Next Article in Journal
Yoda1 Inhibits TGFβ-Induced Cardiac Fibroblast Activation via a BRD4-Dependent Pathway
Previous Article in Journal
Emerging Biomarker Potential of Extracellular Vesicle-Enclosed MicroRNAs for Liver Fibrosis Detection
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Therapeutic Horizons: Gut Microbiome, Neuroinflammation, and Epigenetics in Neuropsychiatric Disorders

by
Shabnam Nohesara
1,†,
Hamid Mostafavi Abdolmaleky
1,2,†,
Ahmad Pirani
3 and
Sam Thiagalingam
1,4,*
1
Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
2
Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
3
Mental Health Research Center, Psychosocial Health Research Institute, Iran University of Medical Sciences, Tehran 14535, Iran
4
Department of Pathology & Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Cells 2025, 14(13), 1027; https://doi.org/10.3390/cells14131027
Submission received: 30 May 2025 / Revised: 29 June 2025 / Accepted: 3 July 2025 / Published: 4 July 2025

Abstract

Neuroinflammation is a hallmark of many neuropsychiatric disorders (NPD), which are among the leading causes of disability worldwide. Emerging evidence highlights the significant role of the gut microbiota (GM)–immune system–brain axis in neuroinflammation and the pathogenesis of NPD, primarily through epigenetic mechanisms. Gut microbes and their metabolites influence immune cell activity and brain function, thereby contributing to neuroinflammation and the development and progression of NPD. The enteric nervous system, the autonomic nervous system, neuroendocrine signaling, and the immune system all participate in bidirectional communication between the gut and the brain. Importantly, the interaction of each of these systems with the GM influences epigenetic pathways. Here, we first explore the intricate relationship among intestinal microbes, microbial metabolites, and immune cell activity, with a focus on epigenetic mechanisms involved in NPD pathogenesis. Next, we provide background information on the association between inflammation and epigenetic aberrations in the context of NPD. Additionally, we review emerging therapeutic strategies—such as prebiotics, probiotics, methyl-rich diets, ketogenic diet, and medications—that may modulate the GM–immune system–brain axis via epigenetic regulation for the prevention or treatment of NPD. Finally, we discuss the challenges and future directions in investigating the critical role of this axis in mental health.

1. Introduction

Neuropsychiatric disorders (NPD) are brain diseases with a complex nature and great degree of genetic heterogeneity that influence millions of people around the world [1].
NPD are conditions that influence both the nervous system and mental health by altering a person’s thoughts, emotions, and behaviors [2]. NPD include neurodevelopmental disorders (e.g., autism spectrum disorder (ASD)), neurodegenerative diseases (e.g., Parkinson’s disease (PD), Alzheimer’s disease (AD)), and psychiatric disorders (e.g., bipolar disorder (BD), schizophrenia (SCZ)) that are commonly characterized by cognitive impairment, behavioral abnormalities, and disturbed emotional and mental functioning [3,4]. Neurodegenerative diseases such as AD and PD are characterized by the progressive loss of neuronal cells and disturbances in their function which in turn give rise to psychological disability and cognitive impairment [5]. However, mental illnesses are associated with dysfunctions in biological factors such as neuromodulators and neurotransmitters that primarily influence emotion, cognition, and behavior [6,7]. Neurodegenerative diseases typically develop over time, and the worsening of symptoms is associated with the disease progression, while mental illnesses are chronic or episodic, with different degrees of severity and duration.
The dysfunction of immune system has been known to be a key player in both the pathogenesis and the treatment outcomes of NPD [8]. The severity of clinical symptoms in NPD is linked to both peripheral and central inflammation [9]. Higher inflammatory responses in patients are connected to poorer response to antipsychotics [10]. Some events of the immune response such as chronic activation of macrophages and T cells, imbalance of T helper cells (Th1 and Th2), and elevated activation of microglia (the brain macrophages) during NPD, lead to impaired neurotransmitter release, derangements in synaptic plasticity and cortisol levels, which in turn may increase the burden of brain diseases [11]. The relationship between NPD and inflammation can be mediated by epigenetic alterations [12]. Accumulating evidence have indicated that aberrations in epigenetic mechanisms, such as DNA methylation, histone modifications, and non-coding RNAs (e.g., microRNAs (miRNAs) and long non-coding RNAs (lncRNA)), contribute to dysregulation of gene expression and determination of immune cell fate [13,14]. For example, the activity of histone deacetylases (HDACs) can regulate macrophage differentiation without lymphocyte stimuli and reduces macrophage plasticity, which further facilitates the pro-inflammatory phenotype [15]. Environmental factors such as lifestyle, diet, gut dysbiosis, and traumatic experiences have potential roles in inducing neuroinflammation and triggering epigenetic shifts, activation or suppression of pertinent genes associated with the onset and/or development of NPD [16,17].
One of the important determinants of health and disease in human is trillions of microorganisms such as bacteria, viruses, fungi, and other microorganisms that are inhabitant in different body organs specially the gastrointestinal (GI) tract [18,19]. Several studies demonstrated that gut bacteria-host interplays determine immune cell maturation, phenotype, and even migration and play a key role in neuroinflammation, the secretion of cytokines, chemokines, neurotransmitters, and microbial by-products [20]. For example, Lopizzo et al. found a specific inflammatory pattern in various brain regions, including dorsal and ventral hippocampus, that was linked to alterations in the gut microbiota (GM) composition [21]. Apart from the GM, altered levels of its metabolites are also connected to the immune-inflammatory response in the central nervous system (CNS) in NPD. For example, short-chain fatty acids (SCFAs) play a key role in regulating microglia maturation, and altered fecal SCFAs concentrations are connected to subclinical inflammation and impaired cognitive function in SCZ subjects [22,23]. Chen et al., also found a significant correlation between the abundance of pro-inflammatory bacteria, including Clostridiales bacterium NK3B98 and Ruminococcus sp. AM07-15 and altered fecal and plasma levels of SCFAs, including acetate, propionate, and butyrate in PD [24]. In AD, a reduction in the abundance of butyrate-producing bacteria like Faecalibacterium and raise in the abundance of lactate-producing genera contributed to immune disturbances and heightened neuroinflammation in the host [25].
Accumulating evidence linking the GM-induced inflammation or neuroinflammation to NPD through the GM–immune–brain axis has paved the way for developing therapies targeting this axis. This review provides an overview of the interplays between the GM, the immune system, and the brain via epigenetic aberrations in NPD. Moreover, various therapeutic interventions, such as prebiotics, probiotics, methyl-rich diets, and medications are highlighted for their role in modulating the GM–immune system–brain axis and alleviating these disorders.

2. Neuroinflammation in the Pathogenesis of Neuropsychiatric Disorders

One of the important etiological factors in NPD is neuroinflammation that may be caused by diverse pathogens or chronic inflammatory diseases [26]. During NPD pathogenesis, perturbations of the blood–brain barrier (BBB) facilitate the entry of peripheral immune cells and inflammatory molecules into the brain. In AD, inflammatory mediators change synaptic function and stimulate neurons and glial cells toward more Aβ production, which in turn lead to cognitive impairment [27]. In PD, dementia risk is connected to neuroinflammation. For example, Kouli et al. found that PD patients with higher dementia risk possess higher inflammation and/or tau accumulation prior to significant decline in cognitive performance [28]. In BD, higher amounts of IL-6 and IL-1β in the hippocampus are associated with more severe neuropsychiatric symptoms [29].
As immune cells and their inflammatory molecules are key players in the pathogenesis of NPD, there are significant changes in the expression of inflammatory genes. Evidence suggests that neuroinflammation in the pathogenesis of NPD is associated with epigenetic and transcriptional alterations in the peripheral immune system. For example, Ramakrishnan et al. found that in AD peripheral immune cells possessed more open chromatin and CD8 T cells exhibited a chromatin modification relevant to CXCR3 expression [30]. Their results also showed that in AD monocytes possessed APOE genotype-specific chromatin modifications, and genes relevant to sporadic AD exhibit chromatin-level changes [30]. Murphy et al. found elevated leukocyte expression of the NF-κB-activating receptors, TLR4 and TNFR2, and the NF-κB subunit, RelB, as well as reduced leukocyte expression of IKKβ and NIK mRNAs in SCZ subjects with higher levels of inflammation [31]. Additionally, the initiation of neuroinflammatory events in autistic BTBR mice is connected to elevation of alarmin S100A9 in the microglia [32]. A recent study indicated that induced pluripotent stem cells (iPSC)-derived astrocytes from patients with autism also exhibited overexpression of immune relevant genes, such as TGFB1, TGFB2, and IL6 that was associated with DNA hypomethylation of TGFB2, IL6, and TNFA gene promoter regions [33]. Megagiannis et al. reported that ASD-associated CHD8 in astrocytes play an important role in the injury-induced reactive gliosis by remodeling chromatin accessibility and alerting gene expression; therefore, targeting CHD8 may represent a promising strategy to alleviate neuroinflammation [34]. Moreover, neuroinflammation-induced depression-like behaviors in mice has been linked to reduced hydroxymethylation of BDNF gene and elevated expression of Iba-1, a marker for microglia activation, in the hippocampus region [35]. Additional studies on altered immune cell composition or inflammatory molecules in specific NPD, along with their consequences, are summarized in Table 1.

3. The Gut Microbiome May Orchestrate the Function and Behavior of Immune System

Several lines of evidence indicates that the GM is a master regulator in modulating CNS inflammation, and disruption in GM composition in NPD is linked to substantial alterations in gut anti-inflammatory and pro-inflammatory bacterial genera [46]. As an interesting example, a subset of astrocytes referred to as “gut-licensed IFNγ+ NK cells drive LAMP1+ TRAIL+ anti-inflammatory astrocytes” in mice can prevent CNS inflammation by the induction of T cell apoptosis mediated by TRAIL–DR5 signaling [47]. While, GM modulates the production of IFNγ (an anti-inflammatory cytokine) by meningeal NK cells, IFNγ induces TRAIL protein expression (encoded by TNFSF10 gene) in astrocytes. However in inflammation, T cells and microglia produce some molecules, which suppress astrocytic expression of TRAIL protein [47]. In other studies, it appears that lactic acid-producing bacteria may play a role in regulation of the function of other immune cells, specially macrophages, since lactate has shown capability for supporting a metabolic–epigenetic link in macrophage polarization [48]. Ling et al. observed that structural dysbiosis in SCZ patients is relevant to the compositional changes in several genera involved in producing immunomodulatory metabolites, which further elevated the amounts of pro-inflammatory cytokines, such as IL-1β and reduced levels of anti-inflammatory cytokines, like IFN-γ [49]. Huang et al. examined the compositional changes in GM and its relation to inflammation in depressed BD patients and found reduced levels of some bacteria (Butyricicoccus, Lachnospiraceae incertae sedis, and Dorea) involved in triggering the secretion of anti-inflammatory cytokines such as IL-4, IL-10, and IL-11 [50]. Lower levels of the anti-inflammatory butyrate-producing bacteria was also reported in untreated BD patients versus healthy controls [51]. Similarly, changes in butyrate-producing genera have been found in SCZ patients [52]. The authors mentioned that patients with SCZ exhibited the depletion of anti-inflammatory butyrate-producing genera, and elevated abundance of some opportunistic bacteria genera [52]. PD is also associated with decreased abundance of anti-inflammatory butyrate-producing bacteria, such as Roseburia, Faecalibacterium, Blautia, and Lachnospira and increased abundance of pro-inflammatory genera such as Streptococcus [53]. Figure 1 depicts how disruption in GM composition due to gut dysbiosis generally contribute to increased neuroinflammation, which in turn promotes the development of NPD.
More studies addressing the relationship between pro- and anti-inflammatory gut bacteria and different NPD are presented in Table 2.
In addition to microbial communities in the GI, metabolites produced by commensal gut flora may mitigate pathogenic activities of microglia and astrocytes by controlling microglia activation and regulating astrocytic transcriptional program via a mechanism mediated by the aryl hydrocarbon receptor [68]. GM-derived metabolites may also play a powerful role in the modulation of the function of immune cells by orchestrating epigenetic modifications. For example, the anti-inflammatory effects of butyrate is associated with the modulation of Mψ (M1/M2) polarization by decreasing the activity of HDAC1 and negative regulation of miR-155 [69]. Moreover, the exogenous acetate produced by some bacteria can contribute to rescuing defective M2 polarization and histone acetylation following mitochondrial pyruvate carrier 1 (MPC1) inhibition or in adenosine triphosphate–citrate lyase (ACLY) deficiency [48]. In another study, both patients with SCZ and major depressive disorder (MDD) exhibited reduced levels of acetic acid, isobutyric acid, and propionic acid, which were associated with higher Positive and Negative Syndrome Scale (PANSS) and Hamilton Depression Rating Scale (HAMD) scores, increased levels of pro-inflammatory cytokines, and reduced levels of anti-inflammatory cytokines (IL-37 and IL-10) [70]

4. Epigenetic Alterations Linked to Immune System and Immune-Relevant Genes in Neuropsychiatric Disorders

4.1. DNA Methylation Patterns

Patients with NPD exhibit noticeable changes in DNA methylation patterns that may affect immune cell composition. For example, Liu et al. reported that the most evident biological functions of differentially methylated CpGs in SCZ subjects are related to inflammatory response of CD224, LAX1, TXK, PRF1, CD7, MPG, and MPO genes that are responsible for activations of T cells, B cells, and natural killer cells [71].
Shindo et al. reported that subjects with untreated MDD exhibited decreased levels of natural killer cells along with epigenetic aging acceleration in HannumAge and GrimAge (two epigenetic clocks—biomarkers representing 71 and 1030 CpG sites, respectively) in blood samples [72]. In a study by Luo et al. a subtype of SCZ consisting almost 40% of patients with higher symptom severity, worst cognitive function, and gray matter thickening exhibited extensive DNA methylation changes among genes linked to immune cell activity, an increase in the proportion of neutrophils, and a decrease in the proportion of lymphocytes [73]. Additionally, they reported that a subgroup of never-treated first-episode SCZ patients (47.5%) had also widespread methylation changes in genes of immune cell activity and increased proportion of neutrophils in blood samples [74]. Table 3 summarizes more studies about associations between altered methylation patterns in immune-relevant genes and developing NPD in humans.

4.2. Histone Modifications

Histone modifications can also contribute to controlling gene expression and regulating inflammatory responses during NPD pathogeneses. The activation of microglia (i.e., inflammatory microglia) confers a sequential chain of events, including hyperexpression of HDAC2 in neurons, reducing amounts of histone acetylation, and inhibiting the transcription of BDNF and c-Fos, which results in memory impairment [88]. The activated microglia can also arrest Nrf2-inducible anti-oxidant defense in astrocytes, which is connected to reduction in histone acetylation [89]. Additionally, the hypothalamic and hippocampal transcription of pro-inflammatory targets by lipopolysaccharide (LPS) in neurons and reactive microglia is mediated by increasing the level of H3S10phK14ac (H3 phosphoserine10 and lysine 14 acetylation) in rat brain [90]. Rodriguez-Zas et al. also reported that inflammation-associated depression-like behaviors in mice are linked to derangements in microglia histone acetylation [91]. Additional examples related to associations between altered histone modifications in immune-relevant genes linked to NPD are provided in Table 4.

4.3. MicroRNAs (miRNAs)

miRNAs, the endogenous and small non-coding RNA molecules (21–25 nucleotides in length), are among other main regulators of gene expression at post-transcriptional level through incomplete pairing with the 3′-untranslated regions (3′-UTRs) of target mRNAs [92]. Numerous inflammatory miRNAs like miR-155 and let-7 play key roles in stimulating neuroinflammation by binding to toll-like receptors (TLRs), activation of microglia and astrocytes, and release of inflammatory mediators [93,94,95]. For example, miR-145-5p is a negative regulator of astrocyte proliferation, and its down-regulation increases smad3 activity and subsequently astrocyte proliferation [96]. There is also a report indicating that elevated levels of miR-223 in the orbitofrontal cortex of SCZ and BD patients are positively and negatively associated with inflammatory and GABAergic gene expression, respectively [97]. Furthermore, Kaurani et al. reported that reduction in the miR-99b-5p level in mice could cause both SCZ-like phenotypes and inflammatory processes, which are relevant to synaptic pruning by microglia [98]. Table 4 summarizes additional studies on the associations between altered miRNAs in immune-relevant genes involved in the pathogenesis of NPD.
Table 4. Altered miRNAs and histone modifications of immune-relevant genes in specific neuropsychiatric conditions in humans or animal studies.
Table 4. Altered miRNAs and histone modifications of immune-relevant genes in specific neuropsychiatric conditions in humans or animal studies.
Neuropsychiatric Disorders/Study SubjectsEpigenetic Study/SamplesImmune-Relevant GenesKey FindingsRef.
SCZ/humanHistone acetylation/peripheral blood mononuclear cells (PBMCs)IL-6 and IFN-γHistone H4 hypoacetylation in PBMCs decreases IL-6 and IFN-γ following 90 days exercise in SCZ vs. day 0.[99]
MDD/humanHistone acetylation/dentate gyrusISG15, IFI44L, IFI6, NR4A1/Nur-77, GABBR1, CCL2/MCP-1, KANSL1Differential expression of 30 genes involved in histone acetylation and inflammation (e.g., CCL2 and KANSL1 in MDD)[100]
MDD/humanHistone methylation/peripheral blood cells (PBCs)TNFAIP3, TLR4, TNIP2, miR-146a, miR-155Lower histone 3 lysine 4 tri-methylation (H3K4me3) levels at the promoters of TNFAIP3, TLR4, TNIP2, miR-146a, and miR-155 in MDD[101]
Alzheimer’s disease (AD)/APP/PS1 miceHistone acetylation/Entorhinal CortexCREB, IL-1β, and TNF-α and NF-kBReduced H3K9K14 acetylation, increasing Aβ deposition, microglia and astrocytes activation, and inflammatory factors via the CREB/BDNF/NF-kB pathway[102]
Postpartum psychosis/humanmiR-146a and miR-212/monocytesADAM17, EGR3, IRAK2, PTGS2, CXCL2, and PTGS2Reduced miR-146a expression in monocytes diminishes natural T regulator cells; reduced expression of miR-212 elevated Adrenomedulin, reduced IL-6, and increased Th2 cells[103]
SCZ/humanmiR-337-3p, miR-127-5p, miR-206, miR-1185-1-3p/human iPSC-derived astrocytes from SCZ patientsIL-1β, LAMTOR4, IL23R, ERBB3, ERBB2, and IRAK1Lower expression of miR-337-3p, miR-127-5p, miR-206, and miR-1185-1-3p in SCZ astrocytes[104]
SCZ/humanhsa-miR-16-5p, hsa-miR-186-5p, hsa-miR-19a-3p, and hsa-miR-19b-3p/blood IL-1β, IL-6, and TNFαHigher PANSS scores is linked to down-regulation of four miRNAs that negatively regulate pro-inflammatory cytokines[105]
Bipolar Disorder (BD)/humanhsa-miR-34a-5p, hsa-miR-152-3p hsa-miR-574-3p, hsa-miR-3128 and hsa-miR-3201/Lymphoblastoid cell linesNF-κB, STAT3, and TNF46 up-regulated and 31 down-regulated miRNAs with immune-related functions in responders vs. non-responders to Lithium in BD[106]
MDD/humanlet-7e, miR-21-5p miR-145, miR-223, miR-146a, and miR-155/PBMCs and monocytesTLR4Lower levels of let-7e, miR-146a, and miR-155 in PBMCs and miR-146a and miR-155 in monocytes in MDD vs. controls[107]
MDD/humanmiRNA-144-5p/plasmaCXCL6, STAMPB, CXCL1, CXCL5, IL-7, MCP-4, MCP-2, MCP_1, MMP-1, IL-8, and IL-18An inverse correlation between miR-144-5p and some inflammatory proteins[108]
MDD/humanmiR-342, miR-146a, and miR-155/PBMCs and plasmaTNF-α, IL-6, and CCL2 (plasma)Positive correlation between miR-342 expression and TNF-α level[109]
Parkinson’s disease (PD)/micemiR-335/serum samplesLRRK2Mitigating neuroinflammation by miR-335 via targeting LRRK2[110]
Autism/micemiRNA profiling/prefrontal cortex (PFC)NF-κB, IRAK1, and TLR7Neuroinflammation is linked to miR-146a, let- 7b, and miR-592[111]
AD/miceSeveral miRNAs/PFC and the hippocampusCst7 and GfapNeuroinflammation is linked to miR-124-3p, miR-125b-5p, miR-21-5p, miR-146a-5p, and miR-155-5p[112]

5. Therapeutic Approach in Neuroinflammatory Diseases

Considering interconnected links between GM, neuroinflammation, and epigenetic alterations in NPD, there are opportunities to target these key interlinked players for the treatment of these diseases. Potential remedies may include prebiotics, probiotics (i.e., nutrition that tailor GM), postbiotics (products of GM) to alleviate inflammation and epigenetic alterations, in addition to psychiatric drugs, which in part influence GM, neuroinflammation and the epigenome.

5.1. Probiotics

Probiotics are microorganisms capable of exerting health benefits when are utilized or consumed in sufficient amounts [113]. Anti-inflammatory effect of probiotics may be associated with enhancing Na+-dependent absorption of luminal butyrate, reshaping GM composition, and altering the expression of genes involved in immune reactions in the prefrontal cortex [114,115]. In a study published in the early 2010 s, the beneficial effects of SCFA-producing bacteria Lactobacillus helveticus R0052 and Bifidobacterium longum R0175 on improving signs of post-myocardial infarction depression in rats were linked to a significant reduction in circulating IL-1β levels and the restoration of intestinal barrier integrity [116]. Lactobacillus plantarum MTCC 9510 supplementation could also mitigate depressive-like behaviors in mice exposed to chronic unpredictable mild stress (CUMS) by reducing serum TNF-α and LPS levels, elevating the abundance of Lactobacillus sp. decreasing the abundance of Enterobacteriaceae, and increasing the production of acetate and butyrate [117].
In another study, researchers served Clostridium butyricum, a butyrate-producing, spore-forming anaerobic bacterium, as a probiotic to examine its antidepressant-like effect in a mouse model of inflammatory depression [66]. Their results indicated that this probiotic could improve depressive-like behaviors by reducing inflammatory factors and normalizing the GM composition [66]. Parra et al. reported that oral administration of a combination of two probiotics, including Lactobacillus rhamnosus GG and Bifidobacterium animalis ssp. lactis Bb12 once daily for 15 days in an inflammatory PD model in rats could improve symptoms by decreasing microglial activation [118]. Valvaikar et al. examined protective effect of probiotic strain, B. breve Bif11, in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD model in rats and their findings showed improvements in the cognitive and motor abnormalities, reductions in pro-inflammatory mediators, oxidative and nitrosative stress in the mid brain of MPTP-lesioned rats by elevating the concentration of epigenetic metabolites, including butyric acid, propionic acid, and acetic acid [119]. Commercial probiotic VSL#3®, including eight bacterial strains (including, different Lactobacillus and Bifidobacterium as well as Streptococcus thermophilus BT01) could ameliorate autistic-like behaviors in a prenatal valproic acid-induced rodent model of autism by elevating serum concentration of the IL-10 and reducing IL-6 [120]. In a clinical study by Reininghaus et al., a four-week probiotic plus biotin supplementation could improve symptoms in MDD patients by increasing the abundance of butyrate-producing bacteria, including Ruminococcus gauvreauii and Coprococcus 3 and by reducing inflammation [121]. In another clinical study a 12-week intervention with a multi-strain probiotic (containing Lactobacillus plantarum PL-02, Bifidobacterium longum subsp. infantis BLI-02, B. animalis subsp. lactis CP-9, B. bifidum VDD088, and B. breve Bv-889) resulted in a 36% increase in BDNF, and anti-oxidant SOD levels but decrease in IL-1β levels in AD [122]. A different recent clinical study indicated a 12-week intervention with two single-strain probiotics (Lacticaseibacillus rhamnosus HA-114 or Bifidobacterium longum R0175) in subjects with mild and moderate AD could improve the serum concentration of GSH, IL-10, and reduce the serum concentration of 8-OHdG, MDA, TNF-α, and IL-6, possibly through producing epigenetic metabolites like butyric acid and propionic acid [123].

5.2. Prebiotics/Postbiotics

Prebiotics are ingredients in food that enhance growth or activity of commensal bacteria [124]. Prebiotics have demonstrated ability for exerting protective effect by reducing mucosal inflammation, enhancing the colonic production of SCFAs involved in mitigating epigenetic aberrations, and regulating the GM via SCFA-induced inhibition of M1 and activation of M2 macrophages [125,126,127,128]. In another study Guo et al. reported that the anti-inflammatory effect of inulin (a soluble dietary fiber) on mice with SCZ phenotype is associated with enhancing intestinal integrity via elevating the expression of tight junction proteins, diminishing inflammatory cytokines, increasing BDNF levels, and raising the abundance of Lactobacillus and Bifidobacterium. These effects are due to the production of anti-inflammatory metabolites, such as butyrate, which modulate the epigenome [129]. Inulin has also been shown to improve behavioral abnormalities in chronically stressed mice by enhancing the production of SCFAs, mitigating CUMS-induced increases in BBB permeability, preventing LPS penetration into the brain, and regulating TLR4/MyD88/NF-κB signaling pathway to attenuate inflammatory responses [130]. Additionally, a 10-week oral natural prebiotics supplementation, including topinambur powder and chicory root inulin could improve anxiety-depressive behaviors, cognitive disorders and dysbiosis in mice exposed to CUMS by elevating the number of bacteria, such as Ruminococcaceae UCG-014, Odoribacter, Roseburia, Anaerotruncus, Intestinimonas, Lachnospiraceae ASF 356, and Clostridielesvadin BB60 involved in the production of metabolites with epigenetic effects [131]. Dietary inulin could also improve depressive and anxiety-like behaviors in constipation induced depression in mice by suppressing neuroinflammation, hampering synaptic ultrastructure damage, reducing the abundance of Muribacalum and Melaminabacteria, and elevating the concentration of different SCFAs like acetate, propionate, and butyrate in the feces [132].
In a recent human study Buchanan et al. reported that a 10-day oligofructose-enriched inulin treatment in SCZ patients could increase plasma butyrate level, which was associated with reduced inflammation [133]. Treatment with Zhi-Zi-Chi decoctions (ZZCD), consisting of Gardeniae Fructus and Semen sojae praeparatum was shown to improve depressive-like behaviors in rats exposed to a depression model of CUMS by increasing the abundance of anti-inflammatory bacteria, diminishing inflammatory and tryptophan-metabolizing bacteria, reducing pro-inflammatory cytokines, and restoring butyrate in the cecal content [134]. Another animal study examined the effect of a prebiotic, including Fructooligosaccharides (FOS) and Galactooligosaccharides (GOS) on inflammatory response and depressive and anxiety-like behaviors in a mouse model fed a high-fat diet (HFD) [135]. The results showed that treatment with FOS and GOS could improve depression and anxiety-like behavior by elevating the levels of acetate-producing bacteria (B. acidifaciens and B. dorei), increasing acetate and GPR43 concentrations in the brain, and subsequently ameliorating chronic peripheral and central inflammation [135]. Shu et al. reported that Xiaoyaosan polysaccharide alleviated depression-like behaviors by modulating the diversity of intestinal butyrate-producing bacteria diversity, including Roseburia sp. and Eubacterium sp. [136]. Another study also showed that antidepressant-like effects of Xiaoyaosan polysaccharides are linked to enriching the diversity and elevating the abundance of the butyrate-producing bacteria Roseburia sp. and Eubacterium sp. [137]. Additionally, raspberry ketone has been shown to reduce gut barrier dysfunction and hamper LPS-induced depression-like behaviors in mice by suppressing TLR-4/NF-κB signaling pathway involving the gut–brain axis and increasing the production of SCFAs [138]. In a valproic acid-induced mouse model of autism, a diet composed of prebiotic fibers, namely, GOS/FOS, could modulate peripheral immune homeostasis, attenuate neuroinflammation in the cerebellum, and improve deficits in social behavior and cognition by improving intestinal integrity, reshaping GM, and restoring the cecal concentrations of SCFA like propionic acid [139]. Sarti et al. also found that the symbiotic treatment with (a multi-extract of fibers and plant complexes, containing inulin/fruit-oligosaccharides) and probiotics (a 50%–50% mixture of Lactobacillus rhamnosus and Lactobacillus paracasei) could attenuate pathological features of AD, including the neuronal degeneration, Aβ deposition and neuritic plaques, and the shrinkage in the cortex of APP/PS1 mice by reducing GFAP expression and microglia activation [140].
In a clinical study conducted by Bedarf et al., a 4-week diet rich in dietary fiber, and the intake of Lactulose prebiotic, could improve PD-dependent gastrointestinal symptoms by elevating Bifidobacteria spp. and restoring epigenetic metabolites such as butyrate and propionate [141].
SCFAs are considered a class of postbiotics. These metabolic byproducts are produced by gut bacteria during the fermentation of dietary fibers, and act as master regulators in host health [142,143]. In the brain, SCFAs can exert anti-inflammatory effects via two major cellular mechanism [144]. The first mechanism involves SCFAs binding to G protein-coupled receptors (GPRs), increasing their activity, and thereby contributing to the maintenance of intestinal immune hemostasis. The second mechanism is the inhibition of HDAC activity, which leads to the suppression of inflammatory cytokines expression in the CNS [145]. Butyrate, a major SCFA produced by the GM through the fermentation of insoluble fibers has been shown to rectify the imbalance in microglia polarization (M1/M2), enhance the expression of intestinal tight junction protein occluding, maintain epithelium barrier integrity, suppress oxidative stress, and diminish LPS-induced inflammation in primary microglia, and neuronal cells in the hippocampus region, and neuronal cells co-cultured with microglial or astrocytes in rats [146,147,148,149,150]. Moreover, butyrate may exert its anti-inflammatory effect by promoting a balance between anti-inflammatory Treg and pro-inflammatory T helper 17 (Th17) cells [151]. As butyrate is involved in facilitating extrathymic generation of Treg cells [151], its anti-inflammatory effects are also mediated by activation of G protein receptor 109a, inhibition of TLR4 signaling and suppression of NF-κB activation [152,153].
In an animal model of acute mania induced by d-amphetamine (d-AMPH), it has been shown that sodium butyrate (SB) could inhibit d-AMPH-related hyperactivity and restore the activity of mitochondrial respiratory-chain complexes as well [154]. Qiu et al. established LPS-induced depression model in mice to examine protective effects of SB. Their findings showed that pretreatment at the dose of 300 mg/kg for 10 days could improve LPS-induced depressive-like behaviors and prevent LPS-induced elevation in pro-inflammatory cytokines, such as IL-1β, IL-6, and TNF-α [155]. In a most recent animal study Wang et al., demonstrated that butyrate could improve depression-like behaviors and mitigated ferroptosis of prefrontal cortex (PFC) neurons induced by acute stress in mice, through modulation of the gut–brain axis and reduction in systemic inflammatory responses [156]. These mechanisms collectively suggest that butyrate supplementation may serve as a promising candidate for alleviating psychological symptoms [157].
Propionate is another SCFA found in both the gut and the bloodstream. Its protective effects in pathological conditions are attributed to the improvement of intestinal barrier function, attenuation of inflammation and oxidative stress, primarily through the modulation of inflammation-related signaling pathways [158,159]. Preoperative supplementation with propionate could enhance intestinal function recovery in rats and ameliorate neuroinflammation and hippocampus-related memory impairment, due to propionate regulatory influence of on Th17 cell-mediated immune responses [160]. Propionate can also enhance neurocognitive function by reducing Th17 cells infiltration into the CNS, decreasing IL-17A expression, and suppressing microglial activation through an IL-17A/IL-17RA-dependent mechanism [160]. Propionate may contribute to boosting de novo Treg-cell generation in the periphery by inhibiting HDAC activity as well [151]. Intravenous administration of low-dose propionate (2 mg/kg body weight/day) was shown to exert antidepressant effects in rats exposed to CUMS [161]. Moreover, short term intrarectal administration of sodium propionate exhibited antidepressant-like effects in CUMS rats [162]. In a recent study, protective effects of oral sodium propionate in CUMS-exposed rats were attributed to reshaping GM and region-specific epigenetic modification of histone 3 [163].

5.3. Methyl Rich Diets and Inflammatory Responses

Some key products of the GM, such as folate and vitamin B12 may exert protective effects against neuroinflammatory disorders by modulating the GM–immune–brain axis, via DNA methylation alterations [164]. Deficiency of these essential products due to gut dysbiosis may contribute to the development of NPD by triggering inflammation. For example, folate deficiency mediates neuroinflammation by inducing NF-ĸB activation [165]. Therefore, their supplementation may be considered a promising strategy for the treatment of immunometabolic form of NPD [166]. In this context, Menegas et al. reported that folic acid, when used as a therapeutic adjunct to lithium, alleviate the manic-like behaviors, oxidative stress, and pro-inflammatory cytokines in a rat model of mania induced by methamphetamine (m-AMPH) [167]. Khosravi et al. found that a healthy dietary pattern decreases the risk of depression by increasing serum levels of the folate and vitamin B12, whereas an unhealthy dietary pattern increases the risk of depression by reducing their serum levels [168]. Jia et al. also reported that neuroprotective effect of folic acid in MPTP-induced PD in mice was linked to suppressing neuroinflammation by inhibiting NLRP3 inflammasome and improving mitochondrial integrity via the p53-PGC-1α pathway [169].
In a clinical study conducted by Chen et al., folic acid supplementation could alleviate AD symptoms by decreasing mRNA levels of pro-inflammatory cytokines such as TNFα in leukocytes [170]. Additionally, Chen et al. reported that folic acid and vitamin B12 supplementation could hamper the progression of cognitive decline in AD patients by decreasing serum concentrations of TNFα [164].
Choline, a methyl donor and precursor of the neurotransmitter acetylcholine, has also been considered a promising intervention for regulating the ratio of pro-to-anti-inflammatory cytokines in the hippocampus and modulating long-term inflammatory tone [171]. Dietary choline intake may be considered a promising candidate to alleviate hallmarks of AD, particularly inflammation [172]. Mechanistically, Egilmez et al. reported that ameliorative influence of choline chloride in the treatment of social behavior in a rat model of LPS-induced autism is connected to reduced levels of TNF-α, IL-2, and IL-17 [173].
In U.S. adults, dietary choline intake is negatively correlated to the risk of depressive symptoms [174]. Furthermore, betaine, one of the choline oxides in the body, may exert an anti-inflammatory effects by suppressing the NF-ĸB signaling pathway, a master regulator of genes involved in the inflammatory response associated with depression [175]. Another mechanism is that betaine helps reduce homocysteine levels by converting it to methionine, and hence exerting antidepressant effects, as elevated homocysteine is linked to an increased risk of depression in humans [176,177]. In addition to anti-inflammatory effects, protective role of betaine is associated with overexpression of tight junction proteins (occludin and ZO-1), promoting the intestinal barrier integrity, and modulation of the GM [178]. Liu et al. also found that betaine alleviates depression and anxiety-like behaviors in mice with dextran sulfate sodium-induced colitis by reducing DNA damage, improving mitochondrial impairment, and suppressing the cGAS-STING signaling pathway involved in the inflammatory responses [179]. Hui et al. reported that betaine could rescue METH-induced depressive-like behavior and cognitive dysfunction by reducing neuroinflammation through inhibition of NLRP3 inflammasome in hippocampal region of mice [180]. Moreover, betaine has demonstrated ability for suppressing AβO-induced neuroinflammation in microglia by mitigating the activation of the NLRP3 inflammasome and NF-κB [181].

5.4. Modified Mediterranean Diet and Ketogenic Diet

Certain specific diets have demonstrated capacity for improving NPD through inhibition of neuroinflammation or modification of the GM structure and composition, thereby affecting epigenetic mechanisms [182,183,184]. Joseph et al. proposed that protective effects of a modified Mediterranean diet promoting the production of SCFAs (e.g., acetate, butyrate, and propionate) by the GM are associated with improvements in immune and metabolic dysfunction in SCZ [185]. The ketogenic diet (KD) is another effective approach to improving certain NPD, such as epilepsy-associated depression, autism bipolar disorder, and SCZ [186]. The KD is a high-fat, low-carbohydrate, and moderate-protein diet that shifts the body’s metabolism toward using fat as its primary energy source instead of carbohydrates [187]. This metabolic state, called ketosis, leads to the production of ketone bodies (such as beta-hydroxybutyrate, an epigenetic modifier) that serve as an alternative fuel, particularly for the brain. In a recent mechanistic study Liang et al. demonstrated that KD could improve CUMS-induced depression-like behaviors in mice by suppressing glial activation markers (Iba-1 and GFAP), hampering the expression of inflammatory cytokines, including IL-1β, TNF-α, and COX-2, inhibiting the TLR4/MyD88/NF-κB signaling pathway, and promoting the BDNF/TrkB/CREB and Wnt/β-catenin signaling pathways [188]. In an AD mouse model, Di Lucente et al. reported that KD could rescue Long-Term-Potentiation (LTP), increase synaptic plasticity, and decrease markers of microgliosis by elevating the concentration of beta-hydroxy-butyrate [189]. Zhu et al. found that the dietary intervention with KD in a LPS-induced rat PD model could reverse the disease progression by preventing certain events, including overexpression of pro-inflammatory mediators (TNF-α, IL-1β, and IL-6), the loss of dopaminergic neurons, and diminishing mGluR5+ microglia cells, elevating TSPO+ microglia cells, decreasing H3K9 acetylation in the mGluR5 promoter region and mGluR5 mRNA expression, and reducing the phosphorylation levels of Akt/GSK-3β/CREB pathway [190].
In a clinical study Allan et al. demonstrated that a modified KD could exert protective effects in ASD children by altering the GM, elevating the expression of butyrate kinase in the gut, reducing the amount of pro-inflammatory cytokines (IL-12p70 and IL-1β), and changing the levels of BDNF-related miRNAs in the plasma [191]. In another clinical study conducted by Schweickart et al., the Mediterranean KD was capable of alleviating risk factors of AD by decreasing the inflammatory marker GlycA, suggesting a reduction in systemic inflammation owing to the effects of this type diet [192].

5.5. Immunomodulatory Effects of Different Drugs in the Treatment of Neuropsychiatric Disorders by Targeting GM via Epigenetic Mechanisms

Accumulating evidence has demonstrated that the beneficial effects of antipsychotics, antidepressants, and antibiotics in the cure of NPD are associated with their ability to down-regulate neuroinflammation by altering GM composition and increasing production of metabolites with anti-inflammatory and epigenetic effects. Figure 2 shows how different types of medications, including antipsychotics, antidepressants, and antibiotics contribute to the treatment of NPD by altering pro- and anti-inflammatory gut bacteria and changing the production of epigenetic metabolites with anti-inflammatory effects.

5.5.1. Psychiatric Medications That Influence GM, Inflammation and the Epigenome

Besides their effects on neurotransmission, psychiatric drugs also exhibit anti-inflammatory properties and modulate the GM. For instance, patients with BD and treated with quetiapine (300 mg/d) for four weeks exhibited a rebound in the abundance of Bifidobacteria and Eubacterium rectale bacteria, which are involved in reducing inflammation by producing butyrate and other SCFAs [193]. In another study Li et al., found that after 24-week risperidone treatment, patients with SCZ exhibited a remarkable increase in serum levels of butyric acid, accompanied by improvements in the PANSS positive symptoms subscale scores [194]. A four-week treatment with amisulpride in SCZ patients was also associated with an increased abundance of SCFA-producing bacteria (such as Dorea and Butyricicoccus), and a decreased abundance of pathogenic bacteria (Actinomyces and Porphyromonas) and a reduction in IL-6 levels [195]. Another recent study demonstrated that treatment with amisulpride exerted anti-inflammatory effects in SCZ patients by elevating serum levels of acetate, increasing the expression of several genes, including HDAC1, GPR109a, GPR43, TLR2, soluble CD14 (sCD14), and N-methyl-d-aspartate receptor (NMDAR), but reducing the expression TLR4 and pregnane X receptor (PXR) [196].
Mood stabilizers, a class of medications used for treating mood swings in BD patients, can also exert their protective effects by reshaping the GM and modulation of immune system function via epigenetic mechanisms. For example, lithium carbonate treatment could mitigate colon inflammation by modulating gut microbial diversity, increasing the abundance of SCFA-producing bacteria, such as Akkermansia muciniphila, and enhancing anti-inflammatory Treg cell activity in colonic lamina propria (LP) through a GPR43-related mechanism [197]. Furthermore, a more recent study showed that the recommended dose of lithium increases the abundance of SCFA-producing bacteria, including Lactobacillus, Ruminococcaceae, Enterorhabdus, Muribaculaceae, and Coprococcus [198].

5.5.2. Antidepressant and Antibiotic Medications

In respect to antidepressants, several studies have demonstrated that these medications may exert their protective effects, in part, by altering the richness, diversity, and composition of the GM in favor of SCFA-producing bacteria, which are known to facilitate epigenetic modifications [199,200]. In an investigation conducted by Zhang et al., the authors reported that antidepressant effects of a 6-week treatment with fluoxetine and amitriptyline in rats exposed to CUMS are linked to increased abundance of anti-inflammatory butyrate-producing bacteria including Butyricimonas [201]. In a human clinical study Jiang et al. reported higher levels of anti-inflammatory bacteria, including Ruminococcus, Bifidobacterium, and Faecalibacterium, which are involved in butyrate production, in responders to serotonin reuptake inhibitors (SSRIs) versus non-responders [202].
There is also evidence that some antibiotics with antidepressant properties my influence inflammation and the GM. For instance, Yang et al. reported that a 4-week minocycline treatment exhibited antidepressant effects in CUMS mice by suppressing neuroinflammation in the hippocampus, reshaping the GM, improving intestinal barrier integrity, and restoring butyrate concentration [203]. Li et al. demonstrated that treating rats exposed to CUMS with 150 mg/kg rifaximin (a broad-spectrum, non-absorbable antibiotic) for 4 weeks enhanced the content of anti-inflammatory mediators secreted by microglia and increased the abundance of Ruminococcaceae and Lachnospiraceae, which are positively associated with elevated butyrate concentration in the brain [204].

6. Conclusions and Future Perspectives

The findings described in this review indicate that disturbances in the GM composition give rise to immune imbalance and subsequently the development of NPD in part by inducing epigenetic aberrations. Knowledge about various communication pathways on the bidirectional gut–brain axis paves the way for researchers to design promising therapeutic strategies for the prevention or treatment of NPD. Specific diets, probiotic/prebiotic supplementations, and various medications are capable of modulating GM, increasing the availability of their metabolites in the intestine, improving immune system function, and hence mitigating the severity of NPD via normalizing epigenetic aberrations. Thus, further research into the interactions between the GM, the immune system, the epigenome, and the brain against a genetic backdrop paves the way for more precise understanding of the pathophysiological mechanisms of NPD and exploring novel and alternative therapeutic targets and strategies for the prevention or treatment of NPD. However, several challenges should be addressed in this field. For instance, the majority of the existing investigations have small sample, which may result in insufficient statistical power to establish robust and reproducible associations. Therefore, gaining deeper insights in the field requires future studies with larger sample sizes and more refined experimental design. To navigate the complexity of this field and develop innovative therapies, collaborative studies involving experts from diverse disciplines—including microbiology, immunology, neuroscience, and computational biology—should be pursued. To clarify the influence of microbial communities in the GI tract on immune system function in the brain, and to gain deeper understanding of the effects of GM-based therapeutics on the GM–immune system–brain axis, large longitudinal clinical studies should be conducted in individuals with NPD rather than relying on animal models.

Author Contributions

Conceptualization, S.T. and H.M.A.; writing—original draft preparation, S.N. and H.M.A.; editing and reorganization of the article, H.M.A., S.T., A.P. and S.N. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We acknowledge Faria Ashrafi for her assistance with the illustrations in this manuscript.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Zuo, Y.; Wei, D.; Zhu, C.; Naveed, O.; Hong, W.; Yang, X. Unveiling the pathogenesis of psychiatric disorders using network models. Genes 2021, 12, 1101. [Google Scholar] [CrossRef]
  2. Ilieva, M.S. Non-coding RNAs in neurological and neuropsychiatric disorders: Unraveling the hidden players in disease pathogenesis. Cells 2024, 13, 1063. [Google Scholar] [CrossRef]
  3. Morozova, A.; Zorkina, Y.; Abramova, O.; Pavlova, O.; Pavlov, K.; Soloveva, K.; Volkova, M.; Alekseeva, P.; Andryshchenko, A.; Kostyuk, G.; et al. Neurobiological highlights of cognitive impairment in psychiatric disorders. Int. J. Mol. Sci. 2022, 23, 1217. [Google Scholar] [CrossRef]
  4. McGrath, T.; Baskerville, R.; Rogero, M.; Castell, L. Emerging evidence for the widespread role of glutamatergic dysfunction in neuropsychiatric diseases. Nutrients 2022, 14, 917. [Google Scholar] [CrossRef]
  5. Piancone, F.; La Rosa, F.; Marventano, I.; Saresella, M.; Clerici, M. The role of the inflammasome in neurodegenerative diseases. Molecules 2021, 26, 953. [Google Scholar] [CrossRef]
  6. Tanaka, M.; Vécsei, L. A Decade of Dedication: Pioneering Perspectives on Neurological Diseases and Mental Illnesses. Biomedicines 2024, 12, 1083. [Google Scholar] [CrossRef]
  7. Łoś, K.; Waszkiewicz, N. Biological markers in anxiety disorders. J. Clin. Med. 2021, 10, 1744. [Google Scholar] [CrossRef]
  8. Cătălina, G.R.; Gheorman, V.; Gheorman, V.; Forțofoiu, M.-C. The Role of Neuroinflammation in the Comorbidity of Psychiatric Disorders and Internal Diseases. Healthcare 2025, 13, 837. [Google Scholar] [CrossRef]
  9. Schmidt-Morgenroth, I.; Michaud, P.; Gasparini, F.; Avrameas, A. Central and peripheral inflammation in mild cognitive impairment in the context of Alzheimer’s disease. Int. J. Mol. Sci. 2023, 24, 10523. [Google Scholar] [CrossRef]
  10. Upthegrove, R.; Khandaker, G.M. Cytokines, oxidative stress and cellular markers of inflammation in schizophrenia. Neuroinflamm. Schizophr. 2019, 44, 49–66. [Google Scholar]
  11. Balõtšev, R.; Koido, K.; Vasar, V.; Janno, S.; Kriisa, K.; Mahlapuu, R.; Ljubajev, U.; Parksepp, M.; Veiksaar, P.; Volke, V.; et al. Inflammatory, cardio-metabolic and diabetic profiling of chronic schizophrenia. Eur. Psychiatry 2017, 39, 1–10. [Google Scholar] [CrossRef]
  12. Lussier, A.A.; Bodnar, T.S.; Weinberg, J. Intersection of epigenetic and immune alterations: Implications for fetal alcohol spectrum disorder and mental health. Front. Neurosci. 2021, 15, 788630. [Google Scholar] [CrossRef]
  13. Lossi, L.; Castagna, C.; Merighi, A. An overview of the epigenetic modifications in the brain under normal and pathological conditions. Int. J. Mol. Sci. 2024, 25, 3881. [Google Scholar] [CrossRef]
  14. Scholz, R.; Brösamle, D.; Yuan, X.; Beyer, M.; Neher, J.J. Epigenetic control of microglial immune responses. Immunol. Rev. 2024, 323, 209–226. [Google Scholar] [CrossRef]
  15. Cabanel, M.; Brand, C.; Oliveira-Nunes, M.C.; Cabral-Piccin, M.P.; Lopes, M.F.; Brito, J.M.; de Oliveira, F.L.; El-Cheikh, M.C.; Carneiro, K. Epigenetic control of macrophage shape transition towards an atypical elongated phenotype by histone deacetylase activity. PLoS ONE 2015, 10, e0132984. [Google Scholar] [CrossRef]
  16. Peedicayil, J. Genome–environment interactions and psychiatric disorders. Biomedicines 2023, 11, 1209. [Google Scholar] [CrossRef]
  17. Bekdash, R.A. Epigenetics, nutrition, and the brain: Improving mental health through diet. Int. J. Mol. Sci. 2024, 25, 4036. [Google Scholar] [CrossRef]
  18. Khalil, M.; Di Ciaula, A.; Mahdi, L.; Jaber, N.; Di Palo, D.M.; Graziani, A.; Baffy, G.; Portincasa, P. Unraveling the role of the human gut Microbiome in Health and diseases. Microorganisms 2024, 12, 2333. [Google Scholar] [CrossRef]
  19. Barathan, M.; Ng, S.L.; Lokanathan, Y.; Ng, M.H.; Law, J.X. The profound influence of gut microbiome and extracellular vesicles on animal health and disease. Int. J. Mol. Sci. 2024, 25, 4024. [Google Scholar] [CrossRef]
  20. O’Riordan, K.J.; Moloney, G.M.; Keane, L.; Clarke, G.; Cryan, J.F. The gut microbiota-immune-brain axis: Therapeutic implications. Cell Rep. Med. 2025, 6, 101982. [Google Scholar] [CrossRef]
  21. Lopizzo, N.; Marizzoni, M.; Begni, V.; Mazzelli, M.; Provasi, S.; Borruso, L.; Riva, M.A.; Cattaneo, A. Social isolation in adolescence and long-term changes in the gut microbiota composition and in the hippocampal inflammation: Implications for psychiatric disorders–Dirk Hellhammer Award Paper 2021. Psychoneuroendocrinology 2021, 133, 105416. [Google Scholar] [CrossRef]
  22. 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]
  23. Kowalski, K.; Szponar, B.; Bochen, P.; Żebrowska-Różańska, P.; Łaczmański, Ł.; Samochowiec, J.; Misiak, B. Altered levels of fecal short-chain fatty acids are associated with subclinical inflammation and worse cognitive performance in patients with schizophrenia. J. Psychiatr. Res. 2023, 165, 298–304. [Google Scholar] [CrossRef]
  24. Chen, S.-J.; Chen, C.-C.; Liao, H.-Y.; Lin, Y.-T.; Wu, Y.-W.; Liou, J.-M.; Wu, M.-S.; Kuo, C.-H.; Lin, C.-H. Association of fecal and plasma levels of short-chain fatty acids with gut microbiota and clinical severity in patients with Parkinson disease. Neurology 2022, 98, e848–e858. [Google Scholar] [CrossRef]
  25. Ling, Z.; Zhu, M.; Yan, X.; Cheng, Y.; Shao, L.; Liu, X.; Jiang, R.; Wu, S. Structural and functional dysbiosis of fecal microbiota in Chinese patients with Alzheimer’s disease. Front. Cell Dev. Biol. 2021, 8, 634069. [Google Scholar] [CrossRef]
  26. Li, Z.; Wang, H.; Yin, Y. Peripheral inflammation is a potential etiological factor in Alzheimer’s disease. Rev. Neurosci. 2024, 35, 99–120. [Google Scholar] [CrossRef]
  27. Zhao, Y.; Sharfman, N.M.; Jaber, V.R.; Lukiw, W.J. Down-regulation of essential synaptic components by GI-tract microbiome-derived lipopolysaccharide (LPS) in LPS-treated human neuronal-glial (HNG) cells in primary culture: Relevance to Alzheimer’s disease (AD). Front. Cell Neurosci. 2019, 13, 314. [Google Scholar] [CrossRef]
  28. Kouli, A.; Spindler, L.R.; Fryer, T.D.; Hong, Y.T.; Malpetti, M.; Aigbirhio, F.I.; White, S.R.; Camacho, M.; O’Brien, J.T.; Williams-Gray, C.H. Neuroinflammation is linked to dementia risk in Parkinson’s disease. Brain 2024, 147, 923–935. [Google Scholar] [CrossRef]
  29. Nascimento, C.; Nunes, P.V.; Leite, R.E.P.; Grinberg, L.T.; Suemoto, C.K.; Lafer, B. The relationship of neuropsychiatric symptoms with inflammatory markers in the hippocampus and cingulate cortex of bipolar disorder subjects: A post-mortem study. J. Psychiatr. Res. 2024, 173, 25–33. [Google Scholar] [CrossRef]
  30. Ramakrishnan, A.; Piehl, N.; Simonton, B.; Parikh, M.; Zhang, Z.; Teregulova, V.; van Olst, L.; Gate, D. Epigenetic dysregulation in Alzheimer’s disease peripheral immunity. Neuron 2024, 112, 1235–1248.e1235. [Google Scholar] [CrossRef]
  31. Murphy, C.E.; Walker, A.K.; O’Donnell, M.; Galletly, C.; Lloyd, A.R.; Liu, D.; Weickert, C.S.; Weickert, T.W. Peripheral NF-κB dysregulation in people with schizophrenia drives inflammation: Putative anti-inflammatory functions of NF-κB kinases. Transl. Psychiatry 2022, 12, 21. [Google Scholar] [CrossRef]
  32. Gong, H.; Lu, Y.; Deng, S.-L.; Lv, K.-Y.; Luo, J.; Luo, Y.; Du, Z.-L.; Wu, L.-F.; Liu, T.-Y.; Wang, X.-Q.; et al. Targeting S100A9 attenuates social dysfunction by modulating neuroinflammation and myelination in a mouse model of autism. Pharmacol. Res. 2025, 211, 107568. [Google Scholar] [CrossRef]
  33. Abdolmaleky, H.M.; Alam, R.; Nohesara, S.; Deth, R.C.; Zhou, J.-R. iPSC-Derived Astrocytes and Neurons Replicate Brain Gene Expression, Epigenetic, Cell Morphology and Connectivity Alterations Found in Autism. Cells 2024, 13, 1095. [Google Scholar] [CrossRef]
  34. Megagiannis, P.; Mei, Y.; Yan, R.E.; Yuan, L.; Wilde, J.J.; Eckersberg, H.; Suresh, R.; Tan, X.; Chen, H.; Farmer, W.T.; et al. Autism-associated CHD8 controls reactive gliosis and neuroinflammation via remodeling chromatin in astrocytes. Cell Rep. 2024, 43, 114637. [Google Scholar] [CrossRef]
  35. Zhao, T.; Piao, L.-H.; Li, D.-P.; Xu, S.-H.; Wang, S.-Y.; Yuan, H.-B.; Zhang, C.-X. BDNF gene hydroxymethylation in hippocampus related to neuroinflammation-induced depression-like behaviors in mice. J. Affect. Disord. 2023, 323, 723–730. [Google Scholar] [CrossRef]
  36. Wang, X.; Chen, X.; Guan, X.; Li, Z. The neutrophil-to-Lymphocyte ratio is associated with clinical symptoms in first-episode medication-naïve patients with schizophrenia. Schizophrenia 2024, 10, 13. [Google Scholar] [CrossRef]
  37. Mendez-Victoriano, G.; Zhu, Y.; Middleton, F.; Massa, P.T.; Ajulu, K.; Webster, M.J.; Weickert, C.S. Increased Parenchymal Macrophages are associated with decreased Tyrosine Hydroxylase mRNA levels in the Substantia Nigra of people with Schizophrenia and Bipolar Disorder. Psychiatry Res. 2024, 340, 116141. [Google Scholar] [CrossRef]
  38. Nadeem, A.; Ahmad, S.F.; Al-Harbi, N.O.; Al-Ayadhi, L.Y.; Sarawi, W.; Attia, S.M.; Bakheet, S.A.; Alqarni, S.A.; Ali, N.; AsSobeai, H.M. Imbalance in pro-inflammatory and anti-inflammatory cytokines milieu in B cells of children with autism. Mol. Immunol. 2022, 141, 297–304. [Google Scholar] [CrossRef]
  39. Yan, Z.; Yang, W.; Wei, H.; Dean, M.N.; Standaert, D.G.; Cutter, G.R.; Benveniste, E.N.; Qin, H. Dysregulation of the adaptive immune system in patients with early-stage Parkinson disease. Neurol. Neuroimmunol. Neuroinflamm. 2021, 8, e1036. [Google Scholar] [CrossRef]
  40. Jiang, S.-S.; Wang, Y.-L.; Xu, Q.-H.; Gu, L.-Y.; Kang, R.-Q.; Yang, W.-Y.; Zhang, B.-R.; Tian, J.; Pu, J.-L. Cytokine and chemokine map of peripheral specific immune cell subsets in Parkinson’s disease. npj Park. Dis. 2023, 9, 117. [Google Scholar] [CrossRef]
  41. Deecke, L.; Goldeck, D.; Ohlei, O.; Homann, J.; Demuth, I.; Bertram, L.; Pawelec, G.; Lill, C.M. Immune cell distributions in the blood of healthy individuals at high genetic risk of Parkinson’s disease. Int. J. Mol. Sci. 2024, 25, 13655. [Google Scholar] [CrossRef]
  42. Lu, Y.; Li, K.; Hu, Y.; Wang, X. Expression of immune related genes and possible regulatory mechanisms in Alzheimer’s disease. Front. Immunol. 2021, 12, 768966. [Google Scholar] [CrossRef]
  43. Xu, H.; Jia, J. Single-cell RNA sequencing of peripheral blood reveals immune cell signatures in Alzheimer’s disease. Front. Immunol. 2021, 12, 645666. [Google Scholar] [CrossRef]
  44. Zhang, H.; Cao, F.; Zhou, Y.; Wu, B.; Li, C. Peripheral immune cells contribute to the pathogenesis of Alzheimer’s disease. Mol. Neurobiol. 2025, 62, 264–270. [Google Scholar] [CrossRef]
  45. Rachayon, M.; Jirakran, K.; Sodsai, P.; Sughondhabirom, A.; Maes, M. T cell activation and deficits in T regulatory cells are associated with major depressive disorder and severity of depression. Sci. Rep. 2024, 14, 11177. [Google Scholar] [CrossRef]
  46. Nikolova, V.L.; Smith, M.R.; Hall, L.J.; Cleare, A.J.; Stone, J.M.; Young, A.H. Perturbations in gut microbiota composition in psychiatric disorders: A review and meta-analysis. JAMA Psychiatry 2021, 78, 1343–1354. [Google Scholar] [CrossRef]
  47. Sanmarco, L.M.; Wheeler, M.A.; Gutiérrez-Vázquez, C.; Polonio, C.M.; Linnerbauer, M.; Pinho-Ribeiro, F.A.; Li, Z.; Giovannoni, F.; Batterman, K.V.; Scalisi, G.; et al. Gut-licensed IFNγ+ NK cells drive LAMP1+ TRAIL+ anti-inflammatory astrocytes. Nature 2021, 590, 473–479. [Google Scholar] [CrossRef]
  48. Noe, J.T.; Rendon, B.E.; Geller, A.E.; Conroy, L.R.; Morrissey, S.M.; Young, L.E.; Bruntz, R.C.; Kim, E.J.; Wise-Mitchell, A.; Rizzo, M.B.d.S.; et al. Lactate supports a metabolic-epigenetic link in macrophage polarization. Sci. Adv. 2021, 7, eabi8602. [Google Scholar] [CrossRef]
  49. Ling, Z.; Jin, G.; Yan, X.; Cheng, Y.; Shao, L.; Song, Q.; Liu, X.; Zhao, L. Fecal dysbiosis and immune dysfunction in Chinese elderly patients with schizophrenia: An observational study. Front. Cell Infect. Microbiol. 2022, 12, 886872. [Google Scholar] [CrossRef]
  50. Huang, T.; Shang, Y.; Dai, C.; Zhang, Q.; Hu, S.; Xie, J. Gut microbiota and its relation to inflammation in patients with bipolar depression: A cross-sectional study. Ann. Gen. Psychiatry 2023, 22, 21. [Google Scholar] [CrossRef]
  51. Hu, S.; Li, A.; Huang, T.; Lai, J.; Li, J.; Sublette, M.E.; Lu, H.; Lu, Q.; Du, Y.; Hu, Z. Gut microbiota changes in patients with bipolar depression. Adv. Sci. 2019, 6, 1900752. [Google Scholar] [CrossRef]
  52. Li, Z.; Tao, X.; Wang, D.; Pu, J.; Liu, Y.; Gui, S.; Zhong, X.; Yang, D.; Zhou, H.; Tao, W.; et al. Alterations of the gut microbiota in patients with schizophrenia. Front. Psychiatry 2024, 15, 1366311. [Google Scholar] [CrossRef]
  53. Nie, S.; Wang, J.; Deng, Y.; Ye, Z.; Ge, Y. Inflammatory microbes and genes as potential biomarkers of Parkinson’s disease. npj Biofilm. Microbiomes 2022, 8, 101. [Google Scholar] [CrossRef]
  54. 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]
  55. Donaldson, A.I.; Fyfe, C.L.; Martin, J.C.; Smith, E.E.; Horgan, G.W.; Myint, P.K.; Johnstone, A.M.; Scott, K.P. Aging Gut-Brain Interactions: Pro-Inflammatory Gut Bacteria Are Elevated in Fecal Samples from Individuals Living with Alzheimer’s Dementia. Geriatrics 2025, 10, 37. [Google Scholar] [CrossRef]
  56. Krueger, M.E.; Boles, J.S.; Simon, Z.D.; Alvarez, S.D.; McFarland, N.R.; Okun, M.S.; Zimmermann, E.M.; Forsmark, C.E.; Tansey, M.G. Comparative analysis of Parkinson’s and inflammatory bowel disease gut microbiomes reveals shared butyrate-producing bacteria depletion. npj Park. Dis. 2025, 11, 50. [Google Scholar] [CrossRef]
  57. Cao, X.; Liu, K.; Liu, J.; Liu, Y.; Xu, L.; Wang, H.; Zhu, Y.; Wang, P.; Li, Z.; Wen, J.; et al. Dysbiotic gut microbiota and dysregulation of cytokine profile in children and teens with autism spectrum disorder. Front Neurosci. 2021, 15, 635925. [Google Scholar] [CrossRef]
  58. Ghorbani, M.; Joseph, G.B.S.; Mei, T.M.; Ramly, S.S.; Rasat, M.A.M.; Croft, L.; Parimannan, S.; Rajandas, H.; Lee, S.Y. Functional associations of the gut microbiome with dopamine, serotonin, and BDNF in schizophrenia: A pilot study. Egypt. J. Neurol. Psychiatry Neurosurg. 2024, 60, 123. [Google Scholar] [CrossRef]
  59. Wu, H.; Liu, Y.; Han, Y.; Liu, B.; Chen, S.; Ye, Z.; Li, J.; Xie, L.; Wu, X. Integrated Analysis of Gut Microbiome, Inflammation, and Neuroimaging Features Supports the Role of Microbiome–Gut–Brain Crosstalk in Schizophrenia. Schizophr. Bull. Open 2024, 5, sgae026. [Google Scholar] [CrossRef]
  60. Painold, A.; Mörkl, S.; Kashofer, K.; Halwachs, B.; Dalkner, N.; Bengesser, S.; Birner, A.; Fellendorf, F.; Platzer, M.; Queissner, R.; et al. A step ahead: Exploring the gut microbiota in inpatients with bipolar disorder during a depressive episode. Bipolar Disord. 2019, 21, 40–49. [Google Scholar] [CrossRef]
  61. Coello, K.; Hansen, T.H.; Sørensen, N.; Munkholm, K.; Kessing, L.V.; Pedersen, O.; Vinberg, M. Gut microbiota composition in patients with newly diagnosed bipolar disorder and their unaffected first-degree relatives. Brain Behav. Immun. 2019, 75, 112–118. [Google Scholar] [CrossRef]
  62. Guo, Z.; Xiao, S.; Chen, G.; Zhong, S.; Zhong, H.; Sun, S.; Chen, P.; Tang, X.; Yang, H.; Jia, Y. Disruption of the gut microbiota-inflammation-brain axis in unmedicated bipolar disorder II depression. Transl. Psychiatry 2024, 14, 495. [Google Scholar] [CrossRef]
  63. Liu, R.T.; Rowan-Nash, A.D.; Sheehan, A.E.; Walsh, R.F.; Sanzari, C.M.; Korry, B.J.; Belenky, P. Reductions in anti-inflammatory gut bacteria are associated with depression in a sample of young adults. Brain Behav. Immun. 2020, 88, 308–324. [Google Scholar] [CrossRef]
  64. Ling, Z.; Cheng, Y.; Chen, F.; Yan, X.; Liu, X.; Shao, L.; Jin, G.; Zhou, D.; Jiang, G.; Li, H.; et al. Changes in fecal microbiota composition and the cytokine expression profile in school-aged children with depression: A case-control study. Front. Immunol. 2022, 13, 964910. [Google Scholar] [CrossRef]
  65. Liu, P.; Gao, M.; Liu, Z.; Zhang, Y.; Tu, H.; Lei, L.; Wu, P.; Zhang, A.; Yang, C.; Li, G.; et al. Gut microbiome composition linked to inflammatory factors and cognitive functions in first-episode, drug-naive major depressive disorder patients. Front. Neurosci. 2022, 15, 800764. [Google Scholar] [CrossRef]
  66. Liu, P.; Liu, Z.; Wang, J.; Wang, J.; Gao, M.; Zhang, Y.; Yang, C.; Zhang, A.; Li, G.; Li, X.; et al. Immunoregulatory role of the gut microbiota in inflammatory depression. Nat. Commun. 2024, 15, 3003. [Google Scholar] [CrossRef]
  67. Cheng, Y.; Zhu, Z.; Yang, Z.; Liu, X.; Qian, X.; Zhu, J.; Hu, X.; Jiang, P.; Cui, T.; Wang, Y.; et al. Alterations in fecal microbiota composition and cytokine expression profiles in adolescents with depression: A case-control study. Sci. Rep. 2025, 15, 12177. [Google Scholar] [CrossRef]
  68. Rothhammer, V.; Borucki, D.M.; Tjon, E.C.; Takenaka, M.C.; Chao, C.-C.; Ardura-Fabregat, A.; De Lima, K.A.; Gutiérrez-Vázquez, C.; Hewson, P.; Staszewski, O.; et al. Microglial control of astrocytes in response to microbial metabolites. Nature 2018, 557, 724–728. [Google Scholar] [CrossRef]
  69. Zhang, L.; Ma, Z.; Zhang, X.; Wang, J.; Tian, W.; Ren, Y.; Liu, Y.; Wang, T.; Li, Y.; Liu, Y. Butyrate alleviates alcoholic liver disease-associated inflammation through macrophage regulation and polarization via the HDAC1/miR-155 axis. Int. Immunopharmacol. 2024, 131, 111852. [Google Scholar] [CrossRef]
  70. Yu, H.; Li, R.; Liang, X.-j.; Yang, W.-M.; Guo, L.; Liu, L.; Tan, Q.-r.R.; Peng, Z.-w. A cross-section study of the comparison of plasma inflammatory cytokines and short-chain fatty acid in patients with depression and schizophrenia. BMC Psychiatry 2024, 24, 834. [Google Scholar] [CrossRef]
  71. Liu, J.; Chen, J.; Ehrlich, S.; Walton, E.; White, T.; Perrone-Bizzozero, N.; Bustillo, J.; Turner, J.A.; Calhoun, V.D. Methylation patterns in whole blood correlate with symptoms in schizophrenia patients. Schizophr. Bull. 2014, 40, 769–776. [Google Scholar] [CrossRef]
  72. Shindo, R.; Tanifuji, T.; Okazaki, S.; Otsuka, I.; Shirai, T.; Mouri, K.; Horai, T.; Hishimoto, A. Accelerated epigenetic aging and decreased natural killer cells based on DNA methylation in patients with untreated major depressive disorder. npj Aging 2023, 9, 19. [Google Scholar] [CrossRef]
  73. Luo, C.; Pi, X.; Hu, N.; Wang, X.; Xiao, Y.; Li, S.; Sweeney, J.A.; Bishop, J.R.; Gong, Q.; Xie, D.; et al. Subtypes of schizophrenia identified by multi-omic measures associated with dysregulated immune function. Mol. Psychiatry 2021, 26, 6926–6936. [Google Scholar] [CrossRef]
  74. Luo, C.; Pi, X.; Zhang, Q.; Hu, N.; Xiao, Y.; Sweeney, J.A.; Bishop, J.R.; Gong, Q.; Xie, D.; Lui, S. A subtype of schizophrenia patients with altered methylation level of genes related to immune cell activity. Psychol. Med. 2024, 54, 2538–2546. [Google Scholar] [CrossRef]
  75. Sabunciyan, S.; Maher, B.; Bahn, S.; Dickerson, F.; Yolken, R.H. Association of DNA methylation with acute mania and inflammatory markers. PLoS ONE 2015, 10, e0132001. [Google Scholar] [CrossRef]
  76. Zhou, C.; Chen, J.; Tang, X.; Feng, X.; Yu, M.; Sha, W.; Wang, X.; Zhang, X.; Yi, H.; Zhang, X. DNA methylation and gene expression of the chemokine (CXC motif) ligand 1 in patients with deficit and non-deficit schizophrenia. Psychiatry Res. 2018, 268, 82–86. [Google Scholar] [CrossRef]
  77. Garcia-Ruiz, B.; Moreno, L.; Muntané, G.; Sánchez-Gistau, V.; Gutiérrez-Zotes, A.; Martorell, L.; Labad, J.; Vilella, E. Leukocyte and brain DDR1 hypermethylation is altered in psychosis and is correlated with stress and inflammatory markers. Epigenomics 2020, 12, 251–265. [Google Scholar] [CrossRef]
  78. Ni, C.; Jiang, W.; Wang, Z.; Wang, Z.; Zhang, J.; Zheng, X.; Liu, Z.; Ou, H.; Jiang, T.; Liang, W. LncRNA-AC006129. 1 reactivates a SOCS3-mediated anti-inflammatory response through DNA methylation-mediated CIC downregulation in schizophrenia. Mol. Psychiatry 2021, 26, 4511–4528. [Google Scholar] [CrossRef]
  79. Tang, Y.; Tan, Y.; Palaniyappan, L.; Yao, Y.; Luo, Q.; Li, Y. Epigenetic profile of the immune system associated with symptom severity and treatment response in schizophrenia. J. Psychiatry Neurosci. 2024, 49, E45–E58. [Google Scholar] [CrossRef]
  80. Mirza, S.; Lima, C.N.C.; Del Favero-Campbell, A.; Rubinstein, A.; Topolski, N.; Cabrera-Mendoza, B.; Kovács, E.H.C.; Blumberg, H.P.; Richards, J.G.; Williams, A.J.; et al. Blood epigenome-wide association studies of suicide attempt in adults with bipolar disorder. Transl. Psychiatry 2024, 14, 70. [Google Scholar] [CrossRef]
  81. Garcia-Ruiz, B.; Jiménez, E.; Aranda, S.; Verdolini, N.; Gutiérrez-Zotes, A.; Sáez, C.; Losantos, E.; Alonso-Lana, S.; Fatjó-Vilas, M.; Sarró, S. Associations of altered leukocyte DDR1 promoter methylation and childhood trauma with bipolar disorder and suicidal behavior in euthymic patients. Mol. Psychiatry 2024, 29, 2478–2486. [Google Scholar] [CrossRef]
  82. Crawford, B.; Craig, Z.; Mansell, G.; White, I.; Smith, A.; Spaull, S.; Imm, J.; Hannon, E.; Wood, A.; Yaghootkar, H.; et al. DNA methylation and inflammation marker profiles associated with a history of depression. Hum. Mol. Genet. 2018, 27, 2840–2850. [Google Scholar] [CrossRef]
  83. Draganov, M.; Arranz, M.J.; Salazar, J.; de Diego-Adeliño, J.; Gallego-Fabrega, C.; Jubero, M.; Carceller-Sindreu, M.; Portella, M.J. Association study of polymorphisms within inflammatory genes and methylation status in treatment response in major depression. Eur. Psychiatry 2019, 60, 7–13. [Google Scholar] [CrossRef]
  84. Rasmusson, A.J.; Gallwitz, M.; Soltanabadi, B.; Ciuculete, D.M.; Mengel-From, J.; Christensen, K.; Nygaard, M.; Soerensen, M.; Boström, A.E.; Fredriksson, R.; et al. Toll-like receptor 4 methylation grade is linked to depressive symptom severity. Transl. Psychiatry 2021, 11, 371. [Google Scholar] [CrossRef]
  85. Han, K.-M.; Choi, K.W.; Kim, A.; Kang, W.; Kang, Y.; Tae, W.-S.; Han, M.-R.; Ham, B.-J. Association of DNA methylation of the NLRP3 gene with changes in cortical thickness in major depressive disorder. Int. J. Mol. Sci. 2022, 23, 5768. [Google Scholar] [CrossRef]
  86. Daily, K.P.; Badr, A.; Eltobgy, M.; Estfanous, S.; Whitham, O.; Tan, M.H.; Carafice, C.; Krause, K.; McNamara, A.; Hamilton, K.; et al. DNA hypomethylation promotes the expression of CASPASE-4 which exacerbates inflammation and amyloid-β deposition in Alzheimer’s disease. Alzheimer’s Res. Ther. 2024, 16, 29. [Google Scholar] [CrossRef]
  87. Alshamrani, A.A.; Alshehri, S.; Alqarni, S.S.; Ahmad, S.F.; Alghibiwi, H.; Al-Harbi, N.O.; Alqarni, S.A.; Al-Ayadhi, L.Y.; Attia, S.M.; Alfardan, A.S.; et al. DNA hypomethylation is associated with increased inflammation in peripheral blood neutrophils of children with autism spectrum disorder: Understanding the role of ubiquitous pollutant Di (2-ethylhexyl) phthalate. Metabolites 2023, 13, 458. [Google Scholar] [CrossRef]
  88. Sun, X.-Y.; Zheng, T.; Yang, X.; Liu, L.; Gao, S.-S.; Xu, H.-B.; Song, Y.-T.; Tong, K.; Yang, L.; Gao, Y.; et al. HDAC2 hyperexpression alters hippocampal neuronal transcription and microglial activity in neuroinflammation-induced cognitive dysfunction. J. Neuroinflamm. 2019, 16, 249. [Google Scholar] [CrossRef]
  89. Correa, F.; Mallard, C.; Nilsson, M.; Sandberg, M. Activated microglia decrease histone acetylation and Nrf2-inducible anti-oxidant defence in astrocytes: Restoring effects of inhibitors of HDACs, p38 MAPK and GSK3β. Neurobiol. Dis. 2011, 44, 142–151. [Google Scholar] [CrossRef]
  90. Rigillo, G.; Vilella, A.; Benatti, C.; Schaeffer, L.; Brunello, N.; Blom, J.M.; Zoli, M.; Tascedda, F. LPS-induced histone H3 phospho (Ser10)-acetylation (Lys14) regulates neuronal and microglial neuroinflammatory response. Brain Behav. Immun. 2018, 74, 277–290. [Google Scholar] [CrossRef]
  91. Rodriguez-Zas, S.L.; Wu, C.; Southey, B.R.; O’Connor, J.C.; Nixon, S.E.; Garcia, R.; Zavala, C.; Lawson, M.; McCusker, R.H.; Romanova, E.V.; et al. Disruption of microglia histone acetylation and protein pathways in mice exhibiting inflammation-associated depression-like symptoms. Psychoneuroendocrinology 2018, 97, 47–58. [Google Scholar] [CrossRef]
  92. Wang, X.; Chen, S.; Ni, J.; Cheng, J.; Jia, J.; Zhen, X. miRNA-3473b contributes to neuroinflammation following cerebral ischemia. Cell Death Dis. 2018, 9, 11. [Google Scholar] [CrossRef]
  93. Slota, J.A.; Booth, S.A. MicroRNAs in neuroinflammation: Implications in disease pathogenesis, biomarker discovery and therapeutic applications. Non-Coding RNA 2019, 5, 35. [Google Scholar] [CrossRef]
  94. Zingale, V.D.; Gugliandolo, A.; Mazzon, E. MiR-155: An important regulator of neuroinflammation. Int. J. Mol. Sci. 2021, 23, 90. [Google Scholar] [CrossRef]
  95. Thomas, K.T.; Zakharenko, S.S. MicroRNAs in the Onset of Schizophrenia. Cells 2021, 10, 2679. [Google Scholar] [CrossRef]
  96. Ye, Y.; Hao, J.; Hong, Z.; Wu, T.; Ge, X.; Qian, B.; Chen, X.; Zhang, F. Downregulation of microRNA-145-5p in activated microglial exosomes promotes astrocyte proliferation by removal of Smad3 inhibition. Neurochem. Res. 2022, 47, 382–393. [Google Scholar] [CrossRef]
  97. Amoah, S.K.; Rodriguez, B.A.; Logothetis, C.N.; Chander, P.; Sellgren, C.M.; Weick, J.P.; Sheridan, S.D.; Jantzie, L.L.; Webster, M.J.; Mellios, N. Exosomal secretion of a psychosis-altered miRNA that regulates glutamate receptor expression is affected by antipsychotics. Neuropsychopharmacology 2020, 45, 656–665. [Google Scholar] [CrossRef]
  98. Kaurani, L.; Islam, M.R.; Heilbronner, U.; Krüger, D.M.; Zhou, J.; Methi, A.; Strauss, J.; Pradhan, R.; Schröder, S.; Burkhardt, S.; et al. Regulation of Zbp1 by miR-99b-5p in microglia controls the development of schizophrenia-like symptoms in mice. EMBO J. 2024, 43, 1420–1444. [Google Scholar] [CrossRef]
  99. Lavratti, C.; Dorneles, G.; Pochmann, D.; Peres, A.; Bard, A.; Schipper, L.d.L.; Lago, P.D.; Wagner, L.C.; Elsner, V.R. Exercise-induced modulation of histone H4 acetylation status and cytokines levels in patients with schizophrenia. Physiol. Behav. 2017, 168, 84–90. [Google Scholar] [CrossRef]
  100. Mahajan, G.J.; Vallender, E.J.; Garrett, M.R.; Challagundla, L.; Overholser, J.C.; Jurjus, G.; Dieter, L.; Syed, M.; Romero, D.G.; Benghuzzi, H.; et al. Altered neuro-inflammatory gene expression in hippocampus in major depressive disorder. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2018, 82, 177–186. [Google Scholar] [CrossRef]
  101. Tseng, C.-C.; Wang, S.-C.; Yang, Y.-C.; Fu, H.-C.; Chou, C.-K.; Kang, H.-Y.; Hung, Y.-Y. Aberrant histone modification of TNFAIP3, TLR4, TNIP2, miR-146a, and miR-155 in major depressive disorder. Mol. Neurobiol. 2023, 60, 4753–4760. [Google Scholar] [CrossRef]
  102. Wang, C.; Shen, D.; Hu, Y.; Chen, J.; Liu, J.; Huang, Y.; Yu, X.; Chu, H.; Zhang, C.; Yin, L.; et al. Selective targeting of class I HDAC reduces microglial inflammation in the entorhinal cortex of young APP/PS1 mice. Int. J. Mol. Sci. 2023, 24, 4805. [Google Scholar] [CrossRef]
  103. Weigelt, K.; Bergink, V.; Burgerhout, K.M.; Pescatori, M.; Wijkhuijs, A.; Drexhage, H.A. Down-regulation of inflammation-protective microRNAs 146a and 212 in monocytes of patients with postpartum psychosis. Brain Behav. Immun. 2013, 29, 147–155. [Google Scholar] [CrossRef]
  104. Akkouh, I.A.; Hughes, T.; Steen, V.M.; Glover, J.C.; Andreassen, O.A.; Djurovic, S.; Szabo, A. Transcriptome analysis reveals disparate expression of inflammation-related miRNAs and their gene targets in iPSC-astrocytes from people with schizophrenia. Brain Behav. Immun. 2021, 94, 235–244. [Google Scholar] [CrossRef]
  105. Miyano, T.; Mikkaichi, T.; Nakamura, K.; Yoshigae, Y.; Abernathy, K.; Ogura, Y.; Kiyosawa, N. Circulating microRNA profiles identify a patient subgroup with high inflammation and severe symptoms in Schizophrenia experiencing Acute psychosis. Int. J. Mol. Sci. 2024, 25, 4291. [Google Scholar] [CrossRef]
  106. Cattane, N.; Courtin, C.; Mombelli, E.; Maj, C.; Mora, C.; Etain, B.; Bellivier, F.; Marie-Claire, C.; Cattaneo, A. Transcriptomics and miRNomics data integration in lymphoblastoid cells highlights the key role of immune-related functions in lithium treatment response in Bipolar disorder. BMC Psychiatry 2022, 22, 665. [Google Scholar] [CrossRef]
  107. Hung, Y.-Y.; Wu, M.-K.; Tsai, M.-C.; Huang, Y.-L.; Kang, H.-Y. Aberrant expression of intracellular let-7e, miR-146a, and miR-155 correlates with severity of depression in patients with major depressive disorder and is ameliorated after antidepressant treatment. Cells 2019, 8, 647. [Google Scholar] [CrossRef]
  108. Sundquist, K.; Memon, A.A.; Palmér, K.; Sundquist, J.; Wang, X. Inflammatory proteins and miRNA-144-5p in patients with depression, anxiety, or stress-and adjustment disorders after psychological treatment. Cytokine 2021, 146, 155646. [Google Scholar] [CrossRef]
  109. Brás, J.P.; Pinto, S.; von Doellinger, O.; Prata, J.; Coelho, R.; Barbosa, M.A.; Almeida, M.I.; Santos, S.G. Combining inflammatory miRNA molecules as diagnostic biomarkers for depression: A clinical study. Front. Psychiatry 2023, 14, 1227618. [Google Scholar] [CrossRef]
  110. Oliveira, S.R.; Dionísio, P.A.; Gaspar, M.M.; Guedes, L.C.; Coelho, M.; Rosa, M.M.; Ferreira, J.J.; Amaral, J.D.; Rodrigues, C.M. miR-335 targets LRRK2 and mitigates inflammation in Parkinson’s disease. Front. Cell Dev. Biol. 2021, 9, 661461. [Google Scholar] [CrossRef]
  111. Mooney, C.; Parlante, A.; Canarutto, G.; Grigoli, A.; Scattoni, M.L.; Ricceri, L.; Jimenez-Mateos, E.M.; Sanz-Rodriguez, A.; Clementi, E.; Piazza, S.; et al. Deregulated mRNA and microRNA Expression Patterns in the Prefrontal Cortex of the BTBR Mouse Model of Autism. Mol. Neurobiol. 2025, 1–21. [Google Scholar] [CrossRef]
  112. Ianni, M.; Corraliza-Gomez, M.; Costa-Coelho, T.; Ferreira-Manso, M.; Inteiro-Oliveira, S.; Alemãn-Serrano, N.; Sebastião, A.M.; Garcia, G.; Diógenes, M.J.; Brites, D. Spatiotemporal Dysregulation of Neuron–Glia Related Genes and Pro-/Anti-Inflammatory miRNAs in the 5xFAD Mouse Model of Alzheimer’s Disease. Int. J. Mol. Sci. 2024, 25, 9475. [Google Scholar] [CrossRef]
  113. Mazziotta, C.; Tognon, M.; Martini, F.; Torreggiani, E.; Rotondo, J.C. Probiotics mechanism of action on immune cells and beneficial effects on human health. Cells 2023, 12, 184. [Google Scholar] [CrossRef]
  114. Borthakur, A.; Anbazhagan, A.N.; Kumar, A.; Raheja, G.; Singh, V.; Ramaswamy, K.; Dudeja, P.K. The probiotic Lactobacillus plantarum counteracts TNF-α-induced downregulation of SMCT1 expression and function. Am. J. Physiol. Gastrointest. Liver Physiol. 2010, 299, G928–G934. [Google Scholar] [CrossRef]
  115. Xu, J.; Zhou, L.; Chen, Z.; Wang, Y.; Xu, F.; Kuang, Q.; Zhang, Y.; Zheng, H. Bacillus coagulans and Clostridium butyricum synergistically alleviate depression in a chronic unpredictable mild stress mouse model through altering gut microbiota and prefrontal cortex gene expression. Front. Pharmacol. 2024, 15, 1393874. [Google Scholar] [CrossRef]
  116. Arseneault-Bréard, J.; Rondeau, I.; Gilbert, K.; Girard, S.-A.; Tompkins, T.A.; Godbout, R.; Rousseau, G. Combination of Lactobacillus helveticus R0052 and Bifidobacterium longum R0175 reduces post-myocardial infarction depression symptoms and restores intestinal permeability in a rat model. Br. J. Nutr. 2012, 107, 1793–1799. [Google Scholar] [CrossRef]
  117. Dhaliwal, J.; Singh, D.; Singh, S.; Pinnaka, A.K.; Boparai, R.; Bishnoi, M.; Kondepudi, K.; Chopra, K. Lactobacillus plantarum MTCC 9510 supplementation protects from chronic unpredictable and sleep deprivation-induced behaviour, biochemical and selected gut microbial aberrations in mice. J. Appl. Microbiol. 2018, 125, 257–269. [Google Scholar] [CrossRef]
  118. Parra, I.; Martínez, I.; Vásquez-Celaya, L.; Gongora-Alfaro, J.L.; Tizabi, Y.; Mendieta, L. Neuroprotective and immunomodulatory effects of probiotics in a rat model of Parkinson’s disease. Neurotox. Res. 2023, 41, 187–200. [Google Scholar] [CrossRef]
  119. Valvaikar, S.; Vaidya, B.; Bishnoi, M.; Kondepudi, K.K.; Sharma, S.S. Supplementation of probiotic Bifidobacterium breve Bif11 reverses neurobehavioural deficits, inflammatory changes and oxidative stress in Parkinson’s disease model. Neurochem. Int. 2024, 174, 105691. [Google Scholar] [CrossRef]
  120. Adıgüzel, E.; Çiçek, B.; Ünal, G.; Aydın, M.F.; Barlak-Keti, D. Probiotics and prebiotics alleviate behavioral deficits, inflammatory response, and gut dysbiosis in prenatal VPA-induced rodent model of autism. Physiol. Behav. 2022, 256, 113961. [Google Scholar] [CrossRef]
  121. Reininghaus, E.Z.; Platzer, M.; Kohlhammer-Dohr, A.; Hamm, C.; Mörkl, S.; Bengesser, S.A.; Fellendorf, F.T.; Lahousen-Luxenberger, T.; Leitner-Afschar, B.; Schöggl, H.; et al. PROVIT: Supplementary probiotic treatment and vitamin B7 in depression—A randomized controlled trial. Nutrients 2020, 12, 3422. [Google Scholar] [CrossRef]
  122. Hsu, Y.-C.; Huang, Y.-Y.; Tsai, S.-Y.; Kuo, Y.-W.; Lin, J.-H.; Ho, H.-H.; Chen, J.-F.; Hsia, K.-C.; Sun, Y. Efficacy of probiotic supplements on brain-derived neurotrophic factor, inflammatory biomarkers, oxidative stress and cognitive function in patients with Alzheimer’s dementia: A 12-week randomized, double-blind active-controlled study. Nutrients 2023, 16, 16. [Google Scholar] [CrossRef]
  123. Akhgarjand, C.; Vahabi, Z.; Shab-Bidar, S.; Anoushirvani, A.; Djafarian, K. The effects of probiotic supplements on oxidative stress and inflammation in subjects with mild and moderate Alzheimer’s disease: A randomized, double-blind, placebo-controlled study. Inflammopharmacology 2024, 32, 1413–1420. [Google Scholar] [CrossRef]
  124. Bevilacqua, A.; Campaniello, D.; Speranza, B.; Racioppo, A.; Sinigaglia, M.; Corbo, M.R. An update on prebiotics and on their health effects. Foods 2024, 13, 446. [Google Scholar] [CrossRef]
  125. Akram, W.; Garud, N.; Joshi, R. Role of inulin as prebiotics on inflammatory bowel disease. Drug Discov. Ther. 2019, 13, 1–8. [Google Scholar] [CrossRef]
  126. Wang, Z.; Zhang, X.; Zhu, L.; Yang, X.; He, F.; Wang, T.; Bao, T.; Lu, H.; Wang, H.; Yang, S. Inulin alleviates inflammation of alcoholic liver disease via SCFAs-inducing suppression of M1 and facilitation of M2 macrophages in mice. Int. Immunopharmacol. 2020, 78, 106062. [Google Scholar] [CrossRef]
  127. Chen, P.; Li, X.; Yu, Y.; Zhang, J.; Zhang, Y.; Li, C.; Li, J.; Li, K. Administration Time and Dietary Patterns Modified the Effect of Inulin on CUMS-Induced Anxiety and Depression. Mol. Nutr. Food Res. 2023, 67, 2200566. [Google Scholar] [CrossRef]
  128. Lin, S.; Li, Q.; Xu, Z.; Chen, Z.; Tao, Y.; Tong, Y.; Wang, T.; Chen, S.; Wang, P. Detection of the role of intestinal flora and tryptophan metabolism involved in antidepressant-like actions of crocetin based on a multi-omics approach. Psychopharmacology 2022, 239, 3657–3677. [Google Scholar] [CrossRef]
  129. Guo, L.; Xiao, P.; Zhang, X.; Yang, Y.; Yang, M.; Wang, T.; Lu, H.; Tian, H.; Wang, H.; Liu, J. Inulin ameliorates schizophrenia via modulation of the gut microbiota and anti-inflammation in mice. Food Funct. 2021, 12, 1156–1175. [Google Scholar] [CrossRef]
  130. Wang, L.; Wang, Z.; Lan, Y.; Tuo, Y.; Ma, S.; Liu, X. Inulin attenuates blood–brain barrier permeability and alleviates behavioral disorders by modulating the TLR4/MyD88/NF-κB pathway in mice with chronic stress. J. Agric. Food Chem. 2023, 71, 13325–13337. [Google Scholar] [CrossRef]
  131. Szala-Rycaj, J.; Szewczyk, A.; Zagaja, M.; Kaczmarczyk-Ziemba, A.; Maj, M.; Andres-Mach, M. The Influence of topinambur and inulin preventive supplementation on microbiota, anxious behavior, cognitive functions and neurogenesis in mice exposed to the chronic unpredictable mild stress. Nutrients 2023, 15, 2041. [Google Scholar] [CrossRef]
  132. Zou, H.; Gao, H.; Liu, Y.; Zhang, Z.; Zhao, J.; Wang, W.; Ren, B.; Tan, X. Dietary inulin alleviated constipation induced depression and anxiety-like behaviors: Involvement of gut microbiota and microbial metabolite short-chain fatty acid. Int. J. Biol. Macromol. 2024, 259, 129420. [Google Scholar] [CrossRef]
  133. Buchanan, R.W.; Werkheiser, A.E.M.; Michel, H.B.; Zaranski, J.M.; Glassman, M.; Adams, H.A.P.; Vyas, G.D.; Blatt, F.; Pilli, N.R.; Pan, Y.; et al. Prebiotic Treatment in People With Schizophrenia. J. Clin. Psychopharmacol. 2024, 44, 457–461. [Google Scholar] [CrossRef]
  134. Liu, J.; Fang, Y.; Cui, L.; Wang, Z.; Luo, Y.; Gao, C.; Ge, W.; Huang, T.; Wen, J.; Zhou, T. Butyrate emerges as a crucial effector of Zhi-Zi-Chi decoctions to ameliorate depression via multiple pathways of brain-gut axis. Biomed. Pharmacother. 2022, 149, 112861. [Google Scholar] [CrossRef]
  135. de Paiva, I.H.R.; Maciel, L.M.; da Silva, R.S.; Mendonça, I.P.; de Souza, J.R.B.; Peixoto, C.A. Prebiotics modulate the microbiota–gut–brain axis and ameliorate anxiety and depression-like behavior in HFD-fed mice. Food Res. Int. 2024, 182, 114153. [Google Scholar] [CrossRef]
  136. Xiong, L.; Mao, M.; Shu, Q. A preliminary study on the diversity of butyrate-producing bacteria in response to the treatment of depression with Xiaoyaosan. Lett. Appl. Microbiol. 2022, 75, 844–856. [Google Scholar] [CrossRef]
  137. Xiong, L.; Wu, Y.; Shu, Q.; Xiong, W. The pharmacological mechanism of Xiaoyaosan polysaccharide reveals improvement of CUMS-induced depression-like behavior by carbon source-triggered butyrate-producing bacteria. J. Appl. Microbiol. 2023, 134, lxad052. [Google Scholar] [CrossRef]
  138. Liu, Y.; Dai, C.; Wang, C.; Wang, J.; Yan, W.; Luo, M.; Dong, J.; Li, X.; Liu, X.; Lan, Y. Raspberry Ketone Prevents LPS-Induced Depression-Like Behaviors in Mice by Inhibiting TLR-4/NF-κB Signaling Pathway via the Gut-Brain Axis. Mol. Nutr. Food Res. 2024, 68, 2400090. [Google Scholar] [CrossRef]
  139. Prince, N.; Marzal, L.N.P.; Markidi, A.; Ahmed, S.; Adolfs, Y.; Pasterkamp, R.J.; Kumar, H.; Roeselers, G.; Garssen, J.; Kraneveld, A.D.; et al. Prebiotic diet normalizes aberrant immune and behavioral phenotypes in a mouse model of autism spectrum disorder. Acta Pharmacol. Sin. 2024, 45, 1591–1603. [Google Scholar] [CrossRef]
  140. Sarti, G.; Traini, C.; Magni, G.; Attorre, S.; Tognozzi, G.; Calussi, E.; Giovannini, M.G.; Vannucchi, M.G.; Lana, D. Chronic administration of prebiotics and probiotics prevent pathophysiological hallmarks of Alzheimer’s disease in the cortex of APP/PS1 mice. Front. Pharmacol. 2025, 16, 1596469. [Google Scholar] [CrossRef]
  141. Bedarf, J.R.; Romano, S.; Heinzmann, S.S.; Duncan, A.; Traka, M.H.; Ng, D.; Segovia-Lizano, D.; Simon, M.-C.; Narbad, A.; Wüllner, U.; et al. A prebiotic dietary pilot intervention restores faecal metabolites and may be neuroprotective in Parkinson’s Disease. npj Park. Dis. 2025, 11, 66. [Google Scholar] [CrossRef]
  142. Maiuolo, J.; Bulotta, R.M.; Ruga, S.; Nucera, S.; Macrì, R.; Scarano, F.; Oppedisano, F.; Carresi, C.; Gliozzi, M.; Musolino, V.; et al. The postbiotic properties of butyrate in the modulation of the gut microbiota: The potential of its combination with polyphenols and dietary fibers. Int. J. Mol. Sci. 2024, 25, 6971. [Google Scholar] [CrossRef]
  143. Głowacka, P.; Oszajca, K.; Pudlarz, A.; Szemraj, J.; Witusik-Perkowska, M. Postbiotics as Molecules Targeting Cellular Events of Aging Brain—The Role in Pathogenesis, Prophylaxis and Treatment of Neurodegenerative Diseases. Nutrients 2024, 16, 2244. [Google Scholar] [CrossRef]
  144. Guo, C.; Huo, Y.-J.; Li, Y.; Han, Y.; Zhou, D. Gut-brain axis: Focus on gut metabolites short-chain fatty acids. World J. Clin. Cases 2022, 10, 1754. [Google Scholar] [CrossRef]
  145. Stilling, R.M.; Van De Wouw, M.; Clarke, G.; Stanton, C.; Dinan, T.G.; Cryan, J.F. The neuropharmacology of butyrate: The bread and butter of the microbiota-gut-brain axis? Neurochem. Int. 2016, 99, 110–132. [Google Scholar] [CrossRef]
  146. Huuskonen, J.; Suuronen, T.; Nuutinen, T.; Kyrylenko, S.; Salminen, A. Regulation of microglial inflammatory response by sodium butyrate and short-chain fatty acids. Br. J. Pharmacol. 2004, 141, 874–880. [Google Scholar] [CrossRef]
  147. Wei, H.; Yu, C.; Zhang, C.; Ren, Y.; Guo, L.; Wang, T.; Chen, F.; Li, Y.; Zhang, X.; Wang, H.; et al. Butyrate ameliorates chronic alcoholic central nervous damage by suppressing microglia-mediated neuroinflammation and modulating the microbiome-gut-brain axis. Biomed. Pharmacother. 2023, 160, 114308. [Google Scholar] [CrossRef]
  148. Valvassori, S.S.; Dal-Pont, G.C.; Steckert, A.V.; Varela, R.B.; Lopes-Borges, J.; Mariot, E.; Resende, W.R.; Arent, C.O.; Carvalho, A.F.; Quevedo, J. Sodium butyrate has an antimanic effect and protects the brain against oxidative stress in an animal model of mania induced by ouabain. Psychiatry Res. 2016, 235, 154–159. [Google Scholar] [CrossRef]
  149. Kalkan, A.E.; BinMowyna, M.N.; Raposo, A.; Ahmad, M.F.; Ahmed, F.; Otayf, A.Y.; Carrascosa, C.; Saraiva, A.; Karav, S. Beyond the Gut: Unveiling Butyrate’s Global Health Impact Through Gut Health and Dysbiosis-Related Conditions: A Narrative Review. Nutrients 2025, 17, 1305. [Google Scholar] [CrossRef]
  150. Chen, G.; Ran, X.; Li, B.; Li, Y.; He, D.; Huang, B.; Fu, S.; Liu, J.; Wang, W. Sodium butyrate inhibits inflammation and maintains epithelium barrier integrity in a TNBS-induced inflammatory bowel disease mice model. EBioMedicine 2018, 30, 317–325. [Google Scholar] [CrossRef]
  151. Arpaia, N.; Campbell, C.; Fan, X.; Dikiy, S.; Van Der Veeken, J.; Deroos, P.; Liu, H.; Cross, J.R.; Pfeffer, K.; Coffer, P.J.; et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature 2013, 504, 451–455. [Google Scholar] [CrossRef]
  152. Fu, S.-P.; Wang, J.-F.; Xue, W.-J.; Liu, H.-M.; Liu, B.-R.; Zeng, Y.-L.; Li, S.-N.; Huang, B.-X.; Lv, Q.-K.; Wang, W.; et al. Anti-inflammatory effects of BHBA in both in vivo and in vitro Parkinson’s disease models are mediated by GPR109A-dependent mechanisms. J. Neuroinflamm. 2015, 12, 9. [Google Scholar] [CrossRef]
  153. Guo, T.-T.; Zhang, Z.; Sun, Y.; Zhu, R.-Y.; Wang, F.-X.; Ma, L.-J.; Jiang, L.; Liu, H.-D. Neuroprotective effects of sodium butyrate by restoring gut microbiota and inhibiting TLR4 signaling in mice with MPTP-induced Parkinson’s disease. Nutrients 2023, 15, 930. [Google Scholar] [CrossRef]
  154. Moretti, M.; Valvassori, S.S.; Varela, R.B.; Ferreira, C.L.; Rochi, N.; Benedet, J.; Scaini, G.; Kapczinski, F.; Streck, E.L.; Zugno, A.I.; et al. Behavioral and neurochemical effects of sodium butyrate in an animal model of mania. Behav. Pharmacol. 2011, 22, 766–772. [Google Scholar] [CrossRef]
  155. Qiu, J.; Liu, R.; Ma, Y.; Li, Y.; Chen, Z.; He, H.; Chen, J.; Tong, L.; Huang, C.; You, Q. Lipopolysaccharide-induced depression-like behaviors is ameliorated by sodium butyrate via inhibiting neuroinflammation and oxido-nitrosative stress. Pharmacology 2020, 105, 550–560. [Google Scholar] [CrossRef]
  156. Wang, Z.; Ma, X.; Shi, W.; Zhu, W.; Feng, X.; Xin, H.; Zhang, Y.; Cong, B.; Li, Y. The Gut Microbiota Metabolite Butyrate Modulates Acute Stress-Induced Ferroptosis in the Prefrontal Cortex via the Gut–Brain Axis. Int. J. Mol. Sci. 2025, 26, 1698. [Google Scholar] [CrossRef]
  157. Firoozi, D.; Masoumi, S.J.; Mohammad-Kazem Hosseini Asl, S.; Fararouei, M.; Jamshidi, S. Effects of Short Chain Fatty Acid-Butyrate Supplementation on the Disease Severity, Inflammation, and Psychological Factors in Patients With Active Ulcerative Colitis: A Double-Blind Randomized Controlled Trial. J. Nutr. Metab. 2025, 2025, 3165876. [Google Scholar] [CrossRef]
  158. Tong, L.-c.; Wang, Y.; Wang, Z.-b.; Liu, W.-y.; Sun, S.; Li, L.; Su, D.-f.; Zhang, L.-c. Propionate ameliorates dextran sodium sulfate-induced colitis by improving intestinal barrier function and reducing inflammation and oxidative stress. Front. Pharmacol. 2016, 7, 253. [Google Scholar] [CrossRef]
  159. Filippone, A.; Lanza, M.; Campolo, M.; Casili, G.; Paterniti, I.; Cuzzocrea, S.; Esposito, E. The anti-inflammatory and antioxidant effects of sodium propionate. Int. J. Mol. Sci. 2020, 21, 3026. [Google Scholar] [CrossRef]
  160. Dai, H.-y.; Zhang, Z.-x.; Tan, C.; Xian, X.; Ji, D.; Yang, J.; Sun, J.; Yao, H. Propionic acid ameliorates cognitive function through immunomodulatory effects on Th17 cells in perioperative neurocognitive disorders. Heliyon 2024, 10, e28817. [Google Scholar] [CrossRef]
  161. Hao, C.; Gao, Z.; Liu, X.; Rong, Z.; Jia, J.; Kang, K.; Guo, W.; Li, J. Intravenous administration of sodium propionate induces antidepressant or prodepressant effect in a dose dependent manner. Sci. Rep. 2020, 10, 19917. [Google Scholar] [CrossRef]
  162. Li, J.; Hou, L.; Wang, C.; Jia, X.; Qin, X.; Wu, C. Short term intrarectal administration of sodium propionate induces antidepressant-like effects in rats exposed to chronic unpredictable mild stress. Front. Psychiatry 2018, 9, 454. [Google Scholar] [CrossRef]
  163. Behrens, L.M.P.; Gasparotto, J.; Rampelotto, P.H.; Escalona, M.A.R.; Silva, L.d.S.d.; Carazza-Kessler, F.G.; Barbosa, C.P.; Campos, M.S.; Dorn, M.; Gelain, D.P. Sodium propionate oral supplementation ameliorates depressive-like behavior through gut microbiome and histone 3 epigenetic regulation. J. Nutr. Biochem. 2024, 130, 109660. [Google Scholar] [CrossRef]
  164. Chen, H.; Liu, S.; Ge, B.; Zhou, D.; Li, M.; Li, W.; Ma, F.; Liu, Z.; Ji, Y.; Huang, G. Effects of folic acid and vitamin B12 supplementation on cognitive impairment and inflammation in patients with Alzheimer’s disease: A randomized, single-blinded, placebo-controlled trial. J. Prev. Alzheimer’s Dis. 2021, 8, 249–256. [Google Scholar] [CrossRef]
  165. Shaikh, A.; Roy, H. Folate deprivation induced neuroinflammation impairs cognition. Neurosci. Lett. 2023, 807, 137264. [Google Scholar] [CrossRef]
  166. Śliwiński, W.; Gawlik-Kotelnicka, O. Circulating B vitamins metabolites in depressive disorders-connections with the microbiota-gut-brain axis. Behav. Brain Res. 2024, 472, 115145. [Google Scholar] [CrossRef]
  167. Menegas, S.; Dal-Pont, G.C.; Cararo, J.H.; Varela, R.B.; Aguiar-Geraldo, J.M.; Possamai-Della, T.; Andersen, M.L.; Quevedo, J.; Valvassori, S.S. Efficacy of folic acid as an adjunct to lithium therapy on manic-like behaviors, oxidative stress and inflammatory parameters in an animal model of mania. Metab. Brain Dis. 2020, 35, 413–425. [Google Scholar] [CrossRef]
  168. Khosravi, M.; Sotoudeh, G.; Amini, M.; Raisi, F.; Mansoori, A.; Hosseinzadeh, M. The relationship between dietary patterns and depression mediated by serum levels of Folate and vitamin B12. BMC Psychiatry 2020, 20, 63. [Google Scholar] [CrossRef]
  169. Jia, Y.; Li, J.; Wang, Y.; Ma, Y.; Chen, L.; Zhang, H.; Xue, M.; Liang, H. Folic acid rescues dopaminergic neurons in MPTP-induced mice by inhibiting the NLRP3 inflammasome and ameliorating mitochondrial impairment. J. Agric. Food Chem. 2024, 72, 5734–5745. [Google Scholar] [CrossRef]
  170. Chen, H.; Liu, S.; Ji, L.; Wu, T.; Ji, Y.; Zhou, Y.; Zheng, M.; Zhang, M.; Xu, W.; Huang, G. Folic acid supplementation mitigates Alzheimer’s disease by reducing inflammation: A randomized controlled trial. Mediat. Inflamm. 2016, 2016, 5912146. [Google Scholar] [CrossRef]
  171. Baker, J.A.; Bodnar, T.S.; Breit, K.R.; Weinberg, J.; Thomas, J.D. Choline supplementation alters hippocampal cytokine levels in adolescence and adulthood in an animal model of fetal alcohol spectrum disorders. Cells 2023, 12, 546. [Google Scholar] [CrossRef]
  172. Dave, N.; Judd, J.M.; Decker, A.; Winslow, W.; Sarette, P.; Espinosa, O.V.; Tallino, S.; Bartholomew, S.K.; Bilal, A.; Sandler, J.; et al. Dietary choline intake is necessary to prevent systems-wide organ pathology and reduce Alzheimer’s disease hallmarks. Aging Cell 2023, 22, e13775. [Google Scholar] [CrossRef]
  173. Egilmez, C.B.; Pazarlar, B.A.; Erdogan, M.A.; Uyanikgil, Y.; Erbas, O. Choline chloride shows gender-dependent positive effects on social deficits, learning/memory impairments, neuronal loss and neuroinflammation in the lipopolysaccharide-induced rat model of autism. Int. J. Dev. Neurosci. 2024, 84, 392–405. [Google Scholar] [CrossRef]
  174. Li, J.; Kang, X.; Zhang, L.; Luo, J.; Zhang, D. Dietary choline is inversely associated with depressive symptoms: A cross-sectional study of the National Health and Nutrition Examination Survey (NHANES) 2011 to 2018. J. Affect. Disord. 2022, 301, 23–29. [Google Scholar] [CrossRef]
  175. Zhao, G.; He, F.; Wu, C.; Li, P.; Li, N.; Deng, J.; Zhu, G.; Ren, W.; Peng, Y. Betaine in inflammation: Mechanistic aspects and applications. Front. Immunol. 2018, 9, 1070. [Google Scholar] [CrossRef]
  176. Ueland, P.M. Choline and betaine in health and disease. J. Inherit. Metab. Dis. 2011, 34, 3–15. [Google Scholar] [CrossRef]
  177. Tolmunen, T.; Hintikka, J.; Voutilainen, S.; Ruusunen, A.; Alfthan, G.; Nyyssönen, K.; Viinamäki, H.; Kaplan, G.A.; Salonen, J.T. Association between depressive symptoms and serum concentrations of homocysteine in men: A population study. Am. J. Clin. Nutr. 2004, 80, 1574–1578. [Google Scholar] [CrossRef]
  178. Zhao, N.; Yang, Y.; Chen, C.; Jing, T.; Hu, Y.; Xu, H.; Wang, S.; He, Y.; Liu, E.; Cui, J. Betaine supplementation alleviates dextran sulfate sodium-induced colitis via regulating the inflammatory response, enhancing the intestinal barrier, and altering gut microbiota. Food Funct. 2022, 13, 12814–12826. [Google Scholar] [CrossRef]
  179. Liu, W.; Zhong, X.; Yi, Y.; Xie, L.; Zhou, W.; Cao, W.; Chen, L. Prophylactic Effects of Betaine on Depression and Anxiety Behaviors in Mice with Dextran Sulfate Sodium-Induced Colitis. J. Agric. Food Chem. 2024, 72, 21041–21051. [Google Scholar] [CrossRef]
  180. Hui, R.; Xu, J.; Zhou, M.; Xie, B.; Zhou, M.; Zhang, L.; Cong, B.; Ma, C.; Wen, D. Betaine improves METH-induced depressive-like behavior and cognitive impairment by alleviating neuroinflammation via NLRP3 inflammasome inhibition. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2024, 135, 111093. [Google Scholar] [CrossRef]
  181. Zhang, Y.; Jia, J. Betaine mitigates amyloid-β-associated neuroinflammation by suppressing the NLRP3 and NF-κB signaling pathways in microglial cells. J. Alzheimer’s Dis. 2023, 94, S9–S19. [Google Scholar] [CrossRef]
  182. Tidman, M.M.; White, D.R.; White, T.A. Impact of a keto diet on symptoms of Parkinson’s disease, biomarkers, depression, anxiety and quality of life: A longitudinal study. Neurodegener. Dis. Manag. 2024, 14, 97–110. [Google Scholar] [CrossRef]
  183. Ozan, E.; Chouinard, V.-A.; Palmer, C.M. The ketogenic diet as a treatment for mood disorders. Curr. Treat. Options Psychiatry 2024, 11, 163–176. [Google Scholar] [CrossRef]
  184. Jiang, Y.; Chen, Y.; Chen, Y.; Gong, X.; Chen, Z.; Zhang, X. Ketogenic Diet and Gut Microbiota: Exploring New Perspectives on Cognition and Mood. Foods 2025, 14, 1215. [Google Scholar] [CrossRef]
  185. Joseph, J.; Depp, C.; Shih, P.-a.B.; Cadenhead, K.S.; Schmid-Schönbein, G. Modified mediterranean diet for enrichment of short chain fatty acids: Potential adjunctive therapeutic to target immune and metabolic dysfunction in schizophrenia? Front. Neurosci. 2017, 11, 155. [Google Scholar] [CrossRef]
  186. Mentzelou, M.; Dakanalis, A.; Vasios, G.K.; Gialeli, M.; Papadopoulou, S.K.; Giaginis, C. The relationship of ketogenic diet with neurodegenerative and psychiatric diseases: A scoping review from basic research to clinical practice. Nutrients 2023, 15, 2270. [Google Scholar] [CrossRef]
  187. Dąbek, A.; Wojtala, M.; Pirola, L.; Balcerczyk, A. Modulation of cellular biochemistry, epigenetics and metabolomics by ketone bodies. Implications of the ketogenic diet in the physiology of the organism and pathological states. Nutrients 2020, 12, 788. [Google Scholar] [CrossRef]
  188. Liang, J.; Zhang, J.; Sun, J.; Liang, Q.; Zhan, Y.; Yang, Z.; Zhang, Y.; Jin, L.; Hu, C.; Zhao, Y.-T. Ketogenic diet attenuates neuroinflammation and restores hippocampal neurogenesis to improve CUMS induced depression-like behavior in mice. Food Funct. 2025, 16, 3408–3422. [Google Scholar] [CrossRef]
  189. Di Lucente, J.; Persico, G.; Zhou, Z.; Jin, L.-W.; Ramsey, J.J.; Rutkowsky, J.M.; Montgomery, C.M.; Tomilov, A.; Kim, K.; Giorgio, M.; et al. Ketogenic diet and BHB rescue the fall of long-term potentiation in an Alzheimer’s mouse model and stimulates synaptic plasticity pathway enzymes. Commun. Biol. 2024, 7, 195. [Google Scholar] [CrossRef]
  190. Zhu, Y.; Tang, X.; Cheng, Z.; Dong, Q.; Ruan, G. The Anti-Inflammatory Effect of Preventive Intervention with Ketogenic Diet Mediated by the Histone Acetylation of mGluR5 Promotor Region in Rat Parkinson’s Disease Model: A Dual-Tracer PET Study. Park. Dis. 2022, 2022, 3506213. [Google Scholar] [CrossRef]
  191. Allan, N.P.; Yamamoto, B.Y.; Kunihiro, B.P.; Nunokawa, C.K.L.; Rubas, N.C.; Wells, R.K.; Umeda, L.; Phankitnirundorn, K.; Torres, A.; Peres, R.; et al. Ketogenic diet induced shifts in the gut microbiome associate with changes to inflammatory cytokines and brain-related miRNAs in children with autism Spectrum disorder. Nutrients 2024, 16, 1401. [Google Scholar] [CrossRef]
  192. Schweickart, A.; Batra, R.; Neth, B.J.; Martino, C.; Shenhav, L.; Zhang, A.R.; Shi, P.; Karu, N.; Huynh, K.; Meikle, P.J.; et al. Serum and CSF metabolomics analysis shows Mediterranean Ketogenic Diet mitigates risk factors of Alzheimer’s disease. NPJ Metab. Health Dis. 2024, 2, 15. [Google Scholar] [CrossRef]
  193. Lu, Q.; Lai, J.; Lu, H.; Ng, C.; Huang, T.; Zhang, H.; Ding, K.; Wang, Z.; Jiang, J.; Hu, J.; et al. Gut microbiota in bipolar depression and its relationship to brain function: An advanced exploration. Front. Psychiatry 2019, 10, 784. [Google Scholar] [CrossRef]
  194. Li, X.; Fan, X.; Yuan, X.; Pang, L.; Hu, S.; Wang, Y.; Huang, X.; Song, X. The role of butyric acid in treatment response in drug-naive first episode schizophrenia. Front. Psychiatry 2021, 12, 724664. [Google Scholar] [CrossRef]
  195. Zheng, J.; Lin, Z.; Ko, C.-Y.; Xu, J.-H.; Lin, Y.; Wang, J. Analysis of gut microbiota in patients with exacerbated symptoms of schizophrenia following therapy with amisulpride: A pilot study. Behav. Neurol. 2022, 2022, 4262094. [Google Scholar] [CrossRef]
  196. Weng, S.; Zheng, J.; Lin, Y.; Fang, H.; Ko, C.-Y. Therapeutic effects of amisulpride in male schizophrenics: Role of short-chain fatty acids and gene expression changes. Physiol. Behav. 2025, 294, 114864. [Google Scholar] [CrossRef]
  197. Huang, S.; Hu, S.; Liu, S.; Tang, B.; Liu, Y.; Tang, L.; Lei, Y.; Zhong, L.; Yang, S.; He, S. Lithium carbonate alleviates colon inflammation through modulating gut microbiota and Treg cells in a GPR43-dependent manner. Pharmacol. Res. 2022, 175, 105992. [Google Scholar] [CrossRef]
  198. Lopes, L.d.S.; da Silva, J.S.; da Luz, J.M.R.; Silva, M.d.C.S.d.; Lima, H.S.; Rocha, G.C.; Mantovani, H.C.; Kasuya, M.C.M. Intestinal microbial diversity of swines fed with different sources of lithium. 3 Biotech 2024, 14, 102. [Google Scholar]
  199. Lukić, I.; Getselter, D.; Ziv, O.; Oron, O.; Reuveni, E.; Koren, O.; Elliott, E. Antidepressants affect gut microbiota and Ruminococcus flavefaciens is able to abolish their effects on depressive-like behavior. Transl. Psychiatry 2019, 9, 133. [Google Scholar] [CrossRef]
  200. Sun, L.; Zhang, H.; Cao, Y.; Wang, C.; Zhao, C.; Wang, H.; Cui, G.; Wang, M.; Pan, Y.; Shi, Y. Fluoxetine ameliorates dysbiosis in a depression model induced by chronic unpredicted mild stress in mice. Int. J. Med. Sci. 2019, 16, 1260. [Google Scholar] [CrossRef]
  201. Zhang, W.; Qu, W.; Wang, H.; Yan, H. Antidepressants fluoxetine and amitriptyline induce alterations in intestinal microbiota and gut microbiome function in rats exposed to chronic unpredictable mild stress. Transl. Psychiatry 2021, 11, 131. [Google Scholar] [CrossRef]
  202. Jiang, Y.; Qu, Y.; Shi, L.; Ou, M.; Du, Z.; Zhou, Z.; Zhou, H.; Zhu, H. The role of gut microbiota and metabolomic pathways in modulating the efficacy of SSRIs for major depressive disorder. Transl. Psychiatry 2024, 14, 493. [Google Scholar] [CrossRef]
  203. Yang, Q.; Luo, L.; Sun, T.; Yang, L.; Cheng, L.-F.; Wang, Y.; Liu, Q.-Q.; Liu, A.; Liu, H.-Y.; Zhao, M.-G.; et al. Chronic minocycline treatment exerts antidepressant effect, inhibits neuroinflammation, and modulates gut microbiota in mice. Psychopharmacology 2020, 237, 3201–3213. [Google Scholar] [CrossRef]
  204. Li, H.; Xiang, Y.; Zhu, Z.; Wang, W.; Jiang, Z.; Zhao, M.; Cheng, S.; Pan, F.; Liu, D.; Ho, R.C.; et al. Rifaximin-mediated gut microbiota regulation modulates the function of microglia and protects against CUMS-induced depression-like behaviors in adolescent rat. J. Neuroinflamm. 2021, 18, 254. [Google Scholar] [CrossRef]
Figure 1. Association between changes in the gut microbiota due to gut dysbiosis and the development of neuropsychiatric disorders through increasing neuroinflammation.
Figure 1. Association between changes in the gut microbiota due to gut dysbiosis and the development of neuropsychiatric disorders through increasing neuroinflammation.
Cells 14 01027 g001
Figure 2. Potential mechanisms of antipsychotics, antidepressants, and antibiotics in the treatment of neuropsychiatric disorders by modulating neuroinflammation through altering the gut microbiome composition and epigenetic mechanisms.
Figure 2. Potential mechanisms of antipsychotics, antidepressants, and antibiotics in the treatment of neuropsychiatric disorders by modulating neuroinflammation through altering the gut microbiome composition and epigenetic mechanisms.
Cells 14 01027 g002
Table 1. Evidence supporting neuroinflammation in different neuropsychiatric disorders.
Table 1. Evidence supporting neuroinflammation in different neuropsychiatric disorders.
Neuropsychiatric DisordersStudy Subjects/SampleImmune CellsMain OutcomesRef.
Schizophrenia (SCZ)Human/bloodNeutrophils and lymphocyteAssociation between greater proportions of neutrophils and neutrophil-to-lymphocyte ratio and higher PANSS-total scores in SCZ[36]
SCZ and Bipolar Disorder (BD)Human/ postmortem midbrain tissueParenchymal CD163+ cellsAssociation between higher proportions of parenchymal CD163+ cells and CD163 protein level, and a reduction in Tyrosine Hydroxylase (TR) in the substantia nigra[37]
Autism spectrum disorder (ASD)Human/bloodB cellsReduced expression of anti-inflammatory cytokine IL-10 and elevated expression of TNF-α and IL-6 due to TLR4 activation in B cells in ASD[38]
Parkinson disease (PD)Human/bloodCentral memory CD4+ T cells, naive CD4+ and naive CD8+ T cellsReductions in naive B cells, naive CD4+ and naive CD8+ T cells; elevation of TNF-α–producing CD19+ B cells, central memory CD4+ T cells, IL-17–producing CD4+ Th17 cells, IL-4–producing CD4+ Th2 cells, and IFN-γ–producing CD8+ T cells in PD [39]
PDHuman/bloodCD8+ T cell and natural killer (NK) cellsNegative association between Montreal Cognitive Assessment scores and Intracellular TNF-α in naïve CD8+ T-cell cluster C16 (CD57− naïve CD8+ T) and NK cell cluster C32 (CD57− CD28− NK)[40]
PDHuman/bloodMyeloid dendritic cells and CD27+ CD4+ memory T cellsRelation between elevated genetic risk for PD and greater levels of myeloid dendritic cells and CD27+ CD4+ memory T cells[41]
Alzheimer’s disease (AD)Human/blood and brain NK cells and CD4 T cellsReduced NK cells and increased CD4 T cells in AD[42]
ADHuman/bloodCD4+ T, CD8+ T, B and, NK cells, monocytes–macrophagesHigh-frequency amplification clonotypes in T and B cells and reduced T cells diversity in AD[43]
ADHuman/bloodMonocytes, regulatory T cells (Treg), and B cellsAD risk in linked to CD33 on CD14+ monocyte; AD risk is inversely linked to secreting CD4 regulatory T cells, %CD4 regulatory T cells and CD25 on switched memory B cells[44]
Major depressive disorder (MDD)Human/bloodT regulatory cellsAcute phase of severe MDD is linked to breakdown of immune tolerance, and CD40L activation; elevated levels of CD3+ CD71+, CD3+ CD40L+, CD4+ CD71+, CD4+ CD40L+, CD4+ HLADR+, and CD8+ HLADR+ T cells in MDD[45]
Table 2. Potential roles of inflammatory gut bacteria in the pathogenesis of specific neuropsychiatric disorders.
Table 2. Potential roles of inflammatory gut bacteria in the pathogenesis of specific neuropsychiatric disorders.
Neuropsychiatric DisordersStudy and Sample Type Alerted Pro- or Anti-Inflammatory Bacteria and Their Epigenetic MetabolitesMain FindingRef.
Alzheimer’s disease (AD)Human/fecal and blood samplesinflammatory bacteria such as Synergistetes and the Christensenellaceae familyIncreased abundance of inflammatory bacteria and elevated levels of inflammatory cytokines in AD[54]
ADHuman/fecal samplesPro-inflammatory (e.g., Escherichia/Shigella, Clostridium_sensu_stricto_1
and anti-inflammatory genera (Faecalibacterium, Blautia, Bacteroides, and Roseburia
Higher levels of pro-inflammatory bacteria and lower levels of anti-inflammatory genera and total SCFAs in AD[55]
Parkinson’s disease (PD)Human/
fecal samples
Anti-inflammatory butyrate-producing bacteria, including Roseburia intestinalis, Faecalibacterium prausnitzii, Anaerostipes hadrus, and Eubacterium rectaleDepletion of anti-inflammatory butyrate-producing bacteria, derangements in SCFA-synthesis, and increased neuro-inflammation due to intestinal inflammation[56]
Autism spectrum disorder (ASD)Human/Stool and blood samplesAnti-inflammatory bacteria like Lachnospiraceae familyNegative correlation between pro-inflammatory cytokines IFN-γ and IL-6 and Lachnospiraceae family[57]
SCZHuman/fecal samplesButyrate-producing and succinate-producing bacteria (Phascolarctobacterium succinatutens and Paraprevotella clara)Association between increased levels of succinate-producing bacteria and inflammation[58]
SCZHuman/fecal and blood samplesPro-inflammatory genera such as Proteus and Succinivibrio and anti-inflammatory butyrate-producing bacteriaReduced levels of butyrate-producing bacteria (e.g., Faecalibacterium, Blautia, Alistipes, Gemmiger, and Butyricicoccus) and elevated levels of genera such as Proteus and Succinivibrio; positive correlations between pro-inflammatory cytokines (IL-1β, IL-2, IL-6, and TNF-α) and Succinivibrio[59]
BDHuman/stool samplesPro-inflammatory genera, like StreptococcusAssociation between higher IL-6 levels and greater abundance of pro-inflammatory bacteria, like Streptococcus[60]
BDHuman/Stool samplesPro-inflammatory genera, like FlavonifractorPositive correlation between Flavonifractor and oxidative stress and inflammation[61]
Depressive BD IIHuman/fecal samplesPro -inflammatory bacteria (e.g., Proteobacteria, Enterobacteriaceae, Porphyromonadaceae, and Pseudescherichia)Higher levels of Proteobacteria, Enterobacteriaceae, Porphyromonadaceae, and Pseudescherichia, along with inflammatory cytokines in unmedicated depressive BD II vs. controls[62]
DepressionHuman/fecal samplesGut anti-inflammatory (Faecalibacterium and Subdoligranulum) and pro-inflammatory (Flavonifractor and Gammaproteobacteria) bacteria Decreased abundance of Faecalibacterium and Subdoligranulum and increased abundance of Flavonifractor and Gammaproteobacteria in depressed vs. control subjects[63]
DepressionHuman/fecal samplePro-inflammatory genera such as Streptococcus and anti-inflammatory genera, like FaecalibacteriumElevated abundance of Streptococcus and Escherichia/Shigella, and reduced abundance of Faecalibacterium; higher levels of pro-inflammatory cytokines like IL-17, and lower levels of anti-inflammatory cytokines, like IFN-γ[64]
DepressionHuman/fecal and blood sampleAnti-inflammatory butyrate-producing bacteria such as Turicibacter, Roseburia, and ClostridiumReduced levels of anti-inflammatory bacteria; negative correlation between Turicibacter and Turicibacteraceae and IL-1β and IL-6 levels[65]
Inflammatory depressionHuman and mouse/fecal, blood, and colon biopsy samplesAnti-inflammatory bacteria such as Clostridium and FaecalibacteriumElevated levels of Bacteroides and reduced levels of Clostridium and Faecalibacterium in inflammatory vs. non-inflammatory depression and HCs; lower levels of propionic and butyric acids in depressed patients vs. HCs[66]
DepressionHuman/fecal samplesPro-inflammatory genera such as Streptococcus and anti-inflammatory genera, like FaecalibacteriumLower α-diversity and richness, changes in β-diversity, elevated abundance of Streptococcus and reduced abundance of Faecalibacterium[67]
Table 3. Associations between altered DNA methylation patterns in immune-relevant genes and developing specific neuropsychiatric disorders in humans.
Table 3. Associations between altered DNA methylation patterns in immune-relevant genes and developing specific neuropsychiatric disorders in humans.
Neuropsychiatric DisordersType of Sample/Study PopulationImmune-Relevant GenesKey FindingsRef.
Acute maniaSerum samples/20 mania and 20 unaffected controlsCYP11A1Relationship between methylation of CYP11A1 and three inflammatory markers in patients[75]
Schizophrenia (SCZ)Blood/deficit SCZ (n = 53), non-deficit SCZ (n = 55), and 63 healthy controls (HCs)CXCL1Hypomethylation of most CpG sites within CXCL1 gene in SCZ vs. HCs[76]
PsychosisLeukocyte/60 non-affective psychosis and 40 HCsDDR1Association between DDR1 hypermethylation and inflammatory markers[77]
SCZPeripheral blood cells (PBCs)/monozygotic twins discordant for SCZSOCS3 and CASP1Reactivating a SOCS3-mediated anti-inflammatory response by LncRNA-AC006129.1 via DNA methylation-mediated down-regulation of Capicua gene[78]
SCZPBCs/469 Han Chinese patients with SCZThe Th1 regulatory-related genes (SLC11A1, TNFSF4, IL27, and IL1R1); L12B, IL27, S100A12, and ZAP70Symptom severity is linked to DNA methylation of immune-relevant genes; hypermethylation of L12B, IL27, S100A12, and ZAP70 correlate to better response to antipsychotics[79]
Bipolar disorder (BD)PBCs/84 BD subjects with a history of suicide attempt (SA) (BD + SA), 79 BD subjects without history of SA (BD–SA)CXCL8, CD300LG, LFNG, TRIM40, RNF14, and HIVEP3Six differentially methylated positions (DMPs) and seven differentially methylated regions (DMRs) in BD + SA vs. BD–SA in immune-related genes[80]
BDLeukocyte/128 BD patients in remission and 141 HCsDDR1DDR1 hypermethylation at cg19215110 and cg23953820, and hypomethylation at cg14279856 and cg03270204 sites are linked to immune and inflammatory mechanisms in BD[81]
Major depressive disorder (MDD)Whole blood and serum samples/self-reported history of depression (n  =  100) vs. no depression (n  =  100)LTB4R2 and IL-6Six DMRs in exon 1 of LTB4R2 gene;
one depression-associated co-methylation module relevant to telomere length and IL-6 levels
[82]
MDDBlood/153 subjects with MDDIL1-β and IL6RHigher methylation percentage of treatment responders in an IL6R CpG island[83]
MDDBlood/52 young patients with MDD in Scandinavian adultsTLR4Reduced methylation of TLR4 in blood is linked to greater depression scores[84]
MDD220 MDD and 82 HCsNLRP3DMPs in NLRP3 are linked to brain structural alterations (NLRP3 DNA methylation may elevate NLRP3 inflammasome-related neuroinflammation)[85]
Alzheimer’s disease (AD)Human brain/5 from AD and age-matched non-dementia controlsCASPASE-4 (CASP4)Hypomethylation of CASP4 in AD, and elevated expression of CASP4, and IL-1β[86]
Autism spectrum disorder (ASD)Peripheral blood neutrophils/52 ASD children and 24 controlsCCR2 and MCP-1DNA hypo-methylation and increased levels of inflammatory mediators (CCR2 and MCP-1)[87]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Nohesara, S.; Mostafavi Abdolmaleky, H.; Pirani, A.; Thiagalingam, S. Therapeutic Horizons: Gut Microbiome, Neuroinflammation, and Epigenetics in Neuropsychiatric Disorders. Cells 2025, 14, 1027. https://doi.org/10.3390/cells14131027

AMA Style

Nohesara S, Mostafavi Abdolmaleky H, Pirani A, Thiagalingam S. Therapeutic Horizons: Gut Microbiome, Neuroinflammation, and Epigenetics in Neuropsychiatric Disorders. Cells. 2025; 14(13):1027. https://doi.org/10.3390/cells14131027

Chicago/Turabian Style

Nohesara, Shabnam, Hamid Mostafavi Abdolmaleky, Ahmad Pirani, and Sam Thiagalingam. 2025. "Therapeutic Horizons: Gut Microbiome, Neuroinflammation, and Epigenetics in Neuropsychiatric Disorders" Cells 14, no. 13: 1027. https://doi.org/10.3390/cells14131027

APA Style

Nohesara, S., Mostafavi Abdolmaleky, H., Pirani, A., & Thiagalingam, S. (2025). Therapeutic Horizons: Gut Microbiome, Neuroinflammation, and Epigenetics in Neuropsychiatric Disorders. Cells, 14(13), 1027. https://doi.org/10.3390/cells14131027

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

Article Metrics

Back to TopTop