Next Article in Journal
Solute Transport through Mitochondrial Porins In Vitro and In Vivo
Next Article in Special Issue
Lipid Rafts: The Maestros of Normal Brain Development
Previous Article in Journal
Rational Design of Key Enzymes to Efficiently Synthesize Phycocyanobilin in Escherichia coli
Previous Article in Special Issue
Methodological Pitfalls of Investigating Lipid Rafts in the Brain: What Are We Still Missing?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Increased NLRP1 mRNA and Protein Expression Suggests Inflammasome Activation in the Dorsolateral Prefrontal and Medial Orbitofrontal Cortex in Schizophrenia

1
Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
2
Wolfson Centre for Inherited Neuromuscular Disease, RJAH Orthopaedic Hospital, Oswestry SY10 7AG, UK
3
School of Pharmacy and Bioengineering, Keele University, Keele ST5 5BG, UK
4
Department of Forensic Medicine, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
5
Department of Psychiatry and Psychological Medicine, University Hospital Center Zagreb, 10000 Zagreb, Croatia
6
University of Zagreb School of Medicine, 10000 Zagreb, Croatia
7
Nash Family Department of Neuroscience, Center for Discovery and Innovation, and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA
8
Department of Psychiatry and Psychotherapy, University of Rostock, Gehlsheimer Str. 20, 18147 Rostock, Germany
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Biomolecules 2024, 14(3), 302; https://doi.org/10.3390/biom14030302
Submission received: 15 January 2024 / Revised: 22 February 2024 / Accepted: 29 February 2024 / Published: 4 March 2024
(This article belongs to the Special Issue Brain Sterols: Biosynthesis and Physiology in Health and Disease)

Abstract

:
Schizophrenia is a complex mental condition, with key symptoms marked for diagnosis including delusions, hallucinations, disorganized thinking, reduced emotional expression, and social dysfunction. In the context of major developmental hypotheses of schizophrenia, notably those concerning maternal immune activation and neuroinflammation, we studied NLRP1 expression and content in the postmortem brain tissue of 10 schizophrenia and 10 control subjects. In the medial orbitofrontal cortex (Brodmann’s area 11/12) and dorsolateral prefrontal cortex (area 46) from both hemispheres of six schizophrenia subjects, the NLRP1 mRNA expression was significantly higher than in six control brains (p < 0.05). As the expression difference was highest for the medial orbitofrontal cortex in the right hemisphere, we assessed NLRP1-immunoreactive pyramidal neurons in layers III, V, and VI in the medial orbitofrontal cortex in the right hemisphere of seven schizophrenia and five control brains. Compared to controls, we quantified a significantly higher number of NLRP1-positive pyramidal neurons in the schizophrenia brains (p < 0.01), suggesting NLRP1 inflammasome activation in schizophrenia subjects. Layer III pyramidal neuron dysfunction aligns with working memory deficits, while impairments of pyramidal neurons in layers V and VI likely disrupt predictive processing. We propose NLRP1 inflammasome as a potential biomarker and therapeutic target in schizophrenia.

1. Introduction

Schizophrenia (SZ) is a persistent, multifactorial mental condition that affects the generation of thoughts, reality perception, cognitive, linguistic, and emotional experience and expression, as well as social relationships [1]. It is characterized by delusions and hallucinations of variable severity (positive symptoms not normally seen in people who do not have SZ) and a disorganized flow of thoughts with consequent incoherent speech. According to the Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition, Text Revision (DSM-5-TR), the diagnosis of SZ requires the presence of two of these three symptoms for a significant portion of time during one month (or less if successfully treated) [2].
The Eleventh Edition of the International Classification of Diseases (ICD-11) by the World Health Organization [3] has harmonized its criteria to better align with the DSM’s criteria. ICD-11 suggests categorizing symptoms into six groups (positive, negative, depressive, manic, psychomotor, and cognitive), requiring the presence of at least two symptoms during a month. Among these two, one must be from the group of so-called basic symptoms (delusions, hallucinations, thought disorder, and distorted perception of reality) [3], facilitating easier comparisons and enhancing clinical usability [4]. Different assessment tools, such as the Clinician-Rated Dimensions of Psychosis Symptom Severity (C-RDPSS) introduced in the DSM-5, are designed to evaluate various groups of symptoms on a scale from 0 (absence of the symptom) to 5 (presence of the symptom in a severe form or to a significant extent), as reported by the individual in the previous seven days, and to monitor treatment success. The main challenge in diagnosing and treating SZ lies in its reliance on symptoms of a psychological nature rather than on clearly delineated pathophysiological or neurobiological mechanisms [5]. The current diagnostic criteria for SZ remain dichotomous, determining the presence or absence of the illness, and do not facilitate early diagnosis; instead, they require the progression of symptomatology before diagnosis. Consequently, there is a crucial need to align these criteria with contemporary scientific knowledge and refine them based on clinical neuroscience, genetic testing, biomarkers, neuroimaging, and personalized pharmacotherapy [6].
SZ is characterized as a disconnection syndrome that arises from the aberrant maturation and connectivity of the prefrontal cortex (PFC). This condition elevates the threshold for conscious perception due to the aberrant synaptic plasticity of glutamate, particularly the hypofunction of N-methyl-D-aspartate receptors (NMDARs), which occurs earlier during or before the juvenile period. This stage is later in adolescence influenced by the dysfunction of various neurotransmitter systems, mainly dopamine, serotonin, acetylcholine, and GABA (for a review, see [7]). Neuroinflammation is increasingly considered a key mechanism involved in this transition. While neuroinflammation plays a key role in the pathogenesis of many brain diseases, including Alzheimer’s disease [8,9,10], Huntington’s disease, Parkinson’s disease, drug use disorders, depression, and anxiety disorders, the relationship between the NLRP inflammasome and schizophrenia, autism, and many other neuropsychiatric disorders remains unknown (for a review, see [11]).
The NMDAR hypofunction hypothesis of SZ is derived from the observation that specific non-competitive NMDAR antagonists, such as phencyclidine and ketamine, elicit behaviors reminiscent of all three types of SZ symptoms (positive, negative, and cognitive) in humans [12,13]. It is posited that this hypofunction is responsible for cognitive and social deficits. This viewpoint is reinforced by the limited efficacy of antipsychotic drugs targeting the dopaminergic system in addressing these deficits in SZ and the development of treatment-resistant SZ (TRS) affecting up to a third of individuals with SZ [14,15]. The consequences of these alterations include deficits in both local circuitry processing within the PFC and its long-range connectivity, causing impairments in predictive processing—the key pathophysiological disturbance in SZ [16,17].
When normal predictive coding, mediated by layer V PFC top-down (feedback) projections to the visual area V4, is experimentally suppressed in monkeys during a task-altering visual predictability task, this intervention results in a lack of the normal increase in α and β electroencephalography (EEG) power that conveys expected inputs (power modulation is stimulus-specific). Simultaneously, unpredicted stimuli do not elicit a rise in spiking and γ band activity, as observed during normal conditions [18]. Similarly, it has been demonstrated that a greater prior expectation of volatility is elevated in individuals with higher paranoia (i.e., the belief that others intend harm) and is associated with persecutory delusion severity in SZ patients—the most common delusions in SZ, representing the outermost edge of the paranoia continuum [19]. Predictive processing not only encompasses sensory systems but is also crucial in approximating the consequences of impending motion [20], language [21,22], and other higher-order functions of the brain. Most of the frontal lobe areas likely serve this function of predictive coding in the motor system, a process termed “active inference” [23]. Probably the only exception to this rule is Brodmann’s area 4 (M1, the primary motor cortex), which does not receive prediction error information and is thus agranular in adults (lacks a well-developed layer IV). The likely underlying reason for this is the postnatal recession of layer IV due to the neurodevelopmental acquisition of motor skills and the fact that prediction errors are efficiently handled at the periphery through the spinal reflex arcs [24] and cerebellum.
Thalamocortical projections and corticothalamocortical loops also play a crucial role in downstream predictive processing, as substantiated by clinical neuroimaging research. For instance, in a study comparing resting-state functional magnetic resonance imaging (rs-fMRI) data from 90 SZ patients with controls, bilaterally excessive functional connectivity between the thalamus and sensory and motor areas of the cerebral cortex was identified in SZ. This excessive connectivity level correlated with the severity of the clinical picture and reduced the connectivity of the thalamus with the PFC, striatum, and cerebellum [25].
Comparably to the diagnostic process, therapy primarily targets individual symptoms of SZ rather than the underlying pathophysiological causes. Consequently, treatment relies on a trial-and-error approach. The predominant use of antipsychotics as the primary intervention to alleviate positive symptoms—delusions and hallucinations—often proves inadequate for many patients, especially considering their long-term effects [26]. Moreover, these medications exhibit limited efficacy against negative symptoms, such as social withdrawal and reduced motivation, as well as cognitive impairments like working memory and goal-directed behavior deficits attributed to the dysfunction of the dorsolateral PFC (DLPFC). Additionally, antipsychotics commonly lead to significant adverse effects, including metabolic, neurological (e.g., subtle but significant gray and white matter loss [27]), and cardiovascular issues and complications, affecting long-term adherence to treatment (for a comprehensive contemplation, see [26]).
Many paradigms of SZ pathogenesis and various causative and risk factors (environmental, psychosocial, developmental, genetic, epigenetic, and drug use disorders) converge to either hyperdopaminergic states in mesocortical projections from the ventral tegmental area to the ventral striatum (including nucleus accumbens) and PFC or neuroinflammatory changes. Hyperdopaminergic states, i.e., uncontrolled increases in presynaptic dopamine levels released without appropriate stimulation, are considered as a basis for positive symptoms of the disease [28]. They lead to misattributing importance to neutral sensory stimuli (aberrant salience), and thus individuals with SZ require longer periods to pay attention to and process irrelevant sensory stimuli compared to control subjects (Simon’s effect) [29,30]. Additionally, patients with SZ have notably decreased structural connectivity between the amygdala and MOFC via the uncinate fasciculus, whereas their increased amygdala activity may have a role in distress and the perception of threat related to auditory hallucinations; individuals with SZ also exhibit altered reward prediction and associated striatal and PFC activation, impaired reward learning, and impaired reward-modulated action selection (for a review, see [31]).
Neuroinflammatory changes play a significant role in SZ (for a review, see [32]). For instance, an increased numerical density of microglial cells in the frontal and temporal lobes has been reported in the brains of individuals with chronic SZ [33]. In animal models, chronic restraint, social isolation, and repeated social defeat lead to elevated microglial activity in the PFC and hippocampus. Calcia and collaborators proposed a two-hit hypothesis, suggesting that chronic and sustained microglial stimulation during prenatal/early life, due to interruptions/changes in the brain’s environment, induces an exaggerated microglial response later on. This primes microglial cells to be more sensitive to minor stimuli such as psychosocial stressors during adolescence/early adulthood [34]; see also [35,36]. Additionally, the humoral activation of brain microglia by patrolling monocytes is believed to affect stress-associated brain regions and amplify pro-inflammatory responses through interoceptive humoral pathways involving vascular endothelial IL-1 receptor type-1 signaling [37,38]. In comparison to healthy controls, a recent report indicates significantly elevated NLRP3, P2RX7, IL-1β, and IL-18 gene expression levels in peripheral blood mononuclear cells of SZ patients, implicating systemic inflammatory changes in SZ [39].
Inflammasomes are protein complexes that assemble in the cytosol after the activation of the cytoplasmic nucleotide-binding oligomerization domain (NOD) of the NOD-like leucine-rich repeat-containing receptors (NLRs) in response to various damage-associated signals, pathogens, harmful substances, and metabolic perturbations [8]. The NLR family pyrin domain-containing 1 protein (NLRP1) is a 1429-amino-acid-long protein known to form an inflammasome complex and activate caspase-1 upon degradation of its N-terminal part by the proteasome in neurons. On the other hand, NLRP3 (NLR family pyrin domain-containing 3 protein) is the main NLRP family member in the brain, predominantly expressed in microglia. Thus far, it has been proposed that the activation of the NLRP3 inflammasome might be relevant to the pathogenesis of SZ [39,40,41,42]. A recent in vitro study on human induced pluripotent stem cells has shown a higher activation of the inflammasome in microglia of the SZ patients, which also impacted neuronal functions and led to a higher loss of the synapses [43].
NLRP3 activation in SZ is observed at the periphery [39,42] and can be triggered by various signals, including minute cholesterol crystals in early atherosclerotic lesions [44]. While the extent to which cholesterol crystals form in vivo remains a topic of ongoing investigations, one study has already shown that exposing bone marrow-derived macrophages to cholesterol-rich myelin debris is sufficient to engage NLRP3 inflammasome [45]. Possibly as a result of failed maturation, imaging and postmortem studies have revealed disturbed myelination and oligodendroglia-related processes in patients with SZ, including irregular gene expression and a reduced number of oligodendrocytes in the DLPFC [46]. This finding is of great potential importance as it links oligodendrocytes and myelin pathology in SZ to the activation of the inflammasome in the myeloid lineage, from which microglial cells are also derived. In line with maternal immune activation (MIA) and the activation of inflammasomes, a substantial number of studies have shown a more robust inflammatory response in patients with SZ (e.g., [47], which is also associated with worse clinical outcomes [48]).
Interestingly, while the NLRP3 inflammasome is dominantly expressed in microglia [8,49], a different type of inflammasome, NLRP1, is mostly expressed in the neurons of the central nervous system [50]. Its involvement in Alzheimer’s disease [9,51], cerebral ischemic [52] and reperfusion-ischemic injury [53], mesial temporal lobe epilepsy [54], and multiple sclerosis [55] has been documented earlier. This study aims to compare the expression of the NLRP1 inflammasome in the neurons of healthy control brains and SZ patients to assess whether the NLRP1 inflammasome plays a role in disease pathogenesis. The comparison of housekeeping gene expression offers additional insight into the underlying biological distinctions between SZ and control brains, contributing to a deeper understanding of the molecular basis of SZ.

2. Materials and Methods

2.1. Human Brain Tissue Samples

For this study, we analyzed brain samples of ten subjects with schizophrenia (SZ; six females and four males, mean age 55.9 ± 10.5 years) and ten control (CON) subjects (three females and seven males, mean age 57.8 ± 10.4 years). All SZ patients met the criteria for a diagnosis of SZ based on the Diagnostic and Statistical Manual of Mental Disorders, Fourth Edition, Text Revision [56], and, until their death, were under long-term treatment with either clozapine, olanzapine, or risperidone, with regular clinical follow-ups by at least one experienced psychiatrist. The symptoms described in the medical records of subjects with SZ whose brain samples were analyzed in our study were quite similar and included delusions, hallucinations, disorganized speech and behavior, as well as negative symptoms such as affective flattening, alogia, and avolition, all lasting for a period beyond 6 months from disease onset. Both SZ and CON brains were carefully selected to ensure that samples for analysis were not taken from subjects with a prior history of head trauma or other major neuropsychiatric disorders. CON samples were obtained from individuals comparable in age to the SZ samples, with no psychiatric or neurological illness. They were chosen to match the SZ group in terms of age and postmortem delay. Demographic data for both SZ and CON subjects are provided in Table 1.
The sampling of brain tissue from routine autopsies was conducted with the approval of the Central Ethical Committee of the University of Zagreb Medical School (Case no. 380-59-10106-23-111/93, Class: 641-01/23-02/01, from 11 December 2015). The postmortem samples included the orbitofrontal Brodmann’s areas 11/12 of the medial orbitofrontal cortex (MOFC) and the mid-frontal Brodmann’s area 46 of the dorsolateral prefrontal cortex (DLPFC) from both hemispheres (Figure 1). These samples were selected from the Zagreb Brain Bank Collection at the Croatian Institute for Brain Research in Zagreb, Croatia [57,58,59]. Each dissected sample had an approximate volume of around 0.5 cm3. The caudal segment of each block underwent staining with cresyl violet (Nissl stain). This staining process was instrumental in verifying the cytoarchitectural features within the analyzed Brodmann’s areas. Area 46 is demarcated dorsally by the granular frontal area 9, extending rostroventrally to the frontopolar area 10, and caudally connecting with the triangular area 45 [60]. On the other hand, area 11 is bordered rostrally and laterally by the frontopolar area 10, the orbital area 47, and the triangular area 45. Its caudal aspect is contiguous with the subgenual area 25. On the medial surface, it extends into the rostral area 12 [61,62,63].

2.2. NLRP1 Expression Analysis Using Microarray Procedure

After dissection, samples were immediately stabilized in RNAlater solution (Thermo Fischer Scientific, Waltham, MA, USA) and stored at −80 °C for subsequent analysis, as previously described [64]. Each microarray utilized 20–30 mg of brain tissue from BA46 or BA11/12. RNA isolation followed the protocol using the RNeasy Plus Mini kit from Qiagen (Venlo, The Netherlands). RNA concentration and quality were assessed using Agilent’s Bioanalyzer 2100 and RNA 6000 Nanochip Kit (both from Agilent Technologies, Santa Clara, CA, USA). RNA degradation was evaluated by 28S/18S ribosomal band peak ratios within the acceptable range of 1.5 to 2.0. The RNA integrity number (RIN) of each sample is given in Table 2. Total RNA underwent reverse transcription and was hybridized onto the Affymetrix HG-U133 Plus 2.0 array, employing the same protocol and gene identification names as the GeneChip Human Exon 1.0ST Arrays (Affymetrix, Santa Clara, CA, USA). This array examines over a million exons, spanning 17,868 NCBI Reference Sequence (RefSeq) transcripts. The procedure involved initial ribosomal RNA removal (RiboMinus Human/Mouse Transcriptome Isolation Kit, Invitrogen-Thermo Fischer Scientific, Waltham, MA, USA) to reduce background noise. Subsequently, double-stranded cDNA was amplified to antisense cRNA, purified, reverse-transcribed, fragmented, labeled, and hybridized into arrays. Gene expression values from microarray data (.cel files) were analyzed using Partek Genomic Suite software ver. 6.5 (Partek Incorporated, St. Louis, MO, USA). Data preparation included background noise correction, log2 transformation, quantile normalization, averaging probe signals by mean, filtering for a fold change of less than 1.5, and a p-value under 0.05 [65]. The entirety of the microarray transcriptome data are collected in our prior study [64]. As they contain all transcripts, including mRNA of the NLRP1 gene listed in the dataset line 18,408 (of the Affymetrix EXON HuEx-1.0-st-v2 chip), we are also attaching them here as Supplementary Table S1 for reference. To find the ID of a gene of interest, one should utilize the conversion tool of the Database for Annotation, Visualization, and Integrated Discovery (DAVID, https://david.ncifcrf.gov/, accessed on 22 June 2023) [66]. The mRNA of the NLRP1 gene corresponds to Entrez Gene ID 3742783 (Homo sapiens, NLR family pyrin domain containing 1, NCBI Reference Sequence: NM_033004.4). The values in dataset line 18,408, representing log2-transformed NLRP1 mRNA signal intensities across all 48 samples, can be transformed back to expression levels of the NLRP1 gene using the reverse procedure from logarithmic to exponential. For instance, a signal intensity of 3.55373 translates to an expression level of 23.55373, which in turn is 11.74301, and so forth (see Table 2 for NLRP1 expression levels across all samples).

2.3. Immunohistochemical Staining

Brain tissue samples were dissected from the MOFC, embedded in paraffin, and cut into 12 μm-thin slices for further immunohistochemical staining. Tissue sections were deparaffinized in xylene and rehydrated in the decreasing concentrations of ethanol (100%–twice, 96%, and 70%). Antigen retrieval was performed in a boiling citrate buffer (anhydrous citric acid solution 10 mM, pH 6), five times shortly (around 1 min) at high microwave power (700 W) and 20 min at low microwave power (300 W). Endogenous peroxidase activity was inhibited by incubating slides in 0.02% H2O2 in methanol (150 mL methanol and 50 mL water) for 30 min. Unspecific signals were blocked with 5% bovine serum albumin (BSA) + 0.5% Triton/PBS for 1 h at RT. The primary antibody (NLRP1, Abcam, Cambridge, UK, AB_776633) was diluted in blocking solution to working concentration (NLRP1 1:100). After overnight incubation with the primary antibody in a humidified chamber at 4 °C, slides were incubated with the goat antirabbit biotinylated secondary antibody (1:200) for 60 min (Vector Laboratories, Newark, CA, USA, AB_2336810, AB_2336811), followed by the application of the ABC complex also for 60 min at RT (Vector Laboratories, Newark, CA, USA, AB_2336810, AB_2336811). 3,3′-diaminobenzidine (Sigma-Aldrich, St. Louis, MO, USA, cat. #D0426) was used as a chromogen for developing peroxidase activity. Negative-control sections were not incubated in the primary antibodies. Sections were dehydrated before mounting in Histomount (Poly-Mount, Polysciences, Warrington, PA, USA, Cat. #08381-120).

2.4. Quantification of NLRP-1-Immunoreactive Pyramidal Neurons

Stained sections were scanned by the Hamamatsu Nanozoomer 2.0. RS in 0.45 µm × 0.45 µm resolution (Hamamatsu Photonics K.K., Hamamatsu City, Japan) and analyzed in NDP.view2 program for digital visualization. Each NLRP1-positive pyramidal neuron was counted as one, and the results were presented as the number of immunoreactive cells per cortical area (in square millimeters) of interest (from the top of layer I to the bottom of layer VI). Cortical layers I-VI were identified based on the size, shape, and distribution of pyramidal cells, and the laminar attribution of labeled cells was determined by measuring the thickness of layers on adjacent Nissl-stained sections. Quantification was performed by D.V., who was blind to the group and the identity of the cases. Section analysis and images were obtained with an Olympus BX53 microscope (Olympus, Tokyo, Japan). The total number of all NLRP1-immunoreactive pyramidal neurons was determined in a selected section with the best staining, assessed using Image J software (National Institute of Health, Bethesda, MD, USA, https://imagej.nih.gov/ij/, accessed on 22 June 2023). The results were presented as the average density of NLRP1-immunoreactive neurons per analyzed area.

2.5. Statistical Analysis

Comparative analysis between SZ and CON groups regarding NLRP1 transcript expression involved employing Student’s t-test. A p-value below the pre-specified α level of 0.05 indicated a statistical difference from the null hypothetical value (p < 0.05). To specifically evaluate if a given dataset followed a normal distribution, we tested using the Shapiro–Wilk test, as it is considered the most powerful one for small-sized samples. Due to a small sample size, differences in the number of NLRP1-positive pyramidal cells were analyzed with an approximation of normal distribution by both a parametric two-sample, two-tailed t-test and a non-parametric two-sample, two-tailed Mann–Whitney U-test (Wilcoxon rank-sum test). All statistical tests and graphs were made in GraphPad Prism version 9.3.1. (GraphPad Software, San Diego, CA, USA).

3. Results

3.1. Expression of NLRP1 mRNA in the DLPFC and MOFC

The results of NLRP1 mRNA expression in the DLPFC and MOFC are summarized in Table 2. Due to a relatively small sample size, we conducted both a parametric t-test and a non-parametric Mann–Whitney test. The results indicated that in the DLPFC and MOFC from both hemispheres of six SZ subjects, the NLRP1 mRNA expression was significantly higher than in six control brains (t-test and Mann–Whitney test p-values were lower than 0.05).

3.2. Immunohistochemical Analysis of the NLRP1 Protein Distribution in the MOFC

Considering that the most substantial mean difference in NLRP1 mRNA expression between control and SZ subjects was found in tissue blocks from the MOFC of the right hemisphere (15.53 in SZ vs. 12.99 in controls, see Table 2), we proceeded with an immunohistochemical analysis on these specific blocks. We chose the best stained, representative MOFC section from each of the 12 blocks (5 from controls and 7 from SZ subjects) and quantified the number of NLRP1-positive pyramidal neurons in layers III, V, and VI. The results of the quantification are presented in Table 3 and Figure 2. They reveal that, in the MOFC of the right hemisphere of seven SZ subjects, NLRP1 protein immunoreactivity was detected in a significantly greater number of pyramidal neurons than in five control brains (both t-test and Mann–Whitney test p-values were lower than 0.01).
NLRP1 protein expression in the MOFC of one selected control sample (CON9) is shown in Figure 3. For comparison, a selected sample of NLRP1 protein expression in the MOFC of one SZ case is presented in Figure 4.

3.3. Expression of Housekeeping Genes

There is no ideal reference housekeeping gene consistently expressed in all experimental conditions, regardless of the tissue type and disease state [67]. Most of the known housekeeping genes we examined, such as B2M, PGK1, PPIA, RLP0, SDHA, TFRC, YWHAZ, and others, exhibited similar expression levels in both our SZ and control samples. However, a smaller subset of recognized housekeeping genes (ACTB, CYY1, EIF4A2, HPRT1, and HBMS) showed significantly lower expression, while TBP and GUSB exhibited higher expression in the SZ samples (Table 4). These findings imply underlying biological distinctions between the two groups that extend beyond the genes potentially associated with SZ. These differences may arise from various other factors present in SZ subjects. Among the most prominent of these factors are inflammation, cellular stress, and alterations in cellular metabolism.

4. Discussion

We found that NLRP1 mRNA expression is substantially higher in the DLPFC and MOFC of six SZ subjects compared to six controls. The immunohistochemical analysis of NLRP1 immunoreactivity in the MOFC from the right hemisphere of seven SZ subjects showed significantly higher numbers of NLRP1-expressing pyramidal cells in layers III, V, and VI compared to five controls, thus supporting our initial observation and suggesting NLRP1 inflammasome activation in SZ subjects. Unlike control samples where only a lower number of layer III pyramidal cells were positive, SZ samples had, on average, a higher number of layers III, V, and VI NLRP1-immunoreactive pyramidal cells. These results are novel, as previous methods and studies did not allow this kind of analysis or have not included the analysis of the NLRP1 inflammasome. Layer III pyramidal neuron dysfunction in SZ aligns with working memory deficits, while impairments of pyramidal neurons in layer V and particularly layer VI are consistent with the disruption of predictive processing. Layer IV was devoid of changes, showing very few, if any, NLRP1-positive cells.
Inside the brain, the majority of immune actions are conducted by microglia, as they control innate immune responses and survey the brain parenchyma [68]. During brain development, microglia play essential roles by regulating cell and synapse numbers, eliminating excess immature synaptic connections, and producing mediators whose profiles vary based on the microglia’s activation state, consequently influencing different neurodevelopmental processes [69,70,71]. The mechanisms to explain the involvement of immune system, including microglia, in the pathogenesis of SZ include changes in the developing fetal brain due to prenatal infection and MIA [35,40,72,73,74,75]. For a recent, comprehensive overview of MIA, refer to [76], and for a potential mechanistic framework toward a deeper understanding of SZ immunophenotype, see [77]. As genes overexpressed in SZ brains were significantly enriched among MIA-induced differentially methylated genes in the fetal brain in a cell-type-specific manner (upregulated genes in layer V pyramidal neurons were highly significantly enriched among hypomethylated genes on gestational days 9 and 17), it is believed that MIA-driven methylation changes during gestation may influence SZ gene expression signatures in the adult brain [78]. MIA can also impact the epigenetic patterns of the genes involved in SZ onset [78]. Indeed, it has been shown that inflammation during early development can change the methylation of some genes and alter their expression in adulthood [78,79,80]. One way of activating inflammatory responses is the activation of the inflammasomes. A recent study has shown that MIA can cause SZ-like behavior in rats due to the activation of Toll-like receptors (TLR) and inflammasomes [40].
Our study indicates significant overexpression, i.e., an activation of the NLRP1 inflammasome in cells of the frontal lobe of individuals with SZ compared to controls. The overexpression of the NLRP1 inflammasome was most pronounced in pyramidal neurons in SZ brains. This led us to conclude that the activation of the “neuronal” NLRP1 inflammasome likely precedes selective atrophy, particularly decreased dendritic arborization and profound dendritic spine loss visualized using the Golgi stain, in deep layer III pyramidal neurons in the PFC in SZ. We hypothesize that this occurs after initial insults during the perinatal or early postnatal period. These initial changes, also known as “the first hit”, might lead to a vulnerability of the cortex to stressors during adolescence and early adulthood, eventually triggering dysfunctional immune pathways and apoptosis [81].
It remains to be examined whether the NLRP1 inflammasome is activated similarly to the NLRP3 inflammasome (as described by [82]). The NLRP3 inflammasome, composed of NLRP3, ASC (apoptosis-associated speck-like protein containing a caspase-activation and recruitment domain), and caspase-1, assembles inside microglia upon activation. This leads to the activation of the NF-κB signaling pathway, increased cleavage and activity of caspase-1, as well as downstream IL-1β and IL-18 release, activation of caspase-6, and cleavage of gasdermin D. To test the hypothesis of early NLRP1 inflammasome activation in SZ, a comprehensive analysis will be required, including appropriate model systems. Since an inflammasome can be activated and reactivated in neighboring cells due to extracellular ASC heteromer uptake, this creates a vicious cycle by promoting discontinued but constant microglial activation and severe inflammation that can spread within affected areas [8,83,84]. This may be one of the reasons why SZ might have a non-linear course characterized by a continuum of typical remissions and exacerbations in many patients.
Deficits in predictive processing are considered the key pathophysiological disturbance in SZ [16,17]. In individuals with SZ, the disruption of predictive processing impacts phenomenal consciousness (also called access consciousness or C0), giving rise to positive symptoms [85,86]. It also significantly affects self-reflection [87] and the experience of one’s body and the sense of “ownership” over it [88], thus undermining the metacognition and self-monitoring of their lived awareness (C1 consciousness), which in healthy individuals gives a subjective sense of error (reality check or C2 consciousness) [89]. Prediction errors update the current understanding of a situation through a search for meaning, with this learning signal strongly modulated by dopamine [90]. If, for any reason, the comparison of reality with internal representations performed through predictive processing by the PFC does not align (resulting in a reality check failure), symptoms of psychotic episodes or disorders may emerge—often within the SZ spectrum. However, a single psychotic episode or disorder can also be triggered, for example, by methamphetamines. Methamphetamines act as elicitors of paranoia in humans, a state associated with a stronger prior on volatility and elevated sensitivity to perceived changes in the task environment. Methamphetamine exposure in rats recapitulates this impaired, uncertainty-driven belief updating and rigid anticipation of a volatile environment [91]. In a mouse model, both chronic and acute methamphetamine treatment upregulated the expression of genes related to dopamine and serotonin metabolism in the striatum and PFC, suggesting a potential mechanism for how methamphetamine elicits an individual’s psychosis risk [92].
Long-term and heavy use of methamphetamine can increase the risk of developing psychotic symptoms, including hallucinations and delusions, which may resemble symptoms of SZ. Intriguingly, both NLRP1 and NLRP3 inflammasome overexpression, along with an increased induction of apoptosis and inflammation, were documented in the hippocampus by Western blotting, immunohistochemistry, and the terminal deoxynucleotidyl transferase dUTP nick end-labeling (TUNEL) method in eleven patients with methamphetamine use disorder [93]. Individuals with SZ also have an increased predilection for addiction, worsened by a loss of top-down control, contributing to more pronounced habitual tendencies and compulsive drug-seeking [94]. Compared to the general population, individuals with SZ are 4.6 times more likely to have a substance abuse diagnosis [95]. Here, it should be noted that this kind of cognitive control is distinct from general (fluid) intelligence, working memory, cognitive flexibility, planning, shifting attention, organizing thoughts, and problem-solving, mediated by networks of the DLPFC [96], and is more closely related to response inhibition and decision-making mediated by networks of the OFC. The DLPFC is especially hypoactive in persons with chronic SZ due to the selective atrophy of the deep layer III pyramidal neurons (for a review, see [81]). The transcranial direct current stimulation (tDCS) of the DLPFC has been shown to enhance working memory and suppress pathological γ power elevations in SZ subjects [97]. The MOFC integrates social and emotional information and its activity is concerned with monitoring, learning, and predicting the likely outcomes of actions related to the reward value of reinforcing stimuli, thus contributing to decision-making processes, especially subjective rewards [98], with more complex or abstract rewards such as monetary gain being represented more anteriorly than less complex ones such as taste, whereas lateral OFC activity is associated with negative reinforcing stimuli [99]. MOFC is considered a key component of the default mode network involved in self-referential thinking and understanding others’ thoughts, beliefs, intentions, and emotions (“theory of mind” abilities) [100,101].
The self-domestication theory suggests that humans have undergone a process of self-selection for traits such as reduced aggression, increased social tolerance, and cooperation [102,103]. Therefore, certain evolutionary pressures favored individuals who could live and work together more harmoniously, leading to the development of traits that are often associated with domesticated animals. One of three major hypotheses for self-domestication as an evolutionary root for SZ [102] considers the neoteny of synaptic spines of the association pyramidal neurons in the PFC [104]. In essence, the excessive generation and developmental remodeling of synaptic spines continue after adolescence under stronger dopaminergic innervation, possibly due to the human-specific expression of TH (tyrosine hydroxylase) gene in a subset of inhibitory neurons in the DLPFC [105] in the second half of the second and the third decade of life before complete stabilization in adult values. This has probably given humans unprecedented opportunity to reach the highest levels of intrinsic motivation and cognitive abilities, while burdening them with increased susceptibility to the development of abnormal neural circuits in adolescence and post-adolescence, manifested in neuropsychiatric disorders such as SZ.
Years of study have emphasized the significance of the optimal activation of cortical dopamine receptors in governing cognition associated with the PFC in humans [106,107]. The expansion of the neocortex in primate evolution has been paralleled by increased innervation by dopamine [108]. An analysis of axon length density to neuron density among species by Raghanti and collaborators revealed that humans and chimpanzees together deviated from macaques in having increased dopaminergic afferents in layers III and V/VI in the PFC [106]. Both the pyramidal and non-pyramidal cells of PFC, which include DLPFC and MOFC, express the D1-like (D1R and D5R) and D2-like (D2R, D3R, and D4 R) families of dopamine receptors, indicating fine regulation that is disturbed in SZ [109,110]. Individuals with SZ often face challenges in distinguishing between their own thoughts and external stimuli, leading to false beliefs or perceptions that are not in line with reality (delusions) and hallucinations (positive symptoms of SZ). These symptoms are associated with the increased dopamine activity of the mesocortical pathway through the nucleus accumbens, augmenting D2-like receptor activation. This effect is blocked by antipsychotics [111]. Besides cognitive deficits, other negative symptoms such as reduced motivation, reduced goal-directed behavior, anhedonia, thought disorder, poverty of speech (alogia), and social withdrawal have been linked to DLPFC dysfunction due to reduced D1R activation [111]. Since cognitive and negative symptoms of SZ are less directly associated with the dysfunction of D2-like receptors, they are more difficult to treat with antipsychotics, especially because some patients may lack insight into the extent and impact of their symptoms [112,113].

5. Limitations of the Study and Conclusions

These initial results showed significantly increased NLRP1 inflammasome activation in the dorsolateral prefrontal and medial orbitofrontal cortex in SZ compared to control brains, which will require detailed analyses of NLRP1 and other inflammasome-related proteins in additional brain regions. Follow-up on new information can be found at: https://www.proteinatlas.org/ENSG00000091592-NLRP1/brain (accessed on 22 February 2024). The primary limitations of this study include a small sample size for NLRP1 expression analysis and immunohistochemical analysis performed only in MOFC brain tissue for the right hemisphere. While elevated levels of NLRP1 mRNA and protein expression can indicate activated inflammasomes, it does not automatically confirm their activation, and it is not the only indicator. NLRP1 is indeed a crucial component of the neuronal inflammasome, but other factors, such as the presence of activating stimuli, the availability of accessory inflammasome components, and the overall cell state, can also play a role in inflammasome activation. That being said, the increase in NLRP1 mRNA and NLRP1 protein expression may indicate a cellular response to various potential threats or stressors. Although inflammasomes are typically activated by certain bacteria, viruses, or pathogens, they can also respond to other stimuli like stress, trauma, or certain drugs. Some published hypotheses regarding the activation of the NLRP1 inflammasome include its activation by various viral proteases [114], possibly during intrauterine development in the case of SZ. Predisposition by certain NLRP1 gene polymorphisms [115] and the prior activation of the NLRP3 inflammasome in microglia, e.g., by extracellular amyloid [82] or templated tau seeds [116], are also suggested, with conceivable spreading by ASC proteins (“specks”) and possibly intracellular amyloid or other intracellularly generated amyloid precursor protein-generated metabolites and other harmful molecules.
Another challenge in interpreting our results is that inflammasome activation involves several steps beyond increased NLRP1 expression. These steps include its assembly into the inflammasome complex, the recruitment of other proteins, and the subsequent cleavage and activation of proinflammatory cytokines. When an inflammasome is activated, it cleaves pro-inflammatory cytokines, such as IL-1β or IL-18, from their inactive precursor forms. These cytokines then can act on a variety of cells to promote the inflammatory response. It should be kept in mind that our analysis is limited to the dorsolateral prefrontal cortex and the medial orbital cortex. Therefore, confirming inflammasome activation requires further investigations, such as assessing the formation of the NLRP1 inflammasome complex and measuring caspase-1 activation and cytokine release. Upstream and downstream alterations due to elevated NLRP1 and associated signaling pathways should also be investigated, as well as inflammatory responses and changes related to neuroinflammation, synaptic dysfunction triggered by inflammation, and neuronal pyroptosis. Despite the multi-faceted nature of inflammasomes, the heterogeneity of pathophysiological alterations, and the consequent variability in clinical presentation, along with all the mentioned limitations of this preliminary research, our results, including the differences in the housekeeping gene expression between the SZ and CON groups, strongly suggest that it is worthwhile to further investigate the role and significance of inflammasome activation in SZ.
Ultimately, all comparative evaluation studies of efficacy and safety to date have supported the use of anti-inflammatory adjuvant therapy over antipsychotics alone. However, despite the recognition of inflammation in individuals affected with SZ, this important discovery and overall significant positive effects of various anti-inflammatory agents (such as acetylsalicylic acid, celecoxib, omega-3 fatty acids, estrogen, selective estrogen receptor modulator, pregnenolone, N-acetylcysteine, minocycline, davunetide, and erythropoietin) in reducing total, positive, and negative symptoms scores in the PANSS, as well as significant cognitive improvements with minocycline and pregnenolone augmentation therapy without significant differences in side effects compared with placebo (for a review, see [117]), have not yet resulted in expected new treatments. The limited success of clinical trials using anti-inflammatory drugs likely stems from the inability to pinpoint the specific inflammatory mechanisms targeted by existing medications. Perhaps, not all patients show initial inflammation. In other words, certain individuals are more prone to heightened inflammation and are also responsive to such treatment (for a review, see [32]). This might also be part of the explanation as to how, despite the substantial heritability of SZ (h2 = 65–79%) [118], the identified risk variants collectively contribute to only a very limited portion (h2SNP = 24%) of the total variability in the susceptibility of the phenotype [119]. In fact, in a longitudinal study on 84 patients with a clinical diagnosis of schizophrenic disorders (ICD-10 “F2x.x”), a multi-layer neural network model based on the backpropagation supervised learning algorithm identified a subgroup of 22.5% of patients with SZ who exhibited a significant correlation between global SZ scores and immunoglobulin M (IgM) levels, along with a correct prediction of the response to therapy in 94.4% of them [120]. Once again, non-steroidal and anti-inflammatory drugs, including acetylsalicylic acid, and COX-2 inhibitors showed significant positive effects as adjunctive treatments in SZ.
In conclusion, further research is needed to determine the neuroinflammation profile in individuals with SZ, involving comprehensive data collection, the molecular analysis of peripheral and central inflammatory biomarkers, and the consideration of the role of the NLRP1 inflammasome as a potential prognostic biomarker and therapeutic target, with a focus on patients with homogeneous clinical profiles for personalized insights into anti-inflammatory treatment effects. Additional specificity in future studies could be made by performing double labeling analyses using specific markers of pyramidal neurons, such as RBP4 and EMX1.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/biom14030302/s1, Table S1: Gene Expression Raw Data.xls.

Author Contributions

Conceptualization, G.Š.; methodology, E.Š.P., D.V., L.L.H., M.M. and G.Š.; software, E.Š.P., M.M. and G.Š.; validation, P.R.H. and G.Š.; formal analysis, E.Š.P., D.V., K.P., M.M. and G.Š.; investigation, E.Š.P., D.V., M.M. and G.Š.; resources, G.Š.; data curation, E.Š.P. and G.Š.; writing—original draft preparation, G.Š.; writing—review and editing, E.Š.P., D.V., H.R.F., L.L.H., D.M. (Davor Mayer), J.K., K.P., M.B.L., M.P., Ž.K., D.M. (Darko Marčinko), M.Š., P.R.H., M.M. and G.Š.; visualization, E.Š.P. and G.Š.; supervision, G.Š.; project administration, E.Š.P., L.L.H. and G.Š.; funding acquisition, G.Š. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Croatian Science Foundation, grant IP-2019-04-3584 (to G.Š.), and by the Scientific Center of Excellence for Basic, Clinical, and Translational Neuroscience CORE-NEURO (GA KK01.1.01.0007) funded by the European Union through the European Regional Development Fund.

Institutional Review Board Statement

Sampling of the brain tissue from routine autopsies was performed with the approval of the Central Ethical committee of the University of Zagreb Medical School (Case no. 380-59-10106-23-111/93, Class: 641-01/23-02/01, from 11 December 2015).

Informed Consent Statement

The informed consent was waived by the ethical committee.

Data Availability Statement

Data are contained within the article and Supplementary Table S1. All data generated or analyzed during this study are available from the corresponding author upon reasonable request.

Conflicts of Interest

The authors declare no conflicts 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. Orsolini, L.; Pompili, S.; Volpe, U. Schizophrenia: A Narrative Review of Etiopathogenetic, Diagnostic and Treatment Aspects. J. Clin. Med. 2022, 11, 5040. [Google Scholar] [CrossRef] [PubMed]
  2. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders, 5th ed.; Text Revision; American Psychiatric Association Publishing: Washington, DC, USA, 2022. [Google Scholar]
  3. World Health Organization. International Classification of Diseases, 11th ed.; WHO: Geneva, Switzerland, 2022. [Google Scholar]
  4. Valle, R. Schizophrenia in ICD-11: Comparison of ICD-10 and DSM-5. Rev. Psiquiatr. Salud. Ment. 2020, 13, 95–104. [Google Scholar] [CrossRef] [PubMed]
  5. Stein, D.J.; Shoptaw, S.J.; Vigo, D.V.; Lund, C.; Cuijpers, P.; Bantjes, J.; Sartorius, N.; Maj, M. Psychiatric Diagnosis and Treatment in the 21st Century: Paradigm Shifts versus Incremental Integration. World Psychiatry 2022, 21, 393–414. [Google Scholar] [CrossRef] [PubMed]
  6. Goldsmith, D.R.; Crooks, C.L.; Walker, E.F.; Cotes, R.O. An Update on Promising Biomarkers in Schizophrenia. Focus (Am. Psychiatr. Publ.) 2018, 16, 153–163. [Google Scholar] [CrossRef]
  7. Gao, W.-J.; Yang, S.-S.; Mack, N.R.; Chamberlin, L.A. Aberrant Maturation and Connectivity of Prefrontal Cortex in Schizophrenia—Contribution of NMDA Receptor Development and Hypofunction. Mol. Psychatry 2022, 27, 731–743. [Google Scholar] [CrossRef]
  8. Španić, E.; Langer Horvat, L.; Hof, P.R.; Šimić, G. Role of Microglial Cells in Alzheimer’s Disease Tau Propagation. Front. Neurosci. 2019, 11, 271. [Google Scholar] [CrossRef]
  9. Španić, E.; Langer Horvat, L.; Ilić, K.; Hof, P.R.; Šimić, G. NLRP1 Inflammasome Activation in the Hippocampal Formation in Alzheimer’s disease: Correlation with Neuropathological Changes and Unbiasedly Estimated Neuronal Loss. Cells 2022, 11, 2223. [Google Scholar] [CrossRef]
  10. Španić Popovački, E.; Babić Leko, M.; Langer Horvat, L.; Brgić, K.; Vogrinc, Ž.; Boban, M.; Borovečki, F.; Šimić, G. Soluble TREM2 Concentrations in the Cerebrospinal Fluid Correlate with the Severity of Neurofibrillary Degeneration, Cognitive Impairment, and Inflammasome Activation in Alzheimer’s Disease. Neurol. Int. 2023, 15, 842–856. [Google Scholar] [CrossRef]
  11. Shen, Y.; Qian, L.; Luo, H.; Li, X.; Ruan, Y.; Fan, R.; Si, Z.; Chen, Y.; Li, L.; Liu, Y. The Significance of NLRP1 Inflammasome in Neuropsychiatric Disorders. Brain Sci. 2022, 12, 1057. [Google Scholar] [CrossRef]
  12. Coyle, J.T.; Tsai, G.; Goff, D. Converging Evidence of NMDA Receptor Hypofunction in the Pathophysiology of Schizophrenia. Ann. N. Y. Acad. Sci. 2003, 1003, 318–327. [Google Scholar] [CrossRef]
  13. Nakazawa, K.; Sapkota, K. The Origin of NMDA Receptor Hypofunction in Schizophrenia. Pharmacol. Ther. 2020, 205, 107426. [Google Scholar] [CrossRef] [PubMed]
  14. Li, P.; Snyder, G.L.; Vanover, K.E. Dopamine Targeting Drugs for the Treatment of Schizophrenia: Past, Present and Future. Curr. Top. Med. Chem. 2016, 16, 3385–3403. [Google Scholar] [CrossRef] [PubMed]
  15. Millgate, E.; Hide, O.; Lawrie, S.M.; Murray, R.M.; MacCabe, J.H.; Kravariti, E. Neuropsychological Differences between Treatment-Resistant and Treatment-Responsive Schizophrenia: A Meta-analysis. Psychol. Med. 2022, 52, 1–13. [Google Scholar] [CrossRef] [PubMed]
  16. Griffin, J.D.; Fletcher, P.C. Predictive Processing, Source Monitoring, and Psychosis. Annu. Rev. Clin. Psychol. 2017, 13, 265–289. [Google Scholar] [CrossRef] [PubMed]
  17. Scheliga, S.; Schwank, R.; Scholle, R.; Habel, U.; Kellermann, T. A Neural Mechanism Underlying Predictive Visual Motion Processing in Patients with Schizophrenia. Psychiatry Res. 2022, 318, 114934. [Google Scholar] [CrossRef] [PubMed]
  18. Bastos, A.M.; Lundquist, M.; Waite, A.S.; Kopell, N.; Miller, E.K. Layer and Rhythm Specificity for Predictive Routing. Proc. Natl. Acad. Sci. USA 2020, 117, 31459–31469. [Google Scholar] [CrossRef]
  19. Sheffield, J.M.; Suthaharan, P.; Laptourgos, P.; Corlett, P.R. Brief Updating and Paranoia in Individuals with Schizophrenia. Biol. Psychatry Cogn. Neurosci. Neuroimaging 2022, 7, 1149–1157. [Google Scholar]
  20. Llinás, R.R.; Roy, S. The ‘Prediction Imperative’ as the Basis for Self-awareness. Phil. Trans. R. Soc. B 2009, 364, 1301–1307. [Google Scholar] [CrossRef]
  21. Cope, T.E.; Sohoglu, E.; Sedley, W.; Patterson, K.; Jones, P.S.; Wiggins, J.; Dawson, C.; Grube, M.; Carlyon, R.P.; Griffiths, T.D.; et al. Evidence for Causal Top-down Frontal Contributions to Predictive Processes in Speech Perception. Nat. Commun. 2017, 8, 2154. [Google Scholar] [CrossRef]
  22. Brouillet, D.; Friston, K. Relative Fluency (Unfelt vs. Felt) in Active Inference. Conscious. Cogn. 2023, 115, 103579. [Google Scholar] [CrossRef]
  23. Alexander, W.H.; Brown, J.W. Frontal Cortex Function as Derived from Hierarchical Predictive Coding. Sci. Rep. 2018, 8, 3843. [Google Scholar] [CrossRef]
  24. Shipp, S.; Adams, R.A.; Friston, K.J. Reflections on Agranular Architecture: Predictive Coding in the Motor Cortex. Trends Neurosci. 2013, 36, 706–716. [Google Scholar] [CrossRef]
  25. Antičević, A.; Cole, M.W.; Repovs, G.; Murray, J.D.; Brumbaugh, M.S.; Winkler, A.M.; Savić, A.; Krystal, J.H.; Pearlson, G.D.; Glahn, D.C. Characterizing Thalamocortical Disturbances in Schizophrenia and Bipolar Illness. Cereb. Cortex 2014, 24, 3116–3130. [Google Scholar] [CrossRef]
  26. Davidson, M. The Debate Regarding Maintenance Treatment with Antipsychotic Drugs in Schizophrenia. Dialogues Clin. Neurosci. 2018, 20, 215–221. [Google Scholar] [CrossRef]
  27. Ho, B.-C.; Andreasen, N.C.; Ziebell, S.; Pierson, R.; Magnotta, V. Long-term Antipsychotic Treatment and Brain Volumes: A Longitudinal Study of First-episode Schizophrenia. Arch. Gen. Psychiatry 2011, 68, 128–137. [Google Scholar] [CrossRef]
  28. Howes, O.D.; Kapur, S. The Dopamine Hypothesis of Schizophrenia: Version III—The Final Common Pathway. Schizophr. Bull. 2009, 35, 549–562. [Google Scholar] [CrossRef]
  29. Simon, J.R. Reactions toward the Source of Stimulation. J. Exp. Psychol. 1969, 81, 174–176. [Google Scholar] [CrossRef]
  30. Berkovitch, L.; Dehaene, S.; Gaillard, R. Disruption of Conscious Access in Schizophrenia. Trends. Cogn. Sci. 2017, 21, 878–892. [Google Scholar] [CrossRef] [PubMed]
  31. Šimić, G.; Tkalčić, M.; Vukić, V.; Mulc, D.; Španić, E.; Šagud, M.; Olucha-Bordonau, F.E.; Vukšić, M.; Hof, P.R. Understanding Emotions: Origins and Roles of the Amygdala. Biomolecules 2021, 11, 823. [Google Scholar] [CrossRef]
  32. Murphy, C.E.; Walker, A.K.; Weickert, C.S. Neuroinflammation in Schizophrenia: The Role of Nuclear Factor Kappa, B. Transl. Psychiatry 2021, 11, 528. [Google Scholar] [CrossRef]
  33. Garey, L. When Cortical Development Goes Wrong: Schizophrenia as a Neurodevelopmental Disease of Microcircuits. J. Anat. 2010, 217, 324–333. [Google Scholar] [CrossRef]
  34. Calcia, M.A.; Bonsall, D.R.; Bloomfield, P.S.; Selvaraj, S.; Barichello, T.; Howes, O.D. Stress and Neuroinflammation: A Systematic Review of the Effects of Stress on Microglia and the Implications for Mental Illness. Psychopharmacology 2016, 233, 1637–1650. [Google Scholar] [CrossRef]
  35. Choudhury, Z.; Lennox, B. Maternal Immune Activation and Schizophrenia—Evidence for an Immune Priming Disorder. Front. Psychiatry 2021, 12, 585742. [Google Scholar] [CrossRef]
  36. Rahimian, R.; Wakid, M.; O’Leary, L.A.; Mechawar, N. The Emerging Tale of Microglia in Psychiatric Disorders. Neurosci. Biobehav. Rev. 2021, 131, 1–29. [Google Scholar] [CrossRef]
  37. Wohleb, E.S.; Delpech, J.C. Dynamic Cross-Talk between Microglia and Peripheral Monocytes Underlies Stress-Induced Neuroinflammation and Behavioral Consequences. Prog. Neuropsychopharmacol. Biol. Psychiatry 2017, 79, 40–48. [Google Scholar] [CrossRef]
  38. Savitz, J.; Harrison, N.A. Interoception and Inflammation in Psychiatric Disorders. Biol. Psychiatry Cogn. Neurosci. Neuroimaging 2018, 3, 514–524. [Google Scholar] [CrossRef]
  39. Unal, G.O.; Ozturk, K.H.; Inci, H.E. Increased NLRP3 Inflammasome Expression in Peripheral Blood Mononuclear Cells of Patients with Schizophrenia: A Case-Control Study. Int. J. Psychiatry Clin. Pract. 2023, 27, 111–117. [Google Scholar] [CrossRef]
  40. Talukdar, P.M.; Abdul, F.; Maes, M.; Berk, M.; Venkatasubramanian, G.; Kutty, B.M.; Debnath, M. A Proof-of-Concept Study of Maternal Immune Activation Mediated Induction of Toll-like Receptor (TLR) and Inflammasome Pathways Leading to Neuroprogressive Changes and Schizophrenia-like Behaviors in Offspring. Eur. Neurophychopharmacol. 2021, 52, 48–61. [Google Scholar] [CrossRef]
  41. Ventura, L.; Freiberger, V.; Thiesen, V.B.; Dias, P.; Dutra, M.L.; Silva, B.B.; Schlindwein, A.D.; Comim, C.M. Involvement of NLRP3 Inflammasome in Schizophrenia-like Behavior in Young Animals after Maternal Immune Activation. Acta Neuropsychiatr. 2020, 32, 321–327. [Google Scholar] [CrossRef]
  42. Szabo, A.; O’Connell, K.S.; Ueland, T.; Sheikh, M.A.; Agartz, I.; Andreou, D.; Aukrust, P.; Boye, B.; Bøen, E.; Drange, O.K.; et al. Increased Circulating IL-18 levels in severe mental disorders indicate systemic inflammasome activation. Brain Behav Immun. 2022, 99, 299–306. [Google Scholar] [CrossRef]
  43. Breitmeyer, R.; Vogel, S.; Heider, J.; Hartmann, S.-M.; Wüst, R.; Keller, A.-L.; Binner, A.; Fitzgerald, J.C.; Fallgatter, A.J.; Volkmer, H. Regulation of Synaptic Connectivity in Schizophrenia Spectrum by Mutual Neuron-Microglia Interaction. Commun. Biol. 2023, 6, 472. [Google Scholar] [CrossRef]
  44. Stowig, T.; Henao-Mejia, J.; Elinav, E.; Flavell, R. Inflammasomes in Health and Disease. Nature 2012, 481, 278–286. [Google Scholar] [CrossRef]
  45. Cantuti-Castelvetri, L.; Fitzner, D.; Bosch-Queralt, M.; Weil, M.-T.; Su, M.; Sen, P.; Ruhwedel, T.; Mitkovski, M.; Trendelenburg, G.; Lütjohann, D.; et al. Defective Cholesterol Clearance Limits Remyelination in the Aged Central Nervous System. Science 2018, 359, 684–688. [Google Scholar] [CrossRef] [PubMed]
  46. Raabe, F.J.; Slapakova, L.; Rossner, M.J.; Cantuti-Castelvetri, L.; Simons, M.; Falkai, P.G.; Schmitt, A. Oligodendrocytes as a New Therapeutic Target in Schizophrenia: From Histopathological Findings to Neuron-Oligodendrocyte Interaction. Cells 2019, 8, 1496. [Google Scholar] [CrossRef] [PubMed]
  47. Khandaker, G.M.; Cousins, L.; Deakin, J.; Lennox, B.R.; Yolken, R.; Jones, P.B. Inflammation and Immunity in Schizophrenia: Implications for Pathophysiology and Treatment. Lancet Psychiatry 2015, 2, 258–270. [Google Scholar] [CrossRef] [PubMed]
  48. Ormerod, M.B.E.G.; Ueland, T.; Werner, M.C.F.; Hjell, F.; Rødevand, L.; Sæther, L.S.; Lunding, S.H.; Johansen, I.T.; Ueland, T.; Lagerberg, T.V.; et al. Composite immune marker scores associated with severe mental disorders and illness course. Brain Behav. Immun. Health 2022, 24, 100483. [Google Scholar] [CrossRef] [PubMed]
  49. Mohamadi, Y.; Borhani-Haghighi, M. TGN020 Application against Aquaporin 4 Improved Multiple Sclerosis by Inhibiting Astrocytes, Microglia, and NLRP3 Inflammasome in a Cuprizone Mouse Model. J. Chem. Neuroanat. 2023, 132, 102306. [Google Scholar] [CrossRef] [PubMed]
  50. Kummer, J.A.; Broekhuizen, R.; Everett, H.; Agostini, L.; Kuijk, L.; Martinon, F.; van Bruggen, R.; Tschopp, J. Inflammasome Components NALP 1 and 3 Show Distinct but Separate expression profiles in Human Tissues Suggesting a Site-Specific Role in the Inflammatory Response. J. Histochem. Cytochem. 2007, 55, 443–452. [Google Scholar] [CrossRef] [PubMed]
  51. Yap, J.K.Y.; Pickard, B.S.; Chan, E.W.L.; Gan, S.Y. The Role of Neuronal NLRP1 Inflammasome in Alzheimer’s Disease: Bringing Neurons into the Neuroinflammation Game. Mol. Neurobiol. 2019, 56, 7741–7753. [Google Scholar] [CrossRef]
  52. Ma, Z.; Li, K.; Chen, P.; Pan, J.; Li, X.; Zhao, G. Propofol Attenuates Inflammatory Damage via Inhibiting NLRP1-Casp1-Casp6 Signaling in Ischemic Brain Injury. Biol. Pharm. Bull. 2020, 43, 1481–1489. [Google Scholar] [CrossRef]
  53. Huang, L.; Li, X.; Liu, Y.; Liang, X.; Ye, H.; Yang, C.; Hua, L.; Zhang, X. Curcumin Alleviates Cerebral Ischemia-reperfusion Injury by Inhibiting NLRP1-dependent Neuronal Pyroptosis. Curr. Neurovasc. Res. 2021, 18, 189–196. [Google Scholar] [CrossRef]
  54. Cristina de Brito Toscano, E.; Leandro Marciano Vieira, E.; Boni Rocha Dias, B.; Vidigal Caliari, M.; Paula Gonçalves, A.; Varela Giannetti, A.; Maurício Sigueira, J.; Suemoto, C.K.; Elaine Paraizo Leite, R.; Nitrini, R.; et al. NLRP3 and NLRP1 Inflammasomes Are Up-regulated in Patients with Mesial Temporal Epilepsy and may Contribute to Overexpression of Caspase-1 and IL-1β in Sclerotic Hippocampi. Brain Res. 2021, 1752, 147230. [Google Scholar] [CrossRef]
  55. Bernales, C.Q.; Encarnacion, M.; Criscuoli, M.G.; Yee, I.M.; Traboulsee, A.L.; Sadovnick, A.D.; Vilariño-Güell. Analysis of NOD-like Receptor NLRP1 in Multiple Sclerosis Families. Immunogenetics 2018, 70, 205–207. [Google Scholar] [CrossRef]
  56. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders, 5th ed.; Text Revision; American Psychiatric Association Publishing: Washington, DC, USA, 2000. [Google Scholar]
  57. Kostović, I.; Judaš, M.; Kostović-Knežević, L.J.; Šimić, G.; Delalle, I.; Chudy, D.; Šajin, B.; Petanjek, Z. Zagreb research collection of human brains for developmental neurobiologists and clinical neuroscientists. Int. J. Dev. Biol. 1991, 35, 215–230. [Google Scholar]
  58. Judaš, M.; Šimić, G.; Petanjek, Z.; Jovanov-Milošević, N.; Pletikos, M.; Vasung, L.; Vukšić, M.; Kostović, I. The Zagreb Collection of Human Brains: A Unique, Versatile, but Underexploited Resource for the Neuroscience Community. Ann. N. Y. Acad. Sci. 2011, 1225 (Suppl. 1), E105–E130. [Google Scholar] [CrossRef] [PubMed]
  59. Krušlin, B.; Džombeta, T.; Bezjak, M.; Sedmak, G.; Petanjek, Z.; Šimić, G.; Judaš, M.; Kostović, I. Congenital Brain Anomalies and Chromosomal Aberrations from the Zagreb Collection of the Human Brains. Transl. Neurosci. 2014, 5, 293–301. [Google Scholar] [CrossRef]
  60. Rajkowska, G.; Goldman-Rakic, P.S. Cytoarchitectonic definition of prefrontal areas in the normal human cortex: II. Variability in locations of areas 9 and 46 and relationship to the Talairach Coordinate System. Cereb. Cortex 1995, 5, 323–337. [Google Scholar] [CrossRef]
  61. Carmichael, S.T.; Price, J.L. Architectonic Subdivision of the Orbital and Medial Prefrontal Corte in the Macaque Monkey. J. Comp. Neurol. 1994, 346, 366–402. [Google Scholar] [CrossRef] [PubMed]
  62. Hof, P.R.; Mufson, E.J.; Morrison, J.H. Human Orbitofrontal Cortex: Cytoarchitecture and Quantitative Immunohistochemical Parcellation. J. Comp. Neurol. 1995, 359, 48–68. [Google Scholar] [CrossRef]
  63. Šimić, G.; Hof, P.R. In Search of the Definitive Brodmann’s Map of Cortical Areas in Human. J. Comp. Neurol. 2015, 523, 5–14. [Google Scholar] [CrossRef] [PubMed]
  64. Mladinov, M.; Sedmak, G.; Fuller, H.R.; Babić Leko, M.; Mayer, D.; Kirincich, J.; Štajduhar, A.; Borovečki, F.; Hof, P.R.; Šimić, G. Gene Expression Profiling of the Dorsolateral and Medial Orbitofrontal Cortex in Schizophrenia. Transl. Neurosci. 2016, 7, 139–150. [Google Scholar] [CrossRef]
  65. Irizarry, R.A.; Hobbs, B.; Collin, F.; Beazer-Barclay, Y.D.; Antonellis, K.J.; Scherf, U.; Speed, T.P. Exploration, Normalization, and Summaries of High-Density Oligonucleotide Array Probe Level Data. Biostatistics 2003, 4, 249–264. [Google Scholar] [CrossRef] [PubMed]
  66. Huang, D.W.; Sherman, B.T.; Lempicki, R.A. Systematic and Integrative Analysis of Large Gene Lists using DAVID Bioinformatics Resources. Nat. Protoc. 2009, 4, 44–57. [Google Scholar] [CrossRef]
  67. Penna, I.; Vella, S.; Gigoni, A.; Russo, C.; Cancedda, R.; Pagano, A. Selection of Candidate Housekeeping Genes for Normalization in Human Postmortem Brain Samples. Int. J. Mol. Sci. 2011, 12, 5461–5470. [Google Scholar] [CrossRef]
  68. Šimić, G.; Španić, E.; Langer Horvat, L.; Hof, P.R. Blood-Brain Barrier and Innate Immunity in the Pathogenesis of Alzheimer’s Disease. Prog. Mol. Biol. Transl. Sci. 2019, 168, 99–145. [Google Scholar]
  69. Cowan, M.; Petri, W.A., Jr. Microglia: Immune Regulators of Neurodevelopment. Front. Immunol. 2018, 9, 2576. [Google Scholar] [CrossRef]
  70. Menassa, D.A.; Muntslag, T.A.O.; Martin-Estebané, M.; Barry-Carroll, L.; Chapman, M.A.; Adorjan, I.; Tyler, T.; Turnbull, B.; Rose-Zerilli, M.J.J.; Nicoll, J.A.R.; et al. The Spatiotemporal Dynamics of Microglia Across the Human Lifespan. Dev. Cell. 2022, 57, 2127–2139. [Google Scholar] [CrossRef] [PubMed]
  71. Matuleviciute, R.; Akinluyi, E.T.; Muntslag, T.A.O.; Dewing, J.M.; Long, K.R.; Vernon, A.C.; Tremblay, M.-E.; Menassa, D.A. Microglial Contribution to the Pathology of Neurodevelopmental Disorders in Humans. Acta Neuropathol. 2023, 146, 663–683. [Google Scholar] [CrossRef] [PubMed]
  72. Canetta, S.E.; Brown, A.S. Prenatal Infection, Maternal Immune Activation, and Risk for Schizophrenia. Transl. Neurosci. 2012, 3, 320–327. [Google Scholar] [CrossRef]
  73. Bergdolt, L.; Dunaevsky, A. Brain Changes in a Maternal Immune Activation Model of Neurodevelopmental Brain Disorders. Prog. Neurobiol. 2019, 175, 1–19. [Google Scholar] [CrossRef]
  74. Ikezu, S.; Yeh, H.; Delpech, J.-C.; Woodbury, M.E.; Van Enoo, A.A.; Ruan, Z.; Sivakumaran, S.; You, Y.; Holland, C.; Guillamon-Vivancos, T.; et al. Inhibition of Colony Stimulating Factor 1 Receptor Corrects Maternal Inflammation-Induced Microglial and Synaptic Dysfunction and Behavioral Abnormalities. Mol. Psychiatry 2021, 26, 1808–1831. [Google Scholar] [CrossRef]
  75. Massrali, A.; Adhya, D.; Srivastava, D.P.; Baron-Cohen, S.; Kotter, M.R. Virus-Induced Maternal Immune Activation as an Environmental Factor in the Etiology of Autism and Schizophrenia. Front. Neurosci. 2022, 16, 834058. [Google Scholar] [CrossRef]
  76. Hall, M.B.; Willis, D.E.; Rodriguez, E.L.; Schwarz, J.M. Maternal Immune Activation as an Epidemiological Risk Ractor for Neurodevelopmental Disorders: Considerations of Timing, Severity, Individual Differences, and Sex in Human and Rodent Studies. Front. Neurosci. 2023, 17, 1135559. [Google Scholar] [CrossRef]
  77. Miller, B.J.; Goldsmith, D.R. Towards an Immunophenotype of Schizophrenia: Progress, Potential Mechanisms, and Future Directions. Neuropsychopharmacology 2017, 42, 299–317. [Google Scholar] [CrossRef]
  78. Johnson, T.; Saatci, D.; Handunnetthi, L. Maternal Immune Activation Induces Methylation Changes in Schizophrenia Genes. PLoS ONE 2022, 17, e0278155. [Google Scholar] [CrossRef]
  79. Nestler, E.J.; Peña, C.J.; Kundakovic, M.; Mitchell, A.; Akbarian, S. Epigenetic Basis of Mental Illness. Neuroscientist 2016, 22, 447–463. [Google Scholar] [CrossRef]
  80. Srivastava, A.; Dada, O.; Qian, J.; Al-Chalabi, N.; Fatemi, A.B.; Gerretsen, P.; Graff, A.; De Luca, V. Epigenetics of Schizophrenia. Psychiatry Res. 2021, 305, 114218. [Google Scholar] [CrossRef]
  81. Datta, D.; Arnsten, A.F.T. Unique Molecular Regulation of Higher-Order Prefrontal Cortical Circuits: Insights into the Neurobiology of Schizophrenia. ASC Chem. Neurosci. 2018, 9, 2127–2145. [Google Scholar] [CrossRef] [PubMed]
  82. Ising, C.; Venegas, C.; Zhang, S.; Scheiblich, H.; Schmidt, S.V.; Vieira-Saecker, A.; Schwartz, S.; Albasset, S.; McManus, R.M.; Tejera, D.; et al. NLRP Inflammasome Activation Drives Tau Pathology. Nature 2019, 575, 669–673. [Google Scholar] [CrossRef] [PubMed]
  83. Franklin, B.S.; Bossaller, L.; De Nardo, D.; Ratter, J.M.; Stutz, A.; Engels, G.; Brenker, C.; Nordhoff, M.; Mirandola, S.R.; Al-Amoudi, A.; et al. The Adaptor ASC has Extracellular and ‘Prionoid’ Activities that Propagate Inflammation. Nat. Immunol. 2014, 15, 727–737. [Google Scholar] [CrossRef] [PubMed]
  84. Stancu, I.-C.; Cremers, N.; Vanrusselt, H.; Couturier, J.; Vanoosthuyse, A.; Kessels, S.; Lodder, C.; Brône, B.; Huaux, F.; Octave, J.-N.; et al. Aggregated Tau Activates NLRP3-ASC Inflammasome Exacerbating Exogenously Seeded and Non-exogenously Seeded Tau Pathology In Vivo. Acta Neuropathol. 2019, 137, 599–617. [Google Scholar] [CrossRef]
  85. Del Cul, A.; Dehaene, S.; Leboyer, M. Preserved Subliminal Processing and Impaired Conscious Access in Schizophrenia. Arch. Gen. Psychiatry 2006, 63, 1313–1323. [Google Scholar] [CrossRef]
  86. Shipp, S. Neural Elements for Predictive Coding. Front. Psychol. 2016, 7, 1792. [Google Scholar] [CrossRef]
  87. Martin, B.; Wittmann, M.; Franck, N.; Cermolacce, M.; Berna, F.; Giersch, A. Temporal Structure of Consciousness and Minimal Self in Schizophrenia. Front. Psychol. 2014, 5, 1175. [Google Scholar] [CrossRef]
  88. Hur, J.-W.; Kwon, J.S.; Lee, T.Y.; Park, S. The Crisis of Minimal Self-awareness in Schizophrenia: A Meta-analytic Review. Schizophr. Res. 2014, 152, 58–64. [Google Scholar] [CrossRef]
  89. Stephen, K.E.; Friston, K.J.; Frith, C.D. Disconnection in Schizophrenia: From Abnormal Synaptic Plasticity to Failures in Self-monitoring. Schizophr. Bull. 2009, 35, 509–527. [Google Scholar] [CrossRef]
  90. Corlett, P.R.; Mollick, J.A.; Kober, H. Meta-analysis of Human Prediction Error for Incentives, Perception, Cognition, and Action. Neuropsychopharmacology 2022, 47, 1339–1349. [Google Scholar] [CrossRef]
  91. Reed, E.J.; Uddenberg, S.; Suthaharan, P.; Mathys, C.D.; Taylor, J.R.; Groman, S.M.; Corlett, P.R. Paranoia is a deficit in non-social belief updating. Elife 2020, 9, e56345. [Google Scholar] [CrossRef]
  92. Casserly, L.; Garton, D.R.; Montaño-Rodrigues, A.; Andreassoo, J.-O. Analysis of Acute and Chronic Methamphetamine Treatment in Mice on Gdnf System Expression Reveals a Potential Mechanism of Schizophrenia Susceptibility. Biomolecules 2023, 13, 1428. [Google Scholar] [CrossRef]
  93. Mahmoudiasl, G.R.; Abbaszadeh, H.A.; Rezaei-Tavirani, M.; Abdollahifar, M.A.; Khoramgah, M.S.; Niknazar, S.; Darabi, S.; Roozbahany, N.A. Nod-like Receptor Protein 3 and Nod-like Receptor Protein 1 Inflammasome Activation in the Hippocampal Region of Postmortem Methamphetamine Chronic User. Bratisl. Lek. Listy 2019, 120, 769–776. [Google Scholar] [CrossRef]
  94. Friedman, N.P.; Robbins, T.W. The Role of the Prefrontal Cortex in Cognitive Control and Executive Function. Neuropsychopharmacology 2022, 47, 72–89. [Google Scholar] [CrossRef]
  95. Winklbaur, B.; Ebner, N.; Sachs, G.; Thau, K.; Fischer, G. Substance Abuse in Patients with Schizophrenia. Dialogues Clin. Neurosci. 2006, 8, 37–43. [Google Scholar] [CrossRef]
  96. Duncan, J. The Multiple-demand (MD) System of the Primate Brain: Mental Programs for Intelligent Behavior. Trends Cog. Sci. 2010, 14, 172–179. [Google Scholar] [CrossRef]
  97. Meiron, O.; Yaniv, A.; Rozenberg, S.; David, J. Transcranial Direct-current Stimulation of the Prefrontal Cortex Enhances Working Memory and Suppresses Pathological Gamma Power Elevation in Schizophrenia. Expert Rev. Neurother. 2023, 12, 1–10. [Google Scholar] [CrossRef]
  98. Pujara, M.S.; Philippi, C.L.; Motzkin, J.C.; Baskaya, M.K. Ventromedial Prefrontal Cortex Damage is Associated with Decreased Ventral Striatum Volume and Response to Reward. J. Neurosci. 2016, 36, 5047–5054. [Google Scholar] [CrossRef]
  99. Šešo-Šimić, Đ.; Sedmak, G.; Hof, P.R.; Šimić, G. Recent advances in the neurobiology of attachment behavior. Transl. Neurosci. 2010, 1, 148–159. [Google Scholar] [CrossRef]
  100. Šimić, G.; Babić, M.; Borovečki, F.; Hof, P.R. Early Failure of the Default-Mode Network and the Pathogenesis of Alzheimer’s Disease. CNS Neurosci. Ther. 2014, 20, 692–698. [Google Scholar] [CrossRef]
  101. DiNicola, L.M.; Braga, M.; Buckner, R.L. Parallel Distributed Networks Dissociate Episodic and Social Functions within the Individual. J. Neurophysiol. 2020, 123, 1144–1179. [Google Scholar] [CrossRef]
  102. Šimić, G.; Vukić, V.; Kopić, J.; Krsnik, Ž.; Hof, P.R. Molecules, Mechanisms, and Disorders of Self-Domestication: Keys for Understanding Emotional and Social Communication from an Evolutionary Perspective. Biomolecules 2021, 11, 2. [Google Scholar] [CrossRef]
  103. Zooh, Y.; Chang, S.W.C.; Crockett, M.J. The Prefrontal Cortex and (Uniquely) Human Cooperation: A Comparative Perspective. Neuropsychopharmacology 2021, 47, 119–133. [Google Scholar] [CrossRef]
  104. Petanjek, Z.; Judaš, M.; Šimić, G.; Rašin, M.R.; Uylings, H.B.; Rakic, P.; Kostović, I. Extraordinary Neoteny of Synaptic Spines in the Human Prefrontal Cortex. Proc. Natl. Acad. Sci. USA 2011, 108, 13281–13286. [Google Scholar] [CrossRef]
  105. Ma, S.; Škarica, M.; Li, Q.; Xu, C.; Risgaard, R.D.; Tebbenkamp, A.T.N.; Mato-Blanco, X.; Kovner, R.; Krsnik, Ž.; Luria, V.; et al. Molecular and Cellular Evolution of the Primate Dorsolateral Prefrontal Cortex. Science 2022, 377, eabo7257. [Google Scholar] [CrossRef] [PubMed]
  106. Raghanti, M.A.; Stimpson, C.D.; Marcinkiewicz, J.L.; Erwin, J.M.; Hof, P.R.; Sherwood, C.C. Cortical Dopaminergic Innervation among Humans, Chimpanzees, and Macaque Monkeys: A Comparative Study. Neuroscience 2008, 155, 203–220. [Google Scholar] [CrossRef]
  107. Jones, D.N.; Raghanti, M.A. The Role of Monoamine Oxidase Enzymes in the Pathophysiology of Neurological Disorders. J. Chem. Neuroanat. 2021, 114, 101957. [Google Scholar] [CrossRef]
  108. Gaspar, P.; Berger, B.; Febvret, A.; Vigny, A.; Henry, J.P. Catecholamine Innervation of the Human Cerebral Cortex as Revealed by Comparative Immunohistochemistry of Tyrosine Hydroxylase and Dopamine-β-hydroxylase. J. Comp. Neurol. 1989, 279, 249–271. [Google Scholar] [CrossRef]
  109. Mladinov, M.; Mayer, D.; Brčić, L.; Wolstencroft, E.; Man, N.T.; Holt, I.; Hof, P.R.; Morris, G.E.; Šimić, G. Astrocyte Expression of D2-like Dopamine Receptors in the Prefrontal Cortex. Transl. Neurosci. 2010, 1, 238–243. [Google Scholar] [CrossRef]
  110. Abi-Dargham, A.; Javitch, J.A.; Slifstein, M.; Antičević, A.; Calkins, M.E.; Cho, Y.T.; Fonteneau, C.; Gil, R.; Girgis, R.; Gur, R.E.; et al. Dopamine D1R Receptor Stimulation as a Mechanistic Pro-cognitive Target for Schizophrenia. Schizophr. Bull. 2022, 48, 199–210. [Google Scholar] [CrossRef]
  111. Shen, L.-H.; Liao, M.-H.; Tseng, Y.-C. Recent advances in imaging of dopaminergic neurons for evaluation of neuropsychiatric disorders. J. Biomed. Biotechnol. 2012, 2012, 259349. [Google Scholar] [CrossRef]
  112. Patel, K.R.; Cherian, J.; Gohil, K.; Atkinson, D. Schizophrenia: Overview and Treatment Options. Pharm. Ther. 2014, 39, 638–645. [Google Scholar]
  113. Kaneko, K. Negative Symptoms and Cognitive Impairments in Schizophrenia: Two Key Symptoms Negatively Influencing Social Functioning. Yonago Acta Med. 2018, 61, 91–102. [Google Scholar] [CrossRef]
  114. Tsu, B.V.; Beierschmitt, C.; Ryan, A.P.; Agarwal, R.; Mitchell, P.S.; Daugherty, M.D. Diverse Viral Proteases Activate the NLRP1 Inflammasome. Elife 2021, 10, e60609. [Google Scholar] [CrossRef] [PubMed]
  115. Pontillo, A.; Catamo, E.; Arosio, B.; Mari, D.; Crovella, S. NALP/NLRP1 Genetic Variants Are Associated with Alzheimer’s Disease. Alzheimer Dis. Assoc. Disord. 2012, 26, 277–281. [Google Scholar] [CrossRef] [PubMed]
  116. Venegas, C.; Kumar, S.; Franklin, B.S.; Dierkes, T.; Brinkschulte, R.; Tejera, D.; Vieira-Saecker, A.; Schwartz, S.; Santarelli, F.; Kummer, M.P.; et al. Microglia-Derived ASC Specks Cross-Seed Amyloid-β in Alzheimer’s Disease. Nature 2017, 552, 355–361. [Google Scholar] [CrossRef] [PubMed]
  117. Cho, M.; Lee, T.Y.; Kwak, Y.B.; Yoon, Y.B.; Kim, M.; Kwon, J.S. Adjunctive Use of Antiinflammatory Drugs for Schizophrenia: A Meta-analytic Investigation of Randomized Controlled Trials. Aust. N. Z. J. Psychiatry 2019, 53, 742–759. [Google Scholar] [CrossRef] [PubMed]
  118. Hilker, R.; Helenius, D.; Fagerlund, B.; Skytthe, A.; Christensen, K.; Werge, T.M.; Nordentoft, M.; Glenthøj, B. Heritability of Schizophrenia and Schizophrenia Spectrum Based on the Nationwide Danish Twin Register. Biol. Psychiatry 2018, 83, 492–498. [Google Scholar] [CrossRef]
  119. Trubetskoy, V.; Pardiñas, A.F.; Qi, T.; Panagiotaropoulou, G.; Awasthi, S.; Bigdeli, T.B.; Bryois, J.; Chen, C.Y.; Dennison, C.A.; Hall, L.S. Mapping Genomic Loci Implicates Genes and Synaptic Biology in Schizophrenia. Nature 2022, 604, 7906. [Google Scholar] [CrossRef]
  120. Stassen, H.H.; Bachmann, S.; Bridler, R.; Cattapan, K.; Herzig, D.; Schneeberger, A.; Seifritz, E. Inflammatory Processes Linked to Major Depression and Schizophrenic Disorders and the Effects of Polypharmacy in Psychiatry: Evidence from a Longitudinal Study of 279 Patients under Therapy. Eur. Arch. Psychiatry Clin. Neurosci. 2021, 271, 507–520. [Google Scholar] [CrossRef]
Figure 1. Locations of the analyzed samples. Blocks of brain tissue were taken from two different areas: (A) the dorsolateral prefrontal cortex (DLPFC, Brodmann’s area 46), and (B) the medial orbitofrontal cortex (MOFC, located between Brodmann’s areas 11 and 12). The specific locations from which these tissue blocks were taken are indicated by transparent red rectangles. The template for the figure is taken from our previous publications on Brodmann’s areas [63] and gene expression profiling in SZ [64].
Figure 1. Locations of the analyzed samples. Blocks of brain tissue were taken from two different areas: (A) the dorsolateral prefrontal cortex (DLPFC, Brodmann’s area 46), and (B) the medial orbitofrontal cortex (MOFC, located between Brodmann’s areas 11 and 12). The specific locations from which these tissue blocks were taken are indicated by transparent red rectangles. The template for the figure is taken from our previous publications on Brodmann’s areas [63] and gene expression profiling in SZ [64].
Biomolecules 14 00302 g001
Figure 2. Graphical representation of the results from Table 3, showing that the average neuronal density of NLRP1-expressing MOFC pyramidal neurons in the RH is significantly higher (p < 0.01) in the group of seven schizophrenia (SZ) samples compared to the group of five control (CON) samples. MOFC, medial orbitofrontal cortex; RH, right hemisphere.
Figure 2. Graphical representation of the results from Table 3, showing that the average neuronal density of NLRP1-expressing MOFC pyramidal neurons in the RH is significantly higher (p < 0.01) in the group of seven schizophrenia (SZ) samples compared to the group of five control (CON) samples. MOFC, medial orbitofrontal cortex; RH, right hemisphere.
Biomolecules 14 00302 g002
Figure 3. Microphotograph of NLRP1 immunocytochemical staining of MOFC from the right hemisphere of a selected control sample (CON9). A few pyramidal neurons in layer III are NLRP1-immunoreactive, while layers V and VI are almost devoid of labeled neurons. Insets (A) and (B) from (1) and (2), respectively, are Nissl-stained adjacent sections that allow cytoarchitecture to be appreciated. Note the thin layer IV. Scale bar = 2.5 mm. Scale bars in the insets (1,2) as well as (A,B) = 500 μm.
Figure 3. Microphotograph of NLRP1 immunocytochemical staining of MOFC from the right hemisphere of a selected control sample (CON9). A few pyramidal neurons in layer III are NLRP1-immunoreactive, while layers V and VI are almost devoid of labeled neurons. Insets (A) and (B) from (1) and (2), respectively, are Nissl-stained adjacent sections that allow cytoarchitecture to be appreciated. Note the thin layer IV. Scale bar = 2.5 mm. Scale bars in the insets (1,2) as well as (A,B) = 500 μm.
Biomolecules 14 00302 g003
Figure 4. Microphotographs of NLRP1 immunocytochemical staining of MOFC from the right hemisphere of an SZ sample (SZ6). Many pyramidal neurons of layer III as well as pyramidal neurons in layers V and VI are NLRP1-immunoreactive. Layer IV is virtually free from NLRP1-immunoreactive cells. Also, note cortical atrophy and the paucity of the white matter relative to the 9-year older control sample shown in Figure 3. Insets (A) and (B) from (1) and (2), respectively, are Nissl-stained adjacent sections that allow cytoarchitecture to be appreciated. Scale bar = 2.5 mm. Scale bars in the insets (14) = 500 μm. Scale bars in (A,B) = 250 μm.
Figure 4. Microphotographs of NLRP1 immunocytochemical staining of MOFC from the right hemisphere of an SZ sample (SZ6). Many pyramidal neurons of layer III as well as pyramidal neurons in layers V and VI are NLRP1-immunoreactive. Layer IV is virtually free from NLRP1-immunoreactive cells. Also, note cortical atrophy and the paucity of the white matter relative to the 9-year older control sample shown in Figure 3. Insets (A) and (B) from (1) and (2), respectively, are Nissl-stained adjacent sections that allow cytoarchitecture to be appreciated. Scale bar = 2.5 mm. Scale bars in the insets (14) = 500 μm. Scale bars in (A,B) = 250 μm.
Biomolecules 14 00302 g004
Table 1. Demographic data of subjects with schizophrenia and control subjects.
Table 1. Demographic data of subjects with schizophrenia and control subjects.
CaseSex (F/M)Age (Years)Cause of Death
Subjects with schizophrenia (SZ)
SZ1F42Drug poisoning
SZ2F47Sudden cardiac arrest
SZ3M50Heart failure
SZ4F50Not known (no autopsy conducted)
SZ5F56Aortic dissection
SZ6F57Sudden cardiac arrest
SZ7F58Suicide by jumping from a height
SZ8M59Suicide by hanging
SZ9M59Suicide by hanging
SZ10M81Not known (no autopsy conducted)
Mean ± SD55.9 ± 10.5
Control subjects (CON)
CON1F40Heart failure
CON2M42Heart failure
CON3M54Heart failure
CON4M55Heart failure
CON5F60Sudden cardiac arrest
CON6F61Heart failure
CON7M63Sudden cardiac arrest
CON8M64Sudden cardiac arrest
CON9M66Sudden cardiac arrest
CON10M73Pulmonary embolism
Mean ± SD57.8 ± 10.4
CON, control subjects; F, female; M, male; SD, standard deviation; SZ, schizophrenia subjects.
Table 2. Tissue characteristics of the control (CON) and schizophrenia (SZ) brain samples analyzed for the expression of the NLRP1 gene (dataset line 18,408 in Supplementary Table S1). RIN was calculated using a proprietary algorithm of Agilent Technologies, Santa Clara, CA, USA. All p-values of the Shapiro–Wilk test greater than α = 0.05 mean that a sample comes from a normally distributed population.
Table 2. Tissue characteristics of the control (CON) and schizophrenia (SZ) brain samples analyzed for the expression of the NLRP1 gene (dataset line 18,408 in Supplementary Table S1). RIN was calculated using a proprietary algorithm of Agilent Technologies, Santa Clara, CA, USA. All p-values of the Shapiro–Wilk test greater than α = 0.05 mean that a sample comes from a normally distributed population.
CON Subject/RINBrain Tissue BlockNLRP1
mRNA
exp. in
LH
DLPFC
NLRP1
mRNA
exp. in
RH
DLPFC
NLRP1
mRNA
exp. in
LH
MOFC
NLRP1
mRNA
exp. in
RH
MOFC
SZ Subject/RINBrain Tissue BlockNLRP1
mRNA
exp. in
LH
DLPFC
NLRP1
mRNA
exp. in
RH
DLPFC
NLRP1
mRNA
exp. in
LH
MOFC
NLRP1
mRNA
exp. in
RH
MOFC
CON3
7.5
LH DLPFC11.7 SZ8
8.7
RH DLPFC 13.2
RH DLPFC 14.2 LH DLPFC12.4
LH MOFC 14.1 RH MOFC 12.9
RH MOFC 11.3LH MOFC 13.6
CON5
6.9
LH DLPFC13.2 SZ5
7.4
RH DLPFC 13.6
LH MOFC 11.7 LH DLPFC13.0
RH DLPFC 13.1 RH MOFC 16.0
RH MOFC 12.4LH MOFC 16.0
CON6
8.6
RH DLPFC 13.2 SZ6
5.0
RH DLPFC 14.6
RH MOFC 11.7LH DLPFC13.7
LH DLPFC13.1 RH MOFC 14.0
LH MOFC 12.4 LH MOFC 12.4
CON1
7.7
RH DLPFC 14.1 SZ3
8.1
RH DLPFC 13.2
LH DLPFC12.2 LH DLPFC12.3
RH MOFC 13.0RH MOFC 16.3
LH MOFC 11.6 LH MOFC 16.5
CON4
8.6
RH DLPFC 12.5 SZ2
8.0
RH DLPFC 17.2
LH DLPFC16.4 LH DLPFC16.6
RH MOFC 15.0RH MOFC 15.0
LH MOFC 14.0 LH MOFC 15.7
CON2
8.8
RH DLPFC 12.4 SZ1
5.0
RH DLPFC 15.7
LH DLPFC15.5 LH DLPFC13.8
RH MOFC 16.7RH MOFC 19.1
LH MOFC 15.6 LH MOFC 14.3
Shapiro–Wilk p0.440.360.480.47 Shapiro–Wilk p0.120.290.760.96
8.0Mean13.6713.2613.2513.997.0Mean13.6314.5714.7515.53
0.7SD1.870.851.582.321.7SD1.601.581.582.14
Total mean13.543 (N = 24) Total mean14.620 (N = 24)
Total SD1.7391 Total SD1.7415
Unpaired two-tailed Student’s t-test: T = −2.1498, d.f. = 46, p = 0.03687
Non-parametric two-sample, two-tailed Mann–Whitney U test: Z = −2.4436, p = 0.01454
CON, control; DLPFC, dorsolateral prefrontal cortex (Brodmann’s area 46); exp., expression; LH, left hemisphere; MOFC, medial orbitofrontal cortex (Brodmann’s area 11/12); RH, right hemisphere; RIN, RNA integrity number; SD, standard deviation; SZ, schizophrenia.
Table 3. Quantitative assessment of the number of NLRP1-expressing pyramidal neurons in the MOFC of control (CON) and schizophrenia (SZ) subjects. MOFC, medial orbitofrontal cortex (Brodmann’s area 11/12); RH, right hemisphere; SD, standard deviation.
Table 3. Quantitative assessment of the number of NLRP1-expressing pyramidal neurons in the MOFC of control (CON) and schizophrenia (SZ) subjects. MOFC, medial orbitofrontal cortex (Brodmann’s area 11/12); RH, right hemisphere; SD, standard deviation.
CON SubjectNumber of NLRP1-Positive Pyramidal Neurons in RH MOFCArea [mm2]Average Density/mm2SZ SubjectNumber of NLRP1-Positive Pyramidal Neurons in RH MOFCArea [mm2]Average Density/mm2
CON825447.85.3SZ5124893.613.3
CON91020184.15.5SZ8122487.813.9
CON1055858.09.6SZ783062.313.3
CONT542289.74.7SZ964835.818.1
CON71064114.79.3SZ101602158.710.1
SZ6103764.416.1
SZ4101083.312.1
Mean663.698.866.88 (N = 5)Mean1085.683.713.84 (N = 7)
SD362.1654.492.37SD310.1338.442.61
Unpaired two-tailed Student’s t-test: T = −4.8068, d.f. = 10, p = 0.00088
Non-parametric two-sample, two-tailed Mann–Whitney U test: Z = −2.7656, p = 0.005681
Table 4. Comparison of housekeeping gene expression between schizophrenia (SZ) and control (CON) samples. Next to the name of each gene in the second column is its Entrez Gene ID number and the dataset line of Supplementary Table S1 in which the mRNA expression of that gene for all samples can be found.
Table 4. Comparison of housekeeping gene expression between schizophrenia (SZ) and control (CON) samples. Next to the name of each gene in the second column is its Entrez Gene ID number and the dataset line of Supplementary Table S1 in which the mRNA expression of that gene for all samples can be found.
GeneEntrez Gene ID/Dataset LineAverage Expression in
24 Samples of
Six CON Subjects ± SD
Average Expression in
24 Samples of
Six SZ Subjects ± SD
Difference
ACTB3036924
8960
6466.17 ± 1028.01> 5356.90 ± 1821.50Significant
(T = 2.60, d.f. = 46, p = 0.013)
B2M3592023
13433
336.17 ± 162.40> 261.70 ± 152.95Non-significant
(T = 1.64, d.f. = 46, p = 0.109)
CYC13120051
6681
155.984 ± 73.52> 116.69 ± 28.53Significant
(T = 2.44, d.f. = 46, p = 0.019)
EIF4A22656738
8617
1648.94 ± 583.95> 1265.58 ± 380.49Significant
(T = 2.69, d.f. = 46, p = 0.010)
GUSB3053691
951
33.98 ± 5.69< 37.50 ± 6.37Significant
(T = −2.02, d.f. = 46, p = 0.049)
HMBS3351841
17437
23.46 ± 4.81> 18.20 ± 3.69Significant
(T = 4.25, d.f. = 46, p = 0.0001)
HPRT13991698
84
520.55 ± 251.05> 354.93 ± 188.48Significant
(T = 2.58, d.f. = 46, p = 0.013)
PGK13982462
14144
3439.35 ± 615.11> 3352.13 ± 577.90Non-significant
(T = 0.51, d.f. = 46, p = 0.506)
PPIA3000073
19231
20.97 ± 7.28< 21.84 ± 5.50Non-significant
(T = −0.47, d.f. = 46, p = 0.642)
RPLP03474344
6859
74.18 ± 14.20< 80.61 ± 20.16Non-significant
(T = −1.28, d.f. = 46, p = 0.208)
SDHA2798538
14738
1249.97 ± 172.91> 1140.73 ± 296.17Non-significant
(T = 1.56, d.f. = 46, p = 0.126)
TBP2937984
9928
102.00 ± 25.81> 118.95 ± 25.85Significant
(T = −2.27, d.f. = 46, p = 0.028)
TFRC2712632
5038
141.99 ± 35.76> 134.22 ± 45.06Non-significant
(T = 0.66, d.f. = 46, p = 0.51)
YWHAZ3146898
9013
1307.58 ± 299.20< 1315.23 ± 334.26Non-significant
(T = −0.08, d.f. = 46, p = 0.934)
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

Španić Popovački, E.; Vogrinc, D.; Fuller, H.R.; Langer Horvat, L.; Mayer, D.; Kopić, J.; Pintarić, K.; Babić Leko, M.; Pravica, M.; Krsnik, Ž.; et al. Increased NLRP1 mRNA and Protein Expression Suggests Inflammasome Activation in the Dorsolateral Prefrontal and Medial Orbitofrontal Cortex in Schizophrenia. Biomolecules 2024, 14, 302. https://doi.org/10.3390/biom14030302

AMA Style

Španić Popovački E, Vogrinc D, Fuller HR, Langer Horvat L, Mayer D, Kopić J, Pintarić K, Babić Leko M, Pravica M, Krsnik Ž, et al. Increased NLRP1 mRNA and Protein Expression Suggests Inflammasome Activation in the Dorsolateral Prefrontal and Medial Orbitofrontal Cortex in Schizophrenia. Biomolecules. 2024; 14(3):302. https://doi.org/10.3390/biom14030302

Chicago/Turabian Style

Španić Popovački, Ena, Dora Vogrinc, Heidi R. Fuller, Lea Langer Horvat, Davor Mayer, Janja Kopić, Klara Pintarić, Mirjana Babić Leko, Mihaela Pravica, Željka Krsnik, and et al. 2024. "Increased NLRP1 mRNA and Protein Expression Suggests Inflammasome Activation in the Dorsolateral Prefrontal and Medial Orbitofrontal Cortex in Schizophrenia" Biomolecules 14, no. 3: 302. https://doi.org/10.3390/biom14030302

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