[go: up one dir, main page]
More Web Proxy on the site http://driver.im/
You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (1,164)

Search Parameters:
Keywords = gut–brain axis

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
17 pages, 1718 KiB  
Systematic Review
The Beneficial Effects of Lactobacillus Strains on Gut Microbiome in Alzheimer’s Disease: A Systematic Review
by Michael Quansah, Monique Antoinette David, Ralph Martins, Emad El-Omar, Silvana Mirella Aliberti, Mario Capunzo, Slade O. Jensen and Mourad Tayebi
Healthcare 2025, 13(1), 74; https://doi.org/10.3390/healthcare13010074 - 3 Jan 2025
Viewed by 252
Abstract
Background/Objectives: Growing evidence suggests that the gut–brain axis influences brain function, particularly the role of intestinal microbiota in modulating cognitive processes. Probiotics may alter brain function and behavior by modulating gut microbiota, with implications for neurodegenerative diseases like Alzheimer’s disease (AD). The purpose [...] Read more.
Background/Objectives: Growing evidence suggests that the gut–brain axis influences brain function, particularly the role of intestinal microbiota in modulating cognitive processes. Probiotics may alter brain function and behavior by modulating gut microbiota, with implications for neurodegenerative diseases like Alzheimer’s disease (AD). The purpose of this review is to systematically review the current literature exploring the effects of probiotic supplementation on gut microbiota and cognitive function in AD and mild cognitive impairment (MCI). Methods: A comprehensive literature search was conducted across PubMed/Medline, Embase, and Scopus to identify relevant randomized controlled trials (RCTs) from inception to 20 August 2024. The search focused on comparing outcomes between intervention and control/placebo groups. Data searches, article selection, data extraction, and risk of bias assessment were performed in accordance with Cochrane guidelines. Systematic Review Registration: PROSPERO registration no: CRD42023446796. Results: Data from four RCTs involving 293 Individuals (AD and MCI patients) receiving mainly Lactobacillus and Bifidobacterium strains showed some beneficial effects on cognitive function, altered gut microbiota composition, and positively affected metabolic biomarkers. However, variability in microbiota assessment across studies limits the interpretation of results. The limited number and quality of the existing studies make it difficult to draw definitive conclusions from the data. Additional high-quality research is clearly needed. Conclusions: Probiotics show promise as an adjunctive intervention for cognitive decline, but larger, long-term trials are needed to confirm their efficacy and clinical applicability in neurodegenerative diseases like AD. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Flow diagram illustrating the identification of studies for inclusion.</p>
Full article ">Figure 2
<p>Risk of bias assessment for the included studies. Risk of bias summary: Green (+): Low risk of bias; Yellow (-): Unclear risk of bias; and Red (X): High risk of bias. [<a href="#B56-healthcare-13-00074" class="html-bibr">56</a>,<a href="#B57-healthcare-13-00074" class="html-bibr">57</a>,<a href="#B58-healthcare-13-00074" class="html-bibr">58</a>,<a href="#B59-healthcare-13-00074" class="html-bibr">59</a>].</p>
Full article ">Figure 3
<p>Risk of bias assessment for the included studies. Risk of bias graph.</p>
Full article ">
20 pages, 1522 KiB  
Review
Alcohol Use Disorder and the Gut–Brain Axis: A Narrative Review of the Role of Gut Microbiota and Implications for Treatment
by Shikha Shukla and Cynthia L. Hsu
Microorganisms 2025, 13(1), 67; https://doi.org/10.3390/microorganisms13010067 - 2 Jan 2025
Viewed by 289
Abstract
Alcohol use disorder (AUD) affects millions of people worldwide and can lead to deleterious physical and social consequences. Recent research has highlighted not only the effect of alcohol on the gut microbiome, but also the role of the gut microbiome and the gut–brain [...] Read more.
Alcohol use disorder (AUD) affects millions of people worldwide and can lead to deleterious physical and social consequences. Recent research has highlighted not only the effect of alcohol on the gut microbiome, but also the role of the gut microbiome and the gut–brain axis in the development and maintenance of alcohol use disorder. This review provides an overview of the reciprocal relationship between alcohol consumption and the gut microbiome, including the effects of alcohol on gut microbial composition, changes in gut microbial metabolites in response to alcohol consumption, and how gut microbial metabolites may modulate alcohol use behavior. We also discuss the gut-mediated mechanisms of neuroinflammation that contribute to and result from AUD, including disruption of the intestinal barrier, toll-like receptor signaling, and the activation of glial cells and immune cells. Finally, we review the current evidence on gut microbial-directed therapies for AUD and discuss the implications of this research for our understanding of the pathophysiology of AUD and future research directions. Full article
(This article belongs to the Section Gut Microbiota)
Show Figures

Figure 1

Figure 1
<p>The gut–brain axis in alcohol use disorder describes a bidirectional communication network where alcohol consumption alters gut microbial composition, leading to changes in gut microbial metabolites that contribute to alcohol-associated neuroinflammation, and ultimately perpetuate a cycle of persistent alcohol consumption.</p>
Full article ">Figure 2
<p>Illustration of the key mechanisms underlying the connection between gut dysbiosis and alcohol use disorder.</p>
Full article ">
25 pages, 427 KiB  
Review
Nutritional and Microbiota-Based Approaches in Amyotrophic Lateral Sclerosis: From Prevention to Treatment
by Francesca Cuffaro, Ingrid Lamminpää, Elena Niccolai and Amedeo Amedei
Nutrients 2025, 17(1), 102; https://doi.org/10.3390/nu17010102 - 30 Dec 2024
Viewed by 469
Abstract
Metabolic alterations, including hypermetabolism, lipid imbalances, and glucose dysregulation, are pivotal contributors to the onset and progression of Amyotrophic Lateral Sclerosis (ALS). These changes exacerbate systemic energy deficits, heighten oxidative stress, and fuel neuroinflammation. Simultaneously, gastrointestinal dysfunction and gut microbiota (GM) dysbiosis intensify [...] Read more.
Metabolic alterations, including hypermetabolism, lipid imbalances, and glucose dysregulation, are pivotal contributors to the onset and progression of Amyotrophic Lateral Sclerosis (ALS). These changes exacerbate systemic energy deficits, heighten oxidative stress, and fuel neuroinflammation. Simultaneously, gastrointestinal dysfunction and gut microbiota (GM) dysbiosis intensify disease pathology by driving immune dysregulation, compromising the intestinal barrier, and altering gut–brain axis (GBA) signaling, and lastly advancing neurodegeneration. Therapeutic and preventive strategies focused on nutrition offer promising opportunities to address these interconnected pathophysiological mechanisms. Diets enriched with antioxidants, omega-3 fatty acids, and anti-inflammatory compounds—such as the Mediterranean diet—have shown potential in reducing oxidative stress and systemic inflammation. Additionally, microbiota-targeted approaches, including probiotics, prebiotics, postbiotics, and fecal microbiota transplantation, are emerging as innovative tools to restore microbial balance, strengthen gut integrity, and optimize GBA function. This review highlights the critical need for personalized strategies integrating immunonutrition and microbiota modulation to slow ALS progression, improve quality of life, and develop preventive measures for neurodegenerative and neuroinflammatory diseases. Future research should prioritize comprehensive dietary and microbiota-based interventions to uncover their therapeutic potential and establish evidence-based guidelines for managing ALS and related disorders. Full article
26 pages, 546 KiB  
Review
The Role of the Gastrointestinal Microbiota in Parkinson’s Disease
by Maurizio Gabrielli, Lorenzo Zileri Dal Verme, Maria Assunta Zocco, Enrico Celestino Nista, Veronica Ojetti and Antonio Gasbarrini
Biomolecules 2025, 15(1), 26; https://doi.org/10.3390/biom15010026 - 28 Dec 2024
Viewed by 330
Abstract
Background/Objectives: Parkinson’s disease (PD) is a progressive neurodegenerative disorder characterized by the loss of dopaminergic neurons leading to debilitating motor and non-motor symptoms. Beyond its well-known neurological features, emerging evidence underscores the pivotal role of the gut–brain axis and gastrointestinal microbiota in PD [...] Read more.
Background/Objectives: Parkinson’s disease (PD) is a progressive neurodegenerative disorder characterized by the loss of dopaminergic neurons leading to debilitating motor and non-motor symptoms. Beyond its well-known neurological features, emerging evidence underscores the pivotal role of the gut–brain axis and gastrointestinal microbiota in PD pathogenesis. Dysbiosis has been strongly linked to PD and is associated with increased intestinal permeability, chronic inflammation, and the production of neurotoxic metabolites that may exacerbate neuronal damage. Methods: This review delves into the complex interplay between PD and dysbiosis, shedding light on two peculiar subsets of dysbiosis, Helicobacter pylori infection and small-intestinal bacterial overgrowth. These conditions may not only contribute to PD progression but also influence therapeutic responses such as L-dopa efficacy. Conclusions: The potential to modulate gut microbiota through probiotics, prebiotics, and synbiotics; fecal microbiota transplantation; and antibiotics represents a promising frontier for innovative PD treatments. Despite this potential, the current evidence is limited by small sample sizes and methodological variability across studies. Rigorous, large-scale, randomized placebo-controlled trials with standardized treatments in terms of composition, dosage, and duration are urgently needed to validate these findings and pave the way for microbiota-based therapeutic strategies in PD management. Full article
Show Figures

Figure 1

Figure 1
<p>Dysbiosis-induced damage to the GI tract triggers local inflammation and compromises the intestinal barrier, exposing the ENS to chronic inflammatory insults. This promotes α-synuclein misfolding, which propagates to the brain via the vagus nerve. Concurrently, chronic GI inflammation leads to the systemic release of inflammatory mediators (single-headed red arrow), weakening the blood–brain barrier. This allows the passage of proinflammatory cytokines, immune cells, antibodies, toxins, and antigens into the central nervous system (CNS), activating microglia and driving neuroinflammation. Microglial activation exacerbates neurodegeneration within the CNS. The double-headed yellow arrow represents the bidirectional communication pathway between the GI tract and the CNS, with the vagus nerve serving as the primary conduit. A self-perpetuating cycle ensues, wherein early alterations in GI motility (from the stomach to the colon), commonly observed in PD, exacerbate dysbiosis, further contributing, as mentioned, to PD pathogenesis.</p>
Full article ">
28 pages, 2050 KiB  
Review
Molecular Mechanisms Linking Omega-3 Fatty Acids and the Gut–Brain Axis
by Anna Zinkow, Wojciech Grodzicki, Malwina Czerwińska and Katarzyna Dziendzikowska
Molecules 2025, 30(1), 71; https://doi.org/10.3390/molecules30010071 - 28 Dec 2024
Viewed by 783
Abstract
The gut–brain axis (GBA) is a complex communication network connecting the gastrointestinal tract (GIT) and the central nervous system (CNS) through neuronal, endocrine, metabolic, and immune pathways. Omega-3 (n-3) fatty acids, particularly eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), are crucial food components [...] Read more.
The gut–brain axis (GBA) is a complex communication network connecting the gastrointestinal tract (GIT) and the central nervous system (CNS) through neuronal, endocrine, metabolic, and immune pathways. Omega-3 (n-3) fatty acids, particularly eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), are crucial food components that may modulate the function of this axis through molecular mechanisms. Derived mainly from marine sources, these long-chain polyunsaturated fatty acids are integral to cell membrane structure, enhancing fluidity and influencing neurotransmitter function and signal transduction. Additionally, n-3 fatty acids modulate inflammation by altering eicosanoid production, reducing proinflammatory cytokines, and promoting anti-inflammatory mediators. These actions help preserve the integrity of cellular barriers like the intestinal and blood–brain barriers. In the CNS, EPA and DHA support neurogenesis, synaptic plasticity, and neurotransmission, improving cognitive functions. They also regulate the hypothalamic–pituitary–adrenal (HPA) axis by reducing excessive cortisol production, associated with stress responses and mental health disorders. Furthermore, n-3 fatty acids influence the composition and function of the gut microbiota, promoting beneficial bacterial populations abundance that contribute to gut health and improve systemic immunity. Their multifaceted roles within the GBA underscore their significance in maintaining homeostasis and supporting mental well-being. Full article
(This article belongs to the Special Issue Bioactive Compounds from Functional Foods, 2nd Edition)
Show Figures

Figure 1

Figure 1
<p>Interaction Between the gut and the brain through the gut–brain axis. HPA axis, hypothalamic-pituitary-adrenal axis; ENS, enteric nervous system; SCFA, short-chain fatty acids. Designed using elements by ©Canva, sparklestroke, Pixeden, iconsy, OpenClipart-Vectors via Canva.com (access date: 18 November 2024).</p>
Full article ">Figure 2
<p>The role of omega-3 fatty acids as components of cell membranes. ↑ indicates an increase, ↓ indicates a decrease. Designed using elements by ©Canva, sparklestroke, Pixeden, iconsy, OpenClipart-Vectors via Canva.com (access date: 18 November 2024).</p>
Full article ">Figure 3
<p>Anti-inflammatory role of n-3 fatty acids. NF-kB—nuclear factor kappa B; COX-2—cyclooxygenase-2, prostaglandin-endoperoxide synthase 2; LOX—lipoxygenase; NOS—nitric oxide synthase; PPAR—peroxisome proliferator-activated receptors; GPR40—G-protein–coupled receptor 40; GPR120—G-protein–coupled receptor 120; RvDn DPA—resolvins derived from DPA n-3; Il-1β—interleukin 1β; IL-2—interleukin 2; IL-6—interleukin 6; INF-γ—interferon-γ; TNF-α—tumor necrosis factor α; TER—trans-epithelial electrical resistance. ↑ indicates an increase, ↓ indicates a decrease. Designed using elements by ©Canva, sparklestroke, Pixeden, iconsy, OpenClipart-Vectors via Canva.com (access date: 18 November 2024).</p>
Full article ">Figure 4
<p>Summary of n-3 PUFAs’ mechanisms of action: a schematic representation of the possible mechanisms through which n-3 PUFAs influence the gut–brain axis. DHA—docosahexaenoic acid; EPA—eicosapentaenoic acid; DPA—n-3 docosapentaenoic acid; HPA—hypothalamic-pituitary-adrenal; BBB—blood–brain barrier; SCFAs—short-chain fatty acids. ↑ indicates an increase, ↓ indicates a decrease. Designed using elements by ©Canva, sparklestroke, Pixeden, iconsy, OpenClipart-Vectors via Canva.com (access date: 18 November 2024).</p>
Full article ">
17 pages, 744 KiB  
Review
Mechanisms and Emerging Regulators of Neuroinflammation: Exploring New Therapeutic Strategies for Neurological Disorders
by Mi Eun Kim and Jun Sik Lee
Curr. Issues Mol. Biol. 2025, 47(1), 8; https://doi.org/10.3390/cimb47010008 - 26 Dec 2024
Viewed by 501
Abstract
Neuroinflammation is a complex and dynamic response of the central nervous system (CNS) to injury, infection, and disease. While acute neuroinflammation plays a protective role by facilitating pathogen clearance and tissue repair, chronic and dysregulated inflammation contributes significantly to the progression of neurodegenerative [...] Read more.
Neuroinflammation is a complex and dynamic response of the central nervous system (CNS) to injury, infection, and disease. While acute neuroinflammation plays a protective role by facilitating pathogen clearance and tissue repair, chronic and dysregulated inflammation contributes significantly to the progression of neurodegenerative disorders such as Alzheimer’s disease, Parkinson’s disease, and Multiple Sclerosis. This review explores the cellular and molecular mechanisms underlying neuroinflammation, focusing on the roles of microglia, astrocytes, and peripheral immune cells. Key signaling pathways, including NF-κB, JAK-STAT, and the NLRP3 inflammasome, are discussed alongside emerging regulators such as non-coding RNAs, epigenetic modifications, and the gut–brain axis. The therapeutic landscape is evolving, with traditional anti-inflammatory drugs like NSAIDs and corticosteroids offering limited efficacy in chronic conditions. Immunomodulators, gene and RNA-based therapeutics, and stem cell methods have all shown promise for more specific and effective interventions. Additionally, the modulation of metabolic states and gut microbiota has emerged as a novel strategy to regulate neuroinflammation. Despite significant progress, challenges remain in translating these findings into clinically viable therapies. Future studies should concentrate on integrated, interdisciplinary methods to reduce chronic neuroinflammation and slowing the progression of neurodegenerative disorders, providing opportunities for revolutionary advances in CNS therapies. Full article
(This article belongs to the Special Issue The Role of Neuroinflammation in Neurodegenerative Diseases)
Show Figures

Figure 1

Figure 1
<p>The major molecular pathways driving neuroinflammation in Alzheimer’s disease, highlighting their interconnected roles in perpetuating chronic inflammation. At the center of the diagram is neuroinflammation, which is fueled by four key pathways. The NF-κB pathway, activated by amyloid beta (Aβ) and tau proteins, promotes the production of pro-inflammatory cytokines such as TNF-α and IL-6, amplifying the inflammatory response. The NLRP3 inflammasome, triggered by Aβ, reactive oxygen species (ROS), and mitochondrial dysfunction, leads to the release of IL-1β and IL-18, further escalating neuroinflammation. The JAK-STAT pathway, induced by cytokines like IL-6, drives the activation of neurotoxic astrocytes, which contribute to neuronal damage. Finally, the MAPK pathway, stimulated by Aβ, tau, and oxidative stress, enhances ROS production and cytokine release, exacerbating oxidative damage and inflammation. Together, these pathways form a complex network that underpins the inflammatory processes observed in Alzheimer’s disease.</p>
Full article ">
23 pages, 3260 KiB  
Article
Clearing the Air on Pollutant Disruptions of the Gut–Brain Axis: Developmental Exposure to Benzo[a]pyrene Disturbs Zebrafish Behavior and the Gut Microbiome in Adults and Subsequent Generations
by Alexandra Alexiev, Ebony Stretch, Kristin D. Kasschau, Lindsay B. Wilson, Lisa Truong, Robyn L. Tanguay and Thomas J. Sharpton
Toxics 2025, 13(1), 10; https://doi.org/10.3390/toxics13010010 - 25 Dec 2024
Viewed by 332
Abstract
Developmental exposure to benzo[a]pyrene (BaP), a ubiquitous environmental pollutant, has been linked to various toxic effects, including multigenerational behavioral impairment. While the specific mechanisms driving BaP neurotoxicity are not fully understood, recent work highlights two important determinants of developmental BaP neurotoxicity: (1) the [...] Read more.
Developmental exposure to benzo[a]pyrene (BaP), a ubiquitous environmental pollutant, has been linked to various toxic effects, including multigenerational behavioral impairment. While the specific mechanisms driving BaP neurotoxicity are not fully understood, recent work highlights two important determinants of developmental BaP neurotoxicity: (1) the aryl hydrocarbon receptor (AHR), which induces host metabolism of BaP, and (2) the gut microbiome, which may interact with BaP to affect its metabolism, or be perturbed by BaP to disrupt the gut–brain axis. We utilized the zebrafish model to explore the role of AHR, the gut microbiome, and their interaction, on BaP-induced neurotoxicity. We tested (1) how developmental BaP exposure and AHR2 perturbation in zebrafish link to adult behavior, (2) how these variables associate with the structure and function of the adult zebrafish gut metagenome, and (3) whether these associations are multigenerational. Our findings reveal a reticulated axis of association between BaP exposure, developmental AHR2 expression, the zebrafish gut metagenome, and behavior. Results indicate that AHR2 is a key modulator of how BaP elicits neurotoxicity and microbiome dysbiosis. Additionally, this axis of association manifests generationally. These findings demonstrate the importance of studying pollutant–microbiome interactions and elucidate the role of specific host genes in neurotoxicity and dysbiosis. Full article
Show Figures

Figure 1

Figure 1
<p>The factorial experimental design and workflow used in this study. The table at the top left shows the sample groups representing the treatments we applied to zebrafish embryos in the F0 generation. The notation in the table is what is used in the graphs and in the text throughout the paper. The bar represents the workflow chronology, with colors showing each generation and red arrows with text for relevant procedures and collections, and ages of fish throughout.</p>
Full article ">Figure 2
<p>Summary heat map of statistical outcomes from behavioral and gut microbiome analyses where experimental treatments were compared relative to the control (AHR2Mo-/BaP-). (<b>A</b>) The behavioral metrics and how they associate with each experimental treatment, per generation. The KS statistic was used and is represented by the color gradient. (<b>B</b>) The gut microbiome alpha diversity metrics (based on 16S [taxonomic] and shotgun metagenomic [pathway] data) associated with the treatments, per generation. (<b>C</b>) The gut microbiome beta diversity metrics (again, for 16S and shotgun metagenomic data) associated with the treatments, per generation. In Panels B and C, the color gradient represents the linear model estimate (i.e., the slope of the association). Ns means not significant, and the asterisk represents significant associations. <a href="#app1-toxics-13-00010" class="html-app">Tables S2 and S3</a> (rows labeled “F0”) show expanded model results and <a href="#toxics-13-00010-t001" class="html-table">Table 1</a> has sample sizes.</p>
Full article ">Figure 3
<p>Box and whisker plot of the differences in zebrafish behavior across experimental groups and generations. Four behavior metrics (free swim distance, shoaling nnd, shoaling iid, and shoaling speed) were measured using a ZebraBox camera, then used to create a cumulative density function (CDF) distribution for each treatment, per generation. CDF distributions were evaluated via a Kolmogorov–Smirnov (KS) test comparing each treatment CDF distribution to that of the control (AHR2Mo-/BaP-), for each generation, per metric. Here, we present box and whisker plots of the area under the curve (AUC) of the CDF distribution for each treatment, organized by generation and metric, to portray trends from the KS test visually. The asterisks indicate significance (<span class="html-italic">p</span> ≤ 0.05) in experimental treatments relative to the control (AHR2Mo-/BaP-, dark purple), derived from the KS test described above (<a href="#app1-toxics-13-00010" class="html-app">Table S1</a>). Each color indicates the treatment. The center line of each box represents the median, the whiskers of each box are the standard deviation. Note that the y-axes for each plot show different scales, in order to more easily visualize trends within each data subset.</p>
Full article ">Figure 4
<p>Plots showing alpha and beta diversity metrics in taxonomic and pathway data per significant treatments (identified via regression approaches) for generation F0. (<b>A</b>) A box and whisker plot showing the ASV richness for each treatment (color) per sex (linear regression: exposure and morpholino interaction, coefficient = −6.8, <span class="html-italic">p</span> = 0.002; morpholino and sex interaction, coefficient = −5.3, <span class="html-italic">p</span> = 8.3 × 10<sup>−5</sup>). The center line is the median and the whiskers show the standard deviation. (<b>B</b>) A box and whisker plot showing pathway richness for each treatment (color) (linear regression: no significant differences). The center line is the median and the whiskers show the standard deviation. (<b>C</b>) A dbRDA ordination showing taxonomic beta diversity (Bray–Curtis dissimilarity) per treatment (color) and sex (shape) (PERMANOVA: <span class="html-italic">p</span> = 0.001 and 0.03, respectively). (<b>D</b>) A dbRDA ordination showing the pathway beta diversity (Bray–Curtis dissimilarity) per treatment (color) (PERMANOVA: <span class="html-italic">p</span> = 0.001 for all three covariates on the vectors).</p>
Full article ">Figure 5
<p>A heatmap of indicator taxa and pathways associated with model terms, per generation, with number (n) of significant associations in parentheses by each model term. (<b>A</b>) This shows indicator taxa listed on the y-axis while (<b>B</b>) shows the indicator pathways, organized by a dendrogram based on the Bray–Curtis matrix (right), and each organized by generation (top labels). Each y-axis lists the model terms used in a negative binomial model of each taxon/pathway relative abundance, along with the number (n) of significantly associated taxa (<b>A</b>) or gene pathways (<b>B</b>) shown in parentheses by the corresponding model term. Log(IRR) is represented by a color scale, which is the strength and direction of the association wherein positive numbers are dark green and show a positive association between the taxon/pathway relative abundance and the model term, and vice versa for negative numbers. The bottom set of colors in panel B indicates the broad category each pathway falls under (curated using available information from BioCyc and literature searches). Asterisks represent significant <span class="html-italic">p</span>-values from the model.</p>
Full article ">Figure 6
<p>Box and whisker plots of alpha diversity metrics per treatment in each generation, for taxonomic and pathway diversity. (<b>A</b>) Taxonomic richness and (<b>B</b>) pathway Shannon index were the only significant metrics (linear regression: <span class="html-italic">p</span> ≤ 0.05) among all alpha diversity metrics tested (see <a href="#app1-toxics-13-00010" class="html-app">Table S1</a> for statistics). Color in both graphs shows generations F0–F2 and x-axis lists the different treatments. The center line is the median and the whiskers show the standard deviation.</p>
Full article ">Figure 7
<p>Bray–Curtis dissimilarity of functional pathways across generations and treatments across the whole dataset. (<b>A</b>) Represents a dbRDA ordination where each point is a zebrafish gut microbiome sample, with color showing treatments and the ordination is faceted by generation. The ellipses show each generation per treatment grouping (n = 12) where color is treatment. (<b>B</b>) A dot plot of the median distance from the points in each generation–treatment combination to the F0 centroid for the corresponding treatment (see methods for details). Distances are taken from the dbRDA ordination in Panel A, normalized to the F0 centroid, and parsed by generation and treatment (color). Lines colored by treatment connect each treatment across generations. The vertical brackets show the standard deviation on each dot in the plot. (<b>C</b>) A box and whisker plot of the dispersion values for each treatment (colors) within each generation. Dispersion values have been log transformed for ease of visualization. The center line is the median and the whiskers show the standard deviation. The legend is common to all three panels.</p>
Full article ">
28 pages, 2501 KiB  
Review
The Microbiota–Gut–Brain Axis: Key Mechanisms Driving Glymphopathy and Cerebral Small Vessel Disease
by Che Mohd Nasril Che Mohd Nassir, Muhammad Danial Che Ramli, Mazira Mohamad Ghazali, Usman Jaffer, Hafizah Abdul Hamid, Muhammad Zulfadli Mehat and Zaw Myo Hein
Life 2025, 15(1), 3; https://doi.org/10.3390/life15010003 - 24 Dec 2024
Viewed by 684
Abstract
The human microbiota constitute a very complex ecosystem of microorganisms inhabiting both the inside and outside of our bodies, in which health maintenance and disease modification are the main regulatory features. The recent explosion of microbiome research has begun to detail its important [...] Read more.
The human microbiota constitute a very complex ecosystem of microorganisms inhabiting both the inside and outside of our bodies, in which health maintenance and disease modification are the main regulatory features. The recent explosion of microbiome research has begun to detail its important role in neurological health, particularly concerning cerebral small vessel disease (CSVD), a disorder associated with cognitive decline and vascular dementia. This narrative review represents state-of-the-art knowledge of the intimate, complex interplay between microbiota and brain health through the gut–brain axis (GBA) and the emerging role of glymphatic system dysfunction (glymphopathy) and circulating cell-derived microparticles (MPs) as mediators of these interactions. We discuss how microbial dysbiosis promotes neuroinflammation, vascular dysfunction, and impaired waste clearance in the brain, which are critical factors in the pathogenesis of CSVD. Further, we discuss lifestyle factors that shape the composition and functionality of the microbiota, focusing on sleep as a modifiable risk factor in neurological disorders. This narrative review presents recent microbiome research from a neuroscientific and vascular perspective to establish future therapeutic avenues in targeting the microbiota to improve brain health and reduce the burden of CSVD. Full article
(This article belongs to the Special Issue Microbiota in Health and Disease)
Show Figures

Figure 1

Figure 1
<p>Schematic diagram of gut–brain–microbiota axis. The brain and gut microbes communicate via many pathways. The central nervous system (CNS) interacts with gut microbes through various direct and indirect gut–brain axis (GBA) pathways. They include the immune pathway (including cytokines), short-chain fatty acids (SCFAs), and microbial metabolites; the neuroactive pathway (including neurotransmitters and neuroactive metabolites); the neural pathway (including the enteric nervous system, vagus nerve, and spinal nerves); and the endocrine pathway, i.e., the hypothalamic–pituitary–adrenal (HPA) axis. The HPA axis response involves hypothalamic neurons that release hormones such as corticotropin receptor hormone (CRH) into the portal circulation of the brain, resulting in the release of adrenocorticotropic hormone (ACTH), which initiates cortisol production and release. Cortisol regulates the neuroimmune signalling reactions.</p>
Full article ">Figure 2
<p>Schematic representation linking the microbiota–gut–brain axis and its potential impact on cerebral small vessel disease (CSVD). An imbalance in gut microbiota, also known as dysbiosis, contributes to decreased short-chain fatty acids (SCFAs) activity, increased secretion of lipopolysaccharides (LPS), trimethylamine-N-oxide (TMAO), and pro-inflammatory cytokines. These alterations can weaken the gut epithelial barrier and promote a systemic inflammatory response and vice versa. Dysbiosis-related molecules such as cytokines and gut-derived LPS enter the systemic circulation, activating the peripheral immune system and leading to further inflammation. These circulating factors influence the central nervous system through vagal signalling and systemic inflammation. In the brain, they contribute to disrupted microcirculation, i.e., endothelial dysfunction, increased production of reactive oxygen species (ROS), elevated cytokine levels, and blood-brain barrier (BBB) damage. This cascade of events ultimately exacerbates CSVD phenotypes by promoting neuroinflammation, vascular impairment, and potential cognitive decline.</p>
Full article ">Figure 3
<p>Schematic illustration of the complex interplay between the microbiota–gut–brain axis, cerebral small vessel disease (CSVD), sleep, and the glymphatic system. Dysbiosis in gut microbiota triggers inflammatory responses, producing cytokines, lipopolysaccharides (LPS) and reducing short-chain fatty acids (SCFAs) that affect the central nervous system (CNS). Disruptions in circadian rhythm impair glymphatic waste clearance, impacting glymphatic flow and microglial activity. Gut-microbial-, circulating cell- and astrocyte-derived microparticles (MPs) and inflammatory mediators contribute to blood–brain barrier (BBB) damage and thrombo-inflammation. These interactions promote CSVD phenotypes, including microbleeds and white matter changes, emphasizing the role of systemic inflammation and neuroimmune dysfunction in CSVD progression.</p>
Full article ">
16 pages, 1027 KiB  
Article
Development and Psychometric Properties of the Pain and Sensitivity Reactivity Scale in a Diverse Sample of Autistic People
by Agustín E. Martínez-González, Matti Cervin, José A. Piqueras, Lidia Infante-Cañete and Susana Pérez-Sánchez
Children 2024, 11(12), 1562; https://doi.org/10.3390/children11121562 - 23 Dec 2024
Viewed by 512
Abstract
Background: Recent studies indicate the need to examine how the gut microbiota–brain axis is implicated in pain, sensory reactivity and gastro-intestinal symptoms in autism spectrum disorder (ASD), but no scale exists that assesses all these constructs simultaneously. Methods: We created a pool of [...] Read more.
Background: Recent studies indicate the need to examine how the gut microbiota–brain axis is implicated in pain, sensory reactivity and gastro-intestinal symptoms in autism spectrum disorder (ASD), but no scale exists that assesses all these constructs simultaneously. Methods: We created a pool of 100 items based on the real-world experience of autistic people, and a multidisciplinary team and stakeholders reduced this pool to 50 items assessing pain, sensory hypersensitivity, and sensory hyposensitivity. In the present study, we present this new assessment tool, the Pain and Sensitivity Reactivity Scale (PSRS), and examine its psychometric properties in a sample of 270 individuals with autism spectrum disorder (ASD; mean age = 9.44, SD = 4.97), of which almost half (45%) had intellectual disability (ID). Results: A factorial model of three factors (pain, hyporeactivity, and hyperreactivity) and five specific factors for sensory hypo- and hyperreactivity, respectively, fitted the data well. Good to excellent internal consistency and adequate test–retest reliability was found for most PSRS scales. Sound psychometric properties were found in individuals with and without ID. Correlations with other measures of sensory sensitivity and pain indicated sound convergent validity. Conclusions: PSRS shows promise as a reliable measure to analyze pain and sensory reactivity in autistic people regardless of whether they have ID or not. The measure overcomes several limitations of previous assessment tools and includes variables that are important for the understanding of the gut microbiota–brain axis in ASD. Full article
Show Figures

Figure 1

Figure 1
<p>Theoretical model and structure of the PSRS. Copyright © 2023 Martinez-González A.E. [<a href="#B41-children-11-01562" class="html-bibr">41</a>].</p>
Full article ">
28 pages, 1980 KiB  
Review
Radiation-Induced Brain Injury: Mechanistic Insights and the Promise of Gut–Brain Axis Therapies
by Mengting Li, Fan Tong, Bian Wu and Xiaorong Dong
Brain Sci. 2024, 14(12), 1295; https://doi.org/10.3390/brainsci14121295 - 23 Dec 2024
Viewed by 692
Abstract
Radiation therapy is widely recognized as an efficacious modality for treating neoplasms located within the craniofacial region. Nevertheless, this approach is not devoid of risks, predominantly concerning potential harm to the neural structures. Adverse effects may encompass focal cerebral necrosis, cognitive function compromise, [...] Read more.
Radiation therapy is widely recognized as an efficacious modality for treating neoplasms located within the craniofacial region. Nevertheless, this approach is not devoid of risks, predominantly concerning potential harm to the neural structures. Adverse effects may encompass focal cerebral necrosis, cognitive function compromise, cerebrovascular pathology, spinal cord injury, and detriment to the neural fibers constituting the brachial plexus. With increasing survival rates among oncology patients, evaluating post-treatment quality of life has become crucial in assessing the benefits of radiation therapy. Consequently, it is imperative to investigate therapeutic strategies to mitigate cerebral complications from radiation exposure. Current management of radiation-induced cerebral damage involves corticosteroids and bevacizumab, with preclinical research on antioxidants and thalidomide. Despite these efforts, an optimal treatment remains elusive. Recent studies suggest the gut microbiota’s involvement in neurologic pathologies. This review aims to discuss the causes and existing treatments for radiation-induced cerebral injury and explore gut microbiota modulation as a potential therapeutic strategy. Full article
(This article belongs to the Section Neurosurgery and Neuroanatomy)
Show Figures

Figure 1

Figure 1
<p>The risk factors of RIBI.</p>
Full article ">Figure 2
<p>The mechanism of RIBI. Vascular damage and initial RN development (<b>a</b>). RN develops within the first 24 h post-radiotherapy due to radiation-induced vascular injury, characterized by endothelial apoptosis and necrosis, followed by capillary collapse and basement membrane thickening. This damage triggers HIF-1α expression, leading to VEGF secretion by glial cells and abnormal angiogenesis. Glial cell dysfunction (<b>b</b>). Radiotherapy induces glial cell activation and neuroinflammation, resulting in astrocyte proliferation and glial scar formation, impaired oligodendrocyte differentiation, and demyelination. Activated microglia release pro-inflammatory cytokines (IL-1, TNF-α, COX-2) and recruit CD8+ T cells, which secrete cytotoxic factors and worsen inflammation. Cellular senescence (<b>c</b>). Radiation induces premature senescence in endothelial and glial cells, marked by increased expression of senescence-associated genes (P21, P39), contributing to radiation-induced tissue aging and delayed RN. Neurogenesis dysfunction (<b>d</b>). Ionizing radiation reduces the self-renewal and differentiation capacity of neural stem cells, leading to decreased neurogenesis and long-term neurocognitive toxicity.</p>
Full article ">Figure 3
<p>The gut–brain axis and relevant disease.</p>
Full article ">
21 pages, 1130 KiB  
Review
Gut Microbiome Modulation of Glutamate Dynamics: Implications for Brain Health and Neurotoxicity
by Benjamin F. Gruenbaum, Kiran S. Merchant, Alexander Zlotnik and Matthew Boyko
Nutrients 2024, 16(24), 4405; https://doi.org/10.3390/nu16244405 - 22 Dec 2024
Viewed by 778
Abstract
The gut–brain axis plays an integral role in maintaining overall health, with growing evidence suggesting its impact on the development of various neuropsychiatric disorders, including depression. This review explores the complex relationship between gut microbiota and glutamate (Glu) regulation, highlighting its effect on [...] Read more.
The gut–brain axis plays an integral role in maintaining overall health, with growing evidence suggesting its impact on the development of various neuropsychiatric disorders, including depression. This review explores the complex relationship between gut microbiota and glutamate (Glu) regulation, highlighting its effect on brain health, particularly in the context of depression following certain neurological insults. We discuss how microbial populations can either facilitate or limit Glu uptake, influencing its bioavailability and predisposing to neuroinflammation and neurotoxicity. Additionally, we examine the role of gut metabolites and their influence on the blood–brain barrier and neurotransmitter systems involved in mood regulation. The therapeutic potential of microbiome-targeted interventions, such as fecal microbiota transplantation, is also highlighted. While much research has explored the role of Glu in major depressive disorders and other neurological diseases, the contribution of gut microbiota in post-neurological depression remains underexplored. Future research should focus on explaining the mechanisms linking the gut microbiota to neuropsychiatric outcomes, particularly in conditions such as post-stroke depression, post-traumatic brain-injury depression, and epilepsy-associated depression. Systematic reviews and human clinical studies are needed to establish causal relationships and assess the efficacy of microbiome-targeted therapies in improving the neuropsychiatric sequalae after neurological insults. Full article
(This article belongs to the Special Issue Diet and Microbiota–Gut–Brain Axis: A Novel Nutritional Therapy)
Show Figures

Figure 1

Figure 1
<p><b>Brain-to-blood glutamate efflux.</b> (<b>1</b>) In the presence of its enzyme co-substrate pyruvate, GPT catalyzes the reversible conversion of glutamate into its inactive form, 2-ketoglutarate, thereby reducing glutamate levels in the blood. This reduction generates a steep concentration gradient between the extracellular fluid and the blood, enhancing the brain-to-blood glutamate efflux rate. This leads to a reduction in elevated glutamate concentrations in the brain. As long as blood glutamate remains low, this efflux persists. Since the reaction converting glutamate to 2-ketoglutarate is reversible, an accumulation of 2-ketoglutarate can drive the enzyme to regenerate glutamate. (<b>2</b>) To sustain glutamate metabolism, 2-ketoglutarate is further degraded by the enzyme 2-ketoglutarate dehydrogenase. By enhancing the concentration gradient between blood and brain glutamate, the brain-to-blood glutamate transport is expedited, thereby mitigating excitotoxicity, associated with elevated brain glutamate levels. AKG, 2-ketoglutarate; AKGH, 2-ketoglutarate dehydrogenase; ALA, alanine; AST, aspartate; CO<sub>2</sub>, carbon dioxide; CoA, Coenzyme A; GLU, glutamate; GOT, glutamate-oxaloacetate transaminase; GPT, glutamate pyruvate transaminase; H<sup>+</sup>, hydrogen ion (proton), NAD<sup>+</sup>, nicotinamide adenine dinucleotide (oxidized form); NADH, nicotinamide adenine dinucleotide (reduced form); OA, oxaloacetate; PYR, pyruvate; succinyl-CoA, succinyl-coenzyme A.</p>
Full article ">Figure 2
<p><b>The disruption of the gut–brain axis and glutamate homeostasis during neurological insults.</b> Neurological insults overwhelm regulatory mechanisms, leading to excess glutamate in the brain, which contributes to neuropsychiatric sequelae, oxidative stress, mitochondrial toxicity, cytotoxic edema, and neuronal death. These interconnected pathways highlight the role of the gut–brain axis in mediating systemic and neurological effects during insults to the central nervous system. CSF, cerebral spinal fluid; GPT, glutamate pyruvate transaminase; GOT, glutamic oxaloacetic transaminase; TBI, traumatic brain injury.</p>
Full article ">
23 pages, 2016 KiB  
Article
The Gut Microbiota’s Role in Neurological, Psychiatric, and Neurodevelopmental Disorders
by Ioannis Alexandros Charitos, Angelo Michele Inchingolo, Laura Ferrante, Francesco Inchingolo, Alessio Danilo Inchingolo, Francesca Castellaneta, Antonella Cotoia, Andrea Palermo, Salvatore Scacco and Gianna Dipalma
Nutrients 2024, 16(24), 4404; https://doi.org/10.3390/nu16244404 - 22 Dec 2024
Viewed by 833
Abstract
Aim: This article aims to explore the role of the human gut microbiota (GM) in the pathogenesis of neurological, psychiatric, and neurodevelopmental disorders, highlighting its influence on health and disease, and investigating potential therapeutic strategies targeting GM modulation. Materials and Methods: A comprehensive [...] Read more.
Aim: This article aims to explore the role of the human gut microbiota (GM) in the pathogenesis of neurological, psychiatric, and neurodevelopmental disorders, highlighting its influence on health and disease, and investigating potential therapeutic strategies targeting GM modulation. Materials and Methods: A comprehensive analysis of the gut microbiota’s composition and its interaction with the human body, particularly, its role in neurological and psychiatric conditions, is provided. The review discusses factors influencing GM composition, including birth mode, breastfeeding, diet, medications, and geography. Additionally, it examines the GM’s functions, such as nutrient absorption, immune regulation, and pathogen defense, alongside its interactions with the nervous system through the gut–brain axis, neurotransmitters, and short-chain fatty acids (SCFAs). Results: Alterations in the GM are linked to various disorders, including Parkinson’s disease, multiple sclerosis, depression, schizophrenia, ADHD, and autism. The GM influences cognitive functions, stress responses, and mood regulation. Antibiotic use disrupts GM diversity, increasing the risk of metabolic disorders, obesity, and allergic diseases. Emerging therapies such as probiotics, prebiotics, and microbiota transplantation show promise in modulating the GM and alleviating symptoms of neurological and psychiatric conditions. Conclusions. The modulation of the GM represents a promising approach for personalized treatment strategies. Further research is needed to better understand the underlying mechanisms and to develop targeted therapies aimed at restoring GM balance for improved clinical outcomes. Full article
(This article belongs to the Special Issue Implications of Diet and the Gut Microbiome in Neuroinflammation)
Show Figures

Figure 1

Figure 1
<p>The main taxa found during childhood. These taxa and some of their species are implicated in neurological and psychiatric diseases due to their increased population in the microbiota, such as <span class="html-italic">Actinomycetota</span> (<span class="html-italic">Bifidobacterium</span> spp.), <span class="html-italic">Verrucomicrobia</span> (<span class="html-italic">Akkermansia</span> spp.), <span class="html-italic">Bacillota</span> (<span class="html-italic">Faecalibacterium</span> spp.), <span class="html-italic">Bacteroidota</span> (such as <span class="html-italic">Prevotella</span> spp.), and <span class="html-italic">Fusobacteriota</span> (such as <span class="html-italic">Fusobacterium</span> spp.). Credits: Original figure by I.A. Charitos.</p>
Full article ">Figure 2
<p>The main bacteria at level of families found during childhood in the gut microbiota. Several species from these families have a connection with neurological and psychiatric diseases or disorders such as <span class="html-italic">Bacteroides</span> spp., <span class="html-italic">Doria</span> spp., <span class="html-italic">Bifidobacteria</span> spp., <span class="html-italic">Prevotella</span> spp. and others.</p>
Full article ">Figure 3
<p>The main genera found in pediatric population.</p>
Full article ">Figure 4
<p>The three enterotypes are recognized based on the predominant bacterium: (1) <span class="html-italic">Bacteroides</span>, (2) <span class="html-italic">Prevotella</span>, and (3) <span class="html-italic">Ruminococcus</span>. In the first intestinal type, <span class="html-italic">Slackia</span>, <span class="html-italic">Parabacteroides</span>, <span class="html-italic">Clostridiales</span>, <span class="html-italic">Alkaliphilus</span>, <span class="html-italic">Lactobacillus</span>, <span class="html-italic">Catenibacterium</span>, and <span class="html-italic">Geobacter coexist</span>. <span class="html-italic">Eggerthella</span>, <span class="html-italic">Veillonella</span>, <span class="html-italic">Ruminococcaceae</span>, <span class="html-italic">Holdemania</span>, <span class="html-italic">Peptostreptococcaceae</span>, <span class="html-italic">Staphylococcus</span>, <span class="html-italic">Leuconostoc</span>, <span class="html-italic">Desulfovibrionaceae</span>, <span class="html-italic">Rhodospirillum</span>, <span class="html-italic">Helicobacter</span>, <span class="html-italic">Escherichia</span>, <span class="html-italic">Shigella</span>, and <span class="html-italic">Akkermansia muciniphila</span> also occur in the second intestinal type. Credits: Original figure by I.A. Charitos The third enteric type also includes <span class="html-italic">Gordonibacter</span>, <span class="html-italic">Sphingobacterium</span>, <span class="html-italic">Staphylococcus</span>, <span class="html-italic">Marvinbryantia</span>, <span class="html-italic">Symbiobacterium</span>, <span class="html-italic">Dialister</span>, and <span class="html-italic">Akkermansia muciniphila</span>. Credits: Original figure by I.A. Charitos.</p>
Full article ">Figure 5
<p>The figure describes the hypotheses of how gut dysbiosis, due to emotional or stressful causes or not, can influence the bidirectional communication of the GBA, causing direct and indirect effects on the ENS and vice versa. Credits: Original figure by I.A. Charitos.</p>
Full article ">
32 pages, 2635 KiB  
Review
The Role of Gut Microbiome in Irritable Bowel Syndrome: Implications for Clinical Therapeutics
by Yucui Zhao, Shixiao Zhu, Yingling Dong, Tian Xie, Zhiqiang Chai, Xiumei Gao, Yongna Dai and Xiaoying Wang
Biomolecules 2024, 14(12), 1643; https://doi.org/10.3390/biom14121643 - 21 Dec 2024
Viewed by 1040
Abstract
Irritable bowel syndrome (IBS) is a functional gastrointestinal disorder (FGID) characterized by chronic or recurrent gastrointestinal symptoms without organic changes, and it is also a common disorder of gut–brain interaction (DGBIs).. The symptoms of IBS not only affect the quality of life for [...] Read more.
Irritable bowel syndrome (IBS) is a functional gastrointestinal disorder (FGID) characterized by chronic or recurrent gastrointestinal symptoms without organic changes, and it is also a common disorder of gut–brain interaction (DGBIs).. The symptoms of IBS not only affect the quality of life for individual patients but also place a significant burden on global healthcare systems. The lack of established and universally applicable biomarkers for IBS, along with the substantial variability in symptoms and progression, presents challenges in developing effective clinical treatments. In recent years, preclinical and clinical studies have linked the pathogenesis of IBS to alterations in the composition and function of the intestinal microbiota. Within the complex microbial community of the gut, intricate metabolic and spatial interactions occur among its members and between microbes and their hosts. Amid the multifaceted pathophysiology of IBS, the role of intestinal microenvironment factors in symptom development has become more apparent. This review aims to delve into the changes in the composition and structure of the gut microbiome in individuals with IBS. It explores how diet-mediated alterations in intestinal microbes and their byproducts play a role in regulating the pathogenesis of IBS by influencing the “brain-gut” axis, intestinal barrier function, immune responses, and more. By doing so, this review seeks to lay a theoretical foundation for advancing the development of clinical therapeutics for IBS. Full article
(This article belongs to the Section Molecular Medicine)
Show Figures

Figure 1

Figure 1
<p>Interactions and connections between the gut microbiota, metabolites, and cellular components with IBS. Imbalance in the intestinal gut microbiota can lead to symptoms such as diarrhea and abdominal pain. Gut microbiota metabolites, such as SCFAs, can directly affect intestinal movement, BAs affect intestinal movement and intestinal barrier function via the FXR/TGR pathway, Trp affects visceral hypersensitivity and intestinal inflammation through conversion to 5-HT and indol, and histamine affects intestinal inflammation through its receptors. Neurotransmitters (NA, DA, GABA), flavonoids, and proteases can also influence visceral hypersensitivity and inflammation. Gut microbiota components, lipopeptide, peptidoglycans, and LPS act on the TLR pathway or stimulate intestinal macrophages to secrete BMP2 and inflammatory factors (COX2, PGE2, NO) to affect intestinal movement.</p>
Full article ">Figure 2
<p>FODMAP classification of common foods.</p>
Full article ">Figure 3
<p>The effect of diet on IBS by acting on the gut microbiota and its metabolites. Low FODMAP promotes Actinobacteria and inhibits <span class="html-italic">B. adolescentis</span> and gas-producing bacteria to improve intestinal barrier dysfunction and inhibit inflammation. A fish and omega-3 oil diet, via balancing intestinal F/B, increases probiotic abundance and plays an antidepressant role. Vitamin D supplementation can reduce the abundance of <span class="html-italic">Pseudomonas</span> spp., <span class="html-italic">Escherichia</span>, and <span class="html-italic">Shigella</span> spp. Iron restriction increases levels of <span class="html-italic">Lactobacillus</span> and <span class="html-italic">Enterococcus</span>, promotes SCFA secretion and anti-anxiety and -inflammation effects, and improves intestinal microenvironment.</p>
Full article ">Figure 4
<p>Dysbiosis within the microbiome triggers IBS via neuroimmune networks. Gut microbes directly produce 5-HT or transmit signals to ECs, inducing TPH transcription, promoting 5-HT synthesis, and participating in brain–gut interactions. Intestinal endocrine cells secrete GLP-1, CCK, and PYY in response to stimulation by microbial metabolites, serving as a signal transducer in the brain–gut–microbiome axis. LPS and PGN enter the brain through systemic circulation, leading to chronic neuroinflammation and visceral hypersensitivity. <span class="html-italic">Campylobacter jejuni</span> activate the vagus nerve, resulting in increased expression of c-Fos. <span class="html-italic">L. rhamnosus GG</span>, <span class="html-italic">L. casei</span>, <span class="html-italic">L. johnsonii</span>, etc., activating macrophages, NK cells, and T cells, thereby enhancing the intestinal mucosal immune system.</p>
Full article ">Figure 5
<p>Gut microbes help protect host intestinal barrier function by directly or indirectly secreting bioactive molecules. <span class="html-italic">Bacteroides</span> spp., <span class="html-italic">Clostridium coccoides</span>, and <span class="html-italic">Lactobacillus</span> spp. can promote the secretion of slgA, while slgA can inhibit segmented filamentous bacteria. Gut microbes secrete bacteriocin to inhibit the colonization and growth of pathogenic bacteria and play an anti-inflammatory role. Mucus-degrading bacteria affect barrier protection function and microbial colonization by altering mucus properties. RegIII-γ and α-defensins expression induced by the MyD88 signaling pathway strengthens the intestinal mucosa against bacterial invasion. The microbial metabolite meprin β promotes the dissociation of MUC2 from goblet cells, releasing it into the intestinal lumen to maintain intestinal homeostasis. <span class="html-italic">L. reuteri</span> can promote intestinal epithelial regeneration and repair of damaged intestinal tract by activating Wnt/β-catenin pathway. Intestinal macrophages and DCs sense microbial signals, prompting differentiation and activation of T cell subsets (Th1, Th2, Th17 or Treg) to produce cytokines.</p>
Full article ">
17 pages, 2717 KiB  
Review
Enzymatic Regulation of the Gut Microbiota: Mechanisms and Implications for Host Health
by Zipeng Jiang, Liang Mei, Yuqi Li, Yuguang Guo, Bo Yang, Zhiyi Huang and Yangyuan Li
Biomolecules 2024, 14(12), 1638; https://doi.org/10.3390/biom14121638 - 20 Dec 2024
Viewed by 648
Abstract
The gut microbiota, a complex ecosystem, is vital to host health as it aids digestion, modulates the immune system, influences metabolism, and interacts with the brain-gut axis. Various factors influence the composition of this microbiota. Enzymes, as essential catalysts, actively participate in biochemical [...] Read more.
The gut microbiota, a complex ecosystem, is vital to host health as it aids digestion, modulates the immune system, influences metabolism, and interacts with the brain-gut axis. Various factors influence the composition of this microbiota. Enzymes, as essential catalysts, actively participate in biochemical reactions that have an impact on the gut microbial community, affecting both the microorganisms and the gut environment. Enzymes play an important role in the regulation of the intestinal microbiota, but the interactions between enzymes and microbial communities, as well as the precise mechanisms of enzymes, remain a challenge in scientific research. Enzymes serve both traditional nutritional functions, such as the breakdown of complex substrates into absorbable small molecules, and non-nutritional roles, which encompass antibacterial function, immunomodulation, intestinal health maintenance, and stress reduction, among others. This study categorizes enzymes according to their source and explores the mechanistic principles by which enzymes drive gut microbial activity, including the promotion of microbial proliferation, the direct elimination of harmful microbes, the modulation of bacterial interaction networks, and the reduction in immune stress. A systematic understanding of enzymes in regulating the gut microbiota and the study of their associated molecular mechanisms will facilitate the application of enzymes to precisely regulate the gut microbiota in the future and suggest new therapeutic strategies and dietary recommendations. In conclusion, this review provides a comprehensive overview of the role of enzymes in modulating the gut microbiota. It explores the underlying molecular and cellular mechanisms and discusses the potential applications of enzyme-mediated microbiota regulation for host gut health. Full article
(This article belongs to the Special Issue Novel Antimicrobial Strategies for Animal Health)
Show Figures

Figure 1

Figure 1
<p>Classification of enzymes and their functions.</p>
Full article ">Figure 2
<p>Mechanisms of enzyme regulation of gut microbes. The main ways in which enzymes regulate the intestinal microbiota include (1) stimulation of microbial growth: enzymes can stimulate the growth of beneficial gut microbes; (2) direct microbial killing: certain enzymes can kill gut microbes directly; (3) interference with microbial networks: enzymes can disrupt microbial networks, such as quorum sensing (QS), which is a communication system used by microbes to coordinate their behavior; and (4) alleviating the immune stress: the use of enzymes to reduce the occurrence of immune stress is through the degradation of resemble immunogenic substances.</p>
Full article ">
32 pages, 6874 KiB  
Article
Alterations in Gut Microbiota Composition Are Associated with Changes in Emotional Distress in Children with Obstructive Sleep Apnea
by Chung-Guei Huang, Wan-Ni Lin, Li-Jen Hsin, Yu-Shu Huang, Li-Pang Chuang, Tuan-Jen Fang, Hsueh-Yu Li, Terry B. J. Kuo, Cheryl C. H. Yang, Chin-Chia Lee and Li-Ang Lee
Microorganisms 2024, 12(12), 2626; https://doi.org/10.3390/microorganisms12122626 - 18 Dec 2024
Viewed by 471
Abstract
Emerging evidence underscores the pivotal role of the gut microbiota in regulating emotional and behavioral responses via the microbiota–gut–brain axis. This study explores associations between pediatric obstructive sleep apnea (OSA), emotional distress (ED), and gut microbiome alterations before and after OSA treatment. Sixty-six [...] Read more.
Emerging evidence underscores the pivotal role of the gut microbiota in regulating emotional and behavioral responses via the microbiota–gut–brain axis. This study explores associations between pediatric obstructive sleep apnea (OSA), emotional distress (ED), and gut microbiome alterations before and after OSA treatment. Sixty-six children diagnosed with OSA via polysomnography participated, undergoing adenotonsillectomy alongside routine educational sessions. ED was assessed using the OSA-18 questionnaire, categorizing participants into high ED (scores ≥ 11, 52%) and low ED (scores < 11, 48%) groups. Gut microbiome analysis revealed significant diversity differences, with high ED linked to a reduced Shannon index (p = 0.03) and increased beta diversity (p = 0.01). Three months post-treatment, significant improvements were observed in OSA symptoms, ED scores, and gut microbiome alpha diversity metrics among 55 participants (all p < 0.04). Moreover, changes in the relative abundances of Veillonella, Bifidobacterium, Flavonifractor, and Agathobacter, as well as ultra-low frequency power and low frequency power of sleep heart rate variability, were independently associated with ED score alterations. These findings underscore the gut microbiome’s critical role in the emotional and behavioral symptoms associated with pediatric OSA, suggesting that microbiome-targeted interventions could complement traditional treatments for ED reduction and emphasizing the need for further research. Full article
(This article belongs to the Section Gut Microbiota)
Show Figures

Figure 1

Figure 1
<p>Case flow diagram. Abbreviations: HRV, heart rate variability; OSA, obstructive sleep apnea.</p>
Full article ">Figure 2
<p>Distributions of the gut microbiota amplicon sequence variants (ASVs) between high emotional distress (ED) and low ED groups in pediatric obstructive sleep apnea. (<b>a</b>) The UpSet plot illustrates the unique and shared ASVs between the low ED group (3485 ASVs) and the high ED group (3258 ASVs) at the pre-treatment stage. (<b>b</b>) The UpSet plot illustrates the unique and shared ASVs between the low ED group (3238 ASVs) and the high ED group (3077 ASVs) at the post-treatment stage.</p>
Full article ">Figure 3
<p>Gut microbial distributions of the top 30 genera between the high and low emotional distress (ED) groups in pediatric obstructive sleep apnea. (<b>a</b>) The heatmap illustrates significant differences in the microbial distribution of the top 30 genera between the high and low ED groups at the pre-treatment stage, with statistical significance confirmed by permutational multivariate analysis of variance with Benjamini–Hochberg correction (<span class="html-italic">p</span> = 0.01). (<b>b</b>) The heatmap illustrates a similar microbial distribution of the top 30 genera between the high and low ED groups at the post-treatment stage (<span class="html-italic">p</span> = 0.33).</p>
Full article ">Figure 3 Cont.
<p>Gut microbial distributions of the top 30 genera between the high and low emotional distress (ED) groups in pediatric obstructive sleep apnea. (<b>a</b>) The heatmap illustrates significant differences in the microbial distribution of the top 30 genera between the high and low ED groups at the pre-treatment stage, with statistical significance confirmed by permutational multivariate analysis of variance with Benjamini–Hochberg correction (<span class="html-italic">p</span> = 0.01). (<b>b</b>) The heatmap illustrates a similar microbial distribution of the top 30 genera between the high and low ED groups at the post-treatment stage (<span class="html-italic">p</span> = 0.33).</p>
Full article ">Figure 4
<p>Gut microbiome stratification between high and low emotional distress (ED) groups in pediatric obstructive sleep apnea. (<b>a</b>) The scatter boxplot illustrates a significantly lower Shannon index in the high ED group compared to the low ED group at the pre-treatment stage (<span class="html-italic">p</span> = 0.03). (<b>b</b>) Principal coordinates analysis (PCoA) highlights significant differences in Bray–Curtis distances between the two ED groups at the pre-treatment stage. (<b>c</b>) The scatter boxplot shows significantly greater microbial community variability in the high ED group compared to the low ED group at the pre-treatment stage (<span class="html-italic">p</span> = 0.01). (<b>d</b>) The scatter boxplot indicates comparable Shannon indices between the two ED groups at the post-treatment stage (<span class="html-italic">p</span> = 0.87). (<b>e</b>) PCoA highlights differences in Bray–Curtis distances between the two ED groups at the post-treatment stage. (<b>f</b>) The scatter boxplot shows a significant increase in microbial community variability within the high ED group compared to the low ED group at the post-treatment stage (<span class="html-italic">p</span> = 0.01). Shannon indices were compared using the independent-samples <span class="html-italic">t</span>-test, and Bray–Curtis distances were compared using permutational multivariate analysis of variance with Benjamini–Hochberg correction.</p>
Full article ">Figure 5
<p>Heatmap of significant associations (−log(q value) × sign(coefficient)) between the top 30 genera and variables of interest. (<b>a</b>) Heatmap showing significant associations across five genera, emotional distress (ED) score, and male sex. (<b>b</b>) Heatmap depicting significant associations across five genera, semi-skimmed milk, whole-meal bread, and rice consumption. (<b>c</b>) Heatmap illustrating significant associations across three genera, very low frequency (VLF) power, and ultra-low frequency (ULF) power. Positive signs indicate enrichment, and negative signs indicate depletion of the corresponding genera relative to increasing variables of interest. The analysis was conducted using the Maaslin2 R package (v1.15.1).</p>
Full article ">
Back to TopTop