Skip to main content
Schizophrenia Bulletin logoLink to Schizophrenia Bulletin
. 2018 Nov 8;45(5):1120–1133. doi: 10.1093/schbul/sby160

A Meta-analysis of Immune Parameters, Variability, and Assessment of Modal Distribution in Psychosis and Test of the Immune Subgroup Hypothesis

Toby Pillinger 1,#,, Emanuele F Osimo 2,3,4,#, Stefan Brugger 4,5,6, Valeria Mondelli 1,7, Robert A McCutcheon 1,4,5, Oliver D Howes 1,4,5,
PMCID: PMC6737479  EMSID: EMS82490  PMID: 30407606

Abstract

Immune parameters are elevated in psychosis, but it is unclear whether alterations are homogenous across patients or heterogeneity exists, consistent with the hypothesis that immune alterations are specific to a subgroup of patients. To address this, we examine whether antipsychotic-naïve first-episode psychosis patients exhibit greater variability in blood cytokines, C-reactive protein, and white cell counts compared with controls, and if group mean differences persist after adjusting for skewed data and potential confounds. Databases were searched for studies reporting levels of peripheral immune parameters. Means and variances were extracted and analyzed using multivariate meta-analysis of mean and variability of differences. Outcomes were (1) variability in patients relative to controls, indexed by variability ratio (VR) and coefficient of variation ratio (CVR); (2) mean differences indexed by Hedges g; (3) Modal distribution of raw immune parameter data using Hartigan’s unimodality dip test. Thirty-five studies reporting on 1263 patients and 1470 controls were included. Variability of interleukin-6 (IL6) (VR = 0.19), tumor necrosis factor-α (TNFα) (VR = 0.36), interleukin-1β (VR = 0.35), interleukin-4 (VR = 0.55), and interleukin-8 (VR = 0.28) was reduced in patients. Results persisted for IL6 and IL8 after mean-scaling. Ninety-four percent and one hundred percent of raw data were unimodally distributed in psychosis and controls, respectively. Mean levels of IL6 (g = 0.62), TNFα (g = 0.56), interferon-γ (IFNγ) (g = 0.32), transforming growth factor-β (g = 0.53), and interleukin-17 (IL17) (g = 0.48) were elevated in psychosis. Sensitivity analyses indicated this is unlikely explained by confounders for IL6, IFNγ, and IL17. These findings show elevated cytokines in psychosis after accounting for confounds, and that the hypothesis of an immune subgroup is not supported by the variability or modal distribution.

Keywords: inflammation, immune, psychosis, variability

Introduction

Schizophrenia and related psychotic disorders have a worldwide lifetime prevalence of approximately 1%.1 They are leading contributors to global disease burden, partly because of inadequate response to antipsychotic treatment in many patients.2 A greater understanding of illness pathophysiology is required to identify novel therapeutic targets, and develop biologically informed diagnoses.3 Converging lines of evidence, including genetic, postmortem, and preclinical data, suggest immune dysregulation may play a role in psychosis pathogenesis.4–22 Supporting this, meta-analyses have shown raised levels of soluble interleukin-2 receptor (sIL2R), interleukin-6 (IL6), interleukin-8 (IL8), interleukin-10 (IL10), interferon-γ (IFNγ), transforming growth factor-β (TGFβ), tumor necrosis factor-α (TNFα), C-reactive protein (CRP) and white cell counts in both antipsychotic-naïve first episode psychosis (FEP) and chronic medicated schizophrenia compared with controls, with large effect sizes.16–22

It has been hypothesized that there are biological subtypes of psychosis,3,23–25 with immune alterations seen only in a proportion of patients and potentially linked to poor response to treatment.26,27 In addition to inflating effect sizes for mean differences between patients and controls, the presence of an immune subgroup would be expected to lead to increased immune parameter variability in patients. We therefore set out to assess this by performing a meta-analysis of variability, as previously employed to examine regional brain structural variability in FEP.28 If immune alterations are seen only in a subgroup of patients then greater immune measure variability in patients relative to controls would be predicted, reflecting heterogeneity in immune dysregulation. Conversely, if immune alterations are a core component of the pathophysiology of psychosis, reduced immune variability in patients compared with controls would be predicted, reflecting homogeneity in immune dysregulation. This approach could however fail to identify subgroups of data contained within the overall data distribution (eg, a bimodal distribution). To address this, we also set out to examine distribution of raw immune parameter data in patients to assess for a multimodal distribution. The presence of latent immune subgroups within the healthy control group27,29 could also influence variability analyses, thus we also set out to assess for multimodality in controls.

Mean differences in immune parameters between patients and controls reported in previous meta-analyses16–22 could be influenced by physiological and environmental confounders, eg, body mass index (BMI),30,31 smoking,32 age,33 gender,34 hypothalamic–pituitary–adrenal (HPA) axis activity,35 and ethnicity.36 Moreover, mean meta-analytic differences may be influenced by data-skew. Many immune parameters are physiologically present at low concentrations, and poor assay sensitivity at these levels can result in a floor effect.37 This results in positive skew, potentially inflating group differences.38,39 Indeed, over half the data sets included in previous meta-analyses examining immune parameters in psychosis show evidence of significant skew,26,40–60 and the Cochrane Collaboration recommends that meta-analyses based on means are appropriate only for data that are at least approximately normally distributed.61 Thus, we also set out to assess if mean differences in immune parameters between patients and controls were robust to sensitivity analyses focusing on studies that matched for physiological/environmental confounds, and with skewed data removed.

To our knowledge, this article represents the first variability meta-analysis of immune parameters, the first study to examine distribution of multiple raw immune parameter data sets, and the first meta-analysis of mean differences to comprehensively examine the role of confounders and data-skew in individuals with psychosis compared with controls.

Methods

Selection Procedures

A systematic review was performed according to Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA)62 and Meta-Analysis of Observational Studies in Epidemiology (MOOSE)63 guidelines (supplementary eAppendix 1), following an a priori protocol (supplementary eAppendix 2). Two reviewers (TP and EO) independently searched the Pubmed, EMBASE, and PsycINFO databases from inception to November (week 2) 2017 using the following keywords: (lymphocytes or T-lymphocytes or B-lymphocytes or monocytes or macrophages or inflammat* or IL-* or cytokine or CRP or C-reactive protein or hs-CRP or hsCRP or interleukin* or tumour necrosis factor or transforming growth factor or interferon) and (schizophren* or psycho*) and (first episode or early or antipsychotic* or drug* or neuroleptic*). Only English-written studies were considered. Abstracts were screened, and full texts of studies retrieved. Where texts were unavailable, authors were contacted and manuscripts requested. We also requested raw data sets from authors, and where appropriate, clarification as to whether patients were antipsychotic naïve. TP and EO selected final studies for meta-analysis.

Selection Criteria

Inclusion criteria were (1) patients with FEP, defined either as first treatment contact/patients recruited from FEP services in line with previous studies64; (2) antipsychotic-naïve; (3) a healthy control group; (4) studies assessing blood cytokines/cytokine receptors, C-reactive protein (CRP) (plasma/serum samples), and white cell counts. The rationale behind focusing on antipsychotic-naïve FEP was to minimize confounding effects of medication and lifestyle habits (eg, diet/exercise) associated with chronic psychotic illness65,66 that may directly (eg, antipsychotics)67 or indirectly (eg, diet/exercise)30,31 alter immune parameters. Exclusion criteria were (1) genetic studies (incomplete translation and post-translational modification means mRNA levels cannot be assumed to reflect protein levels)68; (2) in vitro studies; (3) studies examining stimulated cytokine levels; (4) substance/medication-induced psychosis; (5) absence of data allowing mean and/or standard deviation calculation.

Recorded Variables

Data were extracted as follows: author, publication year, matching criteria, and mean (with standard deviation) measure of immune parameter (table 1 and supplementary eTable 1). See supplementary eAppendix 3 for further details.

Table 1.

Summary of the Designs and Sample Characteristics of the Studies Included in the Meta-analyses

Study Patient, N Control, N Diagnoses Patient Age, Mean (SD) Immune Parameter Matching
Ajami et al40 8 26 Schizophrenia Not specified IL2, IL10, TNFα Not specified
Akiyama69 14 27 Schizophrenia 34.4 (14.0) sIL2R, IL6 Age, gender
Borovcanin et al41 84 35 First-episode psychosis 33.6 (8.8) TGFβ Age
De Berardis et al70 30 30 Schizophrenia (25)
Schizophreniform disorder (2)
Brief psychotic disorder (1)
Delusional disorder (2)
25.9 (6.0) CRP Age, gender
Devanarayanan et al42 22 40 Schizophrenia 29.0 (4.0) CRP Age, gender
Di Nicola et al43 5 24 First-episode psychosis 28.1 (1.1) IL1β, IL2, IL4, IL6, IL8, IL10, TNFα, IFNγ Age, gender, BMI, ethnicity
Ding et al44 69 60 Schizophrenia 27.5 (7.8) IL6, IL17, IFNγ Age, gender, BMI, smoking
El Kissi et al45 10 27 Schizophrenia Not specified IL17, IFNγ, TGFβ Age, gender
Fawzi and Said46 108 200 Schizophrenia 27.2 (10.6) CRP Age, gender, BMI, smoking, ethnicity
Fernandez-Egea et al47 50 50 Schizophrenia (35)
Schizophreniform disorder (8)
Brief psychotic disorder (4)
Delusional disorder (2)
Psychosis NOS (1)
29.4 IL6, CRP Age, gender, BMI, smoking, ethnicity, cortisol
Ganguli and Rabin60 4 57 Schizophrenia Not specified sIL2R Not specified
Ganguli et al71 24 110 Schizophrenia
Schizoaffective disorder
Not specified IL6 Age, gender
Ganguli et al72 33 33 Schizophrenia Not specified IL2 Age, gender
Garcia-Rizo et al48 75 80 First-episode (nonaffective) psychosis 28.0 (6.2) Total lymphocyte count Age, gender, BMI, smoking
Gattaz et al73 10 11 Schizophrenia Not specified IL2, IFNγ Age, gender
Haring et al49
Balotsev et al74
38 37 First-episode psychosis 25.4 (0.9) IL1β, IL2, IL4, IL6, IL8, IL10, IFNγ, TNFα Age, gender, BMI, smoking
Hepgul et al50 4 45 First-episode psychosis Not specified CRP Not specified
Kalmady et al75 25 33 Schizophrenia 29.9 (5.7) IL6 Age, gender
Karanikas et al51 25 23 Schizophrenia (1)
Schizophreniform disorder (16)
Brief psychotic disorder (4)
Psychotic disorder NOS (4)
25.5 (5.4) IL1b, IL2, IL4, IL8, IL10, IFNγ, TNFα Age, BMI, cortisol
Kubistova et al52 25 25 Schizophrenia 32.3 (7.0) IL6, IL8, IL10, TNFα Age, gender
Masserini et al53 7 37 Schizophrenia
Schizophreniform disorder
Not specified Total lymphocyte count Not specified
Mondelli et al26 3 36 First-episode psychosis Not specified IL1β, IL2, IL4, IL6, IL8, IL10, TNFα, IFNγ Age, gender
Noto et al54
Noto et al55
Brinholi et al56
156 58 Schizophrenia Schizophreniform disorder
Brief psychotic disorder
Psychosis NOS
Mania with psychosis
26.2 (7.6) IL4, IL6, IL10, IL17, TNFα, IFNγ Age, gender
Petrikis et al57 39 39 Schizophrenia Schizophreniform disorder
Brief psychotic disorder
27.0
SD not specified
IL6, IL17, TGFβ Age, gender, BMI, smoking
Rapaport and Lohr76 12 14 Schizophrenia 37.9 (11.6) sIL2R Age, gender, ethnicity
Şimşek et al77 30 26 Schizophrenia
Schizophreniform disorder
14.7
1.9
IL2, IL4, IL6, IL10, IL17, TNFα, IFNγ Age, gender, BMI, smoking
Sperner-Unterweger et al58 21 16 Schizophrenia 26.8 (5.5) Total lymphocyte count Not specified
Sirota et al59 6 22 Schizophrenia Not specified sIL2R Age, gender
Song et al78 83 65 Schizophrenia 27.3 (6.7) IL1β, TNFα Age, gender, BMI, smoking
Song et al79
Song et al80
62 60 Schizophrenia 24.7 (5.5) IL1β, IL6, TNFα Age, gender, BMI, smoking
Theodoropoulou et al81 53 62 Schizophrenia Not specified IL1β, IL2, TNFα Age, gender
Xiu et al82 128 62 Schizophrenia 25.8 (9.4) IL10 Age, gender, BMI, smoking, ethnicity

Statistical Analysis

As many studies reported on several parameters, multivariate meta-analysis was used, enabling simultaneous estimation of summary effect sizes across all immune parameters, reducing risk of false positives due to multiple comparisons.83 For all meta-analyses, an omnibus test evaluated significance of model coefficients across immune parameters. Where the omnibus test was significant, we tested the effect separately for each parameter. An unstructured covariance matrix was used owing to uncertainty regarding immune parameter correlations in psychosis. Analyses were only performed if ≥3 studies were identified. A 2-tailed P value <.05 was deemed significant. All analyses were conducted using the metafor package84 in the R statistical programming language.85

Meta-analysis of Variability

To measure variability, the natural log of the ratio of estimates of the population standard deviations for each group was calculated to give the log variability ratio (VR), as previously described (supplementary eAppendix 3).28,86 In biological systems, variance often scales with mean.87 Thus, between group differences in relative variability may, at least partially, be a function of between-group differences in mean. Therefore, a meta-analysis of relative variability of patient compared with control immune parameters scaled to group means was performed: the log coefficient of variation ratio (CVR) (the natural logarithm of the ratio of estimates of population coefficients of variation).28,86 Where the mean is greater in patients than controls, the CVR is a more conservative estimate of variability. To aid interpretation, summary effect sizes for lnVR and lnCVR were transformed back to a linear scale, as previously described.28 Thus, a VR (or CVR) of 1 indicates equal variability in patient and control groups, a VR (or CVR) greater than 1 indicates greater relative variability in patient groups, and a VR (or CVR) less than 1 indicates lower relative variability in patient groups.

Meta-analysis of Mean Differences

A meta-analysis of between group differences in immune parameters was performed, indexed using Hedges g. A random effects model was used owing to expectation of heterogeneity across studies. Data were log-transformed before meta-analysis, since the Cochrane Collaboration recommends log transformation for normalization of positive skew.58 Log-transformed data were either extracted directly from manuscripts, or following provision of raw study-level data by authors which we subsequently log-transformed. Where log-transformed study level/summary data were unavailable, log-transformation estimates were calculated as described by Higgins and colleagues.61

Sensitivity Analyses

To determine if findings were influenced by confounding, we aimed to perform sensitivity analyses to determine if findings remained in studies that matched patients and controls for BMI, smoking, age, gender, ethnicity, and HPA axis activity. We also performed sensitivity analyses excluding poor quality studies.

To determine if findings were influenced by data-skew, we conducted sensitivity analyses after excluding data sets that met Cochrane criteria for skew despite log transformation.61,88 The skew ratio of each immune parameter for patients and controls was determined using the following calculation: lowest possible value for each parameter subtracted from observed mean, divided by standard deviation. A ratio of <1 provides strong evidence of skew,61,88 and, consequently, studies with a ratio <1 were removed in sensitivity analyses.

To determine if there was a difference in proportion of skewed data between patients and controls (which could influence variability analyses), for each immune parameter, the proportion of data sets with severe skew in patients and controls was compared using Fisher’s exact test.89

Consideration of Publication Bias, Study Inconsistency, and Study Quality

Publication bias was assessed for mean differences in all parameters by visual inspection of funnel plots of standard errors against immune residuals. We did not assess for publication bias related to variability, as a selective publication bias is extremely unlikely to exist for such measures. Study quality was assessed using the Newcastle-Ottawa Scale (NOS) (supplementary eAppendix 4).61,90 The thresholds for converting NOS scores into “good,” “fair,” and “poor” quality followed criteria previously described by systematic reviews91 sponsored by the US Agency for Healthcare Research and Quality (supplementary eAppendix 4). Analyses were repeated with poor quality studies removed. Inconsistency between studies was assessed using the I2 statistic (supplementary eAppendix 3).92

Consideration of Raw Data Distribution

For patients and controls, distribution of raw data sets (with >10 data points) for each immune parameter was visually examined using kernel density plots. Studies providing data for the same immune parameter were examined on the same kernel density plot, with values first normalized (mean-scaled). Hartigan’s Dip Test of Unimodality93 was employed to assess the probability of immune parameter data following a unimodal distribution.

Results

Study Selection

Of 3905 citations retrieved, 3751 were excluded after title/abstract review (supplementary eFigure 1). Following manuscript review, 115 studies were excluded based on failure to meet inclusion criteria. All studies included were cross-sectional. IL2 and IL10 data for patients and controls from 2 data sets54–57 were excluded owing to insufficient data to allow mean/standard deviation calculation. The final data set included 35 publications,26,40–60,69–82 making up 32 study data sets providing data on 188 immune measures (number of measures is greater than number of data sets because subjects had more than one measure in many studies). The total sample consisted of 2733 people (1263 patients, 1470 controls; Table 1, supplementary eFigure 1 and supplementary eTable 1), allowing meta-analysis of IL1β, IL2, sIL2R, IL4, IL6, IL8, IL10, IL17, TNFα, IFNγ, TGFβ, CRP, and total lymphocyte count. We received responses from authors of 15 studies, either providing raw data or clarification regarding data modal distribution/medication status.26,40–43,45,49–52,54–57,74 Raw data were obtained for 13 studies.26,41–43,49–52,54–57,74 After exclusion of data with <10 data points,26,43,50 there were sufficient data to separately analyze the distribution of 65 immune measures (32 in patients, 33 in controls) pertaining to IL2, IL4, IL6, IL8, IL10, TGFβ, TNFα, IFNγ, and CRP. The total sample for raw data analysis was 691 (389 patients, 302 controls).

Variability Ratio

We found a significant overall effect of group on log variability ratio across all immune parameters (χ2 = 150.33, P < .0001). Figure 1 shows that the variability of FEP was significantly reduced compared with controls for the following parameters: IL1β (VR = 0.35; 95% CI = 0.17–0.72; P = .004); IL6 (VR = 0.19; 95% CI = 0.09–0.43; P < .0001); IL8 (VR = 0.28; 95% CI = 0.15–0.52; P < .0001); TNFα (VR = 0.36; 95% CI = 0.17–0.75; P = .01); and IL4 (VR = 0.55; 95% CI = 0.32–0.94; P = .03). Variability was not significantly altered for: IL17 (VR = 0.61; 95% CI = 0.35–1.08; P = .09); CRP (VR = 0.82; 95% CI = 0.45–1.47; P = .50); IFNγ (VR = 0.62; 95% CI = 0.25–1.50; P = .29); IL10 (VR = 0.58; 95% CI = 0.27–1.21; P = .15); IL2 (VR = 0.31; 95% CI = 0.09–1.13; P = .08); TGFβ (VR = 0.99; 95% CI = 0.59–1.65; P = .97); sIL2R (VR = 0.79; 95% CI = 0.48–1.32; P = .38); and total lymphocyte cell count (VR = 0.33; 95% CI = 0.08–1.25; P = .10).

Figure 1.

Figure 1.

Forest plot showing effect sizes for variability ratio (VR) of immune parameters in antipsychotic-naïve first-episode psychosis compared with healthy controls. The VR was significantly decreased for interleukin 4 (IL4), interleukin 1beta (IL1β), interleukin 6 (IL6), interleukin 8 (IL8), and tumor necrosis factor alpha (TNFα), indicating lower variability of these immune parameters in patients compared with controls.

Coefficient of Variation Ratio

We found a significant overall effect of group on log variability ratio across all immune parameters (χ2 = 68.62, P < .0001). Figure 2 shows that significant variability differences found with VR remained present using CVR for IL6 (CVR = 0.64; 95% CI = 0.52–0.79; P < .0001) and IL8 (CVR = 0.83; 95% CI = 0.74–0.93; P = .001). There was no significant difference found in variability of CRP (CVR = 1.12; 95% CI = 0.84–1.49; P = .46), IFNγ (CVR = 1.16; 95% CI = 0.83–1.62; P = .39), IL10 (CVR = 0.88; 95% CI = 0.71–1.08; P = .23), IL2 (CVR = 1.00; 95% CI = 0.58–1.73; P = .99), IL17 (CVR = 1.13; 95% CI = 0.96–1.33; P = .13), and total lymphocyte count (CVR = 0.37; 95% CI = 0.10–1.42; P = .15), consistent with VR results. However, differences in variability for IL1β (CVR = 0.87; 95% CI = 0.57–1.33; P = .52), IL4 (CVR = 0.71; 95% CI = 0.48–1.07; P = .10), and TNFα (CVR = 0.97; 95% CI = 0.76–1.23; P = .79), shown to be significantly less variable in patients as per VR results, were not significant using CVR. Moreover, variability of TGFβ, previously shown in VR analysis to be no different between groups, was more variable in patients according to CVR analysis (CVR = 1.41; 95% CI = 1.09–1.83; P = .01). This suggests IL1β, TNFα, IL4, and TGFβ variability analyses are influenced by mean scaling.

Figure 2.

Figure 2.

Forest plot showing effect sizes for mean-scaled variability of immune parameters in antipsychotic-naïve first-episode psychosis compared with healthy controls. The coefficient of variation ratio (CVR) was significant decreased for interleukin 6 (IL6) and interleukin 8 (IL8), indicating lower variability of these immune parameters in patients compared with controls, and significantly increased for transforming growth factor beta (TGFβ), indicating increased variability of this immune parameter in patients compared with controls.

Mean Differences in Immune Measures

We found a significant overall effect of group on mean levels across all immune parameters (χ2 = 114.49, P < .0001). Figure 3 shows that significant elevations in the following parameters were observed in FEP: IFNγ (g = 0.32; 95% CI = 0.11–0.53; P = .003); IL17 (g = 0.48; 95% CI = 0.06–0.89; P = .03); IL6 (g = 0.62; 95% CI = 0.32–0.92; P < .0001); TGFβ (g = 0.53; 95% CI = 0.18–0.88; P = .003); and TNFα (g = 0.56; 95% CI = 0.22–0.90; P = .001). There were no significant differences between groups for: CRP (g = 0.66; 95% CI = −0.03 to 1.34; P = .06); total lymphocyte count (g = 0.31; 95% CI = −0.13 to 0.76; P = .17); IL10 (g = 0.24; 95% CI = −0.13 to 0.62; p = .20); IL1β (g = 0.49; 95% CI = −0.13 to 1.11; P = .12); IL2 (g = −0.07; 95% CI = −0.53 to 0.39; P = .77); IL4 (g = 0.23; 95% CI = −0.05 to 0.51; P = .10); IL8 (g = 0.04; 95% CI = −0.62 to 0.70; P = .90); and sIL2R (g = 2.66; 95% CI = −0.03 to 5.34; P = .05).

Figure 3.

Figure 3.

Forest plot showing effect sizes for mean differences in immune parameters in antipsychotic-naïve first-episode psychosis compared with healthy controls. There was a significant elevation in interferon gamma (IFNγ), interleukin 17 (IL17), interleukin 6 (IL6), transforming growth factor beta (TGFβ), and tumor necrosis factor alpha (TNFα) in patients compared with controls. There was no significant difference in C-reactive protein (CRP), interleukin 10 (IL10), interleukin 1beta (IL1β), interleukin 2 (IL2), interleukin 4 (IL4), interleukin 8 (IL8), soluble interleukin 2 receptor (sIL2R), and total lymphocyte count (TLC) in patients compared with controls.

Sensitivity Analysis of the Influence of Confounders

Restricting analysis of mean differences to studies that matched for age, gender, BMI, and smoking created a data set of 8 studies, covering data on 44 immune measures (supplementary eFigure 2). Analysis showed elevated IL6 (g = 0.83; 95% CI = 0.21–1.45; P = .01), IL17 (g = 0.68; 95% CI = 0.09–1.28; P = .02), and IFNγ (g = 0.54; 95% CI = 0.27–0.80; P < .001) in patients, and no significant difference in TNFα, IL10, or IL1β levels between groups. These findings are consistent with the primary meta-analysis, apart from the TNFα outcome, which was no longer significant, suggesting it could be influenced by confounding. There were insufficient data to analyze IL2, sIL2R, IL4, IL8, TGFβ, CRP, and lymphocyte levels. Sensitivity analyses matching for ethnicity and stress were not possible owing to insufficient studies. We combined CRP and hsCRP data a priori (supplementary eAppendix 3). Poor sensitivity of the CRP assay could conceivably mask hsCRP outcomes. Re-analysis of hsCRP data alone suggests this is not the case, with no difference in hsCRP levels between patients and controls (g = 0.87; 95% CI = 0.17–1.91; P = .10).

Sensitivity Analysis of the Influence of Residual Skew

Removing studies with evidence of persistent skew despite log transformation created a data set of 27 studies, covering data on 118 immune measures (supplementary eFigure 3). Analysis showed elevated IL6 (g = 0.77; 95% CI = 0.15–1.40; P = .02), TNFα (g = 0.58; 95% CI = 0.08–1.09; P = .02), TGFβ (g = 0.68; 95% CI = 0.28–1.08; P < .01), IFNγ (g = 0.24; 95% CI = 0.01–0.48; P = .04), and IL17 (g = 0.64; 95% CI = 0.15–1.14; P = .01) in patients, and no significant difference in IL2, IL4, IL10, CRP, or total lymphocyte count between groups, consistent with our primary meta-analysis. In contrast to the primary analysis, IL1β (g = 0.96; 95% CI = 0.39–1.52; P < .01), sIL2R (g = 0.93; 95% CI = 0.14–1.71; P = .02), and IL8 (g = 0.87; 95% CI = 0.24–1.49; P = .01) were elevated in patients.

For all immune parameters, there was no significant difference in the proportion of immune measures with severe skew in patients compared with controls, either in raw-scaled (P = .20–.99) or log-transformed data sets (P = .21–.99) (supplementary eTable 2).

Study Quality

Newcastle Ottawa Scale quality scores ranged from 0 to 8 (supplementary eTable 3). Of 32 samples, 24 were rated as “good-quality,” 4 as “fair-quality,” and 4 as “poor-quality.”50,53,58,60 Of 3 studies examining total lymphocyte counts, 2 were poor quality.53,58 Excluding the poor-quality paper50 from CRP meta-analysis did not alter outcomes for meta-analysis of mean difference (g = 0.76; 95% CI = −0.01 to 1.53; P = .05). Excluding this study from variability analyses for CRP showed reduced variability in patients (VR = 0.60; 95% CI = 0.45–0.80; P = .001), although there was no difference when mean-scaled (CVR = 1.09; 95% CI = 0.79–1.50; P = .61). Excluding the poor-quality paper60 from sIL2R meta-analysis did not significantly alter outcomes for meta-analysis of mean-difference (g = 3.28; 95% CI = 0.06–6.50; P = .05) nor variability analyses.

Publication Bias and Study Inconsistency

The funnel plot for publication bias did not show evidence of asymmetry (supplementary eFigure 4).26 Higgins’ I2 inconsistency values (supplementary eTable 4) demonstrated a medium-large degree of inconsistency for all parameters, apart from low levels of inconsistency for IFNγ.

Distribution of Raw Immune Parameter Data

Visual inspection of kernel density plots suggested right skewed unimodal distribution for all immune measures in FEP and controls (supplementary eFigure 5). For patients, 30 of 32 immune measures (94%) met Hartigan’s Dip Test criteria for unimodal distribution (supplementary eTable 5), including data sets for IL2, IL4, IL6, IL8, IL10, TGFβ, TNFα, IFNγ, and CRP. For healthy controls, 33 of 33 immune measures (100%) met Hartigan’s Dip Test criteria for unimodal distribution (supplementary eTable 5), including data sets for IL2, IL4, IL6, IL8, IL10, TGFβ, TNFα, IFNγ, and CRP. In the patient group, 1 of the 6 data sets examining IL6 met criteria for a multimodal distribution,54–56 and 1 of the 3 data sets examining IFNγ met criteria for a multimodal distribution.49,74 These 2 data sets did not have overlapping samples and multimodality for IL6 was not accompanied by multimodality for IFNγ (supplementary eTable 5).

Discussion

Summary of Findings

Our first main finding is that that there is a significant reduction in variability of IL1β, IL4, IL6, IL8, and TNFα in FEP patients compared with controls, which is not explained by mean scaling for IL6 and IL8. After adjusting for mean scaling, there was increased heterogeneity of TGFβ in patients compared with controls. As there is no significant difference in proportion of studies with strong evidence of skewed data in patients compared with controls for all parameters, these variability outcomes do not appear to be the result of reduced data skew in patients, and thus may reflect intrinsic differences in immune variability. An examination of raw data did not provide strong evidence for multimodal data distribution of immune parameters in either patients or controls.

We found elevated IL6, TNFα, IFNγ, TGFβ, and IL17 levels in patients compared with controls with small-medium effect sizes (range: g = 0.32–0.62). IL6, IFNγ, and IL17 outcomes were robust to sensitivity analyses, indicating these alterations are unlikely to be driven by key potential confounders and data skew.

The absence of variability elevations of most immune parameters in patients compared to controls and the absence of multimodal distribution of most data is evidence against the existence of an immune subgroup of psychosis. Lower variability of IL6 in patients, coupled with a robust difference in mean concentration, could instead be interpreted as this parameter representing a core (or at least more uniformly present) component in the immunobiology of psychosis.

Strengths and Limitations

By focusing on antipsychotic-naïve FEP, we limited duration of secondary illness-related factors known to influence immune parameters, eg, antipsychotics,67 poor diet, and reduced exercise levels.30,31 Furthermore, sensitivity analyses focusing on studies with strict environmental and physiological matching provides greater confidence that FEP is associated with elevated immune parameters. Relative to previous meta-analyses in the field, log-transformation to reduce influence of skew on summary effect sizes followed by sensitivity analysis excluding data with persistent evidence of skew provides us with robust evidence that immune alterations are present even when influence of data skew is reduced. Moreover, use of a multivariate meta-analytic approach that models the covariance of immune parameters and allows omnibus testing of results thereby reducing multiplicity concerns, is a strength. Gaining access to raw data to examine distribution of multiple data sets is a further strength, complementing findings of the variability meta-analysis. Finally, since in biological systems variance often scales to mean,87 performing a mean-scaled (CVR) variability meta-analysis to complement the primary meta-analysis of variability (VR) provides a conservative approach to assess if primary outcomes are influenced by mean group differences, and provides greater confidence regarding the outcome of increased homogeneity of IL6 and IL8 in patients compared with controls.

It should be noted that there is diagnostic heterogeneity in first-episode samples. In general, about two-thirds of FEP patients have a diagnosis of schizophrenia, while the remainder are diagnosed with other psychotic disorders.94 Of the 32 samples, 18 (56%) included patients with schizophrenia alone (table 1). The remaining samples potentially also included affective psychosis. The proportion of individuals with nonaffective psychosis was not defined, precluding us conducting sensitivity analyses of the effect of diagnosis. Therefore, our findings should not be taken as specific to schizophrenia, but representative of FEP in general. Future studies should provide greater clarity as to whether patients included in analyses presented with an affective or nonaffective psychosis. The combination of affective and nonaffective psychosis in variability analyses should increase heterogeneity in patients and might be expected to influence the modal distribution of patient immune data. However, there was reduced variability of several immune parameters in patients, and no strong evidence for multimodal distribution of patient immune data. Indeed, inclusion of affective psychosis within patient samples may have resulted in under-estimation of increased homogeneity of immune variability in this group, compared with controls.

Inconsistency between studies was moderate to high. This could reflect methodological factors, eg, differences in assay sensitivity, and use of serum/plasma sampling. However, the random effects model used is robust to inconsistency, and would not explain our variability findings, because these reflect within-study variation (where methodologic factors are the same in both patient and control groups in any given study). Moreover, although we were stringent in selecting antipsychotic naïve patients, confirmation of naivety for all psychiatric medications was not universally stated. Thus, use of treatments beyond antipsychotics may have confounded results contributing to inconsistency. Future prospective studies are required that control stringently for medication thereby addressing this potential confound.

Although sample sizes are larger than previous meta-analyses, relatively small sample sizes persist for measures of sIL2R (n = 156), TGFβ (n = 234), and lymphocytes (n = 236). Thus, conclusions regarding these parameters are less secure, and further studies required. Furthermore, only 8 out of 32 (25%) studies matched simultaneously for age, gender, smoking, and BMI, and 2 sensitivity analyses, matching for ethnicity and stress, were not possible owing to insufficient studies. Other factors that could be different between the groups and influence immune measures include recreational substance exposure95 and subclinical physical comorbidity.96–98 Information on these was not included in most studies, precluding sensitivity analysis. Future studies should aim to match patients appropriately to reduce impact of these potential confounds. Moreover, controls may be unusually healthy compared with the general population because they are screened for illnesses, potentially inflating effect sizes for mean differences.99 However, inclusion of super-healthy controls would reduce variability of control immune parameters. In fact, we observed the opposite, and the inclusion of “super controls” could even have led to an under-estimate of reduced variability in immune measures in patients. There is the potential for variability analyses to have been influenced by the presence of latent immune subgroups within the control population. We are however reassured by our findings that 100% of raw control data examined met criteria for unimodal distribution.

Although all studies included in analyses used well-validated quantification techniques (supplementary eTable 6), insufficient assay sensitivity may have limited ability to detect subtle differences in immune parameters between groups, particularly for titers beneath the limit of assay detection. However, where we could assess impact of poor assay sensitivity, examining hsCRP levels separate from CRP levels, results were unaltered. Future developments in immunoassay technology which bring greater assay sensitivity100 will potentially provide greater clarity as to the nature of immune alterations in psychosis.

Positive data skew can inflate standard deviation owing to the presence of outliers within the “tail” of the data.101 We observed evidence of data skew in patients and/or controls for all immune parameters except IL8, TGFβ, and lymphocyte count (supplementary eTable 2). For remaining immune parameters, residual data skew may have influenced variability analyses. However, we demonstrated no significant difference in proportion of skewed data between groups, suggesting that differences in skew in controls compared with patients do not explain variability differences. Moreover, skew does not always inflate standard deviations, since skew describes the shape of data distribution, not scale of spread.94

Comparison With Previous Meta-analyses

A summary of results from previous meta-analyses examining mean differences in immune parameters in FEP16–22 is provided in supplementary eTable 7. There are 2 key differences when compared with previous meta-analyses. First, we failed to observe any differences between patients and controls for mean levels of IL1β, sIL2R, IL4, IL8, IL10, total lymphocyte count, and CRP. However, IL1β, sIL2R, and IL8 levels were elevated in sensitivity analyses excluding data with evidence of skew. These sensitivity analyses are limited by reduced sample size, but suggest the need for future research to determine whether differences in these immune parameters exist between FEP and controls. Second, where differences were observed, effect size estimates were generally smaller than those in previous meta-analyses. For example, Upthegrove et al19 previously reported effect sizes of 2.21 and 0.94 for elevated IL6 and TNFα, respectively in antipsychotic-naïve FEP compared with controls, whereas we observed effect sizes of 0.62 and 0.56, respectively. These differences may be a consequence of several factors, including increased sample size (participant numbers increasing by up to 7-fold), use of log transformed data, and a multivariate meta-analytic approach that models covariance of immune parameters. Given the additional sample size, the focus on antipsychotic naïve patients, the statistical approach employed, and steps we have taken to assess the influence of data-skew and physiological confounds, we suggest that our updated results are likely the most reliable estimates of peripheral immune alterations in FEP to date.

Interpretation and Implications

Our findings suggest that an immune subtype of psychosis, if present, cannot currently be identified through examination of peripheral immune parameter distribution. Indeed, our results could instead be interpreted as supporting the hypothesis that alterations in the immune system are a general feature of psychosis.102

Our results cannot however exclude the possibility that there are alterations in other aspects of the immune system specific to a subgroup of patients. An immune subgroup could manifest itself via various peripheral patterns. Firstly, an immune subgroup might result in wider spread of data points (increased heterogeneity) in patients compared with healthy controls, although, with the potential exception of TGFβ, this is not observed in the current meta-analysis. Secondly, subgroups of patients defined by step increases in immune parameter concentrations could result in multimodal distribution of immune parameter data. Modal analyses in this article suggest a unimodal distribution for most immune parameters, although there was some evidence, albeit weak, for a multimodal distribution of IL6 and IFNγ. Thirdly, a proportion of patients may be more vulnerable to the impact of immune activation, inducing inflammatory and thence psychopathological sequelae in that group (even if proinflammatory titers are of a similar magnitude compared with controls). The observation of increased variability of TGFβ levels in FEP compared with controls following mean scaling could reflect alterations in immune regulatory pathways in a subgroup of individuals with FEP, supporting this model. TGFβ can (although not always)103 perform an anti-inflammatory role, including inhibition of cytokine production from macrophages104 and inhibition of B-lymphocyte proliferation.105 Thus, patients with an impaired TGFβ-mediated immune response could potentially be vulnerable to proinflammatory effects that characterize psychosis (ie, homogenous increases in IL6). Moreover, genetic variants in the TGFβ gene TGFB1 influence susceptibility for schizophrenia.106 Fourthly, immune susceptibility may arise through a unique network-effect of multiple cytokines to bring about inflammatory sequelae and thence psychopathology. Indeed, Weickert and colleagues have employed cluster analysis to divide patients with schizophrenia-spectrum disorders into subgroups based on elevation of multiple cytokines in unison.27,29 They have observed that, compared with controls, a greater proportion of patients clustered within an “elevated cytokine subgroup.” Jeffries and colleagues107 have used graph theory to examine network connectivity of blood proteins related to neuroimmunology across the psychosis spectrum, observing that protein correlation networks can successfully differentiate between controls, and prodromal individuals who transition/do not transition to psychosis. Based on the current meta-analysis, we are unable to comment on whether there is a FEP immune subgroup characterized by an abnormal immune parameter network.

The findings of elevated IL6, IL17, and IFNγ in psychosis appear robust, as elevations are also seen in sensitivity analyses, indicating they are unlikely due to confounding or other nonspecific factors. Elevations of these 3 cytokines could point toward activation of both innate and adaptive immune responses.108,109 Moreover, of these 3 cytokines, IL6 shows reduced variability in patients (including after mean scaling), suggesting this could be a core component of the pathophysiology of psychosis. IL6 is a multifunctional cytokine playing a role in inflammation and the acute phase response,110 immune response,111–113 hematopoiesis,114 glucose and lipid metabolism,115 and bone-turnover.116 It crosses the blood–brain barrier (BBB),117 and there is evidence of increased cerebrospinal fluid (CSF) IL6 levels in psychosis,13 indicating that peripheral IL6 could influence brain function. In the CNS, IL6 induces microglial proliferation118 and activation.119 Microglia play a key role in synaptic remodeling, among other functions.120 Thus, raised peripheral IL6 levels could activate microglia and influence secondary brain changes.4 Supporting this, postmortem studies have reported increased microglial density and morphological changes indicative of microglial activation in schizophrenia,8–10 and some, although not all, positron emission tomography in vivo studies have reported altered binding to a marker expressed on activated microglia in schizophrenia and people at risk of psychosis.11,12 However, it is important to recognize that there are other potential models explaining the association between immune alterations and psychosis,98 and recent studies examining therapeutic potential of anti-IL6 immunotherapy in schizophrenia have been disappointing (although this potentially relates to the inability of the monoclonal antibody to cross the BBB).121,122 Immune alterations could be a consequence of psychosis (eg, psychosocial stress activating an inflammatory response123) or simply an epiphenomenon. Thus, whether peripheral alterations are a cause or a consequence of psychosis remain to be determined.4 Longitudinal studies examining networks of immune parameters in both CSF and blood following individuals in the prodrome through transition to FEP and experimental medicine studies modulating specific aspects of immune function are needed to identify the mechanism underlying the immune alterations we report, and determine the potential for targeting them therapeutically.

Conclusions

Antipsychotic naïve FEP is associated with elevated levels of IL6, IL17, and IFNγ with small-medium effect sizes after accounting for skew and physiological confounds. There is reduced variability in the levels of immune parameters in FEP, and there does not appear to be a clear multimodal distribution of immune parameters in patients. These findings suggest that an immune subgroup of psychosis cannot currently be defined by examination of peripheral immune data spread, and are consistent with elevated immune markers being typical of psychosis.

Summary

  • 1. Antipsychotic-naïve patients with first-episode psychosis show elevated IL6, TNFα, IFNγ, IL17, and TGFβ levels compared with healthy controls.

  • 2. Patients do not show differences in CRP, total lymphocyte counts, IL1β, IL2, sIL2R, IL4, IL8, and IL10 levels compared with healthy controls.

  • 3. Elevated levels of IL6, IL17, and IFNγ are robust to sensitivity analyses which account for data skew, study quality, and physiological/environmental matching.

  • 4. There is reduced variability of immune parameters in patients. Moreover, examination of raw data does not show a multi-modal distribution of immune parameter data in patients.

  • 5. These findings suggest that an immune subgroup of psychosis cannot currently be defined by examination of peripheral immune data spread, and are consistent with elevated immune markers being typical of psychosis.

Funding

This study was funded by grants MC-A656-5QD30 from the Medical Research Council-UK, 666 from the Maudsley Charity 094849/Z/10/Z from the Brain and Behavior Research Foundation, and Wellcome Trust (Dr Howes) and the National Institute for Health Research Biomedical Research Centre at South London and Maudsley National Health Service Foundation Trust and King’s College London. R.M.’s work is supported by the Wellcome Trust (200102/Z/15/Z). V.M. receives funding support from MQ and from the National Institute for Health Research Mental Health Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King’s College London.

Conflict of interest

Dr Howes has received investigator-initiated research funding from and/or participated in advisory/speaker meetings organized by AstraZeneca, Autifony, BMS, Eli Lilly, Heptares, Janssen, Lundbeck, Lyden-Delta, Otsuka, Servier, Sunovion, Rand, and Roche. Drs Pillinger, Osimo, Brugger, Mondelli, and McCutcheon report no conflicts of interest.

Supplementary Material

sby160_suppl_Supplementary_Material

Acknowledgments

Dr Reza Alizadeh-Navei, Dr Abolghasem Ajami, Dr Milica Borovcanin, Dr Hanumanthappa Nandeesha, Dr Yousri El Kissi, Dr Ahlem Mtiraoui, Dr Mariem El Ghardallou, Dr Liina Haring, Dr Evangelos Karanikas, Dr Alzbeta Kubistova, Dr Valeria Mondelli, Dr Petros Petrikis, Dr Cristiano Noto, and Dr Decio Sabbatini Barbosa provided additional data. There was no financial compensation. The funders had no role in the design and conduct of the study; collection, management, analysis, and interpretation of the data; preparation, review, or approval of the manuscript; and decision to submit the manuscript for publication.

References

  • 1. McGrath J, Saha S, Chant D, Welham J. Schizophrenia: a concise overview of incidence, prevalence, and mortality. Epidemiol Rev. 2008;30:67–76. [DOI] [PubMed] [Google Scholar]
  • 2. Howes OD, McCutcheon R, Agid O, et al. Treatment-resistant schizophrenia: Treatment Response and Resistance in Psychosis (TRRIP) Working Group consensus guidelines on diagnosis and terminology. Am J Psychiatry. 2017;174:216–229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Howes OD, Kapur S. A neurobiological hypothesis for the classification of schizophrenia: type A (hyperdopaminergic) and type B (normodopaminergic). Br J Psychiatry. 2014;205:1–3. [DOI] [PubMed] [Google Scholar]
  • 4. Howes OD, McCutcheon R. Inflammation and the neural diathesis-stress hypothesis of schizophrenia: a reconceptualization. Transl Psychiatry. 2017;7:e1024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Ripke S, Neale BM, Corvin A, Walters JTR, Farh KH, Holmans PA et al. Biological insights from 108 schizophrenia-associated genetic loci. Nature 2014;511:421–427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Benros ME, Nielsen PR, Nordentoft M, Eaton WW, Dalton SO, Mortensen PB. Autoimmune diseases and severe infections as risk factors for schizophrenia: a 30-year population-based register study. Am J Psychiatry. 2011;168:1303–1310. [DOI] [PubMed] [Google Scholar]
  • 7. Benros ME, Eaton WW, Mortensen PB. The epidemiologic evidence linking autoimmune diseases and psychosis. Biol Psychiatry. 2014;75:300–306. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Bayer TA, Buslei R, Havas L, Falkai P. Evidence for activation of microglia in patients with psychiatric illnesses. Neurosci Lett. 1999;271:126–128. [DOI] [PubMed] [Google Scholar]
  • 9. Radewicz K, Garey LJ, Gentleman SM, Reynolds R. Increase in HLA-DR immunoreactive microglia in frontal and temporal cortex of chronic schizophrenics. J Neuropathol Exp Neurol. 2000;59:137–150. [DOI] [PubMed] [Google Scholar]
  • 10. Steiner J, Mawrin C, Ziegeler A, et al. Distribution of HLA-DR-positive microglia in schizophrenia reflects impaired cerebral lateralization. Acta Neuropathol. 2006;112:305–316. [DOI] [PubMed] [Google Scholar]
  • 11.Marques TAA, Ashok AH, Pillinger T, et al. Neuroinflammation in schizophrenia: meta-analysis of in-vivo microglial imaging studies. 2018. doi:10.1017/S0033291718003057. [DOI] [PMC free article] [PubMed]
  • 12. Bloomfield PS, Selvaraj S, Veronese M, et al. Microglial activity in people at ultra high risk of psychosis and in schizophrenia: an [(11)C]PBR28 PET brain imaging study. Am J Psychiatry. 2016;173:44–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Wang AK, Miller BJ. Meta-analysis of cerebrospinal fluid cytokine and tryptophan catabolite alterations in psychiatric patients: comparisons between schizophrenia, bipolar disorder, and depression. Schizophr Bull. 2018;44:75–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Matute C, Domercq M, Sánchez-Gómez MV. Glutamate-mediated glial injury: mechanisms and clinical importance. Glia. 2006;53:212–224. [DOI] [PubMed] [Google Scholar]
  • 15. Howes O, McCutcheon R, Stone J. Glutamate and dopamine in schizophrenia: an update for the 21st century. J Psychopharmacol. 2015;29:97–115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Potvin S, Stip E, Sepehry AA, Gendron A, Bah R, Kouassi E. Inflammatory cytokine alterations in schizophrenia: a systematic quantitative review. Biol Psychiatry. 2008;63:801–808. [DOI] [PubMed] [Google Scholar]
  • 17. Miller BJ, Buckley P, Seabolt W, Mellor A, Kirkpatrick B. Meta-analysis of cytokine alterations in schizophrenia: clinical status and antipsychotic effects. Biol Psychiatry. 2011;70:663–671. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Miller BJ, Gassama B, Sebastian D, Buckley P, Mellor A. Meta-analysis of lymphocytes in schizophrenia: clinical status and antipsychotic effects. Biol Psychiatry. 2013;73:993–999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Upthegrove R, Manzanares-Teson N, Barnes NM. Cytokine function in medication-naive first episode psychosis: a systematic review and meta-analysis. Schizophr Res. 2014;155:101–108. [DOI] [PubMed] [Google Scholar]
  • 20. Goldsmith DR, Rapaport MH, Miller BJ. A meta-analysis of blood cytokine network alterations in psychiatric patients: comparisons between schizophrenia, bipolar disorder and depression. Mol Psychiatry. 2016;21:1696–1709. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Fernandes BS, Steiner J, Bernstein HG, et al. C-reactive protein is increased in schizophrenia but is not altered by antipsychotics: meta-analysis and implications. Mol Psychiatry. 2016;21:554–564. [DOI] [PubMed] [Google Scholar]
  • 22. Miller BJ, Culpepper N, Rapaport MH. C-reactive protein levels in schizophrenia: a review and meta-analysis. Clin Schizophr Relat Psychoses. 2014;7:223–230. [DOI] [PubMed] [Google Scholar]
  • 23. Clementz BA, Sweeney JA, Hamm JP, et al. Identification of distinct psychosis biotypes using brain-based biomarkers. Am J Psychiatry. 2016;173:373–384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Kirkpatrick B, Fernandez-Egea E, Garcia-Rizo C, Bernardo M. Differences in glucose tolerance between deficit and nondeficit schizophrenia. Schizophr Res. 2009;107:122–127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. McLean RT, Wilson P, St Clair D, Mustard CJ, Wei J. Differential antibody responses to gliadin-derived indigestible peptides in patients with schizophrenia. Transl Psychiatry. 2017;7:e1121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Mondelli V, Ciufolini S, Belvederi Murri M, et al. Cortisol and inflammatory biomarkers predict poor treatment response in first episode psychosis. Schizophr Bull. 2015;41:1162–1170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Fillman SG, Weickert TW, Lenroot RK, et al. Elevated peripheral cytokines characterize a subgroup of people with schizophrenia displaying poor verbal fluency and reduced Broca’s area volume. Mol Psychiatry. 2016;21:1090–1098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Brugger SP, Howes OD. Heterogeneity and homogeneity of regional brain structure in schizophrenia: a meta-analysis. JAMA Psychiatry. 2017;74:1104–1111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Boerrigter D, Weickert TW, Lenroot R, et al. Using blood cytokine measures to define high inflammatory biotype of schizophrenia and schizoaffective disorder. J Neuroinflammation. 2017;14:188. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Ferrante AW., Jr Obesity-induced inflammation: a metabolic dialogue in the language of inflammation. J Intern Med. 2007;262:408–414. [DOI] [PubMed] [Google Scholar]
  • 31. Visser M, Bouter LM, McQuillan GM, Wener MH, Harris TB. Elevated C-reactive protein levels in overweight and obese adults. JAMA. 1999;282:2131–2135. [DOI] [PubMed] [Google Scholar]
  • 32. Yanbaeva DG, Dentener MA, Creutzberg EC, Wesseling G, Wouters EF. Systemic effects of smoking. Chest. 2007;131:1557–1566. [DOI] [PubMed] [Google Scholar]
  • 33. Franceschi C, Campisi J. Chronic inflammation (inflammaging) and its potential contribution to age-associated diseases. J Gerontol A Biol Sci Med Sci. 2014;69(suppl 1):S4–S9. [DOI] [PubMed] [Google Scholar]
  • 34. Casimir GJ, Mulier S, Hanssens L, Zylberberg K, Duchateau J. Gender differences in inflammatory markers in children. Shock. 2010;33:258–262. [DOI] [PubMed] [Google Scholar]
  • 35. Silverman MN, Sternberg EM. Glucocorticoid regulation of inflammation and its functional correlates: from HPA axis to glucocorticoid receptor dysfunction. Ann N Y Acad Sci. 2012;1261:55–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Paalani M, Lee JW, Haddad E, Tonstad S. Determinants of inflammatory markers in a bi-ethnic population. Ethn Dis. 2011;21:142–149. [PMC free article] [PubMed] [Google Scholar]
  • 37. Amsen D, de Visser KE, Town T. Approaches to determine expression of inflammatory cytokines. Methods Mol Biol. 2009;511:107–142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Imhof A, Fröhlich M, Loewel H, et al. Distributions of C-reactive protein measured by high-sensitivity assays in apparently healthy men and women from different populations in Europe. Clin Chem. 2003;49:669–672. [DOI] [PubMed] [Google Scholar]
  • 39. Ballenberger N, Lluis A, von Mutius E, Illi S, Schaub B. Novel statistical approaches for non-normal censored immunological data: analysis of cytokine and gene expression data. PLoS One. 2012;7:e46423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Ajami A, Abedian F, Hamzeh Hosseini S, Akbarian E, Alizadeh-Navaei R, Taghipour M. Serum TNF-α, IL-10 and IL-2 in schizophrenic patients before and after treatment with risperidone and clozapine. Iran J Immunol. 2014;11:200–209. [PubMed] [Google Scholar]
  • 41. Borovcanin M, Jovanovic I, Radosavljevic G, et al. Elevated serum level of type-2 cytokine and low IL-17 in first episode psychosis and schizophrenia in relapse. J Psychiatr Res. 2012;46:1421–1426. [DOI] [PubMed] [Google Scholar]
  • 42. Devanarayanan S, Nandeesha H, Kattimani S, Sarkar S, Jose J. Elevated copper, hs C-reactive protein and dyslipidemia in drug free schizophrenia: relation with psychopathology score. Asian J Psychiatr. 2016;24:99–102. [DOI] [PubMed] [Google Scholar]
  • 43. Di Nicola M, Cattaneo A, Hepgul N, et al. Serum and gene expression profile of cytokines in first-episode psychosis. Brain Behav Immun. 2013;31:90–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Ding M, Song X, Zhao J, et al. Activation of Th17 cells in drug naïve, first episode schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry. 2014;51:78–82. [DOI] [PubMed] [Google Scholar]
  • 45. El Kissi Y, Samoud S, Mtiraoui A, et al. Increased interleukin-17 and decreased BAFF serum levels in drug-free acute schizophrenia. Psychiatry Res. 2015;225:58–63. [DOI] [PubMed] [Google Scholar]
  • 46. Fawzi MM, Said NS. C-reactive protein serum level in drug-free male Egyptian patients with schizophrenia. Psychiatry Res. 2011;190:91–97. [DOI] [PubMed] [Google Scholar]
  • 47. Fernandez-Egea E, Bernardo M, Donner T, et al. Metabolic profile of antipsychotic-naive individuals with non-affective psychosis. Br J Psychiatry. 2009;194:434–438. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Garcia-Rizo C, Casanovas M, Fernandez-Egea E, et al. Blood cell count in antipsychotic-naive patients with non-affective psychosis. Early Interv Psychiatry. 2017. doi: 10.1111/eip.12456. [DOI] [PubMed] [Google Scholar]
  • 49. Haring L, Koido K, Vasar V, et al. Antipsychotic treatment reduces psychotic symptoms and markers of low-grade inflammation in first episode psychosis patients, but increases their body mass index. Schizophr Res. 2015;169:22–29. [DOI] [PubMed] [Google Scholar]
  • 50. Hepgul N, Pariante CM, Dipasquale S, et al. Childhood maltreatment is associated with increased body mass index and increased C-reactive protein levels in first-episode psychosis patients. Psychol Med. 2012;42:1893–1901. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Karanikas E, Manganaris S, Ntouros E, Floros G, Antoniadis D, Garyfallos G. Cytokines, cortisol and IGF-1 in first episode psychosis and ultra high risk males. Evidence for TNF-α, IFN-γ, ΤNF-β, IL-4 deviation. Asian J Psychiatr. 2017;26:99–103. [DOI] [PubMed] [Google Scholar]
  • 52. Kubistova A, Horacek J, Novak T. Increased interleukin-6 and tumor necrosis factor alpha in first episode schizophrenia patients versus healthy controls. Psychiatr Danub. 2012;24(suppl 1):S153–S156. [PubMed] [Google Scholar]
  • 53. Masserini C, Vita A, Basile R, et al. Lymphocyte subsets in schizophrenic disorders. Relationship with clinical, neuromorphological and treatment variables. Schizophr Res. 1990;3:269–275. [DOI] [PubMed] [Google Scholar]
  • 54. Noto C, Ota VK, Santoro ML, et al. Effects of depression on the cytokine profile in drug naïve first-episode psychosis. Schizophr Res. 2015;164:53–58. [DOI] [PubMed] [Google Scholar]
  • 55. Noto C, Ota VK, Santoro ML, et al. Depression, cytokine, and cytokine by treatment interactions modulate gene expression in antipsychotic naïve first episode psychosis. Mol Neurobiol. 2016;53:5701–5709. [DOI] [PubMed] [Google Scholar]
  • 56. Brinholi FF, Noto C, Maes M, et al. Lowered paraoxonase 1 (PON1) activity is associated with increased cytokine levels in drug naïve first episode psychosis. Schizophr Res. 2015;166:225–230. [DOI] [PubMed] [Google Scholar]
  • 57. Petrikis P, Voulgari PV, Tzallas AT, Archimandriti DT, Skapinakis P, Mavreas V. Cytokine profile in drug-naïve, first episode patients with psychosis. J Psychosom Res. 2015;79:324–327. [DOI] [PubMed] [Google Scholar]
  • 58. Sperner-Unterweger B, Whitworth A, Kemmler G, et al. T-cell subsets in schizophrenia: a comparison between drug-naive first episode patients and chronic schizophrenic patients. Schizophr Res. 1999;38:61–70. [DOI] [PubMed] [Google Scholar]
  • 59. Sirota P, Meiman M, Herschko R, Bessler H. Effect of neuroleptic administration on serum levels of soluble IL-2 receptor-alpha and IL-1 receptor antagonist in schizophrenic patients. Psychiatry Res. 2005;134:151–159. [DOI] [PubMed] [Google Scholar]
  • 60. Ganguli R, Rabin BS. Increased serum interleukin 2 receptor concentration in schizophrenic and brain-damaged subjects. Arch Gen Psychiatry. 1989;46:292. [DOI] [PubMed] [Google Scholar]
  • 61. Higgins JPT, Green S; Cochrane Collaboration Cochrane Handbook for Systematic Reviews of Interventions, Vol 21. Chichester, UK/Hoboken; 2008:649. [Google Scholar]
  • 62. Moher D, Liberati A, Tetzlaff J, Altman DG; PRISMA Group Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. J Clin Epidemiol. 2009;62:1006–1012. [DOI] [PubMed] [Google Scholar]
  • 63. Stroup DF, Berlin JA, Morton SC, et al. Meta-analysis of observational studies in epidemiology: a proposal for reporting. Meta-analysis Of Observational Studies in Epidemiology (MOOSE) group. JAMA. 2000;283:2008–2012. [DOI] [PubMed] [Google Scholar]
  • 64. Breitborde NJ, Srihari VH, Woods SW. Review of the operational definition for first-episode psychosis. Early Interv Psychiatry. 2009;3:259–265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Stubbs B, Firth J, Berry A, et al. How much physical activity do people with schizophrenia engage in? A systematic review, comparative meta-analysis and meta-regression. Schizophr Res. 2016;176:431–440. [DOI] [PubMed] [Google Scholar]
  • 66. Brown S, Birtwistle J, Roe L, Thompson C. The unhealthy lifestyle of people with schizophrenia. Psychol Med. 1999;29:697–701. [DOI] [PubMed] [Google Scholar]
  • 67. Pollmächer T, Haack M, Schuld A, Kraus T, Hinze-Selch D. Effects of antipsychotic drugs on cytokine networks. J Psychiatr Res. 2000;34:369–382. [DOI] [PubMed] [Google Scholar]
  • 68. Shebl FM, Pinto LA, García-Piñeres A, et al. Comparison of mRNA and protein measures of cytokines following vaccination with human papillomavirus-16 L1 virus-like particles. Cancer Epidemiol Biomarkers Prev. 2010;19:978–981. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Akiyama K. Serum levels of soluble IL-2 receptor alpha, IL-6 and IL-1 receptor antagonist in schizophrenia before and during neuroleptic administration. Schizophr Res. 1999;37:97–106. [DOI] [PubMed] [Google Scholar]
  • 70. De Berardis D, Serroni N, Campanella D, et al. C-reactive protein levels and its relationships with suicide risk and alexithymia among newly diagnosed, drug-naive patients with non affective psychosis. Int Clin Psychopharmacol. 2012;28:e35. [Google Scholar]
  • 71. Ganguli R, Yang Z, Shurin G, et al. Serum interleukin-6 concentration in schizophrenia: elevation associated with duration of illness. Psychiatry Res. 1994;51:1–10. [DOI] [PubMed] [Google Scholar]
  • 72. Ganguli R, Brar JS, Chengappa KR, et al. Mitogen-stimulated interleukin-2 production in never-medicated, first-episode schizophrenic patients. The influence of age at onset and negative symptoms. Arch Gen Psychiatry. 1995;52:668–672. [DOI] [PubMed] [Google Scholar]
  • 73. Gattaz WF, Dalgalarrondo P, Schröder HC. Abnormalities in serum concentrations of interleukin-2, interferon-alpha and interferon-gamma in schizophrenia not detected. Schizophr Res. 1992;6:237–241. [DOI] [PubMed] [Google Scholar]
  • 74. Balotsev R, Haring L, Koido K, et al. Antipsychotic treatment is associated with inflammatory and metabolic biomarkers alterations among first-episode psychosis patients: a 7-month follow-up study. Early Interv Psychiatry. 2017. doi: org/10.1111/eip.12457. [DOI] [PubMed] [Google Scholar]
  • 75. Kalmady SV, Venkatasubramanian G, Shivakumar V, et al. Relationship between interleukin-6 gene polymorphism and hippocampal volume in antipsychotic-naïve schizophrenia: evidence for differential susceptibility?PLoS One. 2014;9:e96021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Rapaport MH, Lohr JB. Serum-soluble interleukin-2 receptors in neuroleptic-naive schizophrenic subjects and in medicated schizophrenic subjects with and without tardive dyskinesia. Acta Psychiatr Scand. 1994;90:311–315. [DOI] [PubMed] [Google Scholar]
  • 77. Şimşek Ş, Yıldırım V, Çim A, Kaya S. Serum IL-4 and IL-10 levels correlate with the symptoms of the drug-naive adolescents with first episode, early onset schizophrenia. J Child Adolesc Psychopharmacol. 2016;26:721–726. [DOI] [PubMed] [Google Scholar]
  • 78. Song XQ, Lv LX, Li WQ, Hao YH, Zhao JP. The interaction of nuclear factor-kappa B and cytokines is associated with schizophrenia. Biol Psychiatry. 2009;65:481–488. [DOI] [PubMed] [Google Scholar]
  • 79. Song X, Fan X, Song X, et al. Elevated levels of adiponectin and other cytokines in drug naïve, first episode schizophrenia patients with normal weight. Schizophr Res. 2013;150:269–273. [DOI] [PubMed] [Google Scholar]
  • 80. Song X, Fan X, Li X, et al. Changes in pro-inflammatory cytokines and body weight during 6-month risperidone treatment in drug naïve, first-episode schizophrenia. Psychopharmacology (Berl). 2014;231:319–325. [DOI] [PubMed] [Google Scholar]
  • 81. Theodoropoulou S, Spanakos G, Baxevanis CN, et al. 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. [DOI] [PubMed] [Google Scholar]
  • 82. Xiu MH, Yang GG, Tan YL, et al. Decreased interleukin-10 serum levels in first-episode drug-naïve schizophrenia: relationship to psychopathology. Schizophr Res. 2014;156:9–14. [DOI] [PubMed] [Google Scholar]
  • 83. Bender R, Bunce C, Clarke M, et al. Attention should be given to multiplicity issues in systematic reviews. J Clin Epidemiol. 2008;61:857–865. [DOI] [PubMed] [Google Scholar]
  • 84. Viechtbauer W. Conducting meta-analyses in R with the metafor package. J Stat Softw 2010;36:1–48. [Google Scholar]
  • 85. R Core Team. R: A Language and Environment for Statistical Computing [Software]. Vienna, Austria: R Foundation for Statistical Computing; 2017. [Google Scholar]
  • 86. Nakagawa S, Poulin R, Mengersen K, et al. Meta-analysis of variation: ecological and evolutionary applications and beyond. Methods Ecol Evol. 2015;6:143–152. [Google Scholar]
  • 87. Eisler Z, Bartos I, Kertesz J. Fluctuation scaling in complex systems: Taylor’s law and beyond. Adv Phys. 2008;57:89–142. [Google Scholar]
  • 88. Altman DG, Bland JM. Detecting skewness from summary information. BMJ. 1996;313:1200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Fisher RA. On the interpretation of χ(2) from contingency tables, and the calculation of P. J R Stat Soc. 1922;85:87–94. [Google Scholar]
  • 90. Stang A. Critical evaluation of the Newcastle-Ottawa scale for the assessment of the quality of nonrandomized studies in meta-analyses. Eur J Epidemiol. 2010;25:603–605. [DOI] [PubMed] [Google Scholar]
  • 91. AHRQ. Agency for Healthcare Research and Quality (US) Comparative Effectiveness Reviews. https://www.ncbi.nlm.nih.gov/books/NBK42934/. Accessed June 01, 2018. [Google Scholar]
  • 92. Higgins JP, Thompson SG, Deeks JJ, Altman DG. Measuring inconsistency in meta-analyses. BMJ. 2003;327:557–560. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Hartigan JA, Hartigan PM. The dip test of unimodality. Ann Stat. 1985;13:70–84. [Google Scholar]
  • 94. Groeneveld RA. An influence function-approach to describing the skewness of a distribution. Am Stat. 1991;45:97–102. [Google Scholar]
  • 95. Li YN, Shen RP, Wen GH, et al. Effects of ketamine on levels of inflammatory cytokines IL-6, IL-1 beta, and TNF-alpha in the hippocampus of mice following acute or chronic administration. Front Pharmacol. 2017;8:139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Pillinger T, Beck K, Gobjila C, Donocik JG, Jauhar S, Howes OD. Impaired glucose homeostasis in first-episode schizophrenia: a systematic review and meta-analysis. JAMA Psychiatry. 2017;74:261–269. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Pillinger T, Beck K, Stubbs B, Howes OD. Cholesterol and triglyceride levels in first-episode psychosis: systematic review and meta-analysis. Br J Psychiatry. 2017;211:339–349. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Pillinger T, D’Ambrosio E, McCutcheon R, Howes OD. Is psychosis a multisystem disorder? A meta-review of central nervous system, immune, cardiometabolic, and endocrine alterations in first-episode psychosis and perspective on potential models. Mol Psychiatry. 2018;1–19. doi: 10.1038/s41380-018-0058-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Schwartz S, Susser E. The use of well controls: an unhealthy practice in psychiatric research. Psychol Med. 2011;41:1127–1131. [DOI] [PubMed] [Google Scholar]
  • 100. Woolley CF, Hayes MA. Recent developments in emerging microimmunoassays. Bioanalysis. 2013;5:245–264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Fayers P. Alphas, betas and skewy distributions: two ways of getting the wrong answer. Adv Health Sci Educ Theory Pract. 2011;16:291–296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Müller N, Weidinger E, Leitner B, Schwarz MJ. The role of inflammation in schizophrenia. Front Neurosci. 2015;9:372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Blobe GC, Schiemann WP, Lodish HF. Role of transforming growth factor beta in human disease. N Engl J Med. 2000;342:1350–1358. [DOI] [PubMed] [Google Scholar]
  • 104. Smythies LE, Sellers M, Clements RH, et al. Human intestinal macrophages display profound inflammatory anergy despite avid phagocytic and bacteriocidal activity. J Clin Invest. 2005;115:66–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Li MO, Wan YY, Sanjabi S, Robertson AK, Flavell RA. Transforming growth factor-beta regulation of immune responses. Annu Rev Immunol. 2006;24:99–146. [DOI] [PubMed] [Google Scholar]
  • 106. Frydecka D, Misiak B, Beszlej JA, et al. Genetic variants in transforming growth factor-β gene (TGFB1) affect susceptibility to schizophrenia. Mol Biol Rep. 2013;40:5607–5614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Jeffries CD, Perkins DO, Fournier M, et al. Networks of blood proteins in the neuroimmunology of schizophrenia. Transl Psychiatry. 2018;8:112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Khader SA, Gaffen SL, Kolls JK. Th17 cells at the crossroads of innate and adaptive immunity against infectious diseases at the mucosa. Mucosal Immunol. 2009;2:403–411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. McLoughlin RM, Witowski J, Robson RL, et al. Interplay between IFN-gamma and IL-6 signaling governs neutrophil trafficking and apoptosis during acute inflammation. J Clin Invest. 2003;112:598–607. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Scheller J, Chalaris A, Schmidt-Arras D, Rose-John S. The pro- and anti-inflammatory properties of the cytokine interleukin-6. Biochim Biophys Acta. 2011;1813:878–888. [DOI] [PubMed] [Google Scholar]
  • 111. Bettelli E, Carrier Y, Gao W, et al. Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature. 2006;441:235–238. [DOI] [PubMed] [Google Scholar]
  • 112. Kimura A, Kishimoto T. IL-6: regulator of treg/Th17 balance. Eur J Immunol. 2010;40:1830–1835. [DOI] [PubMed] [Google Scholar]
  • 113. Okada M, Kitahara M, Kishimoto S, Matsuda T, Hirano T, Kishimoto T. IL-6/BSF-2 functions as a killer helper factor in the in vitro induction of cytotoxic T cells. J Immunol. 1988;141:1543–1549. [PubMed] [Google Scholar]
  • 114. Ishibashi T, Kimura H, Shikama Y, et al. Interleukin-6 is a potent thrombopoietic factor in vivo in mice. Blood. 1989;74:1241–1244. [PubMed] [Google Scholar]
  • 115. Glund S, Krook A. Role of interleukin-6 signalling in glucose and lipid metabolism. Acta Physiol (Oxf). 2008;192:37–48. [DOI] [PubMed] [Google Scholar]
  • 116. Kotake S, Sato K, Kim KJ, et al. Interleukin-6 and soluble interleukin-6 receptors in the synovial fluids from rheumatoid arthritis patients are responsible for osteoclast-like cell formation. J Bone Miner Res. 1996;11:88–95. [DOI] [PubMed] [Google Scholar]
  • 117. Banks WA, Kastin AJ, Gutierrez EG. Penetration of interleukin-6 across the murine blood-brain barrier. Neurosci Lett. 1994;179:53–56. [DOI] [PubMed] [Google Scholar]
  • 118. Streit WJ, Hurley SD, McGraw TS, Semple-Rowland SL. Comparative evaluation of cytokine profiles and reactive gliosis supports a critical role for interleukin-6 in neuron-glia signaling during regeneration. J Neurosci Res. 2000;61:10–20. [DOI] [PubMed] [Google Scholar]
  • 119. Klein MA, Möller JC, Jones LL, Bluethmann H, Kreutzberg GW, Raivich G. Impaired neuroglial activation in interleukin-6 deficient mice. Glia. 1997;19:227–233. [DOI] [PubMed] [Google Scholar]
  • 120. Paolicelli RC, Bolasco G, Pagani F, et al. Synaptic pruning by microglia is necessary for normal brain development. Science. 2011;333:1456–1458. [DOI] [PubMed] [Google Scholar]
  • 121. Girgis RR, Ciarleglio A, Choo T, et al. A randomized, double-blind, placebo-controlled clinical trial of tocilizumab, an interleukin-6 receptor antibody, for residual symptoms in schizophrenia. Neuropsychopharmacology. 2018;43:1317–1323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Miller BJ, Dias JK, Lemos HP, Buckley PF. An open-label, pilot trial of adjunctive tocilizumab in schizophrenia. J Clin Psychiatry. 2016;77:275–276. [DOI] [PubMed] [Google Scholar]
  • 123. Calcia MA, Bonsall DR, Bloomfield PS, Selvaraj S, Barichello T, Howes OD. Stress and neuroinflammation: a systematic review of the effects of stress on microglia and the implications for mental illness. Psychopharmacology (Berl). 2016;233:1637–1650. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

sby160_suppl_Supplementary_Material

Articles from Schizophrenia Bulletin are provided here courtesy of Oxford University Press

RESOURCES