Next Article in Journal
The Sweet Side of Constipation: Colonic Motor Dysfunction in Diabetes Mellitus
Previous Article in Journal
Anti-Obesity Potential of Modified Pomelo-Peel Dietary Fiber-Based Pickering Emulsion
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Exploring the Impact and Mechanisms of Coffee and Its Active Ingredients on Depression, Anxiety, and Sleep Disorders

1
School of Public Health, Wuhan University of Science and Technology, Wuhan 430065, China
2
Hubei Provincial Center for Disease Control and Prevention, Wuhan 430079, China
3
Huanggang City Center for Disease Control and Prevention, Huanggang 438000, China
*
Authors to whom correspondence should be addressed.
This authors contributed equally to this work.
Nutrients 2025, 17(19), 3037; https://doi.org/10.3390/nu17193037
Submission received: 18 August 2025 / Revised: 12 September 2025 / Accepted: 19 September 2025 / Published: 24 September 2025
(This article belongs to the Section Nutrition and Neuro Sciences)

Abstract

Depression, anxiety, and accompanying sleep disorders are common mood disorders worldwide, significantly affecting individuals’ physical and mental well-being as well as their quality of life. Coffee is a widely consumed beverage rich in bioactive compounds, including caffeine, polyphenols, tannins, proteins, and minerals, and studies suggest that coffee and its bioactive constituents demonstrate potential benefits for mental health. However, the specific mechanism by which coffee regulates depression, anxiety and sleep disorders is still unclear, and there is a lack of systematic research in this regard. This study searched four databases (PubMed, Embase, Web of Science, and Google Scholar) to summarize the effects of coffee and its active ingredients on depression, anxiety, and sleep disorders and their mechanisms. Keywords included “coffee”, “active ingredients”, “depression”, “anxiety”, “Sleep disorders “and “mechanisms”. This review summarizes 27 animal studies, 11 clinical studies, and 6 epidemiological studies on this topic. The results showed that moderate caffeine intake may improve mood and cognitive performance, while excessive intake may be associated with anxiety, insomnia, and post-traumatic stress disorder (PTSD). Other compounds, such as polyphenols, may contribute to mental health through antioxidant, anti-inflammatory, and neuroprotective mechanisms. Future research is needed to clarify effective dosages, assess long-term safety, improve coffee processing methods, and explore the development of coffee-based functional foods.

Graphical Abstract

1. Introduction

Depression and anxiety often coexist with sleep disturbances [1]. Depression, anxiety and SDs are common mental health problems that significantly interfere with patients’ daily lives, and can lead to other physical health problems [2]. Depression is the leading cause of mental and physical disability, and a major contributor to the global burden of disease worldwide. According to the 2021 WHO, approximately 280 million people (3.8% of the total population) suffered from depression worldwide, and more than 700,000 people died by suicide each year due to depression, with low mood, lack of energy, sadness, insomnia, and the inability to enjoy life [3]. Anxiety is also one of the most common mental health problems worldwide, affecting approximately 25% of the global population, which is characterized by excessive and persistent feelings of fear and worry, that are disproportionate to the actual threats faced, leading to impairment in the patient’s daily life functions [4]. Major depressive disorder (MDD) and anxiety disorders increased by 28% and 26%, respectively, in 2020, with large increases in prevalence among people living in countries severely affected by COVID-19 [5]. SDs are very common, highly treatable conditions that often go untreated, despite their deleterious effects on health and safety. Untreated SDs can reduce alertness and have deleterious effects on cognitive and psychomotor performance [6,7], more than 50% of sleep initiation and maintenance disorders are caused by psychiatric disorders, including addiction [8].
Currently, the treatment and management of depression, anxiety and SDs still face many challenges. In the late 1950s, the first tricyclic drugs (TCAs) were approved for the treatment of depression, mainly imipramine, amitriptyline, clomipramine, desipramine, and doxepin [9]. Other antidepressants include selective 5-HT reuptake inhibitors (SSRIs), fluoxetine, sertraline, paroxetine, and escitalopram [10]; serotonin–norepinephrine (NE) reuptake inhibitors (SNRIs) include milnacipran, duloxetine (DXT), desvenlafaxine succinate (DVS), and venlafaxine [11]. These drugs may cause a range of side effects, including but not limited to weight gain, nausea, sexual dysfunction, and, in certain cases, an increased risk of suicidal tendencies [12]. Canadian clinical treatment guidelines recommend that SSRIs or SNRIs be used as first-line anti-anxiety drug treatment, while benzodiazepines, TCAs, and other drugs are considered second or third-line treatments for panic disorder, generalized anxiety disorder (GAD), and social anxiety disorder (SAD) [13]. However, these treatments are not effective for about a third of patients, who may develop anxiety-related disorders, such as post-traumatic stress disorder (PTSD) and obsessive–compulsive disorder (OCD) [4,14]. Many licensed drugs (including benzodiazepines, daridorexant, suvorexant, and trazodone) are effective for the acute treatment of insomnia, but are poorly tolerated [15]. In recent years, people have been paying increasing attention to natural therapies that utilize natural ingredients from herbs and foods. Such therapies have potential benefits as alternative or adjuvant treatments for mental health problems [16]. For example, in China, Bupleurum has shown some beneficial effects as an adjunctive treatment for post-stroke depression (PSD) [17]. The herb St. John’s wort (hypericum perforatum) may be effective in treating mild to moderate depression [18]. Studies had shown that kava had an anti-anxiety effect on humans [19]. Chinese herbal medicines, such as semen Ziziphus jujube, licorice, and peony had a sedative and hypnotic effect on people with SDs [20].
Coffee, one of the most popular beverages in the world, contains more than a thousand different ingredients, the most well-known of which is caffeine, as well as phenolic acids, diterpenes, trigonelline, tryptophan alkaloids, etc. [21]. Among these identified bioactive ingredients, caffeine can be used as a highly effective stimulant and bronchodilator, while phenolic acids such as chlorogenic acid (CGA), ferulic acid (FA), and caffeic acid (CA) represent the many antioxidant and anti-inflammatory ingredients in coffee. Caffeine remains the most intensively studied compound in coffee, and its effects largely explain why coffee has become such a popular beverage [22]. Coffee has been found to have beneficial effects on human health, such as reduced all-cause and cardiovascular disease-related mortality [23,24], and a lower risk of type 2 diabetes [25,26], Parkinson disease (PD) [27], non-alcoholic fatty liver disease [28], cancer [27,29], and other diseases. Consuming non-toxic doses of caffeine has a variety of positive effects on the brain, including enhanced feelings of well-being, alertness, improved concentration, improved mood, and a reduced risk of depression. However, caffeine may also affect sleep quality in some sensitive individuals and induce anxiety symptoms in some cases [30].
Caffeine is a naturally occurring methylxanthine that acts primarily as a non-selective adenosine A1 and A2A receptor (A1R and A2AR) antagonist and forms functional isomers with dopamine (DA) receptors D1 and D2 in different areas of the brain [31]. Caffeine blocks the binding of A1R/A2AR to adenosine. This effect was manifested in rat experiments with increased secretion of striatal DA after high dose intake [32]. There is also recent evidence that regular consumption of coffee and caffeinated products can affect resting brain connectivity and influence mood, alertness, and readiness for action [33,34]. The specific performance is that coffee intake reduces the functional connectivity between the posterior default mode network (DMN), and the somatosensory/motor network-prefrontal cortex (PFC) [33]. Furthermore, certain functional networks (involving subcortical areas, visual networks, and cerebellum) showed longer connection durations (signature of longer lifespan) [34].
Despite a long history of coffee consumption, the association between coffee and mood disorders remains unclear. The impact mechanisms, long-term effects and dosage relationships of coffee, and its different bioactive components on mood disorders still need to be further explored. This study aims to systematically evaluate the effects of coffee and its active ingredients on mood disorders (including depression, anxiety, and accompanying sleep disorders) and provide a basis for scientific and rational coffee consumption.

2. Methods

2.1. Inclusion Criteria

The inclusion criteria are as follows: (1) included studies need to involve coffee drinking or its main active ingredients (such as caffeine, chlorogenic acid, ferulic acid, caffeic acid, etc.); (2) included studies are required to report at least one outcome related to depression, anxiety, or sleep disorders, including but not limited to clinical diagnostic criteria (such as DSM-IV), scale scores (such as depression/anxiety scales), behavioral tests (such as FST, TST, OFT), or biological indicators (such as neurotransmitters, inflammatory factors); and (3) the literature must be written in English and included in the database, with the search time up to 2025.

2.2. Exclusion Criteria

The exclusion criteria are (1) articles for which an abstract is not available; (2) non-research articles such as conferences, newsletters, and reviews; (3) duplicate articles; (4) no study outcomes related to depression, anxiety, or sleep disorders are included, only discussing other mental and emotional health issues; (5) coffee or its active ingredients are not mentioned; and (6) the full text is not available.

2.3. Search Strategy

We searched electronic databases including PubMed, Web of Science, Embase, and Google Scholar using the keywords “coffee,” “active ingredient,” “depression,” “anxiety,” “sleep disorders,” and “mechanism.” All retrieved study abstracts were read and ranked according to the inclusion and exclusion criteria. The initially included articles were then carefully read and further excluded based on the content of the main articles (Figure 1).

3. Associations Between SDs and Depression, Anxiety

SDs are often a sign of a mood disorder diagnosis [35,36]. It is difficult that SDs enter or maintain sleep, which can affect attention and memory in the short term and reduce immunity in the long term, inducing psychological problems such as anxiety and depression. Improving sleep is important in alleviating these symptoms [37,38,39]. The overlap between depression and anxiety is well established; more than 50% of people with depression reported clinically significant anxiety symptoms, and they were more treatment resistant than depressed people without anxiety [40]. SDs also have complex connections with depression. SDs were risk factor for depressive episodes. It is a common physical symptom in MDD and one of the diagnostic criteria for depression based on the Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition (DSM-IV) [41,42]. In addition, insomnia can affect the trajectory of depression, affecting its severity, duration, and recurrence rate [43]. The prevalence of poor sleep quality is high among patients with PD, PTSD, and GAD. Sleep problems can predict the occurrence of anxiety disorders, and many anxiety disorders (such as GAD) already include SDs such as insomnia as key diagnostic criteria [44].

4. Effects of Coffee and Its Active Ingredients on Depression

4.1. Animal Experimental Study on the Effects of Coffee and Its Active Ingredients on Depression

The sugar preference test (SPT) is used to assess the motivation of mice for a palatable stimulus (1% sucrose solution). Low preference for sucrose (relative to water) is often interpreted as anhedonia in mouse models of depression [45]. The forced swim test (FST) model has been used for decades to test the antidepressant potential of drugs [46]. The tail suspension test (TST) proposed by Steru has a similar depression-inducing mechanism as the FST [47]. The open field test (OFT) is used to determine the depression of model animals based on their activity in unfamiliar open areas [48] (Figure 2A).
Several ingredients of coffee, such as caffeine [49], CGA [50], FA and CA [51], have shown antidepressant effects in many animal and clinical studies. The amounts [52], type [53], and roast degree [54] of coffee consumed may affect the risk of depression. After consuming coffee and decaffeinated coffee, the abundance of beneficial intestinal flora such as S24-7, Lachnospiraceae, Oscillospira and Akkermansia was restored to varying degrees in rats with chronic paradoxical sleep deprivation model, the levels of pro-inflammatory factors such as interleukin (IL)-6 and tumor necrosis factor (TNF)-α were reduced, and the levels of superoxide dismutase (SOD) and glutathione peroxidase (GPx) were increased. Both coffee and decaffeinated coffee can improve depressive-like behaviors induced by sleep deprivation [53]. Caffeinated coffee reduces indoleamine 2,3-dioxygenase (IDO) activity after exposure to lipopolysaccharide (LPS). Increased uric acid (UA)/3-Hydroxykynurenine (3-HK) and UA/tryptophan-KYN)ratios, the balance of the KYN pathway (KP) in the central nervous system (CNS) shifts from a neurotoxic group to a neuroprotective group, the immobility time in both TST and FST was significantly shortened, which may have an antidepressant effect [55]. Caffeine prevents chronic unpredictable stress (CUS)-induced mood (depression) and memory dysfunction by selectively blocking adenosine A2AR mimicry. Treatment of CUS mice with the A2AR antagonist (SCH58261) resulted in reduced densities of syntaxin, SNAP-25, and vGluT1, and reduced amplitudes of long-term potentiation (LTP) in the mouse hippocampus. This suggests that synaptic A2AR plays a key role in modulating the effects of chronic stress on the brain, which is a candidate target for mitigating the effects of chronic stress on brain function [56]. Caffeine also reduced hippocampal inflammatory response by antagonizing A2AR receptors, inhibiting corticosterone (CORT)-induced microglia activation and mitogen-activated protein kinase (MAPK)/extracellular regulated protein kinases (ERK)/nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling pathway, thereby restoring neural stem cell proliferation and differentiation, and ameliorating depressive-like behavior in chronic water immersion restraint stress (CWIRS)mice [57].
In addition, caffeine ameliorated chronic mild stress (CMS)-induced depressive-like behavior in mice by targeting the peroxisome proliferators-activated receptor γ coactivator lα (PGC-1α)-kynurenine aminotransferase (KAT) axis in skeletal muscle, inhibiting the neurotoxic branch metabolism of KYN, and promoting its conversion to neuroprotective KYN acid (KYNA). This mechanism is similar to the molecular pathway of exercise antidepressant, and provides a new theoretical basis for caffeine as a potential antidepressant [58]. A study suggested that caffeine’s role in anxiety and depressive behaviors might be gender-specific. Caffeine showed antidepressant effects in the FST assay, with male rats showing primarily increased swimming time, and female rats showing more struggling behavior. This may be related to neurotransmitter systems (such as 5-HT and NE) as well as neural adaptations in the striatum [59]. In addition, there is now a coffee derivative, SY-2476, which has also been shown to have significant anti-depressant effects, improving sugar-water preference, decreasing immobility time, lowering CORT levels, and increasing antioxidant enzyme levels, as well as modulating adenosine A1/A2A receptor expression in rats [60]. In terms of ingested dose, low doses of caffeine significantly ameliorated LPS-induced depressive-like behavior through anti-inflammatory, antioxidant and up-regulation of brain-derived neurotrophic factor (BDNF) [61]. In terms of coffee intake time, chronic caffeine intake reversed memory deficits but was ineffective against established depressive and anxiety behaviors. Since memory impairment is strongly associated with major depression, caffeine may be better suited as a means of preventing the onset of cognitive decline and depression rather than treating existing symptoms [62,63]. However, the effects of caffeine on depression are not all positive. Studies have shown that caffeine exacerbates depressive-like behaviors in the stress-induced stress (SRS) model of PTSD mice when administered repeatedly (20 and 30 mg/kg). This discrepancy may be due to the use of different mouse models [64]. Caffeine can also improve the subjective well-being of rats by regulating the A1R. A study investigating the health of mice showed that caffeine regulated the function of specific brain regions through A1R, and that the effects differed by sex: the A1R effect was more pronounced in the ventral hippocampus of women, while the effect was more prominent in the dorsolateral striatum of men. Caffeinated coffee improves subjective well-being in both sexes by enhancing A1R inhibition in the ventral hippocampus and striatum, which is manifested by increased sociability in men and reduced rank competition and improved self-care in women [65].
CGA pretreatment at the genus level can regulate the relative abundance changes in key bacteria such as Desulfovibrionales, Desulfovibrio, Klebsiella, Burkholderiales and Bifidobacterium. This optimized the gut microbiome structure of rats treated with adrenocorticotropic hormone (ACTH), which may have a positive impact on its anti-depressant effect [50]. When FA is used in combination with drugs such as fluoxetine, paroxetine and sertraline, it can exert an antidepressant effect through 5-HT receptor activity and significantly shorten the immobility time of mice in the TST model, suggesting that the combination of FA and conventional antidepressants may provide a new option for the treatment of depression [66]. CA can reduce the duration of immobility and freezing caused by forced swimming and conditioned fear stress in mice, and its effects are inhibited byα1Aadrenoreceptor (ADRA1A) antagonists. Indirect regulation of the ADRA1A system may be involved in the antidepressant and/or anxiolytic effects of CA [67] (Table 1).

4.2. Clinical Study on the Effects of Coffee and Its Active Ingredients on Depression

In clinical trials, the effects of coffee and its components on depressive behaviors have shown inconsistent results. A study of depressed patients treated with intermittent burst theta stimulation (iTBS) or sham treatment of the dorsomedial prefrontal cortex (dmPFC) showed that habitual caffeine intake could enhance the antidepressant effect of iTBS by antagonizing adenosine receptors, and its anti-depressant effect may be related to the enhancement of downstream DA activity [81]. Tse WS [82] asked low-caffeine users to drink caffeinated and decaffeinated coffee and evaluated the effects on social behavior and mood. The results suggest that caffeinated coffee may increase social support behaviors and help improve depressive symptoms. Similarly, Dawkins L et al. conducted a double-blind randomized controlled trial (RCT) in 88 college students and habitual coffee drinkers. The intervention group was divided into two groups: caffeinated and decaffeinated coffee. The results showed that the intake of caffeinated coffee improved attention and psychomotor speed, and had an alleviating effect on depression [83].
However, the results of a double-blind RCT by Loke and Meliska [84] showed that the more caffeine consumed, the higher the alertness, which might have a greater impact on cognitive function, but had no significant effect on depression. A prospective, double-blind, placebo-controlled, crossover study conducted by James and Gregg [85] on 48 rested or sleep-restricted participants showed that caffeine intake had no net effect on the mood of rested subjects, nor did it have a net restorative effect on the depressed mood of sleep-restricted subjects, and even weakened the positive effects of sleep on mood in both conditions (Table 2).

4.3. Epidemiological Study on the Effects of Coffee and Its Active Ingredients on Depression

The frequency, amount, and type of coffee intake are all associated with depression. In a prospective study of 9576 Korean adults, those who drank two or more cups of coffee a day had a 32% lower risk of depression than those who did not drink coffee [94]. A prospective study followed 50,739 American women with an average age of 63 for 10 years. The results showed that women who drank 2–3 cups of coffee a day had a 15% lower risk of depression compared to women who drank 1 cup or less a week. Another study also showed that drinking 2–3 cups of coffee a day can reduce the risk of suicide in men and women by about 50% [95,96]. Another cross-sectional study showed that regular coffee and caffeinated coffee intake had a potential protective effect on postpartum women, and consuming more than three cups of caffeinated coffee may reduce the risk of Postpartum Depression (PDD) in women 1–2 years after delivery and in women who are not breastfeeding [97]. A 5-year follow-up study found that the fully adjusted RD (95% CI) for black coffee was −1.7% (−2.6% to −0.7%), suggesting that black coffee might reduce the risk of depression [98] (Table 3).

4.4. The Bidirectional Effects of Coffee and Its Active Ingredients on Depression and Future Research Directions

In summary, current studies have shown that coffee and its active ingredients (such as caffeine, CGA) have a bidirectional regulatory effect on depression: moderate intake may improve depressive behavior through anti-inflammatory, antioxidant and neurotransmitter regulation pathways, while high doses or under specific stress conditions may aggravate symptoms. Therefore, coffee cannot be simply equated with a “natural antidepressant.” Epidemiological evidence shows that drinking 2–3 cups of coffee per day is significantly associated with a reduced risk of depression, but clinical results are inconsistent, which may be related to the intake dose, individual metabolic differences and coffee type (black coffee, decaffeinated coffee).
Future research may focus on (1) the synergistic antidepressant mechanism and dose–effect relationship of different components in coffee or (2) the effects of long-term coffee intake on special populations (such as postpartum women and individuals with chronic stress). It is also important to clarify the impact of coffee processing methods (roasting degree) on the retention of active ingredients and antidepressant efficacy.

5. Effects of Coffee and Its Active Ingredients on Anxiety

5.1. Animal Experimental Study on the Effects of Coffee and Its Active Ingredients on Anxiety

OFT and elevated plus maze test (EPM) are important behavioral methods for assessing animal anxiety level and motor function, and are widely used in neuro-psychopharmacology research [100]. CGA, caffeine, and coffee all improved brain enzyme activity disorders in streptozotocin-induced diabetic rats, prevented the increase in synaptic acetylcholinesterase (AchE) activity in the cerebral cortex, and reduced OS levels by reducing thiobarbituric acid reactive substances (TBARS) levels, thereby improving memory and significantly reducing anxiety [70]. Both acute and chronic low-dose caffeine reverses stress-induced anxiety-like behavior and cognitive deficits, and it is more suitable as an intervention to prevent stress-related cognitive decline and anxiety [68]. Yet another new study suggested that caffeine reduced anxiety behaviors in the short term, but this anxiety-reducing effect faded with long-term intake, though it did enhance memory in the chronic phase [69]. Other study had shown that caffeine (20 and 30 mg/kg, i.p.) injected intra-peritoneally exacerbated anxiety-like behavior induced by the stress model (SRS), as evidenced by mice exploring the open area less in the elevated plus maze (days 14 to 32). Therefore, the effect of caffeine intake on anxiety is not constant, but presents a complex and inconsistent result (Figure 2B).
Isochlorogenic acid (ICCA) improves the function of the cholinergic system and enhances antioxidant and anti-inflammatory capabilities by activating the BDNF/nuclear factor-erythroid 2 related factor 2 (Nrf2)/GPx4 pathway, thereby alleviating lead-induced neuroinflammation, ferroptosis, and OS in mice and significantly reducing anxiety-like behaviors [71]. The researchers used the light/dark box test (LDB), EPM and free exploration test (FET) to explore the anxiolytic effect of CGA in a mouse anxiety model. In LDB, CGA can increase the number of transitions from the dark box to the light box, the cumulative time in the light box, and the motor ability of mice, showing an anti-anxiety effect. In the EPM experiment, the cumulative time and total number of visits in the open arm of the CGA treatment group (20 mg/kg) increased significantly. In FET, mice given CGA (20 mg/kg) and diazepam (1 mg/kg) showed more movement and exploration behavior in unfamiliar environments than the control group. These results confirmed the anxiolytic effect of CGA. Its mechanism of action may be that the pharmacological characteristics of CGA at 20 mg/kg are similar to those of the benzodiazepine diazepam, and it may produce anxiolytic effects by activating benzodiazepine receptors [72]. The combination of CGA and Syringaresinol-di-O-glucoside (SYG) may induce anxiolytic behavior and modulate autonomic nervous system regulation, activate hippocampal BDNF signaling, and attenuate the inhibitory effect on stress-induced peripheral nervous system (PNS) activity [73].
FA restored the 20-carboxy-leukotriene B4 in LPS-induced mice to normal levels, and the ratio of Firmicutes/Bacteroidetes also increased to normal levels. It can also increase the level of 5-hydroxyltryptophan (5-HTP), the precursor of 5-HT, to prevent anxiety [75]. When rats were fed a high-fat diet and exposed to chronic stress, CA modulated the β-catenin/glycogen synthase kinase-3β (GSK-3β) pathway, thereby affecting the function of nerve cells, neurotransmitter levels, neuroplasticity and other related physiological processes, and might have a neuroprotective effect on cognitive changes and anxiety-like behaviors [74] (Table 1).

5.2. Clinical Study on the Effects of Coffee and Its Active Ingredients on Anxiety

A prospective, double-blind, placebo-controlled study of 56 women showed that moderate caffeine intake (low to moderate doses) slightly reduced anxiety in postmenopausal women with underlying overactive bladder (OAB), but had no significant effect on depressive symptoms [86]. In an open-label experimental study by Kimura T et al. [88], 20 patients with frontotemporal lobar degeneration or Lewy body dementia took FA and angelica extract (Feru-guard) 3.0 g daily for 4 weeks. Among them, 19 subjects had a significant decrease in the overall scores of the neuropsychiatric scale and its subscales (including “agitation/aggression”, “anxiety”, etc.), indicating that Feru-guard might have an alleviating effect on anxiety. Quinlan P et al. [87] monitored the effects of caffeine withdrawal on physiological and emotional indicators by having subjects drink hot tea, coffee or water with or without caffeine. The results showed that caffeine can improve mood and reduce anxiety. A RCT of 62 normal adults showed that subjects who consumed caffeine on a daily basis experienced withdrawal symptoms such as depression, anxiety, fatigue and headache after stopping caffeine intake, which might indicate from another perspective that caffeine intake could reduce anxiety reactions [101] (Table 2).
By contrast, a study by Chait LD et al. [102] had smokers take a placebo, different doses of caffeine, or d-amphetamine before smoking and evaluated the effects on their subjective responses. The results showed that caffeine could cause negative subjective reactions, such as nervousness, anxiety and irritability. A double-blind RCT by Nardi AE et al. [103] also showed that patients with PD were more likely to have panic attacks induced by caffeine intake than healthy people, which suggested that caffeine might aggravate anxiety symptoms.

5.3. Epidemiological Study on the Effects of Coffee and Its Active Ingredients on Anxiety

A large prospective cohort study showed that people who drank 2–3 cups of coffee per day had the lowest risk of anxiety. In addition, coffee subtypes including 2–3 cups of ground coffee, milk coffee, and unsweetened coffee were associated with a reduced risk of anxiety [52]. Another cohort study of 180,000 participants aged ≥ 60 years showed that drinking ≥ 1 cup of coffee per day was associated with a lower risk of depression and anxiety disorders [99]. The current results indicate that several proteins (including inflammatory and other proteins) from neural-related tissues (brain, cerebrospinal fluid (CSF) and plasma) interact with coffee and affect anxiety, such as brain protein c-Jun, CSF protein Fas, IL-6, IL-1sRI, and MIP-5. The interaction between CSF protein IL-6 and coffee intake was negatively associated with anxiety, and the interaction between coffee intake and brain protein c-Jun was positively associated with anxiety [104].
However, a cohort study of 941 overweight European adults with subsyndromal depression did not find a relationship between coffee consumption and levels of depression and anxiety [105]. In a recent systematic review, caffeine equivalent to five cups of coffee induced anxiety in both patients with PD and healthy controls [106]. These different conclusions may be attributed to differences in the subjects’ age, gender, physical condition, and different methods of assessing the severity of depression and anxiety (Table 3).

5.4. Bidirectional Regulation of Anxiety by Coffee and Its Active Ingredients Intake and Future Research Directions

Existing animal experiments and clinical studies have shown that coffee and its active ingredients have a bidirectional effect on anxiety: low-dose or short-term intake may relieve anxiety by regulating neurotransmitters and antioxidant pathways, while high-dose or long-term intake may aggravate anxiety symptoms, especially in susceptible populations (such as Parkinson’s disease patients). Epidemiological evidence suggests that moderate coffee consumption (2–3 cups per day) is associated with a reduced risk of anxiety, but the results are influenced by individual differences (metabolic capacity, genotype) and coffee type.
Future studies could further focus on the long-term effects of coffee consumption on anxiety in special populations, such as patients with neurodegenerative diseases. Additionally, the association between specific types of coffee (decaffeinated coffee and milk coffee) and anxiety is also worth exploring.

6. Effects of Coffee and Its Active Ingredients on SDs

6.1. Animal Experimental Study on the Effects of Coffee and Its Active Ingredients on SDs

Habitual caffeine drinkers may have a potential tolerance to caffeine, which may mitigate its deleterious effects on subsequent sleep. Regular caffeine intake will cause the CNS to be continuously exposed to the caffeine environment. When rodents are repeatedly exposed to caffeine, the adenosine system will produce an adaptive response (increased brain adenosine concentration and upregulation of brain adenosine receptors) to cope with caffeine and its metabolites. SDs may be alleviated with repeated caffeine intake [107]. Caffeine disrupts physiological sleep and enhances wakefulness by inhibiting A2AR [76,108]. Caffeine can play a dual protective role by acting on the A2AR of brain vascular endothelial cells: structurally maintaining the integrity of the blood–brain barrier to prevent harmful substances from penetrating into the brain parenchyma; functionally optimizing cerebral blood flow and vascular microenvironment, indirectly supporting the survival and function of neurons [109]. In addition, the neuroprotective effects of caffeine may be related to increased blood flow during sleep, which also helps clear metabolic waste from the brain [77].
Activation of orexin neurons by caffeine through antagonism of adenosine receptors (A1 and A2A) increases arousal levels and sympathetic nerve activity in sleep-deprived mice, while orexin receptor antagonists (OXRs) antagonists block the pro-arousal effects of caffeine, suggesting that the orexin system serves as an important target of caffeine-regulated sleep, providing a potential direction of research for the prevention of caffeine-induced arousal [79]. Both CGA and its metabolite CA can cause a significant increase in sleep latency in rats. The α1-adrenergic receptor (α1-AR) system may contribute to the awakening effect of CA, but neither affects the overall sleep–wake cycle [78] (Table 1).

6.2. Clinical Study on the Effects of Coffee and Its Active Ingredients on SDs

A crossover study conducted on 16 adult men showed that after drinking a beverage containing 300 mg of CGA for 2 consecutive weeks, the CGA group felt less tired when waking up and their sleep quality was significantly improved [89]. In another study, 9 healthy men and women took a drink containing 600 mg of CGA for 5 days. Park et al. found that CGA shortened sleep latency and enhanced parasympathetic nerve activity compared to the control group without adversely affecting sleep quality [90].
A double-blind crossover study in which 12 young people and 12 middle-aged people took 200 mg of caffeine or a placebo showed that caffeine prolonged sleep latency, reduced sleep efficiency and duration, and reduced low-frequency electroencephalogram (EEG) activity and increased high-frequency activity [110]. Marcus GM et al. [111] conducted a randomized case-crossover trial in 100 adults, in which the subjects drank caffeinated coffee or avoided caffeine for 14 days. The results showed that consuming caffeinated coffee significantly reduced the amount of sleep per night (approximately 36 min). Burke TM et al. [91] found that consuming about 200 mg of caffeine 3 h before bedtime can cause a phase delay of about 40 min in the body’s melatonin secretion rhythm, thereby affecting the CR. At the same time, genetic experiments on osteosarcoma cells further confirmed that caffeine could prolong the cycle of circadian clock gene expression in a dose-dependent manner. Ramos-Campo DJ et al. [92] asked 15 middle-and long-distance runners to take 6 mg/kg caffeine or placebo 1 h before an 800-m running test in the evening and evaluated their athletic performance and sleep quality. The results showed that caffeine did not significantly improve running performance, but significantly reduced sleep efficiency, increased the number of awakenings, and impaired subjective sleep quality. However, a study of 20 habitual caffeine consumers who were given either 150 mg of caffeine three times a day or a placebo for 10 consecutive days showed that daily caffeine intake did not significantly affect total sleep time or subjective sleep quality [93] (Table 2).

6.3. Potential Research Directions for Caffeine’s Effects on Sleep

Existing research shows that coffee and its active ingredients have different effects on sleep: caffeine may affect the sleep–wake cycle by regulating adenosine receptors and circadian clock genes, which may prolong the time to fall asleep and reduce sleep efficiency in the short term, but long-term intake may alleviate the negative effects through adaptive mechanisms; CGA shows the potential to improve sleep quality, such as shortening the time to fall asleep and reducing fatigue. Individual responses vary significantly, which may be related to the dose, time of intake and metabolic characteristics.
Future research may further explore (1) the combined effects of the synergistic or antagonistic effects of different components in coffee on sleep; (2) personalized intake recommendations based on individual metabolic differences (genotype, caffeine metabolism rate); and (3) optimizing the coffee intake time window to balance daytime alertness and nighttime sleep quality. In addition, coffee usage strategies for special groups (such as athletes and shift workers) are also worthy of further discussion.

7. Potential Mechanisms of Action of the Main Active Ingredients in Coffee on Depression, Anxiety, and Sleep Disorders

Coffee beverages are complex chemical mixtures extracted from coffee beans. Their bioactive components may produce combined effects in the body through pharmacokinetic interactions (such as absorption, distribution, metabolism and excretion) as well as synergistic or antagonistic pharmacodynamic effects. Therefore, the overall physiological effects of coffee may differ significantly from those observed when studying any of its components in isolation. This study summarizes the potential mechanisms of action of these main active ingredients that have been reported. The effects of coffee beverages on depression, anxiety and sleep disorders may be the result of the interaction of these complex mechanisms (Figure 3).

7.1. Caffeine and Adenosine Receptors

Adenosine is an important nucleoside molecule that plays a key regulatory role in various physiological processes, such as energy metabolism, neurotransmission and immune regulation by binding to four subtypes of adenosine receptors, namely A1, A2A, A2B and A3 [112,113,114]. In the hippocampus, A1R mainly inhibits the release of neurotransmitters before the synapse, reduces synaptic transmission and neuronal excitation, and plays a neuroprotective role [115,116]. It enhances postsynaptic neural plasticity by regulating n-methyl-d-aspartate (NMDA) receptor-dependent LTP. A2A receptor antagonists (such as caffeine) may block A2A receptor-mediated NMDA receptor-dependent LTP and affect synaptic plasticity [117]. A2AR significantly promotes glutamate release when A1R activity is partially inhibited (such as in the presence of N6-Cyclopentyladenosine). It also enhances synaptic transmission by directly regulating glutamate release through the protein kinase C (PKC) pathway, rather than relying on changes in cyclic adenosine monophosphate (cAMP) levels [118]. Stress-induced changes in the adenosine neuromodulatory system are initially an adaptive response to homeostasis, but under chronic stress conditions, persistent changes in the adenosine neuromodulatory system can lead to synaptic dysfunction, which is considered to be the root cause of depressive-like behavior [109,119,120]. Aberrantly increased A2AR signaling in the lateral septal (LS)nucleus is a key upstream regulator of stress-induced depressive-like behavior. Based on this mechanism, A2AR antagonists may become an effective antidepressant [120].
Caffeine’s effects on the brain and CNS are primarily through antagonism of adenosine A1R and A2AR, which blocks the normal inhibitory function of adenosine itself [56,121,122]. Within the safe dosage range, the mechanism of action of caffeine on synaptic function and neural plasticity in the cerebellar hippocampus mainly depend on its antagonistic effect on adenosine receptors. Among them, adenosine A1R mediates the regulation of caffeine on synaptic signal transmission, while adenosine A2AR participates in the caffeine-induced synaptic LTP process through functional regulation [123,124]. Caffeine modulates synaptic function by blocking A2AR, thereby improving mood-related behaviors [56]. The ADORA2A gene is a candidate gene for anxiety, and caffeine may induce more pronounced anxiety responses in people with specific A2AR gene polymorphisms [125]. The rs2298383 single nucleotide polymorphism (SNP) may affect the function of A2A receptor by regulating ADORA2A gene expression. The rs 2298383-TT genotype is associated with a reduced risk of depression, indicating the important role of ADORA2A gene expression in emotion regulation. It is also related to the protective effect of sleep dysfunction [126].

7.2. Monoamine Neurotransmitter Regulation

7.2.1. Effects of Caffeine on DA

Caffeine can stimulate the release of 5-HT in the limbic system and promote the release of DA in the PFC, producing effects similar to those of anti-depression drugs [127,128]. However, the regulatory mechanism of caffeine on DA release is still controversial. Although caffeine may enhance the activity of DA D1 receptors by interfering with the action of adenosine A1R [129], the coexistence of adenosine A2AR in dopaminergic nerve endings may antagonize this effect, and blockade of A2AR will reduce DA release [130,131]. Therefore, the effect of caffeine on DA release is not simply mediated by the A1R, and the final result may depend on the dynamic balance of dual regulation of A1R and A2AR. There are also studies showing that acute exercise-activating doses of caffeine produce a weak antagonistic effect on A2AR, which may not be sufficient to offset caffeine’s antagonistic effect on presynaptic A1R [132]. This may indirectly lead to an increase in DA and glutamate release.

7.2.2. Effects of Coffee and Its Active Ingredients on Monoamine Mechanisms in Depression

Initial evidence supporting the “monoamine hypothesis of depression” was that monoamine oxidase inhibitors and tricyclic antidepressants can improve depressive symptoms by enhancing the activity of 5-HT and NE [133]. The monoamine deficiency hypothesis may not be prevalent in depressed patients, suggesting that other pathways and neurotransmitters are involved in depression [134]. Caffeine has an inhibitory effect on endogenous adenosine, which reduces the release of many neurotransmitters in the CNS, including 5-HT, thereby indirectly protecting the release of 5-HT [135]. One study showed that dietary caffeine administration in rats resulted in increased CNS concentrations of 5-HT, 5-hydroxyindoleacetic acid (a breakdown product of 5-HT),and trypophan (a starting material for the production of 5-HT) [136]. The active ingredients in coffee may also work by regulating monoamine metabolism. Coffee ingredients such as trigonelline, n-methylpyridine, CGA, catechol, and pyrogallol have been shown to increase calcium signaling and DA release, thereby exerting antidepressant effects [49]. Caffeine, FA, and CA inhibit the reuptake of 5-HT, increase the content of synaptic cleft, and affect the occurrence of depression. Chronic caffeine intake (8 mg/kg/d) can also affect the 5-HT level in the hippo-campus [51]. Monoamine oxidase-A (MAO-A), an X-linked mitochondrial enzyme, degrades key neurotransmitters such as 5-HT and DA [137]. CGA in Robusta coffee inhibits MAO-A activity, preventing 5-HT oxidative deamination and thereby elevating synaptic 5-HT levels, this suggests that coffee may be a potential antidepressant [54].

7.2.3. The Effect of Caffeine on Anxiety in the Threat–Fear Circuit

Typically, the neural circuit most closely associated with anxiety disorders is the threat–fear circuit [138]. Caffeine can change the human brain’s response pattern to social threat cues, inducing activation of the threat-related midbrain-periaqueductal gray (PAG) area, which may be linked to the threat-related activity involved in panic attacks and PD [106], and eliminating threat-related activation of the medial prefrontal cortex (mPFC) [139]. Furthermore, the effects of caffeine on the magnitude of threat-related amygdala activation were related to levels of dietary caffeine intake [139]. Deviations in threat prediction may cause the defense system to be over-engaged, leading to anxiety [140].
Caffeine increases threat-related activity in the PAG, which may be linked to the threat-related activity involved in panic attacks and PD [106]. Caffeine modulates hippocampal synaptic plasticity to enhance working and reference memory by blocking adenosine A2AR. A2AR are mainly enriched in the presynaptic terminals of the amygdala, which participate in the formation and consolidation of fear memory by controlling synaptic plasticity, and the downregulation will damage fear memory. Therefore, caffeine may reverse abnormal synaptic plasticity by antagonizing the A2AR, thereby reducing pathological fear memory (such as PTSD) [141].

7.2.4. Mechanisms of Caffeine’s Effects on Sleep and Circadian Rhythm (CR)

Coffee can inhibit adenosine in melatonin secretion, and its secretion level shows specific CR changes. Therefore, caffeine intake will change the CR of melatonin and cause SDs [142]. There is evidence that caffeine intake mainly delays the human melatonin rhythm by antagonizing A1R, increasing cAMP levels, affecting Per1/Per2 transcription through the cAMP-Protein kinases-CREB signaling pathway [91]. Since adenosine receptors are present in the SCN, which acts as a central biological clock and controls the pineal gland through polysynaptic connections, caffeine may cause desynchronization of various circadian systems driven by the SCN. Flavonoids, which are abundant in coffee, have also been shown to inhibit the activity of aryl alkylamine N-acetyltransferase (AANAT) and reduce serum melatonin levels in rats at night [143]. Adenosine acts through the A1R/A2AR signaling pathway, activating the Ca2+-ERK-AP-1 and CREB/CREB-regulated transcription coactivator 1 (CRTC1)-cAMP response element (CRE) pathways, thereby regulating the expression of the clock genes Per1 and Per2. As an adenosine A1R/A2AR antagonist, caffeine can restore the sensitivity of SCN to light through the adenosine receptor pathway and shorten the adjustment time of CR [80]. Adenosine promotes sleep, which inhibits neuronal activity by activating the A1R in the visual cortex, thereby inducing and maintaining non-rapid eye movement sleep (NREM) and rapid eye movement sleep (REM) sleep. As a non-selective adenosine receptor antagonist, caffeine may interfere with this signaling pathway, thereby weakening or completely blocking the sleep aid effect brought by light. Therefore, avoiding caffeine intake may be a key factor in ensuring efficacy in 40 Hz light treatment of SDs based on adenosine mechanism [144].
Caffeine regulates the release and metabolism of monoamine neurotransmitters (DA and 5-HT) by antagonizing adenosine receptors (A1R/A2AR), exerting a complex dual regulatory effect on emotional expression. Therefore, targeted emotion intervention strategies based on receptor subtype specificity (A1R/A2AR) can be further studied in the future. In addition, the impact of individual metabolic differences (genetic polymorphisms) on the neuromodulatory effects of caffeine is also worth exploring.

7.3. Non-Monoamine Neurotransmitter Regulation

7.3.1. Caffeine Regulates Neural Function Through BDNF Signaling Pathway

BDNF is a member of a unique family of neurotrophic growth factors [145]. BDNF plays multiple key roles in brain development and function. It not only promotes the growth, maintenance and protection of neurons, but also participates in the regulation of nerve regeneration and synaptic plasticity. Studies have shown that BDNF plays a key regulatory role in LTP of excitatory synapses in the hippocampus and other brain regions, and plays an important mediating role in the formation of learning and memory [146,147,148,149]. BDNF binds with high affinity to the tropomyosin-related kinase B (TrkB) receptor, thereby activating signaling pathways that regulate many synaptic processes [150]. After BDNF binds to the TrkB receptor, it activates two core signaling pathways, phosphatidy-linositol 3-kinase/mechanistic target of rapamycin (PI3K/mTOR) and mitogenactivated protein kinase (MAPK)/ERK, by phosphorylating Srchomology domain 2 and phospholipase C-γ (PLC-γ) [151,152].
BDNF levels can signal changes in neuronal activity and can also participate in homeostatic plasticity processes, mediating synaptic scaling [153,154,155]. The timing of BDNF release modifies its effects on homeostatic plasticity. Acute administration of BDNF in the nucleus accumbens (NAc) increases the expression of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) subunits, whereas chronic administration leads to downregulation of their expression [154]. TrKB, as a BDNF receptor, not only senses cholesterol through its transmembrane domain and regulates its synaptic effects, but also directly binds to typical and rapid antidepressants, thereby promoting their synaptic localization and BDNF-dependent activation, ultimately mediating neuronal plasticity and antidepressant responses [156].
Caffeine antagonizes presynaptic adenosine A1R and promotes the mobilization of Ca2+ from ryanodine-sensitive intracellular stores. The increase in intracellular Ca2+ concentration may trigger the secretion of BDNF near the synaptic cleft, which in turn activates the TrkB receptor and phosphorylates insulin receptor substrate 2 (IRS2) at the corresponding site, thereby recruiting the PI3K/protein kinases B (Akt/PKB) signaling pathway. Furthermore, caffeine induced an NMDA receptor-independent LTP, termed CAFLTP (caffeine-induced LTP), in the CA1 region of the hippocampus by promoting Ca2+-dependent BDNF secretion. This process is dependent on TrkB-mediated signaling, contributes to CAFLTP expression, and alleviates depressive symptoms [157,158]. Caffeine can reverse the stress-induced decrease in basal phosphorylated calcium calmodulin kinase I (P-CaMKII), total CaMKII and BDNF protein levels in the CA1 region of the hippocampus and restore them to normal levels. Restoration of these signaling molecules may be responsible for the beneficial effects of caffeine on LTP under stressful conditions [159]. BDNF is also believed to be involved in the pathogenesis of anxiety disorders [160]. The role of BDNF in regulating synaptic plasticity may underlie its effects on conditioned fear responses and anxiety regulation [161,162,163]. A2AR deficiency in the hippocampus may impair specific fear conditioning and reduce anxious behavior by interfering with the BDNF-TrkB pathway [163]. Caffeine may be involved in the regulation of BDNF levels in the frontal cortex by antagonizing A2AR, and in the caudate putamen, the effect of caffeine on BDNF expression depends on the individual’s level of anxiety-like behavior, that is, BDNF expression is increased in high anxiety-like behaviors, but not in low anxiety-like behaviors [164].

7.3.2. Caffeine Regulates Neuronal Function via Them Gamma-Aminobutyric Acid (GABA) Pathway

In the human body, GABA mainly acts as a neurotransmitter in the CNS and has the function of inhibiting nerve excitability. In addition, it is also involved in a variety of physiological processes, such as regulating sleep, blood pressure, and anxiety [165]. Cholecystokinin (CCK)-GABA neurons (by injecting AAV-mDlx-DIO-hM4Di-mCherry virus into CCK-Cre mice) can participate in regulating depression-like and anxiety-like behaviors [166]. Traditionally, antidepressant drugs that enhance the function of 5-HT and NE achieve their antidepressant effects by enhancing the function of GABAergic interneurons [167]. The GABAergic output of the anteroventral nucleus of the bed nucleus of the stria terminalis (avBNST) affects the activity of DA neurons in the ventral tegmental area (VTA) and serotonergic neurons in the dorsal raphe nucleus (DRN). Blocking the presynaptic GABAB receptors of the avBNST can inhibit its GABAergic neurons, reduce the inhibitory output to the VTA and DRN, increase the levels of DA and 5-HT in the basal lateral amygdala (BLA), and produce anxiolytic effects [168]. GABAB receptor antagonists are considered effective antidepressants [169]. Caffeine’s enhancing effect on GABA release is mediated by blocking adenosine A1R [170]. Caffeine can temporarily reduce inhibitory postsynaptic currents of GABAergic pathways in hippocampal CA1 pyramidal cells, mainly mediated by the phosphodies-terase pathway. When inhibitory GABAergic signaling is attenuated, it may lead to increased activity of the dopaminergic system [171,172]. Under conditions of caffeine tolerance, the reduction in GABAergic activity can be restored to normal, suggesting that caffeine dose and duration of treatment may play a role in modulating GABAergic signaling [173]. Caffeine-induced protection of GABAergic and Glu neurons mediates the improvement of neurobehavioral responses [170,174].
In summary, caffeine may participate in the regulation of emotional state by regulating the BDNF signaling pathway and GABA pathway. Among them, BDNF’s key role in synaptic plasticity, learning and memory, and emotion regulation makes it an important target for caffeine to affect neural function. While the GABA system is directly related to behavioral manifestations such as anxiety, sleep, and depression. The effects of caffeine on these pathways may be dose-dependent and individual-varying. In the future, further attention can be paid to the specific regulatory patterns of caffeine intake at different doses and durations on the BDNF signaling pathway and the GABA pathway, especially the changes in neural function under stress, anxiety-like behavior and depression.

7.4. Oxidative Stress (OS) and Inflammation

7.4.1. The Mechanism of Action of Coffee and Its Active Ingredients on Emotions Under OS

OS is caused by a disturbance in the normal balance between the production of free radicals (especially reactive oxygen species (ROS)) and antioxidant defenses [175]. Increased reactive oxygen species can modulate hypothalamic–pituitary–adrenal (HPA) axis feedback, inducing its over activation and altering GABA and 5-HT energy transmission [176,177,178]. Activation of the HPA axis in turn promotes the release of glucocorticoids (GCs), which can affect the cell’s redox system through their receptors. Activated glucocorticoid receptors (GRs)can cause increased mitochondrial membrane potential, imbalance of calcium homeostasis, and enhanced mitochondrial oxidation, which further increases the generation of ROS such as superoxide (O2), hydrogen peroxide (H2O2), and hydroxyl radicals (·OH). This cycle ultimately leads to increased OS and may induce cellular oxidative damage [179,180,181]. The hypoxic process disrupts neurohormonal homeostasis in the brain and increases the likelihood of depression by promoting inflammation, apoptosis, 5-HT pathway dysregulation, and mitochondrial OS [181,182]. OS also plays an important role in the pathophysiology of anxiety. The CNS (such as hippocampus, cortex) and peripheral immune cells (such as white blood cells) in anxiety disorders show enhanced OS [183]. Intake of antioxidant supplements such as magnesium, zinc, selenium and Coenzyme Q10 is significantly associated with a decrease in depression and anxiety status, especially with significant effects on improving depression symptoms [184].
Caffeine is a well-proven antioxidant with an activity comparable to glutathione. Caffeine and its metabolites theobromine and xanthine can prevent the production of free radicals such as (·OH), peroxyl radicals (ROO·) and singlet oxygen (1O2), thereby reducing lipid peroxidation in vitro [185,186,187]. After OS caused by hydroxyl and ROO·, the protein oxidation, lipid peroxidation and ROS levels in the synaptic membrane increased significantly. FA can greatly alleviate these oxidative damage and prevent the conformational changes in synaptic membrane proteins caused by free radicals [188]. CA can remove H2O2, superoxide and lipid peroxide, all related to the pathophysiology of depression [185]. CA inhibits ROS-sensitive c-Jun N-terminal kinase 1/2 (JNK 1/2) and p38 MAPK signaling pathways through its antioxidant properties, thereby reducing NF-κB activation and inflammatory response [185,189,190,191].

7.4.2. Mechanisms of Action of Coffee and Its Active Ingredients in Inflammation

Inflammation is considered an important pathological feature of depression and a risk factor for anxiety and SD [192,193,194,195]. Polymorphisms of cytokines and inflammatory mediators (IL-1β, TNF-α, C-C motif chemokine ligand-2 (CCL2) and C-reactive protein (CRP)) are associated with the severity of depression [196]. Proinflammatory cytokines and acute phase proteins are increased in MDD, suggesting that inflammation may be a key disease modifier promoting susceptibility to depression [197]. Peripheral and central inflammatory signals increase the release of proinflammatory cytokines, activate cyclooxygenase (COX-1/COX-2), and increase prostaglandin levels, further aggravating the inflammatory response. In addition, pro-inflammatory cytokines induce enhanced IDO and tryptophan 2,3-dioxygenase (TDO) activity, shifting tryptophan metabolism toward the KP, reducing 5-HT synthesis, and ultimately leading to 5-HT deficiency, exacerbating depression [198]. Inflammatory markers (such as CRP), neutrophils, and white blood cells) may reduce the volume of HPC and PFC regions. Affects CNS signal transduction, changes the levels of neurotransmitters (such as 5-HT, GABA), and aggravates depressive symptoms [199]. Inflammatory responses have been shown to be positively correlated with anxiety arousal, which may influence the overactivation of the amygdala in response to negative stimuli [200]. Under normal conditions, Na+/K+-ATPase (NKA) in microglia forms a complex with the purinergic receptor P2X7R, reducing P2X7R-mediated potass-ium ion (K+) efflux, thereby inhibiting microglia activation, reducing neuroinflammation, and achieving the effect of treating anxiety [201].
Caffeine and its primary metabolite paraxanthine have been demonstrated to inhibit LPS-stimulated TNF-α production in human whole blood. The inhibitory effect is mediated by the cAMP/PKA pathway [202]. CGA can significantly inhibit the release of proinflammatory mediators such as TNF-α in LPS-activated microglia. Its mechanism of action is mainly through blocking the phosphorylation and degradation of inhibitor of nuclear factor kappa-Bα (IκBα), thereby inhibiting the activtion and nuclear translocation of the NF-κB signaling pathway [203]. FA dimer and isoFA can have an anti-inflammatory effect similar to that of non-steroidal anti-inflammatory drugs by inhibiting the gene expression of COX-2 [204,205]. CA (10 μg/mL)can reduce the nitrite concentration in LPS-stimulated Raw 264.7 macrophages to regulate the concentration of inflammation-related substances and exert anti-inflammatory effects. CA showed inhibitory effects on LPS-induced NF-κB activity and the phosphorylation of inflammation-related signaling molecules JNK1/2 and p38 MAPK in the same cell line [189].
In conclusion, coffee and its main active ingredients may play a positive role in regulating mood through antioxidant and anti-inflammatory pathways. Future research can further focus on the specific role of coffee’s antioxidant and anti-inflammatory mechanisms in different human mood disorders, such as its mood regulation effect in chronic low-level inflammatory states.

7.5. Caffeine’s Regulatory Mechanism on the HPA Axis

HPA axis dysregulation is associated with MDD in adults and adolescents [206,207,208,209,210]. Its function is usually assessed by measuring the glucocorticoid cortisol or by the degree of suppression after dexamethasone stimulation [211,212,213]. The etiology of depression is believed to be related to the disorder of the classical stress response, including changes in GRs levels, decreased GRs sensitivity, and impaired negative feedback regulation function [214,215]. The dependence of anxiety-like behaviors on corticotropin releasing hormone (CRH) cells is more related to excitatory glutamatergic projections to a subset of peripheral neurons in the lateral hypothalamic cortex than to endocrine signals [216]. Patients with generalized anxiety, major depression, and other mood disorders often demonstrate HPA axis hyperactivity [217,218]. HPA axis dysregulation in patients with depression may be related to decreased 11β-HSD2 enzyme activity. Impaired function of this enzyme will lead to a decrease in the conversion of cortisol to cortisone, which will increase the level of active cortisol and continuously over activate the GRs, forming a vicious cycle of continued hyperactivity of the HPA axis [210]. Cortisol secretion shows a CR, with the lowest level at night, reaching a peak 30–60 min after waking up, and gradually falling during the day [219]. Poor sleep quality is associated with flatter diurnal cortisol levels, lower wakefulness values, or changes in the cortisol awakening response (CAR) [219,220,221,222].
Caffeine may activate the HPA axis by interacting with centrally located adenosine receptors in the hypothalamic afferent area, ultimately modulating corticotrophin releasing factor (CRF) and HPA axis activity. However, low doses of caffeine do not weaken the peak response of the HPA axis to stress [223]. Caffeine consumption during adolescence alters HPA axis function, leading to increased basal CORT levels during the circadian trough in early adulthood, reduced stress-induced ACTH and CORT release, and reduced adrenal sensitivity to ACTH [224]. Long-term caffeine use may interfere with the negative feedback regulation of the HPA axis by changing the expression of GRs/mineralocorticoid receptors (MRs) in key brain areas, leading to abnormal levels of stress hormones [64]. Regular caffeine intake may induce anxiety symptoms by affecting the functions of brain regions such as the PFC, hippocampus, and amygdala [64,139].
Therefore, future studies may focus on adolescents to explore the long-term effects of caffeine intake on HPA axis function (such as reduced adrenal sensitivity) in adulthood, and further reveal the relationship between this effect and emotion regulation.

7.6. Mechanisms of Coffee and Its Active Ingredients Influencing Depression, Anxiety, and SDs via the Microbiota-Gut–Brain Axis (MGB) Axis

7.6.1. MGB Axis and Depression

The MGB axis connects the intestine to the CNS. Both the MGB axis and intestinal flora disorders are associated with the pathogenesis of a variety of neurological and psychiatric diseases, such as autism spectrum disorder, depression, anxiety, etc. [225,226]. The intestinal flora affects the functions of the immune system, HPA axis and other related systems through the gut–brain axis and the byproducts of metabolism, (such as Short-chain fatty acids (SCFAs)), thereby affecting emotions and behaviors. It also participates in the activation of the HPA axis to trigger immune responses and inflammation, and plays a key role in the development of depression [227]. SCFAs can cross the BBB and have shown anti-depressant and anti-anxiety effects in mouse experiments [228]. The microbial metabolite butyrate has been shown to improve the integrity of the intestinal barrier and BBB by upregulating the expression of hypoxia-inducible factor-1 (HIF-1). CMS combined with probiotic treatment can increase butyrate levels, thereby alleviating depressive behavior [229]. Probiotics can regulate the intestinal barrier function and indirectly stimulate inflammatory immune response, thereby significantly reducing serum cortisol levels and improving depression [230]. MGB axis dysregulation in early life is associated with growth retardation, decreased BDNF, enhanced HPA axis activation, impaired negative feedback of GRs, increased stress reactivity, abnormal brain development, impaired social interaction, anxiety-like manifestations, cognitive deficits and other abnormal behaviors, and may also induce metabolic, immune or psychological disorders inadulth-ood [231]. Rats subjected to chronic unpredictable mild stress (CUMS) and treated with gut microbiota transplantation (FMT) showed increased levels of hippocampal neurochemicals such as 5-HT, GABA, and BDNF, coupled with decreased inflammatory markers, leading to a reduction in depressive symptoms [232].
Coffee-induced changes in gut microbiota composition may translate into changes in effector molecules involved in gut–brain axis signaling, thereby influencing multiple neural and psychological processes [233]. Coffee is closely related to intestinal flora and CNS. It can improve the depressive behavior of rats, increase the levels of SOD and GPx in serum, reduce the levels of IL-6 and TNF-α in serum, and reverse intestinal dysbiosis [53]. Coffee can significantly improve the intestinal flora structure of rats fed a high-fat diet and significantly increase the level of SCFAs [234]. CGA pretreatment improved the depressive-like behaviors and serum biochemical levels (5-HT, DA, IL-6, and TNF-α) of rats with ACTH-induced depression, and also improved the reduction in fecal microbiota diversity in ACTH-treated rats [50].

7.6.2. MGB Axis and Anxiety

Gut microbiota can regulate anxiety by influencing HPC and its related behaviors [192]. Anxiety in mice is accompanied by increased recruitment of hippocampal microglia (Iba1), monocytes (CD11b/CD45), and dendritic cells (CD11b/CD11c), as well as an increase in the number of brain neuron migration, NF-κB activation in the colon and brain, and increased expression of IL-β and TNF-α, leading to increased intestinal barrier permeability, that is, monocyte/macrophage activation triggers intestinal inflammation, and changes in intestinal flora induce anxiety through inflammatory response mechanisms [235]. When mice are exposed to microbial metabolites 4-ethylphenyl sulfate (4EPS), their oligodendrocytes become more immature and their myelin production decreases, which results in a thinning of the insulation around axons and causes anxiety [236].
Caffeine improves neuroinflammation and anxiety during sleep deprivation in rats by inhibiting microglial activation [237]. HPA axis reactivity is part of the bidirectional MGB axis. Caffeine can activate the HPA axis, which in turn stimulates the production of stress catecholamines (NE and AD). These substances are present at the host-microbiome interface such as the intestinal cavity and alveolar fluid, and trigger direct reactions in the microbiome, thereby indirectly affecting related states such as anxiety [238].

7.6.3. Effects of Caffeine on Sleep via Intestinal Flora

CR and feeding behavior jointly influence diurnal variations in community composition and metabolic function in transgenic organisms [239]. The gut and brain communicate through the gut–brain axis, and the gut microbiota may influence areas of the brain that control CR [240]. Both sleep fragmentation and short sleep duration are associated with intestinal dysbiosis, which may be due to activation of the HPA axis. Insufficient sleep can alter the composition of the gut microbiota by increasing hunger and reducing physical activity, immune modulation or HPA axis activation, and subsequent intestinal barrier disruption. The gut microbiome can alter sleep through sleep-inducing LPS and muramyl peptides (MP) translocation, vagal afferent excitation in response to intestinal LPS, regulation of enterochromaffin cells (EC) 5-HT production, and modulation of inflammatory cytokines [241].
Caffeine-induced sleep restriction affects the composition of the gut microbiome and fecal metabolites in a mouse model [242]. Severe sleep deprivation has been shown to lead to ROS accumulation in the gut, and taking antioxidants such as melanoidins found in coffee can prevent this effect [243]. Caffeine can modulate lipid metabolism pathways in the gut microbiota to reduce metabolic disturbances and lipid accumulation induced by sleep restriction [242]. The molecular mechanism of coffee and its active ingredients on depression, anxiety and SD is shown below (Figure 3).
In summary, coffee can relieve depression and anxiety symptoms by changing the composition of intestinal flora and then affecting brain neural activity. Future research can further explore: (1) personalized coffee intake strategies based on individual gut microbiota characteristics. (2) long-term effects of coffee intervention on sleep and mood in special populations (patients with metabolic syndrome). Exploring the effects of coffee processing methods (such as roasting degree) on intestinal flora metabolites can provide a basis for the development of related coffee products.

8. Conclusions and Outlook

This review examined studies on coffee and its bioactive compounds (e.g., caffeine, polyphenols) in relieving depression, anxiety, and SDs, covering animal models, clinical trials, and epidemiological analyses. According to current research results, coffee may affect mood through multiple mechanisms. And coffee has a dual effect on mood: moderate intake can enhance cognitive function and improve mood, but excessive intake may be related to anxiety, insomnia, PTSD, etc.
In addition, the exact mechanisms of coffee’s effects on mental health remain unclear. Future research should focus on the following: (1) There is still a need to further explore the mechanisms by which coffee and its various active ingredients affect brain function and mood. There should be more in-depth research on how coffee and its active ingredients (including metabolites) regulate specific intestinal flora to produce neuroactive substances, thereby exerting a protective effect on brain function. (2) Based on the regulatory effect of caffeine on CR, explore its potential therapeutic strategy for intervening in rhythmic disorders associated with metabolic diseases. (3) Study the effects of different coffee varieties, roasting, and brewing methods on coffee’s bioactive components and their effects on mood, explore the appropriate dosage for drinking coffee, and provide a basis for the development of functional foods containing coffee’s active ingredients.

9. Strengths and Limitations

This study has some strengths as follows: (1) This study explores the association and impact between coffee and its active ingredients and depression, anxiety and SDs, providing valuable guidance for subsequent research. (2) This study systematically reviews the research results on coffee and its ingredients in depression, anxiety and SDs, covering animal experiments, clinical trials and epidemiological studies, and the content is detailed and comprehensive. (3) The description of the mechanism of action of coffee and its components is relatively detailed, including adenosine receptor regulation, regulation of neurotransmitters, antioxidant and anti-inflammatory effects, and so on.
However, there are some limitations: (1) The article discusses the mechanism of action of coffee, but there is still a lack of clear recommendations on the practical application of coffee, such as recommended intake or suitability for specific populations. (2) The article cited a large number of studies, but lacked in-depth evaluation of the limitations and reliability of the methodologies of these studies, such as the sample size of the experimental design and whether the control group setting was adequate, resulting in insufficient depth of critical analysis in the study.

Author Contributions

Conceptualization, J.C. and J.Z.; methodology, Z.S. and J.L.; validation, J.C., J.Z. and Y.Z.; formal analysis, Z.S. and J.L.; investigation, Z.S. and J.L.; resources, J.C. and J.Z.; data curation, G.W., C.M. and W.Z.; writing—original draft preparation, Z.S. and J.L.; writing—review and editing, J.C. and J.Z.; visualization, Z.S., J.L. and L.C.; supervision, J.C., J.Z. and C.X.; project administration, C.X. and T.H.; funding acquisition, J.C. and J.Z. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by grants from National major science and technology project for young scientists (2024ZD0529205), College Student Innovation and Entrepreneurship Project (S202510488171S, S202410488180X), Occupational Hazard Identification and control Key laboratory of Hubei Province open fund (OHIC2024G07, OHIC2024Z04).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

We thank the School of Public Health of Wuhan University of Science and Technology and Hubei Provincial Center for Disease Control and Prevention for their assistance in carrying out this study, especially Zijun Shi, Jin Luan, Yating Zhang, Jianbo Zhan, Jing Chen, and some counselors. We also thank Scidraw (https://scidraw.io/), NIH BioArt Source (https://bioart.niaid.nih.gov/), and Bioicons (https://bioicons.com/) for providing image materials for this study.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
SDsSleep disorders
MDDMajor depressive disorder
TCAs Tricyclic drugs
5-HT Serotonin
SSRIsSelective Serotonin Reuptake Inhibitor(s)
NENorepinephrine
SNRISerotonin–norepinephrine reuptake inhibitors
DXT Duloxetine
DVSDesvenlafaxine succinate
GAD Generalized Anxiety Disorder
SAD Social Anxiety Disorder
PTSD Post-traumatic stress disorder
OCD Obsessive–compulsive disorder
PSD Post-stroke depression
CGA Chlorogenic acid
FA Ferulic acid
CA Caffeic acid
DA Dopamine
PFC Prefrontal Cortex
A1RA1 receptors
A2ARA2A receptors
ACTHadrenocorticotropic hormone
DMNDefault mode network
MAO-AMonoamine oxidase-A
DSM-IVDiagnostic and Statistical Manual of Mental Disorders-IV
SPT Sugar preference test
FST Forced swim test
TST Tail suspension test
OFT Open field test
IL-6Interleukin-6
TNF-αTumor necrosis factor-α
SOD Superoxide dismutase
GPxGlutathione peroxidase
PDDPostpartum Depression
IDOIndoleamine 2, 3-dioxygenase
LPS Lipopolysaccharide
UAUric acid
3-HK3-Hydroxykynurenine
KYN Kynurenine
KPKynurenine pathway
CNS Central Nervous System
CUSChronic unpredictable stress
LTPLong-term potentiation
CORTCorticosterone
MAPKMitogen-activated protein kinase
ERKExtracellular regulated protein kinases
NF-κBNuclear Factor kappa-light-chain-enhancer of activated B cells
CWIRSChronic water immersion restraint stress
CMSChronic Mild Stress
PGC-1αPeroxisome proliferators-activated receptor γ coactivator lα
KATKynurenine aminotransferase
KYNAKynurenic acid
SRSStress-induced stress
PGE2Prostaglandin E2
BrdUBromodeoxyuridine
DCXDoublecortin
IL-1βInterleukin-1β
SNAP-25Synaptosomal-Associated Protein 25
IFN-γInterferon-gamma
MDA Malondialdehyde
BDNFBrain-derived neurotrophic factor
GSHGlutathione
GRGlucocorticoid receptor
ADRA1Aα1Aadrenoreceptor
TBARSThiobarbituric acid reactive substances
AChEAcetylcholinesterase
OSOxidative stress
Nrf2 Nuclear factor-erythroid 2 related factor 2
EPM Elevated plus maze test
LDB Light/dark box test
IATInhibitory avoidance test
FET Free exploration test
PNSPeripheral nervous system
MWMMorris water maze
MBTMarble burying test
5-HTP5-Hydroxytryptophan
REM Rapid eye movement sleep
NREM Non-rapid eye movement sleep
Per1/Per2Period Circadian Regulator 1/2
α1-ARα1-adrenergic receptor
iTBS Intermittent burst theta stimulation
dmPFCDorsomedial prefrontal cortex
SBPSystolic Blood Pressure
DBPDiastolic Blood Pressure
CRCircadian Rhythm
EEG Electroencephalogram
ICCAIsochlorogenic acid
SYGSyringaresinol-di-O-glucoside
GSK-3βGlycogen synthase kinase-3β
OAB Overactive bladder
CSF Cerebrospinal fluid
SCNSuprachiasmatic nucleus
CREBCyclic-AMP response binding protein
OXRsOrexin receptor antagonists
NMDAN-methyl-d-aspartate
PKCProtein kinase C
cAMPCyclic adenosine monophosphate
LSLateral septal
SNPSingle nucleotide polymorphism
PAG Periaqueductal gray
mPFCMedial prefrontal cortex
AANATAryl alkylamine N-acetyltransferase
CRTC1CREB-regulated transcription coactivator 1
CREcAMP response element
PI3KPhosphatidylinositol 3-kinase
mTORMechanistic target of rapamycin
TrkBTropomyosin-related kinase B
PLC-γPhospholipase C-γ
AMPARα-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor
NAcNucleus accumbens
IRS2Insulin receptor substrate 2
CAFLTPCaffeine-induced LTP
P-CaMKIIPhosphorylated calcium calmodulin kinase I
GABAGamma-aminobutyric acid
CCKCholecystokinin
AvBNSTAnteroventral nucleus of the bed nucleus of the stria terminalis
VTAVentral tegmental area
DRNDorsal raphe nucleus
BLABasal lateral amygdala
ROSReactive oxygen species
HPA axishypothalamic–pituitary–adrenal axis
GCsGlucocorticoids
H2O2Hydrogen peroxide
·OHHydroxyl radicals
ROO·Peroxyl radicals
1O2Singlet oxygen
JNK1/2c-Jun N-terminal kinase 1/2
CCL2C-C motif chemokine ligand-2
CRPC-reactive protein
TDOTryptophan 2,3-dioxygenase
NKANa+/K+-ATPase
IκBαInhibitor of nuclear factor kappa-Bα
CRHCorticotropin releasing hormone
CARCortisol awakening response
CRFCorticotrophin releasing factor
MRsMineralocorticoid receptors
MGB axisMicrobiota-Gut–Brain axis
SCFAs Short-chain fatty acids
HIF-1Hypoxia-inducible factor-1
CUMSChronic unpredictable mild stress
FMTFecal microbiota transplantation
4EPS4-Ethylphenyl sulfate
MPMuramyl peptides
ECEnterochromaffin cells

References

  1. Krystal, A.D. Psychiatric disorders and sleep. Neurol. Clin. 2012, 30, 1389–1413. [Google Scholar] [CrossRef]
  2. Borges, C.; Ellis, J.G.; Ruivo Marques, D. The Role of Sleep Effort as a Mediator Between Anxiety and Depression. Psychol. Rep. 2024, 127, 2287–2306. [Google Scholar] [CrossRef]
  3. Cui, R. Editorial: A Systematic Review of Depression. Curr. Neuropharmacol. 2015, 13, 480. [Google Scholar] [CrossRef] [PubMed]
  4. Chellappa, S.L.; Aeschbach, D. Sleep and anxiety: From mechanisms to interventions. Sleep Med. Rev. 2022, 61, 101583. [Google Scholar] [CrossRef]
  5. Santomauro, D.F.; Herrera, A.M.M.; Shadid, J.; Zheng, P.; Ashbaugh, C.; Pigott, D.M.; Abbafati, C.; Adolph, C.; Amlag, J.O.; Aravkin, A.Y.; et al. Global prevalence and burden of depressive and anxiety disorders in 204 countries and territories in 2020 due to the COVID-19 pandemic. Lancet 2021, 398, 1700–1712. [Google Scholar] [CrossRef]
  6. Pilcher, J.J.; Huffcutt, A.I. Effects of sleep deprivation on performance: A meta-analysis. Sleep 1996, 19, 318–326. [Google Scholar] [CrossRef]
  7. Harrison, Y.; Horne, J.A. The impact of sleep deprivation on decision making: A review. J. Exp. Psychol. Appl. 2000, 6, 236–249. [Google Scholar] [CrossRef]
  8. Pollmächer, T.; Wetter, T.C. Schlafstörungen und Psychische Erkrankungen: Eine Einführung für Ärzte und Psychologen; Kohlhammer: Stuttgart, Germany, 2017. [Google Scholar]
  9. Hillhouse, T.M.; Porter, J.H. A brief history of the development of antidepressant drugs: From monoamines to glutamate. Exp. Clin. Psychopharmacol. 2015, 23, 1–21. [Google Scholar] [CrossRef] [PubMed]
  10. Cleare, A.; Pariante, C.M.; Young, A.H.; Anderson, I.M.; Christmas, D.; Cowen, P.J.; Dickens, C.; Ferrier, I.N.; Geddes, J.; Gilbody, S.; et al. Evidence-based guidelines for treating depressive disorders with antidepressants: A revision of the 2008 British Association for Psychopharmacology guidelines. J. Psychopharmacol. 2015, 29, 459–525. [Google Scholar] [CrossRef] [PubMed]
  11. Arakawa, R.; Stenkrona, P.; Takano, A.; Svensson, J.; Andersson, M.; Nag, S.; Asami, Y.; Hirano, Y.; Halldin, C.; Lundberg, J. Venlafaxine ER Blocks the Norepinephrine Transporter in the Brain of Patients with Major Depressive Disorder: A PET Study Using [18F]FMeNER-D2. Int. J. Neuropsychopharmacol. 2019, 22, 278–285. [Google Scholar] [CrossRef]
  12. Morin, C.M.; Vallières, A.; Guay, B.; Ivers, H.; Savard, J.; Mérette, C.; Bastien, C.; Baillargeon, L. Cognitive behavioral therapy, singly and combined with medication, for persistent insomnia: A randomized controlled trial. Jama 2009, 301, 2005–2015. [Google Scholar] [CrossRef] [PubMed]
  13. Katzman, M.A.; Bleau, P.; Blier, P.; Chokka, P.; Kjernisted, K.; Van Ameringen, M.; Antony, M.M.; Bouchard, S.; Brunet, A.; Flament, M.; et al. Canadian clinical practice guidelines for the management of anxiety, posttraumatic stress and obsessive-compulsive disorders. BMC Psychiatry 2014, 14 (Suppl. 1), S1. [Google Scholar] [CrossRef]
  14. Carmi, L.; Tendler, A.; Bystritsky, A.; Hollander, E.; Blumberger, D.M.; Daskalakis, J.; Ward, H.; Lapidus, K.; Goodman, W.; Casuto, L.; et al. Efficacy and Safety of Deep Transcranial Magnetic Stimulation for Obsessive-Compulsive Disorder: A Prospective Multicenter Randomized Double-Blind Placebo-Controlled Trial. Am. J. Psychiatry 2019, 176, 931–938. [Google Scholar] [CrossRef]
  15. De Crescenzo, F.; D’Alò, G.L.; Ostinelli, E.G.; Ciabattini, M.; Di Franco, V.; Watanabe, N.; Kurtulmus, A.; Tomlinson, A.; Mitrova, Z.; Foti, F.; et al. Comparative effects of pharmacological interventions for the acute and long-term management of insomnia disorder in adults: A systematic review and network meta-analysis. Lancet 2022, 400, 170–184. [Google Scholar] [CrossRef]
  16. Rothmore, J. Antidepressant-induced sexual dysfunction. Med. J. Aust. 2020, 212, 329–334. [Google Scholar] [CrossRef]
  17. Zeng, L.F.; Cao, Y.; Wang, L.; Dai, Y.K.; Hu, L.; Wang, Q.; Zhu, L.T.; Bao, W.H.; Zou, Y.P.; Chen, Y.B.; et al. Role of Medicinal Plants for Liver-Qi Regulation Adjuvant Therapy in Post-stroke Depression: A Systematic Review of Literature. Phytother. Res. PTR 2017, 31, 40–52. [Google Scholar] [CrossRef]
  18. Dai, W.; Feng, K.; Sun, X.; Xu, L.; Wu, S.; Rahmand, K.; Jia, D.; Han, T. Natural products for the treatment of stress-induced depression: Pharmacology, mechanism and traditional use. J. Ethnopharmacol. 2022, 285, 114692. [Google Scholar] [CrossRef]
  19. Ernst, E. Herbal remedies for anxiety—A systematic review of controlled clinical trials. Phytomedicine Int. J. Phytother. Phytopharm. 2006, 13, 205–208. [Google Scholar] [CrossRef] [PubMed]
  20. Shi, M.M.; Piao, J.H.; Xu, X.L.; Zhu, L.; Yang, L.; Lin, F.L.; Chen, J.; Jiang, J.G. Chinese medicines with sedative-hypnotic effects and their active components. Sleep Med. Rev. 2016, 29, 108–118. [Google Scholar] [CrossRef]
  21. Hu, G.L.; Wang, X.; Zhang, L.; Qiu, M.H. The sources and mechanisms of bioactive ingredients in coffee. Food Funct. 2019, 10, 3113–3126. [Google Scholar] [CrossRef] [PubMed]
  22. Frary, C.D.; Johnson, R.K.; Wang, M.Q. Food sources and intakes of caffeine in the diets of persons in the United States. J. Am. Diet. Assoc. 2005, 105, 110–113. [Google Scholar] [CrossRef] [PubMed]
  23. Liu, D.; Li, Z.H.; Shen, D.; Zhang, P.D.; Song, W.Q.; Zhang, W.T.; Huang, Q.M.; Chen, P.L.; Zhang, X.R.; Mao, C. Association of Sugar-Sweetened, Artificially Sweetened, and Unsweetened Coffee Consumption With All-Cause and Cause-Specific Mortality: A Large Prospective Cohort Study. Ann. Intern. Med. 2022, 175, 909–917. [Google Scholar] [CrossRef]
  24. O’Keefe, J.H.; Bhatti, S.K.; Patil, H.R.; DiNicolantonio, J.J.; Lucan, S.C.; Lavie, C.J. Effects of habitual coffee consumption on cardiometabolic disease, cardiovascular health, and all-cause mortality. J. Am. Coll. Cardiol. 2013, 62, 1043–1051. [Google Scholar] [CrossRef]
  25. Muley, A.; Muley, P.; Shah, M. Coffee to reduce risk of type 2 diabetes?: A systematic review. Curr. Diabetes Rev. 2012, 8, 162–168. [Google Scholar] [CrossRef]
  26. van Dam, R.M.; Hu, F.B. Coffee consumption and risk of type 2 diabetes: A systematic review. Jama 2005, 294, 97–104. [Google Scholar] [CrossRef]
  27. Iwamoto, H.; Izumi, K.; Natsagdorj, A.; Naito, R.; Makino, T.; Kadomoto, S.; Hiratsuka, K.; Shigehara, K.; Kadono, Y.; Narimoto, K.; et al. Coffee diterpenes kahweol acetate and cafestol synergistically inhibit the proliferation and migration of prostate cancer cells. Prostate 2019, 79, 468–479. [Google Scholar] [CrossRef]
  28. Barré, T.; Fontaine, H.; Ramier, C.; Di Beo, V.; Pol, S.; Carrieri, P.; Marcellin, F.; Cagnot, C.; Dorival, C.; Zucman-Rossi, J.; et al. Elevated coffee consumption is associated with a lower risk of elevated liver fibrosis biomarkers in patients treated for chronic hepatitis B (ANRS CO22 Hepather cohort). Clin. Nutr. 2022, 41, 610–619. [Google Scholar] [CrossRef]
  29. Schmit, S.L.; Rennert, H.S.; Rennert, G.; Gruber, S.B. Coffee Consumption and the Risk of Colorectal Cancer. Cancer Epidemiol. Biomark. Prev. 2016, 25, 634–639. [Google Scholar] [CrossRef] [PubMed]
  30. Nehlig, A. Effects of coffee/caffeine on brain health and disease: What should I tell my patients? Pract. Neurol. 2016, 16, 89–95. [Google Scholar] [CrossRef] [PubMed]
  31. Fredholm, B.B.; Bättig, K.; Holmén, J.; Nehlig, A.; Zvartau, E.E. Actions of caffeine in the brain with special reference to factors that contribute to its widespread use. Pharmacol. Rev. 1999, 51, 83–133. [Google Scholar] [CrossRef]
  32. Yun, H.; Su, W.; You, T.; Wang, J.; Ying, Y.; Wang, C.; Ren, Y.; Lu, B.; Li, Y.; Liu, C. Boosting physical performance in SD rats through brain-targeted delivery of caffeine-loaded transferrin liposomes. Heliyon 2024, 10, e34617. [Google Scholar] [CrossRef]
  33. Picó-Pérez, M.; Magalhães, R.; Esteves, M.; Vieira, R.; Castanho, T.C.; Amorim, L.; Sousa, M.; Coelho, A.; Moreira, P.S.; Cunha, R.A.; et al. Coffee consumption decreases the connectivity of the posterior Default Mode Network (DMN) at rest. Front. Behav. Neurosci. 2023, 17, 1176382. [Google Scholar] [CrossRef]
  34. Magalhães, R.; Picó-Pérez, M.; Esteves, M.; Vieira, R.; Castanho, T.C.; Amorim, L.; Sousa, M.; Coelho, A.; Fernandes, H.M.; Cabral, J.; et al. Habitual coffee drinkers display a distinct pattern of brain functional connectivity. Mol. Psychiatry 2021, 26, 6589–6598. [Google Scholar] [CrossRef]
  35. Benca, R.M.; Okawa, M.; Uchiyama, M.; Ozaki, S.; Nakajima, T.; Shibui, K.; Obermeyer, W.H. Sleep and mood disorders. Sleep Med. Rev. 1997, 1, 45–56. [Google Scholar] [CrossRef]
  36. Neckelmann, D.; Mykletun, A.; Dahl, A.A. Chronic insomnia as a risk factor for developing anxiety and depression. Sleep 2007, 30, 873–880. [Google Scholar] [CrossRef]
  37. Zhai, P.; Liu, H.; Zhang, Y.; Huang, T.; Xiong, C.; Liu, Y.; Wang, G.; Chen, X.; Tan, J.; Jiao, C.; et al. Correlation analysis between sleep quality and the mental health status of female sex workers during the COVID-19 pandemic in Hubei Province. Front. Endocrinol. 2023, 14, 1193266. [Google Scholar] [CrossRef] [PubMed]
  38. Zhang, Y.T.; Huang, T.; Zhou, F.; Huang, A.D.; Ji, X.Q.; He, L.; Geng, Q.; Wang, J.; Mei, C.; Xu, Y.J.; et al. Correlation between Anxiety, Depression, and Sleep Quality in College Students. Biomed. Environ. Sci. BES 2022, 35, 648–651. [Google Scholar] [CrossRef] [PubMed]
  39. Luo, L.; Zhang, Y.; Huang, T.; Zhou, F.; Xiong, C.; Liu, Y.; Zhai, P.; Wang, G.; Tan, J.; Jiao, C.; et al. A description of the current status of chronic fatigue syndrome and associated factors among university students in Wuhan, China. Front. Psychiatry 2022, 13, 1047014. [Google Scholar] [CrossRef]
  40. Fava, M.; Rush, A.J.; Alpert, J.E.; Balasubramani, G.K.; Wisniewski, S.R.; Carmin, C.N.; Biggs, M.M.; Zisook, S.; Leuchter, A.; Howland, R.; et al. Difference in treatment outcome in outpatients with anxious versus nonanxious depression: A STAR*D report. Am. J. Psychiatry 2008, 165, 342–351. [Google Scholar] [CrossRef]
  41. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders; American Psychiatric Association: Washington, DC, USA, 2000. [Google Scholar]
  42. Breslau, N.; Roth, T.; Rosenthal, L.; Andreski, P. Sleep disturbance and psychiatric disorders: A longitudinal epidemiological study of young adults. Biol. Psychiatry 1996, 39, 411–418. [Google Scholar] [CrossRef]
  43. Franzen, P.L.; Buysse, D.J. Sleep disturbances and depression: Risk relationships for subsequent depression and therapeutic implications. Dialogues Clin. Neurosci. 2008, 10, 473–481. [Google Scholar] [CrossRef]
  44. Oakes, D.J.; Pearce, H.A.; Roberts, C.; Gehrman, P.G.; Lewis, C.; Jones, I.; Lewis, K.J.S. Associations between comorbid anxiety and sleep disturbance in people with bipolar disorder: Findings from actigraphy and subjective sleep measures. J. Affect. Disord. 2022, 309, 165–171. [Google Scholar] [CrossRef]
  45. Liu, M.Y.; Yin, C.Y.; Zhu, L.J.; Zhu, X.H.; Xu, C.; Luo, C.X.; Chen, H.; Zhu, D.Y.; Zhou, Q.G. Sucrose preference test for measurement of stress-induced anhedonia in mice. Nat. Protoc. 2018, 13, 1686–1698. [Google Scholar] [CrossRef] [PubMed]
  46. Porsolt, R.D.; Bertin, A.; Jalfre, M. Behavioral despair in mice: A primary screening test for antidepressants. Arch. Int. Pharmacodyn. Ther. 1977, 229, 327–336. [Google Scholar]
  47. Steru, L.; Chermat, R.; Thierry, B.; Simon, P. The tail suspension test: A new method for screening antidepressants in mice. Psychopharmacology 1985, 85, 367–370. [Google Scholar] [CrossRef]
  48. Katz, R.J.; Roth, K.A.; Carroll, B.J. Acute and chronic stress effects on open field activity in the rat: Implications for a model of depression. Neurosci. Biobehav. Rev. 1981, 5, 247–251. [Google Scholar] [CrossRef] [PubMed]
  49. Grosso, G.; Micek, A.; Castellano, S.; Pajak, A.; Galvano, F. Coffee, tea, caffeine and risk of depression: A systematic review and dose-response meta-analysis of observational studies. Mol. Nutr. Food Res. 2016, 60, 223–234. [Google Scholar] [CrossRef]
  50. Song, J.; Zhou, N.; Ma, W.; Gu, X.; Chen, B.; Zeng, Y.; Yang, L.; Zhou, M. Modulation of gut microbiota by chlorogenic acid pretreatment on rats with adrenocorticotropic hormone induced depression-like behavior. Food Funct. 2019, 10, 2947–2957. [Google Scholar] [CrossRef]
  51. Pechlivanova, D.M.; Tchekalarova, J.D.; Alova, L.H.; Petkov, V.V.; Nikolov, R.P.; Yakimova, K.S. Effect of long-term caffeine administration on depressive-like behavior in rats exposed to chronic unpredictable stress. Behav. Pharmacol. 2012, 23, 339–347. [Google Scholar] [CrossRef]
  52. Min, J.; Cao, Z.; Cui, L.; Li, F.; Lu, Z.; Hou, Y.; Yang, H.; Wang, X.; Xu, C. The association between coffee consumption and risk of incident depression and anxiety: Exploring the benefits of moderate intake. Psychiatry Res. 2023, 326, 115307. [Google Scholar] [CrossRef]
  53. Gu, X.; Zhang, S.; Ma, W.; Wang, Q.; Li, Y.; Xia, C.; Xu, Y.; Zhang, T.; Yang, L.; Zhou, M. The Impact of Instant Coffee and Decaffeinated Coffee on the Gut Microbiota and Depression-Like Behaviors of Sleep-Deprived Rats. Front. Microbiol. 2022, 13, 778512. [Google Scholar] [CrossRef]
  54. Grzelczyk, J.; Budryn, G.; Peña-García, J.; Szwajgier, D.; Gałązka-Czarnecka, I.; Oracz, J.; Pérez-Sánchez, H. Evaluation of the inhibition of monoamine oxidase A by bioactive coffee compounds protecting serotonin degradation. Food Chem. 2021, 348, 129108. [Google Scholar] [CrossRef]
  55. Hall, S.; Arora, D.; Anoopkumar-Dukie, S.; Grant, G.D. Effect of Coffee in Lipopolysaccharide-Induced Indoleamine 2,3-Dioxygenase Activation and Depressive-like Behavior in Mice. J. Agric. Food Chem. 2016, 64, 8745–8754. [Google Scholar] [CrossRef]
  56. Kaster, M.P.; Machado, N.J.; Silva, H.B.; Nunes, A.; Ardais, A.P.; Santana, M.; Baqi, Y.; Müller, C.E.; Rodrigues, A.L.; Porciúncula, L.O.; et al. Caffeine acts through neuronal adenosine A2A receptors to prevent mood and memory dysfunction triggered by chronic stress. Proc. Natl. Acad. Sci. USA 2015, 112, 7833–7838. [Google Scholar] [CrossRef]
  57. Mao, Z.F.; Ouyang, S.H.; Zhang, Q.Y.; Wu, Y.P.; Wang, G.E.; Tu, L.F.; Luo, Z.; Li, W.X.; Kurihara, H.; Li, Y.F.; et al. New insights into the effects of caffeine on adult hippocampal neurogenesis in stressed mice: Inhibition of CORT-induced microglia activation. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 2020, 34, 10998–11014. [Google Scholar] [CrossRef]
  58. Fang, C.; Hayashi, S.; Du, X.; Cai, X.; Deng, B.; Zheng, H.; Ishido, S.; Tsutsui, H.; Sheng, J. Caffeine protects against stress-induced murine depression through activation of PPARγC1α-mediated restoration of the kynurenine pathway in the skeletal muscle. Sci. Rep. 2021, 11, 7287. [Google Scholar] [CrossRef]
  59. Turgeon, S.M.; Townsend, S.E.; Dixon, R.S.; Hickman, E.T.; Lee, S.M. Chronic caffeine produces sexually dimorphic effects on amphetamine-induced behavior, anxiety and depressive-like behavior in adolescent rats. Pharmacol. Biochem. Behav. 2016, 143, 26–33. [Google Scholar] [CrossRef] [PubMed]
  60. Khan, I.; Ullah, S.; Ullah, S.; Ali, N.; Huma, Z.; Yaşar, S.; Khan, S.; Haq, R.U.; Khan, A.; Khan, I. Antidepressant effects of SY-2476: A caffeine derivative’s role in A1/A2(A) gene expression modulation in corticosterone-induced depressed rats. Neurosci. Lett. 2025, 845, 138059. [Google Scholar] [CrossRef] [PubMed]
  61. Basu Mallik, S.; Mudgal, J.; Hall, S.; Kinra, M.; Grant, G.D.; Nampoothiri, M.; Anoopkumar-Dukie, S.; Arora, D. Remedial effects of caffeine against depressive-like behaviour in mice by modulation of neuroinflammation and BDNF. Nutr. Neurosci. 2022, 25, 1836–1844. [Google Scholar] [CrossRef] [PubMed]
  62. Machado, N.J.; Simões, A.P.; Silva, H.B.; Ardais, A.P.; Kaster, M.P.; Garção, P.; Rodrigues, D.I.; Pochmann, D.; Santos, A.I.; Araújo, I.M.; et al. Caffeine Reverts Memory But Not Mood Impairment in a Depression-Prone Mouse Strain with Up-Regulated Adenosine A(2A) Receptor in Hippocampal Glutamate Synapses. Mol. Neurobiol. 2017, 54, 1552–1563. [Google Scholar] [CrossRef]
  63. Clark, L.; Chamberlain, S.R.; Sahakian, B.J. Neurocognitive mechanisms in depression: Implications for treatment. Annu. Rev. Neurosci. 2009, 32, 57–74. [Google Scholar] [CrossRef]
  64. Prajapati, S.K.; Dangi, D.S.; Krishnamurthy, S. Repeated caffeine administration aggravates post-traumatic stress disorder-like symptoms in rats. Physiol. Behav. 2019, 211, 112666. [Google Scholar] [CrossRef] [PubMed]
  65. Machado, N.J.; Ardais, A.P.; Nunes, A.; Szabó, E.C.; Silveirinha, V.; Silva, H.B.; Kaster, M.P.; Cunha, R.A. Impact of Coffee Intake on Measures of Wellbeing in Mice. Nutrients 2024, 16, 2920. [Google Scholar] [CrossRef]
  66. Zeni, A.L.; Zomkowski, A.D.; Maraschin, M.; Rodrigues, A.L.; Tasca, C.I. Ferulic acid exerts antidepressant-like effect in the tail suspension test in mice: Evidence for the involvement of the serotonergic system. Eur. J. Pharmacol. 2012, 679, 68–74. [Google Scholar] [CrossRef]
  67. Takeda, H.; Tsuji, M.; Miyamoto, J.; Masuya, J.; Iimori, M.; Matsumiya, T. Caffeic acid produces antidepressive- and/or anxiolytic-like effects through indirect modulation of the alpha 1A-adrenoceptor system in mice. Neuroreport 2003, 14, 1067–1070. [Google Scholar] [CrossRef]
  68. Kasimay Cakir, O.; Ellek, N.; Salehin, N.; Hamamcı, R.; Keleş, H.; Kayalı, D.G.; Akakın, D.; Yüksel, M.; Özbeyli, D. Protective effect of low dose caffeine on psychological stress and cognitive function. Physiol. Behav. 2017, 168, 1–10. [Google Scholar] [CrossRef] [PubMed]
  69. Güvel, M.C.; Aykan, U.; Paykal, G.; Uluoğlu, C. Chronic administration of caffeine, modafinil, AVL-3288 and CX516 induces time-dependent complex effects on cognition and mood in an animal model of sleep deprivation. Pharmacol. Biochem. Behav. 2024, 241, 173793. [Google Scholar] [CrossRef] [PubMed]
  70. Stefanello, N.; Schmatz, R.; Pereira, L.B.; Rubin, M.A.; da Rocha, J.B.; Facco, G.; Pereira, M.E.; Mazzanti, C.M.; Passamonti, S.; Rodrigues, M.V.; et al. Effects of chlorogenic acid, caffeine, and coffee on behavioral and biochemical parameters of diabetic rats. Mol. Cell. Biochem. 2014, 388, 277–286. [Google Scholar] [CrossRef]
  71. Guo, J.T.; Li, H.Y.; Cheng, C.; Shi, J.X.; Ruan, H.N.; Li, J.; Liu, C.M. Isochlorogenic acid A ameliorated lead-induced anxiety-like behaviors in mice by inhibiting ferroptosis-mediated neuroinflammation via the BDNF/Nrf2/GPX4 pathways. Food Chem. Toxicol. Int. J. Publ. Br. Ind. Biol. Res. Assoc. 2024, 190, 114814. [Google Scholar] [CrossRef]
  72. Bouayed, J.; Rammal, H.; Dicko, A.; Younos, C.; Soulimani, R. Chlorogenic acid, a polyphenol from Prunus domestica (Mirabelle), with coupled anxiolytic and antioxidant effects. J. Neurol. Sci. 2007, 262, 77–84. [Google Scholar] [CrossRef]
  73. Miyazaki, S.; Fujita, Y.; Oikawa, H.; Takekoshi, H.; Soya, H.; Ogata, M.; Fujikawa, T. Combination of syringaresinol-di-O-β-D-glucoside and chlorogenic acid shows behavioral pharmacological anxiolytic activity and activation of hippocampal BDNF-TrkB signaling. Sci. Rep. 2020, 10, 18177. [Google Scholar] [CrossRef]
  74. El-Sayed, N.S.; Khalil, N.A.; Saleh, S.R.; Aly, R.G.; Basta, M. The Possible Neuroprotective Effect of Caffeic Acid on Cognitive Changes and Anxiety-Like Behavior Occurring in Young Rats Fed on High-Fat Diet and Exposed to Chronic Stress: Role of β-Catenin/GSK-3B Pathway. J. Mol. Neurosci. 2024, 74, 61. [Google Scholar] [CrossRef]
  75. Deng, L.; Zhou, X.; Tao, G.; Hao, W.; Wang, L.; Lan, Z.; Song, Y.; Wu, M.; Huang, J.Q. Ferulic acid and feruloylated oligosaccharides alleviate anxiety and depression symptom via regulating gut microbiome and microbial metabolism. Food Res. Int. 2022, 162, 111887. [Google Scholar] [CrossRef]
  76. Huang, Z.L.; Qu, W.M.; Eguchi, N.; Chen, J.F.; Schwarzschild, M.A.; Fredholm, B.B.; Urade, Y.; Hayaishi, O. Adenosine A2A, but not A1, receptors mediate the arousal effect of caffeine. Nat. Neurosci. 2005, 8, 858–859. [Google Scholar] [CrossRef] [PubMed]
  77. Aframian, K.; Yousef Yengej, D.; Nwaobi, S.; Raman, S.; Faas, G.C.; Charles, A. Effects of chronic caffeine on patterns of brain blood flow and behavior throughout the sleep-wake cycle in freely behaving mice. PNAS Nexus 2023, 2, pgad303. [Google Scholar] [CrossRef] [PubMed]
  78. Shinomiya, K.; Omichi, J.; Ohnishi, R.; Ito, H.; Yoshida, T.; Kamei, C. Effects of chlorogenic acid and its metabolites on the sleep-wakefulness cycle in rats. Eur. J. Pharmacol. 2004, 504, 185–189. [Google Scholar] [CrossRef]
  79. Li, Y.; Guo, Z.; Cai, C.; Liu, D.; Kang, Y.; Liu, P. The orexinergic system mediates the excitatory effects of caffeine on the arousal and sympathetic activity. Heliyon 2023, 9, e14170. [Google Scholar] [CrossRef] [PubMed]
  80. Jagannath, A.; Varga, N.; Dallmann, R.; Rando, G.; Gosselin, P.; Ebrahimjee, F.; Taylor, L.; Mosneagu, D.; Stefaniak, J.; Walsh, S.; et al. Adenosine integrates light and sleep signalling for the regulation of circadian timing in mice. Nat. Commun. 2021, 12, 2113. [Google Scholar] [CrossRef]
  81. Frick, A.; Persson, J.; Bodén, R. Habitual caffeine consumption moderates the antidepressant effect of dorsomedial intermittent theta-burst transcranial magnetic stimulation. J. Psychopharmacol. 2021, 35, 1536–1541. [Google Scholar] [CrossRef]
  82. Tse, W.S.; Chan, C.C.; Shiu, S.Y.; Chung, P.Y.; Cheng, S.H. Caffeinated coffee enhances co-operative behavior in the Mixed Motive Game in healthy volunteers. Nutr. Neurosci. 2009, 12, 21–27. [Google Scholar] [CrossRef]
  83. Dawkins, L.; Shahzad, F.Z.; Ahmed, S.S.; Edmonds, C.J. Expectation of having consumed caffeine can improve performance and mood. Appetite 2011, 57, 597–600. [Google Scholar] [CrossRef]
  84. Loke, W.H.; Meliska, C.J. Effects of caffeine use and ingestion on a protracted visual vigilance task. Psychopharmacology 1984, 84, 54–57. [Google Scholar] [CrossRef]
  85. James, J.E.; Gregg, M.E. Effects of dietary caffeine on mood when rested and sleep restricted. Hum. Psychopharmacol. 2004, 19, 333–341. [Google Scholar] [CrossRef]
  86. Staack, A.; Distelberg, B.; Moldovan, C.; Belay, R.E.; Sabaté, J. The Impact of Caffeine Intake on Mental Health Symptoms in Postmenopausal Females with Overactive Bladder Symptoms: A Randomized, Double-Blind, Placebo-Controlled Trial. J. Women’s Health 2022, 31, 819–825. [Google Scholar] [CrossRef]
  87. Quinlan, P.; Lane, J.; Aspinall, L. Effects of hot tea, coffee and water ingestion on physiological responses and mood: The role of caffeine, water and beverage type. Psychopharmacology 1997, 134, 164–173. [Google Scholar] [CrossRef] [PubMed]
  88. Kimura, T.; Hayashida, H.; Murata, M.; Takamatsu, J. Effect of ferulic acid and Angelica archangelica extract on behavioral and psychological symptoms of dementia in frontotemporal lobar degeneration and dementia with Lewy bodies. Geriatr. Gerontol. Int. 2011, 11, 309–314. [Google Scholar] [CrossRef] [PubMed]
  89. Ochiai, R.; Tomonobu, K.; Ikushima, I. Effect of chlorogenic acids on fatigue and sleep in healthy males: A randomized, double-blind, placebo-controlled, crossover study. Food Sci. Nutr. 2018, 6, 2530–2536. [Google Scholar] [CrossRef] [PubMed]
  90. Park, I.; Ochiai, R.; Ogata, H.; Kayaba, M.; Hari, S.; Hibi, M.; Katsuragi, Y.; Satoh, M.; Tokuyama, K. Effects of subacute ingestion of chlorogenic acids on sleep architecture and energy metabolism through activity of the autonomic nervous system: A randomised, placebo-controlled, double-blinded cross-over trial. Br. J. Nutr. 2017, 117, 979–984. [Google Scholar] [CrossRef]
  91. Burke, T.M.; Markwald, R.R.; McHill, A.W.; Chinoy, E.D.; Snider, J.A.; Bessman, S.C.; Jung, C.M.; O’Neill, J.S.; Wright, K.P., Jr. Effects of caffeine on the human circadian clock in vivo and in vitro. Sci. Transl. Med. 2015, 7, 305ra146. [Google Scholar] [CrossRef]
  92. Ramos-Campo, D.J.; Pérez, A.; Ávila-Gandía, V.; Pérez-Piñero, S.; Rubio-Arias, J. Impact of Caffeine Intake on 800-m Running Performance and Sleep Quality in Trained Runners. Nutrients 2019, 11, 2040. [Google Scholar] [CrossRef]
  93. Weibel, J.; Lin, Y.S.; Landolt, H.P.; Kistler, J.; Rehm, S.; Rentsch, K.M.; Slawik, H.; Borgwardt, S.; Cajochen, C.; Reichert, C.F. The impact of daily caffeine intake on nighttime sleep in young adult men. Sci. Rep. 2021, 11, 4668. [Google Scholar] [CrossRef]
  94. Kim, J.; Kim, J. Green Tea, Coffee, and Caffeine Consumption Are Inversely Associated with Self-Report Lifetime Depression in the Korean Population. Nutrients 2018, 10, 1201. [Google Scholar] [CrossRef] [PubMed]
  95. Lucas, M.; Mirzaei, F.; Pan, A.; Okereke, O.I.; Willett, W.C.; O’Reilly, É.J.; Koenen, K.; Ascherio, A. Coffee, caffeine, and risk of depression among women. Arch. Intern. Med. 2011, 171, 1571–1578. [Google Scholar] [CrossRef]
  96. Lucas, M.; O’Reilly, E.J.; Pan, A.; Mirzaei, F.; Willett, W.C.; Okereke, O.I.; Ascherio, A. Coffee, caffeine, and risk of completed suicide: Results from three prospective cohorts of American adults. World J. Biol. Psychiatry Off. J. World Fed. Soc. Biol. Psychiatry 2014, 15, 377–386. [Google Scholar] [CrossRef]
  97. Wang, Y.; Wang, Z.; Gui, P.; Zhang, B.; Xie, Y. Coffee and caffeine intake and depression in postpartum women: A cross-sectional study from the National Health and Nutrition Examination Survey 2007-2018. Front. Psychol. 2023, 14, 1134522. [Google Scholar] [CrossRef]
  98. Narita, Z.; Hidese, S.; Kanehara, R.; Tachimori, H.; Hori, H.; Kim, Y.; Kunugi, H.; Arima, K.; Mizukami, S.; Tanno, K.; et al. Association of sugary drinks, carbonated beverages, vegetable and fruit juices, sweetened and black coffee, and green tea with subsequent depression: A five-year cohort study. Clin. Nutr. 2024, 43, 1395–1404. [Google Scholar] [CrossRef]
  99. Xie, J.; Huang, Z.; Mo, Y.; Pan, Y.; Ruan, Y.; Cao, W.; Chen, Y.; Li, Y.; Li, K.; Yu, D.; et al. Ages-specific beverage consumption and its association with depression and anxiety disorders: A prospective cohort study in 188,355 participants. J. Affect. Disord. 2024, 371, 224–233. [Google Scholar] [CrossRef]
  100. Kumar, V.; Bhat, Z.A.; Kumar, D. Animal models of anxiety: A comprehensive review. J. Pharmacol. Toxicol. Methods 2013, 68, 175–183. [Google Scholar] [CrossRef]
  101. Silverman, K.; Evans, S.M.; Strain, E.C.; Griffiths, R.R. Withdrawal syndrome after the double-blind cessation of caffeine consumption. Engl. J. Med. 1992, 327, 1109–1114. [Google Scholar] [CrossRef] [PubMed]
  102. Chait, L.D.; Griffiths, R.R. Effects of caffeine on cigarette smoking and subjective response. Clin. Pharmacol. Ther. 1983, 34, 612–622. [Google Scholar] [CrossRef] [PubMed]
  103. Nardi, A.E.; Valença, A.M.; Nascimento, I.; Freire, R.C.; Veras, A.B.; de-Melo-Neto, V.L.; Lopes, F.L.; King, A.L.; Soares-Filho, G.L.; Mezzasalma, M.A.; et al. A caffeine challenge test in panic disorder patients, their healthy first-degree relatives, and healthy controls. Depress. Anxiety 2008, 25, 847–853. [Google Scholar] [CrossRef]
  104. Liu, L.; Cheng, B.; Ye, J.; Qi, X.; Cheng, S.; Meng, P.; Chen, Y.; Yang, X.; Yao, Y.; Zhang, H.; et al. Understanding the Complex Interactions between Coffee, Tea Intake and Neurologically Relevant Tissues Proteins in the Development of Anxiety and Depression. J. Nutr. Health Aging 2022, 26, 1070–1077. [Google Scholar] [CrossRef]
  105. Castro, A.; Gili, M.; Visser, M.; Penninx, B.; Brouwer, I.A.; Montaño, J.J.; Pérez-Ara, M.; García-Toro, M.; Watkins, E.; Owens, M.; et al. Soft Drinks and Symptoms of Depression and Anxiety in Overweight Subjects: A Longitudinal Analysis of an European Cohort. Nutrients 2023, 15, 3865. [Google Scholar] [CrossRef]
  106. Klevebrant, L.; Frick, A. Effects of caffeine on anxiety and panic attacks in patients with panic disorder: A systematic review and meta-analysis. Gen. Hosp. Psychiatry 2022, 74, 22–31. [Google Scholar] [CrossRef]
  107. Gardiner, C.; Weakley, J.; Burke, L.M.; Roach, G.D.; Sargent, C.; Maniar, N.; Townshend, A.; Halson, S.L. The effect of caffeine on subsequent sleep: A systematic review and meta-analysis. Sleep Med. Rev. 2023, 69, 101764. [Google Scholar] [CrossRef]
  108. Revel, F.G.; Gottowik, J.; Gatti, S.; Wettstein, J.G.; Moreau, J.L. Rodent models of insomnia: A review of experimental procedures that induce sleep disturbances. Neurosci. Biobehav. Rev. 2009, 33, 874–899. [Google Scholar] [CrossRef]
  109. Cunha, R.A. How does adenosine control neuronal dysfunction and neurodegeneration? J. Neurochem. 2016, 139, 1019–1055. [Google Scholar] [CrossRef]
  110. Drapeau, C.; Hamel-Hébert, I.; Robillard, R.; Selmaoui, B.; Filipini, D.; Carrier, J. Challenging sleep in aging: The effects of 200 mg of caffeine during the evening in young and middle-aged moderate caffeine consumers. J. Sleep Res. 2006, 15, 133–141. [Google Scholar] [CrossRef] [PubMed]
  111. Marcus, G.M.; Rosenthal, D.G.; Nah, G.; Vittinghoff, E.; Fang, C.; Ogomori, K.; Joyce, S.; Yilmaz, D.; Yang, V.; Kessedjian, T.; et al. Acute Effects of Coffee Consumption on Health among Ambulatory Adults. Engl. J. Med. 2023, 388, 1092–1100. [Google Scholar] [CrossRef] [PubMed]
  112. Antonioli, L.; Pacher, P.; Haskó, G. Adenosine and inflammation: It’s time to (re)solve the problem. Trends Pharmacol. Sci. 2022, 43, 43–55. [Google Scholar] [CrossRef] [PubMed]
  113. Yegutkin, G.G.; Boison, D. ATP and Adenosine Metabolism in Cancer: Exploitation for Therapeutic Gain. Pharmacol. Rev. 2022, 74, 797–822. [Google Scholar] [CrossRef]
  114. Sarasola, L.I.; Del Torrent, C.L.; Pérez-Arévalo, A.; Argerich, J.; Casajuana-Martín, N.; Chevigné, A.; Fernández-Dueñas, V.; Ferré, S.; Pardo, L.; Ciruela, F. The ADORA1 mutation linked to early-onset Parkinson’s disease alters adenosine A(1)-A(2A) receptor heteromer formation and function. Biomed. Pharmacother. = Biomed. Pharmacother. 2022, 156, 113896. [Google Scholar] [CrossRef]
  115. Dunwiddie, T.V.; Masino, S.A. The role and regulation of adenosine in the central nervous system. Annu. Rev. Neurosci. 2001, 24, 31–55. [Google Scholar] [CrossRef]
  116. Proctor, W.R.; Dunwiddie, T.V. Pre- and postsynaptic actions of adenosine in the in vitro rat hippocampus. Brain Res. 1987, 426, 187–190. [Google Scholar] [CrossRef]
  117. Rebola, N.; Lujan, R.; Cunha, R.A.; Mulle, C. Adenosine A2A receptors are essential for long-term potentiation of NMDA-EPSCs at hippocampal mossy fiber synapses. Neuron 2008, 57, 121–134. [Google Scholar] [CrossRef]
  118. Lopes, L.V.; Cunha, R.A.; Kull, B.; Fredholm, B.B.; Ribeiro, J.A. Adenosine A(2A) receptor facilitation of hippocampal synaptic transmission is dependent on tonic A(1) receptor inhibition. Neuroscience 2002, 112, 319–329. [Google Scholar] [CrossRef] [PubMed]
  119. Duman, R.S.; Aghajanian, G.K. Synaptic dysfunction in depression: Potential therapeutic targets. Science 2012, 338, 68–72. [Google Scholar] [CrossRef]
  120. Wang, M.; Li, P.; Li, Z.; da Silva, B.S.; Zheng, W.; Xiang, Z.; He, Y.; Xu, T.; Cordeiro, C.; Deng, L.; et al. Lateral septum adenosine A(2A) receptors control stress-induced depressive-like behaviors via signaling to the hypothalamus and habenula. Nat. Commun. 2023, 14, 1880. [Google Scholar] [CrossRef]
  121. Zhong, Z.; Dong, H.; Zhou, S.; Lin, C.; Huang, P.; Li, X.; Zhang, J.; Xie, J.; Wu, Y.; Li, P. Caffeine’s Neuroprotective Effect on Memory Impairment: Suppression of Adenosine A(2)A Receptor and Enhancement of Tyrosine Hydroxylase in Dopaminergic Neurons Under Hypobaric Hypoxia Conditions. CNS Neurosci. Ther. 2024, 30, e70134. [Google Scholar] [CrossRef] [PubMed]
  122. Song, X.; Kirtipal, N.; Lee, S.; Malý, P.; Bharadwaj, S. Current therapeutic targets and multifaceted physiological impacts of caffeine. Phytother. Res. PTR 2023, 37, 5558–5598. [Google Scholar] [CrossRef] [PubMed]
  123. Lopes, J.P.; Pliássova, A.; Cunha, R.A. The physiological effects of caffeine on synaptic transmission and plasticity in the mouse hippocampus selectively depend on adenosine A(1) and A(2A) receptors. Biochem. Pharmacol. 2019, 166, 313–321. [Google Scholar] [CrossRef]
  124. Unsal, S.; Sanlier, N. Longitudinal Effects of Lifetime Caffeine Consumption on Levels of Depression, Anxiety, and Stress: A Comprehensive Review. Curr. Nutr. Rep. 2025, 14, 26. [Google Scholar] [CrossRef]
  125. Hamilton, S.P.; Slager, S.L.; De Leon, A.B.; Heiman, G.A.; Klein, D.F.; Hodge, S.E.; Weissman, M.M.; Fyer, A.J.; Knowles, J.A. Evidence for genetic linkage between a polymorphism in the adenosine 2A receptor and panic disorder. Neuropsychopharmacol. Off. Publ. Am. Coll. Neuropsychopharmacol. 2004, 29, 558–565. [Google Scholar] [CrossRef]
  126. Oliveira, S.; Ardais, A.P.; Bastos, C.R.; Gazal, M.; Jansen, K.; de Mattos Souza, L.; da Silva, R.A.; Kaster, M.P.; Lara, D.R.; Ghisleni, G. Impact of genetic variations in ADORA2A gene on depression and symptoms: A cross-sectional population-based study. Purinergic Signal. 2019, 15, 37–44. [Google Scholar] [CrossRef]
  127. Acquas, E.; Tanda, G.; Di Chiara, G. Differential effects of caffeine on dopamine and acetylcholine transmission in brain areas of drug-naive and caffeine-pretreated rats. Neuropsychopharmacol. Off. Publ. Am. Coll. Neuropsychopharmacol. 2002, 27, 182–193. [Google Scholar] [CrossRef]
  128. Szopa, A.; Poleszak, E.; Wyska, E.; Serefko, A.; Wośko, S.; Wlaź, A.; Pieróg, M.; Wróbel, A.; Wlaź, P. Caffeine enhances the antidepressant-like activity of common antidepressant drugs in the forced swim test in mice. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2016, 389, 211–221. [Google Scholar] [CrossRef]
  129. Popoli, P.; Giménez-Llort, L.; Pezzola, A.; Reggio, R.; Martínez, E.; Fuxe, K.; Ferré, S. Adenosine A1 receptor blockade selectively potentiates the motor effects induced by dopamine D1 receptor stimulation in rodents. Neurosci. Lett. 1996, 218, 209–213. [Google Scholar] [CrossRef] [PubMed]
  130. Garção, P.; Szabó, E.C.; Wopereis, S.; Castro, A.A.; Tomé, Â.R.; Prediger, R.D.; Cunha, R.A.; Agostinho, P.; Köfalvi, A. Functional interaction between pre-synaptic α6β2-containing nicotinic and adenosine A2A receptors in the control of dopamine release in the rat striatum. Br. J. Pharmacol. 2013, 169, 1600–1611. [Google Scholar] [CrossRef] [PubMed]
  131. Chen, J.F.; Choi, D.S.; Cunha, R.A. Striatopallidal adenosine A(2A) receptor modulation of goal-directed behavior: Homeostatic control with cognitive flexibility. Neuropharmacology 2023, 226, 109421. [Google Scholar] [CrossRef] [PubMed]
  132. Quarta, D.; Ferré, S.; Solinas, M.; You, Z.B.; Hockemeyer, J.; Popoli, P.; Goldberg, S.R. Opposite modulatory roles for adenosine A1 and A2A receptors on glutamate and dopamine release in the shell of the nucleus accumbens. Effects of chronic caffeine exposure. J. Neurochem. 2004, 88, 1151–1158. [Google Scholar] [CrossRef]
  133. Hamon, M.; Blier, P. Monoamine neurocircuitry in depression and strategies for new treatments. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2013, 45, 54–63. [Google Scholar] [CrossRef] [PubMed]
  134. Kaltenboeck, A.; Harmer, C. The neuroscience of depressive disorders: A brief review of the past and some considerations about the future. Brain Neurosci. Adv. 2018, 2, 2398212818799269. [Google Scholar] [CrossRef] [PubMed]
  135. Mössner, R.; Albert, D.; Persico, A.M.; Hennig, T.; Bengel, D.; Holtmann, B.; Schmitt, A.; Keller, F.; Simantov, R.; Murphy, D.; et al. Differential regulation of adenosine A(1) and A(2A) receptors in serotonin transporter and monoamine oxidase A-deficient mice. Eur. Neuropsychopharmacol. J. Eur. Coll. Neuropsychopharmacol. 2000, 10, 489–493. [Google Scholar] [CrossRef] [PubMed]
  136. Yokogoshi, H.; Iwata, T.; Ishida, K.; Yoshida, A. Effect of amino acid supplementation to low protein diet on brain and plasma levels of tryptophan and brain 5-hydroxyindoles in rats. J. Nutr. 1987, 117, 42–47. [Google Scholar] [CrossRef] [PubMed]
  137. Wang, Y.C.; Wang, X.; Yu, J.; Ma, F.; Li, Z.; Zhou, Y.; Zeng, S.; Ma, X.; Li, Y.R.; Neal, A.; et al. Targeting monoamine oxidase A-regulated tumor-associated macrophage polarization for cancer immunotherapy. Nat. Commun. 2021, 12, 3530. [Google Scholar] [CrossRef]
  138. Robinson, O.J.; Pike, A.C.; Cornwell, B.; Grillon, C. The translational neural circuitry of anxiety. J. Neurol. Neurosurg. Psychiatry 2019, 90, 1353–1360. [Google Scholar] [CrossRef]
  139. Smith, J.E.; Lawrence, A.D.; Diukova, A.; Wise, R.G.; Rogers, P.J. Storm in a coffee cup: Caffeine modifies brain activation to social signals of threat. Soc. Cogn. Affect. Neurosci. 2012, 7, 831–840. [Google Scholar] [CrossRef]
  140. Kenwood, M.M.; Kalin, N.H.; Barbas, H. The prefrontal cortex, pathological anxiety, and anxiety disorders. Neuropsychopharmacol. Off. Publ. Am. Coll. Neuropsychopharmacol. 2022, 47, 260–275. [Google Scholar] [CrossRef]
  141. Simões, A.P.; Machado, N.J.; Gonçalves, N.; Kaster, M.P.; Simões, A.T.; Nunes, A.; Pereira de Almeida, L.; Goosens, K.A.; Rial, D.; Cunha, R.A. Adenosine A(2A) Receptors in the Amygdala Control Synaptic Plasticity and Contextual Fear Memory. Neuropsychopharmacol. Off. Publ. Am. Coll. Neuropsychopharmacol. 2016, 41, 2862–2871. [Google Scholar] [CrossRef]
  142. Riera-Sampol, A.; Rodas, L.; Martínez, S.; Moir, H.J.; Tauler, P. Caffeine Intake among Undergraduate Students: Sex Differences, Sources, Motivations, and Associations with Smoking Status and Self-Reported Sleep Quality. Nutrients 2022, 14, 1661. [Google Scholar] [CrossRef]
  143. Park, J.; Han, J.W.; Lee, J.R.; Byun, S.; Suh, S.W.; Kim, T.; Yoon, I.Y.; Kim, K.W. Lifetime coffee consumption, pineal gland volume, and sleep quality in late life. Sleep 2018, 41, zsy127. [Google Scholar] [CrossRef]
  144. Zhou, X.; He, Y.; Xu, T.; Wu, Z.; Guo, W.; Xu, X.; Liu, Y.; Zhang, Y.; Shang, H.; Huang, L.; et al. 40 Hz light flickering promotes sleep through cortical adenosine signaling. Cell Res. 2024, 34, 214–231. [Google Scholar] [CrossRef]
  145. Barde, Y.A.; Edgar, D.; Thoenen, H. Purification of a new neurotrophic factor from mammalian brain. EMBO J. 1982, 1, 549–553. [Google Scholar] [CrossRef]
  146. Costa, R.O.; Martins, L.F.; Tahiri, E.; Duarte, C.B. Brain-derived neurotrophic factor-induced regulation of RNA metabolism in neuronal development and synaptic plasticity. Wiley Interdiscip. Rev. RNA 2022, 13, e1713. [Google Scholar] [CrossRef] [PubMed]
  147. Leal, G.; Bramham, C.R.; Duarte, C.B. BDNF and Hippocampal Synaptic Plasticity. Vitam. Horm. 2017, 104, 153–195. [Google Scholar] [CrossRef] [PubMed]
  148. Song, M.; Martinowich, K.; Lee, F.S. BDNF at the synapse: Why location matters. Mol. Psychiatry 2017, 22, 1370–1375. [Google Scholar] [CrossRef]
  149. Tanaka, J.; Horiike, Y.; Matsuzaki, M.; Miyazaki, T.; Ellis-Davies, G.C.; Kasai, H. Protein synthesis and neurotrophin-dependent structural plasticity of single dendritic spines. Science 2008, 319, 1683–1687. [Google Scholar] [CrossRef]
  150. Frisén, J.; Verge, V.M.; Fried, K.; Risling, M.; Persson, H.; Trotter, J.; Hökfelt, T.; Lindholm, D. Characterization of glial trkB receptors: Differential response to injury in the central and peripheral nervous systems. Proc. Natl. Acad. Sci. USA 1993, 90, 4971–4975. [Google Scholar] [CrossRef]
  151. Minichiello, L. TrkB signalling pathways in LTP and learning. Nat. Rev. Neurosci. 2009, 10, 850–860. [Google Scholar] [CrossRef]
  152. Wang, C.S.; Kavalali, E.T.; Monteggia, L.M. BDNF signaling in context: From synaptic regulation to psychiatric disorders. Cell 2022, 185, 62–76. [Google Scholar] [CrossRef] [PubMed]
  153. Turrigiano, G.G. The self-tuning neuron: Synaptic scaling of excitatory synapses. Cell 2008, 135, 422–435. [Google Scholar] [CrossRef]
  154. Reimers, J.M.; Loweth, J.A.; Wolf, M.E. BDNF contributes to both rapid and homeostatic alterations in AMPA receptor surface expression in nucleus accumbens medium spiny neurons. Eur. J. Neurosci. 2014, 39, 1159–1169. [Google Scholar] [CrossRef]
  155. Lu, B. BDNF and activity-dependent synaptic modulation. Learn. Mem. 2003, 10, 86–98. [Google Scholar] [CrossRef] [PubMed]
  156. Casarotto, P.C.; Girych, M.; Fred, S.M.; Kovaleva, V.; Moliner, R.; Enkavi, G.; Biojone, C.; Cannarozzo, C.; Sahu, M.P.; Kaurinkoski, K.; et al. Antidepressant drugs act by directly binding to TRKB neurotrophin receptors. Cell 2021, 184, 1299–1313.e1219. [Google Scholar] [CrossRef]
  157. Martín, E.D.; Buño, W. Caffeine-mediated presynaptic long-term potentiation in hippocampal CA1 pyramidal neurons. J. Neurophysiol. 2003, 89, 3029–3038. [Google Scholar] [CrossRef] [PubMed]
  158. Lao-Peregrín, C.; Ballesteros, J.J.; Fernández, M.; Zamora-Moratalla, A.; Saavedra, A.; Gómez Lázaro, M.; Pérez-Navarro, E.; Burks, D.; Martín, E.D. Caffeine-mediated BDNF release regulates long-term synaptic plasticity through activation of IRS2 signaling. Addict. Biol. 2017, 22, 1706–1718. [Google Scholar] [CrossRef] [PubMed]
  159. Alzoubi, K.H.; Srivareerat, M.; Aleisa, A.M.; Alkadhi, K.A. Chronic caffeine treatment prevents stress-induced LTP impairment: The critical role of phosphorylated CaMKII and BDNF. J. Mol. Neurosci. 2013, 49, 11–20. [Google Scholar] [CrossRef]
  160. Zoratto, F.; Fiore, M.; Ali, S.F.; Laviola, G.; Macrì, S. Neonatal tryptophan depletion and corticosterone supplementation modify emotional responses in adult male mice. Psychoneuroendocrinology 2013, 38, 24–39. [Google Scholar] [CrossRef]
  161. Poo, M.M. Neurotrophins as synaptic modulators. Nat. Rev. Neurosci. 2001, 2, 24–32. [Google Scholar] [CrossRef]
  162. Chao, M.V. Neurotrophins and their receptors: A convergence point for many signalling pathways. Nat. Rev. Neurosci. 2003, 4, 299–309. [Google Scholar] [CrossRef]
  163. Wei, C.J.; Augusto, E.; Gomes, C.A.; Singer, P.; Wang, Y.; Boison, D.; Cunha, R.A.; Yee, B.K.; Chen, J.F. Regulation of fear responses by striatal and extrastriatal adenosine A2A receptors in forebrain. Biol. Psychiatry 2014, 75, 855–863. [Google Scholar] [CrossRef]
  164. Florén Lind, S.; Stam, F.; Zelleroth, S.; Meurling, E.; Frick, A.; Grönbladh, A. Acute caffeine differently affects risk-taking and the expression of BDNF and of adenosine and opioid receptors in rats with high or low anxiety-like behavior. Pharmacol. Biochem. Behav. 2023, 227–228, 173573. [Google Scholar] [CrossRef] [PubMed]
  165. Oketch-Rabah, H.A.; Madden, E.F.; Roe, A.L.; Betz, J.M. United States Pharmacopeia (USP) Safety Review of Gamma-Aminobutyric Acid (GABA). Nutrients 2021, 13, 2742. [Google Scholar] [CrossRef] [PubMed]
  166. Asim, M.; Wang, H.; Waris, A.; He, J. Basolateral amygdala parvalbumin and cholecystokinin-expressing GABAergic neurons modulate depressive and anxiety-like behaviors. Transl. Psychiatry 2024, 14, 418. [Google Scholar] [CrossRef] [PubMed]
  167. Fuchs, T.; Jefferson, S.J.; Hooper, A.; Yee, P.H.; Maguire, J.; Luscher, B. Disinhibition of somatostatin-positive GABAergic interneurons results in an anxiolytic and antidepressant-like brain state. Mol. Psychiatry 2017, 22, 920–930. [Google Scholar] [CrossRef]
  168. Li, R.; Yang, Y.; Wang, L.; Tang, G.; Yang, J.; Gao, S.; Liu, J. Blockade of pre- and post-synaptic GABA(B) receptors in the anteroventral bed nucleus of stria terminalis produces anxiolytic-like and anxiety-like effects in parkinsonian rats respectively. Neuropharmacology 2024, 257, 110033. [Google Scholar] [CrossRef]
  169. Jacobson, L.H.; Vlachou, S.; Slattery, D.A.; Li, X.; Cryan, J.F. The Gamma-Aminobutyric Acid B Receptor in Depression and Reward. Biol. Psychiatry 2018, 83, 963–976. [Google Scholar] [CrossRef]
  170. Alasmari, F. Caffeine induces neurobehavioral effects through modulating neurotransmitters. Saudi Pharm. J. SPJ Off. Publ. Saudi Pharm. Soc. 2020, 28, 445–451. [Google Scholar] [CrossRef]
  171. Isokawa, M. Caffeine-Induced Suppression of GABAergic Inhibition and Calcium-Independent Metaplasticity. Neural Plast. 2016, 2016, 1239629. [Google Scholar] [CrossRef]
  172. Yang, J.; Yang, G.; Ren, J.; Zhao, J.; Li, S.J.N. Caffeine suppresses GABA receptor-mediated current in rat primary sensory neurons via inhibition of intracellular phosphodiesterase. Neurophysiology 2015, 47, 108–114. [Google Scholar] [CrossRef]
  173. Mukhopadhyay, S.; Poddar, M.K. Is GABA involved in the development of caffeine tolerance? Neurochem. Res. 1998, 23, 63–68. [Google Scholar] [CrossRef]
  174. Bagga, P.; Chugani, A.N.; Patel, A.B. Neuroprotective effects of caffeine in MPTP model of Parkinson’s disease: A (13)C NMR study. Neurochem. Int. 2016, 92, 25–34. [Google Scholar] [CrossRef] [PubMed]
  175. Pizzino, G.; Irrera, N.; Cucinotta, M.; Pallio, G.; Mannino, F.; Arcoraci, V.; Squadrito, F.; Altavilla, D.; Bitto, A. Oxidative Stress: Harms and Benefits for Human Health. Oxidative Med. Cell. Longev. 2017, 2017, 8416763. [Google Scholar] [CrossRef]
  176. Bhatt, S.; Nagappa, A.N.; Patil, C.R. Role of oxidative stress in depression. Drug Discov. Today 2020, 25, 1270–1276. [Google Scholar] [CrossRef]
  177. Bouayed, J.; Rammal, H.; Soulimani, R. Oxidative stress and anxiety: Relationship and cellular pathways. Oxidative Med. Cell. Longev. 2009, 2, 63–67. [Google Scholar] [CrossRef]
  178. Trifunovic, S.; Stevanovic, I.; Milosevic, A.; Ristic, N.; Janjic, M.; Bjelobaba, I.; Savic, D.; Bozic, I.; Jakovljevic, M.; Tesovic, K.; et al. The Function of the Hypothalamic-Pituitary-Adrenal Axis During Experimental Autoimmune Encephalomyelitis: Involvement of Oxidative Stress Mediators. Front. Neurosci. 2021, 15, 649485. [Google Scholar] [CrossRef]
  179. Du, J.; Wang, Y.; Hunter, R.; Wei, Y.; Blumenthal, R.; Falke, C.; Khairova, R.; Zhou, R.; Yuan, P.; Machado-Vieira, R.; et al. Dynamic regulation of mitochondrial function by glucocorticoids. Proc. Natl. Acad. Sci. USA 2009, 106, 3543–3548. [Google Scholar] [CrossRef]
  180. Spiers, J.G.; Chen, H.J.; Sernia, C.; Lavidis, N.A. Activation of the hypothalamic-pituitary-adrenal stress axis induces cellular oxidative stress. Front. Neurosci. 2014, 8, 456. [Google Scholar] [CrossRef] [PubMed]
  181. Correia, A.S.; Cardoso, A.; Vale, N. Oxidative Stress in Depression: The Link with the Stress Response, Neuroinflammation, Serotonin, Neurogenesis and Synaptic Plasticity. Antioxidants 2023, 12, 470. [Google Scholar] [CrossRef]
  182. Dutta, A.; Sarkar, P.; Shrivastava, S.; Chattopadhyay, A. Effect of Hypoxia on the Function of the Human Serotonin(1A) Receptor. ACS Chem. Neurosci. 2022, 13, 1456–1466. [Google Scholar] [CrossRef] [PubMed]
  183. Rammal, H.; Bouayed, J.; Younos, C.; Soulimani, R. Evidence that oxidative stress is linked to anxiety-related behaviour in mice. Brain Behav. Immun. 2008, 22, 1156–1159. [Google Scholar] [CrossRef]
  184. Wang, H.; Jin, M.; Xie, M.; Yang, Y.; Xue, F.; Li, W.; Zhang, M.; Li, Z.; Li, X.; Jia, N.; et al. Protective role of antioxidant supplementation for depression and anxiety: A meta-analysis of randomized clinical trials. J. Affect. Disord. 2023, 323, 264–279. [Google Scholar] [CrossRef]
  185. Hall, S.; Desbrow, B.; Anoopkumar-Dukie, S.; Davey, A.K.; Arora, D.; McDermott, C.; Schubert, M.M.; Perkins, A.V.; Kiefel, M.J.; Grant, G.D. A review of the bioactivity of coffee, caffeine and key coffee constituents on inflammatory responses linked to depression. Food Res. Int. 2015, 76, 626–636. [Google Scholar] [CrossRef]
  186. Devasagayam, T.P.; Kamat, J.P.; Mohan, H.; Kesavan, P.C. Caffeine as an antioxidant: Inhibition of lipid peroxidation induced by reactive oxygen species. Biochim. Et Biophys. Acta 1996, 1282, 63–70. [Google Scholar] [CrossRef]
  187. Azam, S.; Hadi, N.; Khan, N.U.; Hadi, S.M. Antioxidant and prooxidant properties of caffeine, theobromine and xanthine. Med. Sci. Monit. Int. Med. J. Exp. Clin. Res. 2003, 9, Br325-30. [Google Scholar]
  188. Kanski, J.; Aksenova, M.; Stoyanova, A.; Butterfield, D.A. Ferulic acid antioxidant protection against hydroxyl and peroxyl radical oxidation in synaptosomal and neuronal cell culture systems in vitro: Structure-activity studies. J. Nutr. Biochem. 2002, 13, 273–281. [Google Scholar] [CrossRef]
  189. Búfalo, M.C.; Ferreira, I.; Costa, G.; Francisco, V.; Liberal, J.; Cruz, M.T.; Lopes, M.C.; Batista, M.T.; Sforcin, J.M. Propolis and its constituent caffeic acid suppress LPS-stimulated pro-inflammatory response by blocking NF-κB and MAPK activation in macrophages. J. Ethnopharmacol. 2013, 149, 84–92. [Google Scholar] [CrossRef] [PubMed]
  190. Kim, B.C.; Kim, H.G.; Lee, S.A.; Lim, S.; Park, E.H.; Kim, S.J.; Lim, C.J. Genipin-induced apoptosis in hepatoma cells is mediated by reactive oxygen species/c-Jun NH2-terminal kinase-dependent activation of mitochondrial pathway. Biochem. Pharmacol. 2005, 70, 1398–1407. [Google Scholar] [CrossRef] [PubMed]
  191. Son, Y.; Cheong, Y.K.; Kim, N.H.; Chung, H.T.; Kang, D.G.; Pae, H.O. Mitogen-Activated Protein Kinases and Reactive Oxygen Species: How Can ROS Activate MAPK Pathways? J. Signal Transduct. 2011, 2011, 792639. [Google Scholar] [CrossRef]
  192. Han, Z.; Wang, L.; Zhu, H.; Tu, Y.; He, P.; Li, B. Uncovering the effects and mechanisms of tea and its components on depression, anxiety, and sleep disorders: A comprehensive review. Food Res. Int. 2024, 197, 115191. [Google Scholar] [CrossRef] [PubMed]
  193. Liu, Y.; Wu, Z.; Cheng, L.; Zhang, X.; Yang, H. The role of the intestinal microbiota in the pathogenesis of host depression and mechanism of TPs relieving depression. Food Funct. 2021, 12, 7651–7663. [Google Scholar] [CrossRef]
  194. Dahchour, A. Anxiolytic and antidepressive potentials of rosmarinic acid: A review with a focus on antioxidant and anti-inflammatory effects. Pharmacol. Res. 2022, 184, 106421. [Google Scholar] [CrossRef]
  195. Zielinski, M.R.; Gibbons, A.J. Neuroinflammation, Sleep, and Circadian Rhythms. Front. Cell. Infect. Microbiol. 2022, 12, 853096. [Google Scholar] [CrossRef]
  196. Kofod, J.; Elfving, B.; Nielsen, E.H.; Mors, O.; Köhler-Forsberg, O. Depression and inflammation: Correlation between changes in inflammatory markers with antidepressant response and long-term prognosis. Eur. Neuropsychopharmacol. J. Eur. Coll. Neuropsychopharmacol. 2022, 54, 116–125. [Google Scholar] [CrossRef]
  197. Beurel, E.; Toups, M.; Nemeroff, C.B. The Bidirectional Relationship of Depression and Inflammation: Double Trouble. Neuron 2020, 107, 234–256. [Google Scholar] [CrossRef]
  198. Yin, Y.; Ju, T.; Zeng, D.; Duan, F.; Zhu, Y.; Liu, J.; Li, Y.; Lu, W. “Inflamed” depression: A review of the interactions between depression and inflammation and current anti-inflammatory strategies for depression. Pharmacol. Res. 2024, 207, 107322. [Google Scholar] [CrossRef] [PubMed]
  199. Jiang, R.; Noble, S.; Rosenblatt, M.; Dai, W.; Ye, J.; Liu, S.; Qi, S.; Calhoun, V.D.; Sui, J.; Scheinost, D. The brain structure, inflammatory, and genetic mechanisms mediate the association between physical frailty and depression. Nat. Commun. 2024, 15, 4411. [Google Scholar] [CrossRef]
  200. Boukezzi, S.; Costi, S.; Shin, L.M.; Kim-Schulze, S.; Cathomas, F.; Collins, A.; Russo, S.J.; Morris, L.S.; Murrough, J.W. Exaggerated amygdala response to threat and association with immune hyperactivity in depression. Brain Behav. Immun. 2022, 104, 205–212. [Google Scholar] [CrossRef] [PubMed]
  201. Huang, S.; Dong, W.; Lin, X.; Xu, K.; Li, K.; Xiong, S.; Wang, Z.; Nie, X.; Bian, J.S. Disruption of the Na(+)/K(+)-ATPase-purinergic P2X7 receptor complex in microglia promotes stress-induced anxiety. Immunity 2024, 57, 495–512.e411. [Google Scholar] [CrossRef] [PubMed]
  202. Horrigan, L.A.; Kelly, J.P.; Connor, T.J. Caffeine suppresses TNF-alpha production via activation of the cyclic AMP/protein kinase A pathway. Int. Immunopharmacol. 2004, 4, 1409–1417. [Google Scholar] [CrossRef]
  203. Shen, W.; Qi, R.; Zhang, J.; Wang, Z.; Wang, H.; Hu, C.; Zhao, Y.; Bie, M.; Wang, Y.; Fu, Y.; et al. Chlorogenic acid inhibits LPS-induced microglial activation and improves survival of dopaminergic neurons. Brain Res. Bull. 2012, 88, 487–494. [Google Scholar] [CrossRef]
  204. Hirata, A.; Murakami, Y.; Atsumi, T.; Shoji, M.; Ogiwara, T.; Shibuya, K.; Ito, S.; Yokoe, I.; Fujisawa, S. Ferulic acid dimer inhibits lipopolysaccharide-stimulated cyclooxygenase-2 expression in macrophages. Vivo 2005, 19, 849–853. [Google Scholar]
  205. Hosoda, A.; Ozaki, Y.; Kashiwada, A.; Mutoh, M.; Wakabayashi, K.; Mizuno, K.; Nomura, E.; Taniguchi, H. Syntheses of ferulic acid derivatives and their suppressive effects on cyclooxygenase-2 promoter activity. Bioorganic Med. Chem. 2002, 10, 1189–1196. [Google Scholar] [CrossRef]
  206. Gerritsen, L.; Staufenbiel, S.M.; Penninx, B.; van Hemert, A.M.; Noppe, G.; de Rijke, Y.B.; van Rossum, E.F.C. Long-term glucocorticoid levels measured in hair in patients with depressive and anxiety disorders. Psychoneuroendocrinology 2019, 101, 246–252. [Google Scholar] [CrossRef]
  207. Guerry, J.D.; Hastings, P.D. In search of HPA axis dysregulation in child and adolescent depression. Clin. Child Fam. Psychol. Rev. 2011, 14, 135–160. [Google Scholar] [CrossRef]
  208. Karabatsiakis, A.; de Punder, K.; Manrique, J.-S.; Todt, M.; Dietrich, D.E. Hair Cortisol as a Predictive Biochemical Indicator for the Preventive and Personalised Evaluation of Suicide Risk in Depression? An Exploratory Comparison Study Between Mentally-Stable Control Subjects, Depressed Individuals and Individuals After Completing Suicide; Springer: Berlin/Heidelberg, Germany, 2022. [Google Scholar]
  209. Staufenbiel, S.M.; Penninx, B.W.; de Rijke, Y.B.; van den Akker, E.L.; van Rossum, E.F. Determinants of hair cortisol and hair cortisone concentrations in adults. Psychoneuroendocrinology 2015, 60, 182–194. [Google Scholar] [CrossRef]
  210. Zerroug, Y.; Marin, M.F.; Porter-Vignola, E.; Garel, P.; Herba, C.M. Differences in hair cortisol to cortisone ratio between depressed and non-depressed adolescent women. Stress 2025, 28, 2459726. [Google Scholar] [CrossRef]
  211. Schatzberg, A.F.; Lindley, S. Glucocorticoid antagonists in neuropsychiatric [corrected] disorders. Eur. J. Pharmacol. 2008, 583, 358–364. [Google Scholar] [CrossRef] [PubMed]
  212. Scher, A.; Hall, W.A.; Zaidman-Zait, A.; Weinberg, J. Sleep quality, cortisol levels, and behavioral regulation in toddlers. Dev. Psychobiol. 2010, 52, 44–53. [Google Scholar] [CrossRef] [PubMed]
  213. Rabl, U.; Bartova, L.; Sezen, P.; Keller, J.; Schatzberg, A.; Pezawas, L. HPA axis in psychotic and non-psychotic major depression: Cortisol plasma levels and hippocampal volume. J. Affect. Disord. 2025, 377, 14–22. [Google Scholar] [CrossRef]
  214. Kokras, N.; Hodes, G.E.; Bangasser, D.A.; Dalla, C. Sex differences in the hypothalamic-pituitary-adrenal axis: An obstacle to antidepressant drug development? Br. J. Pharmacol. 2019, 176, 4090–4106. [Google Scholar] [CrossRef]
  215. Dickerson, S.S.; Kemeny, M.E. Acute stressors and cortisol responses: A theoretical integration and synthesis of laboratory research. Psychol. Bull. 2004, 130, 355–391. [Google Scholar] [CrossRef]
  216. Füzesi, T.; Daviu, N.; Wamsteeker Cusulin, J.I.; Bonin, R.P.; Bains, J.S. Hypothalamic CRH neurons orchestrate complex behaviours after stress. Nat. Commun. 2016, 7, 11937. [Google Scholar] [CrossRef]
  217. McEwen, B.S. Physiology and neurobiology of stress and adaptation: Central role of the brain. Physiol. Rev. 2007, 87, 873–904. [Google Scholar] [CrossRef] [PubMed]
  218. Zhao, X.; Hu, W.; Liu, Y.; Guo, K.; Liu, Y.; Yang, J. Separating the influences of means and daily variations of sleep on the stress-induced salivary cortisol response. Psychoneuroendocrinology 2023, 151, 106059. [Google Scholar] [CrossRef] [PubMed]
  219. Okun, M.L.; Segerstrom, S.; Jackman, S.; Ross, K.; Schetter, C.D.; Coussons-Read, M. Variability in perinatal sleep quality is associated with an atypical cortisol awakening response and increased mood symptoms. Psychoneuroendocrinology 2025, 172, 107248. [Google Scholar] [CrossRef] [PubMed]
  220. Hansen, Å.M.; Thomsen, J.F.; Kaergaard, A.; Kolstad, H.A.; Kaerlev, L.; Mors, O.; Rugulies, R.; Bonde, J.P.; Andersen, J.H.; Mikkelsen, S.J.P. Salivary cortisol and sleep problems among civil servants. Psychoneuroendocrinology 2012, 37, 1086–1095. [Google Scholar] [CrossRef]
  221. Huang, T.W.; Cheung, D.S.T.; Xu, X.; Loh, E.W.; Lai, J.H.; Su, W.W.; Wu, S.S.; Lin, C.C. Relationship between diurnal cortisol profile and sleep quality in patients with hepatocellular carcinoma. Biol. Res. Nurs. 2020, 22, 139–147. [Google Scholar] [CrossRef]
  222. Backhaus, J.; Junghanns, K.; Hohagen, F.J.P. Sleep disturbances are correlated with decreased morning awakening salivary cortisol. Psychoneuroendocrinology 2004, 29, 1184–1191. [Google Scholar] [CrossRef]
  223. Patz, M.D.; Day, H.E.; Burow, A.; Campeau, S. Modulation of the hypothalamo-pituitary-adrenocortical axis by caffeine. Psychoneuroendocrinology 2006, 31, 493–500. [Google Scholar] [CrossRef]
  224. O’Neill, C.E.; Newsom, R.J.; Stafford, J.; Scott, T.; Archuleta, S.; Levis, S.C.; Spencer, R.L.; Campeau, S.; Bachtell, R.K. Adolescent caffeine consumption increases adulthood anxiety-related behavior and modifies neuroendocrine signaling. Psychoneuroendocrinology 2016, 67, 40–50. [Google Scholar] [CrossRef]
  225. Cryan, J.F.; O’Riordan, K.J.; Cowan, C.S.M.; Sandhu, K.V.; Bastiaanssen, T.F.S.; Boehme, M.; Codagnone, M.G.; Cussotto, S.; Fulling, C.; Golubeva, A.V.; et al. The Microbiota-Gut-Brain Axis. Physiol. Rev. 2019, 99, 1877–2013. [Google Scholar] [CrossRef]
  226. Morris, G.; Berk, M.; Carvalho, A.; Caso, J.R.; Sanz, Y.; Walder, K.; Maes, M. The Role of the Microbial Metabolites Including Tryptophan Catabolites and Short Chain Fatty Acids in the Pathophysiology of Immune-Inflammatory and Neuroimmune Disease. Mol. Neurobiol. 2017, 54, 4432–4451. [Google Scholar] [CrossRef]
  227. Zhang, Q.; Bi, Y.; Zhang, B.; Jiang, Q.; Mou, C.K.; Lei, L.; Deng, Y.; Li, Y.; Yu, J.; Liu, W.; et al. Current landscape of fecal microbiota transplantation in treating depression. Front. Immunol. 2024, 15, 1416961. [Google Scholar] [CrossRef]
  228. Rothenberg, D.O.; Zhang, L. Mechanisms Underlying the Anti-Depressive Effects of Regular Tea Consumption. Nutrients 2019, 11, 1361. [Google Scholar] [CrossRef]
  229. Radford-Smith, D.E.; Anthony, D.C. Prebiotic and Probiotic Modulation of the Microbiota-Gut-Brain Axis in Depression. Nutrients 2023, 15, 1880. [Google Scholar] [CrossRef] [PubMed]
  230. Venkataraman, R.; Madempudi, R.S.; Neelamraju, J.; Ahire, J.J.; Vinay, H.R.; Lal, A.; Thomas, G.; Stephen, S. Effect of Multi-strain Probiotic Formulation on Students Facing Examination Stress: A Double-Blind, Placebo-Controlled Study. Probiotics Antimicrob. Proteins 2021, 13, 12–18. [Google Scholar] [CrossRef] [PubMed]
  231. Anand, N.; Gorantla, V.R.; Chidambaram, S.B. The Role of Gut Dysbiosis in the Pathophysiology of Neuropsychiatric Disorders. Cells 2022, 12, 54. [Google Scholar] [CrossRef]
  232. Cai, T.; Zheng, S.P.; Shi, X.; Yuan, L.Z.; Hu, H.; Zhou, B.; Xiao, S.L.; Wang, F. Therapeutic effect of fecal microbiota transplantation on chronic unpredictable mild stress-induced depression. Front. Cell. Infect. Microbiol. 2022, 12, 900652. [Google Scholar] [CrossRef]
  233. Saygili, S.; Hegde, S.; Shi, X.Z. Effects of Coffee on Gut Microbiota and Bowel Functions in Health and Diseases: A Literature Review. Nutrients 2024, 16, 3155. [Google Scholar] [CrossRef] [PubMed]
  234. Cowan, T.E.; Palmnäs, M.S.; Yang, J.; Bomhof, M.R.; Ardell, K.L.; Reimer, R.A.; Vogel, H.J.; Shearer, J. Chronic coffee consumption in the diet-induced obese rat: Impact on gut microbiota and serum metabolomics. J. Nutr. Biochem. 2014, 25, 489–495. [Google Scholar] [CrossRef]
  235. Jang, H.M.; Lee, H.J.; Jang, S.E.; Han, M.J.; Kim, D.H. Evidence for interplay among antibacterial-induced gut microbiota disturbance, neuro-inflammation, and anxiety in mice. Mucosal Immunol. 2018, 11, 1386–1397. [Google Scholar] [CrossRef] [PubMed]
  236. Needham, B.D.; Funabashi, M.; Adame, M.D.; Wang, Z.; Boktor, J.C.; Haney, J.; Wu, W.L.; Rabut, C.; Ladinsky, M.S.; Hwang, S.J.; et al. A gut-derived metabolite alters brain activity and anxiety behaviour in mice. Nature 2022, 602, 647–653. [Google Scholar] [CrossRef]
  237. Black, C.N.; Bot, M.; Scheffer, P.G.; Cuijpers, P.; Penninx, B.W. Is depression associated with increased oxidative stress? A systematic review and meta-analysis. Psychoneuroendocrinology 2015, 51, 164–175. [Google Scholar] [CrossRef]
  238. Lyte, J.M. Eating for 3.8 × 10(13): Examining the Impact of Diet and Nutrition on the Microbiota-Gut-Brain Axis Through the Lens of Microbial Endocrinology. Front. Endocrinol. 2018, 9, 796. [Google Scholar] [CrossRef]
  239. Thaiss, C.A.; Zeevi, D.; Levy, M.; Zilberman-Schapira, G.; Suez, J.; Tengeler, A.C.; Abramson, L.; Katz, M.N.; Korem, T.; Zmora, N.; et al. Transkingdom control of microbiota diurnal oscillations promotes metabolic homeostasis. Cell 2014, 159, 514–529. [Google Scholar] [CrossRef] [PubMed]
  240. Gutierrez Lopez, D.E.; Lashinger, L.M.; Weinstock, G.M.; Bray, M.S. Circadian rhythms and the gut microbiome synchronize the host’s metabolic response to diet. Cell Metab. 2021, 33, 873–887. [Google Scholar] [CrossRef] [PubMed]
  241. Matenchuk, B.A.; Mandhane, P.J.; Kozyrskyj, A.L. Sleep, circadian rhythm, and gut microbiota. Sleep Med. Rev. 2020, 53, 101340. [Google Scholar] [CrossRef]
  242. Song, Z.; Liu, L.; Xu, Y.; Cao, R.; Lan, X.; Pan, C.; Zhang, S.; Zhao, H. Caffeine-Induced Sleep Restriction Alters the Gut Microbiome and Fecal Metabolic Profiles in Mice. Int. J. Mol. Sci. 2022, 23, 14837. [Google Scholar] [CrossRef]
  243. Vaccaro, A.; Kaplan Dor, Y.; Nambara, K.; Pollina, E.A.; Lin, C.; Greenberg, M.E.; Rogulja, D. Sleep Loss Can Cause Death through Accumulation of Reactive Oxygen Species in the Gut. Cell 2020, 181, 1307–1328.e1315. [Google Scholar] [CrossRef]
Figure 1. Flowchart of systematic literature search.
Figure 1. Flowchart of systematic literature search.
Nutrients 17 03037 g001
Figure 2. Overview of methods for evaluating depression (A), anxiety (B), and SDs (C) in animal experiments. Notes: The “?” next to the mouse in the picture means the different choices made by the mouse between water and sucrose solution in the SPT.
Figure 2. Overview of methods for evaluating depression (A), anxiety (B), and SDs (C) in animal experiments. Notes: The “?” next to the mouse in the picture means the different choices made by the mouse between water and sucrose solution in the SPT.
Nutrients 17 03037 g002
Figure 3. Potential mechanisms of action of the main active ingredients in coffee on depression, anxiety, and sleep disorders. Notes: Blue “↓” represents downregulation, red “↑” represents upregulation, black arrows represent the path and direction of the mechanism in the figure.
Figure 3. Potential mechanisms of action of the main active ingredients in coffee on depression, anxiety, and sleep disorders. Notes: Blue “↓” represents downregulation, red “↑” represents upregulation, black arrows represent the path and direction of the mechanism in the figure.
Nutrients 17 03037 g003
Table 1. Animal experimental studies on the effects of coffee and its active ingredients on depression, anxiety, SDs.
Table 1. Animal experimental studies on the effects of coffee and its active ingredients on depression, anxiety, SDs.
Mental
Diseases
Coffee and Its
Active Ingredients
Animal ModelsDose and
Duration
Behavioral ManifestationsResults and MechanismsReferences
DepressionCaffeinated and
decaffeinated
coffee
(Coffee Extracts)
PDD inbred male Wistar rats200 mg/kg (p.o.),
7 d
Sports activities↑(OFT)
Sucrose consumption↑
(SPT)
Immobility time↓ (FST)
Immobility time↓ (TST)
IL-6↓ TNF-α↓
SOD↑ GPx↑
Akkermansia (Verrucomicrobia)↑
Phylum Bacteroidetes (Bacteroidetes)↓
[53]
Caffeinated coffee
(Coffee extracts)
Male C57BL/6J mice140 mg/kg (p.o.),
14 d
Sports activities↑ (OFT)
Immobility time↓ (FST)
Immobility time↓ (TST)
IDO↑ PGE2↓
Neopterin/biopterin ratio↑
UA/3-HK, UA/KYN↑
[55]
Caffeine
(Pure ingredients)
Male C57BL/6 mice (10–12 weeks old)6 g/L/week (p.o.),
3 weeks
Immobility time↓ (FST)
Immobility time↓ (TST)
Pleasure deficit-like behavior↓
(SPT)
memory impairment↓
synaptic marker↓
hippocampal density of syntaxin↓
amplitude of hippocampal LTP↓
[56]
Caffeine
(Pure ingredients)
Male C57BL/6J mice (Chronic MildStress)0.5 mg/mL (p.o.),
6 weeks
Immobility time↓ (FST)
Immobility time↓ (TST)
KYN↓ KYNA↑
KAT↑ PGC-1α↑
[58]
Caffeine
(Pure ingredients)
Kunming male mice (Chronic Water Immression Restraint Stress)10, 20 mg/kg (p.o.),
4 weeks
Immobility time↓ (FST)
Immobility time↓ (TST)
5-HT↑
BrdU+/DCX+ cells↑
TNF-α↓ IL-1β↓
[57]
Caffeine
(Pure ingredients)
Sprague-Dawley rats0.25 g/L (p.o.),
14 d
Upright behavior of male rats↓
Immobility time of male rats↓(FST)
Struggling behavior of female rats↑
Changes in 5-HT levels
Regulation of NE
[59]
DepressionCaffeine
(Pure ingredients)
Swiss albino CD1 mice (Helpless Mice)0.3 g/L (p.o.),
4 weeks
Immobility time↑ (FST)
Immobility time↑ (TST)
Time spent in the open arms↓
Length of stay in the central area↓
Memory recovery↑
density of synaptophysin, SNAP-25↓
Restored loss of hippocampal
synaptic markers
[62]
Caffeine
(Pure ingredients)
Male C57BL/6J mice10 mg/kg (p.o.),
14 d
Immobility time↓ (FST)
Immobility time↓ (TST)
TNF-α↓ IL-6↓
IL-1β↓ IFN-γ↓
MDA↓ BDNF↑
[61]
SY-2476 (Caffeine derivative, pure ingredients)Wistar rats10, 30 mg/kg (p.o.),
21 d
Sucrose consumption↑
(SPT)
Immobility time↓ (FST)
Serum cortisol levels↓
MDA↓ SOD↑ GSH↑
A1R expression↑
A2ARexpression↓
[60]
caffeine
(Pure ingredients)
Male Wistar rats (Stress Re-stress)10, 20, 30 mg/kg (i.p.),
24 d
Immobility time (20, 30 mg/kg)↑
(FST)
Plasma CORT levels,
5-HT levels,
GR and MR levels were not changed
[64]
CGA
(Pure ingredients)
Male Wistar rats500 mg kg−1 (p.o.),
14 d
Sucrose consumption↑ (SPT)
Sports activities↑ (OFT)
Immobility time↓ (FST)
Immobility time↓ (TST)
IL-6 and TNF-α↓
Microbiome diversity↑
Serum pro-inflammatory cytokine↓
Serum monoamine neurotransmitter↑
IL-8↓
[50]
FA
(Pure ingredients)
Male Swiss mice0.01, 0.1, 1, 10 mg/kg (p.o.),
1 h
Immobility time↓ (FST)
Immobility time↓ (TST)
5-HT↑ SNRIs↑[66]
CA
(Pure ingredients)
Male ICR mice and ddY mice4 mg/kg (i.p.),
1 d
Immobility time↓ (FST)Indirect regulation of the
ADRA1A system
[67]
AnxietyCaffeine
(Pure ingredients)
Male Sprague Dawley rats3 mg/kg (i.p.), 10 min (acute stress) or 14 d (Chronic variable stress)Head Down Frequency↑Cognitive Differences Score↑
MDA↓ SOD (acute)↑ GSH (acute)↑
[68]
Caffeine
(Pure ingredients)
Male C57BL/6 mice5 mg/kg (i.p.), 24 dOpen arm dwell time (short-term
effect) ↑(EPM)
MDA↓NO↓[69]
Caffeine
(Pure ingredients)
Male Wistar rats (Stress Re-stress)10, 20, 30 mg/kg (i.p.),
24 d
Open arm dwell time (20, 30 mg/kg) ↓(EPM)Plasma CORT levels,
5-HT levels,
GR and MR levels were not changed
[64]
CGA, Caffeine (Pure ingredients)
Coffee
(Coffee Extracts)
Adult male
Wistar rats
CGA: 5 mg/kg (p.o.),
Caffeine: 15 mg/kg (p.o.), Coffee: 0.5 g/kg (p.o.), 29 d
Step-down latencies↑ (IAT)TBARS Level↓
AChE activity↓
Nerve damage↓
[70]
ICCA
(Coffee Extracts)
Male ICR mice20, 40 mg/kg (p.o.), 3 monthsOpen arm dwell time and access
frequency↑ (EPM)
Light box dwell time↑ (LDB)
SH-SY5Y cells, B16 cells,
H9C2 cells↓
OS↓ TNF-α and IL-6↓ Nrf2↑
[71]
CGA
(Coffee Extracts)
Swiss albino
male mice
20 mg/kg (i.p.), 1 hLight box dwell time↑ (LDB)
Time spent in the open arms and
frequency of entering the open arms↑ (EPM)
Feeding behavior, exercise ability↑ (FET)
Benzodiazepine receptors↑
OS↓
[72]
AnxietyCGA
SYG
(Pure ingredients)
Male Sprague Dawley ratsCGA: 40 mg/kg (p.o.),
7 d
SYG: 32 mg/kg (p.o.),
7 d
Time spent in open arms↑
(EPM)
Hippocampal BDNF signaling↑
PNS activity↑
[73]
CGA
(Pure ingredients)
Male Wistar rats50 mg/kg (p.o.),
8 weeks
Average time to reach the
underwater platform↓
The average swimming distance↓ (MWM)
Light box dwell time↑ (LDB)
Number of marbles buried↓ (MBT)
MDA↓ BDNF↑ IL-1β↓
IL-2↓ TNF-α↓ INF-γ↓
[74]
FA
(Pure ingredients)
Male C57BL/6J mice20 mg/kg (p.o.)Sports activities↑ (OFT)
Immobility time↓ (FST)
Immobility time↓ (TST)
Sucrose consumption↑ (SPT)
Open arm dwell time and access
frequency↑ (EPM)
Firmicutes/Bacteroidetes ratio↑
5-HTP↑
[75]
SDsCaffeine
(Pure ingredients)
Male A1R knockout mice, A2AR knockout mice, wild-type C57BL/6 mice2.5, 5, 10, 15 mg/kg (i.p.), 3 hWake up time (5, 10, 15 mg/kg) ↑
NREM and REM↓
A2AR-mediated[76]
Caffeine
(Pure ingredients)
Wild-type
C57Bl6 mice
0.3, 0.6, 0.9 and 1.2 mg/mL (p.o.), 21 dResting phase wakefulness block↓
Quiescent phase↓
Active period↑ REM↓
Sleep delay↑
Fluctuation of adenosine
receptor antagonism
[77]
SDsCGA, CA and
caffeine
(Pure ingredients)
Male Wistar ratsCGA: 50, 100, 200, 500 mg/kg
CA:20, 50, 100, 200 mg/kg; caffeine: 1, 2, 5, 10 mg/kg, (p.o.), 35 d
Sleep latency↑
Wake up time↑
NREM↓
Activation of the
α1-AR system
[78]
Caffeine
(Pure ingredients)
C57BL/6 mice0.13, 0.26 g/kg (i.p.),
12 min
Level of spontaneous activity↓sympathetic nervous
system activity↑
[79]
well-beingCaffeinated and decaffeinated
coffee
(Coffee Extracts)
C57BL/6J mice1 g/L (p.o.),
3 weeks
Caffeinated coffee:
Males: Open arm dwell time ↓(EPM)
Upright behavior and climbing time ↑
Grooming time and social time↑
Number of buried glass spheres↑
Females: Selfcare ↑
Caffeinated: A1R Increased
in striatum (Males)
A1R increased in the ventral
hippocampus (Females)
Males and Females: MDA↓ GSH↑
[80]
Notes: “p.o.”: Per oral, “i.p.”: Intraperitoneal injection, “↓” represents downregulation, “↑” represents upregulation.
Table 2. Clinical studies on the effects of coffee and its active ingredients on depression, anxiety and SDs.
Table 2. Clinical studies on the effects of coffee and its active ingredients on depression, anxiety and SDs.
Mental DiseasesTypes of StudiesCoffee or Its Active IngredientsParticipantsDose and DurationResults and ConclusionsReferences
DepressionRandomized,
double-blind,
sham-controlled trial
caffeineN = 40
(age: 18–59 years)
Self-reported number of cups of coffee and
energy drinks consumed,
10–15 d
Antidepressant effect of
dorsomedial iTBS↑
Depression↓
[81]
Randomized,
double-blind,
crossover study
Caffeinated and decaffeinated coffeeN = 77
(mean age: 20.38 ± 1.28 years)
150 mg (caffeinated coffee), 1 dPost-treatment self-report
questionnaire scores↑
SBP and DBP↑ Depression↓
[82]
Randomized,
double-blind,
controlled trial
Caffeinated and decaffeinated coffeeN = 88
(age: 18–47 years)
75 mg/d (caffeinated coffee), 1 dStroop task accuracy↑
Card arrangement rewards
responsiveness and
sorting speed↑
Depressed mood↓
[83]
AnxietyRandomized,
double-blind,
controlled study
caffeineN = 56
(mean age: 69.2 years, female)
200 mg/d, 400 mg/d,
7 d
Anxiety symptoms↓
Low or moderate caffeine
may reduce anxiety
[86]
Randomized,
controlled,
crossover study
caffeineN = 16
(Caffeine withdrawal subjects)
100 mg/d, 1 dSBP and DBP↑
Anxiety symptoms↓
[87]
Prospective,
open-label trial
Feru-guardN = 20
(patients with
frontotemporal lobar
degeneration or dementia with Lewy bodies)
3 g/d (Feru-guard),
4 weeks
Neuropsychiatric Scale Total Score↓
Dementia, anxiety symptoms↓
May be effective for treating
frontotemporal lobar degeneration
or Lewy body
[88]
SDsRandomized,
double-blind,
controlled,
crossover study
CGAN = 16
(age: 30–54 years, Healthy male)
300 mg/d, 14 dAwakening fatigue↓
Sleep quality↑
Sleep quality↑
[89]
Randomized,
double-blind,
controlled,
crossover study
CGAN = 9
(mean age: 25.7 years, healthy adults)
600 mg/d, 5 dSleep latency↓
Parasympathetic activity↑
No adverse effect on sleep quality
[90]
Randomized,
double-blind,
controlled,
crossover study
caffeineN = 5
(mean age: 24.0 ± 2.8 years)
2.9 mg/kg, 49 dThe CR is delayed[91]
Randomized,
double-blind,
controlled,
crossover study
caffeineN = 15
(mean age: 23.7 ± 8.2 years)
6 mg/kg, 1 dsleep quality↓[92]
Randomized,
double-blind,
controlled,
crossover study
caffeineN = 20
(mean age: 26.4 ± 4 years)
450 mg/d, 9 dEEG power density↓
Nocturnal sleep structure or
subjective sleep quality
is not greatly affected
[93]
Notes: “↓” represents downregulation, “↑” represents upregulation.
Table 3. Epidemiological studies on the effects of coffee and its active ingredients on depression and anxiety.
Table 3. Epidemiological studies on the effects of coffee and its active ingredients on depression and anxiety.
Mental DiseasesParticipantsCoffee or Its Active
Ingredients
ConclusionsReferences
DepressionN = 9576
(age: ≥19 years)
Coffee and CaffeineRegular coffee or caffeine consumption is associated with lower
self-reported lifetime depression prevalence in adults
[94]
N = 50,739
(mean age: 63, female)
Caffeinated coffeeDepression risk decreases with increasing caffeinated coffee intake, with 2–3 cups being most effective[95]
N = 821
(Postpartum women)
Caffeinated coffeeModerate coffee consumption (1–2 cups per day) may reduce the risk of PDD, while higher consumption (3 cups per day or more) has no significant effect[97]
N = 80,497Black CoffeeModerate drinking of black coffee (4–6 times/week) may have a certain preventive effect on depression[98]
AnxietyN = 146,566
(from UK Biobank)
CoffeeModerate coffee consumption (1–4 cups/day) showed a trend of reducing the risk of anxiety[52]
188,355
(age: 37–73, from UK Biobank)
CoffeeFor individuals aged ≥ 60 years, higher coffee intake was negatively associated with anxiety disorders[99]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Shi, Z.; Luan, J.; Zhang, Y.; Wang, G.; Mei, C.; Chen, L.; Zhou, W.; Xiong, C.; Huang, T.; Zhan, J.; et al. Exploring the Impact and Mechanisms of Coffee and Its Active Ingredients on Depression, Anxiety, and Sleep Disorders. Nutrients 2025, 17, 3037. https://doi.org/10.3390/nu17193037

AMA Style

Shi Z, Luan J, Zhang Y, Wang G, Mei C, Chen L, Zhou W, Xiong C, Huang T, Zhan J, et al. Exploring the Impact and Mechanisms of Coffee and Its Active Ingredients on Depression, Anxiety, and Sleep Disorders. Nutrients. 2025; 17(19):3037. https://doi.org/10.3390/nu17193037

Chicago/Turabian Style

Shi, Zijun, Jin Luan, Yating Zhang, Guiping Wang, Can Mei, Linwanyue Chen, Weiji Zhou, Change Xiong, Tao Huang, Jianbo Zhan, and et al. 2025. "Exploring the Impact and Mechanisms of Coffee and Its Active Ingredients on Depression, Anxiety, and Sleep Disorders" Nutrients 17, no. 19: 3037. https://doi.org/10.3390/nu17193037

APA Style

Shi, Z., Luan, J., Zhang, Y., Wang, G., Mei, C., Chen, L., Zhou, W., Xiong, C., Huang, T., Zhan, J., & Cheng, J. (2025). Exploring the Impact and Mechanisms of Coffee and Its Active Ingredients on Depression, Anxiety, and Sleep Disorders. Nutrients, 17(19), 3037. https://doi.org/10.3390/nu17193037

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