Next Article in Journal
Increased Actin Binding Is a Shared Molecular Consequence of Numerous SCA5 Mutations in β-III-Spectrin
Next Article in Special Issue
Concussion: Beyond the Cascade
Previous Article in Journal
Lacking Immunotherapy Biomarkers for Biliary Tract Cancer: A Comprehensive Systematic Literature Review and Meta-Analysis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Microglia and Other Cellular Mediators of Immunological Dysfunction in Schizophrenia: A Narrative Synthesis of Clinical Findings

1
Department of Microbiology and Immunology, Stanford University, Palo Alto, CA 94305, USA
2
Tranquis Therapeutics, Palo Alto, CA 94065, USA
3
Section of Psychiatry, Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, 16126 Genoa, Italy
4
IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
5
Department of Psychiatry, San Maurizio Hospital of Bolzano, 39100 Bolzano, Italy
6
Geriatric-Rehabilitation Department, University Hospital of Parma, 43126 Parma, Italy
7
Department of Psychiatry, Adult Psychiatry Service, University Hospitals of Geneva (HUG), 1207 Geneva, Switzerland
8
Department of Psychiatry, Faculty of Biomedical Sciences, University of Italian Switzerland (USI), 6900 Lugano, Switzerland
9
Department of Psychiatry, Faculty of Medicine, University of Geneva (UNIGE), 1211 Geneva, Switzerland
*
Author to whom correspondence should be addressed.
Cells 2023, 12(16), 2099; https://doi.org/10.3390/cells12162099
Submission received: 11 July 2023 / Revised: 13 August 2023 / Accepted: 17 August 2023 / Published: 19 August 2023

Abstract

:
Schizophrenia is a complex psychiatric condition that may involve immune system dysregulation. Since most putative disease mechanisms in schizophrenia have been derived from genetic association studies and fluid-based molecular analyses, this review aims to summarize the emerging evidence on clinical correlates to immune system dysfunction in this psychiatric disorder. We conclude this review by attempting to develop a unifying hypothesis regarding the relative contributions of microglia and various immune cell populations to the development of schizophrenia. This may provide important translational insights that can become useful for addressing the multifaceted clinical presentation of schizophrenia.

1. Introduction

Schizophrenia (SCZ), a chronic psychiatric illness that affects approximately 24 million people worldwide, is characterized by the hallmark “positive” symptoms of hallucinations and delusions and “negative” symptoms of apathy, anhedonia, avolition, and emotional and cognitive impoverishment [1]. This debilitating disorder imposes a significant risk of physical and mental health complications, ranging from coronary heart disease to suicidal behavior (SB), highlighting reciprocal relationships between somatic psychic implications in neuro-psychiatric conditions [2]. SCZ diagnosis is difficult due to the spectral nature of the illness and the complex progression of its clinical manifestation. Patients with SCZ often present with subtle irritation/behavioral changes in the prodromal phase, followed by the onset of psychosis. Before 2013, SCZ was categorized into various subtypes (paranoid, disorganized, catatonic, undifferentiated, and residual) based on specific clinical presentations. However, this discrete division of the illness was supplemented by the concept of SCZ being a spectral disease that includes schizoaffective, schizophreniform, and schizotypal personality disorders [3].
Various environmental and genetic risk factors have been implicated in the etiology of SCZ, with exposure to toxins/infectious agents, urban lifestyle, pregnancy complications, substance abuse, family history, and male sex reportedly associated with an increase in SCZ risk [4]. However, despite the discovery of many SCZ-associated genetic variants, to date, no single gene has been identified as the dominant causative factor of SCZ development. Currently, there are no effective treatments of SCZ as antipsychotics are only capable of suppressing positive symptoms [5]. Therefore, further research into the cellular and molecular pathology of SCZ is integral to the therapeutic development for this psychiatric disorder.
Consistent with the neuropsychiatric nature of SCZ, various central nervous system (CNS) abnormalities of affected individuals have been observed, including enlarged ventricles, reduced gray matter volume, smaller hippocampus, decreased brain asymmetry, and neurochemical disturbances [6]. Concurrently, accumulating evidence from neuroimmunological studies is pointing to the immune system’s potential involvement in SCZ development, an analogy to other major psychiatric conditions [7,8]. While several prominent hypotheses, including immunocytokine-driven inflammation [9], innate immune-mediated dysfunction in synaptic pruning [10], and antibody-mediated autoimmunity [11], have been suggested, these hypotheses were predominantly derived by extrapolating SCZ-like animal models and human genetic association studies/fluid-based molecular biomarker analyses. Since evidence on cellular abnormalities in SCZ remains scarce and somewhat scattered in the literature, this review aims to provide a comprehensive distillation of clinical findings regarding the potential involvement of various immune cell types in SCZ to facilitate the development of a unifying hypothesis of immune dysregulation in this psychiatric illness.

2. Cellular Constituents of CNS Immunological Aberrations in SCZ

Following reports of elevated neuroinflammation in SCZ, an initial hypothesis stated that CNS immune disturbance may be involved in SCZ pathogenesis. The subsequent discovery of risk factors in genes associated with immune-mediated neurodevelopmental processes and the identity of CNS immune cells provided further support for the immunological origin of SCZ (Figure 1). This section highlights important evidence of CNS immune-related cell-based changes in SCZ and provides a discussion of potential caveats in the interpretation of these findings.

2.1. Microglia

Microglia are the resident brain innate immune cells that have been implicated in host defense against neurotropic pathogens, brain development, and neurodegenerative disorders [12]. The growing importance of these cells in behavioral illnesses is also highlighted by the growing attention they have received in neuroimmunological investigations studying possible alterations in their distribution and function in SCZ. Given the difficulty of sampling live human microglia, cell characterization in SCZ has been mostly conducted in post mortem brain samples. One of the earliest studies of microglia in SCZ was an analysis of embryonic microglia derived from female patients with SCZ in whom these cells displayed a highly phagocytic phenotype compared to healthy controls (HCs) with no psychiatric illnesses [13]. Subsequently, morphologically activated microglia have been observed in the prefrontal cortex (PFC) and visual cortex of paranoid and chronic patients with SCZ in close proximity to dystrophic oligodendrocytes [14,15,16]. Further subcategorization of patients with SCZ revealed that this abnormal microglia activation phenotype might contribute to oligodendrocyte dystrophy in schizophrenia patients with positive symptoms [17,18], providing some of the first morphological evidence for the possible involvement of microglial activation in the development of these SCZ-associated pathologies.
In addition to these morphometric studies, others attempted to localize activation markers on microglia by immunohistochemical analysis and found increases in HLA-DR+ activated microglia in the frontal/temporal cortex and the hippocampus in patients with SCZ [19,20]. Of note, these activated microglia exhibited some degenerating features [21] and were reportedly associated with interleukin IL1β expression in the PFC [22]. Microglia activation was also observed in some of these brain regions in patients with Alzheimer’s disease (AD) and affective disorders [23,24], suggesting the possible existence of microglia reactivity against a common dysfunctional neuronal circuit among various CNS disorders. However, microglia activation in SCZ and affective disorders remains to be validated as some studies failed to detect changes in HLA-DR+ microglia in various brain regions, including PFC, anterior cingulate cortex (ACC), and hippocampus, and/or attribute this microglia activation profile to death by suicide [25,26]. Besides HLA-DR expression, a unique microglial proteome might exist in SCZ. For example, S100 calcium-binding protein (S100) A8/A9 expression [27], an inflammatory marker, was found to be upregulated in frontal cortex microglia, while quinolinic acid expression, a neuroprotective molecule, was suppressed in CA1 hippocampal microglia in patients with SCZ [28]. Furthermore, patterns of expression of the purinergic receptor (P2RY12) were not altered significantly in SCZ microglia compared to HCs, while downregulation of this marker was a cardinal feature of microglia in multiple sclerosis and AD [29].
Microgliosis, marked by increased ionized calcium-binding adapter molecule 1 (IBA-1) staining density [30], has also been linked to the characteristic anatomical lateralization in the ACC of patients with SCZ, as well as patients with bipolar disorder (BD). However, this abnormality was not observed in the PFC of SCZ patients [31], in which microglia showed synaptic pruning activity comparable to HC counterparts. This discrepancy may be attributed to region-specific alterations in microgliosis in SCZ as a significant increase in microgliosis was confirmed in the frontal and temporal cortex, but in no other regions [32]. Alternatively, age-dependent microgliosis may account for the discrepancy between the aforementioned studies [33,34].
To circumvent several methodological shortcomings of post mortem studies, several studies conducted microglia imaging in living patients with SCZ using translocator protein (TSPO) tracer analysis. TSPO is a receptor mainly found on the outer mitochondrial membrane that is expressed throughout the body and brain [35,36]. Among other functions, TSPO can modulate the immune system through modulation of oxidative bursts by neutrophils and macrophages, inhibition of the proliferation of lymphoid cells, and secretion of cytokines by macrophages [37]. Expression of TSPO has also been linked to inflammatory responses that occur after vascular brain injury and in some neurodegenerative or mixed neurodegenerative/vascular diseases, in which interesting links with neuropsychiatric disorders are currently being investigated [24,38]. There are significant inconsistencies among studies using TSPO tracer analysis to investigate possible alterations in the TSPO signal [39,40,41,42,43,44,45], and even studies conducting systematic analyses of single-patient data could not reach a consensus on changes in TSPO signal in SCZ brains [46,47]. This discrepancy may be due to the lack of specificity of TSPO to microglia, as various cell subsets, including astrocytes and endothelial cells, also showed this signal [48].
To date, cell-based evidence of the involvement of microglia in SCZ pathogenesis remains inconclusive. While we cannot discuss this in detail here, numerous association studies also suggested the presence of an increase in SCZ risk in various genes involved in microglia-mediated neuroinflammation [49] and synaptic pruning [50], as well as microglia homeostasis [51]. Of note, many of these molecules are not exclusively related to microglia function and identity, which highlights the urgent need for more accurate characterizations/confirmation of possible changes in various aspects of microglia in this psychiatric illness. Further studies with induced pluripotent stem-cell-derived microglia-like cells may shed some additional light on the role played by microglia genetics in SCZ. Additionally, the development of a rapid, efficient, and reliable method to isolate microglia from post mortem brain tissues for high-throughput proteomic and transcriptomic analyses is expected to provide additional clarification regarding the involvement of this innate immune cell type in SCZ.

2.2. Other Immune Cell Types

Besides microglia, abnormalities in other immune cells have also been detected in CNS samples of SCZ patients(). For example, dynamic trafficking of adaptive immune cells in the CNS has been linked to SCZ. Whole brain immunohistochemical quantitation of T cell and B cell frequencies showed marked increases in these lymphocytes in patients with SCZ and affective disorders compared to HCs [52]. Spatial analyses also revealed region-specific alterations of these lymphocytes in SCZ brain tissues. In this regard, immunohistochemical analysis in the dorsal PFC (DPFC) revealed a reduction in CD3+ T cell density in the leptomeningeal space of subjects with SCZ compared to HCs and no significant difference in the frequencies of these lymphocytes in the gray matter of both groups. Furthermore, analysis of hippocampal T cell (CD3) and B cell (CD20) mRNA transcripts revealed a significant increase in CD20 and CD3 expression in residual SCZ (characterized by negative symptoms) compared to paranoid SCZ (characterized by positive symptoms) and HCs, suggesting that adaptive immune cell trafficking to the hippocampus might be associated with negative symptoms of SCZ [19].
Monophagocyte-related alterations have also been reported in SCZ brains. In the neurogenic subependymal zone (SEZ), an SCZ subgroup with high inflammation (HC) (defined by elevated expression of IL-1β, IL-1R1, serine protease inhibitor member 3 [SERPINA3], and c-x-c motif chemokine ligand 8 [CXCL8] mRNA transcripts) showed higher expression of the identity markers of macrophages (CD163) and monocytes (CD14) than high-inflammation HCs [53]. Notably, increased infiltration of monophagocytes into the SEZ appeared to be a shared pathological feature between patients with SCZ and patients with BD [54]. In the mid brain, immunostaining revealed that CD163+ macrophage density was elevated in high-inflammation SCZ compared to HCs [55]. Along with the close association between these cells and dopaminergic neurons in the substantia nigra, a positive correlation between CD163 and the complement C1 subcomponent A (C1qA) mRNA transcripts was detected in high-inflammation SCZ, suggesting a possible involvement of dysregulated complement-mediated phagocytic activity of these cells in the development of inflammatory pathology in SCZ.
Nevertheless, the exact role played by these brain immune cells in SCZ remains contentious. For instance, immunohistochemical analysis in the DPFC of SCZ patients yielded no evidence of CD163+CD206+ perivascular macrophage infiltration into the brain parenchyma [33]. In contrast, a different study that transcriptionally quantified CD163 mRNA expression in the DPFC showed that macrophage accumulation in this brain region was a signature of high-inflammation SCZ [56]. These discrepancies might be attributed to different analytical approaches employed by these studies (mRNA vs. protein expression), as well as the heterogeneity of the SCZ cohort (i.e., the presence of high inflammation or acute psychosis). Alternatively, procedural differences in tissue collection and storage might also affect the detection of the macrophage marker of interest.
Besides post mortem brain studies, cerebrospinal fluid (CSF) analysis of adaptive and innate immune cells in SCZ is another approach that has been investigated. For example, acute psychotic symptomatology in SCZ patients [57] was associated with an accumulation of monophagocytes in CSF samples. Further analysis revealed that this signature of innate immune alteration was accompanied by an increase in the frequency of lymphocytes with an activated phenotype during psychosis onset in SCZ [58]. Interestingly, this abnormality was responsive to conventional neuroleptic medication, which led to post-treatment normalization of monophagocyte counts in several subjects with SCZ. Alterations in CD4+ and CD8+ T cells have also been reported in acutely psychotic SCZ [59]. Lastly, B cells in the CSF of SCZ patients showed marked differences in their antibody repertoire compared to HCs [60], hinting at a potential involvement of distinct pathogenic B cell subsets in autoimmune-like symptoms of SCZ.
Collectively, these aforementioned studies highlighted the potential involvement of CNS adaptive and innate immune cells in distinct SCZ-associated pathologies. While excessive neuroinflammation may be uniquely linked to changes in monophagocytes, negative symptoms of SCZ may be related to alterations in adaptive lymphocytes. Importantly, perturbations in both cell types might be associated with the onset of psychosis and have been responsive to antipsychotic treatments. Some of these CNS immunological disturbances could also represent an overlapping pathological feature among different neuropsychiatric illnesses.

3. Alterations in Circulating Immune Cells in SCZ

While most studies of CNS immunity have focused on microglia, some also attempted a more diverse characterization of the potential contribution of different peripheral immune cell subsets in SCZ (Table 1). Findings on changes in innate immune cells, including monocytes and neutrophils, along with those related to autoantibodies, presumably produced by specific pathogenic B cell clones, appear to be the most consistent and/or have the lowest risks of bias [61], thus supporting the inflammatory and autoimmune hypotheses of SCZ pathophysiology. Various reports also suggested that natural killer (NK) cells and different T cell populations in SCZ were dysregulated. In this context, this section summarizes the major findings on peripheral immunological changes, including the proposed clinical implications and associated mechanistic insights.

3.1. Monocytes

In the innate immune system, monocytes represent the counterparts in the circulatory system of microglia and are known for their plasticity in responding to environmental changes. There is a growing body of clinical evidence of monocyte alterations in SCZ blood samples [62] (Figure 2), including higher total monocyte counts in SCZ during the first episode of psychosis, although some discrepancies remain as to whether these alterations are linked to disease severity [63,64]. A similar increase in total monocyte number was also observed in patients with non-affective psychosis [65], while elevated counts of classical monocytes and proinflammatory monocytes have been linked to clozapine-treated and recent-onset SCZ, respectively [66,67]. Several studies found higher monocyte-related indices in SCZ. For example, a large study involving over 6000 patients with SCZ revealed that the monocyte to lymphocyte ratio (MLR) was significantly increased in SCZ compared to HCs [68]. Notably, this ratio could be used to distinguish SCZ (during the first episode of psychosis) from HCs or patients during the first episode of depression [69]. Furthermore, while elevated MLR may represent a shared pathological hallmark of SCZ and BD compared to HCs, some discrepancies remain, possibly related to methodological variations and differing patient inclusion criteria [70,71]. Similar relationships were observed between monocyte counts and other metabolic markers, such as the cardioprotective HDL (monocyte to HDL ratio, MHR), hinting at a possible involvement of these cells with the onset of cardiometabolic comorbidities in SCZ and BD [72,73,74,75].
These observations of alterations in various monocyte features represent a significant breakthrough in SCZ research. While many studies inferred a monocyte-associated gene set from bulk immune cell transcriptome profiling, the studies mentioned above typically focused on a phenotypic and functional characterization of the monocytes themselves. For example, an interferon gene signature in isolated monocytes was observed in SCZ, with dynamic changes over the disease course [77]. Interestingly, unique alterations in protein tyrosine phosphatase non-receptor type 7 (PTPN7)/NGFI-A-binding protein 2 (NAB2) were observed between SCZ and BD, while some overlapping gene signatures, characterized by elevations in activating transcription factor 3 (ATF3)/dual specificity phosphatase 2 (DUSP2) and early-growth-response protein 3 (EGR3)/mad-max dimerization protein (MXD1) [78], were shared between both illnesses. Regarding the activation phenotype, a higher expression of triggering receptor expressed on myeloid cell (TREM) 1 and 2 was documented in monocytes from patients with SCZ, with the former having been linked to transcriptional changes in ATF3 and EGR3 [79,80]. Importantly, these changes were specific to SCZ but not patients with major depressive disorder (MDD), thus providing further evidence of the utility of monocyte-related markers as a distinguishing feature between these two neuropsychiatric conditions. Increased expression of a canonical activation marker, HLA-DR, was also observed in SCZ monocytes, along with changes in their phagocytic activity during acute psychotic onset [81,82]. Alterations in cytokine production were also observed in inflammatory mediators, such as the production of IL-1, IL-6, and tumor necrosis factor (TNF-α) [83] from SCZ monocytes, which was accompanied by a higher response to lipopolysaccharide (LPS)/toll-like receptor 4 (TLR4) stimulation [84]. Additionally, concanavalin (Con-A) stimulated IL1 secretion from monocytes in peripheral mononuclear cells from drug-naïve SCZ patients was elevated [85], while polyinosinic-polycytylic acid (poli I:C)/toll-like receptor 3 (TLR3)-stimulated intracellular production of IL1 from monocytes was reduced compared to HCs [86]. Other changes in baseline TLR4 expression and TLR4 downregulation in response to LPS were also observed in monocytes from SCZ with tardive dyskinesia [87] and in those from patients with first episode of psychosis [88].
Of potential clinical utility is the presence of various monocyte-specific markers for treatment response monitoring and differential diagnosis of SCZ. Specifically, reduced glucose transporter (GLUT1) expression in monocytes has been proposed as a key diagnostic feature to distinguish SCZ from BD, MDD, and autistic spectrum disorder [89], while soluble CD14, an identity marker of circulating monocytes, could accurately predict subsequent SCZ diagnosis [90]. A monocytic transcription signature was also proposed as a candidate marker for monitoring beneficial simvastatin response in patients with SCZ [91]. The effectiveness of other antipsychotics, such as haloperidol/perazin and clozapine, can also be predicted by a reduced monocyte production of I-1/TNF-α and reactive oxygen species (ROS), respectively, while the effectiveness of olanzapine could be monitored by pre-treatment monocytic expression of the fatty acid receptor CD36 [83,92,93]. Altogether, these findings on distributional, phenotypic, and functional alterations in monocytes are of high clinical relevance to provide an improved understanding of the cellular mechanisms of SCZ initiation/progression, as well as the regression of its clinical symptoms by currently available pharmacologic agents.

3.2. Granulocytes

Circulating granulocytes consist of three major myeloid cell subsets, namely basophils, eosinophils, and neutrophils. While the first two are rarely discussed in the context of neuropsychiatric illnesses, an increase in neutrophil-related parameters represents one of the most consistent findings regarding changes in peripheral immune cells in SCZ (Figure 2). Several studies involving over 6000 patients found a marked increase in the total number of circulating neutrophils during acute psychotic symptomatology in SCZ patients [68] and in patients with a first episode of psychosis [94,95,96]. Notably, in the latter, the increase in neutrophil count was associated with various CNS anatomical pathologies often observed in SCZ [97], including enlarged ventricles and reduced gray matter volumes, as well as hallmark symptoms of this illnesses, such as hallucination and avolition. Comparable increases in neutrophil count were also noted in other subtypes of SCZ, including paranoid, residual, and non-affective psychotic patients [98,99]. Similarly, the neutrophil to lymphocyte ratio (NLR), an index commonly used for clinical assessment of inflammation, was higher in SCZ patients, with some sex-specific differences [100,101], and in patients affected by MDD and BD [102,103,104,105]. Of note, NLR might represent a common feature of immunological disturbance among various types of neuropsychiatric illnesses, ranging from bipolar disorder and major depression to SCZ, although it remains unclear how the actual magnitudes in NLR change should be interpreted to differentiate SCZ from manic episodes of BD [70,106]. Furthermore, despite the consensus on increased NLR in SCZ, conflicting findings exist regarding its potential association with various symptoms and disease stages of SCZ. For instance, while increased NLR yielded no correlation with SCZ severity and symptoms as assessed by the Brief Psychiatric Rating Scale (BPRS) [107], the correlation became significant once different scales were used, e.g., the Positive and Negative Syndrome Scale (PANSS) [64,108], the Clinical Global Impression-Severity Scale (CGI-S) [109], and the Brief Negative Symptom Scale [100,110]. More specifically, a recent large-scale analysis based on BPRS and CGI-S involving over 1000 SCZ patients demonstrated a significant association between symptoms and NLR. Longitudinal changes in NLR between disease relapse (increased NLR) and remission (reduced NLR) have also been reported [111,112,113]; however, a large-scale study involving 618 patients found no differences between these two disease stages [114]. Collectively, these conflicting results might be attributed to the complexity of the progression of clinical symptoms in SCZ, which is often compounded by the impact of various demographic factors (age/sex) and medication status on neutrophil numbers. In fact, the agranulocytosis effect of clozapine [115] might account for the observed reduction in NLR after treatment in SCZ [109,116], indicating the potential clinical utility of this immunological marker for efficacy monitoring of this atypical antipsychotics.
Besides changes in cell abundance, studies also reported various functional and phenotypic alterations of neutrophils in SCZ [116,117,118,119,120,121], including oxidative stress [108], which showed a positive association with NLR counts in SCZ. Consistent with this finding, some authors observed that SCZ neutrophils exhibited an increased expression of various markers of oxidative stress, such as malonaldehyde [122] and superoxide anion [123,124]. While several studies reported increased phagocytic activity of SCZ neutrophils, the magnitude of the observed increase differed significantly [81,124]. In summary, while certain details regarding the mechanistic involvement of neutrophils in various clinical aspects of SCZ pathogenesis remain to be confirmed in large-scale studies, most of the evidence suggests that these sentinel innate immune cells play an important role in peripheral immune dysfunction/inflammation in SCZ.

3.3. Natural Killer Cells

NK cells are a type of immune cell with both adaptive and innate features. They have been implicated in a wide range of human diseases, ranging from infection and cancer to CNS disorders. Potential abnormalities in both NK cell count and function have been documented in SCZ, although with significant discrepancies among studies. Flow cytometric analysis showed increased counts of NK cells in clozapine-treated chronic SCZ blood samples compared to HCs [67]. In contrast, computational deconvolution based on gene expression yielded lower NK cell numbers in both drug-naïve and medicated SCZ patients [125,126], and this decrease was uncorrelated to psychotic relapse/remission. A different flow cytometric study confirmed these lower NK cell counts in chronic SCZ; however, medication appeared to increase NK cell numbers [127]. These differences could be due to the different quantification methods used (flow cytometry vs. gene expression, whole blood vs. peripheral mononuclear cells). Chronic SCZ subpopulations who received differing regimens of antipsychotic drugs may exhibit distinct NK cell profiles. In fact, an earlier study involving a heterogenous SCZ cohort (more than four subtypes and treatment modalities) failed to detect any abnormalities in immune cell counts in blood samples [128]. Regarding the function of NK cells in SCZ, there have been conflicting reports on NK cell cytolytic activity, possibly related to significant variations in general NK cell lytic function, medication regimes, and SCZ subtypes [129,130,131,132]. With regard to phenotypes, some studies suggested that an elevated expression of NK-cell-activation markers, such as HLA-DR and natural killer group 2C (NKG2C), might be associated with the first episode of psychosis in SCZ patients compared to HCs. However, these features were also observed during the first psychosis of patients with BD [133]. Another inflammatory marker of blood–brain barrier (BBB) disruption, S100B, was reportedly elevated in NK cells of SCZ patients during acute psychotic symptomatology compared to HCs, possibly associated with the activation of stress signaling pathways [134]. In contrast, in medicated SCZ patients, disease remission was linked to a higher NK cell production of IL17 [135]. Given these numerous discrepancies, the involvement of NK cells in SCZ requires further validation studies with a comprehensive analysis of all parameters of NK cell phenotype and function among well-categorized SCZ and appropriately matched HC cohorts.

3.4. B Lymphocytes

Considering that various autoantibody types have been found to be elevated in serum and CSF samples of SCZ patients, B cells, as producers of antibodies, have long been implicated in the autoimmune hypothesis of SCZ (Figure 2). For instance, several small-cohort studies suggested that autoantibodies may act against anti-glutamic acid decarboxylase (GAD), γ-aminobutyric acid A receptor 1 (GABAR1), anti-acetylcholine receptor (A7ChR), and N-metil-D-aspartato receptor (NMDAR) in SCZ pathogenesis [136,137,138,139]. However, large-scale studies (n > 150) failed to confirm the biological significance of these autoantibodies in this disorder [140,141,142], or they detected only a small range of autoantibodies in low concentrations in certain SCZ subsets, such as clinically high-risk SCZ, SCZ with the first episode of psychosis, and SCZ with tardive dyskinesia. Another set of small-cohort studies found a high proportion (>20%) of autoantibodies against various classes of molecules, including DNA/RNA species [143,144], heat-shock proteins [145,146], and neuronal/neurotransmitter targets [147,148,149,150], which currently await further independent validation with larger sample sizes. Finally, several large-scale studies reported the elevated expression of circulating antibodies against IL-1, IL-6, IL-8, CD25, and gliadin (which has also been linked to peripheral inflammation) in SCZ patients, suggesting a possible mechanistic involvement of dysregulated inflammation in the B-cell-driven antibody-mediated autoimmune hypothesis of SCZ [151,152,153,154,155].
Besides autoantibody characterization, only few studies examined potential alterations in circulating B cell subsets in SCZ. Of note, circulating CD19+ B cells were elevated in paranoid SCZ patients during acute psychotic symptomatology, which could be suppressed by treatment [156]. Blood samples of clozapine-treated patients with chronic SCZ showed elevated levels of naive IgD+CD27-CD19+ B cells [67]. CD5+ B cell counts were also elevated in patients with SCZ; however, this increase remained unaffected by antipsychotic withdrawal [157,158]. Furthermore, the association between CD5+ B cells and SCZ was disputed by a study that considered the confounding effect of race, finding that African Americans, regardless of their disease status (SCZ or HC), appeared to have higher levels of CD5+ B cells than Caucasians [159]. Taken together, these findings warrant further multidimensional analyses of B cell repertoire and function in SCZ, which is pivotal to elucidate the mechanistic underpinnings of the various autoimmune-like pathologies in selected SCZ subgroups.

3.5. T Lymphocytes

Numerous phenotypic studies of various circulating T cell subsets have been conducted in SCZ (Figure 2). While the findings were inconsistent with regard to the distribution of total CD3+ T cells, CD4+ helper T cells, and CD8+ cytotoxic T cells [127,156,160,161,162], they mostly agreed with regard to increased T cell activation [66,163,164]. Furthermore, activated T cells in medicated SCZ patients appeared to have higher levels of CD25 than drug-naïve patients. Along with this ex vivo-activated phenotype, several studies observed a reduced responsiveness to in vitro stimulation of IL-2 production in T cells from drug-naïve SCZ patients compared to HCs [165,166,167]. However, this decrease in IL-2 production was not observed in a study of paranoid and residual SCZ [99], possibly due to how patients were stratified in this study and/or the use of purified T cells/peripheral blood mononuclear cells vs. whole blood for in vitro assays. Regarding T-helper (Th) cell subtypes, an increase in Th2 cells was reportedly associated with the SCZ subtype with a pro-inflammatory monocyte feature [66]; however, this alteration could not be confirmed by a different study [99]. The same two studies also yielded inconsistent findings regarding the role of Th1 cells. In contrast, consensus exists regarding the elevated numbers of regulatory T cells (Treg) and IL-17-producing Th cells (Th17) in SCZ [66,135,163,168,169,170]. Of note, the Treg increase in medicated SCZ patients was associated with fewer negative symptoms [169], while Th17 elevation might be linked to psychosis [168,171]. Interestingly, the Th17 increase in SCZ with the first episode of psychosis was suppressed by risperidone treatment [168], while haloperidol or risperidone-treated SCZ patients continued to show elevated Th17 numbers during stable remission [135]. These paradoxical findings warrant further investigations into the longitudinal effects of antipsychotics on Th17 cell counts in SCZ. Mucosal-associated invariant T (MAIT) cells [170] were also elevated in patients with SCZ, supporting the hypothesis of mucosal microbiome involvement in SCZ pathogenesis.
Several studies investigating the T cell phenotype examined the expression of dopamine receptors, the presumed target of clozapine [172]. However, their findings on the expression of dopamine receptors D (DRD) 2 and 4 were inconsistent, likely due to methodological differences (mRNA expression vs. flow cytometry) [173,174]. T-cell-specific oxidative damage in SCZ has also been quantified and showed an increase in mitochondrial dysfunction across different T cell subsets from patients with acute relapse compared to HCs [175]. Furthermore, this cellular pathology appears to be associated with positive symptoms [175]. Lastly, T cell methylation profile and repertoire were reportedly associated with distinct SCZ subtypes. Widespread methylation markers in T cells were observed in SCZ patients with more severe symptoms and cognitive impairment [176]. Additionally, some cis-diagnostic (SCZ specific) and trans-diagnostic (common among several psychiatric illnesses) genetic variants also appeared to be epigenetically active in CD4+ T cells, but not innate immune cells, from SCZ patients, indicating the potential utility of these T-cell-specific biomarkers for individualized therapeutic development in SCZ [177].
Overall, studies examining the role of T cells in SCZ point to possible alterations in the activation status of this lymphocyte subset in SCZ, typically consistent with an increase in the expression of the canonical activation marker CD25 and elevated counts of Treg and Th17. To obtain a more comprehensive understanding of the role of T cells in SCZ, future studies should attempt more detailed characterizations of other T cell subsets, as well as different immunometabolic markers of T cells across the entire spectrum of SCZ patients with different symptoms and treatment statuses.
Table 1. Major immunological dysfunctions in patients with schizophrenia (SCZ).
Table 1. Major immunological dysfunctions in patients with schizophrenia (SCZ).
Central
Nervous System
FeaturesAnatomical
Location
MicrogliaIncreased phagocytosis [13]Whole brain
No change in phagocytosis [31]PFC
Increased IBA-1 [30]ACC
No change in IBA-1 [31]PFC
Increased HLA-DR [19,20]Frontal/temporal cortex, hippocampus
No change in HLA-DR [25,26]PFC, ACC, hippocampus
Increased S100A8/9 [28]Frontal cortex
Activated morphology [14,15,16]PFC, visual cortex
Microgliosis [32]Frontal/temporal cortex
Decreased quinolinic acid [29]Hippocampus
T cellsIncreased CD3+ frequency [19,48]Whole brain, hippocampus
Decreased CD3+ frequency [19]Dorsal PFC
Increased CD4+ frequency [55]CSF
Increased CD8+ frequency [55]CSF
B cellsIncreased CD20+ frequency [19,52]Whole brain, hippocampus
BCR alterations [60]CSF
MacrophagesIncreased CD163 [53,55,56]Dorsal PFC, CSF, SEZ
No change in CD163 [33]Dorsal PFC
Increased CD14 [53]SEZ
Increased frequency [57]CSF
Peripheral BloodFeatures
MonocytesIncreased total monocyte counts [62,63,64]
Increased classical monocyte counts [66]
Increased pro-inflammatory monocyte counts [67]
Increased monocyte to lymphocyte ratio [68]
Increased monocyte to HDL ratio [72,73]
Increased TREM1/2 [79,80]
Increased ATF3/EGR3 [78]
Increased HLA-DR [82]
Increased phagocytosis [81]
Increased IL-1, IL-6, TNF-α [83,85,86]
Increased CD36 [93]
Increased reactive oxygen species [92]
Alterations in TLR4 signaling [84,87,88]
Unique interferon gene signature [77]
Reduced GLUT1 [89]
GranulocytesIncreased neutrophil counts [68,94,95,96,97,98,99]
Increased neutrophil to lymphocyte ratio [100,101]
Increased oxidative stress [108]
Increased malonaldehyde [122]
Increased superoxide anion [123,123]
Increased phagocytosis [81,124]
Natural killer cellsIncreased total counts [68]
Decreased total counts [125,126,127]
No change in total counts [128]
Increased cytotoxicity [132]
Decreased cytotoxicity [129,130]
No change in cytotoxicity [121]
Increased NKG2C [133]
Increased S100B [134]
T CellsIncreased CD3+ and CD4+ T cell counts [153]
Reduced CD3+ T cell counts [150]
Reduced CD4+ T cell counts [146,152]
Increased activation [63,154,155]
Increased CD25 [156]
Reduced IL2 production [156,157,158]
No change in IL2 production [92]
Increased Treg [63,155,159,160,161,162]
Increased Th17 [63,154,155,156,157,158,159,160,161,162]
Increased mucosal-associated invariant T cells [162]
Increased oxidative stress [166]
Altered methylation profile [167]
Altered TCR repertoire [167]
B CellsIncreased CD19+ B cell frequency [156]
Increased IgD+CD27-CD19+ naïve B cell frequency [67]
Increased CD5+ B cell frequency [157,158]
No change in CD5+ B cell frequency [159]
Increased GAD, GABAR1, A7ChR, NMDAR autoantibodies [136,137,138,139]
No changes in GAD, GABAR1, A7ChR, NMDAR autoantibodies [140,141,142]
Increased DNA/RNA autoantibodies [143,144]
Increased heat-shock protein autoantibodies [145,146]
Increased neuronal marker autoantibodies [147,148,149,150]
Increased IL-1/IL-6/IL-8/gliadin autoantibodies [151,152,153,154,155]

4. Implications for Mechanistic Studies and Therapeutic Development

To explore disease mechanisms of SCZ, various in vitro and animal studies have been established, with induced pluripotent stem cells derived from SCZ patients and rodent strains based on human genetics as the most clinically relevant attempts to model SCZ-associated pathology [178,179]. However, most immune-related studies to date have focused exclusively on the role of microglia, but not other peripheral immune cell subsets, in this psychiatric illness. For example, microglia from mice with an overexpression of C4, a genetic risk factor of SCZ, exhibited increased synaptic pruning [180]. Similarly, microglia derived from SCZ patient-derived induced pluripotent stem cells were reportedly more activated and might cause neuronal metabolic disruption [181,182,183]. Building upon these preclinical findings, our synthesis of clinical evidence pertaining to various alterations of other immune cells in peripheral tissues, as well as the CNS, will provide an important impetus for further mechanistic exploration of a potential dynamic interplay among these cell types in different tissue landscapes during SCZ development.
Anomalies in various immune cell subsets and their trafficking patterns in SCZ also suggest the potential utility of cell-type-specific immunotherapy as a novel pharmacological approach for this psychiatric illness [184,185]. In this regard, immunomodulators aiming to inhibit lymphocyte trafficking (fingolimod) and deplete B cells (anti-CD20 monoclonal antibody, rituximab) could significantly improve negative symptoms and general psychopathology of SCZ, respectively [186,187]. Furthermore, cytokine-based immunotherapies, including those aiming to inhibit IL-6 (tocilizumab) and TNF-α (adalimumab), showed promising efficacy in some randomized controlled trials of SCZ [188,189]. Other immunosuppressants, such as azathioprine [190], prednisolone [191], and methotrexate [192], also exhibited preliminary clinical benefits in SCZ patients.
Besides these classical immunotherapies, the potential clinical efficacy in SCZ of other anti-inflammatory treatments, including aspirin, minocycline, N-acetylcysteine, estrogens, telmisartan (an angiotensin receptor 1 antagonist), and pioglitazone (a PPAR-γ antagonist) [193,194,195], has been observed. Interestingly, other anti-inflammatory medications, such as celecoxib, davunetide, dextromethorphan, fatty acids, pregnenolone, statins, and varenicline, did not have a significant impact on SCZ symptoms in a recent meta-analysis, highlighting the presence of inflammatory-pathway-specific abnormalities in SCZ development. Collectively, these findings support the emerging importance of immune-related dysfunction as a targetable pathology of SCZ [196]. However, additional trials are required to confirm these findings as the effectiveness of selected therapies, including anti-IL-6 and anti-CD20 antibodies as well as prednisolone, has been questioned in some clinical studies [197,198,199].

5. Concluding Remarks

Our narrative synthesis of the literature on microglia and other cellular mediators of immunological dysfunction in schizophrenia points to the existence of several immunopathological hallmarks of SCZ in circulation, including major distributional abnormalities in neutrophils, monocytes, immunoregulatory T cell populations, and autoantibody repertoire. In addition, there may be evidence of the occurrence of a dynamic trafficking pattern of both adaptive (T and B cells) and innate (microglia and macrophages) immune cells in various anatomical brain regions during different disease stages of SCZ. We also noted numerous inconsistencies in the clinical evidence, which not only reflect the heterogenous nature of this spectral disorder and other demographic and clinical confounders, but also emphasize the urgent need for the development of high-throughput and high-resolution methodological approaches to facilitate more comprehensive investigations of potential abnormalities in the immune system’s cellular compartments. In this regard, cell-based models of SCZ are of high mechanistic utility as the behavioral nature of this illness is likely difficult to be faithfully recapitulated with animal modelling. Furthermore, such cell-based models would yield a clearer delineation of the roles played by genetic vs. environmental factors in SCZ pathogenesis and provide versatile platforms for therapeutic development to address specific unmet needs in the clinical management of SCZ.

Author Contributions

Conceptualization, K.D.N., L.M. and A.C.; methodology, K.D.N. and L.M.; writing—original draft preparation, K.D.N.; writing—review and editing, A.A. (Andrea Amerio), A.A. (Andrea Aguglia), A.P. and B.C.; supervision, G.S., M.A. and A.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

KDN is the scientific founder of Tranquis Therapeutics, a biotechnology company that develops novel treatments for neuroinflammatory and neurodegenerative diseases. KDN is also a scientific advisor for Tochikunda, a biotechnology company that develops SARS-CoV-2 diagnostic devices. All other authors declare no conflicts of interest and that they have no commercial associations (e.g., consultancies, stock ownership, equity interest, patent/licensing arrangement, etc.) that might pose a conflict of interest in connection with the submitted article.

References

  1. Faden, J.; Citrome, L. Schizophrenia: One name, many different manifestations. Med. Clin. N. Am. 2023, 107, 61–72. [Google Scholar] [CrossRef]
  2. Costanza, A.; Baertschi, M.; Weber, K.; Canuto, A. Maladies neurologiques et suicide: De la neurobiology au manque d’éspoir [neurological diseases and suicide: From neurobiology to hopelessness]. Rev. Med. Suisse 2015, 11, 402–405. [Google Scholar] [PubMed]
  3. Lewine, R.; Hart, M. Schizophrenia spectrum and other psychotic disorders. In Handbook of Clinical Neurology; Elsevier: Amsterdam, The Netherlands, 2020; pp. 315–333. [Google Scholar]
  4. Janoutová, J.; Ambroz, P.; Kovalová, M.; Machaczka, O.; Němeček, K.; Zatloukalová, A.; Mrázková, E.; Košta, O.; Hálová, A.; Hosák, L.; et al. Epidemiology of mild cognitive impairment. Česká A Slov. Neurol. A Neurochir. 2018, 81, 284–289. [Google Scholar] [CrossRef]
  5. Sabe, M.; Chen, C.; Perez, N.; Solmi, M.; Mucci, A.; Galderisi, S.; Strauss, G.P.; Kaiser, S. Thirty years of research on negative symptoms of schizophrenia: A scientometric analysis of hotspots, bursts, and research trends. Neurosci. Biobehav. Rev. 2023, 144, 104979. [Google Scholar] [CrossRef] [PubMed]
  6. Bakhshi, K.; Chance, S.A. The neuropathology of schizophrenia: A selective review of past studies and emerging themes in brain structure and cytoarchitecture. Neuroscience 2015, 303, 82–102. [Google Scholar] [CrossRef] [PubMed]
  7. Costanza, A.; Amerio, A.; Aguglia, A.; Serafini, G.; Amore, M.; Hasler, R.; Ambrosetti, J.; Bondolfi, G.; Sampogna, G.; Berardelli, I.; et al. Hyper/neuroinflammation in COVID-19 and suicide etiopathogenesis: Hypothesis for a nefarious collision? Neurosci. Biobehav. Rev. 2022, 136, 104606. [Google Scholar] [CrossRef]
  8. Serafini, G.; Costanza, A.; Aguglia, A.; Amerio, A.; Trabucco, A.; Escelsior, A.; Sher, L.; Amore, M. The role of inflammation in the pathophysiology of depression and suicidal behavior: Implications for treatment. Med. Clin. N. Am. 2023, 107, 1–29. [Google Scholar] [CrossRef]
  9. Fišar, Z. Biological hypotheses, risk factors, and biomarkers of schizophrenia. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2023, 120, 110626. [Google Scholar] [CrossRef] [PubMed]
  10. Li, J.; Wang, Y.; Yuan, X.; Kang, Y.; Song, X. New insight in the cross-talk between microglia and schizophrenia: From the perspective of neurodevelopment. Front. Psychiatr. 2023, 14, 1126632. [Google Scholar] [CrossRef]
  11. Cao, Y.; Ji, S.; Chen, Y.; Zhang, X.; Ding, G.; Tang, F. Association between autoimmune diseases of the nervous system and schizophrenia: A systematic review and meta-analysis of cohort studies. Compr. Psychiatry 2023, 122, 152370. [Google Scholar] [CrossRef]
  12. Kierdorf, K.; Masuda, T.; Jordão, M.J.C.; Prinz, M. Macrophages at cns interfaces: Ontogeny and function in health and disease. Nat. Rev. Neurosci. 2019, 20, 547–562. [Google Scholar] [CrossRef] [PubMed]
  13. Solov’eva, Z.; Orlovskaia, D.D. Microglia-type cells in normal and pathologic human embryonic brains. Zhurnal Nevropatol. I Psikhiatrii Im. SS Korsakova 1979, 79, 852–857. [Google Scholar]
  14. Uranova, N.A.; Vikhreva, O.V.; Rakhmanova, V.I.; Orlovskaya, D.D. Ultrastructural pathology of oligodendrocytes in the white matter in continuous paranoid schizophrenia: A role for microglia. Zhurnal Nevropatol. I Psikhiatrii Im. SS Korsakova 2017, 117, 76. [Google Scholar] [CrossRef] [PubMed]
  15. Uranova, N.A.; Zimina, I.S.; Vikhreva, O.V.; Krukov, N.O.; Rachmanova, V.I.; Orlovskaya, D.D. Ultrastructural damage of capillaries in the neocortex in schizophrenia. World J. Biol. Psychiatry 2010, 11, 567–578. [Google Scholar] [CrossRef]
  16. Vikhreva, O.V.; Rakhmanova, V.I.; Orlovskaya, D.D.; Uranova, N.A. Ultrastructural pathology of oligodendrocytes in white matter in continuous attack-like schizophrenia and a role for microglia. Zhurnal Nevropatol. I Psikhiatrii Im. SS Korsakova 2018, 118, 69. [Google Scholar] [CrossRef]
  17. Uranova, N.A.; Vikhreva, O.V.; Rakhmanova, V.I.; Orlovskaya, D.D. Ultrastructural pathology of oligodendrocytes adjacent to microglia in prefrontal white matter in schizophrenia. NPJ Schizophr. 2018, 4, 26. [Google Scholar] [CrossRef] [PubMed]
  18. Uranova, N.A.; Vikhreva, O.V.; Rakhmanova, V.I.; Orlovskaya, D.D. Dystrophy of oligodendrocytes and adjacent microglia in prefrontal gray matter in schizophrenia. Front. Psychiatr. 2020, 11, 204. [Google Scholar] [CrossRef] [PubMed]
  19. Busse, S.; Busse, M.; Schiltz, K.; Bielau, H.; Gos, T.; Brisch, R.; Mawrin, C.; Schmitt, A.; Jordan, W.; Müller, U.J.; et al. Different distribution patterns of lymphocytes and microglia in the hippocampus of patients with residual versus paranoid schizophrenia: Further evidence for disease course-related immune alterations? Brain Behav. Immun. 2012, 26, 1273–1279. [Google Scholar] [CrossRef] [PubMed]
  20. Radewicz, K.; Garey, L.J.; Gentleman, S.M.; Reynolds, R. Increase in hla-dr immunoreactive microglia in frontal and temporal cortex of chronic schizophrenics. J. Neuropathol. Exp. Neurol. 2000, 59, 137–150. [Google Scholar] [CrossRef] [PubMed]
  21. Wierzba-Bobrowicz, T.; Lewandowska, E.; Kosno-Kruszewska, E.; Lechowicz, W.; Pasennik, E.; Schmidt-Sidor, B. Degeneration of microglial cells in frontal and temporal lobes of chronic schizophrenics. Folia Neuropathol. 2004, 42, 157–165. [Google Scholar] [PubMed]
  22. Fillman, S.G.; Cloonan, N.; Catts, V.S.; Miller, L.C.; Wong, J.; McCrossin, T.; Cairns, M.; Weickert, C.S. Increased inflammatory markers identified in the dorsolateral prefrontal cortex of individuals with schizophrenia. Mol. Psychiatry 2012, 18, 206–214. [Google Scholar] [CrossRef]
  23. Bayer, T.A.; Buslei, R.; Havas, L.; Falkai, P. Evidence for activation of microglia in patients with psychiatric illnesses. Neurosci. Lett. 1999, 271, 126–128. [Google Scholar] [CrossRef]
  24. Costanza, A.; Xekardaki, A.; Kovari, E.; Gold, G.; Bouras, C.; Giannakopoulos, P. Microvascular burden and alzheimer-type lesions across the age spectrum. J. Alzheimers Dis. 2012, 32, 643–652. [Google Scholar] [CrossRef]
  25. Steiner, J.; Bielau, H.; Brisch, R.; Danos, P.; Ullrich, O.; Mawrin, C.; Bernstein, H.-G.; Bogerts, B. Immunological aspects in the neurobiology of suicide: Elevated microglial density in schizophrenia and depression is associated with suicide. J. Psychiatr. Res. 2008, 42, 151–157. [Google Scholar] [CrossRef]
  26. Steiner, J.; Mawrin, C.; Ziegeler, A.; Bielau, H.; Ullrich, O.; Bernstein, H.-G.; Bogerts, B. Distribution of hla-dr-positive microglia in schizophrenia reflects impaired cerebral lateralization. Acta Neuropathol. 2006, 112, 305–316. [Google Scholar] [CrossRef] [PubMed]
  27. Foster, R.; Kandanearatchi, A.; Beasley, C.; Williams, B.; Khan, N.; Fagerhol, M.K.; Everall, I.P. Calprotectin in microglia from frontal cortex is up-regulated in schizophrenia: Evidence for an inflammatory process? Eur. J. Neurosci. 2006, 24, 3561–3566. [Google Scholar] [CrossRef] [PubMed]
  28. Gos, T.; Myint, A.-M.; Schiltz, K.; Meyer-Lotz, G.; Dobrowolny, H.; Busse, S.; Müller, U.J.; Mawrin, C.; Bernstein, H.-G.; Bogerts, B.; et al. Reduced microglial immunoreactivity for endogenous nmda receptor agonist quinolinic acid in the hippocampus of schizophrenia patients. Brain Behav. Immun. 2014, 41, 59–64. [Google Scholar] [CrossRef] [PubMed]
  29. Mildner, A.; Huang, H.; Radke, J.; Stenzel, W.; Priller, J. P2y(12) receptor is expressed on human microglia under physiological conditions throughout development and is sensitive to neuroinflammatory diseases. Glia 2017, 65, 375–387. [Google Scholar] [CrossRef] [PubMed]
  30. Petrasch-Parwez, E.; Schöbel, A.; Benali, A.; Moinfar, Z.; Förster, E.; Brüne, M.; Juckel, G. Lateralization of increased density of iba1-immunopositive microglial cells in the anterior midcingulate cortex of schizophrenia and bipolar disorder. Eur. Arch. Psychiatry Clin. Neurosci. 2020, 270, 819–828. [Google Scholar] [CrossRef] [PubMed]
  31. Tzioras, M.; Stevenson, A.J.; Boche, D.; Spires-Jones, T.L. Microglial contribution to synaptic uptake in the prefrontal cortex in schizophrenia. Neuropathol. Appl. Neurobiol. 2020, 47, 346–351. [Google Scholar] [CrossRef]
  32. Gober, R.; Ardalan, M.; Shiadeh, S.M.J.; Duque, L.; Garamszegi, S.P.; Ascona, M.; Barreda, A.; Sun, X.; Mallard, C.; Vontell, R.T. Microglia activation in postmortem brains with schizophrenia demonstrates distinct morphological changes between brain regions. Brain Pathol. 2022, 32, e13003. [Google Scholar] [CrossRef] [PubMed]
  33. De Picker, L.J.; Victoriano, G.M.; Richards, R.; Gorvett, A.J.; Lyons, S.; Buckland, G.R.; Tofani, T.; Norman, J.L.; Chatelet, D.S.; Nicoll, J.A.R.; et al. Immune environment of the brain in schizophrenia and during the psychotic episode: A human post-mortem study. Brain Behav. Immun. 2021, 97, 319–327. [Google Scholar] [CrossRef]
  34. Uranova, N.A.; Vikhreva, O.V.; Rakhmanova, V.I. Abnormal microglial reactivity in gray matter of the prefrontal cortex in schizophrenia. Asian J. Psychiatry 2021, 63, 102752. [Google Scholar] [CrossRef] [PubMed]
  35. Mokrov, G.V.; Deeva, O.A.; Gudasheva, T.A. The ligands of translocator protein: Design and biological properties. Curr. Pharm. Des. 2021, 27, 217–237. [Google Scholar] [CrossRef]
  36. Papadopoulos, V.; Baraldi, M.; Guilarte, T.R.; Knudsen, T.B.; Lacapère, J.J.; Lindemann, P.; Norenberg, M.D.; Nutt, D.; Weizman, A.; Zhang, M.R.; et al. Translocator protein (18kda): New nomenclature for the peripheral-type benzodiazepine receptor based on its structure and molecular function. Trends Pharmacol. Sci. 2006, 27, 402–409. [Google Scholar] [CrossRef]
  37. Wolf, A.; Herb, M.; Schramm, M.; Langmann, T. The tspo-nox1 axis controls phagocyte-triggered pathological angiogenesis in the eye. Nat. Commun. 2020, 11, 2709. [Google Scholar] [CrossRef] [PubMed]
  38. Ren, H.; Han, R.; Chen, X.; Liu, X.; Wan, J.; Wang, L.; Yang, X.; Wang, J. Potential therapeutic targets for intracerebral hemorrhage-associated inflammation: An update. J. Cereb. Blood Flow Metab. Off. J. Int. Soc. Cereb. Blood Flow Metab. 2020, 40, 1752–1768. [Google Scholar] [CrossRef] [PubMed]
  39. Collste, K.; Plavén-Sigray, P.; Fatouros-Bergman, H.; Victorsson, P.; Schain, M.; Forsberg, A.; Amini, N.; Aeinehband, S.; Erhardt, S.; Halldin, C.; et al. Lower levels of the glial cell marker tspo in drug-naive first-episode psychosis patients as measured using pet and [11c]pbr28. Mol. Psychiatry 2017, 22, 850–856. [Google Scholar] [CrossRef] [PubMed]
  40. Conen, S.; Gregory, C.J.; Hinz, R.; Smallman, R.; Corsi-Zuelli, F.; Deakin, B.; Talbot, P.S. Neuroinflammation as measured by positron emission tomography in patients with recent onset and established schizophrenia: Implications for immune pathogenesis. Mol. Psychiatry 2021, 26, 5398–5406. [Google Scholar] [CrossRef]
  41. Hafizi, S.; Tseng, H.-H.; Rao, N.; Selvanathan, T.; Kenk, M.; Bazinet, R.P.; Suridjan, I.; Wilson, A.A.; Meyer, J.H.; Remington, G.; et al. Imaging microglial activation in untreated first-episode psychosis: A pet study with [(18)f]feppa. Am. J. Psychiatry 2017, 174, 118–124. [Google Scholar] [CrossRef]
  42. Holmes, S.E.; Hinz, R.; Drake, R.J.; Gregory, C.J.; Conen, S.; Matthews, J.C.; Anton-Rodriguez, J.M.; Gerhard, A.; Talbot, P.S. In vivo imaging of brain microglial activity in antipsychotic-free and medicated schizophrenia: A [11c](r)-pk11195 positron emission tomography study. Mol. Psychiatry 2016, 21, 1672–1679. [Google Scholar] [CrossRef] [PubMed]
  43. Ottoy, J.; De Picker, L.; Verhaeghe, J.; Deleye, S.; Wyffels, L.; Kosten, L.; Sabbe, B.; Coppens, V.; Timmers, M.; van Nueten, L.; et al. (18)f-pbr111 pet imaging in healthy controls and schizophrenia: Test-retest reproducibility and quantification of neuroinflammation. J. Nucl. Med. Off. Publ. Soc. Nucl. Med. 2018, 59, 1267–1274. [Google Scholar]
  44. Selvaraj, S.; Bloomfield, P.S.; Cao, B.; Veronese, M.; Turkheimer, F.; Howes, O.D. Brain tspo imaging and gray matter volume in schizophrenia patients and in people at ultra high risk of psychosis: An [(11)c]pbr28 study. Schizophr. Res. 2018, 195, 206–214. [Google Scholar] [CrossRef] [PubMed]
  45. Van der Doef, T.F.; de Witte, L.D.; Sutterland, A.L.; Jobse, E.; Yaqub, M.; Boellaard, R.; de Haan, L.; Eriksson, J.; Lammertsma, A.A.; Kahn, R.S.; et al. In vivo (r)-[(11)c]pk11195 pet imaging of 18kda translocator protein in recent onset psychosis. NPJ Schizophr. 2016, 2, 16031. [Google Scholar] [CrossRef]
  46. Marques, T.R.; Ashok, A.H.; Pillinger, T.; Veronese, M.; Turkheimer, F.E.; Dazzan, P.; Sommer, I.E.C.; Howes, O.D. Neuroinflammation in schizophrenia: Meta-analysis of in vivo microglial imaging studies. Psychol. Med. 2019, 49, 2186–2196. [Google Scholar] [CrossRef]
  47. Plavén-Sigray, P.; Matheson, G.J.; Collste, K.; Ashok, A.H.; Coughlin, J.M.; Howes, O.D.; Mizrahi, R.; Pomper, M.G.; Rusjan, P.; Veronese, M.; et al. Positron emission tomography studies of the glial cell marker translocator protein in patients with psychosis: A meta-analysis using individual participant data. Biol. Psychiatry 2018, 84, 433–442. [Google Scholar] [CrossRef]
  48. Notter, T.; Coughlin, J.M.; Gschwind, T.; Weber-Stadlbauer, U.; Wang, Y.; Kassiou, M.; Vernon, A.C.; Benke, D.; Pomper, M.G.; Sawa, A.; et al. Translational evaluation of translocator protein as a marker of neuroinflammation in schizophrenia. Mol. Psychiatry 2017, 23, 323–334. [Google Scholar] [CrossRef] [PubMed]
  49. Corley, E.; Holleran, L.; Fahey, L.; Corvin, A.; Morris, D.W.; Donohoe, G. Microglial-expressed genetic risk variants, cognitive function and brain volume in patients with schizophrenia and healthy controls. Transl. Psychiatry 2021, 11, 490. [Google Scholar] [CrossRef]
  50. Mou, T.-C.M.; Lane, M.V.; Ireland, D.D.C.; Verthelyi, D.; Tonelli, L.H.; Clark, S.M. Association of complement component 4 with neuroimmune abnormalities in the subventricular zone in schizophrenia and autism spectrum disorders. Neurobiol. Dis. 2022, 173, 105840. [Google Scholar] [CrossRef]
  51. Snijders, G.J.L.J.; van Zuiden, W.; Sneeboer, M.A.M.; Berdenis van Berlekom, A.; van der Geest, A.T.; Schnieder, T.; MacIntyre, D.J.; Hol, E.M.; Kahn, R.S.; de Witte, L.D. A loss of mature microglial markers without immune activation in schizophrenia. Glia 2021, 69, 1251–1267. [Google Scholar] [CrossRef]
  52. Schlaaff, K.; Dobrowolny, H.; Frodl, T.; Mawrin, C.; Gos, T.; Steiner, J.; Bogerts, B. Increased densities of t and b lymphocytes indicate neuroinflammation in subgroups of schizophrenia and mood disorder patients. Brain Behav. Immun. 2020, 88, 497–506. [Google Scholar] [CrossRef] [PubMed]
  53. North, H.F.; Weissleder, C.; Fullerton, J.M.; Webster, M.J.; Weickert, C.S. Increased immune cell and altered microglia and neurogenesis transcripts in an australian schizophrenia subgroup with elevated inflammation. Schizophr. Res. 2022, 248, 208–218. [Google Scholar] [CrossRef] [PubMed]
  54. North, H.F.; Weissleder, C.; Fullerton, J.M.; Sager, R.; Webster, M.J.; Weickert, C.S. A schizophrenia subgroup with elevated inflammation displays reduced microglia, increased peripheral immune cell and altered neurogenesis marker gene expression in the subependymal zone. Transl. Psychiatry 2021, 11, 635. [Google Scholar] [CrossRef] [PubMed]
  55. Purves-Tyson, T.D.; Robinson, K.; Brown, A.M.; Boerrigter, D.; Cai, H.Q.; Weissleder, C.; Owens, S.J.; Rothmond, D.A.; Shannon Weickert, C. Increased macrophages and c1qa, c3, c4 transcripts in the midbrain of people with schizophrenia. Front. Immunol. 2020, 11, 2002. [Google Scholar] [CrossRef]
  56. Zhu, Y.; Webster, M.J.; Murphy, C.E.; Middleton, F.A.; Massa, P.T.; Liu, C.; Dai, R.; Weickert, C.S. Distinct phenotypes of inflammation associated macrophages and microglia in the prefrontal cortex schizophrenia compared to controls. Front. Neurosci. 2022, 16, 858989. [Google Scholar] [CrossRef]
  57. Nikkilä, H.V.; Müller, K.; Ahokas, A.; Miettinen, K.; Rimón, R.; Andersson, L.C. Accumulation of macrophages in the csf of schizophrenic patients during acute psychotic episodes. Am. J. Psychiatry 1999, 156, 1725–1729. [Google Scholar] [CrossRef]
  58. Nikkilä, H.V.; Müller, K.; Ahokas, A.; Rimón, R.; Andersson, L.C. Increased frequency of activated lymphocytes in the cerebrospinal fluid of patients with acute schizophrenia. Schizophr. Res. 2001, 49, 99–105. [Google Scholar] [CrossRef]
  59. Nikkilä, H.; Müller, K.; Ahokas, A.; Miettinen, K.; Andersson, L.C.; Rimón, R. Abnormal distributions of t-lymphocyte subsets in the cerebrospinal fluid of patients with acute schizophrenia. Schizophr. Res. 1995, 14, 215–221. [Google Scholar] [CrossRef]
  60. Husain-Krautter, S.; Lee, J.; Vos, D.; Gallego, J.A.; Malhotra, A.K.; Rothstein, T.L. Skewing of the antibody repertoire in cerebrospinal fluid b cells from healthy controls and patients with schizophrenia. Behav. Brain Res. 2022, 422, 113743. [Google Scholar] [CrossRef]
  61. Ermakov, E.A.; Melamud, M.M.; Buneva, V.N.; Ivanova, S.A. Immune system abnormalities in schizophrenia: An integrative view and translational perspectives. Front. Psychiatr. 2022, 13, 880568. [Google Scholar] [CrossRef]
  62. Mazza, M.G.; Capellazzi, M.; Lucchi, S.; Tagliabue, I.; Rossetti, A.; Clerici, M. Monocyte count in schizophrenia and related disorders: A systematic review and meta-analysis. Acta Neuropsychiatr. 2020, 32, 229–236. [Google Scholar] [CrossRef]
  63. Orhan, F.; Schwieler, L.; Fatouros-Bergman, H.; Malmqvist, A.; Cervenka, S.; Collste, K.; Flyckt, L.; Farde, L.; Sellgren, C.M.; Piehl, F.; et al. Increased number of monocytes and plasma levels of mcp-1 and ykl-40 in first-episode psychosis. Acta Psychiatr. Scand. 2018, 138, 432–440. [Google Scholar] [CrossRef] [PubMed]
  64. Steiner, J.; Frodl, T.; Schiltz, K.; Dobrowolny, H.; Jacobs, R.; Fernandes, B.S.; Guest, P.C.; Meyer-Lotz, G.; Borucki, K.; Bahn, S.; et al. Innate immune cells and c-reactive protein in acute first-episode psychosis and schizophrenia: Relationship to psychopathology and treatment. Schizophr. Bull. 2020, 46, 363–373. [Google Scholar] [CrossRef] [PubMed]
  65. Miller, B.J.; Kandhal, P.; Rapaport, M.H.; Mellor, A.; Buckley, P. Total and differential white blood cell counts, high-sensitivity c-reactive protein, and cardiovascular risk in non-affective psychoses. Brain Behav. Immun. 2015, 45, 28–35. [Google Scholar] [CrossRef]
  66. Drexhage, R.C.; Hoogenboezem, T.A.; Cohen, D.; Versnel, M.A.; Nolen, W.A.; van Beveren, N.J.M.; Drexhage, H.A. An activated set point of t-cell and monocyte inflammatory networks in recent-onset schizophrenia patients involves both pro- and anti-inflammatory forces. Int. J. Neuropsychopharmacol. 2011, 14, 746–755. [Google Scholar] [CrossRef] [PubMed]
  67. Fernandez-Egea, E.; Vértes, P.E.; Flint, S.M.; Turner, L.; Mustafa, S.; Hatton, A.; Smith, K.G.C.; Lyons, P.A.; Bullmore, E.T. Peripheral immune cell populations associated with cognitive deficits and negative symptoms of treatment-resistant schizophrenia. PLoS ONE 2016, 11, e0155631. [Google Scholar] [CrossRef]
  68. Xu, H.; Wei, Y.; Zheng, L.; Zhang, H.; Luo, T.; Li, H.; Ma, J.; Chen, J. Relation between unconjugated bilirubin and peripheral biomarkers of inflammation derived from complete blood counts in patients with acute stage of schizophrenia. Front. Psychiatr. 2022, 13, 843985. [Google Scholar] [CrossRef] [PubMed]
  69. Yu, Q.; Weng, W.; Zhou, H.; Tang, Y.; Ding, S.; Huang, K.; Liu, Y. Elevated platelet parameter in first-episode schizophrenia patients: A cross-sectional study. J. Interferon Cytokine Res. 2020, 40, 524–529. [Google Scholar] [CrossRef]
  70. Özdin, S.; Sarisoy, G.; Böke, Ö. A comparison of the neutrophil-lymphocyte, platelet-lymphocyte and monocyte-lymphocyte ratios in schizophrenia and bipolar disorder patients—A retrospective file review. Nord. J. Psychiatry 2017, 71, 509–512. [Google Scholar] [CrossRef]
  71. Wei, Y.; Wang, T.; Li, G.; Feng, J.; Deng, L.; Xu, H.; Yin, L.; Ma, J.; Chen, D.; Chen, J. Investigation of systemic immune-inflammation index, neutrophil/high-density lipoprotein ratio, lymphocyte/high-density lipoprotein ratio, and monocyte/high-density lipoprotein ratio as indicators of inflammation in patients with schizophrenia and bipolar disorder. Front. Psychiatr. 2022, 13, 941728. [Google Scholar]
  72. Kılıç, N.; Tasci, G.; Yılmaz, S.; Öner, P.; Korkmaz, S. Monocyte/hdl cholesterol ratios as a new inflammatory marker in patients with schizophrenia. J. Pers. Med. 2023, 13, 276. [Google Scholar] [CrossRef] [PubMed]
  73. Sahpolat, M.; Ayar, D.; Ari, M.; Karaman, M.A. Elevated monocyte to high-density lipoprotein ratios as an inflammation markers for schizophrenia patients. Clin. Psychopharmacol. Neurosci. 2021, 19, 112–116. [Google Scholar] [CrossRef] [PubMed]
  74. Aguglia, A.; Salvi, V.; Amerio, A.; Gari, M.; Dragogna, F.; Mencacci, C.; Volpe, U.; Serafini, G.; Amore, M. Number of episodes and duration of illness associated with hypertension and 10-year cardiovascular risk in patients with bipolar disorder type i. Psychiatry Res. 2022, 308, 114344. [Google Scholar] [CrossRef] [PubMed]
  75. Salvi, V.; Aguglia, A.; Barone-Adesi, F.; Bianchi, D.; Donfrancesco, C.; Dragogna, F.; Palmieri, L.; Serafini, G.; Amore, M.; Mencacci, C. Cardiovascular risk in patients with severe mental illness in italy. Eur. Psychiatry J. Assoc. Eur. Psychiatr. 2020, 63, e96. [Google Scholar] [CrossRef]
  76. Whitcroft, K.L.; Hummel, T. Olfactory dysfunction in COVID-19. JAMA 2020, 323, 2512. [Google Scholar] [CrossRef]
  77. Melbourne, J.K.; Rosen, C.; Chase, K.A.; Feiner, B.; Sharma, R.P. Monocyte transcriptional profiling highlights a shift in immune signatures over the course of illness in schizophrenia. Front. Psychiatr. 2021, 12, 649494. [Google Scholar] [CrossRef] [PubMed]
  78. Drexhage, R.C.; van der Heul-Nieuwenhuijsen, L.; Padmos, R.C.; van Beveren, N.; Cohen, D.; Versnel, M.A.; Nolen, W.A.; Drexhage, H.A. Inflammatory gene expression in monocytes of patients with schizophrenia: Overlap and difference with bipolar disorder. A study in naturalistically treated patients. Int. J. Neuropsychopharmacol. 2010, 13, 1369–1381. [Google Scholar] [CrossRef]
  79. Weigelt, K.; Carvalho, L.A.; Drexhage, R.C.; Wijkhuijs, A.; Wit, H.d.; van Beveren, N.J.M.; Birkenhäger, T.K.; Bergink, V.; Drexhage, H.A. Trem-1 and dap12 expression in monocytes of patients with severe psychiatric disorders. Egr3, atf3 and pu.1 as important transcription factors. Brain Behav. Immun. 2011, 25, 1162–1169. [Google Scholar] [CrossRef]
  80. Yoshino, Y.; Kawabe, K.; Yamazaki, K.; Watanabe, S.; Numata, S.; Mori, Y.; Yoshida, T.; Iga, J.; Ohmori, T.; Ueno, S.-i. Elevated trem2 mrna expression in leukocytes in schizophrenia but not major depressive disorder. J. Neural Transm. 2016, 123, 637–641. [Google Scholar] [CrossRef]
  81. Bonartsev, P.D.; Brusov, O.S.; Morozova, M.A.; Tsutsul’kovskaia, M. Electron-microscopic study of typical lymphocytes and atypical cells of peripheral blood during the treatment of patients with schizophrenia. Zhurnal Nevropatol. I Psikhiatrii Im. SS Korsakova 2009, 109, 54–58. [Google Scholar]
  82. Krause, D.; Wagner, J.; Matz, J.; Weidinger, E.; Obermeier, M.; Riedel, M.; Gruber, R.; Schwarz, M.; Mueller, N. Monocytic hla dr antigens in schizophrenic patients. Neurosci. Res. 2012, 72, 87–93. [Google Scholar] [CrossRef] [PubMed]
  83. Kowalski, J.; Blada, P.; Kucia, K.; Madej, A.; Herman, Z.S. Neuroleptics normalize increased release of interleukin-1β and tumor necrosis factor-α from monocytes in schizophrenia. Schizophr. Res. 2001, 50, 169–175. [Google Scholar] [CrossRef] [PubMed]
  84. Krause, D.L.; Wagner, J.K.; Wildenauer, A.; Matz, J.; Weidinger, E.; Riedel, M.; Obermeier, M.; Gruber, R.; Schwarz, M.; Müller, N. Intracellular monocytic cytokine levels in schizophrenia show an alteration of il-6. Eur. Arch. Psychiatry Clin. Neurosci. 2012, 262, 393–401. [Google Scholar] [CrossRef]
  85. Uranova, N.A.; Bonartsev, P.D.; Androsova, L.V.; Rakhmanova, V.I.; Kaleda, V.G. Impaired monocyte activation in schizophrenia: Ultrastructural abnormalities and increased il-1β production. Eur. Arch. Psychiatry Clin. Neurosci. 2017, 267, 417–426. [Google Scholar] [CrossRef] [PubMed]
  86. 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. [Google Scholar] [CrossRef]
  87. Li, N.; Li, Y.; Huang, J.; Zhang, P.; Tong, J.; Chen, S.; Cui, Y.; Tan, S.; Wang, Z.; Yang, F.; et al. Dysfunctional monocytic toll-like receptor 4 signaling pathway and cognitive deficits in chronic schizophrenia patients with tardive dyskinesia. Neurosci. Lett. 2022, 777, 136581. [Google Scholar] [CrossRef]
  88. Chen, S.; Tian, L.; Chen, N.; Xiu, M.-H.; Wang, Z.-R.; Wang, Y.-C.; Tan, Y.-L.; Wang, C.-Y. More dampened monocytic toll-like receptor 4 response to lipopolysaccharide and its association with cognitive function in chinese han first-episode patients with schizophrenia. Schizophr. Res. 2019, 206, 300–306. [Google Scholar] [CrossRef]
  89. Zaki, J.K.; Lago, S.G.; Rustogi, N.; Gangadin, S.S.; Benacek, J.; van Rees, G.F.; Haenisch, F.; Broek, J.A.; Suarez-Pinilla, P.; Ruland, T.; et al. Diagnostic model development for schizophrenia based on peripheral blood mononuclear cell subtype-specific expression of metabolic markers. Transl. Psychiatry 2022, 12, 457. [Google Scholar] [CrossRef]
  90. Weber, N.S.; Gressitt, K.L.; Cowan, D.N.; Niebuhr, D.W.; Yolken, R.H.; Severance, E.G. Monocyte activation detected prior to a diagnosis of schizophrenia in the us military new onset psychosis project (mnopp). Schizophr. Res. 2018, 197, 465–469. [Google Scholar] [CrossRef]
  91. Aichholzer, M.; Gangadin, S.S.; Sommer, I.E.C.; Wijkhuis, A.; de Witte, L.D.; Kahn, R.S.; Bahn, S.; Drexhage, H.A.; Schiweck, C. Inflammatory monocyte gene signature predicts beneficial within group effect of simvastatin in patients with schizophrenia spectrum disorders in a secondary analysis of a randomized controlled trial. Brain Behav. Immun.—Health 2022, 26, 100551. [Google Scholar] [CrossRef]
  92. Gross, A.; Joffe, G.; Joutsiniemi, S.-L.; Nyberg, P.; Rimón, R.; Appelberg, B. Decreased production of reactive oxygen species by blood monocytes caused by clozapine correlates with eeg slowing in schizophrenic patients. Neuropsychobiology 2003, 47, 73–77. [Google Scholar] [CrossRef]
  93. Tomasik, J.; Schwarz, E.; Lago, S.G.; Rothermundt, M.; Leweke, F.M.; van Beveren, N.J.M.; Guest, P.C.; Rahmoune, H.; Steiner, J.; Bahn, S. Pretreatment levels of the fatty acid handling proteins h-fabp and cd36 predict response to olanzapine in recent-onset schizophrenia patients. Brain Behav. Immun. 2016, 52, 178–186. [Google Scholar] [CrossRef] [PubMed]
  94. Jackson, A.J.; Miller, B.J. Meta-analysis of total and differential white blood cell counts in schizophrenia. Acta Psychiatr. Scand. 2019, 142, 18–26. [Google Scholar] [CrossRef]
  95. Karageorgiou, V.; Milas, G.P.; Michopoulos, I. Neutrophil-to-lymphocyte ratio in schizophrenia: A systematic review and meta-analysis. Schizophr. Res. 2019, 206, 4–12. [Google Scholar] [CrossRef]
  96. Yan, J.; Chen, Y.; Ju, P.; Gao, J.; Zhang, L.; Li, J.; Wang, K.; Zhang, J.; Li, C.; Xia, Q.; et al. Network association of biochemical and inflammatory abnormalities with psychiatric symptoms in first-episode schizophrenia patients. Front. Psychiatr. 2022, 13, 834539. [Google Scholar] [CrossRef] [PubMed]
  97. Núñez, C.; Stephan-Otto, C.; Usall, J.; Bioque, M.; Lobo, A.; González-Pinto, A.; Pina-Camacho, L.; Vieta, E.; Castro-Fornieles, J.; Rodriguez-Jimenez, R.; et al. Neutrophil count is associated with reduced gray matter and enlarged ventricles in first-episode psychosis. Schizophr. Bull. 2019, 45, 846–858. [Google Scholar] [CrossRef] [PubMed]
  98. Garcia-Rizo, C.; Casanovas, M.; Fernandez-Egea, E.; Oliveira, C.; Meseguer, A.; Cabrera, B.; Mezquida, G.; Bioque, M.; Kirkpatrick, B.; Bernardo, M. Blood cell count in antipsychotic-naive patients with non-affective psychosis. Early Interv. Psychiatry 2017, 13, 95–100. [Google Scholar] [CrossRef] [PubMed]
  99. Wilke, I.; Arolt, V.; Rothermundt, M.; Weitzsch, C.; Hornberg, M.; Kirchner, H. Investigations of cytokine production in whole blood cultures of paranoid and residual schizophrenic patients. Eur. Arch. Psychiatry Clin. Neurosci. 1996, 246, 279–284. [Google Scholar] [CrossRef] [PubMed]
  100. Šagud, M.; Madžarac, Z.; Nedic Erjavec, G.; Šimunović Filipčić, I.; Mikulić, F.L.; Rogić, D.; Bradaš, Z.; Bajs Janović, M.; Pivac, N. The associations of neutrophil-lymphocyte, platelet-lymphocyte, monocyte-lymphocyte ratios and immune-inflammation index with negative symptoms in patients with schizophrenia. Biomolecules 2023, 13, 297. [Google Scholar] [CrossRef]
  101. Zhu, X.; Zhou, J.; Zhu, Y.; Yan, F.; Han, X.; Tan, Y.; Li, R. Neutrophil/lymphocyte, platelet/lymphocyte and monocyte/lymphocyte ratios in schizophrenia. Australas. Psychiatry 2021, 30, 95–99. [Google Scholar] [CrossRef] [PubMed]
  102. Dadouli, K.; Janho, M.B.; Hatziefthimiou, A.; Voulgaridi, I.; Piaha, K.; Anagnostopoulos, L.; Ntellas, P.; Mouchtouri, V.A.; Bonotis, K.; Christodoulou, N.; et al. Neutrophil-to-lymphocyte, monocyte-to-lymphocyte, platelet-to-lymphocyte ratio and systemic immune-inflammatory index in different states of bipolar disorder. Brain Sci. 2022, 12, 1034. [Google Scholar] [CrossRef]
  103. Fusar-Poli, L.; Amerio, A.; Cimpoesu, P.; Grimaldi Filioli, P.; Natale, A.; Zappa, G.; Aguglia, E.; Amore, M.; Serafini, G.; Aguglia, A. Gender differences in complete blood count and inflammatory ratios among patients with bipolar disorder. Brain Sci. 2021, 11, 363. [Google Scholar] [CrossRef]
  104. Fusar-Poli, L.; Natale, A.; Amerio, A.; Cimpoesu, P.; Grimaldi Filioli, P.; Aguglia, E.; Amore, M.; Serafini, G.; Aguglia, A. Neutrophil-to-lymphocyte, platelet-to-lymphocyte and monocyte-to-lymphocyte ratio in bipolar disorder. Brain Sci. 2021, 11, 58. [Google Scholar] [CrossRef] [PubMed]
  105. Marazziti, D.; Torrigiani, S.; Carbone, M.G.; Mucci, F.; Flamini, W.; Ivaldi, T.; Dell’Osso, L. Neutrophil/lymphocyte, platelet/lymphocyte, and monocyte/lymphocyte ratios in mood disorders. Curr. Med. Chem. 2022, 29, 5758–5781. [Google Scholar] [CrossRef] [PubMed]
  106. Bulut, N.S.; Yorguner, N.; Çarkaxhiu Bulut, G. The severity of inflammation in major neuropsychiatric disorders: Comparison of neutrophil–lymphocyte and platelet–lymphocyte ratios between schizophrenia, bipolar mania, bipolar depression, major depressive disorder, and obsessive compulsive disorder. Nord. J. Psychiatry 2021, 75, 624–632. [Google Scholar] [CrossRef]
  107. Semiz, M.; Yildirim, O.; Canan, F.; Demir, S.; Hasbek, E.; Tuman, T.C.; Kayka, N.; Tosun, M. Elevated neutrophil/lymphocyte ratio in patients with schizophrenia. Psychiatr. Danub. 2014, 26, 220–225. [Google Scholar]
  108. Kulaksizoglu, B.; Kulaksizoglu, S. Relationship between neutrophil/lymphocyte ratio with oxidative stress and psychopathology in patients with schizophrenia. Neuropsychiatr. Dis. Treat. 2016, 12, 1999–2005. [Google Scholar] [CrossRef] [PubMed]
  109. Kovács, M.Á.; Tényi, T.; Kugyelka, R.; Prenek, L.; Hau, L.; Magyar, É.E.; Herold, R.; Balogh, P.; Simon, D. Elevated osteopontin and interferon gamma serum levels and increased neutrophil-to-lymphocyte ratio are associated with the severity of symptoms in schizophrenia. Front. Psychiatr. 2020, 10, 996. [Google Scholar] [CrossRef]
  110. Zhou, X.; Wang, X.; Li, R.; Yan, J.; Xiao, Y.; Li, W.; Shen, H. Neutrophil-to-lymphocyte ratio is independently associated with severe psychopathology in schizophrenia and is changed by antipsychotic administration: A large-scale cross-sectional retrospective study. Front. Psychiatr. 2020, 11, 581061. [Google Scholar] [CrossRef]
  111. Bioque, M.; Catarina Matias-Martins, A.; Llorca-Bofí, V.; Mezquida, G.; Cuesta, M.J.; Vieta, E.; Amoretti, S.; Lobo, A.; González-Pinto, A.; Moreno, C.; et al. Neutrophil to lymphocyte ratio in patients with a first episode of psychosis: A two-year longitudinal follow-up study. Schizophr. Bull. 2022, 48, 1327–1335. [Google Scholar] [CrossRef]
  112. Bustan, Y.; Drapisz, A.; Ben Dor, D.H.; Avrahami, M.; Schwartz-Lifshitz, M.; Weizman, A.; Barzilay, R. Elevated neutrophil to lymphocyte ratio in non-affective psychotic adolescent inpatients: Evidence for early association between inflammation and psychosis. Psychiatry Res. 2018, 262, 149–153. [Google Scholar] [CrossRef] [PubMed]
  113. Özdin, S.; Böke, Ö. Neutrophil/lymphocyte, platelet/lymphocyte and monocyte/lymphocyte ratios in different stages of schizophrenia. Psychiatry Res. 2019, 271, 131–135. [Google Scholar] [CrossRef] [PubMed]
  114. Balcioglu, Y.H.; Kirlioglu, S.S. C-reactive protein/albumin and neutrophil/albumin ratios as novel inflammatory markers in patients with schizophrenia. Psychiatry Investig. 2020, 17, 902–910. [Google Scholar] [CrossRef] [PubMed]
  115. Mijovic, A.; MacCabe, J.H. Clozapine-induced agranulocytosis. Ann. Hematol. 2020, 99, 2477–2482. [Google Scholar] [CrossRef] [PubMed]
  116. Labonté, C.; Zhand, N.; Park, A.; Harvey, P.D. Complete blood count inflammatory markers in treatment-resistant schizophrenia: Evidence of association between treatment responsiveness and levels of inflammation. Psychiatry Res. 2022, 308, 114382. [Google Scholar] [CrossRef] [PubMed]
  117. Issidorides, M.R.; Stefanis, C.N.; Varsou, E.; Katsorchis, T. Altered chromatin ultrastructure in neutrophils of schizophrenics. Nature 1975, 258, 612–614. [Google Scholar] [CrossRef] [PubMed]
  118. De Campos-Carli, S.M.; Araújo, M.S.; de Oliveira Silveira, A.C.; de Rezende, V.B.; Rocha, N.P.; Ferretjans, R.; Ribeiro-Santos, R.; Teixeira-Carvalho, A.; Martins-Filho, O.A.; Berk, M.; et al. Cannabinoid receptors on peripheral leukocytes from patients with schizophrenia: Evidence for defective immunomodulatory mechanisms. J. Psychiatr. Res. 2017, 87, 44–52. [Google Scholar] [CrossRef] [PubMed]
  119. Rwegellera, G.G.; Fernando, K.A.; Okong’o, O. Bactericidal activity of neutrophils of schizophrenic patients. Med. J. Zamb. 1982, 16, 21–22. [Google Scholar]
  120. Shcherbakova, I.V.; Neshkova, E.A.; Dotsenko, V.L.; Kozlov, L.V.; Mishin, A.A.; Platonova, T.P.; Shcherbakova, E.G.; Iarovaia, G.A. Activation of kallikrein-kinin system, degranulating activity of neutrophils and blood-brain barrier in schizophrenia. Zhurnal Nevropatol. I Psikhiatrii Im. SS Korsakova 1998, 98, 38–41. [Google Scholar]
  121. Tsavaris, N.; Konstantopoulos, K.; Vaidakis, S.; Koumakis, K.; Pangalis, G. Cytochemical determination of monoamine oxidase activity in lymphocytes and neutrophils of schizophrenic patients. Haematologia 1995, 26, 143–146. [Google Scholar]
  122. Srivastava, N.; Barthwal, M.; Dalal, P.; Agarwal, A.; Nag, D.; Srimal, R.; Seth, P.; Dikshit, M. Nitrite content and antioxidant enzyme levels in the blood of schizophrenia patients. Psychopharmacology 2001, 158, 140–145. [Google Scholar] [CrossRef] [PubMed]
  123. Melamed, Y.; Sirota, P.; Dicker, D.R.; Fishman, P. Superoxide anion production by neutrophils derived from peripheral blood of schizophrenic patients. Psychiatry Res. 1998, 77, 29–34. [Google Scholar] [CrossRef] [PubMed]
  124. Sirota, P.; Gavrieli, R.; Wolach, B. Overproduction of neutrophil radical oxygen species correlates with negative symptoms in schizophrenic patients: Parallel studies on neutrophil chemotaxis, superoxide production and bactericidal activity. Psychiatry Res. 2003, 121, 123–132. [Google Scholar] [CrossRef] [PubMed]
  125. Karpiński, P.; Frydecka, D.; Sąsiadek, M.M.; Misiak, B. Reduced number of peripheral natural killer cells in schizophrenia but not in bipolar disorder. Brain Behav. Immun. 2016, 54, 194–200. [Google Scholar] [CrossRef] [PubMed]
  126. Karpiński, P.; Samochowiec, J.; Frydecka, D.; Sąsiadek, M.M.; Misiak, B. Further evidence for depletion of peripheral blood natural killer cells in patients with schizophrenia: A computational deconvolution study. Schizophr. Res. 2018, 201, 243–248. [Google Scholar] [CrossRef]
  127. Sperner-Unterweger, B.; Whitworth, A.; Kemmler, G.; Hilbe, W.; Thaler, J.; Weiss, G.; Fleischhacker, W.W. T-cell subsets in schizophrenia: A comparison between drug-naive first episode patients and chronic schizophrenic patients. Schizophr. Res. 1999, 38, 61–70. [Google Scholar] [CrossRef] [PubMed]
  128. Rudolf, S.; Schlenke, P.; Broocks, A.; Peters, M.; Rothermundt, M.; Arolt, V.; Kirchner, H. Search for atypical lymphocytes in schizophrenia. World J. Biol. Psychiatry 2004, 5, 33–37. [Google Scholar] [CrossRef]
  129. Abdeljaber, M.H.; Nair, M.P.N.; Schork, M.A.; Schwartz, S.A. Depressed natural killer cell activity in schizophrenic patients. Immunol. Investig. 1994, 23, 259–268. [Google Scholar] [CrossRef] [PubMed]
  130. DeLisi, L.E.; Ortaldo, J.R.; Maluish, A.E.; Wyatt, R.J. Deficient natural killer cell (nk) activity and macrophage functioning in schizophrenic patients. J. Neural Transm. 1983, 58, 99–106. [Google Scholar] [CrossRef]
  131. McDaniel, J.; Jewart, R.; Eccard, M.; Pollard, W.; Caudle, J.; Stipetic, M.; Risby, E.; Lewine, R.; Risch, S. Natural killer cell activity in schizophrenia and schizoaffective disorder: A pilot study. Schizophr. Res. 1992, 8, 125–128. [Google Scholar] [CrossRef]
  132. Yovel, G.; Sirota, P.; Mazeh, D.; Shakhar, G.; Rosenne, E.; Ben-Eliyahu, S. Higher natural killer cell activity in schizophrenic patients: The impact of serum factors, medication, and smoking. Brain Behav. Immun. 2000, 14, 153–169. [Google Scholar] [CrossRef] [PubMed]
  133. Tarantino, N.; Leboyer, M.; Bouleau, A.; Hamdani, N.; Richard, J.R.; Boukouaci, W.; Ching-Lien, W.; Godin, O.; Bengoufa, D.; Le Corvoisier, P.; et al. Natural killer cells in first-episode psychosis: An innate immune signature? Mol. Psychiatry 2021, 26, 5297–5306. [Google Scholar] [CrossRef] [PubMed]
  134. Steiner, J.; Westphal, S.; Schroeter, M.L.; Schiltz, K.; Jordan, W.; Müller, U.J.; Bernstein, H.-G.; Bogerts, B.; Schmidt, R.E.; Jacobs, R. Increased s100b+ nk cell counts in acutely ill schizophrenia patients are correlated with the free cortisol index, but not with s100b serum levels. Brain Behav. Immun. 2012, 26, 564–567. [Google Scholar] [CrossRef] [PubMed]
  135. Borovcanin, M.M.; Minic Janicijevic, S.; Jovanovic, I.P.; Gajovic, N.M.; Jurisevic, M.M.; Arsenijevic, N.N. Type 17 immune response facilitates progression of inflammation and correlates with cognition in stable schizophrenia. Diagnostics 2020, 10, 926. [Google Scholar] [CrossRef] [PubMed]
  136. Chandley, M.J.; Miller, M.N.; Kwasigroch, C.N.; Wilson, T.D.; Miller, B.E. Increased antibodies for the α7 subunit of the nicotinic receptor in schizophrenia. Schizophr. Res. 2009, 109, 98–101. [Google Scholar] [CrossRef]
  137. Keshavarz, F.; Soltani, M.; Mokhtarian, K.; Beshkar, P.; Majidi, J.; Azadegan-Dehkordi, F.; Anjomshoa, M.; Bagheri, N. Autoantibodies against central nervous system antigens and the serum levels of il-32 in patients with schizophrenia. Neuroimmunomodulation 2022, 29, 493–499. [Google Scholar] [CrossRef]
  138. Shiwaku, H.; Nakano, Y.; Kato, M.; Takahashi, H. Detection of autoantibodies against gabaarα1 in patients with schizophrenia. Schizophr. Res. 2020, 216, 543–546. [Google Scholar] [CrossRef]
  139. Soltani, M.; Beshkar, P.; Mokhtarian, K.; Anjomshoa, M.; Mohammad Rezaei, M.; Azadegan-Dehkordi, F.; Mirzaei, Y.; Majidi, J.; Bagheri, N. A study of autoantibodies against some central nervous system antigens and the il-35 serum level in schizophrenia. Iran. J. Allergy Asthma Immunol. 2022, 21, 441–448. [Google Scholar] [CrossRef]
  140. Hallford, P.; Clair, D.S.; Halley, L.; Mustard, C.; Wei, J. A study of type-1 diabetes associated autoantibodies in schizophrenia. Schizophr. Res. 2016, 176, 186–190. [Google Scholar] [CrossRef]
  141. Hoffmann, C.; Stevens, J.; Zong, S.; van Kruining, D.; Saxena, A.; Küçükali, C.İ.; Tüzün, E.; Yalçınkaya, N.; De Hert, M.; González-Vioque, E.; et al. Alpha7 acetylcholine receptor autoantibodies are rare in sera of patients diagnosed with schizophrenia or bipolar disorder. PLoS ONE 2018, 13, e0208412. [Google Scholar] [CrossRef]
  142. Zhou, D.; Xie, C.; Li, X.; Song, N.; Kou, Z.; Zhang, T.; Yuan, T.-F. Rare presence of autoantibodies targeting to nmda and gabaa receptors in schizophrenia patients. Schizophr. Res. 2022, 249, 93–97. [Google Scholar] [CrossRef] [PubMed]
  143. Ermakov, E.A.; Ivanova, S.A.; Buneva, V.N.; Nevinsky, G.A. Blood-derived rna- and microrna-hydrolyzing igg antibodies in schizophrenia patients. Biochemistry 2018, 83, 507–526. [Google Scholar] [CrossRef] [PubMed]
  144. Ermakov, E.A.; Smirnova, L.P.; Parkhomenko, T.A.; Dmitrenok, P.S.; Krotenko, N.M.; Fattakhov, N.S.; Bokhan, N.A.; Semke, A.V.; Ivanova, S.A.; Buneva, V.N.; et al. DNA-hydrolysing activity of igg antibodies from the sera of patients with schizophrenia. Open Biol. 2015, 5, 150064. [Google Scholar] [CrossRef] [PubMed]
  145. Kim, J.J.; Lee, S.J.; Toh, K.Y.; Lee, C.U.; Lee, C.; Paik, I.H. Identification of antibodies to heat shock proteins 90 kda and 70 kda in patients with schizophrenia. Schizophr. Res. 2001, 52, 127–135. [Google Scholar] [CrossRef] [PubMed]
  146. Wang, X.-F.; Wang, D.; Zhu, W.; Delrahim, K.K.; Dolnak, D.; Hyman Rapaport, M. Studies characterizing 60 kda autoantibodies in subjects with schizophrenia. Biol. Psychiatry 2003, 53, 361–375. [Google Scholar] [CrossRef]
  147. Endres, D.; von Zedtwitz, K.; Matteit, I.; Bünger, I.; Foverskov-Rasmussen, H.; Runge, K.; Feige, B.; Schlump, A.; Maier, S.; Nickel, K.; et al. Spectrum of novel anti–central nervous system autoantibodies in the cerebrospinal fluid of 119 patients with schizophreniform and affective disorders. Biol. Psychiatry 2022, 92, 261–274. [Google Scholar] [CrossRef]
  148. Klyushnik, T.P.; Danilovskaya, E.V.; Vatolkina, O.E.; Turkova, I.L.; Tsutsul’kovskaya, M.Y.; Orlova, V.A.; Kaleda, V.G. Changes in serum levels of autoantibody to nerve growth factor in patients with schizophrenia. Neurosci. Behav. Physiol. 1999, 29, 355–357. [Google Scholar] [CrossRef]
  149. Sugiura, M.; Yokoi, Y.; Maruyama, S.; Ishido, T.; Tokunaga, Y.; Sasaki, K. Detection of anti-cerebral autoantibodies in schizophrenia and alzheimer’s disease. J. Clin. Lab. Immunol. 1989, 28, 1–3. [Google Scholar]
  150. Tanaka, S.; Matsunaga, H.; Kimura, M.; Tatsumi, K.-i.; Hidaka, Y.; Takano, T.; Uema, T.; Takeda, M.; Amino, N. Autoantibodies against four kinds of neurotransmitter receptors in psychiatric disorders. J. Neuroimmunol. 2003, 141, 155–164, Erratum in J. Neuroimmunol. 2003, 144, 148. [Google Scholar] [CrossRef]
  151. Jin, S.-Z.; Wu, N.; Xu, Q.; Zhang, X.; Ju, G.-Z.; Law, M.H.; Wei, J. A study of circulating gliadin antibodies in schizophrenia among a chinese population. Schizophr. Bull. 2012, 38, 514–518. [Google Scholar] [CrossRef]
  152. Kelly, D.L.; Demyanovich, H.K.; Eaton, W.W.; Cascella, N.; Jackson, J.; Fasano, A.; Carpenter, W.T. Anti gliadin antibodies (aga igg) related to peripheral inflammation in schizophrenia. Brain Behav. Immun. 2018, 69, 57–59. [Google Scholar] [CrossRef]
  153. Li, C.; Whelan, R.; Yang, H.; Jiang, Y.; Qiu, C.; Meng, Q.; Wei, J. Anti-tsnare1 igg plasma levels differ by sex in patients with schizophrenia in a chinese population. FEBS Open Bio 2019, 9, 1705–1712. [Google Scholar] [CrossRef] [PubMed]
  154. Liu, S.; Zhang, X.; Wang, J.; Yang, H.; Jiang, Y.; Qiu, C.; Meng, Q. Analysis of plasma autoantibodies for inflammatory cytokines in patients with first-episode schizophrenia among a chinese population. J. Neuroimmunol. 2020, 341, 577165. [Google Scholar] [CrossRef]
  155. Okusaga, O.; Yolken, R.H.; Langenberg, P.; Sleemi, A.; Kelly, D.L.; Vaswani, D.; Giegling, I.; Hartmann, A.M.; Konte, B.; Friedl, M.; et al. Elevated gliadin antibody levels in individuals with schizophrenia. World J. Biol. Psychiatry 2013, 14, 509–515. [Google Scholar] [CrossRef] [PubMed]
  156. Steiner, J.; Jacobs, R.; Panteli, B.; Brauner, M.; Schiltz, K.; Bahn, S.; Herberth, M.; Westphal, S.; Gos, T.; Walter, M.; et al. Acute schizophrenia is accompanied by reduced t cell and increased b cell immunity. Eur. Arch. Psychiatry Clin. Neurosci. 2010, 260, 509–518. [Google Scholar] [CrossRef] [PubMed]
  157. McAllister, C.G. Increased numbers of cd5 b lymphocytes in schizophrenic patients. Arch. Gen. Psychiatry 1989, 46, 890. [Google Scholar] [CrossRef] [PubMed]
  158. Printz, D.J.; Strauss, D.H.; Goetz, R.; Sadiq, S.; Malaspina, D.; Krolewski, J.; Gorman, J.M. Elevation of cd5+ b lymphocytes in schizophrenia. Biol. Psychiatry 1999, 46, 110–118. [Google Scholar] [CrossRef]
  159. Ganguli, R.; Rabin, B.S. Cd5 positive b lymphocytes in schizophrenia: No alteration in numbers or percentage as compared with control subjects. Psychiatry Res. 1993, 48, 69–78. [Google Scholar] [CrossRef]
  160. Müller, N.; Hofschuster, E.; Ackenheil, M.; Eckstein, R. T-cells and psychopathology in schizophrenia: Relationship to the outcome of neuroleptic therapy. Acta Psychiatr. Scand. 1993, 87, 66–71. [Google Scholar] [CrossRef] [PubMed]
  161. Saĭfulina, N.A.; Arkhipov, G.S.; Stepanov, A.F.; Gusev Iu, A. T- and b-lymphocytes and their clinico-pathogenetic significance in patients with paranoid schizophrenia. Zhurnal Nevropatol. I Psikhiatrii Im. SS Korsakova 1990, 90, 87–90. [Google Scholar]
  162. Achiron, A.; Noy, S.; Pras, E.; Lereya, J.; Hermesh, H.; Laor, N. T-cell subsets in acute psychotic schizophrenic patients. Biol. Psychiatry 1994, 35, 27–31. [Google Scholar] [CrossRef] [PubMed]
  163. Sahbaz, C.; Zibandey, N.; Kurtulmus, A.; Duran, Y.; Gokalp, M.; Kırpınar, I.; Sahin, F.; Guloksuz, S.; Akkoc, T. Reduced regulatory t cells with increased proinflammatory response in patients with schizophrenia. Psychopharmacology 2020, 237, 1861–1871. [Google Scholar] [CrossRef] [PubMed]
  164. Theodoropoulou, S.; Spanakos, G.; Baxevanis, C.N.; Economou, M.; Gritzapis, A.D.; Papamichail, M.P.; Stefanis, C.N. Cytokine serum levels, autologous mixed lymphocyte reaction and surface marker analysis in never medicated and chronically medicated schizophrenic patients. Schizophr. Res. 2001, 47, 13–25. [Google Scholar] [CrossRef] [PubMed]
  165. Bessler, H.; Levental, Z.; Karp, L.; Modai, I.; Djaldetti, M.; Weizman, A. Cytokine production in drug-free and neuroleptic-treated schizophrenic patients. Biol. Psychiatry 1995, 38, 297–302. [Google Scholar] [CrossRef] [PubMed]
  166. Villemain, F.; Chatenoud, L.; Galinowski, A.; Homo-Delarche, F.; Ginestet, D.; Loo, H.; Zarifian, E.; Bach, J.F. Aberrant t cell-mediated immunity in untreated schizophrenic patients: Deficient interleukin-2 production. Am. J. Psychiatry 1989, 146, 609–616. [Google Scholar] [PubMed]
  167. Yang Zhang, X.; Feng Zhou, D.; Yuan Cao, L.; Yan Zhang, P.; Ying Wu, G. Decreased production of interleukin-2 (il-2), il-2 secreting cells and cd4+ cells in medication-free patients with schizophrenia. J. Psychiatr. Res. 2002, 36, 331–336. [Google Scholar] [CrossRef] [PubMed]
  168. Ding, M.; Song, X.; Zhao, J.; Gao, J.; Li, X.; Yang, G.; Wang, X.; Harrington, A.; Fan, X.; Lv, L. Activation of th17 cells in drug naïve, first episode schizophrenia. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2014, 51, 78–82. [Google Scholar]
  169. Kelly, D.L.; Li, X.; Kilday, C.; Feldman, S.; Clark, S.; Liu, F.; Buchanan, R.W.; Tonelli, L.H. Increased circulating regulatory t cells in medicated people with schizophrenia. Psychiatry Res. 2018, 269, 517–523. [Google Scholar] [CrossRef]
  170. Varun, C.N.; Venkataswamy, M.M.; Ravikumar, R.; Nagaraju, R.; Debnath, M.; Varambally, S.; Venkatasubramanian, G.; Ravi, V. Th17 and mait cell mediated inflammation in antipsychotic free schizophrenia patients. Schizophr. Res. 2019, 212, 47–53. [Google Scholar] [CrossRef] [PubMed]
  171. Vergaelen, E.; Schiweck, C.; Van Steeland, K.; Counotte, J.; Veling, W.; Swillen, A.; Drexhage, H.; Claes, S. A pilot study on immuno-psychiatry in the 22q11.2 deletion syndrome: A role for th17 cells in psychosis? Brain Behav. Immun. 2018, 70, 88–95. [Google Scholar] [CrossRef]
  172. Amerio, A.; Magnani, L.; Arduino, G.; Fesce, F.; de Filippis, R.; Parise, A.; Costanza, A.; Nguyen, K.D.; Saverino, D.; De Berardis, D.; et al. Immunomodulatory effects of clozapine: More than just a side effect in schizophrenia. Curr. Neuropharmacol. 2023; in press. [Google Scholar]
  173. Boneberg, E.-M.; von Seydlitz, E.; Pröpster, K.; Watzl, H.; Rockstroh, B.; Illges, H. D3 dopamine receptor mrna is elevated in t cells of schizophrenic patients whereas d4 dopamine receptor mrna is reduced in cd4+-t cells. J. Neuroimmunol. 2006, 173, 180–187. [Google Scholar] [CrossRef] [PubMed]
  174. Brito-Melo, G.E.A.; Nicolato, R.; de Oliveira, A.C.P.; Menezes, G.B.; Lélis, F.J.N.; Avelar, R.S.; Sá, J.; Bauer, M.E.; Souza, B.R.; Teixeira, A.L.; et al. Increase in dopaminergic, but not serotoninergic, receptors in t-cells as a marker for schizophrenia severity. J. Psychiatr. Res. 2012, 46, 738–742. [Google Scholar] [CrossRef]
  175. Hu, A.; Li, F.; Guo, L.; Zhao, X.; Xiang, X. Mitochondrial damage of lymphocytes in patients with acute relapse of schizophrenia: A correlational study with efficacy and clinical symptoms. Neuropsychiatr. Dis. Treat. 2022, 18, 2455–2466. [Google Scholar] [CrossRef]
  176. Luo, C.; Pi, X.; Hu, N.; Wang, X.; Xiao, Y.; Li, S.; Sweeney, J.A.; Bishop, J.R.; Gong, Q.; Xie, D.; et al. Subtypes of schizophrenia identified by multi-omic measures associated with dysregulated immune function. Mol. Psychiatry 2021, 26, 6926–6936. [Google Scholar] [CrossRef] [PubMed]
  177. Lynall, M.-E.; Soskic, B.; Hayhurst, J.; Schwartzentruber, J.; Levey, D.F.; Pathak, G.A.; Polimanti, R.; Gelernter, J.; Stein, M.B.; Trynka, G.; et al. Genetic variants associated with psychiatric disorders are enriched at epigenetically active sites in lymphoid cells. Nat. Commun. 2022, 13, 6102. [Google Scholar] [CrossRef]
  178. Farsi, Z.; Sheng, M. Molecular mechanisms of schizophrenia: Insights from human genetics. Curr. Opin. Neurobiol. 2023, 81, 102731. [Google Scholar] [CrossRef] [PubMed]
  179. Sheridan, S.D.; Horng, J.E.; Perlis, R.H. Patient-Derived In Vitro Models of Microglial Function and Synaptic Engulfment in Schizophrenia. Biol. Psychiatry 2022, 92, 470–479. [Google Scholar] [CrossRef]
  180. Comer, A.L.; Jinadasa, T.; Sriram, B.; Phadke, R.A.; Kretsge, L.N.; Nguyen, T.P.H.; Antognetti, G.; Gilbert, J.P.; Lee, J.; Newmark, E.R.; et al. Increased expression of schizophrenia-associated gene C4 leads to hypoconnectivity of prefrontal cortex and reduced social interaction. PLoS Biol. 2020, 18, e3000604. [Google Scholar] [CrossRef]
  181. Sellgren, C.M.; Gracias, J.; Watmuff, B.; Biag, J.D.; Thanos, J.M.; Whittredge, P.B.; Fu, T.; Worringer, K.; Brown, H.E.; Wang, J.; et al. Increased synapse elimination by microglia in schizophrenia patient-derived models of synaptic pruning. Nat. Neurosci. 2019, 22, 374–385. [Google Scholar] [CrossRef]
  182. Park, G.H.; Noh, H.; Shao, Z.; Ni, P.; Qin, Y.; Liu, D.; Beaudreault, C.P.; Park, J.S.; Abani, C.P.; Park, J.M.; et al. Activated microglia cause metabolic disruptions in developmental cortical interneurons that persist in interneurons from individuals with schizophrenia. Nat. Neurosci. 2020, 23, 1352–1364. [Google Scholar] [CrossRef] [PubMed]
  183. Breitmeyer, R.; Vogel, S.; Heider, J.; Hartmann, S.M.; Wüst, R.; Keller, A.L.; Binner, A.; Fitzgerald, J.C.; Fallgatter, A.J.; Volkmer, H. Regulation of synaptic connectivity in schizophrenia spectrum by mutual neuron-microglia interaction. Commun. Biol. 2023, 6, 472. [Google Scholar] [CrossRef] [PubMed]
  184. Chauhan, P.; Kaur, G.; Prasad, R.; Singh, H. Pharmacotherapy of schizophrenia: Immunological aspects and potential role of immunotherapy. Expert Rev. Neurother. 2021, 21, 1441–1453. [Google Scholar] [CrossRef] [PubMed]
  185. Miller, B.J.; Goldsmith, D.R. Towards an Immunophenotype of Schizophrenia: Progress, Potential Mechanisms, and Future Directions. Neuropsychopharmacology 2017, 42, 299–317. [Google Scholar] [CrossRef] [PubMed]
  186. Bejerot, S.; Sigra Stein, S.; Welin, E.; Eklund, D.; Hylén, U.; Humble, M.B. Rituximab as an Adjunctive Treatment for Schizophrenia Spectrum Disorder or Obsessive-Compulsive Disorder: Two Open-Label Pilot Studies on Treatment-Resistant Patients. J. Psychiatr. Res. 2023, 158, 319–329. [Google Scholar] [CrossRef] [PubMed]
  187. Karbalaee, M.; Jameie, M.; Amanollahi, M.; TaghaviZanjani, F.; Parsaei, M.; Basti, F.A.; Mokhtari, S.; Moradi, K.; Ardakani, M.-R.K.; Akhondzadeh, S. Efficacy and Safety of Adjunctive Therapy with Fingolimod in Patients with Schizophrenia: A Randomized, Double-Blind, Placebo-Controlled Clinical Trial. Schizophr. Res. 2023, 254, 92–98. [Google Scholar] [CrossRef]
  188. Miller, B.J.; Dias, J.K.; Lemos, H.P.; Buckley, P.F. An Open-Label, Pilot Trial of Adjunctive Tocilizumab in Schizophrenia. J. Clin. Psychiatry 2016, 77, 13353. [Google Scholar] [CrossRef] [PubMed]
  189. Motamed, M.; Karimi, H.; Sanjari Moghaddam, H.; Taherzadeh Boroujeni, S.; Sanatian, Z.; Hasanzadeh, A.; Khodaei Ardakani, M.-R.; Akhondzadeh, S. Risperidone Combination Therapy with Adalimumab for Treatment of Chronic Schizophrenia. Int. Clin. Psychopharmacol. 2022, 37, 92–101. [Google Scholar] [CrossRef]
  190. Levine, J.; Gutman, J.; Feraro, R.; Levy, P.; Kimhi, R.; Leykin, I.; Deckmann, M.; Handzel, Z.T.; Shinitzky, M. Side effect profile of azathioprine in the treatment of chronic schizophrenic patients. Neuropsychobiology 1997, 36, 172–176. [Google Scholar] [CrossRef]
  191. Nitta, M.; Kishimoto, T.; Müller, N.; Weiser, M.; Davidson, M.; Kane, J.M.; Correll, C.U. Adjunctive Use of Nonsteroidal Anti-Inflammatory Drugs for Schizophrenia: A Meta-Analytic Investigation of Randomized Controlled Trials. Schizophr. Bull. 2013, 39, 1230–1241. [Google Scholar] [CrossRef]
  192. Chaudhry, I.B.; Husain, M.O.; Khoso, A.B.; Husain, M.I.; Buch, M.H.; Kiran, T.; Fu, B.; Bassett, P.; Qurashi, I.; ur Rahman, R.; et al. A Randomised Clinical Trial of Methotrexate Points to Possible Efficacy and Adaptive Immune Dysfunction in Psychosis. Transl. Psychiatry 2020, 10, 415. [Google Scholar] [CrossRef] [PubMed]
  193. Fan, X.; Song, X.; Zhao, M.; Jarskog, L.F.; Natarajan, R.; Shukair, N.; Freudenreich, O.; Henderson, D.C.; Goff, D.C. The Effect of Adjunctive Telmisartan Treatment on Psychopathology and Cognition in Patients with Schizophrenia. Acta Psychiatr. Scand. 2017, 136, 465–472. [Google Scholar] [CrossRef] [PubMed]
  194. Sagheddu, C.; Melis, M.; Muntoni, A.L.; Pistis, M. Repurposing Peroxisome Proliferator-Activated Receptor Agonists in Neurological and Psychiatric Disorders. Pharmaceuticals 2021, 14, 1025. [Google Scholar] [CrossRef] [PubMed]
  195. Çakici, N.; van Beveren, N.J.M.; Judge-Hundal, G.; Koola, M.M.; Sommer, I.E.C. An update on the efficacy of anti-inflammatory agents for patients with schizophrenia: A meta-analysis. Psychol. Med. 2019, 49, 2307–2319. [Google Scholar] [CrossRef] [PubMed]
  196. Messina, A.; Concerto, C.; Rodolico, A.; Petralia, A.; Caraci, F.; Signorelli, M.S. Is It Time for a Paradigm Shift in the Treatment of Schizophrenia? The Use of Inflammation-Reducing and Neuroprotective Drugs—A Review. Brain Sci. 2023, 13, 957. [Google Scholar] [CrossRef]
  197. Girgis, R.R.; Ciarleglio, A.; Choo, T.; Haynes, G.; Bathon, J.M.; Cremers, S.; Kantrowitz, J.T.; Lieberman, J.A.; Brown, A.S. A Randomized, Double-Blind, Placebo-Controlled Clinical Trial of Tocilizumab, an Interleukin-6 Receptor Antibody, for Residual Symptoms in Schizophrenia. Neuropsychopharmacology 2017, 43, 1317–1323. [Google Scholar] [CrossRef]
  198. Hansen, N.; Malchow, B. Monoclonal Antibody Therapy in Autoantibody-Associated Psychotic Disorders and Schizophrenia: Narrative Review of Past and Current Clinical Trials. Psychiatr. Danub. 2023, 35, 8–15. [Google Scholar] [CrossRef]
  199. Garner, B.; Phillips, L.J.; Bendall, S.; Hetrick, S.E. Antiglucocorticoid and related treatments for psychosis. Cochrane Database Syst. Rev. 2016, 2016, CD006995. [Google Scholar] [CrossRef]
Figure 1. Immunological disturbances in the central nervous system (CNS) of patients with schizophrenia (SCZ). Region-specific immunological changes in CNS tissues of SCZ patients are characterized by (1) elevated expression of various activation markers of microglia (S100/A8, HLA-DR), decreased expression of neuroprotective quinolinic acid (QA), and increased microgliosis (IBA-1 density); (2) increased expression of macrophage markers (CD14 and CD163); (3) dynamic trafficking of various T cell populations (CD3/CD4/CD8); and (4) CD20+ B cell accumulation and altered B cell receptor (BCR) repertoire. Abbreviations: ACC: anterior cingulate cortex; CSF: cerebrospinal fluid; DPFC: dorsal prefrontal cortex; FC: frontal cortex; HP: hippocampus; SEZ: subependymal zone. Red font indicates discrepancies among studies.
Figure 1. Immunological disturbances in the central nervous system (CNS) of patients with schizophrenia (SCZ). Region-specific immunological changes in CNS tissues of SCZ patients are characterized by (1) elevated expression of various activation markers of microglia (S100/A8, HLA-DR), decreased expression of neuroprotective quinolinic acid (QA), and increased microgliosis (IBA-1 density); (2) increased expression of macrophage markers (CD14 and CD163); (3) dynamic trafficking of various T cell populations (CD3/CD4/CD8); and (4) CD20+ B cell accumulation and altered B cell receptor (BCR) repertoire. Abbreviations: ACC: anterior cingulate cortex; CSF: cerebrospinal fluid; DPFC: dorsal prefrontal cortex; FC: frontal cortex; HP: hippocampus; SEZ: subependymal zone. Red font indicates discrepancies among studies.
Cells 12 02099 g001
Figure 2. Peripheral immune alterations in patients with schizophrenia (SCZ). Major changes in immune cell types in blood samples of SCZ patients included: (1) Alterations in monocytes such as increased monocyte-to-lymphocyte and monocyte to HDL ratios (MLR and MHR), changes in expression of various immunometabolic markers (TREM1/2, HLA-DR, CD36, reactive oxygen species [ROS], GLUT1), and abnormalities in phagocytosis and cytokine production; (2) elevated expression of various neutrophil-associated markers such as neutrophil to lymphocyte ratio (NLR), oxidative stress, and phagocytosis; (3) presence of various autoantibody-producing pathogenic B cell clones, as well as increased numbers of different B cell subsets; and (4) increased activation profile of T cells (CD25), alterations in oxidative stress and T cell receptor [76] repertoire, and accumulation of immunoregulatory T lymphocyte populations, such as regulatory T (Treg), IL17-producing T-helper (Th17), and mucosal-associated invariant T (MAIT) cells. Red font indicates discrepancies among studies.
Figure 2. Peripheral immune alterations in patients with schizophrenia (SCZ). Major changes in immune cell types in blood samples of SCZ patients included: (1) Alterations in monocytes such as increased monocyte-to-lymphocyte and monocyte to HDL ratios (MLR and MHR), changes in expression of various immunometabolic markers (TREM1/2, HLA-DR, CD36, reactive oxygen species [ROS], GLUT1), and abnormalities in phagocytosis and cytokine production; (2) elevated expression of various neutrophil-associated markers such as neutrophil to lymphocyte ratio (NLR), oxidative stress, and phagocytosis; (3) presence of various autoantibody-producing pathogenic B cell clones, as well as increased numbers of different B cell subsets; and (4) increased activation profile of T cells (CD25), alterations in oxidative stress and T cell receptor [76] repertoire, and accumulation of immunoregulatory T lymphocyte populations, such as regulatory T (Treg), IL17-producing T-helper (Th17), and mucosal-associated invariant T (MAIT) cells. Red font indicates discrepancies among studies.
Cells 12 02099 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Nguyen, K.D.; Amerio, A.; Aguglia, A.; Magnani, L.; Parise, A.; Conio, B.; Serafini, G.; Amore, M.; Costanza, A. Microglia and Other Cellular Mediators of Immunological Dysfunction in Schizophrenia: A Narrative Synthesis of Clinical Findings. Cells 2023, 12, 2099. https://doi.org/10.3390/cells12162099

AMA Style

Nguyen KD, Amerio A, Aguglia A, Magnani L, Parise A, Conio B, Serafini G, Amore M, Costanza A. Microglia and Other Cellular Mediators of Immunological Dysfunction in Schizophrenia: A Narrative Synthesis of Clinical Findings. Cells. 2023; 12(16):2099. https://doi.org/10.3390/cells12162099

Chicago/Turabian Style

Nguyen, Khoa D., Andrea Amerio, Andrea Aguglia, Luca Magnani, Alberto Parise, Benedetta Conio, Gianluca Serafini, Mario Amore, and Alessandra Costanza. 2023. "Microglia and Other Cellular Mediators of Immunological Dysfunction in Schizophrenia: A Narrative Synthesis of Clinical Findings" Cells 12, no. 16: 2099. https://doi.org/10.3390/cells12162099

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

Article Metrics

Back to TopTop