Glucocorticoid signalling coordinating circadian rhythms between the glymphatic system and sleep contributes to brain health
Sections
Open Access Review
Glucocorticoid signalling coordinating circadian rhythms between the glymphatic system and sleep contributes to brain health

Affiliation:

Laboratory for Neurophysiology, Department of Cell and Chemical Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands

ORCID: https://orcid.org/0009-0000-3570-4153

Laura C. A. van der Zwet

Affiliation:

Laboratory for Neurophysiology, Department of Cell and Chemical Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands

ORCID: https://orcid.org/0000-0001-5506-5037

Stephan Michel

Affiliation:

Laboratory for Neurophysiology, Department of Cell and Chemical Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands

Email: t.de_boer@lumc.nl

ORCID: https://orcid.org/0000-0002-6402-6248

Tom Deboer
*

Explor Endocr Metab Dis. 2025;2:101437 DOl: https://doi.org/10.37349/eemd.2025.101437

Received: March 18, 2025 Accepted: May 28, 2025 Published: July 15, 2025

Academic Editor: Charlotte Steenblock, University Clinic Carl Gustav Carus, Germany

The article belongs to the special issue The HPA Axis in Health and Disease

Abstract

Circadian rhythms are present in almost every cell of the body and play important roles in various physiological processes. The central circadian clock in the suprachiasmatic nucleus (SCN) is synchronized to the environmental light-dark cycle and ensures a temporal order for the peripheral clocks, which in turn modulate tissue and organ function. This temporal organization is crucial for the precise timing of bodily processes, including sleep, glucocorticoid release, and the function of the glymphatic system. Sleep and the glymphatic system are significantly impacted by the rhythmic secretion of glucocorticoids. One important function of the glymphatic system is the clearance of waste metabolites, which most likely happens during sleep. Disruptions within the SCN, glucocorticoid rhythms, sleep, or glymphatic clearance have been implicated in compromised brain health. This review explores the current knowledge on the interdependence of the SCN, glucocorticoids, sleep, and the glymphatic system, and emphasizes their importance in homeostasis and pathology; in particular, Alzheimer’s disease.

Keywords

Circadian rhythms, suprachiasmatic nucleus, glucocorticoids, glymphatic system, clearance, choroid plexus, sleep, Alzheimer’s disease

Introduction

Life has evolved under cyclic environmental conditions, driving organisms to adapt their active period to align with the 24-hour light-dark cycle. As a result, most physiological and biochemical processes, like body temperature, blood flow, and hormone release, are highly regulated and optimized throughout the day [1]. Importantly, these rhythms persist in the absence of external time cues and, since they have a period of approximately but not exactly 24 hours, are termed circadian rhythms. These circadian rhythms are present in nearly every tissue and organ system and are controlled by molecular circadian clocks. To align or entrain all these internal biological clocks to the external light-dark cycle and to maintain the right timing between physiological processes, mammals rely on a central circadian clock in the suprachiasmatic nucleus (SCN), which is located in the hypothalamus [2]. The SCN receives photic input from specialized ganglion cells in the retina to entrain its own circadian rhythm to the light-dark cycle, and it makes use of several neuronal and hormonal pathways to convey this timing information to peripheral tissues (Figure 1). This entrainment ensures that physiological processes occur at optimal times for performance and health.

Overview of the circadian system. Upper: the suprachiasmatic nucleus (SCN), located in the hypothalamus, receives light information from specialized retinal ganglion cells to entrain its circadian rhythm to the external light-dark cycle. The electrical activity of the SCN is highest when there is light input. Lower: the SCN entrains peripheral clocks in structures like the choroid plexus (ChP) (red), lungs, heart, liver, adrenal gland, and muscles through body temperature rhythms, autonomic nervous system signals, and hormones. Peripheral clocks can also be entrained by external zeitgebers, such as exercise (muscle) and nutrient intake (liver). Reciprocally, peripheral clocks interact with the SCN. Created in BioRender. van der Zwet, L. (2025) https://BioRender.com/2t7oo3q

One of the systemic signals that is regulated by the SCN and helps to keep temporal order in physiological processes is the rhythmic release of glucocorticoids (GCs) from the adrenal gland cortex. In humans, GC levels peak at the beginning of the day, likely to anticipate and prepare the body for a period of wakefulness. In nocturnal mice, the GC level also peaks shortly before the active period, and it has been shown that this circadian rhythm in GCs is dependent on a functional SCN [3]. GCs will also cross the blood-brain barrier in a controlled way [4] and modulate a multitude of brain functions.

One brain area under the control of circadian rhythmicity and influenced by GC is the choroid plexus (ChP) [5, 6]. This epithelial tissue lines the walls of the ventricles and is the main source of cerebrospinal fluid (CSF) in the brain, an important component of fluid homeostasis in the brain [7]. The produced CSF eventually moves through the glymphatic system, a network of perivascular spaces that facilitate directional motion of fluid in the brain. This CSF movement is thought to be primarily important for clearing waste metabolites and proteins [8, 9]. In addition, the glymphatic system seems to distribute compounds important for signalling and modulating brain function [8]. The cyclic signals released by the ChP may therefore be used to synchronize circadian rhythms of cellular processes in different brain areas [5, 10, 11]. Recent research has highlighted the connections between the SCN, GCs, and the glymphatic system. For instance, Drapšin et al. [5] demonstrated that disruption of the SCN affects circadian GC regulation and that the ChP’s internal circadian clock is highly sensitive to GCs [6].

Glymphatic influx produced by the ChP and waste clearance are thought to reach peak efficacy during sleep [12], which in turn is also modulated by the central circadian clock. GCs, among others, can influence sleep patterns, and sleep disturbances are commonly reported as side effects by patients treated with exogenous GCs, as well as by patients with Cushing’s syndrome, which is characterized by excessive cortisol production [13]. As the efficacy of the glymphatic system has been associated with sleep in humans [14], sleep-wake cycles may be an important link in the interplay between the SCN, GC release, and the glymphatic system. Disruptions in the crosstalk between these systems may contribute to the etiopathology and pathogenesis of neurodegenerative disease, where less efficient clearing by the glymphatic system may lead to increased deposition of tau, α-synuclein, and amyloid-β, leading to neurological diseases like Parkinson’s and Alzheimer’s disease (AD), highlighting the importance of further investigation. However, a comprehensive review illustrating and critically analysing these interactions has not yet been presented.

So, while the elements of brain homeostasis in health and disease are being thoroughly investigated, the interactions between them, and particularly the influence of the circadian system as a potential regulator and at the same time therapeutic target, receive less attention. Therefore, this review aims to explore the interactions between the SCN and circadian rhythms, GCs, sleep, and the glymphatic system. The first section examines the SCN and its influence on sleep. The second section addresses circadian GC release and explores mechanisms by which GCs affect peripheral clocks. In the third section, the focus will shift to the ChP and the glymphatic system, linking prior sections to discuss how circadian and sleep-driven influences optimize glymphatic clearance. The final section synthesizes the connection between the SCN, GCs, sleep, and the glymphatic system and examines how dysregulation of these systems may contribute to neurodegenerative processes.

The circadian system

Circadian rhythms in mammals are coordinated by the SCN, a hypothalamic bilateral nucleus located just above the optic chiasm [15]. The SCN receives and processes photic information, which it uses to convey external time information to peripheral clocks, thereby synchronizing them to the environment [16]. The next section will examine the SCN and its interactions with sleep.

The SCN: a central clock

In the SCN, as well as in most peripheral cells, a transcriptional-translational feedback loop (TTFL) occurs in the expression of a handful of clock genes, which lasts ~24 hours [17]. The TTFL consists of a positive and negative feedback loop (Figure 2). In the negative feedback loop, a heterodimer composed of BMAL1 and CLOCK or NPAS2 [18] binds to the E-box of three different period (Per) genes and two different cryptochrome (Cry) genes. After translation and post-translational modification, PER and CRY proteins translocate back into the nucleus, where CRY interacts with the CLOCK-BMAL1 heterodimer to inhibit its own transcription. As a result of this negative feedback loop, Bmal1 mRNA reaches peak levels twelve hours in antiphase with Per and Cry mRNA levels [2].

Simplified schematic representation of the transcriptional-translational feedback loop. The CLOCK-BMAL1 heterodimer activates transcription of several clock genes (Cry, Per, Rev-Erbα) by docking onto an E-box, a dimer of CRY and PER, in turn, inhibits CLOCK-BMAL1 heterodimer formation. Created in BioRender. van der Zwet, L. (2025) https://BioRender.com/klj0tg1

Besides Per and Cry, the CLOCK-BMAL1 heterodimers activate Rev-Erbα expression, starting the positive feedback loop. REV-ERBα represses the transcription of Bmal1. Therefore, when CRY inhibits the CLOCK-BMAL1 heterodimer, it also inhibits the expression of Rev-Erbα. This inhibition leads to the de-repression—thus activation—of Clock and Bmal1 transcription and resets the TTFL [2].

The TTFL does not complete in exactly 24 hours. Therefore, the SCN receives afferent information from external zeitgebers to synchronize its clock gene transcription to the time of day [19]. This entrainment allows the SCN to convey accurate temporal cues, in synchrony with the environment, to peripheral clocks.

Light is one of the most potent zeitgebers for the SCN [20]. A subset of intrinsically photosensitive retinal ganglion cells (ipRGCs) expresses melanopsin, a photosensitive pigment. When these ipRGCs detect light or receive photic information from photoreceptor cells in the retina, they process and relay this information to the SCN through the retinohypothalamic tract [2123]. Glutamate, which is the main neurotransmitter of this tract, stimulates NMDA receptors and consequently L-type voltage-dependent Ca2+ channels on the SCN neurons, leading to an increase in intracellular calcium concentration ([Ca2+]i) [2, 2426]. This increased [Ca2+]i can adjust transcription of the clock genes in the TTFL by activating the cAMP response element-binding protein (CREB)/CRE transcriptional pathway [27]. Besides photic input, non-photic input like behavioural activity is also known to be able to modulate the SCN phase [28, 29].

The SCN has historically been considered as the ‘master oscillator’ that forces circadian timing on ‘slave’ peripheral clocks [2]. However, recent perspectives, like that of Starnes and Jones [30], are more nuanced, proposing that interactions between the SCN and peripheral clocks are often bidirectional. Moreover, while some peripheral clocks need constant SCN input, others only require periodic cues to align their rhythm to the environment.

Two-process model of sleep regulation

The two-process model of sleep regulation [31, 32] is often used to explain the timing of sleep within a circadian framework. It describes the interaction between two regulatory processes of sleep: a circadian mechanism (process C) and a sleep-dependent homeostatic mechanism (process S). Process S, which represents the sleep pressure, accumulates during wakefulness and decreases during sleep. Process S is thought to be reflected in the activity of the slow waves in the non-rapid eye movement (NREM) sleep electroencephalogram (EEG). Process C is responsible for sleep/wake initiation and is controlled by the SCN [31]. Sleep onset occurs when the rising process S passes the upper sinusoidal sleep-initiating threshold of process C. During sleep, process S decreases until it intersects with the lower wake-inducing threshold of process C, and wakefulness is initiated. While processes S and C work independently, sleep is optimal when the homeostatic drive for sleep is aligned with the circadian timing of sleep [33, 34].

This entanglement of sleep homeostatic and sleep-regulating circadian processes makes that sleep can be changed by factors that influence the clock. Exposure to light during the night can delay or advance the phase of the circadian clock depending on the timing of light exposure. Light at the beginning of the subjective night will cause a phase delay, while light at the beginning of the subjective day causes a phase advance [20]. Consequently, looking at the model, the sleep homeostatic process S might intersect sooner or later during the day with the thresholds of process C, thus resulting in advanced or delayed sleep onset, respectively.

The flip-flop switch and orexin

The eventual transition from wakefulness to sleep and vice versa depends on a bistable mutually inhibitory system. During wakefulness, arousal-promoting regions in the reticular formation and posterior hypothalamus, like the Raphe nucleus, Locus Coeruleus, and the lateral hypothalamus (LH), are active [3538]. These centres, collectively known as the ascending reticular activation system, inhibit the sleep-promoting neurons in the ventrolateral preoptic nucleus (VLPO) of the hypothalamus [39]. However, when the VLPO is more active, it inhibits the arousal-promoting regions using GABA and rapidly induces sleep [40]. This ‘flip-flop switch’, as it was named by Saper et al. [41], efficiently promotes fast switching between sleep and wakefulness and prevents intermediary states.

The flip-flop switch is thought to be stabilized by orexin, a neuropeptide produced in the LH. Orexin stimulates the arousal-promoting regions, thus reinforcing wakefulness and biasing the switch against sleep. This wake-promoting action is counteracted by the VLPO, which, when active, inhibits the orexin neurons in the LH [42].

The SCN plays an important role in sleep-wake transitions, but has only a few direct neuronal projections to the VLPO or orexin neurons [4345]. Instead, the SCN influences these areas through an indirect, multi-synaptic pathway. The SCN projects to the subparaventricular zone [45, 46], which subsequently relays to the dorsomedial nucleus of the hypothalamus (DMH) [47]. The DMH has extensive connections to the VLPO and orexin neurons [43, 44] and is essential in relaying SCN information to the sleep-wake regulating network [48].

During the day, when SCN neuronal activity is high, the human SCN is thought to indirectly inhibit the VLPO and stimulate orexin production [49, 50] (the opposite then is thought to occur in nocturnal rodents). Both mechanisms drive the switch toward the waking state. By the end of the day, when SCN activity decreases and sleep pressure is highest, the VLPO is activated and sleep is initiated.

Sleep architecture

Sleep consists of two distinct states: rapid eye movement (REM) sleep and NREM sleep. 75% of sleep is spent in NREM sleep, during which the parasympathetic system is most active, leading to decreased heart and respiration rates [51, 52]. During REM sleep, the activity of the sympathetic nervous system dominates, and respiration and heart rate are enhanced [51, 52]. The duration of this ‘dreaming’ state increases through the night and occupies approximately 25% of total sleep [53]. Sleep onset typically begins with a period of NREM sleep, which ends in wakefulness or REM sleep. Humans usually experience 4 to 5 cycles of alternating NREM and REM states during the night [53, 54].

Previously, we have demonstrated that REM sleep significantly increases SCN neuronal activity, and that this enhancement decreases at the end of REM sleep. This effect was observed independent of the circadian phase, and SCN neuronal activity was shown to correlate with EEG slow wave activity [55]. Therefore, the interactions between the SCN and sleep homeostatic mechanisms appear to be bidirectional.

The role of glucocorticoids in circadian timing

Besides timing sleep/wake cycles, the SCN regulates the hypothalamic-pituitary-adrenal axis (HPA-axis), which controls GC production. Although GCs like cortisol are commonly known as stress hormones, they are also of vital importance in non-stress-related physiology. For example, GCs maintain normal metabolism and have potent anti-inflammatory and immunosuppressive properties [5659]. Furthermore, their rhythmic release can synchronize circadian rhythms of peripheral clocks [60, 61]. In the following part of the review, the mechanisms and patterns of GC release will be explored. Then, the influence of the SCN on the HPA-axis will be discussed, followed by a description of GC effects on peripheral clocks.

Mechanisms and patterns of glucocorticoid release

GCs are produced from cholesterol in the zona fasciculata of the adrenal gland cortex through a series of enzymatic conversions [62]. The regulation of GC synthesis and secretion starts in the paraventricular nucleus (PVN), which releases corticotropin-releasing hormone (CRH) into the median eminence either in response to stress or the circadian clock. In the anterior pituitary, CRH binds to its G-protein coupled receptor (GPCR) on corticotropic cells, activating Gαs and raising cAMP concentration, which ultimately causes exocytosis of adrenocorticotropic hormone (ACTH) into the bloodstream [63, 64]. When ACTH binds to its GPCR in the zona fasciculata of the adrenal cortex, intracellular movement of steroid precursors between cytosolic stores and mitochondria is accelerated, and the production of GCs is increased [62]. In humans, the most important GC hormone is cortisol, while corticosterone is the main GC in rodents [65].

To prevent excessive GC production and optimize secretion in unstressed situations, cortisol/corticosterone exerts negative feedback on the PVN and the anterior pituitary [66]. Research in rodents has revealed two types of corticosterone-related negative feedback: a fast nongenomic mechanism and a slower genomic mechanism. The nongenomic mechanism involves GC-mediated suppression of CRH and ACTH release by the PVN and anterior pituitary, respectively [66]. Studies have demonstrated that bilateral adrenalectomy in rats rapidly increases circulating ACTH levels and CRH immunoreactivity in the PVN [67, 68]. Levin et al. [69] add to this observation by demonstrating that exogenously administered corticosterone acts at the PVN to decrease ACTH levels in adrenalectomized rats. Lesioning the PVN before administering corticosterone, in accordance with the proposed mechanism, increases plasma ACTH levels [70].

The genomic mechanism suppresses CRH gene expression and pro-opiomelanocortin—the precursor to ACTH—transcription in pituitary corticotroph cells [66]. The exact mechanisms behind genomic suppression are largely unknown [66], but several rodent studies have shown that GCs inhibit pro-opiomelanocortin gene expression [7173].

CRH is secreted in an ultradian rhythm. The amplitude and frequency of these ultradian peaks are determined by the phase of the circadian rhythm in GC secretion [74]. GC levels are highest just before an animal’s active period, which is early morning for diurnal species and early evening for nocturnal species [7578]. This latter GC peak may permit the body to respond and prepare effectively for physical stressors during the active period [79]. On top of the ultradian rhythm, stressors like hypovolemia or fear can cause acute (~1 hour) peaks in GC release by enhancing the PVN’s CRH secretion [80].

Interactions between the SCN and HPA-axis

Circadian control of the HPA-axis

The SCN is well-established as the regulator of the circadian rhythm in GC release, as studies have consistently demonstrated that lesions to the SCN and exposure to constant light result in the loss of circadian GC rhythmicity [8184]. Despite this knowledge, the precise mechanisms underlying the SCN’s involvement in GC production remain unclear [85].

Several studies with different neuronal tracers have established connectivity between the SCN and the nuclei of the HPA-axis. All showed that the SCN has projections to the PVN/DMH, although the density of these projections remains a topic of debate [45, 46, 86, 87]. Moreover, these PVNCRH neurons show circadian rhythms in clock gene expression and neuronal activity, coordinating peak CRH release around waking [88]. Deletion of BMAL1 in PVNCRH neurons results in irregular GC secretion. Besides clock gene expression, circadian GC release also depends on SCN input to the PVNCRH neurons with inhibitory vasoactive intestinal peptide. GC release rhythms are lost without SCN input [88], thus affirming the tight causal link between the SCN and PVN in regulating circadian GC rhythms.

As some projections from the SCN use arginine vasopressin (AVP) as their neurotransmitter [46, 89], Kalsbeek et al. [90] set out to investigate the effects of AVP on corticosterone release in rats. Following bilateral lesioning of the SCN, leaving the animal arrhythmic, they implanted cannulas aimed at the PVN/DMH area. After perfusing different AVP agonists and antagonists into the PVN/DMH area, blood samples were taken at different time points. The results indicate that AVP inhibits corticosterone release in nocturnal rodents; administration of AVP suppressed corticosterone levels, while AVP antagonists stimulated corticosterone release [90]. The experiment was repeated in the diurnal rodent Arvicanthis ansorgei, which, in contrast to the nocturnal rat, responded to AVP with an elevation in corticosterone levels [91]. These results suggest that the SCN modulates the PVN/DMH induced GC release with AVP differentially in diurnal and nocturnal animals, possibly relating to the difference in temporal patterns in rhythms of GC levels.

Control over rhythmic GC secretion by the SCN seems to involve additional pathways independent of the hypothalamus and ACTH. Studies of ACTH and GC levels have revealed that while GC levels rise drastically before the active period, this is accompanied by only a minor and often insignificant rise in ACTH levels [9294]. The existence of an ACTH-independent pathway is supported by findings that hypophysectomised rats, implanted with constantly releasing ACTH- and thyroxine pellets to maintain basal adrenal function, still display rhythmic corticosterone release [95].

Splanchnic nerve modulation

An alternative pathway suggested for HPA-axis-independent control of GC secretion by the SCN is through the splanchnic nerves [85]. As part of the sympathetic autonomic nervous system, the splanchnic nerves facilitate communication between the central nervous system and visceral organs, like the adrenal gland [96]. The evidence for SCN-controlled GC release through the splanchnic nerve involves two key mechanisms: (1) photic input can modulate GC release through the sympathetic innervation of the adrenal gland, and (2) this sympathetic innervation can modulate adrenal sensitivity to ACTH [85, 97, 98] (Figure 3).

Interactions between the SCN and the HPA-axis. Photic input can modulate the sensitivity of the adrenal cortex to adrenocorticotropic hormone (ACTH) through the splanchnic nerve. Using arginine vasopressin (AVP) as a neurotransmitter, the suprachiasmatic nucleus (SCN) modulates paraventricular nucleus (PVN)/dorsomedial nucleus of the hypothalamus (DMH)-induced glucocorticoid (GC) release. Circadian timing of GC release also depends on the internal molecular clock of the adrenal cortex. Created in BioRender. van der Zwet, L. (2025) https://BioRender.com/ja7kadf

Niijima et al. [97] demonstrated that photic stimuli can alter the activity of the adrenal branch of the splanchnic nerve in rats during the active period. Following light exposure, they observed an increased discharge rate in the adrenal nerve, which was not present in rats with bilateral SCN lesions. Although their very limited sample size of SCN-lesioned animals calls for cautious interpretation, the results suggest that the modulation of the splanchnic nerve activity by photic information is mediated by the SCN [97]. Building on these results, Ishida et al. [98] later showed that photic stimulation in mice during the active period not only elevates the discharge rate in the splanchnic nerve but also rapidly increases corticosterone levels. The absence of a corresponding rise in ACTH reinforces the notion that the hypothalamus and pituitary are not involved in this pathway. Moreover, as their SCN-lesioned mice and mice without splanchnic nerves did not display increased corticosterone levels in response to photic stimuli, Ishida et al. [98] confirm the role of the SCN and splanchnic nerve in light-evoked GC rises.

The GC rise following splanchnic nerve stimulation may be attributed to the role of the splanchnic nerve in modulating adrenal sensitivity to ACTH, a mechanism that has been demonstrated across various animal models [99101]. The sensitivity of the adrenal gland to ACTH is also modulated by a circadian clock. Adrenal sensitivity in rats peaks during their active (dark) phase, while it is low during their inactive (light) phase [94, 102]. Lesioning the SCN eliminates these day/night differences in ACTH sensitivity [103]. Notably, transection of the splanchnic nerve attenuates corticosterone levels in rodents without affecting ACTH levels [101], suggesting that the splanchnic nerve indeed sensitizes the adrenal gland to ACTH.

Altering the activity of the splanchnic nerve, along with direct control of the HPA-axis through the PVN/DMH, allows the SCN to utilize both neural and hormonal signals to fine-tune adrenal responses depending on time of day. However, some doubts remain about the control of the adrenal gland by the SCN. For instance, rats with bilateral SCN lesions still show rhythmic corticosterone secretion [104], and time-restricted feeding produces two GC release peaks, of which only one is SCN-dependent [105]. These results indicate that there are extra-SCN processes that can mediate GC secretion.

It is important to recognize that most cells possess intrinsic molecular clocks, including those in the adrenal gland [106], and can maintain their own autonomous rhythm. Therefore, the main task of the SCN is to synchronize the peripheral rhythms with external stimuli such as light. For example, in the adrenal gland, the CLOCK:BMAL1 heterodimer keeps Steroidogenic Acute Regulatory protein—the rate-limiting enzyme in GC synthesis—under transcriptional control [106]. The expression of this enzyme is unaffected by splanchnic nerve transection [101], demonstrating that the adrenal gland can independently regulate circadian GC production through its molecular clock as well.

Glucocorticoid control of molecular clocks

The earlier presented evidence shows that the SCN and the adrenal molecular clock can regulate GC secretion. GCs themselves appear to act as a mediator to align the rhythms of peripheral clocks to that of the SCN. For instance, when rats are given dexamethasone—a potent GC receptor (GR) agonist—in constant darkness, this injection acts as a zeitgeber for the regulation of intraperitoneal temperature [107]. Furthermore, GC rhythms play a role in coordinating day-night cycles of bladder capacity [108], heart function [109], and metabolism [110].

GCs exert their effects through binding to the GR, a nuclear receptor known as NR3C1. When bound, the complex migrates from the cytoplasm to the nucleus, where it functions as a stimulatory transcription factor by binding to GC response elements (GREs) in the promotor of certain genes. Clock genes Per1 and Per2 were found to contain GREs [111], suggesting a direct impact on the phase of the peripheral molecular clock by GCs. Therefore, it is not surprising that the genes integral to circadian rhythm regulation are particularly sensitive to the loss of GR function, as found in zebrafish models [112].

Interestingly, studies have shown that clock proteins can interact with the GR to alter its function. For example, REV-ERBα affects the subcellular localization of the GR [113], and CLOCK-BMAL1 can acetylate the GR, thereby inhibiting its activity [114]. CRY1 and 2 also seem to oppose the actions of the GR [115]. These mechanisms might help with adjusting the phase of peripheral clocks to that of the external environment. For instance, when GC levels are acutely elevated due to stress, they can disrupt the normal synchronization of peripheral clocks to the SCN [116]. By inhibiting the GR through e.g., CLOCK-BMAL1, unwanted desynchronisation can be mitigated [114].

The GR is expressed in many peripheral cells and in the brain, but has not been found in adult SCN neurons [61, 117]. This finding suggests that GC levels do not influence the rhythmic activity of SCN neurons. However, GRs have been identified in the foetal/neonatal SCN, gradually disappearing as gestation progresses [118]. As maternal GCs can pass the placenta [119], they can entrain the foetal SCN to the maternal circadian rhythm by acting at the GRs [120].

In contrast to adult SCN neurons, GRs were found in the glial cells associated with the SCN and PVN [121, 122]. These glial cells actively control SCN activity by regulating extracellular GABA and glutamate concentrations [123]. GCs have been shown to stimulate the expression of glial fibrillary acidic protein (GFAP), an integral component of glial filaments, in SCN and PVN-associated glial cells [122]. This upregulation suggests a role for GCs in modulating glial plasticity, possibly impacting the SCN network.

Glucocorticoids influence sleep

Stress has profound effects on sleep/wake behaviour, often altering sleep architecture and increasing arousal. In animal models, acute stress and hydrocortisone administration have been shown to suppress sleep [124]. Moreover, conditions characterized by excessive cortisol production, like Cushing’s syndrome, are often associated with sleep disturbances [13]. Chronic corticosterone administration in rodents has been shown to disrupt sleep by influencing the brain regions involved in sleep initiation and/or wake maintenance; corticosterone activates noradrenergic neurons in the locus coeruleus, one of the arousal-promoting areas, and inhibits the GABAergic neurons of the VLPO, thereby promoting wakefulness. These GC-induced sleep disruptions are mediated through the GR, as a GR antagonist can reverse the sleep disturbances [125].

The glymphatic system

The distribution of temporal information from the central clock has been described above using mainly neuronal pathways. However, the SCN also produces and releases a number of neuropeptides, which help to regulate circadian rhythms in physiology and behaviour [126]. The released signals from the SCN can reach peripheral clocks through a specialized vascular circulation pathway to the lamina terminalis, which has recently been described to function as a portal vein [127]. Additionally, the CSF may take up and distribute signalling molecules from the SCN through a system of perivascular channels known as the glymphatic system (Figure 4) [128]. The main function of the glymphatic system was suggested to be the clearance of waste from the brain, the failure of which supposedly contributes to neurological diseases like AD [129]. The SCN, sleep, and GC signalling all seem to influence the glymphatic system, which immediately raises the question about the role of temporal alignment for the proper functioning of the glymphatic system. This part will focus on the function of the glymphatic system and the circadian rhythmicity of an important component, the ChP. Subsequently, the influence of GCs, circadian rhythms and sleep on the glymphatic system will be discussed.

Glymphatic clearance in the brain. Cerebrospinal fluid (CSF) in perivascular spaces enters the brain parenchyma through aquaporin-4 (AQP4) channels located on the endfeet of glial cells. Convective flow through the brain parenchyma facilitates the clearance of waste, which is transported to the perivenous space and into the vein toward the lymphatic system. Arrows indicate the movement of fluids and solutes. Created in BioRender. van der Zwet, L. (2025) https://BioRender.com/bhqajuf

To support the weight of the brain and to nourish it with nutrients, growth factors, ions, and peptides, the brain is bathed in CSF [130]. CSF is produced by the ChPs, which are tufts of highly vascularized, villous structures covered by epithelia that line the brain’s ventricles [131, 132]. The epithelial cells in the ChP are connected by tight junctions, thereby creating a blood-CSF barrier and restricting unwanted movement of water-soluble molecules from the systemic blood circulation into the CSF [133]. Due to the tight junctions and adherence junctions, ChP epithelial cells are polarized and can create osmotic gradients [134]. The extensive system of fenestrated capillary loops in the ChP and the osmotic gradient govern the passive ultrafiltration of plasma into the interstitial fluid of the ChP [135, 136]. To secrete the produced fluid into the ventricles, the ChP epithelium contains aquaporins (AQPs), which are water channels [137]. Ions are actively transported into the CSF by ion transporters [138, 139].

However, increasing buoyancy and nourishing the brain are not the only functions of CSF. A study from 2012 by Iliff et al. [128] demonstrated that CSF from the subarachnoid space can enter the brain parenchyma, mix with interstitial fluid, and drain into the peripheral lymphatic system through perivascular spaces and meningeal lymphatic vessels [140]. This CSF efflux depends on astrocytic AQP4 and is likely modulated by circadian rhythms and sleep/wake cycles [12, 141]. As the system seems to have similar functions as the lymphatic system—clearing waste—and depends on glial cells, it was termed the glymphatic system [128]. The glymphatic system has been shown to clear tau, α-synuclein, and amyloid-β, which are proteins associated with neurodegenerative diseases [128, 142, 143].

A waste-clearing system is most efficient if it has directionality. In the glymphatic system, this direction is from the brain parenchyma toward the peripheral lymphatic system. Several different factors contribute to the directionality of CSF movement. For instance, the rhythmic expansion and contraction of the cerebral arteries propels the fluid through the perivascular space [144, 145], while spontaneous changes in the muscle tone of vessels (vasomotion) support steady CSF movement [146, 147]. At the same time, respiratory cycles create pressure changes that further facilitate CSF flow [148].

The glymphatic system can be compared to a closed hydraulic system, as it relies on fluid pressure and directed flow to clear waste efficiently. Just as poking a hole in a hydraulic system would disrupt its function and pressure, any disruptions in the glymphatic system can compromise its ability to clear waste. Uncareful introduction of tracers or cannulas into the glymphatic system can create leaks that disrupt the closed nature of the system and thereby inactivate it [141, 149]. Likewise, perivascular structures collapse and fill with other fluids postmortem, rendering most histological samples of this system useless [145]. These difficulties in maintaining the glymphatic system’s integrity have caused significant discussion about its existence [150]. The most recent research on the glymphatic system therefore increasingly relies on in vivo imaging techniques like diffusion tensor imaging and positron emission tomography, as this can maintain the system’s integrity and function [151, 152].

Besides the glymphatic system, other pathways have been proposed for clearing brain waste. The intramural periarterial drainage theory suggests that perivascular spaces do not exist. Instead, the CSF enters the brain via arterial pial-glial basement membranes, mixes with interstitial fluid, and leaves along the basement membrane of the smooth muscle cells in the artery [153]. Another theory proposes that convective mixing mechanisms drive the efflux of solutes from the brain parenchyma [154]. As the glymphatic pathway is the most widely accepted waste-clearing theory, this review will exclusively focus on this pathway.

Circadian rhythm in the choroid plexus

The ChP exhibits many functional daily rhythms. CSF production, for instance, was observed in a diffusion MRI study to peak during the night in humans [155]. Moreover, the composition of CSF varies across the day, with Na+ concentrations in human subjects reaching peak values throughout the day and reaching lowest levels during the night [156]. These daily fluctuations can be linked to rhythmic gene expression within the ChP. For example, SNAT3 and NCX4, both sodium transporters, show upregulation during the light phase [157], aligning with the diurnal changes in CSF composition. Additional genes showing high circadian expression are related to the endoplasmic reticulum stress response. Transcriptomic data shows that ER stress response markers peak during the late subjective night in the ChP, optimizing the preparation for stress due to the rapid processing and accumulation of secreted proteins, such as transthyretin [11]. This anticipatory upregulation may prevent ER-stress-related cytotoxicity [158] and fine-tune circadian secretion.

Like most cells in the body, the cells in the ChP contain a molecular clock that can coordinate rhythmic gene expression [159]. However, unlike most peripheral clocks, the cells in the ChP can maintain rhythmic expression of clock genes in vitro without input from the SCN. This autonomous rhythmicity has been demonstrated in organotypic explants of the ChP from PER2::LUC mouse models, which showed consistent rhythmic expression over several days [10, 160]. The clock gene expression in the ChP is not only persistent, but can even show stronger oscillations than the SCN itself [10]. This observation is likely explained by the presence of gap junctions between ChP cells, which allow for strong intercellular coupling [161]. This notion is supported by the finding that inhibiting the ChP gap junctions prolongs the circadian period and reduces the PER2::LUC amplitude [10].

Although the SCN appears to be redundant for the rhythmic expression of ChP clock genes in organotypic slices, in vivo studies indicate that the SCN does play a role in sustaining ChP oscillations [11]. Sládek et al. [11] examined transcriptome profiles in control and SCN-lesioned mice, revealing significantly decreased amplitudes in Clock and other rhythmically expressed ChP genes in the SCN-lesioned group. Besides the SCN-to-ChP communication, a coculturing study has found that the ChP also influences the SCN, likely through signalling molecules in the CSF [10]. This reciprocal influence might enhance the synchronization between the SCN and peripheral clocks.

The influence of glucocorticoids on the choroid plexus

While the precise mechanism behind the SCN-to-ChP communication remains unknown, evidence suggests a role for GCs as a mediator. As described before, GC synchronizes circadian clocks through the GR. When GCs are bound to the GR, it will function as a transcription factor for genes with a GRE, like Per1 and Per2 [111]. Cells of the ChP contain GRs and are therefore susceptible to clock modulation by GCs [6, 162]. This modulation has been meticulously investigated by Liška et al. [6] in a series of in vivo and in vitro experiments. Indeed, after adrenalectomy, the rhythmic expression of Per1 and Per2 in the ChP of mice is abolished and significantly dampened, respectively. However, when these mice were given dexamethasone injections, the rhythmicity in clock genes was restored [6]. Dexamethasone was also able to restore rhythmic clock gene expression in SCN-lesioned mice [11]. In vitro, dexamethasone either delayed or advanced the Per2 rhythm in the ChP depending on the timing of administration [6].

Moreover, the ChP expresses 11β-HSD1, which locally catalyses the conversion from inactive GCs to cortisol/corticosterone [163]. 11β-HSD1 has been found to show circadian expression in the rat’s hippocampus with peak levels occurring around activity onset [164]. Through 11β-HSD1, the ChP might adjust the signals from the HPA-axis to maintain circadian CSF homeostasis. However, as there has been no detailed investigation on diurnal oscillations of 11β-HSD1 in the ChP, the functional role of this protein in ChP physiology remains speculative.

The peak efficacy of the glymphatic system: sleep or circadian-dependent?

While the glymphatic system is increasingly being accepted as an integral part of central nervous system physiology, the diurnal rhythm of its clearance function is still under debate. To date, the majority of evidence suggests a higher flow of CSF and clearance during sleep [12, 155, 165, 166]. Since sleep regulation and the central circadian clock are closely intertwined, as described above, it remains unclear whether this increased clearance is sleep-driven or circadian-driven, and studies are needed to distinguish the effect of sleep states from the potentially direct effects of the circadian clock on glymphatic flow.

Mimicking sleep: the use of anaesthetics

Before exploring the efficacy of the glymphatic system in different vigilance states, it is important to discuss the use of anaesthetics. Most animal experiments investigating the interaction between sleep and the glymphatic system rely on anaesthetic drugs to mimic sleep. However, this approach has limitations. First, although some anaesthetics induce EEG patterns that resemble those seen in NREM sleep [167], sleep and anaesthesia remain two distinct states. For instance, anaesthesia fully inhibits locomotion and autonomic reflexes and produces a more extensive disruption of brain connectivity [168]. Second, different anaesthetic drugs were demonstrated to alter the glymphatic system in different ways, potentially introducing confounding factors to the experiments. Dexmedetomidine, propofol, and pentobarbital were shown to increase glymphatic function in rodents [169, 170], while inhaled isoflurane and ketamine inhibit glymphatic function [171]. Both increases and decreases in glymphatic influx have been observed when ketamine is combined with different doses of xylazine [165, 171]. Thus, the effects of anaesthetic drugs on glymphatic function are both drug- and dose-dependent and cannot be directly compared to a natural sleeping state. Anaesthetics can, however, be used to investigate circadian effects by applying them at different circadian phases.

Evidence in favour of sleep dependency

In 2013, Xie et al. [165] elegantly investigated CSF influx in sleeping and awake mice using in vivo two-photon imaging. While recording electrocorticography (ECoG) and electromyography to determine the mice’s vigilance state, they injected fluorescent tracers into the subarachnoid CSF. Compared to sleeping mice, parenchymal and periarterial tracer influx was sharply reduced in awake mice. This suppressed influx in awake mice appeared to be linked to decreases in interstitial space volume (ISV), as sleeping mice were observed to have an expanded ISV compared to awake mice [165]. The observed changes in ISV might explain the change in CSF influx and the higher efficiency of the glymphatic system during sleep, as increased ISV may decrease resistance to CSF movement, thereby increasing CSF influx [165].

The ECoG of sleeping mice exhibits significantly more slow waves than the ECoG of awake mice. A subsequent study revealed a positive correlation between the ECoG power density of these slow waves and CSF tracer influx in anaesthetized rodents [170]. Notably, the authors used six different anaesthetics, which have varying actions in increasing and decreasing glymphatic influx and likewise in modulation ECoG slow-waves, thereby increasing the reliability of their findings. A study by Fultz et al. [14] has also found coupling between neuronal activity, hemodynamic oscillations, and CSF flow. Additionally, a decreased heart rate, which is a NREM sleep characteristic, was found to positively correlate with tracer influx [170].

Recently, a study from Miao et al. [172] questioned the general consensus on the effects of sleep on brain clearance by stating that brain clearance is reduced during sleep and anaesthesia. Here, the authors measured the diffusion coefficient of their neuronal tracer by tracking its movement from the caudate putamen to the frontal cortex and validated the result in agarose gels. The authors found no difference in diffusion coefficient between waking, sleep, and anaesthesia. Yet, when measuring the clearance of the tracer, a significant clearance reduction was observed in sleeping mice compared to awake mice [172].

However, with their experiments, Miao et al. [172] deviate from commonly used methods to assess glymphatic function. Tracer studies are often performed by injecting the tracer into a ventricle and following its path through the glymphatic system [12, 165, 170]. In contrast, Miao et al. [172] injected their tracer directly into the caudate putamen, which they argue better mimics the location of waste molecules in physiological conditions. Moreover, they used a smaller tracer (4 kDa), compared to previously used tracers (66 kDa) [12, 170], and proteins like the amyloid-β precursor (110–135 kDa) [173]. These methodological discrepancies hamper the comparison to earlier work. Therefore, future research should focus on using similar methods and similar-sized tracers to elucidate whether there are genuine discrepancies in the results obtained or only in the interpretations of the results between the different laboratories.

Evidence in favour of circadian dependency

The evidence that links the efficacy of the glymphatic system to circadian rhythms is mainly focussed on AQP4. AQP4 is a bidirectional water channel in the brain that is most abundant in astrocytes [174]. The importance of AQP4 was established by Iliff et al. [128], who demonstrated through AQP4 KO mice that AQP4 is crucial for coupling CSF influx from para-arterial spaces to clearance of the interstitial fluid. To establish efficient coupling, AQP4 needs to be polarized to the endfeet of the astrocytes [12]. Other studies confirm that changes in AQP4 expression or polarization disturb the glymphatic system [175, 176]. Indeed, both Aqp4 gene expression and polarization show diurnal variation in nocturnal rodents; during their resting phase, Aqp4 expression is upregulated compared to the active phase, and AQP4 is localized at the endfeet of astrocytes [12]. This temporal polarization is likely dependent on the circadian expression of the proteins within the dystrophin-associated complex, a membrane-associated scaffold [12, 177, 178].

Hablitz et al. [12] demonstrated that circadian AQP4 polarization is preserved in constant light conditions. In this experiment, mice were kept in constant light for ten days before being anaesthetized with consistent doses of ketamine/xylazine during the subjective mid-resting or mid-active period. AQP4 polarization and glymphatic influx were found to be highest during the mid-resting phase [12], indicating that AQP4-mediated glymphatic clearance is circadian-dependent.

Moreover, various other circadian-regulated processes may promote glymphatic efficacy during the resting period. For example, CSF production by the ChP is significantly higher during the resting phase compared to the active phase [155]. One problem is that the number of studies investigating circadian functioning of the glymphatic system is limited and none of them is able to separate circadian changes from sleep regulatory influences. Therefore, these studies should be interpreted with caution.

Separating circadian rhythms and sleep

However, although certain aspects that influence glymphatic efficacy can be attributed to circadian or sleep-related processes, it is essential to emphasize that these processes are intimately related. As discussed, the circadian rhythm of the SCN mainly influences sleep timing. Sleep deprivation decreases SCN neuronal activity [179], alters the circadian expression of clock genes, and decreases their amplitude [180182]. Given this interdependence between sleep homeostatic mechanisms and circadian sleep timing, it is necessary to apply sleep-wake protocols where sleep timing is separated from the circadian cycle [183185] to investigate this further.

Brain homeostasis in health and disease

In previous parts of this review, the individual interactions between the SCN, circadian rhythms, GCs, sleep, and the glymphatic system were explored. The following paragraphs will review how these systems work in concert to maintain brain homeostasis in humans, and how disruptions may contribute to neurodegenerative diseases like AD (Figure 5).

Interactions in health and disease. (A) Interactions in health. The circadian rhythm in the suprachiasmatic nucleus (SCN) receives light input and is thereby synchronized (entrained) to the environmental cycle of day and night. The SCN coordinates circadian rhythms in peripheral clocks by means of neuronal or hormonal signalling pathways to create a temporal order. The sleep system is using the time information issued by the SCN to adjust sleep/wake patterns and it also reports its status back to the SCN. The rhythm in glucocorticoid (GC) level is entrained by SCN signals which are mediated through the HPA-axis. GC signalling influence the sleep system as well as the rhythmic production of CSF in the choroid plexus (ChP). The rhythms in the ChP also dependent on SCN function. Rhythmic GC signalling as well as sleep stages (NREM) contributes to a day–night difference in glymphatic flow, which was suggested to lead to a nightly clearance of neurotoxic substances and foster brain homeostasis. The rhythmic production of CSF in the ChP may also contribute to the modulation of flow. (B) Interactions in disease. In diseases like Alzheimer’s, early symptoms include disturbance in the sleep/wake pattern, which lead to a disruption of the rhythms in GCs and glymphatic flow. Circadian disorders caused by irregular activity schedules and light regimes as well as by weakened circadian system as found in the elderly, will contribute to this downward spiral, leading to a less efficient clearance and accumulation of neurotoxins

Brain homeostasis in health

One of the primary signalling pathways the SCN uses to synchronize peripheral clocks to the external environment is rhythmic GC release [8183]. In the early morning, the SCN stimulates GC production by the HPA-axis through the PVN/DMH, and via sympathetic activation of the splanchnic nerve [88, 97, 98]. Notably, the adrenal gland possesses an intrinsic clock, which is aligned by the SCN, but can also independently release GCs [106]. This morning increase in GCs activates the arousal-promoting areas and inhibits the sleep-promoting areas (VLPO) [124, 125]. Simultaneously, the homeostatic sleep pressure reaches its trough and the circadian process promotes wakefulness [31, 32].

During the day, when its activity is highest, the SCN inhibits the VLPO and stimulates orexin production, stabilizing wakefulness throughout the day [49, 50]. As the evening approaches, the activity of the SCN gradually declines, and the inhibition of the VLPO wanes. In addition, the pineal gland, whose circadian rhythm is entrained by the SCN, starts producing melatonin [186, 187]. Melatonin can further attenuate SCN firing [188190], amplifying the decreased inhibition of the VLPO. In addition, sleep pressure, which has been accumulating throughout the day, is thought to reach the sleeping threshold of process C. This series of events facilitates the transition from wakefulness to sleep.

During sleep, the ChP, entrained by GCs, increases CSF production [155]. The prevailing hypothesis indicates that the glymphatic system reaches peak efficacy during sleep, clearing the brain of amyloid-β, tau, and α-synuclein [128, 165]. As the night progresses and ends, the sleep pressure gradually declines, and GC release and SCN activity rise, allowing the cycle to restart.

Brain homeostasis in Alzheimer’s disease

In AD, those processes are disrupted. AD is a progressive neurodegenerative disorder characterized by dementia. The severity of dementia correlates with the amount and distribution of neurofibrillary tangles, which likely cause the clinical symptoms of the disease. The tangles are mainly composed of cytoskeleton-associated tau proteins [191]. The cascade of events that leads to neurofibrillary tangle formation is thought to be initiated by atypical amyloid-β secretion [192].

Sleep and circadian dysfunction

In addition to cognitive decline, AD is also associated with sleep and circadian dysfunction [193, 194]. Rather than solely presenting as symptoms, accumulating evidence suggests that these disruptions contribute to AD pathogenesis [195]. Sleep deprivation increases amyloid-β deposition in both healthy humans and AD mouse models [196, 197]. In a mouse model for AD, depositions could be reduced by injecting the mice with orexin antagonists, which stimulate sleep [197]. Moreover, in humans, high orexin concentrations in CSF have been associated with elevated levels of phosphorylated tau, present in the neurofibrillary tangles [198].

As the glymphatic system seems to clear amyloid-β and tau from the interstitial space most efficiently during sleep, disruptions of sleep and circadian rhythms may contribute to AD pathogenesis through dysfunction of the glymphatic system. Indeed, in AD patients, CSF production is lower than in healthy humans [199], the ChP shows altered secretion of neuroprotective peptides related to amyloid-β [200], and AQP4 polarization to the perivascular endfeet is lost [201].

Furthermore, a post-mortem study has demonstrated that the volume of the SCN and the number of SCN neurons in AD patients are decreased compared to non-AD patients [202]. In humans, the loss of SCN neurons is associated with amplitude-dampened and fragmented locomotor activity [203]. In addition, different brain regions of AD patients lose cohesion in circadian clock gene expression [204], indicating reduced functionality of the SCN. Molecular findings implicate a role of amyloid-β in the desynchronization of brain regions, as it facilitates BMAL1 degradation in neurons [205]. BMAL1 has been shown to regulate the expression of redox defence genes, so disruption of this clock gene can cause neurodegeneration due to oxidative stress [206].

Perhaps due to SCN deterioration, AD patients show reduced melatonin levels and rhythmicity [202, 207]. In addition, the SCN of late-stage AD patients expresses fewer melatonin type 1 receptors [207], the receptor that suppresses SCN firing activity and thereby allows VLPO activation. The reduced melatonin levels, rhythmicity, and ability to give feedback to SCN likely contribute to the sleep disruption observed in AD patients. Importantly, melatonin has been shown to reduce the production of amyloid-β [208]. Loss of proper melatonin levels and rhythmicity may therefore be detrimental to neuronal health in AD.

Circadian HPA-axis dysfunction

Dysregulation of the HPA-axis also plays an important role in the development of AD. AD patients show high basal cortisol levels [209, 210] and higher awakening cortisol levels, which are associated with worse memory performance [211]. In a 6-year follow-up study, AD patients with high cortisol levels declined faster in global cognition, episodic memory, and executive scores compared to AD patients with low cortisol levels [212]. Green et al. [213] have shown that GCs increase amyloid-β formation and neurofibrillary tangle formation in AD mouse models. As GCs also alter sleep architecture and increase arousal, the resulting loss of sleep might be harmful for the maintenance of amyloid-β and tau clearance by the glymphatic system.

Moreover, chronically high GC levels result in a loss of hippocampal volume [214], which is an early biomarker of AD progression [215]. In health, the hippocampus inhibits CRH and thus GC release [216, 217]. Therefore, the withering of the hippocampus results in a vicious cycle of GC release, exacerbating HPA-axis dysfunction. The presence of elevated HPA-axis activity in AD patients is corroborated by the finding that AD patients show increased adrenal sensitivity to exogenous ACTH [218].

In addition, GCs stimulate the expression of GFAP, a marker for abnormal astrocyte proliferation (reactive astrogliosis) [219]. Indeed, abnormally high GFAP levels have been observed in humans with high brain amyloid-β load [219, 220]. GFAP expression correlates with amyloid-β plaque density in AD brain tissue. As astrocytes can degrade amyloid-β [221], the increased GC levels in AD patients may initially serve as a protective mechanism. However, reactive astrogliosis is also closely associated with a decline in glutamate transporters in astrocytes [222], leaving the proximal neurons vulnerable to excitotoxicity.

In conclusion, sleep, circadian rhythms, the glymphatic system and GCs seem to be closely related in the pathogenesis of AD [129]. Further experiments are needed to gain more mechanistic insight on this as the evidence is now mainly correlative. Given the interdependencies of the circadian clock, sleep system, HPA signalling and the glymphatic function, the task is challenging but also holds great therapeutic potential. Treatment of circadian disorders with chronotherapy could normalize sleep patterns and GC signalling and have a positive effect on amyloid clearing by restoring glymphatic flow pattern. This could lead to either delayed onset or slower progression of not only AD, but also other neurodegenerative diseases like Parkinson’s and Huntington’s [223, 224].

Conclusions

Maintaining correct temporal organization among the discussed circadian systems is probably essential for brain homeostasis, with disruptions increasing the risk for neurodegenerative diseases. However, the pathways and mechanisms used by the circadian system to maintain a healthy temporal order are still poorly understood. This review explored the intricate interplay between the circadian system, GC signalling, the glymphatic system, and sleep. While the SCN is traditionally regarded as the controlling factor of circadian rhythms across these systems, emerging evidence suggests that the SCN also receives feedback from the peripheral clocks it entrains. This evolving perspective positions the SCN central in a bidirectionally regulated circadian network, instead of hierarchically above peripheral clocks.

Many open questions remain regarding the interactions between the central clock and peripheral clocks. For instance, the ChP has been shown to exhibit circadian patterns in the transport and degradation of endogenous signalling molecules in CSF [11]. As many brain structures, including the SCN, are exposed to CSF, the ChP may function as a conveyor of circadian signals [10]. For example, there is evidence for rhythmicity of prostaglandin D2 (PGD2) in the CSF, which promotes sleep [11], but the nature of circadian-rhythm-altering molecules in the CSF has not yet been identified. An important question that remains is the relative contribution of sleep and the circadian clock to glymphatic flow, which is important for future treatment possibilities. Should the treatment focus be on enhancing sleep quality or on increasing circadian amplitude? Finally, establishing standardized and reliable methodology will help uncover the exact relationship between sleep, circadian rhythms, and the glymphatic system.

An extensive understanding of the crosstalk between the circadian system, GC signalling, the glymphatic system, and sleep is imperative for preventing and treating circadian morbidity. Currently approved AD medicine focuses on symptom management and directly reducing amyloid-β plaques [225]. However, enhancing the function of the glymphatic system by restoring circadian control and improving sleep may offer an alternative approach by indirectly reducing amyloid-β deposition. Future research should therefore focus on increasing the robustness of the circadian system, and exploring the function of cells that are integral to all discussed systems, like glial cells. These findings may uncover novel therapeutic targets for improving brain health and resilience.

Abbreviations

ACTH: adrenocorticotropic hormone

AD: Alzheimer’s disease

AQP4: aquaporin-4

AVP: arginine vasopressin

ChP: choroid plexus

CRH: corticotropin-releasing hormone

Cry: cryptochrome

CSF: cerebrospinal fluid

DMH: dorsomedial nucleus of the hypothalamus

ECoG: electrocorticography

EEG: electroencephalogram

GC: glucocorticoid

GFAP: glial fibrillary acidic protein

GPCR: G-protein coupled receptor

GR: glucocorticoid receptor

GREs: glucocorticoid response elements

HPA-axis: hypothalamic-pituitary-adrenal axis

ipRGCs: intrinsically photosensitive retinal ganglion cells

ISV: interstitial space volume

LH: lateral hypothalamus

NREM: non-rapid eye movement

Per: period

PVN: paraventricular nucleus

REM: rapid eye movement

SCN: suprachiasmatic nucleus

TTFL: transcriptional-translational feedback loop

VLPO: ventrolateral preoptic nucleus

Declarations

Author contributions

LCAvdZ: Conceptualization, Visualization, Writing—original draft, Writing—review & editing. SM: Conceptualization, Supervision, Visualization, Writing—original draft, Writing—review & editing. TD: Project administration, Supervision, Writing—review & editing.

Conflicts of interest

The authors declare that they have no conflicts of interest.

Ethical approval

Not applicable.

Consent to participate

Not applicable.

Consent to publication

Not applicable.

Availability of data and materials

Not applicable.

Funding

Not applicable.

Copyright

© The Author(s) 2025.

Publisher’s note

Open Exploration maintains a neutral stance on jurisdictional claims in published institutional affiliations and maps. All opinions expressed in this article are the personal views of the author(s) and do not represent the stance of the editorial team or the publisher.

References

Rijo-Ferreira F, Takahashi JS. Genomics of circadian rhythms in health and disease. Genome Med. 2019;11:82. [DOI] [PubMed] [PMC]
Reppert SM, Weaver DR. Coordination of circadian timing in mammals. Nature. 2002;418:93541. [DOI] [PubMed]
Son GH, Cha HK, Chung S, Kim K. Multimodal Regulation of Circadian Glucocorticoid Rhythm by Central and Adrenal Clocks. J Endocr Soc. 2018;2:44459. [DOI] [PubMed] [PMC]
Mason BL, Pariante CM, Jamel S, Thomas SA. Central nervous system (CNS) delivery of glucocorticoids is fine-tuned by saturable transporters at the blood-CNS barriers and nonbarrier regions. Endocrinology. 2010;151:5294305. [DOI] [PubMed] [PMC]
Drapšin M, Dočkal T, Houdek P, Sládek M, Semenovykh K, Sumová A. Circadian clock in choroid plexus is resistant to immune challenge but dampens in response to chronodisruption. Brain Behav Immun. 2024;117:25569. [DOI] [PubMed]
Liška K, Sládek M, Čečmanová V, Sumová A. Glucocorticoids reset circadian clock in choroid plexus via period genes. J Endocrinol. 2021;248:15566. [DOI] [PubMed]
MacAulay N, Keep RF, Zeuthen T. Cerebrospinal fluid production by the choroid plexus: a century of barrier research revisited. Fluids Barriers CNS. 2022;19:26. [DOI] [PubMed] [PMC]
Hablitz LM, Nedergaard M. The Glymphatic System: A Novel Component of Fundamental Neurobiology. J Neurosci. 2021;41:7698711. [DOI] [PubMed] [PMC]
Sehar U, Rawat P, Reddy AP, Kopel J, Reddy PH. Amyloid Beta in Aging and Alzheimer’s Disease. Int J Mol Sci. 2022;23:12924. [DOI] [PubMed] [PMC]
Myung J, Schmal C, Hong S, Tsukizawa Y, Rose P, Zhang Y, et al. The choroid plexus is an important circadian clock component. Nat Commun. 2018;9:1062. [DOI] [PubMed] [PMC]
Sládek M, Houdek P, Myung J, Semenovykh K, Dočkal T, Sumová A. The circadian clock in the choroid plexus drives rhythms in multiple cellular processes under the control of the suprachiasmatic nucleus. Fluids Barriers CNS. 2024;21:46. [DOI] [PubMed] [PMC]
Hablitz LM, Plá V, Giannetto M, Vinitsky HS, Stæger FF, Metcalfe T, et al. Circadian control of brain glymphatic and lymphatic fluid flow. Nat Commun. 2020;11:4411. [DOI] [PubMed] [PMC]
Szmyd B, Rogut M, Białasiewicz P, Gabryelska A. The impact of glucocorticoids and statins on sleep quality. Sleep Med Rev. 2021;55:101380. [DOI] [PubMed]
Fultz NE, Bonmassar G, Setsompop K, Stickgold RA, Rosen BR, Polimeni JR, et al. Coupled electrophysiological, hemodynamic, and cerebrospinal fluid oscillations in human sleep. Science. 2019;366:62831. [DOI] [PubMed] [PMC]
Mieda M. The central circadian clock of the suprachiasmatic nucleus as an ensemble of multiple oscillatory neurons. Neurosci Res. 2020;156:2431. [DOI] [PubMed]
Menaker M, Murphy ZC, Sellix MT. Central control of peripheral circadian oscillators. Curr Opin Neurobiol. 2013;23:7416. [DOI] [PubMed]
Laothamatas I, Rasmussen ES, Green CB, Takahashi JS. Metabolic and chemical architecture of the mammalian circadian clock. Cell Chem Biol. 2023;30:103352. [DOI] [PubMed] [PMC]
Reick M, Garcia JA, Dudley C, McKnight SL. NPAS2: an analog of clock operative in the mammalian forebrain. Science. 2001;293:5069. [DOI] [PubMed]
Refinetti R. Integration of biological clocks and rhythms. Compr Physiol. 2012;2:121339. [DOI] [PubMed]
Hughes ATL, Piggins HD. Feedback actions of locomotor activity to the circadian clock. Prog Brain Res. 2012;199:30536. [DOI] [PubMed]
Do MTH, Kang SH, Xue T, Zhong H, Liao H, Bergles DE, et al. Photon capture and signalling by melanopsin retinal ganglion cells. Nature. 2009;457:2817. [DOI] [PubMed] [PMC]
Hattar S, Liao HW, Takao M, Berson DM, Yau KW. Melanopsin-containing retinal ganglion cells: architecture, projections, and intrinsic photosensitivity. Science. 2002;295:106570. [DOI] [PubMed] [PMC]
Provencio I, Rollag MD, Castrucci AM. Photoreceptive net in the mammalian retina. This mesh of cells may explain how some blind mice can still tell day from night. Nature. 2002;415:493. [DOI] [PubMed]
Irwin RP, Allen CN. Calcium response to retinohypothalamic tract synaptic transmission in suprachiasmatic nucleus neurons. J Neurosci. 2007;27:1174857. [DOI] [PubMed] [PMC]
Colwell CS. NMDA-evoked calcium transients and currents in the suprachiasmatic nucleus: gating by the circadian system. Eur J Neurosci. 2001;13:14208. [DOI] [PubMed] [PMC]
Golombek DA, Rosenstein RE. Physiology of circadian entrainment. Physiol Rev. 2010;90:1063102. [DOI] [PubMed]
Lee B, Li A, Hansen KF, Cao R, Yoon JH, Obrietan K. CREB influences timing and entrainment of the SCN circadian clock. J Biol Rhythms. 2010;25:41020. [DOI] [PubMed] [PMC]
van Oosterhout F, Lucassen EA, Houben T, vanderLeest HT, Antle MC, Meijer JH. Amplitude of the SCN clock enhanced by the behavioral activity rhythm. PLoS One. 2012;7:e39693. [DOI] [PubMed] [PMC]
Mrosovsky N, Salmon PA. A behavioural method for accelerating re-entrainment of rhythms to new light–dark cycles. Nature. 1987;330:3723. [DOI] [PubMed]
Starnes AN, Jones JR. Inputs and Outputs of the Mammalian Circadian Clock. Biology (Basel). 2023;12:508. [DOI] [PubMed] [PMC]
Borbély AA. A two process model of sleep regulation. Hum Neurobiol. 1982;1:195204. [PubMed]
Borbély AA, Daan S, Wirz-Justice A, Deboer T. The two-process model of sleep regulation: a reappraisal. J Sleep Res. 2016;25:13143. [DOI] [PubMed]
Pandi-Perumal SR, Cardinali DP, Zaki NFW, Karthikeyan R, Spence DW, Reiter RJ, et al. Timing is everything: Circadian rhythms and their role in the control of sleep. Front Neuroendocrinol. 2022;66:100978. [DOI] [PubMed]
Deboer T. Sleep homeostasis and the circadian clock: Do the circadian pacemaker and the sleep homeostat influence each other's functioning? Neurobiol Sleep Circadian Rhythms. 2018;5:6877. [DOI] [PubMed] [PMC]
Jones BE. Arousal systems. Front Biosci. 2003;8:s43851. [DOI] [PubMed]
Fornal C, Auerbach S, Jacobs BL. Activity of serotonin-containing neurons in nucleus raphe magnus in freely moving cats. Exp Neurol. 1985;88:590608. [DOI] [PubMed]
Aston-Jones G, Bloom FE. Activity of norepinephrine-containing locus coeruleus neurons in behaving rats anticipates fluctuations in the sleep-waking cycle. J Neurosci. 1981;1:87686. [DOI] [PubMed] [PMC]
Steininger TL, Alam MN, Gong H, Szymusiak R, McGinty D. Sleep-waking discharge of neurons in the posterior lateral hypothalamus of the albino rat. Brain Res. 1999;840:13847. [DOI] [PubMed]
Gallopin T, Fort P, Eggermann E, Cauli B, Luppi PH, Rossier J, et al. Identification of sleep-promoting neurons in vitro. Nature. 2000;404:9925. [DOI] [PubMed]
Sherin JE, Shiromani PJ, McCarley RW, Saper CB. Activation of ventrolateral preoptic neurons during sleep. Science. 1996;271:2169. [DOI] [PubMed]
Saper CB, Scammell TE, Lu J. Hypothalamic regulation of sleep and circadian rhythms. Nature. 2005;437:125763. [DOI] [PubMed]
Inutsuka A, Yamanaka A. The physiological role of orexin/hypocretin neurons in the regulation of sleep/wakefulness and neuroendocrine functions. Front Endocrinol (Lausanne). 2013;4:18. [DOI] [PubMed] [PMC]
Chou TC, Bjorkum AA, Gaus SE, Lu J, Scammell TE, Saper CB. Afferents to the ventrolateral preoptic nucleus. J Neurosci. 2002;22:97790. [DOI] [PubMed] [PMC]
Yoshida K, McCormack S, España RA, Crocker A, Scammell TE. Afferents to the orexin neurons of the rat brain. J Comp Neurol. 2006;494:84561. [DOI] [PubMed] [PMC]
Watts AG, Swanson LW, Sanchez-Watts G. Efferent projections of the suprachiasmatic nucleus: I. Studies using anterograde transport of Phaseolus vulgaris leucoagglutinin in the rat. J Comp Neurol. 1987;258:20429. [DOI] [PubMed]
Watts AG, Swanson LW. Efferent projections of the suprachiasmatic nucleus: II. Studies using retrograde transport of fluorescent dyes and simultaneous peptide immunohistochemistry in the rat. J Comp Neurol. 1987;258:23052. [DOI] [PubMed]
Deurveilher S, Semba K. Indirect projections from the suprachiasmatic nucleus to major arousal-promoting cell groups in rat: implications for the circadian control of behavioural state. Neuroscience. 2005;130:16583. [DOI] [PubMed]
Chou TC, Scammell TE, Gooley JJ, Gaus SE, Saper CB, Lu J. Critical role of dorsomedial hypothalamic nucleus in a wide range of behavioral circadian rhythms. J Neurosci. 2003;23:10691702. [DOI] [PubMed] [PMC]
Saint-Mleux B, Bayer L, Eggermann E, Jones BE, Mühlethaler M, Serafin M. Suprachiasmatic modulation of noradrenaline release in the ventrolateral preoptic nucleus. J Neurosci. 2007;27:64126. [DOI] [PubMed] [PMC]
Zhang S, Zeitzer JM, Yoshida Y, Wisor JP, Nishino S, Edgar DM, et al. Lesions of the suprachiasmatic nucleus eliminate the daily rhythm of hypocretin-1 release. Sleep. 2004;27:61927. [DOI] [PubMed]
Somers VK, Dyken ME, Mark AL, Abboud FM. Sympathetic-nerve activity during sleep in normal subjects. N Engl J Med. 1993;328:3037. [DOI] [PubMed]
Fink AM, Bronas UG, Calik MW. Autonomic regulation during sleep and wakefulness: a review with implications for defining the pathophysiology of neurological disorders. Clin Auton Res. 2018;28:50918. [DOI] [PubMed] [PMC]
Hirshkowitz M. Normal human sleep: an overview. Med Clin North Am. 2004;88:55165, vii. [DOI] [PubMed]
Deboer T. Behavioral and electrophysiological correlates of sleep and sleep homeostasis. Curr Top Behav Neurosci. 2015;25:124. [DOI] [PubMed]
Deboer T, Vansteensel MJ, Détári L, Meijer JH. Sleep states alter activity of suprachiasmatic nucleus neurons. Nat Neurosci. 2003;6:108690. [DOI] [PubMed]
Kuo T, McQueen A, Chen T, Wang J. Regulation of Glucose Homeostasis by Glucocorticoids. Adv Exp Med Biol. 2015;872:99126. [DOI] [PubMed] [PMC]
Mir N, Chin SA, Riddell MC, Beaudry JL. Genomic and Non-Genomic Actions of Glucocorticoids on Adipose Tissue Lipid Metabolism. Int J Mol Sci. 2021;22:8503. [DOI] [PubMed] [PMC]
Syed AP, Greulich F, Ansari SA, Uhlenhaut NH. Anti-inflammatory glucocorticoid action: genomic insights and emerging concepts. Curr Opin Pharmacol. 2020;53:3544. [DOI] [PubMed]
Bereshchenko O, Bruscoli S, Riccardi C. Glucocorticoids, Sex Hormones, and Immunity. Front Immunol. 2018;9:1332. [DOI] [PubMed] [PMC]
Pezük P, Mohawk JA, Wang LA, Menaker M. Glucocorticoids as entraining signals for peripheral circadian oscillators. Endocrinology. 2012;153:477583. [DOI] [PubMed] [PMC]
Balsalobre A, Brown SA, Marcacci L, Tronche F, Kellendonk C, Reichardt HM, et al. Resetting of circadian time in peripheral tissues by glucocorticoid signaling. Science. 2000;289:23447. [DOI] [PubMed]
Kater CE, Giorgi RB, Costa-Barbosa FA. Classic and current concepts in adrenal steroidogenesis: a reappraisal. Arch Endocrinol Metab. 2022;66:7787. [PubMed] [PMC]
Vale W, Spiess J, Rivier C, Rivier J. Characterization of a 41-residue ovine hypothalamic peptide that stimulates secretion of corticotropin and β-endorphin. Science. 1981;213:13947. [DOI] [PubMed]
Tse A, Lee AK, Tse FW. Ca2+ signaling and exocytosis in pituitary corticotropes. Cell Calcium. 2012;51:2539. [DOI] [PubMed]
Joëls M, Karst H, Sarabdjitsingh RA. The stressed brain of humans and rodents. Acta Physiol (Oxf). 2018;223:e13066. [DOI] [PubMed] [PMC]
Gjerstad JK, Lightman SL, Spiga F. Role of glucocorticoid negative feedback in the regulation of HPA axis pulsatility. Stress. 2018;21:40316. [DOI] [PubMed] [PMC]
Dallman MF, Jones MT, Vernikos-Danellis J, Ganong WF. Corticosteroid feedback control of ACTH secretion: rapid effects of bilateral adrenalectomy on plasma ACTH in the rat. Endocrinology. 1972;91:9618. [DOI] [PubMed]
Sawchenko PE. Adrenalectomy-induced enhancement of CRF and vasopressin immunoreactivity in parvocellular neurosecretory neurons: anatomic, peptide, and steroid specificity. J Neurosci. 1987;7:1093106. [DOI] [PubMed] [PMC]
Levin N, Shinsako J, Dallman MF. Corticosterone acts on the brain to inhibit adrenalectomy-induced adrenocorticotropin secretion. Endocrinology. 1988;122:694701. [DOI] [PubMed]
Kovács KJ, Elenkov IJ. Differential dependence of ACTH secretion induced by various cytokines on the integrity of the paraventricular nucleus. J Neuroendocrinol. 1995;7:1523. [DOI] [PubMed]
Gagner JP, Drouin J. Opposite regulation of pro-opiomelanocortin gene transcription by glucocorticoids and CRH. Mol Cell Endocrinol. 1985;40:2532. [DOI] [PubMed]
Eberwine JH, Roberts JL. Glucocorticoid regulation of pro-opiomelanocortin gene transcription in the rat pituitary. J Biol Chem. 1984;259:216670. [PubMed]
Charron J, Drouin J. Glucocorticoid inhibition of transcription from episomal proopiomelanocortin gene promoter. Proc Natl Acad Sci U S A. 1986;83:89037. [DOI] [PubMed] [PMC]
Lightman SL, Birnie MT, Conway-Campbell BL. Dynamics of ACTH and Cortisol Secretion and Implications for Disease. Endocr Rev. 2020;41:bnaa002. [DOI] [PubMed] [PMC]
Gallagher TF, Yoshida K, Roffwarg HD, Fukushima DK, Weitzman ED, Hellman L. ACTH and cortisol secretory patterns in man. J Clin Endocrinol Metab. 1973;36:105868. [DOI] [PubMed]
Qian X, Droste SK, Lightman SL, Reul JMHM, Linthorst ACE. Circadian and ultradian rhythms of free glucocorticoid hormone are highly synchronized between the blood, the subcutaneous tissue, and the brain. Endocrinology. 2012;153:434653. [DOI] [PubMed] [PMC]
Perlow MJ, Reppert SM, Boyar RM, Klein DC. Daily rhythms in cortisol and melatonin in primate cerebrospinal fluid: Effects of constant light and dark. Neuroendocrinology. 1981;32:1936. [DOI] [PubMed]
Cauter EV. Diurnal and ultradian rhythms in human endocrine function: a minireview. Horm Res. 1990;34:4553. [DOI] [PubMed]
Dickmeis T, Weger BD, Weger M. The circadian clock and glucocorticoids – interactions across many time scales. Mol Cell Endocrinol. 2013;380:215. [DOI] [PubMed]
Cunningham ET Jr, Sawchenko PE. Anatomical specificity of noradrenergic inputs to the paraventricular and supraoptic nuclei of the rat hypothalamus. J Comp Neurol. 1988;274:6076. [DOI] [PubMed]
Watanabe K, Hiroshige T. Phase relation between episodic fluctuations of spontaneous locomotor activity and plasma corticosterone in rats with suprachiasmatic nuclei lesions. Neuroendocrinology. 1981;33:529. [DOI] [PubMed]
Waite EJ, McKenna M, Kershaw Y, Walker JJ, Cho K, Piggins HD, et al. Ultradian corticosterone secretion is maintained in the absence of circadian cues. Eur J Neurosci. 2012;36:314250. [DOI] [PubMed]
Moore RY, Eichler VB. Loss of a circadian adrenal corticosterone rhythm following suprachiasmatic lesions in the rat. Brain Res. 1972;42:2016. [DOI] [PubMed]
Claustrat B, Valatx J, Harthé C, Brun J. Effect of constant light on prolactin and corticosterone rhythms evaluated using a noninvasive urine sampling protocol in the rat. Horm Metab Res. 2008;40:398403. [DOI] [PubMed]
Chung S, Son GH, Kim K. Circadian rhythm of adrenal glucocorticoid: its regulation and clinical implications. Biochim Biophys Acta. 2011;1812:58191. [DOI] [PubMed]
Stephan FK, Berkley KJ, Moss RL. Efferent connections of the rat suprachiasmatic nucleus. Neuroscience. 1981;6:262541. [DOI] [PubMed]
Vrang N, Larsen PJ, Mikkelsen JD. Direct projection from the suprachiasmatic nucleus to hypophysiotrophic corticotropin-releasing factor immunoreactive cells in the paraventricular nucleus of the hypothalamus demonstrated by means of Phaseolus vulgaris-leucoagglutinin tract tracing. Brain Res. 1995;684:619. [DOI] [PubMed]
Jones JR, Chaturvedi S, Granados-Fuentes D, Herzog ED. Circadian neurons in the paraventricular nucleus entrain and sustain daily rhythms in glucocorticoids. Nat Commun. 2021;12:5763. [DOI] [PubMed] [PMC]
Hoorneman EM, Buijs RM. Vasopressin fiber pathways in the rat brain following suprachiasmatic nucleus lesioning. Brain Res. 1982;243:23541. [DOI] [PubMed]
Kalsbeek A, Buijs RM, van Heerikhuize JJ, Arts M, van der Woude TP. Vasopressin-containing neurons of the suprachiasmatic nuclei inhibit corticosterone release. Brain Res. 1992;580:627. [DOI] [PubMed]
Kalsbeek A, Verhagen LAW, Schalij I, Foppen E, Saboureau M, Bothorel B, et al. Opposite actions of hypothalamic vasopressin on circadian corticosterone rhythm in nocturnal versus diurnal species. Eur J Neurosci. 2008;27:81827. [DOI] [PubMed]
Akana SF, Cascio CS, Du JZ, Levin N, Dallman MF. Reset of feedback in the adrenocortical system: an apparent shift in sensitivity of adrenocorticotropin to inhibition by corticosterone between morning and evening. Endocrinology. 1986;119:232532. [DOI] [PubMed]
Kaneko M, Hiroshige T, Shinsako J, Dallman MF. Diurnal changes in amplification of hormone rhythms in the adrenocortical system. Am J Physiol. 1980;239:R30916. [DOI] [PubMed]
Wotus C, Lilley TR, Neal AS, Suleiman NL, Schmuck SC, Smarr BL, et al. Forced desynchrony reveals independent contributions of suprachiasmatic oscillators to the daily plasma corticosterone rhythm in male rats. PLoS One. 2013;8:e68793. [DOI] [PubMed] [PMC]
Meier AH. Daily variation in concentration of plasma corticosteroid in hypophysectomized rats. Endocrinology. 1976;98:14759. [DOI] [PubMed]
Engeland WC. Functional innervation of the adrenal cortex by the splanchnic nerve. Horm Metab Res. 1998;30:3114. [DOI] [PubMed]
Niijima A, Nagai K, Nagai N, Akagawa H. Effects of light stimulation on the activity of the autonomic nerves in anesthetized rats. Physiol Behav. 1993;54:55561. [DOI] [PubMed]
Ishida A, Mutoh T, Ueyama T, Bando H, Masubuchi S, Nakahara D, et al. Light activates the adrenal gland: timing of gene expression and glucocorticoid release. Cell Metab. 2005;2:297307. [DOI] [PubMed]
Engeland WC, Gann DS. Splanchnic nerve stimulation modulates steroid secretion in hypophysectomized dogs. Neuroendocrinology. 1989;50:12431. [DOI] [PubMed]
Edwards AV, Jones CT. The effect of splanchnic nerve stimulation on adrenocortical activity in conscious calves. J Physiol. 1987;382:38596. [DOI] [PubMed] [PMC]
Ulrich-Lai YM, Arnhold MM, Engeland WC. Adrenal splanchnic innervation contributes to the diurnal rhythm of plasma corticosterone in rats by modulating adrenal sensitivity to ACTH. Am J Physiol Regul Integr Comp Physiol. 2006;290:R112835. [DOI] [PubMed]
Kaneko M, Kaneko K, Shinsako J, Dallman MF. Adrenal sensitivity to adrenocorticotropin varies diurnally. Endocrinology. 1981;109:705. [DOI] [PubMed]
Sage D, Maurel D, Bosler O. Corticosterone-dependent driving influence of the suprachiasmatic nucleus on adrenal sensitivity to ACTH. Am J Physiol Endocrinol Metab. 2002;282:E45865. [DOI] [PubMed]
Krieger DT, Hauser H, Krey LC. Suprachiasmatic nuclear lesions do not abolish food-shifted circadian adrenal and temperature rhythmicity. Science. 1977;197:3989. [DOI] [PubMed]
Chung S, Lee EJ, Cha HK, Kim J, Kim D, Son GH, et al. Cooperative roles of the suprachiasmatic nucleus central clock and the adrenal clock in controlling circadian glucocorticoid rhythm. Sci Rep. 2017;7:46404. [DOI] [PubMed] [PMC]
Son GH, Chung S, Choe HK, Kim H, Baik S, Lee H, et al. Adrenal peripheral clock controls the autonomous circadian rhythm of glucocorticoid by causing rhythmic steroid production. Proc Natl Acad Sci U S A. 2008;105:209705. [DOI] [PubMed] [PMC]
Horseman ND, Ehret CF. Glucocorticosteroid injection is a circadian zeitgeber in the laboratory rat. Am J Physiol. 1982;243:R3738. [DOI] [PubMed]
Chihara I, Negoro H, Kono J, Nagumo Y, Tsuchiya H, Kojo K, et al. Glucocorticoids coordinate the bladder peripheral clock and diurnal micturition pattern in mice. Commun Biol. 2023;6:81. [DOI] [PubMed] [PMC]
Azmi NASM, Juliana N, Azmani S, Effendy NM, Abu IF, Teng NIMF, et al. Cortisol on Circadian Rhythm and Its Effect on Cardiovascular System. Int J Environ Res Public Health. 2021;18:676. [DOI] [PubMed] [PMC]
BaHammam AS, Pirzada A. Timing Matters: The Interplay between Early Mealtime, Circadian Rhythms, Gene Expression, Circadian Hormones, and Metabolism—A Narrative Review. Clocks Sleep. 2023;5:50735. [DOI] [PubMed] [PMC]
So AY, Bernal TU, Pillsbury ML, Yamamoto KR, Feldman BJ. Glucocorticoid regulation of the circadian clock modulates glucose homeostasis. Proc Natl Acad Sci U S A. 2009;106:175827. [DOI] [PubMed] [PMC]
Eachus H, Oberski L, Paveley J, Bacila I, Ashton J, Esposito U, et al. Glucocorticoid receptor regulates protein chaperone, circadian clock and affective disorder genes in the zebrafish brain. Dis Model Mech. 2023;16:dmm050141. [DOI] [PubMed] [PMC]
Okabe T, Chavan R, Costa SSF, Brenna A, Ripperger JA, Albrecht U. REV-ERBα influences the stability and nuclear localization of the glucocorticoid receptor. J Cell Sci. 2016;129:414354. [DOI] [PubMed] [PMC]
Nader N, Chrousos GP, Kino T. Circadian rhythm transcription factor CLOCK regulates the transcriptional activity of the glucocorticoid receptor by acetylating its hinge region lysine cluster: potential physiological implications. FASEB J. 2009;23:157283. [DOI] [PubMed] [PMC]
Lamia KA, Papp SJ, Yu RT, Barish GD, Uhlenhaut NH, Jonker JW, et al. Cryptochromes mediate rhythmic repression of the glucocorticoid receptor. Nature. 2011;480:5526. [DOI] [PubMed] [PMC]
Kong X, Ota SM, Suchecki D, Lan A, Peereboom AI, Hut RA, et al. Chronic Social Defeat Stress Shifts Peripheral Circadian Clocks in Male Mice in a Tissue-Specific and Time-of-Day Dependent Fashion. J Biol Rhythms. 2022;37:16476. [DOI] [PubMed]
Rosenfeld P, Eekelen JAV, Levine S, Kloet ERD. Ontogeny of the type 2 glucocorticoid receptor in discrete rat brain regions: an immunocytochemical study. Brain Res. 1988;470:11927. [DOI] [PubMed]
Kloet ERD, Rosenfeld P, Eekelen JAV, Sutanto W, Levine S. Stress, glucocorticoids and development. Prog Brain Res. 1988;73:10120. [DOI] [PubMed]
Fowden AL, Vaughan OR, Murray AJ, Forhead AJ. Metabolic Consequences of Glucocorticoid Exposure before Birth. Nutrients. 2022;14:2304. [DOI] [PubMed] [PMC]
Čečmanová V, Houdek P, Šuchmanová K, Sládek M, Sumová A. Development and Entrainment of the Fetal Clock in the Suprachiasmatic Nuclei: The Role of Glucocorticoids. J Biol Rhythms. 2019;34:30722. [DOI] [PubMed]
Lehmann M, Haury K, Oster H, Astiz M. Circadian glucocorticoids throughout development. Front Neurosci. 2023;17:1165230. [DOI] [PubMed] [PMC]
Maurel D, Sage D, Mekaouche M, Bosler O. Glucocorticoids up-regulate the expression of glial fibrillary acidic protein in the rat suprachiasmatic nucleus. Glia. 2000;29:21221. [DOI] [PubMed]
Smyllie NJ, Hastings MH, Patton AP. Neuron-Astrocyte Interactions and Circadian Timekeeping in Mammals. Neuroscientist. 2025;31:6579. [DOI] [PubMed] [PMC]
Vazquez-Palacios G, Velazquez-Moctezuma J. Effect of electric foot shocks, immobilization, and corticosterone administration on the sleep-wake pattern in the rat. Physiol Behav. 2000;71:238. [DOI] [PubMed]
Wang Z, Zhang X, Cui X, Cui S, Yu B, Sheng Z, et al. Glucocorticoid receptors in the locus coeruleus mediate sleep disorders caused by repeated corticosterone treatment. Sci Rep. 2015;5:9442. [DOI] [PubMed] [PMC]
Silver R, LeSauter J, Tresco PA, Lehman MN. A diffusible coupling signal from the transplanted suprachiasmatic nucleus controlling circadian locomotor rhythms. Nature. 1996;382:8103. [DOI] [PubMed]
Roy RK, Yao Y, Green IK, Aitken AV, Biancardi VC, Silver R, et al. Blood flows from the SCN toward the OVLT within a new brain vascular portal pathway. Sci Adv. 2024;10:eadn8350. [DOI] [PubMed] [PMC]
Iliff JJ, Wang M, Liao Y, Plogg BA, Peng W, Gundersen GA, et al. A paravascular pathway facilitates CSF flow through the brain parenchyma and the clearance of interstitial solutes, including amyloid β. Sci Transl Med. 2012;4:147ra111. [DOI] [PubMed] [PMC]
Astara K, Tsimpolis A, Kalafatakis K, Vavougios GD, Xiromerisiou G, Dardiotis E, et al. Sleep disorders and Alzheimer’s disease pathophysiology: The role of the Glymphatic System. A scoping review. Mech Ageing Dev. 2024;217:111899. [DOI] [PubMed]
Spector R, Snodgrass SR, Johanson CE. A balanced view of the cerebrospinal fluid composition and functions: Focus on adult humans. Exp Neurol. 2015;273:5768. [DOI] [PubMed]
Hutton D, Fadelalla MG, Kanodia AK, Hossain-Ibrahim K. Choroid plexus and CSF: an updated review. Br J Neurosurg. 2022;36:30715. [DOI] [PubMed]
Telano LN, Baker S. Physiology, Cerebral Spinal Fluid. StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2025. [PubMed]
Kratzer I, Vasiljevic A, Rey C, Fevre-Montange M, Saunders N, Strazielle N, et al. Complexity and developmental changes in the expression pattern of claudins at the blood–CSF barrier. Histochem Cell Biol. 2012;138:86179. [DOI] [PubMed] [PMC]
Praetorius J, Damkier HH. Transport across the choroid plexus epithelium. Am J Physiol Cell Physiol. 2017;312:C67386. [DOI] [PubMed]
Bitanihirwe BKY, Lizano P, Woo TW. Deconstructing the functional neuroanatomy of the choroid plexus: an ontogenetic perspective for studying neurodevelopmental and neuropsychiatric disorders. Mol Psychiatry. 2022;27:357382. [DOI] [PubMed] [PMC]
Sakka L, Coll G, Chazal J. Anatomy and physiology of cerebrospinal fluid. Eur Ann Otorhinolaryngol Head Neck Dis. 2011;128:30916. [DOI] [PubMed]
Oshio K, Watanabe H, Song Y, Verkman AS, Manley GT. Reduced cerebrospinal fluid production and intracranial pressure in mice lacking choroid plexus water channel Aquaporin-1. FASEB J. 2005;19:768. [DOI] [PubMed]
Brown PD, Davies SL, Speake T, Millar ID. Molecular mechanisms of cerebrospinal fluid production. Neuroscience. 2004;129:95770. [DOI] [PubMed] [PMC]
Damkier HH, Brown PD, Praetorius J. Cerebrospinal fluid secretion by the choroid plexus. Physiol Rev. 2013;93:184792. [DOI] [PubMed]
Yankova G, Bogomyakova O, Tulupov A. The glymphatic system and meningeal lymphatics of the brain: new understanding of brain clearance. Rev Neurosci. 2021;32:693705. [DOI] [PubMed]
Mestre H, Hablitz LM, Xavier AL, Feng W, Zou W, Pu T, et al. Aquaporin-4-dependent glymphatic solute transport in the rodent brain. Elife. 2018;7:e40070. [DOI] [PubMed] [PMC]
Patel TK, Habimana-Griffin L, Gao X, Xu B, Achilefu S, Alitalo K, et al. Dural lymphatics regulate clearance of extracellular tau from the CNS. Mol Neurodegener. 2019;14:11. [DOI] [PubMed] [PMC]
Zou W, Pu T, Feng W, Lu M, Zheng Y, Du R, et al. Blocking meningeal lymphatic drainage aggravates Parkinson’s disease-like pathology in mice overexpressing mutated α-synuclein. Transl Neurodegener. 2019;8:7. [DOI] [PubMed] [PMC]
Iliff JJ, Wang M, Zeppenfeld DM, Venkataraman A, Plog BA, Liao Y, et al. Cerebral arterial pulsation drives paravascular CSF–interstitial fluid exchange in the murine brain. J Neurosci. 2013;33:181909. [DOI] [PubMed] [PMC]
Mestre H, Tithof J, Du T, Song W, Peng W, Sweeney AM, et al. Flow of cerebrospinal fluid is driven by arterial pulsations and is reduced in hypertension. Nat Commun. 2018;9:4878. [DOI] [PubMed] [PMC]
van Veluw SJ, Hou SS, Calvo-Rodriguez M, Arbel-Ornath M, Snyder AC, Frosch MP, et al. Vasomotion as a Driving Force for Paravascular Clearance in the Awake Mouse Brain. Neuron. 2020;105:54961.e5. [DOI] [PubMed] [PMC]
Hauglund NL, Andersen M, Tokarska K, Radovanovic T, Kjaerby C, Sørensen FL, et al. Norepinephrine-mediated slow vasomotion drives glymphatic clearance during sleep. Cell. 2025;188:60622.e17. [DOI] [PubMed]
Kiviniemi V, Wang X, Korhonen V, Keinänen T, Tuovinen T, Autio J, et al. Ultra-fast magnetic resonance encephalography of physiological brain activity – Glymphatic pulsation mechanisms? J Cereb Blood Flow Metab. 2016;36:103345. [DOI] [PubMed] [PMC]
Plog BA, Lou N, Pierre CA, Cove A, Kenney HM, Hitomi E, et al. When the air hits your brain: decreased arterial pulsatility after craniectomy leading to impaired glymphatic flow. J Neurosurg. 2019;133:21023. [DOI] [PubMed] [PMC]
Mestre H, Mori Y, Nedergaard M. The Brain’s Glymphatic System: Current Controversies. Trends Neurosci. 2020;43:45866. [DOI] [PubMed] [PMC]
Butler T, Zhou L, Ozsahin I, Wang XH, Garetti J, Zetterberg H, et al. Glymphatic clearance estimated using diffusion tensor imaging along perivascular spaces is reduced after traumatic brain injury and correlates with plasma neurofilament light, a biomarker of injury severity. Brain Commun. 2023;5:fcad134. [DOI] [PubMed] [PMC]
Okazawa H, Nogami M, Ishida S, Makino A, Mori T, Kiyono Y, et al. PET/MRI multimodality imaging to evaluate changes in glymphatic system function and biomarkers of Alzheimer’s disease. Sci Rep. 2024;14:12310. [DOI] [PubMed] [PMC]
Albargothy NJ, Johnston DA, MacGregor-Sharp M, Weller RO, Verma A, Hawkes CA, et al. Convective influx/glymphatic system: tracers injected into the CSF enter and leave the brain along separate periarterial basement membrane pathways. Acta Neuropathol. 2018;136:13952. [DOI] [PubMed] [PMC]
Hladky SB, Barrand MA. Elimination of substances from the brain parenchyma: efflux via perivascular pathways and via the blood–brain barrier. Fluids Barriers CNS. 2018;15:30. [DOI] [PubMed] [PMC]
Nilsson C, Ståhlberg F, Thomsen C, Henriksen O, Herning M, Owman C. Circadian variation in human cerebrospinal fluid production measured by magnetic resonance imaging. Am J Physiol. 1992;262:R204. [DOI] [PubMed]
Harrington MG, Salomon RM, Pogoda JM, Oborina E, Okey N, Johnson B, et al. Cerebrospinal fluid sodium rhythms. Cerebrospinal Fluid Res. 2010;7:3. [DOI] [PubMed] [PMC]
Edelbo BL, Andreassen SN, Steffensen AB, MacAulay N. Day–night fluctuations in choroid plexus transcriptomics and cerebrospinal fluid metabolomics. PNAS Nexus. 2023;2:pgad262. [DOI] [PubMed] [PMC]
Chen X, Shi C, He M, Xiong S, Xia X. Endoplasmic reticulum stress: molecular mechanism and therapeutic targets. Signal Transduct Target Ther. 2023;8:352. [DOI] [PubMed] [PMC]
Quintela T, Sousa C, Patriarca FM, Gonçalves I, Santos CRA. Gender associated circadian oscillations of the clock genes in rat choroid plexus. Brain Struct Funct. 2015;220:125162. [DOI] [PubMed]
Yamaguchi T, Hamada T, Matsuzaki T, Iijima N. Characterization of the circadian oscillator in the choroid plexus of rats. Biochem Biophys Res Commun. 2020;524:497501. [DOI] [PubMed]
Liddelow SA, Dziegielewska KM, Ek CJ, Habgood MD, Bauer H, Bauer H, et al. Mechanisms that determine the internal environment of the developing brain: a transcriptomic, functional and ultrastructural approach. PLoS One. 2013;8:e65629. [DOI] [PubMed] [PMC]
Martinho A, Gonçalves I, Costa M, Santos CR. Stress and glucocorticoids increase transthyretin expression in rat choroid plexus via mineralocorticoid and glucocorticoid receptors. J Mol Neurosci. 2012;48:113. [DOI] [PubMed]
Sinclair AJ, Onyimba CU, Khosla P, Vijapurapu N, Tomlinson JW, Burdon MA, et al. Corticosteroids, 11β-hydroxysteroid dehydrogenase isozymes and the rabbit choroid plexus. J Neuroendocrinol. 2007;19:61420. [DOI] [PubMed]
Burén J, Bergström S, Loh E, Söderström I, Olsson T, Mattsson C. Hippocampal 11β-hydroxysteroid dehydrogenase type 1 messenger ribonucleic acid expression has a diurnal variability that is lost in the obese Zucker rat. Endocrinology. 2007;148:271622. [DOI] [PubMed]
Xie L, Kang H, Xu Q, Chen MJ, Liao Y, Thiyagarajan M, et al. Sleep drives metabolite clearance from the adult brain. Science. 2013;342:3737. [DOI] [PubMed] [PMC]
Astara K, Pournara C, Natale ERd, Wilson H, Vavougios GD, Lappas AS, et al. A novel conceptual framework for the functionality of the glymphatic system. J Neurophysiol. 2023;129:122836. [DOI] [PubMed]
Franks NP, Wisden W. The inescapable drive to sleep: Overlapping mechanisms of sleep and sedation. Science. 2021;374:5569. [DOI] [PubMed]
Date A, Bashir K, Uddin A, Nigam C. Differences between natural sleep and the anesthetic state. Future Sci OA. 2020;6:FSO664. [DOI]
Lilius TO, Blomqvist K, Hauglund NL, Liu G, Stæger FF, Bærentzen S, et al. Dexmedetomidine enhances glymphatic brain delivery of intrathecally administered drugs. J Control Release. 2019;304:2938. [DOI] [PubMed]
Hablitz LM, Vinitsky HS, Sun Q, Stæger FF, Sigurdsson B, Mortensen KN, et al. Increased glymphatic influx is correlated with high EEG delta power and low heart rate in mice under anesthesia. Sci Adv. 2019;5:eaav5447. [DOI] [PubMed] [PMC]
Gakuba C, Gaberel T, Goursaud S, Bourges J, Palma CD, Quenault A, et al. General Anesthesia Inhibits the Activity of the “Glymphatic System”. Theranostics. 2018;8:71022. [DOI] [PubMed] [PMC]
Miao A, Luo T, Hsieh B, Edge CJ, Gridley M, Wong RTC, et al. Brain clearance is reduced during sleep and anesthesia. Nat Neurosci. 2024;27:104650. [DOI] [PubMed] [PMC]
Selkoe DJ, Podlisny MB, Joachim CL, Vickers EA, Lee G, Fritz LC, et al. Beta-amyloid precursor protein of Alzheimer disease occurs as 110- to 135-kilodalton membrane-associated proteins in neural and nonneural tissues. Proc Natl Acad Sci U S A. 1988;85:73415. [DOI] [PubMed] [PMC]
Rash JE, Yasumura T, Hudson CS, Agre P, Nielsen S. Direct immunogold labeling of aquaporin-4 in square arrays of astrocyte and ependymocyte plasma membranes in rat brain and spinal cord. Proc Natl Acad Sci U S A. 1998;95:119816. [DOI] [PubMed] [PMC]
Kress BT, Iliff JJ, Xia M, Wang M, Wei HS, Zeppenfeld D, et al. Impairment of paravascular clearance pathways in the aging brain. Ann Neurol. 2014;76:84561. [DOI] [PubMed] [PMC]
Munk AS, Wang W, Bèchet NB, Eltanahy AM, Cheng AX, Sigurdsson B, et al. PDGF-B Is Required for Development of the Glymphatic System. Cell Rep. 2019;26:295569.e3. [DOI] [PubMed] [PMC]
Amiry-Moghaddam M, Otsuka T, Hurn PD, Traystman RJ, Haug F, Froehner SC, et al. An α-syntrophin-dependent pool of AQP4 in astroglial end-feet confers bidirectional water flow between blood and brain. Proc Natl Acad Sci U S A. 2003;100:210611. [DOI] [PubMed] [PMC]
Lananna BV, Nadarajah CJ, Izumo M, Cedeño MR, Xiong DD, Dimitry J, et al. Cell-Autonomous Regulation of Astrocyte Activation by the Circadian Clock Protein BMAL1. Cell Rep. 2018;25:19.e5. [DOI] [PubMed] [PMC]
Deboer T, Détári L, Meijer JH. Long term effects of sleep deprivation on the mammalian circadian pacemaker. Sleep. 2007;30:25762. [DOI] [PubMed]
Zhang B, Gao Y, Li Y, Yang J, Zhao H. Sleep Deprivation Influences Circadian Gene Expression in the Lateral Habenula. Behav Neurol. 2016;2016:7919534. [DOI] [PubMed] [PMC]
Möller-Levet CS, Archer SN, Bucca G, Laing EE, Slak A, Kabiljo R, et al. Effects of insufficient sleep on circadian rhythmicity and expression amplitude of the human blood transcriptome. Proc Natl Acad Sci U S A. 2013;110:E113241. [DOI] [PubMed] [PMC]
Curie T, Maret S, Emmenegger Y, Franken P. In Vivo Imaging of the Central and Peripheral Effects of Sleep Deprivation and Suprachiasmatic Nuclei Lesion on PERIOD-2 Protein in Mice. Sleep. 2015;38:138194. [DOI] [PubMed] [PMC]
Dijk DJ, Czeisler CA. Contribution of the circadian pacemaker and the sleep homeostat to sleep propensity, sleep structure, electroencephalographic slow waves, and sleep spindle activity in humans. J Neurosci. 1995;15:352638. [DOI] [PubMed] [PMC]
Yasenkov R, Deboer T. Circadian regulation of sleep and the sleep EEG under constant sleep pressure in the rat. Sleep. 2010;33:63141. [DOI] [PubMed] [PMC]
Wang W, Yuan RK, Mitchell JF, Zitting K, Hilaire MAS, Wyatt JK, et al. Desynchronizing the sleep–wake cycle from circadian timing to assess their separate contributions to physiology and behaviour and to estimate intrinsic circadian period. Nat Protoc. 2023;18:579603. [DOI] [PubMed]
Garidou M, Gauer F, Vivien-Roels B, Sicard B, Pévet P, Simonneaux V. Pineal arylalkylamine N-acetyltransferase gene expression is highly stimulated at night in the diurnal rodent, Arvicanthis ansorgei. Eur J Neurosci. 2002;15:163240. [DOI] [PubMed]
Vivanco P, Ortiz V, Rol MA, Madrid JA. Looking for the keys to diurnality downstream from the circadian clock: role of melatonin in a dual-phasing rodent, Octodon degus. J Pineal Res. 2007;42:28090. [DOI] [PubMed]
Mason R, Brooks A. The electrophysiological effects of melatonin and a putative melatonin antagonist (N-acetyltryptamine) on rat suprachiasmatic neurones in vitro. Neurosci Lett. 1988;95:296301. [DOI] [PubMed]
Shibata S, Cassone VM, Moore RY. Effects of melatonin on neuronal activity in the rat suprachiasmatic nucleus in vitro. Neurosci Lett. 1989;97:1404. [DOI] [PubMed]
Stehle J, Vanecek J, Vollrath L. Effects of melatonin on spontaneous electrical activity of neurons in rat suprachiasmatic nuclei: an in vitro iontophoretic study. J Neural Transm. 1989;78:1737. [DOI] [PubMed]
Savelieff MG, Lee S, Liu Y, Lim MH. Untangling amyloid-β, tau, and metals in Alzheimer’s disease. ACS Chem Biol. 2013;8:85665. [DOI] [PubMed]
Busche MA, Hyman BT. Synergy between amyloid-β and tau in Alzheimer’s disease. Nat Neurosci. 2020;23:118393. [DOI] [PubMed] [PMC]
Hatfield CF, Herbert J, van Someren EJ, Hodges JR, Hastings MH. Disrupted daily activity/rest cycles in relation to daily cortisol rhythms of home-dwelling patients with early Alzheimer’s dementia. Brain. 2004;127:106174. [DOI] [PubMed]
Musiek ES, Bhimasani M, Zangrilli MA, Morris JC, Holtzman DM, Ju YS. Circadian Rest-Activity Pattern Changes in Aging and Preclinical Alzheimer Disease. JAMA Neurol. 2018;75:58290. [DOI] [PubMed] [PMC]
Lim ASP, Kowgier M, Yu L, Buchman AS, Bennett DA. Sleep Fragmentation and the Risk of Incident Alzheimer’s Disease and Cognitive Decline in Older Persons. Sleep. 2013;36:102732. [DOI] [PubMed] [PMC]
Shokri-Kojori E, Wang G, Wiers CE, Demiral SB, Guo M, Kim SW, et al. β-Amyloid accumulation in the human brain after one night of sleep deprivation. Proc Natl Acad Sci U S A. 2018;115:44838. [DOI] [PubMed] [PMC]
Kang J, Lim MM, Bateman RJ, Lee JJ, Smyth LP, Cirrito JR, et al. Amyloid-β dynamics are regulated by orexin and the sleep-wake cycle. Science. 2009;326:10057. [DOI] [PubMed] [PMC]
Osorio RS, Ducca EL, Wohlleber ME, Tanzi EB, Gumb T, Twumasi A, et al. Orexin-A is Associated with Increases in Cerebrospinal Fluid Phosphorylated-Tau in Cognitively Normal Elderly Subjects. Sleep. 2016;39:125360. [DOI] [PubMed] [PMC]
Silverberg GD, Heit G, Huhn S, Jaffe RA, Chang SD, Bronte-Stewart H, et al. The cerebrospinal fluid production rate is reduced in dementia of the Alzheimer’s type. Neurology. 2001;57:17636. [DOI] [PubMed]
Furtado A, Esgalhado AJ, Duarte AC, Costa AR, Costa-Brito AR, Carro E, et al. Circadian rhythmicity of amyloid-beta-related molecules is disrupted in the choroid plexus of a female Alzheimer’s disease mouse model. J Neurosci Res. 2023;101:52440. [DOI] [PubMed]
Zeppenfeld DM, Simon M, Haswell JD, D'Abreo D, Murchison C, Quinn JF, et al. Association of Perivascular Localization of Aquaporin-4 With Cognition and Alzheimer Disease in Aging Brains. JAMA Neurol. 2017;74:919. [DOI] [PubMed]
Swaab DF, Fliers E, Partiman TS. The suprachiasmatic nucleus of the human brain in relation to sex, age and senile dementia. Brain Res. 1985;342:3744. [DOI] [PubMed]
Harper DG, Stopa EG, Kuo-Leblanc V, McKee AC, Asayama K, Volicer L, et al. Dorsomedial SCN neuronal subpopulations subserve different functions in human dementia. Brain. 2008;131:160917. [DOI] [PubMed] [PMC]
Cermakian N, Lamont EW, Boudreau P, Boivin DB. Circadian clock gene expression in brain regions of Alzheimer’s disease patients and control subjects. J Biol Rhythms. 2011;26:16070. [DOI] [PubMed]
Song H, Moon M, Choe HK, Han D, Jang C, Kim A, et al. Aβ-induced degradation of BMAL1 and CBP leads to circadian rhythm disruption in Alzheimer’s disease. Mol Neurodegener. 2015;10:13. [DOI] [PubMed] [PMC]
Musiek ES, Lim MM, Yang G, Bauer AQ, Qi L, Lee Y, et al. Circadian clock proteins regulate neuronal redox homeostasis and neurodegeneration. J Clin Invest. 2013;123:5389400. [DOI] [PubMed] [PMC]
Wu Y, Zhou J, Heerikhuize JV, Jockers R, Swaab DF. Decreased MT1 melatonin receptor expression in the suprachiasmatic nucleus in aging and Alzheimer’s disease. Neurobiol Aging. 2007;28:123947. [DOI] [PubMed]
Zhang Z, Xue P, Bendlin BB, Zetterberg H, Felice FD, Tan X, et al. Melatonin: A potential nighttime guardian against Alzheimer’s. Mol Psychiatry. 2025;30:23750. [DOI] [PubMed] [PMC]
Davis KL, Davis BM, Greenwald BS, Mohs RC, Mathé AA, Johns CA, et al. Cortisol and Alzheimer’s disease, I: Basal studies. Am J Psychiatry. 1986;143:3005. [DOI] [PubMed]
Masugi F, Ogihara T, Sakaguchi K, Otsuka A, Tsuchiya Y, Morimoto S, et al. High plasma levels of cortisol in patients with senile dementia of the Alzheimer’s type. Methods Find Exp Clin Pharmacol. 1989;11:70710. [PubMed]
Holleman J, Adagunodo S, Kåreholt I, Hagman G, Aspö M, Udeh-Momoh CT, et al. Cortisol, cognition and Alzheimer’s disease biomarkers among memory clinic patients. BMJ Neurol Open. 2022;4:e000344. [DOI] [PubMed] [PMC]
Pietrzak RH, Laws SM, Lim YY, Bender SJ, Porter T, Doecke J, et al.; Australian Imaging, Biomarkers and Lifestyle Research Group. Plasma Cortisol, Brain Amyloid-β, and Cognitive Decline in Preclinical Alzheimer’s Disease: A 6-Year Prospective Cohort Study. Biol Psychiatry Cogn Neurosci Neuroimaging. 2017;2:4552. [DOI] [PubMed]
Green KN, Billings LM, Roozendaal B, McGaugh JL, LaFerla FM. Glucocorticoids increase amyloid-β and tau pathology in a mouse model of Alzheimer’s disease. J Neurosci. 2006;26:904756. [DOI] [PubMed] [PMC]
Dronse J, Ohndorf A, Richter N, Bischof GN, Fassbender R, Behfar Q, et al. Serum cortisol is negatively related to hippocampal volume, brain structure, and memory performance in healthy aging and Alzheimer’s disease. Front Aging Neurosci. 2023;15:1154112. [DOI] [PubMed] [PMC]
Eskildsen SF, Coupé P, Fonov VS, Pruessner JC, Collins DL; Alzheimer's Disease Neuroimaging Initiative. Structural imaging biomarkers of Alzheimer’s disease: predicting disease progression. Neurobiol Aging. 2015;36:S2331. [DOI] [PubMed]
Jacobson L, Sapolsky R. The role of the hippocampus in feedback regulation of the hypothalamic-pituitary-adrenocortical axis. Endocr Rev. 1991;12:11834. [DOI] [PubMed]
Cole AB, Montgomery K, Bale TL, Thompson SM. What the hippocampus tells the HPA axis: Hippocampal output attenuates acute stress responses via disynaptic inhibition of CRF+ PVN neurons. Neurobiol Stress. 2022;20:100473. [DOI] [PubMed] [PMC]
O'Brien JT, Ames D, Schweitzer I, Mastwyk M, Colman P. Enhanced adrenal sensitivity to adrenocorticotrophic hormone (ACTH) is evidence of HPA axis hyperactivity in Alzheimer’s disease. Psychol Med. 1996;26:714. [DOI] [PubMed]
Chatterjee P, Pedrini S, Stoops E, Goozee K, Villemagne VL, Asih PR, et al. Plasma glial fibrillary acidic protein is elevated in cognitively normal older adults at risk of Alzheimer’s disease. Transl Psychiatry. 2021;11:27. [DOI] [PubMed] [PMC]
Muramori F, Kobayashi K, Nakamura I. A quantitative study of neurofibrillary tangles, senile plaques and astrocytes in the hippocampal subdivisions and entorhinal cortex in Alzheimer’s disease, normal controls and non-Alzheimer neuropsychiatric diseases. Psychiatry Clin Neurosci. 1998;52:5939. [DOI] [PubMed]
Wyss-Coray T, Loike JD, Brionne TC, Lu E, Anankov R, Yan F, et al. Adult mouse astrocytes degrade amyloid-beta in vitro and in situ. Nat Med. 2003;9:4537. [DOI] [PubMed]
Sompol P, Furman JL, Pleiss MM, Kraner SD, Artiushin IA, Batten SR, et al. Calcineurin/NFAT Signaling in Activated Astrocytes Drives Network Hyperexcitability in Aβ-Bearing Mice. J Neurosci. 2017;37:613248. [DOI] [PubMed] [PMC]
Goedert M, Crowther RA, Scheres SHW, Spillantini MG. Tau and neurodegeneration. Cytoskeleton (Hoboken). 2024;81:95102. [DOI] [PubMed] [PMC]
Ghanizada H, Nedergaard M. The glymphatic system. Handb Clin Neurol. 2025;209:16170. [DOI] [PubMed]
Yiannopoulou KG, Papageorgiou SG. Current and Future Treatments in Alzheimer Disease: An Update. J Cent Nerv Syst Dis. 2020;12:1179573520907397. [DOI] [PubMed] [PMC]
Cite this Article
Export Citation
van der Zwet LCA, Michel S, Deboer T. Glucocorticoid signalling coordinating circadian rhythms between the glymphatic system and sleep contributes to brain health. Explor Endocr Metab Dis. 2025;2:101437. https://doi.org/10.37349/eemd.2025.101437
Article Metrics

View: 422

Download: 22

Times Cited: 0