Next Article in Journal
Dopamine Photochemical Behaviour under UV Irradiation
Next Article in Special Issue
An Adaptive Role for DNA Double-Strand Breaks in Hippocampus-Dependent Learning and Memory
Previous Article in Journal
Molecular Rearrangement of Pyrazino[2,3-c]quinolin-5(6H)-ones during Their Reaction with Isocyanic Acid
Previous Article in Special Issue
Early Life Events and Maturation of the Dentate Gyrus: Implications for Neurons and Glial Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Structural and Functional Deviations of the Hippocampus in Schizophrenia and Schizophrenia Animal Models

1
Department of Cell Morphology and Molecular Neurobiology, Faculty for Biology and Biotechnology, Ruhr-University Bochum, Universitaetsstr. 150, D-44801 Bochum, Germany
2
Department of Psychiatry, LWL University Hospital, Ruhr-University Bochum, Alexandrinenstr. 1, D-44791 Bochum, Germany
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(10), 5482; https://doi.org/10.3390/ijms23105482
Submission received: 12 April 2022 / Revised: 9 May 2022 / Accepted: 11 May 2022 / Published: 13 May 2022

Abstract

:
Schizophrenia is a grave neuropsychiatric disease which frequently onsets between the end of adolescence and the beginning of adulthood. It is characterized by a variety of neuropsychiatric abnormalities which are categorized into positive, negative and cognitive symptoms. Most therapeutical strategies address the positive symptoms by antagonizing D2-dopamine-receptors (DR). However, negative and cognitive symptoms persist and highly impair the life quality of patients due to their disabling effects. Interestingly, hippocampal deviations are a hallmark of schizophrenia and can be observed in early as well as advanced phases of the disease progression. These alterations are commonly accompanied by a rise in neuronal activity. Therefore, hippocampal formation plays an important role in the manifestation of schizophrenia. Furthermore, studies with animal models revealed a link between environmental risk factors and morphological as well as electrophysiological abnormalities in the hippocampus. Here, we review recent findings on structural and functional hippocampal abnormalities in schizophrenic patients and in schizophrenia animal models, and we give an overview on current experimental approaches that especially target the hippocampus. A better understanding of hippocampal aberrations in schizophrenia might clarify their impact on the manifestation and on the outcome of this severe disease.

1. Introduction

In 1893, Emil Kraepelin described schizophrenia as a progressive neurodegenerative disease which leads to an irreversible loss of cognitive functions, and he differentiated it from manic depression [1]. While Kraepelin initially used the term dementia praecox, Eugen Bleuer renamed it to the commonly used designation schizophrenia [2]. Nowadays, it is known that schizophrenia is a complex neuropsychiatric disease with a mean lifetime prevalence of 1%, a reduced life expectancy and a multifactorial cause [3,4,5]. The recent DSM-5 criteria for schizophrenia include delusions, hallucinations, disorganized speech, a grossly disorganized or catatonic behavior and negative symptoms; two or more of these must persist for a period of one month or longer [5]. While schizophrenia is frequently diagnosed in early adulthood, the current state of research posits that the pathogenesis begins in early neurodevelopment [5,6]. This is supported by studies that have observed childhood neurobehavioral deficits in the offspring of schizophrenic parents [7]. In general, the symptoms of schizophrenia can be classified into positive, negative and cognitive symptoms. While the positive symptoms of schizophrenia include hallucinations, delusions, thought disorder and a lack of insight [8], the negative symptoms consist of social withdrawal, poverty of speech, self-neglect, highly reduced motivation and anhedonia [9]. Furthermore, cognitive symptoms are characterized by deficits in attention, working memory, verbal declarative memory and several other executive function impairments [10]. The brain of schizophrenic patients shows structural deviations that include, inter alia, a thinning of the cerebellar cortex [11,12] and a reduced volume of the thalamus and the striatum, accompanied by an enlargement of the lateral ventricles [13,14,15,16]. With a focus on cognitive symptoms that are impairing the life quality of patients, structural aberrations of the hippocampus might especially be of great interest. The hippocampus is located in the allocortex and can be subdivided into the dentate gyrus and the cornu ammonis (CA). It is strongly involved in higher cognitive functions like spatial memory, episodic memory, short-term memory and long-term memory [17]. Interestingly, schizophrenic patients show morphological as well as functional deviations of the hippocampus, as the following review will address. Furthermore, current findings of schizophrenia animal models will give an overview on recent approaches that target the hippocampus as a plastic structure of the central nervous system (CNS).

2. Structural Abnormalities of the Hippocampus in Schizophrenia Patients

For a long while, it has been well-known that structural aberrations of the hippocampus are a common hallmark of schizophrenia (Figure 1). Over decades, the evidence of severe hippocampal abnormalities in schizophrenia patients is mounting and includes a remarkable reduction of the hippocampal volume [18,19,20,21,22,23,24,25], a dysregulation of synaptic protein levels [26,27,28] and a dysconnectivity of the hippocampus from further parts of the CNS [29,30,31,32]. The general reduction of the hippocampal volume can already be observed in patients with a high risk for psychosis [33]. This indicates that the structural deviation occurs in a period before the disease manifestation, supporting the neurodevelopmental model for schizophrenia [34,35,36,37].
While former studies mainly focused on total volume changes of the hippocampus in schizophrenic patients, modern high-resolution magnet resonance imaging (MRI) techniques were utilized for a more detailed and region-specific analysis. Interestingly, these techniques contributed to the observation of so-called incomplete inversion patterns which are characterized by a round and verticalized hippocampal morphology with a deep collateral fissure, and a medial positioning in the coronal plane [38,39,40]. Surprisingly, this structural deviation can be observed in 18–19% of healthy subjects, predominantly in the left hemisphere, and was primarily suspected to contribute to the pathology of epileptic seizures [40,41]. Nevertheless, a recently published study showed that incomplete inversion patterns occur, on the one hand in a more severe manner and on the other hand with a higher frequency in schizophrenic patients [38]. In addition, this study unraveled a link between incomplete inversion patterns and the commonly described reduction of the total hippocampal volume, as well as of the increased volume asymmetry between the left and right hemispheres. Furthermore, the question of whether or not specific incomplete inversion patterns correlate with auditory and visual hallucinations was addressed. Intriguingly, schizophrenic patients with both auditory and visual hallucinations showed a flatter hippocampal morphology in the left hemisphere than healthy subjects and patients suffering exclusively from auditory hallucinations [42].
These observations highly support the neurodevelopmental model for schizophrenia since the inversion of the dentate gyrus and the cornu ammonis occurs in the second trimester of pregnancy [43,44,45]. Ultrasound studies could show that in approximately 50% of preterm neonates, the inversion of the hippocampus is not completely accomplished up to gestation week 24 [45]. However, at gestation week 25, the frequency of incomplete hippocampal inversions is comparable to the frequency of incomplete inversions in the adult population [45]. A genome-wide association study analyzed the heritability of incomplete inversion patterns in adolescents [46]. Here, a high heritability rate was observed with a significantly linked locus identified on chromosome 18q11.2. In addition to this genetic aspect, the second trimester of pregnancy, when the inversion of the hippocampus occurs, appears to be sensitive for environmental factors like inflammation, stress and anxiety [47,48,49]. For this period, it has been described that maternal stress and anxiety clearly have negative effects on neurodevelopmental events and induce structural changes of the hippocampus, as well as a general reduction of the grey matter volume [48,49]. While structural alterations of the hippocampus have been described in very preterm born infants, there is a lack of scientific data about the impact of pre- or perinatal infections and other complications with regard to the disruption of the hippocampal inversion process [50]. Since it is known that a very preterm birth and a low birth weight are risk factors for schizophrenia in later life, it would be of great interest to answer the question of if and how environmental risk factors affect the restriction of the hippocampal inversion process [51,52,53,54]. The identification of potential risk factors could contribute to the prevention of this morphological abnormality. The genetic and environmental risk factors which are suspected to be responsible for the deviations of the hippocampal morphology will be discussed in more detail at a later point of this review.
Despite the restriction of the hippocampal inversion and the general volume reduction, region-specific aberrations could be seen in patients with a high risk for psychosis and in schizophrenia patients. These were especially shown in the anterior and midbody cornu ammonis region 1 (CA1) and 2 (CA2). Narr and colleagues observed that mid- to antero-lateral hippocampal regions show a distinct neuroanatomical volume change in schizophrenic patients and proposed that these regions are more susceptible for the disease progression [55]. In contrast, another study showed a heterogeneous loss of the hippocampal volume at the posterior ends in early-onset schizophrenia (EOS) patients, while the main body volume of the hippocampus was increased [56]. Similarly, childhood-onset schizophrenia patients developed a morphological bilateral inward deformation of the anterior hippocampus which was furthermore related to the severity of the positive symptoms [57]. In this study, healthy siblings of schizophrenic patients were included and tended also to an anterior inward deformation, indicating a genetic vulnerability for this region-specific structural abnormality. The differing region-specific abnormalities of the hippocampus described by various studies might be explained by individual patient-specific risk factors that contributed to the disease manifestation (e.g., genetic factors, prenatal infections, perinatal complications, childhood abuse, drug abuse) or by the individual stage of disease progression (prodromal, acute, residual, childhood-onset). Based on these observations, the morphology of the hippocampus was suspected to give an overview on the disease progression and to be considered for the determination of individual therapeutical strategies. The interesting question is whether or not the volume reduction of the hippocampus in high-risk patients can be consulted for the prediction of a transition towards psychosis and was addressed in recent meta-analyses [58,59]. Importantly, both studies showed no statistical significance between the effect of the hippocampal volume on the transition risk for psychosis. Consequently, the volume of the hippocampus alone is not a sufficient predictor for the transition toward psychosis. Nevertheless, further functional parameters might be considered in addition to the morphological aberrations, as described in the following section of this review.
In addition to the macrostructural alterations of the hippocampus in schizophrenia patients, there are further differences on a synaptic level [60,61]. Here, a recently published study utilized a positron emission tomography (PET) radioligand for the synaptic vesicle protein 2A (SV2A) and observed a significant reduction of synaptic vesicles in the hippocampus of schizophrenia patients [60,62]. In addition, the postsynaptic compartment is affected in a structural manner. A significantly reduced density of dendritic spines in the prefrontal cortex was described in several postmortem studies and additionally confirmed in a recently published meta-analysis [63,64,65,66,67]. While the dendritic spine pathology was frequently described in the prefrontal cortex, less studies focused on dendritic spines in the hippocampus. Interestingly, the above-mentioned meta-analysis observed that the overall size of the effect of studies on the density of postsynaptic elements was unchanged in the hippocampus of schizophrenic patients [63]. Nevertheless, a reduction of the postsynaptic density protein-95 (PSD-95) could be verified on a protein level in the CA1-region and in the dentate molecular layer in postmortem studies [26,28]. Similar results were obtained for the synapse associated protein of 102 kDa (SAP-102) [68]. Contrary to these observations, a quantitative analysis of Golgi-stained hippocampal neurons showed an increase in the spine density on CA3 pyramidal cell dendrites and a rise in the number of thorny excrescences [27]. These observations indicate a necessity of further studies that characterize the dendritic spine densities and morphologies in the hippocampal subfields of schizophrenia patients. Table 1 contains an overview on the different types of structural aberrations in the hippocampus of schizophrenia patients with the corresponding references.

3. Functional Aberrations of the Hippocampus in Schizophrenia Patients

The distinctive feature of total and region-specific volume changes, as well as of a functional dysconnectivity in schizophrenia patients, is frequently accompanied by an electrophysiological and metabolic hyperactivity of the hippocampus. Here, raised levels of glutamate and glutamine were already detected in patients with a high risk for psychosis via functional MRI analysis, as a previously published study can show [69]. Functional MRI studies of schizophrenia patients revealed a hippocampal hyperactivity during resting stages and during minimal cognitive tasks [70,71]. Furthermore, there is the evidence that the hippocampal hyperactivity spreads from a hypermetabolic CA1-region to the subiculum, inducing the transition from a prodromal state toward a psychotic state in patients with psychosis [72]. The hyperactivity of the anterior CA1-region and the subiculum correlates with the severity of positive, negative and cognitive symptoms [70,72,73]. With regard to cognitive parameters, the hippocampal activity was analyzed during tasks addressing specific parameters. Here, a higher hippocampal activation could be observed when schizophrenia patients performed sensory gating tasks or when they were merely exposed to urban noise stimuli [74,75]. Furthermore, schizophrenic patients developed hippocampal deficits in relational memory formation tasks [76,77], as well as face habituation tasks [78,79]. A hyperactivity of the hippocampus frequently goes along with a higher metabolic activity and cerebellar blood volume levels [80]. Measurements of the regional cerebellar blood flow levels in patients who are ultra-high-risk for psychosis showed a significant increase in comparison to healthy subjects [81]. The rise of the cerebellar blood volume levels interestingly vanished when the patients did not meet ultra-high-risk criteria [81]. Regional cerebellar blood volume analyses unraveled that the severity of delusions showed the highest association with increased blood volume levels in the CA1-region of the hippocampus [73]. The increase of the cerebellar blood volume levels in the CA1-subfield furthermore predicted the clinical progression from a prodromal state toward psychosis. While the volume reduction of the hippocampus alone is not sufficient to predict a transition from the prodromal state toward the psychotic state, cerebellar blood volume parameters could be additionally considered to predict an immediate psychotic phase.
In addition to the hyperactivity of the hippocampus in patients at high risk for psychosis and schizophrenia patients, a functional dysconnectivity has been described for the hippocampo-striatal and the hippocampo-prefrontal pathway [29,31,82]. In a recently published study, the connectivity of the anterior hippocampus in patients with a first-episode psychosis is therefore discussed as a useful predictor for the response of an antipsychotic treatment [83]. Another study observed a functional hypoconnectivity of the hippocampus with regions that are involved in episodic memory, such as the medial prefrontal cortex or the parahippocampal gyrus [84]. In addition, functional MRI-scans of unaffected first-degree relatives of schizophrenia patients revealed a hyperconnectivity of the left anterior cingulate cortex with the right hippocampus and a hypoconnectivity of the right anterior cingulate cortex with the right hippocampus, in comparison to healthy controls [85]. This additionally indicates a familiar risk for the schizophrenia susceptibility. The investigation of unaffected first-degree relatives of schizophrenic patients is of great interest since it might draw attention to genetic or environmental factors which increase the susceptibility to this disease. As previously mentioned, individual hippocampal activity levels and connectivity measurements might be useful for therapy options and treatment responses, in addition to the volumetric data and cerebellar blood volume levels. The following figure gives an overview on the most common functional alterations in the hippocampus of schizophrenia patients (Figure 2).

4. Genetic and Environmental Risk Factors Induce Hippocampal Deviations

Although morphological abnormalities of the hippocampus in schizophrenia patients and schizophrenia animal models are well-known, it is still unclear as to how these manifest. A genetic predisposition seems to be a plausible explanation since studies proved a higher risk for the disease manifestation in twin studies [86,87]. Furthermore, the above-mentioned incomplete hippocampal inversion patterns of schizophrenic patients show a high level of heritability [46].
Modern transcriptome techniques were used to investigate the hippocampus of postmortem schizophrenia patients with regard to the up- and downregulation of specific genes. Interestingly, an extensive hippocampal transcriptome-analysis was performed in human schizophrenia patients and revealed subfield-specific dysregulations of schizophrenia-associated genes [88]. Here, an enrichment of excitatory neuronal and synaptic genes (e.g., LMBRD2, MAL2, DLG3, MAP2, VDAC1) was detected in the so-called CA3-M2 module via weighted gene co-expression analysis (WGCNA). The results indicate that the hippocampal CA3-region is genetically affected and possibly contributes to the hippocampal hyperactivity which is frequently described in schizophrenic patients. In addition, another identified module of this study, the so-called CA3-M3 module, included genes associated with microglia (e.g., CSF1R, DOCK2, SYK, APBB1IP), autism (CUL3, GRIA1, BICD1), and further neuron-specific genes (e.g., SLC8A1, CNRIP1, KCNMB2). Importantly, this study also considered the impact of an antipsychotic treatment on the gene expression levels in the hippocampus and observed that 80 genes in the dentate gyrus, 351 genes in the CA3-region and 188 genes in the CA1-region were differentially expressed between treated and untreated patients [88]. Based on this observation, future studies should take into consideration that treatment with antipsychotics clearly affects gene expression and might falsify the conclusions which are made based on the data. A further study precisely focused on the transcriptomic profiling of the granule cell layer in the human dentate gyrus of postmortem schizophrenia patients by laser-capture microdissection, followed by RNA-sequencing [89]. This study identified a different set of schizophrenia-associated genes which were not found in previous studies that utilized the complete hippocampus, including the calcium voltage-gated channel subunit alpha1C (CACNA1C) and the glutamate metabotropic receptor 3 (GRM3). Further transcriptome studies of past years have proven that altered gene expression levels in different brain regions of schizophrenia patients underline the genetic aspect of this disease [89,90,91,92,93,94]. The most recent of these analyses could interestingly show an altered expression of the γ-aminobutyric acid-A (GABA-A) receptor-subunits GABRA1, GABRA2 and GABRB3 in the dorsolateral prefrontal cortex and in the superior temporal gyrus [91,93]. These observations may increase interest on the role of GABA-A receptor-subunits and their role in schizophrenia. Further evidence of a genetic predisposition for hippocampal alterations is provided by the investigation of unaffected first-degree relatives of schizophrenia patients with distinctive hippocampal features, as previously mentioned [57,85].
Despite the genetic risk for the development of schizophrenia, there is growing evidence that environmental factors and inflammatory processes contribute to the manifestation of psychosis and hippocampal aberrations.
A prominent and well-known environmental risk factor is the experience of childhood trauma, which is highly linked to structural changes of the hippocampus in schizophrenia patients [95,96]. In a recently published study, a significant correlation between childhood trauma scores and volume changes across hippocampal subregions could be observed in a sex-dependent manner with significant effects experienced in women [97]. A similar connection between childhood trauma and a reduced hippocampal volume was observed in cases of major depression and posttraumatic stress disorder, with predominant deviations in the left hippocampus [98,99,100]. Furthermore, experiencing childhood maltreatment can induce a volume reduction of the hippocampal CA3-region, of the dentate gyrus and of the subiculum [101]. Here, an inhibition of the hippocampal neurogenesis caused by chronic stress and the release of adrenal steroids induced by the childhood trauma or maltreatment is discussed as a possible mechanism for the hippocampal abnormalities [102].
Interestingly, a current study revealed that the volume of the hippocampus and the amygdala—which are stress-sensitive structures—is affected by the grade of severity of adverse childhood experiences, especially during the periods of pre-adolescence and early adolescence [103]. In hippocampal lesion experiments with rhesus monkeys, it has been shown that early trauma induces effects similar to the symptoms of schizophrenic patients and an altered functional hippocampo-prefrontal network integrity [104,105]. Additional environmental factors that show an impact on the hippocampal morphology are maternal stress and maternal anxiety during mid-gestation, as initially mentioned in the first part of this review [48,49]. A currently published study analyzed the influence of maternal stress at different trimesters of the pregnancy and performed connectivity measurements of the hippocampus with further CNS structures [106]. Interestingly, this study could demonstrate that maternal distress in the third trimester of pregnancy was linked to a weaker hippocampal–cingulate cortex connectivity and to a stronger hippocampal–temporal lobe connectivity. Additionally, the same study revealed that increased cortisol levels in the second but not in the third trimester of pregnancy were associated with a weaker hippocampal–cingulate cortex connectivity and a stronger hippocampal–temporal lobe connectivity [106]. Therefore, the functional connectivity of the hippocampus can be negatively affected by stress, with varying effects depending on the point in time of the pregnancy when stressful periods occur. In studies with rhesus monkeys, it could be furthermore shown that prenatal stress significantly reduced the neurogenesis in the dentate gyrus of juvenile animals [107].
Based on this, genetic, as well as environmental factors seem to play an important role in the occurrence of hippocampal abnormalities in schizophrenia. This aspect might furthermore explain why some patients develop this neuropsychiatric disease without a familiar risk, and vice versa, why subjects do not develop psychosis even though they experienced trauma or similar negative experiences. Furthermore, cumulative effects of genetic and environmental factors may increase the severity of schizophrenia symptoms and impair the outcome of the disease.

5. Inflammatory Processes Contribute to Hippocampal Abnormalities

In past years, evidence has been raised that inflammation is another important key factor in the progression of schizophrenia. The presence of increased inflammatory pathways was proven through the detection of higher peripheral cytokine levels in schizophrenia patients [108]. Interestingly, inflammatory processes could be furthermore identified in the hippocampus of schizophrenic patients. Here, an extensive postmortem transcriptional profiling study revealed a robust enrichment of pro-inflammatory pathways in several parts of the CNS, including the prefrontal cortex, the hippocampus and the striatum [109]. The transcriptional profiling revealed an upregulation of signaling pathways associated with interleukin-6 (IL-6), signal transducer and activator of transcription 3 (Stat3), oncostatin and interferon, as well as a downregulation of signaling pathways associated with GABA-receptors, synaptic long-term potentiation and Ephrin-B [109]. Another study could detect an increase of inflammatory-related genes like interferon-induced transmembrane protein 1, 2 and 3 (IFITM1,2,3), apolipoprotein L1 (APOL1), adenosine receptor A2a (ADORA2A), insulin-like growth factor-binding protein 4 (IGFBP4) and a cluster of differentiation 163 (CD163) in the hippocampus of patients via mRNA-sequencing [110].
Since microglia are the resident immune cells of the CNS, the hypothesis arose that this cell type might contribute to the disease’s manifestation and progression by the release of inflammatory factors and an extensive synapse elimination [111]. Microglia were first described by Pio del Rio-Hortega in 1919 as the fourth element of the CNS [112]. During early embryonic development, microglia originate from erythromyeloid progenitors (EMPs) located in the extra-embryonic yolk sac and migrate into the CNS [113,114,115]. In the CNS, microglia form a stable population that undergoes slow proliferation and apoptosis. Nowadays, it is known that microglia contribute to the elimination of excessive synapses, to the dendritic maturation and to the postmitotic differentiation of neurons [116,117,118,119,120]. With regard to the role of the hippocampus for schizophrenia, microglia might be of greater interest because they show a regional heterogeneity with a higher density in the hippocampus compared to other regions of the CNS [121]. Furthermore, both neurons and microglia possess functional receptors for the released factors of the others [122,123,124,125,126,127,128]. Therefore, a bidirectional interaction between neurons and microglia exists as discussed by Szepesi and colleagues [129]. Recently published findings support this idea by demonstrating that microglia act as negative feedback regulators of neuronal activity and consequently protect the CNS from excessive activation [130]. A dysregulation of microglia might consequently increase the neuronal activity and serve as another possible explanation for the hyperactive state of the hippocampus in schizophrenia patients. On the other hand, a hyperactive neuronal network might vice versa affect the activation state of microglia and induce the release of inflammatory factors or matrix-degrading enzymes.
Indeed, several studies observed an increased number or a higher activation state of microglia in the hippocampus of schizophrenia patients [131,132]. Interestingly, the analysis of schizophrenia patient-derived microglia-like cells revealed a higher synapse engulfment and elimination on neuronal cells in vitro [133]. In this context, important studies identified the major histocompatibility complex (MHC), and especially the complement system, as an important mediator of synapse elimination in schizophrenia [134,135,136,137]. While a higher reactive state of microglia could be a plausible explanation for the hippocampal deviations, positron emission tomography (PET) studies of recent years that targeted the translocator 18 kDa protein (TSPO) did not find significant—or found only minor microglial—changes in recent-onset or established schizophrenia patients [138,139,140,141]. However, there is increasing doubt that TSPO is a reliable marker for activated microglia since it is not correlated with other microglial activation markers and furthermore not restricted to microglia [142]. In addition, further PET studies with variable markers could verify activated microglia in the CNS of schizophrenic patients [143,144]. Considering these conflicting observations, a carefully validated marker for activated microglia is highly desirable for future PET studies and investigations.
Notwithstanding, inflammation seems to be an important contributor to this neuropsychiatric disease. Therefore, several clinical studies considered this aspect and administered an additive treatment of patients with the neuroprotective and anti-inflammatory antibiotic minocycline. Surprisingly, opposing observations were made when an adjunctive treatment with minocycline was performed. While several studies proved beneficial effects for the negative and cognitive deficits, as well as for the pro-inflammatory cytokine-levels [145,146,147,148,149,150,151], other studies did not observe a benefit for schizophrenia patients [152,153]. These opposing results do not necessarily mean that minocycline has no beneficial effects. The point in time when minocycline is administered could be a critical aspect, as previously discussed by Kishimoto and colleagues [154]. Furthermore, the additive treatment with minocycline might be useful for a subgroup of schizophrenia patients that clearly show a rise of inflammatory pathways. Therefore, it could be useful to initially screen for inflammatory factors in blood samples before a minocycline treatment is planned. A summary on the previously mentioned risk factors is given in the following schematic depiction (Figure 3).
In addition to microglia, astrocytes are another glial cell type that might be of interest for the pathophysiology of schizophrenia. Here, various studies described region-specific differences either in the number of astrocytes or in the expression levels of astrocyte-specific markers like the glial fibrillary acidic protein (GFAP), the aldehyde dehydrogenase 1 L1 (ALDH1L1) or the glutamate transporter-1 (GLT-1) in postmortem tissue samples of schizophrenia patients [155,156,157]. Interestingly, a genome-wide association study identified six astrocyte gene sets that are strongly associated with schizophrenia, indicating a possible role of astrocytes for the disease’s manifestation and progression [158]. However, no significant differences were described concerning the properties of astrocytes in the hippocampus of schizophrenic patients. Here, the analysis of the astrocyte density in posterior hippocampal subregions of postmortem schizophrenia patients revealed no deviations [159]. In another postmortem study, a similar number of phosphorylated-GFAP (pGFAP) positive cells was observed in the hippocampus of healthy subjects and schizophrenic patients [160]. These observations are furthermore supported by a transcriptomic analysis that revealed no significantly altered expression profiles of astrocytes in subcortical regions, including the hippocampus [161]. Although astrocytes seem to be unaffected in the hippocampus of postmortem schizophrenia patients, their possible influence should not be underestimated since a strong impact of genetically modified astrocytes on hippocampal neurons could be shown. Here, the use of an astrocyte-specific mutant version of the disrupted in schizophrenia 1 gene (DISC1) significantly reduced the proliferation rate of neural progenitors and the dendrite growth of newborn neurons in the adult hippocampus [162]. These observations were furthermore associated with elevated levels of anxiety, attenuated social behaviors, and impaired hippocampus-dependent learning and memory [162]. Therefore, additional astrocyte-specific studies are necessary to fully clarify the role of this glial cell type for schizophrenia.

6. Structural and Functional Hippocampus Deviations in Animal Models for Schizophrenia

Different animal models were established to shed light on the mechanisms responsible for the pathophysiology of schizophrenia. As previously mentioned, prominent region-specific effects were observed in the human anterior hippocampus that corresponds to the ventral hippocampus in rodents. The structural organization of the hippocampus in mice and rats is very similar, as a recent flatmap study has shown [163]. Here, a spatial similarity was demonstrated by rescaled comparisons. Furthermore, a similar distribution of parvalbumin-positive interneurons in the murine and rat hippocampus was shown [164]. Nevertheless, it has been observed that there are more adult-born hippocampal neurons in rats than in mice that interestingly mature faster and are more strongly associated with animal behavior [165]. On a functional level, the hippocampal formations of mice and rats show similar macroscopic physiological patterns with a similar spatial and regional distribution of extracellular voltage changes, frequencies of oscillations and correlations between local field activities and animal behaviors [166]. However, significant differences were observed with regard to the place cell activities in the murine and rat hippocampus [167]. These observations should be considered when newly generated data are interpreted and discussed in future studies. Although the majority of schizophrenia animal models are based on rodents, there is a growing interest in studies also utilizing the zebrafish as a model organism [168]. Since the brains of zebrafish do not contain a typical hippocampal formation, this organism will not be discussed in more detail within the scope of this review. However, a recently published study performed a phenotypical landscaping of schizophrenia-associated genes in the zebrafish and revealed an association of some risk genes with a lowered volume of the pallium, the homologue structure of the hippocampus in mammals [169,170]. The following section reviews the findings of established schizophrenia models, including (1) prenatal immune activation or prenatal neurotoxicity animal models, (2) studies with treatment-induced symptoms related to schizophrenia and (3) knockout models and current optogenetic approaches.
Maternal immune activation (MIA) models for schizophrenia are based on epidemiological studies that unraveled a link between prenatal infections during influenza pandemics and a higher occurrence of schizophrenia cases in the adult offspring [171,172,173,174,175,176]. Pregnant mice treated with a sublethal dose of the human influenza virus gave birth to animals with a significant upregulation of the schizophrenia and autism related gene Forkhead box P2 (Foxp2) in the hippocampus at P35 and P56 [177]. This was accompanied by an atrophy of different brain regions and an altered gene expression in the hippocampus of offspring animals [177]. Interestingly, additional studies showed that further pathogens can also increase the risk for schizophrenia in the offspring [178,179,180,181,182,183]. Here, infections during the second trimester of pregnancy were especially proven to increase schizophrenia risk. The varying trimester-dependent effects of the maternal immune challenge on CNS development and on behavioral abnormalities in the offspring were furthermore observed in animal models [184,185,186]. Since the placenta is impenetrable for most pathogens, the induction of the maternal immune response and the associated release of cytokines is highly suspected to be responsible for the previously mentioned effects [187,188,189,190]. Therefore, MIA models for schizophrenia frequently induce an activation of the maternal immune response by prenatal injections with immunostimulants like polyinosinic–polycytidylic acid (Poly I:C) or lipopolysaccharide (LPS). The most common cognitive and behavioral abnormalities in these models were observed for parameters like prepulse inhibition [191,192,193,194,195,196,197], latent inhibition [195,198] and social interaction [193,199,200]. In rhesus monkeys, a Poly I:C-induced maternal immune challenge increased repetitive behavior patterns and reduced communication, as well as social interactions in the offspring [201]. Furthermore, a smaller average soma size of neurons, which was, however, not significant, could be observed in a prenatal maternal immune activation study on rhesus monkeys [202].
Interestingly, the specific analysis of the hippocampus in prenatal immune activation models revealed structural and functional abnormalities which share similarities with the hippocampal deviations in schizophrenic patients. The activation of the maternal immune system via the administration of Poly I:C at gestation day 15.5 in pregnant mice induced a significantly reduced hippocampal volume, a decreased number of parvalbumin-positive interneurons and a lowered synaptic inhibition in mature dentate gyrus neurons of three-month-old male offspring animals [203]. Furthermore, the induction of a maternal immune challenge with Poly I:C decreased the serotonin and taurine levels in the hippocampus of offspring animals and affected neurotransmitter levels in other parts of the CNS [204]. On a functional level, the treatment of pregnant rat dams with LPS at embryonic day 15 and 16 led to smaller evoked field excitatory postsynaptic potentials (fEPSPs), but to a higher intrinsic excitability of CA1 pyramidal neurons in hippocampal slices of 20–25 day-old offspring animals [205]. Similarly, a reduced firing frequency and increased amplitudes of miniature excitatory postsynaptic currents (mEPSPs) in the hippocampal CA1-region of mice derived from Poly I:C- treated dams could be observed in hippocampal slices [206]. In addition, dissociated hippocampal neurons isolated from embryonic mice of Poly I:C-treated dams developed a network hyperactivity and disruptions of perineuronal nets (PNNs), as well as reduced soma areas of PNN-wearing neurons in vitro [207]. PNNs are a specialized and condensed form of the extracellular matrix (ECM) that stabilize synaptic contacts but prevent the formation of new connections [208]. Finally, a study that induced an immune challenge in the early postnatal phase instead of the prenatal phase could observe hippocampal long-term effects and an impairment of learning and memory [209].
Based on these observations, different studies addressed the question of whether or not the effects which were induced by a maternal immune activation can be prevented or ameliorated. Here, beneficial results were achieved when offspring animals received a treatment with minocycline [210,211], different neuroleptics [212] or IL-6-blocking antibodies [195]. Anti-inflammatory agents might be of great interest since a microglial activation could be shown in a Poly I:C mouse model for schizophrenia [213]. Regarding the role of IL-6 for the structural integrity of the hippocampus, a recently published study could demonstrate that there is a correlation between increased levels of IL-6 and a decrease of the total hippocampal volume [214]. Additionally, it has been shown that IL-6 can pass the placenta more efficiently during the mid-gestation period than during the late-gestation period and induce the release of fetal stress hormones [215]. A currently published study analyzed if the effects of a prenatal maternal immune activation can be enhanced by an environmental enriched housing during the experimental procedures [216]. This study provided evidence that the expression of the stress-associated markers oxytocin receptor (Oxtr), corticotropin-releasing hormone (Crh) and nuclear receptor subfamily 3 group C member 1 (Nr3c1) was significantly increased in the ventral hippocampus of MIA offspring animals. Regarding the oxytocin receptors in the hippocampus, it has been shown that neonatal stress negatively affects the development of this system in rats [217]. Furthermore, increased levels of corticosterone were identified in plasma specimens in offspring animals, indicating a dysregulation of the hypothalamic–pituitary–adrenal (HPA)-axis [216]. The housing in an environmentally enriched surrounding reversed the MIA-induced effects in the offspring [216]. In conclusion, different independently performed studies observed structural, as well as functional deviations of the hippocampus in the offspring of dams exposed to maternal immune activation. This aspect and new preventive therapies might be of great interest with regard to the COVID-19 pandemic and its possible consequences regarding neuropsychiatric disorders in the future.
In contrast to maternal immune activation models, the prenatal methylazoxymethanol acetate (MAM)-model directly induces neurotoxic effects and is consequently a developmental disruption model for schizophrenia [218]. Interestingly, studies utilizing the MAM-model revealed a reduction of the hippocampal volume in the offspring with schizophrenia-related behavioral abnormalities [219,220]. Proteomic, as well as metabolomic analyses produced strong evidence that a treatment of rats with MAM at embryonic day 17 is responsible for deficits primarily in the hippocampus, affecting the glutamatergic neurotransmission [221]. Here, electrophysiological recordings of the CA1-region in the hippocampus revealed a significant reduction of fEPSPs and a deficit of the AMPA-receptor-mediated synaptic transmission. Furthermore, animals that received a prenatal MAM-treatment developed hyperactivity in the ventral hippocampus in later stages of development [222,223]. On a cellular level, a previously published study could identify a loss of parvalbumin-positive interneurons in combination with a reduced gamma-band response [224]. Interestingly, an additional published study showed that a MAM-treatment at gestational day 16 and 17 induced a reduction of the hippocampal volume, an impaired contextual fear memory and a significantly reduced long-term potentiation (LTP) in the synapses of the CA1-region in mice [225]. Furthermore, this study focused on sex-specific differences and could demonstrate that male offspring mice that previously suffered from a MAM-treatment at gestation day 16 developed a decreased parvalbumin-expression in the hippocampus and deficits in the delayed alternation task. The loss of parvalbumin-positive interneurons in the hippocampus of MAM-treated animals was also observed in an additional study and could, interestingly, be prevented by a peripubertal treatment with diazepam [226]. The peripubertal treatment with diazepam prevented the hyperresponsivity of the dopamine system in the MAM-model [227]. As previously described for prenatal immune activation models, the housing of animals in an environmentally enriched surrounding within a prepubertal time window prevented dopamine dysregulation and hippocampal hyperactivity in a murine MAM-model [228]. The experiments that utilized an enhanced environmental enrichment might be interesting for new preventive therapeutical approaches.
In the ketamine model for schizophrenia, acute psychosis is induced by injections in rodents. Here, ketamine acts as a N-methyl-D-aspartate receptor (NMDAR) antagonist and induces behavioral abnormalities which are similar to the positive and negative symptoms of schizophrenia patients [229,230,231]. Similar deviations were observed when other NMDAR antagonists, like phencyclidine or MK-801 were given, as well as in cases of autoimmune anti-NMDA receptor encephalitis [232,233,234,235]. A treatment of animals with ketamine showed strong effects on the hippocampal function and structure. Here, a repeated exposure to ketamine shifted the hippocampus towards a hypermetabolic basal state with an accompanying atrophy and a disruption of parvalbumin-expressing interneurons [72]. Interestingly, a currently published study analyzed hippocampal–prefrontal cortex (HPC-PFC) local-field potentials before and after the administration of ketamine [236]. The results showed that ketamine promotes an abnormal delta-high-gamma cross-frequency-coupling in the PFC and a rise of responses in the hippocampus. Furthermore, the authors could observe that LTP-induction before the ketamine treatment prevented the increase of the gamma amplitude [236]. Therefore, a rise of the glutamatergic synaptic efficiency is suspected to enhance cognitive impairments in animal models for psychosis, as discussed by the authors. Furthermore, it has been described that a subclass-imbalance of parvalbumin-expressing GABAergic interneurons occurs in the ketamine mouse model with disturbances of PNNs [237]. The disruption of PNNs in the ketamine model for schizophrenia was supported by a high-resolution confocal microscopic study that observed a significantly altered PNN fine-structure in a ketamine rat model [238]. In this study, ketamine-treated animals developed a significantly reduced mean area of PNN-units and so-called ECM-enriched vertices [238]. Although this study focused on the deep layers IV and V of the prelimbic cortex, similar PNN alterations might also occur in the hippocampus since it is a highly ketamine-affected structure. The role of parvalbumin-positive PNN-carrying neurons will be considered in more detail in a later paragraph of this review. Besides the manipulation of the glutamatergic synapse transmission, both electrophysiological or other pharmacological NMDAR-stimulation of the hippocampus induced a disruption of the prepulse inhibition in rats [239,240]. As already indicated previously, an impairment of prepulse inhibition can be frequently seen in schizophrenic patients, which is caused by sensory gating deficits [241]. Here, the activity level of the ventral hippocampus seems to play an important role in this deviation [242].
Studies of recent years that utilized knockout models and optogenetic manipulations allowed for the analysis of subfield-specific, as well as receptor-specific mechanisms which possibly contribute to the schizophrenia-related hippocampal alterations.
Interestingly, a subfield-specific GluN1-knockout mouse model with a disease-like disruption specifically in the dentate gyrus of the hippocampus showed several functional abnormalities [243]. Here, a psychosis-like behavior could be observed which was accompanied by a CA3-specific hyperactivity and an increased glutamate transmission at synapses between mossy fibers and CA3 neurons. Furthermore, an increased number of cFos-activated pyramidal neurons in the CA3-region could be seen [243]. Another recently published study revealed that an early ablation of corticolimbic NMDA receptors on interneurons induces a significantly reduced functional connectivity between the ventral hippocampus and the prefrontal cortex before and after adolescence [244]. These observations highly underline the important role of glutamate receptor subunits for psychosis, as well as for schizophrenia. Other studies investigated cyclin D2 mutant mice (Ccnd2) and observed deficits in parvalbumin-positive interneurons and a raised hippocampal excitability, including increased metabolic activity [245]. This study also described cognitive impairments when cyclin D2 was missing.
As previously indicated, optogenetics can be used for a region-specific manipulation of the neuronal activity and were discussed as a useful tool for the analysis of the disease-underlying mechanisms [246]. Interestingly, the optogenetic activation of excitatory neurons in the ventral hippocampus of mice induced a hyperlocomotion which is known as a rodent correlate of the positive symptoms of schizophrenia [247]. The optogenetic activation of the ventral hippocampus furthermore impaired performance on the spatial novelty preference test of short-term memory, indicating a cognitive impairment [247]. Conversely, the optogenetic inhibition of ventral hippocampal neurons enhanced motor learning dysfunction in a phencyclidine rat model for schizophrenia [248]. A currently published study utilized excitatory chemogenetic constructs in order to specifically induce a hyperactivation of mossy cells in the ventral dentate gyrus that are targeting granule cells and interneurons in the dorsal part of the dentate gyrus [249]. Here, the authors could show that a chemogenetic activation of the ventral mossy cells increased the activity of dorsal granule cells and significantly impaired the test performance in an object location memory task [249]. Considering these observations, the specific optogenetic manipulation of neuronal circuits emerges as a promising method which will unravel new aspects and important circuits of this neuropsychiatric disease in future studies.

7. Interneuron-Abnormalities in the Hippocampus of Schizophrenic Patients and Schizophrenia Animal Models

With regard to the electrophysiological hyperactivity, it has been proposed that parvalbumin-positive interneurons are hypofunctional, and this consequently results in an overstimulation of excitatory neurons [250]. Possibly, this idea might be adopted for the interneurons in the hippocampus. Interestingly, studies on postmortem schizophrenia patients unraveled a reduction of somatostatin- and parvalbumin-positive interneurons in the hippocampus, which was furthermore confirmed by qRT-PCR analysis [251,252,253]. As previously mentioned, a hippocampal transcriptome analysis also observed a significant reduction of GABAergic signaling pathways in schizophrenic patients [109]. Somatostatin- and parvalbumin-positive interneurons fulfill an important function by contributing to rhythmic gamma-oscillations in the CNS [254,255,256,257]. In schizophrenia patients, disturbances of these gamma-oscillations were frequently described by several independent studies [258,259,260,261,262]. Therefore, disruptions of interneurons might be the possible cause and inducer of the hippocampal hyperactivity in schizophrenic patients. The inhibition of the parvalbumin- and GAD65- positive interneuron population induced behavioral deficiencies like an impaired prepulse inhibition and startle reactivity [263]. With regard to therapeutical strategies, a recently published study investigated the gamma oscillations in schizophrenia patients and discussed them as a possible biomarker for the efficacy of targeted cognitive training (TCT) [264]. Here, the baseline of the gamma power predicted cognitive benefits after a full course of TCT. Furthermore, a change of the gamma power after 1 h of TCT predicted the enhancement of both positive and negative symptoms [264]. Interestingly, exercise in the form of treadmill running also had positive effects on the interneuron activation-dependent adult hippocampal neurogenesis in a MK801-induced schizophrenia-like animal model, as a current study demonstrated [265]. Additional interesting approaches of recent years focused on interneuron or interneuron precursor cell transplants in the hippocampus in different animal models for schizophrenia. Astonishingly, transplants consisting of stem-cell derived interneurons reduced the hippocampal hyperactivity and normalized the aberrant characteristics of dopaminergic neurons in a MAM-rat model for schizophrenia [266,267]. Similar results were achieved with interneuron-precursor transplants in adult Ccnd2 knockout mice [268]. Here, Gilani and colleagues observed that mice developed a lowered metabolic hippocampal hyperactivity and an improved context-depending learning and memory up to six months after transplantation of the interneuron precursors [268]. Interestingly, the beneficial effects of interneuron transplants were also observed in an animal model for autism, as a currently published study could show [269]. Here, the social behavioral deficits were rescued by the transplantation of interneurons [269]. Furthermore, a prenatal immune activation model exhibited a reduction of parvalbumin-positive interneurons and an impaired GABAergic transmission in the dentate gyrus in offspring animals [270]. Here, a treatment with minocycline or with the microglia-specific arginase-1 prevented this effect. Interestingly, another current study revealed that a regular physical exercise significantly increased the neurogenesis of parvalbumin-positive interneurons with ameliorating effects for schizophrenia-like phenotypes [265]. These results show that targeting and manipulating interneurons in the hippocampus of schizophrenic patients could be a promising way to weaken the symptoms of this disease. Furthermore, this aspect might be especially interesting for cognitive impairments of schizophrenic patients which highly disturb life quality. A summary of beneficial experimental approaches with regard to hippocampal hyperactivity is given in Figure 4.
A subclass of parvalbumin-positive interneurons is covered by a dense and lattice-like structure of the ECM around the soma and proximal dendrites. These structures are the so-called perineuronal nets which were first described by Camillo Golgi more than a century ago [273]. Interestingly, perineuronal net-wearing neurons are represented in the hippocampus and can be observed, inter alia, in the CA1-, CA2- and CA3-subfield. Nowadays, several studies observed an important involvement of perineuronal nets for the regulation of the synaptic plasticity [271,274,275,276,277,278,279]. Furthermore, PNNs unfold a neuroprotective effect on parvalbumin-positive interneurons and fulfill ion-sorting and buffering functions [280,281,282,283]. Concomitant with the previously mentioned abnormalities of parvalbumin-positive interneurons, different studies showed disruptions of PNNs in schizophrenia patients. Here, postmortem analyses of schizophrenic patients especially revealed either a significant reduction in the amount of PNN-wearing neurons or in the PNN-staining intensity in the prefrontal cortex [284,285], in the amygdala [286,287] and in the inferior colliculus [288]. Contrarily, a currently published study did not find disturbances of parvalbumin-positive interneurons or PNNs in postmortem tissue of schizophrenia patients, although a negative correlation between the PNN density and the occurrence of psychosis could be shown [289]. Most of the human postmortem studies and animal models focused on PNN abnormalities in the cortex and unfortunately less is known about PNN alterations in the hippocampus of schizophrenic patients or schizophrenia animal models. Based on this, there are extensive reviews which focus on the role of perineuronal nets in schizophrenia [290,291,292,293,294]. Nevertheless, a currently published study utilized a double-hit model with a perinatal injection of the NMDAR antagonist MK801, combined with a post-weaning social isolation as an early-life stress event [295]. Interestingly, the authors could observe a region-specific reduction in the number of PNNs and PNN-wearing parvalbumin-positive neurons especially in the CA1-subfield of the hippocampus [295]. In addition, the presence of atypical perineuronal nets in the CA2-subfield was observed to be associated with a dysfunction of the social memory [296]. In this perspective, the targeting of perineuronal nets might be an interesting approach with regard to the functional deficits of the hippocampus in schizophrenia and schizophrenia animal models. The manipulation of perineuronal nets in the CNS was frequently performed by the stereotactic administration of bacterially-derived chondroitinase ABC (ChABC) or hyaluronidase in rodents [271,274,275,296,297,298,299,300]. Intriguingly, the experimental digestion of PNNs by ChABC-injections in the ventral hippocampus raised the general hippocampal activity, increased the activity of dopaminergic neurons and augmented the locomotor response to amphetamine indicating possible implications for schizophrenia [301]. With a focus on the hippocampus, as well as on cultured hippocampal neurons, electrophysiological alterations were observed when perineuronal nets were disrupted in vivo and in vitro [271,302,303,304,305,306]. Interestingly, there is a growing evidence that microglia are involved in the maintenance of perineuronal nets [307]. In disease models for Alzheimer’s and Huntington’s, a contribution of microglia to the loss of perineuronal nets could be proven [308,309]. Furthermore, an in vitro study revealed a disruption of perineuronal nets in cultured hippocampal neurons after an incubation with microglia conditioned medium which was accompanied by altered structural synapse numbers and electrophysiological network properties [305]. In contrast, a depletion of microglia induced more dense perineuronal nets [310]. As previously introduced, microglia and inflammatory processes are suspected to play an important role in schizophrenia. Therefore, the targeting of microglia might be an interesting option for reversing perineuronal net disturbances and restoring a synaptic balance.
While the procedure of PNN-digestion can be performed in animals, it is problematic for a therapeutical strategy in humans. However, a currently published study achieved a modulation of perineuronal nets by the oral administration of 4-methylumbelliferone (4-MU), an inhibitor of the hyaluronic acid synthesis [311]. Here, a reduction of perineuronal nets was accompanied by an enhancement of the memory retention when mice were treated for 6 months with 4-MU [311]. This approach might be interesting since it allows for the manipulation of perineuronal nets in the CNS by an oral treatment. However, in schizophrenia animal models, an increase and a strengthening of the PNNs is of greater interest than a disruption. Here, another mild approach could allow for new therapeutical strategies. Several studies could show that an enhanced environmental enrichment increased the density of perineuronal nets in the developing striatum [272], in the prefrontal cortex [312] and importantly in the CA2-subfield of the hippocampus [271]. As previously described in this review, current studies revealed that the housing in an environmentally enriched surrounding improved the deficits in different animal models for schizophrenia [216,228]. Therefore, the environmental enrichment could be used as a non-invasive method for the manipulation of perineuronal nets in regions of the CNS that are involved in the pathology of schizophrenia and allow for new therapeutic strategies.
Based on this, interneurons especially play an important role for the functional deviations in the hippocampus of schizophrenic patients. Future studies might combine schizophrenia animal models with PNN-modulating agents or environmental enriched strategies to shed light on enhancing, as well as worsening effects for cognitive symptoms of schizophrenia or other neuropsychiatric, as well as neurological disorders.

8. Conclusions

In summary, hippocampal deviations are a common hallmark of the severe neuropsychiatric disorder schizophrenia. Although a majority of schizophrenia studies focus on the prefrontal cortex and the striatum in patients, hippocampal formation should not be underestimated since it contributes to higher cognitive functions. While former experiments have already unraveled a volume reduction of the hippocampus in schizophrenic patients, novel techniques allow for region-specific, morphological and functional analyses that provide a better overview on various hippocampal parameters in patients. Furthermore, hippocampal deviations, similar to those of schizophrenic patients, were observed in different animal models for schizophrenia. In recent years, several interesting experimental approaches were utilized in these models that targeted the hippocampus especially on a functional level. Here, a reduction of the hyperactivity went along with an amelioration of the behavioral and cognitive deficiencies in these models. In patients, hippocampal parameters could give valuable information about the disease’s progress, the risk of an immediate psychotic phase and the efficacy of different therapeutical strategies.

Author Contributions

Conceptualization, D.W. and A.F.; writing—original draft preparation, D.W.; writing—review and editing, A.F. and G.J.; visualization, D.W.; supervision, D.W., A.F. and G.J.; funding acquisition, A.F. and G.J. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by the German Research Foundation (DFG FA 159/22-1, grant number 290189690). We acknowledge support from the DFG Open Access Publication Funds of the Ruhr-Universität Bochum.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Adityanjee; Aderibigbe, Y.A.; Theodoridis, D.; Vieweg, V.R. Dementia praecox to schizophrenia: The first 100 years. Psychiatry Clin. Neurosci. 1999, 53, 437–448. [Google Scholar] [CrossRef] [PubMed]
  2. Moskowitz, A.; Heim, G. Eugen Bleuler’s Dementia praecox or the group of schizophrenias (1911): A centenary appreciation and reconsideration. Schizophr. Bull. 2011, 37, 471–479. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Saha, S.; Chant, D.; Welham, J.; McGrath, J. A systematic review of the prevalence of schizophrenia. PLoS Med. 2005, 2, e141. [Google Scholar] [CrossRef] [PubMed]
  4. Hjorthøj, C.; Stürup, A.E.; McGrath, J.J.; Nordentoft, M. Years of potential life lost and life expectancy in schizophrenia: A systematic review and meta-analysis. Lancet Psychiatry 2017, 4, 295–301. [Google Scholar] [CrossRef]
  5. McCutcheon, R.A.; Reis Marques, T.; Howes, O.D. Schizophrenia-An Overview. JAMA Psychiatry 2020, 77, 201–210. [Google Scholar] [CrossRef]
  6. Murray, R.M.; Lewis, S.W. Is schizophrenia a neurodevelopmental disorder? Br. Med. J. 1988, 296, 63. [Google Scholar] [CrossRef] [Green Version]
  7. Erlenmeyer-Kimling, L.; Rock, D.; Roberts, S.A.; Janal, M.; Kestenbaum, C.; Cornblatt, B.; Adamo, U.H.; Gottesman, I.I. Attention, memory, and motor skills as childhood predictors of schizophrenia-related psychoses: The New York High-Risk Project. Am. J. Psychiatry 2000, 157, 1416–1422. [Google Scholar] [CrossRef]
  8. Picchioni, M.M.; Murray, R.M. Schizophrenia. BMJ 2007, 335, 91–95. [Google Scholar] [CrossRef]
  9. Correll, C.U.; Schooler, N.R. Negative Symptoms in Schizophrenia: A Review and Clinical Guide for Recognition, Assessment, and Treatment. Neuropsychiatr. Dis. Treat. 2020, 16, 519–534. [Google Scholar] [CrossRef] [Green Version]
  10. Bowie, C.R.; Harvey, P.D. Cognitive deficits and functional outcome in schizophrenia. Neuropsychiatr. Dis. Treat. 2006, 2, 531–536. [Google Scholar] [CrossRef] [Green Version]
  11. Van Haren, N.E.M.; Schnack, H.G.; Cahn, W.; van den Heuvel, M.P.; Lepage, C.; Collins, L.; Evans, A.C.; Hulshoff Pol, H.E.; Kahn, R.S. Changes in cortical thickness during the course of illness in schizophrenia. Arch. Gen. Psychiatry 2011, 68, 871–880. [Google Scholar] [CrossRef] [PubMed]
  12. Nesvåg, R.; Lawyer, G.; Varnäs, K.; Fjell, A.M.; Walhovd, K.B.; Frigessi, A.; Jönsson, E.G.; Agartz, I. Regional thinning of the cerebral cortex in schizophrenia: Effects of diagnosis, age and antipsychotic medication. Schizophr. Res. 2008, 98, 16–28. [Google Scholar] [CrossRef] [PubMed]
  13. Gaser, C.; Nenadic, I.; Buchsbaum, B.R.; Hazlett, E.A.; Buchsbaum, M.S. Ventricular enlargement in schizophrenia related to volume reduction of the thalamus, striatum, and superior temporal cortex. Am. J. Psychiatry 2004, 161, 154–156. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Andreasen, N.C.; Olsen, S.A.; Dennert, J.W.; Smith, M.R. Ventricular enlargement in schizophrenia: Relationship to positive and negative symptoms. Am. J. Psychiatry 1982, 139, 297–302. [Google Scholar] [CrossRef] [PubMed]
  15. Horga, G.; Bernacer, J.; Dusi, N.; Entis, J.; Chu, K.; Hazlett, E.A.; Haznedar, M.M.; Kemether, E.; Byne, W.; Buchsbaum, M.S. Correlations between ventricular enlargement and gray and white matter volumes of cortex, thalamus, striatum, and internal capsule in schizophrenia. Eur. Arch. Psychiatry Clin. Neurosci. 2011, 261, 467–476. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Narr, K.L.; Bilder, R.M.; Woods, R.P.; Thompson, P.M.; Szeszko, P.; Robinson, D.; Ballmaier, M.; Messenger, B.; Wang, Y.; Toga, A.W. Regional specificity of cerebrospinal fluid abnormalities in first episode schizophrenia. Psychiatry Res. 2006, 146, 21–33. [Google Scholar] [CrossRef]
  17. Bird, C.M.; Burgess, N. The hippocampus and memory: Insights from spatial processing. Nat. Rev. Neurosci. 2008, 9, 182–194. [Google Scholar] [CrossRef]
  18. Bogerts, B.; Falkai, P.; Haupts, M.; Greve, B.; Ernst, S.; Tapernon-Franz, U.; Heinzmann, U. Post-mortem volume measurements of limbic system and basal ganglia structures in chronic schizophrenics. Initial results from a new brain collection. Schizophr. Res. 1990, 3, 295–301. [Google Scholar] [CrossRef]
  19. Bogerts, B.; Lieberman, J.A.; Ashtari, M.; Bilder, R.M.; Degreef, G.; Lerner, G.; Johns, C.; Masiar, S. Hippocampus-amygdala volumes and psychopathology in chronic schizophrenia. Biol. Psychiatry 1993, 33, 236–246. [Google Scholar] [CrossRef]
  20. Szeszko, P.R.; Goldberg, E.; Gunduz-Bruce, H.; Ashtari, M.; Robinson, D.; Malhotra, A.K.; Lencz, T.; Bates, J.; Crandall, D.T.; Kane, J.M.; et al. Smaller anterior hippocampal formation volume in antipsychotic-naive patients with first-episode schizophrenia. Am. J. Psychiatry 2003, 160, 2190–2197. [Google Scholar] [CrossRef]
  21. Velakoulis, D.; Pantelis, C.; McGorry, P.D.; Dudgeon, P.; Brewer, W.; Cook, M.; Desmond, P.; Bridle, N.; Tierney, P.; Murrie, V.; et al. Hippocampal volume in first-episode psychoses and chronic schizophrenia: A high-resolution magnetic resonance imaging study. Arch. Gen. Psychiatry 1999, 56, 133–141. [Google Scholar] [CrossRef] [PubMed]
  22. Walter, A.; Studerus, E.; Smieskova, R.; Kuster, P.; Aston, J.; Lang, U.E.; Radue, E.-W.; Riecher-Rössler, A.; Borgwardt, S. Hippocampal volume in subjects at high risk of psychosis: A longitudinal MRI study. Schizophr. Res. 2012, 142, 217–222. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Lawrie, S.M.; Whalley, H.; Kestelman, J.N.; Abukmeil, S.S.; Byrne, M.; Hodges, A.; Rimmington, J.E.; Best, J.J.; Owens, D.G.; Johnstone, E.C. Magnetic resonance imaging of brain in people at high risk of developing schizophrenia. Lancet 1999, 353, 30–33. [Google Scholar] [CrossRef]
  24. Shenton, M.E.; Dickey, C.C.; Frumin, M.; McCarley, R.W. A review of MRI findings in schizophrenia. Schizophr. Res. 2001, 49, 1–52. [Google Scholar] [CrossRef] [Green Version]
  25. Roeske, M.J.; Konradi, C.; Heckers, S.; Lewis, A.S. Hippocampal volume and hippocampal neuron density, number and size in schizophrenia: A systematic review and meta-analysis of postmortem studies. Mol. Psychiatry 2021, 26, 3524–3535. [Google Scholar] [CrossRef] [PubMed]
  26. Matosin, N.; Fernandez-Enright, F.; Lum, J.S.; Engel, M.; Andrews, J.L.; Gassen, N.C.; Wagner, K.V.; Schmidt, M.V.; Newell, K.A. Molecular evidence of synaptic pathology in the CA1 region in schizophrenia. NPJ Schizophr. 2016, 2, 16022. [Google Scholar] [CrossRef] [Green Version]
  27. Li, W.; Ghose, S.; Gleason, K.; Begovic, A.; Perez, J.; Bartko, J.; Russo, S.; Wagner, A.D.; Selemon, L.; Tamminga, C.A. Synaptic proteins in the hippocampus indicative of increased neuronal activity in CA3 in schizophrenia. Am. J. Psychiatry 2015, 172, 373–382. [Google Scholar] [CrossRef] [Green Version]
  28. Toro, C.; Deakin, J.F.W. NMDA receptor subunit NRI and postsynaptic protein PSD-95 in hippocampus and orbitofrontal cortex in schizophrenia and mood disorder. Schizophr. Res. 2005, 80, 323–330. [Google Scholar] [CrossRef]
  29. Avery, S.N.; Rogers, B.P.; Heckers, S. Hippocampal Network Modularity Is Associated with Relational Memory Dysfunction in Schizophrenia. Biol. Psychiatry Cogn. Neurosci. Neuroimaging 2018, 3, 423–432. [Google Scholar] [CrossRef]
  30. Du, Y.; Hao, H.; Wang, S.; Pearlson, G.D.; Calhoun, V.D. Identifying commonality and specificity across psychosis sub-groups via classification based on features from dynamic connectivity analysis. Neuroimage Clin. 2020, 27, 102284. [Google Scholar] [CrossRef]
  31. Edmiston, E.K.; Song, Y.; Chang, M.; Yin, Z.; Zhou, Q.; Zhou, Y.; Jiang, X.; Wei, S.; Xu, K.; Tang, Y.; et al. Hippocampal Resting State Functional Connectivity in Patients with Schizophrenia and Unaffected Family Members. Front. Psychiatry 2020, 11, 278. [Google Scholar] [CrossRef] [PubMed]
  32. Nelson, E.A.; Kraguljac, N.V.; Maximo, J.O.; Briend, F.; Armstrong, W.; Hoef, L.W.V.; Johnson, V.; Lahti, A.C. Hippocampal Dysconnectivity and Altered Glutamatergic Modulation of the Default Mode Network: A Combined Resting-State Connectivity and Magnetic Resonance Spectroscopy Study in Schizophrenia. Biol. Psychiatry Cogn. Neurosci. Neuroimaging 2022, 7, 108–118. [Google Scholar] [CrossRef] [PubMed]
  33. Harrisberger, F.; Buechler, R.; Smieskova, R.; Lenz, C.; Walter, A.; Egloff, L.; Bendfeldt, K.; Simon, A.E.; Wotruba, D.; Theodoridou, A.; et al. Alterations in the hippocampus and thalamus in individuals at high risk for psychosis. NPJ Schizophr. 2016, 2, 16033. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Lewis, S.W.; Murray, R.M. Obstetric complications, neurodevelopmental deviance, and risk of schizophrenia. J. Psychiatr. Res. 1987, 21, 413–421. [Google Scholar] [CrossRef]
  35. Waddington, J.L.; Torrey, E.F.; Crow, T.J.; Hirsch, S.R. Schizophrenia, neurodevelopment, and disease. The Fifth Biannual Winter Workshop on Schizophrenia, Badgastein, Austria, January 28 to February 3, 1990. Arch. Gen. Psychiatry 1991, 48, 271–273. [Google Scholar] [CrossRef]
  36. Rapoport, J.L.; Addington, A.M.; Frangou, S.; Psych, M.R.C. The neurodevelopmental model of schizophrenia: Update 2005. Mol. Psychiatry 2005, 10, 434–449. [Google Scholar] [CrossRef]
  37. Fatemi, S.H.; Folsom, T.D. The neurodevelopmental hypothesis of schizophrenia, revisited. Schizophr. Bull. 2009, 35, 528–548. [Google Scholar] [CrossRef]
  38. Roeske, M.J.; McHugo, M.; Vandekar, S.; Blackford, J.U.; Woodward, N.D.; Heckers, S. Incomplete hippocampal inversion in schizophrenia: Prevalence, severity, and impact on hippocampal structure. Mol. Psychiatry 2021, 26, 5407–5416. [Google Scholar] [CrossRef]
  39. Cury, C.; Toro, R.; Cohen, F.; Fischer, C.; Mhaya, A.; Samper-González, J.; Hasboun, D.; Mangin, J.-F.; Banaschewski, T.; Bokde, A.L.W.; et al. Incomplete Hippocampal Inversion: A Comprehensive MRI Study of Over 2000 Subjects. Front. Neuroanat. 2015, 9, 160. [Google Scholar] [CrossRef]
  40. Bajic, D.; Wang, C.; Kumlien, E.; Mattsson, P.; Lundberg, S.; Eeg-Olofsson, O.; Raininko, R. Incomplete inversion of the hippocampus--a common developmental anomaly. Eur. Radiol. 2008, 18, 138–142. [Google Scholar] [CrossRef]
  41. Bajic, D.; Kumlien, E.; Mattsson, P.; Lundberg, S.; Wang, C.; Raininko, R. Incomplete hippocampal inversion-is there a relation to epilepsy? Eur. Radiol. 2009, 19, 2544–2550. [Google Scholar] [CrossRef] [PubMed]
  42. Cachia, A.; Cury, C.; Brunelin, J.; Plaze, M.; Delmaire, C.; Oppenheim, C.; Medjkane, F.; Thomas, P.; Jardri, R. Deviations in early hippocampus development contribute to visual hallucinations in schizophrenia. Transl. Psychiatry 2020, 10, 102. [Google Scholar] [CrossRef] [PubMed]
  43. Arnold, S.E.; Trojanowski, J.Q. Human fetal hippocampal development: I. Cytoarchitecture, myeloarchitecture, and neuronal morphologic features. J. Comp. Neurol. 1996, 367, 274–292. [Google Scholar] [CrossRef]
  44. Kier, E.L.; Kim, J.H.; Fulbright, R.K.; Bronen, R.A. Embryology of the human fetal hippocampus: MR imaging, anatomy, and histology. AJNR Am. J. Neuroradiol. 1997, 18, 525–532. [Google Scholar] [PubMed]
  45. Bajic, D.; Ewald, U.; Raininko, R. Hippocampal development at gestation weeks 23 to 36. An ultrasound study on preterm neonates. Neuroradiology 2010, 52, 489–494. [Google Scholar] [CrossRef]
  46. Cury, C.; Scelsi, M.A.; Toro, R.; Frouin, V.; Artiges, E.; Grigis, A.; Heinz, A.; Lemaître, H.; Martinot, J.-L.; Poline, J.-B.; et al. Genome wide association study of incomplete hippocampal inversion in adolescents. PLoS ONE 2020, 15, e0227355. [Google Scholar] [CrossRef] [Green Version]
  47. Allswede, D.M.; Cannon, T.D. Prenatal inflammation and risk for schizophrenia: A role for immune proteins in neurodevelopment. Dev. Psychopathol. 2018, 30, 1157–1178. [Google Scholar] [CrossRef]
  48. Buss, C.; Davis, E.P.; Muftuler, L.T.; Head, K.; Sandman, C.A. High pregnancy anxiety during mid-gestation is associated with decreased gray matter density in 6–9-year-old children. Psychoneuroendocrinology 2010, 35, 141–153. [Google Scholar] [CrossRef] [Green Version]
  49. Qiu, A.; Rifkin-Graboi, A.; Chen, H.; Chong, Y.-S.; Kwek, K.; Gluckman, P.D.; Fortier, M.V.; Meaney, M.J. Maternal anxiety and infants’ hippocampal development: Timing matters. Transl. Psychiatry 2013, 3, e306. [Google Scholar] [CrossRef] [Green Version]
  50. Thompson, D.K.; Adamson, C.; Roberts, G.; Faggian, N.; Wood, S.J.; Warfield, S.K.; Doyle, L.W.; Anderson, P.J.; Egan, G.F.; Inder, T.E. Hippocampal shape variations at term equivalent age in very preterm infants compared with term controls: Perinatal predictors and functional significance at age 7. Neuroimage 2013, 70, 278–287. [Google Scholar] [CrossRef] [Green Version]
  51. Nosarti, C.; Reichenberg, A.; Murray, R.M.; Cnattingius, S.; Lambe, M.P.; Yin, L.; MacCabe, J.; Rifkin, L.; Hultman, C.M. Preterm birth and psychiatric disorders in young adult life. Arch. Gen. Psychiatry 2012, 69, 610–617. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Vanes, L.D.; Murray, R.M.; Nosarti, C. Adult outcome of preterm birth: Implications for neurodevelopmental theories of psychosis. Schizophr. Res. 2021. [Google Scholar] [CrossRef] [PubMed]
  53. Rifkin, L.; Lewis, S.; Jones, P.; Toone, B.; Murray, R. Low birth weight and schizophrenia. Br. J. Psychiatry 1994, 165, 357–362. [Google Scholar] [CrossRef] [PubMed]
  54. Foerster, A.; Lewis, S.W.; Owen, M.J.; Murray, R.M. Low birth weight and a family history of schizophrenia predict poor premorbid functioning in psychosis. Schizophr. Res. 1991, 5, 13–20. [Google Scholar] [CrossRef]
  55. Narr, K.L.; Thompson, P.M.; Szeszko, P.; Robinson, D.; Jang, S.; Woods, R.P.; Kim, S.; Hayashi, K.M.; Asunction, D.; Toga, A.W.; et al. Regional specificity of hippocampal volume reductions in first-episode schizophrenia. Neuroimage 2004, 21, 1563–1575. [Google Scholar] [CrossRef]
  56. Nugent, T.F.; Herman, D.H.; Ordonez, A.; Greenstein, D.; Hayashi, K.M.; Lenane, M.; Clasen, L.; Jung, D.; Toga, A.W.; Giedd, J.N.; et al. Dynamic mapping of hippocampal development in childhood onset schizophrenia. Schizophr. Res. 2007, 90, 62–70. [Google Scholar] [CrossRef]
  57. Johnson, S.L.M.; Wang, L.; Alpert, K.I.; Greenstein, D.; Clasen, L.; Lalonde, F.; Miller, R.; Rapoport, J.; Gogtay, N. Hippocampal shape abnormalities of patients with childhood-onset schizophrenia and their unaffected siblings. J. Am. Acad. Child Adolesc. Psychiatry 2013, 52, 527–536.e2. [Google Scholar] [CrossRef] [Green Version]
  58. Hinney, B.; Walter, A.; Aghlmandi, S.; Andreou, C.; Borgwardt, S. Does Hippocampal Volume Predict Transition to Psychosis in a High-Risk Group? A Meta-Analysis. Front. Psychiatry 2020, 11, 614659. [Google Scholar] [CrossRef]
  59. Walter, A.; Suenderhauf, C.; Harrisberger, F.; Lenz, C.; Smieskova, R.; Chung, Y.; Cannon, T.D.; Bearden, C.E.; Rapp, C.; Bendfeldt, K.; et al. Hippocampal volume in subjects at clinical high-risk for psychosis: A systematic review and meta-analysis. Neurosci. Biobehav. Rev. 2016, 71, 680–690. [Google Scholar] [CrossRef]
  60. Radhakrishnan, R.; Skosnik, P.D.; Ranganathan, M.; Naganawa, M.; Toyonaga, T.; Finnema, S.; Hillmer, A.T.; Esterlis, I.; Huang, Y.; Nabulsi, N.; et al. In vivo evidence of lower synaptic vesicle density in schizophrenia. Mol. Psychiatry 2021, 26, 7690–7698. [Google Scholar] [CrossRef]
  61. Osimo, E.F.; Beck, K.; Reis Marques, T.; Howes, O.D. Synaptic loss in schizophrenia: A meta-analysis and systematic review of synaptic protein and mRNA measures. Mol. Psychiatry 2019, 24, 549–561. [Google Scholar] [CrossRef] [PubMed]
  62. Li, W.; Lv, L.; Luo, X.-J. In vivo study sheds new light on the dendritic spine pathology hypothesis of schizophrenia. Mol. Psychiatry 2022, 1–3. [Google Scholar] [CrossRef] [PubMed]
  63. Van Berdenis Berlekom, A.; Muflihah, C.H.; Snijders, G.J.L.J.; MacGillavry, H.D.; Middeldorp, J.; Hol, E.M.; Kahn, R.S.; de Witte, L.D. Synapse Pathology in Schizophrenia: A Meta-analysis of Postsynaptic Elements in Postmortem Brain Studies. Schizophr. Bull. 2020, 46, 374–386. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Garey, L.J.; Ong, W.Y.; Patel, T.S.; Kanani, M.; Davis, A.; Mortimer, A.M.; Barnes, T.R.; Hirsch, S.R. Reduced dendritic spine density on cerebral cortical pyramidal neurons in schizophrenia. J. Neurol. Neurosurg. Psychiatry 1998, 65, 446–453. [Google Scholar] [CrossRef] [PubMed]
  65. Glantz, L.A.; Lewis, D.A. Decreased dendritic spine density on prefrontal cortical pyramidal neurons in schizophrenia. Arch. Gen. Psychiatry 2000, 57, 65–73. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Konopaske, G.T.; Lange, N.; Coyle, J.T.; Benes, F.M. Prefrontal cortical dendritic spine pathology in schizophrenia and bipolar disorder. JAMA Psychiatry 2014, 71, 1323–1331. [Google Scholar] [CrossRef] [Green Version]
  67. Kolluri, N.; Sun, Z.; Sampson, A.R.; Lewis, D.A. Lamina-specific reductions in dendritic spine density in the prefrontal cortex of subjects with schizophrenia. Am. J. Psychiatry 2005, 162, 1200–1202. [Google Scholar] [CrossRef]
  68. Toyooka, K.; Iritani, S.; Makifuchi, T.; Shirakawa, O.; Kitamura, N.; Maeda, K.; Nakamura, R.; Niizato, K.; Watanabe, M.; Kakita, A.; et al. Selective reduction of a PDZ protein, SAP-97, in the prefrontal cortex of patients with chronic schizophrenia. J. Neurochem. 2002, 83, 797–806. [Google Scholar] [CrossRef]
  69. Provenzano, F.A.; Guo, J.; Wall, M.M.; Feng, X.; Sigmon, H.C.; Brucato, G.; First, M.B.; Rothman, D.L.; Girgis, R.R.; Lieberman, J.A.; et al. Hippocampal Pathology in Clinical High-Risk Patients and the Onset of Schizophrenia. Biol. Psychiatry 2020, 87, 234–242. [Google Scholar] [CrossRef]
  70. Tregellas, J.R.; Smucny, J.; Harris, J.G.; Olincy, A.; Maharajh, K.; Kronberg, E.; Eichman, L.C.; Lyons, E.; Freedman, R. Intrinsic hippocampal activity as a biomarker for cognition and symptoms in schizophrenia. Am. J. Psychiatry 2014, 171, 549–556. [Google Scholar] [CrossRef]
  71. Tregellas, J.R.; Ellis, J.; Shatti, S.; Du, Y.P.; Rojas, D.C. Increased hippocampal, thalamic, and prefrontal hemodynamic response to an urban noise stimulus in schizophrenia. Am. J. Psychiatry 2009, 166, 354–360. [Google Scholar] [CrossRef] [Green Version]
  72. Schobel, S.A.; Chaudhury, N.H.; Khan, U.A.; Paniagua, B.; Styner, M.A.; Asllani, I.; Inbar, B.P.; Corcoran, C.M.; Lieberman, J.A.; Moore, H.; et al. Imaging patients with psychosis and a mouse model establishes a spreading pattern of hippocampal dysfunction and implicates glutamate as a driver. Neuron 2013, 78, 81–93. [Google Scholar] [CrossRef] [Green Version]
  73. Schobel, S.A.; Lewandowski, N.M.; Corcoran, C.M.; Moore, H.; Brown, T.; Malaspina, D.; Small, S.A. Differential targeting of the CA1 subfield of the hippocampal formation by schizophrenia and related psychotic disorders. Arch. Gen. Psychiatry 2009, 66, 938–946. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Tregellas, J.R.; Davalos, D.B.; Rojas, D.C.; Waldo, M.C.; Gibson, L.; Wylie, K.; Du, Y.P.; Freedman, R. Increased hemodynamic response in the hippocampus, thalamus and prefrontal cortex during abnormal sensory gating in schizophrenia. Schizophr. Res. 2007, 92, 262–272. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Tregellas, J. Connecting brain structure and function in schizophrenia. Am. J. Psychiatry 2009, 166, 134–136. [Google Scholar] [CrossRef] [PubMed]
  76. Weiss, A.P.; Schacter, D.L.; Goff, D.C.; Rauch, S.L.; Alpert, N.M.; Fischman, A.J.; Heckers, S. Impaired hippocampal recruitment during normal modulation of memory performance in schizophrenia. Biol. Psychiatry 2003, 53, 48–55. [Google Scholar] [CrossRef]
  77. Ongür, D.; Cullen, T.J.; Wolf, D.H.; Rohan, M.; Barreira, P.; Zalesak, M.; Heckers, S. The neural basis of relational memory deficits in schizophrenia. Arch. Gen. Psychiatry 2006, 63, 356–365. [Google Scholar] [CrossRef]
  78. Williams, L.E.; Blackford, J.U.; Luksik, A.; Gauthier, I.; Heckers, S. Reduced habituation in patients with schizophrenia. Schizophr. Res. 2013, 151, 124–132. [Google Scholar] [CrossRef] [Green Version]
  79. Holt, D.J.; Kunkel, L.; Weiss, A.P.; Goff, D.C.; Wright, C.I.; Shin, L.M.; Rauch, S.L.; Hootnick, J.; Heckers, S. Increased medial temporal lobe activation during the passive viewing of emotional and neutral facial expressions in schizophrenia. Schizophr. Res. 2006, 82, 153–162. [Google Scholar] [CrossRef]
  80. Talati, P.; Rane, S.; Kose, S.; Blackford, J.U.; Gore, J.; Donahue, M.J.; Heckers, S. Increased hippocampal CA1 cerebral blood volume in schizophrenia. Neuroimage Clin. 2014, 5, 359–364. [Google Scholar] [CrossRef] [Green Version]
  81. Allen, P.; Chaddock, C.A.; Egerton, A.; Howes, O.D.; Bonoldi, I.; Zelaya, F.; Bhattacharyya, S.; Murray, R.; McGuire, P. Resting Hyperperfusion of the Hippocampus, Midbrain, and Basal Ganglia in People at High Risk for Psychosis. Am. J. Psychiatry 2016, 173, 392–399. [Google Scholar] [CrossRef] [PubMed]
  82. Ghoshal, A.; Conn, P.J. The hippocampo-prefrontal pathway: A possible therapeutic target for negative and cognitive symptoms of schizophrenia. Future Neurol. 2015, 10, 115–128. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Blessing, E.M.; Murty, V.P.; Zeng, B.; Wang, J.; Davachi, L.; Goff, D.C. Anterior Hippocampal-Cortical Functional Connectivity Distinguishes Antipsychotic Naïve First-Episode Psychosis Patients from Controls and May Predict Response to Second-Generation Antipsychotic Treatment. Schizophr. Bull. 2020, 46, 680–689. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Zhou, Y.; Shu, N.; Liu, Y.; Song, M.; Hao, Y.; Liu, H.; Yu, C.; Liu, Z.; Jiang, T. Altered resting-state functional connectivity and anatomical connectivity of hippocampus in schizophrenia. Schizophr. Res. 2008, 100, 120–132. [Google Scholar] [CrossRef] [PubMed]
  85. Xi, Y.-B.; Li, C.; Cui, L.-B.; Liu, J.; Guo, F.; Li, L.; Liu, T.-T.; Liu, K.; Chen, G.; Xi, M.; et al. Anterior Cingulate Cortico-Hippocampal Dysconnectivity in Unaffected Relatives of Schizophrenia Patients: A Stochastic Dynamic Causal Modeling Study. Front. Hum. Neurosci. 2016, 10, 383. [Google Scholar] [CrossRef] [Green Version]
  86. Sullivan, P.F.; Kendler, K.S.; Neale, M.C. Schizophrenia as a complex trait: Evidence from a meta-analysis of twin studies. Arch. Gen. Psychiatry 2003, 60, 1187–1192. [Google Scholar] [CrossRef] [Green Version]
  87. Cardno, A.G.; Gottesman, I.I. Twin studies of schizophrenia: From bow-and-arrow concordances to star wars Mx and functional genomics. Am. J. Med. Genet. 2000, 97, 12–17. [Google Scholar] [CrossRef]
  88. Perez, J.M.; Berto, S.; Gleason, K.; Ghose, S.; Tan, C.; Kim, T.-K.; Konopka, G.; Tamminga, C.A. Hippocampal subfield transcriptome analysis in schizophrenia psychosis. Mol. Psychiatry 2020, 26, 2577–2589. [Google Scholar] [CrossRef]
  89. Jaffe, A.E.; Hoeppner, D.J.; Saito, T.; Blanpain, L.; Ukaigwe, J.; Burke, E.E.; Collado-Torres, L.; Tao, R.; Tajinda, K.; Maynard, K.R.; et al. Profiling gene expression in the human dentate gyrus granule cell layer reveals insights into schizophrenia and its genetic risk. Nat. Neurosci. 2020, 23, 510–519. [Google Scholar] [CrossRef]
  90. Etemadikhah, M.; Niazi, A.; Wetterberg, L.; Feuk, L. Transcriptome analysis of fibroblasts from schizophrenia patients reveals differential expression of schizophrenia-related genes. Sci. Rep. 2020, 10, 630. [Google Scholar] [CrossRef] [Green Version]
  91. Hall, L.S.; Medway, C.W.; Pain, O.; Pardiñas, A.F.; Rees, E.G.; Escott-Price, V.; Pocklington, A.; Bray, N.J.; Holmans, P.A.; Walters, J.T.R.; et al. A transcriptome-wide association study implicates specific pre- and post-synaptic abnormalities in schizophrenia. Hum. Mol. Genet. 2020, 29, 159–167. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Gusev, A.; Mancuso, N.; Won, H.; Kousi, M.; Finucane, H.K.; Reshef, Y.; Song, L.; Safi, A.; McCarroll, S.; Neale, B.M.; et al. Transcriptome-wide association study of schizophrenia and chromatin activity yields mechanistic disease insights. Nat. Genet. 2018, 50, 538–548. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Frajman, A.; Maggio, N.; Muler, I.; Haroutunian, V.; Katsel, P.; Yitzhaky, A.; Weiser, M.; Hertzberg, L. Gene expression meta-analysis reveals the down-regulation of three GABA receptor subunits in the superior temporal gyrus of patients with schizophrenia. Schizophr. Res. 2020, 220, 29–37. [Google Scholar] [CrossRef] [PubMed]
  94. Li, Z.; Chen, J.; Yu, H.; He, L.; Xu, Y.; Zhang, D.; Yi, Q.; Li, C.; Li, X.; Shen, J.; et al. Genome-wide association analysis identifies 30 new susceptibility loci for schizophrenia. Nat. Genet. 2017, 49, 1576–1583. [Google Scholar] [CrossRef] [PubMed]
  95. Read, J.; van Os, J.; Morrison, A.P.; Ross, C.A. Childhood trauma, psychosis and schizophrenia: A literature review with theoretical and clinical implications. Acta Psychiatr. Scand. 2005, 112, 330–350. [Google Scholar] [CrossRef]
  96. Hoy, K.; Barrett, S.; Shannon, C.; Campbell, C.; Watson, D.; Rushe, T.; Shevlin, M.; Bai, F.; Cooper, S.; Mulholland, C. Childhood trauma and hippocampal and amygdalar volumes in first-episode psychosis. Schizophr. Bull. 2012, 38, 1162–1169. [Google Scholar] [CrossRef] [Green Version]
  97. Du Plessis, S.; Scheffler, F.; Luckhoff, H.; Asmal, L.; Kilian, S.; Phahladira, L.; Emsley, R. Childhood trauma and hippocampal subfield volumes in first-episode schizophrenia and healthy controls. Schizophr. Res. 2020, 215, 308–313. [Google Scholar] [CrossRef]
  98. Vythilingam, M.; Heim, C.; Newport, J.; Miller, A.H.; Anderson, E.; Bronen, R.; Brummer, M.; Staib, L.; Vermetten, E.; Charney, D.S.; et al. Childhood trauma associated with smaller hippocampal volume in women with major depression. Am. J. Psychiatry 2002, 159, 2072–2080. [Google Scholar] [CrossRef]
  99. Frodl, T.; Reinhold, E.; Koutsouleris, N.; Reiser, M.; Meisenzahl, E.M. Interaction of childhood stress with hippocampus and prefrontal cortex volume reduction in major depression. J. Psychiatr. Res. 2010, 44, 799–807. [Google Scholar] [CrossRef]
  100. Bremner, J.D.; Vythilingam, M.; Vermetten, E.; Southwick, S.M.; McGlashan, T.; Nazeer, A.; Khan, S.; Vaccarino, L.V.; Soufer, R.; Garg, P.K.; et al. MRI and PET study of deficits in hippocampal structure and function in women with childhood sexual abuse and posttraumatic stress disorder. Am. J. Psychiatry 2003, 160, 924–932. [Google Scholar] [CrossRef] [Green Version]
  101. Teicher, M.H.; Anderson, C.M.; Polcari, A. Childhood maltreatment is associated with reduced volume in the hippocampal subfields CA3, dentate gyrus, and subiculum. Proc. Natl. Acad. Sci. USA 2012, 109, E563–E572. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Gould, E.; Tanapat, P. Stress and hippocampal neurogenesis. Biol. Psychiatry 1999, 46, 1472–1479. [Google Scholar] [CrossRef]
  103. Herzog, J.I.; Thome, J.; Demirakca, T.; Koppe, G.; Ende, G.; Lis, S.; Rausch, S.; Priebe, K.; Müller-Engelmann, M.; Steil, R.; et al. Influence of Severity of Type and Timing of Retrospectively Reported Childhood Maltreatment on Female Amygdala and Hippocampal Volume. Sci. Rep. 2020, 10, 1903. [Google Scholar] [CrossRef]
  104. Li, C.-X.; Li, Z.; Hu, X.; Zhang, X.; Bachevalier, J. Altered hippocampal-prefrontal functional network integrity in adult macaque monkeys with neonatal hippocampal lesions. Neuroimage 2021, 227, 117645. [Google Scholar] [CrossRef] [PubMed]
  105. Beauregard, M.; Bachevalier, J. Neonatal insult to the hippocampal region and schizophrenia: A review and a putative animal model. Can. J. Psychiatry 1996, 41, 446–456. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Scheinost, D.; Spann, M.N.; McDonough, L.; Peterson, B.S.; Monk, C. Associations between different dimensions of prenatal distress, neonatal hippocampal connectivity, and infant memory. Neuropsychopharmacology 2020, 45, 1272–1279. [Google Scholar] [CrossRef] [Green Version]
  107. Coe, C.L.; Kramer, M.; Czéh, B.; Gould, E.; Reeves, A.J.; Kirschbaum, C.; Fuchs, E. Prenatal stress diminishes neurogenesis in the dentate gyrus of juvenile rhesus monkeys. Biol. Psychiatry 2003, 54, 1025–1034. [Google Scholar] [CrossRef]
  108. Lesh, T.A.; Careaga, M.; Rose, D.R.; McAllister, A.K.; van de Water, J.; Carter, C.S.; Ashwood, P. Cytokine alterations in first-episode schizophrenia and bipolar disorder: Relationships to brain structure and symptoms. J. Neuroinflammation 2018, 15, 165. [Google Scholar] [CrossRef] [Green Version]
  109. Lanz, T.A.; Reinhart, V.; Sheehan, M.J.; Rizzo, S.J.S.; Bove, S.E.; James, L.C.; Volfson, D.; Lewis, D.A.; Kleiman, R.J. Postmortem transcriptional profiling reveals widespread increase in inflammation in schizophrenia: A comparison of prefrontal cortex, striatum, and hippocampus among matched tetrads of controls with subjects diagnosed with schizophrenia, bipolar or major depressive disorder. Transl. Psychiatry 2019, 9, 151. [Google Scholar] [CrossRef] [Green Version]
  110. Hwang, Y.; Kim, J.; Shin, J.Y.; Kim, J., II; Seo, J.S.; Webster, M.J.; Lee, D.; Kim, S. Gene expression profiling by mRNA sequencing reveals increased expression of immune/inflammation-related genes in the hippocampus of individuals with schizophrenia. Transl. Psychiatry 2013, 3, e321. [Google Scholar] [CrossRef] [Green Version]
  111. Monji, A.; Kato, T.; Kanba, S. Cytokines and schizophrenia: Microglia hypothesis of schizophrenia. Psychiatry Clin. Neurosci. 2009, 63, 257–265. [Google Scholar] [CrossRef] [PubMed]
  112. Sierra, A.; de Castro, F.; Del Río-Hortega, J.; Rafael Iglesias-Rozas, J.; Garrosa, M.; Kettenmann, H. The “Big-Bang” for modern glial biology: Translation and comments on Pío del Río-Hortega 1919 series of papers on microglia. Glia 2016, 64, 1801–1840. [Google Scholar] [CrossRef] [PubMed]
  113. Prinz, M.; Priller, J. Microglia and brain macrophages in the molecular age: From origin to neuropsychiatric disease. Nat. Rev. Neurosci. 2014, 15, 300–312. [Google Scholar] [CrossRef] [PubMed]
  114. Kierdorf, K.; Erny, D.; Goldmann, T.; Sander, V.; Schulz, C.; Perdiguero, E.G.; Wieghofer, P.; Heinrich, A.; Riemke, P.; Hölscher, C.; et al. Microglia emerge from erythromyeloid precursors via Pu.1- and Irf8-dependent pathways. Nat. Neurosci. 2013, 16, 273–280. [Google Scholar] [CrossRef]
  115. Ginhoux, F.; Greter, M.; Leboeuf, M.; Nandi, S.; See, P.; Gokhan, S.; Mehler, M.F.; Conway, S.J.; Ng, L.G.; Stanley, E.R.; et al. Fate mapping analysis reveals that adult microglia derive from primitive macrophages. Science 2010, 330, 841–845. [Google Scholar] [CrossRef] [Green Version]
  116. Paolicelli, R.C.; Bolasco, G.; Pagani, F.; Maggi, L.; Scianni, M.; Panzanelli, P.; Giustetto, M.; Ferreira, T.A.; Guiducci, E.; Dumas, L.; et al. Synaptic pruning by microglia is necessary for normal brain development. Science 2011, 333, 1456–1458. [Google Scholar] [CrossRef] [Green Version]
  117. Zhan, Y.; Paolicelli, R.C.; Sforazzini, F.; Weinhard, L.; Bolasco, G.; Pagani, F.; Vyssotski, A.L.; Bifone, A.; Gozzi, A.; Ragozzino, D.; et al. Deficient neuron-microglia signaling results in impaired functional brain connectivity and social behavior. Nat. Neurosci. 2014, 17, 400–406. [Google Scholar] [CrossRef]
  118. Ji, K.; Akgul, G.; Wollmuth, L.P.; Tsirka, S.E. Microglia actively regulate the number of functional synapses. PLoS ONE 2013, 8, e56293. [Google Scholar] [CrossRef] [Green Version]
  119. Weinhard, L.; Di Bartolomei, G.; Bolasco, G.; Machado, P.; Schieber, N.L.; Neniskyte, U.; Exiga, M.; Vadisiute, A.; Raggioli, A.; Schertel, A.; et al. Microglia remodel synapses by presynaptic trogocytosis and spine head filopodia induction. Nat. Commun. 2018, 9, 1228. [Google Scholar] [CrossRef] [Green Version]
  120. Hattori, Y.; Naito, Y.; Tsugawa, Y.; Nonaka, S.; Wake, H.; Nagasawa, T.; Kawaguchi, A.; Miyata, T. Transient microglial absence assists postmigratory cortical neurons in proper differentiation. Nat. Commun. 2020, 11, 1631. [Google Scholar] [CrossRef]
  121. Tan, Y.-L.; Yuan, Y.; Tian, L. Microglial regional heterogeneity and its role in the brain. Mol. Psychiatry 2020, 25, 351–367. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Pocock, J.M.; Kettenmann, H. Neurotransmitter receptors on microglia. Trends Neurosci. 2007, 30, 527–535. [Google Scholar] [CrossRef] [PubMed]
  123. York, E.M.; Bernier, L.-P.; MacVicar, B.A. Microglial modulation of neuronal activity in the healthy brain. Dev. Neurobiol. 2018, 78, 593–603. [Google Scholar] [CrossRef] [PubMed]
  124. Kettenmann, H.; Hanisch, U.-K.; Noda, M.; Verkhratsky, A. Physiology of microglia. Physiol. Rev. 2011, 91, 461–553. [Google Scholar] [CrossRef] [PubMed]
  125. Viviani, B.; Bartesaghi, S.; Gardoni, F.; Vezzani, A.; Behrens, M.M.; Bartfai, T.; Binaglia, M.; Corsini, E.; Di Luca, M.; Galli, C.L.; et al. Interleukin-1beta enhances NMDA receptor-mediated intracellular calcium increase through activation of the Src family of kinases. J. Neurosci. 2003, 23, 8692–8700. [Google Scholar] [CrossRef]
  126. Vezzani, A.; Viviani, B. Neuromodulatory properties of inflammatory cytokines and their impact on neuronal excitability. Neuropharmacology 2015, 96, 70–82. [Google Scholar] [CrossRef]
  127. Gardoni, F.; Boraso, M.; Zianni, E.; Corsini, E.; Galli, C.L.; Cattabeni, F.; Marinovich, M.; Di Luca, M.; Viviani, B. Distribution of interleukin-1 receptor complex at the synaptic membrane driven by interleukin-1β and NMDA stimulation. J. Neuroinflammation 2011, 8, 14. [Google Scholar] [CrossRef] [Green Version]
  128. Stellwagen, D.; Beattie, E.C.; Seo, J.Y.; Malenka, R.C. Differential regulation of AMPA receptor and GABA receptor trafficking by tumor necrosis factor-alpha. J. Neurosci. 2005, 25, 3219–3228. [Google Scholar] [CrossRef] [Green Version]
  129. 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]
  130. Badimon, A.; Strasburger, H.J.; Ayata, P.; Chen, X.; Nair, A.; Ikegami, A.; Hwang, P.; Chan, A.T.; Graves, S.M.; Uweru, J.O.; et al. Negative feedback control of neuronal activity by microglia. Nature 2020, 586, 417–423. [Google Scholar] [CrossRef]
  131. Bayer, T.A.; Buslei, R.; Havas, L.; Falkai, P. Evidence for activation of microglia in patients with psychiatric illnesses. Neurosci. Lett. 1999, 271, 126–128. [Google Scholar] [CrossRef]
  132. Busse, S.; Busse, M.; Schiltz, K.; Bielau, H.; Gos, T.; Brisch, R.; Mawrin, C.; Schmitt, A.; Jordan, W.; Müller, U.J.; et al. Different distribution patterns of lymphocytes and microglia in the hippocampus of patients with residual versus paranoid schizophrenia: Further evidence for disease course-related immune alterations? Brain Behav. Immun. 2012, 26, 1273–1279. [Google Scholar] [CrossRef] [PubMed]
  133. Sellgren, C.M.; Gracias, J.; Watmuff, B.; Biag, J.D.; Thanos, J.M.; Whittredge, P.B.; Fu, T.; Worringer, K.; Brown, H.E.; Wang, J.; et al. Increased synapse elimination by microglia in schizophrenia patient-derived models of synaptic pruning. Nat. Neurosci. 2019, 22, 374–385. [Google Scholar] [CrossRef] [PubMed]
  134. Schizophrenia Working Group of the Psychiatric Genomics Consortium. Biological insights from 108 schizophrenia-associated genetic loci. Nature 2014, 511, 421–427. [Google Scholar] [CrossRef] [Green Version]
  135. Yilmaz, M.; Yalcin, E.; Presumey, J.; Aw, E.; Ma, M.; Whelan, C.W.; Stevens, B.; McCarroll, S.A.; Carroll, M.C. Overexpression of schizophrenia susceptibility factor human complement C4A promotes excessive synaptic loss and behavioral changes in mice. Nat. Neurosci. 2021, 24, 214–224. [Google Scholar] [CrossRef]
  136. Comer, A.L.; Jinadasa, T.; Sriram, B.; Phadke, R.A.; Kretsge, L.N.; Nguyen, T.P.H.; Antognetti, G.; Gilbert, J.P.; Lee, J.; Newmark, E.R.; et al. Increased expression of schizophrenia-associated gene C4 leads to hypoconnectivity of prefrontal cortex and reduced social interaction. PLoS Biol. 2020, 18, e3000604. [Google Scholar] [CrossRef] [Green Version]
  137. Sekar, A.; Bialas, A.R.; de Rivera, H.; Davis, A.; Hammond, T.R.; Kamitaki, N.; Tooley, K.; Presumey, J.; Baum, M.; van Doren, V.; et al. Schizophrenia risk from complex variation of complement component 4. Nature 2016, 530, 177–183. [Google Scholar] [CrossRef] [Green Version]
  138. Conen, S.; Gregory, C.J.; Hinz, R.; Smallman, R.; Corsi-Zuelli, F.; Deakin, B.; Talbot, P.S. Neuroinflammation as measured by positron emission tomography in patients with recent onset and established schizophrenia: Implications for immune pathogenesis. Mol. Psychiatry 2020, 26, 5398–5406. [Google Scholar] [CrossRef]
  139. Di Biase, M.A.; Zalesky, A.; O’keefe, G.; Laskaris, L.; Baune, B.T.; Weickert, C.S.; Olver, J.; McGorry, P.D.; Amminger, G.P.; Nelson, B.; et al. PET imaging of putative microglial activation in individuals at ultra-high risk for psychosis, recently diagnosed and chronically ill with schizophrenia. Transl. Psychiatry 2017, 7, e1225. [Google Scholar] [CrossRef] [Green Version]
  140. Marques, T.R.; Ashok, A.H.; Pillinger, T.; Veronese, M.; Turkheimer, F.E.; Dazzan, P.; Sommer, I.E.C.; Howes, O.D. Neuroinflammation in schizophrenia: Meta-analysis of in vivo microglial imaging studies. Psychol. Med. 2019, 49, 2186–2196. [Google Scholar] [CrossRef] [Green Version]
  141. Holmes, S.E.; Hinz, R.; Drake, R.J.; Gregory, C.J.; Conen, S.; Matthews, J.C.; Anton-Rodriguez, J.M.; Gerhard, A.; Talbot, P.S. In vivo imaging of brain microglial activity in antipsychotic-free and medicated schizophrenia: A 11C(R)-PK11195 positron emission tomography study. Mol. Psychiatry 2016, 21, 1672–1679. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Sneeboer, M.A.M.; van der Doef, T.; Litjens, M.; Psy, N.B.B.; Melief, J.; Hol, E.M.; Kahn, R.S.; de Witte, L.D. Microglial activation in schizophrenia: Is translocator 18 kDa protein (TSPO) the right marker? Schizophr. Res. 2020, 215, 167–172. [Google Scholar] [CrossRef] [PubMed]
  143. Van Berckel, B.N.; Bossong, M.G.; Boellaard, R.; Kloet, R.; Schuitemaker, A.; Caspers, E.; Luurtsema, G.; Windhorst, A.D.; Cahn, W.; Lammertsma, A.A.; et al. Microglia activation in recent-onset schizophrenia: A quantitative (R)-11CPK11195 positron emission tomography study. Biol. Psychiatry 2008, 64, 820–822. [Google Scholar] [CrossRef]
  144. Bloomfield, P.S.; Selvaraj, S.; Veronese, M.; Rizzo, G.; Bertoldo, A.; Owen, D.R.; Bloomfield, M.A.; Bonoldi, I.; Kalk, N.; Turkheimer, F.; et al. Microglial Activity in People at Ultra High Risk of Psychosis and in Schizophrenia: An (11)CPBR28 PET Brain Imaging Study. Am. J. Psychiatry 2016, 173, 44–52. [Google Scholar] [CrossRef] [Green Version]
  145. Zhang, L.; Zheng, H.; Wu, R.; Kosten, T.R.; Zhang, X.-Y.; Zhao, J. The effect of minocycline on amelioration of cognitive deficits and pro-inflammatory cytokines levels in patients with schizophrenia. Schizophr. Res. 2019, 212, 92–98. [Google Scholar] [CrossRef]
  146. Levkovitz, Y.; Mendlovich, S.; Riwkes, S.; Braw, Y.; Levkovitch-Verbin, H.; Gal, G.; Fennig, S.; Treves, I.; Kron, S. A double-blind, randomized study of minocycline for the treatment of negative and cognitive symptoms in early-phase schizophrenia. J. Clin. Psychiatry 2010, 71, 138–149. [Google Scholar] [CrossRef]
  147. Miyaoka, T.; Yasukawa, R.; Yasuda, H.; Hayashida, M.; Inagaki, T.; Horiguchi, J. Minocycline as adjunctive therapy for schizophrenia: An open-label study. Clin. Neuropharmacol. 2008, 31, 287–292. [Google Scholar] [CrossRef]
  148. Chaudhry, I.B.; Hallak, J.; Husain, N.; Minhas, F.; Stirling, J.; Richardson, P.; Dursun, S.; Dunn, G.; Deakin, B. Minocycline benefits negative symptoms in early schizophrenia: A randomised double-blind placebo-controlled clinical trial in patients on standard treatment. J. Psychopharmacol. 2012, 26, 1185–1193. [Google Scholar] [CrossRef]
  149. Solmi, M.; Veronese, N.; Thapa, N.; Facchini, S.; Stubbs, B.; Fornaro, M.; Carvalho, A.F.; Correll, C.U. Systematic review and meta-analysis of the efficacy and safety of minocycline in schizophrenia. CNS Spectr. 2017, 22, 415–426. [Google Scholar] [CrossRef] [Green Version]
  150. Xiang, Y.-Q.; Zheng, W.; Wang, S.-B.; Yang, X.-H.; Cai, D.-B.; Ng, C.H.; Ungvari, G.S.; Kelly, D.L.; Xu, W.-Y.; Xiang, Y.-T. Adjunctive minocycline for schizophrenia: A meta-analysis of randomized controlled trials. Eur. Neuropsychopharmacol. 2017, 27, 8–18. [Google Scholar] [CrossRef]
  151. Zheng, W.; Zhu, X.-M.; Zhang, Q.-E.; Cheng, G.; Cai, D.-B.; He, J.; Ng, C.H.; Ungvari, G.S.; Peng, X.-J.; Ning, Y.-P.; et al. Adjunctive minocycline for major mental disorders: A systematic review. J. Psychopharmacol. 2019, 33, 1215–1226. [Google Scholar] [CrossRef] [PubMed]
  152. Deakin, B.; Suckling, J.; Barnes, T.R.E.; Byrne, K.; Chaudhry, I.B.; Dazzan, P.; Drake, R.J.; Giordano, A.; Husain, N.; Jones, P.B.; et al. The benefit of minocycline on negative symptoms of schizophrenia in patients with recent-onset psychosis (BeneMin): A randomised, double-blind, placebo-controlled trial. Lancet Psychiatry 2018, 5, 885–894. [Google Scholar] [CrossRef] [Green Version]
  153. Weiser, M.; Levi, L.; Burshtein, S.; Chiriță, R.; Cirjaliu, D.; Gonen, I.; Yolken, R.; Davidson, M.; Zamora, D.; Davis, J.M. The effect of minocycline on symptoms in schizophrenia: Results from a randomized controlled trial. Schizophr. Res. 2019, 206, 325–332. [Google Scholar] [CrossRef]
  154. Kishimoto, T.; Horigome, T.; Takamiya, A. Minocycline as a treatment for schizophrenia: Is the discussion truly finished? Lancet Psychiatry 2018, 5, 856–857. [Google Scholar] [CrossRef] [Green Version]
  155. Tarasov, V.V.; Svistunov, A.A.; Chubarev, V.N.; Sologova, S.S.; Mukhortova, P.; Levushkin, D.; Somasundaram, S.G.; Kirkland, C.E.; Bachurin, S.O.; Aliev, G. Alterations of Astrocytes in the Context of Schizophrenic Dementia. Front. Pharmacol. 2019, 10, 1612. [Google Scholar] [CrossRef] [Green Version]
  156. Kim, R.; Healey, K.L.; Sepulveda-Orengo, M.T.; Reissner, K.J. Astroglial correlates of neuropsychiatric disease: From astrocytopathy to astrogliosis. Prog. Neuropsychopharmacol. Biol. Psychiatry 2018, 87, 126–146. [Google Scholar] [CrossRef]
  157. Notter, T. Astrocytes in schizophrenia. Brain Neurosci. Adv. 2021, 5, 1–8. [Google Scholar] [CrossRef]
  158. Goudriaan, A.; de Leeuw, C.; Ripke, S.; Hultman, C.M.; Sklar, P.; Sullivan, P.F.; Smit, A.B.; Posthuma, D.; Verheijen, M.H.G. Specific glial functions contribute to schizophrenia susceptibility. Schizophr. Bull. 2014, 40, 925–935. [Google Scholar] [CrossRef] [Green Version]
  159. Schmitt, A.; Steyskal, C.; Bernstein, H.-G.; Schneider-Axmann, T.; Parlapani, E.; Schaeffer, E.L.; Gattaz, W.F.; Bogerts, B.; Schmitz, C.; Falkai, P. Stereologic investigation of the posterior part of the hippocampus in schizophrenia. Acta Neuropathol. 2009, 117, 395–407. [Google Scholar] [CrossRef] [Green Version]
  160. Webster, M.J.; Knable, M.B.; Johnston-Wilson, N.; Nagata, K.; Inagaki, M.; Yolken, R.H. Immunohistochemical localization of phosphorylated glial fibrillary acidic protein in the prefrontal cortex and hippocampus from patients with schizophrenia, bipolar disorder, and depression. Brain Behav. Immun. 2001, 15, 388–400. [Google Scholar] [CrossRef] [Green Version]
  161. Toker, L.; Mancarci, B.O.; Tripathy, S.; Pavlidis, P. Transcriptomic Evidence for Alterations in Astrocytes and Parvalbumin Interneurons in Subjects with Bipolar Disorder and Schizophrenia. Biol. Psychiatry 2018, 84, 787–796. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Terrillion, C.E.; Abazyan, B.; Yang, Z.; Crawford, J.; Shevelkin, A.V.; Jouroukhin, Y.; Yoo, K.H.; Cho, C.H.; Roychaudhuri, R.; Snyder, S.H.; et al. DISC1 in Astrocytes Influences Adult Neurogenesis and Hippocampus-Dependent Behaviors in Mice. Neuropsychopharmacology 2017, 42, 2242–2251. [Google Scholar] [CrossRef] [PubMed]
  163. Swanson, L.W.; Hahn, J.D. A qualitative solution with quantitative potential for the mouse hippocampal cortex flatmap problem. Proc. Natl. Acad. Sci. USA 2020, 117, 3220–3231. [Google Scholar] [CrossRef]
  164. Bjerke, I.E.; Yates, S.C.; Laja, A.; Witter, M.P.; Puchades, M.A.; Bjaalie, J.G.; Leergaard, T.B. Densities and numbers of calbindin and parvalbumin positive neurons across the rat and mouse brain. iScience 2021, 24, 101906. [Google Scholar] [CrossRef] [PubMed]
  165. Snyder, J.S.; Choe, J.S.; Clifford, M.A.; Jeurling, S.I.; Hurley, P.; Brown, A.; Kamhi, J.F.; Cameron, H.A. Adult-born hippocampal neurons are more numerous, faster maturing, and more involved in behavior in rats than in mice. J. Neurosci. 2009, 29, 14484–14495. [Google Scholar] [CrossRef] [Green Version]
  166. Buzsáki, G.; Buhl, D.L.; Harris, K.D.; Csicsvari, J.; Czéh, B.; Morozov, A. Hippocampal network patterns of activity in the mouse. Neuroscience 2003, 116, 201–211. [Google Scholar] [CrossRef]
  167. Mou, X.; Cheng, J.; Yu, Y.S.W.; Kee, S.E.; Ji, D. Comparing Mouse and Rat Hippocampal Place Cell Activities and Firing Sequences in the Same Environments. Front. Cell. Neurosci. 2018, 12, 332. [Google Scholar] [CrossRef] [Green Version]
  168. Langova, V.; Vales, K.; Horka, P.; Horacek, J. The Role of Zebrafish and Laboratory Rodents in Schizophrenia Research. Front. Psychiatry 2020, 11, 703. [Google Scholar] [CrossRef]
  169. Mueller, T.; Dong, Z.; Berberoglu, M.A.; Guo, S. The dorsal pallium in zebrafish, Danio rerio (Cyprinidae, Teleostei). Brain Res. 2011, 1381, 95–105. [Google Scholar] [CrossRef] [Green Version]
  170. Thyme, S.B.; Pieper, L.M.; Li, E.H.; Pandey, S.; Wang, Y.; Morris, N.S.; Sha, C.; Choi, J.W.; Herrera, K.J.; Soucy, E.R.; et al. Phenotypic Landscape of Schizophrenia-Associated Genes Defines Candidates and Their Shared Functions. Cell 2019, 177, 478–491.e20. [Google Scholar] [CrossRef] [Green Version]
  171. Mednick, S.A.; Machon, R.A.; Huttunen, M.O.; Bonett, D. Adult schizophrenia following prenatal exposure to an influenza epidemic. Arch. Gen. Psychiatry 1988, 45, 189–192. [Google Scholar] [CrossRef] [PubMed]
  172. McGrath, J.J.; Pemberton, M.R.; Welham, J.L.; Murray, R.M. Schizophrenia and the influenza epidemics of 1954, 1957 and 1959: A southern hemisphere study. Schizophr. Res. 1994, 14, 1–8. [Google Scholar] [CrossRef]
  173. O’Callaghan, E.; Sham, P.; Takei, N.; Glover, G.; Murray, R.M. Schizophrenia after prenatal exposure to 1957 A2 influenza epidemic. Lancet 1991, 337, 1248–1250. [Google Scholar] [CrossRef]
  174. Adams, W.; Kendell, R.E.; Hare, E.H.; Munk-Jørgensen, P. Epidemiological evidence that maternal influenza contributes to the aetiology of schizophrenia. An analysis of Scottish, English, and Danish data. Br. J. Psychiatry 1993, 163, 522–534. [Google Scholar] [CrossRef] [PubMed]
  175. Limosin, F.; Rouillon, F.; Payan, C.; Cohen, J.-M.; Strub, N. Prenatal exposure to influenza as a risk factor for adult schizophrenia. Acta Psychiatr. Scand. 2003, 107, 331–335. [Google Scholar] [CrossRef] [PubMed]
  176. Brown, A.S.; Derkits, E.J. Prenatal infection and schizophrenia: A review of epidemiologic and translational studies. Am. J. Psychiatry 2010, 167, 261–280. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Fatemi, S.H.; Reutiman, T.J.; Folsom, T.D.; Huang, H.; Oishi, K.; Mori, S.; Smee, D.F.; Pearce, D.A.; Winter, C.; Sohr, R.; et al. Maternal infection leads to abnormal gene regulation and brain atrophy in mouse offspring: Implications for genesis of neurodevelopmental disorders. Schizophr. Res. 2008, 99, 56–70. [Google Scholar] [CrossRef] [Green Version]
  178. Brown, A.S.; Schaefer, C.A.; Quesenberry, C.P.; Liu, L.; Babulas, V.P.; Susser, E.S. Maternal exposure to toxoplasmosis and risk of schizophrenia in adult offspring. Am. J. Psychiatry 2005, 162, 767–773. [Google Scholar] [CrossRef]
  179. Brown, A.S.; Cohen, P.; Harkavy-Friedman, J.; Babulas, V.; Malaspina, D.; Gorman, J.M.; Susser, E.S. Prenatal rubella, premorbid abnormalities, and adult schizophrenia. Biol. Psychiatry 2001, 49, 473–486. [Google Scholar] [CrossRef]
  180. Buka, S.L.; Tsuang, M.T.; Torrey, E.F.; Klebanoff, M.A.; Bernstein, D.; Yolken, R.H. Maternal infections and subsequent psychosis among offspring. Arch. Gen. Psychiatry 2001, 58, 1032–1037. [Google Scholar] [CrossRef] [Green Version]
  181. Buka, S.L.; Cannon, T.D.; Torrey, E.F.; Yolken, R.H. Maternal exposure to herpes simplex virus and risk of psychosis among adult offspring. Biol. Psychiatry 2008, 63, 809–815. [Google Scholar] [CrossRef] [PubMed]
  182. Mortensen, P.B.; Nørgaard-Pedersen, B.; Waltoft, B.L.; Sørensen, T.L.; Hougaard, D.; Torrey, E.F.; Yolken, R.H. Toxoplasma gondii as a risk factor for early-onset schizophrenia: Analysis of filter paper blood samples obtained at birth. Biol. Psychiatry 2007, 61, 688–693. [Google Scholar] [CrossRef] [PubMed]
  183. Suvisaari, J.; Haukka, J.; Tanskanen, A.; Hovi, T.; Lönnqvist, J. Association between prenatal exposure to poliovirus infection and adult schizophrenia. Am. J. Psychiatry 1999, 156, 1100–1102. [Google Scholar] [CrossRef] [PubMed]
  184. Meyer, U.; Nyffeler, M.; Engler, A.; Urwyler, A.; Schedlowski, M.; Knuesel, I.; Yee, B.K.; Feldon, J. The time of prenatal immune challenge determines the specificity of inflammation-mediated brain and behavioral pathology. J. Neurosci. 2006, 26, 4752–4762. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Meyer, U.; Yee, B.K.; Feldon, J. The neurodevelopmental impact of prenatal infections at different times of pregnancy: The earlier the worse? Neuroscientist 2007, 13, 241–256. [Google Scholar] [CrossRef]
  186. Meyer, U. Developmental neuroinflammation and schizophrenia. Prog. Neuropsychopharmacol. Biol. Psychiatry 2013, 42, 20–34. [Google Scholar] [CrossRef]
  187. Ashdown, H.; Dumont, Y.; Ng, M.; Poole, S.; Boksa, P.; Luheshi, G.N. The role of cytokines in mediating effects of prenatal infection on the fetus: Implications for schizophrenia. Mol. Psychiatry 2006, 11, 47–55. [Google Scholar] [CrossRef]
  188. Gilmore, J.H.; Fredrik Jarskog, L.; Vadlamudi, S.; Lauder, J.M. Prenatal infection and risk for schizophrenia: IL-1beta, IL-6, and TNFalpha inhibit cortical neuron dendrite development. Neuropsychopharmacology 2004, 29, 1221–1229. [Google Scholar] [CrossRef]
  189. Patterson, P.H. Immune involvement in schizophrenia and autism: Etiology, pathology and animal models. Behav. Brain Res. 2009, 204, 313–321. [Google Scholar] [CrossRef]
  190. Goldstein, J.M.; Cherkerzian, S.; Seidman, L.J.; Donatelli, J.-A.L.; Remington, A.G.; Tsuang, M.T.; Hornig, M.; Buka, S.L. Prenatal maternal immune disruption and sex-dependent risk for psychoses. Psychol. Med. 2014, 44, 3249–3261. [Google Scholar] [CrossRef] [Green Version]
  191. Mattei, D.; Ivanov, A.; Ferrai, C.; Jordan, P.; Guneykaya, D.; Buonfiglioli, A.; Schaafsma, W.; Przanowski, P.; Deuther-Conrad, W.; Brust, P.; et al. Maternal immune activation results in complex microglial transcriptome signature in the adult offspring that is reversed by minocycline treatment. Transl. Psychiatry 2017, 7, e1120. [Google Scholar] [CrossRef] [PubMed]
  192. Luchicchi, A.; Lecca, S.; Melis, M.; de Felice, M.; Cadeddu, F.; Frau, R.; Muntoni, A.L.; Fadda, P.; Devoto, P.; Pistis, M. Maternal Immune Activation Disrupts Dopamine System in the Offspring. Int. J. Neuropsychopharmacol. 2016, 19, pyw007. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Hui, C.W.; St-Pierre, A.; El Hajj, H.; Remy, Y.; Hébert, S.S.; Luheshi, G.N.; Srivastava, L.K.; Tremblay, M.-È. Prenatal Immune Challenge in Mice Leads to Partly Sex-Dependent Behavioral, Microglial, and Molecular Abnormalities Associated with Schizophrenia. Front. Mol. Neurosci. 2018, 11, 13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Ding, S.; Hu, Y.; Luo, B.; Cai, Y.; Hao, K.; Yang, Y.; Zhang, Y.; Wang, X.; Ding, M.; Zhang, H.; et al. Age-related changes in neuroinflammation and prepulse inhibition in offspring of rats treated with Poly I:C in early gestation. Behav. Brain Funct. 2019, 15, 3. [Google Scholar] [CrossRef]
  195. Smith, S.E.P.; Li, J.; Garbett, K.; Mirnics, K.; Patterson, P.H. Maternal immune activation alters fetal brain development through interleukin-6. J. Neurosci. 2007, 27, 10695–10702. [Google Scholar] [CrossRef] [Green Version]
  196. Wolff, A.R.; Bilkey, D.K. Immune activation during mid-gestation disrupts sensorimotor gating in rat offspring. Behav. Brain Res. 2008, 190, 156–159. [Google Scholar] [CrossRef]
  197. Eßlinger, M.; Wachholz, S.; Manitz, M.-P.; Plümper, J.; Sommer, R.; Juckel, G.; Friebe, A. Schizophrenia associated sensory gating deficits develop after adolescent microglia activation. Brain Behav. Immun. 2016, 58, 99–106. [Google Scholar] [CrossRef]
  198. Zuckerman, L.; Rehavi, M.; Nachman, R.; Weiner, I. Immune activation during pregnancy in rats leads to a postpubertal emergence of disrupted latent inhibition, dopaminergic hyperfunction, and altered limbic morphology in the offspring: A novel neurodevelopmental model of schizophrenia. Neuropsychopharmacology 2003, 28, 1778–1789. [Google Scholar] [CrossRef] [Green Version]
  199. Wu, Y.; Qi, F.; Song, D.; He, Z.; Zuo, Z.; Yang, Y.; Liu, Q.; Hu, S.; Wang, X.; Zheng, X.; et al. Prenatal influenza vaccination rescues impairments of social behavior and lamination in a mouse model of autism. J. Neuroinflammation 2018, 15, 228. [Google Scholar] [CrossRef]
  200. Fernández de Cossío, L.; Guzmán, A.; van der Veldt, S.; Luheshi, G.N. Prenatal infection leads to ASD-like behavior and altered synaptic pruning in the mouse offspring. Brain Behav. Immun. 2017, 63, 88–98. [Google Scholar] [CrossRef]
  201. Bauman, M.D.; Iosif, A.-M.; Smith, S.E.P.; Bregere, C.; Amaral, D.G.; Patterson, P.H. Activation of the maternal immune system during pregnancy alters behavioral development of rhesus monkey offspring. Biol. Psychiatry 2014, 75, 332–341. [Google Scholar] [CrossRef] [Green Version]
  202. Weir, R.K.; Forghany, R.; Smith, S.E.P.; Patterson, P.H.; McAllister, A.K.; Schumann, C.M.; Bauman, M.D. Preliminary evidence of neuropathology in nonhuman primates prenatally exposed to maternal immune activation. Brain Behav. Immun. 2015, 48, 139–146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  203. Zhang, Z.; van Praag, H. Maternal immune activation differentially impacts mature and adult-born hippocampal neurons in male mice. Brain Behav. Immun. 2015, 45, 60–70. [Google Scholar] [CrossRef] [PubMed]
  204. Winter, C.; Djodari-Irani, A.; Sohr, R.; Morgenstern, R.; Feldon, J.; Juckel, G.; Meyer, U. Prenatal immune activation leads to multiple changes in basal neurotransmitter levels in the adult brain: Implications for brain disorders of neurodevelopmental origin such as schizophrenia. Int. J. Neuropsychopharmacol. 2009, 12, 513–524. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  205. Lowe, G.C.; Luheshi, G.N.; Williams, S. Maternal infection and fever during late gestation are associated with altered synaptic transmission in the hippocampus of juvenile offspring rats. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2008, 295, R1563–R1571. [Google Scholar] [CrossRef] [Green Version]
  206. Ito, H.T.; Smith, S.E.P.; Hsiao, E.; Patterson, P.H. Maternal immune activation alters nonspatial information processing in the hippocampus of the adult offspring. Brain Behav. Immun. 2010, 24, 930–941. [Google Scholar] [CrossRef] [Green Version]
  207. Wegrzyn, D.; Manitz, M.-P.; Kostka, M.; Freund, N.; Juckel, G.; Faissner, A. Poly I:C-induced maternal immune challenge reduces perineuronal net area and raises spontaneous network activity of hippocampal neurons in vitro. Eur. J. Neurosci. 2020, 53, 3920–3941. [Google Scholar] [CrossRef]
  208. Celio, M.R.; Blümcke, I. Perineuronal nets—A specialized form of extracellular matrix in the adult nervous system. Brain Res. Brain Res. Rev. 1994, 19, 128–145. [Google Scholar] [CrossRef]
  209. Baghel, M.S.; Singh, B.; Dhuriya, Y.K.; Shukla, R.K.; Patro, N.; Khanna, V.K.; Patro, I.K.; Thakur, M.K. Postnatal exposure to poly (I:C) impairs learning and memory through changes in synaptic plasticity gene expression in developing rat brain. Neurobiol. Learn. Mem. 2018, 155, 379–389. [Google Scholar] [CrossRef]
  210. Mattei, D.; Djodari-Irani, A.; Hadar, R.; Pelz, A.; de Cossío, L.F.; Goetz, T.; Matyash, M.; Kettenmann, H.; Winter, C.; Wolf, S.A. Minocycline rescues decrease in neurogenesis, increase in microglia cytokines and deficits in sensorimotor gating in an animal model of schizophrenia. Brain Behav. Immun. 2014, 38, 175–184. [Google Scholar] [CrossRef] [Green Version]
  211. Zhu, F.; Zheng, Y.; Liu, Y.; Zhang, X.; Zhao, J. Minocycline alleviates behavioral deficits and inhibits microglial activation in the offspring of pregnant mice after administration of polyriboinosinic-polyribocytidilic acid. Psychiatry Res. 2014, 219, 680–686. [Google Scholar] [CrossRef]
  212. Reisinger, S.; Khan, D.; Kong, E.; Berger, A.; Pollak, A.; Pollak, D.D. The poly(I:C)-induced maternal immune activation model in preclinical neuropsychiatric drug discovery. Pharmacol. Ther. 2015, 149, 213–226. [Google Scholar] [CrossRef] [Green Version]
  213. Juckel, G.; Manitz, M.P.; Brüne, M.; Friebe, A.; Heneka, M.T.; Wolf, R.J. Microglial activation in a neuroinflammational animal model of schizophrenia--a pilot study. Schizophr. Res. 2011, 131, 96–100. [Google Scholar] [CrossRef]
  214. Miller, B.J.; Herzig, K.-H.; Jokelainen, J.; Karhu, T.; Keinänen-Kiukaanniemi, S.; Järvelin, M.-R.; Veijola, J.; Viinamäki, H.; Päivikki, T.; Jääskeläinen, E.; et al. Inflammation, hippocampal volume, and cognition in schizophrenia: Results from the Northern Finland Birth Cohort 1966. Eur. Arch. Psychiatry Clin. Neurosci. 2020, 271, 609–622. [Google Scholar] [CrossRef]
  215. Dahlgren, J.; Samuelsson, A.-M.; Jansson, T.; Holmäng, A. Interleukin-6 in the maternal circulation reaches the rat fetus in mid-gestation. Pediatr. Res. 2006, 60, 147–151. [Google Scholar] [CrossRef] [Green Version]
  216. Zhao, X.; Mohammed, R.; Tran, H.; Erickson, M.; Kentner, A.C. Poly (I:C)-induced maternal immune activation modifies ventral hippocampal regulation of stress reactivity: Prevention by environmental enrichment. Brain Behav. Immun. 2021, 95, 203–215. [Google Scholar] [CrossRef]
  217. Noonan, L.R.; Caldwell, J.D.; Li, L.; Walker, C.H.; Pedersen, C.A.; Mason, G.A. Neonatal stress transiently alters the development of hippocampal oxytocin receptors. Brain Res. Dev. Brain Res. 1994, 80, 115–120. [Google Scholar] [CrossRef]
  218. Lodge, D.J. The MAM rodent model of schizophrenia. Curr. Protoc. Neurosci. 2013, 63, 9.43.1–9.43.7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  219. Featherstone, R.E.; Rizos, Z.; Nobrega, J.N.; Kapur, S.; Fletcher, P.J. Gestational methylazoxymethanol acetate treatment impairs select cognitive functions: Parallels to schizophrenia. Neuropsychopharmacology 2007, 32, 483–492. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  220. Moore, H.; Jentsch, J.D.; Ghajarnia, M.; Geyer, M.A.; Grace, A.A. A neurobehavioral systems analysis of adult rats exposed to methylazoxymethanol acetate on E17: Implications for the neuropathology of schizophrenia. Biol. Psychiatry 2006, 60, 253–264. [Google Scholar] [CrossRef] [Green Version]
  221. Hradetzky, E.; Sanderson, T.M.; Tsang, T.M.; Sherwood, J.L.; Fitzjohn, S.M.; Lakics, V.; Malik, N.; Schoeffmann, S.; O’Neill, M.J.; Cheng, T.M.; et al. The methylazoxymethanol acetate (MAM-E17) rat model: Molecular and functional effects in the hippocampus. Neuropsychopharmacology 2012, 37, 364–377. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  222. Lodge, D.J.; Grace, A.A. Aberrant hippocampal activity underlies the dopamine dysregulation in an animal model of schizophrenia. J. Neurosci. 2007, 27, 11424–11430. [Google Scholar] [CrossRef] [PubMed]
  223. Lodge, D.J.; Grace, A.A. Gestational methylazoxymethanol acetate administration alters proteomic and metabolomic markers of hippocampal glutamatergic transmission. Neuropsychopharmacology 2012, 37, 319–320. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  224. Lodge, D.J.; Behrens, M.M.; Grace, A.A. A loss of parvalbumin-containing interneurons is associated with diminished oscillatory activity in an animal model of schizophrenia. J. Neurosci. 2009, 29, 2344–2354. [Google Scholar] [CrossRef]
  225. Chalkiadaki, K.; Velli, A.; Kyriazidis, E.; Stavroulaki, V.; Vouvoutsis, V.; Chatzaki, E.; Aivaliotis, M.; Sidiropoulou, K. Development of the MAM model of schizophrenia in mice: Sex similarities and differences of hippocampal and prefrontal cortical function. Neuropharmacology 2019, 144, 193–207. [Google Scholar] [CrossRef]
  226. Du, Y.; Grace, A.A. Loss of Parvalbumin in the Hippocampus of MAM Schizophrenia Model Rats Is Attenuated by Peripubertal Diazepam. Int. J. Neuropsychopharmacol. 2016, 19, pyw065. [Google Scholar] [CrossRef] [Green Version]
  227. Du, Y.; Grace, A.A. Peripubertal diazepam administration prevents the emergence of dopamine system hyperresponsivity in the MAM developmental disruption model of schizophrenia. Neuropsychopharmacology 2013, 38, 1881–1888. [Google Scholar] [CrossRef]
  228. Zhu, X.; Grace, A.A. Prepubertal Environmental Enrichment Prevents Dopamine Dysregulation and Hippocampal Hyperactivity in MAM Schizophrenia Model Rats. Biol. Psychiatry 2021, 89, 298–307. [Google Scholar] [CrossRef]
  229. Lee, G.; Zhou, Y. NMDAR Hypofunction Animal Models of Schizophrenia. Front. Mol. Neurosci. 2019, 12, 185. [Google Scholar] [CrossRef] [Green Version]
  230. Bondi, C.; Matthews, M.; Moghaddam, B. Glutamatergic animal models of schizophrenia. Curr. Pharm. Des. 2012, 18, 1593–1604. [Google Scholar] [CrossRef]
  231. Frohlich, J.; van Horn, J.D. Reviewing the ketamine model for schizophrenia. J. Psychopharmacol. 2014, 28, 287–302. [Google Scholar] [CrossRef] [PubMed]
  232. Kayser, M.S.; Dalmau, J. Anti-NMDA Receptor Encephalitis, Autoimmunity, and Psychosis. Focus 2016, 14, 510–515. [Google Scholar] [CrossRef] [PubMed]
  233. Omdal, R.; Brokstad, K.; Waterloo, K.; Koldingsnes, W.; Jonsson, R.; Mellgren, S.I. Neuropsychiatric disturbances in SLE are associated with antibodies against NMDA receptors. Eur. J. Neurol. 2005, 12, 392–398. [Google Scholar] [CrossRef] [PubMed]
  234. Javitt, D.C.; Zukin, S.R. Recent advances in the phencyclidine model of schizophrenia. Am. J. Psychiatry 1991, 148, 1301–1308. [Google Scholar] [CrossRef] [PubMed]
  235. Carlsson, M.; Carlsson, A. The NMDA antagonist MK-801 causes marked locomotor stimulation in monoamine-depleted mice. J. Neural Transm. 1989, 75, 221–226. [Google Scholar] [CrossRef] [PubMed]
  236. Lopes-Aguiar, C.; Ruggiero, R.N.; Rossignoli, M.T.; Esteves, I.d.M.; Peixoto-Santos, J.E.; Romcy-Pereira, R.N.; Leite, J.P. Long-term potentiation prevents ketamine-induced aberrant neurophysiological dynamics in the hippocampus-prefrontal cortex pathway in vivo. Sci. Rep. 2020, 10, 7167. [Google Scholar] [CrossRef]
  237. Fujikawa, R.; Yamada, J.; Jinno, S. Subclass imbalance of parvalbumin-expressing GABAergic neurons in the hippocampus of a mouse ketamine model for schizophrenia, with reference to perineuronal nets. Schizophr. Res. 2020, 229, 80–93. [Google Scholar] [CrossRef]
  238. Kaushik, R.; Lipachev, N.; Matuszko, G.; Kochneva, A.; Dvoeglazova, A.; Becker, A.; Paveliev, M.; Dityatev, A. Fine structure analysis of perineuronal nets in the ketamine model of schizophrenia. Eur. J. Neurosci. 2020, 53, 3988–4004. [Google Scholar] [CrossRef]
  239. Bast, T.; Zhang, W.N.; Heidbreder, C.; Feldon, J. Hyperactivity and disruption of prepulse inhibition induced by N-methyl-D-aspartate stimulation of the ventral hippocampus and the effects of pretreatment with haloperidol and clozapine. Neuroscience 2001, 103, 325–335. [Google Scholar] [CrossRef]
  240. Howland, J.G.; MacKenzie, E.M.; Yim, T.T.; Taepavarapruk, P.; Phillips, A.G. Electrical stimulation of the hippocampus disrupts prepulse inhibition in rats: Frequency- and site-dependent effects. Behav. Brain Res. 2004, 152, 187–197. [Google Scholar] [CrossRef]
  241. San-Martin, R.; Castro, L.A.; Menezes, P.R.; Fraga, F.J.; Simões, P.W.; Salum, C. Meta-Analysis of Sensorimotor Gating Deficits in Patients With Schizophrenia Evaluated by Prepulse Inhibition Test. Schizophr. Bull. 2020, 46, 1482–1497. [Google Scholar] [CrossRef] [PubMed]
  242. Swerdlow, N.R.; Shoemaker, J.M.; Noh, H.R.; Ma, L.; Gaudet, I.; Munson, M.; Crain, S.; Auerbach, P.P. The ventral hippocampal regulation of prepulse inhibition and its disruption by apomorphine in rats are not mediated via the fornix. Neuroscience 2004, 123, 675–685. [Google Scholar] [CrossRef] [PubMed]
  243. Segev, A.; Yanagi, M.; Scott, D.; Southcott, S.A.; Lister, J.M.; Tan, C.; Li, W.; Birnbaum, S.G.; Kourrich, S.; Tamminga, C.A. Reduced GluN1 in mouse dentate gyrus is associated with CA3 hyperactivity and psychosis-like behaviors. Mol. Psychiatry 2020, 25, 2832–2843. [Google Scholar] [CrossRef]
  244. Alvarez, R.J.; Pafundo, D.E.; Zold, C.L.; Belforte, J.E. Interneuron NMDA Receptor Ablation Induces Hippocampus-Prefrontal Cortex Functional Hypoconnectivity after Adolescence in a Mouse Model of Schizophrenia. J. Neurosci. 2020, 40, 3304–3317. [Google Scholar] [CrossRef] [PubMed]
  245. Grimm, C.M.; Aksamaz, S.; Schulz, S.; Teutsch, J.; Sicinski, P.; Liss, B.; Kätzel, D. Schizophrenia-related cognitive dysfunction in the Cyclin-D2 knockout mouse model of ventral hippocampal hyperactivity. Transl. Psychiatry 2018, 8, 212. [Google Scholar] [CrossRef] [Green Version]
  246. Peled, A. Optogenetic neuronal control in schizophrenia. Med. Hypotheses 2011, 76, 914–921. [Google Scholar] [CrossRef]
  247. Wolff, A.R.; Bygrave, A.M.; Sanderson, D.J.; Boyden, E.S.; Bannerman, D.M.; Kullmann, D.M.; Kätzel, D. Optogenetic induction of the schizophrenia-related endophenotype of ventral hippocampal hyperactivity causes rodent correlates of positive and cognitive symptoms. Sci. Rep. 2018, 8, 12871. [Google Scholar] [CrossRef]
  248. Fan, Z.-L.; Wu, B.; Wu, G.-Y.; Yao, J.; Li, X.; Hu, K.-H.; Zhou, Z.-H.; Sui, J.-F. Optogenetic inhibition of ventral hippocampal neurons alleviates associative motor learning dysfunction in a rodent model of schizophrenia. PLoS ONE 2019, 14, e0227200. [Google Scholar] [CrossRef]
  249. Bauer, J.P.; Rader, S.L.; Joffe, M.E.; Kwon, W.; Quay, J.; Seanez, L.; Zhou, C.; Conn, P.J.; Lewis, A.S. Modeling intrahippocampal effects of anterior hippocampal hyperactivity relevant to schizophrenia using chemogenetic excitation of long axis-projecting mossy cells in the mouse dentate gyrus. Biol. Psychiatry Glob. Open Sci. 2021, 1, 101–111. [Google Scholar] [CrossRef]
  250. Gonzalez-Burgos, G.; Lewis, D.A. GABA neurons and the mechanisms of network oscillations: Implications for understanding cortical dysfunction in schizophrenia. Schizophr. Bull. 2008, 34, 944–961. [Google Scholar] [CrossRef] [Green Version]
  251. Konradi, C.; Yang, C.K.; Zimmerman, E.I.; Lohmann, K.M.; Gresch, P.; Pantazopoulos, H.; Berretta, S.; Heckers, S. Hippocampal interneurons are abnormal in schizophrenia. Schizophr. Res. 2011, 131, 165–173. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  252. Zhang, Z.; Sun, J.; Reynolds, G.P. A selective reduction in the relative density of parvalbumin-immunoreactive neurons in the hippocampus in schizophrenia patients. Chin. Med. J. 2002, 115, 819–823. [Google Scholar] [PubMed]
  253. Knable, M.B.; Barci, B.M.; Webster, M.J.; Meador-Woodruff, J.; Torrey, E.F. Molecular abnormalities of the hippocampus in severe psychiatric illness: Postmortem findings from the Stanley Neuropathology Consortium. Mol. Psychiatry 2004, 9, 609–620; 544. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  254. Antonoudiou, P.; Tan, Y.L.; Kontou, G.; Upton, A.L.; Mann, E.O. Parvalbumin and Somatostatin Interneurons Contribute to the Generation of Hippocampal Gamma Oscillations. J. Neurosci. 2020, 40, 7668–7687. [Google Scholar] [CrossRef]
  255. Craig, M.T.; McBain, C.J. Fast gamma oscillations are generated intrinsically in CA1 without the involvement of fast-spiking basket cells. J. Neurosci. 2015, 35, 3616–3624. [Google Scholar] [CrossRef] [Green Version]
  256. Cardin, J.A.; Carlén, M.; Meletis, K.; Knoblich, U.; Zhang, F.; Deisseroth, K.; Tsai, L.-H.; Moore, C.I. Driving fast-spiking cells induces gamma rhythm and controls sensory responses. Nature 2009, 459, 663–667. [Google Scholar] [CrossRef] [Green Version]
  257. Sohal, V.S.; Zhang, F.; Yizhar, O.; Deisseroth, K. Parvalbumin neurons and gamma rhythms enhance cortical circuit performance. Nature 2009, 459, 698–702. [Google Scholar] [CrossRef] [Green Version]
  258. Spencer, K.M.; Niznikiewicz, M.A.; Shenton, M.E.; McCarley, R.W. Sensory-evoked gamma oscillations in chronic schizophrenia. Biol. Psychiatry 2008, 63, 744–747. [Google Scholar] [CrossRef] [Green Version]
  259. Ferrarelli, F.; Massimini, M.; Peterson, M.J.; Riedner, B.A.; Lazar, M.; Murphy, M.J.; Huber, R.; Rosanova, M.; Alexander, A.L.; Kalin, N.; et al. Reduced evoked gamma oscillations in the frontal cortex in schizophrenia patients: A TMS/EEG study. Am. J. Psychiatry 2008, 165, 996–1005. [Google Scholar] [CrossRef]
  260. Gallinat, J.; Winterer, G.; Herrmann, C.S.; Senkowski, D. Reduced oscillatory gamma-band responses in unmedicated schizophrenic patients indicate impaired frontal network processing. Clin. Neurophysiol. 2004, 115, 1863–1874. [Google Scholar] [CrossRef]
  261. Farzan, F.; Barr, M.S.; Levinson, A.J.; Chen, R.; Wong, W.; Fitzgerald, P.B.; Daskalakis, Z.J. Evidence for gamma inhibition deficits in the dorsolateral prefrontal cortex of patients with schizophrenia. Brain 2010, 133, 1505–1514. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  262. Lewis, D.A.; Curley, A.A.; Glausier, J.R.; Volk, D.W. Cortical parvalbumin interneurons and cognitive dysfunction in schizophrenia. Trends Neurosci. 2012, 35, 57–67. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  263. Nguyen, R.; Morrissey, M.D.; Mahadevan, V.; Cajanding, J.D.; Woodin, M.A.; Yeomans, J.S.; Takehara-Nishiuchi, K.; Kim, J.C. Parvalbumin and GAD65 interneuron inhibition in the ventral hippocampus induces distinct behavioral deficits relevant to schizophrenia. J. Neurosci. 2014, 34, 14948–14960. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  264. Molina, J.L.; Thomas, M.L.; Joshi, Y.B.; Hochberger, W.C.; Koshiyama, D.; Nungaray, J.A.; Cardoso, L.; Sprock, J.; Braff, D.L.; Swerdlow, N.R.; et al. Gamma oscillations predict pro-cognitive and clinical response to auditory-based cognitive training in schizophrenia. Transl. Psychiatry 2020, 10, 405. [Google Scholar] [CrossRef]
  265. Yi, Y.; Song, Y.; Lu, Y. Parvalbumin Interneuron Activation-Dependent Adult Hippocampal Neurogenesis Is Required for Treadmill Running to Reverse Schizophrenia-Like Phenotypes. Front. Cell Dev. Biol. 2020, 8, 24. [Google Scholar] [CrossRef] [Green Version]
  266. Donegan, J.J.; Tyson, J.A.; Branch, S.Y.; Beckstead, M.J.; Anderson, S.A.; Lodge, D.J. Stem cell-derived interneuron transplants as a treatment for schizophrenia: Preclinical validation in a rodent model. Mol. Psychiatry 2017, 22, 1492–1501. [Google Scholar] [CrossRef] [Green Version]
  267. Perez, S.M.; Lodge, D.J. Hippocampal interneuron transplants reverse aberrant dopamine system function and behavior in a rodent model of schizophrenia. Mol. Psychiatry 2013, 18, 1193–1198. [Google Scholar] [CrossRef] [Green Version]
  268. Gilani, A.I.; Chohan, M.O.; Inan, M.; Schobel, S.A.; Chaudhury, N.H.; Paskewitz, S.; Chuhma, N.; Glickstein, S.; Merker, R.J.; Xu, Q.; et al. Interneuron precursor transplants in adult hippocampus reverse psychosis-relevant features in a mouse model of hippocampal disinhibition. Proc. Natl. Acad. Sci. USA 2014, 111, 7450–7455. [Google Scholar] [CrossRef] [Green Version]
  269. Southwell, D.G.; Seifikar, H.; Malik, R.; Lavi, K.; Vogt, D.; Rubenstein, J.L.; Sohal, V.S. Interneuron Transplantation Rescues Social Behavior Deficits without Restoring Wild-Type Physiology in a Mouse Model of Autism with Excessive Synaptic Inhibition. J. Neurosci. 2020, 40, 2215–2227. [Google Scholar] [CrossRef]
  270. Xia, Y.; Zhang, Z.; Lin, W.; Yan, J.; Zhu, C.; Yin, D.; He, S.; Su, Y.; Xu, N.; Caldwell, R.W.; et al. Modulating microglia activation prevents maternal immune activation induced schizophrenia-relevant behavior phenotypes via arginase 1 in the dentate gyrus. Neuropsychopharmacology 2020, 45, 1896–1908. [Google Scholar] [CrossRef]
  271. Carstens, K.E.; Phillips, M.L.; Pozzo-Miller, L.; Weinberg, R.J.; Dudek, S.M. Perineuronal Nets Suppress Plasticity of Excitatory Synapses on CA2 Pyramidal Neurons. J. Neurosci. 2016, 36, 6312–6320. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  272. O’Connor, A.M.; Burton, T.J.; Mansuri, H.; Hand, G.R.; Leamey, C.A.; Sawatari, A. Environmental Enrichment From Birth Impacts Parvalbumin Expressing Cells and Wisteria Floribunda Agglutinin Labelled Peri-Neuronal Nets Within the Developing Murine Striatum. Front. Neuroanat. 2019, 13, 90. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  273. Celio, M.R.; Spreafico, R.; de Biasi, S.; Vitellaro-Zuccarello, L. Perineuronal nets: Past and present. Trends Neurosci. 1998, 21, 510–515. [Google Scholar] [CrossRef]
  274. Pizzorusso, T.; Medini, P.; Berardi, N.; Chierzi, S.; Fawcett, J.W.; Maffei, L. Reactivation of ocular dominance plasticity in the adult visual cortex. Science 2002, 298, 1248–1251. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  275. Gogolla, N.; Caroni, P.; Lüthi, A.; Herry, C. Perineuronal nets protect fear memories from erasure. Science 2009, 325, 1258–1261. [Google Scholar] [CrossRef] [Green Version]
  276. Frischknecht, R.; Heine, M.; Perrais, D.; Seidenbecher, C.I.; Choquet, D.; Gundelfinger, E.D. Brain extracellular matrix affects AMPA receptor lateral mobility and short-term synaptic plasticity. Nat. Neurosci. 2009, 12, 897–904. [Google Scholar] [CrossRef]
  277. Gottschling, C.; Wegrzyn, D.; Denecke, B.; Faissner, A. Elimination of the four extracellular matrix molecules tenascin-C, tenascin-R, brevican and neurocan alters the ratio of excitatory and inhibitory synapses. Sci. Rep. 2019, 9, 13939. [Google Scholar] [CrossRef] [Green Version]
  278. Lensjø, K.K.; Lepperød, M.E.; Dick, G.; Hafting, T.; Fyhn, M. Removal of Perineuronal Nets Unlocks Juvenile Plasticity Through Network Mechanisms of Decreased Inhibition and Increased Gamma Activity. J. Neurosci. 2017, 37, 1269–1283. [Google Scholar] [CrossRef] [Green Version]
  279. Pizzorusso, T.; Medini, P.; Landi, S.; Baldini, S.; Berardi, N.; Maffei, L. Structural and functional recovery from early monocular deprivation in adult rats. Proc. Natl. Acad. Sci. USA 2006, 103, 8517–8522. [Google Scholar] [CrossRef] [Green Version]
  280. Cabungcal, J.-H.; Steullet, P.; Morishita, H.; Kraftsik, R.; Cuenod, M.; Hensch, T.K.; Do, K.Q. Perineuronal nets protect fast-spiking interneurons against oxidative stress. Proc. Natl. Acad. Sci. USA 2013, 110, 9130–9135. [Google Scholar] [CrossRef] [Green Version]
  281. Härtig, W.; Derouiche, A.; Welt, K.; Brauer, K.; Grosche, J.; Mäder, M.; Reichenbach, A.; Brückner, G. Cortical neurons immunoreactive for the potassium channel Kv3.1b subunit are predominantly surrounded by perineuronal nets presumed as a buffering system for cations. Brain Res. 1999, 842, 15–29. [Google Scholar] [CrossRef]
  282. Morawski, M.; Brückner, M.K.; Riederer, P.; Brückner, G.; Arendt, T. Perineuronal nets potentially protect against oxidative stress. Exp. Neurol. 2004, 188, 309–315. [Google Scholar] [CrossRef] [PubMed]
  283. Morawski, M.; Reinert, T.; Meyer-Klaucke, W.; Wagner, F.E.; Tröger, W.; Reinert, A.; Jäger, C.; Brückner, G.; Arendt, T. Ion exchanger in the brain: Quantitative analysis of perineuronally fixed anionic binding sites suggests diffusion barriers with ion sorting properties. Sci. Rep. 2015, 5, 16471. [Google Scholar] [CrossRef] [PubMed]
  284. Enwright, J.F.; Sanapala, S.; Foglio, A.; Berry, R.; Fish, K.N.; Lewis, D.A. Reduced Labeling of Parvalbumin Neurons and Perineuronal Nets in the Dorsolateral Prefrontal Cortex of Subjects with Schizophrenia. Neuropsychopharmacology 2016, 41, 2206–2214. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  285. Mauney, S.A.; Athanas, K.M.; Pantazopoulos, H.; Shaskan, N.; Passeri, E.; Berretta, S.; Woo, T.-U.W. Developmental pattern of perineuronal nets in the human prefrontal cortex and their deficit in schizophrenia. Biol. Psychiatry 2013, 74, 427–435. [Google Scholar] [CrossRef] [Green Version]
  286. Pantazopoulos, H.; Woo, T.-U.W.; Lim, M.P.; Lange, N.; Berretta, S. Extracellular matrix-glial abnormalities in the amygdala and entorhinal cortex of subjects diagnosed with schizophrenia. Arch. Gen. Psychiatry 2010, 67, 155–166. [Google Scholar] [CrossRef] [Green Version]
  287. Pantazopoulos, H.; Markota, M.; Jaquet, F.; Ghosh, D.; Wallin, A.; Santos, A.; Caterson, B.; Berretta, S. Aggrecan and chondroitin-6-sulfate abnormalities in schizophrenia and bipolar disorder: A postmortem study on the amygdala. Transl. Psychiatry 2015, 5, e496. [Google Scholar] [CrossRef] [Green Version]
  288. Kilonzo, V.W.; Sweet, R.A.; Glausier, J.R.; Pitts, M.W. Deficits in Glutamic Acid Decarboxylase 67 Immunoreactivity, Parvalbumin Interneurons, and Perineuronal Nets in the Inferior Colliculus of Subjects with Schizophrenia. Schizophr. Bull. 2020, 46, 1053–1059. [Google Scholar] [CrossRef]
  289. Alcaide, J.; Guirado, R.; Crespo, C.; Blasco-Ibáñez, J.M.; Varea, E.; Sanjuan, J.; Nacher, J. Alterations of perineuronal nets in the dorsolateral prefrontal cortex of neuropsychiatric patients. Int. J. Bipolar Disord. 2019, 7, 24. [Google Scholar] [CrossRef]
  290. Wen, T.H.; Binder, D.K.; Ethell, I.M.; Razak, K.A. The Perineuronal ‘Safety’ Net? Perineuronal Net Abnormalities in Neurological Disorders. Front. Mol. Neurosci. 2018, 11, 270. [Google Scholar] [CrossRef]
  291. Berretta, S.; Pantazopoulos, H.; Markota, M.; Brown, C.; Batzianouli, E.T. Losing the sugar coating: Potential impact of perineuronal net abnormalities on interneurons in schizophrenia. Schizophr. Res. 2015, 167, 18–27. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  292. Bitanihirwe, B.K.Y.; Woo, T.-U.W. Perineuronal nets and schizophrenia: The importance of neuronal coatings. Neurosci. Biobehav. Rev. 2014, 45, 85–99. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  293. Bitanihirwe, B.K.Y.; Mauney, S.A.; Woo, T.-U.W. Weaving a Net of Neurobiological Mechanisms in Schizophrenia and Unraveling the Underlying Pathophysiology. Biol. Psychiatry 2016, 80, 589–598. [Google Scholar] [CrossRef] [Green Version]
  294. Sorg, B.A.; Berretta, S.; Blacktop, J.M.; Fawcett, J.W.; Kitagawa, H.; Kwok, J.C.F.; Miquel, M. Casting a Wide Net: Role of Perineuronal Nets in Neural Plasticity. J. Neurosci. 2016, 36, 11459–11468. [Google Scholar] [CrossRef] [Green Version]
  295. Klimczak, P.; Rizzo, A.; Castillo-Gómez, E.; Perez-Rando, M.; Gramuntell, Y.; Beltran, M.; Nacher, J. Parvalbumin Interneurons and Perineuronal Nets in the Hippocampus and Retrosplenial Cortex of Adult Male Mice After Early Social Isolation Stress and Perinatal NMDA Receptor Antagonist Treatment. Front. Synaptic Neurosci. 2021, 13, 733989. [Google Scholar] [CrossRef]
  296. Cope, E.C.; Zych, A.D.; Katchur, N.J.; Waters, R.C.; Laham, B.J.; Diethorn, E.J.; Park, C.Y.; Meara, W.R.; Gould, E. Atypical perineuronal nets in the CA2 region interfere with social memory in a mouse model of social dysfunction. Mol. Psychiatry 2021, 1–5. [Google Scholar] [CrossRef]
  297. Christensen, A.C.; Lensjø, K.K.; Lepperød, M.E.; Dragly, S.-A.; Sutterud, H.; Blackstad, J.S.; Fyhn, M.; Hafting, T. Perineuronal nets stabilize the grid cell network. Nat. Commun. 2021, 12, 253. [Google Scholar] [CrossRef]
  298. Romberg, C.; Yang, S.; Melani, R.; Andrews, M.R.; Horner, A.E.; Spillantini, M.G.; Bussey, T.J.; Fawcett, J.W.; Pizzorusso, T.; Saksida, L.M. Depletion of perineuronal nets enhances recognition memory and long-term depression in the perirhinal cortex. J. Neurosci. 2013, 33, 7057–7065. [Google Scholar] [CrossRef] [Green Version]
  299. Yang, S.; Cacquevel, M.; Saksida, L.M.; Bussey, T.J.; Schneider, B.L.; Aebischer, P.; Melani, R.; Pizzorusso, T.; Fawcett, J.W.; Spillantini, M.G. Perineuronal net digestion with chondroitinase restores memory in mice with tau pathology. Exp. Neurol. 2015, 265, 48–58. [Google Scholar] [CrossRef] [Green Version]
  300. Xue, Y.-X.; Xue, L.-F.; Liu, J.-F.; He, J.; Deng, J.-H.; Sun, S.-C.; Han, H.-B.; Luo, Y.-X.; Xu, L.-Z.; Wu, P.; et al. Depletion of perineuronal nets in the amygdala to enhance the erasure of drug memories. J. Neurosci. 2014, 34, 6647–6658. [Google Scholar] [CrossRef] [Green Version]
  301. Shah, A.; Lodge, D.J. A loss of hippocampal perineuronal nets produces deficits in dopamine system function: Relevance to the positive symptoms of schizophrenia. Transl. Psychiatry 2013, 3, e215. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  302. Sun, Z.Y.; Bozzelli, P.L.; Caccavano, A.; Allen, M.; Balmuth, J.; Vicini, S.; Wu, J.-Y.; Conant, K. Disruption of perineuronal nets increases the frequency of sharp wave ripple events. Hippocampus 2018, 28, 42–52. [Google Scholar] [CrossRef] [PubMed]
  303. Hayani, H.; Song, I.; Dityatev, A. Increased Excitability and Reduced Excitatory Synaptic Input into Fast-Spiking CA2 Interneurons After Enzymatic Attenuation of Extracellular Matrix. Front. Cell. Neurosci. 2018, 12, 149. [Google Scholar] [CrossRef] [Green Version]
  304. Shi, W.; Wei, X.; Wang, X.; Du, S.; Liu, W.; Song, J.; Wang, Y. Perineuronal nets protect long-term memory by limiting activity-dependent inhibition from parvalbumin interneurons. Proc. Natl. Acad. Sci. USA 2019, 116, 27063–27073. [Google Scholar] [CrossRef] [PubMed]
  305. Wegrzyn, D.; Freund, N.; Faissner, A.; Juckel, G. Poly I:C Activated Microglia Disrupt Perineuronal Nets and Modulate Synaptic Balance in Primary Hippocampal Neurons in vitro. Front. Synaptic Neurosci. 2021, 13, 637549. [Google Scholar] [CrossRef] [PubMed]
  306. Pyka, M.; Wetzel, C.; Aguado, A.; Geissler, M.; Hatt, H.; Faissner, A. Chondroitin sulfate proteoglycans regulate astrocyte-dependent synaptogenesis and modulate synaptic activity in primary embryonic hippocampal neurons. Eur. J. Neurosci. 2011, 33, 2187–2202. [Google Scholar] [CrossRef]
  307. Crapser, J.D.; Arreola, M.A.; Tsourmas, K.I.; Green, K.N. Microglia as hackers of the matrix: Sculpting synapses and the extracellular space. Cell. Mol. Immunol. 2021, 18, 2472–2488. [Google Scholar] [CrossRef]
  308. Crapser, J.D.; Ochaba, J.; Soni, N.; Reidling, J.C.; Thompson, L.M.; Green, K.N. Microglial depletion prevents extracellular matrix changes and striatal volume reduction in a model of Huntington’s disease. Brain 2020, 143, 266–288. [Google Scholar] [CrossRef]
  309. Crapser, J.D.; Spangenberg, E.E.; Barahona, R.A.; Arreola, M.A.; Hohsfield, L.A.; Green, K.N. Microglia facilitate loss of perineuronal nets in the Alzheimer’s disease brain. EBioMedicine 2020, 58, 102919. [Google Scholar] [CrossRef]
  310. Strackeljan, L.; Baczynska, E.; Cangalaya, C.; Baidoe-Ansah, D.; Wlodarczyk, J.; Kaushik, R.; Dityatev, A. Microglia Depletion-Induced Remodeling of Extracellular Matrix and Excitatory Synapses in the Hippocampus of Adult Mice. Cells 2021, 10, 1862. [Google Scholar] [CrossRef]
  311. Dubisova, J.; Burianova, J.S.; Svobodova, L.; Makovicky, P.; Martinez-Varea, N.; Cimpean, A.; Fawcett, J.W.; Kwok, J.C.F.; Kubinova, S. Oral treatment of 4-methylumbelliferone reduced perineuronal nets and improved recognition memory in mice. Brain Res. Bull. 2022, 181, 144–156. [Google Scholar] [CrossRef] [PubMed]
  312. Slaker, M.; Barnes, J.; Sorg, B.A.; Grimm, J.W. Impact of Environmental Enrichment on Perineuronal Nets in the Prefrontal Cortex following Early and Late Abstinence from Sucrose Self-Administration in Rats. PLoS ONE 2016, 11, e0168256. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. An overview of the most common structural hippocampus deviations in schizophrenia. Patients suffering from schizophrenia frequently develop a reduction of the general hippocampal volume, as well as a higher volume asymmetry between the left and right hippocampus. Furthermore, subfield-specific volume changes were especially described for the CA1- and CA2-region. Besides volume reductions, morphological abnormalities are known and include incomplete inversion patterns and inward deformations. Lastly, connectivity studies unraveled an impaired connection between the hippocampus of schizophrenic patients with further parts of the CNS.
Figure 1. An overview of the most common structural hippocampus deviations in schizophrenia. Patients suffering from schizophrenia frequently develop a reduction of the general hippocampal volume, as well as a higher volume asymmetry between the left and right hippocampus. Furthermore, subfield-specific volume changes were especially described for the CA1- and CA2-region. Besides volume reductions, morphological abnormalities are known and include incomplete inversion patterns and inward deformations. Lastly, connectivity studies unraveled an impaired connection between the hippocampus of schizophrenic patients with further parts of the CNS.
Ijms 23 05482 g001
Figure 2. An overview of the functional deviations in the hippocampus of schizophrenia patients. In schizophrenia, the basal hippocampal activity is frequently increased and develops a hypermetabolic state with increased glutamate- and glutamine-levels and a higher cerebellar blood volume (CBV). Additionally, the hippocampo-striatal, as well as the hippocampo-prefrontal pathway, show a reduced functional connectivity.
Figure 2. An overview of the functional deviations in the hippocampus of schizophrenia patients. In schizophrenia, the basal hippocampal activity is frequently increased and develops a hypermetabolic state with increased glutamate- and glutamine-levels and a higher cerebellar blood volume (CBV). Additionally, the hippocampo-striatal, as well as the hippocampo-prefrontal pathway, show a reduced functional connectivity.
Ijms 23 05482 g002
Figure 3. The possible risk factors for hippocampal deviations in schizophrenia. Besides genetic factors that increase the risk for hippocampal alterations, several environmental factors have been identified that directly influence hippocampal integrity. Here, especially prenatal, perinatal and early-life stress have negative effects regarding structural, as well as functional properties of the hippocampus. Furthermore, childhood trauma and inflammatory processes can negatively affect the development and the physiology.
Figure 3. The possible risk factors for hippocampal deviations in schizophrenia. Besides genetic factors that increase the risk for hippocampal alterations, several environmental factors have been identified that directly influence hippocampal integrity. Here, especially prenatal, perinatal and early-life stress have negative effects regarding structural, as well as functional properties of the hippocampus. Furthermore, childhood trauma and inflammatory processes can negatively affect the development and the physiology.
Ijms 23 05482 g003
Figure 4. The intervening strategies that enhance hippocampal deviations in animal models for schizophrenia. Several studies targeted the hippocampus in schizophrenia animal models and observed ameliorating effects induced by different experimental approaches. Here, the transplantation of interneuron and interneuron-precursor transplants resulted in a reduction of the hippocampal hyperactivity [266,267,268]. Additionally, a reduction of the increased hippocampal activity could be achieved using optogenetic techniques [248]. Physical exercise [265], as well as an enhanced environmental enrichment [216,271,272] during the experimental procedures showed beneficial and protecting effects regarding the hippocampal activity. In maternal immune activation models for schizophrenia, a treatment with anti-inflammatory agents enhanced hippocampal parameters in the offspring [270]. Lastly, a manipulation of the extracellular matrix, especially of the interneuron-surrounding perineuronal nets, could be another possible approach for schizophrenia since this neuronal subtype shows disruptions in patients and goes along with impaired gamma-waves.
Figure 4. The intervening strategies that enhance hippocampal deviations in animal models for schizophrenia. Several studies targeted the hippocampus in schizophrenia animal models and observed ameliorating effects induced by different experimental approaches. Here, the transplantation of interneuron and interneuron-precursor transplants resulted in a reduction of the hippocampal hyperactivity [266,267,268]. Additionally, a reduction of the increased hippocampal activity could be achieved using optogenetic techniques [248]. Physical exercise [265], as well as an enhanced environmental enrichment [216,271,272] during the experimental procedures showed beneficial and protecting effects regarding the hippocampal activity. In maternal immune activation models for schizophrenia, a treatment with anti-inflammatory agents enhanced hippocampal parameters in the offspring [270]. Lastly, a manipulation of the extracellular matrix, especially of the interneuron-surrounding perineuronal nets, could be another possible approach for schizophrenia since this neuronal subtype shows disruptions in patients and goes along with impaired gamma-waves.
Ijms 23 05482 g004
Table 1. An overview on the structural abnormalities of the hippocampus in schizophrenia patients.
Table 1. An overview on the structural abnormalities of the hippocampus in schizophrenia patients.
Type of Structural DeviationExperimental ApproachReference
general volume reduction of the hippocampuspostmortem morphometric
measurement
[18]
general volume reduction of the hippocampusCoronal MRI/
high-resolution MRI
[19,21,22,55,56]
volume reduction of the left amygdala/hippocampal
complex (AHC)
Coronal MRI/
high-resolution MRI
[23]
volume reduction of
hippocampal subfields in the left hippocampus
Meta-analysis of
postmortem studies
[25]
higher rate of incomplete hippocampal inversions with a reduced hippocampal
volume
Coronal MRI/
high-resolution MRI
[38,39]
association of visual
hallucinations with specific
inversion patterns
Coronal MRI/
high-resolution MRI
[42]
volume reduction of anterior and midbody CA1- and CA2- regions with increased
peri-hippocampal CSF levels
Coronal MRI/
high-resolution MRI
[20,55]
bilateral inward deformation of the anterior hippocampusCoronal MRI/
high-resolution MRI
[57]
reduction of SV2A-positive synaptic vesicles in the
hippocampus
PET-scan
analysis
[60]
unchanged density of postsynaptic elements in the hippocampusMeta-analysis of
postmortem studies
[63]
reduced PSD-95 levels in the CA1-region/dentate
molecular layer
Immunoblot-analysis of
postmortem samples/
immuno-autoradiography
[26,28]
reduced SAP-102 levels in the hippocampusWestern blot analysis of
postmortem samples
[68]
increased spine density and PSD-95 levels on CA3 pyramidal cell dendritesWestern blot analysis of
postmortem samples/
Golgi staining
[27]
abnormal resting-state
cortico-hippocampal
network coherence
Functional connectivity
analysis
[29]
reduced connectivity
between the hippocampus and the striatum
Functional MRI analysis[31]
functional hypoconnectivity to regions of the default mode network and hyperconnectivity to the lateral
occipital cortex
Connectivity and
Magnetic Resonance
Spectroscopy Study
[32]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Wegrzyn, D.; Juckel, G.; Faissner, A. Structural and Functional Deviations of the Hippocampus in Schizophrenia and Schizophrenia Animal Models. Int. J. Mol. Sci. 2022, 23, 5482. https://doi.org/10.3390/ijms23105482

AMA Style

Wegrzyn D, Juckel G, Faissner A. Structural and Functional Deviations of the Hippocampus in Schizophrenia and Schizophrenia Animal Models. International Journal of Molecular Sciences. 2022; 23(10):5482. https://doi.org/10.3390/ijms23105482

Chicago/Turabian Style

Wegrzyn, David, Georg Juckel, and Andreas Faissner. 2022. "Structural and Functional Deviations of the Hippocampus in Schizophrenia and Schizophrenia Animal Models" International Journal of Molecular Sciences 23, no. 10: 5482. https://doi.org/10.3390/ijms23105482

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