- 1Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
- 2Department of Neuroregulation Center, Southwest Rehabilitation Hospital, Chengdu, China
- 3School of Education, University of New South Wales, Sydney, NSW, Australia
- 4Dalian Key Laboratory for Nuclear Receptors in Major Metabolic Diseases, Dalian, China
Schizophrenia (SZ) is a severe neuropsychiatric disorder arising from complex interactions between genetic susceptibility and environmental factors. There is growing evidence that immune dysregulation and neuroinflammation are central to its pathogenesis, with the microbiota-gut-brain (MGB) axis playing a critical role. This review synthesizes clinical and preclinical findings to elucidate the relationship between gut microbiota dysbiosis and aberrant inflammatory signaling in the periphery and central nervous system in schizophrenia. We detail how alterations in gut microbiota metabolites, following dysbiosis disrupt blood-brain barrier (BBB) integrity and exacerbate neuroinflammation, ultimately leading to the neuropathology of SZ. The review further explores how gut dysbiosis activates innate immune pathways, including the complement system (e.g., C4) and Toll-like receptors (e.g., TLR4), and examines the bidirectional relationship between cytokine imbalances and gut microbiota. A key focus is placed on the dysregulation of the kynurenine pathway of tryptophan metabolism, which mechanistically links immune activation to neurotransmitter imbalances. Collectively, these findings demonstrate that gut microbiota dysbiosis contributes to the pathophysiology of schizophrenia through multifaceted immune-neuro-endocrine pathways, highlighting the MGB axis as a promising target for novel therapeutic strategies.
1 Introduction
Schizophrenia (SZ) is a chronic brain disorder characterized by genetic heterogeneity and neuropathological alterations, with high mortality rates (1). It impairs higher-order brain functions, leading to multifaceted disabilities and incoordination of mental activities. Its clinical symptoms primarily include positive symptoms, negative symptoms, and cognitive deficits (2, 3). Evidence supports the interplay between genetic and environmental factors plays a crucial role in the pathogenesis of schizophrenia (4–6).
Current evidence from genetics, molecular neuropathology, and clinical studies underscores the importance of immune factors in both the pathogenesis and treatment of schizophrenia (7–9). Notably, genetic data from multiple large-scale patient cohorts and genome-wide association studies have shown that single nucleotide polymorphisms located within the major immune-related region on chromosome 6 are associated with schizophrenia (10–12). This finding provides strong genetic support for the immune hypothesis of schizophrenia. Furthermore, immune dysfunction may indirectly increase the risk of developing schizophrenia (see Figure 1). This dysfunction can be triggered by intrinsic host factors, such as autoimmune diseases (13–15), as well as extrinsic environmental factors, such as maternal infection and environmental exposures to infections during early life, childhood, and around the first episode of psychosis (16, 17).
Figure 1. Immune-related risk factors that may induce schizophrenia and pathophysiological consequences. The figure provides an overview of the process by which immune-related risk factors disrupt the balance of the immune system, ultimately contributing to the development of SZ. The risk factors associated with immune system imbalance include intrinsic factors (such as autoimmune diseases) and extrinsic environmental factors (such as maternal infections, environmental exposures during early life, childhood, and the first episode of psychosis, as well as gut microbiota dysbiosis). Immune system imbalance is primarily reflected in abnormalities of immune cells, immune organs, and immune-related molecules. The pathological mechanisms by which immune system imbalance may lead to SZ include neurotransmitter dysregulation (abnormalities in dopamine/glutamate/GABA/5-HT), neurodevelopmental impairments (such as abnormal synaptic pruning and brain structural changes), inflammatory responses, autoimmune reactions, and related clinical symptoms (positive symptoms, negative symptoms, and cognitive deficits). CMV, Cytomegalovirus; HSV-2, Herpes Simplex Virus Type 2; MS, Multiple Sclerosis; GBS, Guillain-Barré Syndrome; CNVs, Copy Number Variations; SNPs, Single Nucleotide Polymorphism.
Among these risk factors, infectious pathogens play a significant role as environmental risk factors in SZ. Clinical evidence (see Table 1) and meta-analyses (30) show a significant association between schizophrenia and a history of infection with various pathogens (for example, Chlamydophila psittaci, Chlamydia pneumoniae, Human Herpesvirus 2, Borna disease virus, and Human Endogenous Retrovirus W). Exposure to these infectious pathogens and inflammatory stimuli can profoundly affect the brain and behavior (31). A prominent example is Toxoplasma gondii infection, which, in animal models, alters behavior and increases the release of neurotransmitters such as dopamine, and the amount of dopamine release is correlated with the number of infected cells (32). In humans, it is associated with symptoms similar to those observed in schizophrenia (33). Notably, such infections can induce a complex neuroimmune response involving cytokine production by microglia, astrocytes, and neurons (34). Furthermore, they can dysregulate key immune components; for example, CD8+T cells, which are crucial for sustaining lasting immunity, have been found to be downregulated in schizophrenia patients (35). It is noteworthy that Toxoplasma gondii infection has been shown to affect the gut microbiome in mice (36). This suggests that the immune and neurochemical disruptions may be partially mediated through gut-related pathways. This interplay highlights the intricate interactions between genetic and environmental factors in disease development (37).
In fact, the total number of non-redundant genes encoded by the microbiota far exceeds that of the human host, and the composition and function of the microbiota are significantly influenced by environmental factors (38, 39). This genomic feature highlights the potentially critical role of the microbiome in mediating gene-environment interactions in humans.
A growing number of studies indicate that levels of natural antibodies against Gram-negative bacteria and lipopolysaccharide (LPS), such as IgA, IgM, and IgG, are elevated in patients with SZ. The production of these antibodies may be related to gut microbiota dysbiosis and increased intestinal permeability (40). Recent research has shown that gut microbes and their metabolites can enter the systemic circulation and affect the central nervous system, which may be closely related to the mechanisms underlying the symptoms of schizophrenia. Together, this evidence positions the Microbiota-Gut-Brain (MGB) axis as a key pathway through which environmental and immune factors contribute to the pathophysiology of schizophrenia, a premise we will explore in detail throughout this review.
2 The microbiota-gut-brain axis and schizophrenia: evidence from clinical and preclinical studies
During fetal development, microbial communities are already present in the maternal placenta, amniotic fluid, and umbilical cord (41–43). The perinatal period represents a critical window for the development of both the gut microbiota and the brain. Factors such as perinatal maternal stress, mode of delivery, environmental exposures, and genetic factors significantly influence the composition of the offspring’s gut microbiota and can alter behavior and Central nervous system (CNS) structure (41–50). Historically, a complex bidirectional communication system between the gastrointestinal tract, its microbiota, and the CNS—comprising neural, hormonal, and immune pathways—has been recognized and termed the MGB axis (51).
In recent years, numerous clinical studies have demonstrated that patients with schizophrenia exhibit characteristic gut dysbiosis, manifested as significant disruptions in microbial relative abundance and reduced α-diversity (52–57). Specifically, these alterations include an increased abundance of Proteobacteria and Lactobacillus, along with decreased levels of anti-inflammatory commensals such as Prevotella (58). This dysbiosis reflects a pro-inflammatory state within the gastrointestinal tract, characterized by an elevated abundance of Lachnoclostridium and reduced levels of short-chain fatty acid-producing Blautia spp. and Ruminococcus spp. (59). These microbial changes correlate with higher lipopolysaccharide (LPS) and lower superoxide dismutase-1 levels (59, 60), which can lead to microbial translocation, systemic inflammation, and increased permeability of both the intestinal and blood-brain barriers (BBB), creating a vicious cycle (61–65). These alterations are associated with specific SZ phenotypes, symptom severity, and treatment response (39, 66–68). For instance, a meta-analysis by Murray et al. indicated that abnormal proliferation of specific microbial taxa (e.g., Bifidobacterium and Lactobacillus) is significantly correlated with core clinical features, including worsened negative symptoms, metabolic pathway disturbances, and reduced cortical gray matter volume (57).
3 Microbial metabolites in SZ pathology: BBB disruption and neuroinflammation
The alterations in microbial composition described above are closely linked to functional disturbances in the gut ecosystem of patients with schizophrenia. Influenced by the altered gut microbiota composition, SZ patients show an increased incidence of gastrointestinal barrier dysfunction, food antigen sensitivity, inflammation, and metabolic syndrome (56, 69, 70). This may result from severe disruption of functional capacity in the gut microbiota of SZ patients, leading to a marked increase in pro-inflammatory metabolites and a significant decrease in anti-inflammatory metabolites (71). In preclinical and clinical metabolomics studies, approximately 10 metabolites have been identified to exhibit significant alterations in patients with SZ. These include N-acetylaspartate, lactate, tryptophan, kynurenine, glutamate, creatine, linoleic acid, D-serine, glutathione, and 3-hydroxybutyrate, among others (72). A recent study found that elevated levels of lactate and cortisol in the peripheral blood of SZ patients were significantly correlated with decreased immune parameters (such as reactive lymphocytes) and altered concentrations of cerebral metabolites like glutamate and N-acetylaspartate (73). Another study further identified correlations between specific gut bacteria (e.g., Streptococcus-Sobrinus) and metabolites (such as 7-aminomethyl-7-carbaguanine and vitamin D2), and discovered that these microbial features were closely associated with patients’ cognitive function (74). Furthermore, the gut microbiome is associated with changes in brain structure and function in SZ patients, as evidenced by neuroimaging studies linking microbial α-diversity to alterations in gray matter volume and regional homogeneity (75). These alterations in brain structure may facilitate the entry of toxic substances and peripheral inflammatory mediators into the brain, thereby triggering neuroinflammation. This process can involve gut microbiota metabolites crossing the blood-brain barrier, modulating the microbiota-gut-brain axis, and regulating microglial activity and cytokine release.
3.1 Bile acids
The potential role of bile acids (BAs) metabolism dysregulation in SZ is likely mediated through interactions with the gut-brain axis (76). Clinical studies have confirmed significant alterations in both the gut microbiota composition and BAs profiles in SZ patients (77), highlighting a potential correlation between BAs dysregulation and the disorder. A metagenome-wide association study further identified gut bacteria unique to SZ patients, including Alkalibacterium, Enterococcus faecium, and Lactobacillus fermentum (78). Cross-sectional studies found that certain gut microbes such as Collinsella, Corynebacterium, Lactobacillus, and Succinivibrio (67); Lachnospiraceae (79); and Veillonella (75) were positively correlated with the severity of SZ. This association between gut microbiota dysbiosis and SZ severity might be mediated through abnormalities in the BAs decoupling process. Metabolomic analysis of serum BAss in SZ patients revealed significantly altered BAs profiles compared to healthy controls (80). Notably, levels of primary Bas, such as CDCA, were generally elevated in patients, whereas levels of secondary Bas (e.g., DCA and LCA) were reduced (80). These altered BAs profiles may subsequently affect the composition of the gut microbiota, thereby interfering with neural function and contributing to the development and progression of SZ. A recent case report described a 39-year-old Persian male with treatment-resistant SZ who, after receiving 300 mg UDCA daily for 12 weeks, showed significant improvement in both positive and negative symptoms, along with enhanced cognitive abilities. Importantly, UDCA treatment was not only effective but also well-tolerated, with no adverse reactions reported during the treatment period, underscoring the safety and efficacy of UDCA supplementation (81). Animal studies have revealed that secondary bile acids may impair the integrity of the BBB. Among these, DCA has been demonstrated to damage endothelial tight junctions, leading to increased BBB permeability (82). This disruption allows toxic substances and peripheral inflammatory mediators to enter the brain, triggering neuroinflammation.
3.2 Polyamines
Polyamines (PAs) are important metabolites produced by gut bacteria. Research has confirmed that the gut microbiota is a significant source of polyamine synthesis, with its production of putrescine, spermidine, and spermine playing a key role in maintaining intestinal polyamine homeostasis (83). Historically, PAs were implicated in the etiology of schizophrenia because certain antipsychotic and antimalarial drugs contain structural components resembling spermidine and were associated with extrapyramidal symptoms and psychosis (84). A multi-omics analysis revealed significant disturbances in the polyamine biosynthesis pathway (85). Currently, elevated blood concentrations of PAs (primarily spermine and/or spermidine) have been observed in cases across various subtypes of schizophrenia (86–88). However, data on PA concentrations in the brain remain limited. One study on human brain tissue found no difference in polyamine levels in the frontal cortex or hippocampus of schizophrenia patients compared to control subjects (89). Another study reported significantly elevated agmatine concentrations in both the plasma and postmortem frontal cortex tissue of first-episode and chronic schizophrenia patients (90–93), and found that antipsychotic treatment could reduce blood agmatine levels (94). Elevated spermidine and total PA concentrations were also detected in fibroblasts obtained from SZ patients (95). Furthermore, studies on serum from SZ patients showed elevated levels of polyamine oxidase (the enzyme responsible for degrading PAs) (96, 97), while the activities of three enzymes involved in PA synthesis—ornithine aminotransferase, antizyme inhibitor 1 (AZIN1), and ornithine cyclodeaminase —were found to be reduced in the prefrontal cortex of both treated and untreated patients (98), ultimately potentially leading to disrupted polyamine homeostasis. A translational convergent functional genomics study identified the gene AZIN1, which encodes AZIN1, as a candidate gene for schizophrenia (99), providing genetic support for these findings. The pathophysiological role of PAs in SZ is primarily thought to be mediated through the dopamine pathway and by altering the function of the N-methyl-D-aspartate receptor (100), although the precise mechanisms remain unclear.
Under pathological conditions such as cerebral ischemia and trauma, stress states can induce significant disruption in polyamine metabolism, particularly the abnormal accumulation of putrescine. This alteration is closely associated with vasogenic edema and the disruption of the BBB (101). Research has confirmed that inhibiting polyamine biosynthesis with the ornithine decarboxylase inhibitor α-difluoromethylornithine significantly mitigates increased BBB permeability, Evans blue extravasation, and brain tissue water content in models of cerebral ischemia (102). Notably, this protective effect can be reversed by the administration of exogenous putrescine. This indicates that upregulation of endogenous polyamines, especially putrescine, serves as a critical mediator in BBB injury.
3.3 Short-chain fatty acids
The structural integrity of the BBB is maintained by tightly joined brain endothelial cells, astrocytic end-feet, pericytes, and various transport proteins, which stringently regulate the penetration of substances from the blood into the brain tissue (103). SCFAs—such as acetate, propionate, and butyrate—produced by gut microbial fermentation, can enter the systemic circulation, reach the brain, and are present in considerable concentrations in the cerebrospinal fluid (104).
Recent research has shown that SCFAs play a crucial role in modulating the structure and function of the BBB. On the one hand, SCFAs can enhance BBB integrity by regulating the expression of tight junction proteins. For instance, studies in germ-free or antibiotic-treated mouse and rhesus monkey models have demonstrated reduced levels of BBB tight junction proteins (e.g., claudin-5, occludin) and increased BBB permeability; colonization with SCFA-producing strains (such as Clostridium tyrobutyricum and Bacteroides) reversed these effects (105–108). On the other hand, SCFAs can protect the BBB directly or indirectly through anti-inflammatory mechanisms. SCFAs can interact with free fatty acid receptor 3 present on brain endothelial cells (109), suppress inflammatory responses, and alleviate oxidative stress, thereby promoting BBB stability. For example, propionate exerts anti-inflammatory and antioxidant effects by reducing cell surface CD14 expression and influencing the translocation of Nuclear Factor Erythroid 2-Related Factor 2 (110). Peripherally, certain SCFAs (e.g., acetate, propionate, and butyrate) have also been identified to possess anti-inflammatory properties (111–113).
4 Gut microbiota dysbiosis and abnormal activation of the innate immune system
4.1 Evidence for the association between complement components and schizophrenia
Current studies have identified elevated transcript levels of complement components (C1qA, C3, C4, C5) in patients with schizophrenia (114) (see Table 2), suggesting, albeit not entirely consistently, that increased activation of the classical complement pathway may be associated with SZ.
4.2 Gut microbiota dysbiosis and abnormal activation of complement C4
Genetic susceptibility to schizophrenia is significantly associated with polymorphisms in genes related to the complement system, largely attributable to alleles of the complement C4 gene located within the major histocompatibility complex region on chromosome 6 (132). The C4 allele demonstrates the strongest association with SZ risk (132)and is closely linked to increased synaptic phagocytosis and elimination by microglia (8). Furthermore, recent research has found that C4 gene overexpression triggers impaired GluR1 trafficking through an intracellular mechanism involving the endosomal protein SNX27, leading to pathological synaptic loss.
Pathogen exposure has long been recognized as a risk factor for the development of schizophrenia (142). Immune-related environmental variables, such as pathogen infection and gut microbiota dysbiosis, may interact with complement system dysfunction (143). Particularly noteworthy is the finding that genetic polymorphisms of complement components C4A and C4B are significantly associated with various microbial environmental variables in schizophrenia patients, including a history of pathogen exposure and gut ecological imbalance (143). Among these, a negative correlation was observed between plasma lipopolysaccharide-binding protein (LBP) levels and C4A gene copy number was observed in patients with schizophrenia, but not in healthy controls (143). This discovery is significant because LBP, a marker of bacterial translocation, reflects the host’s response to gut microbial translocation and circulating bacterial LPS (144). Together, these results suggest that the complement system, particularly C4A, may play a key role in the gene-environment interactions of schizophrenia by modulating the gut microbiota and systemic immune activation status.
Furthermore, the negative correlation between C4A and C4B copy numbers in SZ patients may reflect a non-random association between these loci. Further analysis revealed that C4A and C4B haplotypes are significantly associated not only with the diagnosis of schizophrenia and environmental factors but also with psychiatric symptoms and cognitive function (143). While multiple previous studies have confirmed the association between the C4A gene and an increased risk of schizophrenia (132), the intrinsic mechanism underlying the negative correlation between C4B and schizophrenia risk remains unclear. It is currently unknown whether a lower C4B copy number has a protective effect against the disease or merely reflects an associated pathological protein deficiency. The negative correlations between C4A and C4B, as well as between C4L and C4S copy numbers, more likely indicate that these loci reside at opposing ends of a risk (C4A and C4L) and protective (C4B and C4S) spectrum, existing in a state of linkage disequilibrium (143). Biochemically, C4A exhibits higher affinity for amino groups, whereas C4B binds more readily to hydroxyl groups, suggesting that C4A may be more involved in binding immune complexes and protein antigens, while C4B plays a more important role in binding carbohydrate-rich microbial antigens (145). Consequently, a deficiency in C4B protein in patients may impair its ability to bind microbial antigens. Conversely, another possibility is that a lower C4B copy number might protect the stability of the host microbiome to some extent by reducing the excessive clearance of beneficial microbes mediated by C4B. A study on pediatric inflammatory bowel disease indicated that individuals with low C4B copy numbers had milder inflammation and higher gut microbial diversity compared to those with high C4B copy numbers (146).
As research on the gut microbiome in schizophrenia expands, integrating C4 genotyping with microbiome analysis is of great importance. Furthermore, these haplotype associations require further analysis to exclude nearby HLA gene variants—which may influence the observed immune phenotype due to linkage disequilibrium with specific C4 alleles (123).
5 Potential pathophysiological mechanisms of toll-like receptors and gut bacterial translocation in schizophrenia and psychiatric disorders
Toll-like receptors (TLRs) coordinate the activation of innate immune responses alongside the complement system and play a significant role in the neuroimmune mechanisms of schizophrenia. Studies indicate that activation of TLR2/3/4/5 triggers the NF-κB/NLRP3 pathway and the PI3K/Akt/mTORC1 signaling pathway, leading to microglial activation, neuroinflammation, and neuronal damage (147, 148). Furthermore, thickness changes in the limbic system and cortical brain regions of schizophrenia patients are correlated with abnormal expression of specific TLRs, suggesting their involvement in brain structural remodeling (149). Stimulation of whole blood cells from SZ patients with selective TLR agonists results in enhanced release of pro-inflammatory cytokines (including IL-1β, IL-6, IL-8, and TNF-α) (150), further supporting the role of TLRs in regulating neuroinflammation in schizophrenia.
TLR4, a key receptor for recognizing pathogen-associated molecular patterns such as LPS, activates downstream signaling through both MyD88-dependent and independent pathways, inducing the production of various cytokines and participating in the regulation of neuroinflammation and cellular function (151, 152). In recent years, studies on SZ bodily fluids have consistently reported increased numbers of TLR4-positive monocytes and elevated TLR4 expression in the peripheral blood of schizophrenia patients (149, 153–155), suggesting that TLR4 upregulation is a key factor in the immunopathological process of schizophrenia. Postmortem results show increased TLR4 protein expression in the prefrontal cortex of schizophrenia patients (152), which is associated with activation of the MyD88 and NF-κB pathways (156). Recent research has identified the TLR4/MyD88/NF-κB pathway as playing a central role in various neurological disease models, suggesting its relevance in schizophrenia pathogenesis. For instance, regulatory T cells modulate neuroinflammation and microglial pyroptosis in LPC-induced demyelination via this pathway, thereby alleviating myelin loss and cognitive dysfunction (157). Additionally, TLR4 regulates hippocampal neurogenesis and synaptic function through this pathway, and is involved in neuroinflammation and neuronal apoptosis (158). Conversely, inhibiting TLR4 can suppress microglial activation, alleviate neuroinflammation, improve cognitive function, and mitigate synaptic plasticity impairments and depression-like symptoms via this pathway (159, 160).
The pathological processes described above are often initiated in schizophrenia by a breach of the intestinal barrier, creating a conduit for gut-derived immune activation. Alterations in the microbial flora, gut inflammation, increased intestinal barrier permeability (forming a “leaky gut”), bacterial translocation, and exposure to stressful environments in schizophrenia (144, 152, 156), may trigger innate immunity via TLR4 stimulation. The specific mechanism may involve a compromised intestinal barrier allowing antigens from the gut microbiota to penetrate and contact IgG antibodies, forming immune complexes that circulate in the bloodstream, including to the choroid plexus of the CNS. The chronic accumulation of these complexes can initiate inflammation and contribute to the progression of chronic disease. When a “leaky gut” is present, intestinal barrier permeability is increased, pro-inflammatory substances like LPS may activate inflammatory pathways, be recognized and activated by the TLR4 receptor, and mediate inflammatory responses. This indirect influence of gut microbes on the innate immune system leads to changes in the circulating levels of pro- and anti-inflammatory cytokines, subsequently directly affecting brain function (161).
Exposure to microbial products such as LPS, which can translocate from a leaky gut, activates TLR4 signaling, which is closely intertwined with gut-brain axis interactions. This is corroborated by numerous animal models. Pretreatment with paliperidone (an atypical antipsychotic drug) inhibits TLR4 activation and neuroinflammatory responses in the prefrontal cortex of stressed rats. The mechanism involves modulating stress-induced gut inflammation and reducing plasma LPS levels, thereby influencing brain TLR4 signaling pathways. This result suggests that the therapeutic effects of paliperidone extend beyond its impact on dopamine and serotonin neurotransmission systems (162). Another rat study found that acute restraint stress can upregulate TLR4 gene expression in the frontal cortex by inducing gut microbiota translocation, while intervention with antibiotics or the TLR4 specific inhibitor TAK-242 effectively suppresses this process and reduces the accumulation of inflammatory and oxidative/nitrosative mediators (163). Furthermore, inhibiting TLR4 can also modulate gut microbiota homeostasis and the MyD88/NF-κB axis in ulcerative colitis (164), indicating that TLR4 acts not only as a key sensor of innate immunity but may also regulate neuroimmune crosstalk and gut microenvironment homeostasis. Given that SZ patients also experience loss of gut microbiota homeostasis, targeting the TLR4 signaling pathway and focusing on bacterial translocation and microbiota may offer new avenues for immunomodulatory therapy in schizophrenia.
Specifically, in the “leaky gut” state, gut microbiota dysbiosis occurs, the expression of intestinal epithelial junction proteins (e.g., tight junction proteins) decreases, and intestinal permeability increases, forming a “leaky gut” (165). This breach facilitates bacterial translocation, as evidenced by the significant increase in markers such as soluble CD14 and LBP in the blood of individuals with schizophrenia (144, 166). Upon entering the systemic circulation, microbial products like LPS are recognized by and activate the TLR4 receptor on innate immune cells. This triggers downstream signaling pathways (e.g., NF-κB and MAPK), driving the massive release of pro-inflammatory cytokines, including IL-1β, IL-6, and TNF-α (167–170). These peripheral inflammatory mediators can, in turn, compromise the blood-brain barrier and activate central immune cells, thereby linking gut-derived innate immune stimulation to the neuroinflammation characteristic of schizophrenia. This cascade establishes a self-perpetuating vicious cycle, wherein systemic inflammation exacerbates gut barrier dysfunction and dysbiosis, which then further fuels the inflammatory response.
6 Gut microbiota dysbiosis and cytokine abnormalities
6.1 Evidence for the association between cytokines and schizophrenia
Given the dysfunction of the BBB in patients with schizophrenia, changes in cytokine levels in the periphery (peripheral blood) and the center (cerebrospinal fluid, CSF) are valuable for assessing the CNS inflammatory state. Numerous studies have identified abnormal alterations in various cytokines in the CSF and peripheral blood of SZ patients (see Table 3), most of which are pro-inflammatory cytokines (IL-1β, IL-2, IL-8, TNF-α, and IFN-γ) (188–191), with a smaller proportion being anti-inflammatory cytokines (including TGF-β1, IL-4, and IL-10) (192). The prevailing research view is that a dynamic imbalance between pro-inflammatory and anti-inflammatory cytokines may contribute to the pathogenesis of schizophrenia and subsequent psychopathological symptoms (193–196).
Table 3. Summary of meta-analysis of cytokines in peripheral blood and cerebrospinal fluid of SZ patients.
According to Table 3, a characteristic pro-inflammatory state can be observed in SZ patients, manifested by persistently elevated levels of pro-inflammatory cytokines such as IL-6, IL-1β, and TNF-α in both peripheral blood and CSF, accompanied by a relative deficiency of anti-inflammatory cytokines like IL-10 and TGF-β. This cytokine imbalance is closely related to symptom severity, cognitive deficits, and treatment response. This aligns with current mainstream research; for example, the TLR4/NF-κB/IL-1β signaling pathway is activated in chronic SZ patients (197), and clozapine reduces the expression of pro-inflammatory genes such as IL-1β and IL-6 by inhibiting the TLR4/NF-κB pathway (198). Additionally, IL-8 mediates the migration and survival of neural stem cells and oligodendrocyte progenitor cells early in life, and elevated CSF cytokines IL-1β and IL-6 suggest the possibility of microglial activation (199–201).
Aggregating data from Table 3 reveals consistent cross-barrier alterations in cytokine levels in both CSF and peripheral blood across different clinical stages (first-episode/FES, acute/ASZ, and chronic/CSZ), which hold clinical translational significance: 1). Significantly elevated levels of IL-1β, IL-6, and IL-8 in FES and ASZ SZ patients suggest they may be state markers for the onset or acute exacerbation of SZ; 2). The dose-dependent decrease in pro-inflammatory cytokine levels (e.g., IL-1β, IFN-γ, IL-6, TNF-α) following antipsychotic treatment (risperidone and clozapine) indicates value for monitoring treatment response; meanwhile, a significant increase in anti-inflammatory cytokine levels (e.g., sIL-2R) was observed in some patients (181, 184), a finding suggesting that antipsychotic drugs may exert anti-inflammatory-like effects.
6.2 Potential pathophysiological mechanisms of gut microbiota and metabolite SCFAs in cytokine abnormalities
SCFAs (such as acetate, propionate, and butyrate) activate receptors like FFAR2/3 and Gpr109a and inhibit histone deacetylase (HDAC), collectively suppressing the activation of the NF-κB signaling pathway, thereby downregulating the expression of pro-inflammatory cytokines (202–204). It is well-known that the NF-κB signaling pathway is typically activated in inflammatory and autoimmune diseases (205–207). Interestingly, all three cytokines elevated in SZ patients in Table 1 (IL-1β, IL-6, and IL-8) are regulated via the NF-κB pathway. Since cytokines can also regulate the activity of tryptophan catabolism in astrocytes and microglia, this finding corresponds to the alterations in the kynurenine pathway observed in the brains of individuals with schizophrenia (186).
SCFAs play an important regulatory role on various immune cells, particularly those central to maintaining immune homeostasis and anti-inflammatory responses, by inhibiting HDAC. For instance, SCFAs can inhibit NF-κB activation and the secretion of inflammatory cytokines like TNF-α in peripheral blood monocytes, neutrophils, and macrophages, thereby mitigating excessive immune responses. In dendritic cells, butyrate and propionate can impede their normal differentiation and induce an immune tolerant phenotype. Furthermore, SCFAs upregulate the expression of Foxp3, a key transcription factor for regulatory T cells, through HDAC inhibition (208, 209), promoting Treg differentiation and suppressing the production of pro-inflammatory cytokines (210). In SZ patients, the abundance of SCFA-producing bacteria is often altered, leading to reduced SCFA levels, which weakens the capacity to suppress inflammation; the abundance of SCFA-producing bacteria also changes post-treatment (211, 212).
6.3 The kynurenine pathway: an interactive mechanism linking gut microbiota, immunity, and neurotransmitters
Specific neurotransmitters, such as GABA, dopamine, glutamate, and serotonin (5-HT), are derived from precursors tyrosine and tryptophan, which are transported across the BBB into the CNS and subsequently converted into neurotransmitters (213). Tryptophan is primarily absorbed in the gut and metabolized by the gut microbiota through three downstream pathways: the 5-HT pathway, the kynurenine pathway, and the indole pathway (214). The kynurenine pathway of tryptophan metabolism represents a critical link through which the MGB axis participates in the immunopathology of schizophrenia. In schizophrenia, immune activation upregulates pro-inflammatory cytokines (e.g., IFN-γ, IL-1, TNF-α), which activate indoleamine 2,3-dioxygenase (IDO) via both IFN-γ receptor (IFN-γR)-dependent and independent pathways (e.g., synergistic action of TLR4, IL-1R, TNF-αR) (215). Enhanced activity of IDO enzymes (IDO-1 and IDO-2) shifts tryptophan metabolism towards the kynurenine pathway, leading to increased concentrations of kynurenic acid (KYNA) within the CNS (216–220). Kynurenine (KYN) is transported across the BBB and metabolized by glial cells into KYNA and quinolinic acid (221, 222) ultimately resulting in excessive KYNA production. Acting as an endogenous antagonist, KYNA inhibits NMDA receptor and α7 nicotinic receptor function (223), suppresses glutamate and acetylcholine neurotransmission (223, 224), and leads to an imbalance in the glutamate, dopamine, and acetylcholine systems, thereby affecting neurotransmission, synaptic organization, and brain connectivity (225, 226).
On the other hand, activation of the KP results in substantial tryptophan consumption, reducing the substrate available for 5-HT synthesis (see Figure 2). Over 90% of the body’s 5-HT is synthesized in the gut by enterochromaffin cells, which absorb tryptophan from dietary proteins as a substrate for 5-HT synthesis; this process is regulated by SCFAs and the kynurenine synthesis pathway (227, 228). Studies in animal models with gut microbiota depletion have shown increased plasma tryptophan, elevated brain serotonin concentrations, and reduced kynurenine pathway activity, all of which normalize following microbiota restoration (229–231). Gut microbiota dysbiosis and immune activation can further enhance IDO activity, exacerbating the conversion of tryptophan to KYNA. This leads to decreased peripheral and central 5-HT levels, impaired receptor function, reduced concentrations of tryptophan and 5-HT (232), worsened symptoms of affective disorders, elevated levels of tryptophan catabolites (233, 234), and increased concentrations of toxic metabolites in the CNS (235).
Figure 2. The interactive mechanism of the kynurenine pathway among gut microbiota, immunity, and neurotransmitters. TRP, Tryptophan; KYN, Kynurenine; KYNA, Kynurenic acid; 3-HK, 3-Hydroxykynurenine; QUIN, Quinolinic acid; 3-HAA, 3-Hydroxyanthranilic acid;α7nAChR, α7-nicotinic acetylcholine receptors; NMDAR, N-methyl-D-aspartate receptor; GABA, Gamma-Aminobutyric Acid; 5-HT, 5-Hydroxytryptamine, Serotonin; 5-HTP, 5-Hydroxytryptophan; IDO, Indoleamine 2,3-Dioxygenase; TDO, Tryptophan 2,3-Dioxygenase; KAT, Kynurenine Aminotransferase; KMO, Kynurenine Monooxygenase; 3-HAO, 3-Hydroxyanthranilic Acid Oxygenase; HAAO, 3-Hydroxyanthranilate-3,4-dioxygenase; XA, Xanthurenic acid.
Animal experiments have demonstrated that acute tryptophan depletion reduces brain tryptophan concentration by 70%, leading to decreased serotonin levels, diminished 5-HT receptor binding (236, 237), and effects on compulsive behavior (238). Similar results have been observed in human cerebrospinal fluid (CSF) studies (239). This suggests that immune dysregulation-induced gut microbiota dysbiosis and activation of the kynurenine pathway cause acute tryptophan depletion in the gut. This, in turn, leads to reduced brain tryptophan concentration, decreased 5-HT levels, and diminished 5-HT receptor binding, which may underlie the 5-HT system dysfunction, impaired neurotransmission, and negative cognitive effects observed in SZ.
6.4 Limitations
This study has several limitations. First, despite conducting an extensive literature search, we cannot entirely exclude the possibility that some published studies may have been overlooked. Second, the multiple meta-analyses and systematic reviews cited in this article may carry a risk of accumulated type I errors (240). Furthermore, although we endeavored to include studies that performed stratified analyses of patients with schizophrenia, the interpretability and generalizability of the results may still be constrained by limited sample sizes and considerable population heterogeneity—such as confounding factors including age, gender, ethnicity, smoking history, and BMI (125).
7 Conclusions and discussion
7.1 Integrated mechanisms of the microbiota-gut-brain-immune axis in schizophrenia
By integrating the evidence discussed throughout this review, we propose a comprehensive model illustrating the MGB-immune interactions in schizophrenia (See Figure 3). At the peripheral level, gut microbiota dysbiosis and a reduction in microbial metabolites—particularly SCFAs—promote increased antigen exposure and activation of innate immune responses. This leads to the activation of immune cells (e.g., T cells, B cells, NK cells, monocytes/macrophages) and dysfunction of glial cells, culminating in the release of pro-inflammatory cytokines and complement components such as C4. Accompanied by impaired BBB integrity, these peripheral immune factors gain access to the CNS, where they mediate neuroinflammatory responses, resulting in synaptic damage and neuronal dysfunction.
Figure 3. Integrated mechanisms of the microbiota-gut-brain-immune axis in schizophrenia. HPA axis, Hypothalamic-Pituitary-Adrenal Axis; SCFAs, Short-Chain Fatty Acids; PRRs, Pattern Recognition Receptors; CRP, C-Reactive Protein; TLRs, Toll-like receptors; NLRs, Nucleotide-binding Oligomerization Domain-like Receptors; CSF, Cerebrospinal Fluid; MHC, Major Histocompatibility Complex; BBB, Blood-Brain Barrier; C3/C4/C1q, Complement Component 3/4/1q.
In the CNS, dysbiosis directly or indirectly modulates the function of microglia and astrocytes via metabolites including SCFAs. Aberrantly activated glial cells exacerbate neuroinflammation and influence synaptic pruning processes, with complement C4-mediated synaptic phagocytosis playing a critical role. Concurrently, pro-inflammatory cytokines regulate the activity of key enzymes in tryptophan metabolism—indoleamine IDO and TDO—promoting the production of kynurenine pathway metabolites such as KYNA. This disrupts multiple neurotransmitter systems, including dopamine, GABA, glutamate, and serotonin, further exacerbating neurotransmission dysfunction. Moreover, compromised intestinal barrier integrity and gut-derived microbial components such as LPS activate TLR/NF-κB/NLRP3 pathways, which not only drive systemic inflammation but also modulate gut-derived tryptophan metabolism and serotonin synthesis, thereby coupling immune regulation with neuromodulatory functions.
The gut microbiota, as a critical “microbial organ,” has garnered extensive attention for its role in neuropsychiatric disorders such as SZ. However, the causal relationships among microbial dysbiosis, immune dysregulation, and disease onset remain elusive. It is still unclear whether dysbiosis acts as a driver, a consequence, or both. Elucidating this issue is complicated by multiple confounding factors, including population heterogeneity, medication use, dietary and lifestyle variations, disease staging, and inconsistencies in research methodologies. Although small-scale studies and disparities in technical procedures—such as sample processing, sequencing, and bioinformatic analyses—limit the reliability and generalizability of the findings. Therefore, further large-scale and better-standardized clinical studies with stratified populations are urgently needed to provide more robust data on the association between altered gut microbial features and schizophrenia.
7.2 Comparative analysis of gut microbiota in neuropsychiatric disorders
It is well established that the gut microbiota regulates brain function and behavior. The immune-mediated dysregulation of the MGB axis detailed in this review is observed not only in schizophrenia but also across various neurological disorders (241), including neurodevelopmental disorders (242), epilepsy (243), and depression (244), among others. Major depressive disorder is characterized by an elevated Bacteroidetes/Firmicutes ratio (5), accompanied by enrichment of Bacteroides and depletion of Blautia, Faecalibacterium, and Coprococcus, alongside increased abundance of Eggerthella and elevated levels of pro-inflammatory genera such as Escherichia (245). In contrast, anxiety disorders demonstrate an increased Firmicutes/Bacteroidetes ratio (246), reduced abundance of SCFA-producing genera including Bifidobacterium and Lactobacillus, while Akkermansia abundance shows a negative correlation with anxiety severity (247, 248). Bipolar disorder similarly exhibits disruption of the Firmicutes/Bacteroidetes ratio and reduced α-diversity, featuring increased Streptococcaceae and Bacteroidaceae abundance contrasting with depletion of anti-inflammatory commensals such as Faecalibacterium (249).Among neurodegenerative disorders, Parkinson’s disease shows increased abundance of Lactobacillus and Bifidobacterium with concurrent reduction in Faecalibacterium, Coprococcus, and Blautia (250, 251), where decreased Blautia abundance correlates with clinical severity and reduced fecal butyrate levels (252). Alzheimer’s disease manifests through reduced beneficial bacteria including Bifidobacterium and elevated opportunistic pathogens such as Escherichia and Clostridium (253, 254). Autism spectrum disorder(ASD) presents with reduced microbial diversity yet increased biomass in pediatric populations (255, 256), featuring a shift from beneficial microorganisms toward spore-forming, antibiotic-resistant, and/or neurotoxin-producing species (257). Specific alterations include reductions in Prevotella, Coprococcus, and Veillonellaceae, alongside overgrowth of Desulfovibrio (positively correlated with Autism spectrum disorder severity) (258), Sutterella, Ruminococcus, Clostridium, Megamonas, and Candida (259, 260), accompanied by an elevated Firmicutes/Bacteroidetes ratio (255). Notably, Candida overgrowth and associated toxin production may exacerbate neurobehavioral symptoms (37). Epilepsy research similarly reveals substantial gut microbiota dysbiosis, with most studies demonstrating reduced α-diversity in treatment-resistant epilepsy (261–264). Phylum-level analyses indicate predominant Firmicutes with relative Bacteroidetes reduction in some studies (264, 265), while others report increased Actinobacteria, Verrucomicrobia, or pro-inflammatory Proteobacteria, alongside potential reduction of beneficial Bacteroidetes and Actinobacteria (262, 266).
In summary, these disorders share fundamental microbial alterations: 1) Structural dysbiosis manifested through disrupted Bacteroidetes/Firmicutes ratios and reduced α-diversity; 2) Functional impairment characterized by universal depletion of anti-inflammatory, SCFA-producing genera including Faecalibacterium, Blautia, and Bifidobacterium; 3) Common pathophysiological mechanisms involving impaired SCFA production, immune-inflammatory activation, and dysregulated neuroactive metabolite metabolism. These alterations promote systemic low-grade inflammation through an imbalance between elevated pro-inflammatory cytokines (e.g., IL-6, TNF-α) and anti-inflammatory mediators (e.g., IL-10) (267–269). This systemic inflammation, akin to the processes described in schizophrenia, can traverse the blood-brain barrier or transmit via vagal afferents, subsequently activating microglia and exacerbating neuroinflammatory processes while impairing prefrontal cortex-mediated executive functions including decision-making and emotional regulation (270). Pro-inflammatory cytokines disrupt neurotransmitter metabolism through mechanisms such as inhibited tryptophan conversion to serotonin while driving microglial activation, thereby amplifying neuroinflammation (271). Activated microglia release chemokines, cytokines, and reactive oxygen species, crucially contributing to neuroinflammatory cascades while disrupting neurotransmitter balance and synaptic plasticity through pro-inflammatory phenotypic transformation. Notwithstanding these commonalities, disorder specific patterns emerge. Major depressive disorder demonstrates Bacteroidetes/Firmicutes ratio elevation contrasting with anxiety disorders, while ASD presents the distinct profile of reduced diversity with increased biomass and concurrent expansion of multiple opportunistic pathogens. These shared and distinct features collectively illustrate the multifaceted involvement of gut microbiota dysbiosis in neuropsychiatric disorders. Beyond inflammation, psychological stress represents another prevalent pathophysiological feature in microbiota-associated diseases. Stress contributes to depression (272), schizophrenia (273), autism spectrum disorder (274), epilepsy (275), and migraine (276). Significant comorbidity exists among these conditions, exemplified by the frequent co-occurrence of depression and ASD with epilepsy (277), the common comorbidity of depression with migraine (277), and the elevated prevalence of inflammatory bowel disease or irritable bowel syndrome among migraine patients (278).
8 Future perspectives
The gut microbiota can influence drug bioavailability and efficacy through metabolism, while most psychotropic medications possess anti-inflammatory properties and may directly alter microbial composition. A recent study revealed that gut microbiome abnormalities in schizophrenia patients were primarily associated with resistance to antipsychotic treatment, whereas this correlation was significantly weaker in patients who responded well to atypical antipsychotics (279). This suggests that structural changes in the gut microbiota may serve more as a potential biomarker for clozapine resistance rather than an intrinsic feature of schizophrenia itself. Underlying mechanisms may involve microbial-mediated drug metabolism, transformation, and modulation of intestinal barrier function.
Furthermore, current immunomodulatory therapies, including immunosuppressants and biologics, remain predominantly palliative. Long-term administration often leads to drug tolerance and opportunistic infections (280). The substantial comorbidity burden—approximately 30% of inflammatory bowel disease patients develop anxiety or depression—highlights the urgent need for dual-effect therapies that simultaneously address intestinal inflammation and gut-brain axis modulation. Combinatorial treatment strategies targeting the gut-brain interplay represent a paradigm shift in managing psychiatric comorbidities in systemic disorders.
By assessing baseline microbiome profiles, it becomes possible to identify individuals at high risk for poor response to relevant medications prior to treatment initiation. This enables the development of personalized dosing regimens or adjunctive microecological interventions (e.g., probiotic/prebiotic supplementation) to enhance therapeutic efficacy while reducing the incidence of drug resistance and adverse effects. These advances underscore the translational value of microbiome research in the precision medicine of mental disorders.
Although microbial-based interventions, such as probiotics, demonstrate therapeutic potential—for instance, certain strains of Lactobacillus and Bifidobacterium have shown preliminary efficacy in alleviating depressive symptoms (281)—their benefits in schizophrenia remain inconsistent and lack high-quality clinical support (282). Therapeutic strategies targeting the MGB axis, including prebiotics, synbiotics, and fecal microbiota transplantation, are not universally applicable. Instead, they should be tailored based on disease subtype, patient stratification, and bacterial functionality. Their clinical utility urgently requires validation through well-designed, large-scale randomized controlled trials.
Author contributions
B-WS: Conceptualization, Visualization, Writing – original draft, Writing – review & editing. YL: Investigation, Writing – original draft. L-YY: Writing – review & editing. H-XY: Visualization, Writing – review & editing. W-HW: Writing – review & editing. H-WR: Writing – review & editing. Y-NB: Writing – review & editing. J-YL: Writing – review & editing. Z-LL: Funding acquisition, Supervision, Writing – review & editing.
Funding
The author(s) declare financial support was received for the research and/or publication of this article. This work was supported by Basic Scientific Research Fund of Education Department of Liaoning Province, China (LJ212410161100 to Z-LL), Natural Science Foundation of Liaoning Province, China (2022-MS-326 to Z-LL), and Dalian Medical University's "Integrated Traditional Chinese and Western Medicine " Open Fund Project (ZXY2025JC04 to Z-LL).
Acknowledgments
Figure 1 was partly created using figures provided by Servier Medical Art (https://smart.servier.com/), licensed under a Creative Commons Attribution 4.0 Unported License. Figures 2 and 3 are drawn using Figdraw (https://www.figdraw.com).
Conflict of interest
The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
Generative AI statement
The author(s) declare that no Generative AI was used in the creation of this manuscript.
Any alternative text (alt text) provided alongside figures in this article has been generated by Frontiers with the support of artificial intelligence and reasonable efforts have been made to ensure accuracy, including review by the authors wherever possible. If you identify any issues, please contact us.
Publisher’s note
All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.
References
1. Solmi M, Croatto G, Fornaro M, Schneider LK, Rohani-Montez SC, Fairley L, et al. Regional differences in mortality risk and in attenuating or aggravating factors in schizophrenia: A systematic review and meta-analysis. Eur Neuropsychopharmacol. (2024) 80:55–69. doi: 10.1016/j.euroneuro.2023.12.010
2. Owen MJ, Sawa A, and Mortensen PB. Schizophrenia. Lancet. (2016) 388:86–97. doi: 10.1016/s0140-6736(15)01121-6
3. McCutcheon RA, Reis Marques T, and Howes OD. Schizophrenia-an overview. JAMA Psychiatry. (2020) 77:201–10. doi: 10.1001/jamapsychiatry.2019.3360
4. Tsuang M. Schizophrenia: genes and environment. Biol Psychiatry. (2000) 47:210–20. doi: 10.1016/s0006-3223(99)00289-9
5. Demjaha A, MacCabe JH, and Murray RM. How genes and environmental factors determine the different neurodevelopmental trajectories of schizophrenia and bipolar disorder. Schizophr Bull. (2012) 38:209–14. doi: 10.1093/schbul/sbr100
6. Modinos G, Iyegbe C, Prata D, Rivera M, Kempton MJ, Valmaggia LR, et al. Molecular genetic gene-environment studies using candidate genes in schizophrenia: A systematic review. Schizophr Res. (2013) 150:356–65. doi: 10.1016/j.schres.2013.09.010
7. Ripke S, Neale BM, Corvin A, Walters JTR, Farh K-H, Holmans PA, et al. Biological insights from 108 schizophrenia-associated genetic loci. Nature. (2014) 511:421–7. doi: 10.1038/nature13595
8. Sellgren CM, Gracias J, Watmuff B, Biag JD, Thanos JM, Whittredge PB, et al. Increased synapse elimination by microglia in schizophrenia patient-derived models of synaptic pruning. Nat Neurosci. (2019) 22:374–85. doi: 10.1038/s41593-018-0334-7
9. Müller N, Myint AM, Krause D, Weidinger E, and Schwarz MJ. Anti-inflammatory treatment in schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry. (2013) 42:146–53. doi: 10.1016/j.pnpbp.2012.11.008
10. Shi J, Levinson DF, Duan J, Sanders AR, Zheng Y, Pe’er I, et al. Common variants on chromosome 6p22.1 are associated with schizophrenia. Nature. (2009) 460:753–7. doi: 10.1038/nature08192
11. Stefansson H, Ophoff RA, Steinberg S, Andreassen OA, Cichon S, Rujescu D, et al. Common variants conferring risk of schizophrenia. Nature. (2009) 460:744–7. doi: 10.1038/nature08186
12. Purcell SM, Wray NR, Stone JL, Visscher PM, O’Donovan MC, Sullivan PF, et al. Common polygenic variation contributes to risk of schizophrenia and bipolar disorder. Nature. (2009) 460:748–52. doi: 10.1038/nature08185
13. Kridin K, Zelber-Sagi S, Comaneshter D, and Cohen AD. Association between schizophrenia and an autoimmune bullous skin disease-pemphigus: A population-based large-scale study. Epidemiol Psychiatr Sci. (2019) 28:191–8. doi: 10.1017/s204579601700052x
14. Maślińska M, Trędzbor B, and Krzystanek M. Dysbiosis, gut-blood barrier rupture and autoimmune response in rheumatoid arthritis and schizophrenia. Reumatologia. (2021) 59:180–7. doi: 10.5114/reum.2021.107588
15. Wang LY, Chen SF, Chiang JH, Hsu CY, and Shen YC. Autoimmune diseases are associated with an increased risk of schizophrenia: A nationwide population-based cohort study. Schizophr Res. (2018) 202:297–302. doi: 10.1016/j.schres.2018.06.033
16. Tandon R, Nasrallah H, Akbarian S, Carpenter WT Jr., DeLisi LE, Gaebel W, et al. The schizophrenia syndrome, circa 2024: what we know and how that informs its nature. Schizophr Res. (2024) 264:1–28. doi: 10.1016/j.schres.2023.11.015
17. Massrali A, Adhya D, Srivastava DP, Baron-Cohen S, and Kotter MR. Virus-induced maternal immune activation as an environmental factor in the etiology of autism and schizophrenia. Front Neurosci. (2022) 16:834058. doi: 10.3389/fnins.2022.834058
18. Liu T, Gao P, Bu D, and Liu D. Association between toxoplasma gondii infection and psychiatric disorders: A cross-sectional study in China. Sci Rep. (2022) 12:15092. doi: 10.1038/s41598-022-16420-y
19. Oncu-Oner T and Can S. Meta-analysis of the relationship between toxoplasma gondii and schizophrenia. Ann Parasitol. (2022) 68:103–10. doi: 10.17420/ap6801.414
20. Zhu YT, Yang XH, Chen MR, Hu Y, Chang YF, and Wu X. Research progress on the association between schizophrenia and toxoplasma gondii infection. Biomed Environ sciences: BES. (2024) 37:647–60. doi: 10.3967/bes2024.071
21. Montazeri M, Moradi E, Moosazadeh M, Hosseini SH, and Fakhar M. Relationship between toxoplasma gondii infection and psychiatric disorders in Iran: A systematic review with meta-analysis. PLoS One. (2023) 18:e0284954. doi: 10.1371/journal.pone.0284954
22. Fellerhoff B, Laumbacher B, and Wank R. High risk of schizophrenia and other mental disorders associated with chlamydial infections: hypothesis to combine drug treatment and adoptive immunotherapy. Med Hypotheses. (2005) 65:243–52. doi: 10.1016/j.mehy.2005.03.013
23. Park MH, Kwon YJ, Jeong HY, Lee HY, Hwangbo Y, Yoon HJ, et al. Association between intracellular infectious agents and schizophrenia. Clin Psychopharmacol Neurosci. (2012) 10:117–23. doi: 10.9758/cpn.2012.10.2.117
24. Krause D, Matz J, Weidinger E, Wagner J, Wildenauer A, Obermeier M, et al. The association of infectious agents and schizophrenia. World J Biol Psychiatry. (2010) 11:739–43. doi: 10.3109/15622971003653246
25. Fellerhoff B, Laumbacher B, Mueller N, Gu S, and Wank R. Associations between chlamydophila infections, schizophrenia and risk of hla-A10. Mol Psychiatry. (2007) 12:264–72. doi: 10.1038/sj.mp.4001925
26. Soleimani MF, Ayubi E, Khosronezhad S, Hasler G, Amiri MR, Beikpour F, et al. Human endogenous retroviruses type W (Herv-) activation and schizophrenia: A meta-analysis. Schizophr Res. (2024) 271:220–7. doi: 10.1016/j.schres.2024.07.014
27. Rangel SC, da Silva MD, Natrielli Filho DG, Santos SN, do Amaral JB, Victor JR, et al. Herv-W upregulation expression in bipolar disorder and schizophrenia: unraveling potential links to systemic immune/inflammation status. Retrovirology. (2024) 21:7. doi: 10.1186/s12977-024-00640-3
28. Mohagheghi M, Eftekharian MM, Taheri M, and Alikhani MY. Determining the igm and igg antibodies titer against hsv1, hsv2 and cmv in the serum of schizophrenia patients. Hum antibodies. (2018) 26:87–93. doi: 10.3233/hab-170325
29. Azami M, Jalilian FA, Khorshidi A, Mohammadi Y, and Tardeh Z. The association between borna disease virus and schizophrenia: A systematic review and meta-analysis. Asian J Psychiatry. (2018) 34:67–73. doi: 10.1016/j.ajp.2017.11.026
30. Arias I, Sorlozano A, Villegas E, de Dios Luna J, McKenney K, Cervilla J, et al. Infectious agents associated with schizophrenia: A meta-analysis. Schizophr Res. (2012) 136:128–36. doi: 10.1016/j.schres.2011.10.026
31. Spencer SJ and Meyer U. Perinatal programming by inflammation. Brain Behav Immun. (2017) 63:1–7. doi: 10.1016/j.bbi.2017.02.007
32. Prandovszky E, Gaskell E, Martin H, Dubey JP, Webster JP, and McConkey GA. The neurotropic parasite toxoplasma gondii increases dopamine metabolism. PLoS One. (2011) 6:e23866. doi: 10.1371/journal.pone.0023866
33. Torrey EF and Yolken RH. Toxoplasma gondii and schizophrenia. Emerg Infect Dis. (2003) 9:1375–80. doi: 10.3201/eid0911.030143
34. Carruthers VB and Suzuki Y. Effects of toxoplasma gondii infection on the brain. Schizophr Bull. (2007) 33:745–51. doi: 10.1093/schbul/sbm008
35. Bhadra R, Cobb DA, Weiss LM, and Khan IA. Psychiatric disorders in toxoplasma seropositive patients–the cd8 connection. Schizophr Bull. (2013) 39:485–9. doi: 10.1093/schbul/sbt006
36. Molloy MJ, Grainger JR, Bouladoux N, Hand TW, Koo LY, Naik S, et al. Intraluminal containment of commensal outgrowth in the gut during infection-induced dysbiosis. Cell Host Microbe. (2013) 14:318–28. doi: 10.1016/j.chom.2013.08.003
37. Tsuang MT, Stone WS, and Faraone SV. Genes, environment and schizophrenia. Br J Psychiatry Suppl. (2001) 40:s18–24. doi: 10.1192/bjp.178.40.s18
38. Guarner F, Hooper LV, and Núñez G. Understanding the microbiota in the midst of renaissance architecture and olive groves. Nat Immunol. (2013) 14:101–5. doi: 10.1038/ni.2512
39. Dinan TG, Borre YE, and Cryan JF. Genomics of schizophrenia: time to consider the gut microbiome? Mol Psychiatry. (2014) 19:1252–7. doi: 10.1038/mp.2014.93
40. Ermakov EA, Melamud MM, Buneva VN, and Ivanova SA. Immune system abnormalities in schizophrenia: an integrative view and translational perspectives. Front Psychiatry. (2022) 13:880568. doi: 10.3389/fpsyt.2022.880568
41. DiGiulio DB. Diversity of microbes in amniotic fluid. Semin Fetal Neonatal Med. (2012) 17:2–11. doi: 10.1016/j.siny.2011.10.001
42. Jiménez E, Fernández L, Marín ML, Martín R, Odriozola JM, Nueno-Palop C, et al. Isolation of commensal bacteria from umbilical cord blood of healthy neonates born by cesarean section. Curr Microbiol. (2005) 51:270–4. doi: 10.1007/s00284-005-0020-3
43. Aagaard K, Ma J, Antony KM, Ganu R, Petrosino J, and Versalovic J. The placenta harbors a unique microbiome. Sci Transl Med. (2014) 6:237ra65. doi: 10.1126/scitranslmed.3008599
44. Clemente JC, Ursell LK, Parfrey LW, and Knight R. The impact of the gut microbiota on human health: an integrative view. Cell. (2012) 148:1258–70. doi: 10.1016/j.cell.2012.01.035
45. Maynard CL, Elson CO, Hatton RD, and Weaver CT. Reciprocal interactions of the intestinal microbiota and immune system. Nature. (2012) 489:231–41. doi: 10.1038/nature11551
46. Lozupone CA, Stombaugh JI, Gordon JI, Jansson JK, and Knight R. Diversity, stability and resilience of the human gut microbiota. Nature. (2012) 489:220–30. doi: 10.1038/nature11550
47. Parfrey LW and Knight R. Spatial and temporal variability of the human microbiota. Clin Microbiol Infect. (2012) 18 Suppl 4:8–11. doi: 10.1111/j.1469-0691.2012.03861.x
48. Golubeva AV, Crampton S, Desbonnet L, Edge D, O’Sullivan O, Lomasney KW, et al. Prenatal stress-induced alterations in major physiological systems correlate with gut microbiota composition in adulthood. Psychoneuroendocrinology. (2015) 60:58–74. doi: 10.1016/j.psyneuen.2015.06.002
49. Gur TL, Shay L, Palkar AV, Fisher S, Varaljay VA, Dowd S, et al. Prenatal stress affects placental cytokines and neurotrophins, commensal microbes, and anxiety-like behavior in adult female offspring. Brain Behav Immun. (2017) 64:50–8. doi: 10.1016/j.bbi.2016.12.021
50. Jašarević E, Howard CD, Misic AM, Beiting DP, and Bale TL. Stress during pregnancy alters temporal and spatial dynamics of the maternal and offspring microbiome in a sex-specific manner. Sci Rep. (2017) 7:44182. doi: 10.1038/srep44182
51. Jin Y, Wu S, Zeng Z, and Fu Z. Effects of environmental pollutants on gut microbiota. Environ pollut. (2017) 222:1–9. doi: 10.1016/j.envpol.2016.11.045
52. Nuncio-Mora L, Lanzagorta N, Nicolini H, Sarmiento E, Ortiz G, Sosa F, et al. The role of the microbiome in first episode of psychosis. Biomedicines. (2023) 11:1770. doi: 10.3390/biomedicines11061770
53. Xu R, Wu B, Liang J, He F, Gu W, Li K, et al. Altered gut microbiota and mucosal immunity in patients with schizophrenia. Brain Behav Immun. (2020) 85:120–7. doi: 10.1016/j.bbi.2019.06.039
54. Wang Y, Bi S, Li X, Zhong Y, and Qi D. Perturbations in gut microbiota composition in schizophrenia. PLoS One. (2024) 19:e0306582. doi: 10.1371/journal.pone.0306582
55. Zhuang Z, Yang R, Wang W, Qi L, and Huang T. Associations between gut microbiota and alzheimer’s disease, major depressive disorder, and schizophrenia. J Neuroinflamm. (2020) 17:288. doi: 10.1186/s12974-020-01961-8
56. Ling Z, Lan Z, Cheng Y, Liu X, Li Z, Yu Y, et al. Altered gut microbiota and systemic immunity in chinese patients with schizophrenia comorbid with metabolic syndrome. J Transl Med. (2024) 22:729. doi: 10.1186/s12967-024-05533-9
57. Murray N, Al Khalaf S, Bastiaanssen TFS, Kaulmann D, Lonergan E, Cryan JF, et al. Compositional and functional alterations in intestinal microbiota in patients with psychosis or schizophrenia: A systematic review and meta-analysis. Schizophr Bull. (2023) 49:1239–55. doi: 10.1093/schbul/sbad049
58. Wu H, Jiawei X, Wen Z, Han Y, Liu Y, Chen S, et al. Microbiome-gut-brain profiles in schizophrenia and their potential link to cognitive performance: findings from a case-control study. Schizophr Bull. (2025). doi: 10.1093/schbul/sbaf028
59. Yuan X, Wang Y, Li X, Jiang J, Kang Y, Pang L, et al. Gut microbial biomarkers for the treatment response in first-episode, drug-naïve schizophrenia: A 24-week follow-up study. Transl Psychiatry. (2021) 11:422. doi: 10.1038/s41398-021-01531-3
60. Coughlin JM, Ishizuka K, Kano SI, Edwards JA, Seifuddin FT, Shimano MA, et al. Marked reduction of soluble superoxide dismutase-1 (Sod1) in cerebrospinal fluid of patients with recent-onset schizophrenia. Mol Psychiatry. (2013) 18:10–1. doi: 10.1038/mp.2012.6
61. Dickerson F, Severance E, and Yolken R. The microbiome, immunity, and schizophrenia and bipolar disorder. Brain Behav Immun. (2017) 62:46–52. doi: 10.1016/j.bbi.2016.12.010
62. Severance EG, Alaedini A, Yang S, Halling M, Gressitt KL, Stallings CR, et al. Gastrointestinal inflammation and associated immune activation in schizophrenia. Schizophr Res. (2012) 138:48–53. doi: 10.1016/j.schres.2012.02.025
63. Severance EG, Gressitt KL, Yang S, Stallings CR, Origoni AE, Vaughan C, et al. Seroreactive marker for inflammatory bowel disease and associations with antibodies to dietary proteins in bipolar disorder. Bipolar Disord. (2014) 16:230–40. doi: 10.1111/bdi.12159
64. Severance EG, Prandovszky E, Castiglione J, and Yolken RH. Gastroenterology issues in schizophrenia: why the gut matters. Curr Psychiatry Rep. (2015) 17:27. doi: 10.1007/s11920-015-0574-0
65. Severance EG, Yolken RH, and Eaton WW. Autoimmune diseases, gastrointestinal disorders and the microbiome in schizophrenia: more than a gut feeling. Schizophr Res. (2016) 176:23–35. doi: 10.1016/j.schres.2014.06.027
66. Nguyen TT, Kosciolek T, Eyler LT, Knight R, and Jeste DV. Overview and systematic review of studies of microbiome in schizophrenia and bipolar disorder. J Psychiatr Res. (2018) 99:50–61. doi: 10.1016/j.jpsychires.2018.01.013
67. Li S, Zhuo M, Huang X, Huang Y, Zhou J, Xiong D, et al. Altered gut microbiota associated with symptom severity in schizophrenia. PeerJ. (2020) 8:e9574. doi: 10.7717/peerj.9574
68. Schwarz E, Maukonen J, Hyytiäinen T, Kieseppä T, Orešič M, Sabunciyan S, et al. Analysis of microbiota in first episode psychosis identifies preliminary associations with symptom severity and treatment response. Schizophr Res. (2018) 192:398–403. doi: 10.1016/j.schres.2017.04.017
69. Xing M, Gao H, Yao L, Wang L, Zhang C, Zhu L, et al. Profiles and diagnostic value of intestinal microbiota in schizophrenia patients with metabolic syndrome. Front Endocrinol (Lausanne). (2023) 14:1190954. doi: 10.3389/fendo.2023.1190954
70. Nemani K, Hosseini Ghomi R, McCormick B, and Fan X. Schizophrenia and the gut-brain axis. Prog Neuropsychopharmacol Biol Psychiatry. (2015) 56:155–60. doi: 10.1016/j.pnpbp.2014.08.018
71. Fan Y, Gao Y, Ma Q, Yang Z, Zhao B, He X, et al. Multi-omics analysis reveals aberrant gut-metabolome-immune network in schizophrenia. Front Immunol. (2022) 13:812293. doi: 10.3389/fimmu.2022.812293
72. Li C, Wang A, Wang C, Ramamurthy J, Zhang E, Guadagno E, et al. Metabolomics in patients with psychosis: A systematic review. Am J Med Genet B Neuropsychiatr Genet. (2018) 177:580–8. doi: 10.1002/ajmg.b.32662
73. Krzyściak W, Bystrowska B, Karcz P, Chrzan R, Bryll A, Turek A, et al. Association of blood metabolomics biomarkers with brain metabolites and patient-reported outcomes as a new approach in individualized diagnosis of schizophrenia. Int J Mol Sci. (2024) 25:2294. doi: 10.3390/ijms25042294
74. Shi L, Ju P, Meng X, Wang Z, Yao L, Zheng M, et al. Intricate role of intestinal microbe and metabolite in schizophrenia. BMC Psychiatry. (2023) 23:856. doi: 10.1186/s12888-023-05329-z
75. Li S, Song J, Ke P, Kong L, Lei B, Zhou J, et al. The gut microbiome is associated with brain structure and function in schizophrenia. Sci Rep. (2021) 11:9743. doi: 10.1038/s41598-021-89166-8
76. Fogelson KA, Dorrestein PC, Zarrinpar A, and Knight R. The gut microbial bile acid modulation and its relevance to digestive health and diseases. Gastroenterology. (2023) 164:1069–85. doi: 10.1053/j.gastro.2023.02.022
77. Liang Y, Shi X, Shen Y, Huang Z, Wang J, Shao C, et al. Enhanced intestinal protein fermentation in schizophrenia. BMC Med. (2022) 20:67. doi: 10.1186/s12916-022-02261-z
78. Zhu F, Ju Y, Wang W, Wang Q, Guo R, Ma Q, et al. Metagenome-wide association of gut microbiome features for schizophrenia. Nat Commun. (2020) 11:1612. doi: 10.1038/s41467-020-15457-9
79. Nguyen TT, Kosciolek T, Daly RE, Vázquez-Baeza Y, Swafford A, Knight R, et al. Gut microbiome in schizophrenia: altered functional pathways related to immune modulation and atherosclerotic risk. Brain Behav Immun. (2021) 91:245–56. doi: 10.1016/j.bbi.2020.10.003
80. Qing Y, Wang P, Cui G, Zhang J, Liang K, Xia Z, et al. Targeted metabolomics reveals aberrant profiles of serum bile acids in patients with schizophrenia. Schizophr (Heidelb). (2022) 8:65. doi: 10.1038/s41537-022-00273-5
81. Khosravi M. Ursodeoxycholic acid augmentation in treatment-refractory schizophrenia: A case report. J Med Case Rep. (2020) 14:137. doi: 10.1186/s13256-020-02484-9
82. Quinn M, McMillin M, Galindo C, Frampton G, Pae HY, and DeMorrow S. Bile acids permeabilize the blood brain barrier after bile duct ligation in rats via rac1-dependent mechanisms. Dig Liver Dis. (2014) 46:527–34. doi: 10.1016/j.dld.2014.01.159
83. Tofalo R, Cocchi S, and Suzzi G. Polyamines and gut microbiota. Front Nutr. (2019) 6:16. doi: 10.3389/fnut.2019.00016
84. Richardson-Andrews RC. A central role for the polyamines in the aetiology of schizophrenia. Med Hypotheses. (1983) 11:157–66. doi: 10.1016/0306-9877(83)90059-2
85. Spathopoulou A, Sauerwein GA, Marteau V, Podlesnic M, Lindlbauer T, Kipura T, et al. Integrative metabolomics-genomics analysis identifies key networks in a stem cell-based model of schizophrenia. Mol Psychiatry. (2024) 29:3128–40. doi: 10.1038/s41380-024-02568-8
86. Leppik L, Kriisa K, Koido K, Koch K, Kajalaid K, Haring L, et al. Profiling of Amino Acids and Their Derivatives Biogenic Amines before and after Antipsychotic Treatment in First-Episode Psychosis. Front Psychiatry. (2018) 9:155. doi: 10.3389/fpsyt.2018.00155
87. Svinarev VI. Serum spermidine levels of schizophrenic patients. Zh Nevropatol Psikhiatr Im S S Korsakova. (1987) 87:732–4.
88. Ramchand CN, Das I, Gliddon A, and Hirsch SR. Role of polyamines in the membrane pathology of schizophrenia. A study using fibroblasts from schizophrenic patients and normal controls. Schizophr Res. (1994) 13:249–53. doi: 10.1016/0920-9964(94)90049-3
89. Gilad GM, Gilad VH, Casanova MF, and Casero RA Jr. Polyamines and their metabolizing enzymes in human frontal cortex and hippocampus: preliminary measurements in affective disorders. Biol Psychiatry. (1995) 38:227–34. doi: 10.1016/0006-3223(94)00256-3
90. Garip B, Kayir H, and Uzun O. L-Arginine Metabolism before and after 10 Weeks of Antipsychotic Treatment in First-Episode Psychotic Patients. Schizophr Res. (2019) 206:58–66. doi: 10.1016/j.schres.2018.12.015
91. Uzbay T, Goktalay G, Kayir H, Eker SS, Sarandol A, Oral S, et al. Increased plasma agmatine levels in patients with schizophrenia. J Psychiatr Res. (2013) 47:1054–60. doi: 10.1016/j.jpsychires.2013.04.004
92. Liu P, Jing Y, Collie ND, Dean B, Bilkey DK, and Zhang H. Altered brain arginine metabolism in schizophrenia. Transl Psychiatry. (2016) 6:e871. doi: 10.1038/tp.2016.144
93. Knox LT, Jing Y, Bawazier-Edgecombe J, Collie ND, Zhang H, and Liu P. Effects of withdrawal from repeated phencyclidine administration on behavioural function and brain arginine metabolism in rats. Pharmacol Biochem Behav. (2017) 153:45–59. doi: 10.1016/j.pbb.2016.12.006
94. Syatkin S, Smirnova IP, Kuznetsova O, Lobaeva TA, Blagonravov M, Ryskina EA, et al. The Levels of Polyamines in Autopsy Materials of Some Structures of Brain Lymbic System and Reticular Formation of Patients with Schizophrenia. Research Journal of Pharmaceutical, Biological and Chemical Sciences. (2014) 5:1486–90.
95. Bernstein HG and Müller M. The cellular localization of the L-ornithine decarboxylase/polyamine system in normal and diseased central nervous systems. Prog Neurobiol. (1999) 57:485–505. doi: 10.1016/s0301-0082(98)00065-3
96. Flayeh KA. Spermidine oxidase activity in serum of normal and schizophrenic subjects. Clin Chem. (1988) 34:401–3. doi: 10.1093/clinchem/34.2.401
97. Dahel KA, Al-Saffar NM, and Flayeh KA. Polyamine oxidase activity in sera of depressed and schizophrenic patients after ect treatment. Neurochem Res. (2001) 26:415–8. doi: 10.1023/a:1010959300545
98. Middleton FA, Mirnics K, Pierri JN, Lewis DA, and Levitt P. Gene expression profiling reveals alterations of specific metabolic pathways in schizophrenia. J Neurosci. (2002) 22:2718–29. doi: 10.1523/jneurosci.22-07-02718.2002
99. Ayalew M, Le-Niculescu H, Levey DF, Jain N, Changala B, Patel SD, et al. Convergent functional genomics of schizophrenia: from comprehensive understanding to genetic risk prediction. Mol Psychiatry. (2012) 17:887–905. doi: 10.1038/mp.2012.37
100. Williams K. Modulation and block of ion channels: A new biology of polyamines. Cell Signal. (1997) 9:1–13. doi: 10.1016/s0898-6568(96)00089-7
101. Schmitz MP, Combs DJ, and Dempsey RJ. Difluoromethylornithine decreases postischemic brain edema and blood-brain barrier breakdown. Neurosurgery. (1993) 33:882–7. doi: 10.1227/00006123-199311000-00016
102. Temiz C, Dogan A, Baskaya MK, and Dempsey RJ. Effect of difluoromethylornithine on reperfusion injury after temporary middle cerebral artery occlusion. J Clin Neurosci. (2005) 12:449–52. doi: 10.1016/j.jocn.2004.05.019
103. Wu D, Chen Q, Chen X, Han F, Chen Z, and Wang Y. The blood-brain barrier: structure, regulation, and drug delivery. Signal Transduct Target Ther. (2023) 8:217. doi: 10.1038/s41392-023-01481-w
104. Wishart DS, Feunang YD, Marcu A, Guo AC, Liang K, Vázquez-Fresno R, et al. Hmdb 4.0: the human metabolome database for 2018. Nucleic Acids Res. (2018) 46:D608–d17. doi: 10.1093/nar/gkx1089
105. Braniste V, Al-Asmakh M, Kowal C, Anuar F, Abbaspour A, Tóth M, et al. The gut microbiota influences blood-brain barrier permeability in mice. Sci Transl Med. (2014) 6:263ra158. doi: 10.1126/scitranslmed.3009759
106. Sun N, Hu H, Wang F, Li L, Zhu W, Shen Y, et al. Antibiotic-induced microbiome depletion in adult mice disrupts blood-brain barrier and facilitates brain infiltration of monocytes after bone-marrow transplantation. Brain Behav Immun. (2021) 92:102–14. doi: 10.1016/j.bbi.2020.11.032
107. Chenghan M, Wanxin L, Bangcheng Z, Yao H, Qinxi L, Ting Z, et al. Short-chain fatty acids mediate gut microbiota-brain communication and protect the blood-brain barrier integrity. Ann N Y Acad Sci. (2025) 1545:116–31. doi: 10.1111/nyas.15299
108. Wu Q, Zhang Y, Zhang Y, Xia C, Lai Q, Dong Z, et al. Potential effects of antibiotic-induced gut microbiome alteration on blood-brain barrier permeability compromise in rhesus monkeys. Ann N Y Acad Sci. (2020) 1470:14–24. doi: 10.1111/nyas.14312
109. Uhlén M, Fagerberg L, Hallström BM, Lindskog C, Oksvold P, Mardinoglu A, et al. Proteomics. Tissue-based map of the human proteome. Science. (2015) 347:1260419. doi: 10.1126/science.1260419
110. Hoyles L, Snelling T, Umlai UK, Nicholson JK, Carding SR, Glen RC, et al. Microbiome-host systems interactions: protective effects of propionate upon the blood-brain barrier. Microbiome. (2018) 6:55. doi: 10.1186/s40168-018-0439-y
111. Ji J, Shu D, Zheng M, Wang J, Luo C, Wang Y, et al. Microbial metabolite butyrate facilitates M2 macrophage polarization and function. Sci Rep. (2016) 6:24838. doi: 10.1038/srep24838
112. Nastasi C, Candela M, Bonefeld CM, Geisler C, Hansen M, Krejsgaard T, et al. The effect of short-chain fatty acids on human monocyte-derived dendritic cells. Sci Rep. (2015) 5:16148. doi: 10.1038/srep16148
113. Tan J, McKenzie C, Potamitis M, Thorburn AN, Mackay CR, and Macia L. The role of short-chain fatty acids in health and disease. Adv Immunol. (2014) 121:91–119. doi: 10.1016/b978-0-12-800100-4.00003-9
114. Purves-Tyson TD, Robinson K, Brown AM, Boerrigter D, Cai HQ, Weissleder C, et al. Increased macrophages and C1qa, C3, C4 transcripts in the midbrain of people with schizophrenia. Front Immunol. (2020) 11:2002. doi: 10.3389/fimmu.2020.02002
115. Severance EG, Gressitt KL, Buka SL, Cannon TD, and Yolken RH. Maternal complement C1q and increased odds for psychosis in adult offspring. Schizophr Res. (2014) 159:14–9. doi: 10.1016/j.schres.2014.07.053
116. Kopczynska M, Zelek W, Touchard S, Gaughran F, Di Forti M, Mondelli V, et al. Complement system biomarkers in first episode psychosis. Schizophr Res. (2019) 204:16–22. doi: 10.1016/j.schres.2017.12.012
117. Jenkins AK, Lewis DA, and Volk DW. Altered expression of microglial markers of phagocytosis in schizophrenia. Schizophr Res. (2023) 251:22–9. doi: 10.1016/j.schres.2022.12.005
118. Yu H, Ni P, Tian Y, Zhao L, Li M, Li X, et al. Association of elevated levels of peripheral complement components with cortical thinning and impaired logical memory in drug-naïve patients with first-episode schizophrenia. Schizophr (Heidelb). (2023) 9:79. doi: 10.1038/s41537-023-00409-1
119. Cao Y, Xu Y, Xia Q, Shan F, and Liang J. Peripheral complement factor-based biomarkers for patients with first-episode schizophrenia. Neuropsychiatr Dis Treat. (2023) 19:1455–62. doi: 10.2147/ndt.S420475
120. Koskuvi M, Malwade S, Gracias Lekander J, Hörbeck E, Bruno S, Holmen Larsson J, et al. Lower complement C1q levels in first-episode psychosis and in schizophrenia. Brain Behav Immun. (2024) 117:313–9. doi: 10.1016/j.bbi.2024.01.219
121. Laskaris L, Zalesky A, Weickert CS, Di Biase MA, Chana G, Baune BT, et al. Investigation of peripheral complement factors across stages of psychosis. Schizophr Res. (2019) 204:30–7. doi: 10.1016/j.schres.2018.11.035
122. Li H, Zhang Q, Li N, Wang F, Xiang H, Zhang Z, et al. Plasma levels of th17-related cytokines and complement C3 correlated with aggressive behavior in patients with schizophrenia. Psychiatry Res. (2016) 246:700–6. doi: 10.1016/j.psychres.2016.10.061
123. Kamitaki N, Sekar A, Handsaker RE, de Rivera H, Tooley K, Morris DL, et al. Complement genes contribute sex-biased vulnerability in diverse disorders. Nature. (2020) 582:577–81. doi: 10.1038/s41586-020-2277-x
124. Xia XW, Li LJ, Chen ZC, Qiu Y, Zhao JS, Wu JY, et al. Correlation of the peripheral serum complement protein levels and cognitive function in first-episode drug-naive patients with schizophrenia. Zhonghua Yi Xue Za Zhi. (2020) 100:3081–5. doi: 10.3760/cma.j.cn112137-20200425-01316
125. Mohd Asyraf AJ, Nour El Huda AR, Hanisah MN, Norsidah KZ, and Norlelawati AT. Relationship of selective complement markers with schizophrenia. J Neuroimmunol. (2022) 363:577793. doi: 10.1016/j.jneuroim.2021.577793
126. Borbye-Lorenzen N, Zhu Z, Agerbo E, Albiñana C, Benros ME, Bian B, et al. The correlates of neonatal complement component 3 and 4 protein concentrations with a focus on psychiatric and autoimmune disorders. Cell Genom. (2023) 3:100457. doi: 10.1016/j.xgen.2023.100457
127. Ji E, Boerrigter D, Cai HQ, Lloyd D, Bruggemann J, O’Donnell M, et al. Peripheral complement is increased in schizophrenia and inversely related to cortical thickness. Brain Behav Immun. (2022) 101:423–34. doi: 10.1016/j.bbi.2021.11.014
128. Chen Y, Zhao Z, Lin F, Wang L, Lin Z, and Yue W. Associations between genotype and peripheral complement proteins in first-episode psychosis: evidences from C3 and C4. Front Genet. (2021) 12:647246. doi: 10.3389/fgene.2021.647246
129. Kalinowski A, Liliental J, Anker LA, Linkovski O, Culbertson C, Hall JN, et al. Increased activation product of complement 4 protein in plasma of individuals with schizophrenia. Transl Psychiatry. (2021) 11:486. doi: 10.1038/s41398-021-01583-5
130. Zhang S, Zhou N, Liu R, Rao W, Yang M, Cao B, et al. Association between polymorphisms of the complement 3 gene and schizophrenia in a han chinese population. Cell Physiol Biochem. (2018) 46:2480–6. doi: 10.1159/000489654
131. Nsaiba MJ, Lapointe M, Mabrouk H, Douki W, Gaha L, Pérusse L, et al. C3 polymorphism influences circulating levels of C3, asp and lipids in schizophrenic patients. Neurochem Res. (2015) 40:906–14. doi: 10.1007/s11064-015-1543-z
132. Sekar A, Bialas AR, de Rivera H, Davis A, Hammond TR, Kamitaki N, et al. Schizophrenia risk from complex variation of complement component 4. Nature. (2016) 530:177–83. doi: 10.1038/nature16549
133. Yilmaz M, Yalcin E, Presumey J, Aw E, Ma M, Whelan CW, et al. Overexpression of schizophrenia susceptibility factor human complement C4a promotes excessive synaptic loss and behavioral changes in mice. Nat Neurosci. (2021) 24:214–24. doi: 10.1038/s41593-020-00763-8
134. Comer AL, Jinadasa T, Sriram B, Phadke RA, Kretsge LN, Nguyen TPH, et al. Increased expression of schizophrenia-associated gene C4 leads to hypoconnectivity of prefrontal cortex and reduced social interaction. PLoS Biol. (2020) 18:e3000604. doi: 10.1371/journal.pbio.3000604
135. Zhang T, Tang Y, Yang X, Wang X, Ding S, Huang K, et al. Expression of gsk3β, pick1, nefl, C4, nkcc1 and synaptophysin in peripheral blood mononuclear cells of the first-episode schizophrenia patients. Asian J Psychiatr. (2021) 55:102520. doi: 10.1016/j.ajp.2020.102520
136. Rey R, Suaud-Chagny MF, Bohec AL, Dorey JM, d’Amato T, Tamouza R, et al. Overexpression of complement component C4 in the dorsolateral prefrontal cortex, parietal cortex, superior temporal gyrus and associative striatum of patients with schizophrenia. Brain Behav Immun. (2020) 90:216–25. doi: 10.1016/j.bbi.2020.08.019
137. Prasad KM, Chowdari KV, D’Aiuto LA, Iyengar S, Stanley JA, and Nimgaonkar VL. Neuropil contraction in relation to complement C4 gene copy numbers in independent cohorts of adolescent-onset and young adult-onset schizophrenia patients-a pilot study. Transl Psychiatry. (2018) 8:134. doi: 10.1038/s41398-018-0181-z
138. Chen CC, Howie J, Ebrahimi M, Teymouri K, Woo JJ, Tiwari AK, et al. Analysis of the complement component C4 gene with schizophrenia subphenotypes. Schizophr Res. (2024) 271:309–18. doi: 10.1016/j.schres.2024.07.039
139. Ji RN, Zhang LL, Zhao MF, He HF, Bai W, Duan RX, et al. Decreased serum complement component 4 levels in patients with schizophrenia. Psychiatr Genet. (2019) 29:127–9. doi: 10.1097/ypg.0000000000000226
140. Gracias J, Orhan F, Hörbeck E, Holmén-Larsson J, Khanlarkani N, Malwade S, et al. Cerebrospinal fluid concentration of complement component 4a is increased in first episode schizophrenia. Nat Commun. (2022) 13:6427. doi: 10.1038/s41467-022-33797-6
141. Ishii T, Hattori K, Miyakawa T, Watanabe K, Hidese S, Sasayama D, et al. Increased cerebrospinal fluid complement C5 levels in major depressive disorder and schizophrenia. Biochem Biophys Res Commun. (2018) 497:683–8. doi: 10.1016/j.bbrc.2018.02.131
142. Severance EG and Yolken RH. From infection to the microbiome: an evolving role of microbes in schizophrenia. Curr Top Behav Neurosci. (2020) 44:67–84. doi: 10.1007/7854_2018_84
143. Severance EG, Leister F, Lea A, Yang S, Dickerson F, and Yolken RH. Complement C4 associations with altered microbial biomarkers exemplify gene-by-environment interactions in schizophrenia. Schizophr Res. (2021) 234:87–93. doi: 10.1016/j.schres.2021.02.001
144. Severance EG, Gressitt KL, Stallings CR, Origoni AE, Khushalani S, Leweke FM, et al. Discordant patterns of bacterial translocation markers and implications for innate immune imbalances in schizophrenia. Schizophr Res. (2013) 148:130–7. doi: 10.1016/j.schres.2013.05.018
145. Presumey J, Bialas AR, and Carroll MC. Complement system in neural synapse elimination in development and disease. Adv Immunol. (2017) 135:53–79. doi: 10.1016/bs.ai.2017.06.004
146. Nissilä E, Korpela K, Lokki AI, Paakkanen R, Jokiranta S, de Vos WM, et al. C4b gene influences intestinal microbiota through complement activation in patients with paediatric-onset inflammatory bowel disease. Clin Exp Immunol. (2017) 190:394–405. doi: 10.1111/cei.13040
147. Ifuku M, Hinkelmann L, Kuhrt LD, Efe IE, Kumbol V, Buonfiglioli A, et al. Activation of toll-like receptor 5 in microglia modulates their function and triggers neuronal injury. Acta Neuropathol Commun. (2020) 8:159. doi: 10.1186/s40478-020-01031-3
148. Schilling S, Chausse B, Dikmen HO, Almouhanna F, Hollnagel JO, Lewen A, et al. Tlr2- and tlr3-activated microglia induce different levels of neuronal network dysfunction in a context-dependent manner. Brain Behav Immun. (2021) 96:80–91. doi: 10.1016/j.bbi.2021.05.013
149. Weickert TW, Ji E, Galletly C, Boerrigter D, Morishima Y, Bruggemann J, et al. Toll-like receptor mrna levels in schizophrenia: association with complement factors and cingulate gyrus cortical thinning. Schizophr Bull. (2024) 50:403–17. doi: 10.1093/schbul/sbad171
150. McKernan DP, Dennison U, Gaszner G, Cryan JF, and Dinan TG. Enhanced peripheral toll-like receptor responses in psychosis: further evidence of a pro-inflammatory phenotype. Transl Psychiatry. (2011) 1:e36. doi: 10.1038/tp.2011.37
151. Akira S and Takeda K. Toll-like receptor signalling. Nat Rev Immunol. (2004) 4:499–511. doi: 10.1038/nri1391
152. García Bueno B, Caso JR, Madrigal JL, and Leza JC. Innate immune receptor toll-like receptor 4 signalling in neuropsychiatric diseases. Neurosci Biobehav Rev. (2016) 64:134–47. doi: 10.1016/j.neubiorev.2016.02.013
153. Balaji R, Subbanna M, Shivakumar V, Abdul F, Venkatasubramanian G, and Debnath M. Pattern of expression of toll like receptor (Tlr)-3 and -4 genes in drug-naïve and antipsychotic treated patients diagnosed with schizophrenia. Psychiatry Res. (2020) 285:112727. doi: 10.1016/j.psychres.2019.112727
154. Kéri S, Szabó C, and Kelemen O. Antipsychotics influence toll-like receptor (Tlr) expression and its relationship with cognitive functions in schizophrenia. Brain Behav Immun. (2017) 62:256–64. doi: 10.1016/j.bbi.2016.12.011
155. Müller N, Wagner JK, Krause D, Weidinger E, Wildenauer A, Obermeier M, et al. Impaired monocyte activation in schizophrenia. Psychiatry Res. (2012) 198:341–6. doi: 10.1016/j.psychres.2011.12.049
156. García-Bueno B, Gassó P, MacDowell KS, Callado LF, Mas S, Bernardo M, et al. Evidence of activation of the toll-like receptor-4 proinflammatory pathway in patients with schizophrenia. J Psychiatry Neurosci. (2016) 41:E46–55. doi: 10.1503/jpn.150195
157. Wang Y, Sadike D, Huang B, Li P, Wu Q, Jiang N, et al. Regulatory T cells alleviate myelin loss and cognitive dysfunction by regulating neuroinflammation and microglial pyroptosis via tlr4/myd88/nf-Κb pathway in lpc-induced demyelination. J Neuroinflamm. (2023) 20:41. doi: 10.1186/s12974-023-02721-0
158. Zhong Q, Zou Y, Liu H, Chen T, Zheng F, Huang Y, et al. Toll-like receptor 4 deficiency ameliorates Β2-microglobulin induced age-related cognition decline due to neuroinflammation in mice. Mol Brain. (2020) 13:20. doi: 10.1186/s13041-020-0559-8
159. Dai QD, Wu KS, Xu LP, Zhang Y, Lin N, Jiang Y, et al. Toll-like receptor 4 deficiency ameliorates propofol-induced impairments of cognitive function and synaptic plasticity in young mice. Mol Neurobiol. (2024) 61:519–32. doi: 10.1007/s12035-023-03606-2
160. Xiao X, Zhang H, Ning W, Yang Z, Wang Y, and Zhang T. Knockdown of fstl1 inhibits microglia activation and alleviates depressive-like symptoms through modulating tlr4/myd88/nf-Κb pathway in cums mice. Exp Neurol. (2022) 353:114060. doi: 10.1016/j.expneurol.2022.114060
161. Kim YK and Shin C. The microbiota-gut-brain axis in neuropsychiatric disorders: pathophysiological mechanisms and novel treatments. Curr Neuropharmacol. (2018) 16:559–73. doi: 10.2174/1570159x15666170915141036
162. MacDowell KS, Caso JR, Martín-Hernández D, Madrigal JL, Leza JC, and García-Bueno B. Paliperidone prevents brain toll-like receptor 4 pathway activation and neuroinflammation in rat models of acute and chronic restraint stress. Int J Neuropsychopharmacol. (2014) 18. doi: 10.1093/ijnp/pyu070
163. Gárate I, García-Bueno B, Madrigal JL, Caso JR, Alou L, Gómez-Lus ML, et al. Toll-like 4 receptor inhibitor tak-242 decreases neuroinflammation in rat brain frontal cortex after stress. J Neuroinflamm. (2014) 11:8. doi: 10.1186/1742-2094-11-8
164. Chen Y, Li D, Sun L, Qi K, and Shi L. Pharmacological inhibition of toll-like receptor 4 with tlr4-in-C34 modulates the intestinal flora homeostasis and the myd88/nf-Κb axis in ulcerative colitis. Eur J Pharmacol. (2022) 934:175294. doi: 10.1016/j.ejphar.2022.175294
165. Cryan JF, O’Riordan KJ, Cowan CSM, Sandhu KV, Bastiaanssen TFS, Boehme M, et al. The microbiota-gut-brain axis. Physiol Rev. (2019) 99:1877–2013. doi: 10.1152/physrev.00018.2018
166. González-Blanco L, Dal Santo F, García-Portilla MP, Alfonso M, Hernández C, Sánchez-Autet M, et al. Intestinal permeability biomarkers in patients with schizophrenia: additional support for the impact of lifestyle habits. Eur Psychiatry. (2024) 67:e84. doi: 10.1192/j.eurpsy.2024.1765
167. Schirmer M, Smeekens SP, Vlamakis H, Jaeger M, Oosting M, Franzosa EA, et al. Linking the human gut microbiome to inflammatory cytokine production capacity. Cell. (2016) 167:1125–36.e8. doi: 10.1016/j.cell.2016.10.020
168. Yarandi SS, Peterson DA, Treisman GJ, Moran TH, and Pasricha PJ. Modulatory effects of gut microbiota on the central nervous system: how gut could play a role in neuropsychiatric health and diseases. J Neurogastroenterol Motil. (2016) 22:201–12. doi: 10.5056/jnm15146
169. Mhanna A, Martini N, Hmaydoosh G, Hamwi G, Jarjanazi M, Zaifah G, et al. The correlation between gut microbiota and both neurotransmitters and mental disorders: A narrative review. Med (Baltimore). (2024) 103:e37114. doi: 10.1097/md.0000000000037114
170. Zheng P, Zeng B, Liu M, Chen J, Pan J, Han Y, et al. The gut microbiome from patients with schizophrenia modulates the glutamate-glutamine-gaba cycle and schizophrenia-relevant behaviors in mice. Sci Adv. (2019) 5:eaau8317. doi: 10.1126/sciadv.aau8317
171. González-Castro TB, Tovilla-Zárate CA, Juárez-Rojop IE, Hernández-Díaz Y, López-Narváez ML, and Ortiz-Ojeda RF. Effects of il-6/il-6r axis alterations in serum, plasma and cerebrospinal fluid with the schizophrenia: an updated review and meta-analysis of 58 studies. Mol Cell Biochem. (2024) 479:525–37. doi: 10.1007/s11010-023-04747-7
172. Yan J, Xia Q, Sun X, Yang P, Gao H, Pan Z, et al. Dysregulation of interleukin-8 is involved in the onset and relapse of schizophrenia: an independent validation and meta-analysis. Prog Neuropsychopharmacol Biol Psychiatry. (2024) 133:111018. doi: 10.1016/j.pnpbp.2024.111018
173. Patlola SR, Donohoe G, and McKernan DP. Anti-inflammatory effects of 2nd generation antipsychotics in patients with schizophrenia: A systematic review and meta-analysis. J Psychiatr Res. (2023) 160:126–36. doi: 10.1016/j.jpsychires.2023.01.042
174. Zhang Y, Wang J, Ye Y, Zou Y, Chen W, Wang Z, et al. Peripheral cytokine levels across psychiatric disorders: A systematic review and network meta-analysis. Prog Neuropsychopharmacol Biol Psychiatry. (2023) 125:110740. doi: 10.1016/j.pnpbp.2023.110740
175. Halstead S, Siskind D, Amft M, Wagner E, Yakimov V, Shih-Jung Liu Z, et al. Alteration patterns of peripheral concentrations of cytokines and associated inflammatory proteins in acute and chronic stages of schizophrenia: A systematic review and network meta-analysis. Lancet Psychiatry. (2023) 10:260–71. doi: 10.1016/s2215-0366(23)00025-1
176. Dunleavy C, Elsworthy RJ, Upthegrove R, Wood SJ, and Aldred S. Inflammation in first-episode psychosis: the contribution of inflammatory biomarkers to the emergence of negative symptoms, a systematic review and meta-analysis. Acta Psychiatr Scand. (2022) 146:6–20. doi: 10.1111/acps.13416
177. Çakici N, Sutterland AL, Penninx B, de Haan L, and van Beveren NJM. Changes in peripheral blood compounds following psychopharmacological treatment in drug-naïve first-episode patients with either schizophrenia or major depressive disorder: A meta-analysis. Psychol Med. (2021) 51:538–49. doi: 10.1017/s0033291721000155
178. Çakici N, Sutterland AL, Penninx B, Dalm VA, de Haan L, and van Beveren NJM. Altered peripheral blood compounds in drug-naïve first-episode patients with either schizophrenia or major depressive disorder: A meta-analysis. Brain Behav Immun. (2020) 88:547–58. doi: 10.1016/j.bbi.2020.04.039
179. Park S and Miller BJ. Meta-analysis of cytokine and C-reactive protein levels in high-risk psychosis. Schizophr Res. (2020) 226:5–12. doi: 10.1016/j.schres.2019.03.012
180. Fraguas D, Díaz-Caneja CM, Ayora M, Hernández-Álvarez F, Rodríguez-Quiroga A, Recio S, et al. Oxidative stress and inflammation in first-episode psychosis: A systematic review and meta-analysis. Schizophr Bull. (2019) 45:742–51. doi: 10.1093/schbul/sby125
181. Romeo B, Brunet-Lecomte M, Martelli C, and Benyamina A. Kinetics of cytokine levels during antipsychotic treatment in schizophrenia: A meta-analysis. Int J Neuropsychopharmacol. (2018) 21:828–36. doi: 10.1093/ijnp/pyy062
182. Zhang J, Luo W, Huang P, Peng L, and Huang Q. Maternal C-reactive protein and cytokine levels during pregnancy and the risk of selected neuropsychiatric disorders in offspring: A systematic review and meta-analysis. J Psychiatr Res. (2018) 105:86–94. doi: 10.1016/j.jpsychires.2018.09.002
183. Capuzzi E, Bartoli F, Crocamo C, Clerici M, and Carrà G. Acute variations of cytokine levels after antipsychotic treatment in drug-naïve subjects with a first-episode psychosis: A meta-analysis. Neurosci Biobehav Rev. (2017) 77:122–8. doi: 10.1016/j.neubiorev.2017.03.003
184. Goldsmith DR, Rapaport MH, and Miller BJ. A meta-analysis of blood cytokine network alterations in psychiatric patients: comparisons between schizophrenia, bipolar disorder and depression. Mol Psychiatry. (2016) 21:1696–709. doi: 10.1038/mp.2016.3
185. Upthegrove R, Manzanares-Teson N, and Barnes NM. Cytokine function in medication-naive first episode psychosis: A systematic review and meta-analysis. Schizophr Res. (2014) 155:101–8. doi: 10.1016/j.schres.2014.03.005
186. Wang AK and Miller BJ. Meta-analysis of cerebrospinal fluid cytokine and tryptophan catabolite alterations in psychiatric patients: comparisons between schizophrenia, bipolar disorder, and depression. Schizophr Bull. (2018) 44:75–83. doi: 10.1093/schbul/sbx035
187. Gallego JA, Blanco EA, Husain-Krautter S, Madeline Fagen E, Moreno-Merino P, Del Ojo-Jiménez JA, et al. Cytokines in cerebrospinal fluid of patients with schizophrenia spectrum disorders: new data and an updated meta-analysis. Schizophr Res. (2018) 202:64–71. doi: 10.1016/j.schres.2018.07.019
188. Pape K, Tamouza R, Leboyer M, and Zipp F. Immunoneuropsychiatry - novel perspectives on brain disorders. Nat Rev Neurol. (2019) 15:317–28. doi: 10.1038/s41582-019-0174-4
189. Pillinger T, Osimo EF, Brugger S, Mondelli V, McCutcheon RA, and Howes OD. A meta-analysis of immune parameters, variability, and assessment of modal distribution in psychosis and test of the immune subgroup hypothesis. Schizophr Bull. (2019) 45:1120–33. doi: 10.1093/schbul/sby160
190. Lv Y, Wen L, Hu W-J, Deng C, Ren H-W, Bao Y-N, et al. Schizophrenia in the genetic era: A review from development history, clinical features and genomic research approaches to insights of susceptibility genes. Metab Brain Dis. (2023) 39:147–171. doi: 10.1007/s11011-023-01271-x
191. Lv H, Guo M, Guo C, and He K. The interrelationships between cytokines and schizophrenia: A systematic review. Int J Mol Sci. (2024) 25:8477. doi: 10.3390/ijms25158477
192. Liang PY, Diao LH, Huang CY, Lian RC, Chen X, Li GG, et al. The pro-inflammatory and anti-inflammatory cytokine profile in peripheral blood of women with recurrent implantation failure. Reprod BioMed Online. (2015) 31:823–6. doi: 10.1016/j.rbmo.2015.08.009
193. Momtazmanesh S, Zare-Shahabadi A, and Rezaei N. Cytokine alterations in schizophrenia: an updated review. Front Psychiatry. (2019) 10:892. doi: 10.3389/fpsyt.2019.00892
194. Wang Y, Wei Y, Edmiston EK, Womer FY, Zhang X, Duan J, et al. Altered structural connectivity and cytokine levels in schizophrenia and genetic high-risk individuals: associations with disease states and vulnerability. Schizophr Res. (2020) 223:158–65. doi: 10.1016/j.schres.2020.05.044
195. Clark SM, Notarangelo FM, Li X, Chen S, Schwarcz R, and Tonelli LH. Maternal immune activation in rats blunts brain cytokine and kynurenine pathway responses to a second immune challenge in early adulthood. Prog Neuropsychopharmacol Biol Psychiatry. (2019) 89:286–94. doi: 10.1016/j.pnpbp.2018.09.011
196. Mondelli V, Blackman G, Kempton MJ, Pollak TA, Iyegbe C, Valmaggia LR, et al. Serum immune markers and transition to psychosis in individuals at clinical high risk. Brain Behav Immun. (2023) 110:290–6. doi: 10.1016/j.bbi.2023.03.014
197. Li H, Chen W, Gou M, Li W, Tong J, Zhou Y, et al. The relationship between tlr4/nf-Κb/il-1β Signaling, cognitive impairment, and white-matter integrity in patients with stable chronic schizophrenia. Front Psychiatry. (2022) 13:966657. doi: 10.3389/fpsyt.2022.966657
198. Jeon S, Kim SH, Shin SY, and Lee YH. Clozapine reduces toll-like receptor 4/nf-Κb-mediated inflammatory responses through inhibition of calcium/calmodulin-dependent akt activation in microglia. Prog Neuropsychopharmacol Biol Psychiatry. (2018) 81:477–87. doi: 10.1016/j.pnpbp.2017.04.012
199. Banati R and Hickie IB. Therapeutic signposts: using biomarkers to guide better treatment of schizophrenia and other psychotic disorders. Med J Aust. (2009) 190:S26–32. doi: 10.5694/j.1326-5377.2009.tb02371.x
200. Doorduin J, de Vries EF, Willemsen AT, de Groot JC, Dierckx RA, and Klein HC. Neuroinflammation in schizophrenia-related psychosis: A pet study. J Nucl Med. (2009) 50:1801–7. doi: 10.2967/jnumed.109.066647
201. van Berckel BN, Bossong MG, Boellaard R, Kloet R, Schuitemaker A, Caspers E, et al. Microglia activation in recent-onset schizophrenia: A quantitative (R)-[11c]Pk11195 positron emission tomography study. Biol Psychiatry. (2008) 64:820–2. doi: 10.1016/j.biopsych.2008.04.025
202. Lee SU, In HJ, Kwon MS, Park BO, Jo M, Kim MO, et al. Β-arrestin 2 mediates G protein-coupled receptor 43 signals to nuclear factor-Κb. Biol Pharm Bull. (2013) 36:1754–9. doi: 10.1248/bpb.b13-00312
203. Place RF, Noonan EJ, and Giardina C. Hdac inhibition prevents nf-kappa B activation by suppressing proteasome activity: down-regulation of proteasome subunit expression stabilizes I kappa B alpha. Biochem Pharmacol. (2005) 70:394–406. doi: 10.1016/j.bcp.2005.04.030
204. Zandi-Nejad K, Takakura A, Jurewicz M, Chandraker AK, Offermanns S, Mount D, et al. The role of hca2 (Gpr109a) in regulating macrophage function. FASEB J. (2013) 27:4366–74. doi: 10.1096/fj.12-223933
205. Pace TW and Miller AH. Cytokines and glucocorticoid receptor signaling. Relevance to major depression. Ann N Y Acad Sci. (2009) 1179:86–105. doi: 10.1111/j.1749-6632.2009.04984.x
206. Thomas R. The traf6-nf kappa B signaling pathway in autoimmunity: not just inflammation. Arthritis Res Ther. (2005) 7:170–3. doi: 10.1186/ar1784
207. Derry HM, Fagundes CP, Andridge R, Glaser R, Malarkey WB, and Kiecolt-Glaser JK. Lower subjective social status exaggerates interleukin-6 responses to a laboratory stressor. Psychoneuroendocrinology. (2013) 38:2676–85. doi: 10.1016/j.psyneuen.2013.06.026
208. Tao R, de Zoeten EF, Ozkaynak E, Chen C, Wang L, Porrett PM, et al. Deacetylase inhibition promotes the generation and function of regulatory T cells. Nat Med. (2007) 13:1299–307. doi: 10.1038/nm1652
209. Rooks MG and Garrett WS. Gut microbiota, metabolites and host immunity. Nat Rev Immunol. (2016) 16:341–52. doi: 10.1038/nri.2016.42
210. Wang L, de Zoeten EF, Greene MI, and Hancock WW. Immunomodulatory effects of deacetylase inhibitors: therapeutic targeting of foxp3+ Regulatory T cells. Nat Rev Drug Discov. (2009) 8:969–81. doi: 10.1038/nrd3031
211. Peng H, Ouyang L, Li D, Li Z, Yuan L, Fan L, et al. Short-chain fatty acids in patients with schizophrenia and ultra-high risk population. Front Psychiatry. (2022) 13:977538. doi: 10.3389/fpsyt.2022.977538
212. Li Z, Qing Y, Cui G, Li M, Liu T, Zeng Y, et al. Shotgun metagenomics reveals abnormal short-chain fatty acid-producing bacteria and glucose and lipid metabolism of the gut microbiota in patients with schizophrenia. Schizophr Res. (2023) 255:59–66. doi: 10.1016/j.schres.2023.03.005
213. Chen Y, Xu J, and Chen Y. Regulation of neurotransmitters by the gut microbiota and effects on cognition in neurological disorders. Nutrients. (2021) 13:2099. doi: 10.3390/nu13062099
214. Agus A, Planchais J, and Sokol H. Gut microbiota regulation of tryptophan metabolism in health and disease. Cell Host Microbe. (2018) 23:716–24. doi: 10.1016/j.chom.2018.05.003
215. Savitz J. The kynurenine pathway: A finger in every pie. Mol Psychiatry. (2020) 25:131–47. doi: 10.1038/s41380-019-0414-4
216. Golimbet VE, Korovaĭtseva GI, Gabaeva MV, Velikaia NV, Snegireva AA, Kasparov SV, et al. a study of il-1β and ido gene polymorphisms in patients with schizophrenia. Zh Nevrol Psikhiatr Im S S Korsakova. (2014) 114:46–9.
217. O’Connor JC, André C, Wang Y, Lawson MA, Szegedi SS, Lestage J, et al. Interferon-gamma and tumor necrosis factor-alpha mediate the upregulation of indoleamine 2,3-dioxygenase and the induction of depressive-like behavior in mice in response to bacillus calmette-guerin. J Neurosci. (2009) 29:4200–9. doi: 10.1523/jneurosci.5032-08.2009
218. Babcock TA and Carlin JM. Transcriptional activation of indoleamine dioxygenase by interleukin 1 and tumor necrosis factor alpha in interferon-treated epithelial cells. Cytokine. (2000) 12:588–94. doi: 10.1006/cyto.1999.0661
219. Connor TJ, Starr N, O’Sullivan JB, and Harkin A. Induction of indolamine 2,3-dioxygenase and kynurenine 3-monooxygenase in rat brain following a systemic inflammatory challenge: A role for ifn-gamma? Neurosci Lett. (2008) 441:29–34. doi: 10.1016/j.neulet.2008.06.007
220. Zunszain PA, Anacker C, Cattaneo A, Choudhury S, Musaelyan K, Myint AM, et al. Interleukin-1β: A new regulator of the kynurenine pathway affecting human hippocampal neurogenesis. Neuropsychopharmacology. (2012) 37:939–49. doi: 10.1038/npp.2011.277
221. Martín-Hernández D, Tendilla-Beltrán H, Madrigal JLM, García-Bueno B, Leza JC, and Caso JR. Chronic mild stress alters kynurenine pathways changing the glutamate neurotransmission in frontal cortex of rats. Mol Neurobiol. (2019) 56:490–501. doi: 10.1007/s12035-018-1096-7
222. Kennedy PJ, Cryan JF, Dinan TG, and Clarke G. Kynurenine pathway metabolism and the microbiota-gut-brain axis. Neuropharmacology. (2017) 112:399–412. doi: 10.1016/j.neuropharm.2016.07.002
223. Albuquerque EX and Schwarcz R. Kynurenic acid as an antagonist of Α7 nicotinic acetylcholine receptors in the brain: facts and challenges. Biochem Pharmacol. (2013) 85:1027–32. doi: 10.1016/j.bcp.2012.12.014
224. Moroni F, Cozzi A, Sili M, and Mannaioni G. Kynurenic acid: A metabolite with multiple actions and multiple targets in brain and periphery. J Neural Transm (Vienna). (2012) 119:133–9. doi: 10.1007/s00702-011-0763-x
225. Kanchanatawan B, Sirivichayakul S, Thika S, Ruxrungtham K, Carvalho AF, Geffard M, et al. Physio-somatic symptoms in schizophrenia: association with depression, anxiety, neurocognitive deficits and the tryptophan catabolite pathway. Metab Brain Dis. (2017) 32:1003–16. doi: 10.1007/s11011-017-9982-7
226. Müller N. Neuroprogression in schizophrenia and psychotic disorders: the possible role of inflammation. Mod Trends Pharmacopsychiatry. (2017) 31:1–9. doi: 10.1159/000470802
227. Bailey MT and Cryan JF. The microbiome as a key regulator of brain, behavior and immunity: commentary on the 2017 named series. Brain Behav Immun. (2017) 66:18–22. doi: 10.1016/j.bbi.2017.08.017
228. Reigstad CS, Salmonson CE, Rainey JF 3rd, Szurszewski JH, Linden DR, Sonnenburg JL, et al. Gut microbes promote colonic serotonin production through an effect of short-chain fatty acids on enterochromaffin cells. FASEB J. (2015) 29:1395–403. doi: 10.1096/fj.14-259598
229. Roth W, Zadeh K, Vekariya R, Ge Y, and Mohamadzadeh M. Tryptophan metabolism and gut-brain homeostasis. Int J Mol Sci. (2021) 22:2973. doi: 10.3390/ijms22062973
230. Desbonnet L, Clarke G, Traplin A, O’Sullivan O, Crispie F, Moloney RD, et al. Gut microbiota depletion from early adolescence in mice: implications for brain and behaviour. Brain Behav Immun. (2015) 48:165–73. doi: 10.1016/j.bbi.2015.04.004
231. Clarke G, Stilling RM, Kennedy PJ, Stanton C, Cryan JF, and Dinan TG. Minireview: gut microbiota: the neglected endocrine organ. Mol Endocrinol. (2014) 28:1221–38. doi: 10.1210/me.2014-1108
232. Myint AM and Kim YK. Network beyond ido in psychiatric disorders: revisiting neurodegeneration hypothesis. Prog Neuropsychopharmacol Biol Psychiatry. (2014) 48:304–13. doi: 10.1016/j.pnpbp.2013.08.008
233. Zádor F, Joca S, Nagy-Grócz G, Dvorácskó S, Szűcs E, Tömböly C, et al. Pro-inflammatory cytokines: potential links between the endocannabinoid system and the kynurenine pathway in depression. Int J Mol Sci. (2021) 22:5903. doi: 10.3390/ijms22115903
234. Cryan JF and Dinan TG. Mind-altering microorganisms: the impact of the gut microbiota on brain and behaviour. Nat Rev Neurosci. (2012) 13:701–12. doi: 10.1038/nrn3346
235. Wichers MC and Maes M. The role of indoleamine 2,3-dioxygenase (Ido) in the pathophysiology of interferon-alpha-induced depression. J Psychiatry Neurosci. (2004) 29:11–7. doi: 10.1139/jpn.0402
236. Lieben CK, Blokland A, Westerink B, and Deutz NE. Acute tryptophan and serotonin depletion using an optimized tryptophan-free protein-carbohydrate mixture in the adult rat. Neurochem Int. (2004) 44:9–16. doi: 10.1016/s0197-0186(03)00102-5
237. Cahir M, Ardis T, Reynolds GP, and Cooper SJ. Acute and chronic tryptophan depletion differentially regulate central 5-ht1a and 5-ht 2a receptor binding in the rat. Psychopharmacol (Berl). (2007) 190:497–506. doi: 10.1007/s00213-006-0635-5
238. Merchán A, Navarro SV, Klein AB, Aznar S, Campa L, Suñol C, et al. Tryptophan depletion affects compulsive behaviour in rats: strain dependent effects and associated neuromechanisms. Psychopharmacol (Berl). (2017) 234:1223–36. doi: 10.1007/s00213-017-4561-5
239. Moreno FA, Parkinson D, Palmer C, Castro WL, Misiaszek J, El Khoury A, et al. Csf neurochemicals during tryptophan depletion in individuals with remitted depression and healthy controls. Eur Neuropsychopharmacol. (2010) 20:18–24. doi: 10.1016/j.euroneuro.2009.10.003
240. Owens JK. Systematic reviews: brief overview of methods, limitations, and resources. Nurse Author Editor. (2021) 31:69–72. doi: 10.1111/nae2.28
241. Nakhal MM, Yassin LK, Alyaqoubi R, Saeed S, Alderei A, Alhammadi A, et al. The microbiota-gut-brain axis and neurological disorders: A comprehensive review. Life (Basel). (2024) 14:1234. doi: 10.3390/life14101234
242. Loh JS, Mak WQ, Tan LKS, Ng CX, Chan HH, Yeow SH, et al. Microbiota-gut-brain axis and its therapeutic applications in neurodegenerative diseases. Signal Transduct Target Ther. (2024) 9:37. doi: 10.1038/s41392-024-01743-1
243. Ding M, Lang Y, Shu H, Shao J, and Cui L. Microbiota-gut-brain axis and epilepsy: A review on mechanisms and potential therapeutics. Front Immunol. (2021) 12:742449. doi: 10.3389/fimmu.2021.742449
244. Zhu F, Tu H, and Chen T. The microbiota-gut-brain axis in depression: the potential pathophysiological mechanisms and microbiota combined antidepression effect. Nutrients. (2022) 14:2081. doi: 10.3390/nu14102081
245. Liu P, Liu Z, Wang J, Wang J, Gao M, Zhang Y, et al. Immunoregulatory role of the gut microbiota in inflammatory depression. Nat Commun. (2024) 15:3003. doi: 10.1038/s41467-024-47273-w
246. Lai Y and Xiong P. Analysis of gut microbiota and depression and anxiety: mendelian randomization from three datasets. Gen Hosp Psychiatry. (2025) 94:206–18. doi: 10.1016/j.genhosppsych.2025.03.012
247. Pan M, Qian C, Huo S, Wu Y, Zhao X, Ying Y, et al. Gut-derived lactic acid enhances tryptophan to 5-hydroxytryptamine in regulation of anxiety via akkermansia muciniphila. Gut Microbes. (2025) 17:2447834. doi: 10.1080/19490976.2024.2447834
248. Zhang X, Hou Y, Li Y, Wei W, Cai X, Shao H, et al. Taxonomic and metabolic signatures of gut microbiota for assessing the severity of depression and anxiety in major depressive disorder patients. Neuroscience. (2022) 496:179–89. doi: 10.1016/j.neuroscience.2022.06.024
249. Sublette ME, Cheung S, Lieberman E, Hu S, Mann JJ, Uhlemann AC, et al. Bipolar disorder and the gut microbiome: A systematic review. Bipolar Disord. (2021) 23:544–64. doi: 10.1111/bdi.13049
250. Hasegawa S, Goto S, Tsuji H, Okuno T, Asahara T, Nomoto K, et al. Intestinal dysbiosis and lowered serum lipopolysaccharide-binding protein in parkinson’s disease. PLoS One. (2015) 10:e0142164. doi: 10.1371/journal.pone.0142164
251. Unger MM, Spiegel J, Dillmann KU, Grundmann D, Philippeit H, Bürmann J, et al. Short chain fatty acids and gut microbiota differ between patients with parkinson’s disease and age-matched controls. Parkinsonism Relat Disord. (2016) 32:66–72. doi: 10.1016/j.parkreldis.2016.08.019
252. Liu J, Lv X, Ye T, Zhao M, Chen Z, Zhang Y, et al. Microbiota-microglia crosstalk between blautia producta and neuroinflammation of parkinson’s disease: A bench-to-bedside translational approach. Brain Behav Immun. (2024) 117:270–82. doi: 10.1016/j.bbi.2024.01.010
253. Chen C, Liao J, Xia Y, Liu X, Jones R, Haran J, et al. Gut microbiota regulate alzheimer’s disease pathologies and cognitive disorders via pufa-associated neuroinflammation. Gut. (2022) 71:2233–52. doi: 10.1136/gutjnl-2021-326269
254. Kesika P, Suganthy N, Sivamaruthi BS, and Chaiyasut C. Role of gut-brain axis, gut microbial composition, and probiotic intervention in alzheimer’s disease. Life Sci. (2021) 264:118627. doi: 10.1016/j.lfs.2020.118627
255. Kang DW, Park JG, Ilhan ZE, Wallstrom G, Labaer J, Adams JB, et al. Reduced incidence of prevotella and other fermenters in intestinal microflora of autistic children. PLoS One. (2013) 8:e68322. doi: 10.1371/journal.pone.0068322
256. Srikantha P and Mohajeri MH. The possible role of the microbiota-gut-brain-axis in autism spectrum disorder. Int J Mol Sci. (2019) 20:2115. doi: 10.3390/ijms20092115
257. Socała K, Doboszewska U, Szopa A, Serefko A, Włodarczyk M, Zielińska A, et al. The role of microbiota-gut-brain axis in neuropsychiatric and neurological disorders. Pharmacol Res. (2021) 172:105840. doi: 10.1016/j.phrs.2021.105840
258. Tomova A, Husarova V, Lakatosova S, Bakos J, Vlkova B, Babinska K, et al. Gastrointestinal microbiota in children with autism in Slovakia. Physiol Behav. (2015) 138:179–87. doi: 10.1016/j.physbeh.2014.10.033
259. Wang L, Christophersen CT, Sorich MJ, Gerber JP, Angley MT, and Conlon MA. Increased abundance of sutterella spp. And ruminococcus torques in feces of children with autism spectrum disorder. Mol Autism. (2013) 4:42. doi: 10.1186/2040-2392-4-42
260. Parracho HM, Bingham MO, Gibson GR, and McCartney AL. Differences between the gut microflora of children with autistic spectrum disorders and that of healthy children. J Med Microbiol. (2005) 54:987–91. doi: 10.1099/jmm.0.46101-0
261. Xie G, Zhou Q, Qiu CZ, Dai WK, Wang HP, Li YH, et al. Ketogenic diet poses a significant effect on imbalanced gut microbiota in infants with refractory epilepsy. World J Gastroenterol. (2017) 23:6164–71. doi: 10.3748/wjg.v23.i33.6164
262. Gong X, Liu X, Chen C, Lin J, Li A, Guo K, et al. Alteration of gut microbiota in patients with epilepsy and the potential index as a biomarker. Front Microbiol. (2020) 11:517797. doi: 10.3389/fmicb.2020.517797
263. Lee K, Kim N, Shim JO, and Kim GH. Gut bacterial dysbiosis in children with intractable epilepsy. J Clin Med. (2020) 10:5. doi: 10.3390/jcm10010005
264. Lee H, Lee S, Lee DH, and Kim DW. A comparison of the gut microbiota among adult patients with drug-responsive and drug-resistant epilepsy: an exploratory study. Epilepsy Res. (2021) 172:106601. doi: 10.1016/j.eplepsyres.2021.106601
265. Peng A, Qiu X, Lai W, Li W, Zhang L, Zhu X, et al. Altered composition of the gut microbiome in patients with drug-resistant epilepsy. Epilepsy Res. (2018) 147:102–7. doi: 10.1016/j.eplepsyres.2018.09.013
266. Şafak B, Altunan B, Topçu B, and Eren Topkaya A. The gut microbiome in epilepsy. Microb Pathog. (2020) 139:103853. doi: 10.1016/j.micpath.2019.103853
267. Rao J, Qiu P, Zhang Y, and Wang X. Gut microbiota trigger host liver immune responses that affect drug-metabolising enzymes. Front Immunol. (2024) 15:1511229. doi: 10.3389/fimmu.2024.1511229
268. Wang X, Ye C, Xun T, Mo L, Tong Y, Ni W, et al. Bacteroides fragilis polysaccharide a ameliorates abnormal voriconazole metabolism accompanied with the inhibition of tlr4/nf-Κb pathway. Front Pharmacol. (2021) 12:663325. doi: 10.3389/fphar.2021.663325
269. Deng X, Li Y, Jiang L, Xie X, and Wang X. 1-methylnicotinamide modulates il-10 secretion and voriconazole metabolism. Front Immunol. (2025) 16:1529660. doi: 10.3389/fimmu.2025.1529660
270. Wang YL, Chen L, Zhong XL, Liu QS, Li WQ, Cheng Y, et al. Antidepressant effects of ershiwei roudoukou pills and its active ingredient macelignan: multiple mechanisms involving oxidative stress, neuroinflammation and synaptic plasticity. Transl Psychiatry. (2025) 15:163. doi: 10.1038/s41398-025-03378-4
271. Park SY, Kim YH, Kim Y, and Lee SJ. Aromatic-turmerone’s anti-inflammatory effects in microglial cells are mediated by protein kinase a and heme oxygenase-1 signaling. Neurochem Int. (2012) 61:767–77. doi: 10.1016/j.neuint.2012.06.020
272. Willner P, Scheel-Krüger J, and Belzung C. The neurobiology of depression and antidepressant action. Neurosci Biobehav Rev. (2013) 37:2331–71. doi: 10.1016/j.neubiorev.2012.12.007
273. Golofast B and Vales K. The connection between microbiome and schizophrenia. Neurosci Biobehav Rev. (2020) 108:712–31. doi: 10.1016/j.neubiorev.2019.12.011
274. Theoharides TC, Kavalioti M, and Tsilioni I. Mast cells, stress, fear and autism spectrum disorder. Int J Mol Sci. (2019) 20:3611. doi: 10.3390/ijms20153611
275. Kotwas I, McGonigal A, Bastien-Toniazzo M, Bartolomei F, and Micoulaud-Franchi JA. Stress regulation in drug-resistant epilepsy. Epilepsy Behav. (2017) 71:39–50. doi: 10.1016/j.yebeh.2017.01.025
276. Goadsby PJ, Holland PR, Martins-Oliveira M, Hoffmann J, Schankin C, and Akerman S. Pathophysiology of migraine: A disorder of sensory processing. Physiol Rev. (2017) 97:553–622. doi: 10.1152/physrev.00034.2015
277. Paudel YN, Shaikh MF, Shah S, Kumari Y, and Othman I. Role of inflammation in epilepsy and neurobehavioral comorbidities: implication for therapy. Eur J Pharmacol. (2018) 837:145–55. doi: 10.1016/j.ejphar.2018.08.020
278. Amoozegar F. Depression comorbidity in migraine. Int Rev Psychiatry. (2017) 29:504–15. doi: 10.1080/09540261.2017.1326882
279. Vasileva SS, Yang Y, Baker A, Siskind D, Gratten J, and Eyles D. Associations of the gut microbiome with treatment resistance in schizophrenia. JAMA Psychiatry. (2024) 81:292–302. doi: 10.1001/jamapsychiatry.2023.5371
280. Starr AE, Deeke SA, Ning Z, de Nanassy J, Singleton R, Benchimol EI, et al. Associations between cellular energy and pediatric inflammatory bowel disease patient response to treatment. J Proteome Res. (2021) 20:4393–404. doi: 10.1021/acs.jproteome.1c00341
281. Mörkl S, Butler MI, Holl A, Cryan JF, and Dinan TG. Probiotics and the microbiota-gut-brain axis: focus on psychiatry. Curr Nutr Rep. (2020) 9:171–82. doi: 10.1007/s13668-020-00313-5
Keywords: schizophrenia, microbiota-gut-brain axis, gut microbiota metabolites, immune dysregulation, neuroinflammation, kynurenine pathway, complement system, toll-like receptors
Citation: Su B-W, Li Y, Yang L-Y, Yang H-X, Wang W-H, Ren H-W, Bao Y-N, Lao J-Y and Luan Z-L (2025) The role of the microbiota-gut-brain axis in schizophrenia: an immunological perspective. Front. Immunol. 16:1711756. doi: 10.3389/fimmu.2025.1711756
Received: 23 September 2025; Accepted: 31 October 2025;
Published: 19 November 2025.
Edited by:
Jean-louis Mege, Aix-Marseille Université, FranceReviewed by:
Nadja Freund, Ruhr University Bochum, GermanyNamrata Anand, University of Chicago Medical Center, United States
Copyright © 2025 Su, Li, Yang, Yang, Wang, Ren, Bao, Lao and Luan. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
*Correspondence: Zhi-Lin Luan, bHVhbnpsQGRtdS5lZHUuY24=
†These authors have contributed equally to this work
Le-Ying Yang3