Skip to main content

The role of infections and inflammation in schizophrenia: review of the evidence

Abstract

Background

Schizophrenia is a severe mental illness affecting approximately 1% of the population worldwide. While its exact causes remain unknown, emerging evidence suggests that infections and inflammation may contribute to disease development in a subset of individuals. This review comprehensively summarizes the evidence linking infections, immune system dysfunction, and schizophrenia risk.

Main body

Several population-based studies have linked serious prenatal or childhood infections requiring hospitalization to increased risk of later schizophrenia diagnosis, especially in individuals with genetic predisposition. Both central nervous system infections and systemic infections appear to confer risk. Specific pathogens including Toxoplasma gondii, herpesviruses, Chlamydophila, and more have been implicated. Autoimmune diseases are also associated with increased schizophrenia susceptibility, possibly due to blood-brain barrier disruption allowing brain-reactive antibodies access. The recent Coronavirus disease 2019 (COVID-19) pandemic raises questions about SARS-CoV-2 as a risk factor for new-onset psychosis. The mechanisms underlying the infection-schizophrenia link likely involve inflammation, cytokines, microglial activation, and tryptophan/kynurenine pathway modulation. Treatments targeting inflammation showed some efficacy in schizophrenia, further supporting an inflammation hypothesis. While the epidemiological and mechanistic evidence is substantial, further research is needed to conclusively determine the exact mechanisms linking immune dysfunction to schizophrenia requires further study.

Conclusion

The evidence suggests immune system abnormalities likely play a role, perhaps by interacting with genetic and environmental factors, in instigating schizophrenia pathophysiology in a subset of patients. More research is needed to understand these connections more clearly which may aid future prevention and personalized treatment approaches tailored to specific illness subtypes.

Background

Although the exact causes of schizophrenia (SCZ) remain elusive, emerging evidence suggests that infections and inflammation may play a role in the development of the disease in a subset of individuals. The correlation between recent hospital infections and the risk of schizophrenia exhibits an upward trend. Throughout history, both bacterial and viral infections, particularly following the 1918 influenza pandemic, have been acknowledged for their potential to trigger psychosis and symptoms resembling schizophrenia [1, 2]. Additionally, Swedish researchers identified a connection between hospital visits and schizophrenia spectrum disorders in 2014 [3].

This comprehensive review aims to summarize the evidence connecting infections, and immune system dysfunction, with the risk of schizophrenia. The review encompasses hypotheses linking infections to schizophrenia, epidemiological research implicating specific pathogens, maternal immune activation, potential mechanisms such as inflammation and cytokine pathways, immunological markers, and reviewing the clinical studies examining neuroinflammation and anti-inflammatory treatments in schizophrenia. The evidence suggests that immune system abnormalities likely contribute, possibly via interacting with genetic and environmental factors, to initiate schizophrenia in a subset of patients. Further research is essential to gain a clearer understanding of these connections, paving the way for future prevention and personalized treatment approaches tailored to specific illness subtypes.

Hypotheses about the relationship of infection and inflammation with schizophrenia

Historically, two concepts of studies have examined the potential connection between immunological disorders and severe psychoses. The first concept was based on the direct involvement of infectious agents, whereas the second concept considered the dysregulation of immuno-inflammatory systems [4].

The infective hypothesis

Schizophrenia has historically been linked to viral infections such as cytomegalovirus and herpes viruses, which have adversely affected the development of the central nervous system (CNS) [5]. Flu infection during pregnancy raises the likelihood of developing schizophrenia later, perhaps because of an imbalance in GABAergic transmission [6].

It was discovered that the levels of toxoplasma antibodies in schizophrenia patients were higher compared to those in the general population [7, 8]. Toxoplasma may contribute to the development of schizophrenia by elevating the levels of kynurenic acid in the central nervous system. This acid acts as an antagonist to the glutamatergic N-methyl-D-aspartate receptor (NMDA), which is involved in the regulation of neurotransmission [9].

Immuno-inflammatory dysregulation hypothesis

The concept of inflammatory dysregulation in schizophrenia was initially proposed based on the observation of the beneficial effects of anti-inflammatory medication and the decreased incidence of rheumatoid arthritis in schizophrenia patients compared to the general population [10].

Wright and Gilvarry (1996) linked type 1 diabetes to autoimmune thyrotoxicosis and SCZ in various families [11]. Moreover, Danish researchers also identified a 10% increase in SCZ and a 6% increase in autoimmune diseases among parents of individuals with SCZ. Furthermore, parental autoimmune diseases such as autoimmune hepatitis, type 1 diabetes, Sjögren’s syndrome, iridocyclitis, multiple sclerosis, psoriasis vulgaris, and dermato-polymyositis were associated with an elevated risk of SCZ [12].

Early ‘stressors’, such as problems during childbirth, are believed to be linked to the excessive production of certain genetic variations (e.g., inflammatory cytokines) that are responsible for the immunological dysregulation associated with schizophrenia [13].

The biological alterations associated with schizophrenia, such as immunological abnormalities, have usually been described by two perspectives [4]:

  • According to a neurodevelopmental hypothesis, the occurrence of symptoms in adulthood can be attributed to a disturbance in brain development that took place during early life.

  • According to a progressive hypothesis, symptoms and disruption of neurotransmitters are believed to cause changes in the immune system that can be noticed during an illness [14].

Evidence of the role of infection in immune dysregulation in schizophrenia

Specific pathogens implicated in increasing schizophrenia risk

Genetic predisposition

In populations genetically predisposed to the condition, maternal immune responses to viral illnesses may disrupt fetal brain development, contributing to the onset of SCZ [15]. Women with SCZ have been found to have higher levels of postpartum antibodies [16]. The largest Danish study, encompassing 3722 SCZ patients, indicated that maternal infections increased the risk of infant SCZ by 39%, decreasing to 23% after adjusting for parental mental illness [17].

This risk persisted whether infections occurred during or outside pregnancy, suggesting a genetic component [12]. Factors such as infections, autoimmune diseases, and genes related to inflammation may increase the risk of SCZ [18]. HLA genes, which play a role in regulating immunity, have been linked to both SCZ and autoimmune diseases [18]. In large-scale genome-wide association studies (GWAS), most of the associations between SCZ and HLA genes were found on chromosome 6 [18]. When stratifying studies on autoimmune diseases and infections, a family history of mental illness did not increase the risk of SCZ. Several autoimmune diseases share HLA markers, which may explain their connection to SCZ. The inverse association between SCZ and rheumatoid arthritis may have a genetic basis [19, 20].

Ascertainment bias or the anti-inflammatory and analgesic properties of antipsychotic medications may account for this weak association [21, 22]. The association between the HLA region and SCZ is supported by the complement component 4 (C4) gene, which is involved in synaptic pruning during postnatal development [23]. Interestingly, the polygenic risk score for SCZ did not increase the risk of infections [24]. This is intriguing because parents with SCZ had a higher risk of immune-related diseases outside of pregnancy, suggesting a genetic link. However, shared genes related to susceptibility to infections that are not included in the polygenic risk score for SCZ may influence this relationship. The polygenic risk score for SCZ was found to increase by 2.30-fold in the highest quartile of genetic load and by 3.39-fold in individuals with infections [24]. Another study found that the SCZ polygenic risk score predicted the risk of type 1 diabetes, rheumatoid arthritis, and Crohn’s disease [25]. Researchers have examined inflammation-related genes in blood, lymphoblastoid cell lines, and postmortem brains of individuals with SCZ using microarrays. The largest postmortem brain transcriptome analysis found no changes in 600 inflammation-related genes [26, 27]. However, other unbiased transcriptome profiles have identified alterations in immune-related gene expression in the blood and brains of individuals with SCZ. The largest GWAS for SCZ did not identify immune loci beyond the major histocompatibility complex [28]. While C-reactive protein has been associated with SCZ in studies of interleukin-6 (IL-6), this association was not confirmed in Mendelian randomization studies [29, 30].

Specific pathogens implicated in increasing schizophrenia risk

In a Danish study, encompassing a younger population and involving comprehensive follow-up of all hospital contacts from birth, it was revealed that 45% of psychotic patients had a prior history of hospital infections. This antecedent increased the risk of SCZ by 41% and the risk of bacterial infections by 63% [31]. Köhler et al. (2017) determined that less severe infections treated by general practitioners (GPs) also heightened the risk of schizophrenia, albeit not to the same extent as hospitalizations. Notably, most GP-treated bacterial illnesses, particularly those necessitating broad-spectrum antibiotics, exhibited an associated increased risk [32].

Since the 1918–1919 influenza pandemic, our understanding of influenza and schizophrenia has expanded significantly. Studies suggest a season-of-birth effect in schizophrenia, with maternal influenza infection increasing the likelihood of psychosis in offspring [33]. Stem cell technology could analyze the impact of influenza infection on the induced pluripotent stem-cell microglia-like cells, providing insights into neurodevelopment [34].

Toxoplasma gondii [35], human herpesvirus 2, Borna disease virus, human endogenous retrovirus, and Chlamydophila psittaci have all been found to increase the risk of SCZ [36]. In a large population-based study, Toxoplasma gondii serum titres were found to be dose-dependently associated with an increased risk of SCZ [37].

Herpes simplex virus infection has been linked to SCZ [38]. Patients with SCZ exhibited higher serum cytomegalovirus (CMV) antibody titres, particularly among those newly diagnosed and untreated [2, 39]. Although the brains of individuals with SCZ did not display neuropathological signs of CMV, elevated CMV levels were identified in their cerebrospinal fluid [40]. Additionally, individuals with SCZ were found to have higher levels of Borna virus in their serum and retroviral products [41, 42].

Furthermore, mental health has been significantly impacted by the recent Coronavirus disease 2019 (COVID-19) pandemic. While some case reports describe psychosis following COVID-19, uncertainties about causality arise due to confounding factors such as pre-existing conditions. Nevertheless, there is biological plausibility for an association between COVID-19 and psychosis [43, 44]. SARS-CoV-2 may adversely affect the brain through direct infection and inflammation [45]. The impact of COVID-19 on regions involved in psychosis, such as dopamine and glutamate pathways, is noteworthy [46, 47]. Inflammation resulting from COVID-19 could disrupt the blood-brain barrier, facilitating immune activation in the brain. Stress induced by COVID-19 might also prime vulnerability in individuals predisposed to psychosi s[48, 49]. Features of COVID-associated psychosis include a later onset and a positive response to antipsychotics [50]. Overall, while anecdotal data exists, robust evidence confirming a causal link between SARS-CoV-2 and new-onset psychosis is still lacking. More research is needed to unravel the complex interplay between COVID-19, inflammation, and psychotic illness [45, 51,52,53].

Regarding bacterial infection, patients with SCZ also reported higher rates of Chlamydophila infections, which were correlated with genetic markers related to the immune system [54]. Postmortem analysis revealed higher levels of Chlamydophila DNA in the brains of individuals with SCZ [55]. Population-based studies linked pneumococcal disease to psychosis [56]. Evidence of Bartonella infection in the blood of patients with schizophrenia and schizoaffective disorder was reported [57]. Notably, state mental hospitals even employed antibiotics in the treatment of neurosyphilis-induced mental illnesses [58].

Maternal immune activation

Maternal immune activation (MIA) disrupts normal fetal brain development [59,60,61], potentially causing over 30% of schizophrenia cases [62]. MIA is primarily caused by bacterial, viral, or parasitic illnesses, and antibodies against influenza or toxoplasmosis in maternal serum during pregnancy are linked to an elevated risk of schizophrenia in the offspring [63]. These infections elicit a maternal immunological response that includes the activation of different cytokine pathways such as IL-1, IL-6, TNFα, and IFNγ [59]. The effect of MIA on the fetal brain does not require direct infection: even in the absence of the virus, cytokine induction with polyI:C (a synthetic dsRNA) is enough to produce long-lasting effects, implying that the mother's response to the infection is important for altering fetal brain development [64].

Pathological findings in schizophrenia patients include increased GABAA receptor 2 immunoreactivity, dopamine hyperfunction, delayed hippocampal myelination, decreased NMDA receptor expression in the hippocampus, decreased numbers of reelin- and parvalbumin-positive cells, decreased dopamine D1 and D2 receptors in the prefrontal cortex, and increased tyrosine hydroxylase in striatal structures, and similar changes are present in the adult offspring of MIA-exposed mice [65]. Postmortem brain transcriptome studies are crucial for detecting disrupted genes and pathways caused by genetic predispositions and environmental insults [66, 67].

Factors leading to confusion between schizophrenia and immunological illnesses

Psychological stress before a diagnosis can increase the risk of infections and immunological issues in patients with SCZ [68, 69]. It is important to note that while inflammation and immune-related illnesses may not directly cause SCZ, psychological stress might precede the development of both psychiatric and immunological problems that can be explained by the duration of untreated conditions [70, 71]. Some chronic autoimmune illnesses, when considered alone, do not significantly raise the risk of SCZ. It is worth mentioning that the use of anti-autoimmune steroids and interferon therapy might increase the risk of psychosis, although such iatrogenic consequences are relatively rare. Surprisingly, steroids, in some cases, can actually lower the risk of psychosis [72].

Antibiotics can also influence the gut microbiota, which can have an impact on both the immune system and the risk of SCZ [73]. Additionally, antipsychotic medications can affect immunological responses, potentially increasing the risk of autoimmune disorders and infections [74]. Unhealthy lifestyle choices, such as smoking, drinking, and drug abuse, can weaken the immune system in individuals without SCZ, making them more susceptible to infections and autoimmune conditions. Furthermore, certain social and behavioral traits may prevent individuals with SCZ from seeking help or adhering to their prescribed medications, which could lead to autoimmune illnesses and hospitalization due to infections. However, having a family history of mental illness or a history of substance addiction did not appear to increase the risk of infection or autoimmune diseases [75]. Also, there is a role of hormones [76, 77] and drugs [78,79,80] that affect the immune system with mental illness.

The association between NMDAR antibodies and the glutamate/NMDAR hypofunction hypothesis of psychotic disorders is also of interest [81, 82]. Anti-NMDAR encephalitis is a unique and complex autoimmune encephalitis that is the most common type [83]. It is caused by IgG antibodies targeting the NMDAR [84]. Due to early psychiatric symptoms, around 80% of cases first present to psychiatrists, and over 60% are initially admitted to psychiatric units [83, 85]. As a result, psychiatrists play a critical role in diagnosing this disorder and are encouraged to frequently consider it in their practice [86]. Also, Hammer et al. established a correlation between NMDAR antibody seropositivity and influenza virus A or B IgG [87].

During systemic inflammation, substances like autoantibodies, cytokines, and certain T-cell subsets involved in brain surveillance can enter the blood-brain barrier [88]. There is also evidence to suggest that an imbalance in the Th1-Th2 immune system can lead to the development of autoimmune and atopic illnesses in individuals with SCZ. Inflammation can stimulate peripheral neurons or activate pathways like the tryptophan-kynurenine pathway, which can impact neurotransmitters like glutamate, serotonin, and potentially dopamine [89]. Additionally, low-grade brain inflammation resulting from infections and autoimmune diseases may contribute to specific subtypes of SCZ [90]. When activated, microglia, which are immune cells in the brain, can alter brain signaling during inflammation [91]. It's important to note that the immune system might not always recognize infectious agents, and latent infections that emerge after acute infections or inflammation can lead to symptoms [55].

Furthermore, the gut microbiota has been found to influence brain function and behavior through various processes involving neurons, endocrine signaling, and the immune system [73]. It is also possible that immunological factors and brain inflammation might predispose genetically susceptible individuals to SCZ. The observed familial connection between autoimmune diseases and SCZ suggests that certain subgroups of patients with SCZ might exhibit autoimmune symptoms [75, 92]. Hence, both SCZ and autoimmunity share genetic and pathogenic factors [93].

Neuroimmunology markers

Immune dysregulation appears to play some role in the pathophysiology and outcome of major psychoses along with genetic factors and neurodevelopmental disturbances [94, 95]. For this reason, in recent years, researchers have tried to identify neuroimmunology markers, defined as any genetically or environmentally biological parameter, with the aim of identifying individuals at risk and monitoring the progression of illness [96].

Microglia

Immune cells in the brain, play a critical role in these processes. The link between psychiatric disorders and immune system dysfunction has long been recognized, dating back to observations by Kraepelin and Menninger in the late nineteenth and early twentieth centuries [97, 98]. Advances in immunology and genetics have furthered our understanding of how the immune system impacts brain function and contributes to conditions like depression, SCZ, autism spectrum disorders, and bipolar disorder. Neuroinflammation, characterized by abnormal cytokine and chemokine levels and changes in astrocytes and microglia, is common in these disorders [97, 98].

The concept of inflammation has existed for over 2000 years, defined by Celsus as redness, swelling, heat, and discomfort. Inflammation is critical for tissue recovery after injury [99]. Inflammation in CNS, termed neuroinflammation, involves immune cells and molecules working to fight infections and promote healing, similar to multiple sclerosis, stroke, traumatic brain injury, and CNS infections [97, 98]. However, oversimplifying neuroinflammation can impede research efforts in mental and neurodegenerative diseases [100].

Microglia, the resident immune cells of the brain, play a key role in neuroinflammation. First described by Nissl and del Rio Hortega in the late nineteenth/early twentieth centuries, they are dynamic cells that monitor and respond to their environment [101]. Recent research has revealed that microglia originate from primitive yolk sac macrophages and have a complex developmental process. They enter the CNS during embryonic development and remain there throughout life. Unlike fetal monocytes, microglia become part of the CNS after the blood-brain barrier has already formed [102, 103].

Microglia have diverse functions in neurodevelopment, regulating neural progenitors, neurogenesis, synaptic pruning, and phagocytosis. They also remodel synapses and support adult neurogenesis [104,105,106,107]. Microglia possess receptor networks enabling them to sense neurons and respond via phagocytosis, cytokine release, and more [108, 109]. Beyond immune activities, they impact sleep, learning, memory, plasticity, and neurogenesis through the release of cytokines like IL-1β and TNFα [110]. Dysregulated microglial cytokines may impair cognition in psychiatric patients. In summary, the historical and modern research highlights the critical interactions between microglia and neurons, and the connections between immune dysfunctions and psychiatric illness. Further study of proinflammatory cytokines and their roles is needed.

Cytokines

Cytokines are signaling molecules released by both innate and adaptive immune cells, playing a role in both the brain and the peripheral immune system. These molecules are responsible for coordinating various immune responses and maintaining the balance between defending the body against pathogens and tolerating self-antigens and beneficial microorganisms. In SCZ, peripheral cytokine changes may impact the brain via several mechanisms [111].

Studying medication-naive patients provides insights into inherent cytokine changes in SCZ [112,113,114,115]. Meta-analyses found mixed cytokine alterations in first-episode psychosis, including lower IL-1β, IL-6, and TNFα [116]. Cytokine levels appear to increase during acute episodes and decrease with treatment response [117,118,119,120]. Different psychiatric disorders show increased inflammatory cytokines [121,122,123]. Chronic SCZ patients exhibit elevated TNFα, IL-12, IFNγ, and IL-6 [122,123,124]. Cerebrospinal fluid cytokine changes mirror blood findings [125, 126].

Oxidative stress from glutathione depletion may contribute to SCZ pathophysiology [127,128,129,130]. Glutamate dysfunction is implicated in SCZ and depression [131,132,133]. Inflammation can increase reactive oxygen species, impacting glutathione defense [134,135,136]. In summary, cytokines are involved in SCZ onset and progression, with intricate links to oxidative stress, glutamate disruption, and inflammation. Table 1 provides an overview of the major cytokines that have been implicated in SCZ research.

Microbiome and gut inflammation

Table 1 Overview of cytokine dysregulation in schizophrenia (key findings from the literature)

SCZ has long been associated with gastrointestinal (GI) issues like irritable bowel syndrome, gluten sensitivity, ulcerative colitis, and Crohn’s disease. Patients of SCZ especially those in the early stages and those who are medication-naïve, tend to have elevated antibodies against Saccharomyces cerevisiae (ASCA) that link antigenic foods, gut bacteria, and SCZ [139,140,141,142,143,144]. Moreover, a previous study found a relationship between cognitive function and gut bacteria [145].

Studies link SCZ to markers like sCD14, LBP, CRP, and food antigen antibodies suggesting microbial translocation causes inflammation and GI problems [146]. A healthy gut is crucial for digestion, absorption, immunity, and maintaining the gut-blood barrier. Factors like stress, drugs, infections, and genetic susceptibility can disrupt this balance [147].

Considering the gut microbiome dysbiosis in SCZ is key, as it can cause neuroinflammation. Translocated GI products can have detrimental effects on brain connections [148]. SCZ may alter endothelial cells and blood-brain barrier permeability. This can cause inflammation and inflammatory cell translocation into the brain [111, 149].

Germ-free rodent models show gut flora impacts brain maturation and function. Fortunately, modifying the bacterial composition, performing vagotomies, and employing probiotic or antibiotic therapies can help mitigate these effects [150]. Changes in the microbiota can affect blood-brain barrier permeability. Metagenomic and 16S rRNA gene sequencing studies reveal differences in the microbiomes of SCZ patients compared to controls. Interestingly, they found that patients of SCZ had more lactobacilli and bifidobacteria in their oropharyngeal microbiomes, which can contribute to inflammation [151].

Imbalances in commensal bacteria and yeast, often from medications and diet, can lead to dysbiosis and inflammation in SCZ [152]. The gut-brain connection and its role in SCZ involves many intricate factors that require further research.

Immune cell count

The inconsistency of the findings stems from the fact that most studies analyzed the impact of antipsychotics on drug-naïve patients rather than their lymphocyte distribution. Nevertheless, the findings of most studies are consistent in that drug-naïve patients have a decrease in T cells and an increase in B cells, whereas patients who are medicated exhibit the opposite pattern [153].

  1. a)

    Polyunsaturated fatty acids

Several studies have reported that individuals with schizophrenia have reduced levels of polyunsaturated fatty acids (PUFA) in their brain and peripheral membranes [154]. There is a hypothesis that this decline may be attributed to heightened arachidonic acid degradation caused by compromised immune function and excessive prostaglandin production, specifically prostaglandin E [155].

Antioxidant defence system

Neuron injury in schizophrenic patients may be attributable to a deficiency in the antioxidant defense system (AODS) [156]. In fact, AODS inhibits the cellular damage caused by the free radicals that proliferate excessively during inflammatory processes. AODS comprises both enzymatic and non-enzymatic antioxidants (superoxide dismutase, catalase, glutathione peroxidase, albumin, bilirubin, uric acid, ascorbic acid, tocopherol, glutathione) [156]. Reduced cellular and plasma antioxidant levels are anticipated in patients with schizophrenia. Schizophrenics exhibited reduced levels of the following non-enzymatic antioxidants in their plasma: albumin [157], bilirubin [158], uric acid [159], ascorbic acid [160], glutathione [161], and tocopherol [162]. Divergent findings were observed with respect to the concentrations of superoxide dismutase in serum and cells.

One hypothesis posits that chronic patients may exhibit elevated levels of this enzyme as a compensatory mechanism against oxidative stress [156]. Nitric oxide, an essential regulator and modulator of numerous inflammatory conditions, is purportedly elevated in schizophrenic patients due to its function as a source of free radicals. However, the lack of consistency in the results [163, 164] precludes the use of NO as a diagnostic indicator to differentiate schizophrenics from healthy controls.

Clinical evidence of neuroinflammation in schizophrenia

Imaging studies

Imaging studies have shown chronic inflammation in the brains of patients with SCZ, with low concentrations of a peripheral benzodiazepine receptor found in the brain by positron emission tomography (PET) scan. This correlated with the chronic inflammation hypothesis as elevated concentrations of a peripheral benzodiazepine receptor are attributed to a microglial response during active neuroinflammation [165, 166].

Although there is a lack of research examining the correlation between peripheral cytokine levels and neuroinflammation in SCZ, studies have found a correlation between elevated IL-6 gene expression in blood leukocytes and reduced left hippocampal volume in SCZ [167]. This suggests that IL-6, brain-derived neurotrophic factor (BDNF), and cortisol may have a synergistic effect on hippocampal volume [168]. However, the absence of evidence for gliosis in SCZ remains incongruous despite the vast body of literature. Schneider and Dwork [169] have conducted an exhaustive literature review on gliosis and SCZ postmortem. Although some negative findings were also documented, the review incorporated positive studies by Bayer et al. [170], Radewicz et al. [171], and Steiner et al. [172] reported increased immunoreactive microglia in SCZ.

Management studies

Immunotherapies which are targeting distinct symptoms associated with SCZ can be categorized into various classes, each addressing specific aspects of the disease progression:

Anti-inflammatory drugs

Nonsteroidal anti-inflammatory agents, including aspirin, celecoxib, and minocycline, have demonstrated anti-inflammatory effects in SCZ [173, 174]. The anti-inflammatory effects of these medications in SCZ are summarised in Table 2. Further investigation is crucial to comprehend their anti-inflammatory qualities and clinical significance.

Antioxidants

Table 2 Summary of emerging evidence of different medications with anti-inflammatory and antioxidant action used in SCZ

Antioxidants are substances that protect cells from free radicals. While they may not provide a cure for SCZ, antioxidants can improve overall cell health. Antioxidants, such as N-acetylcysteine (NAC), ascorbic acid, α-tocopherol, EPA, DHA, melatonin, and L-Theanine, have been explored in SCZ research [174,175,176,177]. The role in SCZ is summarised in Table 2.

Antipsychotics with anti-inflammatory effects

For more than half a century, antipsychotic medications have been an essential component in the management of psychosis. First-generation antipsychotics (FGAs) and second-generation antipsychotics (SGAs) are the two classifications applicable to these medications. SGAs have been observed to exert an influence on dopamine and serotonin receptors. Additionally, they may indirectly affect glutamatergic receptors, specifically NMDA receptors, through alpha-adrenergic, histaminic, and cholinergic receptors in certain instances. The principal indication for the efficacy of antipsychotics is the treatment of acute positive psychotic symptoms, such as disorganization, hallucinations, and delusions. Additionally, they might offer certain advantages in reducing cognitive and negative symptoms; however, their effectiveness may differ [185].

In the context of the relationship between schizophrenia and inflammation and infection, it was found that some psychotropic drugs have antiviral properties [186, 187]. It is of interest that psychotropic compounds commonly used to treat mental disorders exercise a putative preventive effect against the most catastrophic outcomes related to SARS-CoV-2 infection [187]. Regarding antipsychotics, chlorpromazine (phenothiazine) is a well-established antipsychotic medication that has recently been proposed to have antiviral activity against SARS-CoV-2 [186]. Other antipsychotic medications (risperidone and olanzapine, clozapine, and haloperidol) also have been studied. The increasing evidence of antipsychotic anti-inflammatory effects is summarised in Table 3.

Table 3 Summary of emerging evidence of different antipsychotic drugs for their anti-inflammatory and anti-infection action

Other agents

Other agents have shown early anti-inflammatory promise for SCZ, but further research is still needed. They include biological therapy, peroxisome proliferator-activated receptors (PPARs), neuroprotectors, and herbs.

Conclusion

The collective evidence suggests that infections, inflammation, and immune system abnormalities may play an etiological role in schizophrenia. Both prenatal infections and immune insults later in neurodevelopment appear contributory in some cases. There are likely complex interactions between genetic susceptibility and environmental immune triggers influencing schizophrenia risk. However, more research focused on understanding the precise immunological mechanisms and the involved pathways is still needed.

Availability of data and materials

Not applicable.

Abbreviations

SCZ:

Schizophrenia

CNS:

Central nervous system

CMV:

Cytomegalovirus

GWAS:

Genome-wide association studies

DNA:

Deoxyribonucleic acid

COVID-19:

Coronavirus disease 2019

PET:

Positron emission tomography

NAC:

N-acetylcysteine

FGAs:

First-generation antipsychotics

SGAs:

Second-generation antipsychotics

C4:

the complement component 4

FEP:

First episode psychosis

GI:

Gastrointestinal

NAC:

N-acetylcysteine

PPARs:

Peroxisome proliferator-activated receptors

References

  1. Noll R (2007) Kraepelin’slost biological psychiatry’? Autointoxication, organotherapy and surgery for dementia praecox. Hist Psychiatry 18(3):301–320

    Article  PubMed  Google Scholar 

  2. Torrey EF, Leweke MF, Schwarz MJ, Mueller N, Bachmann S, Schroeder J et al (2006) Cytomegalovirus and schizophrenia. CNS Drugs 20:879–885

    Article  PubMed  Google Scholar 

  3. Blomström Å, Karlsson H, Svensson A, Frisell T, Lee BK, Dal H et al (2014) Hospital admission with infection during childhood and risk for psychotic illness—a population-based cohort study. Schizophr Bull 40(6):1518–1525

    Article  PubMed  Google Scholar 

  4. Altamura AC, Buoli M, Pozzoli S (2014) Role of immunological factors in the pathophysiology and diagnosis of bipolar disorder: comparison with schizophrenia. Psychiatry Clin Neurosci 68(1):21–36. https://doi.org/10.1111/pcn.12089

    Article  PubMed  CAS  Google Scholar 

  5. Krause D, Matz J, Weidinger E, Wagner J, Wildenauer A, Obermeier M et al (2010) The association of infectious agents and schizophrenia. World J Biol Psychiatry 11(5):739–743. https://doi.org/10.3109/15622971003653246

    Article  PubMed  Google Scholar 

  6. Nyffeler M, Meyer U, Yee BK, Feldon J, Knuesel I (2006) Maternal immune activation during pregnancy increases limbic GABAA receptor immunoreactivity in the adult offspring: implications for schizophrenia. Neuroscience. 143(1):51–62. https://doi.org/10.1016/j.neuroscience.2006.07.029

    Article  PubMed  CAS  Google Scholar 

  7. Torrey EF, Bartko JJ, Lun ZR, Yolken RH (2007) Antibodies to Toxoplasma gondii in patients with schizophrenia: a meta-analysis. Schizophr Bull 33(3):729–736. https://doi.org/10.1093/schbul/sbl050

    Article  PubMed  Google Scholar 

  8. Ahmed G, Khalifa H, Hassaan S, Hussein M, Shaaban I, Farrag H (2020) Seroprevalence of Toxoplasma gondii among patients with schizophrenia and bipolar disorder in Upper Egypt: a comparative study with a control group. Ann Parasitol  https://doi.org/10.17420/ap6602.253

  9. Schwarcz R, Hunter CA (2007) Toxoplasma gondii and schizophrenia: linkage through astrocyte-derived kynurenic acid? Schizophr Bull 33(3):652–653. https://doi.org/10.1093/schbul/sbm030

    Article  PubMed  PubMed Central  Google Scholar 

  10. Gorwood P, Pouchot J, Vinceneux P, Puéchal X, Flipo RM, De Bandt M et al (2004) Rheumatoid arthritis and schizophrenia: a negative association at a dimensional level. Schizophr Res 66(1):21–29. https://doi.org/10.1016/s0920-9964(03)00017-3

    Article  PubMed  CAS  Google Scholar 

  11. Wright P, Sham PC, Gilvarry CM, Jones PB, Cannon M, Sharma T et al (1996) Autoimmune diseases in the pedigrees of schizophrenic and control subjects. Schizophr Res 20(3):261–267

    Article  PubMed  CAS  Google Scholar 

  12. Eaton WW, Pedersen MG, Nielsen PR, Mortensen PB (2010) Autoimmune diseases, bipolar disorder, and non-affective psychosis. Bipolar Disord 12(6):638–646

    Article  PubMed  PubMed Central  Google Scholar 

  13. Di Nicola M, Cattaneo A, Hepgul N, Di Forti M, Aitchison KJ, Janiri L et al (2013) Serum and gene expression profile of cytokines in first-episode psychosis. Brain Behav Immun 31:90–95. https://doi.org/10.1016/j.bbi.2012.06.010

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Murray RM, Sham P, Van Os J, Zanelli J, Cannon M, McDonald C (2004) A developmental model for similarities and dissimilarities between schizophrenia and bipolar disorder. Schizophr Res 71(2–3):405–416. https://doi.org/10.1016/j.schres.2004.03.002

    Article  PubMed  Google Scholar 

  15. Ozawa K, Hashimoto K, Kishimoto T, Shimizu E, Ishikura H, Iyo M (2006) Immune activation during pregnancy in mice leads to dopaminergic hyperfunction and cognitive impairment in the offspring: a neurodevelopmental animal model of schizophrenia. Biol Psychiatry 59(6):546–554

    Article  PubMed  CAS  Google Scholar 

  16. Buka SL, Tsuang MT, Torrey EF, Klebanoff MA, Bernstein D, Yolken RH (2001) Maternal infections and subsequent psychosis among offspring. Arch Gen Psychiatry 58(11):1032–1037

    Article  PubMed  CAS  Google Scholar 

  17. Nielsen PR, Laursen TM, Mortensen PB (2013) Association between parental hospital-treated infection and the risk of schizophrenia in adolescence and early adulthood. Schizophr Bull 39(1):230–237

    Article  PubMed  Google Scholar 

  18. Ripke S, Neale BM, Corvin A, Walters JTR, Farh K-H, Holmans PA et al (2014) Biological insights from 108 schizophrenia-associated genetic loci. Nature. 511(7510):421–427. https://doi.org/10.1038/nature13595

    Article  PubMed Central  CAS  Google Scholar 

  19. Benros ME, Mortensen PB, Eaton WW (2012) Autoimmune diseases and infections as risk factors for schizophrenia. Ann N Y Acad Sci 1262(1):56–66

    Article  PubMed  Google Scholar 

  20. Eaton WW, Hayward C, Ram R (1992) Schizophrenia and rheumatoid arthritis: a review. Schizophr Res 6(3):181–192

    Article  PubMed  CAS  Google Scholar 

  21. Torrey EF, Yolken RH (2001) The schizophrenia–rheumatoid arthritis connection: infectious, immune, or both? Brain Behav Immun 15(4):401–410

    Article  PubMed  CAS  Google Scholar 

  22. Sellgren C, Frisell T, Lichtenstein P, Landèn M, Askling J (2014) The association between schizophrenia and rheumatoid arthritis: a nationwide population-based Swedish study on intraindividual and familial risks. Schizophr Bull 40(6):1552–1559

    Article  PubMed  PubMed Central  Google Scholar 

  23. Sekar A, Bialas AR, De Rivera H, Davis A, Hammond TR, Kamitaki N et al (2016) Schizophrenia risk from complex variation of complement component 4. Nature. 530(7589):177–183

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  24. Benros ME, Trabjerg BB, Meier S, Mattheisen M, Mortensen PB, Mors O et al (2016) Influence of polygenic risk scores on the association between infections and schizophrenia. Biol Psychiatry 80(8):609–616. https://doi.org/10.1016/j.biopsych.2016.04.008

    Article  PubMed  Google Scholar 

  25. Stringer S, Kahn RS, de Witte LD, Ophoff RA, Derks EM (2014) Genetic liability for schizophrenia predicts risk of immune disorders. Schizophr Res 159(2–3):347–352

    Article  PubMed  Google Scholar 

  26. Birnbaum R, Jaffe A, Chen Q, Shin J, Kleinman J, Hyde T et al (2018) Investigating the neuroimmunogenic architecture of schizophrenia. Mol Psychiatry 23(5):1251–1260

    Article  PubMed  CAS  Google Scholar 

  27. Gandal MJ, Haney JR, Parikshak NN, Leppa V, Ramaswami G, Hartl C et al (2018) Shared molecular neuropathology across major psychiatric disorders parallels polygenic overlap. Science. 359(6376):693–697

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  28. Pouget JG, Gonçalves VF, Spain SL, Finucane HK, Raychaudhuri S, Kennedy JL et al (2016) Genome-wide association studies suggest limited immune gene enrichment in schizophrenia compared to 5 autoimmune diseases. Schizophr Bull 42(5):1176–1184. https://doi.org/10.1093/schbul/sbw059

    Article  PubMed  PubMed Central  Google Scholar 

  29. Hartwig FP, Borges MC, Horta BL, Bowden J, Smith GD (2017) Inflammatory biomarkers and risk of schizophrenia: a 2-sample mendelian randomization study. JAMA Psychiatry 74(12):1226–1233

    Article  PubMed  PubMed Central  Google Scholar 

  30. Prins BP, Abbasi A, Wong A, Vaez A, Nolte I, Franceschini N et al (2016) Investigating the causal relationship of C-reactive protein with 32 complex somatic and psychiatric outcomes: a large-scale cross-consortium Mendelian randomization study. PLoS Med 13(6):e1001976

    Article  PubMed  PubMed Central  Google Scholar 

  31. Nielsen PR, Benros ME, Mortensen PB (2014) Hospital contacts with infection and risk of schizophrenia: a population-based cohort study with linkage of Danish national registers. Schizophr Bull 40(6):1526–1532

    Article  PubMed  Google Scholar 

  32. Köhler O, Petersen L, Mors O, Mortensen P, Yolken R, Gasse C et al (2017) Infections and exposure to anti-infective agents and the risk of severe mental disorders: a nationwide study. Acta Psychiatr Scand 135(2):97–105

    Article  PubMed  Google Scholar 

  33. Kępińska AP, Iyegbe CO, Vernon AC, Yolken R, Murray RM, Pollak TA (2020) Schizophrenia and influenza at the centenary of the 1918-1919 Spanish influenza pandemic: mechanisms of psychosis risk. Front Psych 11:72. https://doi.org/10.3389/fpsyt.2020.00072

    Article  Google Scholar 

  34. Sellgren CM, Gracias J, Watmuff B, Biag JD, Thanos JM, Whittredge PB et al (2019) Increased synapse elimination by microglia in schizophrenia patient-derived models of synaptic pruning. Nat Neurosci 22(3):374–385. https://doi.org/10.1038/s41593-018-0334-7

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Hussein EAM, Khalifa H, Ramadan GK, Hassaan SH, Shaaban I, Farrag HMM (2020) Seroprevalence of Toxoplasma gondii among patients with schizophrenia and bipolar disorder in Upper Egypt: a comparative study with a control group. Ann Parasitol 66(2):183–192. https://doi.org/10.17420/ap6602.253

    Article  PubMed  Google Scholar 

  36. Arias I, Sorlozano A, Villegas E, de Dios LJ, McKenney K, Cervilla J et al (2012) Infectious agents associated with schizophrenia: a meta-analysis. Schizophr Res 136(1–3):128–136. https://doi.org/10.1016/j.schres.2011.10.026

    Article  PubMed  Google Scholar 

  37. Pedersen MG, Stevens H, Pedersen CB, Nørgaard-Pedersen B, Mortensen PB (2011) Toxoplasma infection and later development of schizophrenia in mothers. Am J Psychiatr 168(8):814–821

    Article  PubMed  Google Scholar 

  38. Niebuhr DW, Millikan AM, Yolken R, Li Y, Weber NS (2008) Results from a hypothesis generating case-control study: herpes family viruses and schizophrenia among military personnel. Schizophr Bull 34(6):1182–1188

    Article  PubMed  Google Scholar 

  39. Leweke FM, Gerth CW, Koethe D, Klosterkötter J, Ruslanova I, Krivogorsky B et al (2004) Antibodies to infectious agents in individuals with recent onset schizophrenia. Eur Arch Psychiatry Clin Neurosci 254:4–8

    Article  PubMed  Google Scholar 

  40. Torrey EF, Yolken RH, Winfrey CJ (1982) Cytomegalovirus antibody in cerebrospinal fluid of schizophrenic patients detected by enzyme immunoassay. Science. 216(4548):892–894

    Article  PubMed  CAS  Google Scholar 

  41. Karlsson H, Bachmann S, Schröder J, McArthur J, Torrey EF, Yolken RH (2001) Retroviral RNA identified in the cerebrospinal fluids and brains of individuals with schizophrenia. Proc Natl Acad Sci 98(8):4634–4639

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  42. Hart DJ, Heath RG, Sautter FJ Jr, Schwartz BD, Garry RF, Choi B et al (1999) Antiretroviral antibodies: implications for schizophrenia, schizophrenia spectrum disorders, and bipolar disorder. Biol Psychiatry 45(6):704–714

    Article  PubMed  CAS  Google Scholar 

  43. Shehata RR, Ahmed GK, Hussien AARM, Mahmoud MA (2023) Does post-acute COVID-19 syndrome women’s sex problems link to psychiatry after 6 months? The Egyptian Journal of Neurology. Psychiatry Neurosurg 59(1):119. https://doi.org/10.1186/s41983-023-00722-7

    Article  Google Scholar 

  44. Ahmed GK, Khedr EM, Hamad DA, Meshref TS, Hashem MM, Aly MM (2021) Long term impact of Covid-19 infection on sleep and mental health: a cross-sectional study. Psychiatry Res 305:114243. https://doi.org/10.1016/j.psychres.2021.114243

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  45. Moccia L, Kotzalidis GD, Bartolucci G, Ruggiero S, Monti L, Biscosi M et al (2023) COVID-19 and new-onset psychosis: a comprehensive review. J Pers Med 13(1):104

    Article  PubMed  PubMed Central  Google Scholar 

  46. Popoli M, Yan Z, McEwen BS, Sanacora G (2012) The stressed synapse: the impact of stress and glucocorticoids on glutamate transmission. Nat Rev Neurosci 13(1):22–37. https://doi.org/10.1038/nrn3138

    Article  CAS  Google Scholar 

  47. Pruessner M, Cullen AE, Aas M, Walker EF (2017) The neural diathesis-stress model of schizophrenia revisited: An update on recent findings considering illness stage and neurobiological and methodological complexities. Neurosci Biobehav Rev 73:191–218. https://doi.org/10.1016/j.neubiorev.2016.12.013

    Article  PubMed  Google Scholar 

  48. Ahmed GK, Ramadan HK-A, Refay SM, Khashbah MA (2021) Comparison of knowledge, attitude, socioeconomic burden, and mental health disorders of COVID-19 pandemic between general population and health care workers in Egypt. Egypt J Neurol Psychiatry Neurosurg 57(1):25. https://doi.org/10.1186/s41983-021-00280-w

    Article  Google Scholar 

  49. Osman DM, Khalaf FR, Ahmed GK, Abdelbadee AY, Abbas AM, Mohammed HM (2022) Worry from contracting COVID-19 infection and its stigma among Egyptian health care providers. J Egypt Public Health Assoc 97(1):2. https://doi.org/10.1186/s42506-021-00099-6

    Article  PubMed  PubMed Central  Google Scholar 

  50. Chaudhary AMD, Musavi NB, Saboor S, Javed S, Khan S, Naveed S (2022) Psychosis during the COVID-19 pandemic: A systematic review of case reports and case series. J Psychiatr Res 153:37–55. https://doi.org/10.1016/j.jpsychires.2022.06.041

    Article  PubMed  PubMed Central  Google Scholar 

  51. Ahmed GK, Elbeh K, Gomaa HM, Soliman S (2021) Does COVID-19 infection have an impact on children’s psychological problems? Middle East Curr Psychiatry 28(1):77. https://doi.org/10.1186/s43045-021-00155-z

    Article  Google Scholar 

  52. Ahmed GK, Mostafa S, Elbeh K, Gomaa HM, Soliman S (2022) Effect of COVID-19 infection on psychological aspects of pre-schooler children: a cross-sectional study. Middle East Curr Psychiatry 29(1):42. https://doi.org/10.1186/s43045-022-00207-y

    Article  Google Scholar 

  53. Gk A, Salman SA, Elbeh K, Amer ZS, Abbas AM (2022) Correlation between psychiatric impact of COVID-19 during pregnancy and fetal outcomes in Egyptian women. Psychiatry Res 317:114920. https://doi.org/10.1016/j.psychres.2022.114920

    Article  Google Scholar 

  54. Fellerhoff B, Laumbacher B, Mueller N, Gu S, Wank R (2007) Associations between Chlamydophila infections, schizophrenia and risk of HLA-A10. Mol Psychiatry 12(3):264–272

    Article  PubMed  CAS  Google Scholar 

  55. Fellerhoff B, Wank R (2011) Increased prevalence of Chlamydophila DNA in post-mortem brain frontal cortex from patients with schizophrenia. Schizophr Res 129(2–3):191–195

    Article  PubMed  Google Scholar 

  56. Crump C, Winkleby MA, Sundquist K, Sundquist J (2013) Comorbidities and mortality in persons with schizophrenia: a Swedish national cohort study. Am J Psychiatr 170(3):324–333

    Article  PubMed  Google Scholar 

  57. Lashnits E, Maggi R, Jarskog F, Bradley J, Breitschwerdt E (2021) Frohlich F. Schizophrenia and Bartonella spp. Infection: A Pilot Case-Control Study. Vector Borne Zoonotic Dis 21(6):413–421. https://doi.org/10.1089/vbz.2020.2729

    Article  PubMed  PubMed Central  Google Scholar 

  58. Sullivan PF, Daly MJ, O’donovan M. (2012) Genetic architectures of psychiatric disorders: the emerging picture and its implications. Nat Rev Genet 13(8):537–551

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  59. Shi L, Fatemi SH, Sidwell RW, Patterson PH (2003) Maternal influenza infection causes marked behavioral and pharmacological changes in the offspring. J Neurosci 23(1):297–302. https://doi.org/10.1523/JNEUROSCI.23-01-00297.2003

    Article  PubMed  PubMed Central  Google Scholar 

  60. Nawa H, Takei N (2006) Recent progress in animal modeling of immune inflammatory processes in schizophrenia: implication of specific cytokines. Neurosci Res 56(1):2–13. https://doi.org/10.1016/j.neures.2006.06.002

    Article  PubMed  CAS  Google Scholar 

  61. Nawa H, Yamada K (2012) Experimental schizophrenia models in rodents established with inflammatory agents and cytokines. Methods Mol Biol 829:445–451. https://doi.org/10.1007/978-1-61779-458-2_28

    Article  PubMed  CAS  Google Scholar 

  62. Brown AS, Derkits EJ (2010) Prenatal infection and schizophrenia: a review of epidemiologic and translational studies. Am J Psychiatry 167(3):261–280. https://doi.org/10.1176/appi.ajp.2009.09030361

    Article  PubMed  PubMed Central  Google Scholar 

  63. Brown AS, Schaefer CA, Quesenberry CP Jr, Liu L, Babulas VP, Susser ES (2005) Maternal exposure to toxoplasmosis and risk of schizophrenia in adult offspring. Am J Psychiatry 162(4):767–773. https://doi.org/10.1176/appi.ajp.162.4.767

    Article  PubMed  Google Scholar 

  64. Smith SE, Li J, Garbett K, Mirnics K, Patterson PH (2007) Maternal immune activation alters fetal brain development through interleukin-6. J Neurosci 27(40):10695–10702. https://doi.org/10.1523/JNEUROSCI.2178-07.2007

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  65. Boksa P (2010) Effects of prenatal infection on brain development and behavior: a review of findings from animal models. Brain Behav Immun 24(6):881–897. https://doi.org/10.1016/j.bbi.2010.03.005

    Article  PubMed  Google Scholar 

  66. Horváth S, Janka Z, Mirnics K (2011) Analyzing schizophrenia by DNA microarrays. Biol Psychiatry 69(2):157–162. https://doi.org/10.1016/j.biopsych.2010.07.017

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  67. Mirnics K, Levitt P, Lewis DA (2006) Critical appraisal of DNA microarrays in psychiatric genomics. Biol Psychiatry 60(2):163–176. https://doi.org/10.1016/j.biopsych.2006.02.003

    Article  PubMed  CAS  Google Scholar 

  68. Pedersen A, Zachariae R, Bovbjerg DH (2010) Influence of psychological stress on upper respiratory infection—a meta-analysis of prospective studies. Psychosom Med 72(8):823–832

    Article  PubMed  Google Scholar 

  69. Ader R, Cohen N, Felten D (1995) Psychoneuroimmunology: interactions between the nervous system and the immune system. Lancet 345(8942):99–103. https://doi.org/10.1016/s0140-6736(95)90066-7

    Article  PubMed  CAS  Google Scholar 

  70. Ahmed GK, Elbeh K, Khalifa H, Samaan MR (2021) Impact of duration of untreated illness in bipolar I disorder (manic episodes) on clinical outcome, socioecnomic burden in Egyptian population. Psychiatry Res 296:113659. https://doi.org/10.1016/j.psychres.2020.113659

    Article  PubMed  Google Scholar 

  71. Zoghbi AW, Lieberman JA, Girgis RR (2023) The neurobiology of duration of untreated psychosis: a comprehensive review. Mol Psychiatry 28(1):168–190. https://doi.org/10.1038/s41380-022-01718-0

    Article  PubMed  Google Scholar 

  72. Laan W, Smeets H, de Wit NJ, Kahn RS, Grobbee DE, Burger H (2009) Glucocorticosteroids associated with a decreased risk of psychosis. J Clin Psychopharmacol 29(3):288–290

    Article  PubMed  CAS  Google Scholar 

  73. Cryan JF, Dinan TG (2012) Mind-altering microorganisms: the impact of the gut microbiota on brain and behaviour. Nat Rev Neurosci 13(10):701–712

    Article  PubMed  CAS  Google Scholar 

  74. Goldsmith CAW, Rogers DP (2008) The case for autoimmunity in the etiology of schizophrenia. Pharmacotherapy: J Hum Pharmacol Drug Ther 28(6):730–741

    Article  CAS  Google Scholar 

  75. Benros ME, Nielsen PR, Nordentoft M, Eaton WW, Dalton SO, Mortensen PB (2011) Autoimmune diseases and severe infections as risk factors for schizophrenia: a 30-year population-based register study. Am J Psychiatr 168(12):1303–1310

    Article  PubMed  Google Scholar 

  76. Ahmed GK, Elbeh K, Shams RM, Malek MAA, Ibrahim AK (2021) Prevalence and predictors of postpartum depression in Upper Egypt: A multicenter primary health care study. J Affect Disord 290:211–218. https://doi.org/10.1016/j.jad.2021.04.046

    Article  PubMed  Google Scholar 

  77. Khedr EM, Ramadan ES, Osman MN, Ahmed GK (2023) Risk factors-related first episode postpartum psychosis among Egyptian women: the role of psychosocial and the biological factors. Egypt J Neurol Psychiatry Neurosurg 59(1):51. https://doi.org/10.1186/s41983-023-00653-3

    Article  Google Scholar 

  78. Hassaan SH, Darwish AM, Khalifa H, Ramadan HKA, Hassany SM, Ahmed GK et al (2019) Assessment of cognitive functions and psychiatric symptoms in hepatitis C patients receiving pegylated interferon alpha and ribavirin: A prospective cohort study. Int J Psychiatry Med 54(6):424–440. https://doi.org/10.1177/0091217419858277

    Article  PubMed  Google Scholar 

  79. Shehata GA, Ahmed GK, Hassan EA, Rehim ASE-DA, Mahmoud SZ, Masoud NA et al (2022) Impact of direct-acting antivirals on neuropsychiatric and neurocognitive dysfunction in chronic hepatitis C patients. Egypt J Neurol Psychiatry Neurosurg 58(1):143. https://doi.org/10.1186/s41983-022-00568-5

    Article  Google Scholar 

  80. Mahran ZG, Khalifa H, Makhlouf NA, Mostafa DK, Aboalam HS, Moustafa EF et al (2022) Effect of gender difference on psychiatric outcomes for hepatitis C virus patients receiving direct-acting antivirals in Egyptian population: a cohort study. Egypt J Neurol Psychiatry Neurosurg 58(1):155. https://doi.org/10.1186/s41983-022-00585-4

    Article  Google Scholar 

  81. Jézéquel J, Rogemond V, Pollak T, Lepleux M, Jacobson L, Gréa H et al (2017) Cell- and Single Molecule-Based Methods to Detect Anti-N-Methyl-D-Aspartate Receptor Autoantibodies in Patients With First-Episode Psychosis From the OPTiMiSE Project. Biol Psychiatry 82(10):766–772. https://doi.org/10.1016/j.biopsych.2017.06.015

    Article  PubMed  CAS  Google Scholar 

  82. Jézéquel J, Johansson EM, Dupuis JP, Rogemond V, Gréa H, Kellermayer B et al (2017) Dynamic disorganization of synaptic NMDA receptors triggered by autoantibodies from psychotic patients. Nat Commun 8(1):1791. https://doi.org/10.1038/s41467-017-01700-3

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  83. Bost C, Pascual O, Honnorat J (2016) Autoimmune encephalitis in psychiatric institutions: current perspectives. Neuropsychiatr Dis Treat 2775-87

  84. Dalmau J, Gleichman AJ, Hughes EG, Rossi JE, Peng X, Lai M et al (2008) Anti-NMDA-receptor encephalitis: case series and analysis of the effects of antibodies. Lancet Neurol 7(12):1091–1098. https://doi.org/10.1016/S1474-4422(08)70224-2

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  85. Abdel Aziz K, Stip E, Arnone D (2021) More than just anti-NMDAR: the many facets of autoimmune encephalitis. BJPsych Bull 46(4):1–5. https://doi.org/10.1192/bjb.2021.113

    Article  PubMed  Google Scholar 

  86. Abdel Aziz K, AlSuwaidi A, Al-Ammari A, Al Khoori A, AlBloushi A, Al-Nuaimi N et al (2021) When should psychiatrists think of anti-NMDA receptor encephalitis? A systematic approach in clinical reasoning. Asian J Psychiatr 56:102524. https://doi.org/10.1016/j.ajp.2020.102524

    Article  PubMed  Google Scholar 

  87. Coutinho E, Menassa DA, Jacobson L, West SJ, Domingos J, Moloney TC et al (2017) Persistent microglial activation and synaptic loss with behavioral abnormalities in mouse offspring exposed to CASPR2-antibodies in utero. Acta Neuropathol 134(4):567–583. https://doi.org/10.1007/s00401-017-1751-5

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  88. Goverman J (2009) Autoimmune T cell responses in the central nervous system. Nat Rev Immunol 9(6):393–407

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  89. Muller N, Schwarz J, M. (2010) The role of immune system in schizophrenia. Curr Immunol Rev 6(3):213–220

    Article  PubMed  PubMed Central  Google Scholar 

  90. Bechter K (2013) Updating the mild encephalitis hypothesis of schizophrenia. Prog Neuro-Psychopharmacol Biol Psychiatry 42:71–91

    Article  CAS  Google Scholar 

  91. Hickie IB, Banati R, Stewart CH, Lloyd AR (2009) Are common childhood or adolescent infections risk factors for schizophrenia and other psychotic disorders? Med J Aust 190(S4):S17–S21

    Article  PubMed  Google Scholar 

  92. Benros ME, Pedersen MG, Rasmussen H, Eaton WW, Nordentoft M, Mortensen PB (2014) A nationwide study on the risk of autoimmune diseases in individuals with a personal or a family history of schizophrenia and related psychosis. Am J Psychiatr 171(2):218–226

    Article  PubMed  Google Scholar 

  93. Rose NR (1998) The role of infection in the pathogenesis of autoimmune disease. Semin Immunol 10(1):5–13. https://doi.org/10.1006/smim.1997.0100

    Article  PubMed  CAS  Google Scholar 

  94. Müller N, Riedel M, Ackenheil M, Schwarz MJ (1999) The role of immune function in schizophrenia: an overview. Eur Arch Psychiatry Clin Neurosci 249(Suppl 4):62–68. https://doi.org/10.1007/pl00014187

    Article  PubMed  Google Scholar 

  95. Mundo E, Altamura AC, Vismara S, Zanardini R, Bignotti S, Randazzo R et al (2005) MCP-1 gene (SCYA2) and schizophrenia: a case-control association study. Am J Med Genet B Neuropsychiatr Genet 132b(1):1–4. https://doi.org/10.1002/ajmg.b.30100

    Article  PubMed  Google Scholar 

  96. Stober G, Ben-Shachar D, Cardon M, Falkai P, Fonteh AN, Gawlik M et al (2009) Schizophrenia: from the brain to peripheral markers. A consensus paper of the WFSBP task force on biological markers. World J Biol Psychiatry 10(2):127–155. https://doi.org/10.1080/15622970902898980

    Article  PubMed  Google Scholar 

  97. Graeber MB (2014) Neuroinflammation: no rose by any other name. Brain Pathol 24(6):620

    Article  PubMed  PubMed Central  Google Scholar 

  98. Masgrau R, Guaza C, Ransohoff RM, Galea E (2017) Should we stop saying ‘glia’and ‘neuroinflammation’? Trends Mol Med 23(6):486–500

    Article  PubMed  CAS  Google Scholar 

  99. Schwartz M, Baruch K (2014) The resolution of neuroinflammation in neurodegeneration: leukocyte recruitment via the choroid plexus. EMBO J 33(1):7–22

    Article  PubMed  CAS  Google Scholar 

  100. Estes ML, McAllister AK (2014) Alterations in immune cells and mediators in the brain: it’s not always neuroinflammation! Brain Pathol 24(6):623–630

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  101. Helmut K, Hanisch UK, Noda M, Verkhratsky A (2011) Physiology of microglia. Physiol Rev 91(2):461–553

    Article  Google Scholar 

  102. Ginhoux F, Greter M, Leboeuf M, Nandi S, See P, Gokhan S et al (2010) Fate mapping analysis reveals that adult microglia derive from primitive macrophages. Science. 330(6005):841–845. https://doi.org/10.1126/science.1194637

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  103. Daneman R, Zhou L, Kebede AA, Barres BA (2010) Pericytes are required for blood-brain barrier integrity during embryogenesis. Nature. 468(7323):562–566. https://doi.org/10.1038/nature09513

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  104. Schafer DP, Lehrman EK, Kautzman AG, Koyama R, Mardinly AR, Yamasaki R et al (2012) Microglia sculpt postnatal neural circuits in an activity and complement-dependent manner. Neuron. 74(4):691–705

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  105. Sierra A, Beccari S, Diaz-Aparicio I, Encinas JM, Comeau S, Tremblay M-È (2014) Surveillance, phagocytosis, and inflammation: how never-resting microglia influence adult hippocampal neurogenesis. Neural Plast 2014

  106. Sierra A, Encinas JM, Deudero JJ, Chancey JH, Enikolopov G, Overstreet-Wadiche LS et al (2010) Microglia shape adult hippocampal neurogenesis through apoptosis-coupled phagocytosis. Cell Stem Cell 7(4):483–495

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  107. Chen Z, Palmer TD (2013) Differential roles of TNFR1 and TNFR2 signaling in adult hippocampal neurogenesis. Brain Behav Immun 30:45–53. https://doi.org/10.1016/j.bbi.2013.01.083

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  108. Liu H, Leak RK, Hu X (2016) Neurotransmitter receptors on microglia. Stroke Vasc Neurol 1(2)

  109. Krabbe G, Matyash V, Pannasch U, Mamer L, Boddeke HW, Kettenmann H (2012) Activation of serotonin receptors promotes microglial injury-induced motility but attenuates phagocytic activity. Brain Behav Immun 26(3):419–428

    Article  PubMed  CAS  Google Scholar 

  110. Parkin J, Cohen B (2001) An overview of the immune system. Lancet 357(9270):1777–1789

    Article  PubMed  CAS  Google Scholar 

  111. Khandaker GM, Dantzer R (2016) Is there a role for immune-to-brain communication in schizophrenia? Psychopharmacology 233:1559–1573

    Article  PubMed  CAS  Google Scholar 

  112. Drzyzga Ł, Obuchowicz E, Marcinowska A, Herman ZS (2006) Cytokines in schizophrenia and the effects of antipsychotic drugs. Brain Behav Immun 20(6):532–545

    Article  PubMed  CAS  Google Scholar 

  113. Røge R, Møller BK, Andersen CR, Correll CU, Nielsen J (2012) Immunomodulatory effects of clozapine and their clinical implications: what have we learned so far? Schizophr Res 140(1–3):204–213

    Article  PubMed  Google Scholar 

  114. Löffler S, Löffler-Ensgraber M, Fehsel K, Klimke A (2010) Clozapine therapy raises serum concentrations of high sensitive C-reactive protein in schizophrenic patients. Int Clin Psychopharmacol 25(2):101–106

    Article  PubMed  Google Scholar 

  115. Kluge M, Schuld A, Schacht A, Himmerich H, Dalal MA, Wehmeier PM et al (2009) Effects of clozapine and olanzapine on cytokine systems are closely linked to weight gain and drug-induced fever. Psychoneuroendocrinology. 34(1):118–128

    Article  PubMed  CAS  Google Scholar 

  116. Noto C, Ota VK, Santoro ML, Ortiz BB, Rizzo LB, Higuchi CH et al (2015) Effects of depression on the cytokine profile in drug naive first-episode psychosis. Schizophr Res 164(1–3):53–58

    Article  PubMed  Google Scholar 

  117. Miller BJ, Buckley P, Seabolt W, Mellor A, Kirkpatrick B (2011) Meta-analysis of cytokine alterations in schizophrenia: clinical status and antipsychotic effects. Biol Psychiatry 70(7):663–671

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  118. Khandaker GM, Pearson RM, Zammit S, Lewis G, Jones PB (2014) Association of serum interleukin 6 and C-reactive protein in childhood with depression and psychosis in young adult life: a population-based longitudinal study. JAMA Psychiatry 71(10):1121–1128

    Article  PubMed  PubMed Central  Google Scholar 

  119. Baumeister D, Akhtar R, Ciufolini S, Pariante CM, Mondelli V (2016) Childhood trauma and adulthood inflammation: a meta-analysis of peripheral C-reactive protein, interleukin-6 and tumour necrosis factor-α. Mol Psychiatry 21(5):642–649

    Article  PubMed  CAS  Google Scholar 

  120. Mondelli V, Ciufolini S, Belvederi Murri M, Bonaccorso S, Di Forti M, Giordano A et al (2015) Cortisol and inflammatory biomarkers predict poor treatment response in first episode psychosis. Schizophr Bull 41(5):1162–1170

    Article  PubMed  PubMed Central  Google Scholar 

  121. Johnsen E, Fathian F, Kroken RA, Steen VM, Jørgensen HA, Gjestad R et al (2016) The serum level of C-reactive protein (CRP) is associated with cognitive performance in acute phase psychosis. BMC Psychiatry 16(1):1–11

    Article  Google Scholar 

  122. Miller BJ, Culpepper N, Rapaport MH (2014) C-reactive protein levels in schizophrenia: a review and meta-analysis. Clin Schizophr Relat Psychoses 7(4):223–230. https://doi.org/10.3371/csrp.micu.020813

    Article  PubMed  Google Scholar 

  123. Dickerson F, Stallings C, Origoni A, Vaughan C, Khushalani S, Yang S et al (2013) C-reactive protein is elevated in schizophrenia. Schizophr Res 143(1):198–202. https://doi.org/10.1016/j.schres.2012.10.041

    Article  PubMed  Google Scholar 

  124. Dickerson F, Stallings C, Origoni A, Boronow J, Yolken R (2007) C-reactive protein is associated with the severity of cognitive impairment but not of psychiatric symptoms in individuals with schizophrenia. Schizophr Res 93(1–3):261–265. https://doi.org/10.1016/j.schres.2007.03.022

    Article  PubMed  Google Scholar 

  125. Wang AK, Miller BJ (2018) Meta-analysis of cerebrospinal fluid cytokine and tryptophan catabolite alterations in psychiatric patients: comparisons between schizophrenia, bipolar disorder, and depression. Schizophr Bull 44(1):75–83. https://doi.org/10.1093/schbul/sbx035

    Article  PubMed  Google Scholar 

  126. Coughlin JM, Wang Y, Ambinder EB, Ward RE, Minn I, Vranesic M et al (2016) In vivo markers of inflammatory response in recent-onset schizophrenia: a combined study using [11C]DPA-713 PET and analysis of CSF and plasma. Translational. Psychiatry. 6(4):e777-e. https://doi.org/10.1038/tp.2016.40

    Article  CAS  Google Scholar 

  127. Flatow J, Buckley P, Miller BJ (2013) Meta-analysis of oxidative stress in schizophrenia. Biol Psychiatry 74(6):400–409. https://doi.org/10.1016/j.biopsych.2013.03.018

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  128. Wang L-J, Lin P-Y, Lee Y, Huang Y-C, Wu C-C, Hsu S-T et al (2018) Increased serum levels of cysteine in patients with schizophrenia: a potential marker of cognitive function preservation. Schizophr Res 192:391–397. https://doi.org/10.1016/j.schres.2017.03.041

    Article  PubMed  Google Scholar 

  129. Traynelis SF, Wollmuth LP, McBain CJ, Menniti FS, Vance KM, Ogden KK et al (2010) Glutamate receptor ion channels: structure, regulation, and function. Pharmacol Rev 62(3):405–496. https://doi.org/10.1124/pr.109.002451

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  130. Fraguas D, Gonzalez-Pinto A, Micó JA, Reig S, Parellada M, Martínez-Cengotitabengoa M et al (2012) Decreased glutathione levels predict loss of brain volume in children and adolescents with first-episode psychosis in a two-year longitudinal study. Schizophr Res 137(1):58–65. https://doi.org/10.1016/j.schres.2012.01.040

    Article  PubMed  Google Scholar 

  131. Jiménez-Fernández S, Gurpegui M, Díaz-Atienza F, Pérez-Costillas L, Gerstenberg M, Correll CU (2015) Oxidative stress and antioxidant parameters in patients with major depressive disorder compared to healthy controls before and after antidepressant treatment: results from a meta-analysis. J Clin Psychiatry 76(12):1658–1667. https://doi.org/10.4088/JCP.14r09179

    Article  PubMed  Google Scholar 

  132. Raffa M, Atig F, Mhalla A, Kerkeni A, Mechri A (2011) Decreased glutathione levels and impaired antioxidant enzyme activities in drug-naive first-episode schizophrenic patients. BMC Psychiatry 11:124. https://doi.org/10.1186/1471-244x-11-124

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  133. Wood SJ, Yücel M, Pantelis C, Berk M (2009) Neurobiology of schizophrenia spectrum disorders: the role of oxidative stress. Ann Acad Med Singap 38(5):396–396

    Article  PubMed  Google Scholar 

  134. Marsman A, van den Heuvel MP, Klomp DW, Kahn RS, Luijten PR, Hulshoff Pol HE (2013) Glutamate in schizophrenia: a focused review and meta-analysis of 1H-MRS studies. Schizophr Bull 39(1):120–129. https://doi.org/10.1093/schbul/sbr069

    Article  PubMed  Google Scholar 

  135. Khandaker GM, Dantzer R, Jones PB (2017) Immunopsychiatry: important facts. Psychol Med 47(13):2229–2237. https://doi.org/10.1017/s0033291717000745

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  136. McGinty J, Sayeed Haque M, Upthegrove R (2018) Depression during first episode psychosis and subsequent suicide risk: a systematic review and meta-analysis of longitudinal studies. Schizophr Res 195:58–66. https://doi.org/10.1016/j.schres.2017.09.040

    Article  PubMed  Google Scholar 

  137. Fernandes BS, Steiner J, Bernstein HG, Dodd S, Pasco JA, Dean OM et al (2016) C-reactive protein is increased in schizophrenia but is not altered by antipsychotics: meta-analysis and implications. Mol Psychiatry 21(4):554-64. https://doi.org/10.1038/mp.2015.87

  138. Goldsmith DR, Rapaport MH, Miller BJ (2016) A meta-analysis of blood cytokine network alterations in psychiatric patients: comparisons between schizophrenia, bipolar disorder and depression. Mol Psychiatry 21(12):1696-709. https://doi.org/10.1038/mp.2016.3

  139. Severance EG, Alaedini A, Yang S, Halling M, Gressitt KL, Stallings CR et al (2012) Gastrointestinal inflammation and associated immune activation in schizophrenia. Schizophr Res 138(1):48–53. https://doi.org/10.1016/j.schres.2012.02.025

    Article  PubMed  PubMed Central  Google Scholar 

  140. Oshitani N, Hato F, Matsumoto T, Jinno Y, Sawa Y, Hara J et al (2000) Decreased anti-Saccharomyces cerevisiae antibody titer by mesalazine in patients with Crohn’s disease. J Gastroenterol Hepatol 15(12):1400–1403. https://doi.org/10.1046/j.1440-1746.2000.02357.x

    Article  PubMed  CAS  Google Scholar 

  141. Mallant-Hent R, Mary B, von Blomberg E, Yüksel Z, Wahab PJ, Gundy C et al (2006) Disappearance of anti-Saccharomyces cerevisiae antibodies in coeliac disease during a gluten-free diet. Eur J Gastroenterol Hepatol 18(1):75–78. https://doi.org/10.1097/00042737-200601000-00013

    Article  PubMed  CAS  Google Scholar 

  142. Kotze LMS, Nisihara RM, Utiyama SRR, Kotze PG, Theiss PM, Olandoski M (2010) Antibodies anti-Saccharomyces cerevisiae (ASCA) do not differentiate Crohn’s disease from celiac disease. Arq Gastroenterol 47(3):242–245

    Article  PubMed  Google Scholar 

  143. Desplat-Jégo S, Johanet C, Escande A, Goetz J, Fabien N, Olsson N et al (2007) Update on Anti-Saccharomyces cerevisiae antibodies, anti-nuclear associated anti-neutrophil antibodies and antibodies to exocrine pancreas detected by indirect immunofluorescence as biomarkers in chronic inflammatory bowel diseases: results of a multicenter study. World J Gastroenterol 13(16):2312–2318. https://doi.org/10.3748/wjg.v13.i16.2312

    Article  PubMed  PubMed Central  Google Scholar 

  144. Ashorn S, Välineva T, Kaukinen K, Ashorn M, Braun J, Raukola H et al (2009) Serological responses to microbial antigens in celiac disease patients during a gluten-free diet. J Clin Immunol 29(2):190–195. https://doi.org/10.1007/s10875-008-9255-7

    Article  PubMed  Google Scholar 

  145. Khedr EM, Omeran N, Karam-Allah Ramadan H, Ahmed GK, Abdelwarith AM (2022) Alteration of gut microbiota in Alzheimer’s disease and their relation to the cognitive impairment. J Alzheimers Dis 88:1103–1114. https://doi.org/10.3233/JAD-220176

    Article  PubMed  CAS  Google Scholar 

  146. Severance EG, Gressitt KL, Stallings CR, Origoni AE, Khushalani S, Leweke FM et al (2013) Discordant patterns of bacterial translocation markers and implications for innate immune imbalances in schizophrenia. Schizophr Res 148(1–3):130–137. https://doi.org/10.1016/j.schres.2013.05.018

    Article  PubMed  PubMed Central  Google Scholar 

  147. Sandhya P, Danda D, Sharma D, Scaria V (2016) Does the buck stop with the bugs?: an overview of microbial dysbiosis in rheumatoid arthritis. Int J Rheum Dis 19(1):8–20. https://doi.org/10.1111/1756-185x.12728

    Article  PubMed  Google Scholar 

  148. Kannan G, Gressitt KL, Yang S, Stallings CR, Katsafanas E, Schweinfurth LA et al (2017) Pathogen-mediated NMDA receptor autoimmunity and cellular barrier dysfunction in schizophrenia. Transl Psychiatry 7(8):e1186. https://doi.org/10.1038/tp.2017.162

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  149. D’Mello C, Swain MG (2014) Liver–brain interactions in inflammatory liver diseases: Implications for fatigue and mood disorders. Brain Behav Immun 35:9–20. https://doi.org/10.1016/j.bbi.2013.10.009

    Article  PubMed  CAS  Google Scholar 

  150. Braniste V, Al-Asmakh M, Kowal C, Anuar F, Abbaspour A, Tóth M et al (2014) The gut microbiota influences blood-brain barrier permeability in mice. Sci Transl Med 6(263):263ra158. https://doi.org/10.1126/scitranslmed.3009759

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  151. Castro-Nallar E, Bendall ML, Pérez-Losada M, Sabuncyan S, Severance EG, Dickerson FB et al (2015) Composition, taxonomy and functional diversity of the oropharynx microbiome in individuals with schizophrenia and controls. PeerJ 3:e1140. https://doi.org/10.7717/peerj.1140

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  152. Kim J, Sudbery P (2011) Candida albicans, a major human fungal pathogen. J Microbiol (Seoul, Korea) 49(2):171–177. https://doi.org/10.1007/s12275-011-1064-7

    Article  Google Scholar 

  153. Steiner J, Jacobs R, Panteli B, Brauner M, Schiltz K, Bahn S et al (2010) Acute schizophrenia is accompanied by reduced T cell and increased B cell immunity. Eur Arch Psychiatry Clin Neurosci 260(7):509–518. https://doi.org/10.1007/s00406-010-0098-x

    Article  PubMed  Google Scholar 

  154. van der Kemp WJ, Klomp DW, Kahn RS, Luijten PR, Hulshoff Pol HE (2012) A meta-analysis of the polyunsaturated fatty acid composition of erythrocyte membranes in schizophrenia. Schizophr Res 141(2–3):153–161. https://doi.org/10.1016/j.schres.2012.08.014

    Article  PubMed  Google Scholar 

  155. Yao JK, van Kammen DP (2004) Membrane phospholipids and cytokine interaction in schizophrenia. Int Rev Neurobiol 59:297–326. https://doi.org/10.1016/S0074-7742(04)59012-8

    Article  PubMed  CAS  Google Scholar 

  156. Yao JK, Keshavan MS (2011) Antioxidants, redox signaling, and pathophysiology in schizophrenia: an integrative view. Antioxid Redox Signal 15(7):2011–2035. https://doi.org/10.1089/ars.2010.3603

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  157. Pae CU, Paik IH, Lee C, Lee SJ, Kim JJ, Lee CU (2004) Decreased plasma antioxidants in schizophrenia. Neuropsychobiology 50(1):54–56. https://doi.org/10.1159/000077942

    Article  PubMed  CAS  Google Scholar 

  158. Vítek L, Novotná M, Lenícek M, Novotný L, Eberová J, Petrásek J et al (2010) Serum bilirubin levels and UGT1A1 promoter variations in patients with schizophrenia. Psychiatry Res 178(2):449–450. https://doi.org/10.1016/j.psychres.2009.12.008

    Article  PubMed  CAS  Google Scholar 

  159. Yao JK, Reddy R, van Kammen DP (1998) Reduced level of plasma antioxidant uric acid in schizophrenia. Psychiatry Res 80(1):29–39. https://doi.org/10.1016/s0165-1781(98)00051-1

    Article  PubMed  CAS  Google Scholar 

  160. Suboticanec K, Folnegović-Smalc V, Korbar M, Mestrović B, Buzina R (1990) Vitamin C status in chronic schizophrenia. Biol Psychiatry 28(11):959–966. https://doi.org/10.1016/0006-3223(90)90061-6

    Article  PubMed  CAS  Google Scholar 

  161. Raffa M, Mechri A, Othman LB, Fendri C, Gaha L, Kerkeni A (2009) Decreased glutathione levels and antioxidant enzyme activities in untreated and treated schizophrenic patients. Prog Neuro-Psychopharmacol Biol Psychiatry 33(7):1178–1183. https://doi.org/10.1016/j.pnpbp.2009.06.018

    Article  CAS  Google Scholar 

  162. Brown K, Reid A, White T, Henderson T, Hukin S, Johnstone C et al (1998) Vitamin E, lipids, and lipid peroxidation products in tardive dyskinesia. Biol Psychiatry 43(12):863–867. https://doi.org/10.1016/s0006-3223(97)00197-2

    Article  PubMed  CAS  Google Scholar 

  163. Minutolo G, Petralia A, Dipasquale S, Aguglia E (2012) Nitric oxide in patients with schizophrenia: the relationship with the severity of illness and the antipsychotic treatment. Expert Opin Pharmacother 13(14):1989–1997. https://doi.org/10.1517/14656566.2012.713346

    Article  PubMed  CAS  Google Scholar 

  164. Zhang XY, Zhou DF, Shen YC, Zhang PY, Zhang WF, Liang J et al (2012) Effects of risperidone and haloperidol on superoxide dismutase and nitric oxide in schizophrenia. Neuropharmacology 62(5–6):1928–1934. https://doi.org/10.1016/j.neuropharm.2011.12.014

    Article  PubMed  CAS  Google Scholar 

  165. Myers R, Manjil LG, Cullen BM, Price GW, Frackowiak RS, Cremer JE (1991) Macrophage and astrocyte populations in relation to [3H]PK 11195 binding in rat cerebral cortex following a local ischaemic lesion. J Cereb Blood Flow Metab 11(2):314–322. https://doi.org/10.1038/jcbfm.1991.64

    Article  PubMed  CAS  Google Scholar 

  166. Benavides J, Cornu P, Dennis T, Dubois A, Hauw JJ, MacKenzie ET et al (1988) Imaging of human brain lesions with an omega 3 site radioligand. Ann Neurol 24(6):708–712. https://doi.org/10.1002/ana.410240603

    Article  PubMed  CAS  Google Scholar 

  167. Mondelli V, Cattaneo A, Murri MB, Di Forti M, Handley R, Hepgul N et al (2011) Stress and inflammation reduce brain-derived neurotrophic factor expression in first-episode psychosis: a pathway to smaller hippocampal volume. J Clin Psychiatry 72(12):1677–1684. https://doi.org/10.4088/JCP.10m06745

    Article  PubMed  PubMed Central  Google Scholar 

  168. Marsland AL, Gianaros PJ, Abramowitch SM, Manuck SB, Hariri AR (2008) Interleukin-6 covaries inversely with hippocampal grey matter volume in middle-aged adults. Biol Psychiatry 64(6):484–490. https://doi.org/10.1016/j.biopsych.2008.04.016

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  169. Schnieder TP, Dwork AJ (2011) Searching for neuropathology: gliosis in schizophrenia. Biol Psychiatry 69(2):134–139. https://doi.org/10.1016/j.biopsych.2010.08.027

    Article  PubMed  Google Scholar 

  170. Bayer TA, Buslei R, Havas L, Falkai P (1999) Evidence for activation of microglia in patients with psychiatric illnesses. Neurosci Lett 271(2):126–128. https://doi.org/10.1016/s0304-3940(99)00545-5

    Article  PubMed  CAS  Google Scholar 

  171. Radewicz K, Garey LJ, Gentleman SM, Reynolds R (2000) Increase in HLA-DR immunoreactive microglia in frontal and temporal cortex of chronic schizophrenics. J Neuropathol Exp Neurol 59(2):137–150. https://doi.org/10.1093/jnen/59.2.137

    Article  PubMed  CAS  Google Scholar 

  172. Steiner J, Mawrin C, Ziegeler A, Bielau H, Ullrich O, Bernstein HG et al (2006) Distribution of HLA-DR-positive microglia in schizophrenia reflects impaired cerebral lateralization. Acta Neuropathol 112(3):305–316. https://doi.org/10.1007/s00401-006-0090-8

    Article  PubMed  CAS  Google Scholar 

  173. Nitta M, Kishimoto T, Müller N, Weiser M, Davidson M, Kane JM et al (2013) Adjunctive use of nonsteroidal anti-inflammatory drugs for schizophrenia: a meta-analytic investigation of randomized controlled trials. Schizophr Bull 39(6):1230–1241. https://doi.org/10.1093/schbul/sbt070

    Article  PubMed  PubMed Central  Google Scholar 

  174. Sommer IE, van Westrhenen R, Begemann MJ, de Witte LD, Leucht S, Kahn RS (2014) Efficacy of anti-inflammatory agents to improve symptoms in patients with schizophrenia: an update. Schizophr Bull 40(1):181–191. https://doi.org/10.1093/schbul/sbt139

    Article  PubMed  Google Scholar 

  175. Arroll MA, Wilder L, Neil J (2014) Nutritional interventions for the adjunctive treatment of schizophrenia: a brief review. Nutr J 13:91. https://doi.org/10.1186/1475-2891-13-91

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  176. McGorry PD, Nelson B, Markulev C, Yuen HP, Schäfer MR, Mossaheb N et al (2017) Effect of ω-3 polyunsaturated fatty acids in young people at ultrahigh risk for psychotic disorders: the NEURAPRO randomized clinical trial. JAMA Psychiatry 74(1):19–27. https://doi.org/10.1001/jamapsychiatry.2016.2902

    Article  PubMed  Google Scholar 

  177. Joy CB, Mumby-Croft R, Joy LA (2006) Polyunsaturated fatty acid supplementation for schizophrenia. Cochrane Database Syst Rev 2006(3):Cd001257. https://doi.org/10.1002/14651858.CD001257.pub2

    Article  PubMed  PubMed Central  Google Scholar 

  178. Girgis RR, Ciarleglio A, Choo T, Haynes G, Bathon JM, Cremers S et al (2018) A Randomized, Double-Blind, Placebo-Controlled Clinical Trial of Tocilizumab, An Interleukin-6 Receptor Antibody, For Residual Symptoms in Schizophrenia. Neuropsychopharmacology 43(6):1317-23. https://doi.org/10.1038/npp.2017.258

  179. Miller BJ, Buckley PF (2016) The Case for Adjunctive Monoclonal Antibody Immunotherapy in Schizophrenia. The Psychiatric clinics of North America 39(2):187-98

  180. Rolland B, Deguil J, Jardri R, Cottencin O, Thomas P, Bordet R (2013) Therapeutic prospects of PPARs in psychiatric disorders: a comprehensive review. Curr Drug Targets 14(7):724-32. https://doi.org/10.2174/1389450111314070002

  181. Cox D, Chan MK, Bahn S (2015) The potential of immune biomarkers to advance personalized medicine approaches for schizophrenia. The Journal of nervous and mental disease 203(5):393-9. https://doi.org/10.1097/nmd.0000000000000289

  182. Dickerson FB, Stallings C, Origoni A, Katsafanas E, Savage CL, Schweinfurth LA et al (2014) Effect of probiotic supplementation on schizophrenia symptoms and association with gastrointestinal functioning: a randomized, placebo-controlled trial. Prim Care Companion CNS Disord 16(1). https://doi.org/10.4088/PCC.13m01579

  183. Tomasik J, Yolken RH, Bahn S, Dickerson FB (2015) Immunomodulatory Effects of Probiotic Supplementation in Schizophrenia Patients: A Randomized, Placebo-Controlled Trial. Biomark Insights 10:47-54. https://doi.org/10.4137/bmi.s22007

  184. Pandurangi AK, Buckley PF (2020) Inflammation, Antipsychotic Drugs, and Evidence for Effectiveness of Anti-inflammatory Agents in Schizophrenia. In: Khandaker GM, Meyer U, Jones PB (eds). Neuroinflammation and Schizophrenia. Springer International Publishing, Cham, p 227-44

  185. Kane JM, Correll CU (2010) Past and present progress in the pharmacologic treatment of schizophrenia. J Clin Psychiatry 71(9):1115–1124. https://doi.org/10.4088/JCP.10r06264yel

    Article  PubMed  PubMed Central  Google Scholar 

  186. Stip E, Rizvi TA, Mustafa F, Javaid S, Aburuz S, Ahmed NN et al (2020) The large action of chlorpromazine: translational and transdisciplinary considerations in the face of COVID-19. Front Pharmacol 11:577678

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  187. Stip E, Arnone D, Abdel Aziz K, Javaid SF (2021) Diversity of mechanism of action of psychotropic drugs in their anti-COVID-19 properties. Mol Psychiatry 26(12):7093–7097. https://doi.org/10.1038/s41380-021-01222-x

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  188. Bumb JM, Enning F, Leweke FM (2015) Drug repurposing and emerging adjunctive treatments for schizophrenia. Expert Opin Pharmacother 16(7):1049-67. https://doi.org/10.1517/14656566.2015.1032248

  189. Chan MK, Guest PC, Levin Y, Umrania Y, Schwarz E, Bahn S et al (2011) Converging evidence of blood-based biomarkers for schizophrenia: an update. International review of neurobiology 101:95-144. https://doi.org/10.1016/b978-0-12-387718-5.00005-5

  190. Chen ML, Tsai TC, Wang LK, Lin YY, Tsai YM, Lee MC et al (2012) Risperidone modulates the cytokine and chemokine release of dendritic cells and induces TNF-α-directed cell apoptosis in neutrophils. Int Immunopharmacol 12(1):197-204. https://doi.org/10.1016/j.intimp.2011.11.011

  191. de Witte L, Tomasik J, Schwarz E, Guest PC, Rahmoune H, Kahn RS et al (2014) Cytokine alterations in first-episode schizophrenia patients before and after antipsychotic treatment. Schizophr Res 154(1-3):23-9. https://doi.org/10.1016/j.schres.2014.02.005

  192. Debnath M, Venkatasubramanian G (2013) Recent advances in psychoneuroimmunology relevant to schizophrenia therapeutics. Current Opinion in Psychiatry 26(5)

  193. Diaz FJ, Pérez-Iglesias R, Mata I, Martínez-Garcia O, Vázquez-Barquero JL, de Leon J et al (2010) Possible effects of some antipsychotic drugs on C-reactive protein in a drug-naïve psychotic sample. Schizophr Res 121(1-3):207-12. https://doi.org/10.1016/j.schres.2010.06.002

  194. Maes M, Delange J, Ranjan R, Meltzer HY, Desnyder R, Cooremans W et al (1997) Acute phase proteins in schizophrenia, mania and major depression: modulation by psychotropic drugs. Psychiatry Research 66(1):1-11

  195. Müller N, Myint AM, Krause D, Weidinger E, Schwarz MJ (2013) Anti-inflammatory treatment in schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry 42:146-53. https://doi.org/10.1016/j.pnpbp.2012.11.008

  196. Potvin S, Stip E, Sepehry AA, Gendron A, Bah R, Kouassi E (2008) Inflammatory cytokine alterations in schizophrenia: a systematic quantitative review. Biol Psychiatry 63(8):801-8. https://doi.org/10.1016/j.biopsych.2007.09.024

Download references

Acknowledgements

None

Funding

No sources of funding were required.

Author information

Authors and Affiliations

Authors

Contributions

GA conceived the original idea of this work and took the lead in writing the manuscript. HR and NE contributed equally to the literature review and writing of the manuscript. KE was responsible for revision. The final version of the manuscript was reviewed and approved by all co-authors.

Corresponding author

Correspondence to Gellan K. Ahmed.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ahmed, G.K., Ramadan, H.KA., Elbeh, K. et al. The role of infections and inflammation in schizophrenia: review of the evidence. Middle East Curr Psychiatry 31, 9 (2024). https://doi.org/10.1186/s43045-024-00397-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s43045-024-00397-7

Keywords