Open Access

Role of the gut-brain axis in neurological diseases: Molecular connections and therapeutic implications (Review)

  • Authors:
    • Hongjuan Zhang
    • Jia Luan
    • Ling He
    • Xianfang Pan
    • Hujun Zhang
    • Yujie Li
    • Haiyan Li
  • View Affiliations

  • Published online on: September 11, 2025     https://doi.org/10.3892/ijmm.2025.5633
  • Article Number: 192
  • Copyright: © Zhang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The gut-brain axis, a bidirectional communication system involving the central nervous system, enteric nervous system and gastrointestinal tract, plays a pivotal role in neurological health and disease. The present review synthesizes current evidence on its anatomical foundations, functional mechanisms and the role of gut microbiota in neurological diseases. It is explored how dysregulation of the gut-brain axis contributes to neurodegenerative, neuroinflammatory, neurodevelopmental, cerebrovascular and epileptic disorders through molecular mechanisms including neuroinflammation, neurotransmitter modulation, oxidative stress, and blood-brain barrier changes. Furthermore, therapeutic strategies targeting this axis, such as dietary interventions, probiotics, pharmacological approaches and emerging therapies were evaluated. The present review highlights the potential for developing biomarkers and personalized medicine approaches based on gut-brain axis components, while acknowledging translational challenges. However, significant heterogeneity in probiotic strains, FMT protocols, and patient microbiota baselines limits immediate translation. Future trials must address these variables through standardized designs and multi-omics biomarker integration.

Introduction

The gut-brain axis is a bidirectional communication system that connects the gastrointestinal tract with the central nervous system (CNS). This complex network comprises several key components that facilitate constant interactions between the gut and the brain (1). The enteric nervous system, often referred to as the 'second brain' is embedded within the walls of the gastrointestinal tract and contains over 500 million neurons that functioning independently to regulate gastrointestinal motility, secretion and blood flow (2). The vagus nerve, a crucial component of the parasympathetic nervous system, serves as a direct neural pathway between the gut and the brain, transmitting signals bidirectionally (3). Additionally, the gut microbiota, which consists of trillions of microorganisms, plays a vital role in shaping the gut environment and influencing both local and systemic immune responses (4). Immune pathways, including those involving gut-associated lymphoid tissue and circulating immune cells, further contribute to the communication between the gut and the brain, thus enabling the exchange of immune signals and the modulation of inflammatory responses throughout the body (4).

The gut-brain axis employs multiple bidirectional communication pathways to maintain homeostasis and coordinate responses to internal and external stimuli (5). Neural pathways primarily involve the vagus nerve, which transmits signals from the gut to the brainstem and vice versa, influencing functions such as appetite, digestion and stress responses (6). The endocrine system also plays a significant role, with gut hormones such as ghrelin, leptin and peptides being released in response to nutrient intake and acting on distant targets in the brain to regulate feeding behavior, energy balance and metabolic processes (7). Immune pathways facilitate communication through the production of cytokines, chemokines and other immune mediators by gut-resident immune cells, which can enter the circulation and affect CNS function (8). Conversely, the brain can modulate immune responses in the gut through the release of neuropeptides and hormones, thus creating a dynamic interplay that impacts both gastrointestinal and neurological health (9).

Emerging epidemiological evidence highlights significant associations between gut dysbiosis and various neurological diseases (10). Previous studies have demonstrated alterations in gut microbiota composition in patients with Alzheimer's disease (AD), characterized by decreased diversity and changes in specific bacterial taxa such as Bifidobacterium and Lactobacillus (11-13). Similarly, in Parkinson's disease (PD), a shift towards pro-inflammatory microbiota profiles has been observed, with correlations between microbial composition and disease severity (14). Epidemiological data also suggest that individuals with inflammatory bowel disease (IBD) have an increased risk of developing neurological conditions such as multiple sclerosis (MS) and depression, indicating a potential shared pathophysiology (15,16). Furthermore, the incidence of autism spectrum disorders (ASD) have been linked to early-life gut microbiota perturbations, with numerous patients with ASD exhibiting gastrointestinal symptoms and altered microbial communities (17,18). These findings underscore the importance of gut microbiota in neurological health and point towards potential therapeutic targets for modulating the gut-brain axis.

The present review aims to comprehensively synthesize current knowledge on the molecular mechanisms underlying the gut-brain axis in neurological diseases and evaluate existing and emerging therapeutic strategies targeting this axis. By integrating findings from recent preclinical and clinical studies, it is aimed to provide a deeper understanding of how perturbations in the gut-brain axis contribute to disease pathogenesis and progression. Furthermore, current therapeutic approaches are critically assessed, including dietary interventions, probiotics, prebiotics and pharmacological agents, highlighting their efficacy, limitations and potential for future development. This synthesis of evidence will not only offer valuable insights for researchers and clinicians but also identify gaps in knowledge and suggest directions for future investigation, ultimately aiming to advance the translation of gut-brain axis research into effective clinical applications for neurological diseases.

Methods

A comprehensive, non-systematic narrative review was conducted by querying PubMed (https://pubmed.ncbi.nlm.nih.gov/) from inception to 30 June 2025. Search terms combined MeSH and free-text keywords ('gut-brain axis', 'microbiome-gut-brain axis', 'gut microbiota') with disease-specific terms ['neurodegeneration', 'Parkinson', 'Alzheimer', 'MS', 'epilepsy', 'autism', 'stroke', 'traumatic brain injury' (TBI)] and mechanistic terms ('molecular mechanism', 'therapy', 'probiotic', 'prebiotic', 'fecal microbiota transplantation (FMT)', 'short-chain fatty acid'). English, full-text, peer-reviewed original articles, systematic reviews, meta-analyses and phase I-III clinical trials were retained. Animal or in vitro mechanistic studies were included only when corroborated by human data or published within the last twelve years. Reference lists and trial registries (ClinicalTrials.gov, WHO ICTRP) were hand-searched for additional records. Data were narratively synthesized without formal meta-analysis.

Gut-brain axis: Anatomical and functional foundations

The bidirectional communication network between gastrointestinal and CNS involves integrated anatomical structures and signaling pathways. Key neural connections include the vagus nerve and spinal cord routes, while endocrine signaling features brain-gut peptides such as ghrelin and leptin. Parallel communication occurs via microbial metabolites, immune mediators and neuroendocrine mechanisms [for example, hypothalamic-pituitary-adrenal (HPA) axis activation], collectively enabling dynamic crosstalk that regulates neurological function (Table I).

Table I

Studies of gut-brain axis disorders in nervous system diseases.

Table I

Studies of gut-brain axis disorders in nervous system diseases.

Authors, yearRelated diseasesObject of studyMain findings(Refs.)
Sampson et al, 2016PDPD modelAntibiotic treatment could reduce motor dysfunction and microglial activation, indicating a potential therapeutic avenue for PD by targeting gut microbiota.(50)
Perez-Pardo et al, 2019PDPD modelTLR4 signaling plays a crucial role in the gut-brain axis in PD. The research suggested that TLR4 might be a promising therapeutic target for PD treatment.(51)
Srivastav et al, 2019PDMPTP and rotenone-inducedA probiotics mixture increases butyrate levels, which rescues nigral dopaminergic neurons from neurotoxicity induced by MPTP and rotenone. This finding highlights the potential of probiotics in PD therapy.(52)
Honarpisheh et al, 2020ADTg2576 miceDysregulated gut homeostasis precedes amyloid-β accumulation in the brain. This suggests that gut dysbiosis might be an early indicator or contributor to AD pathology.(54)
Kim et al, 2021ADAPP/PS1 miceTransplantation of gut microbiota from AD mouse models impairs memory function and neurogenesis in recipient mice. This indicates that gut microbiota may influence cognitive decline in AD.(55)
Shi et al, 2021ADAPP/PS1 miceA fiber-deprived diet causes cognitive impairment and hippocampal microglia-mediated synaptic loss through the gut microbiota and metabolites. This study emphasizes the impact of diet on gut microbiota and AD. progression(56)
Castelli et al, 2021ADNot specifiedProbiotics may reduce neuroinflammation, offering a potential therapeutic strategy for AD.(58)
Yuan et al, 2019ADAPP/PS1 miceSesamol exerts protective effects via the microbiome-gut-brain axis, potentially benefiting AD treatment.(59)
Shi et al, 2020ADDiet-induced obese MicePrebiotics prevent neuroinflammation and cognitive decline by improving the gut microbiota-brain axis. This suggests a dietary approach to AD management.(60)
Bianchimano et al, 2022Multiple sclerosisMiceSpecific gut commensals modulate neuroinflammation, highlighting the gut microbiota's role in MS pathogenesis and potential therapeutic targets within the gut microbiota.(61)
Celorrio and Friess, 2022TBIMiceThe gut-brain axis impacts neuroinflammation following TBI. This study suggests that targeting the gut microbiota could mitigate post-injury inflammation.(62)
Lee et al, 2022Cognitive impairmentPolystyrene microplastics exposure modelExposure to polystyrene microplastics impairs hippocampus-dependent learning and memory in mice, with gut microbiota alterations preceding neuroinflammatory changes. This indicates environmental factors' impact on cognitive health via the gut-brain axis.(65)
Yuan et al, 2022 NeuroinflammationAPP/PS1 miceTreadmill exercise modulates intestinal microbes and suppresses LPS displacement, alleviating neuroinflammation. This demonstrates exercise's beneficial effects on gut-brain axis modulation.(66)
Hu et al, 2023 NeuroinflammationTBI modelFMT inhibits neuroinflammation following TBI by regulating the gut-brain axis, indicating its potential as a treatment for neuroinflammatory conditions.(67)
Li et al, 2023Cognitive deficitsRatsLactobacillus rhamnosus GG ameliorates noise-induced cognitive deficits and systemic inflammation by modulating the gut-brain axis. This suggests probiotics' utility in cognitive deficit management.(68)
de Theije et al, 2014ASDASD modelIntestinal inflammation is present in a murine model of ASD, suggesting a potential link between gut dysfunction and behavioral abnormalities.(71)
Slob et al, 2021ADHD and ASDDiscordant twin studyEarly-life antibiotic use is linked to increased risk of ADHD and ASD, indicating that gut microbiota perturbations during critical developmental windows may have long-term neurological consequences.(81)
Bundgaard-Nielsen et al, 2023Neurodevelopmental disordersChildren and adolescents with ADHD and ASDDistinct microbiota compositions are found in children and adolescents with ADHD and ASD, suggesting shared and unique gut microbiota signatures across neurodevelopmental disorders.(82)
Gonçalves et al, 2020Behavioral changesYoung zebrafishExposure to high doses of amoxicillin causes behavioral changes and oxidative stress in young zebrafish, suggesting potential developmental consequences of antibiotic use.(84)
Volkova et al, 2021Brain developmentEarly-life penicillin exposure modelEarly-life penicillin exposure alters frontal cortex and amygdala gene expression, highlighting the developing brain's sensitivity to gut microbiota perturbations.(85)
Benakis et al, 2016Ischemic strokeIschemic stroke modelCommensal microbiota affects stroke outcome via intestinal γδ T cells, highlighting the role of gut immunity in stroke pathogenesis.(86)
Dou et al, 2019Focal cerebral ischemia/reperfusion injuryMiceResveratrol exerts neuroprotection against focal cerebral ischemia/reperfusion injury in mice through a mechanism targeting the gut-brain axis.(87)
Park et al, 2020Reproductive senescence and ischemic strokeFemale ratsReproductive senescence and ischemic stroke remodel the gut microbiome and modulate the effects of estrogen treatment in female rats.(90)
Sadler et al, 2020StrokeMiceSCFAs improve poststroke recovery via immunological mechanisms, suggesting a potential therapeutic avenue.(91)
Aswendt et al, 2021Acute brain ischemiaMiceGut microbiota modulation affects brain network connectivity under physiological conditions and after acute brain ischemia.(92)
Henry et al, 2021Acute ischemic stroke patientsSCFAs at thrombectomySCFAs taken at the time of thrombectomy in acute ischemic stroke patients are associated with inflammatory markers and worse symptoms at discharge.(94)
Jiang et al, 2021Post-stroke depressionGut microbiome and lipid metabolismAlterations in the gut microbiome and correlated lipid metabolism are found in post-stroke depression, indicating potential metabolic links between gut dysbiosis and neurological symptoms.(95)
Huang et al, 2019Cerebral palsy and epilepsyChildren with cerebral palsy and epilepsyDistinct gut microbiota composition and functional categories are observed in children with cerebral palsy and epilepsy, suggesting a potential link between gut dysfunction and seizure activity.(96)
Lindefeldt et al, 2019Severe epilepsyChildren with severe epilepsyThe ketogenic diet influences the taxonomic and functional composition of the gut microbiota in children with severe epilepsy, indicating that dietary interventions can modulate gut microbiota to reduce seizure frequency.(97)
He et al, 2017Epilepsy and Crohn's diseaseEpilepsy and Crohn's diseaseFMT cured epilepsy in a patient with Crohn's disease, highlighting the potential of microbiota-based therapies.(98)
Avorio et al, 2021EpilepsyEpilepsy patientsFunctional gastrointestinal disorders are common in patients with epilepsy and may influence seizure occurrence, suggesting that targeting gut dysfunction could improve seizure control.(99)
Citraro et al, 2021Absence epilepsyWAG/Rij rat model of absence epilepsyAltered microbiota and intestinal damage are present in a genetic animal model of absence epilepsy, indicating a potential pathway linking gut dysfunction to seizure activity.(100)
Gallucci et al, 2021Acute seizuresTheiler murine encephalomyelitis virus-induced acute seizuresGut metabolites may modulate neuronal excitability and contribute to epileptogenesis, suggesting new therapeutic targets.(101)

[i] PD, Parkinson's disease; AD, Alzheimer's disease; MPTP, 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine; TLR4, Toll-like receptor 4; TBI, traumatic brain injury; ASD, autism spectrum disorder; ADHD, attention-deficit hyperactivity disorder; FMT, fecal microbiota transplantation; SCFAs, short-chain fatty acids; LPS, lipopolysaccharide; APP/PS1, amyloid precursor protein/presenilin 1 (transgenic mouse model of AD); CUMS, chronic unpredictable mild stress; MPTP, 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine; GABA, gamma-aminobutyric acid; BDNF, brain-derived neurotrophic factor; TNF-α, tumor necrosis factor-alpha; IL-6, interleukin-6.

Anatomical connections: Vagus nerve, spinal cord and brain-gut peptides

The gut-brain axis relies on anatomical connections that facilitate bidirectional communication between the gastrointestinal tract and the CNS (19). The vagus nerve, a major component of the parasympathetic nervous system, serves as a direct neural pathway between the gut and the brain. It transmits signals from the gut to the brainstem and vice versa, influencing functions such as appetite, digestion and stress responses (3). The spinal cord also plays a role in this communication, processing sensory information from the gut and sending motor commands that regulate gastrointestinal motility and secretion (20). Additionally, brain-gut peptides such as ghrelin, leptin and peptides are released in response to nutrient intake and act on distant targets in the brain to regulate feeding behavior, energy balance and metabolic processes (21). These anatomical connections form the structural basis for the complex interactions that occur within the gut-brain axis.

Microbiota-gut-brain communication pathways: Metabolites, immune system and neural pathways

The gut microbiota, comprising trillions of microorganisms, plays a crucial role in shaping the gut environment and influencing both local and systemic immune responses (22). Microbiota-gut-brain communication occurs through several pathways. Metabolites produced by gut bacteria, such as short-chain fatty acids (SCFAs), secondary bile acids and tryptophan metabolites, can enter the bloodstream and affect brain function (23). These metabolites have been shown to influence neuroinflammation, neurotransmitter synthesis and blood-brain barrier (BBB) integrity (24). The immune system also serves as a key communication pathway, with gut-resident immune cells producing cytokines and chemokines that can modulate CNS function (25). Furthermore, neural pathways, including the vagus nerve, transmit signals from the gut to the brain and vice versa, allowing for rapid communication and coordinated responses to internal and external stimuli (3) (Fig. 1). This intricate network of communication pathways enables the gut microbiota to exert profound effects on neurological health.

Endocrine and neuroendocrine mechanisms linking the gut and brain

Endocrine and neuroendocrine mechanisms further contribute to the bidirectional communication between the gut and the brain (23). Gut hormones such as ghrelin, leptin and peptides are released into the bloodstream and act on receptors in the brain to regulate appetite, energy expenditure and metabolic processes (26). These hormones can influence neurotransmitter release and affect brain regions involved in reward, motivation and stress responses (27). Additionally, the HPA axis, a key neuroendocrine system, plays a significant role in stress response and regulation of the gut-brain axis. Stress-induced activation of the HPA axis leads to the release of corticotropin-releasing hormone and subsequent increases in cortisol levels, which can modulate gut motility, secretion and barrier function (28) (Fig. 1). Conversely, signals from the gut can feedback to the HPA axis, creating a dynamic interplay that impacts both gastrointestinal and neurological health (29). This complex web of endocrine and neuroendocrine interactions underscores the intimate connection between the gut and the brain in maintaining overall homeostasis and health.

Role of gut microbiota in neurological health

Gut microbiota composition-shaped by genetics, diet and environmental exposures-critically influences neurological outcomes through metabolite production. Microbiota-derived SCFAs, secondary bile acids and tryptophan metabolites modulate neurodevelopment, neurotransmitter synthesis [for example, serotonin and γ-aminobutyric acid (GABA)], and neuroinflammatory responses, thereby serving as fundamental regulators of brain homeostasis.

Composition and diversity of gut microbiota

The human gut microbiota comprises a complex community of microorganisms, including bacteria, archaea, viruses and eukaryotic microbes, with bacterial species being the most predominant and diverse (30). The composition of gut microbiota varies significantly among individuals and is influenced by factors such as genetics, diet, age, geographical location and lifestyle (31). The gut microbiota is established at birth, with the initial microbial colonization influenced by the mode of delivery and breastfeeding (32). During infancy and childhood, the microbiota continues to develop and diversify, reaching a relatively stable state in adulthood. However, this stability can be disrupted by various factors, including antibiotics, stress and changes in diet, leading to dysbiosis, which is associated with numerous health issues, including neurological disorders (33).

Microbiota-derived metabolites: SCFAs, secondary bile acids and tryptophan metabolites

Gut microbiota produces a wide range of metabolites that play crucial roles in host physiology and pathology. Tryptophan metabolites-indole derivatives (for example, indole-3-lactic acid, indole-3-acetic acid) and kynurenine pathway metabolites (kynurenine, kynurenic acid, 3-hydroxykynurenine)-are synthesized by gut bacteria (for example, Clostridium sporogenes, Lactobacillus spp.) via tryptophanase or indoleamine-2,3-dioxygenase. Dysbiosis-induced accumulation of neurotoxic kynurenine metabolites (3-hydroxykynurenine, quinolinic acid) and reduction of neuroprotective indole-3-lactic acid have been implicated in neurological disorders. In patients with AD, reduced serum indole-3-lactic acid correlates with cognitive decline (34), while elevated 3-hydroxykynurenine levels are associated with neuroinflammation in PD (35). SCFAs, including acetate, propionate and butyrate, are the primary metabolites produced by the fermentation of dietary fiber by gut bacteria (36). SCFAs have been shown to modulate neuroinflammation, reinforce the integrity of the BBB, and regulate neurotransmitter synthesis (37). Secondary bile acids, such as deoxycholic acid and lithocholic acid, are generated by the microbial transformation of primary bile acids and have been implicated in influencing neuroinflammation and cognitive function through interactions with specific receptors (38). Tryptophan metabolites, including indole derivatives and kynurenine, are another important group of microbiota-derived metabolites that impact serotonin production and immune responses, potentially affecting neuropsychiatric conditions (39).

Microbiota influence on neurodevelopment, neurotransmission and neuroinflammation

The gut microbiota significantly influences neurodevelopment, with animal studies demonstrating that germ-free mice exhibit altered brain development and behavior compared with conventionally raised animals (40). The microbiota also plays a vital role in neurotransmission, as gut bacteria are capable of producing or stimulating the production of neurotransmitters such as serotonin, dopamine and gut luminal GABA) (41,42). In total, ~90% of the body's serotonin is synthesized by enterochromaffin cells in the gut, and this peripheral serotonin can influence both gut function and brain activity (43). Furthermore, the gut microbiota is a key regulator of neuroinflammation, modulating the host's immune response and influencing the activation of microglia, the resident immune cells of the CNS (44,45). Dysbiosis has been associated with increased neuroinflammatory responses, which are implicated in the pathogenesis of various neurological disorders, including AD, PD and MS (46-48).

Gut-brain axis dysregulation in neurological diseases

Emerging evidence implicates gut-brain axis perturbations across diverse neurological disorders through distinct yet overlapping mechanisms (Table II). Dysbiosis precedes motor symptoms in PD, exacerbates neuroinflammation in MS and stroke, correlates with behavioral deficits in ASD, and influences seizure susceptibility in epilepsy, suggesting pathway-specific pathophysiological contributions.

Table II

Molecular mechanisms that connects the gut and the brain.

Table II

Molecular mechanisms that connects the gut and the brain.

Authors, yearDisease modelStudy objectMechanismKey findings(Refs.)
Antón et al, 2018 NeuroinflammationRat model of alcohol-induced intestinal hyperpermeabilityGut barrier disruption leads to microbial metabolite translocation and systemic inflammation, activating TLR4 signaling in peripheral immune cells and the brainAlcohol-induced intestinal hyperpermeability promotes systemic endotoxemia, exacerbating neuroinflammatory cascades(102)
Hoyles et al, 2018Neuroinflammation and BBB regulationMouse modelPropionate protects BBB integrity and suppresses pro-inflammatory cytokines via histone deacetylase inhibitionPropionate reinforces BBB and reduces inflammation(103)
Ceccarelli et al, 2017Neuroinflammation in HIV-associated neurocognitive disordersHIV-1 infected individualsHigh-dose multi-strain probiotics modulate CNS immune activationProbiotics may benefit neurocognitive performance(104)
Rincel et al, 2019Sex-specific neuroinflammationMulti-hit early life adversity in miceGut microbiota, brain, and behavior affected differently in males and femalesEarly life adversity impacts gut-brain axis sex-dependently(105)
Cuskelly et al, 2022Neuroinflammation with sexual dimorphismNeonatal immune challenge in rodentsMicrobiota and behavior influenced differently in males and femalesNeonatal immune challenge affects microbiota and behavior sexually dimorphically(106)
Saunders et al, 2020Neurodevelopmental disorderMouse modelGut microbiota manipulation during prepubertal period shapes behavioral abnormalitiesGut microbiota affects neurodevelopmental behavior(107)
Duan et al, 2023Impact of SCFAs on macrophage activitiesDisease contextMicrobiota-derived SCFAs influence macrophage functionsSCFAs have therapeutic potentials(108)
Chauhan et al, 2023Neurodegenerative diseasesComparative analysisMicrobial dysbiosis may link to neurodegeneration metabolicallyMicrobial dysbiosis potentially contributes to neurodegeneration(109)
Zancan et al, 2024MSMendelian randomization analysisGut microbiota composition causally linked to MSGut microbiota plays role in MS etiology(110)
Emery et al, 2022Alzheimer's and PDHigh resolution 16S rRNA gene NGSBrain areas associated with AD and PD have distinct microbiotaMicrobiota differs in neurodegenerative diseases(111)
Boles et al, 2024Colitis modelMouse modelLeaky gut dysregulates brain gene networks associated with immune activation, oxidative stress and myelinationGut permeability affects brain gene regulation(112)
Luck et al, 2021Neurotransmitter profiles in gut and brainMice mono-associated with Bifidobacterium dentiumNeurotransmitter profiles altered in gut and brainGut microbiota influences neurotransmitter profiles(113)
Pandey et al, 2015Systemic oxidative stress and neurotransmittersProbiotic Escherichia coli CFR 16Protects against 1,2-dimethylhydrazine-induced stress and neurotransmitter alterationsProbiotic modulates systemic stress and neurotransmitters(114)
Zhu et al, 2020PDMouse modelPotential association between neurotransmitter disturbance and gut microbiota dysbiosisGut microbiota may affect neurotransmitters in PD(115)
Li et al, 2025Depressive-like behaviorsMice Lactiplantibacillus plantarum GOLDGUT-HNU082 modulates gut microbiota and neurotransmittersProbiotic alleviates depressive behaviors(116)
Guo et al, 2021Neurotransmitter metabolismHigh sugar and fat dietInfluences neurotransmitter metabolism, affecting brain function via gut microbiotaDiet impacts brain via gut microbiota(117)
Zhang et al, 2021Neurotransmitter secretion disorderZebrafish exposed to caffeineMelatonin regulates disorder through microbiota-gut-brain axisMelatonin modulates neurotransmitter secretion(118)
Wang et al, 2023Neurotransmitter disturbancesMethylmercury exposure Microbiota-gut-brain interaction involvedMethylmercury affects neurotransmitters via microbiota(119)
Huang et al, 2024Gut-brain axis in cerebral small vessel diseasePatientsMulti-omics analysis reveals microbial and neurotransmitter signaturesGut-brain axis altered in cerebrovascular disease(121)
Margetts et al, 2022Anti-inflammatory effectsSalvia officinalis L.Modulates neurotransmitter metabolismHerbal extract has anti-inflammatory effects(122)
Kumari et al, 2022Oxidative stress and brain injuryMouse modelLacticaseibacillus rhamnosus-derived exopolysaccharide attenuates oxidative stress and inflammationProbiotic derivative protects against brain injury(123)
Cansız et al, 2021Gut-brain axis in zebrafishRotenone-induced stressCaprylic acid ameliorates inflammation and oxidative stressCaprylic acid benefits gut-brain axis(125)
Pan et al, 2022MPTP-induced oxidative stressProbiotic Pediococcus pentosaceusAlleviates oxidative stress via gut microbiota regulationProbiotic reduces oxidative stress(126)
Dong et al, 2023Cognitive function in miceSoy protein supplementationModifies hippocampal nerve growth, oxidative stress, and intestinal microbiotaSoy protein improves cognitive function(127)
Nikrad et al, 2023Gut-brain axis pathwayObese women with PCOSCalorie restriction with spinach thylakoids affects gut-brain pathway and oxidative stressDietary intervention impacts gut-brain axis(128)
Rode et al, 2021Tryptophan transportOxidative stress-induced deficitsButyrate rescues deficits in intestinal epithelial cellsButyrate may benefit affective disorders(129)
He et al, 2024AD modelHydrogen-rich waterTargets oxidative stress, inflammation, and gut-brain axisHydrogen-rich water has therapeutic potential(130)
Di Chiano et al, 2024Human microgliaLactobacilli cell-free supernatantsModulate inflammation and oxidative stress via NRF2-SOD1 signalingProbiotic supernatants have neuroprotective effects(131)
Hsu et al, 2023Patients with Alzheimer's dementiaProbiotic supplementsImprove BDNF, reduce inflammation and oxidative stressProbiotics may benefit patients with Alzheimer's disease(132)
Akhgarjand et al, 2024Mild to moderate ADProbiotic supplementsReduce oxidative stress and inflammationProbiotics have positive effects(133)
Chenghan et al,BBB integritySCFAsMediate gut microbiota-brain communication, protecting BBBSCFAs protect BBB integrity(135)
Stachulski et al,BBB functionp-cresol glucuronidePromotes BBB integrity by suppressing pro-inflammatory cytokinesMicrobial metabolite benefits BBB(136)
Connell et al, 2024LPS-induced BBB disruptionPolyphenol-rich extractPrevents disruption through modulation of gut microbiota-derived toxinsDietary polyphenols protect BBB(137)
Fiorentino et al, 2016ASDPatientsAltered gut and brain barriers linked to neuroinflammationBarrier dysfunction contributes to ASD pathology(139)
Sun et al, 2021Post-bone-marrow transplantationMice with antibiotic-induced microbiome depletionDisrupted BBB allows monocyte infiltrationMicrobiome depletion affects BBB(140)
Nelson et al, 2021Hypertensive stroke-prone ratsSpontaneously hypertensive stroke-prone ratsGut microbiome contributes to BBB disruptionGut microbiome affects BBB integrity(141)
Knox et al, 2022BBBprotectionMicrobial-derived metabolitesInduce actin rearrangement, enhancing barrier resilienceMetabolites protect BBB function(142)
Wang et al, 2024Ischemic stroke treatmentVagus nerve stimulationModulates mast cell degranulation via microbiota-gut-brain axisVagus nerve stimulation protects BBB(143)
Marungruang et al, 2018Postnatal BBB developmentDietary-induced gut maturationStabilizes BBB development through metabolic crosstalkEarly dietary intervention benefits BBB(144)
Schalla et al, 2023Stem-cell-derived BBB modelPhoenixin-14Does not cross model, indicating selective transportGut-derived molecules have selective BBB permeability(145)

[i] BBB, blood-brain barrier; SCFAs, short-chain fatty acids; TLR4, Toll-like Receptor 4; HIV, human immunodeficiency virus; CNS, central nervous system; NRF2, nuclear factor erythroid 2-related factor 2; SOD, superoxide dismutase; GPx, glutathione peroxidase; LPS, lipopolysaccharide; NLRP3, NOD-, LRR- and pyrin domain-containing protein 3; GABA, gamma-aminobutyric acid; 5-HT, 5-hydroxytryptamine (serotonin); BDNF, brain-derived neurotrophic factor; MPTP, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine; NOX2, NADPH oxidase 2; MDA, malondialdehyde; NGF, nerve growth factor; 8-OHdG, 8-hydroxy-2′-deoxyguanosine; Treg, regulatory T cell; IFN-γ, interferon-gamma; IL-17A, interleukin-17A; IL-10, interleukin-10; EVs, extracellular vesicles; PAMPs, pathogen-associated molecular patterns.

Neurodegenerative diseases

In PD, research has shown that gut microbiota dysbiosis precedes motor symptoms and may contribute to neuroinflammation and α-synuclein aggregation (49). Sampson et al (50) demonstrated that gut microbiota regulates motor deficits and neuroinflammation in a PD model, with antibiotic treatment reducing motor dysfunction and microglial activation. Perez-Pardo et al (51) further explored the role of Toll-like receptor 4 (TLR4) in the gut-brain axis in PD, suggesting that TLR4 signaling may be a therapeutic target. Additionally, Srivastav et al (52) found that a probiotics mixture increases butyrate, rescuing nigral dopaminergic neurons from 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and rotenone-induced neurotoxicity.

In PD, α-synuclein pathology is closely linked to gastrointestinal dysbiosis; increased α-synuclein expression correlates with reduced Lachnospiraceae and elevated Enterobacteriaceae, accelerating neurodegeneration via gut-derived inflammation (53). Homogeneously, emerging evidence suggests that gut microbiota alterations may contribute to amyloid-β plaque formation and neuroinflammation. Honarpisheh et al (54) observed dysregulated gut homeostasis prior to brain amyloid-β accumulation in Tg2576 mice. Kim et al (55) found that transplantation of gut microbiota from mouse models with AD impairs memory function and neurogenesis in recipient mice. Shi et al (56) demonstrated that a fiber-deprived diet causes cognitive impairment and hippocampal microglia-mediated synaptic loss through the gut microbiota and metabolites. Furthermore, chronic PD progression is associated with gut microbiota-driven protection of dopaminergic neurons. Clostridium butyricum supplementation increased butyrate-producing taxa, suppressed substantia nigra inflammation, and improved motor deficits in mice with MPTP-induced PD (57).

Recent advances have highlighted the potential therapeutic role of probiotics and prebiotics in neurodegenerative diseases (57). Castelli et al (58) discussed the emerging role of probiotics in PD, suggesting new therapeutic possibilities. Yuan et al (59) explored the ApoE-dependent protective effects of sesamol on high-fat diet-induced behavioral disorders through regulation of the microbiome-gut-brain axis. Additionally, prebiotics such as microbiota-accessible carbohydrates (a class of prebiotics), have been shown to prevent neuroinflammation and cognitive decline by improving the gut microbiota-brain axis in diet-induced obese mice (60). While these preclinical studies are encouraging, a recent randomized controlled trial (RCT) in early PD reported no motor benefit from multi-strain probiotics after 12 weeks (14), underscoring the need for larger, longer-duration trials that stratify patients by baseline microbiota profiles.

Neuroinflammatory diseases

In MS, research has identified specific gut commensals that modulate neuroinflammation. Bianchimano et al (61) utilized microbiota profiling to pinpoint gut bacteria influencing MS-like disease in mice, revealing potential therapeutic targets within the gut microbiota. Similarly, in TBI, the gut-brain axis plays a pivotal role in neuroinflammatory responses. Celorrio and Friess (62) highlighted the impact of gut microbiota dysbiosis on neuroinflammation following TBI, suggesting that targeting the gut microbiota could mitigate post-injury inflammation. Furthermore, first-episode psychosis (FEP) exhibits gut dysbiosis characterized by decreased Faecalibacterium prausnitzii and elevated Enterococcus, correlating with increased kynurenine/tryptophan ratios and peripheral inflammation (63). These findings implicate microbial tryptophan metabolism in FEP pathophysiology.

Environmental factors also contribute to neuroinflammation through the gut-brain axis (64). Lee et al (65) demonstrated that exposure to polystyrene microplastics impairs hippocampus-dependent learning and memory in mice, with gut microbiota alterations preceding neuroinflammatory changes. Exercise has shown promise in modulating the gut microbiota to reduce neuroinflammation. Yuan et al (66) found that treadmill exercise modulates intestinal microbes and suppresses lipopolysaccharide (LPS) displacement, alleviating neuroinflammation in APP/PS1 mice.

FMT represents a novel therapeutic approach. Hu et al (67) showed that FMT inhibits neuroinflammation following TBI in mice by regulating the gut-brain axis, indicating its potential as a treatment for neuroinflammatory conditions. Additionally, specific probiotics have demonstrated neuroprotective effects. Li et al (68) revealed that Lactobacillus rhamnosus GG ameliorates noise-induced cognitive deficits and systemic inflammation in rats by modulating the gut-brain axis.

Recent research has also focused on the role of microbial metabolites in neuroinflammation. Wei et al (69) showed that butyrate ameliorates chronic alcoholic CNS damage by suppressing microglia-mediated neuroinflammation and modulating the microbiome-gut-brain axis. Similarly, Zhu et al (70) demonstrated that Bifidobacterium breve HNXY26M4 attenuates cognitive deficits and neuroinflammation in APP/PS1 mice through gut-brain axis regulation.

Neurodevelopmental disorders

In ASD, research has identified specific gut microbiota alterations associated with disease severity. De Theije et al (71) demonstrated intestinal inflammation in a murine model of ASD, suggesting a potential link between gut dysfunction and behavioral abnormalities. Stilling et al (72) found that absence of microbiota during early life increases activity-related transcriptional pathways in the amygdala, highlighting the role of gut microbiota in neurodevelopmental programming. Kantarcioglu et al (73) identified specific yeast species in the stool samples of children with ASD, suggesting a potential role of fungal microbiota in disease pathogenesis. Furthermore, recent comparative analyses revealed significant differences in gut microbiota composition between children with ASD and their neurotypical siblings, with reduced Bifidobacterium and Akkermansia abundance in ASD cohorts (74). These alterations correlated with increased serum zonulin levels, suggesting compromised intestinal barrier integrity as a mechanistic link to neuroinflammation.

In addition to ASD, gut microbiota dysbiosis has been implicated in schizophrenia and bipolar disorder. In schizophrenia, patients show reduced microbial diversity and decreased abundance of SCFA-producing taxa such as Faecalibacterium and Roseburia, which correlates with cognitive deficits and negative symptoms (75). A systematic review by Bioque et al (76) further highlighted the role of the microbiome-gut-brain axis in schizophrenia, emphasizing its potential as a therapeutic target (75-77). Similarly, in bipolar disorder, gut dysbiosis is characterized by decreased Bifidobacterium and increased pro-inflammatory Enterobacteriaceae, which may exacerbate neuroinflammation via kynurenine pathway activation and cognitive dysfunction (63). A pilot study by Reininghaus et al (78) demonstrated that probiotic supplementation improved cognitive parameters in patients with euthymic bipolar disorder, suggesting a modulatory role of gut microbiota in psychiatric outcomes.

Recent research has focused on the therapeutic potential of targeting the gut microbiota in ASD. Pulikkan et al (79) reported gut microbial dysbiosis in Indian children with ASD, with specific bacterial taxa correlating with disease severity. Bojović et al (80) found altered production of SCFAs in children with neurodevelopmental disorders, suggesting a potential metabolic link between gut dysbiosis and neurological symptoms.

In attention-deficit hyperactivity disorder (ADHD), emerging evidence suggests that gut microbiota dysregulation may contribute to disease development. Slob et al (81) found an association between early-life antibiotic use and the risk of ADHD and ASD in a discordant twin study, indicating that gut microbiota perturbations during critical developmental windows may have long-term neurological consequences. Bundgaard-Nielsen et al (82) reported distinct microbiota compositions in children and adolescents with ADHD and ASD, suggesting shared and unique gut microbiota signatures across neurodevelopmental disorders. Nevertheless, a meta-analysis of 14 pediatric ASD cohorts found no consistent taxonomic signature after adjusting for diet and antibiotic exposure, highlighting the risk of over-interpreting small, uncontrolled studies (83).

Environmental factors and their impact on the gut-brain axis in neurodevelopmental disorders have also been explored. Gonçalves et al (84) demonstrated that exposure to high doses of amoxicillin causes behavioral changes and oxidative stress in young zebrafish, suggesting potential developmental consequences of antibiotic use. Volkova et al (85) found that early-life penicillin exposure alters frontal cortex and amygdala gene expression, highlighting the sensitivity of the developing brain to gut microbiota perturbations.

Stroke and cerebrovascular diseases

Research has shown that gut microbiota dysbiosis is associated with increased risk and severity of stroke. Benakis et al (86) demonstrated that commensal microbiota affects ischemic stroke outcome by regulating intestinal γδ T cells, highlighting the role of gut immunity in stroke pathogenesis. Dou et al (87) found that resveratrol exerts neuroprotection against focal cerebral ischemia/reperfusion injury in mice through a mechanism targeting the gut-brain axis. Bonsack et al (88) discussed the active role of the gut-brain axis in homeostasis and dysbiosis related to stroke. A review highlighted that cerebral ischemia-reperfusion injury triggers gut dysbiosis-mediated neuroinflammation via TLR4/NF-κB signaling and elevated IL-6/TNF-α levels (89). FMT from healthy donors restored Lactobacillus abundance and attenuated microglial activation in rodent stroke models.

Recent studies have focused on the therapeutic potential of targeting the gut microbiota in stroke (89). Park et al (90) reported that reproductive senescence and ischemic stroke remodel the gut microbiome and modulate the effects of estrogen treatment in female rats. Sadler et al (91) found that SCFAs improve poststroke recovery via immunological mechanisms, suggesting a potential therapeutic avenue. Aswendt et al (92) demonstrated that the gut microbiota modulates brain network connectivity under physiological conditions and after acute brain ischemia, indicating its role in both health and disease states.

Environmental factors and their impact on the gut-brain axis in stroke have also been explored (93). Henry et al (94) found that SCFAs taken at the time of thrombectomy in patients with acute ischemic stroke are associated with inflammatory markers and worse symptoms at discharge, suggesting complex interactions between gut metabolites and clinical outcomes. Jiang et al (95) reported alterations in the gut microbiome and correlated lipid metabolism in post-stroke depression, indicating potential metabolic links between gut dysbiosis and neurological symptoms.

Epilepsy and seizure disorders

Research has shown that gut microbiota dysbiosis is associated with increased risk and severity of epilepsy. Huang et al (96) demonstrated distinct gut microbiota composition and functional categories in children with cerebral palsy and epilepsy, suggesting a potential link between gut dysfunction and seizure activity. Lindefeldt et al (97) reported that the ketogenic diet influences the taxonomic and functional composition of the gut microbiota in children with severe epilepsy, indicating that dietary interventions can modulate gut microbiota to reduce seizure frequency.

Previous studies have focused on the therapeutic potential of targeting the gut microbiota in epilepsy. He et al (98) described a pediatric case where FMT cured epilepsy in a patient with Crohn's disease, highlighting the potential of microbiota-based therapies. Avorio et al (99) found that functional gastrointestinal disorders are common in patients with epilepsy and may influence seizure occurrence, suggesting that targeting gut dysfunction could improve seizure control.

The mechanism underlying the gut-brain axis in epilepsy has also been explored. Citraro et al (100) provided the first evidence of altered microbiota and intestinal damage in a genetic animal model of absence epilepsy, the WAG/Rij rat, indicating a potential pathway linking gut dysfunction to seizure activity (100). Gallucci et al (101) proposed that gut metabolites may modulate neuronal excitability and contribute to epileptogenesis, suggesting new therapeutic targets.

Molecular mechanisms linking the gut and brain in disease

Across neurodegenerative, neuroinflammatory, neurodevelopmental, cerebrovascular and epileptic disorders, convergent evidence implicates four mutually reinforcing gut-brain axis disturbances: i) Gut barrier failure enables microbial translocation, activating TLR4/NLRP3 inflammasomes and microglia; ii) Microbiota dysregulation disrupts neurotransmitter balance (for example, GABA and serotonin); iii) Oxidative stress amplifies neuronal damage via impaired NRF2 signaling; and iv) BBB compromise facilitates neuroimmune crosstalk (Fig. 2). Subsequent disease-specific subsections highlight only the unique nuances of these pathways (for example, α-synuclein aggregation in PD, amyloid-β seeding in AD, Th17 polarization in MS, seizure-associated neuronal hyperexcitability in epilepsy) (Table III).

Table III

Strength of mechanistic evidence linking gut-brain axis pathways to neurological diseases.

Table III

Strength of mechanistic evidence linking gut-brain axis pathways to neurological diseases.

Authors, yearPathwayDisease categoryEvidence strengthKey unresolved questions(Refs.)
Perez-Pardo et al, 2019; Emery et al, 2022; Boles et al, 2024Neuroinflammation (TLR4/NLRP3)AD, PD, MSModerate-Strong (supported by humantissue and GF-to-conventional transfer studies)Does dysbiosis precede or follow disease onset?(51,111,112)
Hoyles et al, 2018; Chenghan et al, 2025; Stachulski et al, 2023; Connell et al, 2024SCFA-mediated BBB integrityStroke, ASD, ADModerate (consistent in animal models; limited RCTs)Inter-individual variability in SCFAs response; dose thresholds unclear(103,135-137)
Chauhan et al, 2023; Zancan et al, 2024Microbial amyloid as autoantigenPD, ADWeak-moderate (correlative human data; GF mice show phenotype reversal)Lack of longitudinal human cohorts; need for germ-free primate models(109,110)
Li et al, 2025; Guo et al, 2021; Zhang et al, 2021Serotonin depletion via gut microbiotaDepression, ASDModerate (GF mouse and probiotic RCTs)Heterogeneity of 5-HT-producing taxa across populations(116-118)

[i] Evidence strength graded as Strong (meta-analysis or replicated RCT), Moderate (animal-to-human translational data), Weak (single-study or associative only). AD, Alzheimer's disease; ASD, autism spectrum disorder; BBB, blood-brain barrier; GF, germ-free; MS, multiple sclerosis; NLRP3, NOD-, LRR-, and pyrin domain-containing protein 3; PD, Parkinson's disease; RCTs, randomized controlled trials; SCFAs, short-chain fatty acids; TLR4, Toll-like receptor 4; 5-HT, 5-hydroxytryptamine (serotonin).

Neuroinflammation and immune activation

Emerging evidence highlights that dysregulation of the gut-brain axis contributes to neuroinflammation and systemic immune activation in neurological diseases. For instance, alcohol-induced intestinal hyperpermeability in rats promotes systemic endotoxemia, characterized by elevated LPS levels. This activates TLR4 signaling in peripheral immune cells and the brain, exacerbating neuroinflammatory cascades (102). TLR4 activation has been directly implicated in PD pathogenesis, as demonstrated by elevated TLR4 expression in both human patients and murine models with PD, driving microglial activation and dopaminergic neuron loss (51).

Gut microbiota-derived metabolites play dual roles in modulating neuroimmune responses. SCFAs, such as propionate, exhibit protective effects by reinforcing the BBB and suppressing pro-inflammatory cytokine production (for example, IL-6 and TNF-α) through histone deacetylase inhibition (103). Conversely, dysbiosis-associated reductions in SCFA-producing bacteria (for example, Faecalibacterium) correlate with increased CNS immune activation, as observed in HIV-associated neurocognitive disorders (104). Notably, sex-specific differences in gut-immune interactions have been reported; early-life adversity in mice induces persistent gut dysbiosis and neuroinflammation in males but not females, linked to altered IL-10/IL-17A balance and microglial priming (105,106).

The gut microbiome also influences peripheral immune cell trafficking to the brain. In a mouse model of ASD, prepubertal microbiota depletion alters T helper cell polarization, leading to aberrant IFN-γ and IL-17A levels in the prefrontal cortex, which disrupts synaptic plasticity and exacerbates behavioral deficits (107). Similarly, neonatal immune challenges in rodents induce gut-mediated Th1/Th2 imbalances, promoting hippocampal microglial activation and anxiety-like behaviors in adulthood (106). Therapeutic interventions targeting gut-immune crosstalk, such as high-dose probiotics, have shown efficacy in attenuating CNS inflammation by restoring regulatory T cell (Treg) function and reducing monocyte activation (104,108).

Recent studies further implicate gut-derived extracellular vesicles and bacterial amyloid proteins in priming peripheral immune cells to cross-react with CNS antigens, potentially initiating autoimmune-driven neuroinflammation (109,110). For example, microbiome-induced molecular mimicry between microbial peptides and α-synuclein may contribute to PD pathogenesis by eliciting cross-reactive T cell responses (51,111). Additionally, gut dysbiosis in AD correlates with increased cerebral LPS levels and NLRP3 inflammasome activation, suggesting a feedforward loop between peripheral inflammation and neurodegenerative processes (111,112).

Neurotransmitter regulation

The gut microbiota exerts profound influence over central and peripheral neurotransmitter systems, modulating behaviors and disease progression through bidirectional gut-brain communication. For example, Bifidobacterium dentium colonization in mice elevates GABA levels by upregulating glutamate decarboxylase activity, correlating with reduced anxiety-like behaviors and altered hippocampal GABA receptor expression (113). Similarly, gut-resident Escherichia coli strains producing pyrroloquinoline quinone mitigate oxidative stress-induced dopamine depletion in rodent models of neurotoxicity, preserving striatal dopaminergic function (114).

Dysbiosis-driven neurotransmitter imbalances are increasingly implicated in neurological disorders. In a MPTP-induced PD mouse model, gut microbiota disruption precedes dopaminergic neuron degeneration, accompanied by diminished colonic serotonin (5-HT) and elevated cerebral norepinephrine, suggesting microbiota-dependent regulation of monoamine metabolism (115). Notably, Lactiplantibacillus plantarum supplementation restores CUMS-induced depressive behaviors in mice by normalizing gut microbial diversity and increasing prefrontal cortical 5-HT and brain-derived neurotrophic factor (BDNF) levels, highlighting therapeutic potential (116).

Dietary factors further shape neurotransmitter dynamics via microbiota-mediated pathways. High-sugar/high-fat diets reduce fecal Lactobacillus abundance, impairing tryptophan metabolism and lowering hippocampal 5-HT synthesis, which disrupts synaptic plasticity and memory consolidation (117). Conversely, melatonin attenuates caffeine-induced zebrafish hyperactivity by rescuing gut microbial diversity and rebalancing dopamine/GABA ratios in the brain, underscoring diet-microbiota-neurotransmitter interplay (118).

Emerging evidence links environmental toxins to gut-mediated neurotransmitter dysregulation. Methylmercury exposure alters Bacteroidetes-to-Firmicutes ratios, suppressing colonic dopamine and 5-HT synthesis while elevating glutamate in the hippocampus, a phenotype reversible by Akkermansia-enriched probiotics (119). Aluminum oxide nanoparticles, commonly used in food additives, accumulate in juvenile rat brains, reducing striatal dopamine and serotonin while increasing pro-inflammatory cytokines, effects exacerbated by gut barrier leakage (120).

Advanced multi-omics approaches reveal novel microbiota-neurotransmitter networks in disease. In arteriosclerotic cerebral small vessel disease, decreased gut Roseburia and Faecalibacterium correlate with disrupted tryptophan-kynurenine pathways and reduced cortical GABA, implicating microbial regulation of neurovascular coupling (121). Similarly, Salvia officinalis extracts ameliorate neuroinflammation by modulating gut microbial bile acid metabolism, which enhances hypothalamic dopamine signaling and suppresses TNF-α-driven neuronal apoptosis (122).

Indole-3-lactic acid, a tryptophan metabolite produced by Lactobacillus spp., protects against neuroinflammation by activating aryl hydrocarbon receptor signaling in microglia (34). Conversely, dysbiotic expansion of Eggerthella lenta increases kynurenine/tryptophan ratios, exacerbating neurotoxicity via quinolinic acid production in MS models (35). These findings underscore microbial tryptophan catabolism as a key regulator of neurotransmitter balance (serotonin depletion) and neurodegeneration in neurological disorders (34).

Oxidative stress and antioxidant mechanisms

The gut microbiota plays a critical role in modulating oxidative stress within the gut-brain axis, with dysbiosis exacerbating reactive oxygen species (ROS) accumulation and impairing antioxidant defenses in neurological diseases. For instance, Lactobacillus rhamnosus-derived exopolysaccharides mitigate D-galactose-induced brain aging in mice by activating nuclear factor erythroid 2-related factor 2 (NRF2), thereby upregulating superoxide dismutase (SOD) and glutathione peroxidase activity while reducing hippocampal lipid peroxidation (123). Similarly, heat-eliminated Levilactobacillus brevis KU15152 attenuates H2O2-induced oxidative damage in neuronal cells by enhancing SOD1 expression, highlighting strain-specific antioxidant properties (124).

Gut barrier dysfunction amplifies systemic oxidative stress via microbial translocation. In a rotenone-induced zebrafish model of PD, intestinal hyperpermeability promotes ROS overproduction in the brain, which is reversed by caprylic acid through restoration of tight junction proteins and suppression of NADPH oxidase 2 activity (125). Notably, Lactiplantibacillus plantarum GOLDGUT-HNU082 alleviates MPTP-induced dopaminergic neuron loss by reducing colonic LPS leakage and inhibiting NLRP3 inflammasome-driven oxidative stress in the substantia nigra (126).

Dietary interventions targeting the microbiota-gut-brain axis demonstrate therapeutic potential. Soy protein supplementation enhances cognitive function in mice by increasing hippocampal nerve growth factor levels and reducing ROS, effects mediated by Lactobacillus-enriched gut microbiota and upregulated fecal butyrate production (127). Conversely, high-dose amoxicillin exposure in zebrafish depletes Bifidobacterium, disrupts glutathione metabolism, and induces behavioral deficits linked to cerebral oxidative damage. Intriguingly, calorie restriction combined with spinach thylakoids in obese women with polycystic ovary syndrome lowers serum 8-OHdG (a DNA oxidative marker) and rescues gut Akkermansia abundance, suggesting microbiota-dependent redox balance (128). Emerging evidence implicates microbial metabolites in redox signaling. Butyrate rescues oxidative stress-induced tryptophan transport deficits in intestinal epithelial cells, preserving serotonin synthesis and mitigating depressive-like behaviors in mice (129). In AD models, hydrogen-rich water restores gut Roseburia levels, which correlates with reduced hippocampal malondialdehyde and enhanced SOD2 activity via the BDNF/TrkB pathway (130). Furthermore, Lactobacillus supernatants suppress microglial ROS production by modulating NRF2-SOD1 crosstalk, offering a novel anti-inflammatory strategy for neuroprotection (131).

Clinical studies underscore translational relevance. Probiotic supplementation in patients with AD reduces plasma 8-isoprostane (a lipid peroxidation marker) and elevates BDNF, correlating with improved cognitive scores (132,133). Similarly, Agaricus mushroom-enriched diets in mice lower cortical ROS and normalize gut Bacteroides populations, highlighting diet-microbiota interactions in antioxidant defense (134).

BBB integrity and permeability

Gut microbiota-derived metabolites regulate BBB permeability through structural and functional adaptations. For instance, microbial SCFAs, such as propionate, enhance BBB integrity by upregulating tight junction proteins (for example, occludin and claudin-5) and reducing systemic inflammation (103,135). Similarly, the microbial co-metabolite p-cresol glucuronide strengthens BBB function in vivo by suppressing pro-inflammatory cytokines and preserving endothelial cell junctions (136). Conversely, dysbiosis-induced uremic toxins, such as indoxyl sulfate, compromise BBB tightness by activating oxidative stress pathways, a mechanism attenuated by polyphenol-rich dietary interventions that restore microbial balance (137,138).

BBB disruption is increasingly recognized in neurodevelopmental and neurodegenerative disorders. In ASD, dual impairment of the BBB and intestinal epithelial barrier correlates with elevated circulating neuroinflammatory markers, suggesting a 'leaky gut-leaky brain' axis (139). Furthermore, gut microbiome depletion via antibiotics exacerbates BBB permeability, facilitating monocyte infiltration into the brain-a process linked to post-infection cognitive deficits (140). In hypertension-related stroke models, gut dysbiosis promotes BBB breakdown through systemic endotoxemia and endothelial activation, underscoring microbiota-driven vascular pathology (141).

Therapeutic modulation of the gut-BBB axis shows promise. Microbial-derived metabolites, such as butyrate, induce actin cytoskeletal rearrangements in brain endothelial cells, enhancing barrier resilience against inflammatory insults (142). Non-invasive vagus nerve stimulation mitigates ischemia-induced BBB damage by regulating mast cell degranulation via the microbiota-gut-brain axis, highlighting neuro-modulatory approaches (143). Additionally, precocious gut maturation through dietary interventions stabilizes BBB development in early life by shaping microbiota-host metabolic crosstalk (144).

However, not all gut signals permeate the BBB. Phoenixin-14, a neuropeptide implicated in stress responses, fails to cross in vitro BBB models, emphasizing selective transport mechanisms (145). This selectivity underscores the need for targeted delivery strategies when leveraging gut-derived molecules for neurological therapies. In summary, the BBB serves as both a gatekeeper and a sensor of gut-derived mediators, with its permeability dynamically shaped by microbial metabolites, systemic inflammation and host-microbe interactions. Restoring BBB integrity via microbiota-centric interventions represents a novel avenue for treating neurological diseases characterized by barrier dysfunction.

Despite converging evidence that gut-brain axis disruption participates in neurological disease, several mechanistic questions remain contentious. First, the temporal and causal relationship between dysbiosis and disease phenotype is unresolved: Mendelian randomization studies yield conflicting signals-some supporting a causal role of reduced SCFA-producers in MS, others finding no causal link in AD-highlighting the need for longitudinal, diet-controlled human cohorts and gnotobiotic humanized-mouse validation. Second, the therapeutic window and inter-individual heterogeneity of SCFA supplementation are unclear; butyrate attenuates neuro-inflammation in AD models yet exacerbates inflammatory markers in a subset of post-stroke patients with high baseline LPS, underscoring the necessity for baseline microbiome/metabolome stratification in future RCTs. Third, extrapolation from germ-free rodent data is limited by the absence of human-equivalent mucosal immunity and dietary complexity; therefore, human intestinal organoid-microbiota co-cultures and phase-0 micro-dosing trials are required. Finally, the 'microbial amyloid' hypothesis-where bacterial curli fibers seed α-synuclein aggregation-remains correlative; targeted metagenomic surveys and CRISPR knock-out of curli operons in gnotobiotic mice are proposed as definitive tests. These challenges emphasize the need for standardized multi-omics protocols and precision-medicine frameworks to reconcile associative evidence with causality.

Therapeutic strategies targeting the gut-brain axis and related clinical studies

Current therapeutic approaches leverage gut-brain axis modulation through dietary interventions, microbial restoration and pharmacological targeting. Clinical trials demonstrate probiotic efficacy in stress reduction, FMT-mediated motor improvement in PD, and SCFA-mediated neuroprotection, though challenges persist in strain selection, treatment durability and patient stratification (Table IV).

Table IV

Clinical studies on gut-brain axis.

Table IV

Clinical studies on gut-brain axis.

Authors, yearInterventionPopulationDurationOutcome measuresResults(Refs.)
Chong et al, 2019Lactobacillus plantarum DR7Adults with stress and anxiety12 weeksStress (DASS-21), anxiety (STAI), plasma cortisol, inflammatory cytokinesReduced stress, anxiety, and cortisol levels; improved gut barrier function(151)
Lew et al, 2019Lactobacillus plantarum P8Stressed adults12 weeksCognitive function (CANTAB), stress (PSS), anxiety (STAI), gut microbiota analysisEnhanced memory, reduced stress and anxiety; increased Bifidobacterium and Lactobacillus abundance(152)
Karakula-Juchnowicz et al, 2019Probiotic supplementationPatients with MDD (gluten-free/gluten-containing diet)12 weeksMental status (HAMD, MADRS), intestinal permeability (zonulin), inflammatory markersImproved mental status, reduced inflammation; no significant difference between dietary groups(153)
Cheng et al, 2023FMTPD patients12 weeksMotor symptoms (UPDRS), gut microbiota composition, quality of life (PDQ-39)Improved motor symptoms and quality of life; restored microbial diversity and SCFA production(154)
Dalile et al, 2020Colon-delivered SCFAsHealthy men under psychosocial stress4 weeksCortisol response, psychological stress (VAS), gut microbiotaAttenuated cortisol response to stress; modulated gut microbial composition(155)
Lei et al, 2022Disulfiram (bile acid modulation)Nonalcoholic steatohepatitis patients24 weeksLiver function, gut microbiota, bile acid profilesImproved liver function; restored gut microbiota and bile acid homeostasis(156)
Fleck et al, 2021Dietary conjugated linoleic acidPatients with multiple sclerosis12 weeksCNS autoimmunity markers, intestinal inflammation, gut barrier integrityReduced CNS inflammation; improved gut barrier function and microbiota diversity(157)
Martin et al, 2024Probiotic (Lactobacillus strain)Irritable bowel syndrome patients8 weeksMetabolomic, profiling psychological symptoms (HADS)Improved psychological comorbidities; altered gut metabolome linked to serotonin pathways(158)
Mysonhimer et al, 2023Prebiotic supplementationHealthy adults6 weeksMicrobiota biomarkers (cortisol), mental health questionnairesModulated microbiota composition; no significant changes in stress/inflammation biomarkers or mental health(159)
Li et al, 2024Bifidobacterium breve 207-1Healthy adults8 weeksLifestyle behaviors, mental wellness (DASS-21), gut microbiotaImproved mental wellness; increased Bifidobacterium abundance and SCFA levels(160)
Casertano et al 2024γ-aminobutyric acid-producing LactobacillusHealthy adults4 weeksCognitive reactivity (LEIDS-R), mood (POMS), gut-brain metabolitesReduced cognitive reactivity to negative mood; no improvement in cognitive performance(161)

[i] DASS-21, depression anxiety stress scales; STAI, state-trait anxiety inventory; CANTAB, Cambridge neuropsychological test automated battery; PSS, perceived stress scale; HAMD, Hamilton depression rating scale; MADRS: Montgomery-Åsberg depression rating scale; UPDRS, unified Parkinson's disease rating scale; PDQ-39, Parkinson's disease questionnaire; VAS, visual analog scale; HADS, hospital anxiety and depression scale; LEIDS-R, Leiden index of depression sensitivity-revised; POMS, profile of mood states.

Dietary interventions

Dietary interventions have emerged as a promising approach to modulate the gut-brain axis and improve neurological health. The Mediterranean diet, rich in vegetables, fruits, whole grains, legumes and olive oil, has been associated with beneficial effects on cognitive function and a reduced risk of neurodegenerative diseases. This diet promotes a healthy gut microbiota composition, increasing the abundance of beneficial bacteria such as Bifidobacterium and Lactobacillus, which produce SCFAs that exert anti-inflammatory effects and enhance gut barrier function. For example, Solch et al (146) found that adherence to the Mediterranean diet was associated with a lower risk of AD. The mechanisms underlying this effect may involve the production of SCFAs, which can modulate neuroinflammation, reinforce the BBB, and regulate neurotransmitter synthesis.

The ketogenic diet, high in fats and low in carbohydrates, has also gained attention for its neuroprotective effects, particularly in epilepsy and neurodegenerative disorders. By inducing ketosis, this diet alters the metabolic landscape of the brain, providing alternative energy substrates and reducing oxidative stress. Lussier et al (147) demonstrated that the ketogenic diet could inhibit tumor growth in a mouse model of glioblastoma multiforme. In addition to its direct metabolic effects, the ketogenic diet can also influence gut microbiota composition, increase the relative abundance of beneficial bacteria and reduce pathogenic species. This modulation of the gut microbiota may contribute to the diet's therapeutic effects on neurological diseases.

Fiber supplementation and prebiotics represent another avenue for targeting the gut-brain axis. Berding et al (148) demonstrated that supplementing with microbiota-accessible carbohydrates can prevent neuroinflammation and cognitive decline by improving the gut microbiota-brain axis in diet-induced obese mice. This evidence underscores the potential of fiber supplementation and prebiotics in modulating the gut-brain axis for neurological health. Alcohol addiction exacerbates gut dysbiosis in patients with minimal hepatic encephalopathy, further impairing cognitive function through increased gut-derived ammonia and systemic inflammation (149).

Probiotics and microbial therapies

Specific probiotic strains have been investigated for their effects on neurodegenerative and neuroinflammatory diseases. For instance, Lactobacillus and Bifidobacterium strains have demonstrated the ability to produce neurotransmitters such as GABA, serotonin and dopamine, which can influence brain function and behavior. Bravo et al (150) found that administration of Lactobacillus rhamnosus to mice reduced anxiety-like behavior and altered GABA receptor expression in the brain. In animal models of AD, probiotic supplementation with strains such as Lactobacillus plantarum and Bifidobacterium longum has been shown to reduce amyloid-beta pathology and improve cognitive performance. Srivastav et al (52) demonstrated that a probiotics mixture increased butyrate levels, which rescued nigral dopaminergic neurons from MPTP and rotenone-induced neurotoxicity.

Clinical trials in humans have also provided encouraging results. An RCT by Chong et al (151) found that Lactobacillus plantarum DR7 alleviated stress and anxiety in adults. Lew et al (152) showed that Lactobacillus plantarum P8 alleviated stress and anxiety while enhancing memory and cognition in stressed adults. In patients with major depressive disorder, probiotic supplementation has been associated with improvements in mental status, inflammation and intestinal barrier function. Karakula-Juchnowicz et al (153) evaluated the effect of probiotic supplementation on the mental status, inflammation and intestinal barrier in patients with major depressive disorder using a gluten-free or gluten-containing diet. The results showed that probiotic supplementation improved mental status and reduced inflammation in both groups.

FMT, the transfer of microbiota from a healthy donor to a recipient, has gained interest as a therapeutic approach for neurological diseases. FMT aims to restore a healthy gut microbiota composition and function, potentially reversing dysbiosis-associated neurological pathology. Preclinical studies have shown promising results in various neurological disease models. Cheng et al (154) investigated the efficacy of FMT in patients with PD. The results showed that FMT improved motor symptoms and quality of life in patients with PD. These preliminary data (n=47) lack blinding and showed transient engraftment (<8 weeks) in 30% of recipients. Adverse events including bloating (23%) and transient delirium (4%) further caution against uncritical adoption outside controlled trials.

However, translating these findings to clinical practice presents several challenges. The mechanisms underlying FMT's therapeutic effects remain incompletely understood, and there is a need for standardized protocols regarding donor selection, preparation of fecal microbiota, and administration routes. Additionally, safety concerns related to the transmission of pathogens or harmful metabolites necessitate rigorous screening of donors and donor materials. Despite these challenges, ongoing clinical trials are exploring the efficacy and safety of FMT in neurological conditions such as MS, AD and PD, which may provide valuable insights into its therapeutic potential in the near future.

Pharmacological approaches

Pharmacological strategies targeting microbiota-derived metabolites offer another avenue for modulating the gut-brain axis. SCFAs analogs and bile acid sequestrants represent two classes of agents with potential therapeutic applications in neurological diseases. Dalile et al (155) demonstrated that colon-delivered SCFAs attenuated the cortisol response to psychosocial stress in healthy men.

Bile acid sequestrants, such as cholestyramine, bind to bile acids in the gut, altering their enterohepatic circulation and potentially modulating bile acid signaling pathways. Recent research has shown that bile acids can influence gut microbiota composition and immune responses, suggesting that sequestrants may indirectly affect the gut-brain axis. In animal models of PD, cholestyramine treatment reduced alpha-synuclein pathology and improved motor function, highlighting the potential of this approach in neurodegenerative diseases. Lei et al (156) demonstrated that disulfiram ameliorated nonalcoholic steatohepatitis by modulating the gut microbiota and bile acid metabolism.

Immune modulators and anti-inflammatory agents represent a broad category of pharmacological interventions with potential applications in targeting the gut-brain axis. For instance, anti-tumor necrosis factor (TNF) agents, such as infliximab and adalimumab, have been used in the treatment of IBD and have shown promise in ameliorating neurological symptoms in patients with comorbid IBD and neurological disorders. Similarly, interleukin (IL)-17 and IL-23 inhibitors are being investigated for their potential to modulate neuroinflammation in conditions such as MS and PD. Fleck et al (157) found that dietary conjugated linoleic acid links reduced intestinal inflammation to amelioration of CNS autoimmunity.

Recent clinical trials have explored the use of these agents in neurological diseases. A phase II trial of the IL-17 inhibitor secukinumab in patients with relapsing-remitting MS demonstrated a reduction in disease activity and MRI lesions, suggesting a potential role for targeting IL-17 signaling in this condition. Additionally, the TNF inhibitor etanercept has shown beneficial effects in patients with PD, with improvements in motor symptoms and quality of life. These findings highlight the potential of immune modulators in targeting gut-brain pathways for neurological health.

Emerging therapies

Advances in synthetic biology and genetic engineering have paved the way for the development of engineered microbiota with tailored functions for therapeutic applications. These engineered bacteria can be designed to produce specific metabolites, enzymes, or anti-inflammatory molecules that target key pathways in neurological diseases. Martin et al (158) demonstrated that metabolome-associated psychological comorbidities improved in patients with irritable bowel syndrome receiving a probiotic. Moreover, synthetic biology approaches allow for the creation of bacteria with sensing and responsive capabilities, enabling them to detect specific disease biomarkers and release therapeutic agents in a targeted manner. This precision medicine approach could enhance the efficacy and safety of microbiota-based therapies for neurological disorders. Mysonhimer et al (159) found that prebiotic consumption alters microbiota but not biological markers of stress and inflammation or mental health symptoms in healthy adults.

Nanomedicine and advanced drug delivery systems offer innovative strategies to enhance the targeting and efficacy of therapeutics aimed at the gut-brain axis. Nanoparticles can be engineered to encapsulate drugs, probiotics, or bioactive compounds, protecting them from degradation and enabling controlled release at specific sites along the gastrointestinal tract. For instance, chitosan nanoparticles loaded with curcumin, a polyphenol with anti-inflammatory and antioxidant properties, have shown enhanced bioavailability and therapeutic efficacy in animal models of AD and PD. These nanoparticles can traverse the BBB and deliver curcumin to the brain, where it exerts neuroprotective effects. Similarly, lipid-based nanoparticles have been used to deliver probiotics to the gut, improving their survival and colonization efficiency. Li et al (160) investigated the effects of Bifidobacterium breve 207-1 on regulating lifestyle behaviors and mental wellness in healthy adults based on the microbiome-gut-brain axis. The results showed that Bifidobacterium breve 207-1 improved mental wellness and regulated lifestyle behaviors in healthy adults.

Additionally, targeted drug delivery systems can be designed to specifically interact with gut microbiota or immune cells, modulating their function in a precise manner. For example, antibody-conjugated nanoparticles can target specific bacterial strains or immune cells, delivering therapeutic agents directly to the site of action. These approaches hold promise for improving the therapeutic index of existing drugs and developing novel interventions for neurological diseases by leveraging the gut-brain axis. Casertano et al (161) demonstrated that GABA-producing lactobacilli boost cognitive reactivity to negative mood without improving cognitive performance.

Future directions and challenges

The gut-brain axis serves as a promising source of biomarkers for neurological diseases. Recent studies have identified specific gut microbiota signatures associated with conditions such as AD, PD and MS (162-164). These signatures can potentially be used for early diagnosis, disease progression monitoring and treatment response prediction. For example, certain bacterial taxa such as Bifidobacterium and Lactobacillus have been found to be decreased in patients with AD, whereas an increase in pathogenic bacteria such as Escherichia coli has been observed (165). Similarly, in PD, specific microbial metabolites such as SCFAs and secondary bile acids have shown potential as biomarkers because of their altered levels in patient sera (166). The development of these biomarkers is crucial for improving diagnostic accuracy and personalizing treatment approaches.

Personalized medicine holds considerable promise for treating neurological disorders by tailoring treatments to individual patients on the basis of their unique gut microbiota profiles. This approach involves a comprehensive analysis of a patient's gut microbiota composition, genetic makeup and metabolic status to design targeted therapeutic interventions (14). For example, patients with specific microbial imbalances could be prescribed customized probiotic formulations or dietary plans to restore microbial balance and improve neurological outcomes (45). Additionally, integrating gut microbiota data with other omics data (genomic, proteomic and metabolomic data) can provide a more holistic understanding of disease mechanisms and enable the development of multitargeted therapies (14). Clinical trials are underway to validate the efficacy of such personalized approaches and establish standardized protocols for implementation in clinical practice.

Emerging comparative analyses indicate that precision-medicine paradigms outperform universal interventions across neurological disorders. Multi-strain probiotics co-administered with dietary substrates (prebiotics) currently yield the strongest clinical signal for stress-related disorders and early PD, achieving significant reductions in anxiety and Unified PD Rating Scale motor scores in randomized trials lasting ≥12 weeks (167). In AD, SCFA-enriched Mediterranean diets outperform single-probiotic regimens by simultaneously improving gut barrier integrity, systemic inflammation and cognitive composite scores (167,168). FMT retains promise for refractory epilepsy and pediatric ASD, yet pooled data demonstrate 30-40% responder rates only when donor selection is paired with hostmicrobiome matching algorithms and baseline α-diversity is low (19). In diseases, compared with non-personalized probiotics, intervention measures integrating multi-omics stratification (baseline microbiota, metabolome, host genetics) have improved effects (167). Consequently, combination strategies that couple dietary modification with targeted microbial restoration appear most effective, whereas single-agent probiotics or unpersonalized FMT offer limited and transient benefits.

Despite promising preclinical evidence, the translation of gut-brain axis research into clinical practice faces several significant challenges (169). One major hurdle is establishing a definitive causal relationship between gut microbiota alterations and neurological diseases. Numerous studies have identified correlations, but determining whether these effects are causative, consequential, or merely bystander responses remains difficult (170). Another challenge is the complexity of the gut-brain axis itself, with its multitude of interacting components and pathways making it challenging to identify specific targets for intervention (171). Furthermore, existing animal models do not fully recapitulate human pathophysiology, thus limiting the translational value of preclinical findings (172). Additionally, there are technical limitations in accurately measuring and manipulating the gut microbiota among humans, as well as regulatory and ethical considerations regarding the use of microbiota-based therapies (173). Addressing these challenges requires multidisciplinary collaboration, rigorous clinical trials and the development of advanced technologies for microbiota analysis and manipulation.

Numerous clinical trials are currently ongoing or planned to investigate therapies targeting the gut-brain axis in neurological disorders. These trials encompass a range of approaches, including probiotics, prebiotics, FMT, dietary interventions and pharmacological agents. For example, several trials are evaluating the efficacy of specific probiotic strains in improving cognitive function in patients with AD and motor symptoms in patients with PD (174-176). Other trials are exploring the potential of FMT to modulate the gut microbiota and reduce disease activity in patients with MS and IBD with neurological complications (177,178). Additionally, research is ongoing to develop novel drugs that can specifically target gut-brain communication pathways, such as inhibitors of microbial metabolite production or modulators of immune signaling (179). These clinical trials are essential for validating the therapeutic potential of gut-brain axis-targeted strategies and establishing evidence-based treatment guidelines.

Despite emerging evidence, the gut-brain axis field faces three critical gaps: Causality vs. correlation, as most human studies remain associative, and Mendelian randomization and long-term gnotobiotic models are needed to clarify whether dysbiosis initiates or merely mirrors neurological disease; microbiome stability, as the rate of probiotic engraftment falls <30% after 4 weeks and high interindividual variability undermining reproducibility (180); and clinical heterogeneity, exemplified by recent negative RCTs in AD (NCT04168974) and early PD that reveal 'one-size-fits-all' interventions to be ineffective, thereby requiring precision-medicine frameworks integrating host genetics, metabolomics and real-time dietary data. Furthermore, there is the lack of standardized protocols for FMT-donor selection, dosing and delivery routes vary widely, leading to inconsistent engraftment and safety signals. Adopting consensus guidelines such as those proposed by the International Scientific Association for Probiotics and Prebiotics will be essential to harmonize endpoints, biomarker panels and adverse-event reporting, ultimately determining whether microbiota-directed therapeutics can deliver reproducible neurological benefits.

Conclusion

In conclusion, the gut-brain axis represents a fascinating and promising frontier in neurological research with significant clinical implications. By developing biomarkers based on gut-brain axis components, adopting personalized medicine approaches, overcoming translational challenges and conducting rigorous clinical trials, the therapeutic potential of the gut-brain axis can be harnessed to improve outcomes for patients with neurological diseases. Future research should continue to explore the intricate mechanisms underlying gut-brain communication and expand our understanding of how to effectively modulate this axis for therapeutic benefit.

Availability of data and materials

Not applicable.

Authors' contributions

HZ and JL contributed equally to the research and writing process. LH provided substantial input on the clinical laboratory aspects. XP offered valuable insights from the neurological department's perspective. HZ, JL, LH, XP, HZ, YL and HL participated in the critical revision of the manuscript for important intellectual content. All authors read and approved the final version of the manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgements

Not applicable.

Funding

No funding was received.

References

1 

Morais LH, Schreiber HL 4th and Mazmanian SK: The gut microbiota-brain axis in behaviour and brain disorders. Nat Rev Microbiol. 19:241–255. 2021. View Article : Google Scholar

2 

Chanpong A, Borrelli O and Thapar N: Recent advances in understanding the roles of the enteric nervous system. Fac Rev. 11:72022. View Article : Google Scholar : PubMed/NCBI

3 

Carabotti M, Scirocco A, Maselli MA and Severi C: The gut-brain axis: Interactions between enteric microbiota, central and enteric nervous systems. Ann Gastroenterol. 28:203–209. 2015.PubMed/NCBI

4 

Schluter J, Peled JU, Taylor BP, Markey KA, Smith M, Taur Y, Niehus R, Staffas A, Dai A, Fontana E, et al: The gut microbiota is associated with immune cell dynamics in humans. Nature. 588:303–307. 2020. View Article : Google Scholar : PubMed/NCBI

5 

Gwak MG and Chang SY: Gut-brain connection: Microbiome, gut barrier, and environmental sensors. Immune Netw. 21:e202021. View Article : Google Scholar : PubMed/NCBI

6 

Appleton J: The Gut-brain axis: Influence of microbiota on mood and mental health. Integr Med (Encinitas). 17:28–32. 2018.

7 

Pucci A and Batterham RL: Endocrinology of the gut and the regulation of body weight and metabolism. Endotext. Feingold KR, Anawalt B, Blackman MR, et al: MDText. com, Inc. Copyright © 2000-2025. MDText.com, Inc.; South Dartmouth (MA): 2000

8 

Konsman JP: Cytokines in the Brain and Neuroinflammation: We Didn't Starve the Fire! Pharmaceuticals (Basel). 15:1402022. View Article : Google Scholar : PubMed/NCBI

9 

Köhler A, Delbauve S, Smout J, Torres D and Flamand V: Very early-life exposure to microbiota-induced TNF drives the maturation of neonatal pre-cDC1. Gut. 70:511–521. 2021. View Article : Google Scholar

10 

Hrncir T: Gut microbiota dysbiosis: Triggers, consequences, diagnostic and therapeutic options. Microorganisms. 10:5782022. View Article : Google Scholar : PubMed/NCBI

11 

Quansah M, David MA, Martins R, El-Omar E, Aliberti SM, Capunzo M, Jensen SO and Tayebi M: The beneficial effects of lactobacillus strains on gut microbiome in Alzheimer's disease: A systematic review. Healthcare. 13:742025. View Article : Google Scholar : PubMed/NCBI

12 

Zhang X, Wang Y, Liu W, Wang T, Wang L, Hao L, Ju M and Xiao R: Diet quality, gut microbiota, and microRNAs associated with mild cognitive impairment in middle-aged and elderly Chinese population. Am J Clin Nutr. 114:429–440. 2021. View Article : Google Scholar : PubMed/NCBI

13 

Li B, He Y, Ma J, Huang P, Du J, Cao L, Wang Y, Xiao Q, Tang H and Chen S: Mild cognitive impairment has similar alterations as Alzheimer's disease in gut microbiota. Alzheimers Dement. 15:1357–1366. 2019. View Article : Google Scholar : PubMed/NCBI

14 

Tan AH, Lim SY and Lang AE: The microbiome-gut-brain axis in Parkinson disease-from basic research to the clinic. Nat Rev Neurol. 18:476–495. 2022. View Article : Google Scholar : PubMed/NCBI

15 

Casella G, Tontini GE, Bassotti G, Pastorelli L, Villanacci V, Spina L, Baldini V and Vecchi M: Neurological disorders and inflammatory bowel diseases. World J Gastroenterol. 20:8764–8782. 2014.PubMed/NCBI

16 

Yaqubi K, Kostev K, Klein I, Schüssler S, May P, Luedde T, Roderburg C and Loosen SH: Inflammatory bowel disease is associated with an increase in the incidence of multiple sclerosis: A retrospective cohort study of 24,934 patients. Eur J Med Res. 29:1862024. View Article : Google Scholar : PubMed/NCBI

17 

Hughes HK, Rose D and Ashwood P: The gut microbiota and dysbiosis in autism spectrum disorders. Curr Neurol Neurosci Rep. 18:812018. View Article : Google Scholar : PubMed/NCBI

18 

Naspolini NF, Natividade AP, Asmus CIF, Moreira JC, Dominguez-Bello MG and Meyer A: Early-life gut microbiome is associated with behavioral disorders in the Rio birth cohort. Sci Rep. 15:86742025. View Article : Google Scholar : PubMed/NCBI

19 

Doenyas C, Clarke G and Cserjési R: Gut-brain axis and neuropsychiatric health: Recent advances. Sci Rep. 15:34152025. View Article : Google Scholar : PubMed/NCBI

20 

Holmes GM and Blanke EN: Gastrointestinal dysfunction after spinal cord injury. Exp Neurol. 320:1130092019. View Article : Google Scholar : PubMed/NCBI

21 

Su Y, Wang D, Liu N, Yang J, Sun R and Zhang Z: Clostridium butyricum improves cognitive dysfunction in ICV-STZ-induced Alzheimer's disease mice via suppressing TLR4 signaling pathway through the gut-brain axis. PLoS One. 18:e02860862023. View Article : Google Scholar : PubMed/NCBI

22 

Fan Y and Pedersen O: Gut microbiota in human metabolic health and disease. Nat Rev Microbiol. 19:55–71. 2021. View Article : Google Scholar

23 

Ahmed H, Leyrolle Q, Koistinen V, Kärkkäinen O, Layé S, Delzenne N and Hanhineva K: Microbiota-derived metabolites as drivers of gut-brain communication. Gut Microbes. 14:21028782022. View Article : Google Scholar : PubMed/NCBI

24 

Oleskin AV and Shenderov BA: Neuromodulatory effects and targets of the SCFAs and gasotransmitters produced by the human symbiotic microbiota. Microb Ecol Health Dis. 27:309712016.PubMed/NCBI

25 

Ramesh G, MacLean AG and Philipp MT: Cytokines and chemokines at the crossroads of neuroinflammation, neurodegeneration, and neuropathic pain. Mediators Inflamm. 2013:4807392013. View Article : Google Scholar : PubMed/NCBI

26 

Person H and Keefer L: Psychological comorbidity in gastrointestinal diseases: Update on the brain-gut-microbiome axis. Prog Neuropsychopharmacol Biol Psychiatry. 107:1102092021. View Article : Google Scholar :

27 

Mayer EA, Nance K and Chen S: The gut-brain axis. Annu Rev Med. 73:439–453. 2022. View Article : Google Scholar

28 

Makris AP, Karianaki M, Tsamis KI and Paschou SA: The role of the gut-brain axis in depression: Endocrine, neural, and immune pathways. Hormones (Athens). 20:1–12. 2021. View Article : Google Scholar

29 

Ohara TE and Hsiao EY: Microbiota-neuroepithelial signalling across the gut-brain axis. Nat Rev Microbiol. 23:371–384. 2025. View Article : Google Scholar : PubMed/NCBI

30 

Rastogi S and Singh A: Gut microbiome and human health: Exploring how the probiotic genus Lactobacillus modulate immune responses. Front Pharmacol. 13:10421892022. View Article : Google Scholar : PubMed/NCBI

31 

Parizadeh M and Arrieta MC: The global human gut microbiome: Genes, lifestyles, and diet. Trends Mol Med. 29:789–801. 2023. View Article : Google Scholar : PubMed/NCBI

32 

Procházková N, Laursen MF, La Barbera G, Tsekitsidi E, Jørgensen MS, Rasmussen MA, Raes J, Licht TR, Dragsted LO and Roager HM: Gut physiology and environment explain variations in human gut microbiome composition and metabolism. Nat Microbiol. 9:3210–3225. 2024. View Article : Google Scholar : PubMed/NCBI

33 

Gomaa EZ: Human gut microbiota/microbiome in health and diseases: A review. Antonie Van Leeuwenhoek. 113:2019–2040. 2020. View Article : Google Scholar : PubMed/NCBI

34 

Kearns R: Gut-brain axis and neuroinflammation: The role of gut permeability and the kynurenine pathway in neurological disorders. Cell Mol Neurobiol. 44:642024. View Article : Google Scholar : PubMed/NCBI

35 

Kearns R: The kynurenine pathway in gut permeability and inflammation. Inflammation. 48:1063–1077. 2025. View Article : Google Scholar

36 

Koh A, De Vadder F, Kovatcheva-Datchary P and Bäckhed F: From dietary fiber to host physiology: Short-chain fatty acids as key bacterial metabolites. Cell. 165:1332–1345. 2016. View Article : Google Scholar : PubMed/NCBI

37 

Fock E and Parnova R: Mechanisms of Blood-brain barrier protection by Microbiota-derived short-chain fatty acids. Cells. 12:6572023. View Article : Google Scholar : PubMed/NCBI

38 

Jia M, Fan Y, Ma Q, Yang D, Wang Y, He X, Zhao B, Zhan X, Qi Z, Ren Y, et al: Gut microbiota dysbiosis promotes cognitive impairment via bile acid metabolism in major depressive disorder. Transl Psychiatry. 14:5032024. View Article : Google Scholar : PubMed/NCBI

39 

Hou Y, Li J and Ying S: Tryptophan metabolism and gut microbiota: A novel regulatory axis integrating the microbiome, immunity, and cancer. Metabolites. 13:11662023. View Article : Google Scholar : PubMed/NCBI

40 

Luczynski P, McVey Neufeld KA, Oriach CS, Clarke G, Dinan TG and Cryan JF: Growing up in a Bubble: Using Germ-free animals to assess the influence of the gut microbiota on brain and behavior. Int J Neuropsychopharmacol. 19:pyw0202016. View Article : Google Scholar : PubMed/NCBI

41 

Mhanna A, Martini N, Hmaydoosh G, Hamwi G, Jarjanazi M, Zaifah G, Kazzazo R, Haji Mohamad A and Alshehabi Z: The correlation between gut microbiota and both neurotransmitters and mental disorders: A narrative review. Medicine. 103:e371142024. View Article : Google Scholar : PubMed/NCBI

42 

Gu F, Wu Y, Liu Y, Dou M, Jiang Y and Liang H: Lactobacillus casei improves depression-like behavior in chronic unpredictable mild stress-induced rats by the BDNF-TrkB signal pathway and the intestinal microbiota. Food Funct. 11:6148–6157. 2020. View Article : Google Scholar : PubMed/NCBI

43 

Hwang YK and Oh JS: Interaction of the vagus nerve and serotonin in the Gut-brain axis. Int J Mol Sci. 26:11602025. View Article : Google Scholar : PubMed/NCBI

44 

Sorboni SG, Moghaddam HS, Jafarzadeh-Esfehani R and Soleimanpour S: A comprehensive review on the role of the gut microbiome in human neurological disorders. Clin Microbiol Rev. 35:e00338202022. View Article : Google Scholar : PubMed/NCBI

45 

Cryan JF, O'Riordan KJ, Sandhu K, Peterson V and Dinan TG: The gut microbiome in neurological disorders. Lancet Neurol. 19:179–194. 2020. View Article : Google Scholar

46 

Liu C, Yang SY, Wang L and Zhou F: The gut microbiome: Implications for neurogenesis and neurological diseases. Neural Regen Res. 17:53–58. 2022. View Article : Google Scholar

47 

Socała K, Doboszewska U, Szopa A, Serefko A, Włodarczyk M, Zielińska A, Poleszak E, Fichna J and Wlaź P: The role of microbiota-gut-brain axis in neuropsychiatric and neurological disorders. Pharmacol Res. 172:1058402021. View Article : Google Scholar

48 

Hirschberg S, Gisevius B, Duscha A and Haghikia A: Implications of diet and the gut microbiome in neuroinflammatory and neurodegenerative diseases. Int J Mol Sci. 20:31092019. View Article : Google Scholar : PubMed/NCBI

49 

Ivan IF, Irincu VL, Diaconu Ș, Falup-Pecurariu O, Ciopleiaș B and Falup-Pecurariu C: Gastro-intestinal dysfunctions in Parkinson's disease (review). Exp Ther Med. 22:10832021. View Article : Google Scholar : PubMed/NCBI

50 

Sampson TR, Debelius JW, Thron T, Janssen S, Shastri GG, Ilhan ZE, Challis C, Schretter CE, Rocha S, Gradinaru V, et al: Gut microbiota regulate motor deficits and neuroinflammation in a model of Parkinson's disease. Cell. 167:1469–1480.e12. 2016. View Article : Google Scholar : PubMed/NCBI

51 

Perez-Pardo P, Dodiya HB, Engen PA, Forsyth CB, Huschens AM, Shaikh M, Voigt RM, Naqib A, Green SJ, Kordower JH, et al: Role of TLR4 in the gut-brain axis in Parkinson's disease: A translational study from men to mice. Gut. 68:829–843. 2019. View Article : Google Scholar

52 

Srivastav S, Neupane S, Bhurtel S, Katila N, Maharjan S, Choi H, Hong JT and Choi DY: Probiotics mixture increases butyrate, and subsequently rescues the nigral dopaminergic neurons from MPTP and rotenone-induced neurotoxicity. J Nutr Biochem. 69:73–86. 2019. View Article : Google Scholar : PubMed/NCBI

53 

Lei Q, Wu T, Wu J, Hu X, Guan Y, Wang Y, Yan J and Shi G: Roles of α-synuclein in gastrointestinal microbiome dysbiosis-related Parkinson's disease progression (review). Mol Med Rep. 24:7342021. View Article : Google Scholar

54 

Honarpisheh P, Reynolds CR, Blasco Conesa MP, Moruno Manchon JF, Putluri N, Bhattacharjee MB, Urayama A, McCullough LD and Ganesh BP: Dysregulated gut homeostasis observed prior to the accumulation of the brain amyloid-β in Tg2576 mice. Int J Mol Sci. 21:17112020. View Article : Google Scholar

55 

Kim N, Jeon SH, Ju IG, Gee MS, Do J, Oh MS and Lee JK: Transplantation of gut microbiota derived from Alzheimer's disease mouse model impairs memory function and neurogenesis in C57BL/6 mice. Brain Behav Immun. 98:357–365. 2021. View Article : Google Scholar : PubMed/NCBI

56 

Shi H, Ge X, Ma X, Zheng M, Cui X, Pan W, Zheng P, Yang X, Zhang P, Hu M, et al: A fiber-deprived diet causes cognitive impairment and hippocampal microglia-mediated synaptic loss through the gut microbiota and metabolites. Microbiome. 9:2232021. View Article : Google Scholar : PubMed/NCBI

57 

Zhang T, Wang T, Chen X, Zhao Z and Chen Z: Gut microbiota relieves inflammation in the substantia nigra of chronic Parkinson's disease by protecting the function of dopamine neurons. Exp Ther Med. 23:522022. View Article : Google Scholar

58 

Castelli V, d'Angelo M, Quintiliani M, Benedetti E, Cifone MG and Cimini A: The emerging role of probiotics in neurodegenerative diseases: New hope for Parkinson's disease? Neural Regen Res. 16:628–634. 2021. View Article : Google Scholar

59 

Yuan T, Chu C, Shi R, Cui T, Zhang X, Zhao Y, Shi X, Hui Y, Pan J, Qian R, et al: ApoE-dependent protective effects of sesamol on High-Fat Diet-induced behavioral disorders: Regulation of the microbiome-Gut-brain axis. J Agric Food Chem. 67:6190–6201. 2019. View Article : Google Scholar : PubMed/NCBI

60 

Shi H, Wang Q, Zheng M, Hao S, Lum JS, Chen X, Huang XF, Yu Y and Zheng K: Supplement of microbiota-accessible carbohydrates prevents neuroinflammation and cognitive decline by improving the gut microbiota-brain axis in diet-induced obese mice. J Neuroinflammation. 17:772020. View Article : Google Scholar : PubMed/NCBI

61 

Bianchimano P, Britton GJ, Wallach DS, Smith EM, Cox LM, Liu S, Iwanowski K, Weiner HL, Faith JJ, Clemente JC and Tankou SK: Mining the microbiota to identify gut commensals modulating neuroinflammation in a mouse model of multiple sclerosis. Microbiome. 10:1742022. View Article : Google Scholar : PubMed/NCBI

62 

Celorrio M and Friess SH: Gut-brain axis in traumatic brain injury: Impact on neuroinflammation. Neural Regen Res. 17:1007–1008. 2022. View Article : Google Scholar

63 

Theleritis C, Stefanou MI, Demetriou M, Alevyzakis E, Triantafyllou K, Smyrnis N, Spandidos DA and Rizos E: Association of gut dysbiosis with first-episode psychosis (review). Mol Med Rep. 30:1302024. View Article : Google Scholar

64 

Torres-Chávez ME, Torres-Carrillo NM, Monreal-Lugo AV, Garnés-Rancurello S, Murugesan S, Gutiérrez-Hurtado IA, Beltrán-Ramírez JR, Sandoval-Pinto E and Torres-Carrillo N: Association of intestinal dysbiosis with susceptibility to multiple sclerosis: Evidence from different population studies (review). Biomed Rep. 19:932023. View Article : Google Scholar : PubMed/NCBI

65 

Lee CW, Hsu LF, Wu IL, Wang YL, Chen WC, Liu YJ, Yang LT, Tan CL, Luo YH, Wang CC, et al: Exposure to polystyrene microplastics impairs hippocampus-dependent learning and memory in mice. J Hazard Mater. 430:1284312022. View Article : Google Scholar : PubMed/NCBI

66 

Yuan S, Yang J, Jian Y, Lei Y, Yao S, Hu Z, Liu X, Tang C and Liu W: Treadmill exercise modulates intestinal microbes and suppresses LPS displacement to alleviate neuroinflammation in the brains of APP/PS1 Mice. Nutrients. 14:41342022. View Article : Google Scholar : PubMed/NCBI

67 

Hu X, Jin H, Yuan S, Ye T, Chen Z, Kong Y, Liu J, Xu K and Sun J: Fecal microbiota transplantation inhibited neuroinflammation of traumatic brain injury in mice via regulating the gut-brain axis. Front Cell Infect Microbiol. 13:12546102023. View Article : Google Scholar : PubMed/NCBI

68 

Li X, Zheng P, Cao W, Cao Y, She X, Yang H, Ma K, Wu F, Gao X, Fu Y, et al: Lactobacillus rhamnosus GG ameliorates noise-induced cognitive deficits and systemic inflammation in rats by modulating the gut-brain axis. Front Cell Infect Microbiol. 13:10673672023. View Article : Google Scholar : PubMed/NCBI

69 

Wei H, Yu C, Zhang C, Ren Y, Guo L, Wang T, Chen F, Li Y, Zhang X, Wang H and Liu J: Butyrate ameliorates chronic alcoholic central nervous damage by suppressing microglia-mediated neuroinflammation and modulating the microbiome-gut-brain axis. Biomed Pharmacother. 160:1143082023. View Article : Google Scholar : PubMed/NCBI

70 

Zhu G, Zhao J, Wang G and Chen W: Bifidobacterium breve HNXY26M4 attenuates cognitive deficits and neuroinflammation by regulating the Gut-brain axis in APP/PS1 mice. J Agric Food Chem. 71:4646–4655. 2023. View Article : Google Scholar : PubMed/NCBI

71 

de Theije CG, Koelink PJ, Korte-Bouws GA, Lopes da Silva S, Korte SM, Olivier B, Garssen J and Kraneveld AD: Intestinal inflammation in a murine model of autism spectrum disorders. Brain Behav Immun. 37:240–247. 2014. View Article : Google Scholar

72 

Stilling RM, Ryan FJ, Hoban AE, Shanahan F, Clarke G, Claesson MJ, Dinan TG and Cryan JF: Microbes & neurodevelopment-Absence of microbiota during early life increases activity-related transcriptional pathways in the amygdala. Brain Behav Immun. 50:209–220. 2015. View Article : Google Scholar : PubMed/NCBI

73 

Kantarcioglu AS, Kiraz N and Aydin A: Microbiota-gut-brain axis: Yeast species isolated from stool samples of children with suspected or diagnosed autism spectrum disorders and in vitro susceptibility against nystatin and fluconazole. Mycopathologia. 181:1–7. 2016. View Article : Google Scholar

74 

Abuljadayel D, Alotibi A, Algothmi K, Basingab F, Alhazmi S, Almuhammadi A, Alharthi A, Alyoubi R and Bahieldin A: Gut microbiota of children with autism spectrum disorder and healthy siblings: A comparative study. Exp Ther Med. 28:4302024. View Article : Google Scholar : PubMed/NCBI

75 

Dai W, Liu J, Qiu Y, Teng Z, Li S, Yuan H, Huang J, Xiang H, Tang H, Wang B, et al: Gut microbial dysbiosis and cognitive impairment in bipolar disorder: Current evidence. Front Pharmacol. 13:8935672022. View Article : Google Scholar : PubMed/NCBI

76 

Bioque M, González-Rodríguez A, Garcia-Rizo C, Cobo J, Monreal JA, Usall J, Soria V; PNECAT Group; Labad J: Targeting the microbiome-gut-brain axis for improving cognition in schizophrenia and major mood disorders: A narrative review. Prog Neuropsychopharmacol Biol Psychiatry. 105:1101302021. View Article : Google Scholar

77 

Reddy-Thootkur M, Kraguljac NV and Lahti AC: The role of glutamate and GABA in cognitive dysfunction in schizophrenia and mood disorders-A systematic review of magnetic resonance spectroscopy studies. Schizophr Res. 249:74–84. 2022. View Article : Google Scholar

78 

Reininghaus EZ, Wetzlmair LC, Fellendorf FT, Platzer M, Queissner R, Birner A, Pilz R, Hamm C, Maget A, Koidl C, et al: The impact of probiotic supplements on cognitive parameters in euthymic individuals with bipolar disorder: A pilot study. Neuropsychobiology. 1–8. 2018. View Article : Google Scholar : Epub ahead of print. PubMed/NCBI

79 

Pulikkan J, Maji A, Dhakan DB, Saxena R, Mohan B, Anto MM, Agarwal N, Grace T and Sharma VK: Gut microbial dysbiosis in indian children with autism spectrum disorders. Microb Ecol. 76:1102–1114. 2018. View Article : Google Scholar : PubMed/NCBI

80 

Bojović K, Ignjatović ÐI, Soković Bajić S, Vojnović Milutinović D, Tomić M, Golić N and Tolinački M: Gut microbiota dysbiosis associated with altered production of short chain fatty acids in children with neurodevelopmental disorders. Front Cell Infect Microbiol. 10:2232020. View Article : Google Scholar

81 

Slob EM A, Brew BK, Vijverberg SJH, Dijs T, van Beijsterveldt CEM, Koppelman GH, Bartels M, Dolan CV, Larsson H, Lundström S, et al: Early-life antibiotic use and risk of attention-deficit hyperactivity disorder and autism spectrum disorder: Results of a discordant twin study. Int J Epidemiol. 50:475–484. 2021. View Article : Google Scholar

82 

Bundgaard-Nielsen C, Lauritsen MB, Knudsen JK, Rold LS, Larsen MH, Hindersson P, Villadsen AB, Leutscher PDC, Hagstrøm S, Nyegaard M and Sørensen S: Children and adolescents with attention deficit hyperactivity disorder and autism spectrum disorder share distinct microbiota compositions. Gut Microbes. 15:22119232023. View Article : Google Scholar : PubMed/NCBI

83 

Liu S, Gao J, Zhu M, Liu K and Zhang HL: Gut microbiota and dysbiosis in Alzheimer's disease: Implications for pathogenesis and treatment. Mol Neurobiol. 57:5026–5043. 2020. View Article : Google Scholar : PubMed/NCBI

84 

Gonçalves CL, Vasconcelos FFP, Wessler LB, Lemos IS, Candiotto G, Lin J, Matias MBD, Rico EP and Streck EL: Exposure to a high dose of amoxicillin causes behavioral changes and oxidative stress in young zebrafish. Metab Brain Dis. 35:1407–1416. 2020. View Article : Google Scholar : PubMed/NCBI

85 

Volkova A, Ruggles K, Schulfer A, Gao Z, Ginsberg SD and Blaser MJ: Effects of early-life penicillin exposure on the gut microbiome and frontal cortex and amygdala gene expression. iScience. 24:1027972021. View Article : Google Scholar : PubMed/NCBI

86 

Benakis C, Brea D, Caballero S, Faraco G, Moore J, Murphy M, Sita G, Racchumi G, Ling L, Pamer EG, et al: Commensal microbiota affects ischemic stroke outcome by regulating intestinal γδ T cells. Nat Med. 22:516–523. 2016. View Article : Google Scholar : PubMed/NCBI

87 

Dou Z, Rong X, Zhao E, Zhang L and Lv Y: Neuroprotection of resveratrol against focal cerebral ischemia/reperfusion injury in mice through a mechanism targeting Gut-brain axis. Cell Mol Neurobiol. 39:883–898. 2019. View Article : Google Scholar : PubMed/NCBI

88 

Bonsack B, Jiang RH and Borlongan CV: A gut feeling about stroke reveals gut-brain axis' active role in homeostasis and dysbiosis. J Cereb Blood Flow Metab. 40:1132–1134. 2020. View Article : Google Scholar : PubMed/NCBI

89 

Zhang Y, Yang H, Hou S, Xia Y and Wang YQ: Influence of the brain-gut axis on neuroinflammation in cerebral ischemia-reperfusion injury (Review). Int J Mol Med. 53:302024. View Article : Google Scholar :

90 

Park MJ, Pilla R, Panta A, Pandey S, Sarawichitr B, Suchodolski J and Sohrabji F: Reproductive senescence and ischemic stroke remodel the gut microbiome and modulate the effects of estrogen treatment in female rats. Transl Stroke Res. 11:812–830. 2020. View Article : Google Scholar

91 

Sadler R, Cramer JV, Heindl S, Kostidis S, Betz D, Zuurbier KR, Northoff BH, Heijink M, Goldberg MP, Plautz EJ, et al: Short-chain fatty acids improve poststroke recovery via immunological mechanisms. J Neurosci. 40:1162–1173. 2020. View Article : Google Scholar : PubMed/NCBI

92 

Aswendt M, Green C, Sadler R, Llovera G, Dzikowski L, Heindl S, Gomez de Agüero M, Diedenhofen M, Vogel S, Wieters F, et al: The gut microbiota modulates brain network connectivity under physiological conditions and after acute brain ischemia. iScience. 24:1030952021. View Article : Google Scholar : PubMed/NCBI

93 

Ji W, Zhu Y, Kan P, Cai Y, Wang Z, Wu Z and Yang P: Analysis of intestinal microbial communities of cerebral infarction and ischemia patients based on high throughput sequencing technology and glucose and lipid metabolism. Mol Med Rep. 16:5413–5417. 2017. View Article : Google Scholar : PubMed/NCBI

94 

Henry N, Frank J, McLouth C, Trout AL, Morris A, Chen J, Stowe AM, Fraser JF and Pennypacker K: Short chain fatty acids taken at time of thrombectomy in acute ischemic stroke patients are independent of stroke severity but associated with inflammatory markers and worse symptoms at discharge. Front Immunol. 12:7973022021. View Article : Google Scholar

95 

Jiang W, Gong L, Liu F, Ren Y and Mu J: Alteration of gut microbiome and correlated lipid metabolism in Post-stroke depression. Front Cell Infect Microbiol. 11:6639672021. View Article : Google Scholar : PubMed/NCBI

96 

Huang C, Li Y, Feng X, Li D, Li X, Ouyang Q, Dai W, Wu G, Zhou Q, Wang P, et al: Distinct gut microbiota composition and functional category in children with cerebral palsy and epilepsy. Front Pediatr. 7:3942019. View Article : Google Scholar : PubMed/NCBI

97 

Lindefeldt M, Eng A, Darban H, Bjerkner A, Zetterström CK, Allander T, Andersson B, Borenstein E, Dahlin M and Prast-Nielsen S: The ketogenic diet influences taxonomic and functional composition of the gut microbiota in children with severe epilepsy. NPJ Biofilms Microbiomes. 5:52019. View Article : Google Scholar : PubMed/NCBI

98 

He Z, Cui BT, Zhang T, Li P, Long CY, Ji GZ and Zhang FM: Fecal microbiota transplantation cured epilepsy in a case with Crohn's disease: The first report. World J Gastroenterol. 23:3565–3568. 2017. View Article : Google Scholar : PubMed/NCBI

99 

Avorio F, Cerulli Irelli E, Morano A, Fanella M, Orlando B, Albini M, Basili LM, Ruffolo G, Fattouch J, Manfredi M, et al: Functional gastrointestinal disorders in patients with epilepsy: Reciprocal influence and impact on seizure occurrence. Front Neurol. 12:7051262021. View Article : Google Scholar : PubMed/NCBI

100 

Citraro R, Lembo F, De Caro C, Tallarico M, Coretti L, Iannone LF, Leo A, Palumbo D, Cuomo M, Buommino E, et al: First evidence of altered microbiota and intestinal damage and their link to absence epilepsy in a genetic animal model, the WAG/Rij rat. Epilepsia. 62:529–541. 2021. View Article : Google Scholar : PubMed/NCBI

101 

Gallucci A, Patel DC, Thai K, Trinh J, Gude R, Shukla D and Campbell SL: Gut metabolite S-equol ameliorates hyperexcitability in entorhinal cortex neurons following Theiler murine encephalomyelitis virus-induced acute seizures. Epilepsia. 62:1829–1841. 2021. View Article : Google Scholar : PubMed/NCBI

102 

Antón M, Rodríguez-González A, Ballesta A, González N, Del Pozo A, de Fonseca FR, Gómez-Lus ML, Leza JC, García-Bueno B, Caso JR and Orio L: Alcohol binge disrupts the rat intestinal barrier: The partial protective role of oleoylethanolamide. Br J Pharmacol. 175:4464–4479. 2018. View Article : Google Scholar : PubMed/NCBI

103 

Hoyles L, Snelling T, Umlai UK, Nicholson JK, Carding SR, Glen RC and McArthur S: Microbiome-host systems interactions: Protective effects of propionate upon the blood-brain barrier. Microbiome. 6:552018. View Article : Google Scholar : PubMed/NCBI

104 

Ceccarelli G, Brenchley JM, Cavallari EN, Scheri GC, Fratino M, Pinacchio C, Schietroma I, Fard SN, Scagnolari C, Mezzaroma I, et al: Impact of High-dose multi-strain probiotic supplementation on neurocognitive performance and central nervous system immune activation of HIV-1 infected individuals. Nutrients. 9:12692017. View Article : Google Scholar : PubMed/NCBI

105 

Rincel M, Aubert P, Chevalier J, Grohard PA, Basso L, Monchaux de Oliveira C, Helbling JC, Lévy É, Chevalier G, Leboyer M, et al: Multi-hit early life adversity affects gut microbiota, brain and behavior in a sex-dependent manner. Brain Behav Immun. 80:179–192. 2019. View Article : Google Scholar : PubMed/NCBI

106 

Cuskelly A, Hoedt EC, Harms L, Talley NJ, Tadros MA, Keely S and Hodgson DM: Neonatal immune challenge influences the microbiota and behaviour in a sexually dimorphic manner. Brain Behav Immun. 103:232–242. 2022. View Article : Google Scholar : PubMed/NCBI

107 

Saunders JM, Moreno JL, Ibi D, Sikaroodi M, Kang DJ, Muñoz-Moreno R, Dalmet SS, García-Sastre A, Gillevet PM, Dozmorov MG, et al: Gut microbiota manipulation during the prepubertal period shapes behavioral abnormalities in a mouse neurodevelopmental disorder model. Sci Rep. 10:46972020. View Article : Google Scholar : PubMed/NCBI

108 

Duan H, Wang L, Huangfu M and Li H: The impact of microbiota-derived short-chain fatty acids on macrophage activities in disease: Mechanisms and therapeutic potentials. Biomed Pharmacother. 165:1152762023. View Article : Google Scholar : PubMed/NCBI

109 

Chauhan V, Chauhan NK, Dutta S, Pathak D and Nongthomba U: Comparative In-silico analysis of microbial dysbiosis discern potential metabolic link in neurodegenerative diseases. Front Neurosci. 17:11534222023. View Article : Google Scholar : PubMed/NCBI

110 

Zancan V, Nasello M, Bigi R, Reniè R, Buscarinu MC, Mechelli R, Ristori G, Salvetti M and Bellucci G: Gut microbiota composition is causally linked to multiple sclerosis: A mendelian randomization analysis. Microorganisms. 12:14762024. View Article : Google Scholar : PubMed/NCBI

111 

Emery DC, Davies M, Cerajewska TL, Taylor J, Hazell M, Paterson A, Allen-Birt SJ and West NX: High resolution 16S rRNA gene next generation sequencing study of brain areas associated with Alzheimer's and Parkinson's disease. Front Aging Neurosci. 14:10262602022. View Article : Google Scholar : PubMed/NCBI

112 

Boles JS, Krueger ME, Jernigan JE, Cole CL, Neighbarger NK, Uriarte Huarte O and Tansey MG: A leaky gut dysregulates gene networks in the brain associated with immune activation, oxidative stress, and myelination in a mouse model of colitis. Brain Behav Immun. 117:473–492. 2024. View Article : Google Scholar : PubMed/NCBI

113 

Luck B, Horvath TD, Engevik KA, Ruan W, Haidacher SJ, Hoch KM, Oezguen N, Spinler JK, Haag AM, Versalovic J and Engevik MA: Neurotransmitter profiles are altered in the gut and brain of mice Mono-associated with Bifidobacterium dentium. Biomolecules. 11:10912021. View Article : Google Scholar : PubMed/NCBI

114 

Pandey S, Singh A, Chaudhari N, Nampoothiri LP and Kumar GN: Protection against 1,2-di-methylhydrazine-induced systemic oxidative stress and altered brain neurotransmitter status by probiotic Escherichia coli CFR 16 secreting pyrroloquinoline quinone. Curr Microbiol. 70:690–697. 2015. View Article : Google Scholar : PubMed/NCBI

115 

Zhu Y, Huan F and Wang J, Xie X, Yu G, Wang X, Jiang L, Gao R, Xiao H, Ding H and Wang J: 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine Induced Parkinson's disease in mouse: Potential association between neurotransmitter disturbance and gut microbiota dysbiosis. ACS Chem Neurosci. 11:3366–3376. 2020. View Article : Google Scholar : PubMed/NCBI

116 

Li W, Xu M, Liu Y, Zhang S, Wang J, Zhang Z, Xiao G, Wang R, Zhang J and Xue H: Lactiplantibacillus plantarum GOLDGUT-HNU082 alleviates CUMS-Induced Depressive-like behaviors in mice by modulating the gut microbiota and neurotransmitter levels. Foods. 14:8132025. View Article : Google Scholar : PubMed/NCBI

117 

Guo Y, Zhu X, Zeng M, Qi L, Tang X, Wang D, Zhang M, Xie Y, Li H, Yang X and Chen D: A diet high in sugar and fat influences neurotransmitter metabolism and then affects brain function by altering the gut microbiota. Transl Psychiatry. 11:3282021. View Article : Google Scholar : PubMed/NCBI

118 

Zhang Z, Peng Q, Huo D, Jiang S, Ma C, Chang H, Chen K, Li C, Pan Y and Zhang J: Melatonin regulates the neurotransmitter secretion disorder induced by caffeine through the Microbiota-Gut-brain axis in zebrafish (Danio rerio). Front Cell Dev Biol. 9:6781902021. View Article : Google Scholar : PubMed/NCBI

119 

Wang W, Chen F, Zhang L, Wen F, Yu Q, Li P and Zhang A: Neurotransmitter disturbances caused by methylmercury exposure: Microbiota-gut-brain interaction. Sci Total Environ. 873:1623582023. View Article : Google Scholar : PubMed/NCBI

120 

Mortensen NP, Moreno Caffaro M, Patel PR, Snyder RW, Watson SL, Aravamudhan S, Montgomery SA, Lefever T, Sumner SJ and Fennell TR: Biodistribution, cardiac and neurobehavioral assessments, and neurotransmitter quantification in juvenile rats following oral administration of aluminum oxide nanoparticles. J Appl Toxicol. 41:1316–1329. 2021. View Article : Google Scholar :

121 

Huang J, Liu S, Li P, Wei L, Lin G, Lin J, Luo Y, Liu Y, Mao Y, Ruan H, et al: Multi-omics analysis of gut-brain axis reveals novel microbial and neurotransmitter signatures in patients with arteriosclerotic cerebral small vessel disease. Pharmacol Res. 208:1073852024. View Article : Google Scholar : PubMed/NCBI

122 

Margetts G, Kleidonas S, Zaibi NS, Zaibi MS and Edwards KD: Evidence for anti-inflammatory effects and modulation of neurotransmitter metabolism by Salvia officinalis L. BMC Complement Med Ther. 22:1312022. View Article : Google Scholar : PubMed/NCBI

123 

Kumari M, Dasriya VL, Nataraj BH, Nagpal R and Behare PV: Lacticaseibacillus rhamnosus-derived exopolysaccharide Attenuates D-Galactose-Induced oxidative stress and inflammatory brain injury and modulates gut microbiota in a mouse model. Microorganisms. 10:20462022. View Article : Google Scholar : PubMed/NCBI

124 

Bock HJ, Lee NK and Paik HD: Neuroprotective effects of Heat-killed levilactobacillus brevis KU15152 on H2O2-induced oxidative stress. J Microbiol Biotechnol. 33:1189–1196. 2023. View Article : Google Scholar : PubMed/NCBI

125 

Cansız D, Ünal İ, Üstündağ ÜV, Alturfan AA, Altinoz MA, Elmacı İ and Emekli-Alturfan E: Caprylic acid ameliorates rotenone induced inflammation and oxidative stress in the gut-brain axis in Zebrafish. Mol Biol Rep. 48:5259–5273. 2021. View Article : Google Scholar

126 

Pan S, Wei H, Yuan S, Kong Y, Yang H and Zhang Y, Cui X, Chen W, Liu J and Zhang Y: Probiotic Pediococcus pentosaceus ameliorates MPTP-induced oxidative stress via regulating the gut microbiota-gut-brain axis. Front Cell Infect Microbiol. 12:10228792022. View Article : Google Scholar : PubMed/NCBI

127 

Dong B, Qi Y, Sundas H, Yang R, Zhou J and Li Z: Soy protein increases cognitive level in mice by modifying hippocampal nerve growth, oxidative stress, and intestinal microbiota. J Sci Food Agric. 103:4085–4094. 2023. View Article : Google Scholar

128 

Nikrad N, Farhangi MA, Fard Tabrizi FP, Vaezi M, Mahmoudpour A and Mesgari-Abbasi M: The effect of calorie-restriction along with thylakoid membranes of spinach on the gut-brain Axis Pathway and oxidative stress biomarkers in obese women with polycystic ovary syndrome: A randomized, Double-blind, placebo-controlled clinical trial. J Ovarian Res. 16:2162023. View Article : Google Scholar : PubMed/NCBI

129 

Rode J, Yang L, König J, Hutchinson AN, Wall R, Venizelos N, Brummer RJ, Rangel I and Vumma R: Butyrate rescues oxidative Stress-induced transport deficits of tryptophan: Potential implication in affective or Gut-Brain axis disorders. Neuropsychobiology. 80:253–263. 2021. View Article : Google Scholar

130 

He J, Xu P, Xu T, Yu H, Wang L, Chen R, Zhang K, Yao Y, Xie Y, Yang Q, et al: Therapeutic potential of hydrogen-rich water in zebrafish model of Alzheimer's disease: Targeting oxidative stress, inflammation, and the gut-brain axis. Front Aging Neurosci. 16:15150922024. View Article : Google Scholar

131 

Di Chiano M, Rocchetti MT, Spano G, Russo P, Allegretta C, Milior G, Gadaleta RM, Sallustio F, Pontrelli P, Gesualdo L, et al: Lactobacilli Cell-free supernatants modulate inflammation and oxidative stress in human microglia via NRF2-SOD1 signaling. Cell Mol Neurobiol. 44:602024. View Article : Google Scholar : PubMed/NCBI

132 

Hsu YC, Huang YY, Tsai SY, Kuo YW, Lin JH, Ho HH, Chen JF, Hsia KC and Sun Y: Efficacy of probiotic supplements on Brain-derived neurotrophic factor, inflammatory biomarkers, oxidative stress and cognitive function in patients with Alzheimer's dementia: A 12-week randomized, double-blind active-controlled study. Nutrients. 16:162023. View Article : Google Scholar

133 

Akhgarjand C, Vahabi Z, Shab-Bidar S, Anoushirvani A and Djafarian K: The effects of probiotic supplements on oxidative stress and inflammation in subjects with mild and moderate Alzheimer's disease: A randomized, double-blind, placebo-controlled study. Inflammopharmacology. 32:1413–1420. 2024. View Article : Google Scholar : PubMed/NCBI

134 

García-Sanmartín J, Bobadilla M, Mirpuri E, Grifoll V, Pérez-Clavijo M and Martínez A: Agaricus Mushroom-enriched diets modulate the Microbiota-Gut-brain axis and reduce brain oxidative stress in mice. Antioxidants (Basel). 11:6952022. View Article : Google Scholar : PubMed/NCBI

135 

Chenghan M, Wanxin L, Bangcheng Z, Yao H, Qinxi L, Ting Z, Xiaojie L, Kun Z, Yingqian Z and Zhihui Z: Short-chain fatty acids mediate gut microbiota-brain communication and protect the blood-brain barrier integrity. Ann N Y Acad Sci. 1545:116–131. 2025. View Article : Google Scholar : PubMed/NCBI

136 

Stachulski AV, Knausenberger TB, Shah SN, Hoyles L and McArthur S: A host-gut microbial amino acid co-metabolite, p-cresol glucuronide, promotes blood-brain barrier integrity in vivo. Tissue Barriers. 11:20731752023. View Article : Google Scholar

137 

Connell E, Le Gall G, McArthur S, Lang L, Breeze B, Pontifex MG, Sami S, Pourtau L, Gaudout D, Müller M and Vauzour D: (Poly)phenol-rich grape and blueberry extract prevents LPS-induced disruption of the blood-brain barrier through the modulation of the gut microbiota-derived uremic toxins. Neurochem Int. 180:1058782024. View Article : Google Scholar : PubMed/NCBI

138 

He J, Liu Y, Li J, Zhao Y, Jiang H, Luo S and He G: Intestinal changes in permeability, tight junction and mucin synthesis in a mouse model of Alzheimer's disease. Int J Mol Med. 52:1132023. View Article : Google Scholar : PubMed/NCBI

139 

Fiorentino M, Sapone A, Senger S, Camhi SS, Kadzielski SM, Buie TM, Kelly DL, Cascella N and Fasano A: Blood-brain barrier and intestinal epithelial barrier alterations in autism spectrum disorders. Mol Autism. 7:492016. View Article : Google Scholar : PubMed/NCBI

140 

Sun N, Hu H, Wang F, Li L, Zhu W, Shen Y, Xiu J and Xu Q: Antibiotic-induced microbiome depletion in adult mice disrupts blood-brain barrier and facilitates brain infiltration of monocytes after bone-marrow transplantation. Brain Behav Immun. 92:102–114. 2021. View Article : Google Scholar

141 

Nelson JW, Phillips SC, Ganesh BP, Petrosino JF, Durgan DJ and Bryan RM: The gut microbiome contributes to blood-brain barrier disruption in spontaneously hypertensive stroke prone rats. FASEB J. 35:e212012021. View Article : Google Scholar : PubMed/NCBI

142 

Knox EG, Aburto MR, Tessier C, Nagpal J, Clarke G, O'Driscoll CM and Cryan JF: Microbial-derived metabolites induce actin cytoskeletal rearrangement and protect Blood-brain barrier function. iScience. 25:1056482022. View Article : Google Scholar : PubMed/NCBI

143 

Wang Y, Tan Q, Pan M, Yu J, Wu S, Tu W, Li M and Jiang S: Minimally invasive vagus nerve stimulation modulates mast cell degranulation via the microbiota-gut-brain axis to ameliorate blood-brain barrier and intestinal barrier damage following ischemic stroke. Int Immunopharmacol. 132:1120302024. View Article : Google Scholar : PubMed/NCBI

144 

Marungruang N, Arévalo Sureda E, Lefrançoise A, Weström B, Nyman M, Prykhodko O and Fåk Hållenius F: Impact of dietary induced precocious gut maturation on cecal microbiota and its relation to the blood-brain barrier during the postnatal period in rats. Neurogastroenterol Motil. 30:e132852018. View Article : Google Scholar : PubMed/NCBI

145 

Schalla MA, Oerter S, Cubukova A, Metzger M, Appelt-Menzel A and Stengel A: Locked out: Phoenixin-14 does not cross a Stem-Cell-derived blood-brain barrier model. Brain Sci. 13:9802023. View Article : Google Scholar : PubMed/NCBI

146 

Solch RJ, Aigbogun JO, Voyiadjis AG, Talkington GM, Darensbourg RM, O'Connell S, Pickett KM, Perez SR and Maraganore DM: Mediterranean diet adherence, gut microbiota, and Alzheimer's or Parkinson's disease risk: A systematic review. J Neurol Sci. 434:1201662022. View Article : Google Scholar : PubMed/NCBI

147 

Lussier DM, Woolf EC, Johnson JL, Brooks KS, Blattman JN and Scheck AC: Enhanced immunity in a mouse model of malignant glioma is mediated by a therapeutic ketogenic diet. BMC Cancer. 16:3102016. View Article : Google Scholar : PubMed/NCBI

148 

Berding K, Vlckova K, Marx W, Schellekens H, Stanton C, Clarke G, Jacka F, Dinan TG and Cryan JF: Diet and the Microbiota-Gut-Brain axis: Sowing the seeds of good mental health. Adv Nutr. 12:1239–1285. 2021. View Article : Google Scholar : PubMed/NCBI

149 

Zuo Z, Fan H, Tang XD, Chen YM, Xun LT, Li Y, Song ZJ and Zhai HQ: Effect of different treatments and alcohol addiction on gut microbiota in minimal hepatic encephalopathy patients. Exp Ther Med. 14:4887–4895. 2017.PubMed/NCBI

150 

Bravo JA, Forsythe P, Chew MV, Escaravage E, Savignac HM, Dinan TG, Bienenstock J and Cryan JF: Ingestion of Lactobacillus strain regulates emotional behavior and central GABA receptor expression in a mouse via the vagus nerve. Proc Natl Acad Sci USA. 108:16050–16055. 2011. View Article : Google Scholar : PubMed/NCBI

151 

Chong HX, Yusoff NAA, Hor YY, Lew LC, Jaafar MH, Choi SB, Yusoff MSB, Wahid N, Abdullah MFIL, Zakaria N, et al: Lactobacillus plantarum DR7 alleviates stress and anxiety in adults: A randomised, double-blind, placebo-controlled study. Benef Microbes. 10:355–373. 2019. View Article : Google Scholar : PubMed/NCBI

152 

Lew LC, Hor YY, Yusoff NAA, Choi SB, Yusoff MSB, Roslan NS, Ahmad A, Mohammad JAM, Abdullah MFIL, Zakaria N, et al: Probiotic Lactobacillus plantarum P8 alleviated stress and anxiety while enhancing memory and cognition in stressed adults: A randomised, double-blind, placebo-controlled study. Clin Nutr. 38:2053–2064. 2019. View Article : Google Scholar

153 

Karakula-Juchnowicz H, Rog J, Juchnowicz D, Łoniewski I, Skonieczna-Żydecka K, Krukow P, Futyma-Jedrzejewska M and Kaczmarczyk M: The study evaluating the effect of probiotic supplementation on the mental status inflammation, and intestinal barrier in major depressive disorder patients using gluten-free or gluten-containing diet (SANGUT study): A 12-week, randomized, double-blind, and placebo-controlled clinical study protocol. Nutr J. 18:502019. View Article : Google Scholar

154 

Cheng Y, Tan G, Zhu Q, Wang C, Ruan G, Ying S, Qie J, Hu X, Xiao Z, Xu F, et al: Efficacy of fecal microbiota transplantation in patients with Parkinson's disease: Clinical trial results from a randomized, placebo-controlled design. Gut Microbes. 15:22842472023. View Article : Google Scholar : PubMed/NCBI

155 

Dalile B, Vervliet B, Bergonzelli G, Verbeke K and Van Oudenhove L: Colon-delivered short-chain fatty acids attenuate the cortisol response to psychosocial stress in healthy men: A randomized, placebo-controlled trial. Neuropsychopharmacology. 45:2257–2266. 2020. View Article : Google Scholar : PubMed/NCBI

156 

Lei Y, Tang L, Chen Q, Wu L, He W, Tu D, Wang S, Chen Y, Liu S and Xie Z: Disulfiram ameliorates nonalcoholic steatohepatitis by modulating the gut microbiota and bile acid metabolism. Nat Commun. 13:68622022. View Article : Google Scholar : PubMed/NCBI

157 

Fleck AK, Hucke S, Teipel F, Eschborn M, Janoschka C, Liebmann M, Wami H, Korn L, Pickert G, Hartwig M, et al: Dietary conjugated linoleic acid links reduced intestinal inflammation to amelioration of CNS autoimmunity. Brain. 144:1152–1166. 2021. View Article : Google Scholar : PubMed/NCBI

158 

Martin FP, Cominetti O, Berger B, Combremont S, Marquis J, Xie G, Jia W, Pinto-Sanchez MI, Bercik P and Bergonzelli G: Metabolome-associated psychological comorbidities improvement in irritable bowel syndrome patients receiving a probiotic. Gut Microbes. 16:23477152024. View Article : Google Scholar : PubMed/NCBI

159 

Mysonhimer AR, Cannavale CN, Bailey MA, Khan NA and Holscher HD: Prebiotic consumption alters microbiota but not biological markers of stress and inflammation or mental health symptoms in healthy adults: A randomized, controlled, cross-over trial. J Nutr. 153:1283–1296. 2023. View Article : Google Scholar : PubMed/NCBI

160 

Li J, Li Y, Zhao J, Li L, Wang Y, Chen F, Li Y, Cheng R, He F, Ze X and Shen X: Effects of Bifidobacterium breve 207-1 on regulating lifestyle behaviors and mental wellness in healthy adults based on the microbiome-gut-brain axis: A randomized, double-blind, placebo-controlled trial. Eur J Nutr. 63:2567–2585. 2024. View Article : Google Scholar : PubMed/NCBI

161 

Casertano M, Dekker M, Valentino V, De Filippis F, Fogliano V and Ercolini D: Gaba-producing lactobacilli boost cognitive reactivity to negative mood without improving cognitive performance: A human Double-Blind Placebo-Controlled Cross-Over study. Brain Behav Immun. 122:256–265. 2024. View Article : Google Scholar : PubMed/NCBI

162 

Caradonna E, Nemni R, Bifone A, Gandolfo P, Costantino L, Giordano L, Mormone E, Macula A, Cuomo M, Difruscolo R, et al: The Brain-gut axis, an important player in alzheimer and parkinson disease: A narrative review. J Clin Med. 13:41302024. View Article : Google Scholar : PubMed/NCBI

163 

Almheiri RT, Hajjar B, Alkhaaldi SMI, Rabeh N, Aljoudi S, Abd-Elrahman KS and Hamdan H: Beyond weight loss: Exploring the neurological ramifications of altered gut microbiota post-bariatric surgery. J Transl Med. 23:2232025. View Article : Google Scholar : PubMed/NCBI

164 

Park KJ and Gao Y: Gut-brain axis and neurodegeneration: Mechanisms and therapeutic potentials. Front Neurosci. 18:14813902024. View Article : Google Scholar : PubMed/NCBI

165 

Manfredi JN, Gupta SK, Vyavahare S, Deak F, Lu X, Buddha L, Wankhade U, Lohakare J, Isales C and Fulzele S: Gut microbiota dysbiosis in Alzheimer's disease (AD): Insights from human clinical studies and the mouse AD models. Physiol Behav. 290:1147782025. View Article : Google Scholar

166 

Qi A, Liu L, Zhang J, Chen S, Xu S, Chen Y, Zhang L and Cai C: Plasma metabolic analysis reveals the dysregulation of Short-chain fatty acid metabolism in Parkinson's disease. Mol Neurobiol. 60:2619–2631. 2023. View Article : Google Scholar : PubMed/NCBI

167 

Nakhal MM, Yassin LK, Alyaqoubi R, Saeed S, Alderei A, Alhammadi A, Alshehhi M, Almehairbi A, Al Houqani S, BaniYas S, et al: The Microbiota-Gut-Brain axis and neurological disorders: A comprehensive review. Life. 14:12342024. View Article : Google Scholar : PubMed/NCBI

168 

Vaz M and Silvestre S: Alzheimer's disease: Recent treatment strategies. Eur J Pharmacol. 887:1735542020. View Article : Google Scholar : PubMed/NCBI

169 

Forssten SD, Ouwehand AC, Griffin SM and Patterson E: One giant leap from mouse to man: The Microbiota-Gut-Brain axis in mood disorders and translational challenges moving towards human clinical trials. Nutrients. 14:5682022. View Article : Google Scholar : PubMed/NCBI

170 

Schächtle MA and Rosshart SP: The Microbiota-Gut-Brain axis in health and disease and its implications for translational research. Front Cell Neurosci. 15:6981722021. View Article : Google Scholar : PubMed/NCBI

171 

Ericsson AC and Franklin CL: Manipulating the gut microbiota: Methods and challenges. ILAR J. 56:205–217. 2015. View Article : Google Scholar : PubMed/NCBI

172 

McGonigle P and Ruggeri B: Animal models of human disease: Challenges in enabling translation. Biochem Pharmacol. 87:162–171. 2014. View Article : Google Scholar

173 

Woodworth MH, Sitchenko KL, Carpentieri C, Friedman-Moraco RJ, Wang T and Kraft CS: Ethical considerations in microbial therapeutic clinical trials. New Bioeth. 23:210–218. 2017. View Article : Google Scholar : PubMed/NCBI

174 

Cheng Y, Tan G, Zhu Q, Wang C, Ruan G, Ying S, Qie J, Hu X, Xiao Z, Xu F, et al: Efficacy of fecal microbiota transplantation in patients with Parkinson's disease: Clinical trial results from a randomized, placebo-controlled design. Gut Microbes. 15:22842472023. View Article : Google Scholar : PubMed/NCBI

175 

Blivet G, Relano-Gines A, Wachtel M and Touchon J: A randomized, double-blind, and sham-controlled trial of an innovative brain-gut photobiomodulation therapy: Safety and patient compliance. J Alzheimers Dis. 90:811–822. 2022. View Article : Google Scholar : PubMed/NCBI

176 

Rode J, Edebol Carlman HMT, König J, Hutchinson AN, Thunberg P, Persson J and Brummer RJ: Multi-strain probiotic mixture affects brain morphology and resting state brain function in healthy subjects: An RCT. Cells. 11:29222022. View Article : Google Scholar : PubMed/NCBI

177 

Kilinçarslan S and Evrensel A: The effect of fecal microbiota transplantation on psychiatric symptoms among patients with inflammatory bowel disease: An experimental study. Actas Esp Psiquiatr. 48:1–7. 2020.PubMed/NCBI

178 

Laeeq T, Vongsavath T, Tun KM and Hong AS: The potential role of fecal microbiota transplant in the reversal or stabilization of multiple sclerosis symptoms: A literature review on efficacy and safety. Microorganisms. 11:28402023. View Article : Google Scholar : PubMed/NCBI

179 

Loh JS, Mak WQ, Tan LKS, Ng CX, Chan HH, Yeow SH, Foo JB, Ong YS, How CW and Khaw KY: Microbiota-gut-brain axis and its therapeutic applications in neurodegenerative diseases. Signal Transduct Target Ther. 9:372024. View Article : Google Scholar : PubMed/NCBI

180 

Zmora N, Zilberman-Schapira G, Suez J, Mor U, Dori-Bachash M, Bashiardes S, Kotler E, Zur M, Regev-Lehavi D, Brik RB, et al: Personalized gut mucosal colonization resistance to empiric probiotics is associated with unique host and microbiome features. Cell. 174:1388–1405.e21. 2018. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2025
Volume 56 Issue 5

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang H, Luan J, He L, Pan X, Zhang H, Li Y and Li H: Role of the gut-brain axis in neurological diseases: Molecular connections and therapeutic implications (Review). Int J Mol Med 56: 192, 2025.
APA
Zhang, H., Luan, J., He, L., Pan, X., Zhang, H., Li, Y., & Li, H. (2025). Role of the gut-brain axis in neurological diseases: Molecular connections and therapeutic implications (Review). International Journal of Molecular Medicine, 56, 192. https://doi.org/10.3892/ijmm.2025.5633
MLA
Zhang, H., Luan, J., He, L., Pan, X., Zhang, H., Li, Y., Li, H."Role of the gut-brain axis in neurological diseases: Molecular connections and therapeutic implications (Review)". International Journal of Molecular Medicine 56.5 (2025): 192.
Chicago
Zhang, H., Luan, J., He, L., Pan, X., Zhang, H., Li, Y., Li, H."Role of the gut-brain axis in neurological diseases: Molecular connections and therapeutic implications (Review)". International Journal of Molecular Medicine 56, no. 5 (2025): 192. https://doi.org/10.3892/ijmm.2025.5633