The Potential Role of Toll-like Receptors in Schizophrenia: History
Please note this is an old version of this entry, which may differ significantly from the current revision.
Contributor: , ,

Toll-like receptors (TLRs) are a family of pattern recognition receptors (PRRs) ubiquitously expressed in the human body. They protect the brain and central nervous system from self and foreign antigens/pathogens. The immune response elicited by these receptors culminates in the release of cytokines, chemokines, and interferons causing an inflammatory response, which can be both beneficial and harmful to neurodevelopment. Such changes due to TLRs are shown to be associated with alterations in cognitive functions in various neurodegenerative and psychiatric disorders, and schizophrenia is one such disorder where multiple genetic and environmental factors contribute to alterations associated with TLRs.

  • inflammation
  • Toll-like receptor
  • schizophrenia
  • cognition

1. Introduction

Schizophrenia is a debilitating psychiatric disorder affecting 0.45% of adults worldwide, according to the World Health Organisation, [1]. According to 2019 data, the prevalence of schizophrenia ranges from 0.2 to 0.5% across nations worldwide [2]. Emerging evidence suggests the role of the immune system and its various components in the risk of schizophrenia, including cytokines, C-reactive protein, chemokines, antibodies, etc. [3]. Multiple studies show altered protein and mRNA expression of Toll-like receptors (TLRs) in schizophrenia compared to healthy adults [4][5][6]. Human postmortem studies carried out on individuals diagnosed with schizophrenia, brain samples showed lower tlr4 mRNA and protein levels, and downregulation of il6, il10, and tnfa mRNA in the prefrontal cortex (PFC) region [7][8], and elevated TLR4 protein levels in the cerebellum region [7]. On the contrary, high tlr4 and myd88 mRNA expression was observed in the PFC region of the human brain [9]. On the flip side, studies on peripheral blood show downregulation of tlr3 and 5 mRNA levels and upregulation of il6 and il10 mRNA levels in patients with schizophrenia [10], which is in contrast to evidence from brain samples. Studies of acute and first-episode psychosis (FEP) patient groups reported higher expression (measured by mean fluorescence intensity (MFI)) of TLR3, 4, and 5, but not TLR2, in monocytes [11][12]. Although Keri et al. observed higher TLR4 MFI in schizophrenia post-TLR4 stimulation, the MFI expression of IL-6, TNF-α, and IL-1β was lower than controls [12]. Similar results were also observed by Muller et al. with IL-1β. This was interpreted to represent an increase in TLR expression to compensate for the functional deficit [11]. In contrast, whole blood stimulation studies in schizophrenia showed elevated concentrations of IL-6, TNF-α, and IL-1β post-TLR2 stimulation, and increased IL-1β alone post-TLR4 and 8 stimulation [13]. Individual blood cell studies in FEP patients showed lower TLR protein expression in monocytes, B cells, and T cells [6]. Overall, researchers can infer that dysregulation of TLR expression exists in patients with schizophrenia and varies at various stages of disease progression (Table 1), but researchers do not have sufficient evidence to implicate their extent of involvement in the immunopathology of schizophrenia.

Characteristics of Human studies illustrating TLRs in various models and cognition. PFC—prefrontal cortex, CB—cerebellum, PBMC—peripheral blood mononuclear cells, LPS—lipopolysaccharide, TLR—Toll-like receptor, RBANS—Repeatable Battery for the Assessment of Neuropsychological Status, MCCB—Measurement and Treatment Research to Improve Cognition in Schizophrenia (MATRICS™) Consensus Cognitive Battery, MyD88—myeloid differentiation primary response 88, NFkB—nuclear factor kappa B, IkBa—, poly (IC)—polyinosinic:polycytidylic acid, LPS—lipopolysaccharide, DSM-IV—Diagnostic and Statistical Manual of Mental Disorders-IV, SCID-CV—Structured Clinical Interview for DSM-Clinical version, ICD-10—International Classification of Diseases-10.

2. Maternal Immune Activation (MIA) and Early Life Exposure

Increased risk for psychosis-like symptoms following MIA (inflammatory stimuli due to TLR activation), conventionally associated with increased production of cytokines and chemokines, in offspring has been theorised for a long time [20][21]. Infections such as cytomegalovirus, influenza, herpes simplex virus, and coronavirus lead to upregulation of TLR mRNA (2–4 and 7–9). This is also accompanied by an increase in interferon and cytokine production causing complications in pregnancy, miscarriage, or foetal abnormalities [22][23]. These elevated cytokines and chemokines can migrate from the placenta into the foetus, and such exposure could cause changes to the CNS, leading to abnormal neurodevelopment [24]. Moreover, conditions such as diabetes mellitus, obesity, and systemic lupus erythematosus (SLE) in pregnancy were linked to high expression of TLRs, cytokines, and chemokines in peripheral blood, and similar levels were reflected in the placenta [25][26][27]. This suggests that conditions other than viral/bacterial stimuli could lead to an elevation in proinflammatory cytokines that could cross the placenta leading to prenatal immune activation increasing the risk of schizophrenia.
One of the earliest epidemiological studies linking infection with schizophrenia was in 1989, which was based on the data of subjects born during the epidemic period (1918 and 1957 influenza epidemic) reporting schizophrenia. A significant risk (1.4 risk ratio) of schizophrenia was observed in the subjects whose mothers were exposed to infection during 5–7 months of pregnancy compared to other months of pregnancy in the Scottish population. The same was not observed if all United Kingdom subjects were considered [28]. Similarly, another study conducted on patients diagnosed with schizophrenia showed that the risk of schizophrenia greatly increased in offspring with elevated maternal antibodies against type 2 HSV infection during pregnancy [16]. Despite the small sample size (N = 27/50 case/controls), it was reported that elevated maternal serum TNF-α during pregnancy had an association with the risk (8.5 odds ratio (OR)) of schizophrenia later in their offspring [29]. Animal models show that neural progenitor cells (NPCs) (NPCs undergo multiple changes to develop into neurons) expressed TLR3 and IRF3, and these cells responded to both in vitro and in vivo (dams) stimulation with poly I:C. Although conflicting results were observed from both models, in vivo results showed a significant decrease in cell proliferation post-treatment [30]. Similarly, irregular differentiation (high cholinergic neuronal differentiation and undifferentiated progenitors) and neurodevelopment were observed when forebrain from rat embryos were cultured with microglial conditioned medium stimulated by LPS [31]. Such evidence points to the detrimental neurodevelopmental effects on the foetus during pregnancy postexposure to infections or stimuli (TLR ligands), which could later affect the offspring.
For a long time, TLRs have been considered a target for vaccine adjuvants to increase the efficacy of the vaccine, where they act by low-level stimulation of the target TLRs and activate the interferon pathway [32]. It was observed in animals that vaccination for influenza promoted better neural development (indicated by increased cell proliferation, neural differentiation, and expression of brain-derived neurotrophic factor (BDNF) in pups) and provided protective function in the mother and offspring through TLR4 stimulation [33]. The aforementioned evidence indicates that the extent of activation of TLRs is determined by the severity of the infection in mothers, further affecting the neurodevelopment in the offspring while appropriate care such as vaccination, where the virus cannot replicate, can promote protective effects through TLRs on the offspring.
Animal models show a more detailed picture of MIA (Table 2). Intraperitoneal administration of Poly I:C and LPS in dams led to upregulation of TLR3 and 4 mRNA expression in the hippocampus tissue of the offspring. Moreover, this upregulation was associated with sociocognitive deficits and behaviour abnormalities in them [34]. In another study, post-LPS treatment in dams, mice showed upregulation of TLR2 and 4 mRNA in addition to COX-2 (cyclooxygenase), iNOS (inducible nitric oxide synthase), and CCL2 (chemokine C-C motif ligand) in the foetal stage, and later in adolescence. This was suggested to affect the microglial and astrocyte development in the amygdala region of the brain [35].
Table 2. Characteristics of animal studies.
Study Animal
(Type of Cells/Culture)
Model TLR Analysed Analysis Results
Talukdar et al., 2021 [34] Rats MIA (Poly IC and LPS) TLR2, 3, and 4 mRNA Elevated TLR genes were highly associated with a decrease in sociability and normal behaviour in the offspring.
Lee et al., 2016 [36] Mice MIA (Zymosan) TLR2 TLR2 stimulation showed decreased cognitive and behavioural changes only in female offspring.
O’Loughlin et al., 2017 [35] Mice MIA (LPS) TLR2 and 4 mRNA Elevated TLR genes in pre/postnatal stages after stimulation.
These were associated with morphological changes in microglia, suggesting activation.
Connolly et al., 2021 [37] Mice TLR 4 antagonism TLR4 TLR4 antagonism showed beneficial cognitive effects
(enhanced spatial learning and memory) only in young female mice.
Drouin-Ouellet
et al., 2012 [38]
Mice MyD88 KO TLR The absence of MyD88 in mice led to spatial learning and working memory deficits, and reduced motor activity.
Ibi et al., 2009 [39] Mice Poly IC stimulation TLR3 TLR3 stimulation showed decreased social interaction, cognitive, and behavioural changes.
Liu et al., 2013 [40] Mice TLR7 KO TLR7 TLR 7 activation reduced dendrite growth mediated by IL-6 in mice.
TLR 7 KO reduced dendrite growth and reduced behavioural activity.
Madar et al., 2015 Mice TLR2 KO and TLR2+ TLR2 TLR2 KO decreased mice activity and impaired spatial learning. Neonatal TLR2/1 activation led to impaired spatial learning in adulthood.
Park et al., 2015 Mice TLR2 KO TLR2 TLR2 KO mice showed cognitive and social recognition impairment.
Melnik et al., 2012 Mice In utero (poly IC stimulation) TLR3 mRNA TLR3 stimulation leads to decreased proliferation after Poly IC treatment unlike, in vitro studies.
Mice
(DG NPC)
Poly IC treatment TLR3 mRNA TLR3 stimulation leads to increased proliferation after 24 and 48 h of poly IC treatment.
Liu et al., 2013 [40] Mice
(cortical neurons)
  TLR 7 TLR7 activation produced cytokines and hindered dendrite growth. TLR7 KO promoted axon and dendrite growth.
Nakajima et al., 2014 [41] Mice
(astrocyte cell culture)
LPS stimulated TLR3 and 4 mRNA LPS stimulation showed significantly elevated TLR3 expression up to 24 h and dropped until 72 h.
No significant change was seen with TLR4 expression, but a trend of upregulation was observed from 3 to 72 h.
Zhou et al., 2020 [42] Mice
(BV2 microglial cells)
LPS treatment TLR4 protein TLR4 and NF-kB upregulation post-treatment.
Characteristics of animal studies showcasing the effect on TLRs in various models and cognition. DG-NPC—Dentate gyrus-neural progenitor cells, LPS—lipopolysaccharide; TLR—Toll-like receptor, MIA—maternal immune activation, KO—knockout, IL-6—interleukin-6.

3. Factors Influencing Toll-like Receptors in Schizophrenia

3.1. Diet and Gut Microbiome

There are many diseases and disorders that are associated with improper diet. Studies showed that prenatal malnutrition during famine throughout the world in various eras increased the risk of nervous system-related disorders and mental illnesses such as schizophrenia, antisocial behaviours, other psychiatric disorders, and congenital anomalies such as neural tube defects [51]. Although it is unclear if this was due to the direct result of malnutrition (diet change) or other underlying causes such as stress, it was observed in Dutch and Chinese populations who experienced the ‘Dutch hunger of 1944–1945’ and ‘Great leap forward of 1958–1962’, respectively, that the risk of acquiring mental illness, especially schizophrenia, was two-fold higher in the populations born during these great famines [51][52].
Similar to how dietary deficits can be dangerous to mental health, an imbalanced diet lifestyle can affect the same. The nutritional psychiatry division of Harvard reports that the type of food can affect the inflammatory status and oxidative stress in the body. It was observed that the traditional diet from Asian and Mediterranean areas proved to have a 25–30% lower risk of depression when compared to the western diet [53]. Moreover, studies show that saturated fatty acids (SFAs) are capable of activating microglia and inducing proinflammatory cytokines, which is facilitated by TLR2 and 4 [54]. The mRNA levels of il6, il1b, and tnfa were found to be elevated post-SFA treatment in mouse cell culture, and their upregulated expression was similar to that of LPS stimulation [55]. Furthermore, in in vitro studies (mouse and human), TLR2- and 4-dependent activations were observed with SFAs. TLR2, 3, 5, and 9 activations by their respective agonists were attenuated by ω-3- and ω-6-polyunsaturated fatty acids (PUFAs), indicating that some fatty acids activate TLRs, and some others inhibit their activity [54][56][57].
A high-fat diet is closely related to obesity [58], and it was observed that a high-fat diet elevated proinflammatory cytokine expression via the TLR pathway (NF-kB activation), causing an inflammatory state which correlated with weight gain [59][60]. This high-fat diet can also dysregulate the gut microbiome [61], due to bacterial metabolites migrating to the brain and crossing the BBB [62], and inducing inflammatory responses in the brain. This further affects neurodevelopment (discussed in detail elsewhere [63][64]). The “leaky gut” MIA model suggests that metabolites from maternal microbiome dysbiosis can act as inflammatory stimuli on the foetus [64] and, in turn, potentially increase the risk of psychiatric disorders such as schizophrenia.
Evidence also suggests that the presence of unwanted or harmful microbes such as Saccharomyces cerevisiae (induced gastrointestinal inflammation) is associated with schizophrenia in both FEP and chronic patients [64][65]. Furthermore, schizophrenia was found to be associated with gastrointestinal dysfunction symptoms. The prevalence rate of such symptoms and conditions such as irritable bowel syndrome is higher (~19%) in these patients over healthy individuals [66]. This suggests that the microbiome and diet might have a role in the immunopathology of schizophrenia.

3.2. Drugs

Antipsychotic drugs or neuroleptics have both intended (therapeutic) and unintended (adverse/side effects) effects. For instance, the antipsychotic drug clozapine’s main target is dopamine and serotonin receptors, and this interaction leads to the alleviation of positive and negative symptoms in many patients. In addition, it acts partially on muscarinic, adrenergic, and histamine receptors, which leads to side effects such as drowsiness and dizziness [67]. Similarly, there is a vast literature that implicates antipsychotic medication as having another function, which is to alter the cytokine network in the schizophrenia population [68][69][70][71]. It is not yet understood how this medication alters cytokine levels, but researchers can hypothesise two possibilities: Firstly, they could affect the cytokine levels directly by unknown means, or they alter the TLR expression/protein levels and regulators in their downstream signalling, further influencing the cytokine expression. It could possibly be the latter, as TLRs regulate cytokine expression. In congruence with this statement, a study showed that paliperidone [72] and clozapine [73] reduced the inflammatory response (LPS-induced) associated with TLR4 by partly inhibiting the calcium/calmodulin-dependent Akt (protein kinase B) in a rat microglial cell line. Inhibition of this enzyme prevents Akt-dependent IKKα phosphorylation, further preventing NF-kB nuclear translocation [73]. Moreover, the MIA model of mice using poly I:C reported that paliperidone prevented the upregulation of TLR3 protein in the frontal cortex region [74]. On the contrary, another antipsychotic treatment, olanzapine, resulted in a significant rapid elevation of inflammatory cytokines (IL-6, TNF-α, IL-1β) and TLR4 protein expression in rat PFC [75]. A study on unmedicated patients reported significantly higher expression (MFI) of TLR2 in monocytes post-antipsychotics treatment, whereas TLR4 and 5 MFI were significantly higher before medication, and no difference after [76]. These studies indicate that drugs alter TLR downstream signalling, which leads to cytokine imbalance. On the other hand, studies reported that antipsychotic-treated patients showed higher expression of tlr4 mRNA [9][77] and myd88 mRNA [9] in peripheral blood and postmortem brain tissue samples, respectively, over antipsychotic-free patients (schizophrenia). Interestingly, no significant differences were observed in the other downstream signalling protein mRNA levels such as nf-kb, ikbα, il-1β, and il-6 [9]. On the other hand, 3 months of antipsychotic treatment resulted in higher TLR6 protein expression, and a trend towards higher TLR8 and 9 in monocytes, T cells, and B cells of FEP patients [6]. Given the evidence, there is a disparity in the current literature regarding the influence and relationship between antipsychotic drugs and TLRs and cytokines which is not yet clearly understood, and studies are ongoing to find the same.
Studies have reported that recreational drugs such as alcohol consumption (binge) and frequent or continuous use of amphetamines and methamphetamines induces psychosis-like symptoms in nearly half the users [78][79]. It was found that methamphetamine elevated IL-1β and IL-18 in primary mouse astrocyte cultures, and this was mediated by TLR4, NF-kB, and caspase-11 pathways, causing neuroinflammation [80]. This is the most probable reason why most of these users are at significant risk of schizophrenia where some of them develop psychosis, which eventually transforms into schizophrenia [81].

3.3. Genetics

Genetic polymorphisms represent DNA variation in a specific population that may/may not lead to a change in protein expression. One of the commonly occurring polymorphisms is a single nucleotide polymorphism (SNP), which has been used as a genetic target to study multiple diseases [82]. SNPs within multiple immune-related gene networks have now been investigated in terms of their association with disease risk. Among these, it was found that polymorphisms within genes encoding tlrs and their pathways were associated with either increased or decreased protection against various diseases and infections. Moreover, altered expression of cytokines in response to infections was associated with tlr gene polymorphisms [83].
In schizophrenia, gene polymorphisms associated with pathways of inflammation, immune response, neurodevelopment, and cell death have been identified. Alterations of such pathways due to polymorphisms are significantly associated with schizophrenia risk, and the TLR pathway is suggested to be a modulator of these pathways [84]. Studies also show that TLR polymorphisms are associated with schizophrenia (Table 3), such as tlr2 polymorphism in the Tunisian population [85]. An insertion/deletion polymorphism of an SNP (rs111200466) located in the promoter region of the tlr2 gene showed to be protective in healthy females, as this polymorphism was not observed in the schizophrenia group [85]. Missense polymorphisms (rs5743708 and rs121917864) located in the third exon of tlr2 gene were considered to increase the risk and susceptibility to schizophrenia. The aforementioned three SNPs were previously associated with decreased activation and expression of TLR2 and cytokines [85]. In another study, two SNPs, rs3804099 and rs3804100 (of unknown function), located in the third exon of tlr2 gene, were reported to be significantly associated with poor concentration in the Korean schizophrenia population. Although these 2 SNPs were found to be associated with cognition, none were associated with either positive or negative symptoms [86]. Patients belonging to north Indian ethnicity showed a similar association with tlr2 SNP (rs3804099) [87]. This indicates the broad (multiethnic) implications of TLR2 polymorphism in schizophrenia. TLR4 polymorphism also showed an association with schizophrenia risk; four SNPs of TLR4 were investigated, of which three (rs11536889-3′UTR, rs1927911-intron, and rs1927914-5′UTR) showed an association with schizophrenia. One of these investigated SNPs was previously associated with multiple diseases and disorders [88]. Changes in the TLR downstream signalling proteins such as myd88 SNP (rs7744-3′UTR) were reported to be less prominent in patients with schizophrenia than in controls. Moreover, a model utilising 5 SNPs (3 TLR downstream signalling protein genes (myd88, irak1, and nfkb1) and 2 cytokines (il6 and il1b)) for a five-way analysis identifies different combinations of these five genes, predicting the risk of schizophrenia in a given population. This model showed that people having a specific combination of these five genes were at higher risk (OR—6.9) of developing schizophrenia [9]. This evidence indicates the immune–genetic predisposition in schizophrenia, and it can be understood that multiple polymorphisms of various genes associated with the TLR pathway collectively contribute towards the incidence of schizophrenia (Table 3).
Table 3. Characteristics of genetic studies.
Study Population Gene (SNPs) Location of SNP Region of SNP SNP Function Analysis Results
Aflouk et al., 2021 [85] Tunisian tlr2 (rs111200466) Chr4. −196 to −174 Insertion/Deletion Promoter region Decreased activation and
expression of TLR2 and cytokines
Protective action in females, as this
polymorphism was not observed in the schizophrenia group (females).
tlr2 (rs5743708) Chr4. G > A Missense variant Third exon (functional) Increases the risk and susceptibility to schizophrenia.
tlr2 (rs121917864) Chr4. C > T Missense variant
Kang et al., 2013 [86] Korean tlr2 (rs3804099) Chr4. T > C Synonymous Variant Third exon (functional) Unknown function C’ allele associated with
cognition (poor concentration).
tlr2 (rs3804100) Chr4. T > A/T > C Missense variant
Sharma et al., 2022 [87] Indian tlr2 (rs3804099) Chr4. T > C Synonymous Variant Third exon (functional) Unknown function Increases the risk of schizophrenia.
Mostafa et al., 2022 [88] Egyptian tlr4 (rs11536889) Chr9. G > A/G > C UTR Variant 3′UTR Interferes with the translation process and increase the expression of TLR4 A strong association between these
genes and schizophrenia.
tlr4 (rs1927911) Chr9. A > C/G/T Intron -
tlr4 (rs1927914) Chr9. G > A 5′UTR Regulation of gene expression and protein levels
García-Bueno et al., 2016 [9] Spanish myd88 (rs7744) Chr3. A > G/A > T Noncoding Transcript Variant 3′UTR Previously associated with autoimmune or inflammatory processes These two polymorphisms were found to be crucial in the schizophrenia predictive model for this population.
il6 (rs1800795) Chr7. C > G/C > T Promoter region Increases IL-6 levels
Characteristics of genetic studies illustrating polymorphisms in schizophrenia group in various populations. Chr—chromosome, A/G/T/C—nucleotide bases (adenine (A), cytosine (C), guanine (G), and thymine (T)), tlr—Toll-like receptor, il6—interleukin-6, myd88—myeloid differentiation primary response 88 protein, rsxxxxxx—reference SNP “ID”, SNP—single nucleotide polymorphism, rs—reference SNP, UTR—untranslated region.

This entry is adapted from the peer-reviewed paper 10.3390/biom13081188

References

  1. WHO. Schizophrenia. Available online: https://www.who.int/news-room/fact-sheets/detail/schizophrenia#:~:text=Schizophrenia%20affects%20approximately%2024%20million,%25)%20among%20adults%20(2) (accessed on 10 August 2022).
  2. Saloni, D.; Hannah, R.; Max, R. Mental Health. Available online: https://ourworldindata.org/mental-health#schizophrenia (accessed on 11 August 2022).
  3. Ermakov, E.A.; Melamud, M.M.; Buneva, V.N.; Ivanova, S.A. Immune System Abnormalities in Schizophrenia: An Integrative View and Translational Perspectives. Front. Psychiatry 2022, 13, 880568.
  4. Kozłowska, E.; Agier, J.; Wysokiński, A.; Łucka, A.; Sobierajska, K.; Brzezińska-Błaszczyk, E. The expression of toll-like receptors in peripheral blood mononuclear cells is altered in schizophrenia. Psychiatry Res. 2019, 272, 540–550.
  5. Chase, K.A.; Melbourne, J.K.; Rosen, C.; McCarthy-Jones, S.; Jones, N.; Feiner, B.M.; Sharma, R.P. Traumagenics: At the intersect of childhood trauma, immunity and psychosis. Psychiatry Res. 2019, 273, 369–377.
  6. Juncal-Ruiz, M.; Riesco-Davila, L.; Vazquez-Bourgon, J.; Ortiz-Garcia de la Foz, V.; Mayoral-Van Son, J.; Ayesa-Arriola, R.; Setien-Suero, E.; Leza, J.C.; Lopez-Hoyos, M.; Crespo-Facorro, B. Expression and functionality study of 9 toll-like receptors in 33 drug-naive non-affective first episode psychosis individuals: A 3-month study. Int. J. Mol. Sci. 2020, 21, 6106.
  7. MacDowell, K.S.; Pinacho, R.; Leza, J.C.; Costa, J.; Ramos, B.; García-Bueno, B. Differential regulation of the TLR4 signalling pathway in post-mortem prefrontal cortex and cerebellum in chronic schizophrenia: Relationship with SP transcription factors. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2017, 79, 481–492.
  8. López-González, I.; Pinacho, R.; Vila, È.; Escanilla, A.; Ferrer, I.; Ramos, B. Neuroinflammation in the dorsolateral prefrontal cortex in elderly chronic schizophrenia. Eur. Neuropsychopharmacol. 2019, 29, 384–396.
  9. García-Bueno, B.; Gassó, P.; MacDowell, K.S.; Callado, L.F.; Mas, S.; Bernardo, M.; Lafuente, A.; Meana, J.J.; Leza, J.C. Evidence of activation of the Toll-like receptor-4 proinflammatory pathway in patients with schizophrenia. J. Psychiatry Neurosci. 2016, 41, E46.
  10. Chang, S.-H.; Chiang, S.-Y.; Chiu, C.-C.; Tsai, C.-C.; Tsai, H.-H.; Huang, C.-Y.; Hsu, T.-C.; Tzang, B.-S. Expression of anti-cardiolipin antibodies and inflammatory associated factors in patients with schizophrenia. Psychiatry Res. 2011, 187, 341–346.
  11. Müller, N.; Wagner, J.K.; Krause, D.; Weidinger, E.; Wildenauer, A.; Obermeier, M.; Dehning, S.; Gruber, R.; Schwarz, M.J. Impaired monocyte activation in schizophrenia. Psychiatry Res. 2012, 198, 341–346.
  12. Kéri, S.; Szabó, C.; Kelemen, O. Uniting the neurodevelopmental and immunological hypotheses: Neuregulin 1 receptor ErbB and Toll-like receptor activation in first-episode schizophrenia. Sci. Rep. 2017, 7, 4147.
  13. McKernan, D.P.; Dennison, U.; Gaszner, G.; Cryan, J.F.; Dinan, T.G. Enhanced peripheral toll-like receptor responses in psychosis: Further evidence of a pro-inflammatory phenotype. Transl. Psychiatry 2011, 1, e36.
  14. Menninger, K.A. Influenza and schizophrenia: An analysis of post-influenzal “dementia precox”, as of 1918, and five years later. Am. J. Psychiatry 1926, 82, 469–529.
  15. Kępińska, A.P.; Iyegbe, C.O.; Vernon, A.C.; Yolken, R.; Murray, R.M.; Pollak, T.A. Schizophrenia and Influenza at the Centenary of the 1918-1919 Spanish Influenza Pandemic: Mechanisms of Psychosis Risk. Front. Psychiatry 2020, 11, 72.
  16. Buka, S.L.; Tsuang, M.T.; Torrey, E.F.; Klebanoff, M.A.; Bernstein, D.; Yolken, R.H. Maternal Infections and Subsequent Psychosis Among Offspring. Arch. Gen. Psychiatry 2001, 58, 1032–1037.
  17. Dickerson, F.; Jones-Brando, L.; Ford, G.; Genovese, G.; Stallings, C.; Origoni, A.; O’Dushlaine, C.; Katsafanas, E.; Sweeney, K.; Khushalani, S.; et al. Schizophrenia is Associated with an Aberrant Immune Response to Epstein–Barr Virus. Schizophr. Bull. 2018, 45, 1112–1119.
  18. Burgdorf, K.S.; Trabjerg, B.B.; Pedersen, M.G.; Nissen, J.; Banasik, K.; Pedersen, O.B.; Sørensen, E.; Nielsen, K.R.; Larsen, M.H.; Erikstrup, C.; et al. Large-scale study of Toxoplasma and Cytomegalovirus shows an association between infection and serious psychiatric disorders. Brain Behav. Immun. 2019, 79, 152–158.
  19. Choudhury, Z.; Lennox, B. Maternal Immune Activation and Schizophrenia-Evidence for an Immune Priming Disorder. Front. Psychiatry 2021, 12, 585742.
  20. Ashdown, H.; Dumont, Y.; Ng, M.; Poole, S.; Boksa, P.; Luheshi, G.N. The role of cytokines in mediating effects of prenatal infection on the fetus: Implications for schizophrenia. Mol. Psychiatry 2006, 11, 47–55.
  21. Meyer, U.; Feldon, J.; Yee, B.K. A Review of the Fetal Brain Cytokine Imbalance Hypothesis of Schizophrenia. Schizophr. Bull. 2008, 35, 959–972.
  22. Hamilton, S.T.; Scott, G.; Naing, Z.; Iwasenko, J.; Hall, B.; Graf, N.; Arbuckle, S.; Craig, M.E.; Rawlinson, W.D. Human Cytomegalovirus-Induces Cytokine Changes in the Placenta with Implications for Adverse Pregnancy Outcomes. PLoS ONE 2013, 7, e52899.
  23. Liu, X.-R.; Wei, X.-W.; Lin, Y.; Wang, Y.-H. Toll-Like Receptor-Dependent Antiviral Responses at the Maternal-Fetal Interface. Reprod. Dev. Med. 2020, 04, 251–256.
  24. Yockey, L.J.; Iwasaki, A. Interferons and Proinflammatory Cytokines in Pregnancy and Fetal Development. Immunity 2018, 49, 397–412.
  25. Kuzmicki, M.; Telejko, B.; Wawrusiewicz-Kurylonek, N.; Lipinska, D.; Pliszka, J.; Wilk, J.; Zielinska, A.; Skibicka, J.; Szamatowicz, J.; Kretowski, A. The expression of genes involved in NF-kB activation in peripheral blood mononuclear cells of patients with gestational diabetes. Eur. J. Endocrinol. 2013, 168, 419–427.
  26. Qu, X.; Yu, X.; Liu, J.; Wang, J. Pro-inflammatory cytokines are elevated in pregnant women with systemic lupus erythematosus in association with the activation of TLR4. Clin. Lab. 2016, 62, 535–544.
  27. Yang, X.; Li, M.; Haghiac, M.; Catalano, P.M.; O’Tierney-Ginn, P.; Hauguel-de Mouzon, S. Causal relationship between obesity-related traits and TLR4-driven responses at the maternal–fetal interface. Diabetologia 2016, 59, 2459–2466.
  28. Kendell, R.E.; Kemp, I.W. Maternal Influenza in the Etiology of Schizophrenia. Arch. Gen. Psychiatry 1989, 46, 878–882.
  29. Buka, S.L.; Tsuang, M.T.; Torrey, E.F.; Klebanoff, M.A.; Wagner, R.L.; Yolken, R.H. Maternal cytokine levels during pregnancy and adult psychosis. Brain Behav. Immun. 2001, 15, 411–420.
  30. Melnik, A.; Tauber, S.; Dumrese, C.; Ullrich, O.; Wolf, S.A. Murine adult neural progenitor cells alter their proliferative behavior and gene expression after the activation of toll-like-receptor 3. Eur. J. Microbiol. Immunol. 2012, 2, 239–248.
  31. Ni, L.; Acevedo, G.; Muralidharan, B.; Padala, N.; To, J.; Jonakait, G.M. Toll-like receptor ligands and CD154 stimulate microglia to produce a factor(s) that promotes excess cholinergic differentiation in the developing rat basal forebrain: Implications for neurodevelopmental disorders. Pediatr. Res. 2007, 61, 15–20.
  32. Hennessy, C.; McKernan, D.P. Anti-Viral Pattern Recognition Receptors as Therapeutic Targets. Cells 2021, 10, 2258.
  33. Xia, Y.; Qi, F.; Zou, J.; Yang, J.; Yao, Z. Influenza vaccination during early pregnancy contributes to neurogenesis and behavioral function in offspring. Brain Behav. Immun. 2014, 42, 212–221.
  34. Talukdar, P.M.; Abdul, F.; Maes, M.; Berk, M.; Venkatasubramanian, G.; Kutty, B.M.; Debnath, M. A proof-of-concept study of maternal immune activation mediated induction of Toll-like receptor (TLR) and inflammasome pathways leading to neuroprogressive changes and schizophrenia-like behaviours in offspring. Eur. Neuropsychopharmacol. 2021, 52, 48–61.
  35. O’Loughlin, E.; Pakan, J.M.; Yilmazer-Hanke, D.; McDermott, K.W. Acute in utero exposure to lipopolysaccharide induces inflammation in the pre-and postnatal brain and alters the glial cytoarchitecture in the developing amygdala. J. Neuroinflamm. 2017, 14, 212.
  36. Lee, Y.; Jeon, S.J.; Choi, J.W.; Ryu, J.H. Activation of maternal toll-like receptor-2 causes social deficits and memory impairment in female offspring. NeuroReport 2016, 27, 224–229.
  37. Connolly, M.G.; Potter, O.V.; Sexton, A.R.; Kohman, R.A. Effects of Toll-like receptor 4 inhibition on spatial memory and cell proliferation in male and female adult and aged mice. Brain Behav. Immun. 2021, 97, 383–393.
  38. Drouin-Ouellet, J.; LeBel, M.; Filali, M.; Cicchetti, F. MyD88 deficiency results in both cognitive and motor impairments in mice. Brain Behav. Immun. 2012, 26, 880–885.
  39. Ibi, D.; Nagai, T.; Kitahara, Y.; Mizoguchi, H.; Koike, H.; Shiraki, A.; Takuma, K.; Kamei, H.; Noda, Y.; Nitta, A.; et al. Neonatal polyI:C treatment in mice results in schizophrenia-like behavioral and neurochemical abnormalities in adulthood. Neurosci. Res. 2009, 64, 297–305.
  40. Liu, H.-Y.; Hong, Y.-F.; Huang, C.-M.; Chen, C.-Y.; Huang, T.-N.; Hsueh, Y.-P. TLR7 negatively regulates dendrite outgrowth through the Myd88–c-Fos–IL-6 pathway. J. Neurosci. 2013, 33, 11479–11493.
  41. Nakajima, A.; Ibi, D.; Nagai, T.; Yamada, S.; Nabeshima, T.; Yamada, K. Induction of interferon-induced transmembrane protein 3 gene expression by lipopolysaccharide in astrocytes. Eur. J. Pharmacol. 2014, 745, 166–175.
  42. Zhou, X.Y.; Liu, J.; Xu, Z.P.; Fu, Q.; Wang, P.Q.; Wang, J.H.; Zhang, H. Dexmedetomidine ameliorates postoperative cognitive dysfunction by inhibiting Toll-like receptor 4 signaling in aged mice. Kaohsiung J. Med. Sci. 2020, 36, 721–731.
  43. Manly, J.T.; Kim, J.E.; Rogosch, F.A.; Cicchetti, D. Dimensions of child maltreatment and children’s adjustment: Contributions of developmental timing and subtype. Dev. Psychopathol. 2001, 13, 759–782.
  44. Jurewicz, J.; Polanska, K.; Hanke, W. Chemical exposure early in life and the neurodevelopment of children–an overview of current epidemiological evidence. Ann. Agric. Environ. Med. 2013, 20, 3.
  45. Khandaker, G.M.; Zimbron, J.; Dalman, C.; Lewis, G.; Jones, P.B. Childhood infection and adult schizophrenia: A meta-analysis of population-based studies. Schizophr. Res. 2012, 139, 161–168.
  46. Baghel, M.S.; Singh, B.; Dhuriya, Y.K.; Shukla, R.K.; Patro, N.; Khanna, V.K.; Patro, I.K.; Thakur, M.K. Postnatal exposure to poly (I:C) impairs learning and memory through changes in synaptic plasticity gene expression in developing rat brain. Neurobiol. Learn. Mem. 2018, 155, 379–389.
  47. Singh, B.; Dhuriya, Y.K.; Patro, N.; Thakur, M.K.; Khanna, V.K.; Patro, I.K. Early life exposure to poly I: C impairs striatal DA-D2 receptor binding, myelination and associated behavioural abilities in rats. J. Chem. Neuroanat. 2021, 118, 102035.
  48. King, S.; Holleran, L.; Mothersill, D.; Patlola, S.; Rokita, K.; McManus, R.; Kenyon, M.; McDonald, C.; Hallahan, B.; Corvin, A.; et al. Early life Adversity, functional connectivity and cognitive performance in Schizophrenia: The mediating role of IL-6. Brain Behav. Immun. 2021, 98, 388–396.
  49. Flouri, E.; Francesconi, M.; Papachristou, E.; Midouhas, E.; Lewis, G. Stressful life events, inflammation and emotional and behavioural problems in children: A population-based study. Brain Behav. Immun. 2019, 80, 66–72.
  50. Song, R.; Gao, Y.; Dozmorov, I.; Malladi, V.; Saha, I.; McDaniel, M.M.; Parameswaran, S.; Liang, C.; Arana, C.; Zhang, B.; et al. IRF1 governs the differential interferon-stimulated gene responses in human monocytes and macrophages by regulating chromatin accessibility. Cell Rep. 2021, 34, 108891.
  51. Lumey, L.H.; Stein, A.D.; Susser, E. Prenatal famine and adult health. Annu. Rev. Public. Health 2011, 32, 237–262.
  52. Huang, C.; Phillips, M.R.; Zhang, Y.; Zhang, J.; Shi, Q.; Song, Z.; Ding, Z.; Pang, S.; Martorell, R. Malnutrition in early life and adult mental health: Evidence from a natural experiment. Soc. Sci. Med. 2013, 97, 259–266.
  53. Selhub, E. Nutritional psychiatry: Your brain on food. Harv. Health Blog 2020, 16, 11.
  54. Lee, J.Y.; Zhao, L.; Youn, H.S.; Weatherill, A.R.; Tapping, R.; Feng, L.; Lee, W.H.; Fitzgerald, K.A.; Hwang, D.H. Saturated fatty acid activates but polyunsaturated fatty acid inhibits Toll-like receptor 2 dimerized with Toll-like receptor 6 or 1. J. Biol. Chem. 2004, 279, 16971–16979.
  55. Wang, Z.; Liu, D.; Wang, F.; Liu, S.; Zhao, S.; Ling, E.-A.; Hao, A. Saturated fatty acids activate microglia via Toll-like receptor 4/NF-κB signalling. Br. J. Nutr. 2012, 107, 229–241.
  56. Lee, J.Y.; Plakidas, A.; Lee, W.H.; Heikkinen, A.; Chanmugam, P.; Bray, G.; Hwang, D.H. Differential modulation of Toll-like receptors by fatty acids: Preferential inhibition by n-3 polyunsaturated fatty acids. J. Lipid Res. 2003, 44, 479–486.
  57. Fessler, M.B.; Rudel, L.L.; Brown, J.M. Toll-like receptor signaling links dietary fatty acids to the metabolic syndrome. Curr. Opin. Lipidol. 2009, 20, 379–385.
  58. Wang, L.; Wang, H.; Zhang, B.; Popkin, B.M.; Du, S. Elevated Fat Intake Increases Body Weight and the Risk of Overweight and Obesity among Chinese Adults: 1991–2015 Trends. Nutrients 2020, 12, 3272.
  59. Ding, S.; Chi, M.M.; Scull, B.P.; Rigby, R.; Schwerbrock, N.M.J.; Magness, S.; Jobin, C.; Lund, P.K. High-fat diet: Bacteria interactions promote intestinal inflammation which precedes and correlates with obesity and insulin resistance in mouse. PLoS ONE 2010, 5, e12191.
  60. Jialal, I.; Kaur, H.; Devaraj, S. Toll-like Receptor Status in Obesity and Metabolic Syndrome: A Translational Perspective. J. Clin. Endocrinol. Metab. 2014, 99, 39–48.
  61. Marrone, M.C.; Coccurello, R. Dietary Fatty Acids and Microbiota-Brain Communication in Neuropsychiatric Diseases. Biomolecules 2019, 10, 12.
  62. Arentsen, T.; Qian, Y.; Gkotzis, S.; Femenia, T.; Wang, T.; Udekwu, K.; Forssberg, H.; Diaz Heijtz, R. The bacterial peptidoglycan-sensing molecule Pglyrp2 modulates brain development and behavior. Mol. Psychiatry 2017, 22, 257–266.
  63. Severance, E.G.; Yolken, R.H.; Eaton, W.W. Autoimmune diseases, gastrointestinal disorders and the microbiome in schizophrenia: More than a gut feeling. Schizophr. Res. 2016, 176, 23–35.
  64. R Caso, J.; Balanza-Martinez, V.; Palomo, T.; Garcia-Bueno, B. The microbiota and gut-brain axis: Contributions to the immunopathogenesis of schizophrenia. Curr. Pharm. Des. 2016, 22, 6122–6133.
  65. Severance, E.G.; Alaedini, A.; Yang, S.; Halling, M.; Gressitt, K.L.; Stallings, C.R.; Origoni, A.E.; Vaughan, C.; Khushalani, S.; Leweke, F.M. Gastrointestinal inflammation and associated immune activation in schizophrenia. Schizophr. Res. 2012, 138, 48–53.
  66. Fadgyas-Stanculete, M.; Buga, A.-M.; Popa-Wagner, A.; Dumitrascu, D.L. The relationship between irritable bowel syndrome and psychiatric disorders: From molecular changes to clinical manifestations. J. Mol. Psychiatry 2014, 2, 4.
  67. Wishart, D.S.; Feunang, Y.D.; Guo, A.C.; Lo, E.J.; Marcu, A.; Grant, J.R.; Sajed, T.; Johnson, D.; Li, C.; Sayeeda, Z.; et al. DrugBank 5.0: A major update to the DrugBank database for 2018. Nucleic Acids Res. 2018, 46, D1074–D1082.
  68. Miller, B.J.; Buckley, P.; Seabolt, W.; Mellor, A.; Kirkpatrick, B. Meta-analysis of cytokine alterations in schizophrenia: Clinical status and antipsychotic effects. Biol. Psychiatry 2011, 70, 663–671.
  69. Tourjman, V.; Kouassi, E.; Koue, M.-E.; Rocchetti, M.; Fortin-Fournier, S.; Fusar-Poli, P.; Potvin, S. Antipsychotics’ effects on blood levels of cytokines in schizophrenia: A meta-analysis. Schizophr. Res. 2013, 151, 43–47.
  70. Marcinowicz, P.; Więdłocha, M.; Zborowska, N.; Dębowska, W.; Podwalski, P.; Misiak, B.; Tyburski, E.; Szulc, A. A meta-analysis of the influence of antipsychotics on cytokines levels in first episode psychosis. J. Clin. Med. 2021, 10, 2488.
  71. Patlola, S.R.; Donohoe, G.; McKernan, D.P. Anti-inflammatory effects of 2nd generation antipsychotics in patients with schizophrenia: A systematic review and meta-analysis. J. Psychiatr. Res. 2023, 160, 126–136.
  72. MacDowell, K.; Caso, J.; Martin-Hernandez, D.; Madrigal, J.; Leza, J.; Garcia-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. 2015, 18.
  73. Jeon, S.; Kim, S.H.; Shin, S.Y.; Lee, Y.H. Clozapine reduces Toll-like receptor 4/NF-κB-mediated inflammatory responses through inhibition of calcium/calmodulin-dependent Akt activation in microglia. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2018, 81, 477–487.
  74. MacDowell, K.S.; Munarriz-Cuezva, E.; Caso, J.R.; Madrigal, J.L.; Zabala, A.; Meana, J.J.; García-Bueno, B.; Leza, J.C. Paliperidone reverts Toll-like receptor 3 signaling pathway activation and cognitive deficits in a maternal immune activation mouse model of schizophrenia. Neuropharmacology 2016, 116, 196–207.
  75. Li, W.-T.; Huang, X.-F.; Deng, C.; Zhang, B.-H.; Qian, K.; He, M.; Sun, T.-L. Olanzapine Induces inflammation and immune response via activating ER stress in the rat prefrontal cortex. Curr. Med. Sci. 2021, 41, 788–802.
  76. Kéri, S.; Szabó, C.; Kelemen, O. Antipsychotics influence Toll-like receptor (TLR) expression and its relationship with cognitive functions in schizophrenia. Brain Behav. Immun. 2017, 62, 256–264.
  77. Balaji, R.; Subbanna, M.; Shivakumar, V.; Abdul, F.; Venkatasubramanian, G.; 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.
  78. Bramness, J.G.; Gundersen, Ø.H.; Guterstam, J.; Rognli, E.B.; Konstenius, M.; Løberg, E.-M.; Medhus, S.; Tanum, L.; Franck, J. Amphetamine-induced psychosis—A separate diagnostic entity or primary psychosis triggered in the vulnerable? BMC Psychiatry 2012, 12, 221.
  79. Yang, Z.; Xiong, G.L. Alcohol-Related Psychosis. Available online: https://emedicine.medscape.com/article/289848-overview (accessed on 11 August 2022).
  80. Du, S.-H.; Qiao, D.-F.; Chen, C.-X.; Chen, S.; Liu, C.; Lin, Z.; Wang, H.; Xie, W.-B. Toll-Like Receptor 4 Mediates Methamphetamine-Induced Neuroinflammation through Caspase-11 Signaling Pathway in Astrocytes. Front. Mol. Neurosci. 2017, 10, 409.
  81. Medhus, S.; Rognli, E.B.; Gossop, M.; Holm, B.; Mørland, J.; Bramness, J.G. Amphetamine-induced psychosis: Transition to schizophrenia and mortality in a small prospective sample. Am. J. Addict. 2015, 24, 586–589.
  82. Sukhumsirichart, W. Polymorphisms. In Genetic Diversity and Disease Susceptibility; IntechOpen: London, UK, 2018.
  83. Medvedev, A.E. Toll-like receptor polymorphisms, inflammatory and infectious diseases, allergies, and cancer. J. Interferon Cytokine Res. 2013, 33, 467–484.
  84. Crisafulli, C.; Drago, A.; Calabrò, M.; Spina, E.; Serretti, A. A molecular pathway analysis informs the genetic background at risk for schizophrenia. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2015, 59, 21–30.
  85. Aflouk, Y.; Inoubli, O.; Saoud, H.; Zaafrane, F.; Gaha, L.; Bel Hadj Jrad, B. Association between TLR2 polymorphisms (− 196–174 Ins/Del, R677W, R753Q, and P631H) and schizophrenia in a Tunisian population. Immunol. Res. 2021, 69, 541–552.
  86. Kang, W.S.; Park, J.K.; Lee, S.M.; Kim, S.K.; Park, H.J.; Kim, J.W. Association between genetic polymorphisms of Toll-like receptor 2 (TLR2) and schizophrenia in the Korean population. Gene 2013, 526, 182–186.
  87. Sharma, I.; Priya, I.; Sharma, S.; Gupta, S.; Arora, M.; Mahajan, R.; Kapoor, N. Association of toll-like receptor 2 gene polymorphism (rs3804099) with susceptibility to Schizophrenia risk in the Dogra population of Jammu region, North India. Eur. J. Psychiatry 2022, 36, 106–113.
  88. Mostafa, M.; Elwasify, M.; Fathy, A.A.; Abdelsalam, M. Toll-Like Receptor 4 Gene Polymorphisms and Susceptibility to Schizophrenia: A Case-Control Study. Immunol. Investig. 2022, 51, 2009–2024.
More
This entry is offline, you can click here to edit this entry!
Video Production Service