Next Article in Journal
Dietary Interventions for Adults with Type 1 Diabetes: Clinical Outcomes, Guideline Alignment, and Research Gaps—A Scoping Review
Previous Article in Journal
Protein and Macronutrient Metabolism in Liver Cirrhosis: About Sarcopenia
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Gut Microbiome and Its Impact on Mood and Decision-Making: A Mechanistic and Therapeutic Review

by
Pierluigi Diotaiuti
*,
Francesco Misiti
,
Giulio Marotta
,
Lavinia Falese
,
Giovanna Elisa Calabrò
and
Stefania Mancone
Department of Human Sciences, Society and Health, University of Cassino and Southern Lazio, 03043 Cassino, Italy
*
Author to whom correspondence should be addressed.
Nutrients 2025, 17(21), 3350; https://doi.org/10.3390/nu17213350
Submission received: 15 September 2025 / Revised: 13 October 2025 / Accepted: 22 October 2025 / Published: 24 October 2025
(This article belongs to the Section Prebiotics and Probiotics)

Abstract

Background/Objectives: The gut microbiome is increasingly recognized as a key modulator of central nervous system function through the gut–brain axis. Dysbiosis has been associated with neuropsychiatric disorders such as depression, anxiety, impulsivity, cognitive decline, and addiction. This review aims to synthesize mechanistic insights and therapeutic perspectives on how gut microbiota influence mood regulation, decision-making, and cognitive processes. Methods: A comprehensive narrative review was conducted using peer-reviewed articles retrieved from PubMed, Scopus, and Web of Science up to August 2025. Studies were included if they explored microbiota-related effects on behavior, mood, cognition, or decision-making using human or animal models. Emphasis was placed on molecular mechanisms, microbiome-targeted therapies, and multi-omics approaches. Results: Evidence indicates that gut microbiota modulate neurochemical pathways involving serotonin, dopamine, GABA, and glutamate, as well as immune and endocrine axes. Microbial imbalance contributes to low-grade systemic inflammation, impaired neuroplasticity, and altered stress responses, all of which are linked to mood and cognitive disturbances. Specific microbial taxa, dietary patterns, and interventions such as probiotics, prebiotics, psychobiotics, and fecal microbiota transplantation (FMT) have shown promise in modulating these outcomes. The review highlights methodological advances including germ-free models, metagenomic profiling, and neuroimaging studies that clarify causal pathways. Conclusions: Gut microbiota play a foundational role in shaping emotional and cognitive functions through complex neuroimmune and neuroendocrine mechanisms. Microbiome-based interventions represent a promising frontier in neuropsychiatric care, although further translational research is needed to define optimal therapeutic strategies and address individual variability.

1. Introduction

The human microbiome, comprising bacteria, viruses, fungi, and archaea, forms a mutually beneficial ecosystem with the host, supporting essential physiological functions such as metabolism, immune regulation, intestinal barrier integrity, and neurophysiological signaling through dense trophic and molecular communication networks [1,2,3,4,5,6,7,8,9,10,11,12,13,14,15,16]. Throughout this review, we explicitly name representative probiotic strains, e.g., Lactobacillus rhamnosus GG, Lactobacillus plantarum PS128, Bifidobacterium longum 1714, and Saccharomyces boulardii, alongside key prebiotic substrates such as inulin-type fructans, galacto-oligosaccharides (GOS), resistant starch, arabinoxylans, and β-glucans, referencing exemplar clinical and mechanistic studies where relevant. Key commensals with well-described beneficial effects include Faecalibacterium prausnitzii (a major butyrate producer with anti-inflammatory activity), Akkermansia muciniphila (supports mucus-layer integrity and metabolic homeostasis), Lactobacillus rhamnosus GG and Lactobacillus plantarum PS128 (psychobiotic strains linked to improvements in stress, mood, and performance), and Clostridium butyricum (butyrate-mediated neuroprotection); in addition, Lactobacillus reuteri has emerged as a promising target for mechanistic clinical trials in the gut–brain axis [12,17,18,19,20].
In particular, the gut microbial community contributes to immune cell homeostasis, enteric nervous system regulation, angiogenesis, vitamin synthesis, energy balance, xenobiotic metabolism, and pathogen exclusion [1,2,4,6,9,10,11,12,15]. When this complex ecosystem becomes imbalanced, commonly referred to as dysbiosis, it has been linked to conditions such as non-alcoholic fatty liver disease and inflammatory bowel disorders [21,22,23].
Through the gut–brain axis, microbial states correlate closely with central nervous system (CNS) signaling pathways that influence mood, cognition, and behavior [12,13,14,15,24,25,26]. Gut microbes actively modulate host neurotransmitter systems, including serotonin and dopamine, which are critically involved in emotional regulation and psychological well-being. It also contributes to the detoxification of xenobiotics, modulation of bile acids, and synthesis of neuroactive metabolites that influence both local and systemic physiology [27,28,29,30,31,32].
Imbalances in gut microbiota composition have been associated with irritability and mood instability, often mediated by cytokine signaling that reduces tryptophan availability and serotonin synthesis [33,34,35]. Diet-induced microbiota changes also affect lipid and energy metabolism, with downstream effects on emotional states [36,37,38,39,40]. Bidirectional gut–brain communication influences brain development, behavior, and cognitive performance across the lifespan [24,25,26].
Increasing evidence also connects gut dysbiosis with cognitive deficits and impaired decision-making, through mechanisms involving neurotransmitter imbalances, neuroinflammation, and metabolic disruptions [41,42,43,44,45,46,47,48,49]. Both human and preclinical studies suggest that microbial diversity is positively linked to cognitive function in aging, with Mediterranean dietary patterns and targeted supplementation (e.g., prebiotics, probiotics) showing particular promise [44,45,46,49,50,51].
Alterations in the gut microbiome have been implicated in diabetes-related cognitive changes and forms of dementia, with emerging attention to the role of the gut mycobiome [52,53,54,55].

Scope and Rationale

Against this backdrop, numerous studies have begun to examine the relationship between microbial composition and physiological domains such as glucose regulation, systemic inflammation, intestinal barrier function, adipose metabolism, bile-acid and hormonal signaling, and xenobiotic biotransformation. These systems intersect with central neural circuits involved in stress, reward, and emotional processing [10,11,12,13,14,15]. Despite a rapidly expanding literature, current reviews seldom provide a mechanism-anchored synthesis that links named probiotic strains and defined prebiotic substrates to CNS-relevant human outcomes (stress, mood, attention/cognition, performance) through clearly articulated pathways (SCFAs/butyrate, barrier integrity, bile-acid and tryptophan metabolism, vagal/HPA and neuroimmune signaling). Evidence is often summarized at the genus/species level, blurring strain specificity and dosing/duration considerations, while heterogeneity in outcomes, small samples, short interventions, and incomplete control of modifiers (dietary fiber, baseline microbiome, sleep/circadian factors, medications, physical activity) limit comparability and translation. Mapping from preclinical signals to human endpoints and integrating emerging domains such as decision-making and attentional bias remain underdeveloped, with little emphasis on precision (who responds, under what conditions). To address these gaps, this review offers a named-agent, mechanism-first synthesis, explicitly connecting key strains (e.g., LGG, PS128, B. longum 1714, S. boulardii) and substrates (inulin-type fructans, GOS, RS2/RS3, arabinoxylans, β-glucans) to CNS-relevant outcomes while appraising study quality and sources of heterogeneity. For comprehensive overviews and background perspectives on microbiome–brain interactions, readers may also consult recent reviews and position papers [56,57,58,59]. This review seeks to integrate mechanistic and therapeutic perspectives on the gut microbiome’s impact on mood and cognition. Particular emphasis is placed on methodological quality, translational challenges, and future directions to help bridge the gap between basic research and clinical applications.

2. Materials and Methods

This narrative review was conducted by synthesizing evidence from peer-reviewed literature to explore the mechanisms through which the gut microbiome modulates mood, cognition, and decision-making. A comprehensive literature search was carried out across major academic databases including PubMed, Scopus, and Web of Science. The search included articles published from 1 January 2000 to 31 August 2025 and used keywords such as gut–brain axis, gut microbiota, mood disorders, cognitive function, neuroinflammation, psychobiotics, and fecal microbiota transplantation. In data extraction we systematically tracked named probiotic strains and prebiotic substrates (when reported at strain/substrate level) and recorded their primary putative mechanisms and outcome domains to harmonize terminology across sections. Both preclinical (animal model) and clinical (human subject) studies were considered, with particular emphasis on mechanistic investigations and therapeutic interventions. To enhance transparency, we summarize the study selection process in a PRISMA-style flow diagram (Figure 1), including numbers identified, screened, excluded with reasons, and included in the narrative synthesis. We followed a PRISMA 2020-style flow [60] to enhance transparency of the study selection process, while conducting a narrative (non-systematic) review without meta-analysis. Screening proceeded in two stages: first, titles and abstracts were screened to remove clearly ineligible records; second, full texts were retrieved and assessed for eligibility, with reasons for exclusion documented (e.g., not CNS-relevant outcomes, no mechanistic link, in vitro/cell-line only, methodological limitations, other). We excluded in vitro and cell-line-only studies from screening and data extraction because they do not capture whole-organism interactions required for CNS-relevant outcomes (e.g., neuroendocrine, immune, vagal/HPA pathways). Findings from in vitro work were considered only as narrative mechanistic background when directly supportive of in vivo pathways; they were not counted as included records and were not part of the PRISMA flow.
Inclusion criteria comprised (1) studies examining gut microbiota influence on neuropsychological or behavioral outcomes; (2) use of multi-omics technologies (e.g., metagenomics, metabolomics, metaproteomics); (3) intervention studies involving probiotics, prebiotics, psychobiotics, or dietary manipulation; (4) publications in English from peer-reviewed journals. Exclusion criteria included (1) non-peer-reviewed articles; (2) opinion pieces lacking empirical data; and (3) studies focused solely on gastrointestinal outcomes unrelated to neurobehavioral effects.
The selected literature was critically appraised for methodological rigor, sample size, reproducibility, and translational relevance. Special consideration was given to studies that incorporated causality models (e.g., germ-free animal designs, fecal transplantation) and those using integrated omics or neuroimaging to elucidate host–microbiome interactions. No formal meta-analysis was performed due to heterogeneity in methodologies, outcome measures, and microbial assessment techniques across studies.

3. Results

Gut microbes are known to synthesize a range of neuroactive compounds, such as gamma-aminobutyric acid (GABA), and to modulate immune–endocrine mediators that are highly relevant to mood disorders including depression and anxiety. These effects may be mediated by low-grade inflammation and reduced tryptophan availability, which limit central serotonin synthesis and alter emotional regulation [61,62,63,64,65,66,67,68,69,70]. Microbial imbalances, particularly dysbiosis, have been implicated in the disruption of sleep patterns and circadian rhythms, thereby amplifying symptoms related to chronic stress and emotional instability [64,65,66,69,70,71]. Taken together, this body of evidence supports the notion that the gut microbiome is a critical modulator of neurochemical activity and emotional regulation, playing a central role in the etiology of mood disorders [67,68,69,70].
An overview of key agents is provided in Box 1, with detailed mechanisms and outcomes summarized in Table 1 below.
Box 1. Key probiotics and prebiotics referenced in this review.
Probiotics (exemplar strains): Lactobacillus rhamnosus GG (ATCC 53103); Lactobacillus plantarum PS128; Bifidobacterium longum 1714; Saccharomyces boulardii.
Prebiotics (exemplar substrates): Inulin-type fructans; Galacto-oligosaccharides (GOS); Resistant starch (types 2–3); Arabinoxylans; β-glucans.
Principal mechanisms (overview): Short-chain fatty acid generation (notably butyrate), reinforcement of epithelial barrier and mucus layer, bile-acid and tryptophan pathway modulation, immune signaling, and vagal/neuroendocrine interfaces.
Importantly, the influence of the gut microbiota is not confined to the gastrointestinal tract. Instead, microbial communities exert systemic effects on distant organs such as the lungs, heart, and liver, as well as on the central nervous system, via endocrine, immune, and neural pathways [17,72,73,74,75]. A compelling illustration of this systemic interplay is offered by fecal microbiota transplantation (FMT), which has demonstrated causal influence on host phenotypes and is currently being explored for its potential impact on neurobehavioral conditions, including Alzheimer’s disease and inflammatory pain syndromes [76,77,78,79,80]. These associations also extend to the renal system, particularly within the framework of the gut–kidney axis, where microbial states have been linked to chronic kidney disease progression [81,82].
From a mechanistic perspective, microbial pathways play a vital role in the synthesis and regulation of neurochemicals such as GABA, serotonin, dopamine, and their respective precursors. These metabolites influence receptor expression and CNS signaling, particularly via vagal pathways and modulation of precursor bioavailability [83,84,85,86,87,88,89,90,91]. Notably, experimental studies using germ-free animal models have revealed profound shifts in fecal and serum neurotransmitter levels, underscoring the foundational role of microbiota in setting neurochemical baselines [87,88,89,90]. In addition, emerging research has begun to investigate the immunomodulatory effects of gut microbes in the context of psychiatric disorders such as schizophrenia, further broadening the relevance of microbiota to mental health [92].
A substantial body of literature has also established a link between low-grade systemic inflammation and mood disorders, including depression and bipolar disorder. These conditions frequently present with disruptions in glutamatergic signaling, and it is noteworthy that many psychotropic medications exhibit ancillary anti-inflammatory properties [93,94,95,96,97,98,99]. Such findings highlight plausible biological bridges connecting dysbiosis, immune dysfunction, and affective symptomatology.
The microbiota–gut–brain axis represents a highly integrated system that includes enteroendocrine signaling, neural pathways (notably involving the vagus nerve), and immune communication [100,101,102,103,104,105,106]. Dysbiosis within this axis has been proposed not only as a biomarker but also as a modifiable therapeutic target across a range of neurological and psychiatric disorders, including Alzheimer’s disease, Parkinson’s disease, irritable bowel syndrome (IBS)-related comorbidities, and bipolar disorder. Mechanistic pathways likely involve toll-like receptor 4 (TLR4) activation, lipopolysaccharide (LPS)-mediated signaling, intestinal barrier integrity, and neuroimmune crosstalk [101,102,103,104,105].

3.1. Effects on Decision-Making and Cognition

Beyond descriptive associations, converging evidence supports causal routes by which gut-derived signals may shape decision-making processes. First, short-chain fatty acids (SCFAs), notably butyrate, modulate microglial tone, synaptic plasticity, and blood–brain barrier integrity, with downstream effects on prefrontal efficiency and cognitive control; these pathways provide a biological bridge from fiber-fermenting prebiotics (e.g., inulin-type fructans, GOS, RS2/RS3) and butyrogenic commensals to performance on attention/executive tasks and reduced negative attentional bias in humans [51,52,53,54,55,61,62,63,64,65,78,79,80,81,82,83,84,85,86]. Second, tryptophan metabolism links the microbiome to central serotonergic and kynurenine pathways that influence valuation, impulsivity, and flexibility; in parallel, bile-acid signaling via FXR/TGR5 interfaces with dopaminergic and energy-balance networks relevant to frontostriatal decision systems [78,79,80,81,82,83,84,85,86,93,94,95,96,97,98,99]. Third, vagal afferents and HPA-axis dynamics transmit interoceptive and stress signals that bias risk/effort trade-offs and executive control, offering a plausible route for psychobiotic strains (e.g., Lactobacillus rhamnosus GG, L. plantarum PS128, Bifidobacterium longum 1714, Saccharomyces boulardii) to influence perceived stress, attentional control, and task engagement [12,17,18,19,20,100,101,102,103,104,105,106]. Immune–inflammatory mediators (e.g., low-grade cytokine activity) alter corticostriatal function and reward processing, providing a mechanistic rationale for observed links between microbiome modulation, inflammation lowering, and shifts in decision-relevant behaviors (e.g., vigilance, cognitive persistence) [78,79,80,81,82,83,84,85,86,93,94,95,96,97,98,99,100,101,102,103,104,105,106]. Taken together, these pathways outline a mechanism-anchored model in which named strains and defined prebiotic substrates (see Box 1 and Table 1) target tractable biological levers that align with task-level outcomes central to decision-making and cognition in humans [12,17,18,19,20,51,52,53,54,55,61,62,63,64,65].

3.2. Influence on Cognitive Function

Molecular routes link microbes to cognition. Converging evidence indicates that short-chain fatty acids (SCFAs), especially butyrate, promote synaptic plasticity, microglial homeostasis, and blood–brain barrier integrity, supporting attention/executive performance and memory; these effects mechanistically connect butyrogenic commensals (e.g., Faecalibacterium prausnitzii, Clostridium butyricum) and prebiotics that drive SCFA production (inulin-type fructans, GOS, RS2/RS3, arabinoxylans, β-glucans) to human cognitive endpoints [51,52,53,54,55,61,62,63,64,65,78,79,80,81,82,83,84,85,86].
In parallel, tryptophan–serotonin/kynurenine and bile-acid (FXR/TGR5) pathways interface with frontostriatal dopamine and cortical networks relevant to working memory, flexibility, and valuation, while vagal/HPA-axis signaling and immune–inflammatory tone (low-grade cytokines) further bias cognitive control and processing efficiency [78,79,80,81,82,83,84,85,86,93,94,95,96,97,98,99,100,101,102,103,104,105,106]. Several taxa/strains modulate neurotransmitters (e.g., GABA, glutamate) and stress biology in ways consistent with cognitive benefits: L. rhamnosus GG, L. plantarum PS128, B. longum 1714, and S. boulardii; Akkermansia muciniphila (barrier/mucus layer) and emerging L. reuteri add mechanistic plausibility via barrier and neuroimmune pathways [12,17,18,19,20,100,101,102,103,104,105,106]. These routes align with the named-agent synthesis in Box 1/Table 1 and are consistent with neuroimaging and multi-omics links between microbial features and brain networks underlying cognition [20,100,101,102,103,104,105,106,107,108,109,110,111]. Additional cohort- and review-level evidence complements these findings and frames cognitive outcomes within broader microbiome–behavior links [112].
The pioneering work of Foster & McVey Neufeld [107] highlights the gut–brain axis’s role in regulating anxiety and depression, setting the stage for a broader understanding of the microbiome’s impact on higher cognitive functions such as reasoning, attention, memory, decision-making, learning, and problem-solving. Mayer et al. [108] further cement the paradigm shift in neuroscience by emphasizing the gut–brain axis’s significant influence on cognitive function.
Recent neuroimaging findings by Liu et al. [20] update our understanding of the gut microbiota’s interaction with cognitive function, while Liang et al. [109] integrate multi-omics data to elucidate the gut microbiome’s effects on cognitive function and brain structure. The regulation of Alzheimer’s disease pathologies and cognitive disorders through gut microbiota-mediated neuroinflammation, as demonstrated by Bairamian et al. [110], marks a critical advance in our understanding of neurodegenerative diseases. Wasser et al. [111] explore the associations between gut microbiota and cognitive performance in Huntington’s disease, offering insights into clinical outcomes.
The connection between Western diet consumption, the microbiome, and cognitive impairment presented by Noble et al. [42] illustrates the nutritional and lifestyle factors contributing to gut–brain dysbiosis. Smith et al. [113] introduce an intriguing association between gut microbiome diversity and sleep physiology, suggesting a broader physiological relevance of the microbiome. The emerging field of neuromicrobiology, as proposed by Miri et al. [114], and the insights into the microbiome’s effect on synaptic plasticity expand the scope of microbiome research into neurometabolic processes and synaptic malleability [115].
Investigations into specific interventions, such as the impact of American Ginseng on mood and cognition [116] and the psychological benefits of LGG probiotic supplementation [18], offer promising evidence of modulating the gut microbiome to enhance cognitive and psychological health. Also, the relationship between wine consumption, diet, and microbiome modulation in Alzheimer’s disease [51] opens a novel avenue for dietary interventions in neurodegenerative disease management.
This body of research provides compelling evidence that the gut microbiome significantly influences brain function and structure, cognitive performance, and neurological health outcomes. It underscores the potential of microbiome-targeted interventions as therapeutic strategies for cognitive disorders, psychological status, and overall brain health, marking a significant shift in our approach to neuroscience and mental health.

3.3. Impact on Risk Assessment

In this review, risk assessment denotes decision-making under risk, i.e., choices among options with explicit, known probabilities and outcome magnitudes, and, where noted, decision-making under ambiguity, where probabilities are only partially specified. Empirical work operationalizes these constructs through lottery/expected-value tasks, probability-discounting paradigms, and the Balloon Analogue Risk Task (BART) for risk-taking; the Iowa Gambling Task (IGT) primarily indexes decision-making under ambiguity rather than pure risk. Within this framework, signals attributed to psychobiotic strains (e.g., Lactobacillus rhamnosus GG, L. plantarum PS128, Bifidobacterium longum 1714, Saccharomyces boulardii) and prebiotics (inulin-type fructans, GOS, RS2/RS3, arabinoxylans, β-glucans) are interpreted via mechanism-anchored pathways, notably SCFA/butyrate effects on microglia and synaptic plasticity, barrier integrity, tryptophan–serotonin and bile-acid (FXR/TGR5) signaling with downstream dopaminergic influences, and vagal/HPA–immune interfaces that modulate arousal and stress responsivity, each of which can shift risk preference, probability weighting, and choice consistency on these tasks [12,17,18,19,20,51,52,53,54,55,61,62,63,64,65,78,79,80,81,82,83,84,85,86,93,94,95,96,97,98,99,100,101,102,103,104,105,106]. Accordingly, where we report changes in “risk taking” we refer specifically to task-level indices under known or partially known probabilities rather than generic behavioral descriptors; results are cross-referenced to Box 1/Table 1 to maintain traceability to the underlying named agents and mechanisms [12,17,18,19,20,51,52,53,54,55,61,62,63,64,65].
Fecal microbiome analysis has identified decreased abundance of Firmicutes, specifically Faecalibacterium, in patients presenting with bipolar disorder, which also correlated with self-reporting symptom severity, indicating a potential link between gut microbiome composition and psychiatric symptoms [50]. Studies have demonstrated that the gut microbiome influences dopamine levels in the frontal cortex and striatum in rodents, brain areas involved in executive functions, suggesting a potential mechanism through which the microbiome may impact risk-related behaviors [117]. Other findings suggest that targeting the microbiome may represent a novel approach for treating neurodevelopmental disorders such as autism, further emphasizing the potential role of the gut microbiome in shaping behaviors and psychiatric conditions [118]. Animal studies have also shown that the presence of the gut flora in rodents may impose behavioral changes with wider implications for psychiatric disorders in humans [119]. The gut microbiome has been implicated in various psychiatric conditions, including major depressive disorder, bipolar affective disorder, autism spectrum disorder, anxiety disorders, schizophrenia, and psychotic disorders, highlighting the broad impact of the microbiome on mental health [63].
Microbiome has been associated with metabolic syndrome, obesity, diabetes, dyslipidemia, and hypertension, suggesting a potential link between the microbiome and physiological factors that may contribute to psychiatric disorders [120,121]. Taken together, the pattern suggests modest, mechanism-consistent shifts in risk-sensitive choice, most plausibly mediated by stress/HPA attenuation, inflammation lowering, and serotonergic/dopaminergic pathway engagement, rather than a uniform change in “riskiness” per se, reinforcing the need for standardized, probability-explicit tasks in future trials [78,79,80,81,82,83,84,85,86,93,94,95,96,97,98,99,100,101,102,103,104,105,106].

3.4. Connection to Impulsive Behavior

In this review, impulsivity refers to a multidimensional tendency toward premature or poorly controlled action and can be conceptualized as both a trait (stable, dispositional) and a state (stress- or context-sensitive). We consider three principal facets that are commonly quantified in human and preclinical work: (1) motor impulsivity (deficits in response inhibition, typically assessed with go/no-go or stop-signal tasks); (2) choice impulsivity (a preference for smaller-sooner over larger-later rewards, measured via delay-discounting paradigms); and (3) reflection impulsivity (reduced information gathering before choice). This construct is distinct from risk-taking, which concerns choices under known or partially known probabilities (treated separately in our “Risk Assessment” section). Within this framework, gut-derived pathways—SCFA/butyrate effects on plasticity and microglial tone, tryptophan–serotonin/kynurenine and bile-acid signaling (FXR/TGR5) with downstream dopaminergic influences, and vagal/HPA–immune interfaces affecting arousal and stress responsivity—offer biologically plausible routes by which named probiotics (e.g., LGG, PS128, B. longum 1714, S. boulardii) and defined prebiotics (inulin-type fructans, GOS, RS2/RS3, arabinoxylans, β-glucans) could modulate task-level indices of impulsivity [12,17,18,19,20,51,52,53,54,55,61,62,63,64,65,78,79,80,81,82,83,84,85,86,93,94,95,96,97,98,99,100,101,102,103,104,105,106,122,123,124,125,126,127,128,129,130,131,132,133,134]. Broader syntheses and related primary studies further contextualize impulsivity-related outcomes within microbiome research [135,136,137,138,139,140,141].
The relationship between the gut microbiome and impulsive behavior is an area of growing interest in the field of neuroscience and psychology. Several studies have highlighted the potential impact of the gut microbiome on cognitive functions, decision-making, and impulsive behaviors [122,123,124,125,126,127,128,129]. The gut microbiome has been shown to influence various aspects of behavior, including impulsivity, attention, reward-learning, and locomotor response to novelty [130]. Alterations in the gut flora have been correlated with increased impulsivity and vulnerability to substance use disorders [131]. These findings suggest a potential link between the gut microbiome and impulsive behavior. Research has also indicated that the gut microbiome may covary with the processing in frontoparietal circuits associated with cognitive control and self-regulation, which are essential for regulating impulsive behaviors [132]. Studies in animal models have suggested that changes in the microbiome can be associated with alterations in cognition and behavior, including impulse transmission along the vagus nerve, which connects the intestinal tract and the central nervous system [133].
The microbiome has been implicated in the modulation of mood and affect, which are closely related to impulsive behavior [129]. Traits associated with sensitivity to drug stimuli in animals have been linked to alterations in the microbiome, further emphasizing the potential role of the gut microbiome in impulsive behaviors [134].
The potential impact of the gut microbiome on impulsive behavior has significant implications for various fields, including psychiatry, neurology, and addiction medicine. Understanding the mechanisms through which the gut microbiome influences impulsive behavior could lead to novel therapeutic interventions targeting the microbiome to modulate impulsive tendencies and improve cognitive control.

3.5. Role in Addiction and Substance Abuse

Several reviews and primary studies converge on the microbiome’s contribution to reward processing and substance-related phenotypes, offering frameworks that complement the targeted reports summarized below [135,136,137,138,139,140,141,142,143,144,145,146,147]. Forouzan, McGrew, and Kosten [148] delve into the relationship between negative affect, psychostimulant use and withdrawal, and the microbiome, providing insights into how disruptions in gut flora may exacerbate psychological distress associated with addiction. García-Cabrerizo et al. [130] further elucidate the microbiota–gut–brain axis’s regulatory capacity on reward processes, suggesting that gut microbiota may influence the brain’s reward circuitry, thereby affecting addictive behaviors. Russell et al. [149] review the gut microbiome’s role in substance use disorder, proposing that alterations in gut flora could influence the predisposition to and outcomes of addiction treatments. Cryan et al. [50] provide a comprehensive overview of the microbiota–gut–brain axis, emphasizing its significance in brain diseases and, implicitly, in the modulation of behaviors related to addiction. Research by Browning et al. [150] on the potential role of the gut microbiome in substance use disorders highlights the connection between gut dysbiosis, illicit drug use, and the immune system’s role in addiction. Similarly, studies like that of Molavi et al. [151] investigate the effects of probiotic supplementation on opioid-related disorders, suggesting that modifying the gut microbiota could offer therapeutic benefits in addiction treatment. Emerging research, such as that by Fu et al. [152], points to the microbiome–gut–brain axis as a promising therapeutic target for substance-related disorders, indicating that interventions aimed at restoring gut microbial balance may mitigate addiction behaviors. Studies like that of Ren & Lotfipour [153] show that the gut microbiota composition can vary significantly with opioid use, impacting circulating levels of hormones such as leptin and oxytocin, which are known to influence addiction pathways. The role of diet in modulating the gut microbiota and, consequently, addiction behaviors cannot be overstated, as evidenced by Yang et al. [154], who explore the impact of dietary nutrients on gut flora. This line of research suggests that nutritional interventions could alter the gut microbiome in a manner that supports addiction recovery processes.
The literature underscores the gut microbiome’s profound influence on addiction behaviors through various pathways, including the modulation of the gut–brain axis, immune responses, and dietary impacts on gut flora. These findings pave the way for innovative approaches to addiction treatment, emphasizing the potential of microbiome-targeted therapies to complement traditional interventions. The complexity of the microbiome’ s role in addiction underscores the need for further research to unravel the intricate mechanisms by which the gut influences brain health and behavior, offering hope for more effective and personalized addiction treatment strategies.

3.6. Factors Shaping Microbiome Composition

The composition and functional diversity of the gut microbiome are highly dynamic and sensitive to a variety of internal and external influences. Among the most powerful of these are dietary patterns, which exert profound effects on microbial populations, modulating both inflammatory tone and overall disease risk. Diets rich in plant-based foods and dietary fibers, such as fruits, vegetables, legumes, and whole grains, have consistently been associated with favorable outcomes in terms of metabolic health and neurocognitive performance [37,40]. In contrast, diets low in fiber and high in refined sugars and saturated fats tend to promote dysbiosis and inflammation. Importantly, exposures during early life, including to allergens, nutritional components, and microbial environments, can shape the long-term trajectory of the gut microbiome, with lasting implications for health and behavior [155,156,157].
Beyond nutrition, psychological and pharmacological stressors represent additional key modulators of gut microbial ecology. Chronic stress, traumatic experiences, and the use of certain psychotropic or somatic medications can induce shifts in microbiota composition, leading to downstream effects on pain sensitivity, metabolic processes, and mental health across the lifespan [158,159,160,161,162,163,164,165]. Notably, early-life stress has been shown to exert long-term programming effects on both the microbiome and behavioral outcomes, reinforcing the concept of a sensitive developmental window for host–microbe interactions [161,162,163].
Pharmacological agents, particularly antibiotics but also non-antibiotic drugs, can significantly perturb microbial ecosystems too. Such disturbances often result in the depletion of beneficial taxa and reduced microbial stability. In experimental models, these ecological disruptions have even been causally linked to increased mortality, highlighting the critical role of microbial homeostasis for host resilience [166,167,168].
Another factor that influences microbiota composition is physical activity, especially when performed at high intensity. While moderate exercise can have beneficial effects, excessive exertion may transiently increase systemic inflammation and compromise intestinal barrier function, a phenomenon often referred to as “leaky gut”. However, this effect can be mitigated through targeted nutritional strategies and probiotic supplementation, which help buffer oxidative stress and promote adaptive recovery responses. Also, disruption of circadian rhythms, including irregular sleep patterns, has been shown to alter microbial diversity and composition, with downstream effects on hormonal regulation and emotional reactivity [19,169,170,171,172].
These findings underscore the multifactorial nature of microbiome regulation, suggesting that individualized interventions targeting diet, stress management, medication use, and physical activity could offer integrated strategies to promote gut and brain health.

3.7. Methods to Study Microbiome–Mood Links

The investigation of microbiome–brain interactions has benefited immensely from recent advances in multi-omics technologies, which allow researchers to unravel complex structure–function relationships within microbial ecosystems. Among the most widely used techniques are 16S rRNA gene sequencing, shotgun metagenomics, metatranscriptomics, metaproteomics, and metabolomics. These tools provide detailed insights into the taxonomic composition, gene expression patterns, protein synthesis, and metabolic profiles of gut microbiota [173,174,175,176,177,178,179,180,181,182].
To make sense of the massive and multidimensional datasets generated by these approaches, researchers increasingly rely on compositional data analysis techniques and machine learning models that can uncover hidden patterns and functional associations. In particular, network-based approaches and Gaussian process models are proving valuable in capturing dependencies across multiple omics layers, such as transcript–metabolite correlations or protein–taxon relationships [183,184]. However, for findings to be replicable and translatable, the standardization of analytical pipelines remains a critical priority. Certified workflows, quality control metrics, and harmonized data formats are essential for achieving cross-study comparability and advancing the field [185,186].
In addition to human cohort studies, animal models continue to serve as indispensable platforms for establishing causal relationships between microbial colonization patterns and behavioral phenotypes. Experimental manipulations, such as germ-free environments, microbial transfers, or selective antibiotic exposure, enable researchers to directly link microbiota configurations to changes in neurotransmitter levels, particularly serotonin and GABA, and corresponding alterations in affective and cognitive behavior [90,187,188].
While animal models provide critical insights into mechanistic pathways, translating these findings to human populations requires caution due to interspecies differences in microbial architecture, neurodevelopment, and behavior. On the human side, researchers often evaluate the impact of interventions such as antibiotics, prebiotics, probiotics, and controlled diets on emotional well-being and cognitive performance. These interventions are complemented by emerging strategies such as remote cognitive testing, which enhances the feasibility of studying neuropsychological outcomes even in vulnerable or hard-to-reach populations, including individuals with severe mental illness [189,190,191,192,193].

3.8. Therapeutic Approaches

Growing evidence supports the use of microbiome-targeted therapies as promising adjuncts in the treatment of mood and cognitive disorders. This includes not only mood and anxiety disorders but also emerging indications such as autism spectrum conditions, multiple sclerosis, and Parkinson’s disease, where preliminary evidence suggests that microbiome-based interventions may modulate disease trajectory. Among these, probiotics, defined as live microorganisms that confer health benefits to the host, have shown the capacity to modulate the gut–brain axis, particularly in relation to stress biology and emotional regulation. Clinical trials have reported improvements in mood and anxiety symptoms following probiotic or prebiotic supplementation, underscoring their potential role as safe and accessible interventions. For clarity, examples cited throughout include probiotics such as Lactobacillus rhamnosus GG, L. plantarum PS128, Bifidobacterium longum 1714, and Saccharomyces boulardii, and prebiotics such as inulin/GOS, resistant starch, arabinoxylans, and β-glucans (WHO; [194,195,196,197,198,199,200,201,202,203,204,205,206,207,208,209,210]). Also, fermented foods, which naturally contain probiotic strains, may serve as complementary strategies within broader dietary patterns.
Another therapeutic avenue is fecal microbiota transplantation (FMT), which involves the introduction of gut microbiota from a healthy donor to a recipient via endoscopic delivery or oral capsules. While FMT is already established as an effective treatment for recurrent Clostridioides difficile infection, it is now being actively investigated for its potential impact on neuropsychiatric and neurodegenerative conditions. Preliminary studies suggest promising outcomes, although further research is needed to clarify mechanistic pathways, long-term effects, and safety protocols [194,211,212,213].
In parallel, dietary composition, particularly the balance of macronutrients and availability of fermentable substrates, plays a critical role in shaping the gut microbiota and influencing host physiology. Adjusting diet can alter microbial metabolism, reduce inflammatory markers, and impact emotional states. Importantly, aligning dietary strategies with cultural and individual preferences improves adherence and enhances psychological outcomes, thereby maximizing therapeutic potential [195,196,197].
Emerging within this context is the concept of psychobiotics, a class of probiotics and prebiotics specifically selected for their impact on central nervous system function. Randomized controlled trials (RCTs) have documented psychobiotic-induced reductions in cortisol levels, perceived stress, and depressive symptoms, suggesting that these agents may serve as low-risk adjuncts or, in some cases, alternatives to conventional treatments [198,199,200,201,202,203,204,205]. However, for psychobiotics to gain broader clinical acceptance, challenges related to regulatory approval, quality assurance, and strain-specific efficacy must be systematically addressed.

4. Discussion

Across the evidence base reviewed, the most consistently studied probiotics include Lactobacillus rhamnosus GG, L. plantarum PS128, Bifidobacterium longum 1714, and Saccharomyces boulardii, while prebiotics most frequently linked to beneficial stress/mood and cognition-relevant outcomes include inulin-type fructans, GOS, resistant starch (types 2–3), arabinoxylans, and β-glucans [12,17,18,19,20,51,52,53,54,55,61,62,63,64,65]. Mechanistically, convergent pathways span short-chain fatty acid (SCFA) production, especially butyrate-barrier reinforcement, bile-acid and tryptophan signaling, immune tone modulation, and vagal/HPA-axis interfaces [78,79,80,81,82,83,84,85,86,93,94,95,96,97,98,99,100,101,102,103,104,105,106]. Taken together, these findings suggest strain- and substrate-specific effects rather than a generic “microbiome” signal, with small-to-moderate, population-dependent benefits most consistently observed for perceived stress, mood indices, and selected attentional/cognitive endpoints [51,52,53,54,55,61,62,63,64,65].
Most important signals from human studies: Trials using LGG and PS128 report reductions in perceived stress and inflammatory markers, with PS128 additionally showing benefits in performance/recovery contexts; B. longum 1714 demonstrates stress-attenuating and attentional effects in human experimental paradigms. For prebiotics, GOS shows the clearest human signal for cortisol modulation and reduced negative attentional bias in young adults, whereas inulin-type fructans and resistant starch (RS2/RS3) consistently act as bifidogenic/butyrogenic substrates that translate into improvements in stress proxies, inflammatory/metabolic markers, and GI function, all domains plausibly connected to affect and cognition [12,17,18,19,20,51,52,53,54,55,61,62,63,64,65,78,79,80,81,82,83,84,85,86]. These agent-specific profiles align with the synthesis presented in Box 1 and Table 1 in the Results.
Mechanistic coherence and translational plausibility: Across agents, the most plausible molecular levers for cognition are butyrate-centric SCFA signaling (epigenetic/BDNF-linked plasticity; BBB/microglia), tryptophan–serotonin/kynurenine and bile-acid crosstalk (with downstream effects on dopamine and cortical–striatal networks), vagal/HPA interfaces (stress responsivity and arousal), immune–inflammatory recalibration, and GABA/glutamate modulation. These pathways provide a coherent rationale for the cognitive signals seen with L. rhamnosus GG, L. plantarum PS128, B. longum 1714, and S. boulardii, and with prebiotics that increase butyrate (inulin-type fructans, GOS, RS2/RS3, arabinoxylans, β-glucans); commensals such as F. prausnitzii and Akkermansia contribute via butyrate production and barrier reinforcement [12,17,18,19,20,51,52,53,54,55,61,62,63,64,65,78,79,80,81,82,83,84,85,86,93,94,95,96,97,98,99,100,101,102,103,104,105,106]. Human neuroimaging and multi-omics studies add convergent validity by linking microbiome features to attention and decision-making networks, reinforcing a mechanism-anchored interpretation of cognitive findings in this review [20,100,101,102,103,104,105,106,107,108,109,110,111]. Evidence linking butyrate-centric metabolism and barrier integrity to improved affective outcomes is supported by studies showing shifts in neurotransmitter precursors and neuroimmune mediators (e.g., tryptophan/serotonin availability, cytokines) and by emerging work implicating vagal/HPA-axis signaling in stress responsivity [78,79,80,81,82,83,84,85,86,93,94,95,96,97,98,99,100,101,102,103,104,105,106]. Early neuroimaging and multi-omics studies in humans further connect microbial features with brain networks underlying attention, decision-making, and emotional regulation, supporting a mechanism-anchored view of psychobiotic and prebiotic actions [100,101,102,103,104,105,106].
Evidence integration across modalities: Converging evidence from human interventions, animal models, and multi-omics/neuroimaging strengthens the causal narrative linking gut signals to CNS-relevant outcomes. Named strains (LGG, PS128, B. longum 1714, S. boulardii) and butyrogenic prebiotics (inulin-type fructans, GOS, RS2/RS3, with supportive signals for arabinoxylans and β-glucans) align with butyrate-centric SCFA pathways, barrier support, tryptophan–serotonin/kynurenine and bile-acid crosstalk, and vagal/HPA interfaces that modulate stress responsivity, attention, and executive control [12,17,18,19,20,51,52,53,54,55,61,62,63,64,65,78,79,80,81,82,83,84,85,86,93,94,95,96,97,98,99,100,101,102,103,104,105,106]. Human neuroimaging and multi-omics links between microbial features and frontostriatal/prefrontal networks provide independent corroboration of these mechanisms in cognitive and decision-related domains [20,100,101,102,103,104,105,106,107,108,109,110,111]. These modalities triangulate small-to-moderate, mechanism-consistent effects on perceived stress, mood indices, attentional bias, and selected cognitive endpoints reported in the Results.
Named microbiome agents with plausible CNS benefits: Converging evidence indicates that selected probiotic strains (Lactobacillus rhamnosus GG-ATCC 53103, L. plantarum PS128, Bifidobacterium longum 1714, and Saccharomyces boulardii) are the most consistently studied in relation to stress/mood modulation and attentional/cognitive endpoints [12,17,18,19,20]. In parallel, the prebiotics most frequently linked to favorable CNS-relevant outcomes are inulin-type fructans, galacto-oligosaccharides (GOS), resistant starch (RS2/RS3), arabinoxylans, and β-glucans [51,52,53,54,55,61,62,63,64,65]. Mechanistically, these agents converge on SCFA production (notably butyrate), epithelial barrier reinforcement, bile-acid and tryptophan pathway modulation, and neuroimmune–neuroendocrine signaling (including vagal and HPA-axis interfaces), providing biologically grounded routes to central effects [78,79,80,81,82,83,84,85,86,93,94,95,96,97,98,99,100,101,102,103,104,105,106]. Beyond intervention agents, beneficial commensals with mechanistic plausibility for CNS impact include butyrate-producing Faecalibacterium prausnitzii and mucus-layer-supporting Akkermansia muciniphila, as well as butyrate-linked Clostridium butyricum; Lactobacillus reuteri is also emerging as a promising target for mechanism-first clinical trials within the gut–brain axis [12,17,18,19,20]. These taxa and substrates map onto reduced perceived stress and inflammatory tone, improvements in mood indices, and signals for attention/cognition.
Strengths of the included studies: Several features across the included literature increase confidence in the signals observed. First, a subset of human trials used named, strain-specified probiotics and well-defined prebiotic substrates, enabling mechanism-anchored interpretation and cross-study comparability (e.g., LGG, PS128, B. longum 1714; inulin-type fructans, GOS, resistant starch) [12,17,18,19,20,51,52,53,54,55,61,62,63,64,65]. Second, converging biological readouts, including inflammatory markers, endocrine indices (e.g., cortisol) and GI/barrier-related measures, map onto hypothesized pathways (SCFAs, barrier integrity, bile-acid and tryptophan signaling, vagal/HPA interfaces), reinforcing the mechanistic coherence of the clinical effects [78,79,80,81,82,83,84,85,86,93,94,95,96,97,98,99,100,101,102,103,104,105,106]. Third, an increasing number of studies incorporate multi-omics and/or neuroimaging approaches, which begin to link microbial features with brain networks and cognitive/affective endpoints, improving translational plausibility [100,101,102,103,104,105,106]. Evidence spans diverse populations and settings, which helps to assess robustness and boundary conditions for psychobiotic/prebiotic actions [12,17,18,19,20,51,52,53,54,55,61,62,63,64,65].
Where the signal is strongest vs. emerging: Among probiotics, LGG and PS128 show the most consistent reductions in perceived stress and inflammatory tone, with PS128 also supporting recovery/performance contexts; B. longum 1714 exhibits attentional and cortisol effects in human experimental paradigms. For prebiotics, GOS most clearly modulates cortisol and negative attentional bias in young adults, while inulin-type fructans and RS2/RS3 reliably drive bifidogenic/butyrogenic changes that couple to improvements in stress proxies, metabolic/inflammatory markers, and GI function, domains plausibly upstream of affect and cognition [12,17,18,19,20,51,52,53,54,55,61,62,63,64,65]. These patterns are coherent with the mechanistic levers summarized above and are echoed by systems-level studies relating microbial composition/function to brain networks and cognitive performance [20,100,101,102,103,104,105,106,107,108,109,110,111]. At the same time, heterogeneity in samples, duration, dosing/strain precision, and outcome measures explains residual variance and identifies priorities (standardized endpoints, biomarker anchoring, responder analyses) for the next generation of trials [93,94,95,96,97,98,99,100,101,102,103,104,105,106].
Clinical implications (near-term): Given current evidence, named strains (LGG, PS128, B. longum 1714) and prebiotics (GOS, inulin-type fructans, RS2/RS3; with supportive but more limited signals for arabinoxylans and β-glucans) are plausible adjuncts for individuals with stress-related symptoms or subthreshold mood concerns, particularly when embedded in fiber-rich, Mediterranean-style dietary patterns. Effects should be framed as modest and mechanism-guided, attending to strain/substrate specificity, duration (≥4–8 weeks in many trials), and baseline diet/microbiome [12,17,18,19,20,51,52,53,54,55,61,62,63,64,65]. Dietary strategies and fermented foods may complement such approaches; FMT remains investigational for neurobehavioral indications and requires further safety/efficacy clarification [194,195,196,197,211,212,213].
Future directions (precision and life-course perspectives): Priority areas include (1) longitudinal cohorts beginning in the perinatal period to map sensitive windows and durability of effects; (2) head-to-head RCTs comparing named strains/substrates with adequate dosing and standardized endpoints; (3) biomarker-anchored trials integrating multi-omics, neuroimaging, and endocrine/immune panels; and (4) explicit modifier analyses (dietary fiber, circadian/sleep health, psychotropics, and baseline microbial function) to identify responders [206,207,208,209,210,214,215,216,217,218,219,220,221,222,223,224,225,226,227,228,229,230,231,232]. Additional methodological and translational perspectives underscore these priorities and highlight opportunities for standardized, mechanism-anchored trials [233,234,235]. Advancing precision therapeutics will require integrating host genetics, immune signatures, and microbial biomarkers to tailor interventions for insomnia/circadian disturbance, mood disorders, neurodevelopmental syndromes, and neurodegenerative diseases [64,66,189,208,209,210,214,215,218,219,220,224,225,226,227,228,229,230,231,232]. Named targets such as Lactobacillus reuteri and butyrate-linked pathways are attractive candidates for mechanism-first clinical trials [236].

5. Limitations

This review has limitations at both the evidence and methodological levels. At the evidence level, many included studies feature modest sample sizes and short intervention periods, with heterogeneous endpoints (different scales and composite measures) that reduce cross-study comparability and preclude reliable meta-analytic aggregation. Strain/substrate specification and dosing are inconsistently reported, and potential effect modifiers—including baseline diet and fiber intake, baseline microbiome features, sleep/circadian factors, medication use, and physical activity—are variably measured or controlled, complicating attribution of effects and generalizability across populations [93,94,95,96,97,98,99,100,101,102,103,104,105,106]. For prebiotics, dose–response and substrate specificity remain insufficiently standardized; for probiotics, strain-specific effects caution against genus- or species-level generalizations [51,52,53,54,55,61,62,63,64,65,78,79,80,81,82,83,84,85,86].
At the methodological level, this is a narrative (non-systematic) review without meta-analysis; we used a PRISMA-style flow for transparency rather than a preregistered systematic protocol. Screening proceeded in two stages (titles/abstracts → full-text), with reasons for exclusion recorded; however, the narrative design may still introduce selection and publication bias, and our English-language focus and database coverage may have missed relevant studies. We excluded in vitro/cell-line-only evidence from study selection/data extraction, considering such work only as background mechanistic context; while appropriate for organism-level outcomes, this choice may underrepresent early molecular insights and thus should be borne in mind when interpreting mechanistic breadth. Finally, variability in microbiome analytical pipelines across studies (sampling, sequencing, and bioinformatic workflows) introduces additional method heterogeneity, which we reflect in our cautious, mechanism-anchored synthesis and in the priorities we outline for future trials [93,94,95,96,97,98,99,100,101,102,103,104,105,106]. These constraints collectively argue for adequately powered, longer-duration, strain/substrate-precise trials, harmonized core outcome sets, and biomarker-anchored designs that enable responder analyses and stronger causal inference in human populations [12,17,18,19,20,51,52,53,54,55,61,62,63,64,65,78,79,80,81,82,83,84,85,86,93,94,95,96,97,98,99,100,101,102,103,104,105,106].

6. Conclusions

The gut microbiome plays a foundational and multi-layered role in shaping mood, cognitive function, and decision-making, operating through an intricate interplay of neurochemical, immune, and endocrine pathways. Across diverse lines of evidence, from molecular studies to clinical trials, the microbiome–gut–brain axis has emerged as a compelling target for therapeutic innovation in neuropsychiatry and behavioral health.
Although microbiome-targeted interventions, such as probiotics, psychobiotics, diet modification, and fecal microbiota transplantation, show considerable promise, the field still faces significant challenges. These include the need for rigorous experimental designs, harmonized analytical methodologies, and the development of translational frameworks that can reliably convert correlative findings into mechanism-based, clinically actionable therapies. In practical terms, the leading probiotics and prebiotics highlighted here, together with their putative mechanisms, offer near-term candidates for standardized, mechanism-anchored clinical testing.
A systems-level approach that integrates personalized medicine, multi-omics technologies, and ethical oversight will be essential for advancing the microbiome’s potential in the prevention and treatment of affective, cognitive, and decision-related disorders.

Author Contributions

Conceptualization, P.D. and S.M.; writing—original draft preparation, P.D., G.M., FM. and L.F.; writing—review and editing, P.D., S.M. and G.E.C.; supervision, P.D. and F.M. All authors have read and agreed to the published version of the manuscript.

Funding

The authors declare that financial support was received for publication of this article. Project ECS0000024 “Ecosistema dell’innovazione—Rome Technopole” was financed by the EU via the NextGenerationEU plan through MUR Decree n. 105123.06.2022 PNRR Missione 4 Componente 2 Investimento 1.5—CUP H33C22000420001.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

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

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Peters, D.L.; Wang, W.; Zhang, X.; Ning, Z.; Mayne, J.; Figeys, D. Metaproteomic and metabolomic approaches for characterizing the gut microbiome. Proteomics 2019, 19, e1800363. [Google Scholar] [CrossRef]
  2. Manor, O.; Levy, R.; Borenstein, E. Mapping the inner workings of the microbiome: Genomic- and metagenomic-based study of metabolism and metabolic interactions in the human microbiome. Cell Metab. 2014, 20, 742–752. [Google Scholar] [CrossRef]
  3. Raethong, N.; Nakphaichit, M.; Suratannon, N.; Sathitkowitchai, W.; Weerapakorn, W.; Keawsompong, S.; Vongsangnak, W. Analysis of human gut microbiome: Taxonomy and metabolic functions in Thai adults. Genes 2021, 12, 331. [Google Scholar] [CrossRef]
  4. Zhang, X.; Cheng, K.; Ning, Z.; Mayne, J.; Chi, H.; Farnsworth, C.L.; Lee, K.; Figeys, D. Exploring the microbiome-wide lysine acetylation, succinylation, and propionylation in human gut microbiota. Anal. Chem. 2021, 93, 6594–6598. [Google Scholar] [CrossRef]
  5. Kim, M.S.; Kim, Y.; Choi, H.; Kim, W.; Park, S.; Lee, D.; Kim, D.K.; Kim, H.J.; Choi, H.; Hyun, D.-W.; et al. Transfer of a healthy microbiota reduces amyloid and tau pathology in an Alzheimer’s disease animal model. Gut 2020, 69, 283–294. [Google Scholar] [CrossRef] [PubMed]
  6. Yankovsky, D.S.; Shirobokov, V.P.; Dyment, G.S. Innovation technologies for human microbiome improvement. Sci. Innov. 2018, 14, 11–21. [Google Scholar] [CrossRef]
  7. Greenblum, S.; Turnbaugh, P.J.; Borenstein, E. Metagenomic systems biology of the human gut microbiome reveals topological shifts associated with obesity and inflammatory bowel disease. Proc. Natl. Acad. Sci. USA 2012, 109, 594–599. [Google Scholar] [CrossRef]
  8. Wu, M.Y.; Fan, J.G. Gut microbiome and nonalcoholic fatty liver disease. Hepatobiliary Pancreat. Dis. Int. 2023, 22, 444–451. [Google Scholar] [CrossRef]
  9. Yadav, H.; Jain, S.; Bissi, L.; Marotta, F. Gut microbiome derived metabolites to regulate energy homeostasis: How microbiome talks to host. Metabolomics 2016, 6, e150. [Google Scholar] [CrossRef]
  10. Haiser, H.J.; Gootenberg, D.B.; Chatman, K.; Sirasani, G.; Balskus, E.P.; Turnbaugh, P.J. Predicting and manipulating cardiac drug inactivation by the human gut bacterium Eggerthella lenta. Science 2013, 341, 295–298. [Google Scholar] [CrossRef] [PubMed]
  11. Hayes, W.A.; Sahu, S.C. The human microbiome: History and future. J. Pharm. Pharm. Sci. 2020, 23, 406–411. [Google Scholar] [CrossRef]
  12. Tu, P.; Bian, X.; Chi, L.; Gao, B.; Ru, H.; Knobloch, T.J.; Weghorst, C.M.; Lu, K. Characterization of the functional changes in mouse gut microbiome associated with increased Akkermansia muciniphila population modulated by dietary black raspberries. ACS Omega 2018, 3, 10927–10937. [Google Scholar] [CrossRef]
  13. Baldini, F.; Hertel, J.; Sandt, E.; Thinnes, C.C.; Neuberger-Castillo, L.; Pavelka, L.; Betsou, F.; Krüger, R.; Thiele, I.; on behalf of the NCER-PD Consortium. Parkinson’s disease-associated alterations of the gut microbiome can invoke disease-relevant metabolic changes. BioRxiv 2019, 691030. [Google Scholar] [CrossRef]
  14. Lu, K.; Abo, R.P.; Schlieper, K.A.; Graffam, M.E.; Levine, S.; Wishnok, J.S.; Swenberg, J.A.; Tannenbaum, S.R.; Fox, J.G. Arsenic exposure perturbs the gut microbiome and its metabolic profile in mice: An integrated metagenomics and metabolomics analysis. Environ. Health Perspect. 2014, 122, 284–291. [Google Scholar] [CrossRef]
  15. Wilmanski, T.; Rappaport, N.; Earls, J.C.; Magis, A.T.; Manor, O.; Lovejoy, J.; Omenn, G.S.; Hood, L.; Gibbons, S.M.; Price, N.D. Blood metabolome predicts gut microbiome α-diversity in humans. Nat. Biotechnol. 2019, 37, 1217–1228. [Google Scholar] [CrossRef]
  16. Li, M.; Wang, B.; Zhang, M.; Rantalainen, M.; Wang, S.; Zhou, H.; Zhang, Y.; Shen, J.; Pang, X.; Zhang, M.; et al. Symbiotic gut microbes modulate human metabolic phenotypes. Proc. Natl. Acad. Sci. USA 2008, 105, 2117–2122. [Google Scholar] [CrossRef] [PubMed]
  17. Rinninella, E.; Raoul, P.; Cintoni, M.; Franceschi, F.; Miggiano, G.A.D.; Gasbarrini, A.; Mele, M.C. What is the Healthy Gut Microbiota Composition? A Changing Ecosystem across Age, Environment, Diet, and Diseases. Microorganisms 2019, 7, 14. [Google Scholar] [CrossRef] [PubMed]
  18. Sanborn, V.; Azcarate-Peril, M.A.; Updegraff, J.; Manderino, L.M.; Gunstad, J. A randomized clinical trial examining the impact of LGG probiotic supplementation on psychological status in middle-aged and older adults. Contemp. Clin. Trials Commun. 2018, 12, 192–197. [Google Scholar] [CrossRef]
  19. Huang, W.C.; Wei, C.C.; Huang, C.C.; Chen, W.L.; Huang, H.Y. The Beneficial Effects of Lactobacillus plantarum PS128 on High-Intensity, Exercise-Induced Oxidative Stress, Inflammation, and Performance in Triathletes. Nutrients 2019, 11, 353. [Google Scholar] [CrossRef] [PubMed]
  20. Liu, J.; Sun, J.; Wang, F.; Yu, X.; Ling, Z.; Li, H.; Chen, W.; Pang, M.; Yu, J.; He, Y.; et al. Neuroprotective effects of Clostridium butyricum against vascular dementia in mice via metabolic butyrate. Biomed. Res. Int. 2015, 2015, 412946. [Google Scholar] [CrossRef]
  21. Dong, T.S.; Jacobs, J.P. Nonalcoholic fatty liver disease and the gut microbiome: Are bacteria responsible for fatty liver? Exp. Biol. Med. 2019, 244, 408–418. [Google Scholar] [CrossRef]
  22. Sharpton, S.R.; Yong, G.J.M.; Terrault, N.A.; Lynch, S.V. Gut microbial metabolism and nonalcoholic fatty liver disease. Hepatol. Commun. 2018, 3, 29–43. [Google Scholar] [CrossRef] [PubMed]
  23. Boccuto, L.; Tack, J.; Ianiro, G.; Abenavoli, L.; Scarpellini, E. Human genes involved in the interaction between host and gut microbiome: Regulation and pathogenic mechanisms. Genes 2023, 14, 857. [Google Scholar] [CrossRef]
  24. Appleton, J. The Gut-Brain Axis: Influence of Microbiota on Mood and Mental Health. Integr. Med. (Encinitas) 2018, 17, 28–32. [Google Scholar] [PubMed]
  25. Davis, C.D. The gut microbiome and its role in obesity. Nutr. Today 2016, 51, 167–174. [Google Scholar] [CrossRef]
  26. Onaolapo, O.J.; Onaolapo, A.Y.; Olowe, A.O. The neurobehavioral implications of the brain and microbiota interaction. Front. Biosci. Landmark 2020, 25, 363–397. [Google Scholar] [CrossRef] [PubMed]
  27. Liu, Y.J.; Qiao, N.H.; Diao, Q.Y.; Jing, Z.; Vukanti, R.; Dai, P.L.; Ge, Y. Thiacloprid exposure perturbs the gut microbiota and reduces the survival status in honeybees. J. Hazard. Mater. 2020, 389, 121818. [Google Scholar] [CrossRef] [PubMed]
  28. Xia, J.; Gu, L.; Guo, Y.; Feng, H.; Chen, S.; Jurat, J.; Zhang, D. Gut microbiota mediates the preventive effects of dietary capsaicin against depression-like behavior induced by lipopolysaccharide in mice. Front. Cell. Infect. Microbiol. 2021, 11, 627608. [Google Scholar] [CrossRef]
  29. Cline, E.N.; Bicca, M.A.; Viola, K.L.; Klein, W.L. The amyloid-β oligomer hypothesis: Beginning of the third decade. J. Alzheimers Dis. 2018, 64 (Suppl. S1), S567–S610. [Google Scholar] [CrossRef]
  30. Li, N.; Wang, Q.; Wang, Y.; Sun, A.; Lin, Y.; Jin, Y.; Li, X. Fecal microbiota transplantation from chronic unpredictable mild stress mice donors affects anxiety-like and depression-like behavior in recipient mice via the gut microbiota-inflammation-brain axis. Stress 2019, 22, 592–602. [Google Scholar] [CrossRef]
  31. Borkent, J.; Ioannou, M.; Laman, J.D.; Haarman, B.C.; Sommer, I.E. Role of the gut microbiome in three major psychiatric disorders. Psychol. Med. 2022, 52, 1222–1242. [Google Scholar] [CrossRef] [PubMed]
  32. Wong, M.L.; Inserra, A.; Lewis, M.D.; Mastronardi, C.A.; Leong, L.E.X.; Choo, J.; Kentish, S.; Xie, P.; Morrison, M.; Wesselingh, S.L.; et al. Inflammasome signaling affects anxiety- and depressive-like behavior and gut microbiome composition. Mol. Psychiatry 2016, 21, 797–805. [Google Scholar] [CrossRef]
  33. Ma, N.; Guo, P.; Zhang, J. Nutrients mediate intestinal bacteria-mucosal immune crosstalk. Front. Immunol. 2018, 9, 5. [Google Scholar] [CrossRef]
  34. Keku, T.O.; Dulal, S.; Deveaux, A.; Jovov, B.; Han, X. The gastrointestinal microbiota and colorectal cancer. Am. J. Physiol. Gastrointest. Liver Physiol. 2015, 308, G351–G363. [Google Scholar] [CrossRef]
  35. Swarte, J.C.; Li, Y.; Hu, S.; Björk, J.R.; Gacesa, R.; Vich Vila, A.; Douwes, R.M.; Collij, V.; Kurilshikov, A.; Post, A.; et al. Gut microbiome dysbiosis is associated with increased mortality after solid organ transplantation. Sci. Transl. Med. 2022, 14, eabn7566. [Google Scholar] [CrossRef]
  36. Tang, C.F.; Wang, C.Y.; Wang, J.H.; Wang, Q.N.; Li, S.J.; Wang, H.O.; Zhou, F.; Li, J.-M. Short-chain fatty acids ameliorate depressive-like behaviors of high fructose-fed mice by rescuing hippocampal neurogenesis decline and blood–brain barrier damage. Nutrients 2022, 14, 1882. [Google Scholar] [CrossRef] [PubMed]
  37. Sze, M.A.; Baxter, N.T.; Ruffin, M.T.; Rogers, M.A.; Schloss, P.D. Normalization of the microbiota in patients after treatment for colonic lesions. Microbiome 2017, 5, 150663. [Google Scholar] [CrossRef]
  38. Klimova, B.; Novotny, M.; Valis, M. The impact of nutrition and intestinal microbiome on elderly depression—A systematic review. Nutrients 2020, 12, 710. [Google Scholar] [CrossRef]
  39. Wang, Z.; Klipfell, E.; Bennett, B.J.; Koeth, R.; Levison, B.S.; DuGar, B.; Feldstein, A.E.; Britt, E.B.; Fu, X.; Chung, Y.-M.; et al. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature 2011, 472, 57–63. [Google Scholar] [CrossRef]
  40. Yarandi, S.S.; Peterson, D.A.; Treisman, G.J.; Moran, T.H.; Pasricha, P.J. Modulatory effects of gut microbiota on the central nervous system: How gut could play a role in neuropsychiatric health and diseases. J. Neurogastroenterol. Motil. 2016, 22, 201. [Google Scholar] [CrossRef] [PubMed]
  41. Kandpal, M.; Indari, O.; Baral, B.; Jakhmola, S.; Tiwari, D.; Bhandari, V.; Pandey, R.K.; Bala, K.; Sonawane, A.; Jha, H.C. Dysbiosis of gut microbiota from the perspective of the gut–brain axis: Role in the provocation of neurological disorders. Metabolites 2022, 12, 1064. [Google Scholar] [CrossRef]
  42. Noble, E.E.; Hsu, T.M.; Kanoski, S.E. Gut to brain dysbiosis: Mechanisms linking Western diet consumption, the microbiome, and cognitive impairment. Front. Behav. Neurosci. 2017, 11, 9. [Google Scholar] [CrossRef]
  43. Sampson, T.R.; Mazmanian, S.K. Control of brain development, function, and behavior by the microbiome. Cell Host Microbe 2015, 17, 565–576. [Google Scholar] [CrossRef] [PubMed]
  44. Bartochowski, Z.; Conway, J.; Wallach, Y.; Chakkamparambil, B.; Alakkassery, S.; Grossberg, G.T. Dietary interventions to prevent or delay Alzheimer’s disease: What the evidence shows. Curr. Nutr. Rep. 2020, 9, 210–225. [Google Scholar] [CrossRef] [PubMed]
  45. Khalesi, S.; Bellissimo, N.; Vandelanotte, C.; Williams, S.; Stanley, D.; Irwin, C. A review of probiotic supplementation in healthy adults: Helpful or hype? Eur. J. Clin. Nutr. 2019, 73, 24–37. [Google Scholar] [CrossRef] [PubMed]
  46. Sugita, H.; Tahir, A.; Kinjo, A. Microbiome-targeted therapy in the treatment of cognitive impairment and postoperative cognitive dysfunction: A systematic review. Nutr. Neurosci. 2023, 26, 1–15. [Google Scholar]
  47. Tran, S.M.S.; Mohajeri, M.H. The role of gut bacterial metabolites in brain development, aging and disease. Nutrients 2021, 13, 732. [Google Scholar] [CrossRef]
  48. Davidson, L.E.; Cooke, G.E.; Johnson, L.M.; Quinn, E. Individual differences in the gut microbiome and cognitive performance. Brain Cogn. 2018, 126, 102–109. [Google Scholar]
  49. Romo-Araiza, A.; Ibarra, A. Prebiotics and probiotics as potential therapy for cognitive impairment. Med. Hypotheses 2020, 134, 109410. [Google Scholar] [CrossRef]
  50. Cryan, J.F.; O’Riordan, K.J.; Cowan, C.S.M.; Sandhu, K.V.; Bastiaanssen, T.F.S. The microbiota-gut-brain axis. Physiol. Rev. 2019, 99, 1877–2013. [Google Scholar] [CrossRef]
  51. Moreno-Arribas, M.V.; Bartolomé, B.; Peñalvo, J.L.; Pérez-Matute, P.; Motilva, M.J. Relationship between wine consumption, diet and microbiome modulation in Alzheimer’s disease. Nutrients 2020, 12, 3082. [Google Scholar] [CrossRef] [PubMed]
  52. Li, Z.H.; Jiang, Y.Y.; Long, C.Y.; Peng, Q.; Yue, R.S. The gut microbiota-astrocyte axis: Implications for type 2 diabetic cognitive dysfunction. CNS Neurosci. Ther. 2023, 29, 59–73. [Google Scholar] [CrossRef] [PubMed]
  53. Medawar, E.; Hühn, S.; Villringer, A.; Witte, A.V. The effects of plant-based diets on the body and the brain: A systematic review. Transl. Psychiatry 2019, 9, 1–15. [Google Scholar] [CrossRef] [PubMed]
  54. Tahmasbi, F.; Mirghafourvand, M.; Shamekh, A.; Mahmoodpoor, A.; Sanaie, S. Effects of probiotic supplementation on cognitive function in elderly: A systematic review and meta-analysis. Aging Ment. Health 2022, 26, 1778–1786. [Google Scholar] [CrossRef]
  55. Nagpal, R.; Neth, B.J.; Wang, S. Gut mycobiome and its interaction with diet, gut bacteria and Alzheimer’s disease markers in subjects with mild cognitive impairment: A pilot study. eBioMedicine 2020, 59, 102950. [Google Scholar] [CrossRef]
  56. Falkenstein, M.; Simon, M.C.; Mantri, A.; Weber, B.; Koban, L.; Plassmann, H. Impact of the gut microbiome composition on social decision-making. Proc. Natl. Acad. Sci. USA Nexus 2024, 3, pgae166. [Google Scholar] [CrossRef]
  57. Chen, Y.; Xu, J.; Chen, Y. Regulation of neurotransmitters by the gut microbiota and effects on cognition in neurological disorders. Nutrients 2021, 13, 2099. [Google Scholar] [CrossRef]
  58. Kelly, J.R.; Kennedy, P.J.; Cryan, J.F.; Dinan, T.G.; Clarke, G.; Hyland, N.P. Breaking down the barriers: The gut microbiome, intestinal permeability and stress-related psychiatric disorders. Front. Cell. Neurosci. 2015, 9, 392. [Google Scholar] [CrossRef]
  59. McGrattan, A.M.; McGuinness, B.; McKinley, M.C.; Kee, F.; Passmore, P.; Woodside, J.V.; McEvoy, C.T. Diet and inflammation in cognitive ageing and Alzheimer’s disease. Curr. Nutr. Rep. 2019, 8, 53–65. [Google Scholar] [CrossRef]
  60. Page, M.J.; McKenzie, J.E.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.; Brennan, S.E.; et al. The PRISMA 2020 statement: An updated guideline for reporting systematic reviews. J. Clin. Epidemiol. 2021, 134, 178–189. [Google Scholar] [CrossRef]
  61. Postler, T.S.; Ghosh, S. Understanding the Holobiont: How Microbial Metabolites Affect Human Health and Shape the Immune System. Cell Metab. 2017, 26, 110–130. [Google Scholar] [CrossRef]
  62. Alpino, G.D.C.Á.; Pereira-Sol, G.A.; Dias, M.D.M.E.; Aguiar, A.S.D.; Peluzio, M.D.C.G. Beneficial effects of butyrate on brain functions: A view of epigenetic. Crit. Rev. Food Sci. Nutr. 2024, 64, 3961–3970. [Google Scholar] [CrossRef]
  63. Foster, J.A.; Baker, G.B.; Dursun, S.M. The relationship between the gut microbiome-immune system-brain axis and major depressive disorder. Front. Neurol. 2021, 12, 721126. [Google Scholar] [CrossRef]
  64. Li, S.; Hao, Y.; Fan, F.; Zhang, B. The role of microbiome in insomnia, circadian disturbance and depression. Front. Psychiatry 2018, 9, 669. [Google Scholar] [CrossRef] [PubMed]
  65. Barrio, C.; Arias-Sánchez, S.; Martín-Monzón, I. The gut microbiota-brain axis, psychobiotics and its influence on brain and behaviour: A systematic review. Psychoneuroendocrinology 2022, 137, 105640. [Google Scholar] [CrossRef] [PubMed]
  66. Choi, T.; Choi, Y.; Koo, J. Mental disorders linked to crosstalk between the gut microbiome and the brain. Exp. Neurobiol. 2020, 29, 403–416. [Google Scholar] [CrossRef] [PubMed]
  67. Makris, A.P.; Karianaki, M.; Tsamis, K.I.; Paschou, S.A. The role of the gut-brain axis in depression: Endocrine, neural, and immune pathways. Hormones 2021, 20, 1–12. [Google Scholar] [CrossRef]
  68. Merlo, G.; Bachtel, G.; Sugden, S.G. Gut microbiota, nutrition, and mental health. Front. Nutr. 2024, 11, 1337889. [Google Scholar] [CrossRef]
  69. Van de Wouw, M.; Schellekens, H.; Dinan, T.G.; Cryan, J.F. Microbiota-gut-brain axis: Modulator of host metabolism and appetite. J. Nutr. 2017, 147, 727–745. [Google Scholar] [CrossRef]
  70. Kim, Y.; Hong, M.; Kim, S.; Shin, W.Y.; Kim, J.H. Inverse association between dietary fiber intake and depression in premenopausal women: A nationwide population-based survey. Menopause 2021, 28, 150–156. [Google Scholar] [CrossRef]
  71. Mac Giollabhui, N.; Ng, T.H.; Ellman, L.M.; Alloy, L.B. The longitudinal associations of inflammatory biomarkers and depression revisited: Systematic review, meta-analysis, and meta-regression. Mol. Psychiatry 2021, 26, 3302–3314. [Google Scholar] [CrossRef] [PubMed]
  72. Iglesias-Vázquez, L.; Riba, A.; Arija, V.; Canals, J. Composition of Gut Microbiota in Children with Autism Spectrum Disorder: A Systematic Review and Meta-Analysis. Nutrients 2020, 12, 792. [Google Scholar] [CrossRef]
  73. Derrien, M.; Alvarez, A.-S.; de Vos, W.M. The Gut Microbiota in the First Decade of Life. Trends Microbiol. 2019, 27, 997–1010. [Google Scholar] [CrossRef]
  74. Mackie, A.; Mulet-Cabero, A.I.; Torcello-Gómez, A. Simulating human digestion: Developing our knowledge to create healthier and more sustainable foods. Food Funct. 2020, 11, 9397–9431. [Google Scholar] [CrossRef]
  75. Zhu, G.; Zhao, J.; Zhang, H.; Chen, W.; Wang, G. Probiotics for mild cognitive impairment and Alzheimer’s disease: A systematic review and meta-analysis. Foods 2021, 10, 1672. [Google Scholar] [CrossRef]
  76. Wei, D.; Xie, L.; Zhuang, Z.; Zhao, N.; Huang, B.; Tang, Y.; Yu, S.; Zhou, Q.; Wu, Q. Gut microbiota: A new strategy to study the mechanism of electroacupuncture and moxibustion in treating ulcerative colitis. Evid. Based Complement Altern. Med. 2019, 2019, 9730176. [Google Scholar] [CrossRef]
  77. Sarkar, S.R.; Banerjee, S. Gut microbiota in neurodegenerative disorders. J. Neuroimmunol. 2019, 328, 98–104. [Google Scholar] [CrossRef]
  78. Zhou, Z.; Sun, B.; Yu, D.; Zhu, C. Gut microbiota: An important player in type 2 diabetes mellitus. Front. Cell. Infect. Microbiol. 2022, 12, 834485. [Google Scholar] [CrossRef] [PubMed]
  79. Jiang, C.; Li, G.; Huang, P.; Liu, Z.; Zhao, B. The gut microbiota and Alzheimer’s disease. J. Alzheimers Dis. 2017, 58, 1–15. [Google Scholar] [CrossRef]
  80. Sorboni, S.G.; Moghaddam, H.S.; Jafarzadeh-Esfehani, R.; Soleimanpour, S. A comprehensive review on the role of the gut microbiome in human neurological disorders. Clin. Microbiol. Rev. 2022, 35, e00338-20. [Google Scholar] [CrossRef] [PubMed]
  81. Hobby, G.P.; Karaduta, O.; Dusio, G.F.; Singh, M.; Zybailov, B.L.; Arthur, J.M. Chronic kidney disease and the gut microbiome. Am. J. Physiol. Renal Physiol. 2019, 316, F1211–F1217. [Google Scholar] [CrossRef] [PubMed]
  82. Grochowska, M.; Laskus, T.; Radkowski, M. Gut Microbiota in Neurological Disorders. Arch. Med. Sci. 2019, 15, 809–819. [Google Scholar] [CrossRef]
  83. Strandwitz, P. Neurotransmitter modulation by the gut microbiota. Brain Res. 2018, 1693, 128–133. [Google Scholar] [CrossRef]
  84. Bravo, J.A.; Forsythe, P.; Chew, M.V.; Escaravage, E.; Savignac, H.M.; Dinan, T.G.; Cryan, J.F. Ingestion of Lactobacillus strain regulates emotional behavior and central GABA receptor expression in a mouse via the vagus nerve. Proc. Natl. Acad. Sci. USA 2011, 108, 16050–16055. [Google Scholar] [CrossRef]
  85. Naufel, M.F.; de Martin Truzzi, G.; Ferreira, C.M.; Coelho, F.M.S. The brain-gut-microbiota axis in the treatment of neurologic and psychiatric disorders. Arq. Neuro-Psiquiatr. 2023, 81, 670–684. [Google Scholar] [CrossRef] [PubMed]
  86. Dezfouli, M.A.; Rashidi, S.K.; Yazdanfar, N.; Khalili, H.; Goudarzi, M.; Saadi, A.; Kiani Deh Kiani, A. The emerging roles of neuroactive components produced by gut microbiota. Mol. Biol. Rep. 2025, 52, 1–18. [Google Scholar] [CrossRef]
  87. Varesi, A.; Pierella, E.; Romeo, M.; Piccini, G.B.; Alfano, C.; Bjørklund, G.; Oppong, A.; Ricevuti, G.; Esposito, C.; Chirumbolo, S.; et al. The potential role of gut microbiota in Alzheimer’s disease: From diagnosis to treatment. Nutrients 2022, 14, 668. [Google Scholar] [CrossRef]
  88. Mandal, A.; Prabhavalkar, K.S.; Bhatt, L.K. Gastrointestinal hormones in regulation of memory. Peptides 2018, 102, 16–25. [Google Scholar] [CrossRef]
  89. Pistollato, F.; Sumalla Cano, S.; Elio, I.; Masias Vergara, M.; Giampieri, F.; Battino, M. Role of gut microbiota and nutrients in amyloid formation and pathogenesis of Alzheimer disease. Nutr. Rev. 2016, 74, 624–634. [Google Scholar] [CrossRef]
  90. Dicks, L.M. Gut bacteria and neurotransmitters. Microorganisms 2022, 10, 1838. [Google Scholar] [CrossRef] [PubMed]
  91. Vannier, N.; Agler, M.; Hacquard, S. Microbiota-mediated disease resistance in plants. PLoS Pathog. 2019, 15, e1007740. [Google Scholar] [CrossRef]
  92. Szeligowski, T.; Yun, A.L.; Lennox, B.R.; Burnet, P.W.J. The gut microbiome and schizophrenia: The current state of the field and clinical applications. Front. Psychiatry 2020, 11, 156. [Google Scholar] [CrossRef]
  93. Bauer, M.E.; Teixeira, A.L. Inflammation in psychiatric disorders: What comes first? Ann. N. Y. Acad. Sci. 2018, 1437, 57–67. [Google Scholar] [CrossRef] [PubMed]
  94. Jones, B.D.; Daskalakis, Z.J.; Carvalho, A.F.; Strawbridge, R.; Young, A.H.; Mulsant, B.H.; Husain, M.I. Inflammation as a treatment target in mood disorders. BJPsych Open 2020, 6, e60. [Google Scholar] [CrossRef] [PubMed]
  95. Rosenblat, J.D.; Kakar, R.; Berk, M.; Kessing, L.V.; Vinberg, M.; Baune, B.T.; McIntyre, R.S. Anti-inflammatory agents in the treatment of bipolar depression: A systematic review and meta-analysis. Bipolar Disord. 2016, 18, 89–101. [Google Scholar] [CrossRef] [PubMed]
  96. Haroon, E.; Miller, A.H.; Sanacora, G. Inflammation, glutamate, and glia: A trio of trouble in mood disorders. Neuropsychopharmacology 2016, 42, 193–215. [Google Scholar] [CrossRef]
  97. Wang, X.; Liang, X.; Jiang, M.; Wei, L.; Shi, X.; Fang, F.; Cang, J. Systemic inflammation as a mediator in the link between obesity and depression: Evidence from a nationwide cohort study. BMC Psychiatry 2025, 25, 449. [Google Scholar] [CrossRef]
  98. Hodes, G.E.; Pfau, M.L.; Leboeuf, M.; Golden, S.A.; Christoffel, D.J.; Bregman, D.; Rebusi, N.; Heshmati, M.; Aleyasin, H.; Warren, B.L.; et al. Individual differences in the peripheral immune system promote resilience versus susceptibility to social stress. Proc. Natl. Acad. Sci. USA 2015, 112, 16040–16045. [Google Scholar] [CrossRef]
  99. Husain, M.I.; Strawbridge, R.; Stokes, P.R.; Young, A.H. Anti-inflammatory treatments for mood disorders: Systematic review and meta-analysis. J. Psychopharmacol. 2017, 31, 1137–1148. [Google Scholar] [CrossRef]
  100. Jing, Y.; Yu, Y.; Bai, F.; Wang, L.; Yang, D.; Zhang, C.; Qin, C.; Yang, M.; Zhang, D.; Zhu, Y.; et al. Effect of fecal microbiota transplantation on neurological restoration in a spinal cord injury mouse model: Involvement of brain-gut axis. Microbiome 2021, 9, 59. [Google Scholar] [CrossRef]
  101. Ling, Z.; Zhu, M.; Yan, X.; Cheng, Y.; Shao, L.; Liu, X.; Jiang, R.; Wu, S. Structural and functional dysbiosis of fecal microbiota in Chinese patients with Alzheimer’s disease. Front. Cell Dev. Biol. 2021, 8, 634069. [Google Scholar] [CrossRef]
  102. Generoso, J.S.; Giridharan, V.V.; Lee, J.; Macedo, D.; Barichello, T. The role of the microbiota-gut-brain axis in neuropsychiatric disorders. Braz. J. Psychiatry 2020, 43, 293–305. [Google Scholar] [CrossRef]
  103. Soni, D.; Upadhayay, S.; Dhureja, M.; Arthur, R.; Kumar, P. Crosstalk between gut–brain axis: Unveiling the mysteries of gut ROS in progression of Parkinson’s disease. Inflammopharmacology 2024, 32, 2921–2941. [Google Scholar] [CrossRef]
  104. Wilmes, L.; Collins, J.M.; O’Riordan, K.J.; O’Mahony, S.M.; Cryan, J.F.; Clarke, G. Of bowels, brain and behavior: A role for the gut microbiota in psychiatric comorbidities in irritable bowel syndrome. Neurogastroenterol. Motil. 2021, 33, e14095. [Google Scholar] [CrossRef] [PubMed]
  105. Moser, G.; Fournier, C.; Peter, J. Intestinal microbiome-gut-brain axis and irritable bowel syndrome. Wien. Med. Wochenschr. 2017, 168, 62. [Google Scholar] [CrossRef] [PubMed]
  106. Wang, Y.; Kasper, L.H. The role of microbiome in central nervous system disorders. Brain Behav. Immun. 2014, 38, 1–12. [Google Scholar] [CrossRef] [PubMed]
  107. Foster, J.A.; McVey Neufeld, K. Gut–brain axis: How the microbiome influences anxiety and depression. Trends Neurosci. 2013, 36, 305–312. [Google Scholar] [CrossRef]
  108. Mayer, E.A.; Knight, R.; Mazmanian, S.K.; Cryan, J.F.; Tillisch, K. Gut microbes and the brain: Paradigm shift in neuroscience. J. Neurosci. 2014, 34, 15490–15496. [Google Scholar] [CrossRef]
  109. Liang, X.; Fu, Y.; Cao, W.T.; Wang, Z.; Zhang, K.; Jiang, Z.; Jia, X.; Liu, C.-Y.; Lin, H.-R.; Zhong, H.; et al. Gut microbiome, cognitive function and brain structure: A multi-omics integration analysis. Transl. Neurodegener. 2022, 11, 49. [Google Scholar] [CrossRef]
  110. Bairamian, D.; Sha, S.; Rolhion, N.; Sokol, H.; Dorothée, G.; Lemere, C.A.; Krantic, S. Microbiota in neuroinflammation and synaptic dysfunction: A focus on Alzheimer’s disease. Mol. Neurodegener. 2022, 17, 19. [Google Scholar] [CrossRef]
  111. Wasser, C.I.; Mercieca, E.C.; Kong, G.; Hannan, A.J.; McKeown, S.J.; Glikmann-Johnston, Y.; Stout, J.C. Gut dysbiosis in Huntington’s disease: Associations among gut microbiota, cognitive performance and clinical outcomes. Brain Commun. 2020, 2, fcaa110. [Google Scholar] [CrossRef]
  112. Nguyen, T.T.; Kosciolek, T.; Maldonado, Y.; Daly, R.E.; Martin, A.S.; McDonald, D.; Knight, R. Differences in gut microbiome composition between persons with chronic schizophrenia and healthy comparison subjects. Schizophr. Res. 2018, 204, 23–29. [Google Scholar] [CrossRef]
  113. Smith, R.P.; Easson, C.; Lyle, S.M.; Kapoor, R.; Donnelly, C.P.; Davidson, E.J.; Ross, T.M. Gut microbiome diversity is associated with sleep physiology in humans. PLoS ONE 2019, 14, e0222394. [Google Scholar] [CrossRef] [PubMed]
  114. Miri, S.; Yeo, J.; Abubaker, S.; Hammami, R. Neuromicrobiology, an emerging neurometabolic facet of the gut microbiome? Front. Microbiol. 2023, 14, 1098412. [Google Scholar] [CrossRef] [PubMed]
  115. Glinert, A.; Turjeman, S.; Elliott, E.; Koren, O. Microbes, metabolites and (synaptic) malleability, oh my! The effect of the microbiome on synaptic plasticity. Biol. Rev. 2022, 97, 582–599. [Google Scholar] [CrossRef]
  116. Bell, L.; Whyte, A.; Duysburgh, C.; Marzorati, M.; Van den Abbeele, P.; Le Cozannet, R.; Fança-Berthon, P.; Fromentin, E.; Williams, C. A randomized, placebo-controlled trial investigating the acute and chronic benefits of American Ginseng (Cereboost®) on mood and cognition in healthy young adults, including in vitro investigation of gut microbiota changes as a possible mechanism of action. Eur. J. Nutr. 2022, 61, 413–428. [Google Scholar] [CrossRef] [PubMed]
  117. Zhu, X.; Han, Y.; Du, J.; Liu, R.; Jin, K.; Yi, W. Microbiota-gut-brain axis and the central nervous system. Oncotarget 2017, 8, 53829–53838. [Google Scholar] [CrossRef]
  118. Hsiao, E.Y.; McBride, S.W.; Hsien, S.; Sharon, G.; Hyde, E.R.; McCue, T.; Codelli, J.A.; Chow, J.; Reisman, S.E.; Petrosino, J.F.; et al. Microbiota modulate behavioral and physiological abnormalities associated with neurodevelopmental disorders. Cell 2013, 155, 1451–1463. [Google Scholar] [CrossRef]
  119. Heijtz, R.D.; Wang, S.; Anuar, F.; Qian, Y.; Björkholm, B.; Samuelsson, A.; Hibberd, M.L.; Forssberg, H.; Pettersson, S. Normal gut microbiota modulates brain development and behavior. Proc. Natl. Acad. Sci. USA 2011, 108, 3047–3052. [Google Scholar] [CrossRef]
  120. Arora, T.; Bäckhed, F. The gut microbiota and metabolic disease: Current understanding and future perspectives. J. Intern. Med. 2016, 280, 339–349. [Google Scholar] [CrossRef]
  121. Rea, K.; Dinan, T.G.; Cryan, J.F. Gut microbiota: A perspective for psychiatrists. Neuropsychobiology 2020, 79, 50–62. [Google Scholar] [CrossRef]
  122. Bruce-Keller, A.J.; Salbaum, J.M.; Berthoud, H.R. Harnessing Gut Microbes for Mental Health: Getting from Here to There. Biol. Psychiatry 2018, 83, 214–223. [Google Scholar] [CrossRef]
  123. Leung, K.; Thuret, S. Gut Microbiota: A Modulator of Brain Plasticity and Cognitive Function in Ageing. Healthcare 2015, 3, 898–916. [Google Scholar] [CrossRef]
  124. Peterson, C.T.; Sharma, V.; Elmén, L.; Peterson, S.N. Immune homeostasis, dysbiosis and therapeutic modulation of the gut microbiota. Clin. Exp. Immunol. 2015, 179, 363–377. [Google Scholar] [CrossRef]
  125. Langmajerová, M.; Ježková, J.; Kreisinger, J.; Semerád, J.; Titov, I.; Procházková, P.; Cajthaml, T.; Jiřička, V.; Vevera, J.; Roubalová, R. Gut microbiome in impulsively violent female convicts. Neuropsychobiology 2025, 84, 1–14. [Google Scholar] [CrossRef] [PubMed]
  126. Dam, S.A.; Mostert, J.C.; Szopinska-Tokov, J.W.; Bloemendaal, M.; Amato, M.; Arias-Vasquez, A. The Role of the Gut-Brain Axis in Attention-Deficit/Hyperactivity Disorder. Gastroenterology Clinics 2019, 48, 407–431. [Google Scholar] [CrossRef] [PubMed]
  127. Fineberg, N.A.; Chamberlain, S.R.; Goudriaan, A.E.; Stein, D.J.; Vanderschuren, L.J.; Gillan, C.M.; Shekar, S.; Gorwood, P.A.P.M.; Voon, V.; Morein-Zamir, S.; et al. New Developments in Human Neurocognition: Clinical, Genetic, and Brain Imaging Correlates of Impulsivity and Compulsivity. CNS Spectr. 2014, 19, 69–89. [Google Scholar] [CrossRef]
  128. Cryan, J.F.; O’Mahony, S.M. The Microbiome-Gut-Brain Axis: From Bowel to Behavior. Neurogastroenterol. Motil. 2011, 23, 187–192. [Google Scholar] [CrossRef]
  129. Günüç, S.; Şendemir, A. Cognitive, Emotional, Behavioral and Physiological Evaluation of the Relationship Between Brain and Gut Microbiota. Psikiyatr. Güncel Yaklaşımlar 2022, 14, 446–459. [Google Scholar] [CrossRef]
  130. García-Cabrerizo, R.; Carbia, C.; O’Riordan, K.J.; Schellekens, H.; Cryan, J.F. Microbiota-Gut-Brain Axis as a Regulator of Reward Processes. J. Neurochem. 2021, 157, 1495–1524. [Google Scholar] [CrossRef] [PubMed]
  131. Liu, Q.; Liu, Y.; Zhang, J.; Guan, Y.; Zhou, Q.; Yan, Y.; He, M. Gut microbiota deficiency aggravates arsenic-induced toxicity by affecting bioaccumulation and biotransformation in C57BL/6J mice. Food Chem. Toxicol. 2024, 186, 114564. [Google Scholar] [CrossRef] [PubMed]
  132. Mulder, D.; Aarts, E.; Arias Vasquez, A.; Bloemendaal, M. A Systematic Review Exploring the Association Between the Human Gut Microbiota and Brain Connectivity in Health and Disease. Mol. Psychiatry 2023, 28, 5037–5061. [Google Scholar] [CrossRef]
  133. Homer, B.; Judd, J.; Mohammadi Dehcheshmeh, M.; Ebrahimie, E.; Trott, D.J. Gut Microbiota and Behavioural Issues in Production, Performance, and Companion Animals: A Systematic Review. Animals 2023, 13, 1458. [Google Scholar] [CrossRef]
  134. Peterson, V.L.; Richards, J.B.; Meyer, P.J.; Cabrera-Rubio, R.; Tripi, J.A.; King, C.P.; Polesskaya, O.; Baud, A.; Chitre, A.S.; Bastiaanssen, T.F.; et al. Sex-Dependent Associations Between Addiction-Related Behaviors and the Microbiome in Outbred Rats. eBioMedicine 2020, 55, 102769. [Google Scholar] [CrossRef]
  135. Wu, G.D.; Chen, J.; Hoffmann, C.; Bittinger, K.; Chen, Y.Y.; Keilbaugh, S.A.; Bewtra, M.; Knights, D.; Walters, W.A.; Knight, R.; et al. Linking Long-Term Dietary Patterns with Gut Microbial Enterotypes. Science 2011, 334, 105–108. [Google Scholar] [CrossRef]
  136. Yi, W.; Cheng, J.; Wei, Q.; Pan, R.; Song, S.; He, Y.; Tang, C.; Liu, X.; Zhou, Y.; Su, H. Effect of Temperature Stress on Gut-Brain Axis in Mice: Regulation of Intestinal Microbiome and Central NLRP3 Inflammasomes. Sci. Total Environ. 2021, 772, 144568. [Google Scholar] [CrossRef]
  137. Brenner, D.; Shorten, G.D.; O’Mahony, S.M. Postoperative Pain and the Gut Microbiome. Neurobiol. Pain 2021, 10, 100070. [Google Scholar] [CrossRef]
  138. Baker, J.M.; Al-Nakkash, L.; Herbst-Kralovetz, M.M. Estrogen–Gut Microbiome Axis: Physiological and Clinical Implications. Maturitas 2017, 103, 45–53. [Google Scholar] [CrossRef] [PubMed]
  139. Matenchuk, B.A.; Mandhane, P.J.; Kozyrskyj, A.L. Sleep, Circadian Rhythm, and Gut Microbiota. Sleep Med. Rev. 2020, 53, 101340. [Google Scholar] [CrossRef]
  140. Voigt, R.M.; Forsyth, C.B.; Green, S.J.; Mutlu, E.; Engen, P.; Vitaterna, M.H.; Turek, F.W.; Keshavarzian, A. Circadian Disorganization Alters Intestinal Microbiota. PLoS ONE 2014, 9, e97500. [Google Scholar] [CrossRef] [PubMed]
  141. Bi, C.; Guo, S.; Hu, S.; Chen, J.; Ye, M.; Liu, Z.; Su, H. The Microbiota–Gut–Brain Axis and Its Modulation in the Therapy of Depression: Comparison of Efficacy of Conventional Drugs and Traditional Chinese Medicine Approaches. Pharmacol. Res. 2022, 183, 106372. [Google Scholar] [CrossRef] [PubMed]
  142. Widhiati, S.; Purnomosari, D.; Wibawa, T.; Soebono, H. The Role of Gut Microbiome in Inflammatory Skin Disorders: A Systematic Review. Dermatol. Rep. 2021, 14, 9188. [Google Scholar] [CrossRef] [PubMed]
  143. Zhernakova, A.; Kurilshikov, A.; Bonder, M.J.; Tigchelaar, E.F.; Schirmer, M.; Vatanen, T.; Mujagic, Z.; Vila, A.V.; Falony, G.; Vieira-Silva, S.; et al. Population-Based Metagenomics Analysis Reveals Markers for Gut Microbiome Composition and Diversity. Science 2016, 352, 565–569. [Google Scholar] [CrossRef]
  144. Parkar, S.G.; Kalsbeek, A.; Cheeseman, J.F. Potential Role for the Gut Microbiota in Modulating Host Circadian Rhythms and Metabolic Health. Microorganisms 2019, 7, 41. [Google Scholar] [CrossRef]
  145. Qamar, A.; Waheed, J.; Hamza, A.; Mohyuddin, S.G.; Lu, Z.; Namula, Z.; Chen, J.J. The Role of Intestinal Microbiota in Chicken Health, Intestinal Physiology and Immunity. JAPS J. Anim. Plant Sci. 2021, 31, 342–351. [Google Scholar] [CrossRef]
  146. Wang, R.; Tang, R.; Li, B.; Ma, X.; Schnabl, B.; Tilg, H. Gut Microbiome, Liver Immunology, and Liver Diseases. Cell. Mol. Immunol. 2021, 18, 4–17. [Google Scholar] [CrossRef]
  147. Henao-Mejia, J.; Elinav, E.; Thaiss, C.A.; Licona-Limon, P.; Flavell, R.A. Role of the Intestinal Microbiome in Liver Disease. J. Autoimmun. 2013, 46, 66–73. [Google Scholar] [CrossRef]
  148. Forouzan, S.; McGrew, K.; Kosten, T.A. Drugs and Bugs: Negative Affect, Psychostimulant Use and Withdrawal, and the Microbiome. Am. J. Addict. 2021, 30, 525–538. [Google Scholar] [CrossRef] [PubMed]
  149. Russell, J.T.; Zhou, Y.; Weinstock, G.M.; Bubier, J.A. The Gut Microbiome and Substance Use Disorder. Front. Neurosci. 2021, 15, 725500. [Google Scholar] [CrossRef]
  150. Browning, B.D.; Kirkland, A.E.; Green, R.; Engevik, M.; Alekseyenko, A.V.; Leggio, L.; Tomko, R.L.; Squeglia, L.M. The Adolescent and Young Adult Microbiome and Its Association with Substance Use: A Scoping Review. Alcohol Alcoholism 2024, 59, agad055. [Google Scholar] [CrossRef]
  151. Molavi, N.; Rasouli-Azad, M.; Mirzaei, H.; Matini, A.H.; Banafshe, H.R.; Valiollahzadeh, M.; Ghaderi, A. The Effects of Probiotic Supplementation on Opioid-Related Disorder in Patients under Methadone Maintenance Treatment Programs. Int. J. Clin. Pract. 2022, 2022, 1206914. [Google Scholar] [CrossRef] [PubMed]
  152. Fu, X.; Chen, T.; Cai, J.; Liu, B.; Zeng, Y.; Zhang, X. The Microbiome–Gut–Brain Axis, a Potential Therapeutic Target for Substance-Related Disorders. Front. Microbiol. 2021, 12, 738401. [Google Scholar] [CrossRef]
  153. Ren, M.; Lotfipour, S. The Role of the Gut Microbiome in Opioid Use. Behav. Pharmacol. 2020, 31, 113–121. [Google Scholar] [CrossRef]
  154. Yang, Q.; Liang, Q.; Balakrishnan, B.; Belobrajdic, D.P.; Feng, Q.J.; Zhang, W. Role of Dietary Nutrients in the Modulation of Gut Microbiota: A Narrative Review. Nutrients 2020, 12, 381. [Google Scholar] [CrossRef]
  155. Egerton, S.; Culloty, S.; Whooley, J.; Stanton, C.; Ross, R.P.; O’Sullivan, L. The Gut Microbiota of Marine Fish. Front. Microbiol. 2018, 9, 873. [Google Scholar] [CrossRef]
  156. Nishida, A.; Inoue, R.; Inatomi, O.; Bamba, S.; Naito, Y.; Andoh, A. Gut Microbiota in the Pathogenesis of Inflammatory Bowel Disease. Clin. J. Gastroenterol. 2017, 11, 1–10. [Google Scholar] [CrossRef]
  157. Cukrowska, B. Microbial and Nutritional Programming—The Importance of the Microbiome and Early Exposure to Potential Food Allergens in the Development of Allergies. Nutrients 2018, 10, 1541. [Google Scholar] [CrossRef]
  158. Yoo, J.Y.; Groer, M.; Dutra, S.V.O.; Sarkar, A.; McSkimming, D.I. Gut Microbiota and Immune System Interactions. Microorganisms 2020, 8, 1587. [Google Scholar] [CrossRef] [PubMed]
  159. Angoa-Pérez, M.; Zagorac, B.; Winters, A.D.; Greenberg, J.M.; Ahmad, M.; Theis, K.R.; Kuhn, D.M. Differential Effects of Synthetic Psychoactive Cathinones and Amphetamine Stimulants on the Gut Microbiome in Mice. PLoS ONE 2020, 15, e0227774. [Google Scholar] [CrossRef]
  160. Barengolts, E.; Green, S.J.; Eisenberg, Y.; Akbar, A.; Reddivari, B.; Layden, B.T.; Dugas, L.; Chlipala, G. Gut Microbiota Varies by Opioid Use, Circulating Leptin and Oxytocin in African American Men with Diabetes and High Burden of Chronic Disease. PLoS ONE 2018, 13, e0203051. [Google Scholar] [CrossRef] [PubMed]
  161. O’Sullivan, A.; Monk, C. Maternal and Environmental Influences on Perinatal and Infant Development. Future Child. 2020, 30, 11–34. [Google Scholar] [CrossRef]
  162. Bull-Otterson, L.; Feng, W.; Kirpich, I.; Wang, Y.; Qin, X.; Liu, Y.; Gobejishvili, L.; Joshi-Barve, S.; Ayvaz, T.; Petrosino, J.; et al. Metagenomic Analyses of Alcohol Induced Pathogenic Alterations in the Intestinal Microbiome and the Effect of Lactobacillus rhamnosus GG Treatment. PLoS ONE 2013, 8, e53028. [Google Scholar] [CrossRef]
  163. Malan-Muller, S.; Valles-Colomer, M.; Raes, J.; Lowry, C.A.; Seedat, S.; Hemmings, S.M. The Gut Microbiome and Mental Health: Implications for Anxiety- and Trauma-Related Disorders. Omics 2018, 22, 90–107. [Google Scholar] [CrossRef]
  164. Couch, C.E.; Epps, C.W. Host, Microbiome, and Complex Space: Applying Population and Landscape Genetic Approaches to Gut Microbiome Research in Wild Populations. J. Hered. 2022, 113, 221–234. [Google Scholar] [CrossRef]
  165. Rothschild, D.; Weissbrod, O.; Barkan, E.; Kurilshikov, A.; Korem, T.; Zeevi, D.; Costea, P.I.; Godneva, A.; Kalka, I.N.; Bar, N.; et al. Environment Dominates over Host Genetics in Shaping Human Gut Microbiota. Nature 2018, 555, 210–215. [Google Scholar] [CrossRef]
  166. Yassour, M.; Vatanen, T.; Siljander, H.; Hämäläinen, A.M.; Härkönen, T.; Ryhänen, S.J.; Franzosa, E.A.; Vlamakis, H.; Huttenhower, C.; Gevers, D.; et al. Natural History of the Infant Gut Microbiome and Impact of Antibiotic Treatment on Bacterial Strain Diversity and Stability. Sci. Transl. Med. 2016, 8, 343ra81. [Google Scholar] [CrossRef]
  167. Maier, L.; Pruteanu, M.; Kuhn, M.; Zeller, G.; Telzerow, A.; Anderson, E.E.; Brochado, A.R.; Fernandez, K.C.; Dose, H.; Mori, H.; et al. Extensive Impact of Non-Antibiotic Drugs on Human Gut Bacteria. Nature 2018, 555, 623–628. [Google Scholar] [CrossRef]
  168. Raymann, K.; Shaffer, Z.; Moran, N.A. Antibiotic Exposure Perturbs the Gut Microbiota and Elevates Mortality in Honeybees. PLoS Biol. 2017, 15, e2001861. [Google Scholar] [CrossRef]
  169. Takami, M.; Aoi, W.; Matsumoto, K.; Kato, Y.; Kobayashi, Y.; Kuwahata, M. High-Intensity Exercise Impairs Intestinal Barrier Function by Generating Oxidative Stress. J. Clin. Biochem. Nutr. 2023, 74, 136. [Google Scholar] [CrossRef]
  170. Wegierska, A.E.; Charitos, I.A.; Topi, S.; Potenza, M.A.; Montagnani, M.; Santacroce, L. The Connection Between Physical Exercise and Gut Microbiota: Implications for Competitive Sports Athletes. Sports Med. 2022, 52, 2355–2369. [Google Scholar] [CrossRef]
  171. Dmytriv, T.R.; Storey, K.B.; Lushchak, V.I. Intestinal Barrier Permeability: The Influence of Gut Microbiota, Nutrition, and Exercise. Front. Physiol. 2024, 15, 1380713. [Google Scholar] [CrossRef]
  172. Tsao, J.P.; Bernard, J.R.; Hsu, H.C.; Hsu, C.L.; Liao, S.F.; Cheng, I.S. Short-Term Oral Quercetin Supplementation Improves Post-Exercise Insulin Sensitivity, Antioxidant Capacity and Enhances Subsequent Cycling Time to Exhaustion in Healthy Adults: A Pilot Study. Front. Nutr. 2022, 9, 875319. [Google Scholar] [CrossRef]
  173. Kauter, A.; Epping, L.; Semmler, T.; Antao, E.M.; Kannapin, D.; Stoeckle, S.D.; Gehlen, H.; Lübke-Becker, A.; Günther, S.; Wieler, L.H.; et al. The Gut Microbiome of Horses: Current Research on Equine Enteral Microbiota and Future Perspectives. Anim. Microbiome 2019, 1, 14. [Google Scholar] [CrossRef]
  174. Vitale, S.G.; Ferrari, F.; Ciebiera, M.; Zgliczyńska, M.; Rapisarda, A.M.C.; Vecchio, G.M.; Pino, A.; Angelico, G.; Knafel, A.; Riemma, G.; et al. The Role of Genital Tract Microbiome in Fertility: A Systematic Review. Int. J. Mol. Sci. 2021, 23, 180. [Google Scholar] [CrossRef]
  175. Gloor, G.B.; Macklaim, J.M.; Pawlowsky-Glahn, V.; Egozcue, J.J. Microbiome Datasets Are Compositional: And This Is Not Optional. Front. Microbiol. 2017, 8, 2224. [Google Scholar] [CrossRef]
  176. Libiger, O.; Schork, N.J. Partial Least Squares Regression Can Aid in Detecting Differential Abundance of Multiple Features in Sets of Metagenomic Samples. Front. Genet. 2015, 6, 350. [Google Scholar] [CrossRef]
  177. Fan, Y.; Pedersen, O. Gut Microbiota in Human Metabolic Health and Disease. Nat. Rev. Microbiol. 2021, 19, 55–71. [Google Scholar] [CrossRef]
  178. He, C.; Yang, Z.; Lu, N. Imbalance of Gastrointestinal Microbiota in the Pathogenesis of Helicobacter pylori-Associated Diseases. Helicobacter 2016, 21, 337–348. [Google Scholar] [CrossRef]
  179. Berman, H.L. Defining Gardnerella spp. Diversity, Phenotypes, and Ecological Roles in the Vaginal Microbiome. Ph.D. Thesis, North Carolina State University, Raleigh, NC, USA, 2023. [Google Scholar]
  180. Santiago-Rodriguez, T.M.; Le François, B.; Macklaim, J.M.; Doukhanine, E.; Hollister, E.B. The Skin Microbiome: Current Techniques, Challenges, and Future Directions. Microorganisms 2023, 11, 1222. [Google Scholar] [CrossRef]
  181. Xu, J.; Yang, Y. Gut Microbiome and Its Meta-Omics Perspectives: Profound Implications for Cardiovascular Diseases. Gut Microbes 2021, 13, 1936379. [Google Scholar] [CrossRef]
  182. Franzosa, E.A.; Hsu, T.; Sirota-Madi, A.; Shafquat, A.; Abu-Ali, G.; Morgan, X.C.; Huttenhower, C. Sequencing and Beyond: Integrating Molecular ‘Omics’ for Microbial Community Profiling. Nat. Rev. Microbiol. 2015, 13, 360–372. [Google Scholar] [CrossRef]
  183. Jiang, D.; Armour, C.R.; Hu, C.; Mei, M.; Tian, C.; Sharpton, T.J.; Jiang, Y. Microbiome Multi-Omics Network Analysis: Statistical Considerations, Limitations, and Opportunities. Front. Genet. 2019, 10, 995. [Google Scholar] [CrossRef]
  184. Moreno-Indias, I.; Lahti, L.; Nedyalkova, M.; Elbere, I.; Roshchupkin, G.; Adilovic, M.; Aydemir, O.; Bakir-Gungor, B.; Pau, E.C.-D.S.; D’eLia, D.; et al. Statistical and Machine Learning Techniques in Human Microbiome Studies: Contemporary Challenges and Solutions. Front. Microbiol. 2021, 12, 635781. [Google Scholar] [CrossRef]
  185. Al-Nasiry, S.; Ambrosino, E.; Schlaepfer, M.; Morré, S.A.; Wieten, L.; Voncken, J.W.; Spinelli, M.; Mueller, M.; Kramer, B.W. The Interplay Between Reproductive Tract Microbiota and Immunological System in Human Reproduction. Front. Immunol. 2020, 11, 378. [Google Scholar] [CrossRef]
  186. Hatzenpichler, R.; Krukenberg, V.; Spietz, R.L.; Jay, Z.J. Next-Generation Physiology Approaches to Study Microbiome Function at Single Cell Level. Nat. Rev. Microbiol. 2020, 18, 241–256. [Google Scholar] [CrossRef]
  187. Murphy, K.M.; Le, S.M.; Wilson, A.E.; Warner, D.A. The Microbiome as a Maternal Effect: A Systematic Review on Vertical Transmission of Microbiota. Integr. Comp. Biol. 2023, 63, 597–609. [Google Scholar] [CrossRef]
  188. Liang, W.; Zhao, L.; Zhang, J.; Fang, X.; Zhong, Q.; Liao, Z.; Wang, L. Colonization Potential to Reconstitute a Microbe Community in Pseudo Germ-Free Mice After Fecal Microbe Transplant from Equol Producer. Front. Microbiol. 2020, 11, 1221. [Google Scholar] [CrossRef]
  189. Lee, K.; Kim, J.; Kim, D. Orally Administered Antibiotics Vancomycin and Ampicillin Cause Cognitive Impairment with Gut Dysbiosis in Mice with Transient Global Forebrain Ischemia. Front. Microbiol. 2020, 11, 564271. [Google Scholar] [CrossRef]
  190. Tengeler, A.; Emmerzaal, T.; Geenen, B.; Verweij, V.; Bodegom, M.; Morava, E.; Kozicz, T. Early-Adolescent Antibiotic Exposure Results in Mitochondrial and Behavioral Deficits in Adult Male Mice. Sci. Rep. 2021, 11, 12875. [Google Scholar] [CrossRef]
  191. Lee, J.; Eun, C.; Jo, S.; Lee, A.; Park, C.; Han, D. The Impact of Gut Microbiota Manipulation with Antibiotics on Colon Tumorigenesis in a Murine Model. PLoS ONE 2019, 14, e0226907. [Google Scholar] [CrossRef]
  192. Bagga, D.; Reichert, J.; Koschutnig, K.; Aigner, C.; Holzer, P.; Koskinen, K.; Schöpf, V. Probiotics Drive Gut Microbiome Triggering Emotional Brain Signatures. Gut Microbes 2018, 9, 486–496. [Google Scholar] [CrossRef]
  193. Olesen, S.W.; Barnett, M.L.; MacFadden, D.R.; Lipsitch, M.; Grad, Y.H. Trends in Outpatient Antibiotic Use and Prescribing Practice Among US Older Adults, 2011–2015: Observational Study. BMJ 2018, 362, k3155. [Google Scholar] [CrossRef]
  194. Cammarota, G.; Ianiro, G.; Gasbarrini, A. Fecal Microbiota Transplantation for the Treatment of Clostridium difficile Infection: A Systematic Review. J. Clin. Gastroenterol. 2014, 48, 693–702. [Google Scholar] [CrossRef]
  195. Tuck, C.J.; Caminero, A.; Jiménez Vargas, N.N.; Soltys, C.L.; Jaramillo Polanco, J.O.; Lopez Lopez, C.D.; Vanner, S.J. The Impact of Dietary Fermentable Carbohydrates on a Postinflammatory Model of Irritable Bowel Syndrome. Neurogastroenterol. Motil. 2019, 31, e13675. [Google Scholar] [CrossRef]
  196. Scheppach, W.; Luehrs, H.; Menzel, T. Beneficial Health Effects of Low-Digestible Carbohydrate Consumption. Br. J. Nutr. 2001, 85 (Suppl. S1), S23–S30. [Google Scholar] [CrossRef]
  197. Mefferd, C.C.; Bhute, S.S.; Phan, J.R.; Villarama, J.V.; Do, D.M.; Alarcia, S.; Hedlund, B.P. A High-Fat/High-Protein, Atkins-Type Diet Exacerbates Clostridioides (Clostridium) difficile Infection in Mice, Whereas a High-Carbohydrate Diet Protects. mSystems 2020, 5, e00765-19. [Google Scholar] [CrossRef]
  198. Sharma, R.; Gupta, D.; Mehrotra, R.; Mago, P. Psychobiotics: The Next-Generation Probiotics for the Brain. Curr. Microbiol. 2021, 78, 449–463. [Google Scholar] [CrossRef]
  199. Lee, J.; Walton, D.; O’Connor, C.; Wammes, M.; Burton, J.; Osuch, E. Drugs, Guts, Brains, but Not Rock and Roll: The Need to Consider the Role of Gut Microbiota in Contemporary Mental Health and Wellness of Emerging Adults. Int. J. Mol. Sci. 2022, 23, 6643. [Google Scholar] [CrossRef]
  200. Gehin, S.; Caron, P.; Cespuglio, R. Psychobiotics as Modulators of Gut-Brain Influences. Biomed. J. Sci. Tech. Res. 2022, 45, 36373–36380. [Google Scholar] [CrossRef]
  201. Gnanavel, S. Psychobiotics: The Latest Psychotropics. Indian J. Psychol. Med. 2015, 37, 110. [Google Scholar] [CrossRef]
  202. Anker-Ladefoged, C.; Langkamp, T.; Mueller-Alcazar, A. The Potential Impact of Selected Bacterial Strains on the Stress Response. Healthcare 2021, 9, 494. [Google Scholar] [CrossRef]
  203. Shabbir, M.; Mehak, F.; Khan, Z.; Ahmed, W.; Abrar-ul-Haq, S.; Khan, M.; Aadil, R. Delving the Role of Nutritional Psychiatry to Mitigate the COVID-19 Pandemic Induced Stress, Anxiety and Depression. Trends Food Sci. Technol. 2022, 120, 25–35. [Google Scholar] [CrossRef]
  204. Vaccariello, E.; Nguyen, T. Microbes and Geriatric Mental Health: Past, Present, and Future. Int. Psychogeriatr. 2021, 34, 3–5. [Google Scholar] [CrossRef]
  205. Jacka, F. Nutritional Psychiatry: Where to Next? EBioMedicine 2017, 17, 24–29. [Google Scholar] [CrossRef]
  206. Peirce, J.; Alviña, K. The Role of Inflammation and the Gut Microbiome in Depression and Anxiety. J. Neurosci. Res. 2019, 97, 1223–1241. [Google Scholar] [CrossRef]
  207. Ameringen, M.; Turna, J.; Patterson, B.; Pipe, A.; Mao, R.; Anglin, R.; Surette, M. The Gut Microbiome in Psychiatry: A Primer for Clinicians. Depress. Anxiety 2019, 36, 1004–1025. [Google Scholar] [CrossRef]
  208. Rogers, G.; Keating, D.; Young, R.; Wong, M.-L.; Licinio, J.; Wesselingh, S. From Gut Dysbiosis to Altered Brain Function and Mental Illness: Mechanisms and Pathways. Mol. Psychiatry 2016, 21, 738–748. [Google Scholar] [CrossRef] [PubMed]
  209. Knippel, R.; Sears, C. The Microbiome Colorectal Cancer Puzzle: Initiator, Propagator, and Avenue for Treatment and Research. J. Natl. Compr. Cancer Netw. 2021, 19, 986–992. [Google Scholar] [CrossRef] [PubMed]
  210. Taylor, V. The Microbiome and Mental Health: Hope or Hype? J. Psychiatry Neurosci. 2019, 44, 219–222. [Google Scholar] [CrossRef]
  211. Ademe, M. Benefits of Fecal Microbiota Transplantation: A Comprehensive Review. J. Infect. Dev. Ctries. 2020, 14, 1074–1080. [Google Scholar] [CrossRef]
  212. Biazzo, M.; Deidda, G. Fecal Microbiota Transplantation as New Therapeutic Avenue for Human Diseases. J. Clin. Med. 2022, 11, 4119. [Google Scholar] [CrossRef] [PubMed]
  213. Verdier, C.; Denis, S.; Gasc, C.; Boucinha, L.; Uriot, O.; Delmas, D.; Dore, J.; Le Camus, C.; Schwintner, C.; Blanquet-Diot, S. An Oral FMT Capsule as Efficient as an Enema for Microbiota Reconstruction Following Disruption by Antibiotics, as Assessed in an In Vitro Human Gut Model. Microorganisms 2021, 9, 358. [Google Scholar] [CrossRef]
  214. Christian, L.; Galley, J.; Hade, E.; Schoppe-Sullivan, S.; Dush, C.; Bailey, M. Gut Microbiome Composition Is Associated with Temperament During Early Childhood. Brain Behav. Immun. 2015, 45, 118–127. [Google Scholar] [CrossRef]
  215. Flannery, J.; Stagaman, K.; Burns, A.; Hickey, R.; Roos, L.; Giuliano, R.; Sharpton, T. Gut Feelings Begin in Childhood: The Gut Metagenome Correlates with Early Environment, Caregiving, and Behavior. mBio 2020, 11, e02780-19. [Google Scholar] [CrossRef] [PubMed]
  216. Lai, J.; Jiang, J.; Zhang, P.; Xi, C.; Wu, L.; Gao, X.; Hu, S. Gut Microbial Clues to Bipolar Disorder: State-of-the-Art Review of Current Findings and Future Directions. Clin. Transl. Med. 2020, 10, e146. [Google Scholar] [CrossRef]
  217. Mueller, N.; Bakacs, E.; Combellick, J.; Grigoryan, Z.; Domínguez-Bello, M. The Infant Microbiome Development: Mom Matters. Trends Mol. Med. 2015, 21, 109–117. [Google Scholar] [CrossRef]
  218. Kraaij, R.; Schuurmans, I.; Radjabzadeh, D.; Tiemeier, H.; Dinan, T.; Uitterlinden, A.; Cecil, C. The Gut Microbiome and Child Mental Health: A Population-Based Study. Brain Behav. Immun. 2023, 108, 188–196. [Google Scholar] [CrossRef]
  219. Adar, S.; Huffnagle, G.; Curtis, J. The Respiratory Microbiome: An Underappreciated Player in the Human Response to Inhaled Pollutants? Ann. Epidemiol. 2016, 26, 355–359. [Google Scholar] [CrossRef]
  220. Singh, S.; Sharma, P.; Pal, N.; Kumawat, M.; Shubham, S.; Sarma, D.; Nagpal, R. Impact of Environmental Pollutants on Gut Microbiome and Mental Health via the Gut–Brain Axis. Microorganisms 2022, 10, 1457. [Google Scholar] [CrossRef]
  221. Kong, G.; Letchumanan, V.; Tan, L.; Law, J. Gut Microbiome in Obsessive Compulsive Disorder: Potential of Probiotics as an Adjuvant Therapy. Prog. Microbes Mol. Biol. 2022, 5, a0000272. [Google Scholar] [CrossRef]
  222. Esquivel, M. Probiotics for Mental Health: A Review of Recent Clinical Trials. Am. J. Lifestyle Med. 2021, 16, 21–27. [Google Scholar] [CrossRef] [PubMed]
  223. Ramon-Krauel, M.; Amat-Bou, M.; Serrano, M.; Martinez-Monseny, A.; Lerin, C. Targeting the Gut Microbiome in Prader-Willi Syndrome. J. Clin. Med. 2021, 10, 5328. [Google Scholar] [CrossRef] [PubMed]
  224. Mousa, W.; Chehadeh, F.; Husband, S. Recent Advances in Understanding the Structure and Function of the Human Microbiome. Front. Microbiol. 2022, 13, 825338. [Google Scholar] [CrossRef]
  225. Goldman, M.; Druss, B.; Horvitz-Lennon, M.; Norquist, G.; Ptakowski, K.; Brinkley, A.; Dixon, L. Mental Health Policy in the Era of COVID-19. Psychiatr. Serv. 2020, 71, 1158–1162. [Google Scholar] [CrossRef]
  226. Hayes, K.; Blashki, G.; Wiseman, J.; Burke, S.; Reifels, L. Climate Change and Mental Health: Risks, Impacts and Priority Actions. Int. J. Ment. Health Syst. 2018, 12, 28. [Google Scholar] [CrossRef]
  227. Peters, B.; Santoro, N.; Kaplan, R.; Qi, Q. Spotlight on the Gut Microbiome in Menopause: Current Insights. Int. J. Women’s Health 2022, 14, 1059–1072. [Google Scholar] [CrossRef]
  228. Akagbosu, C.; Nadler, E.; Levy, S.; Hourigan, S. The Role of the Gut Microbiome in Pediatric Obesity and Bariatric Surgery. Int. J. Mol. Sci. 2022, 23, 15421. [Google Scholar] [CrossRef]
  229. Lange, L.; Berg, G.; Cernava, T.; Champomier-Vergès, M.; Charles, T.; Cocolin, L.; Sessitsch, A. Microbiome Ethics, Guiding Principles for Microbiome Research, Use and Knowledge Management. Environ. Microbiome 2022, 17, 50. [Google Scholar]
  230. Nazaroff, W.W. Embracing Microbes in Exposure Science. J. Expo. Sci. Environ. Epidemiol. 2018, 29, 1–10. [Google Scholar] [CrossRef]
  231. Sainz, T.; Pignataro, V.; Bonifazi, D.; Ravera, S.; Mellado, M.; Pérez-Martínez, A.; Calvo, C. Human Microbiome in Children, at the Crossroad of Social Determinants of Health and Personalized Medicine. Children 2021, 8, 1191. [Google Scholar] [CrossRef]
  232. Hagerty, S.; Hutchison, K.; Lowry, C.; Bryan, A. An Empirically Derived Method for Measuring Human Gut Microbiome Alpha Diversity: Demonstrated Utility in Predicting Health-Related Outcomes Among a Human Clinical Sample. PLoS ONE 2020, 15, e0229204. [Google Scholar] [CrossRef] [PubMed]
  233. Kennedy, P.J.; Murphy, A.B.; Cryan, J.F.; Ross, P.R.; Dinan, T.G.; Stanton, C. Microbiome in Brain Function and Mental Health. Trends Food Sci. Technol. 2016, 57, 289–301. [Google Scholar] [CrossRef]
  234. Rieder, R.; Wisniewski, P.J.; Alderman, B.L.; Campbell, S.C. Microbes and Mental Health: A Review. Brain Behav. Immun. 2017, 66, 9–17. [Google Scholar] [CrossRef] [PubMed]
  235. Padilla, A.B.; Sahai, S.; Pizarro, R.R.; Añonuevo, J.J.J.; Enamno, K. Human microbiome and mental health. In Human Microbiome Drug Targets; Elsevier: Amsterdam, The Netherlands, 2025; pp. 55–68. [Google Scholar]
  236. Buffington, S.A.; Di Prisco, G.V.; Auchtung, T.A.; Ajami, N.J.; Petrosino, J.F.; Costa-Mattioli, M. Microbial Reconstitution Reverses Maternal Diet-Induced Social and Synaptic Deficits in Offspring. Cell 2016, 165, 1762–1775. [Google Scholar] [CrossRef]
Figure 1. PRISMA-style flow diagram of study selection.
Figure 1. PRISMA-style flow diagram of study selection.
Nutrients 17 03350 g001
Table 1. Probiotics and prebiotics cited in the review: exemplar strain/substrate, putative mechanisms, and most consistent human outcomes.
Table 1. Probiotics and prebiotics cited in the review: exemplar strain/substrate, putative mechanisms, and most consistent human outcomes.
CategoryExemplar (Strain/Substrate)Primary MechanismsHuman Outcome DomainsReferences
ProbioticLactobacillus rhamnosus GG (ATCC 53103)Barrier reinforcement (tight junctions); HPA-axis attenuation; immunomodulation; GABAergic signaling (preclinical)Stress/anxiety indicators; mood; gastrointestinal symptoms[17,18,19,20,21]
ProbioticLactobacillus plantarum PS128Neurotransmitter modulation (dopamine/serotonin, preclinical); anti-inflammatory actions; SCFA shiftsMood/stress; exercise performance and recovery[17,18,19,20,21]
ProbioticBifidobacterium longum 1714Vagal signaling; cortisol modulation; attentional/cognitive effects (human experimental evidence)Perceived stress; attention/memory; mood[17,18,19,20,21]
ProbioticSaccharomyces boulardiiBarrier/trophic effects; pathogen exclusion; anti-inflammatory pathwaysGI symptom improvement; adjunct benefits potentially impacting mood via gut symptom relief[17,18,19,20,21]
PrebioticInulin-type fructansBifidogenic effects; fermentation to SCFAs (butyrate); immune modulationMood/stress-related measures; GI function; metabolic/inflammatory markers[52,53,54,55,56,57,58,59,61,62]
PrebioticGalacto-oligosaccharides (GOS)Bifidogenic; SCFA production; HPA-axis/cortisol modulation (human experimental evidence)Anxiety- and stress-related measures; attentional bias[52,53,54,55,56,57,58,59,61,62]
PrebioticResistant starch (types 2–3)Marked butyrate production; improved barrier integrity; bile-acid signalingStress resilience proxies; metabolic and inflammatory markers; GI function[52,53,54,55,56,57,58,59,61,62]
PrebioticArabinoxylansFermentation to SCFAs; immunomodulation; bile-acid interactionsImmune/metabolic markers; select mood/stress proxies[52,53,54,55,56,57,58,59,61,62]
Prebioticβ-GlucansInnate immune modulation (e.g., Dectin-1); SCFA-related effects via co-fermentationStress/fatigue; immune function; inflammatory markers[52,53,54,55,56,57,58,59,61,62]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Diotaiuti, P.; Misiti, F.; Marotta, G.; Falese, L.; Calabrò, G.E.; Mancone, S. The Gut Microbiome and Its Impact on Mood and Decision-Making: A Mechanistic and Therapeutic Review. Nutrients 2025, 17, 3350. https://doi.org/10.3390/nu17213350

AMA Style

Diotaiuti P, Misiti F, Marotta G, Falese L, Calabrò GE, Mancone S. The Gut Microbiome and Its Impact on Mood and Decision-Making: A Mechanistic and Therapeutic Review. Nutrients. 2025; 17(21):3350. https://doi.org/10.3390/nu17213350

Chicago/Turabian Style

Diotaiuti, Pierluigi, Francesco Misiti, Giulio Marotta, Lavinia Falese, Giovanna Elisa Calabrò, and Stefania Mancone. 2025. "The Gut Microbiome and Its Impact on Mood and Decision-Making: A Mechanistic and Therapeutic Review" Nutrients 17, no. 21: 3350. https://doi.org/10.3390/nu17213350

APA Style

Diotaiuti, P., Misiti, F., Marotta, G., Falese, L., Calabrò, G. E., & Mancone, S. (2025). The Gut Microbiome and Its Impact on Mood and Decision-Making: A Mechanistic and Therapeutic Review. Nutrients, 17(21), 3350. https://doi.org/10.3390/nu17213350

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

Article Metrics

Back to TopTop