Next Article in Journal
Paraneoplastic Neurological Syndromes of the Central Nervous System: Pathophysiology, Diagnosis, and Treatment
Next Article in Special Issue
The Effects of the S1P Receptor Agonist Fingolimod (FTY720) on Central and Peripheral Myelin in Twitcher Mice
Previous Article in Journal
Association of TP53 Single Nucleotide Polymorphisms with Prostate Cancer in a Racially Diverse Cohort of Men
Previous Article in Special Issue
Low Temperature Delays the Effects of Ischemia in Bergmann Glia and in Cerebellar Tissue Swelling
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Insights into the Potential Impact of Quetiapine on the Microglial Trajectory and Inflammatory Response in Organotypic Cortical Cultures Derived from Rat Offspring

Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St., 31-343 Kraków, Poland
*
Author to whom correspondence should be addressed.
Biomedicines 2023, 11(5), 1405; https://doi.org/10.3390/biomedicines11051405
Submission received: 9 March 2023 / Revised: 27 April 2023 / Accepted: 5 May 2023 / Published: 9 May 2023

Abstract

:
Atypical antipsychotics currently constitute the first-line medication for schizophrenia, with quetiapine being one of the most commonly prescribed representatives of the group. Along with its specific affinity for multiple receptors, this compound exerts other biological characteristics, among which anti-inflammatory effects are strongly suggested. Simultaneously, published data indicated that inflammation and microglial activation could be diminished by stimulation of the CD200 receptor (CD200R), which takes place by binding to its ligand (CD200) or soluble CD200 fusion protein (CD200Fc). Therefore, in the present study, we sought to evaluate whether quetiapine could affect certain aspects of microglial activity, including the CD200-CD200R and CX3CL1-CX3CR1 axes, which are involved in the regulation of neuron–microglia interactions, as well as the expression of selected markers of the pro- and anti-inflammatory profile of microglia (Cd40, Il-1β, Il-6, Cebpb, Cd206, Arg1, Il-10 and Tgf-β). Concurrently, we examined the impact of quetiapine and CD200Fc on the IL-6 and IL-10 protein levels. The abovementioned aspects were investigated in organotypic cortical cultures (OCCs) prepared from the offspring of control rats (control OCCs) or those subjected to maternal immune activation (MIA OCCs), which is a widely implemented approach to explore schizophrenia-like disturbances in animals. The experiments were performed under basal conditions and after additional exposure to the bacterial endotoxin lipopolysaccharide (LPS), according to the “two-hit” hypothesis of schizophrenia. The results of our research revealed differences between control and MIA OCCs under basal conditions and in response to treatment with LPS in terms of lactate dehydrogenase and nitric oxide release as well as Cd200r, Il-1β, Il-6 and Cd206 expression. The additional stimulation with the bacterial endotoxin resulted in a notable change in the mRNA levels of pro- and anti-inflammatory microglial markers in both types of OCCs. Quetiapine diminished the influence of LPS on Il-1β, Il-6, Cebpb and Arg1 expression in control OCCs as well as on IL-6 and IL-10 levels in MIA OCCs. Moreover, CD200Fc reduced the impact of the bacterial endotoxin on IL-6 production in MIA OCCs. Thus, our results demonstrated that quetiapine, as well as the stimulation of CD200R by CD200Fc, beneficially affected LPS-induced neuroimmunological changes, including microglia-related activation.

1. Introduction

Antipsychotics are a group of compounds widely implemented in the treatment of mental disorders with the main indication being the treatment of schizophrenia [1]. Along with their affinity to dopamine (DA) receptors, these drugs impact various other targets, including serotonin (5-HT), muscarinic, adrenergic and histamine receptors [2]. Based on differences in the mechanisms of action, antipsychotics are generally categorized into two main groups, typical (first-generation) and atypical, in which two subcategories are distinguished: second- and third-generation [3,4]. Currently, atypical antipsychotics are considered the first-line medication for schizophrenia due to numerous claims regarding their higher efficacy, safety and tolerability when compared to typical drugs [5,6].
One of the most commonly prescribed representatives of second-generation atypical antipsychotics is quetiapine. Along with a broad range of clinical utility across several neuropsychiatric disorders [7,8,9], this compound has a lower potential to produce extrapyramidal symptoms or hyperprolactinemia [10,11]. In schizophrenic patients, quetiapine has shown efficacy against both positive and negative symptoms and has benefits in improving cognitive impairment, aggression, hostility and affective disturbances [12]. The favourable action of this drug has also been described in animal models of schizophrenia-like alterations. In rats after basolateral amygdala lesions generated using quinolinic acid [13], as well as in a rodent model based on prenatal exposure to polyinosinic-polycytidylic acid [14], quetiapine normalized deficits in prepulse inhibition of sensorimotor gating. A similar phenomenon was reported for rat offspring subjected to lipopolysaccharide (LPS) in the prenatal period [15]. Furthermore, treatment with quetiapine attenuated behaviours resembling this neurological condition (e.g., hyperactivity, spatial working memory impairments and sensorimotor gating deficits) in MK-801-injected mice [16].
At the molecular level, quetiapine acts as an antagonist in various pathways, including DA transmission, 5-HT2A, 5-HT2B, 5-HT2C, α1- and α2-adrenergic as well as H1-histamine receptors [17,18]. Additionally, this drug has an affinity for the 5-HT1A receptor as a partial agonist [18].
Multiple preclinical studies have demonstrated that in addition to its antipsychotic activity, quetiapine exerts other biological effects, among which its impact on the immune response is of particular interest [19,20,21,22]. It has been proven that quetiapine inhibited the release of nitric oxide (NO) and tumour necrosis factor-α (TNF-α) from interferon-γ-activated microglia in vitro [19], and ameliorated higher production of interleukin-6 (IL-6) and TNF-α in mice following cuprizone exposure [20]. Moreover, this antipsychotic diminished microglial cell numbers in the hippocampus and reduced Aβ-generated glial activation in a transgenic mouse model of Alzheimer’s disease [21,22].
Microglia possess a myriad of properties that make them an attractive candidate effector to maintain homeostasis in the brain, among others, by being under the control of immunomodulators such as CD200 and/or CX3CL1 [23]. Both ligands, secreted mainly by neurons, participate in the modulation of microglial activation by interacting with specific receptors (CD200R and CX3CR1, respectively), the localization of which is largely limited to microglial cells [24].
To cast more light on the potential involvement of these neuron–microglia axes, in particular CD200-CD200R, in the mechanisms of quetiapine action, we sought to evaluate whether this drug could affect the expression of the Cd200-Cd200r and Cx3cl1-Cx3cr1 pairs as well as the protein levels of both receptors. Additionally, we examined the impact of quetiapine on certain aspects of inflammatory processes, including selected markers of the pro- and anti-inflammatory profile of microglial activity (Cd40, Il-1β, Il-6, Cebpb, Cd206, Arg1, Il-10 and Tgf-β) and cytokine release (IL-6, IL-10). In the final stage of our research, we introduced a soluble CD200 fusion protein containing the ectodomain of CD200 bound to a murine IgG2a module (CD200Fc), which mimics the effects of the ligand CD200 [25,26], to determine whether the modulation of CD200R could influence the IL-6 and IL-10 protein levels.
Considering the specificity of our research, the study described in this article was performed using organotypic cortical cultures (OCCs), which constitute an ex vivo system preserving several aspects of the structural and synaptic organization of the original tissue, including neuron–microglia interactions [27]. Herein, we employed OCCs prepared from control rat offspring (control OCCs) and animals that were subjected to maternal immune activation (MIA OCCs) with LPS, which is one of the widely implemented approaches to explore schizophrenia-like disturbances in animals [28,29,30,31]. The experiments were performed under basal conditions and after additional exposure to the bacterial endotoxin (LPS), complying with the “two-hit” hypothesis of schizophrenia [32].

2. Materials and Methods

2.1. Animals

Adult Wistar rats were purchased from Charles River (Sulzfeld, Germany) and housed under standard conditions with a room temperature of 23 °C, 12/12 h light/dark cycle (lights on at 6:00 am) and ad libitum access to water and food. A set of pregnant females (n = 22) was generated as previously described [30,33] and randomly divided into two equal groups: (1) control and (2) MIA. All procedures were performed under the approval of the Animal Care Committee of the Maj Institute of Pharmacology, Polish Academy of Sciences, Cracow and followed the recommendations of the International Council for Laboratory Animal Science and Guide for the Care and Use of Laboratory Animals (consent numbers: 236/2016 and 128/2018). All possible efforts were made to minimize the number of animals used and their suffering.

2.2. Prenatal Exposure to LPS

MIA was induced by the administration of LPS to pregnant rats as previously reported [29,30,33,34,35]. LPS from Escherichia coli 026:B6 (Sigma-Aldrich, St. Louis, MO, USA) was dissolved to obtain a concentration of 2 mg/kg in 1 mL of saline. The solution was subcutaneously administered to females of the MIA group on alternate days starting from the 7th day of pregnancy between 9:00 and 10:00 a.m. Control pregnant animals underwent the same treatment regimen with the corresponding volume (1 mL/kg) of saline. After delivery, the pups were housed with dams until postnatal days 6–7 (PND6–7). No litter size or weight differences were noted between the control and MIA offspring.

2.3. Organotypic Cortical Cultures

Organotypic cultures were prepared based on the procedure of Stoppini et al. [36], with slight modifications, from the frontal cortices of pups at PND6–7 from both the control (control OCCs) and MIA (MIA OCCs) groups. The animals were decapitated, the brains were aseptically removed and placed in an ice-cold working buffer consisting of 96% Hanks’ balanced salt solution (HBSS) without salts, 3.5% glucose, 0.4% penicillin and streptomycin solution and HEPES (to maintain the pH) (all from Gibco, Paisley, UK). Then, the frontal cortices were dissected, transferred to Teflon discs, transversely cut into 350 μm slices using a McIlwain tissue chopper and situated on ThinCertsTM—TC Inserts (Greiner bio-one, Kremsmünster, Austria) with 0.4 μm pore size transparent membranes in 6-well plates. The sections were cultured with 1 mL of Dulbecco’s Modified Eagle Medium (DMEM) + GlutaMax™-I (50%; pH 7.4) supplemented with 20.5% HBSS with salts, 25% horse serum (HS), 0.1 mg/mL glucose, 1% amphotericin B, 0.4% penicillin and streptomycin solution, 1% B-27 supplement and HEPES (to maintain pH at 7.4) (all from Gibco, Paisley, UK) in a humidified 5% CO2 incubator at 37 °C. After 24 h, half of the medium volume (0.5 mL) was replaced, and then after the next 48 h, 1 mL of medium was changed to a fresh one. Later, the medium was substituted (1 mL) every second day with a new one containing a gradually reduced amount of HS (25% and 10%). On the 7th day of in vitro culture, the medium was replaced with a serum-free 1% N2-supplemented mixture of 50% DMEM F-12 (pH 7.4), 44% HBSS with salts, 0.1 mg/mL glucose, 1% amphotericin B, 0.4% penicillin and streptomycin solution, 1% B-27 and HEPES (to maintain pH at 7.4) (all from Gibco, Paisley, UK).

2.4. Chemicals and Drugs

Recombinant mouse CD200 Fc chimera protein (CD200Fc; R&D Systems, Minneapolis, MN, USA) was reconstituted in sterile phosphate-buffered saline (PBS; Sigma-Aldrich, St. Louis, MO, USA) (the final concentration in the well was 5 µg/mL) [37]. Quetiapine (Carbosynth Limited, Berkshire, UK) was dissolved in dimethyl sulfoxide (BioShop, Burlington, ON, Canada) to a 10 mM stock solution and was freshly diluted before each use in PBS (Sigma-Aldrich, St. Louis, MO, USA) (the final concentration in the well was 1, 5 or 10 µM). LPS (from Escherichia coli 0111:B4; Sigma-Aldrich, St. Louis, MO, USA) was prepared in PBS (Sigma-Aldrich, St. Louis, MO, USA) and the final concentration in the well was 1 μg/mL [38].

2.5. Treatment of the OCCs

An overview of the treatment schedule is illustrated in Figure 1. Thirty minutes after the last medium change, control and MIA OCCs were stimulated in the manner described hereafter.
  • Experiment 1
In the experiments comparing control and MIA OCCs in terms of response to LPS exposure, the slices were subjected to this bacterial endotoxin for 24 h, and afterwards, the appropriate analyses were performed.
  • Experiment 2
In the next set of experiments determining quetiapine doses for administration in further studies and assessing the impact of the drug on mRNA expression of Cd200-Cd200r and Cx3cl1-Cx3cr1 axes and markers of the pro- and anti-inflammatory profile of microglia (Cd40, Il-1β, Il-6, Cebpb, Cd206, Arg1, Il-10 and Tgf-β) as well as on CD200R and CX3CR1 levels, the OCCs were incubated with the antipsychotic for 90 min and later additionally exposed to LPS for 24 h.
  • Experiment 3
In the last set of experiments comparing the potential influence of CD200Fc and quetiapine effects on MIA OCCs, the slices were treated with CD200Fc for 12 h or exposed to the drug for 90 min and finally incubated with LPS for 24 h.
Control groups in all experiments were subjected to the appropriate solvent in the corresponding volume and regimen.

2.6. Culture Collection and Sample Preparation

At the end of each experiment, culture media were collected for the evaluation of lactate dehydrogenase (LDH) and NO release as well as for enzyme-linked immunosorbent assay (ELISA).
Simultaneously, the slices intended for ELISA analyses were lysed in RIPA lysis buffer enriched with protease inhibitor cocktail, phosphatase inhibitor cocktail, 1 mM sodium orthovanadate and 1 mM phenylmethanesulfonyl fluoride (all from Sigma-Aldrich, St. Louis, MO, USA). The protein concentration in the prepared samples was determined utilizing a PierceTM BCA Protein Assay Kit (Thermo Fisher, Rockford, IL, USA) according to the manufacturer’s instructions. Bovine serum albumin from the kit served as a standard, and the absorbance for each sample was examined at a wavelength of 562 nm in a Tecan Infinite 200 Pro spectrophotometer (Tecan, Mannedorf, Germany).
To prepare samples for quantitative real-time polymerase chain reaction (qRT-PCR), the slices were transferred to TRI Reagent® (Sigma-Aldrich, St. Louis, MO, USA), and total RNA was extracted using the Chomczynski method [39]. Instantly after extraction, RNA concentration was evaluated with a NanoDrop Spectrophotometer (ND/1000 UV/Vis, Thermo Fisher NanoDrop, Waltham, MA, USA).

2.7. Lactate Dehydrogenase Release

The LDH assay was performed as previously described [38] based on a colorimetric method using a commercially available Cytotoxicity Detection Kit (Roche, Mannheim, Germany) following the manufacturer’s instructions. The optical density of the red-colored solution of formazan formed during the test was measured at a wavelength of 490 nm in a Tecan Infinite 200 Pro spectrophotometer (Tecan, Mannedorf, Germany) and was proportional to the LDH level. The results were normalized to the LDH release calculated for the vehicle group from control OCCs (Experiment 1) or the vehicle group from the corresponding type of OCCs (Experiment 2) and are displayed as a percentage of the control ± standard error of the mean (SEM).

2.8. Nitric Oxide Release

The level of NO secreted into the culture medium was determined by the Griess reaction as previously described [38]. Equal volumes of supernatant, Griess reagent A [0.1% N-(1-naphthyl)ethylenediamine dihydrochloride] and Griess reagent B (1% sulfanilamide in 5% phosphoric acid) (both from Sigma-Aldrich, St. Louis, MO, USA) were mixed, and the absorbance of the prepared solution was measured immediately at a wavelength of 540 nm in a Tecan Infinite 200 Pro spectrophotometer (Tecan, Mannedorf, Germany). The data were normalized to the level of NO released from the vehicle group in control OCCs (Experiment 1) or the vehicle group from the corresponding type of OCCs (Experiment 2) and are presented as the percentage of the control ± SEM.

2.9. Enzyme-Linked Immunosorbent Assay

The protein levels of CD200R and CX3CR1 (both from Cusabio, Houston, TX, USA) were established in the OCC lysates, whereas levels of IL-6 and IL-10 (both from BD Biosciences, San Diego, CA, USA) were quantified in the supernatants using commercially available ELISA kits following the manufacturer’s instructions. The outcomes of ELISA experiments were calculated as pg/mg of protein or optical density at 450 nm.

2.10. Quantitative Real-Time Polymerase Chain Reaction

Equal amounts of RNA (0.5 μg) were reverse transcribed into complementary DNA (cDNA) employing an NG dART RT kit (EURx, Gdańsk, Poland). The cDNA was amplified with a FastStart Universal Probe Master (Rox) kit (Roche, Basel, Switzerland) and TaqMan probes (Life Technologies, Carlsbad, CA, USA) for the following genes: Cd200r (Rn00576646_m1), Cd200 (Rn01646320_m1), Cx3cr1 (Rn00591798_m1), Cx3cl1 (Rn00593186_m1), Cd40 (Rn01423583_m1), Il-1β (Rn00580432_m1), Il-6 (Rn01410330_m1), Cebpb (Rn00824635_s1), Cd206 (Rn01487342_m1), Arg1 (Rn00691090_m1), Il-10 (Rn01644839_m1), Tgf-β (Rn00572010_m1), and, as a reference, Gapdh (Rn01775763_g1). The PCR products were generated in mixtures consisting of cDNA (diluted 10 times in PCR grade distilled water) used as the PCR template (3 μL), TaqMan probe (0.5 μL), 1× FastStart Universal Probe Master (Rox) mix containing 250 nM hydrolysis probe labelled with the fluorescent reporter dye (fluorescein) at the 5′-end and a quenching dye at the 3′-end (5 μL), and finally, the remainder of PCR grade distilled water to reach a total volume of 10 μL. Thermocycling conditions were as follows: initial denaturation at 95 °C for 10 min, 40 cycles of denaturation at 95 °C for 15 s, annealing at 60 °C for 1 min and extension at 50 °C for 2 min. The threshold value (Ct) for each sample was set in the exponential phase of PCR and the ∆∆Ct method was used for data analysis.

2.11. Statistical Analysis

Statistical analysis of the data was executed using Statistica 13.0 software (StatSoft, Palo Alto, CA, USA). All biochemical experiments were carried out under the same conditions, regardless of the treatment or culture (control OCCs vs. MIA OCCs). The results of all analyses are displayed as the mean ± SEM. When applicable, statistical outliers were identified using Grubbs’ test. The normal distribution and the homogeneity of the variance were examined using the Shapiro–Wilk test and Levene’s test, respectively. Comparisons of variables between groups were performed using factorial analysis of variance (factorial ANOVA) with Duncan’s post hoc test or planned comparisons via a one-way ANOVA (contrast analysis). The results were considered statistically significant when the p value was less than 0.05. All graphs were prepared with GraphPad Prism 7 software (San Diego, CA, USA).

3. Results

3.1. Release of Lactate Dehydrogenase and Nitric Oxide in Control and MIA OCCs under Basal and LPS-Induced Conditions

In the initial experiments, we examined the basal and LPS-evoked levels of LDH and NO in OCCs obtained from control and MIA offspring (Figure 2).
Figure 2. The basal and LPS-induced levels of LDH (A) and NO (B) in control and MIA OCCs. n = 21 in each group. The results were calculated as a percentage of the release obtained for the vehicle group from control OCCs (% of control) and are presented as the means ± standard errors of the means (SEM). Statistical analysis was performed using factorial ANOVA with Duncan’s post hoc test. * p < 0.05 vs. control OCCs + vehicle, # p < 0.05 vs. control OCCs + LPS, ^ p < 0.05 vs. MIA OCCs + vehicle.
Figure 2. The basal and LPS-induced levels of LDH (A) and NO (B) in control and MIA OCCs. n = 21 in each group. The results were calculated as a percentage of the release obtained for the vehicle group from control OCCs (% of control) and are presented as the means ± standard errors of the means (SEM). Statistical analysis was performed using factorial ANOVA with Duncan’s post hoc test. * p < 0.05 vs. control OCCs + vehicle, # p < 0.05 vs. control OCCs + LPS, ^ p < 0.05 vs. MIA OCCs + vehicle.
Biomedicines 11 01405 g002
As shown in Figure 2A, the nonstimulated release of LDH in MIA OCCs was significantly higher (p = 0.0166) than that in control OCCs. The 24-h incubation with the bacterial endotoxin resulted in the increased efflux of LDH from the slices of both control (p = 0.0001) and MIA (p = 0.0001) OCCs (Figure 2A). Regarding basal NO secretion, we did not observe any difference between the two types of OCCs (Figure 2B). Exposure to LPS elevated the level of NO in both control (p = 0.0001) and MIA (p = 0.0001) OCCs. Simultaneously, the effect of treatment with the bacterial endotoxin on NO release was more pronounced for MIA (p = 0.0004) than for control OCCs (Figure 2B).

3.2. The Impact of Different Doses of Quetiapine on Lactate Dehydrogenase and Nitric Oxide Release in Control and MIA OCCs under Basal and LPS-Induced Conditions

In the next step, LDH and NO assays were applied to determine the dose of quetiapine to be used in further experiments (Figure 3).
Figure 3. LDH (A,B) and NO (C,D) levels in control (A,C) and MIA (B,D) OCCs after treatment with different doses of quetiapine (1, 5 or 10 µM) under basal and LPS-induced conditions. n = 6–17 (LDH) or 7–16 (NO) in control OCCs, n = 8–26 (LDH) or 8–17 (NO) in MIA OCCs. The results were calculated as a percentage of the release observed for the vehicle group from the corresponding type of OCCs (% of control) and are presented as the means ± standard errors of the means (SEM). Statistical analysis was performed using planned comparisons via a one-way ANOVA (contrast analysis). * p < 0.05 vs. control OCCs + vehicle, # p < 0.05 vs. control OCCs + LPS, ^ p < 0.05 vs. MIA OCCs + vehicle, $ p < 0.05 vs. MIA OCCs + LPS.
Figure 3. LDH (A,B) and NO (C,D) levels in control (A,C) and MIA (B,D) OCCs after treatment with different doses of quetiapine (1, 5 or 10 µM) under basal and LPS-induced conditions. n = 6–17 (LDH) or 7–16 (NO) in control OCCs, n = 8–26 (LDH) or 8–17 (NO) in MIA OCCs. The results were calculated as a percentage of the release observed for the vehicle group from the corresponding type of OCCs (% of control) and are presented as the means ± standard errors of the means (SEM). Statistical analysis was performed using planned comparisons via a one-way ANOVA (contrast analysis). * p < 0.05 vs. control OCCs + vehicle, # p < 0.05 vs. control OCCs + LPS, ^ p < 0.05 vs. MIA OCCs + vehicle, $ p < 0.05 vs. MIA OCCs + LPS.
Biomedicines 11 01405 g003
Incubation of the slices with LPS (p = 0.0377), but also with the drug at a concentration of 5 μM enhanced LDH secretion in control OCCs (p = 0.0171) (Figure 3A). Concurrently, in MIA OCCs, stimulation with LPS (p < 0.0001) as well as with the antipsychotic at a dose of 10 µM (p = 0.0070) upregulated LDH efflux (Figure 3B). Pretreatment with 5 µM quetiapine (p = 0.0156) intensified LDH release in response to the addition of the bacterial endotoxin in MIA OCCs. The outcome of the NO test revealed that exposure to LPS led to the increased release of the measured compound both in control (p < 0.0001) and MIA (p < 0.0001) OCCs (Figure 3C,D). This effect was aggravated when the slices were preincubated with quetiapine at a concentration of 1 (control OCCs: p = 0.0021; MIA OCCs: p = 0.0154) or 5 µM (control OCCs: p = 0.0022; MIA OCCs: p < 0.0001) in both types of OCCs. In contrast, 10 µM quetiapine prevented the increase in NO secretion after LPS stimulation in both control (p = 0.0001) and MIA (p = 0.0001) OCCs (Figure 3C,D). Therefore, based on the above-described results showing that among the tested doses, only quetiapine at 10 µM did not enhance the effect of the bacterial endotoxin on LDH levels and at the same time reduced NO release after treatment with LPS in both types of OCCs, further experiments were carried out using the drug at a concentration of 10 µM.

3.3. The Impact of Quetiapine on the mRNA Expression of the Cd200-Cd200r and Cx3cl1-Cx3cr1 Axes in Control and MIA OCCs under Basal and LPS-Induced Conditions

The data suggested that microglia might be intimately engaged in MIA-triggered disturbances, resembling schizophrenia [40], and that antipsychotics may affect the activation of these cells [22]. Accordingly, we evaluated the gene expression of two microglial receptors (Cd200r and Cx3cr1) and their corresponding neuronal ligands (Cd200 and Cx3cl1, respectively) in control and MIA OCCs under basal conditions and when subjected to quetiapine and/or additional LPS treatment (Figure 4).
Figure 4. The gene expression of Cd200r (A), Cd200 (B), Cx3cr1 (C) and Cx3cl1 (D) in control and MIA OCCs after quetiapine (10 µM) treatment under basal and LPS-induced conditions. n = 4–8 in control OCCs and n = 3–6 in MIA OCCs (Cx3cr1), n = 4–7 in control OCCs and n = 3–6 in MIA OCCs (Cd200r, Cd200, Cx3cl1). The results were calculated as the average fold change and are presented as the means ± standard errors of the means (SEM). Statistical analysis was performed using factorial ANOVA with Duncan’s post hoc test or planned comparisons via a one-way ANOVA (contrast analysis). * p < 0.05 vs. control OCCs + vehicle, # p < 0.05 vs. control OCCs + LPS, ^ p < 0.05 vs. MIA OCCs + vehicle.
Figure 4. The gene expression of Cd200r (A), Cd200 (B), Cx3cr1 (C) and Cx3cl1 (D) in control and MIA OCCs after quetiapine (10 µM) treatment under basal and LPS-induced conditions. n = 4–8 in control OCCs and n = 3–6 in MIA OCCs (Cx3cr1), n = 4–7 in control OCCs and n = 3–6 in MIA OCCs (Cd200r, Cd200, Cx3cl1). The results were calculated as the average fold change and are presented as the means ± standard errors of the means (SEM). Statistical analysis was performed using factorial ANOVA with Duncan’s post hoc test or planned comparisons via a one-way ANOVA (contrast analysis). * p < 0.05 vs. control OCCs + vehicle, # p < 0.05 vs. control OCCs + LPS, ^ p < 0.05 vs. MIA OCCs + vehicle.
Biomedicines 11 01405 g004
In the case of the Cd200-Cd200r axis, we observed a lower level of the receptor (p = 0.0002) in MIA OCCs than in control OCCs. Simultaneously, MIA OCCs were more susceptible than control OCCs to incubation with the bacterial endotoxin in terms of Cd200r (p = 0.0010) expression (Figure 4A). When evaluating the Cx3cl1-Cx3cr1 dyad, we detected a significant reduction in Cx3cr1 mRNA levels in both control (p = 0.0055) and MIA (p = 0.0099) OCCs exposed to LPS (Figure 4C). No effect of incubation with quetiapine and/or bacterial endotoxin on the expression of the examined ligands was revealed in any of the investigated types of OCCs (Figure 4).

3.4. The Impact of Quetiapine on the CD200R and CX3CR1 Protein Levels in Control and MIA OCCs under Basal and LPS-Induced Conditions

Considering the alterations in the gene expression of the systems supervising neuron–microglia interactions, in the next step of the study, we explored the protein levels of selected microglial receptors (CD200R and CX3CR1) in control and MIA OCCs after treatment with quetiapine and/or additional LPS (Figure 5). As illustrated in Figure 5, we did not notice any difference between the two types of OCCs or the impact of incubation with the drug and/or bacterial endotoxin on the levels of CD200R or CX3CR1.
Figure 5. CD200R (A) and CX3CR1 (B) protein levels in control and MIA OCCs after treatment with quetiapine (10 µM) under basal and LPS-induced conditions. n = 3–6 in each group. The results were calculated as pg/mg of protein and are presented as the means ± standard errors of the means (SEM). Statistical analysis was performed using factorial ANOVA with Duncan’s post hoc test.
Figure 5. CD200R (A) and CX3CR1 (B) protein levels in control and MIA OCCs after treatment with quetiapine (10 µM) under basal and LPS-induced conditions. n = 3–6 in each group. The results were calculated as pg/mg of protein and are presented as the means ± standard errors of the means (SEM). Statistical analysis was performed using factorial ANOVA with Duncan’s post hoc test.
Biomedicines 11 01405 g005

3.5. The Impact of Quetiapine on the mRNA Expression of Microglial Markers in Control and MIA OCCs under Basal and LPS-Induced Conditions

The response of microglia to disturbances in brain homeostasis is highly associated with the release of multiple pro- and anti-inflammatory factors as well as various CD antigen levels [41]. Therefore, we measured the expression of genes (Cd40, Il-1β, Il-6, Cebpb, Cd206, Arg1, Il-10 and Tgf-β) that are considered essential for microglial activity (Figure 6).
Figure 6. The gene expression of Cd40 (A), Il-1β (B), Il-6 (C), Cebpb (D), Cd206 (E), Arg1 (F), Il-10 (G) and Tgf-β (H) in control and MIA OCCs after quetiapine (10 µM) treatment under basal and LPS-induced conditions. n = 4–8 in control OCCs and n = 3–6 in MIA OCCs (Cd40, Il-1β, Il-6, Cebpb, Cd206, Il-10 and Tgf-β), n = 3–7 in control OCCs and n = 3–6 in MIA OCCs (Arg1). The results were calculated as the average fold change and are presented as the means ± standard errors of the means (SEM). Statistical analysis was performed using planned comparisons via a one-way ANOVA (contrast analysis). * p < 0.05 vs. control OCCs + vehicle, # p < 0.05 vs. control OCCs + LPS, ^ p < 0.05 vs. MIA OCCs + vehicle.
Figure 6. The gene expression of Cd40 (A), Il-1β (B), Il-6 (C), Cebpb (D), Cd206 (E), Arg1 (F), Il-10 (G) and Tgf-β (H) in control and MIA OCCs after quetiapine (10 µM) treatment under basal and LPS-induced conditions. n = 4–8 in control OCCs and n = 3–6 in MIA OCCs (Cd40, Il-1β, Il-6, Cebpb, Cd206, Il-10 and Tgf-β), n = 3–7 in control OCCs and n = 3–6 in MIA OCCs (Arg1). The results were calculated as the average fold change and are presented as the means ± standard errors of the means (SEM). Statistical analysis was performed using planned comparisons via a one-way ANOVA (contrast analysis). * p < 0.05 vs. control OCCs + vehicle, # p < 0.05 vs. control OCCs + LPS, ^ p < 0.05 vs. MIA OCCs + vehicle.
Biomedicines 11 01405 g006
Among the tested markers, the mRNA levels of Cd40 (p = 0.0201), Il-1β (p < 0.0001), Il-6 (p < 0.0001), Cebpb (p < 0.0001), Arg1 (p = 0.0007) and Il-10 (p < 0.0001) were increased, whereas Tgf-β (p = 0.0001) expression was decreased after LPS stimulation in control OCCs (Figure 6A–D,F–H). Pretreatment of the slices with quetiapine diminished the impact of the bacterial endotoxin on Il-1β (p = 0.0145), Il-6 (p = 0.0052), Cebpb (p = 0.0031) and Arg1 (p = 0.0386) levels (Figure 6B–D,F).
qRT-PCR analysis of MIA OCCs demonstrated that the expression of Il-1β (p < 0.0001), Il-6 (p < 0.0001), Cebpb (p < 0.0001), Arg1 (p = 0.0007) and Il-10 (p < 0.0001) was elevated, and simultaneously, Tgf-β (p = 0.0001) levels were reduced in response to LPS exposure (Figure 6B–D,F–H). The effect of the bacterial endotoxin in MIA OCCs on Il-1β (p = 0.0001) and Il-6 (p = 0.0014) expression was less distinct, whereas on Cd206 (p = 0.0323), levels was more pronounced than that in control OCCs (Figure 6B,C,E). Furthermore, quetiapine preincubation upregulated the mRNA expression of Cd206 (p = 0.0256) in MIA OCCs (Figure 6E).

3.6. The Impact of Quetiapine and CD200Fc on IL-6 and IL-10 Protein Levels in Control and MIA OCCs under Basal and LPS-Induced Conditions

In the final experiments, we attempted to investigate whether the limited response of MIA OCCs to quetiapine, and thus, its slight modulating effect on the gene expression of pro- and anti-inflammatory cytokines, was related to the changed susceptibility of this type of OCCs as a result of MIA. For this purpose, we compared the impact of the drug to CD200Fc for which anti-inflammatory properties have been suggested. We evaluated the protein levels of pro-inflammatory IL-6 and anti-inflammatory IL-10 after stimulation with CD200Fc or quetiapine under basal and LPS-induced conditions in MIA OCCs (Figure 7).
Figure 7. IL-6 (A) and IL-10 (B) protein levels in MIA OCCs after treatment with CD200Fc or quetiapine (10 µM) under basal and LPS-induced conditions. n = 4–14. The results were calculated as the optical density at 450 nm and are presented as the means ± standard errors of the means (SEM). Statistical analysis was performed using planned comparisons via a one-way ANOVA (contrast analysis). ^ p < 0.05 vs. MIA OCCs + vehicle, $ p < 0.05 vs. MIA OCCs + LPS.
Figure 7. IL-6 (A) and IL-10 (B) protein levels in MIA OCCs after treatment with CD200Fc or quetiapine (10 µM) under basal and LPS-induced conditions. n = 4–14. The results were calculated as the optical density at 450 nm and are presented as the means ± standard errors of the means (SEM). Statistical analysis was performed using planned comparisons via a one-way ANOVA (contrast analysis). ^ p < 0.05 vs. MIA OCCs + vehicle, $ p < 0.05 vs. MIA OCCs + LPS.
Biomedicines 11 01405 g007
Treatment with the bacterial endotoxin elevated IL-6 production (p < 0.0001), which was reduced by both CD200Fc (p = 0.0050) and quetiapine (p = 0.0002) preincubation (Figure 7A). In the case of the IL-10 level, which was also increased in response to LPS exposure (p < 0.0001), the stimulation with the drug (p = 0.0381) diminished, while CD200Fc addition did not influence this disturbance (Figure 7B).

4. Discussion

In the present study, we demonstrated that MIA had an adverse effect on the biochemical status of OCCs and the expression of microglial receptors involved in neuron–microglia interactions. The changes were observed under basal conditions and/or after additional treatment with LPS applied in our research as a “second hit” activation. Specifically, MIA intensified the basal secretion of LDH and simultaneously NO release when the cultures were exposed to the bacterial endotoxin. Another important consequence of MIA was the strong limitation of Cd200r expression under nonstimulated conditions. The deficit was further enhanced by the introduction of the “second hit” to the experimental setup, which concurrently resulted in decreases of Cx3cr1 levels in both types of OCCs. Nevertheless, the alterations in the gene expression of both receptors due to MIA were not followed by disturbances in the mRNA profile of the evaluated pro- and anti-inflammatory markers. Regarding quetiapine, although this atypical antipsychotic did not directly affect the expression of the ligand-receptor axes or the protein levels of CD200R and CX3CR1, drug administration upregulated Cd206 expression and inhibited the induction of the “second hit” increase in IL-6 and IL-10 protein levels in MIA OCCs. The beneficial impact of quetiapine was also present in control OCCs, where the antipsychotic prevented Il-1β, Il-6, Cebpb and Arg1 expression from rising after LPS treatment. Therefore, our observations suggest that MIA can influence the biochemical and immune profile of OCCs and determine the response to quetiapine treatment.

4.1. Maternal Immune Activation Affects Lactate Dehydrogenase Release in Organotypic Cortical Cultures and Partially Their Response to Stimulation with Lipopolysaccharide

MIA-generated behavioural, neurological and immunological alterations described in animals’ offspring at juvenile and adult stages of life are considered to reflect the aetiology of neuropsychiatric disorders in humans [42,43,44]. Nonetheless, the molecular mechanisms leading to these changes are still ambiguous, justifying a constant need for developing useful models to broaden the knowledge about these phenomena and for identifying new targets for potentially more effective pharmacotherapies.
Our present research was conducted in a neurodevelopmental model of schizophrenia based on the administration of bacterial endotoxin (specifically, LPS) to female rats throughout pregnancy [15,29,30,33,34,45]. MIA applied in this paradigm produces age-dependent behavioural deficits in sensorimotor gating [29,30], disturbances in exploration [33,45], spontaneous and amphetamine-induced locomotor activity [35] and social interactions [45] as well as the presence of anxiety behaviour [33]. Concurrently, MIA with LPS generates various long-lasting biochemical consequences, including peripheral and central immune disturbances [29,30,33,46,47]. Data have indicated that proper neuron–microglia communication is vital for the control of the immune response, whereas dysfunction of this dynamic crosstalk results in microglial activation and exaggerated inflammation [23,48,49]. In a proof-of-concept study, we revealed alterations in the CX3CL1-CX3CR1 and CD200-CD200R axes in the MIA model, and we have suggested that some of these can be essential for the development of behavioural disturbances [30].
As a consequence of the abovementioned data, the first primary objective of the present study was to establish whether MIA might also affect the basal and additionally stimulated by the bacterial endotoxin biochemical and immunological statuses of the offspring’s frontal cortex in ex vivo conditions. To answer this question, we employed organotypic cultures prepared from this brain structure, as its role in schizophrenia-like pathomechanisms is widely accepted [50,51,52]. The main advantages of organotypic cultures are their ability to partially retain a three-dimensional organization as well as the fact that they are anatomically intact and reflect, to some extent, their brain area of origin [53]. Since neurons and glia in OCCs represent the populations of corresponding cells found in vivo [54,55], this type of culture can constitute an alternative tool to certain in vivo experiments, allowing for the reduction in the number of animals required for studies [56] on cellular and molecular processes of the brain ex vivo [53].
In our study, we showed that both control and MIA OCCs were susceptible to LPS exposure, as confirmed by not only the higher secretion of LDH and NO but also the extensive immunological response (an increase in Il-1β, Il-6, Cebpb, Arg1 and Il-10 as well as a decrease in Tgf-β mRNA expression in both types of OCCs and additionally elevated Cd40 level in control OCCs) to treatment with this bacterial endotoxin. Comparable observations regarding the effectiveness of LPS stimulation were described for control organotypic cultures prepared from the hippocampus (OHCs), in which incubation with the bacterial endotoxin resulted in the upregulated efflux of LDH, raised propidium iodide uptake, enhanced levels of nitrite, iNOS, IL-1β and TNF-α as well as decreased thiazolyl blue tetrazolium bromide reduction [57,58]. Moreover, in the present research, we demonstrated that MIA led to higher LDH release and increased vulnerability of OCCs to external stimulation with LPS, as revealed by the elevated secretion of NO. These results may indicate that MIA negatively affected the elementary biochemical processes of the cultures and intensified their susceptibility to the adverse influence of the additional stimulus. A similar phenomenon resembling the “priming” effect to subsequent LPS treatment was presented for OHCs prepared from the offspring of female rats with experimentally induced diabetes [59]. In this type of culture, incubation with the bacterial endotoxin caused a more pronounced inflammatory response and upregulated propidium iodide uptake than in control OHCs. All the abovementioned information further affirms the significance of the events occurring in the prenatal period on the developmental trajectory of the central nervous system and their possible long-lasting consequences for the offspring.
Gene expression analyses conducted herein showed that MIA significantly reduced the Cd200r mRNA level in OCCs. Exposure to the bacterial endotoxin (applied as a “second hit”) enhanced the impact of MIA and simultaneously triggered a deficit in Cx3cr1 expression in both types of cultures. In previous studies, we revealed that MIA-generated malfunctions in the neuron–microglia axes were already present in the brains of the offspring at the early stages of life, specifically when rats were 7 days old [30]. Hence, our current data, corresponding partially with the observations obtained in the in vivo MIA model, seem to support the translational nature of the OCC technique.
In the present research, disturbances in gene expression were not accompanied by altered protein levels of CD200R or CX3CR1 in MIA OCCs under basal conditions or after additional LPS stimulation. The incompatible influence of MIA on these parameters might emerge from alterations in the regulation of various stages of mRNA expression, starting with changes in chromatin conformation, gene activation in response to external stimuli and control of the transcription process [60,61].
CD200-CD200R signalling constitutes one of the well-characterized checkpoint mechanisms that restrains the immune activity of microglia [62,63]. Through binding with the cognate receptor, CD200 initiates an intracellular signalling cascade, resulting in a general inhibition of microglial pro-inflammatory marker expression and cytokine responses to immune stimuli [64]. Malfunction of CD200-CD200R communication, as well as the CX3CL1-CX3CR1 pair, another critical neuron–microglia axis, limited the resolution of inflammation, potentiated microglial pro-inflammatory activity and exacerbated disease severity and/or progression in several models of neuroinflammatory diseases [15,65,66]. Herein, we detected deficits in Cd200r and/or Cx3cr1 expression in MIA OCCs, yet we did not find significant changes in the mRNA levels of microglial markers (Cd40, Cd206, Arg1 and Cebpb) or the expression profiles of pro- (Il-1β and Il-6) and anti-inflammatory (Il-10 and Tgf-β) cytokines. Furthermore, the “second hit” did not particularly affect the microglial phenotype. In line with these observations, some researchers have suggested that disturbances in microglial activation are not a prerequisite for the presence of various deficits, including behavioural ones in MIA models [67,68].

4.2. Quetiapine Exerts Immunomodulatory Effects in Organotypic Cortical Cultures

In the second part of our study, we attempted to determine whether the anomalies in the expression of microglial receptors in MIA OCCs could represent a relevant target for quetiapine treatment. Unexpectedly, we did not observe an impact of quetiapine administration on the expression of the Cd200-Cd200r and Cx3cl1-Cx3cr1 systems in either control or MIA OCCs. Concurrently, the downregulation of Cd200r and Cx3cr1 gene expression resulting from the “second hit” with LPS was not modified by this drug. The lack of an effect of quetiapine treatment on neuron–microglia interactions was also displayed using ELISAs, quantifying the protein levels of both receptors in MIA OCCs.
At the mRNA level, the anti-inflammatory potential of quetiapine was presented in MIA OCCs in the form of the upregulation of Cd206 expression. The key role of this mannose receptor, among others, is to regulate the secretion of molecules during the inflammatory response [69,70]. Therefore, generally, CD206 expression during inflammation occurs at low levels, while in the course of the resolution of inflammation, it increases to ensure the clearance of dismissed inflammatory factors [71,72].
Regarding the influence of quetiapine on cytokine release, the antipsychotic prevented the boost of IL-6 production in MIA OCCs after the additional stimulation with LPS. Comparatively, Grolli et al. [73] demonstrated the suppressive quetiapine potential on IL-6 synthesis in the serum of rats submitted to chronic stress. The regulation of IL-6-mediated processes is highly precise, with several intracellular pathways being involved [74,75]. IL-6 forms a system with a specific IL-6R and can bind with two glycoprotein 130 (gp130) subunits with high affinity, creating a tetramer or hexamer IL-6/IL-6R/gp130 complex [75]. The gp130 molecule conducts biological signals through two major pathways, including the JAK/STAT and Ras/MAPK signalling cascades. Among the JAK kinases, the role of JAK1 with its main substrate, transcription factor STAT3, is of particular significance in the inflammatory response [76]. Additionally, quetiapine can inhibit the activation of NFκB signalling in neurons [77,78] and glial cells, simultaneously reducing the release of IL-1β and TNF-α [22].
Exposure to bacterial endotoxins constitutes one of the most potent inducers of not only pro- but also anti-inflammatory factors, including IL-10. Generally, this cytokine plays a critical role in balancing immune responses to prevent chronic inflammatory diseases [79]. In our research, pretreatment with quetiapine diminished the production of this negative modulator of inflammation in MIA OCCs subjected to LPS stimulation. This observation corresponds to some extent with the findings described by Jaehne et al. [80], who revealed that both quetiapine and its primary active metabolite norquetiapine appeared to affect the increase in IL-10 expression noted at 24 h post incubation with the bacterial endotoxin. Therefore, the suppressive impact of quetiapine on IL-10 level is at least partially an adverse effect leading to prolongation of the inflammatory reaction and preventing a return to homeostasis after LPS treatment. Based on the literature data [81,82,83], the role of LPS-induced activation of common signalling pathways for IL-6 and IL-10, including the JAK/STAT system, may be crucial to this phenomenon. However, this hypothesis demands further confirmation.
Furthermore, in our study, the potential of quetiapine was displayed in control OCCs in the form of suppression of Il-1β, Il-6, Cebpb and Arg1 expression under incubation with the bacterial endotoxin. To date, data concerning the anti-inflammatory action of quetiapine have varied and depended on the experimental model used, the dose and the administration method of the drug. Previously published reports underlined a double-edged immunomodulatory mechanism of this antipsychotic [15], which could be crucial in the context of the efficacy of quetiapine, as it was also influenced by the prior inflammatory activation state of cells [84]. Thus, the modulatory potential of quetiapine might be related to the inhibition of pro-inflammatory response rather than the stimulation of its anti-inflammatory components. Nevertheless, the mechanism of this action requires further research, particularly in ex vivo conditions.
To consolidate the results of the current research regarding the immunomodulatory properties of quetiapine and the detected deficiency in Cd200r expression in MIA OCCs, we introduced the positive regulation of CD200R via its agonist, specifically CD200Fc [85,86], to our experimental design. The notable finding described within this article is the suppressive effect of CD200Fc treatment on the IL-6 release in MIA OCCs in the presence of the “second hit”. The external modulation of CD200R via its ligand did not produce a more prominent impact on the investigated parameters in MIA OCCs than quetiapine stimulation. To the best of our knowledge, these results are the first that compared the action of quetiapine and CD200R modulation in MIA OCCs, providing new information on this phenomenon.

4.3. Limitations of the Study

We are fully aware of the limitations of the present research. First, only one dose of quetiapine (10 µM) was selected for the main experiments in our study, which undoubtedly reduced the basis for unambiguous conclusions regarding the mechanism of action of this antipsychotic. Second, we performed the analyses by applying only one selected concentration of LPS (1 µg/mL), the effect of which as the “second hit” stimulation was limited in some experiments and could subsequently influence the further effectiveness of quetiapine treatment. Moreover, in terms of most of the examined immunological parameters, we quantified only mRNA expression without measuring their protein levels. Furthermore, a broader perspective on the CD200-CD200R dyad would be achieved by additional investigation of the activation of signalling pathways related to this axis. Finally, the matter of high variability in group sizes including a couple of small groups in some experiments should be noted.

5. Conclusions

The present study demonstrated that MIA had an adverse effect on the biochemical status of OCCs and the expression of microglial receptors engaged in neuron–microglia communication under basal conditions and/or after “second hit” stimulation. The MIA procedure determined the OCCs’ response to quetiapine treatment, the immunomodulatory potential of which was expressed mainly as the inhibition of the negative impact of LPS on IL-6 and IL-10 release. In the current research, we did not observe the direct involvement of the CD200-CD200R axis in the mechanism of quetiapine action. Therefore, the question of whether the effects of CD200R modulation via CD200Fc and antipsychotic administration may be interrelated remains open, supporting further research on the involvement of this microglial receptor in the immunoregulating properties of quetiapine. Undoubtedly, our data strengthened the utility of the MIA model and OCC technique for investigating the processes underlying prenatal immune activation and searching for new targets for the pharmacological treatment of MIA-related conditions.

Author Contributions

Conceptualization, K.C. (Katarzyna Chamera) and A.B.-K.; methodology, K.C. (Katarzyna Chamera), K.C. (Katarzyna Curzytek), K.K., E.T., M.L. and K.T.; formal analysis, K.C. (Katarzyna Chamera) and K.C. (Katarzyna Curzytek); investigation, K.C. (Katarzyna Chamera), K.C. (Katarzyna Curzytek), K.K., E.T., M.L., K.T. and M.R.; resources, A.B.-K.; data curation, K.C. (Katarzyna Chamera) and K.C. (Katarzyna Curzytek); writing—original draft preparation, K.C. (Katarzyna Chamera) and A.B.-K.; writing—review and editing, K.C. (Katarzyna Chamera) and A.B.-K.; supervision, A.B.-K.; project administration, A.B.-K.; funding acquisition, A.B.-K. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by the National Science Centre, Poland, grant no. 2015/19/B/NZ7/02394 (OPUS), and the statutory funds of the Maj Institute of Pharmacology, Polish Academy of Sciences.

Institutional Review Board Statement

All procedures were performed under the approval of the Animal Care Committee of the Maj Institute of Pharmacology, Polish Academy of Sciences, Cracow and followed the recommendations of the International Council for Laboratory Animal Science and Guide for the Care and Use of Laboratory Animals (consent numbers: 236/2016 and 128/2018).

Data Availability Statement

All data supporting the conclusions of this manuscript are provided in the text and figures.

Acknowledgments

We would like to express our appreciation to Adam Roman for consulting on the statistical analysis of the data.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

References

  1. Hudepohl, N.S.; Nasrallah, H.A. Antipsychotic Drugs. Handb. Clin. Neurol. 2012, 106, 657–667. [Google Scholar]
  2. Kaar, S.J.; Natesan, S.; McCutcheon, R.; Howes, O.D. Antipsychotics: Mechanisms underlying clinical response and side-effects and novel treatment approaches based on pathophysiology. Neuropharmacology 2020, 172, 107704. [Google Scholar] [CrossRef]
  3. Meltzer, H.Y. Update on Typical and Atypical Antipsychotic Drugs. Annu. Rev. Med. 2013, 64, 393–406. [Google Scholar] [CrossRef]
  4. Stępnicki, P.; Kondej, M.; Kaczor, A.A. Current Concepts and Treatments of Schizophrenia. Molecules 2018, 23, 2087. [Google Scholar] [CrossRef]
  5. Kane, J.M.; Correll, C.U. Past and Present Progress in the Pharmacologic Treatment of Schizophrenia. J. Clin. Psychiatry 2010, 71, 1115–1124. [Google Scholar] [CrossRef]
  6. Kusumi, I.; Boku, S.; Takahashi, Y. Psychopharmacology of atypical antipsychotic drugs: From the receptor binding profile to neuroprotection and neurogenesis. Psychiatry Clin. Neurosci. 2015, 69, 243–258. [Google Scholar] [CrossRef]
  7. Adityanjee; Schulz, S.C. Clinical use of quetiapine in disease states other than schizophrenia. J. Clin. Psychiatry 2002, 63 (Suppl. S1), 32–38. [Google Scholar]
  8. Suttajit, S.; Srisurapanont, M.; Maneeton, N.; Maneeton, B. Quetiapine for acute bipolar depression: A systematic review and meta-analysis. Drug Des. Dev. Ther. 2014, 8, 827–838. [Google Scholar] [CrossRef]
  9. Chen, J.J.; Hua, H.; Massihi, L.; Portillo, I.; Alipour, A.; Ondo, W.; Dashtipour, K. Systematic Literature Review of Quetiapine for the Treatment of Psychosis in Patients With Parkinsonism. J. Neuropsychiatry Clin. Neurosci. 2019, 31, 188–195. [Google Scholar] [CrossRef]
  10. Kasper, S.; Tauscher, J.; Heiden, A. Quetiapine: Efficacy and tolerability in schizophrenia. Eur. Neuropsychopharmacol. 2001, 11, S405–S413. [Google Scholar] [CrossRef]
  11. Leucht, S.; Cipriani, A.; Spineli, L.; Mavridis, D.; Örey, D.; Richter, F.; Samara, M.; Barbui, C.; Engel, R.R.; Geddes, J.R.; et al. Comparative efficacy and tolerability of 15 antipsychotic drugs in schizophrenia: A multiple-treatments meta-analysis. Lancet 2013, 382, 951–962. [Google Scholar] [CrossRef]
  12. Cheer, S.M.; Wagstaff, A.J. Quetiapine. CNS Drugs 2004, 18, 173–199. [Google Scholar] [CrossRef]
  13. Shoemaker, J.M.; Pitcher, L.; Noh, H.R.; Swerdlow, N.R. Quetiapine produces a prolonged reversal of the sensorimotor gating-disruptive effects of basolateral amygdala lesions in rats. Behav. Neurosci. 2003, 117, 136–143. [Google Scholar] [CrossRef]
  14. Carreño, F.; Helfer, V.E.; Staudt, K.J.; Paese, K.; Meyer, F.S.; Herrmann, A.P.; Guterres, S.S.; Rates, S.M.K.; Dalla Costa, T. Quetiapine lipid core nanocapsules restore prepulse inhibition deficits in a neurodevelopmental model of schizophrenia in male and female rats. Schizophr. Res. 2020, 218, 173–179. [Google Scholar] [CrossRef]
  15. Chamera, K.; Curzytek, K.; Kamińska, K.; Trojan, E.; Basta-Kaim, A. Quetiapine Ameliorates MIA-Induced Impairment of Sensorimotor Gating: Focus on Neuron-Microglia Communication and the Inflammatory Response in the Frontal Cortex of Adult Offspring of Wistar Rats. Cells 2022, 11, 2788. [Google Scholar] [CrossRef]
  16. He, J.; Zu, Q.; Wen, C.; Liu, Q.; You, P.; Li, X.; Wang, W. Quetiapine Attenuates Schizophrenia-Like Behaviors and Demyelination in a MK-801–Induced Mouse Model of Schizophrenia. Front. Psychiatry 2020, 11, 843. [Google Scholar] [CrossRef]
  17. Baldwin, C.M.; Scott, L.J. Quetiapine Extended Release. CNS Drugs 2009, 23, 261–269. [Google Scholar] [CrossRef]
  18. Mauri, M.C.; Paletta, S.; Maffini, M.; Colasanti, A.; Dragogna, F.; Di Pace, C.; Altamura, A.C. Clinical pharmacology of atypical antipsychotics: An update. EXCLI J. 2014, 13, 1163–1191. [Google Scholar]
  19. Bian, Q.; Kato, T.; Monji, A.; Hashioka, S.; Mizoguchi, Y.; Horikawa, H.; Kanba, S. The effect of atypical antipsychotics, perospirone, ziprasidone and quetiapine on microglial activation induced by interferon-γ. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2008, 32, 42–48. [Google Scholar] [CrossRef]
  20. Shao, Y.; Peng, H.; Huang, Q.; Kong, J.; Xu, H. Quetiapine mitigates the neuroinflammation and oligodendrocyte loss in the brain of C57BL/6 mouse following cuprizone exposure for one week. Eur. J. Pharmacol. 2015, 765, 249–257. [Google Scholar] [CrossRef]
  21. Zhao, Z.; Luo, G.; Liu, M.; Guo, H.; Xue, M.; Wang, X.; Li, X.-M.; He, J. Quetiapine reduces microglial number in the hippocampus of a transgenic mouse model of Alzheimer’s disease. Neuroreport 2014, 25, 870–874. [Google Scholar] [CrossRef]
  22. Zhu, S.; Shi, R.; Li, V.; Wang, J.-F.; Zhang, R.; Tempier, A.; He, J.; Kong, J.; Wang, J.-F.; Li, X.-M. Quetiapine Attenuates Glial Activation and Proinflammatory Cytokines in APP/PS1 Transgenic Mice via Inhibition of Nuclear Factor-κB Pathway. Int. J. Neuropsychopharmacol. 2015, 18, pyu022. [Google Scholar] [CrossRef]
  23. Szepesi, Z.; Manouchehrian, O.; Bachiller, S.; Deierborg, T. Bidirectional Microglia–Neuron Communication in Health and Disease. Front. Cell. Neurosci. 2018, 12, 323. [Google Scholar] [CrossRef]
  24. Chamera, K.; Trojan, E.; Szuster-Głuszczak, M.; Basta-Kaim, A. The Potential Role of Dysfunctions in Neuron-Microglia Communication in the Pathogenesis of Brain Disorders. Curr. Neuropharmacol. 2020, 18, 408–430. [Google Scholar] [CrossRef]
  25. Gorczynski, R.M.; Cattral, M.S.; Chen, Z.; Hu, J.; Lei, J.; Min, W.P.; Yu, G.; Ni, J. An immunoadhesin incorporating the molecule OX-2 is a potent immunosuppressant that prolongs allo- and xenograft survival. J. Immunol. 1999, 163, 1654–1660. [Google Scholar] [CrossRef]
  26. Erin, N.; Tanrıöver, G.; Curry, A.; Akman, M.; Duymuş, Ö.; Gorczynski, R. CD200fc enhances anti-tumoral immune response and inhibits visceral metastasis of breast carcinoma. Oncotarget 2018, 9, 19147–19158. [Google Scholar] [CrossRef]
  27. Humpel, C. Organotypic Brain Slice Cultures. Curr. Protoc. Immunol. 2018, 123, e59. [Google Scholar] [CrossRef]
  28. Romero, E.; Ali, C.; Molina-Holgado, E.; Castellano, B.; Guaza, C.; Borrell, J. Neurobehavioral and Immunological Consequences of Prenatal Immune Activation in Rats. Influence of Antipsychotics. Neuropsychopharmacology 2007, 32, 1791–1804. [Google Scholar] [CrossRef]
  29. Basta-Kaim, A.; Szczęsny, E.; Leśkiewicz, M.; Głombik, K.; Ślusarczyk, J.; Budziszewska, B.; Regulska, M.; Kubera, M.; Nowak, W.; Wędzony, K.; et al. Maternal immune activation leads to age-related behavioral and immunological changes in male rat offspring—The effect of antipsychotic drugs. Pharmacol. Rep. 2012, 64, 1400–1410. [Google Scholar] [CrossRef]
  30. Chamera, K.; Kotarska, K.; Szuster-Głuszczak, M.; Trojan, E.; Skórkowska, A.; Pomierny, B.; Krzyżanowska, W.; Bryniarska, N.; Basta-Kaim, A. The prenatal challenge with lipopolysaccharide and polyinosinic:polycytidylic acid disrupts CX3CL1-CX3CR1 and CD200-CD200R signalling in the brains of male rat offspring: A link to schizophrenia-like behaviours. J. Neuroinflamm. 2020, 17, 247. [Google Scholar] [CrossRef]
  31. Vojtechova, I.; Maleninska, K.; Kutna, V.; Klovrza, O.; Tuckova, K.; Petrasek, T.; Stuchlik, A. Behavioral Alterations and Decreased Number of Parvalbumin-Positive Interneurons in Wistar Rats after Maternal Immune Activation by Lipopolysaccharide: Sex Matters. Int. J. Mol. Sci. 2021, 22, 3274. [Google Scholar] [CrossRef] [PubMed]
  32. Monte, A.S.; Mello, B.S.F.; Borella, V.C.M.; da Silva Araujo, T.; da Silva, F.E.R.; de Sousa, F.C.F.; de Oliveira, A.C.P.; Gama, C.S.; Seeman, M.V.; Vasconcelos, S.M.M.; et al. Two-hit model of schizophrenia induced by neonatal immune activation and peripubertal stress in rats: Study of sex differences and brain oxidative alterations. Behav. Brain Res. 2017, 331, 30–37. [Google Scholar] [CrossRef] [PubMed]
  33. Chamera, K.; Szuster-Głuszczak, M.; Trojan, E.; Basta-Kaim, A. Maternal Immune Activation Sensitizes Male Offspring Rats to Lipopolysaccharide-Induced Microglial Deficits Involving the Dysfunction of CD200–CD200R and CX3CL1–CX3CR1 Systems. Cells 2020, 9, 1676. [Google Scholar] [CrossRef] [PubMed]
  34. Basta-Kaim, A.; Fijał, K.; Budziszewska, B.; Regulska, M.; Leśkiewicz, M.; Kubera, M.; Gołembiowska, K.; Lasoń, W.; Wędzony, K. Prenatal lipopolysaccharide treatment enhances MK-801-induced psychotomimetic effects in rats. Pharmacol. Biochem. Behav. 2011, 98, 241–249. [Google Scholar] [CrossRef]
  35. Basta-Kaim, A.; Budziszewska, B.; Leśkiewicz, M.; Fijał, K.; Regulska, M.; Kubera, M.; Wȩdzony, K.; Lasoń, W. Hyperactivity of the hypothalamus-pituitary-adrenal axis in lipopolysaccharide-induced neurodevelopmental model of schizophrenia in rats: Effects of antipsychotic drugs. Eur. J. Pharmacol. 2011, 650, 586–595. [Google Scholar] [CrossRef]
  36. Stoppini, L.; Buchs, P.-A.; Muller, D. A simple method for organotypic cultures of nervous tissue. J. Neurosci. Methods 1991, 37, 173–182. [Google Scholar] [CrossRef]
  37. Nicholls, S.M.; Copland, D.A.; Vitova, A.; Kuffova, L.; Forrester, J.V.; Dick, A.D. Local targeting of the CD200-CD200R axis does not promote corneal graft survival. Exp. Eye Res. 2015, 130, 1–8. [Google Scholar] [CrossRef]
  38. Trojan, E.; Tylek, K.; Leśkiewicz, M.; Lasoń, W.; Brandenburg, L.-O.; Leopoldo, M.; Lacivita, E.; Basta-Kaim, A. The N-Formyl Peptide Receptor 2 (FPR2) Agonist MR-39 Exhibits Anti-Inflammatory Activity in LPS-Stimulated Organotypic Hippocampal Cultures. Cells 2021, 10, 1524. [Google Scholar] [CrossRef]
  39. Chomczynski, P. A reagent for the single-step simultaneous isolation of RNA, DNA and proteins from cell and tissue samples. Biotechniques 1993, 15, 532–534, 536–537. [Google Scholar]
  40. Comer, A.L.; Carrier, M.; Tremblay, M.-È.; Cruz-Martín, A. The Inflamed Brain in Schizophrenia: The Convergence of Genetic and Environmental Risk Factors That Lead to Uncontrolled Neuroinflammation. Front. Cell. Neurosci. 2020, 14, 274. [Google Scholar] [CrossRef]
  41. Kabba, J.A.; Xu, Y.; Christian, H.; Ruan, W.; Chenai, K.; Xiang, Y.; Zhang, L.; Saavedra, J.M.; Pang, T. Microglia: Housekeeper of the Central Nervous System. Cell. Mol. Neurobiol. 2018, 38, 53–71. [Google Scholar] [CrossRef] [PubMed]
  42. Boksa, P. Effects of prenatal infection on brain development and behavior: A review of findings from animal models. Brain. Behav. Immun. 2010, 24, 881–897. [Google Scholar] [CrossRef] [PubMed]
  43. Estes, M.L.; McAllister, A.K. Maternal immune activation: Implications for neuropsychiatric disorders. Science. 2016, 353, 772–777. [Google Scholar] [CrossRef] [PubMed]
  44. Brown, A.S.; Meyer, U. Maternal Immune Activation and Neuropsychiatric Illness: A Translational Research Perspective. Am. J. Psychiatry 2018, 175, 1073–1083. [Google Scholar] [CrossRef]
  45. Basta-Kaim, A.; Fijał, K.; Ślusarczyk, J.; Trojan, E.; Głombik, K.; Budziszewska, B.; Leśkiewicz, M.; Regulska, M.; Kubera, M.; Lasoń, W.; et al. Prenatal administration of lipopolysaccharide induces sex-dependent changes in glutamic acid decarboxylase and parvalbumin in the adult rat brain. Neuroscience 2015, 287, 78–92. [Google Scholar] [CrossRef]
  46. Zager, A.; Pinheiro, M.L.; Ferraz-de-Paula, V.; Ribeiro, A.; Palermo-Neto, J. Increased cell-mediated immunity in male mice offspring exposed to maternal immune activation during late gestation. Int. Immunopharmacol. 2013, 17, 633–637. [Google Scholar] [CrossRef]
  47. Savareh, E.; Davoodian, N.; Mousaviyan, R.; Ghasemi-Kasman, M.; Atashabparvar, A.; Eftekhar, E. Prenatal Zinc Supplementation Ameliorates Hippocampal Astrocytes Activation and Inflammatory Cytokines Expression Induced by Lipopolysaccharide in a Rat Model of Maternal Immune Activation. Basic Clin. Neurosci. J. 2022, 13, 335–348. [Google Scholar] [CrossRef]
  48. Paolicelli, R.C.; Bisht, K.; Tremblay, M.-Ã. Fractalkine regulation of microglial physiology and consequences on the brain and behavior. Front. Cell. Neurosci. 2014, 8, 129. [Google Scholar] [CrossRef]
  49. Wohleb, E.S. Neuron–Microglia Interactions in Mental Health Disorders: “For Better, and For Worse”. Front. Immunol. 2016, 7, 544. [Google Scholar] [CrossRef]
  50. Amodeo, D.A.; Lai, C.-Y.; Hassan, O.; Mukamel, E.A.; Behrens, M.M.; Powell, S.B. Maternal immune activation impairs cognitive flexibility and alters transcription in frontal cortex. Neurobiol. Dis. 2019, 125, 211–218. [Google Scholar] [CrossRef]
  51. Tendilla-Beltrán, H.; del Carmen Sanchez-Islas, N.; Marina-Ramos, M.; Leza, J.C.; Flores, G. The prefrontal cortex as a target for atypical antipsychotics in schizophrenia, lessons of neurodevelopmental animal models. Prog. Neurobiol. 2021, 199, 101967. [Google Scholar] [CrossRef] [PubMed]
  52. Hanson, K.L.; Grant, S.E.; Funk, L.H.; Schumann, C.M.; Bauman, M.D. Impact of Maternal Immune Activation on Nonhuman Primate Prefrontal Cortex Development: Insights for Schizophrenia. Biol. Psychiatry 2022, 92, 460–469. [Google Scholar] [CrossRef] [PubMed]
  53. Humpel, C. Organotypic brain slice cultures: A review. Neuroscience 2015, 305, 86–98. [Google Scholar] [CrossRef] [PubMed]
  54. Beach, R.L.; Bathgate, S.L.; Cotman, C.W. Identification of cell types in rat hippocampal slices maintained in organotypic cultures. Dev. Brain Res. 1982, 3, 3–20. [Google Scholar] [CrossRef]
  55. Staal, J.A.; Alexander, S.R.; Liu, Y.; Dickson, T.D.; Vickers, J.C. Characterization of Cortical Neuronal and Glial Alterations during Culture of Organotypic Whole Brain Slices from Neonatal and Mature Mice. PLoS ONE 2011, 6, e22040. [Google Scholar] [CrossRef]
  56. Croft, C.L.; Futch, H.S.; Moore, B.D.; Golde, T.E. Organotypic brain slice cultures to model neurodegenerative proteinopathies. Mol. Neurodegener. 2019, 14, 45. [Google Scholar] [CrossRef]
  57. Ślusarczyk, J.; Piotrowski, M.; Szczepanowicz, K.; Regulska, M.; Leśkiewicz, M.; Warszyński, P.; Budziszewska, B.; Lasoń, W.; Basta-Kaim, A. Nanocapsules with Polyelectrolyte Shell as a Platform for 1,25-dihydroxyvitamin D3 Neuroprotection: Study in Organotypic Hippocampal Slices. Neurotox. Res. 2016, 30, 581–592. [Google Scholar] [CrossRef]
  58. Basta-Kaim, A.; Ślusarczyk, J.; Szczepanowicz, K.; Warszyński, P.; Leśkiewicz, M.; Regulska, M.; Trojan, E.; Lasoń, W. Protective effects of polydatin in free and nanocapsulated form on changes caused by lipopolysaccharide in hippocampal organotypic cultures. Pharmacol. Rep. 2019, 71, 603–613. [Google Scholar] [CrossRef]
  59. Głombik, K.; Trojan, E.; Kurek, A.; Budziszewska, B.; Basta-Kaim, A. Inflammatory Consequences of Maternal Diabetes on the Offspring Brain: A Hippocampal Organotypic Culture Study. Neurotox. Res. 2019, 36, 357–375. [Google Scholar] [CrossRef]
  60. Doma, M.K.; Parker, R. RNA Quality Control in Eukaryotes. Cell 2007, 131, 660–668. [Google Scholar] [CrossRef]
  61. Houseley, J.; Tollervey, D. The Many Pathways of RNA Degradation. Cell 2009, 136, 763–776. [Google Scholar] [CrossRef] [PubMed]
  62. Frank, M.G.; Annis, J.L.; Watkins, L.R.; Maier, S.F. Glucocorticoids mediate stress induction of the alarmin HMGB1 and reduction of the microglia checkpoint receptor CD200R1 in limbic brain structures. Brain. Behav. Immun. 2019, 80, 678–687. [Google Scholar] [CrossRef] [PubMed]
  63. Le, C.; Hao, X.; Li, J.; Zhong, J.; Lin, H.; Zhou, Y.; Travis, Z.D.; Tong, L.; Gao, F. CD200Fc Improves Neurological Function by Protecting the Blood–brain Barrier after Intracerebral Hemorrhage. Cell Transplant. 2019, 28, 1321–1328. [Google Scholar] [CrossRef] [PubMed]
  64. Zhang, S.; Cherwinski, H.; Sedgwick, J.D.; Phillips, J.H. Molecular Mechanisms of CD200 Inhibition of Mast Cell Activation. J. Immunol. 2004, 173, 6786–6793. [Google Scholar] [CrossRef]
  65. Frank, M.G.; Fonken, L.K.; Annis, J.L.; Watkins, L.R.; Maier, S.F. Stress disinhibits microglia via down-regulation of CD200R: A mechanism of neuroinflammatory priming. Brain. Behav. Immun. 2018, 69, 62–73. [Google Scholar] [CrossRef]
  66. Chamera, K.; Trojan, E.; Kotarska, K.; Szuster-Głuszczak, M.; Bryniarska, N.; Tylek, K.; Basta-Kaim, A. Role of Polyinosinic:Polycytidylic Acid-Induced Maternal Immune Activation and Subsequent Immune Challenge in the Behaviour and Microglial Cell Trajectory in Adult Offspring: A Study of the Neurodevelopmental Model of Schizophrenia. Int. J. Mol. Sci. 2021, 22, 1558. [Google Scholar] [CrossRef]
  67. Smolders, S.; Smolders, S.M.T.; Swinnen, N.; Gärtner, A.; Rigo, J.-M.; Legendre, P.; Brône, B. Maternal immune activation evoked by polyinosinic:polycytidylic acid does not evoke microglial cell activation in the embryo. Front. Cell. Neurosci. 2015, 9, 301. [Google Scholar] [CrossRef]
  68. Giovanoli, S.; Weber-Stadlbauer, U.; Schedlowski, M.; Meyer, U.; Engler, H. Prenatal immune activation causes hippocampal synaptic deficits in the absence of overt microglia anomalies. Brain. Behav. Immun. 2016, 55, 25–38. [Google Scholar] [CrossRef]
  69. Tidball, J.G.; Villalta, S.A. Regulatory interactions between muscle and the immune system during muscle regeneration. Am. J. Physiol. Integr. Comp. Physiol. 2010, 298, R1173–R1187. [Google Scholar] [CrossRef]
  70. Stakenborg, M.; Abdurahiman, S.; De Simone, V.; Goverse, G.; Stakenborg, N.; van Baarle, L.; Wu, Q.; Pirottin, D.; Kim, J.-S.; Chappell-Maor, L.; et al. Enteric glial cells favor accumulation of anti-inflammatory macrophages during the resolution of muscularis inflammation. Mucosal Immunol. 2022, 15, 1296–1308. [Google Scholar] [CrossRef]
  71. Lee, S.J.; Evers, S.; Roeder, D.; Parlow, A.F.; Risteli, J.; Risteli, L.; Lee, Y.C.; Feizi, T.; Langen, H.; Nussenzweig, M.C. Mannose Receptor-Mediated Regulation of Serum Glycoprotein Homeostasis. Science 2002, 295, 1898–1901. [Google Scholar] [CrossRef] [PubMed]
  72. Porcheray, F.; Viaud, S.; Rimaniol, A.-C.; Léone, C.; Samah, B.; Dereuddre-Bosquet, N.; Dormont, D.; Gras, G. Macrophage activation switching: An asset for the resolution of inflammation. Clin. Exp. Immunol. 2005, 142, 481–489. [Google Scholar] [CrossRef] [PubMed]
  73. Grolli, R.E.; Bertollo, A.G.; Behenck, J.P.; de Araujo Borba, L.; Plissari, M.E.; Soares, S.J.B.; Manica, A.; da Silva Joaquim, L.; Petronilho, F.; Quevedo, J.; et al. Quetiapine effect on depressive-like behaviors, oxidative balance, and inflammation in serum of rats submitted to chronic stress. Naunyn-Schmiedeberg’s. Arch. Pharmacol. 2023; Online ahead of print. [Google Scholar] [CrossRef]
  74. Wolf, J.; Rose-John, S.; Garbers, C. Interleukin-6 and its receptors: A highly regulated and dynamic system. Cytokine 2014, 70, 11–20. [Google Scholar] [CrossRef] [PubMed]
  75. Aliyu, M.; Zohora, F.T.; Anka, A.U.; Ali, K.; Maleknia, S.; Saffarioun, M.; Azizi, G. Interleukin-6 cytokine: An overview of the immune regulation, immune dysregulation, and therapeutic approach. Int. Immunopharmacol. 2022, 111, 109130. [Google Scholar] [CrossRef]
  76. Hunter, C.A.; Jones, S.A. IL-6 as a keystone cytokine in health and disease. Nat. Immunol. 2015, 16, 448–457. [Google Scholar] [CrossRef]
  77. Bi, X.; Yan, B.; Fang, S.; Yang, Y.; He, J.; Li, X.-M.; Kong, J. Quetiapine regulates neurogenesis in ischemic mice by inhibiting NF- κ B p65/p50 expression. Neurol. Res. 2009, 31, 159–166. [Google Scholar] [CrossRef]
  78. Panizzutti, B.; Bortolasci, C.C.; Spolding, B.; Kidnapillai, S.; Connor, T.; Richardson, M.F.; Truong, T.T.T.; Liu, Z.S.J.; Morris, G.; Gray, L.; et al. Transcriptional Modulation of the Hippo Signaling Pathway by Drugs Used to Treat Bipolar Disorder and Schizophrenia. Int. J. Mol. Sci. 2021, 22, 7164. [Google Scholar] [CrossRef]
  79. Burmeister, A.R.; Marriott, I. The Interleukin-10 Family of Cytokines and Their Role in the CNS. Front. Cell. Neurosci. 2018, 12, 458. [Google Scholar] [CrossRef]
  80. Jaehne, E.J.; Corrigan, F.; Toben, C.; Jawahar, M.C.; Baune, B.T. The effect of the antipsychotic drug quetiapine and its metabolite norquetiapine on acute inflammation, memory and anhedonia. Pharmacol. Biochem. Behav. 2015, 135, 136–144. [Google Scholar] [CrossRef]
  81. Ahmed, S.T.; Ivashkiv, L.B. Inhibition of IL-6 and IL-10 Signaling and Stat Activation by Inflammatory and Stress Pathways. J. Immunol. 2000, 165, 5227–5237. [Google Scholar] [CrossRef]
  82. Minogue, A.M.; Barrett, J.P.; Lynch, M.A. LPS-induced release of IL-6 from glia modulates production of IL-1β in a JAK2-dependent manner. J. Neuroinflamm. 2012, 9, 629. [Google Scholar] [CrossRef] [PubMed]
  83. Zhou, Y.; Wang, J.; Yang, W.; Qi, X.; Lan, L.; Luo, L.; Yin, Z. Bergapten prevents lipopolysaccharide-induced inflammation in RAW264.7 cells through suppressing JAK/STAT activation and ROS production and increases the survival rate of mice after LPS challenge. Int. Immunopharmacol. 2017, 48, 159–168. [Google Scholar] [CrossRef] [PubMed]
  84. Turra, B.O.; Barbisan, F.; Azzolin, V.F.; Teixeira, C.F.; Flores, T.; Braun, L.E.; de Oliveira Nerys, D.A.; Rissi, V.B.; de Oliveira Alves, A.; Assmann, C.E.; et al. Unmetabolized quetiapine exerts an in vitro effect on innate immune cells by modulating inflammatory response and neutrophil extracellular trap formation. Biomed. Pharmacother. 2020, 131, 110497. [Google Scholar] [CrossRef] [PubMed]
  85. Cox, F.F.; Carney, D.; Miller, A.-M.; Lynch, M.A. CD200 fusion protein decreases microglial activation in the hippocampus of aged rats. Brain. Behav. Immun. 2012, 26, 789–796. [Google Scholar] [CrossRef]
  86. Jiang, L.; Xu, F.; He, W.; Chen, L.; Zhong, H.; Wu, Y.; Zeng, S.; Li, L.; Li, M. CD200Fc reduces TLR4-mediated inflammatory responses in LPS-induced rat primary microglial cells via inhibition of the NF-κB pathway. Inflamm. Res. 2016, 65, 521–532. [Google Scholar] [CrossRef]
Figure 1. Schedule of the experimental procedure applied in the study. Thirty minutes after the last medium change, both control and MIA OCCs were subjected to one of three treatments. Experiment 1: OCCs were incubated with LPS (1 µg/mL) for 24 h, and LDH and NO assays were then performed. The corresponding results are presented in Figure 2. Experiment 2: OCCs were first exposed to quetiapine (1, 5 or 10 µM) for 90 min and later to LPS (1 µg/mL) for 24 h. LDH and NO tests (with quetiapine at all doses), ELISAs and qRT-PCR (with quetiapine at 10 µM) were carried out, and the results of these analyses are displayed in Figure 3, Figure 4, Figure 5 and Figure 6. Experiment 3: MIA OCCs were treated with CD200Fc (5 µg/mL) for 12 h. Quetiapine (10 µM) was added to MIA OCCs for 90 min. Then, LPS (1 µg/mL) was introduced to the medium for 24 h. The ELISA method was used, and the corresponding results are shown in Figure 7.
Figure 1. Schedule of the experimental procedure applied in the study. Thirty minutes after the last medium change, both control and MIA OCCs were subjected to one of three treatments. Experiment 1: OCCs were incubated with LPS (1 µg/mL) for 24 h, and LDH and NO assays were then performed. The corresponding results are presented in Figure 2. Experiment 2: OCCs were first exposed to quetiapine (1, 5 or 10 µM) for 90 min and later to LPS (1 µg/mL) for 24 h. LDH and NO tests (with quetiapine at all doses), ELISAs and qRT-PCR (with quetiapine at 10 µM) were carried out, and the results of these analyses are displayed in Figure 3, Figure 4, Figure 5 and Figure 6. Experiment 3: MIA OCCs were treated with CD200Fc (5 µg/mL) for 12 h. Quetiapine (10 µM) was added to MIA OCCs for 90 min. Then, LPS (1 µg/mL) was introduced to the medium for 24 h. The ELISA method was used, and the corresponding results are shown in Figure 7.
Biomedicines 11 01405 g001
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

Chamera, K.; Curzytek, K.; Kamińska, K.; Trojan, E.; Leśkiewicz, M.; Tylek, K.; Regulska, M.; Basta-Kaim, A. Insights into the Potential Impact of Quetiapine on the Microglial Trajectory and Inflammatory Response in Organotypic Cortical Cultures Derived from Rat Offspring. Biomedicines 2023, 11, 1405. https://doi.org/10.3390/biomedicines11051405

AMA Style

Chamera K, Curzytek K, Kamińska K, Trojan E, Leśkiewicz M, Tylek K, Regulska M, Basta-Kaim A. Insights into the Potential Impact of Quetiapine on the Microglial Trajectory and Inflammatory Response in Organotypic Cortical Cultures Derived from Rat Offspring. Biomedicines. 2023; 11(5):1405. https://doi.org/10.3390/biomedicines11051405

Chicago/Turabian Style

Chamera, Katarzyna, Katarzyna Curzytek, Kinga Kamińska, Ewa Trojan, Monika Leśkiewicz, Kinga Tylek, Magdalena Regulska, and Agnieszka Basta-Kaim. 2023. "Insights into the Potential Impact of Quetiapine on the Microglial Trajectory and Inflammatory Response in Organotypic Cortical Cultures Derived from Rat Offspring" Biomedicines 11, no. 5: 1405. https://doi.org/10.3390/biomedicines11051405

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