An Updated and Comprehensive Review Exploring the Gut–Brain Axis in Neurodegenerative Disorders and Neurotraumas: Implications for Therapeutic Strategies
Abstract
1. Introduction
2. Search Strategy
3. Molecular Mechanisms of GBA in Neuroinflammatory-Based Disorders
4. NDDs
4.1. Pathophysiology of AD
Therapeutic Strategies Focusing on the GBA in AD
Intervention | Key Findings—Related Targets and Pathways | Model/Limitations | Effect on AD | Ref. |
---|---|---|---|---|
Compared germ-free mice to normal mice; tested effects of gut microbiota colonization and SCFA treatment on BBB permeability | Germ-free mice had leaky BBB with reduced tight junction proteins (occludin, claudin-5); gut microbiota and SCFAs restored BBB integrity | Mouse model/Short time intervention, Species differences between human and mouse model | Gut microbiota helps maintain BBB integrity; disruption may contribute to AD progression through increased brain vulnerability | [42] |
Aanalyzing gut microbiota composition and blood cytokine levels in AD patients | AD patients exhibited a decreased Firmicutes/Bacteroidetes ratio, indicating gut dysbiosis. Elevated levels of proinflammatory cytokines, including IL-1β, IL-6, IL-8, and TNF-α, were detected in the blood of AD patients | Human subjects/Sample size, Does not prove the causality between dysbiosis and AD | Gut dysbiosis may contribute to systemic inflammation, which could exacerbate neuroinflammatory processes involved in AD pathogenesis | [44] |
Gut microbial SoLs | SoLs bind to TLR4 and inhibit LPS binding. This action suppresses TLR4-mediated NF-κB signaling, leading to reduced production of pro-inflammatory cytokines (e.g., TNF-α, IL-6) and inhibition of macrophage M1 polarization and as a result neuroinflammation | Mouse model/Translational Gap, Complexity of Gut Microbiota, Limited Scope on Long-term Effects | Given the role of TLR4-mediated neuroinflammation in AD pathogenesis, SoLs’ ability to modulate TLR4 signaling suggests a potential protective effect against AD by reducing neuroinflammatory responses | [48] |
Gut microbiota modulation and intervention with Huanglian Jiedu Decoction (HLJDD) | Altered SCFA and tryptophan-kynurenine metabolism; reduced pro-inflammatory cytokines and immune imbalance via gut–brain axis | Mouse Model/Limited Behavioral Analysis, short duration, Translation to Humans | Reduced neuroinflammation, decreased Aβ plaques, and improved cognitive function in AD model mice | [49] |
Measurement of fecal calprotectin levels (a marker of intestinal inflammation) | Calprotectin levels increase with age, which was associated with more brain amyloid burden in AD patients | Human Cohort Study/The sample size, Biomarker Limitations, Potential confounders, No longitudinal data | Intestinal inflammation may contribute to amyloid accumulation and Alzheimer’s progression, but the study does not prove causality | [50,51,52] |
Anvestigating GALT in 5xFAD transgenic mice, a model for AD | IL-17 protein production was reduced in gut-associated tissues (Peyer’s Patches and Mesenteric Lymph Nodes) of 5xFAD mice, despite normal Th17 cell numbers and IL-17 mRNA levels. In addition to downregulation of miR-155 | 5xFAD transgenic mouse model/focused only on IL-17, limited scope of immune factors, No longitudinal behavioral or cognitive correlation. | Impaired IL-17 production may weaken gut immune defense, promote dysbiosis, and worsen systemic and brain inflammation, potentially accelerating AD progression. Enhancing IL-17 signaling could be a therapeutic target | [53,54] |
Analyzing the relation between APOE4 genotype, Aβ deposition, and episodic memory decline using data from the AD Research Initiative | APOE4 carriers exhibit a more rapid decline in episodic memory correlated with the duration of amyloid positivity, suggesting increased susceptibility to Aβ-related neurotoxicity | Human Cohort Study/Observational design, potential confounding factors, limit generalizability | The presence of the APOE4 allele accelerates cognitive decline in individuals with amyloid accumulation | [55,56] |
4.2. Pathophysiology of PD
Therapeutic Interventions Targeting GBA in PD
Intervention | Key Findings—Related Targets and Pathways | Model/Limitations | Effect on PD | Ref. |
---|---|---|---|---|
Effect of FMT from healthy donors into MPTP-induced PD mouse models. and the action of SCFAs through their receptors, FFAR2 and FFAR3, in modulating CNS functions | FMT increased gut levels of SCFAs in PD mice, upregulated SCFA receptors FFAR2 and FFAR3 in the CNS, enhanced dopamine and serotonin levels in the striatum, promoted the survival of dopaminergic neurons in the substantia nigra, and reduced glial cell activation | Mouse model/Most evidence is correlational not causality, limit generalizability, Species-specific microbiota differences, No human validation | FMT restored SCFA levels, improved dopaminergic neuron survival, reduced neuroinflammation, and increased dopamine and serotonin levels in the brain, leading to improved motor and neurological functions in PD mice | [66] |
Administration of α7nAChR agonists, GTS-21 and PNU-282987 to subacute MPTP-induced PD mouse models | Treatment reduced α-synuclein accumulation in the brain and colon, activated AMPK-mTOR–mediated autophagy, promoting autophagic clearance of α-synuclein, suppressed pro-inflammatory mediators such as iNOS, IL-6, and TNF-α, in both brain and gut tissues, enhanced dopaminergic signaling and motor function, and exerted effects via α7nAChR, as confirmed by MLA inhibition | Mouse Model/Short duration, Mechanism not fully clear, Limited behavioral data, Focus on α-synuclein only | Activation of α7nAChRs mitigated α-synuclein pathology, reduced neuroinflammation, and preserved dopaminergic neurons, leading to improved motor function in PD mouse models | [67] |
Prospective metabolome-wide association study (MWAS) analyzing pre-diagnostic plasma samples from the EPIC4PD cohort to identify microbial metabolites associated with future PD risk | Identified 13 microbial metabolites associated with PD risk, including amino acids, bile acids, indoles, and hydroxy acids, with Pathway analyses highlighting branched-chain amino acid degradation, such as valine, leucine, and isoleucine and SCFA metabolism, and stronger associations observed in men, smokers, and individuals with obesity | Human Cohort Study/Observational design only, limited population diversity, No causal inference | Alterations in gut microbial metabolites may serve as early biomarkers for PD risk, highlighting the potential role of the gut–brain axis in PD pathogenesis and offering avenues for early intervention strategies | [68] |
Utilization of a gut-originated PD mouse model to investigate the role of gut microbiota in the propagation of pathological α-syn from the gut to the brain | Gut microbiota promoted the vagus nerve-mediated spread of pathological α-synuclein from the gut to the brain, triggered neuroinflammation through microglial activation and cytokine release, and worsened motor deficits compared to germ-free mice | Mouse Model/No human validation, short duration of study, Microbiota complexity underestimated | The study demonstrates that gut microbiota plays a crucial role in promoting the propagation of α-syn pathology from the gut to the brain, thereby exacerbating neuroinflammation and motor deficits characteristic of PD | [69] |
A deep learning model used gut microbiota data and combines Combined Ranking using Random Forest Scores (CRFS) for feature selection, LSTM-penultimate to SVM Input Method (LSIM) for classification, and soft voting for prediction, based on data from 39 PD patients and their healthy spouses | PGPM demonstrated high predictive accuracy (85%) and AUC (0.92) in distinguishing PD patients from controls, with CRFS identifying key microbial features and the LSIM architecture enhancing classification through combined LSTM and SVM modeling | Computational Model with Human Microbiome Data/No experimental validation, Correlational model only, Potential overfitting risk, Lacks biological mechanisms | The study demonstrates that analyzing gut microbiota using advanced deep learning techniques can effectively predict Parkinson’s disease, highlighting the potential of gut microbiota as a non-invasive biomarker for early diagnosis | [70] |
Oral supplementation of Akkermansia muciniphila strain Akk11 in mice induced with PD using MPTP | Akk11 suppressed activation of the NLRP3 inflammasome in microglia, reducing neuroinflammation, preserved dopaminergic neurons in the substantia nigra, improved MPTP-induced motor deficits, and protected colonic integrity | Mouse Model/Animal Model limitation, Short Duration of Treatment, Limited Sample Size, Focus on Specific Outcomes | Akk11 supplementation mitigated neurodegeneration and motor dysfunction in PD models by modulating neuroinflammatory pathways and enhancing gut health. | [71] |
Oral administration of Lactiplantibacillus plantarum SG5 to mice induced with PD using MPTP | SG5 improved motor function and dopaminergic neuron survival in the substantia nigra, reduced α-synuclein levels, suppressed overactivation of microglia and astrocytes, restored BBB and gut barrier integrity, modulated gut microbiota, and activated the GLP-1/PGC-1α pathway effects, which was validated by pathway-specific inhibitors | Mouse Mode/Animal Model Limitations, short duration of treatment, Potential Confounding Factors | SG5 supplementation mitigated neurodegeneration and motor dysfunction in PD models by modulating neuroinflammatory pathways, enhancing gut health, and activating the GLP-1/PGC-1α signaling pathway | [72] |
Oral administration of a 91.23% brazilin-enriched extract from Caesalpinia sappan L. (SE) to MPTP/p-induced PD mice | SE ameliorated motor deficits and improved dopaminergic neuron survival, reduced oxidative stress and neuroinflammation in the brain, restored gut microbiota balance by increasing Firmicutes and decreasing Bacteroidetes, increased SCFA (especially butyric acid) levels, enhanced intestinal barrier integrity via upregulation of ZO-1 and occludin, and reduced LPS leakage and systemic inflammation | Mouse Model/Animal Model Limitations, short duration of treatment, lack of Control Comparisons | SE mitigated neurodegeneration and motor dysfunction in PD models by modulating the gut–brain axis, reducing neuroinflammation, and restoring intestinal barrier integrity | [73] |
Administration of 1-methyl-tryptophan (1-MT), an IDO-1 inhibitor, to MPTP-induced PD mice | IDO-1 inhibition improved motor behavior and dopaminergic neuron survival in the substantia nigra, reduced serum quinolinic acid and aryl hydrocarbon receptor levels in the striatum and colon, suppressed TLR4/NF-κB mediated neuroinflammation, leading to decreased iNOS and COX2 expression, promoted hippocampal neurogenesis indicated by increased DCX+ and SOX2+ cells, likely via activation of the BDNF/TrkB pathway, normalized SCFA levels, and required gut microbiota for its neuroprotective effects | Mouse Model/Animal Model Limitations, short duration of treatment, Translational relevance uncertain, Gut microbiota variability | IDO-1 inhibition mitigated neurodegeneration and motor dysfunction in PD models by modulating neuroinflammatory pathways, promoting neurogenesis, and regulating gut microbiota composition | [74] |
Oral administration of Enterococcus faecalis (EF) to MPTP-induced PD mice | EF treatment alleviated neurofunctional impairments and gastrointestinal disorders associated with PD in mice, reversed dysbiosis of PD-related microbial communities induced by MPTP, and exerted its neuroprotective effects via the vagus nerve, as shown by diminished benefits following vagotomy | Mouse Model/Animal Model Limitations, short duration of treatment, Mouse-specific neural pathways, Vagus mechanism uncertain | EF supplementation mitigated neurodegeneration and motor dysfunction in PD models by modulating neuroinflammatory pathways and enhancing gut health through vagus nerve-mediated mechanisms | [75] |
Administration of FLZ, a neuroprotective agent, to MPTP-induced PD mice | FLZ modulated gut microbiota, by downregulating Clostridium innocuum, increased glycoursodeoxycholic acid levels through enhanced bile salt hydrolase (BSH) enzymes activity, activated the Nrf2 antioxidant pathway, reduced neuroinflammation, protected dopaminergic neurons in the substantia nigra, and exerted its effects via the microbiota-GBA | Mouse Model/Animal Model Limitations, short duration of treatment, Single bacterial target | FLZ mitigated neurodegeneration and motor dysfunction in PD models by modulating gut microbiota composition, enhancing beneficial metabolites like GUDCA, and activating neuroprotective pathways | [76] |
Co-administration of Vinpocetine and Lactobacillus in rotenone-induced PD rat model | Treatment with Vinpocetine and Lactobacillus increased tyrosine hydroxylase (TH) expression to restore dopamine synthesis, reduced oxidative stress (lower MDA, higher GSH), suppressed pro-inflammatory cytokines (IL-1β, TNF-α) and nitrite levels, and decreased accumulation of α-synuclein and tau proteins | Mouse Model/Animal model limitations, short duration of treatment, Absence of Mechanistic Insights | Ameliorated dopaminergic neurodegeneration, improved motor function, reduced inflammation and oxidative stress, and inhibited Lewy body formation in PD models | [77] |
Engagement in an active lifestyle characterized by regular physical activity in individuals with PD | Physically active individuals showed increased SCFA production, reduced intestinal inflammation (lower fecal calprotectin), improved constipation symptoms, and potential beneficial modulation of the gut–brain axis in PD patients | preliminary Human Observational Study/Small Sample Size, Lack of Control for Physical Activity Types, No Evidence of Sex-Specific Differences, No microbiota analysis | The active lifestyle may alleviate gastrointestinal symptoms and reduce intestinal inflammation in PD patients, potentially contributing to improved overall health and quality of life | [78] |
An analysis of peripheral blood mononuclear cells (PBMCs) from individuals with PD or IBD, focusing on iron metabolism and the influence of NSAIDs | PBMCs from PD and IBD patients show impaired iron storage and transport, leading to mitochondrial dysfunction characterized by dysmorphic mitochondria, suggesting a link to ferroptosis, with shared inflammatory and metabolic signatures; NSAID uses may modulate these transcriptional abnormalities | Human Clinical Sample Analysis/Sample size and selection, Cross-sectional design, NSAID effects variable, No microbiota profiling | The study suggests that iron dysregulation and mitochondrial dysfunction in peripheral immune cells may contribute to PD pathogenesis. NSAID-mediated modulation of these pathways indicates potential therapeutic avenues for PD | [79] |
Administration of rotenone (a mitochondrial complex I inhibitor) to rats to model PD | Rotenone exposure disrupted metabolic homeostasis by altering peptide levels (decreased GLP-1, C-peptide, amylin; increased insulin, leptin, pancreatic polypeptide, peptide YY, gastric inhibitory polypeptide), induced systemic inflammation via TNF-α–expressing CD4+ T cells, and increased body weight, linking mitochondrial dysfunction to metabolic and immune disturbances | Rat Model/Species differences between rats and humans, Focus on short-term effects, No behavioral assessments | The study suggests that rotenone-induced mitochondrial dysfunction leads to metabolic peptide dysregulation and systemic inflammation, which may contribute to the pathogenesis of PD | [80] |
4.3. Etiopathogenesis of MS
Evaluating Gut–Brain-Based Treatments in MS
Intervention | Key Findings—Related Targets and Pathways | Model/Limitations | Effect on MS | Ref. |
---|---|---|---|---|
Measurement of fecal lipocalin-2 (Lcn-2) levels in patients with relapsing-remitting multiple sclerosis (RRMS) and in experimental autoimmune encephalomyelitis (EAE) mouse models | RRMS patients showed elevated fecal Lcn-2 levels, which correlated with reduced microbial diversity and increased intestinal inflammation, findings that were mirrored in EAE mouse models, supporting a link between gut dysbiosis and inflammation. | humanized transgenic mouse model/Small sample size, Heterogeneity of Microbiome, Focus on Specific Biomarkers, Short-term Observations, Limited clinical correlation | Fecal Lcn-2 serves as a sensitive, non-invasive biomarker for detecting gut dysbiosis and intestinal inflammation in MS. This highlights the role of gut health in MS pathogenesis and suggests potential for monitoring disease progression or response to therapies targeting the gut microbiome | [95] |
Implementation of a Mediterranean diet in patients with MS. | Adherence to the Mediterranean diet improved physical and mental health-related quality of life measures, reduced disability levels, and likely mitigated MS symptoms through its anti-inflammatory effects | Human Clinical Study/Small sample size, Single healthcare setting, No control group, Lack of biochemical parameters, Self-reported dietary data | The Mediterranean diet appears to positively influence disease outcomes in MS patients by improving quality of life and reducing disability, potentially through its anti-inflammatory effects | [100] |
Monthly FMT administered to patients with relapsing-remitting multiple sclerosis (RRMS) over a six-month period | FMT was safe and well-tolerated in RRMS patients, led to improved intestinal permeability in some cases, induced beneficial donor-specific shifts in gut microbiota, but showed no significant changes in Expanded Disability Status Scale scores or MRI outcomes | Human Randomized controlled trial (pilot RCT)/Small sample size, Lack of control group, Short-term follow-up, Limited efficacy data | The study suggests that FMT may improve gut-related parameters in MS patients, such as intestinal permeability and microbiota composition, without adverse effects. However, due to the small sample size and early termination of the study, further research is needed to assess its impact on neurological outcomes | [101,102] |
Administration of tryptamine, a naturally occurring monoamine derived from tryptophan, in mouse models of EAE, which is commonly used to study MS | Tryptamine activates the aryl hydrocarbon receptor (AHR), suppresses neuroinflammation and paralysis in EAE models via AHR-dependent pathways, tryptamine effect was absent in mice lacking AHR in T cells, and modulates gut microbiota to increase anti-inflammatory butyrate production | Murine model/Not directly translating to human data, Animal model constraints | The study suggests that tryptamine, through AHR activation, can modulate immune responses and gut microbiota composition, leading to reduced neuroinflammation and amelioration of MS-like symptoms in animal models. This points to the potential of targeting the AHR pathway as a therapeutic strategy in MS | [103] |
Treatment of RAW264.7 macrophage cells with microbe-derived antioxidants (MAs) to assess their effect on LPS-induced inflammation | Microbial antioxidants (MAs) activate the Nrf2 pathway, enhancing antioxidant enzyme expression and reducing ROS, while inhibiting NLRP3 inflammasome activation and associated cytokine release, with their anti-inflammatory effects dependent on Nrf2 signaling | In Vitro RAW264.7 cells/Cell line specificity, Lack of in vivo validation, Simplified inflammatory model, Translational relevance uncertain | MAs attenuate LPS-induced inflammatory responses by activating the Nrf2 pathway, which in turn inhibits the ROS/NLRP3/IL-1β signaling axis. This mechanism suggests potential therapeutic applications of MAs in managing inflammation-related diseases | [105,106] |
Investigation of microglial development and function in germ-free (GF) mice, mice with limited microbiota complexity, and mice recolonized with complex microbiota | Germ-free (GF) mice show impaired microglial maturation and function due to microbiota absence, with restoration partially achieved through complex microbiota or SCFA supplementation, highlighting SCFAs and FFAR2 signaling as key regulators of microglial homeostasis | Mouse Model/Mouse model constraints, Translational relevance uncertain, Microbiota-microglia mechanism unclear | The study demonstrates that host microbiota is vital for the proper maturation and function of microglia, suggesting that disruptions in gut microbiota could impact CNS immunity and potentially contribute to neurological disorders | [107] |
Administration of butyrate, a microbial fermentation product, to assess its effect on the differentiation of colonic Treg cells in mice | Butyrate promotes colonic Treg cell differentiation by enhancing histone H3 acetylation at the Foxp3 locus and ameliorates colitis in a CD4⁺ CD45RBhigh T cell transfer model in Rag1⁻/⁻ mice, highlighting its epigenetic and immunomodulatory effects | Mouse Model and in vitro cell assays/small sample sizeLimited cell types analyzed, Model limitations | The study demonstrates that butyrate, produced by commensal microbes, plays a crucial role in maintaining intestinal immune homeostasis by promoting the differentiation of colonic Treg cells, thereby suppressing inflammatory responses | [108,109] |
Investigation of gut microbiota-specific IgA⁺ B cells in CNS of individuals with active MS | IgA⁺ B cells specific to gut microbiota were found in the CNS during active MS, indicating that gut-derived immune responses can mediate CNS inflammation and may serve as biomarkers or therapeutic targets | Human Subjects and mouse model/Human sample variability, Cross-sectional design, small sample size and Diversity | The study indicates that gut microbiota-specific IgA⁺ B cells contribute to CNS inflammation in active MS, highlighting a novel gut–brain immune axis that may be pivotal in disease progression | [110] |
4.4. Pathophysiology of ALS
Targeting the GBA in ALS: Emerging Therapeutic Strategies
4.5. Overview of HD
Modulating the GBA in HD: Insights into Therapeutic Approaches
Intervention | Key Findings—Related Targets and Pathways | Model/Limitations | Effect on HD | Ref. |
---|---|---|---|---|
Analysis of gut microbiota composition in 42 HD gene expansion carriers (19 manifest and 23 premanifest) compared to 36 age- and gender-matched healthy controls | HD gene expansion carriers exhibited significantly reduced alpha diversity (species richness and evenness; p = 0.001) and distinct beta diversity (microbial community structure; p = 0.001) compared to controls. Taxonomic analyses revealed shifts in microbial phyla and families, alongside differences in functional pathways and enzyme profiles. Specific gut microbes were linked to cognitive performance and clinical outcomes, suggesting microbiota involvement in HD pathophysiology | Human cohort/Small sample size, Lack of longitudinal data, No functional or mechanistic insights | NOT AVAILABLE | [134] |
Utilization of 16S rRNA amplicon sequencing to characterize the gut microbiome in R6/1 transgenic HD mice compared to wild-type littermate controls at 12 weeks of age | HD mouse models showed gut dysbiosis characterized by increased Bacteroidetes, reduced Firmicutes, sex-specific microbial diversity changes, and physiological alterations, including impaired weight gain, motor deficits, and increased fecal water content | Transgenic mice model/Mouse model limitations, small Sample size, Limited scope of investigation, No mechanistic or metabolic follow-up | The observed gut dysbiosis in HD mice was associated with systemic physiological changes, including weight loss and motor deficits, indicating that alterations in the gut microbiome may contribute to the progression and severity of Huntington’s disease symptoms | [135] |
Administration of triheptanoin (1 g/kg/day) for 1 month to early-stage HD patients | Triheptanoin improved brain energy metabolism in HD patients by restoring Pi/PCr ratios during stimulation and supporting mitochondrial function through its anaplerotic action on the Krebs cycle | Human patients/Small patient sample size, Lack of placebo control group, Open-Label Design, Metabolic improvements not directly linked to clinical outcomes | Improved brain energy metabolism was associated with stabilization or improvement in motor function, suggesting potential therapeutic benefits in slowing HD progression | [136] |
A 6-week double-blind, placebo-controlled trial tested probiotics on 41 HD gene expansion carriers (HDGECs), consisting of 19 early manifest and 22 premanifest individuals, compared to 36 healthy controls | HDGECs exhibited altered gut microbiome diversity compared to healthy controls, indicating gut dysbiosis; however, probiotic treatment failed to improve microbial composition or clinical outcomes such as cognition, mood, and gastrointestinal symptoms | Human randomized controlled trial/No metabolomic or mechanistic data | The probiotic intervention did not produce measurable benefits in gut microbiota composition or clinical symptoms in HDGECs. However, the study underscores the potential of the gut as a therapeutic target in HD, warranting further exploration of alternative gut-targeted interventions | [138] |
Fecal microbiota transplantation (FMT) from wild-type mice into HD mice | FMT from wild-type mice into HD mice restored gut microbiota diversity and composition. It improved cognitive function, particularly in female HD mice, while male HD mice showed less efficient FMT engraftment, potentially due to greater gut microbial instability | Transgenic mice model/Small sample size limits generalizability, Mechanistic pathways linking microbiota changes to cognition unclear | The findings suggest that FMT can modulate gut microbiota and improve cognitive function in HD mice, highlighting the potential of gut–brain axis interventions in neurodegenerative diseases | [139] |
Genetic ablation of kynurenine 3-monooxygenase (KMO) in R6/2 mice | In R6/2 mice, significant increases in plasma cytokines (TNFα, IL1β, IL4, IL6, and IL10) were observed compared to wild-type controls, but KMO ablation normalized these elevated levels to near wild-type levels, while also increasing neuroprotective kynurenic acid (KYNA) and decreasing neurotoxic 3-hydroxykynurenine (3-HK) in the brain and periphery | Transgenic mice model/animal model limitations, focus on single enzyme limits broader pathway understanding | While KMO ablation led to normalization of inflammatory cytokine levels and favorable shifts in kynurenine pathway metabolites, it did not result in improvements in behavioral phenotypes or disease progression in R6/2 mice | [141] |
5. Neurotrauma
5.1. Pathophysiological Processes of TBI
GBA in TBI: Insights into Novel Therapies
Intervention | Key Findings—Related Targets and Pathways | Model/Limitations | Effect on TBI | Ref. |
---|---|---|---|---|
Administration of broad-spectrum antibiotics (vancomycin, neomycin-sulfate, ampicillin, and metronidazole) to induce gut microbial dysbiosis in mice before, during, and after TBI | Antibiotic-induced dysbiosis modulated the immune response after TBI by reducing Ly6Chigh monocyte and T cell infiltration into the brain, increasing microglial activation (TLR4, MHCII), impairing neurogenesis in the dentate gyrus, and leading to cognitive deficits evidenced by altered fear memory behavior | Mouse Model/Lack of control groups, Variability in results, Limited behavioral assessment | Induced dysbiosis post-TBI led to increased neuronal loss in the hippocampal CA3 region, persistent microglial activation driving chronic neuroinflammation, and long-term cognitive impairments, especially in fear memory performance | [154] |
Male C57BL/6J mice were administered a broad-spectrum antibiotic cocktail (ampicillin, gentamicin, metronidazole, and vancomycin) via oral gavage for 2 days to deplete gut microbiota, followed by traumatic brain injury (TBI) induction using CCI model | Antibiotic treatment led to significant shifts in gut microbiota diversity and abundance during both acute and chronic phases post-TBI, and to reduced neuroinflammation, marked by decreased microglial activation, resulting in improved neurological outcomes | Mouse model/Short duration of observation, Mouse model limits direct human translation, Functional impacts unclear | Antibiotic-induced microbiota depletion improved neurological recovery post-TBI, indicating a neuroprotective effect likely mediated through modulation of the brain–gut axis and attenuation of neuroinflammatory responses | [153] |
Administration of probiotics: Lactobacillus helveticus and Bifidobacterium longum | Probiotic treatment reversed TBI-induced reductions in hippocampal mRNA expression of CaMKII and CREB, improved glucose metabolism, altered gut microbiota composition, and partially restored disrupted hepatic lipid profiles, while having no significant effect on BDNF mRNA levels or neuroinflammatory markers | Mouse model/Pre-treatment design, not including both sexes in the experimental design, No motor/neurological functional analysis, Translational gap | Probiotic treatment in TBI mice improved memory performance, glucose tolerance, and neuroplasticity markers (CaMKII and CREB), but did not significantly reduce TBI-induced pro-inflammatory cytokine levels | [155] |
Administration of an 8% inulin-enriched diet to male mice, starting 3 months post-injury (mpi) and continuing for 8 weeks | Inulin supplementation enhanced gut microbiota by increasing beneficial bacteria and SCFAs, while also restoring cerebral blood flow in both hippocampi and left thalamus and improving white matter integrity, as indicated by MRI assessments | Mouse model/Mouse model limits direct human translation, Lack of motor and cognitive testing, small sample size, Clinical relevance requires human trials | The findings suggest that inulin supplementation during the chronic phase post-mild TBI can modulate the gut microbiota, leading to improved brain vascular and structural integrity, as well as enhanced cognitive function | [156] |
Administration of Omega-3 Polyunsaturated Fatty Acids (ω-3 PUFAs), specifically docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA), in a mouse model of TBI | ω-3 PUFAs reduced levels of pro-inflammatory cytokines (TNF-α, IL-1β, IL-6) and increased anti-inflammatory cytokine IL-10, shifted microglial activation from pro-inflammatory M1 phenotype to anti-inflammatory M2 phenotype, and inhibited the HMGB1/NF-κB signaling pathway through SIRT1-mediated deacetylation, reducing neuroinflammation, and suppressed necroptosis by downregulating RIP1 and RIP3 expression | Mouse model/lack of behavior tests, Translational gap to humans | The findings suggest that omega-3 PUFA supplementation can mitigate early brain injury following TBI by modulating inflammatory responses and inhibiting necroptotic pathways, thereby improving neurological outcomes | [157] |
Transplantation of gut microbiota from healthy donor rats into male rats with TBI induced by gas explosion | FMT restored microbial diversity and balance, correcting dysbiosis caused by TBI, enhanced expression of tight junction proteins (Claudin-1, Occludin, ZO-1) to improve intestinal barrier integrity, improved BBB function by reducing permeability and potential neuroinflammation, and increased levels of regulatory T cell (Treg)-related factors (IL-10, PD-1, FoxP3), indicating an anti-inflammatory response. | Rat model/Lack of functional analysis, Focus on male rats, Rat model may not fully replicate human TBI. | The findings suggest that FMT can mitigate cognitive impairments following TBI by restoring gut microbiota balance, enhancing barrier functions, and modulating immune responses, highlighting its potential as a therapeutic strategy for TBI-related cognitive deficits. | [159] |
Administration of fecal microbiota from healthy donor mice to TBI-induced mice. | FMT inhibited microglial activation and reduced pro-inflammatory cytokine TNF-α levels, increased expression of synaptic proteins PSD-95 and synapsin I, indicating improved synaptic integrity, and enhanced population of regulatory T cells (Tregs) in the spleen, suggesting modulation of systemic immune response | Mouse model/Mouse model limits human applicability, short intervention duration, the number of mice used doesn’t specified, No sex-specific reporting | The findings suggest that FMT can modulate the gut–brain axis to suppress neuroinflammation and promote neurological recovery following TBI | [160] |
Administration of fecal microbiota from healthy donor rats to male Sprague-Dawley rats subjected to CCI to induce TBI. FMT was performed for 7 consecutive days | TBI induced significant changes in gut microbiota diversity and composition; FMT restored these alterations, TBI increased levels of trimethylamine (TMA) in feces and trimethylamine N-oxide (TMAO) in the brain and serum; FMT reduced these levels, and TBI decreased antioxidant enzyme methionine sulfoxide reductase A (MsrA) and increased oxidative stress markers; FMT reversed these effects | Rat model/Limited sample size, Lack of control over microbiota composition, No motor behavior data, Male-only subjects | The findings suggest that FMT can restore gut microbiota balance and alleviate neurological deficits following TBI, potentially through modulation of the gut–brain axis and reduction of oxidative stress | [161,162] |
Administration of fecal microbiota from healthy donor mice to C57Bl/6 mice subjected to severe TBI via controlled cortical impact. FMT was performed weekly starting 1 h post-injury | TBI led to significant reductions in species richness and evenness; FMT restored these parameters, indicating reversal of dysbiosis. Histological analysis showed that FMT reduced microglial activation, as evidenced by decreased Iba1 expression, suggesting attenuation of neuroinflammatory responses. MRI assessments demonstrated that FMT mitigated ventriculomegaly and preserved white matter integrity, as indicated by improved fractional anisotropy values | Mouse model/Small sample size, Short-term observations, Animal model limits direct human applicability | The findings suggest that FMT can effectively restore gut microbiota balance and alleviate both functional and structural deficits following TBI, potentially through modulation of the gut–brain axis and reduction of neuroinflammation | [161,162] |
5.2. Neurobiology of SCI
Therapeutic Strategies for GBA Modulation in SCI
Intervention | Key Findings—Related Targets and Pathways | Model/Limitations | Effect on SCI | Ref. |
---|---|---|---|---|
Oral administration of nanoparticles composed of pectin and zein encapsulating indole-3-propionic acid (IPA) to mice with spinal cord injury | The intervention reduced pro-inflammatory cytokines (IL-6, TNF-α, IL-1β) and shifted microglial polarization from the M1 to M2 phenotype, indicating attenuation of neuroinflammation. It also activated the AKT/Nrf-2 signaling pathway, thereby enhancing the expression of antioxidant proteins such as HO-1, NQO1, and SOD2, which contributed to a reduction in oxidative stress. Additionally, it modulated gut microbiota composition by decreasing L-methionine-producing bacteria (e.g., Clostridia_UCG-014) and increasing beneficial bacteria (e.g., Parasutterella), leading to reduced L-methionine accumulation. Furthermore, intestinal barrier function was improved through the upregulation of tight junction proteins (ZO-1, Occludin) and inhibition of the NF-κB signaling pathway | Mouse model/Small sample size, Nanoparticle safety and long-term effects need assessment, short half-Life of IPA, Sex not reported | The findings suggest that Pectin-Zein-IPA nanoparticles can effectively promote functional recovery and alleviate neuroinflammation after spinal cord injury by modulating gut microbiota, reducing oxidative stress, and enhancing neuronal regeneration | [169] |
Increased generation of H2S in the colon following chronic constriction injury (CCI) in rodents | Elevated colonic H2S levels lead to activation of spinal N-methyl-D-aspartate (NMDA) receptors, contributing to neuropathic pain. This pain response is attenuated by the administration of H2S synthesis inhibitors or NMDA receptor antagonists (e.g., MK-801), highlighting the pivotal role of H2S-NMDA receptor interaction in pain modulation | Mouse model/Lack of direct mechanistic link, Sex unspecified, Translational applicability unclear | Colon-derived H2S induces neuropathic pain behaviors in rodents, suggesting that targeting its production or spinal NMDA receptor activation could offer potential therapeutic strategies for pain management | [170] |
Implementation of EAW training in patients with motor-complete SCI (T2–L1) to assess its impact on bowel function and gut microbiota | EAW training altered the abundance of intestinal flora, notably increasing beneficial bacteria, suggesting gut microbiota modulation, which may be linked to improvements in intestinal function through changes in the brain–gut axis, though the exact mechanisms remain unclear | Human Pilot clinical study/Small sample size, Pilot study design; no control group, Short duration | The findings suggest that EAW may serve as a promising intervention to alleviate bowel dysfunction in individuals with motor-complete spinal cord injuries, potentially through modulation of the gut microbiota and the brain–gut axis | [171] |
Administration of a PPARα agonist (GW7647) and antagonist (GW6471) in mice with chronic constriction injury (CCI) to assess the role of PPARα in neuropathic pain | PPARα activation altered the abundance, homogeneity, and composition of the gut microbiome, as revealed by 16S rRNA gene sequencing, and led to significant changes in spinal cord metabolites, including nicotinamide, benzimidazole, eicosanoids, and pyridine, that correlated with specific gut microbiota alterations; additionally, it promoted M2-type microglia polarization, thereby reducing neuroinflammation in the spinal cord | Mouse model/Small sample size, Limited scope of microbial analysis, Sex unspecified | The findings suggest that PPARα plays a crucial role in modulating neuropathic pain by influencing gut microbiota composition, spinal cord metabolite profiles, and microglial activation states. Targeting PPARα may offer a promising therapeutic strategy for managing neuropathic pain | [172] |
Oral administration of GV-971, a marine-derived oligosaccharide approved in China for mild-to-moderate Alzheimer’s disease, to NMOSD murine models induced by NMO-IgG and complement | GV-971 remodeled gut microbiota composition and restored microbial balance disrupted by NMOSD. It reduced peripheral inflammation and metabolic disorders, as shown by cytokine and metabolomics analyses. Histological evaluations revealed reduced neuroinflammation and neural injury in the spinal cord. These findings suggest GV-971 exerts therapeutic effects through gut-immune-CNS axis modulation | Mouse model/Mixed disease induction strategies which limit clarity on mechanism, Focus on young and aged mice | The findings suggest that GV-971 may attenuate the progression of NMOSD by modulating the gut microbiota and reducing peripheral and central inflammation | [173] |
6. Conclusions and Future Perspectives
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
Abbreviations
Gut–brain axis | GBA |
Central nervous system | CNS |
Neurodegenerative diseases | NDDs |
Short-chain fatty acids | SCFAs |
Parkinson’s disease | PD |
Alzheimer’s disease | AD |
Amyotrophic lateral sclerosis | ALS |
Huntington’s disease | HD |
Traumatic brain injury | TBI |
Multiple sclerosis | MS |
Indoleamine 2,3-dioxygenase 1 | IDO-1 |
Enteric nervous system | ENS |
Fecal microbiota transplantation | FMT |
Blood–brain barrier | BBB |
Aryl hydrocarbon receptor | AhR |
Lipopolysaccharides | LPS |
Gut-associated lymphoid tissue | GALT |
Beta-amyloid plaques | Aβ |
Gamma-aminobutyric acid | GABA |
Toll-like receptor 4 | TLR4 |
Low-density lipoprotein | LDL |
Parkinson Gut Prediction Method | PGPM |
Non-steroidal anti-inflammatory drugs | NSAIDs |
Glucagon-like peptide-1 | GLP-1 |
Inflammatory bowel disease | IBD |
Interferon-gamma | INF-γ |
Interleukin-6 | IL-6 |
Interleukin-10 | IL-10 |
Interleukin-1β | IL-1β |
Tumor necrosis factor-alpha | TNF-α |
Transforming growth factor beta | TGF-β |
Gastrointestinal | GI |
Galactooligosaccharides | GOS |
Fructo-oligosaccharides | FOS |
References
- Scalese, G.; Severi, C. The pathophysiology of gut–brain connection. In The Complex Interplay Between Gut-Brain, Gut-Liver, and Liver-Brain Axes; Elsevier: Amsterdam, The Netherlands, 2021; pp. 3–16. [Google Scholar]
- Arneth, B.M. Gut-brain axis biochemical signalling from the gastrointestinal tract to the central nervous system: Gut dysbiosis and altered brain function. Postgrad. Med. J. 2018, 94, 446–452. [Google Scholar] [CrossRef] [PubMed]
- Bhatt, B.; Patel, K.; Lee, C.N.; Moochhala, S. The Microbial Blueprint: The Impact of Your Gut on Your Well-being; Partridge Publishing: Singapore, 2024. [Google Scholar]
- O’Riordan, K.J.; Collins, M.K.; Moloney, G.M.; Knox, E.G.; Aburto, M.R.; Fülling, C.; Morley, S.J.; Clarke, G.; Schellekens, H.; Cryan, J.F. Short chain fatty acids: Microbial metabolites for gut-brain axis signalling. Mol. Cell. Endocrinol. 2022, 546, 111572. [Google Scholar] [CrossRef] [PubMed]
- Warren, A.; Nyavor, Y.; Zarabian, N.; Mahoney, A.; Frame, L.A. The microbiota-gut-brain-immune interface in the pathogenesis of neuroinflammatory diseases: A narrative review of the emerging literature. Front. Immunol. 2024, 15, 1365673. [Google Scholar] [CrossRef] [PubMed]
- Loh, J.S.; Mak, W.Q.; Tan, L.K.S.; Ng, C.X.; Chan, H.H.; Yeow, S.H.; Foo, J.B.; Ong, Y.S.; How, C.W.; Khaw, K.Y. Microbiota–gut–brain axis and its therapeutic applications in neurodegenerative diseases. Signal Transduct. Target. Ther. 2024, 9, 37. [Google Scholar]
- Di Vincenzo, F.; Del Gaudio, A.; Petito, V.; Lopetuso, L.R.; Scaldaferri, F. Gut microbiota, intestinal permeability, and systemic inflammation: A narrative review. Intern. Emerg. Med. 2024, 19, 275–293. [Google Scholar] [CrossRef]
- Hanscom, M.; Loane, D.J.; Shea-Donohue, T. Brain-gut axis dysfunction in the pathogenesis of traumatic brain injury. J. Clin. Invest. 2021, 131, e143777. [Google Scholar] [CrossRef]
- Yaqub, M.O.; Jain, A.; Joseph, C.E.; Edison, L.K. Microbiome-Driven Therapeutics: From Gut Health to Precision Medicine. Gastrointest. Disord. 2025, 7, 7. [Google Scholar] [CrossRef]
- Guo, B.; Zhang, J.; Zhang, W.; Chen, F.; Liu, B. Gut microbiota-derived short chain fatty acids act as mediators of the gut–brain axis targeting age-related neurodegenerative disorders: A narrative review. Crit. Rev. Food Sci. Nutr. 2025, 65, 265–286. [Google Scholar] [CrossRef]
- Cuesta, C.M.; Guerri, C.; Ureña, J.; Pascual, M. Role of microbiota-derived extracellular vesicles in gut-brain communication. Int. J. Mol. Sci. 2021, 22, 4235. [Google Scholar] [CrossRef]
- Portincasa, P.; Bonfrate, L.; Vacca, M.; De Angelis, M.; Farella, I.; Lanza, E.; Khalil, M.; Wang, D.Q.-H.; Sperandio, M.; Di Ciaula, A. Gut microbiota and short chain fatty acids: Implications in glucose homeostasis. Int. J. Mol. Sci. 2022, 23, 1105. [Google Scholar] [CrossRef]
- Ikeda, T.; Nishida, A.; Yamano, M.; Kimura, I. Short-chain fatty acid receptors and gut microbiota as therapeutic targets in metabolic, immune, and neurological diseases. Pharmacol. Ther. 2022, 239, 108273. [Google Scholar] [CrossRef] [PubMed]
- La Rosa, G.; Lonardo, M.S.; Cacciapuoti, N.; Muscariello, E.; Guida, B.; Faraonio, R.; Santillo, M.; Damiano, S. Dietary polyphenols, microbiome, and multiple sclerosis: From molecular anti-inflammatory and neuroprotective mechanisms to clinical evidence. Int. J. Mol. Sci. 2023, 24, 7247. [Google Scholar] [CrossRef] [PubMed]
- Fock, E.; Parnova, R. Mechanisms of blood–brain barrier protection by microbiota-derived short-chain fatty acids. Cells 2023, 12, 657. [Google Scholar] [CrossRef] [PubMed]
- Zhou, Y.; Chen, Y.; He, H.; Peng, M.; Zeng, M.; Sun, H. The role of the indoles in microbiota-gut-brain axis and potential therapeutic targets: A focus on human neurological and neuropsychiatric diseases. Neuropharmacology 2023, 239, 109690. [Google Scholar] [CrossRef]
- Pickard, J.M.; Zeng, M.Y.; Caruso, R.; Núñez, G. Gut microbiota: Role in pathogen colonization, immune responses, and inflammatory disease. Immunol. Rev. 2017, 279, 70–89. [Google Scholar] [CrossRef]
- Yang, J.; Wise, L.; Fukuchi, K.-I. TLR4 cross-talk with NLRP3 inflammasome and complement signaling pathways in Alzheimer’s disease. Front. Immunol. 2020, 11, 724. [Google Scholar] [CrossRef]
- Majd, S.; Power, J.H.; Grantham, H.J. Neuronal response in Alzheimer’s and Parkinson’s disease: The effect of toxic proteins on intracellular pathways. BMC Neurosci. 2015, 16, 69. [Google Scholar] [CrossRef]
- Johansson-Lindbom, B.; Svensson, M.; Wurbel, M.-A.; Malissen, B.; Márquez, G.; Agace, W. Selective generation of gut tropic T cells in gut-associated lymphoid tissue (GALT) requirement for GALT dendritic cells and adjuvant. J. Exp. Med. 2003, 198, 963–969. [Google Scholar] [CrossRef]
- Su, X.; Yin, X.; Liu, Y.; Yan, X.; Zhang, S.; Wang, X.; Lin, Z.; Zhou, X.; Gao, J.; Wang, Z.; et al. Gut Dysbiosis Contributes to the Imbalance of Treg and Th17 Cells in Graves’ Disease Patients by Propionic Acid. J. Clin. Endocrinol. Metab. 2020, 105, 3526–3547. [Google Scholar] [CrossRef]
- Maciak, K.; Pietrasik, S.; Dziedzic, A.; Redlicka, J.; Saluk-Bijak, J.; Bijak, M.; Włodarczyk, T.; Miller, E. Th17-related cytokines as potential discriminatory markers between neuromyelitis optica (Devic’s disease) and multiple sclerosis—A review. Int. J. Mol. Sci. 2021, 22, 8946. [Google Scholar] [CrossRef]
- Oemcke, L.A.; Anderson, R.C.; Altermann, E.; Roy, N.C.; McNabb, W.C. The role of segmented filamentous bacteria in immune barrier maturation of the small intestine at weaning. Front. Nutr. 2021, 8, 759137. [Google Scholar] [CrossRef] [PubMed]
- Aljeradat, B.; Kumar, D.; Abdulmuizz, S.; Kundu, M.; Almealawy, Y.F.; Batarseh, D.R.; Atallah, O.; Ennabe, M.; Alsarafandi, M.; Alan, A. Neuromodulation and the gut–brain axis: Therapeutic mechanisms and implications for gastrointestinal and neurological disorders. Pathophysiology 2024, 31, 244–268. [Google Scholar] [CrossRef] [PubMed]
- Jameson, K.G.; Kazmi, S.A.; Ohara, T.E.; Son, C.; Yu, K.B.; Mazdeyasnan, D.; Leshan, E.; Vuong, H.E.; Paramo, J.; Lopez-Romero, A. Select microbial metabolites in the small intestinal lumen regulates vagal activity via receptor-mediated signaling. Iscience 2025, 28, 111699. [Google Scholar] [CrossRef] [PubMed]
- Quadt, L.; Critchley, H.; Nagai, Y. Cognition, emotion, and the central autonomic network. Auton. Neurosci. 2022, 238, 102948. [Google Scholar] [CrossRef]
- Bravo, J.A.; Forsythe, P.; Chew, M.V.; Escaravage, E.; Savignac, H.M.; Dinan, T.G.; Bienenstock, J.; Cryan, J.F. Ingestion of Lactobacillus strain regulates emotional behavior and central GABA receptor expression in a mouse via the vagus nerve. Proc. Natl. Acad. Sci. USA 2011, 108, 16050–16055. [Google Scholar] [CrossRef]
- Van Autryve, B. The Role of the Vagus Nerve in Neuroinflammation. Master’s Thesis, Ghent University, Gent, Belgian, 2024. [Google Scholar]
- Kearns, R. Gut–brain axis and neuroinflammation: The role of gut permeability and the kynurenine pathway in neurological disorders. Cell. Mol. Neurobiol. 2024, 44, 64. [Google Scholar] [CrossRef]
- Zhao, B.; Yin, Q.; Fei, Y.; Zhu, J.; Qiu, Y.; Fang, W.; Li, Y. Research progress of mechanisms for tight junction damage on blood–brain barrier inflammation. Arch. Physiol. Biochem. 2022, 128, 1579–1590. [Google Scholar] [CrossRef]
- Munir, M.U.; Ali, S.A.; Chung, K.H.K.; Kakinen, A.; Javed, I.; Davis, T.P. Reverse engineering the Gut-Brain Axis and microbiome-metabolomics for symbiotic/pathogenic balance in neurodegenerative diseases. Gut Microbes 2024, 16, 2422468. [Google Scholar] [CrossRef]
- Ardizzone, A.; Capra, A.P.; Repici, A.; Lanza, M.; Bova, V.; Palermo, N.; Paterniti, I.; Esposito, E. Rebalancing NOX2/Nrf2 to limit inflammation and oxidative stress across gut-brain axis in migraine. Free Radic. Biol. Med. 2024, 213, 65–78. [Google Scholar] [CrossRef]
- Casili, G.; Lanza, M.; Campolo, M.; Siracusa, R.; Paterniti, I.; Ardizzone, A.; Scuderi, S.A.; Cuzzocrea, S.; Esposito, E. Synergic therapeutic potential of PEA-um treatment and NAAA enzyme silencing in the management of neuroinflammation. Int. J. Mol. Sci. 2020, 21, 7486. [Google Scholar] [CrossRef]
- DeTure, M.A.; Dickson, D.W. The neuropathological diagnosis of Alzheimer’s disease. Mol. Neurodegener. 2019, 14, 32. [Google Scholar] [CrossRef] [PubMed]
- Spillantini, M.G.; Goedert, M. Tau pathology and neurodegeneration. Lancet Neurol. 2013, 12, 609–622. [Google Scholar] [CrossRef] [PubMed]
- Gatz, M.; Reynolds, C.A.; Fratiglioni, L.; Johansson, B.; Mortimer, J.A.; Berg, S.; Fiske, A.; Pedersen, N.L. Role of genes and environments for explaining Alzheimer disease. Arch. Gen. Psychiatry 2006, 63, 168–174. [Google Scholar] [CrossRef] [PubMed]
- Rabinovici, G.D.; Carrillo, M.C.; Forman, M.; DeSanti, S.; Miller, D.S.; Kozauer, N.; Petersen, R.C.; Randolph, C.; Knopman, D.S.; Smith, E.E. Multiple comorbid neuropathologies in the setting of Alzheimer’s disease neuropathology and implications for drug development. Alzheimer’s Dement. Transl. Res. Clin. Interv. 2017, 3, 83–91. [Google Scholar] [CrossRef]
- Rullmann, M.; Brendel, M.; Schroeter, M.L.; Saur, D.; Levin, J.; Perneczky, R.G.; Tiepolt, S.; Patt, M.; Mueller, A.; Villemagne, V.L. Multicenter 18F-PI-2620 PET for in vivo Braak staging of tau pathology in Alzheimer’s disease. Biomolecules 2022, 12, 458. [Google Scholar] [CrossRef]
- Jack Jr, C.R.; Bennett, D.A.; Blennow, K.; Carrillo, M.C.; Dunn, B.; Haeberlein, S.B.; Holtzman, D.M.; Jagust, W.; Jessen, F.; Karlawish, J. NIA-AA research framework: Toward a biological definition of Alzheimer’s disease. Alzheimer’s Dement. 2018, 14, 535–562. [Google Scholar] [CrossRef]
- Pasupalak, J.K.; Rajput, P.; Gupta, G.L. Gut microbiota and Alzheimer’s disease: Exploring natural product intervention and the Gut–Brain axis for therapeutic strategies. Eur. J. Pharmacol. 2024, 984, 177022. [Google Scholar] [CrossRef]
- Nakhal, M.M.; Yassin, L.K.; Alyaqoubi, R.; Saeed, S.; Alderei, A.; Alhammadi, A.; Alshehhi, M.; Almehairbi, A.; Al Houqani, S.; BaniYas, S. The microbiota–gut–brain axis and neurological disorders: A comprehensive review. Life 2024, 14, 1234. [Google Scholar] [CrossRef]
- Braniste, V.; Al-Asmakh, M.; Kowal, C.; Anuar, F.; Abbaspour, A.; Tóth, M.; Korecka, A.; Bakocevic, N.; Ng, L.G.; Kundu, P. The gut microbiota influences blood-brain barrier permeability in mice. Sci. Transl. Med. 2014, 6, 263ra158. [Google Scholar] [CrossRef]
- Khan, I.M.; Nassar, N.; Chang, H.; Khan, S.; Cheng, M.; Wang, Z.; Xiang, X. The microbiota: A key regulator of health, productivity, and reproductive success in mammals. Front. Microbiol. 2024, 15, 1480811. [Google Scholar] [CrossRef]
- Kozhakhmetov, S.; Kaiyrlykyzy, A.; Jarmukhanov, Z.; Vinogradova, E.; Zholdasbekova, G.; Alzhanova, D.; Kunz, J.; Kushugulova, A.; Askarova, S. Inflammatory Manifestations Associated with Gut Dysbiosis in Alzheimer’s Disease. Int. J. Alzheimer’s Dis. 2024, 2024, 9741811. [Google Scholar] [CrossRef] [PubMed]
- Quansah, M.; David, M.A.; Martins, R.; El-Omar, E.; Aliberti, S.M.; Capunzo, M.; Jensen, S.O.; Tayebi, M. The Beneficial Effects of Lactobacillus Strains on Gut Microbiome in Alzheimer’s Disease: A Systematic Review. Healthcare 2025, 13, 74. [Google Scholar] [CrossRef] [PubMed]
- Łysiak, K.; Łysiak, A. The role of the gut microbiome in Alzheimer’s disease. Prospect. Pharm. Sci. 2024, 22, 81–85. [Google Scholar] [CrossRef]
- Kim, Y.; Lim, J.; Oh, J. Taming neuroinflammation in Alzheimer’s disease: The protective role of phytochemicals through the gut− brain axis. Biomed. Pharmacother. 2024, 178, 117277. [Google Scholar] [CrossRef]
- Older, E.A.; Zhang, J.; Ferris, Z.E.; Xue, D.; Zhong, Z.; Mitchell, M.K.; Madden, M.; Wang, Y.; Chen, H.; Nagarkatti, P. Biosynthetic enzyme analysis identifies a protective role for TLR4-acting gut microbial sulfonolipids in inflammatory bowel disease. Nat. Commun. 2024, 15, 9371. [Google Scholar] [CrossRef]
- Gu, X.; Fan, M.; Zhou, Y.; Zhang, Y.; Wang, L.; Gao, W.; Li, T.; Wang, H.; Si, N.; Wei, X. Intestinal endogenous metabolites affect neuroinflammation in 5× FAD mice by mediating “gut-brain” axis and the intervention with Chinese Medicine. Alzheimer’s Res. Ther. 2024, 16, 222. [Google Scholar] [CrossRef]
- Heston, M.B.; Hanslik, K.L.; Zarbock, K.R.; Harding, S.J.; Davenport-Sis, N.J.; Kerby, R.L.; Chin, N.; Sun, Y.; Hoeft, A.; Deming, Y. Gut inflammation associated with age and Alzheimer’s disease pathology: A human cohort study. Sci. Rep. 2023, 13, 18924. [Google Scholar] [CrossRef]
- Yang, J.; Liang, J.; Hu, N.; He, N.; Liu, B.; Liu, G.; Qin, Y. The gut microbiota modulates neuroinflammation in Alzheimer’s disease: Elucidating crucial factors and mechanistic underpinnings. CNS Neurosci. Ther. 2024, 30, e70091. [Google Scholar] [CrossRef]
- Solanki, R.; Karande, A.; Ranganathan, P. Emerging role of gut microbiota dysbiosis in neuroinflammation and neurodegeneration. Front. Neurol. 2023, 14, 1149618. [Google Scholar] [CrossRef]
- Saksida, T.; Koprivica, I.; Vujičić, M.; Stošić-Grujičić, S.; Perović, M.; Kanazir, S.; Stojanović, I. Impaired IL-17 production in gut-residing immune cells of 5xFAD mice with Alzheimer’s disease pathology. J. Alzheimer’s Dis. 2017, 61, 619–630. [Google Scholar] [CrossRef]
- Bemark, M.; Pitcher, M.J.; Dionisi, C.; Spencer, J. Gut-associated lymphoid tissue: A microbiota-driven hub of B cell immunity. Trends Immunol. 2024, 45, 211–223. [Google Scholar] [CrossRef] [PubMed]
- Vanderlip, C.R.; Stark, C.E. APOE4 Increases Susceptibility to Amyloid, Accelerating Episodic Memory Decline. bioRxiv 2024. [Google Scholar] [CrossRef]
- Ciurleo, G.C.; de Azevedo, O.G.; Carvalho, C.G.; Vitek, M.P.; Warren, C.A.; Guerrant, R.L.; Oriá, R.B. Apolipoprotein E4 and Alzheimer’s disease causality under adverse environments and potential intervention by senolytic nutrients. Clin. Nutr. ESPEN 2024, 64, 16–20. [Google Scholar] [CrossRef] [PubMed]
- Ardizzone, A.; Bova, V.; Casili, G.; Filippone, A.; Campolo, M.; Lanza, M.; Esposito, E.; Paterniti, I. SUN11602, a bFGF mimetic, modulated neuroinflammation, apoptosis and calcium-binding proteins in an in vivo model of MPTP-induced nigrostriatal degeneration. J. Neuroinflamm. 2022, 19, 107. [Google Scholar] [CrossRef]
- Berg, D.; Borghammer, P.; Fereshtehnejad, S.-M.; Heinzel, S.; Horsager, J.; Schaeffer, E.; Postuma, R.B. Prodromal Parkinson disease subtypes—Key to understanding heterogeneity. Nat. Rev. Neurol. 2021, 17, 349–361. [Google Scholar] [CrossRef]
- Borghammer, P.; Van Den Berge, N. Brain-first versus gut-first Parkinson’s disease: A hypothesis. J. Park. Dis. 2019, 9, S281–S295. [Google Scholar] [CrossRef]
- Beekes, M. The Neural Gut–Brain Axis of Pathological Protein Aggregation in Parkinson’s Disease and Its Counterpart in Peroral Prion Infections. Viruses 2021, 13, 1394. [Google Scholar] [CrossRef]
- Hirayama, M.; Ohno, K. Parkinson’s disease and gut microbiota. Ann. Nutr. Metab. 2021, 77, 28–35. [Google Scholar] [CrossRef]
- Heravi, F.S.; Naseri, K.; Hu, H. Gut microbiota composition in patients with neurodegenerative disorders (Parkinson’s and Alzheimer’s) and healthy controls: A systematic review. Nutrients 2023, 15, 4365. [Google Scholar] [CrossRef]
- Heidari, A.; Yazdanpanah, N.; Rezaei, N. The role of Toll-like receptors and neuroinflammation in Parkinson’s disease. J. Neuroinflamm. 2022, 19, 135. [Google Scholar] [CrossRef]
- Kline, E.M.; Houser, M.C.; Herrick, M.K.; Seibler, P.; Klein, C.; West, A.; Tansey, M.G. Genetic and environmental factors in P arkinson’s disease converge on immune function and inflammation. Mov. Disord. 2021, 36, 25–36. [Google Scholar] [CrossRef] [PubMed]
- Liang, X.; Huang, G.; Wang, Y.; Andrikopoulos, N.; Tang, H.; Ding, F.; Li, Y.; Ke, P.C. Polystyrene Nanoplastics Hitch-Hike the Gut–Brain Axis to Exacerbate Parkinson’s Pathology. ACS Nano 2025, 19, 5475–5492. [Google Scholar] [CrossRef] [PubMed]
- Ni, Y.; Tong, Q.; Xu, M.; Gu, J.; Ye, H. Gut microbiota-induced modulation of the central nervous system function in Parkinson’s disease through the gut-brain Axis and short-chain fatty acids. Mol. Neurobiol. 2025, 62, 2480–2492. [Google Scholar] [CrossRef] [PubMed]
- Leem, Y.-H.; Park, J.-E.; Park, J.-S.; Kim, D.-Y.; Park, J.-M.; Kim, S.-E.; Kang, J.L.; Kim, H.-S. Activation of α7nAch receptors ameliorates α-synuclein pathology in the brain and gut of a subacute MPTP mouse model of Parkinson’s disease. Biomed. Pharmacother. 2025, 184, 117871. [Google Scholar] [CrossRef]
- Zhao, Y.; Lai, Y.; Darweesh, S.K.; Bloem, B.R.; Forsgren, L.; Hansen, J.; Katzke, V.A.; Masala, G.; Sieri, S.; Sacerdote, C. Gut Microbial Metabolites and Future Risk of Parkinson’s Disease: A Metabolome-Wide Association Study. Mov. Disord. 2024, 40, 556–560. [Google Scholar] [CrossRef]
- Wu, J.; Li, C.-S.; Huang, W.-Y.; Zhou, S.-Y.; Zhao, L.-P.; Li, T.; Li, M.-A.; Zhang, M.-X.; Qiao, C.-M.; Zhao, W.-J. Gut microbiota promote the propagation of pathologic α-syn from gut to brain in a gut-originated mouse model of Parkinson’s disease. Brain Behav. Immun. 2025, 128, 152–169. [Google Scholar] [CrossRef]
- Yu, B.; Zhang, H.; Zhang, M. Deep learning-based differential gut flora for prediction of Parkinson’s. PLoS ONE 2025, 20, e0310005. [Google Scholar] [CrossRef]
- Wang, W.; Shi, S.; Su, M.; Li, Y.; Yao, X.; Jiang, J.; Yao, W.; Qin, X.; Wang, Z.; Tang, C. Akkermansia muciniphila Akk11 Supplementation Attenuates MPTP-Induced Neurodegeneration by Inhibiting Microglial NLRP3 Inflammasome. Probiotics Antimicrob. Proteins, 2025; Online ahead of print. [Google Scholar] [CrossRef]
- Qi, Y.; Dong, Y.; Chen, J.; Xie, S.; Ma, X.; Yu, X.; Yu, Y.; Wang, Y. Lactiplantibacillus plantarum SG5 inhibits neuroinflammation in MPTP-induced PD mice through GLP-1/PGC-1α pathway. Exp. Neurol. 2025, 383, 115001. [Google Scholar] [CrossRef]
- Gao, W.; Wu, X.; Wang, Y.; Lu, F.; Liu, F. Brazilin-Rich Extract from Caesalpinia sappan L. Attenuated the Motor Deficits and Neurodegeneration in MPTP/p-Induced Parkinson’s Disease Mice by Regulating Gut Microbiota and Inhibiting Inflammatory Responses. ACS Chem. Neurosci. 2024, 16, 181–194. [Google Scholar] [CrossRef]
- Qiao, C.-M.; Ma, X.-Y.; Tan, L.-L.; Xia, Y.-M.; Li, T.; Wu, J.; Cui, C.; Zhao, W.-J.; Shen, Y.-Q. Indoleamine 2, 3-dioxygenase 1 inhibition mediates the therapeutic effects in Parkinson’s disease mice by modulating inflammation and neurogenesis in a gut microbiota dependent manner. Exp. Neurol. 2025, 385, 115142. [Google Scholar] [CrossRef]
- Shao, X.; Wu, T.; Li, M.; Zheng, M.; Lin, H.; Qi, X. Enterococcus faecalis Exerts Neuroprotective Effects via the Vagus Nerve in a Mouse Model of Parkinson’s Disease. Mol. Neurobiol. 2025, 62, 7875–7891. [Google Scholar] [CrossRef] [PubMed]
- Shang, M.; Ning, J.; Zang, C.; Ma, J.; Yang, Y.; Jiang, Y.; Chen, Q.; Dong, Y.; Wang, J.; Li, F. FLZ attenuates Parkinson’s disease pathological damage by increasing glycoursodeoxycholic acid production via down-regulating Clostridium innocuum. Acta Pharm. Sin. B 2025, 15, 973–990. [Google Scholar] [CrossRef] [PubMed]
- Hassan, H.M.; Abou-Hany, H.O.; Shata, A.; Hellal, D.; El-Baz, A.M.; ElSaid, Z.H.; Haleem, A.A.; Morsy, N.E.; Abozied, R.M.; Elbrolosy, B.M. Vinpocetine and Lactobacillus Attenuated Rotenone-Induced Parkinson’s Disease and Restored Dopamine Synthesis in Rats through Modulation of Oxidative Stress, Neuroinflammation, and Lewy Bodies Inclusion. J. Neuroimmune Pharmacol. 2025, 20, 22. [Google Scholar] [CrossRef] [PubMed]
- Chtioui, N.; Duval, C.; St-Pierre, D.H. The impact of an active lifestyle on markers of intestinal inflammation in Parkinson’s disease: Preliminary findings. Clin. Park. Relat. Disord. 2025, 12, 100301. [Google Scholar] [CrossRef]
- Bolen, M.L.; Gomes, B.N.; Gill, B.; Menees, K.B.; Staley, H.; Jernigan, J.; McFarland, N.R.; Zimmermann, E.M.; Forsmark, C.E.; Tansey, M.G. Peripheral blood immune cells from individuals with Parkinson’s disease or inflammatory bowel disease share deficits in iron storage and transport that are modulated by non-steroidal anti-inflammatory drugs. Neurobiol. Dis. 2025, 207, 106794. [Google Scholar] [CrossRef]
- Zaman, V.; Matzelle, D.; Banik, N.L.; Haque, A. Dysregulation of Metabolic Peptides Precedes Hyperinsulinemia and Inflammation Following Exposure to Rotenone in Rats. Cells 2025, 14, 124. [Google Scholar] [CrossRef]
- Kempuraj, D.; Thangavel, R.; Natteru, P.; Selvakumar, G.; Saeed, D.; Zahoor, H.; Zaheer, S.; Iyer, S.; Zaheer, A. Neuroinflammation induces neurodegeneration. J. Neurol. Neurosurg. Spine 2016, 1, 1003. [Google Scholar]
- Miner, A.E.; Dastgheyb, N.; Palomino, M.; Graves, J.S. The Genetics of Multiple Sclerosis. In Neuroimmunology: Multiple Sclerosis, Autoimmune Neurology and Related Diseases; Springer: Berlin/Heidelberg, Germany, 2021; pp. 155–172. [Google Scholar]
- Beard, K.; Srivastava, S.; Sharma, K.; Jaiswal, S.; Reddy, S.P.; Lisak, R.P.; Sriwastava, S. Epidemiology, epigenetics, and etiological factors in multiple sclerosis. In Clinical Aspects of Multiple Sclerosis Essentials and Current Updates; Elsevier: Amsterdam, The Netherlands, 2024; pp. 67–96. [Google Scholar]
- Garg, N.; Smith, T.W. An update on immunopathogenesis, diagnosis, and treatment of multiple sclerosis. Brain Behav. 2015, 5, e00362. [Google Scholar] [CrossRef]
- Bjørklund, G.; Wallace, D.R.; Hangan, T.; Butnariu, M.; Gurgas, L.; Peana, M. Cerebral iron accumulation in multiple sclerosis: Pathophysiology and therapeutic implications. Autoimmun. Rev. 2025, 24, 103741. [Google Scholar] [CrossRef]
- Falcão, A.M.; van Bruggen, D.; Marques, S.; Meijer, M.; Jäkel, S.; Agirre, E.; Samudyata, n.; Floriddia, E.M.; Vanichkina, D.P.; Ffrench-Constant, C. Disease-specific oligodendrocyte lineage cells arise in multiple sclerosis. Nat. Med. 2018, 24, 1837–1844. [Google Scholar] [CrossRef]
- Ghasemi, N.; Razavi, S.; Nikzad, E. Multiple sclerosis: Pathogenesis, symptoms, diagnoses and cell-based therapy. Cell J. (Yakhteh) 2017, 19, 1. [Google Scholar]
- Zierfuss, B.; Larochelle, C.; Prat, A. Blood–brain barrier dysfunction in multiple sclerosis: Causes, consequences, and potential effects of therapies. Lancet Neurol. 2024, 23, 95–109. [Google Scholar] [CrossRef] [PubMed]
- Msheik, A.N.; Al Mokdad, Z.; Hamed, F.; Assi, F.; Jibbawi, A.; Saad, J.-P.; Mohanna, R.; Khoury, A.; Farhat, M.; Atat, R. Epstein–Barr virus flare: A multiple sclerosis attack. Surg. Neurol. Int. 2024, 15, 355. [Google Scholar] [CrossRef] [PubMed]
- Hedström, A.K. Risk factors for multiple sclerosis in the context of Epstein-Barr virus infection. Front. Immunol. 2023, 14, 1212676. [Google Scholar] [CrossRef]
- Bakhshi, A.; Eslami, N.; Norouzi, N.; Letafatkar, N.; Amini-Salehi, E.; Hassanipour, S. The association between various viral infections and multiple sclerosis: An umbrella review on systematic review and meta-analysis. Rev. Med. Virol. 2024, 34, e2494. [Google Scholar] [CrossRef]
- Giordano, A.; Clarelli, F.; Pignolet, B.; Mascia, E.; Sorosina, M.; Misra, K.; Ferrè, L.; Bucciarelli, F.; Manouchehrinia, A.; Moiola, L. Vitamin D affects the risk of disease activity in multiple sclerosis. J. Neurol. Neurosurg. Psychiatry 2025, 96, 170–176. [Google Scholar] [CrossRef]
- Wu, J.; Olsson, T.; Alfredsson, L.; Hedström, A.K. Association between sun exposure habits and disease progression in multiple sclerosis. Eur. J. Neurol. 2024, 31, e16269. [Google Scholar] [CrossRef]
- Ouyang, Q.; Yu, H.; Xu, L.; Yu, M.; Zhang, Y. Relationship between gut microbiota and multiple sclerosis: A scientometric visual analysis from 2010 to 2023. Front. Immunol. 2024, 15, 1451742. [Google Scholar]
- Yadav, S.K.; Ito, N.; Mindur, J.E.; Kumar, H.; Youssef, M.; Suresh, S.; Kulkarni, R.; Rosario, Y.; Balashov, K.E.; Dhib-Jalbut, S. Fecal Lcn-2 level is a sensitive biological indicator for gut dysbiosis and intestinal inflammation in multiple sclerosis. Front. Immunol. 2022, 13, 1015372. [Google Scholar] [CrossRef]
- Doenyas, C.; Clarke, G.; Cserjési, R. Gut–brain axis and neuropsychiatric health: Recent advances. Sci. Rep. 2025, 15, 3415. [Google Scholar] [CrossRef]
- Cryan, J.F.; O’Riordan, K.J.; Cowan, C.S.; Sandhu, K.V.; Bastiaanssen, T.F.; Boehme, M.; Codagnone, M.G.; Cussotto, S.; Fulling, C.; Golubeva, A.V. The microbiota-gut-brain axis. Physiol. Rev. 2019, 99, 1877–2013. [Google Scholar] [CrossRef] [PubMed]
- Mostafavi Abdolmaleky, H.; Zhou, J.-R. Gut microbiota dysbiosis, oxidative stress, inflammation, and epigenetic alterations in metabolic diseases. Antioxidants 2024, 13, 985. [Google Scholar] [CrossRef] [PubMed]
- Mowry, E.M.; Glenn, J.D. The dynamics of the gut microbiome in multiple sclerosis in relation to disease. Neurol. Clin. 2018, 36, 185–196. [Google Scholar] [CrossRef] [PubMed]
- Metaxouli, K.; Tsiou, C.; Dokoutsidou, E.; Margari, N. Nutritional Intervention in Patients with Multiple Sclerosis, Correlation with Quality of Life and Disability—A Prospective and Quasi-Experimental Study. NeuroSci 2025, 6, 4. [Google Scholar] [CrossRef]
- Chu, F.; Shi, M.; Lang, Y.; Shen, D.; Jin, T.; Zhu, J.; Cui, L. Gut microbiota in multiple sclerosis and experimental autoimmune encephalomyelitis: Current applications and future perspectives. Mediat. Inflamm. 2018, 2018, 8168717. [Google Scholar] [CrossRef]
- Al, K.F.; Craven, L.J.; Gibbons, S.; Parvathy, S.N.; Wing, A.C.; Graf, C.; Parham, K.A.; Kerfoot, S.M.; Wilcox, H.; Burton, J.P. Fecal microbiota transplantation is safe and tolerable in patients with multiple sclerosis: A pilot randomized controlled trial. Mult. Scler. J. Exp. Transl. Clin. 2022, 8, 20552173221086662. [Google Scholar] [CrossRef]
- Dopkins, N.; Becker, W.; Miranda, K.; Walla, M.; Nagarkatti, P.; Nagarkatti, M. Tryptamine attenuates experimental multiple sclerosis through activation of aryl hydrocarbon receptor. Front. Pharmacol. 2021, 11, 619265. [Google Scholar] [CrossRef]
- Melnikov, M.; Kasatkin, D.; Lopatina, A.; Spirin, N.; Boyko, A.; Pashenkov, M. Serotonergic drug repurposing in multiple sclerosis: A new possibility for disease-modifying therapy. Front. Neurol. 2022, 13, 920408. [Google Scholar] [CrossRef]
- Muñoz-Jurado, A.; Escribano, B.M.; Caballero-Villarraso, J.; Galván, A.; Agüera, E.; Santamaría, A.; Túnez, I. Melatonin and multiple sclerosis: Antioxidant, anti-inflammatory and immunomodulator mechanism of action. Inflammopharmacology 2022, 30, 1569–1596. [Google Scholar] [CrossRef]
- Shen, C.; Luo, Z.; Ma, S.; Yu, C.; Gao, Q.; Zhang, M.; Zhang, H.; Zhang, J.; Xu, W.; Yao, J. Microbe-derived antioxidants reduce lipopolysaccharide-induced inflammatory responses by activating the Nrf2 pathway to inhibit the ROS/NLRP3/IL-1β signaling pathway. Int. J. Mol. Sci. 2022, 23, 12477. [Google Scholar] [CrossRef]
- Erny, D.; Hrabě de Angelis, A.L.; Jaitin, D.; Wieghofer, P.; Staszewski, O.; David, E.; Keren-Shaul, H.; Mahlakoiv, T.; Jakobshagen, K.; Buch, T. Host microbiota constantly control maturation and function of microglia in the CNS. Nat. Neurosci. 2015, 18, 965–977. [Google Scholar] [CrossRef] [PubMed]
- Yu, H.; Bai, S.; Hao, Y.; Guan, Y. Fatty acids role in multiple sclerosis as “metabokines”. J. Neuroinflamm. 2022, 19, 157. [Google Scholar] [CrossRef] [PubMed]
- Furusawa, Y.; Obata, Y.; Fukuda, S.; Endo, T.A.; Nakato, G.; Takahashi, D.; Nakanishi, Y.; Uetake, C.; Kato, K.; Kato, T. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature 2013, 504, 446–450. [Google Scholar] [CrossRef] [PubMed]
- Pröbstel, A.-K.; Zhou, X.; Baumann, R.; Wischnewski, S.; Kutza, M.; Rojas, O.L.; Sellrie, K.; Bischof, A.; Kim, K.; Ramesh, A. Gut microbiota–specific IgA+ B cells traffic to the CNS in active multiple sclerosis. Sci. Immunol. 2020, 5, eabc7191. [Google Scholar] [CrossRef]
- Dhasmana, S.; Dhasmana, A.; Narula, A.S.; Jaggi, M.; Yallapu, M.M.; Chauhan, S.C. The panoramic view of amyotrophic lateral sclerosis: A fatal intricate neurological disorder. Life Sci. 2022, 288, 120156. [Google Scholar] [CrossRef]
- Van Es, M.A.; Hardiman, O.; Chio, A.; Al-Chalabi, A.; Pasterkamp, R.J.; Veldink, J.H.; Van den Berg, L.H. Amyotrophic lateral sclerosis. Lancet 2017, 390, 2084–2098. [Google Scholar] [CrossRef]
- Ludolph, A.C.; Dietrich, J.; Dreyhaupt, J.; Kassubek, J.; Del Tredici, K.; Rosenbohm, A. Clinical spreading of muscle weakness in amyotrophic lateral sclerosis (ALS): A study in 910 patients. J. Neurol. 2024, 271, 5357–5367. [Google Scholar] [CrossRef]
- Xu, Z.; Xu, R. Current potential diagnostic biomarkers of amyotrophic lateral sclerosis. Rev. Neurosci. 2024, 35, 917–931. [Google Scholar] [CrossRef]
- Ciervo, Y.; Ning, K.; Jun, X.; Shaw, P.J.; Mead, R.J. Advances, challenges and future directions for stem cell therapy in amyotrophic lateral sclerosis. Mol. Neurodegener. 2017, 12, 85. [Google Scholar] [CrossRef]
- Al-Khayri, J.M.; Ravindran, M.; Banadka, A.; Vandana, C.D.; Priya, K.; Nagella, P.; Kukkemane, K. Amyotrophic Lateral Sclerosis: Insights and New Prospects in Disease Pathophysiology, Biomarkers and Therapies. Pharmaceuticals 2024, 17, 1391. [Google Scholar] [CrossRef]
- Song, L.; Gao, Y.; Zhang, X.; Le, W. Galactooligosaccharide improves the animal survival and alleviates motor neuron death in SOD1G93A mouse model of amyotrophic lateral sclerosis. Neuroscience 2013, 246, 281–290. [Google Scholar] [CrossRef] [PubMed]
- Labarre, A.; Guitard, E.; Tossing, G.; Forest, A.; Bareke, E.; Labrecque, M.; Tétreault, M.; Ruiz, M.; Alex Parker, J. Fatty acids derived from the probiotic Lacticaseibacillus rhamnosus HA-114 suppress age-dependent neurodegeneration. Commun. Biol. 2022, 5, 1340. [Google Scholar] [CrossRef] [PubMed]
- Chen, P.-C.; Kaur, K.; Ko, M.-W.; Huerta-Yepez, S.; Jain, Y.; Jewett, A. Regulation of Cytotoxic Immune Effector Function by AJ3 Probiotic Bacteria in Amyotrophic Lateral Sclerosis (ALS). Crit. Rev. Immunol. 2023, 43, 13–26. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Y.; Xia, Y.; Sun, J. Probiotics and microbial metabolites maintain barrier and neuromuscular functions and clean protein aggregation to delay disease progression in TDP43 mutation mice. Gut Microbes 2024, 16, 2363880. [Google Scholar] [CrossRef]
- Zhang, Y.-g.; Wu, S.; Yi, J.; Xia, Y.; Jin, D.; Zhou, J.; Sun, J. Target intestinal microbiota to alleviate disease progression in amyotrophic lateral sclerosis. Clin. Ther. 2017, 39, 322–336. [Google Scholar] [CrossRef]
- Ogbu, D.; Zhang, Y.; Claud, K.; Xia, Y.; Sun, J. Target metabolites to slow down progression of amyotrophic lateral sclerosis in mice. Metabolites 2022, 12, 1253. [Google Scholar] [CrossRef]
- Ryu, H.; Smith, K.; Camelo, S.I.; Carreras, I.; Lee, J.; Iglesias, A.H.; Dangond, F.; Cormier, K.A.; Cudkowicz, M.E.; Brown, R.H. Sodium phenylbutyrate prolongs survival and regulates expression of anti-apoptotic genes in transgenic amyotrophic lateral sclerosis mice. J. Neurochem. 2006, 96, 908. [Google Scholar] [CrossRef]
- Del Signore, S.J.; Amante, D.J.; Kim, J.; Stack, E.C.; Goodrich, S.; Cormier, K.; Smith, K.; Cudkowicz, M.E.; Ferrante, R.J. Combined riluzole and sodium phenylbutyrate therapy in transgenic amyotrophic lateral sclerosis mice. Amyotroph. Lateral Scler. 2009, 10, 85–94. [Google Scholar] [CrossRef]
- Yao, J.-y.; Liu, T.; Hu, X.-r.; Sheng, H.; Chen, Z.-h.; Zhao, H.-y.; Li, X.-j.; Wang, Y.; Hao, L. An insight into allele-selective approaches to lowering mutant huntingtin protein for Huntington’s disease treatment. Biomed. Pharmacother. 2024, 180, 117557. [Google Scholar] [CrossRef]
- Landles, C.; Osborne, G.F.; Phillips, J.; Pico, M.C.; Nita, I.M.; Ali, N.; Greene, J.R.; Sathasivam, K.; Bates, G.P. Mutant HTT protein decreases with CAG repeat expansion: Implications for therapeutics and bioassays. Brain Commun. 2024, 6, fcae410. [Google Scholar] [CrossRef]
- Gil, J.M.; Rego, A.C. Mechanisms of neurodegeneration in Huntington’s disease. Eur. J. Neurosci. 2008, 27, 2803–2820. [Google Scholar] [CrossRef] [PubMed]
- Miguez, A.; Gomis, C.; Vila, C.; Monguió-Tortajada, M.; Fernández-García, S.; Bombau, G.; Galofré, M.; García-Bravo, M.; Sanders, P.; Fernández-Medina, H. Soluble mutant huntingtin drives early human pathogenesis in Huntington’s disease. Cell. Mol. Life Sci. 2023, 80, 238. [Google Scholar] [CrossRef] [PubMed]
- Dhingra, H.; Gaidhane, S.A. Huntington’s disease: Understanding its novel drugs and treatments. Cureus 2023, 15, e47526. [Google Scholar] [CrossRef] [PubMed]
- Pauli, W.M.; O’Reilly, R.C.; Yarkoni, T.; Wager, T.D. Regional specialization within the human striatum for diverse psychological functions. Proc. Natl. Acad. Sci. USA 2016, 113, 1907–1912. [Google Scholar] [CrossRef]
- Aguirre, C.G.; Tshilenge, K.-T.; Battistoni, E.; Lopez-Ramirez, A.; Naphade, S.; Perez, K.; Song, S.; Mooney, S.D.; Melov, S.; Ehrlich, M.E. Transcriptomic characterization reveals disrupted medium spiny neuron trajectories in Huntington’s disease and possible therapeutic avenues. bioRxiv 2023. [Google Scholar] [CrossRef]
- Dai, Y.; Wang, H.; Lian, A.; Li, J.; Zhao, G.; Hu, S.; Li, B. A comprehensive perspective of Huntington’s disease and mitochondrial dysfunction. Mitochondrion 2023, 70, 8–19. [Google Scholar] [CrossRef]
- Cavalcante, F.S.B.M.; de Araújo, L.L.; Gomes, G.F.; Galvão, S.L.; Hipólito, T.C.S.a.E.; de Campos Piagentini, M.; dos Santos, J.C.C. Role of Gut-Microbiome-Brain-Axis in Neurodegenerative Diseases: A Review on Mechanisms and Potential Therapeutics. Braz. J. Clin. Med. Rev. 2025, 3, bjcmr5. [Google Scholar] [CrossRef]
- Wasser, C.I.; Mercieca, E.-C.; Kong, G.; Hannan, A.J.; McKeown, S.J.; Glikmann-Johnston, Y.; Stout, J.C. Gut dysbiosis in Huntington’s disease: Associations among gut microbiota, cognitive performance and clinical outcomes. Brain Commun. 2020, 2, fcaa110. [Google Scholar] [CrossRef]
- Kong, G.; Lê Cao, K.-A.; Judd, L.M.; Li, S.; Renoir, T.; Hannan, A.J. Microbiome profiling reveals gut dysbiosis in a transgenic mouse model of Huntington’s disease. Neurobiol. Dis. 2020, 135, 104268. [Google Scholar] [CrossRef]
- Adanyeguh, I.M.; Rinaldi, D.; Henry, P.-G.; Caillet, S.; Valabregue, R.; Durr, A.; Mochel, F. Triheptanoin improves brain energy metabolism in patients with Huntington disease. Neurology 2015, 84, 490–495. [Google Scholar] [CrossRef]
- Kacher, R.; Mounier, C.; Caboche, J.; Betuing, S. Altered cholesterol homeostasis in Huntington’s disease. Front. Aging Neurosci. 2022, 14, 797220. [Google Scholar] [CrossRef] [PubMed]
- Wasser, C.I.; Mercieca, E.-C.; Kong, G.; Hannan, A.J.; Allford, B.; McKeown, S.J.; Stout, J.C.; Glikmann-Johnston, Y. A randomized controlled trial of probiotics targeting gut dysbiosis in Huntington’s disease. J. Huntington’s Dis. 2023, 12, 43–55. [Google Scholar] [CrossRef] [PubMed]
- Gubert, C.; Choo, J.M.; Love, C.J.; Kodikara, S.; Masson, B.A.; Liew, J.J.; Wang, Y.; Kong, G.; Narayana, V.K.; Renoir, T. Faecal microbiota transplant ameliorates gut dysbiosis and cognitive deficits in Huntington’s disease mice. Brain Commun. 2022, 4, fcac205. [Google Scholar] [CrossRef] [PubMed]
- Pathak, S.; Nadar, R.; Kim, S.; Liu, K.; Govindarajulu, M.; Cook, P.; Watts Alexander, C.S.; Dhanasekaran, M.; Moore, T. The influence of kynurenine metabolites on neurodegenerative pathologies. Int. J. Mol. Sci. 2024, 25, 853. [Google Scholar] [CrossRef]
- Bondulich, M.K.; Fan, Y.; Song, Y.; Giorgini, F.; Bates, G.P. Ablation of kynurenine 3-monooxygenase rescues plasma inflammatory cytokine levels in the R6/2 mouse model of Huntington’s disease. Sci. Rep. 2021, 11, 5484. [Google Scholar] [CrossRef]
- Rauchman, S.H.; Zubair, A.; Jacob, B.; Rauchman, D.; Pinkhasov, A.; Placantonakis, D.G.; Reiss, A.B. Traumatic brain injury: Mechanisms, manifestations, and visual sequelae. Front. Neurosci. 2023, 17, 1090672. [Google Scholar] [CrossRef]
- Cederberg, D.; Siesjö, P. What has inflammation to do with traumatic brain injury? Child’s Nerv. Syst. 2010, 26, 221–226. [Google Scholar] [CrossRef]
- Rock, K.L.; Latz, E.; Ontiveros, F.; Kono, H. The sterile inflammatory response. Annu. Rev. Immunol. 2009, 28, 321–342. [Google Scholar] [CrossRef]
- Corps, K.N.; Roth, T.L.; McGavern, D.B. Inflammation and neuroprotection in traumatic brain injury. JAMA Neurol. 2015, 72, 355–362. [Google Scholar] [CrossRef]
- Karve, I.P.; Taylor, J.M.; Crack, P.J. The contribution of astrocytes and microglia to traumatic brain injury. Br. J. Pharmacol. 2016, 173, 692–702. [Google Scholar] [CrossRef]
- Stewart, I.J.; Howard, J.T.; Amuan, M.E.; Kennedy, E.; Balke, J.E.; Poltavskiy, E.; Walker, L.E.; Haigney, M.; Pugh, M.J. Traumatic brain injury is associated with the subsequent risk of atrial fibrillation or atrial flutter. Heart Rhythm. 2024, 22, 661–667. [Google Scholar] [CrossRef] [PubMed]
- Tsymbalyuk, O.; Gerzanich, V.; Simard, J.M.; Rathinam, C.V. Traumatic brain injury alters dendritic cell differentiation and distribution in lymphoid and non-lymphoid organs. J. Neuroinflamm. 2022, 19, 238. [Google Scholar] [CrossRef] [PubMed]
- Kerr, N.A.; de Rivero Vaccari, J.P.; Abbassi, S.; Kaur, H.; Zambrano, R.; Wu, S.; Dietrich, W.D.; Keane, R.W. Traumatic brain injury-induced acute lung injury: Evidence for activation and inhibition of a neural-respiratory-inflammasome axis. J. Neurotrauma 2018, 35, 2067–2076. [Google Scholar] [CrossRef] [PubMed]
- Kharrazian, D. Traumatic brain injury and the effect on the brain-gut axis. Altern. Ther. Health Med. 2015, 21, 28–32. [Google Scholar]
- Urban, R.J.; Pyles, R.B.; Stewart, C.J.; Ajami, N.; Randolph, K.M.; Durham, W.J.; Danesi, C.P.; Dillon, E.L.; Summons, J.R.; Singh, C.K. Altered fecal microbiome years after traumatic brain injury. J. Neurotrauma 2020, 37, 1037–1051. [Google Scholar] [CrossRef]
- Godinho, D.B.; da Silva Fiorin, F.; Oliveira, M.S.; Furian, A.F.; Fighera, M.R.; Royes, L.F.F. The immunological influence of physical exercise on TBI-induced pathophysiology: Crosstalk between the spleen, gut, and brain. Neurosci. Biobehav. Rev. 2021, 130, 15–30. [Google Scholar] [CrossRef]
- Flinn, H.; Marshall, A.; Holcomb, M.; Cruz, L.; Soriano, S.; Treangen, T.J.; Villapol, S. Antibiotic treatment induces microbiome dysbiosis and reduction of neuroinflammation following traumatic brain injury in mice. Res. Sq. 2024, rs.3.rs-4475195. [Google Scholar] [CrossRef]
- Celorrio, M.; Abellanas, M.A.; Rhodes, J.; Goodwin, V.; Moritz, J.; Vadivelu, S.; Wang, L.; Rodgers, R.; Xiao, S.; Anabayan, I. Gut microbial dysbiosis after traumatic brain injury modulates the immune response and impairs neurogenesis. Acta Neuropathol. Commun. 2021, 9, 40. [Google Scholar] [CrossRef]
- Amaral, W.Z.; Kokroko, N.; Treangen, T.J.; Villapol, S.; Gomez-Pinilla, F. Probiotic therapy modulates the brain-gut-liver microbiota axis in a mouse model of traumatic brain injury. Biochim. Biophys. Acta (BBA)-Mol. Basis Dis. 2024, 1870, 167483. [Google Scholar] [CrossRef]
- Yanckello, L.M.; Fanelli, B.; McCulloch, S.; Xing, X.; Sun, M.; Hammond, T.C.; Colwell, R.; Gu, Z.; Ericsson, A.C.; Chang, Y.-H. Inulin supplementation mitigates gut dysbiosis and brain impairment induced by mild traumatic brain injury during chronic phase. J. Cell. Immunol. 2022, 4, 50. [Google Scholar]
- Wu, Y.; Zhang, J.; Feng, X.; Jiao, W. Omega-3 polyunsaturated fatty acids alleviate early brain injury after traumatic brain injury by inhibiting neuroinflammation and necroptosis. Transl. Neurosci. 2023, 14, 20220277. [Google Scholar] [CrossRef]
- Bakkar, N.-M.Z.; Ibeh, S.; AlZaim, I.; El-Yazbi, A.F.; Kobeissy, F. High-fat diets in traumatic brain injury: A ketogenic diet resolves what the Western diet messes up neuroinflammation and beyond. In Diet and Nutrition in Neurological Disorders; Elsevier: Amsterdam, The Netherlands, 2023; pp. 175–197. [Google Scholar]
- Dong, X.; Su, Y.; Luo, Z.; Li, C.; Gao, J.; Han, X.; Yao, S.; Wu, W.; Tian, L.; Bai, Y. Fecal microbiota transplantation alleviates cognitive impairment by improving gut microbiome composition and barrier function in male rats of traumatic brain injury following gas explosion. Front. Microbiol. 2024, 15, 1485936. [Google Scholar] [CrossRef] [PubMed]
- Hu, X.; Jin, H.; Yuan, S.; Ye, T.; Chen, Z.; Kong, Y.; Liu, J.; Xu, K.; Sun, J. Fecal microbiota transplantation inhibited neuroinflammation of traumatic brain injury in mice via regulating the gut–brain axis. Front. Cell. Infect. Microbiol. 2023, 13, 1254610. [Google Scholar] [CrossRef] [PubMed]
- Du, D.; Tang, W.; Zhou, C.; Sun, X.; Wei, Z.; Zhong, J.; Huang, Z. Fecal microbiota transplantation is a promising method to restore gut microbiota dysbiosis and relieve neurological deficits after traumatic brain injury. Oxidative Med. Cell. Longev. 2021, 2021, 5816837. [Google Scholar] [CrossRef] [PubMed]
- Davis IV, B.T.; Chen, Z.; Islam, M.B.; Timken, M.E.; Procissi, D.; Schwulst, S.J. Fecal microbiota transfer attenuates gut dysbiosis and functional deficits after traumatic brain injury. Shock 2022, 57, 251–259. [Google Scholar] [CrossRef]
- Ardizzone, A.; Bova, V.; Casili, G.; Filippone, A.; Lanza, M.; Repici, A.; Esposito, E.; Paterniti, I. bFGF-like Activity Supported Tissue Regeneration, Modulated Neuroinflammation, and Rebalanced Ca(2+) Homeostasis following Spinal Cord Injury. Int. J. Mol. Sci. 2023, 24, 14654. [Google Scholar] [CrossRef]
- Tang, H.; Gu, Y.; Jiang, L.; Zheng, G.; Pan, Z.; Jiang, X. The role of immune cells and associated immunological factors in the immune response to spinal cord injury. Front. Immunol. 2023, 13, 1070540. [Google Scholar] [CrossRef]
- Hasan, A.; Repici, A.; Capra, A.P.; Mannino, D.; Bova, V.; Catalfamo, A.; Campolo, M.; Paterniti, I.; Esposito, E.; Ardizzone, A. CCR1 antagonist as a potential modulator of inflammatory, autophagic, and apoptotic markers in spinal cord injury. Neuropharmacology 2025, 264, 110239. [Google Scholar] [CrossRef]
- Moura, M.M.; Monteiro, A.; Salgado, A.J.; Silva, N.A.; Monteiro, S. Disrupted autonomic pathways in spinal cord injury: Implications for the immune regulation. Neurobiol. Dis. 2024, 195, 106500. [Google Scholar] [CrossRef]
- de la Cruz-Castillo, E.; García-Vences, E. Beyond the Quality of Life in Bowel Dysfunction after Spinal Cord Injury: Approaches to the Consequences in Motility, Immune. Paraplegia 2021, 1, 127. [Google Scholar]
- Mou, Y.; Du, Y.; Zhou, L.; Yue, J.; Hu, X.; Liu, Y.; Chen, S.; Lin, X.; Zhang, G.; Xiao, H. Gut microbiota interact with the brain through systemic chronic inflammation: Implications on neuroinflammation, neurodegeneration, and aging. Front. Immunol. 2022, 13, 796288. [Google Scholar] [CrossRef]
- Chen, X.; Wang, B.; Al Mamun, A.; Du, K.; Wang, S.; Hu, Q.; Chen, X.; Lu, Y.; Du, A.; Wu, Y. Pectin-Zein-IPA nanoparticles promote functional recovery and alleviate neuroinflammation after spinal cord injury. J. Nanobiotechnol. 2025, 23, 152. [Google Scholar] [CrossRef] [PubMed]
- Wang, J.; Zhang, N.; Liu, H.-Z.; Wang, J.-L.; Zhang, Y.-B.; Su, D.-D.; Zhang, L.-M.; Li, B.-D.; Miao, H.-T.; Miao, J. Hydrogen Sulfide (H2S) Generated in the Colon Induces Neuropathic Pain by Activating Spinal NMDA Receptors in a Rodent Model of Chronic Constriction Injury. Neurochem. Res. 2025, 50, 90. [Google Scholar] [CrossRef] [PubMed]
- Hu, X.; Feng, J.; Lu, J.; Pang, R.; Zhang, A.; Liu, J.; Gou, X.; Bai, X.; Wang, J.; Chang, C. Effects of exoskeleton-assisted walking on bowel function in motor-complete spinal cord injury patients: Involvement of the brain–gut axis, a pilot study. Front. Neurosci. 2024, 18, 1395671. [Google Scholar] [CrossRef] [PubMed]
- Wu, Z.-J.; Zhao, Y.-Y.; Hao, S.-J.; Dong, B.-B.; Zheng, Y.-X.; Liu, B.; Li, J. Combining fecal 16 S rRNA sequencing and spinal cord metabolomics analysis to explain the modulatory effect of PPARα on neuropathic pain. Brain Res. Bull. 2024, 211, 110943. [Google Scholar] [CrossRef]
- Yang, X.; Zhangyi, Z.; Yu, A.; Zhou, Q.; Xia, A.; Qiu, J.; Cai, M.; Chu, X.; Li, L.; Feng, Z. GV-971 attenuates the progression of neuromyelitis optica in murine models and reverses alterations in gut microbiota and associated peripheral abnormalities. CNS Neurosci. Ther. 2024, 30, e14847. [Google Scholar] [CrossRef]
Intervention | Key Findings—Related Targets and Pathways | Model/Limitations | Effect on ALS | Ref. |
---|---|---|---|---|
Administration of galactooligosaccharide (GOS) to SOD1G93A transgenic mice, a widely used animal model of ALS | GOS treatment extended survival, preserved spinal motor neurons, reduced neuroinflammation through decrease activation of microglia and astrocytes, and mitigated oxidative stress in SOD1G93A ALS mice | Mouse Model/Mouse model specificity, Gut–CNS link unclear | The findings suggest that GOS has neuroprotective effects in the SOD1G93A mouse model of ALS, potentially through its anti-inflammatory and antioxidant properties, leading to delayed disease progression and extended survival | [117] |
Administration of Lacticaseibacillus rhamnosus HA-114 or its fatty acid extracts to Caenorhabditis elegans models of neurodegenerative diseases, including ALS HD | HA-114 supplementation reduced age-dependent paralysis and motor neuron degeneration in C. elegans models expressing mutant human proteins associated with ALS (FUSS57Δ and TDP-43A315T) via fatty acid-mediated neuroprotection, requiring mitochondrial β-oxidation genes such as acdh-1 (ACADSB), kat-1 (ACAT1), and elo-6 (ELOVL3/6) and restoring neuronal lipid and energy homeostasis through a carnitine-independent mechanism | Caenorhabditis elegans models/non-mammalian model, Limited translational relevance, Simplified nervous system, No human data, Lack of Specialized Tissues in C. elegans | The study suggests that fatty acids produced by L. rhamnosus HA-114 can mitigate neurodegenerative phenotypes in C. elegans models by enhancing mitochondrial function and energy metabolism, highlighting a potential therapeutic avenue for age-related neurodegenerative diseases | [118] |
Administration of the probiotic formulation AJ3 (Al-Pro) to assess its impact on immune cell activity in the context of ALS | AJ3 treatment reduced elevated IFN-γ levels secreted by natural killer (NK) and CD8⁺ T cells, contributing to immune-mediated damage in the central nervous system and peripheral organs and shifted the IFN-γ/IL-10 ratio toward an anti-inflammatory profile in ALS, outperforming other probiotic formulations in suppressing cytotoxic immune responses | ALS patient-derived samples And in vitro immune cell assays/Lacks in vivo validation, Human sample variability, Mechanism not fully explored, Translation to ALS uncertain, Patient heterogeneity considerations | The findings suggest that AJ3 may alleviate autoimmunity in ALS by modulating the activity of cytotoxic immune effectors, thereby potentially preventing motor neuron death and disease progression | [119] |
Administration of butyrate (a short-chain fatty acid) or the probiotic formulation VSL#3 to TDP-43A315T mutant mice, starting at 9 weeks of age | Treatment improved motor and gut function, restored barrier integrity by increase expression of tight junction proteins (ZO-1 and Claudin-5) and α-smooth muscle actin (α-SMA) in the colon, spinal cord, and brain, reduced mutant TDP-43 aggregation and glial activation, modulated gut microbiota toward butyrate production, and attenuated systemic inflammation by reducing serum levels of inflammatory cytokines (IL-6, IL-17, and IFN-γ) and LPS | Transgenic mice model/using a specific mouse model which limits the generalizability of the findings, lacking long-term follow-up, lacking human validation | The interventions delayed disease onset and progression in TDP-43 mutant mice by restoring gut and blood–brain barrier integrity, reducing neuroinflammation and protein aggregation, and modulating gut microbiota composition | [120] |
Administration of 2% sodium butyrate (a natural bacterial product) in drinking water to G93A transgenic mice, a model for human ALS | Butyrate treatment in ALS models restored gut microbiota by increasing beneficial butyrate-producing bacteria, such as Butyrivibrio fibrisolvens, Bacteroides, Odoribacter, and Eubacterium, improved intestinal barrier integrity through upregulation of tight junction protein ZO-1, and normalized Paneth cell function by enhancing antimicrobial peptide-like lysozyme 1 and defensin 5 alpha expression. Additionally, it reduced the aggregation of mutant SOD1G93A protein in both intestinal tissues and human epithelial cells, highlighting its therapeutic potential through gut–CNS axis modulation | Transgenic mice model/Single genetic mutation, Lack of mechanistic studies, Focused mainly on butyrate metabolite | Butyrate treatment in ALS models delayed disease onset, extended survival by an average of 38 days, and preserved neuromuscular function, indicating systemic benefits mediated through gut microbiota modulation and gut-CNS axis interaction | [121] |
Administration of specific metabolites or compounds, including butyrate, lanthionine ketimine ethyl ester (LKE), and methionine sulfoximine (MSO), to ALS mouse models | Butyrate, LKE, and MSO each demonstrated therapeutic benefits in SOD1G93A ALS mouse models by targeting distinct pathological mechanisms: butyrate reduced systemic inflammation by reducing serum levels of IL-17 and LPS, and restored gut-CNS communication; LKE promoted neuroprotection and functional recovery; and MSO mitigated excitotoxicity by lowering glutamate levels, thereby delaying disease onset and extending survival | Transgenic mice model/Animal model limitations, focus on specific metabolites, short treatment duration, small sample size | Administration of specific metabolites or compounds, including butyrate, lanthionine ketimine ethyl ester (LKE), and methionine sulfoximine (MSO), to ALS mouse models | [122] |
Administration of sodium phenylbutyrate (SPB), a histone deacetylase (HDAC) inhibitor, to SOD1G93A transgenic mice, a commonly used ALS model | SPB treatment in ALS mice significantly prolonged survival, improved motor function, and delayed disease onset. Mechanistically, SPB enhanced histone acetylation and upregulated anti-apoptotic genes (Bcl-2, Bcl-xL), indicating epigenetic regulation as a key mode of neuroprotection | Transgenic mice model/Mouse model may not fully replicate human ALS, the study did not assess long-term toxicity, adverse effects, Lack of dosing optimization. | The study demonstrates that SPB exerts neuroprotective effects in ALS by modulating epigenetic markers and gene expression, leading to delayed disease progression and extended survival in the mouse model | [123] |
Combined administration of riluzole and sodium phenylbutyrate (NaPB) to SOD1G93A transgenic mice, a commonly used ALS model | Combination therapy in ALS models extended survival by 21.5%, outperforming riluzole or NaPB alone. It improved neuropathology by reducing spinal cord atrophy, neuronal death, and astrogliosis. Mechanistically, it enhanced histone H4 acetylation and NF-κB p50 nuclear translocation, promoting anti-apoptotic gene expression through epigenetic modulation | Transgenic mice model/Focus on Specific Mechanism, clinical trials limitations, Long-term safety and efficacy requires further validation | The combined therapy demonstrated a synergistic neuroprotective effect, leading to prolonged survival, improved motor function, and reduced neurodegeneration in the ALS mouse model | [124] |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2025 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Hasan, A.; Scuderi, S.A.; Capra, A.P.; Giosa, D.; Bonomo, A.; Ardizzone, A.; Esposito, E. An Updated and Comprehensive Review Exploring the Gut–Brain Axis in Neurodegenerative Disorders and Neurotraumas: Implications for Therapeutic Strategies. Brain Sci. 2025, 15, 654. https://doi.org/10.3390/brainsci15060654
Hasan A, Scuderi SA, Capra AP, Giosa D, Bonomo A, Ardizzone A, Esposito E. An Updated and Comprehensive Review Exploring the Gut–Brain Axis in Neurodegenerative Disorders and Neurotraumas: Implications for Therapeutic Strategies. Brain Sciences. 2025; 15(6):654. https://doi.org/10.3390/brainsci15060654
Chicago/Turabian StyleHasan, Ahmed, Sarah Adriana Scuderi, Anna Paola Capra, Domenico Giosa, Andrea Bonomo, Alessio Ardizzone, and Emanuela Esposito. 2025. "An Updated and Comprehensive Review Exploring the Gut–Brain Axis in Neurodegenerative Disorders and Neurotraumas: Implications for Therapeutic Strategies" Brain Sciences 15, no. 6: 654. https://doi.org/10.3390/brainsci15060654
APA StyleHasan, A., Scuderi, S. A., Capra, A. P., Giosa, D., Bonomo, A., Ardizzone, A., & Esposito, E. (2025). An Updated and Comprehensive Review Exploring the Gut–Brain Axis in Neurodegenerative Disorders and Neurotraumas: Implications for Therapeutic Strategies. Brain Sciences, 15(6), 654. https://doi.org/10.3390/brainsci15060654