), 74–89. 10.1038/s41386-019-0411-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  356. Robertson JA, Purple RJ, Cole P, Zaiwalla Z, Wulff K, & Pattinson KTS (2016). Sleep disturbance in patients taking opioid medication for chronic back pain. Anaesthesia, 71(11), 1296–1307. 10.1111/anae.13601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  357. Rodd ZA, McKinzie DL, Bell RL, McQueen VK, Murphy JM, Schoepp DD, et al. (2006). The metabotropic glutamate 2/3 receptor agonist LY404039 reduces alcohol-seeking but not alcohol self-administration in alcohol-preferring (P) rats. Behavioural Brain Research, 171(2), 207–215. 10.1016/j.bbr.2006.03.032. [DOI] [PubMed] [Google Scholar]
  358. Rodriguez AL, Nong Y, Sekaran NK, Alagille D, Tamagnan GD, & Conn PJ (2005). A close structural analog of 2-methyl-6-(phenylethynyl)-pyridine acts as a neutral allosteric site ligand on metabotropic glutamate receptor subtype 5 and blocks the effects of multiple allosteric modulators. Molecular Pharmacology, 68(6), 1793–1802. 10.1124/mol.105.016139. [DOI] [PubMed] [Google Scholar]
  359. Roehrs T, Sibai M, & Roth T (2021). Sleep and alertness disturbance and substance use disorders: A bi-directional relation. Pharmacology Biochemistry and Behavior, 203, 173153. 10.1016/j.pbb.2021.173153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  360. Rogawski MA, & Wenk GL (2003). The neuropharmacological basis for the use of memantine in the treatment of Alzheimer’s disease. CNS Drug Reviews, 9(3),275–308. 10.1111/j.1527-3458.2003.tb00254.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  361. Romanella SM, Roe D, Tatti E, Cappon D, Paciorek R, Testani E, et al. (2021). The sleep side of aging and Alzheimer’s disease. Sleep Medicine, 77, 209–225. 10.1016/j.sleep.2020.05.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  362. Romano C, Sesma MA, McDonald CT, O’Malley K, van den Pol AN, & Olney JW (1995). Distribution of metabotropic glutamate receptor mGluR5 immunoreactivity in rat brain. The Journal of Comparative Neurology, 355(3), 455–469. 10.1002/cne.903550310. [DOI] [PubMed] [Google Scholar]
  363. Rondina R 2nd, Olsen RK, McQuiggan DA, Fatima Z, Li L, Oziel E, et al. (2016). Age-related changes to oscillatory dynamics in hippocampal and neocortical networks. Neurobiology of Learning and Memory, 134, 15–30. 10.1016/j.nlm.2015.11.017. [DOI] [PubMed] [Google Scholar]
  364. Ronesi JA, & Huber KM (2008). Homer interactions are necessary for metabotropic glutamate receptor-induced long-term depression and translational activation. The Journal of Neuroscience, 28(2), 543–547. 10.1523/JNEUROSCI.5019-07.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  365. Rosen IM, Aurora RN, Kirsch DB, Carden KA, Malhotra RK, Ramar K, et al. (2019). Chronic opioid therapy and sleep: An American academy of sleep medicine position statement. Journal of Clinical Sleep Medicine, 15(11), 1671–1673. 10.5664/jcsm.8062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  366. Rosenwasser AM, & Turek FW (2015). Neurobiology of circadian rhythm regulation. Sleep Medicine Clinics, 10(4), 403–412. 10.1016/j.jsmc.2015.08.003. [DOI] [PubMed] [Google Scholar]
  367. Rousseau PF, Vallat R, Coste O, Cadis H, Nicolas F, Trousselard M, et al. (2021). Sleep parameters improvement in PTSD soldiers after symptoms remission. Scientific Reports, 11(1), 1–13. 10.1038/s41598-021-88337-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  368. Salling MC, Grassetti A, Ferrera VP, Martinez D, & Foltin RW (2021). Negative allosteric modulation of metabotropic glutamate receptor 5 attenuates alcohol self-administration in baboons. Pharmacology, Biochemistry, and Behavior, 208, 173227. 10.1016/j.pbb.2021.173227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  369. Sanacora G, Treccani G, & Popoli M (2012). Towards a glutamate hypothesis of depression: An emerging frontier of neuropsychopharmacology for mood disorders. Neuropharmacology, 62(1), 63–77. 10.1016/j.neuropharm.2011.07.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  370. Sansig G, Bushell TJ, Clarke VR, Rozov A, Burnashev N, Portet C, et al. (2001). Increased seizure susceptibility in mice lacking metabotropic glutamate receptor 7. The Journal of Neuroscience: The Official Journal of the Society for Neuroscience, 21(22), 8734–8745. 10.1523/JNEUROSCI.21-22-08734.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  371. Saper CB, & Fuller PM (2017). Wake-sleep circuitry: An overview. Current Opinion in Neurobiology, 44, 186–192. 10.1016/j.conb.2017.03.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  372. Saper CB, Fuller PM, Pedersen NP, Lu J, & Scammell TE (2010). Sleep state switching. Neuron, 68(6), 1023–1042. 10.1016/j.neuron.2010.11.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  373. Saper CB, Scammell TE, & Lu J (2005). Hypothalamic regulation of sleep and circadian rhythms. Nature, 437(7063), 1257–1263. 10.1038/nature04284. [DOI] [PubMed] [Google Scholar]
  374. Scammell TE, Arrigoni E, & Lipton JO (2017). Neural circuitry of wakefulness and sleep. Neuron, 93(4), 747–765. 10.1016/j.neuron.2017.01.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  375. Schierenbeck T, Riemann D, Berger M, & Hornyak M (2008). Effect of illicit recreational drugs upon sleep: Cocaine, ecstasy and marijuana. Sleep Medicine Reviews, 12(5), 381–389. 10.1016/j.smrv.2007.12.004. [DOI] [PubMed] [Google Scholar]
  376. Scullin MK, & Bliwise DL (2015a). Is cognitive aging associated with levels of REM sleep or slow wave sleep? Sleep, 38(3), 335–336. 10.5665/sleep.4482. [DOI] [PMC free article] [PubMed] [Google Scholar]
  377. Scullin MK, & Bliwise DL (2015b). Sleep, cognition, and normal aging: Integrating a half century of multidisciplinary research. Perspectives on Psychological Science: A Journal of the Association for Psychological Science, 10(1), 97–137. 10.1177/1745691614556680. [DOI] [PMC free article] [PubMed] [Google Scholar]
  378. Sekiya Y, Nakagawa T, Ozawa T, Minami M, & Satoh M (2004). Facilitation of morphine withdrawal symptoms and morphine-induced conditioned place preference by a glutamate transporter inhibitor DL-threo-β-benzyloxyaspartate in rats. European Journal of Pharmacology, 485(1–3), 201–210. 10.1016/j.ejphar.2003.11.062. [DOI] [PubMed] [Google Scholar]
  379. Sengmany K, & Gregory KJ (2016). Metabotropic glutamate receptor subtype 5: Molecular pharmacology, allosteric modulation and stimulus bias. British Journal of Pharmacology, 173(20), 3001–3017. 10.1111/bph.13281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  380. Shankar GM, Bloodgood BL, Townsend M, Walsh DM, Selkoe DJ, & Sabatini BL (2007). Natural oligomers of the Alzheimer amyloid-β protein induce reversible synapse loss by modulating an NMDA-type glutamate receptor-dependent signaling pathway. Journal of Neuroscience, 27(11), 2866–2875. 10.1523/JNEUROSCI.4970-06.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  381. Sharkey KM, Kurth ME, Anderson BJ, Corso RP, Millman RP, & Stein MD (2011). Assessing sleep in opioid dependence: A comparison of subjective ratings, sleep diaries, and home polysomnography in methadone maintenance patients. Drug and Alcohol Dependence, 113(2–3), 245–248. 10.1016/j.drugalcdep.2010.08.007.Assessing. [DOI] [PMC free article] [PubMed] [Google Scholar]
  382. Sharma P (2016). Excessive daytime sleepiness in Schizophrenia: A naturalistic clinical study. Journal of Clinical and Diagnostic Research, 10–12. 10.7860/jcdr/2016/21272.8627. [DOI] [PMC free article] [PubMed] [Google Scholar]
  383. Shaw IR, Lavigne G, & Mayer P (2006). Acute intravenous administration of morphine perturbs sleep architecture in healthy painfree young adults: A preliminary study. Sleep, 28(6), 677–682. 10.1093/sleep/28.6.677. [DOI] [PubMed] [Google Scholar]
  384. Shepherd JD (2012). Memory, plasticity and sleep—A role for calcium permeable AMPA receptors? Frontiers in Molecular Neuroscience, 5, 49. 10.3389/fnmol.2012.00049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  385. Sherin JE, Elmquist JK, Torrealba F, & Saper CB (1998). Innervation of histaminergic tuberomammillary neurons by GABAergic and galaninergic neurons in the ventrolateral preoptic nucleus of the rat. The Journal of Neuroscience, 18(12), 4705–4721. 10.1523/JNEUROSCI.18-12-04705.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  386. Shi G, Yin C, Fan Z, Xing L, Mostovoy Y, Kwok PY, et al. (2021). Mutations in metabotropic glutamate receptor 1 contribute to natural short sleep trait. Current Biology, 31(1), 13–24.e4. 10.1016/j.cub.2020.09.071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  387. Shigemoto R, Nakanishi S, & Mizuno N (1992). Distribution of the mRNA for a metabotropic glutamate receptor (mGluR1) in the central nervous system: An in situ hybridization study in adult and developing rat. The Journal of Comparative Neurology, 322(1), 121–135. 10.1002/cne.903220110. [DOI] [PubMed] [Google Scholar]
  388. Shigemoto R, Nomura S, Ohishi H, Sugihara H, Nakanishi S, & Mizuno N (1993). Immunohistochemical localization of a metabotropic glutamate receptor, mGluR5, in the rat brain. Neuroscience Letters, 163(1), 53–57. 10.1016/0304-3940(93)90227-c. [DOI] [PubMed] [Google Scholar]
  389. Shin H-Y, Gadzhanova S, Roughead EE, Ward MB, & Pont LG (2016). The use of antipsychotics among people treated with medications for dementia in residential aged care facilities. International Psychogeriatrics, 28(6), 977–982. 10.1017/S1041610215002434. [DOI] [PubMed] [Google Scholar]
  390. Shiraishi-Yamaguchi Y, & Furuichi T (2007). The Homer family proteins. Genome Biology, 8(2), 1–12. 10.1186/gb-2007-8-2-206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  391. Siciliano CA, Ferris MJ, & Jones SR (2016). Cocaine self-administration disrupts mesolimbic dopamine circuit function and attenuates dopaminergic responsiveness to cocaine. Behavioural Neuroscience, 42(4), 2091–2096. 10.1111/ejn.12970. [DOI] [PMC free article] [PubMed] [Google Scholar]
  392. Sokolenko E, Hudson MR, Nithianantharajah J, & Jones NC (2019). The mGluR2/3 agonist LY379268 reverses NMDA receptor antagonist effects on cortical gamma oscillations and phase coherence, but not working memory impairments, in mice. Journal of Psychopharmacology, 33(12), 1588–1599. 10.1177/0269881119875976. [DOI] [PubMed] [Google Scholar]
  393. Song Y, Blackwell T, Yaffe K, Ancoli-Israel S, Redline S, & Stone KL (2015). Relationships between sleep stages and changes in cognitive function in older men: The MrOS Sleep Study. Sleep, 38(3), 411–421. 10.5665/sleep.4500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  394. Sprecher KE, Ferrarelli F, & Benca RM (2015). Sleep and plasticity in schizophrenia. Current Topics in Behavioral Neurosciences, 25, 433–458. 10.1007/7854_2014_366. [DOI] [PMC free article] [PubMed] [Google Scholar]
  395. Srinivasan V, Pandi-Perumal SR, Cardinali DP, Poeggeler B, & Hardeland R (2006). Melatonin in Alzheimer’s disease and other neurodegenerative disorders. Behavioral and Brain Functions, 2, 1–23. 10.1186/1744-9081-2-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  396. Steiger A, & Kimura M (2010). Wake and sleep EEG provide biomarkers in depression. Journal of Psychiatric Research, 44(4), 242–252. 10.1016/j.jpsychires.2009.08.013. [DOI] [PubMed] [Google Scholar]
  397. Steiger A, & Pawlowski M (2019). Depression and sleep. International Journal of Molecular Sciences, 20(3), 1–14. 10.3390/ijms20030607. [DOI] [PMC free article] [PubMed] [Google Scholar]
  398. Steiger A, Pawlowski M, & Kimura M (2015). Sleep electroencephalography as a biomarker in depression. ChronoPhysiology and Therapy, 15. 10.2147/cpt.s41760. [DOI] [Google Scholar]
  399. Stein MD, Herman DS, Bishop S, Lassor JA, Weinstock M, Anthony J, et al. (2004). Sleep disturbances among methadone maintained patients. Journal of Substance Abuse Treatment, 26(3), 175–180. 10.1016/S0740-5472(03)00191-0. [DOI] [PubMed] [Google Scholar]
  400. Steriade M, Domich L, Oakson G, & Deschfines M (1987). The deafferented reticular thalamic nucleus generates spindle rhythmicity. Journal of Neurophysiology, 57(1), 260–273. 10.1152/jn.1987.57.1.260. [DOI] [PubMed] [Google Scholar]
  401. Steriade M, Iosif G, & Apostol V (1968). Responsiveness of thalamic and cortical motor relays during arousal and various stages of sleep. Journal of Neurophysiology, 32(2), 251–265. 10.1152/jn.1969.32.2.251. [DOI] [PubMed] [Google Scholar]
  402. Stopa EG, Volicer L, Kuo-Leblanc V, Harper D, Lathi D, Tate B, et al. (1999). Pathologic evaluation of the human suprachiasmatic nucleus in severe dementia. Neuropathology and Experimental Neurology, 58(1), 29–39. 10.1097/00005072-199901000-00004. [DOI] [PubMed] [Google Scholar]
  403. Straus LD, Acheson DT, Risbrough VB, Sean PA, Diego S, Diego S, et al. (2018). Sleep deprivation disrupts recall of conditioned fear extinction. Biological psychiatry. Cognitive Neuroscience and Neuroimaging, 2(2), 123–129. 10.1016/j.bpsc.2016.05.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  404. Sun YG, Rupprecht V, Zhou L, Dasgupta R, Seibt F, & Beierlein M (2016). MGluR1 and mGluR5 synergistically control cholinergic synaptic transmission in the thalamic reticular nucleus. Journal of Neuroscience, 36(30), 7886–7896. 10.1523/JNEUROSCI.0409-16.2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  405. Suntsova N, Szymusiak R, Alam MN, Guzman-Marin R, & McGinty D (2002). Sleep-waking discharge patterns of median preoptic nucleus neurons in rats. The Journal of Physiology, 543, 665–677. 10.1113/jphysiol.2002.023085. [DOI] [PMC free article] [PubMed] [Google Scholar]
  406. Swanson CJ, Baker DA, Carson D, Worley PF, & Kalivas PW (2001). Repeated cocaine administration attenuates group I metabotropic glutamate receptor-mediated glutamate release and behavioral activation: A potential role for homer. The Journal of Neuroscience: The Official Journal of the Society for Neuroscience, 21(22), 9043–9052. 10.1523/JNEUROSCI.21-22-09043.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  407. Sweeten BLW, Adkins AM, Wellman LL, & Sanford LD (2021). Group II metabotropic glutamate receptor activation in the basolateral amygdala mediates individual differences in stress-induced changes in rapid eye movement sleep. Progress in Neuro-Psychopharmacology & Biological Psychiatry, 104, 110014. 10.1016/j.pnpbp.2020.110014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  408. Swift KM (2020). Sleep and PTSD: Delving deeper to understand a complicated relationship. Sleep, 43(9), 1–3. 10.1093/sleep/zsaa074. [DOI] [Google Scholar]
  409. Tadavarty R, Rajput PS, Wong JM, Kumar U, & Sastry BR (2011). Sleep-deprivation induces changes in GABAb and mglu receptor expression and has consequences for synaptic long-term depression. PLoS One, 6(9). 10.1371/journal.pone.0024933. [DOI] [PMC free article] [PubMed] [Google Scholar]
  410. Taylor DL, Diemel LT, Cuzner ML, & Pocock JM (2002). Activation of group II metabotropic glutamate receptors underlies microglial reactivity and neurotoxicity following stimulation with chromogranin A, a peptide up-regulated in Alzheimer’s disease. Journal of Neurochemistry, 82(5), 1179–1191. 10.1046/j.1471-4159.2002.01062.x. [DOI] [PubMed] [Google Scholar]
  411. Taylor DJ, Pruiksma KE, Hale W, McLean CP, Zandberg LJ, Brown L, et al. (2020). Sleep problems in active duty military personnel seeking treatment for post-traumatic stress disorder: Presence, change, and impact on outcomes. Sleep, 43(10), 1–13. 10.1093/sleep/zsaa065. [DOI] [PubMed] [Google Scholar]
  412. Tesler N, Gerstenberg M, Franscini M, Jenni OG, Walitza S, & Huber R (2016). Increased frontal sleep slow wave activity in adolescents with major depression. NeuroImage: Clinical, 10, 250–256. 10.1016/j.nicl.2015.10.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  413. Tok S, Ahnaou A, & Drinkenburg W (2021). Functional neurophysiological biomarkers of early-stage Alzheimer’s Disease: A perspective of network hyperexcitability in disease progression. Journal of Alzheimer’s Disease, 1–28. 10.3233/jad-210397. [DOI] [PMC free article] [PubMed] [Google Scholar]
  414. Tokuyama S, Wakabayashi H, & Ho IK (1995). Direct evidence for a role of glutamate in the expression of the opioid withdrawal syndrome. European Journal of Pharmacology, 295, 123–129. 10.1007/978-1-4020-5614-7_3754. [DOI] [PubMed] [Google Scholar]
  415. Tononi G, & Cirelli C (2014). Sleep and the price of plasticity: From synaptic and cellular homeostasis to memory consolidation and integration. Neuron, 81(1), 12–34. 10.1016/j.neuron.2013.12.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  416. Tripathi R, Rao R, Dhawan A, Jain R, & Sinha S (2020). Opioids and sleep—A review of literature. Sleep Medicine, 67, 269–275. 10.1016/j.sleep.2019.06.012. [DOI] [PubMed] [Google Scholar]
  417. Troxel WM, Rodriguez A, Seelam R, Tucker JS, Shih RA, Dong L, et al. (2021). Longitudinal associations of sleep problems with alcohol and cannabis use from adolescence to emerging adulthood. Sleep, 44(10). 10.1093/sleep/zsab102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  418. Trullas R, & Skolnick P (1990). Functional antagonists at the NMDA receptor complex exhibit antidepressant actions. European Journal of Pharmacology, 185(1), 1–10. 10.1016/0014-2999(90)90204-J. [DOI] [PubMed] [Google Scholar]
  419. Um JW, Kaufman AC, Kostylev M, Heiss JK, Stagi M, Takahashi H, et al. (2013). Metabotropic glutamate receptor 5 is a coreceptor for Alzheimer Aβ oligomer bound to cellular prion protein. Neuron, 79(5), 887–902. 10.1016/j.neuron.2013.06.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  420. Umbricht D, Niggli M, Sanwald-Ducray P, Deptula D, Moore R, Grünbauer W, et al. (2020). Randomized, double-blind, placebo-controlled trial of the mGlu2/3 negative allosteric modulator decoglurant in partially refractory major depressive disorder. Journal of Clinical Psychiatry, 81(4). 10.4088/JCP.18m12470. [DOI] [PubMed] [Google Scholar]
  421. Valentino RJ, & Volkow ND (2020). Drugs, sleep, and the addicted brain. Neuropsychopharmacology, 45(1), 3–5. 10.1038/s41386-019-0465-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  422. Valladares EM, & Irwin MR (2007). Polysomnographic sleep dysregulation in cocaine dependence. The Scientific World Journal, 7(Suppl. 2), 213–216. 10.1100/tsw.2007.264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  423. van der Kam EL, de Vry J, & Tzschentke TM (2007). Effect of 2-methyl-6-(phenylethynyl) pyridine on intravenous self-administration of ketamine and heroin in the rat. Behavioural Pharmacology, 18(8), 717–724. 10.1097/FBP.0b013e3282f18d58. [DOI] [PubMed] [Google Scholar]
  424. van Huijstee AN, & Mansvelder HD (2014). Glutamatergic synaptic plasticity in the mesocorticolimbic system in addiction. Frontiers in Cellular Neuroscience, 8, 466. 10.3389/fncel.2014.00466. [DOI] [PMC free article] [PubMed] [Google Scholar]
  425. van Liempt S, van Zuiden M, Westenberg H, Super A, & Vermetten E (2013). Impact of impaired sleep on the development of PTSD symptoms in combat veterans: A prospective longitudinal cohort study. Depression and Anxiety, 30(5), 469–474. 10.1002/da.22054. [DOI] [PubMed] [Google Scholar]
  426. Veatch OJ, Sutcliffe JS, Warren ZE, Keenan BT, Potter MH, & Malow BA (2017). Shorter sleep duration is associated with social impairment and comorbidities in ASD. Autism Research: Official Journal of the International Society for Autism Research, 10(7), 1221–1238. 10.1002/aur.1765. [DOI] [PMC free article] [PubMed] [Google Scholar]
  427. Veeneman MMJ, Boleij H, Broekhoven MH, Snoeren EMS, Guitart Masip M, Cousijn J, et al. (2011). Dissociable roles of mGlu5 and dopamine receptors in the rewarding and sensitizing properties of morphine and cocaine. Psychopharmacology, 214(4), 863–876. 10.1007/s00213-010-2095-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  428. Verret L, Goutagny R, Fort P, Cagnon L, Salvert D, Leger L, et al. (2003). A role of melanin-concentrating hormone producing neurons in the central regulation of paradoxical sleep. BMC Neuroscience, 4, 19. 10.1186/1471-2202-4-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  429. Vyazovskiy V, Cirelli C, Pfister-Genskow M, Faraguna U, & Tononi G (2008). Molecular and electrophysiological evidence for net synaptic potentiation in wake and depression in sleep. Nature Neuroscience, 11(2), 200–208. 10.1038/nn2035. [DOI] [PubMed] [Google Scholar]
  430. Walker AG, Wenthur CJ, Xiang Z, Rook JM, Emmitte KA, Niswender CM, et al. (2015). Metabotropic glutamate receptor 3 activation is required for long-term depression in medial prefrontal cortex and fear extinction. Proceedings of the National Academy of Sciences of the United States of America, 112(4), 1196–1201. 10.1073/pnas.1416196112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  431. Wamsley EJ, Tucker MA, Shinn AK, Ono KE, McKinley SK, Ely AV, et al. (2012). Reduced sleep spindles and spindle coherence in schizophrenia: Mechanisms of impaired memory consolidation? Biological Psychiatry, 71(2), 154–161. 10.1016/j.biopsych.2011.08.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  432. Wang HE, Campbell-Sills L, Kessler RC, Sun X, Heeringa SG, Nock MK, et al. (2019). Pre-deployment insomnia is associated with post-deployment post-traumatic stress disorder and suicidal ideation in US Army soldiers. Sleep, 42(2), 1–9. 10.1093/sleep/zsy229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  433. Wang YQ, Liu WY, Li L, Qu WM, & Huang ZL (2021). Neural circuitry underlying REM sleep: A review of the literature and current concepts. Progress in Neurobiology, 204, 102106. 10.1016/j.pneurobio.2021.102106. [DOI] [PubMed] [Google Scholar]
  434. Wang D, & Teichtahl H (2007). Opioids, sleep architecture and sleep-disordered breathing. Sleep Medicine Reviews, 11(1), 35–46. 10.1016/j.smrv.2006.03.006. [DOI] [PubMed] [Google Scholar]
  435. Wang H, & Zhuo M (2012). Group I metabotropic glutamate receptor-mediated gene transcription and implications for synaptic plasticity and diseases. Frontiers in Pharmacology, 3, 1–8. 10.3389/fphar.2012.00189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  436. Watanabe M, Nakamura M, Sato K, Kano M, Simon MI, & Inoue Y (1998). Patterns of expression for the mRNA corresponding to the four isoforms of phospholipase Cβ in mouse brain. European Journal of Neuroscience, 10(6), 2016–2025. 10.1046/j.1460-9568.1998.00213.x. [DOI] [PubMed] [Google Scholar]
  437. Watson CJ, Lydic R, & Baghdoyan HA (2011). Sleep duration varies as a function of glutamate and GABA in rat pontine reticular formation. Journal of Neurochemistry, 118(4), 571–580. 10.1111/j.1471-4159.2011.07350.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  438. Weber F, & Dan Y (2016). Circuit-based interrogation of sleep control. Nature, 538(7623), 51–59. 10.1038/nature19773. [DOI] [PubMed] [Google Scholar]
  439. Weigend S, Holst SC, Treyer V, O’Gorman Tuura RL, Meier J, Ametamey SM, et al. (2019). Dynamic changes in cerebral and peripheral markers of glutamatergic signaling across the human sleep-wake cycle. Sleep, 42(11), 1–11. 10.1093/sleep/zsz161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  440. Welsch L, Bailly J, Darcq E, & Kieffer BL (2020). The negative affect of protracted opioid abstinence: Progress and perspectives from rodent models. Biological Psychiatry, 87(1), 54–63. 10.1016/j.biopsych.2019.07.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  441. Weng YY, Lei X, & Yu J (2020). Sleep spindle abnormalities related to Alzheimer’s disease: A systematic mini-review. Sleep Medicine, 75, 37–44. 10.1016/j.sleep.2020.07.044. [DOI] [PubMed] [Google Scholar]
  442. Westermeyer J, Khawaja IS, Freerks M, Sutherland RJ, Engle K, Johnson D, et al. (2010). Quality of sleep in patients with posttraumatic stress disorder. Psychiatry (Edgemont), 7(9), 21–27. [PMC free article] [PubMed] [Google Scholar]
  443. Wichniak A, Antczak J, Wierzbicka A, & Jernajczyk W (2002). Alterations in pattern of rapid eye movement activity during REM sleep in depression. Acta Neurobiologiae Experimentalis, 62(4), 243–250. [DOI] [PubMed] [Google Scholar]
  444. Wichniak A, Wierzbicka A, & Jernajczyk W (2013). Sleep as a biomarker for depression. International Review of Psychiatry, 25(5), 632–645. 10.3109/09540261.2013.812067. [DOI] [PubMed] [Google Scholar]
  445. Wichniak A, Wierzbicka A, Walęcka M, & Jernajczyk W (2017). Effects of antidepressants on sleep. Current Psychiatry Reports, 19(9), 1–7. 10.1007/s11920-017-0816-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  446. Williams JA, Comisarow J, Day J, Fibiger HC, & Reiner PB (1994). State-dependent release of acetylcholine in rat thalamus measured by in vivo microdialysis. The Journal of Neuroscience: The Official Journal of the Society for Neuroscience, 14(9), 5236–5242. 10.1523/JNEUROSCI.14-09-05236.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  447. Winkelman JW, & de Lecea L (2020). Sleep and neuropsychiatric illness. Neuropsychopharmacology, 45(1). 10.1038/s41386-019-0514-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  448. Winship IR, Dursun SM, Baker GB, Balista PA, Kandratavicius L, Maiade-Oliveira JP, et al. (2019). An overview of animal models related to Schizophrenia. Canadian Journal of Psychiatry, 64(1), 5–17. 10.1177/0706743718773728. [DOI] [PMC free article] [PubMed] [Google Scholar]
  449. Witkin JM, Monn JA, Schoepp DD, Li X, Overshiner C, Mitchell SN, et al. (2016). The rapidly acting antidepressant ketamine and the mGlu2/3 receptor antagonist LY341495 rapidly engage dopaminergic mood circuits. Journal of Pharmacology and Experimental Therapeutics, 358(1), 71–82. 10.1124/jpet.116.233627. [DOI] [PubMed] [Google Scholar]
  450. Wolf ME (2016). Synaptic mechanisms underlying persistent cocaine craving. Nature Reviews Neuroscience, 17(6), 351–365. Nature Publishing Group; 10.1038/nrn.2016.39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  451. Wong B, Zimbelman AR, Milovanovic M, Wolf ME, & Stefanik MT (2022). GluA2-lacking AMPA receptors in the nucleus accumbens core and shell contribute to the incubation of oxycodone craving in male rats. Addiction Biology, 27(6), e13237. 10.1111/adb.13237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  452. Wood CM, Wafford KA, McCarthy AP, Hewes N, Shanks E, Lodge D, et al. (2018). Investigating the role of mGluR2 versus mGluR3 in antipsychotic-like effects, sleep-wake architecture and network oscillatory activity using novel Han Wistar rats lacking mGluR2 expression. Neuropharmacology, 140, 246–259. 10.1016/j.neuropharm.2018.07.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  453. Woodward SH, Leskin GA, & Sheikh JI (2002). Movement during sleep: Associations with posttraumatic stress disorder, nightmares, and comorbid panic disorder. Sleep, 25(6), 681–688. 10.1093/sleep/25.6.669. [DOI] [PubMed] [Google Scholar]
  454. Wright RA, Arnold MB, Wheeler WJ, Ornstein PL, & Schoepp DD (2001). [3H]LY341495 binding to group II metabotropic glutamate receptors in rat brain. The Journal of Pharmacology and Experimental Therapeutics, 298(2), 453–460. [PubMed] [Google Scholar]
  455. Wulff K, Gatti S, Wettstein JG, & Foster RG (2010). Sleep and circadian rhythm disruption in psychiatric and neurodegenerative disease. Nature Reviews Neuroscience, 11(8), 589–599. 10.1038/nrn2868. [DOI] [PubMed] [Google Scholar]
  456. Xie L, Kang H, Xu Q, Chen M, Liao Y, Thiyagarajan M, et al. (2013). Sleep drives metabolite clearance from the adult brain. NIH, 342(6156). 10.1126/science.1241224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  457. Yeh MSL, Poyares D, Coimbra BM, Mello AF, Tufik S, & Mello MF (2021). Subjective and objective sleep quality in young women with posttraumatic stress disorder following sexual assault: A prospective study. European Journal of Psychotraumatology, 12-(1). 10.1080/20008198.2021.1934788. [DOI] [PMC free article] [PubMed] [Google Scholar]
  458. Yokoi M, Kobayashi K, Manabe T, Takahashi T, Sakaguchi I, Katsuura G, et al. (1996). Impairment of hippocampal mossy fiber LTD in mice lacking mGluR2. Science (New York, N.Y.), 273(5275), 645–647. 10.1126/science.273.5275.645. [DOI] [PubMed] [Google Scholar]
  459. Yoo S-S, Gujar N, Hu P, Jolesz FA, & Walker MP (2007). The human emotional brain without sleep—A prefrontal amygdala disconnect. Current Biology: CB, 17(20), R877–R878. 10.1016/j.cub.2007.08.007. [DOI] [PubMed] [Google Scholar]
  460. Yu X, Li W, Ma Y, Tossell K, Harris JJ, Harding EC, et al. (2019). GABA and glutamate neurons in the VTA regulate sleep and wakefulness. Nature Neuroscience, 22(1), 106–119. 10.1038/s41593-018-0288-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  461. Zarate CA, Singh JB, Carlson PJ, Brutsche NE, Ameli R, Luckenbaugh DA, et al. (2006). A randomized trial of an N-methyl-D-aspartate antagonist in treatment-resistant major depression. North, 63(8), 793–802. [DOI] [PubMed] [Google Scholar]
  462. Zhai J, Tian MT, Wang Y, Yu JL, Köster A, Baez M, et al. (2002). Modulation of lateral perforant path excitatory responses by metabotropic glutamate 8 (mGlu8) receptors. Neuropharmacology, 43(2), 223–230. 10.1016/S0028-3908(02)00087-4. [DOI] [PubMed] [Google Scholar]
  463. Zhang Y, Ren R, Yang L, Zhang H, Shi Y, Okhravi HR, et al. (2022). Sleep in Alzheimer’s disease: A systematic review and meta-analysis of polysomnographic findings. Translational Psychiatry, 12(1). 10.1038/s41398-022-01897-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  464. Zhao W, van Someren EJW, Li C, Chen X, Gui W, Tian Y, et al. (2021). EEG spectral analysis in insomnia disorder: A systematic review and meta-analysis. In Vol. 59. Sleep Medicine Reviews W.B. Saunders Ltd. 10.1016/j.smrv.2021.101457. [DOI] [PubMed] [Google Scholar]
  465. Zhu H, Rockhold RW, & Ho IK (1998). The role of glutamate in physical dependence on opioids. Japanese Journal of Pharmacology, 76(1), 1–14. 10.1254/jjp.76.1. [DOI] [PubMed] [Google Scholar]

RESOURCES