Next Article in Journal
Integrated Evaluation of Dual-Functional DPP-IV and ACE Inhibitory Effects of Peptides Derived from Sericin Hydrolysis and Their Stabilities during In Vitro-Simulated Gastrointestinal and Plasmin Digestions
Next Article in Special Issue
Preparation, Identification and Preliminary Application of the Fenvalerate Monoclonal Antibody in Six Kinds of Dark Tea
Previous Article in Journal
Bioaccessibility and Intestinal Transport of Tebuconazole in Table Grape by Using In Vitro Digestion Models
Previous Article in Special Issue
Adsorption Behavior of the L-Theanine onto Cation Exchange Resin ZGSPC106Na and D001SD
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

New Perspectives on Sleep Regulation by Tea: Harmonizing Pathological Sleep and Energy Balance under Stress

1
Key Laboratory of Tea Science of Ministry of Education, Changsha 410128, China
2
National Research Center of Engineering and Technology for Utilization of Botanical Functional Ingredients, Changsha 410128, China
3
Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Changsha 410128, China
4
College of Physical Education, Hunan City University, Yiyang 413002, China
*
Author to whom correspondence should be addressed.
Foods 2022, 11(23), 3930; https://doi.org/10.3390/foods11233930
Submission received: 2 November 2022 / Revised: 25 November 2022 / Accepted: 26 November 2022 / Published: 5 December 2022

Abstract

:
Sleep, a conservative evolutionary behavior of organisms to adapt to changes in the external environment, is divided into natural sleep, in a healthy state, and sickness sleep, which occurs in stressful environments or during illness. Sickness sleep plays an important role in maintaining energy homeostasis under an injury and promoting physical recovery. Tea, a popular phytochemical-rich beverage, has multiple health benefits, including lowering stress and regulating energy metabolism and natural sleep. However, the role of tea in regulating sickness sleep has received little attention. The mechanism underlying tea regulation of sickness sleep and its association with the maintenance of energy homeostasis in injured organisms remains to be elucidated. This review examines the current research on the effect of tea on sleep regulation, focusing on the function of tea in modulating energy homeostasis through sickness sleep, energy metabolism, and damage repair in model organisms. The potential mechanisms underlying tea in regulating sickness sleep are further suggested. Based on the biohomology of sleep regulation, this review provides novel insights into the role of tea in sleep regulation and a new perspective on the potential role of tea in restoring homeostasis from diseases.

Graphical Abstract

1. Introduction

Sleep, a spontaneous and reversible resting state in mammals, is essential in optimizing energy conservation or allocation, regulating core molecular and cellular processes, and enhancing brain functions [1,2,3]. Both quality and quantity are fundamental components of sleep. Poor sleep quality contributes to chronic diseases such as kidney disease, hypertension, obesity, and diabetes mellitus [4,5,6]. Meanwhile, lack of sleep can induce substantial short- and long-term memory impairment and is a risk factor for anxiety and depression [7,8,9,10].
There are two types of sleep: natural sleep and sickness sleep. Sickness sleep is the sensation of being sleepy and fatigued under stress, infection, or disease conditions. In lower animals, sickness sleep, known as stress-induced sleep (SIS), has a non-negligible role in supporting recovery from injury [11]. In Caenorhabditis elegans, ALA neuron-dependent SIS is important to increase survival after cellular stress [12]. Increasing sleep increases survival after oxidative challenge in Drosophila [13]. Sleep plays an equal role in recovery from exhaustion and illness in higher organisms. In mice, deeper resting leads to a faster recovery [14]. In a symptom assessment of 59 burnt-out employees taking extended sick leave, sleep was found to play an important role both in symptom improvement and in return to work [15].
Energy homeostasis is essential for the normal physiological activities of the body. Damage repair is induced by stress and promotes sleep; however, it also results in a disequilibrium of energy homeostasis due to increased energy requirements. Cellular energy charge and glycogen levels, induced by energy metabolism, may operate as signaling mechanisms. This may be when energy stores reallocate energy away from high-energy demands and gene expression during the waking state toward biological processes favoring sleep [16].
As one of the most widely consumed non-alcoholic beverages, tea has numerous recognized health benefits, such as resistance to stress and sleep regulation [17,18]. Tea effectively improves resistance to various stresses, including heat, oxidative, and ultraviolet (UV) stress [19,20,21]. Oolong tea consumption has been reported to improve stress symptoms and alleviate the elevated plasma lipid peroxidation levels caused by nighttime stress in study subjects [22]. Green tea has anti-stress effects, in which theanine, Epigallocatechin (EGC), and arginine synergistically eliminate the antagonistic effects of caffeine and Epigallocatechin gallate (EGCG) on adrenal hypertrophy induced by psychosocial stress in mice [23]. Sleep duration and quality are affected by the frequency and amount of tea consumed [24,25,26,27,28]. A recent study that investigated the effects of tea on alleviating acute alcohol intoxication (AAI) in mice found that tea reduced AAI and regulated sleep by inhibiting oxidative stress and inflammation [29].
Obesity and diabetes are caused by the imbalance of the body’s energy homeostasis. Tea has been found to reduce obesity and ease diabetes. Oolong tea extract reduces lipid accumulation by regulating lipid metabolism and intestinal flora distribution, thus inhibiting high-diet-induced obesity in mice [30]. Green tea components are involved in protein interactions and cell signaling pathways that regulate energy metabolism, including glycogen synthesis and glucose reabsorption, thereby reducing glycogen accumulation and improving the pathological features in diabetic mice [31].
Previous studies have illustrated the role of tea in regulating stress injury response, healthy sleep, and energy homeostasis; whether and how tea regulates sickness sleep under stress or illness remains unclear. Therefore, this review covers recent discoveries on the effect of tea on sickness sleep and explores whether the chemical properties and consumption of tea can influence energy homeostasis and sickness sleep in stressful environments. This review also explores the potential link between tea consumption and the effects of sleep under stress.

2. Tea and Sleep

2.1. Tea

Tea has originated in China and since spread worldwide. People in more than 160 countries and regions habitually drink tea. Based on the degree of fermentation during processing, tea is classified into six types: green, yellow, white, oolong, dark, and black. Tea is rich in active ingredients, including polyphenols, amino acids, alkaloids, aromatic substances, and sugars [32]. Table 1 lists the degree of fermentation, key processes, and recent studies related to the respective health benefits of tea. It is worth noting that despite the different processing processes of the six tea types, the health benefits are all centered on their antibacterial and antiviral activities; neuroprotection; protection against cancer, obesity, diabetes, and cardiovascular diseases; and sleep regulation [33].

2.2. Sleep

Sleep is primarily classified as either natural or sickness, of which natural sleep is regulated by the interaction of circadian and homeostatic mechanisms [11]. In mammals and avian species, natural sleep is identified based on altered brain electrical activity, recorded on an electroencephalogram (EEG); it comprises non-rapid eye movement sleep (NREMS) and rapid eye movement sleep (REMS) [54]. During NREMS, the EEG pattern shows slow oscillations, referred to as slow waves, and the muscle tone and brain activity are at rest [55,56,57]. Conversely, REMS (also termed fast-wave sleep) is characterized by reduced amplitude and faster frequency in cortical EEG [56]. Mammals breathe irregularly and with relaxed muscles in REMS, which have specific effects on the respiratory system and motor neurons [58,59].
As non-mammalians do not have differentiated EEG, sleep is usually defined according to the following behavioral states: spontaneous circadian motion quiescence, decreased reactivity, increased arousal threshold, and rapid reversibility [60,61]. In zebrafish, the sleep state is characterized by a reversible circadian rhythm and an increased arousal threshold [62,63]. In Drosophila, sleep is typically characterized as an inactive period spanning 5 min or more and heightened arousal thresholds [64,65]. Developmentally timed sleep (DTS) in C. elegans is the 2–3 h of quiescence during the transition between larval stages [60].

2.3. Sickness Sleep

Sickness sleep occurs in stressful environments or diseases as a sensation of being sleepy and fatigued. Sleepiness is common following traumatic injury, particularly traumatic brain injury, with more severe injuries resulting in greater sleepiness in humans [66]. Inflammation has been suggested as a potential contributor to the development of fatigue [67]. Stress activates sleep-related brain regions and induces sleep-like inactivity in mice [68]. Murine gammaherpesvirus 68 alters sleep, activity, and temperature in a manner suggestive of fatigue in mice [69]. Sickness sleep is easier to induce in non-mammals and is called SIS. The sleep duration in Drosophila melanogaster increases following aseptic injury [70]. Long-term radiofrequency radiation exposure enhances the heat stress response and affects the expression of the circadian clock and neurotransmitter genes, thereby prolonging sleep duration [71]. SIS in C. elegans can be triggered by conditions of cellular stress, including noxious heat, cold, hyperosmolarity, ultraviolet irradiation, and mechanical injury [72], and exhibits behavioral quiescence similar to DTS [73]. Sleep is a conserved evolutionary behavior of organisms, whether mammals or non-mammals, to adapt to changes in the external environment [74].

3. Tea Effects on Sleep

Previous studies have found that people suffer from insomnia and fatigue after consuming a high intake of green tea [75]. Short sleep duration was associated with a higher intake of black tea [76]. However, other studies have suggested that tea can calm nerves and promote sleep [25,77]. For example, after consuming black tea with γ-aminobutyric acid (GABA), the sleeping time with sodium pentobarbital was significantly prolonged, and the quality was improved in mice [78]. Green tea extracts can improve sleep disturbances and stabilize mood in humans [79,80]. Fragrant compounds in oolong tea have tranquilizing effects on the brains of mice [81]. Different active ingredients in tea have various implications for sleep (as shown in Figure 1).
Tea polyphenols play an essential role in regulating sleep and mood [82]. After consuming polyphenols, sleep duration is prolonged, and the quality is significantly improved in humans [83]. Meanwhile, tea polyphenols treatment effectively enhances cognitive impairment, memory impairment, and anxiety-like behaviors in sleep-disordered mice [82,84].
Theanine, the main amino acid in tea, promotes sleep. Studies have shown that following treatment with L-theanine, the Pittsburgh Sleep Quality Index (PSQI) subscale scores for sleep latency and satisfaction are improved in participants with no major psychiatric illness [85,86]. L-theanine increases sleep duration and shorten sleep latency in mice [87], while L-theanine/GABA mixture significantly increases NREMS and REMS in the mice [88].
Caffeine, the primary alkaloid in tea, is used to combat high sleep pressures. After 10 days of caffeine intake, a significant reduction in the gray matter volume in the medial temporal lobe was observed [89]. Oral caffeine administration significantly increases sleep latency and decreases the amount of NREMS in mice [90].
Tea contains various aromatic substances. Although natural aromatic components are not in high concentration in tea, they can be used as sleep aids [91]. One of the aromatic substances abundantly present in tea is linalool, which has the aroma of lily or magnolia flowers, and Linalyl acetate has the aroma of green lemons. Studies have shown that essential oils composed of linalool and linalyl acetate can significantly reduce sleep latency and prolong sleep duration in mice [92]. Jasmine lactone, an aromatic substance, constitutes oolong tea’s floral and fruity aroma. Studies have shown that inhalation of cis-jasmone or methyl jasmonate significantly increases the sleeping time in mice induced by pentobarbital and has a tranquilizing effect on their brains [81].

4. Does Tea Affect Sickness Sleep and Maintain Energy Homeostasis?

The relationship between sleep and energy is well-discussed, and the energy allocation model proposes that animal species share a universal sleep function. Sleep conserves energy by (i) reducing energy requirements for core thermoregulatory defenses and skeletal muscle tone when the external environment changes and (ii) enhancing energy appropriation for somatic and CNS-related processes [16,93,94].
Energy homeostasis in animals is maintained through energy metabolism via a dynamic balance between consumption and supply. However, animals in complex environments are subjected to daily stresses, such as solar radiation, pathogenic bacterial infections, and mechanical damage. Therefore, they suffer from organismal damage and must increase their energy expenditure for damage repair, thus breaking the energy balance for a short period. Therefore, animals have evolved sickness sleep behaviors to maintain energy homeostasis in stressful environments, a strategy for conserving energy consumption and optimizing energy allocation [2,54,74,95].
Many studies have shown that tea effectively improves stress resistance, including heat, oxidative, and UV [19,20,21]. Previous research in the laboratory found that an aqueous extract of black tea promoted SIS and prolonged the lifespan of C. elegans compared with the control group under ultraviolet irradiation. This suggests a link between lifespan and SIS, in which tea may play a role. However, the mechanism by which tea regulates sickness sleep remains unclear. In this review, a possible mechanism for this phenomenon is proposed. Tea may promote sickness sleep and maintain energy homeostasis, stimulating the organism’s health by acting on damage repair and energy metabolism (Figure 2).

4.1. Tea Acts on the Brain–Gut Axis to Regulate Sickness Sleep

The active substances of tea are known to act on the brain–gut axis to regulate sleep [96]. This review summarizes the relevant research and hypotheses regarding tea regulation of sickness sleep under stress via the brain–gut axis (Figure 3).

4.1.1. Tea Acts on the Nervous System to Regulate Sickness Sleep

The nervous system is mainly composed of neurons and glia wrapped around neurons. Neurotransmitters are released between synapses to convey information that controls sleep behavior. Neurons regulate sleep and waking [95,97]. Two types of neurons that control sleep have been identified: sleep-active neurons, such as GABAergic/peptidergic neurons in the preoptic area of mammals, and sleep-promoting neurons, such as RIM and PVC neurons in C. elegans [56,98]. Tea effectively protects neuronal tissues and promotes growth and differentiation under stressful conditions. A previous study showed that oolong tea extracts had a protective effect against the death of neuronal cells (Neuro-2a and HT22) [99]. This is because oolong tea extracts reduce the accumulation of intracellular reactive oxygen species (ROS) and induce gene expression of cellular antioxidant enzymes. These extracts also increased average neurite length in Neuro-2a cells. Another study found that theaflavins enhanced PC12 cell survival following H2O2-induced toxicity and increased cell viability [45]. This study suggested that the neuroprotective effects of theaflavins against oxidative stress in PC12 cells are derived from the suppression of oxidant enzyme activity. Furthermore, green tea protects against hippocampal neuronal apoptosis by inhibiting the JNK/MLCK pathway [100]. The catechins EGCg and GCg effectively protect nerve cells against H2O2 or Aβ1-42-induced injury [101,102]. EGCg and its degradation products induce neuronal differentiation and neurite outgrowth by upregulating synaptophysin gene expression and reducing DNA methylation [103,104]. Water-soluble flavonoids enhance the neuronal differentiation of neural stem cells in a dose-dependent manner and significantly enhance neurite growth in mice [105].
Glial cells, including astrocytes, oligodendrocytes, and microglia, have neurotransmitter receptors and ion channels wrapped in neurons and play an essential role in regulating behaviors, such as movement and sleep [106]. Astroglial calcium activity changes dynamically across vigilance states and is proportional to sleep requirements. Astrocytic Gi- and Gq-coupled G-protein-coupled receptor signaling controls NREMS depth and duration, respectively [107,108,109]. Depleting microglial cells disrupts the brain tissue’s circadian rhythmicity, increases the duration of NREMS, and reduces hippocampal excitatory neurotransmission in mice [110,111]. Tea protects glial cells under stress. The pathological activation of astrocytes and other glia in mice was inhibited by green tea extract [112,113]. Chlorogenic acid increased ω-7 palmitoleic fatty acid, which was associated with an IL-6 decrease, and effectively alleviated inflammation of glial cells in mice [113,114].
Neurotransmitters mainly include acetylcholine, monoamines (dopamine and serotonin), amino acids (excitatory transmitters, such as glutamate, and inhibitory transmitters, such as GABA), and neuropeptides. Multiple neurotransmitters regulate the sleep-wake cycle [115]. FMRFamide (Phe-Met-Arg-Phe-NH2), also known as FMRFamide-related neuropeptides, and their receptors play a conserved and vital role in regulating SIS in response to cellular stress [11,70]. In C. elegans, heat stress-induced sleep requires ALA depolarization and the release of FMRFamide-like neuropeptides release encoded by the flp-13 gene [116]. Tea can regulate neurotransmitter levels [117,118]. Theanine is a derivative of glutamine that is structurally analogous to glutamate. It inhibits glutamine uptake in the glutamine-glutamate cycle via SLC38A1. This controls the balance of glutamate and glutamine in the brain to regulate sleep [119,120]. Additionally, theanine significantly increased the concentrations of acetylcholine and GABA and decreased the concentration of serotonin in the brain [87,121]. Polyphenols can prevent the reuptake of monoamine neurotransmitters and increase cerebral blood flow [122]. Aromatic substances, such as homeopathic jasmine, jasmine lactone, linalool oxide, and methyl jasmonate, significantly enhanced the expression of GABA receptors in Xenopus oocytes and increased the content of 5-HT and GABA in mouse brains [81,92].

4.1.2. Tea Regulates Intestinal Flora to Mediate Sickness Sleep

The intestinal microbiota regulates host sleep and mental states through the microbiota-gut-brain axis [123]. In antibiotic-induced microbiota-depleted mice, the time spent in NREMS was reduced, while the number of REMS episodes increased; this was accompanied by frequent transitions from NREMS to REMS [124]. In addition, compared with healthy individuals, people with poor sleep quality have a higher relative abundance of Firmicutes and a lower relative abundance of Bacteroidetes in the intestinal flora [125,126,127]. Bioactive metabolites produced by the intestinal flora play an essential role in sickness sleep regulation. Lipopolysaccharide (LPS), lipoteichoic acid (LTA), and peptidoglycans induce sleep, fever, and anorexia. LTA may play a role in developing disease responses to gram-positive bacterial infections and in sleep signaling through the commensal gut microbiota [128].
Tea can effectively improve the proportion of beneficial to harmful microorganisms and regulate microbial diversity and metabolite production in the gut. Oolong, yellow, black, and dark teas have been reported to regulate intestinal microbiota. Black tea significantly promotes gastrointestinal transit and colonization of beneficial Bifidobacterium, Lactobacillus, and Bacteroides and inhibits the growth of harmful Firmicutes, Escherichia coli, and Enterococcus [129,130,131]. Yellow tea extracts altered the gut microbiota composition and increased community diversity and richness [132]. Green tea increases the abundance of Flavonifractor plautii (FP) in the gut microbiota, and LTA from FP was identified as the active component mediating IL-17 inhibition [133]. Tea compounds affect the growth of bacterial species involved in inflammatory processes, such as the release of LPS [134]. ECg promotes the release of LTA from the plasma membrane of Staphylococcal cells [135]. L-theanine improved intestinal dysbiosis by decreasing the ratio of Firmicutes/Bacteroidetes, along with increased fecal SCFA concentrations [136].

4.2. Tea Acts on Damage Repair to Mediate Sickness Sleep

Damage repair is considered a biological function of SIS. Sleep regulates repair mechanisms and immune responses in the body and promotes neural repair, metabolite clearance, and circuit reorganization [137]. In addition, sleep affects both humoral and cellular immunities. In mice, it significantly increases the number of monocytes in the blood and spleen and enhances the ability of monocytes and neutrophils to produce reactive oxygen species (ROS) [138]. Recent research on the interaction between sleep and damage repair has found that melatonergic regulators, which regulate circadian rhythms and sleep, inhibit the DNA damage response and activate the RAS/MAPK signaling pathways [139]. The antimicrobial peptide (AMP) neuropeptide-like protein (NLP)-29 in C. elegans acts through the neuropeptide receptor NPR-12 in locomotion-controlling neurons RIM and PVC, which are presynaptic to RIS neurons and depolarize this to induce sleep [140].

4.2.1. DNA Damage Repair

External carcinogens and endogenous cellular processes cause DNA damage. The major endogenous sources of DNA damage are errors in DNA replication and spontaneous chemical changes produced by ROS, carbonyl stress, and hydrolysis of the glycosylic bonds. Exogenous damage is caused by exogenous agents, such as ultraviolet radiation, ionizing radiation, and various chemicals [141,142,143,144]. In long-term evolution, organisms have developed a series of DNA damage repair mechanisms, such as DNA repair and DNA damage response (DDR), to maintain genetic integrity [142,145,146].
The components of tea may play a role in DNA damage repair mechanisms. Regular intake of tea polyphenols rescued UVB-induced miR-29 depletion and prevented tumor growth by maintaining reduced DNA hypermethylation [147]. EGCg and its degradation products ensure normal neuronal differentiation and synaptic growth by reducing DNA methylation of the synaptophysin promoter [103,104]. In addition, EGCg acts as an antioxidant that protects embryos from oxidative damage by restoring the expression of ribosome/tumor-related proteins [148]. The antibacterial activity of green tea catechins results from various mechanisms, including DNA damage [149].
Previous studies have found a strong association between DNA damage repair and sleep. For instance, in zebrafish and mice, the activity of the DDR initiator poly polymerase 1 (Parp1) increases following sleep deprivation, while the activity of the DDR proteins, Rad52 and Ku80, increases during sleep, revealing that DNA damage triggers sleep [150]. CEP-1, which encodes proteins in the DDR pathway, acts downstream or parallel to ALA activation to promote SIS in C. elegans [151]. Sleep also promotes the repair of DNA damage. Participants who worked overnight after sleep deprivation had a lower baseline DNA repair gene expression and more DNA breaks [152]. Sleep increases chromosome dynamics, which is necessary to reduce the number of DNA double-strand breaks in zebrafish [153].

4.2.2. Immune Response

To maintain homeostasis, animals have evolved mechanisms that include physical barriers and behavioral and immune responses to defend against and eliminate pathogen infection [154]. During an immune response, various cytokines drive immune cells to sites of infection for clearance and repair. The inflammatory response to treatment with pro-inflammatory cytokines is mediated by p38 mitogen-activated protein kinase (MAPK) [155,156,157,158]. Systemic inflammation significantly affects metabolism and induces characteristic sleep responses [159]. Pro-inflammatory cytokines such as tumor necrosis factor-alpha (TNFα) and interleukin-1 (IL-1) are assumed to mediate increased sleep under inflammatory conditions [160]. TNFα knockout mice had increased REMS, and IL-1 receptor accessory protein (AcP) is required for NREMS [161].
Tea regulates pro-inflammatory cytokines during inflammatory responses. Studies have shown that Pu-erh tea and white tea act on the p38/MAPK pathway to mediate inflammatory responses [162,163,164]. Tea polyphenols can effectively alleviate inflammation by downregulating the level of TNFα-converting enzymes and reducing the expression of pro-inflammatory cytokines, such as IL-1β. L-theanine has been suggested to inhibit heat stress-induced imbalance in oxidative stress and inflammatory responses by reducing inflammatory factors, such as TNF-α, IL-6, and IL-1β, via the p38/MAPK pathway [84,113,114]. In addition, theanine reduced synaptic scaling by downregulating TNFα-induced AMPA receptor phosphorylation, which upregulated Homer1a expression, thereby improving sleep [19].

4.3. Tea Acts on Energy Metabolism to Mediate Sickness Sleep

The energy metabolism pathways mainly include lipid metabolism, adenosine monophosphate-activated protein kinase (AMPK), insulin/IGF-1 signaling (IIS), and the mammalian/mechanistic target of rapamycin (mTOR) signaling pathways. Studies have found that individuals with obesity showed lower fat oxidation and higher carbohydrate oxidative catabolism during sleep and experienced shorter sleep duration than normal-weight individuals. This indicates that energy metabolism and sleep are mutually regulated [165].

4.3.1. Lipid Metabolism

Lipid metabolism can regulate sleep in living organisms. Brown adipose tissue and uncoupling proteins are essential for maintaining energy homeostasis and body temperature. Studies have shown that the pharmacological activation of brown adipose tissue promotes sleep. Uncoupling protein 1 (UCP1), which promotes thermogenesis in brown adipocytes, is necessary for increasing NREMS [166]. In C. elegans, the transcription factor ETS-5 promotes roaming and inhibits quiescence by regulating a complex network of serotonergic and neuropeptide signaling pathways through fat regulation [167].
Lipid metabolism is also regulated during sleep. Long sleep durations were significantly associated with low high-density lipoprotein (HDL) cholesterol levels [168]. Morning-to-evening-regulated pathways of carbohydrate and lipid metabolism are sensitive to sleep loss [169]. Lipid levels in Drosophila are altered considerably during sleep [159]. Recently, weight loss during sleep has become an important issue. Studies have found that sleep extension can significantly reduce energy intake and result in a negative energy balance to reduce weight [170].
Tea has been shown to regulate lipid metabolism. Yellow and oolong tea inhibits obesity by increasing energy expenditure and fatty acid oxidation [171]. In contrast, green, white, Fuzhuan, and raw Pu-erh tea inhibit fatty acid synthesis [171]. Among them, green tea leaf powder reduces body weight and total cholesterol in mice on a high-fat diet (HFD) by decreasing the expression of fatty acid synthase and sterol regulatory element binding protein-1c (SREBP-1c) [172]. Pu-erh tea treatment significantly reduces free fatty acid (FFA) synthesis and increases the expression of genes involved in FFA uptake and β-oxidation in HFD-induced obese mice [173,174]. L-theanine promotes the metabolic activity of brown adipose tissue and subcutaneous white fat by enhancing thermogenic gene expression [136].

4.3.2. AMPK

AMPK, an AMP-activated protein kinase that controls cellular metabolic decisions, is activated by increasing the AMP/ATP ratio in the body under cellular stress, exercise, and hormones. AMPK mediates the interaction between energy and sleep. The AMPK/SIRT1/PGC-1α pathway regulates the expression of skeletal muscle clock genes and the circadian locomotor output cycle kaput (Clock) [175]. AMPK knockdown in neuropeptide leucokinin (Lk) neurons inhibits sleep in Drosophila [176]. Salt-inducible kinases (SIKs) are essential members of the AMPK family. A SIK family kinase 3 (SIK3) deletion mutation in a well-conserved protein kinase A (PKA) phosphorylation site, S551, caused a lethargic phenotype in mice, characterized by a reduced wake time and increased NREMS time and delta density [177,178]. In addition, a gain-of-function sleepy mutation in SIK3 can also increase NREMS power and amount [179]. KIN-29 in C. elegans is a homolog of SIK, which acts upstream of fat regulation and sleep-controlling neurons to transduce low cellular energy charges into the mobilization of fat stores, thus promoting sleep [180].
Tea acts on AMPK, which mediates energy metabolism and affects physiological activities, including lipid and glucose metabolism. Yellow and raw Pu-erh tea significantly upregulated AMPK (p-AMPK) in HFD-induced obese mice [171]. Green, yellow, and black tea combined with citrus, can activate the AMP-activated protein kinase (AMPK)/acetyl-CoA carboxylase (ACC) signaling pathway and upregulate the expression of p-AMPK, p-ACC, and CPT-1 proteins, thereby inhibiting fat accumulation [181]. The combination of white tea and jiaogulan significantly suppresses hepatic glucose 6-phosphatase (G6Pase) expression by activating the AMPK pathway, thereby inhibiting gluconeogenesis [162]. Catechins, which increase AMPK activity and reduce ACC activity in metabolic tissues, affect lipid metabolism by reducing triglyceride levels and lipid droplet formation [182,183].

4.3.3. IIS Signaling Pathway

The insulin/IGF signaling pathway (IIS), an evolutionarily conserved hormonal pathway, comprises insulin, insulin-like growth factor (IGF), or insulin-like peptide, insulin receptor IR/IGFR, serine-threonine kinase AKT, and downstream target forkhead box O (FOXO) transcription factors. It is vital in regulating energy metabolism, growth, and stress resistance [184,185].
Tea can regulate energy metabolism by activating downstream targets through the IIS pathway. Tea polyphenolics reportedly possess blood glucose-lowering properties by improving insulin sensitivity [186]. One study has shown that green tea extract attenuates downstream signaling of the insulin-like growth factor receptor [187]. In C. elegans, complex I inhibition by EGCg and ECg induced a transient drop in cellular ATP levels and a temporary ROS burst, resulting in SKN-1 and FOXO/DAF-16 activation [188]. Linalool, an aroma substance in tea, activates downstream sod-3 and hsp-12.6 gene expression through FOXO/DAF-16 and affects fat accumulation in C. elegans [189]. Tea also regulates the IIS pathway to play an anti-oxidative stress role. Oolong tea extract enhanced IGF/IR/p-AKT mechanism in the IIS pathway to aid cellular adaptation against hypoxic challenges [47].
The IIS pathway plays a vital role in sleep regulation. In the treatment of patients with circadian rhythm sleep-wake disorders, improvements in symptoms were found to be strongly associated with increased serum concentrations of insulin-like growth factor (IGF-1) [190]. In-depth studies have found that the activity of orexin neurons is modulated by IGF-1, which controls sleep duration and architecture [191]. In Drosophila, insulin-like peptide 2 (DILP2) is required for starvation-induced changes in sleep depth [192]. The adipokinetic hormone (AKH)-FOXO pathway has been shown to respond to energy changes and adjust Drosophila’s sleep by remodeling the dorsal projections of the small ventral lateral neurons [193]. In C. elegans, RIS neurons are activated by the conserved insulin receptors IR/DAF-2 and FOXO/DAF-16, thereby inducing sleep to conserve energy during extended food deprivation [194,195].

4.3.4. mTOR Pathway

The mTOR is a serine/threonine kinase. The mTOR signaling pathway, the downstream target of AKT in the IIS pathway, plays a vital role in integrating nutrition, energy metabolism, growth, and proliferation [196,197]. Recently, the mTOR signaling pathway was found to be closely related to sleep. Both mTOR activity and orexin expression were increased in the hypothalamic sections and cultured hypothalamic neurons of Tsc1GFAPCKO mice, which showed sleep disorders. Both sleep abnormalities and increased orexin expression restore mTOR activation [198]. Food deprivation can inhibit SIS through the target of rapamycin and transforming growth factor-β (TGF-β) nutrient signaling pathways in C. elegans [199].
Tea affects energy metabolism through the mTOR signaling pathway. Fuzhuan tea aqueous extract alleviated insulin resistance by activating the insulin signaling Akt/GLUT4, FOXO1, and mTOR/S6K1 pathways in the skeletal muscles [200]. Studies have shown that tea may regulate neuro-mechanisms through the mTOR signaling pathway. A study found that polyphenols significantly improve sleep deprivation-induced contextual memory deficits, possibly through activating the cAMP-response element binding protein (CREB) and mTOR signaling pathways [201]. Theanine can upregulate SLC38A1 expression to activate the intracellular mTOR signaling pathway required to replicate and form neurons and neuron orientation [202]. In contrast, tea polysaccharides repressed the proliferation of colon cancer line HCT116 cells by targeting lysosomes to induce cytotoxic autophagy, which might be achieved through mTOR- transcription factor EB (TFEB) signaling [203].

5. Conclusions and Remarks

This review summarizes the role and mechanisms of tea in regulating sickness sleep. The potential efficacy of tea in promoting recovery under stress is further discussed, and the mechanisms by which tea regulates sickness sleep and maintains energy homeostasis are postulated.
Firstly, the regulation of sickness sleep is related to the brain–gut axis as well as damage repair. Among them, the brain–gut axis directly regulates sickness sleep, and damage repair acts as a sleep motive to indirectly induce sickness sleep. Tea may have the potential to regulate sickness sleep with its vital function in relieving stress and regulating healthy sleep. However, current research on tea in sickness sleep is deficient, and it is unclear whether and how tea regulates sickness sleep. Based on the mechanisms of sickness sleep regulation and the health benefits of tea, possible mechanisms of tea modulation of sickness sleep have been proposed, including acting on the nervous system, regulating intestinal flora, mediating DNA damage repair, and immune response. Secondly, according to the role of sickness sleep in promoting energy homeostasis, this review discusses the role of tea in linking sickness sleep and energy homeostasis, with lipid metabolism and AMPK, IIS, and mTOR pathways as potential targets.
The role and regulatory mechanisms of sickness sleep are more extensively studied in lower organisms. Owing to the more complex regulatory mechanisms, the role of sickness sleep is still not well elucidated in higher organisms. Excessive sleepiness under stress or illness has been found to be detrimental to patient recovery; hence, exploring the role of tea in regulating sickness sleep cannot be too one-sided. Further animal experiments and clinical validation are still needed in the future to focus on systematically and comprehensively describing the positive and negative effects of tea on sleep regulation under different conditions. Negative emotions under stress and in illness, such as anxiety and depression, can affect biological sleep while being detrimental to health recovery. Theanine in tea acts as a sedative and calming agent, and aromatic substances are widely used as sleep aids. Therefore, it would be a promising application to develop the beneficial components of tea into a calming and tranquilizing product and use it for patient recovery.
Whether, and how, sickness sleep and energy homeostasis under stress are influenced by tea warrants further investigation. Mitochondria may be an entry point to explore this question. Owing to the intermediary role of mitochondria in energy metabolism and response to stress, research on how tea affects mitochondrial function during sleep regulation, including mitochondrial dynamics, mitochondrial autophagy, and mitochondrial biosynthesis, might be a great direction for future research.
Overall, new insights on tea regulation of sickness sleep will enrich the health benefits of tea, while the potential of tea to harmonize sleep and energy balance under stress will provide insights into improving disease treatment recovery. A warm cup of tea and a nap during stress or discomfort may provide enough energy for a better recovery.

Author Contributions

Writing—original draft, J.O.; Writing—review & editing, Y.G.; Conceptualization, J.O. and Y.G.; Visualization, Y.P.; Funding acquisition, Y.G. All authors have read and agreed to the published version of the manuscript.

Funding

National Natural Science Foundation of China, grant number: 32072155; The Key R&D Program of Hunan Province, grant number: 2020NK2047; Scientific Research Fund of Hunan Provincial Education Department, grant number: 20C0346.

Data Availability Statement

The data that support the findings of this study are available from the corresponding author upon reasonable request.

Conflicts of Interest

The authors declare that the publication of this paper has no conflict of interest.

Abbreviations

List of abbreviations
AcronymFull name
UVUltraviolet
EGCEpigallocatechin
EGCGEpigallocatechin gallate
AAIAcute alcohol intoxication
EEGElectroencephalogram
NREMSNon-rapid eye movement sleep
REMSRapid eye movement sleep
DTSDevelopmentally timed sleep
SISStress-induced sleep
GABAγ-aminobutyric acid
PSQIPittsburgh sleep quality index
ROSReactive oxygen species
LPSLipopolysaccharide
LTALipoteichoic acid
FPFlavonifractor plautii
SCFAShort-chain fatty acid
AMPAntimicrobial peptide
NLPNeuropeptide-like protein
DDRDNA damage response
MAPKMitogen-activated protein kinase
TNFαTumor necrosis factor-alpha
IL-1Interleukin-1
AMPKAdenosine monophosphate-activated protein kinase
IISInsulin/IGF-1 signaling
mTORMammalian/mechanistic target of rapamycin
UCP1Uncoupling protein 1
HDLHigh-density lipoprotein
HFDHigh-fat diet
SREBP-1cSterol regulatory element binding protein-1c
FFAFree fatty acid
LkLeucokinin
SIKsSalt-inducible kinases
PKAProtein kinase A
ACCAcetyl-CoA carboxylase
G6PaseGlucose 6-phosphatase
IGFInsulin-like growth factor
FOXOForkhead box O
AKHAdipokinetic hormone
CREBcAMP-response element binding protein
TFEBTranscription factor EB
mtROSMitochondrial reactive oxygen species

References

  1. Bringmann, H. Genetic sleep deprivation: Using sleep mutants to study sleep functions. EMBO Rep. 2019, 20, 14. [Google Scholar] [CrossRef] [PubMed]
  2. Cirelli, C.; Tononi, G. Is sleep essential? PLoS Biol. 2008, 6, e216. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Mignot, E. Why we sleep: The temporal organization of recovery. PLoS Biol. 2008, 6, e106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Feng, X.J.; Liu, Q.Q.; Li, Y.J.; Zhao, F.F.; Chang, H.; Lyu, J. Longitudinal study of the relationship between sleep duration and hypertension in Chinese adult residents (CHNS 2004–2011). Sleep Med. 2019, 58, 88–92. [Google Scholar] [CrossRef] [PubMed]
  5. Hopkinson, N.S.; Rossi, N.; El-Sayed Moustafa, J.; Laverty, A.A.; Quint, J.K.; Freidin, M.; Visconti, A.; Murray, B.; Modat, M.; Ourselin, S.; et al. Multiple, objectively measured sleep dimensions including hypoxic burden and chronic kidney disease: Findings from the Multi-Ethnic Study of Atherosclerosis. Thorax 2021, 76, 714–722. [Google Scholar] [CrossRef]
  6. Suriagandhi, V.; Nachiappan, V. Protective Effects of Melatonin against Obesity-Induced by Leptin Resistance. Behav. Brain Res. 2022, 417, 9. [Google Scholar] [CrossRef]
  7. Carpena, M.X.; Matijasevich, A.; Loret de Mola, C.; Santos, I.S.; Munhoz, T.N.; Tovo-Rodrigues, L. The effects of persistent sleep disturbances during early childhood over adolescent ADHD, and the mediating effect of attention-related executive functions: Data from the 2004 Pelotas Birth Cohort. J. Affect. Disord. 2022, 296, 175–182. [Google Scholar] [CrossRef]
  8. Kang, X.; Jiang, L.; Lan, F.; Tang, Y.-Y.; Zhang, P.; Zou, W.; Chen, Y.-J.; Tang, X.-Q. Hydrogen sulfide antagonizes sleep deprivation-induced depression- and anxiety-like behaviors by inhibiting neuroinflammation in a hippocampal Sirt1-dependent manner. Brain Res. Bull. 2021, 177, 194–202. [Google Scholar] [CrossRef]
  9. Lau, E.Y.Y.; Wong, M.L.; Lam, Y.C.; Lau, K.N.T.; Chung, K.F.; Rusak, B. Sleep and Inhibitory Control Over Mood-Congruent Information in Emerging Adults With Depressive Disorder. Psychosom. Med. 2021, 83, 1004–1012. [Google Scholar] [CrossRef]
  10. Ma, T.; Wang, Y.-Y.; Lu, Y.; Feng, L.; Yang, Y.-T.; Li, G.-H.; Li, C.; Chu, Y.; Wang, W.; Zhang, H. Inhibition of Piezo1/Ca2+/calpain signaling in the rat basal forebrain reverses sleep deprivation-induced fear memory impairments. Behav. Brain Res. 2022, 417. [Google Scholar] [CrossRef]
  11. Davis, K.C.; Raizen, D.M. A mechanism for sickness sleep: Lessons from invertebrates. J. Physiol. 2017, 595, 5415–5424. [Google Scholar] [CrossRef] [Green Version]
  12. Konietzka, J.; Fritz, M.; Spiri, S.; McWhirter, R.; Leha, A.; Palumbos, S.; Costa, W.S.; Oranth, A.; Gottschalk, A.; Miller, D.M., III; et al. Epidermal Growth Factor Signaling Promotes Sleep through a Combined Series and Parallel Neural Circuit. Curr. Biol. 2020, 30, 1–16.e13. [Google Scholar] [CrossRef] [Green Version]
  13. Hill, V.M.; O’Connor, R.M.; Sissoko, G.B.; Irobunda, I.S.; Leong, S.; Canman, J.C.; Stavropoulos, N.; Shirasu-Hiza, M. A bidirectional relationship between sleep and oxidative stress in Drosophila. PLoS Biol. 2018, 16, e2005206. [Google Scholar] [CrossRef]
  14. Tanaka, S.; Toyoda, H.; Honda, Y.; Seki, Y.; Sakurai, T.; Honda, K.; Kodama, T. Hypocretin/orexin prevents recovery from sickness. Biomed. Rep. 2015, 3, 648–650. [Google Scholar] [CrossRef] [Green Version]
  15. Sonnenschein, M.; Sorbi, M.J.; Verbraak, M.; Schaufeli, W.B.; Maas, C.J.M.; van Doomen, L.J.P. Influence of sleep on symptom improvement and return to work in clinical burnout. Scand. J. Work Environ. Health 2008, 34, 23–32. [Google Scholar] [CrossRef] [Green Version]
  16. Schmidt, M.H. The energy allocation function of sleep: A unifying theory of sleep, torpor, and continuous wakefulness. Neurosci. Biobehav. Rev. 2014, 47, 122–153. [Google Scholar] [CrossRef] [Green Version]
  17. Hou, S.-J.; Tsai, S.-J.; Kuo, P.-H.; Lin, W.-Y.; Liu, Y.-L.; Yang, A.C.; Lin, E.; Lan, T.-H. An association study in the Taiwan Biobank elicits the GABAA receptor genes GABRB3, GABRA5, and GABRG3 as candidate loci for sleep duration in the Taiwanese population. BMC Med. Genom. 2021, 14, 223. [Google Scholar] [CrossRef]
  18. Winiarska-Mieczan, A.; Tomaszewska, E.; Jachimowicz, K. Antioxidant, Anti-Inflammatory, and Immunomodulatory Properties of Tea-The Positive Impact of Tea Consumption on Patients with Autoimmune Diabetes. Nutrients 2021, 13, 3972. [Google Scholar] [CrossRef]
  19. Kehong, L.; Enshuo, L.; Ling, L.; Yuan, H.; Yong, Y.; Wenjun, X. L-Theanine mediates the p38MAPK signaling pathway to alleviate heat-induced oxidative stress and inflammation in mice. Food Funct. 2022, 13, 2120–2130. [Google Scholar] [CrossRef]
  20. Wu, Z.; Sun, L.; Chen, R.; Wen, S.; Li, Q.; Lai, X.; Zhang, Z.; Cao, F.; Sun, S. Chinese Tea Alleviates CCl4-Induced Liver Injury through the NF-kappaBorNrf2Signaling Pathway in C57BL-6J Mice. Nutrients 2022, 14, 972. [Google Scholar] [CrossRef]
  21. Prasanth, M.I.; Sivamaruthi, B.S.; Chaiyasut, C.; Tencomnao, T. A Review of the Role of Green Tea (Camellia sinensis) in Antiphotoaging, Stress Resistance, Neuroprotection, and Autophagy. Nutrients 2019, 11, 474. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Kurihara, H.; Ling, C.; Bi-Feng, Z.; Zhao-Dong, H.; Shibata, H.; Kiso, Y.; Tanaka, T.; Xin-Sheng, Y. Anti-stress effect of oolong tea in women loaded with vigil. J. Health Sci. 2003, 49, 436–443. [Google Scholar] [CrossRef] [Green Version]
  23. Unno, K.; Hara, A.; Nakagawa, A.; Iguchi, K.; Ohshio, M.; Morita, A.; Nakamura, Y. Anti-stress effects of drinking green tea with lowered caffeine and enriched theanine, epigallocatechin and arginine on psychosocial stress induced adrenal hypertrophy in mice. Phytomedicine 2016, 23, 1365–1374. [Google Scholar] [CrossRef] [PubMed]
  24. Baradaran Mahdavi, S.; Mansourian, M.; Shams, E.; Qorbani, M.; Heshmat, R.; Motlagh, M.E.; Ziaodini, H.; Dashti, R.; Taheri, M.; Kelishadi, R. Association of Sunlight Exposure with Sleep Hours in Iranian Children and Adolescents: The CASPIAN-V Study. J. Trop. Pediatr. 2020, 66, 4–14. [Google Scholar] [CrossRef] [PubMed]
  25. Jansen, E.C.; Corcoran, K.; Perng, W.; Dunietz, G.L.; Cantoral, A.; Zhou, L.; Tellez-Rojo, M.M.; Peterson, K.E. Relationships of beverage consumption and actigraphy-assessed sleep parameters among urban-dwelling youth from Mexico. Public Health Nutr. 2021, 25, 1844–1853. [Google Scholar] [CrossRef] [PubMed]
  26. Manzar, M.D.; Alghadir, A.H.; Khan, M.; Salahuddin, M.; Albougami, A.; Maniago, J.D.; Vasquez, B.A.; Pandi-Perumal, S.R.; Bahammam, A.S. Anxiety Symptoms Are Associated With Higher Psychological Stress, Poor Sleep, and Inadequate Sleep Hygiene in Collegiate Young Adults-A Cross-Sectional Study. Front. Psychiatry 2021, 12, 677136. [Google Scholar] [CrossRef]
  27. Althakafi, K.A.; Alrashed, A.A.; Aljammaz, K.I.; Abdulwahab, I.J.; Hamza, R.; Hamad, A.F.; Alhejaili, K.S. Prevalence of short sleep duration and effect of co-morbid medical conditions—A cross-sectional study in Saudi Arabia. J. Fam. Med. Prim. Care 2019, 8, 3334–3339. [Google Scholar] [CrossRef]
  28. Hou, S.J.; Tsai, S.J.; Kuo, P.H.; Liu, Y.L.; Yang, A.C.; Lin, E.; Lan, T.H. An association study in the Taiwan Biobank reveals RORA as a novel locus for sleep duration in the Taiwanese Population. Sleep Med. 2020, 73, 70–75. [Google Scholar] [CrossRef]
  29. Lai, X.F.; Wang, X.R.; Wen, S.; Sun, L.L.; Chen, R.H.; Zhang, Z.B.; Li, Q.H.; Cao, J.X.; Lai, Z.X.; Li, Z.G.; et al. Six Types of Tea Reduce Acute Alcoholism in Mice by Enhancing Ethanol Metabolism, Suppressing Oxidative Stress and Inflammation. Front. Nutr. 2022, 9, 848918. [Google Scholar] [CrossRef]
  30. Tung, Y.C.; Liang, Z.R.; Yang, M.J.; Ho, C.T.; Pan, M.H. Oolong tea extract alleviates weight gain in high-fat diet-induced obese rats by regulating lipid metabolism and modulating gut microbiota. Food Funct. 2022, 13, 2846–2856. [Google Scholar] [CrossRef]
  31. Ladeira, L.C.M.; dos Santos, E.C.; Santos, T.A.; da Silva, J.; Lima, G.D.D.; Machado-Neves, M.; da Silva, R.C.; Freitas, M.B.; Maldonado, I. Green tea infusion prevents diabetic nephropathy aggravation in recent-onset type 1 diabetes regardless of glycemic control. J. Ethnopharmacol. 2021, 274, 114032. [Google Scholar] [CrossRef]
  32. Mo, T.; Zhang, W.; Li, P. The change and regulation mechanism of key components during tea processing. J. Chin. Inst. Food Sci. Technol. 2011, 11, 176–180. [Google Scholar]
  33. Sanlier, N.; Gokcen, B.B.; Altug, M. Tea consumption and disease correlations. Trends Food Sci. Technol. 2018, 78, 95–106. [Google Scholar] [CrossRef]
  34. Liu, S.; Zhang, Q.; Li, H.; Qiu, Z.; Yu, Y. Comparative Assessment of the Antibacterial Efficacies and Mechanisms of Different Tea Extracts. Foods 2022, 11, 620. [Google Scholar] [CrossRef]
  35. Kan, Z.P.; Wang, Y.J.; Chen, Q.; Tang, X.Y.; Thompson, H.J.; Huang, J.B.; Zhang, J.S.; Gao, F.; Shen, Y.; Wan, X.C. Green Tea Suppresses Amyloid beta Levels and Alleviates Cognitive Impairment by Inhibiting APP Cleavage and Preventing Neurotoxicity in 5XFAD Mice. Mol. Nutr. Food Res. 2021, 65, 2100626. [Google Scholar] [CrossRef]
  36. Perdices, L.; Fuentes-Broto, L.; Segura, F.; Cavero, A.; Orduna-Hospital, E.; Insa-Sanchez, G.; Sanchez-Cano, A.I.; Fernandez-Sanchez, L.; Cuenca, N.; Pinilla, I. Systemic epigallocatechin gallate protects against retinal degeneration and hepatic oxidative stress in the P23H-1 rat. Neural Regen. Res. 2022, 17, 625–631. [Google Scholar] [CrossRef]
  37. Li, M.Z.; Duan, Y.J.; Wang, Y.; Chen, L.; Abdelrahim, M.E.A.; Yan, J. The effect of Green green tea consumption on body mass index, lipoprotein, liver enzymes, and liver cancer: An updated systemic review incorporating a meta-analysis. Crit. Rev. Food Sci. Nutr. 2022, 1–9. [Google Scholar] [CrossRef]
  38. Colonetti, L.; Grande, A.J.; Toreti, I.R.; Ceretta, L.B.; da Rosa, M.I.; Colonetti, T. Green tea promotes weight loss in women with polycystic ovary syndrome: Systematic review and meta-analysis. Nutr. Res. 2022, 104, 1–9. [Google Scholar] [CrossRef]
  39. Kwak, J.; Shin, D. Association between Green Tea Consumption and Abdominal Obesity Risk in Middle-Aged Korean Population: Findings from the Korean Genome and Epidemiology Study. Int. J. Environ. Res. Public Health 2022, 19, 2735. [Google Scholar] [CrossRef]
  40. Bedran, T.B.L.; Morin, M.P.; Spolidorio, D.P.; Grenier, D. Black tea extract and its theaflavin derivatives inhibit the growth of periodontopathogens and modulate interleukin-8 and beta-defensin secretion in oral epithelial cells. PLoS ONE 2015, 10, e0143158. [Google Scholar] [CrossRef]
  41. Korystova, A.F.; Kublik, L.N.; Samokhvalova, T.V.; Shaposhnikova, V.V.; Korystov, Y.N. Black tea is more effective than green tea in prevention of radiation-induced oxidative stress in the aorta of rats. Biomed. Pharmacother. 2021, 142, 112064. [Google Scholar] [CrossRef] [PubMed]
  42. Yusufoglu, B.; Karakus, E.; Yaman, M. Determining the amount and bioaccessibility of methylglyoxal and glyoxal in functional snack foods with herbal teas: Effect of different herbal teas on alpha-Dicarbonyls. Food Sci. Technol. 2022, 42, e82621. [Google Scholar] [CrossRef]
  43. Wang, Q.; Zhao, X.; Qian, Y.; Wang, R. In vitro antioxidative activity of yellow tea and its in vivo preventive effect on gastric injury. Exp. Ther. Med. 2013, 6, 423–426. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Yun, T.; Daxiang, L.; Ponmari, G.; Na, X.; Zhongwen, X. Dietary supplement of large yellow tea ameliorates metabolic syndrome and attenuates hepatic steatosis in db/db Mice. Nutrients 2018, 10, 75. [Google Scholar] [CrossRef] [Green Version]
  45. Shaoxiong, Z.; Chatrawee, D.; Tewin, T.; Jianghong, L.; Jinke, L.; Wink, M. Neuroprotective effects of oolong tea extracts against glutamate-induced toxicity in cultured neuronal cells and beta-amyloid-induced toxicity in Caenorhabditis elegans. Food Funct. 2020, 11, 8179–8192. [Google Scholar] [CrossRef]
  46. Wang, Q.; Guan, X.F.; Lai, C.C.; Gao, H.Y.; Zheng, Y.F.; Huang, J.Q.; Lin, B. Selenium enrichment improves anti-proliferative effect of oolong tea extract on human hepatoma HuH-7 cells. Food Chem. Toxicol. 2021, 147, 111873. [Google Scholar] [CrossRef]
  47. Asokan Shibu, M.; Chia-Hua, K.; Bih-Cheng, C.; Da-Tong, J.; Ray-Jade, C.; Chao-Hung, L.; Pei-Jane, H.; Padma Viswanadha, V.; Wei-Wen, K.; Chih-Yang, H. Oolong tea prevents cardiomyocyte loss against hypoxia by attenuating p-JNK mediated hypertrophy and enhancing P-IGF1R, p-akt, and p-Badser136 activity and by fortifying NRF2 antioxidation system. Environ. Toxicol. 2018, 33, 220–233. [Google Scholar] [CrossRef]
  48. Duangjan, C.; Curran, S.P. Oolonghomobisflavans from Camellia sinensis increase Caenorhabditis elegans lifespan and healthspan. Geroscience 2022, 44, 533–545. [Google Scholar] [CrossRef]
  49. Santana-Rios, G.; Orner, G.A.; Xu, M.; Izquierdo-Pulido, M.; Dashwood, R.H. Inhibition by white tea of 2-amino-1-methyl-6-phenylimidazo 4,5-b pyridine-induced colonic aberrant crypts in the F344 rat. Nutr. Cancer 2001, 41, 98–103. [Google Scholar] [CrossRef] [Green Version]
  50. He, M.R.; Lyu, X.H. Application of BRAFO-tiered approach for health benefit-risk assessment of dark tea consumption in China. Food Chem. Toxicol. 2021, 158, 112615. [Google Scholar] [CrossRef]
  51. Siqiang, W.; Zhuoting, G.; Beichao, W.; Na, Z.; Kun, L.; Ting, Y. Effect of brewing conditions on polyphenols in the dark tea (Camellia sinensis L.) infusions: Content, composition and antioxidant activities. Food Sci. Technol. Cienc. Tecnol. Aliment. 2022, 42, e36322. [Google Scholar] [CrossRef]
  52. Wang, C.Q.; Hu, M.H.; Yi, Y.H.; Wen, X.N.; Lv, C.H.; Shi, M.; Zeng, C.X. Multiomic analysis of dark tea extract on glycolipid metabolic disorders in db/db mice. Front. Nutr. 2022, 9, 1006517. [Google Scholar] [CrossRef]
  53. Liming, W.; Ting, L. Effect of Eurotium cristatum fermented dark tea extract on body weight and blood lipid in rats. Curr. Dev. Nutr. 2018, 2, A77. [Google Scholar] [CrossRef] [Green Version]
  54. Campbell, S.S.; Tobler, I. Animal sleep: A review of sleep duration across phylogeny. Neurosci. Biobehav. Rev. 1984, 8, 269–300. [Google Scholar] [CrossRef]
  55. Aserinsky, E.; Kleitman, N. Regularly occurring periods of eye motility, and concomitant phenomena, during sleep. Science 1953, 118, 273–274. [Google Scholar] [CrossRef] [Green Version]
  56. Bringmann, H. Sleep-Active Neurons: Conserved Motors of Sleep. Genetics 2018, 208, 1279–1289. [Google Scholar] [CrossRef] [Green Version]
  57. Vorster, A.P.; Born, J. Sleep and memory in mammals, birds and invertebrates. Neurosci. Biobehav. Rev. 2015, 50, 103–119. [Google Scholar] [CrossRef] [Green Version]
  58. Orem, J.; Lovering, A.T.; Dunin-Barkowski, W.; Vidruk, E.H. Endogenous excitatory drive to the respiratory system in rapid eye movement sleep in cats. J. Physiol. 2000, 527 Pt 2, 365–376. [Google Scholar] [CrossRef]
  59. Peever, J.; Fuller, P.M. The Biology of REM Sleep. Curr. Biol. 2017, 27, R1237–R1248. [Google Scholar] [CrossRef]
  60. Trojanowski, N.F.; Nelson, M.D.; Flavell, S.W.; Fang-Yen, C.; Raizen, D.M. Distinct Mechanisms Underlie Quiescence during Two Caenorhabditis elegans Sleep-Like States. J. Neurosci. 2015, 35, 14571–14584. [Google Scholar] [CrossRef] [Green Version]
  61. Arnason, B.B.; Thornorsteinsson, H.; Karlsson, K.A.E. Absence of rapid eye movements during sleep in adult zebrafish. Behav. Brain Res. 2015, 291, 189–194. [Google Scholar] [CrossRef] [PubMed]
  62. Yokogawa, T.; Marin, W.; Faraco, J.; Pezeron, G.; Appelbaum, L.; Zhang, J.; Rosa, F.; Mourrain, P.; Mignot, E. Characterization of sleep in zebrafish and insomnia in hypocretin receptor mutants. PLoS Biol. 2007, 5, e277. [Google Scholar] [CrossRef] [PubMed]
  63. Rihel, J.; Prober, D.A.; Schier, A.F. Monitoring sleep and arousal in zebrafish. Methods Cell Biol. 2010, 100, 281–294. [Google Scholar] [CrossRef] [PubMed]
  64. Nitz, D.A.; van Swinderen, B.; Tononi, G.; Greenspan, R.J. Electrophysiological correlates of rest and activity in Drosophila melanogaster. Curr. Biol. 2002, 12, 1934–1940. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Shaw, P.J.; Cirelli, C.; Greenspan, R.J.; Tononi, G. Correlates of sleep and waking in Drosophila melanogaster. Science 2000, 287, 1834–1837. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Watson, N.F.; Dikmen, S.; Machamer, J.; Doherty, M.; Temkin, N. Hypersomnia Following Traumatic Brain Injury. J. Clin. Sleep Med. 2007, 3, 363–368. [Google Scholar] [CrossRef] [Green Version]
  67. Karshikoff, B.; Sundelin, T.; Lasselin, J. Role of inflammation in Human Fatigue: Relevance of Multidimensional Assessments and Potential Neuronal Mechanisms. Front. Immunol. 2017, 8, 21. [Google Scholar] [CrossRef] [Green Version]
  68. Nagai, M.; Nagai, H.; Numa, C.; Furuyashiki, T. Stress-induced sleep-like inactivity modulates stress susceptibility in mice. Sci. Rep. 2020, 10, 1–11. [Google Scholar] [CrossRef]
  69. Olivadoti, M.D.; Weinberg, J.B.; Toth, L.A.; Opp, M.R. Sleep and fatigue in mice infected with murine gammaherpesvirus 68. Brain Behav. Immun. 2011, 25, 696–705. [Google Scholar] [CrossRef] [Green Version]
  70. Lenz, O.; Xiong, J.; Nelson, M.D.; Raizen, D.M.; Williams, J.A. FMRFamide signaling promotes stress-induced sleep in Drosophila. Brain Behav. Immun. 2015, 47, 141–148. [Google Scholar] [CrossRef] [Green Version]
  71. Wang, Y.H.; Zhang, H.Y.; Zhang, Z.Y.; Sun, B.Q.; Tang, C.; Zhang, L.; Jiang, Z.H.; Ding, B.; Liao, Y.Y.; Cai, P. Simulated mobile communication frequencies (3.5 GHz) emitted by a signal generator affects the sleep of Drosophila melanogaster. Environ. Pollut. 2021, 283, 117087. [Google Scholar] [CrossRef]
  72. Goetting, D.L.; Mansfield, R.; Soto, R.; Van Buskirk, C. Cellular damage, including wounding, drives C. elegans stress-induced sleep. J. Neurogenet. 2020, 34, 430–439. [Google Scholar] [CrossRef]
  73. Hill, A.J.; Mansfield, R.; Lopez, J.M.N.G.; Raizen, D.M.; Van Buskirk, C. Cellular Stress Induces a Protective Sleep-like State in C. elegans. Curr. Biol. 2014, 24, 2399–2405. [Google Scholar] [CrossRef] [Green Version]
  74. Freiberg, A.S. Why We Sleep: A Hypothesis for an Ultimate or Evolutionary Origin for Sleep and Other Physiological Rhythms. J. Circadian Rhythm. 2020, 18, 2. [Google Scholar] [CrossRef] [Green Version]
  75. Filippini, T.; Malavolti, M.; Borrelli, F.; Izzo, A.A.; Fairweather-Tait, S.J.; Horneber, M.; Vinceti, M. Green tea (Camellia sinensis) for the prevention of cancer. Cochrane Database Syst. Rev. 2020, 2021, CD005004. [Google Scholar] [CrossRef] [Green Version]
  76. Kleiser, C.; Wawro, N.; Stelmach-Mardas, M.; Boeing, H.; Gedrich, K.; Himmerich, H.; Linseisen, J. Are sleep duration, midpoint of sleep and sleep quality associated with dietary intake among Bavarian adults? Eur. J. Clin. Nutr. 2017, 71, 631–637. [Google Scholar] [CrossRef]
  77. Xue, L.; Zhang, J.; Shen, H.; Ai, L.; Wu, R. A randomized controlled pilot study of the effectiveness of magnolia tea on alleviating depression in postnatal women. Food Sci. Nutr. 2020, 8, 1554–1561. [Google Scholar] [CrossRef] [Green Version]
  78. Zhao, W.F.; Li, Y.; Ma, W.; Ge, Y.Z.; Huang, Y.H. A study on quality components and sleep-promoting effects of GABA black tea. Food Funct. 2015, 6, 3393–3398. [Google Scholar] [CrossRef]
  79. Kolar, M.H.; Plajnsek, K.T.; Dimitrijevic, D.; Gay, T.; Cohen, O.O. Mixture for Reducing Body Mass. WO 2015/099616 A1, 21 August 2015. [Google Scholar]
  80. Sun, J. Morning/evening menopausal formula relieves menopausal symptoms: A pilot study. J. Altern. Complement. Med. 2003, 9, 403–409. [Google Scholar] [CrossRef]
  81. Hossain, S.J.; Aoshima, H.; Koda, H.; Kiso, Y. Fragrances in oolong tea that enhance the response of GABAA receptors. Biosci. Biotechnol. Biochem. 2004, 68, 1842–1848. [Google Scholar] [CrossRef] [Green Version]
  82. Forouzanfar, F.; Gholami, J.; Foroughnia, M.; Payvar, B.; Nemati, S.; Khodadadegan, M.A.; Saheb, M.; Hajali, V. The beneficial effects of green tea on sleep deprivation-induced cognitive deficits in rats: The involvement of hippocampal antioxidant defense. Heliyon 2021, 7, e08336. [Google Scholar] [CrossRef] [PubMed]
  83. Romain, C.; Alcaraz, P.E.; Chung, L.H.; Cases, J. Regular consumption of HolisFiit, a polyphenol-rich extract-based food supplement, improves mind and body well-being of overweight and slightly obese volunteers: A randomized, double-blind, parallel trial. Int. J. Food Sci. Nutr. 2017, 68, 840–848. [Google Scholar] [CrossRef] [PubMed]
  84. Yang, H.; Xie, J.; Mu, W.; Ruan, X.; Zhang, J.; Yao, L.; Diao, Z.; Wu, M.; Li, Y.; Ren, W.; et al. Tea polyphenols protect learning and memory in sleep-deprived mice by promoting AMPA receptor internalization. Neuroreport 2020, 31, 857–864. [Google Scholar] [CrossRef] [PubMed]
  85. Sarris, J.; Byrne, G.J.; Cribb, L.; Oliver, G.; Murphy, J.; Macdonald, P.; Nazareth, S.; Karamacoska, D.; Galea, S.; Short, A.; et al. L-theanine in the adjunctive treatment of generalized anxiety disorder: A double-blind, randomised, placebo-controlled trial. J. Psychiatr. Res. 2019, 110, 31–37. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Hidese, S.; Ogawa, S.; Ota, M.; Ishida, I.; Yasukawa, Z.; Ozeki, M.; Kunugi, H. Effects of L-Theanine Administration on Stress-Related Symptoms and Cognitive Functions in Healthy Adults: A Randomized Controlled Trial. Nutrients 2019, 11, 2362. [Google Scholar] [CrossRef] [Green Version]
  87. Zhang, Y.; Jia, X.Z.; Chen, X.M.; Liu, Y.J.; Zhao, Z.F.; Hao, J.Y.; Wu, R.; Feng, H.T.; Ren, X.N. L-theanine and Neumentix mixture improves sleep quality and modulates brain neurotransmitter levels in mice. Ann. Palliat. Med. 2021, 10, 4572–4581. [Google Scholar] [CrossRef]
  88. Kim, S.; Jo, K.; Hong, K.B.; Han, S.H.; Suh, H.J. GABA and L-theanine mixture decreases sleep latency and improves NREM sleep. Pharm. Biol. 2019, 57, 65–73. [Google Scholar] [CrossRef] [Green Version]
  89. Lin, Y.S.A.; Weibel, J.; Landolt, H.P.; Santini, F.; Meyer, M.; Brunmair, J.; Meier-Menches, S.M.; Gerner, C.; Borgwardt, S.; Cajochen, C.; et al. Daily Caffeine Intake Induces Concentration-Dependent Medial Temporal Plasticity in Humans: A Multimodal Double-Blind Randomized Controlled Trial. Cerebral Cortex 2021, 31, 3096–3106. [Google Scholar] [CrossRef]
  90. Kwon, S.; Yoon, M.; Lee, J.; Moon, K.D.; Kim, D.; Kim, S.B.; Cho, S. A Standardized Phlorotannin Supplement Attenuates Caffeine-Induced Sleep Disruption in Mice. Nutrients 2019, 11, 556. [Google Scholar] [CrossRef] [Green Version]
  91. Ohno, A. Sleep Aid. WO 2020/059808 A1, 28 May 2020. [Google Scholar]
  92. Zhong, Y.; Zheng, Q.; Hu, P.; Huang, X.; Yang, M.; Ren, G.; Du, Q.; Luo, J.; Zhang, K.; Li, J.; et al. Sedative and hypnotic effects of compound Anshen essential oil inhalation for insomnia. BMC Complement. Altern. Med. 2019, 19, 306. [Google Scholar] [CrossRef]
  93. Komagata, N.; Latifi, B.; Rusterholz, T.; Bassetti, C.L.A.; Adamantidis, A.; Schmidt, M.H. Dynamic REM Sleep Modulation by Ambient Temperature and the Critical Role of the Melanin-Concentrating Hormone System. Curr. Biol. 2019, 29, 1976–1987.e4. [Google Scholar] [CrossRef]
  94. Latifi, B.; Adamantidis, A.; Bassetti, C.; Schmidt, M.H. Sleep-Wake Cycling and Energy Conservation: Role of Hypocretin and the Lateral Hypothalamus in Dynamic State-Dependent Resource Optimization. Front. Neurol. 2018, 9, 16. [Google Scholar] [CrossRef]
  95. Bandaru, S.S.; Khanday, M.A.; Ibrahim, N.; Naganuma, F.; Vetrivelan, R. Sleep-Wake Control by Melanin-Concentrating Hormone (MCH) Neurons: A Review of Recent Findings. Curr. Neurol. Neurosci. 2020, 20, 1–12. [Google Scholar] [CrossRef]
  96. Wei, Y.; Xu, J.; Miao, S.; Wei, K.; Peng, L.; Wang, Y.; Wei, X. Recent advances in the utilization of tea active ingredients to regulate sleep through neuroendocrine pathway, immune system and intestinal microbiota. Crit. Rev. Food Sci. Nutr. 2022, 1–29. [Google Scholar] [CrossRef]
  97. Tsunematsu, T. Elucidation of Neural Circuits Involved in the Regulation of Sleep/Wakefulness Using Optogenetics. In Optogenetics: Light-Sensing Proteins and Their Applications in Neuroscience and Beyond, 2nd ed.; Springer: Singapore, 2021; Volume 1293, pp. 391–406. [Google Scholar] [CrossRef]
  98. Maluck, E.; Busack, I.; Besseling, J.; Masurat, F.; Turek, M.; Busch, K.E.; Bringmann, H. A wake-active locomotion circuit depolarizes a sleep-active neuron to switch on sleep. PLoS Biol. 2020, 18, e3000361. [Google Scholar] [CrossRef] [Green Version]
  99. Zhang, J.; Cai, S.; Li, J.; Xiong, L.; Tian, L.; Liu, J.; Huang, J.; Liu, Z. Neuroprotective Effects of Theaflavins Against Oxidative Stress-Induced Apoptosis in PC12 Cells. Neurochem. Res. 2016, 41, 3364–3372. [Google Scholar] [CrossRef]
  100. Xu, Y.J.; Liu, S.J.; Zhu, L.Y.; Dai, L.G.; Qian, W.; Zhang, J.Z.; Li, X.; Pan, W. Green tea protects against hippocampal neuronal apoptosis in diabetic encephalopathy by inhibiting JNK/MLCK signaling. Mol. Med. Rep. 2021, 24, 1–13. [Google Scholar] [CrossRef]
  101. Zhang, W.; Li, X.; Hua, F.; Chen, W.; Wang, W.; Chu, G.X.; Bao, G.H. Interaction between Ester-Type Tea Catechins and Neutrophil Gelatinase-Associated Lipocalin: Inhibitory Mechanism. J. Agric. Food Chem. 2018, 66, 1147–1156. [Google Scholar] [CrossRef]
  102. Kim, J.; Funayama, S.; Izuo, N.; Shimizu, T. Dietary supplementation of a high-temperature-processed green tea extract attenuates cognitive impairment in PS2 and Tg2576 mice. Biosci. Biotechnol. Biochem. 2019, 83, 2364–2371. [Google Scholar] [CrossRef]
  103. Unno, K.; Nakamura, Y. Green Tea Suppresses Brain Aging. Molecules 2021, 26, 4897. [Google Scholar] [CrossRef]
  104. Bae, H.J.; Kim, J.; Jeon, S.J.; Kim, J.; Goo, N.; Jeong, Y.; Cho, K.; Cai, M.; Jung, S.Y.; Kwon, K.J.; et al. Green tea extract containing enhanced levels of epimerized catechins attenuates scopolamine-induced memory impairment in mice. J. Ethnopharmacol. 2020, 258, 112923. [Google Scholar] [CrossRef] [PubMed]
  105. Masood, M.I.; Schafer, K.H.; Naseem, M.; Weyland, M.; Meiser, P. Troxerutin flavonoid has neuroprotective properties and increases neurite outgrowth and migration of neural stem cells from the subventricular zone. PLoS ONE 2020, 15, e0237025. [Google Scholar] [CrossRef] [PubMed]
  106. Nagai, J.; Yu, X.Z.; Papouin, T.; Cheong, E.J.; Freeman, M.R.; Monk, K.R.; Hastings, M.H.; Haydon, P.G.; Rowitch, D.; Shaham, S.; et al. Behaviorally consequential astrocytic regulation of neural circuits. Neuron 2021, 109, 576–596. [Google Scholar] [CrossRef] [PubMed]
  107. Blum, I.D.; Keles, M.F.; Baz, E.S.; Han, E.; Park, K.; Luu, S.; Issa, H.; Brown, M.; Ho, M.C.W.; Tabuchi, M.; et al. Astroglial Calcium Signaling Encodes Sleep Need in Drosophila. Curr. Biol. 2021, 31, 150–162.e7. [Google Scholar] [CrossRef] [PubMed]
  108. Ingiosi, A.M.; Hayworth, C.R.; Harvey, D.O.; Singletary, K.G.; Rempe, M.J.; Wisor, J.P.; Frank, M.G. A Role for Astroglial Calcium in Mammalian Sleep and Sleep Regulation. Curr. Biol. 2020, 30, 4373–4383.e7. [Google Scholar] [CrossRef] [PubMed]
  109. Vaidyanathan, T.V.; Collard, M.; Yokoyama, S.; Reitman, M.E.; Poskanzer, K.E. Cortical astrocytes independently regulate sleep depth and duration via separate GPCR pathways. eLife 2021, 10, e63329. [Google Scholar] [CrossRef]
  110. Choudhury, M.E.; Miyanishi, K.; Takeda, H.; Tanaka, J. Microglia and the Aging Brain: Are Geriatric Microglia Linked to Poor Sleep Quality? Int. J. Mol. Sci. 2021, 22, 7824. [Google Scholar] [CrossRef]
  111. Corsi, G.; Picard, K.; di Castro, M.A.; Garofalo, S.; Tucci, F.; Chece, G.; del Percio, C.; Golia, M.T.; Raspa, M.; Scavizzi, F.; et al. Microglia modulates hippocampal synaptic transmission and sleep duration along the light/dark cycle. GLIA 2022, 70, 89–105. [Google Scholar] [CrossRef]
  112. Barenys, M.; Gassmann, K.; Baksmeier, C.; Heinz, S.; Reverte, I.; Schmuck, M.; Temme, T.; Bendt, F.; Zschauer, T.C.; Rockel, T.D.; et al. Epigallocatechin gallate (EGCG) inhibits adhesion and migration of neural progenitor cells in vitro. Arch. Toxicol. 2017, 91, 827–837. [Google Scholar] [CrossRef]
  113. Ren, J.L.; Yu, Q.X.; Liang, W.C.; Leung, P.Y.; Ng, T.K.; Chu, W.K.; Pang, C.P.; Chan, S.O. Green tea extract attenuates LPS-induced retinal inflammation in rats. Sci. Rep. 2018, 8, 429. [Google Scholar] [CrossRef]
  114. Cittadini, M.C.; Garcia-Estevez, I.; Escribano-Bailon, M.T.; Rivas-Gonzalo, J.C.; Valentich, M.A.; Repossi, G.; Soria, E.A. Modulation of Fatty Acids and Interleukin-6 in Glioma Cells by South American Tea Extracts and their Phenolic Compounds. Nutr. Cancer 2018, 70, 267–277. [Google Scholar] [CrossRef] [PubMed]
  115. Pak, K.; Kim, J.; Kim, K.; Kim, S.J.; Kim, I.J. Sleep and Neuroimaging. Nucl. Med. Mol. Imaging 2020, 54, 98–104. [Google Scholar] [CrossRef] [PubMed]
  116. Nelson, M.D.; Lee, K.H.; Churgin, M.A.; Hill, A.J.; Van Buskirk, C.; Fang-Yen, C.; Raizen, D.M. FMRFamide-like FLP-13 Neuropeptides Promote Quiescence following Heat Stress in Caenorhabditis elegans. Curr. Biol. 2014, 24, 2406–2410. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Shanshan, H.; Liyong, L.; Xintong, B.; Rui Hai, L.; Sibo, Z.; Yu, C.; Kang, S.; Jielin, J.; Zhonghua, L.; Liang, Z. Pu-erh tea restored circadian rhythm disruption by regulating tryptophan metabolism. J. Agric. Food Chem. 2022, 70, 5610–5623. [Google Scholar] [CrossRef]
  118. Ouyang, S.H.; Zhai, Y.J.; Wu, Y.P.; Xie, G.; Wang, G.E.; Mao, Z.F.; Hu, H.H.; Luo, X.H.; Sun, W.Y.; Liang, L.; et al. Theacrine, a Potent Antidepressant Purine Alkaloid from a Special Chinese Tea, Promotes Adult Hippocampal Neurogenesis in Stressed Mice. J. Agric. Food Chem. 2021, 69, 7016–7027. [Google Scholar] [CrossRef]
  119. Kawada, K.; Kuramoto, N.; Mimori, S. Possibility that the Onset of Autism Spectrum Disorder is Induced by Failure of the Glutamine-Glutamate Cycle. Curr. Mol. Pharmacol. 2021, 14, 170–174. [Google Scholar] [CrossRef]
  120. Deb, S.; Dutta, A.; Phukan, B.C.; Manivasagam, T.; Thenmozhi, A.J.; Bhattacharya, P.; Paul, R.; Borah, A. Neuroprotective attributes of L-theanine, a bioactive amino acid of tea, and its potential role in Parkinson’s disease therapeutics. Neurochem. Int. 2019, 129, 104478. [Google Scholar] [CrossRef]
  121. Raj, K.; Gupta, G.D.; Singh, S. l-Theanine ameliorates motor deficit, mitochondrial dysfunction, and neurodegeneration against chronic tramadol induced rats model of Parkinson’s disease. Drug Chem Toxicol 2021, 45, 2097–2108. [Google Scholar] [CrossRef]
  122. Wightman, E.L.; Jackson, P.A.; Khan, J.; Forster, J.; Heiner, F.; Feistel, B.; Suarez, C.G.; Pischel, I.; Kennedy, D.O. The Acute and Chronic Cognitive and Cerebral Blood Flow Effects of a Sideritis scardica (Greek Mountain Tea) Extract: A Double Blind, Randomized, Placebo Controlled, Parallel Groups Study in Healthy Humans. Nutrients 2018, 10, 955. [Google Scholar] [CrossRef] [Green Version]
  123. Wagner-Skacel, J.; Dalkner, N.; Moerkl, S.; Kreuzer, K.; Farzi, A.; Lackner, S.; Painold, A.; Reininghaus, E.Z.; Butler, M.I.; Bengesser, S. Sleep and Microbiome in Psychiatric Diseases. Nutrients 2020, 12, 2198. [Google Scholar] [CrossRef]
  124. Ogawa, Y.; Miyoshi, C.; Obana, N.; Yajima, K.; Hotta-Hirashima, N.; Ikkyu, A.; Kanno, S.; Soga, T.; Fukuda, S.; Yanagisawa, M. Gut microbiota depletion by chronic antibiotic treatment alters the sleep/wake architecture and sleep EEG power spectra in mice. Sci. Rep. 2020, 10, 1–11. [Google Scholar] [CrossRef]
  125. Yao, Z.W.; Zhao, B.C.; Yang, X.; Lei, S.H.; Jiang, Y.M.; Liu, K.X. Relationships of sleep disturbance, intestinal microbiota, and postoperative pain in breast cancer patients: A prospective observational study. Sleep Breath. 2021, 25, 1655–1664. [Google Scholar] [CrossRef]
  126. Zhang, R.R.; Gao, S.J.; Wang, S.H.; Zhang, J.W.; Bai, Y.Y.; He, S.; Zhao, P.; Zhang, H.J. Gut Microbiota in Patients with Type 1 Narcolepsy. Nat. Sci. Sleep 2021, 13, 2007–2018. [Google Scholar] [CrossRef]
  127. Valentini, F.; Evangelisti, M.; Arpinelli, M.; Di Nardo, G.; Borro, M.; Simmaco, M.; Villa, M.P. Gut microbiota composition in children with obstructive sleep apnoea syndrome: A pilot study. Sleep Med. 2020, 76, 140–147. [Google Scholar] [CrossRef]
  128. Szentirmai, E.; Massie, A.R.; Kapas, L. Lipoteichoic acid, a cell wall component of Gram-positive bacteria, induces sleep and fever and suppresses feeding. Brain Behav. Immun. 2021, 92, 184–192. [Google Scholar] [CrossRef]
  129. Wang, C.; Gao, Z.; Qian, Y.; Li, X.; Wang, J.; Ma, J.; Guo, J.; Fu, F. Effects of Different Concentrations of Ganpu Tea on Fecal Microbiota and Short Chain Fatty Acids in Mice. Nutrients 2021, 13, 3715. [Google Scholar] [CrossRef]
  130. Li, B.Y.; Mao, Q.Q.; Zhou, D.D.; Luo, M.; Gan, R.Y.; Li, H.Y.; Huang, S.Y.; Saimaiti, A.; Shang, A.; Li, H.B. Effects of Tea against Alcoholic Fatty Liver Disease by Modulating Gut Microbiota in Chronic Alcohol-Exposed Mice. Foods 2021, 10, 1232. [Google Scholar] [CrossRef]
  131. Gong, Z.P.; Ouyang, J.; Wu, X.L.; Zhou, F.; Lu, D.M.; Zhao, C.J.; Liu, C.F.; Zhu, W.; Zhang, J.C.; Li, N.X.; et al. Dark tea extracts: Chemical constituents and modulatory effect on gastrointestinal function. Biomed. Pharmacother. 2020, 130, 110514. [Google Scholar] [CrossRef]
  132. Cao, P.Q.; Li, X.P.; Jian, O.Y.; Jiang, R.G.; Huang, F.F.; Wen, B.B.; Zhang, X.N.; Huang, J.A.; Liu, Z.H. The protective effects of yellow tea extract against loperamide-induced constipation in mice. Food Funct. 2021, 12, 5621–5636. [Google Scholar] [CrossRef]
  133. Mikami, A.; Ogita, T.; Namai, F.; Shigemori, S.; Sato, T.; Shimosato, T. Oral Administration of Flavonifractor plautii, a Bacteria Increased With Green Tea Consumption, Promotes Recovery From Acute Colitis in Mice via Suppression of IL-17. Front. Nutr. 2020, 7, 610946. [Google Scholar] [CrossRef]
  134. Perez-Burillo, S.; Navajas-Porras, B.; Lopez-Maldonado, A.; Hinojosa-Nogueira, D.; Pastoriza, S.; Rufian-Henares, J.A. Green Tea and Its Relation to Human Gut Microbiome. Molecules 2021, 26, 3907. [Google Scholar] [CrossRef] [PubMed]
  135. Stapleton, P.D.; Shah, S.; Ehlert, K.; Hara, Y.; Taylor, P.W. The beta-lactam-resistance modifier (−)-epicatechin gallate alters the architecture of the cell wall of Staphylococcus aureus. Microbiology 2007, 153, 2093–2103. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. He, J.; Chen, J.; He, Q.; Li, S.; Jian, L.; Xie, F.; Dong, C.; Bai, G.Y.; Wang, Z.R.; Zou, T.D.; et al. Oral L-theanine administration promotes fat browning and prevents obesity in mice fed high-fat diet associated with the modulation of gut microbiota. J. Funct. Foods 2021, 81, 104476. [Google Scholar] [CrossRef]
  137. Cao, J.; Herman, A.B.; West, G.B.; Poe, G.; Savage, V.M. Unraveling why we sleep: Quantitative analysis reveals abrupt transition from neural reorganization to repair in early development. Sci. Adv. 2020, 6, eaba0398. [Google Scholar] [CrossRef] [PubMed]
  138. Hahn, J.; Gunter, M.; Schuhmacher, J.; Bieber, K.; Poschel, S.; Schutz, M.; Engelhardt, B.; Oster, H.; Sina, C.; Lange, T.; et al. Sleep enhances numbers and function of monocytes and improves bacterial infection outcome in mice. Brain Behav. Immun. 2020, 87, 329–338. [Google Scholar] [CrossRef]
  139. Zhang, J.; Jiang, H.; Du, K.; Xie, T.; Wang, B.; Chen, C.; Reiter, R.J.; Cen, B.; Yuan, Y. Pan-cancer analyses reveal genomics and clinical characteristics of the melatonergic regulators in cancer. J. Pineal Res. 2021, 71, e12758. [Google Scholar] [CrossRef]
  140. Sinner, M.P.; Masurat, F.; Ewbank, J.J.; Pujol, N.; Bringmann, H. Innate Immunity Promotes Sleep through Epidermal Antimicrobial Peptides. Curr. Biol. 2021, 31, 27. [Google Scholar] [CrossRef]
  141. Droge, W. Free radicals in the physiological control of cell function. Physiol. Rev. 2002, 82, 47–95. [Google Scholar] [CrossRef] [Green Version]
  142. Gupta, S.; You, P.N.; SenGupta, T.; Nilsen, H.; Sharma, K. Crosstalk between Different DNA Repair Pathways Contributes to Neurodegenerative Diseases. Biology 2021, 10, 163. [Google Scholar] [CrossRef]
  143. Halliwell, B. Antioxidants in human health and disease. Annu. Rev. Nutr. 1996, 16, 33–50. [Google Scholar] [CrossRef]
  144. Miao, L.; Daozhou, L.; Ying, C.; Qibing, M.; Siyuan, Z. A resveratrol-loaded nanostructured lipid carrier hydrogel to enhance the anti-UV irradiation and anti-oxidant efficacy. Colloids Surf. B Biointerfaces 2021, 204, 111786. [Google Scholar] [CrossRef]
  145. Hou, X.; Shao, C.; Sun, K.; Li, R.; Gao, L.; Meng, Y.; Jing, Y.; Wei, L. Autophagy deficiency downregulates O(6)methylguanine-DNA methyltransferase and increases chemosensitivity of liver cancer cells. Aging 2021, 13, 14289–14303. [Google Scholar] [CrossRef]
  146. Rodriguez-Rocha, H.; Garcia-Garcia, A.; Panayiotidis, M.I.; Franco, R. DNA damage and autophagy. Mutat. Res.-Fundam. Mol. Mech. Mutagen. 2011, 711, 158–166. [Google Scholar] [CrossRef] [Green Version]
  147. Kansal, V.; Agarwal, A.; Harbour, A.; Farooqi, H.; Singh, V.K.; Prasad, R. Regular Intake of Green Tea Polyphenols Suppresses the Development of Nonmelanoma Skin Cancer through miR-29-Mediated Epigenetic Modifications. J. Clin. Med. 2022, 11, 398. [Google Scholar] [CrossRef]
  148. Huang, Y.; Li, Z.; Lin, E.; He, P.; Ru, G. Oxidative damage-induced hyperactive ribosome biogenesis participates in tumorigenesis of offspring by cross-interacting with the Wnt and TGF-beta1 pathways in IVF embryos. Exp. Mol. Med. 2021, 53, 1792–1806. [Google Scholar] [CrossRef]
  149. Renzetti, A.; Betts, J.W.; Fukumoto, K.; Rutherford, R.N. Antibacterial green tea catechins from a molecular perspective: Mechanisms of action and structure-activity relationships. Food Funct. 2020, 11, 9370–9396. [Google Scholar] [CrossRef]
  150. Zada, D.; Sela, Y.; Matosevich, N.; Monsonego, A.; Lerer-Goldshtein, T.; Nir, Y.; Appelbaum, L. Parp1 promotes sleep, which enhances DNA repair in neurons. Mol. Cell 2021, 81, 4979–4993.e7. [Google Scholar] [CrossRef]
  151. DeBardeleben, H.K.; Lopes, L.E.; Nessel, M.P.; Raizen, D.M. Stress-Induced Sleep After Exposure to Ultraviolet Light Is Promoted by p53 in Caenorhabditis elegans. Genetics 2017, 207, 571–582. [Google Scholar] [CrossRef] [Green Version]
  152. Cheung, V.; Yuen, V.M.; Wong, G.T.C.; Choi, S.W. The effect of sleep deprivation and disruption on DNA damage and health of doctors. Anaesthesia 2019, 74, 434–440. [Google Scholar] [CrossRef] [Green Version]
  153. Zada, D.; Bronshtein, I.; Lerer-Goldshtein, T.; Garini, Y.; Appelbaum, L. Sleep increases chromosome dynamics to enable reduction of accumulating DNA damage in single neurons. Nat. Commun. 2019, 10, 1–12. [Google Scholar] [CrossRef]
  154. Sellegounder, D.; Liu, Y.; Wibisono, P.; Chen, C.-H.; Leap, D.; Sun, J. Neuronal GPCR NPR-8 regulates C. elegans defense against pathogen infection. Sci. Adv. 2019, 5, eaaw4717. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Burton, J.C.; Antoniades, W.; Okalova, J.; Roos, M.M.; Grimsey, N.J. Atypical p38 Signaling, Activation, and Implications for Disease. Int. J. Mol. Sci. 2021, 22, 4183. [Google Scholar] [CrossRef] [PubMed]
  156. Eckert, R.L.; Efimova, T.; Balasubramanian, S.; Crish, J.F.; Bone, F.; Dashti, S. p38 Mitogen-activated protein kinases on the body surface—A function for p38 delta. J. Investig. Dermatol. 2003, 120, 823–828. [Google Scholar] [CrossRef] [PubMed]
  157. Enslen, H.; Raingeaud, J.; Davis, R.J. Selective activation of p38 mitogen-activated protein (MAP) kinase isoforms by the MAP kinase kinases MKK3 and MKK6. J. Biol. Chem. 1998, 273, 1741–1748. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Lin, X.; Wang, M.; Zhang, J.; Xu, R. p38 MAPK: A Potential Target of Chronic Pain. Curr. Med. Chem. 2014, 21, 4405–4418. [Google Scholar] [CrossRef]
  159. Sarikaya, D.P.; Cridland, J.; Tarakji, A.; Sheehy, H.; Davis, S.; Kochummen, A.; Hatmaker, R.; Khan, N.; Chiu, J.; Begun, D.J. Phenotypic coupling of sleep and starvation resistance evolves in D. melanogaster. BMC Evol. Biol. 2020, 20, 126. [Google Scholar] [CrossRef]
  160. Szentirmai, E.; Kapas, L. Sleep and body temperature in TNF alpha knockout mice: The effects of sleep deprivation, beta 3-AR stimulation and exogenous TNF alpha. Brain Behav. Immun. 2019, 81, 260–271. [Google Scholar] [CrossRef]
  161. Nguyen, J.; Gibbons, C.M.; Dykstra-Aiello, C.; Ellingsen, R.; Koh, K.M.S.; Taishi, P.; Krueger, J.M. Interleukin-1 receptor accessory proteins are required for normal homeostatic responses to sleep deprivation. J. Appl. Physiol. 2019, 127, 770–780. [Google Scholar] [CrossRef]
  162. Xia, X.; Xu, J.; Wang, X.; Wang, H.; Lin, Z.; Shao, K.; Fang, L.; Zhang, C.; Zhao, Y. Jiaogulan tea (Gpostemma pentaphyllum) potentiates the antidiabetic effect of white tea via the AMPK and PI3K pathways in C57BL/6 mice. Food Funct. 2020, 11, 4339–4355. [Google Scholar] [CrossRef]
  163. Xiong, L.G.; Pan, L.Y.; Gong, Y.S.; Huang, J.A.; Liu, Z.H. Fuzhuan Tea protects Caenorhabditis elegans from glucose and advanced glycation end products via distinct pathways. J. Funct. Foods 2019, 59, 148–155. [Google Scholar] [CrossRef]
  164. Hu, S.; Li, S.; Liu, Y.; Sun, K.; Luo, L.; Zeng, L. Aged Ripe Pu-erh Tea Reduced Oxidative Stress-Mediated Inflammation in Dextran Sulfate Sodium-Induced Colitis Mice by Regulating Intestinal Microbes. J. Agric. Food Chem. 2021, 69, 10592–10605. [Google Scholar] [CrossRef]
  165. Cai, X.; Quan, H.; Chang, D.; Bi, J.; Zhang, K. Metabolic rate and substrate oxidation of young males with obesity at the different sleep stages. Obes. Res. Clin. Pract. 2022, 16, 17–22. [Google Scholar] [CrossRef]
  166. Szentirmai, E.; Kapas, L. Brown adipose tissue plays a central role in systemic inflammation-induced sleep responses. PLoS ONE 2018, 13, e0197409. [Google Scholar] [CrossRef] [Green Version]
  167. Juozaityte, V.; Pladevall-Morera, D.; Podolska, A.; Norgaard, S.; Neumann, B.; Pocock, R. The ETS-5 transcription factor regulates activity states in Caenorhabditis elegans by controlling satiety. Proc. Natl. Acad. Sci. USA 2017, 114, E1651–E1658. [Google Scholar] [CrossRef] [Green Version]
  168. Wilms, B.; Leineweber, E.M.; Molle, M.; Chamorro, R.; Pommerenke, C.; Salinas-Riester, G.; Sina, C.; Lehnert, H.; Oster, H.; Schmid, S.M. Sleep Loss Disrupts Morning-to-Evening Differences in Human White Adipose Tissue Transcriptome. J. Clin. Endocrinol. Metab. 2019, 104, 1687–1696. [Google Scholar] [CrossRef] [Green Version]
  169. Song, Q.; Liu, X.; Zhou, W.; Wu, S.; Wang, X. Night sleep duration and risk of each lipid profile abnormality in a Chinese population: A prospective cohort study. Lipids Health Dis. 2020, 19, 185. [Google Scholar] [CrossRef]
  170. Tasali, E.; Wroblewski, K.; Kahn, E.; Kilkus, J.; Schoeller, D.A. Effect of Sleep Extension on Objectively Assessed Energy Intake Among Adults With Overweight in Real-life Settings: A Randomized Clinical Trial. JAMA Intern. Med. 2022, 182, 365–374. [Google Scholar] [CrossRef]
  171. Liu, C.; Guo, Y.; Sun, L.; Lai, X.; Li, Q.; Zhang, W.; Xiang, L.; Sun, S.; Cao, F. Six types of tea reduce high-fat-diet-induced fat accumulation in mice by increasing lipid metabolism and suppressing inflammation. Food Funct. 2019, 10, 2061–2074. [Google Scholar] [CrossRef]
  172. Wang, J.; Li, P.; Liu, S.; Zhang, B.W.; Hu, Y.Z.; Ma, H.; Wang, S. Green tea leaf powder prevents dyslipidemia in high-fat diet-fed mice by modulating gut microbiota. Food Nutr. Res. 2020, 64, 3672. [Google Scholar] [CrossRef]
  173. Huang, F.; Wang, S.; Zhao, A.; Zheng, X.; Zhang, Y.; Lei, S.; Ge, K.; Qu, C.; Zhao, Q.; Yan, C.; et al. Pu-erh Tea Regulates Fatty Acid Metabolism in Mice Under High-Fat Diet. Front. Pharmacol. 2019, 10, 63. [Google Scholar] [CrossRef]
  174. Zhou, M.X.; Tian, X.; Wu, Z.Q.; Li, K.; Li, Z.J. Fuzhuan brick tea supplemented with areca nuts: Effects on serum and gut microbiota in mice. J. Food Biochem. 2021, 45, e13737. [Google Scholar] [CrossRef] [PubMed]
  175. Liu, Y.; Lang, H.; Zhou, M.; Huang, L.; Hui, S.; Wang, X.; Chen, K.; Mi, M. The Preventive Effects of Pterostilbene on the Exercise Intolerance and Circadian Misalignment of Mice Subjected to Sleep Restriction. Mol. Nutr. Food Res. 2020, 64, e1900991. [Google Scholar] [CrossRef] [PubMed]
  176. Yurgel, M.E.; Kakad, P.; Zandawala, M.; Nassel, D.R.; Godenschwege, T.A.; Keene, A.C. A single pair of leucokinin neurons are modulated by feeding state and regulate sleep-metabolism interactions. PLoS Biol. 2019, 17, e2006409. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Funato, H.; Miyoshi, C.; Fujiyama, T.; Kanda, T.; Kanda, T.; Sato, M.; Wang, Z.; Ma, J.; Nakane, S.; Tomita, J.; et al. Forward-genetics analysis of sleep in randomly mutagenized mice. Nature 2016, 539, 378–383. [Google Scholar] [CrossRef]
  178. Park, M.; Miyoshi, C.; Fujiyama, T.; Kakizaki, M.; Ikkyu, A.; Honda, T.; Choi, J.; Asano, F.; Mizuno, S.; Takahashi, S.; et al. Loss of the conserved PKA sites of SIK1 and SIK2 increases sleep need. Sci. Rep. 2020, 10, 1–14. [Google Scholar] [CrossRef]
  179. Iwasaki, K.; Fujiyama, T.; Nakata, S.; Park, M.; Miyoshi, C.; Hotta-Hirashima, N.; Ikkyu, A.; Kakizaki, M.; Sugiyama, F.; Mizuno, S.; et al. Induction of Mutant Sik3(Sleepy) Allele in Neurons in Late Infancy Increases Sleep Need. J. Neurosci. 2021, 41, 2733–2746. [Google Scholar] [CrossRef]
  180. Grubbs, J.J.; Lopes, L.E.; van der Linden, A.M.; Raizen, D.M. A salt-induced kinase is required for the metabolic regulation of sleep. PLoS Biol. 2020, 18, e3000220. [Google Scholar] [CrossRef] [Green Version]
  181. An, R.; Wen, S.; Li, D.L.; Li, Q.H.; Lai, X.F.; Zhang, W.J.; Chen, R.H.; Cao, J.X.; Li, Z.G.; Huang, Q.S.; et al. Mixtures of Tea and Citrus maxima (pomelo) Alleviate Lipid Deposition in HepG2 Cells Through the AMPK/ACC Signaling Pathway. J. Med. Food 2020, 23, 943–951. [Google Scholar] [CrossRef]
  182. Mika, M.; Wikiera, A.; Antonczyk, A.; Grabacka, M. The impact of catechins included in high fat diet on AMP-dependent protein kinase in apoE knock-out mice. Int. J. Food Sci. Nutr. 2021, 72, 348–356. [Google Scholar] [CrossRef]
  183. Wang, Y.J.; Pan, H.T.; Chen, D.; Guo, D.D.; Wang, X.Y. Targeting at cancer energy metabolism and lipid droplet formation as new treatment strategies for epigallocatechin-3-gallate (EGCG) in colorectal cancer cells. J. Funct. Foods 2021, 83, 104570. [Google Scholar] [CrossRef]
  184. Karp, X. Hormonal Regulation of Diapause and Development in Nematodes, Insects, and Fishes. Front. Ecol. Evol. 2021, 9, 735924. [Google Scholar] [CrossRef]
  185. Wang, L.; Chen, H.; Wang, L.L.; Song, L.S. An insulin-like peptide serves as a regulator of glucose metabolism in the immune response of Chinese mitten crab Eriocheir sinensis. Dev. Comp. Immunol. 2020, 108, 103686. [Google Scholar] [CrossRef]
  186. Li, J.T.S.; Hon, K.L.; Leung, A.K.C.; Lee, V.W.Y. Pharmacologic Evidence of Green Tea in Targeting Tyrosine Kinases. Curr. Rev. Clin. Exp. Pharmacol. 2021, 16, 239–246. [Google Scholar] [CrossRef]
  187. Ni, D.; Ai, Z.; Munoz-Sandoval, D.; Suresh, R.; Ellis, P.R.; Yuqiong, C.; Sharp, P.A.; Butterworth, P.J.; Yu, Z.; Corpe, C.P. Inhibition of the facilitative sugar transporters (GLUTs) by tea extracts and catechins. FASEB J. 2020, 34, 9995–10010. [Google Scholar] [CrossRef]
  188. Tian, J.; Geiss, C.; Zarse, K.; Madreiter-Sokolowski, C.T.; Ristow, M. Green tea catechins EGCG and ECG enhance the fitness and lifespan of Caenorhabditis elegans by complex I inhibition. Aging 2021, 13, 22629–22648. [Google Scholar] [CrossRef]
  189. Hirano, N.; Sakamoto, K. Linalool odor stimulation improves heat stress tolerance and decreases fat accumulation in nematodes. Biosci. Biotechnol. Biochem. 2019, 83, 148–154. [Google Scholar] [CrossRef] [Green Version]
  190. Kimura, S.; Toyoura, M.; Toyota, Y.; Takaoka, Y. Serum concentrations of insulin-like growth factor-1 as a biomarker of improved circadian rhythm sleep-wake disorder in school-aged children. J. Clin. Sleep Med. 2020, 16, 2073–2078. [Google Scholar] [CrossRef]
  191. Zegarra-Valdivia, J.A.; Pignatelli, J.; Fernandez de Sevilla, M.E.; Fernandez, A.M.; Munive, V.; Martinez-Rachadell, L.; Nunez, A.; Torres Aleman, I. Insulin-like growth factor I modulates sleep through hypothalamic orexin neurons. FASEB J. 2020, 34, 15975–15990. [Google Scholar] [CrossRef]
  192. Brown, E.B.; Shah, K.D.; Faville, R.; Kottler, B.; Keene, A.C. Drosophila insulin-like peptide 2 mediates dietary regulation of sleep intensity. PLoS Genet. 2020, 16, e1008270. [Google Scholar] [CrossRef]
  193. He, Q.; Du, J.; Wei, L.; Zhao, Z. AKH-FOXO pathway regulates starvation-induced sleep loss through remodeling of the small ventral lateral neuron dorsal projections. PLoS Genet. 2020, 16, e1009181. [Google Scholar] [CrossRef]
  194. Kelly, M.L.; Collins, S.P.; Lesku, J.A.; Hemmi, J.M.; Collin, S.P.; Radford, C.A. Energy conservation characterizes sleep in sharks. Biol. Lett. 2022, 18, 20210259. [Google Scholar] [CrossRef] [PubMed]
  195. Wu, Y.; Masurat, F.; Preis, J.; Bringmann, H. Sleep Counteracts Aging Phenotypes to Survive Starvation-Induced Developmental Arrest in C. elegans. Curr. Biol. 2018, 28, 3610–3624.e8. [Google Scholar] [CrossRef] [Green Version]
  196. Hwang, S.K.; Kim, H.H. The functions of mTOR in ischemic diseases. BMB Rep. 2011, 44, 506–511. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  197. Shams, R.; Ito, Y.; Miyatake, H. Mapping of mTOR drug targets: Featured platforms for anti-cancer drug discovery. Pharmacol. Ther. 2021, 232, 108012. [Google Scholar] [CrossRef] [PubMed]
  198. Zhang, B.; Guo, D.J.; Han, L.R.; Rensing, N.; Satoh, A.; Wong, M. Hypothalamic orexin and mechanistic target of rapamycin activation mediate sleep dysfunction in a mouse model of tuberous sclerosis complex. Neurobiol. Dis. 2019, 134, 104615. [Google Scholar] [CrossRef]
  199. Goetting, D.L.; Soto, R.; Van Buskirk, C. Food-Dependent Plasticity in Caenorhabditis elegans Stress-Induced Sleep Is Mediated by TOR-FOXA and TGF- Signaling. Genetics 2018, 209, 1183–1195. [Google Scholar] [CrossRef] [Green Version]
  200. Du, H.; Wang, Q.; Yang, X. Fu Brick Tea Alleviates Chronic Kidney Disease of Rats with High Fat Diet Consumption through Attenuating Insulin Resistance in Skeletal Muscle. J. Agric. Food Chem. 2019, 67, 2839–2847. [Google Scholar] [CrossRef]
  201. Zhao, W.; Wang, J.; Bi, W.; Ferruzzi, M.; Yemul, S.; Freire, D.; Mazzola, P.; Ho, L.; Dubner, L.; Pasinetti, G.M. Novel application of brain-targeting polyphenol compounds in sleep deprivation-induced cognitive dysfunction. Neurochem. Int. 2015, 89, 191–197. [Google Scholar] [CrossRef] [Green Version]
  202. Yoneda, Y.; Kuramoto, N.; Kawada, K. The role of glutamine in neurogenesis promoted by the green tea amino acid theanine in neural progenitor cells for brain health. Neurochem. Int. 2019, 129, 104505. [Google Scholar] [CrossRef]
  203. Zhou, Y.; Zhou, X.; Huang, X.; Hong, T.; Zhang, K.; Qi, W.; Guo, M.; Nie, S. Lysosome-Mediated Cytotoxic Autophagy Contributes to Tea Polysaccharide-Induced Colon Cancer Cell Death via mTOR-TFEB Signaling. J. Agric. Food Chem. 2021, 69, 686–697. [Google Scholar] [CrossRef]
Figure 1. Main active ingredients of tea that affect sleep.
Figure 1. Main active ingredients of tea that affect sleep.
Foods 11 03930 g001
Figure 2. Tea coordinates sickness sleep, and energy homeostasis under stress. (A). Tea may act on damage repair and energy metabolism to promote sickness sleep and maintain energy homeostasis. (B). The energy homeostasis of animals is maintained under the dynamic balance of consumption and supply under stress. Sickness sleep, as a strategy for conserving energy consumption and optimizing energy allocation, helps to regulate damage repair and energy metabolism to maintain energy homeostasis under stress.
Figure 2. Tea coordinates sickness sleep, and energy homeostasis under stress. (A). Tea may act on damage repair and energy metabolism to promote sickness sleep and maintain energy homeostasis. (B). The energy homeostasis of animals is maintained under the dynamic balance of consumption and supply under stress. Sickness sleep, as a strategy for conserving energy consumption and optimizing energy allocation, helps to regulate damage repair and energy metabolism to maintain energy homeostasis under stress.
Foods 11 03930 g002
Figure 3. Tea regulates sickness sleep through the brain–gut axis. Components of tea act on the nervous system under stress. These components reduce neuronal ROS levels and cell apoptosis and promote neuronal growth and differentiation; inhibit glial pathology and reduce inflammation; and regulate the secretion of neurotransmitters, including ACH, GABA, and 5-HT. Furthermore, tea affects the intestinal flora, specifically by reducing harmful bacteria and promoting the production of beneficial bacteria metabolism.
Figure 3. Tea regulates sickness sleep through the brain–gut axis. Components of tea act on the nervous system under stress. These components reduce neuronal ROS levels and cell apoptosis and promote neuronal growth and differentiation; inhibit glial pathology and reduce inflammation; and regulate the secretion of neurotransmitters, including ACH, GABA, and 5-HT. Furthermore, tea affects the intestinal flora, specifically by reducing harmful bacteria and promoting the production of beneficial bacteria metabolism.
Foods 11 03930 g003
Table 1. The main health benefits of the six major tea types.
Table 1. The main health benefits of the six major tea types.
Types of TeaDegree of FermentationKey Processing TechnologyMajor Health Benefits
Green teaNon-fermentedFixing Antibacterial [34]
Suppressing the amyloid beta levels and alleviating cognitive impairment in 5XFAD mice [35]
Reducing lipid peroxidation and increasing total antioxidant capacity, and reducing oxidative damage [36]
Significantly lowering the risk of developing liver cancer and improving the effect on body mass index, liver enzymes, and lipoprotein [37]
Preventing obesity [38,39]
Black teaFully fermentedFermentationExerting antibacterial activity against major periodontopathogens, attenuating the secretion of IL-8, and inducing hBD secretion in oral epithelial cells [40]
Preventing radiation-induced increase of ACE activity and oxidative stress in the aorta [41]
Limiting the formation of glycation products [42]
Yellow teaSlightly fermentedYellowingAntioxidant and preventing gastric injury [43]
Reducing blood glucose levels, increasing glucose tolerance, and preventing fatty liver in diabetes mice [44]
Oolong teaSemi-fermentedRotatingNeurodegenerative and neurite outgrowth-promoting [45]
Inhibiting cancer cell proliferation [46]
Providing cardio-protective benefits during hypoxic conditions [47]
Prolonging lifespan and improving health span by curtailing the age-related decline in muscle activity and the accumulation of age pigment in C. elegans [48]
White teaSlightly fermentedWitheringInhibiting PhlP-induced aberrant crypt foci by altering the expression of carcinogen-metabolizing enzymes in rats [49]
Dark teaPost-fermentedPile fermentationDecreasing risks of coronary heart disease and diabetes [50]
Scavenging of DPPH and ABTS free radicals [51]
Regulating the glycolipid metabolic disorders [52]
Decreasing body weight and serum triglycerides for SD rats [53]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Ouyang, J.; Peng, Y.; Gong, Y. New Perspectives on Sleep Regulation by Tea: Harmonizing Pathological Sleep and Energy Balance under Stress. Foods 2022, 11, 3930. https://doi.org/10.3390/foods11233930

AMA Style

Ouyang J, Peng Y, Gong Y. New Perspectives on Sleep Regulation by Tea: Harmonizing Pathological Sleep and Energy Balance under Stress. Foods. 2022; 11(23):3930. https://doi.org/10.3390/foods11233930

Chicago/Turabian Style

Ouyang, Jin, Yuxuan Peng, and Yushun Gong. 2022. "New Perspectives on Sleep Regulation by Tea: Harmonizing Pathological Sleep and Energy Balance under Stress" Foods 11, no. 23: 3930. https://doi.org/10.3390/foods11233930

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop