Next Article in Journal
The Receptor Binding Domain of SARS-CoV-2 Lambda Variant Has a Better Chance Than the Delta Variant in Evading BNT162b2 COVID-19 mRNA Vaccine-Induced Humoral Immunity
Previous Article in Journal
Modeling of DNA Damage Repair and Cell Response in Relation to p53 System Exposed to Ionizing Radiation
Previous Article in Special Issue
Insulin-like Growth Factor 2 (IGF-2) and Insulin-like Growth Factor Binding Protein 7 (IGFBP-7) Are Upregulated after Atypical Antipsychotics in Spanish Schizophrenia Patients
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Cytokine Imbalance as a Biomarker of Treatment-Resistant Schizophrenia

by
Natalia A. Shnayder
1,2,*,
Aiperi K. Khasanova
1,
Anna I. Strelnik
3,4,
Mustafa Al-Zamil
5,
Andrey P. Otmakhov
6,7,
Nikolay G. Neznanov
1,
German A. Shipulin
8,
Marina M. Petrova
2,
Natalia P. Garganeeva
9 and
Regina F. Nasyrova
1,3,*
1
Institute of Personalized Psychiatry and Neurology, Shared Core Facilities, V.M. Bekhterev National Medical Research Centre for Psychiatry and Neurology, 192019 Saint Petersburg, Russia
2
Shared Core Facilities “Molecular and Cell Technologies”, V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia
3
International Centre for Education and Research in Neuropsychiatry, Samara State Medical University, 443016 Samara, Russia
4
Department of Psychiatry, Narcology and Psychotherapy, Samara State Medical University, 443016 Samara, Russia
5
Department of Physiotherapy, Faculty of Continuing Medical Education, Peoples’ Friendship University of Russia, 117198 Moscow, Russia
6
Basic Department of Psychological and Social Support, St. Petersburg State Institute of Psychology and Social Work, 199178 Saint Petersburg, Russia
7
St. Nikolay Psychiatric Hospital, 190121 Saint Petersburg, Russia
8
Centre for Strategic Planning and Management of Biomedical Health Risks Management, 119121 Moscow, Russia
9
Department of General Medical Practice and Outpatient Therapy, Siberian State Medical University, 634050 Tomsk, Russia
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(19), 11324; https://doi.org/10.3390/ijms231911324
Submission received: 1 September 2022 / Revised: 19 September 2022 / Accepted: 21 September 2022 / Published: 26 September 2022
(This article belongs to the Special Issue Molecular Advances in Schizophrenia)

Abstract

:
Treatment-resistant schizophrenia (TRS) is an important and unresolved problem in biological and clinical psychiatry. Approximately 30% of cases of schizophrenia (Sch) are TRS, which may be due to the fact that some patients with TRS may suffer from pathogenetically “non-dopamine” Sch, in the development of which neuroinflammation is supposed to play an important role. The purpose of this narrative review is an attempt to summarize the data characterizing the patterns of production of pro-inflammatory and anti-inflammatory cytokines during the development of therapeutic resistance to APs and their pathogenetic and prognostic significance of cytokine imbalance as TRS biomarkers. This narrative review demonstrates that the problem of evaluating the contribution of pro-inflammatory and anti-inflammatory cytokines to maintaining or changing the cytokine balance can become a new key in unlocking the mystery of “non-dopamine” Sch and developing new therapeutic strategies for the treatment of TRS and psychosis in the setting of acute and chronic neuroinflammation. In addition, the inconsistency of the results of previous studies on the role of pro-inflammatory and anti-inflammatory cytokines indicates that the TRS biomarker, most likely, is not the serum level of one or more cytokines, but the cytokine balance. We have confirmed the hypothesis that cytokine imbalance is one of the most important TRS biomarkers. This hypothesis is partially supported by the variable response to immunomodulators in patients with TRS, which were prescribed without taking into account the cytokine balance of the relation between serum levels of the most important pro-inflammatory and anti-inflammatory cytokines for TRS.

1. Introduction

Schizophrenia (Sch) is a common socially significant mental disorder associated with premature mortality and reduced life expectancy of patients [1,2]. Epidemiological studies show that the life expectancy of patients with serious mental disorders, including Sch, is reduced by 7–24 years [3]. This is partly due to the development of serious adverse reactions to drugs (primarily metabolic syndrome caused by antipsychotics (APs)) and therapeutic resistance to APs [4].
Therapeutic resistance (TR) is a condition in which a mental disorder cannot be treated or corrected despite an adequate course of treatment. Currently, the TR problem remains relevant in relation to a wide range of mental disorders: depressive disorder, obsessive-compulsive disorder, bipolar affective disorder, Sch, etc. [5].
Due to the fact that APs of the first and new generations affect different mechanisms of action in the treatment of Sch, the risk of developing TR to some APs of new generations remains high, almost similar to APs of the first generation. This may be due to the fact that the pathophysiological mechanisms of TRS development are more complex than previously thought. All currently available APs are able to antagonize dopamine D2 receptors, and the APs’ therapeutic effects in psychosis are related to their action on the limbic system reducing dopamine transmission [6]. This is confirmed by several reports that therapeutic doses of typical APs block D2 receptors in 70–89% of cases in young adults, while atypical APs block D2 receptors in 38–63% of cases [7]. In addition to the effect of AP on dopaminergic neurons, other possible mechanisms are being considered [6,8].
TR to APs in patients with Sch or treatment-resistant Sch (TRS) is one of the urgent problems of psychiatry and clinical pharmacology and is far from being resolved, despite the development and use in real clinical practice of new generations of APs (Figure 1). The average incidence of TRS occurs in approximately 30% of individuals diagnosed with Sch [9]. However, according to a meta-analysis by Suzuki et al. [10], the frequency of occurrence of TRS varies from 0 to 76%. This may be due to both the difference in the methodology and design of epidemiological studies of TRS, and different definitions of TRS, which have been revised several times. In 2004, the American Psychiatric Association considered TRS to be a minor or no symptomatic response to multiple (at least two) APs given for an adequate duration (at least 6 weeks) and in an adequate therapeutic dose (therapeutic range), while at least one AP must be of a new generation [11]. The 2012 World Federation of Societies for Biological Psychiatry (WFSBP) Biological Treatment of Sch guidelines define TRS as a disorder in which no significant improvement in psychopathological symptoms and/or other target symptoms has been achieved despite treatment with at least two different APs from two different chemical classes (at least one must be an atypical AP) at recommended therapeutic doses for a treatment period of at least 2–8 weeks [12]. The National Institute for Health and Clinical Excellence (NICE) in 2014 defined the criteria for TRS as insufficient response to at least two different consecutively prescribed APs at appropriate doses taken over an appropriate period of time (4–6 weeks); however, at least one AP must be new-generation non-clozapine APs [13]. According to the Diagnostic and Terminology Working Group Guidelines, Treatment Response and Resistance in Psychosis (TRRIP), TRS is considered to be at least moderately severe, with <20% improvement in Sch symptoms, at least moderate functional impairment (based on the appropriate approved scale), and confirmed adherence to APs by measuring the concentration of APs by taking two different APs at adequate therapeutic doses for at least 6 weeks, while at least one AP is a prolonged injectable form [14,15]. In addition to the above criteria, TRS criteria have also been proposed by other Sch treatment guidelines, such as The Texas Treatment Algorithm Project [16] and the International Psychopharmacological Algorithm Project (IPAP, 2006) [17]. All these definitions of TRS are different and subject to a wide range of interpretations, which can lead to inconsistent clinical management and inaccurate treatment [18], as well as variable results of epidemiological studies.
Sch and TRS Clinical Experts in 2017 reviewed the main areas of TRS research. They concluded that the diagnosis of TRS required an inadequate response to two different APs, each taken at an adequate dose and for an adequate duration. In each course of treatment, it is recommended to use objective Sch symptom scores to assess response to APs to ensure adherence to APs therapy. Once no response has been established (after ≥12 weeks for positive symptoms [2 courses of ARs lasting ≥6 weeks]), it is recommended that the Sch treatment plan be reviewed and alternative pharmacological or non-pharmacological treatments be considered [9].
In addition, TRRIP has developed current criteria for defining TRS and a consensus has been reached on “minimum requirements”. However, any definition of TRS must indicate that the Sch patient received an adequate course of APs in terms of dosage (equivalent to or greater than 600 mg chlorpromazine per day), two courses of two different APs lasting 6 weeks (each course) at a therapeutic dose, active control of adherence to treatment (≥80% of prescribed doses), as well as the use of structured clinical assessments to establish the presence and severity of symptoms [20].
The mechanisms of TRS development are heterogeneous (Table 1) and have been actively studied for many years, but there is no single view. Several hypotheses have been proposed (Figure 2), among which the TRS inflammatory hypothesis is of particular scientific and clinical interest, since the use of APs is ineffective in approximately 30% of all patients with Sch. This may be due to the fact that some patients with TRS may suffer from pathogenetically “non-dopamine” Sch [21]. In each case of TRS, it is important to rule out pseudo-resistance, the underlying mechanism of which may lie in an imbalance of pro-inflammatory and anti-inflammatory cytokines (cytokine status).
The purpose of this narrative review is an attempt to summarize the data characterizing the patterns of production of pro-inflammatory and anti-inflammatory cytokines during the development of therapeutic resistance to APs and their pathogenetic and prognostic significance of cytokine imbalance as TRS biomarkers.

2. Pathogenetic Aspect of Inflammation in Treatment Resistance Schizophrenia

There are many factors leading to a chronic neuroinflammatory process in Sch. Research in this area has led to the formation of several mechanisms of TRS (Figure 3), which will be discussed in detail later.

2.1. Changes in the Functional Activity of Microglia in Treatment Resistance Schizophrenia

Microglia account for 10–20% of all cells found in the brain and are an important component of the CNS immune system [47]. Microglia play an important role in neuroinflammation, providing protection in the event of damage or disease to the CNS. When neuroinfection occurs, activation of microglia, synthesis and release of central pro-inflammatory cytokines, which leads to various mental and behavioral disorders [48]. There is now evidence that aging [49], neurodegeneration [50] and stress [51] lead to “sensitization” or “priming” of microglia, which subsequently causes an exaggerated immune response. Exposure of primed microglia to, for example, minor systemic inflammation leads to proliferation and increased production of pro-inflammatory cytokines [52], which, in turn, can exacerbate the immune response in the CNS and be expressed in a change in behavior [53]. One drug thought to reduce microglial activation is minocycline, a broad-spectrum tetracycline antibiotic with broad anti-inflammatory activity [54]. First and second generations of APs regulate the secretory profile of microglia in vitro. They inhibit the release of proinflammatory cytokines from activated microglia and alleviate oxidative stress [55]. However, some recent reports have shown conflicting results on the effect of some APs on the release of pro-inflammatory cytokines [56]. At the same time, not all APs have an anti-inflammatory effect, which may be due to the role of microglia in the development of TRS.

2.2. Sensitization or Kindling in Treatment Resistance Schizophrenia

“Firing”/“sensitization” refers to the process by which the initial immune response to some stimulus (stress or infection) raises or lowers the threshold to respond to the next exposure to the same stimulus. At the same time, a weaker stimulus is required to activate the immune response or release cytokines than with the initial exposure to an unfavorable (damaging) factor. It is believed that the memory function of the acquired immune system is responsible for this process [51]. The action of factors such as systemic inflammation or stress on healthy people leads to the stimulation of the immune response. As a result, cell proliferation is activated, an increase in the production and release of pro-inflammatory cytokines is observed [57]. “Firing up”/“sensitization” supports the hypothesis that neuroinfection in early childhood may lead to increased release of cytokines when the immune system is activated later in life. These processes lead to neurotransmitter disorders. Stress induces a pro-inflammatory immune response in CNS. However, this is usually reduced after a stressful event. Psychopathological symptoms and neuroinflammation are associated with the immune response of CNS cells to stress, and neuroinflammation is involved in stress-related behavioral changes induced by cytokines and mediated by neurotransmitters. Studies have found that after exposure to chronic stress or repeated stressful events, the threshold for physiological responses of the CNS to stress decreases. As a result, less stimulus is enough to activate an immune or neurotransmitter response. In an animal study, it was shown that with age, the brain is in a sensitized state and produces more cytokines for inflammatory stimuli than the brain of young animals [52]. Repeated exposure to pro-inflammatory cytokines leads to increased neurotransmitter responses [58] as, for example, with tumor necrosis factor alpha (TNF-α) [59]. Stress causes activation and proliferation of microglia in the CNS, which may possibly mediate these cytokine effects [52]. Chronic stress is known to affect the glutamatergic system, ionotropic and metabotropic glutamate receptors and excitatory amino acid transporters [60], which may also play a role in the development of TRS, as it is associated with higher levels of glutamate in the anterior cingulate cortex [61].

2.3. Vulnerability-Stress-Inflammation in Treatment Resistance Schizophrenia

The risk of developing TRS increases with stressful life events or psychological stress, especially those that act at key periods in the development of the CNS. The Sch vulnerability-stress model was first proposed by Zubin and Vesna [62], who suggested that stress above the vulnerability threshold in humans contributes to the development of a psychotic episode. It is important to add inflammation to this model, forming the vulnerability-stress-inflammation model, since neuroinflammation plays an important role in the pathogenesis of TRS and can in turn be caused by stress [63]. For example, if an inflammatory response in the CNS is stimulated in a second trimester of pregnancy or offspring while the CNS is still developing, the offspring may be a risk of developing Sch. Animal studies have shown that exposure to stress at an early age leads to an increase in the level of pro-inflammatory cytokines [64], which play an important role in the development of Sch and TRS. Vulnerability-stress-inflammation-induced immune dysregulation is associated with dysregulation of many neurotransmitter systems that APs cannot therapeutically address. Thus, the development of TRS is likely associated with stress-induced inflammation [5].

2.4. Prenatal, Perinatal and Postnatal Infection in Treatment Resistance Schizophrenia

Existing epidemiological studies give us the idea that prenatal exposure to maternal infection is associated with an increased risk of Sch in the offspring [65]. The risk of developing Sch may be related to the direct effects of neuroinfection (e.g., disruption of structure due to cyst formation, exposure to inflammatory factors) as well as neurochemical changes such as increased dopamine levels associated with poor performance. Catechol-O-methyl transferase (COMT) and increased dopamine synthesis caused by Toxoplasma gondii infection [66]. Exposure to viruses and other infectious agents—influenza, herpes simplex virus type 2, Coronavirus Disease 2019 (COVID-19) during pregnancy and at the time of conception—is associated with a greater risk of psychotic disorders [67]. Given the changes in pro-inflammatory cytokine production in pregnant women with COVID-19, schizophrenic and psychotic disorders may potentially be a long-term risk in the offspring of pregnant women who have experienced COVID-19 [68]. Animal studies have also provided evidence for the role of pre- and perinatal infections in the later development of Sch [69]. For example, after prenatal exposure to viruses, offspring show typical symptoms of Sch, such as cognitive impairment or startle reflex abnormalities [70]. Maternal bacterial infection during pregnancy is closely associated with the development of psychosis in the offspring and varied depending on the severity of the infection and the sex of the offspring. At the same time, the effect of a multisystem bacterial infection was almost two times higher than that of a less severe localized bacterial infection [71]. Of interest are studies that have demonstrated the association of Sch development with prenatal or early childhood exposure to various viruses [72], respiratory infections [73] and infections of the genital organs or reproductive tract [74]. Because Sch develops more frequently during adolescence or adulthood, it is important to establish a possible mechanism for the association between early infection and Sch in adults. Studies in animal models show that early infection or immune activation affects several processes of neurogenesis, including dopaminergic and glutamatergic neurotransmission [75]. The study of bacterial [71] and some other infections in humans [76] are examples that highlight this connection. The risk of developing TRS is also indicated by an increased level of C-reactive protein (CRP) or cytokines in childhood [77]. In addition, neuroinfection at a later age has been shown to be associated with an increased risk of developing TRS. A large epidemiological study conducted in Denmark showed that autoimmune disorders, as well as severe infections, increase the risk of developing Sch and Sch spectrum disorders. This is especially true for patients with both risk factors for TRS [78].

2.5. Cytokine Imbalance in Treatment Resistance Schizophrenia

Based on the meta-analyses by Momtazmanesh et al. [79], it is possible to conditionally classify cytokines according to their serum levels in patients with TRS into four groups: group 1—elevated cytokines, including interleukin 6 (IL-6), tumor necrosis factor alpha (TNF-α), interleukin 1 beta (IL-1β), interleukin 12 (IL-12) and transforming growth factor beta (TGF-β); group 2—unchanged cytokines, including interleukin 2 (IL-2), interleukin 4 (IL-4) and interleukin 17 (IL-17); group 3—elevated or unchanged cytokines, including interleukin 8 (IL-8) and interferon gamma (IFN-γ); group 4—interleukin 10 (IL-10) with increased, decreased and unchanged serum levels. However, this grouping is not unambiguous and includes mainly pro-inflammatory cytokines. In addition, the authors did not provide an analysis of the relationship between the levels of pro-inflammatory and anti-inflammatory cytokines in patients with TRS.
Higher serum levels of pro-inflammatory cytokines are characteristic of both patients with the first episode of Sch and patients with relapse and TRS, compared with the control group [80]. IL-1β, IL-6 and TGF-β were elevated at the first psychotic episode, and Sch flare normalized after AP treatment. Conversely, the levels of IL-12, IFN-γ, TNF-α and soluble interleukin 2 receptor (sIL-2R) remained elevated during exacerbations and during AP therapy [81]. A study of interleukins in the cerebrospinal fluid (CSF) showed that the levels of interleukin 6 (IL-6) and IL-8 were increased in Sch, but not significantly increased in affective disorders [82]. A meta-analysis of CSF cytokines showed higher levels of pro-inflammatory cytokines and lower levels of anti-inflammatory cytokines in patients with Sch and TRS [83].
It is known that dopaminergic dysfunction is a significant feature in the pathophysiology of TRS [84]. Interactions between cytokines and neurotransmitters in certain areas of the brain, and also during brain development, are important in the pathophysiology of TRS. Apparently, the pro-inflammatory cytokine IL-1β, which induces the transformation of rat mesencephalic progenitor cells into a dopaminergic phenotype [85], and IL-6, which reduces the survival of serotonergic neurons in the fetal brain, seem to play an important role in influencing neurotransmitter systems in TRS. [86]. Studies have found abnormalities in the cytokine system in patients with TRS [87,88]. There is evidence that the levels of IL-2 and IL-6 were elevated in patients with TRS, which is probably associated with the activation of the inflammatory response system (IRS). Moreover, serum IL-2 or IL-6 and cortisol are positively correlated with Sch, supporting the hypothesis that hypercortisolemia may also be caused by pro-inflammatory cytokines [89,90].
So, a summary of the results of studies on the role of neuroinflammation in the development of TRS is presented in Table 2.

3. Cytokines Alteration in Treatment-Resistant Schizophrenia

Cytokines, which comprise a family of proteins—interleukins (IL), lymphokines, monokines, interferons and chemokines—are important components of the immune system (Table 3).
Cytokines act in concert with specific cytokine inhibitors and soluble cytokine receptors to regulate the human immune response [91]. Their physiologic role in inflammation and pathologic role in systemic inflammatory states are now well recognized. An imbalance in cytokine production or cytokine receptor expression and/or dysregulation of a cytokine process contributes to various pathological disorders, including Sch [92]. Cytokines are classified as pro-inflammatory and anti-inflammatory. The time-dependent pro- and anti-inflammatory imbalance determines the outcome of an inflammatory response in development of TRS [93]. It should be clarified that the division of cytokines into pro- and anti-inflammatory is very conditional, because depending on the conditions, the cytokine can behave as a pro- or anti-inflammatory cytokine (for example, IL-6) [94]. Indeed, the number of cytokines, the nature of the activating signal, the nature of the target cell, the nature of the cytokines produced, the timing, the sequence of action of cytokines and even the experimental model are parameters that strongly affect the properties of cytokines [95].

3.1. Pro-Inflammatory Cytokines

Pro-inflammatory cytokines play a central role in neuroinflammatory disorders of infectious or noninfectious origin. Pro-inflammatory cytokines are produced predominantly by activated macrophages and are involved in the upregulation of inflammatory reactions [96]. These cytokines serve to contain and resolve the inflammatory foci through activation of local and systemic inflammatory responses. Pro-inflammatory cytokines may directly modulate neuronal activity in various classes of neurons in CNS, including dopaminergic neurons [97]. The major pro-inflammatory cytokines that are responsible for early responses are interleukin 1 alpha (IL1-α), interleukin 1 β (IL1-β), interleukin 6 (IL-6) and tumor necrosis factor alpha (TNF-α). Other pro-inflammatory mediators include members of the interleukin 20 (IL-20) family, interleukin 33 (IL-33), leukemia inhibitory factor (LIF), interferon gamma (IFN-γ), oncostatin M (OSM), ciliary neurotrophic factor (CNTF), transforming growth factor beta (TGF-β), granulocytic-macrophage colony-stimulating factor (GM-CSF), interleukin 11 (IL-11), interleukin 12 (IL-12), interleukin 17 (IL-17), interleukin 18 (IL-18), interleukin 18 (IL-8) and a variety of other chemokines that chemoattract inflammatory cells. These cytokines either act as endogenous pyrogens (IL-1, IL-6, TNF-α), upregulate the synthesis of secondary mediators and pro-inflammatory cytokines by both macrophages and mesenchymal cells, stimulate the production of acute phase proteins or attract inflammatory cells [98]. IL-1β, TNFα, IFN-γ, IL-12 and interleukin 11 (IL-18) are well characterized as pro-inflammatory cytokines.

3.1.1. Interleukin 1 β

IL-1β is produced by myeloid blood cells, pathogenic lymphocytes, resident microglia and CNS astrocytes in autoimmune diseases, neurodegeneration and metabolic diseases. It is a key pro-inflammatory cytokine that is involved in the regulation of the innate immune response [99]. IL-1β is a pleiotropic cytokine that can activate microglia and astrocytes and lead to subsequent synthesis of other pro-inflammatory cytokines and chemotactic mediators in the CNS [100]. IL-1β leads to aberrant release and accumulation of glutamate, which subsequently leads to neuronal death in most neurodegenerative diseases [101]. In a cross-sectional study by Enache et al. [102], investigating the association of plasma cytokine levels with TRS, no association of IL-1β with TRS was found. However, other studies have conflicting results [103].

3.1.2. Tumor Necrosis Factor Alpha

TNF-α regulates several processes, including sleep, learning and memory, synaptic plasticity and astrocytic-induced synaptic reinforcement in the healthy CNS [103]. The biological functions of TNF-α are mediated through its two main receptors: tumor necrosis factor receptor 1 (TNFR1 or p55) and tumor necrosis factor receptor 2 (TNFR2 or p75). TNFR1 activation initiates inflammatory, apoptotic and degenerative cascades, while TNF-α signaling through TNFR2 is anti-inflammatory and cytoprotective, resulting in induction of proliferation, differentiation, angiogenesis and tissue repair [104]. TNF-α is also an important pro-inflammatory cytokine produced by both neurons and glial cells. Genetic association studies have provided evidence of Sch-associated gene variations in the innate and adaptive immune systems [105]. In a recent genetic study, which examined the relationship between TNF-α polymorphism–238 G/A and response to APs treatment, it was shown that while TNF-α polymorphisms–238 G/A and -308 G /A were not associated with Sch, TNF-α–238 G/A polymorphism may be associated with treatment resistance and suicide attempts in patients with Sch in the Turkish population [106]. Another study on the prognosis of TRS using immune-inflammatory biomarkers reported that TRS is associated with a specific cytokine-chemokine profile, i.e., elevated levels of C-C motif chemokine ligand 11 (CCL11), macrophage inflammatory protein-1 alpha (MIP-1α), soluble tumor necrosis factor receptors 1 (sTNF-R1) and soluble tumor necrosis factor receptors 2 (sTNF-R2), as well as decreased levels of interferon gamma induced protein 10 (IP-10), TNF-α, IL-2 and IL-4 [107]. Data from a 2021 crossover study showed, however, that both TRS and ultra-treatment-resistant Sch (UTRS) patients tended to increase TNFα expression, which, however, did not reach statistical significance [108].

3.1.3. Interferon Gamma

IFN-γ is a soluble cytokine that is predominantly released from T helper type 1 (Th1), cytotoxic T lymphocytes and natural killer cells. IFN-γ serves to prime microglia, which is associated with various cellular adaptations, including changes in morphology, upregulation of receptors and increased levels of pro-inflammatory cytokines [38]. Data regarding the level of IFN-γ and TRS remain contradictory. For example, in a study by Upthegrove et al. [109], evidence has been obtained that elevated IFN-γ levels are associated with TRS. However, another study reported that IFN-γ was not associated with response to APs therapy [110].

3.1.4. Interleukin 12

IL-12 secreted mainly by macrophages and dendritic cells in response to components of the bacterial cell wall. IL-12 stimulates proliferation, and also activates and increases the cytotoxicity of natural killer cells (NK cells) and T cells, promoting the differentiation of the latter into Th1. It is also known to induce the secretion of IFN-γ and TNF-α and has a synergistic effect with Interleukin 18 (IL-18) [92]. When examining the plasma level of IL-12, it was found to be elevated in TRS and UTRS [109].

3.1.5. Interleukin 18

Research on the presence of IL-18 in the CNS began shortly after its discovery as a stimulator of inf-γ production in the immune system. IL-18 has been investigated for its similarity to IL-1β as a possible mediator of disease behavior and local inflammatory responses associated with neuronal injury. IL-18 promotes loss of appetite, sleep, and inhibition of long-term potentiation (LTP), and is also produced by and active in microglial cells and possibly contributes to neurodegeneration. IL-18 represents a link between the immune and nervous systems, since IL-18 and its receptors in the CNS mediate neuroinflammation by modulating homeostasis and behavior [111]. There is some evidence that IL-18 levels are elevated in patients with Sch but do not appear to be the cause of the disorder itself [112], although it is likely that elevated serum IL-18 levels may be a biomarker for TRS and UTRS.

3.1.6. Interleukin 8

IL-8 is secreted predominantly in response to an antigen by macrophages, T-lymphocytes, neutrophils, and other cells; IL-8 is also the most potent human chemokine [113]. IL-8, being a pro-inflammatory cytokine, enhances the migration of neutrophils, T-lymphocytes and monocytes, whose enzymes produce free oxygen radicals and, thus, increase oxidative stress, which can lead to neuronal death [114]. Studies have shown that IL-8 significantly predicted non-response to APs therapy and positively correlated with negative Sch symptoms [102] and can be considered as a potential TRS biomarker.

3.1.7. Interleukin 17

IL-17 is secreted by helper lymphocytes 17 (Th17) and stimulates macrophages and microglia to secrete pro-inflammatory cytokines [115]. According to some data, no effect of APs on peripheral levels of IL-17 has been demonstrated [116]. However, it has been reported that activation of the IL-17 pathways may be present from the onset of Sch and appears to increase with disease progression up to the development of TRS and UTRS. The IL-23/IL-17 pathway is being considered as a therapeutic target for patients with TRS, especially since many anti-inflammatory drugs have been proposed as adjuncts to treat Sch symptoms, such as N-acetylcysteine, which appears to reduce they produce IL-17 [38].
Summary, the role of pro-inflammatory cytokines in TRS is presented in the Table 4.

3.2. Anti-Inflammatory Cytokines

The anti-inflammatory (immunosuppressive) cytokines are a series of immunoregulatory molecules that control the pro-inflammatory cytokine response. The anti-inflammatory cytokines act in concert with specific pro-inflammatory cytokine inhibitors and soluble cytokine receptors to regulate the human immune response. Major anti-inflammatory cytokines include interleukin 1 receptor antagonist (IL-1Ra), IL-4, IL-6, IL-10, IL-11, interleukin 13 (IL-13) and TGF-β. Specific cytokine receptors for IL-1, TNFα and IL-18 also function as pro-inflammatory cytokine inhibitors: IL-1Ra as an interleukin 1 alpha (IL-1α) and IL-1β antagonist; Interleukin-18-binding protein (IL-18BP) as an IL-18 antagonist [109]. Several newly found cytokines, such as IL-33, interleukin 35 (IL-35) and interleukin 37 (IL-37), also participate in regulating the function of neurons and neuroglia. Anti-inflammatory cytokines, in particular IL-10, inhibit pro-inflammatory cytokine synthesis and adhesion molecule expression, while increasing the levels of specific cytokine inhibitors [117]. IL-1Ra, IL-4, IL-6 and interleukin 10 (IL-10) are well characterized as anti-inflammatory cytokines.

3.2.1. Interleukin 4

IL-4 is produced by activated Th lymphocytes, mainly Th2 helper lymphocytes, natural killer T cells (NK cells), mast cells and basophils. Its role is to promote the differentiation of Th into Th2 lymphocytes, as well as to increase their cytotoxicity. IL-4 affects macrophages and microglial cells and may be neuroprotective by reducing their ability to induce oxidative stress. In addition, IL-4 also plays a role in cognitive processes [118]. A study of serum IL-4 levels in patients with TRS did not reveal a significant difference in IL-4 levels between the three groups—patients with TRS, patients without TRS and a healthy control group [119]. In another study, Şükrü et al. [120] also found no significant differences between patients with TRS and the control group in terms of serum IL-4 levels.

3.2.2. Interleukin 6

IL-6 is a multifunctional pro-inflammatory cytokine that is secreted primarily through monocytes and macrophages. They play a key role in processes related to immunity and neuroinflammation. IL-6 regulates the transmission of neuronal excitability, metabolism and sensitivity of CNS neurotransmitters to them. High concentrations of IL-6 at baseline have been associated with the development of TRS and prolonged hospital stay [121]. Mongan et al. [122] showed that serum IL-6 was significantly higher in TRS patients than in normal volunteers, while Sch patients without TRS showed intermediate values. In addition, elevated levels of IL-6 were found in not only patients with TRS, but also in patients with UTRS, confirming the significant predictive role of IL-6 in TRS. IL-6 is one of the main effector cytokines of Th17 cells. However, it is also possible that the high levels of IL-6 observed in the UTRS subgroup may indicate a bias towards the Th2 pathway associated with the chronicity of the schizophrenic process [123]. TRS is also accompanied by signs of IRS and compensatory immunoregulatory system (CIRS), including activation of M1 cells (especially IL-6 and TNFα trans-signaling) [96]. Clear immune abnormalities are seen in TRS patients, and IL-6 may be an important marker of TRS [124].

3.2.3. Interleukin 10

IL-10 and receptors to IL-10 (IL-10R) are synthesized in the CNS, including by microglia and astrocytes; they can be considered an important modulator of neuroinflammation [125]. After IL-10 binds to its receptor, this cytokine initiates its cellular effects through canonical Janus kinase (JAK)/signal transducer and transcriptional activator (STAT), which includes JAK1 and STAT3, which subsequently induces the expression of genes associated with immunosuppression [126]. Taken together, the evidence suggests that IL-10 plays a critical role in limiting inflammation in the CNS, similar to that seen in peripheral sites, by altering the ability of resident glia and infiltrating leukocytes to respond to activating stimuli and decreasing the production of inflammatory mediators by these cells [127]. Patients with TRS show high levels of IL-10. The upregulation of this potent anti-inflammatory cytokines may reflect the induction of contractive homeostatic processes [38]. A meta-analysis by Marcinowicz et al. [116] demonstrated a decrease in serum IL-10 levels in patients with a first psychotic episode after APs.
A summary of the role of anti-inflammatory cytokines in TRS is presented in the Table 5.

4. Correction of Cytokine Status Imbalance as a Promising Therapeutic Strategy for Treatment-Resistant Schizophrenia

Current treatment of Sch considers the use of APs as the first line of therapy [128,129]. This is often followed by the use of non-steroidal anti-inflammatory drugs (NSAIDs) in addition to nutrients (vitamins, minerals, plant and animal products) that affect inflammation and the immune system [130]. The effect of APs and NSAIDs on the level of cytokines has also been shown with a decrease in the expression levels of pro-inflammatory cytokines, such as IL-18, IL-1β, IL-6 and IL-8 [131,132].
The heterogeneity of the phenotypes underlying Sch and the high likelihood of adverse drug reactions (ADRs) with APs support the need to explore new treatment strategies for TRS, most of which are under study. Therapy for TRS with monoclonal antibodies, intravenous immunoglobulins (IVIG), NSAIDs, corticosteroids, tetracycline antibiotics, antioxidants and statins have been described as possible strategies for disease-modifying therapy [130]. Prospects also have a correction of the cytokine imbalance in patients with TRS. Thus, this issue remains open and is actively studied in connection with the relevance of the task (Table 6).

5. Discussion

The delicate balance between pro-inflammatory and anti-inflammatory cytokines determines the net effect of a neuroinflammatory response in patients with TRS. Perturbations in this equilibrium can drive the patient defense immune response towards chronic neuroinflammation (pro-inflammatory) or towards healing (anti-inflammatory). Thus, a cytokine imbalance may be beneficial to the patient with Sch by initiating the neuroinflammatory response. However, overproduction or underproduction of pro-inflammatory or anti-inflammatory endogenous mediators (cytokines) may actually be deleterious to the patient with “non-dopamine’’ Sch (Figure 4).
In addition, chronic neuroinflammation, supported by an imbalance between pro-inflammatory and anti-inflammatory cytokines, and persistent dopaminergic neurotransmission disorder can be considered as an overlap syndrome in patients with “dopamine” Sch.
A genetic predisposition that determines the balance of pro-inflammatory and anti-inflammatory cytokines and, hence, susceptibility to TRS is very important in the patients with Sch. Various single nucleotide variants (SNVs) have been identified within pro-inflammatory and anti-inflammatory cytokine and cytokine receptor genes that alter their expression. These SNVs of cytokine and cytokine receptor genes may determine the imbalance of pro-inflammatory and anti-inflammatory cytokines in the neuroinflammatory response in patients with “non dopamine” and “dopamine” Sch.
To date, therapeutic strategies targeting pro-inflammatory cytokines may be effective in treating TRS. Pro-inflammatory cytokines are known to be crucial for initiating a neuroinflammatory response. However, their level in the CNS may have reached its absolute or relative peak before the clinical signs of TRS became apparent.
In addition, therapy that blocks pro-inflammatory cytokines, paradoxically, may lead to increased inflammation [151]. Various inflammation paradoxes have been reported, including new inflammations ocurring when: (1) particular cytokine and inflammatory regulators encoding genes are mutated [152]; (2) patients experience somatic mutations [153] and inflammation [154]; (3) pro-inflammatory cytokines are weakened due to SNVs of the genes encoding them [155]; (4) pro-inflammatory cytokine blockage therapies are used; etc. [144]. Pro-inflammatory cytokines and regulators are interconnected through evolution; single cytokine blockade therapies may result in significant upregulation of a long list of genes and signaling pathways, presumably the “second wave of inflammation” [156]. For example, the second wave of inflammation may be the main mechanism of ADRs observed in patients receiving Mab therapy that blocks pro-inflammatory cytokines [151].
Nevertheless, our narrative review provides a new insight into the role of imbalance between pro-inflammatory and anti-inflammatory cytokines in the pathogenesis of TRS and in new approaches to predicting and early diagnosis of the development of TR to APs, as well as new targets for future therapeutic interventions in “non-dopamine” Sch.

6. Conclusions

Our narrative review demonstrates that the problem of evaluating the contribution of pro-inflammatory and anti-inflammatory cytokines to maintaining or changing the cytokine balance can become a new key in unlocking the mystery of “non-dopamine” Sch and developing new therapeutic strategies for the treatment of TRS and psychosis in acute and chronic neuroinflammation. In addition, the inconsistency of the results of previous studies on the role of pro-inflammatory and anti-inflammatory cytokines indicates that the TRS biomarker, most likely, is not the serum level of one or several cytokines, but the cytokine balance. We have demonstrated a hypothesis that the cytokine imbalance is one of the most important TRS biomarkers. Partially, this hypothesis is supported by the variable response to immunomodulators in patients with TRS, which were prescribed without taking into account the cytokine balance of the relation between serum levels of the most important pro-inflammatory and anti-inflammatory cytokines for TRS.

Author Contributions

Conceptualization, R.F.N., N.A.S. and M.M.P.; methodology, N.A.S.; software, A.K.K.; validation, A.K.K., A.I.S. and N.P.G.; formal analysis, A.K.K. and A.I.S.; investigation, A.K.K. and A.I.S.; resources, N.A.S. and R.F.N.; data curation, N.A.S. and G.A.S.; writing—original draft preparation, A.K.K., A.I.S. and A.P.O.; writing—review and editing, N.A.S., G.A.S. and R.F.N.; visualization, A.K.K., N.A.S. and M.A.-Z.; supervision, N.A.S. and R.F.N.; project administration, N.G.N. and R.F.N. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors are grateful to Ruslan S. Khasanov for his help in the design of all figures.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Saha, S.; Chant, D.; McGrath, J. A systematic review of mortality in schizophrenia: Is the differential mortality gap worsening over time? Arch. Gen. Psychiatry 2007, 64, 1123–1131. [Google Scholar] [CrossRef] [PubMed]
  2. Laursen, T.M. Life expectancy among persons with schizophrenia or bipolar affective disorder. Schizophr. Res. 2011, 131, 101–104. [Google Scholar] [CrossRef] [PubMed]
  3. Chesney, E.; Goodwin, G.M.; Fazel, S. Risks of all-cause and suicide mortality in mental disorders: A meta-review. World Psychiatry 2014, 13, 153–160. [Google Scholar] [CrossRef] [PubMed]
  4. Khasanova, A.K.; Dobrodeeva, V.S.; Shnayder, N.A.; Petrova, M.M.; Pronina, E.A.; Bochanova, E.N.; Lareva, N.V.; Garganeeva, N.P.; Smirnova, D.A.; Nasyrova, R.F. Blood and urinary biomarkers of antipsychotic-induced metabolic syndrome. Metabolites 2022, 12, 726. [Google Scholar] [CrossRef] [PubMed]
  5. Howes, O.D.; Thase, M.E.; Pillinger, T. Treatment resistance in psychiatry: State of the art and new directions. Mol. Psychiatry 2022, 27, 58–72. [Google Scholar] [CrossRef] [PubMed]
  6. Vaiman, E.E.; Shnayder, N.A.; Khasanova, A.K.; Strelnik, A.I.; Gayduk, A.J.; Al-Zamil, M.; Sapronova, M.R.; Zhukova, N.G.; Smirnova, D.A.; Nasyrova, R.F. Pathophysiological mechanisms of antipsychotic-induced parkinsonism. Biomedicines 2022, 10, 2010. [Google Scholar] [CrossRef] [PubMed]
  7. Nyberg, S.; Dencker, S.J.; Malm, U.; Dahl, M.-L.; Svetnson, J.-O.; Halldin, C.; Naskashima, Y.; Farde, L. D(2)- and 5-Ht(2) receptor occupancy in high-dose neuroleptictreated patients. Int. J. Neuropsychopharmacol. 1998, 1, 95–101. [Google Scholar] [CrossRef]
  8. Grunder, G.; Carlsson, A.; Wong, D.F. Mechanism of new antipsychotic medications: Occupancy is not just antagonism. Arch. Gen. Psychiatry 2003, 60, 974–977. [Google Scholar] [CrossRef]
  9. Kane, J.M.; Agid, O.; Baldwin, M.L.; Howes, O.; Lindenmayer, J.P.; Marder, S.; Olfson, M.; Potkin, S.G.; Correll, C.U. Clinical Guidance on the Identification and Management of Treatment-Resistant Schizophrenia. J. Clin. Psychiatry 2019, 80, 2783. [Google Scholar] [CrossRef]
  10. Suzuki, T.; Remington, G.; Mulsant, B.H.; Rajji, T.K.; Uchida, H.; Graff-Guerrero, A.; Mamo, D.C. Treatment resistant schizophrenia and response to antipsychotics: A review. Schizophr. Res. 2011, 133, 54–62. [Google Scholar] [CrossRef]
  11. Work Group on Schizophrenia. Practice Guideline for the Treatment of Patients with Schizophrenia, 2nd ed.; American Psychiatric Press: Arlington, VA, USA, 2004. [Google Scholar]
  12. Hasan, A.; Falkai, P.; Wobrock, T.; Lieberman, J.; Glenthoj, B.; Gattaz, W.F.; Thibaut, F.; Möller, H.J.; World Federation of Societies of Biological Psychiatry (WFSBP) Task Force on Treatment Guidelines for Schizophrenia. World Federation of Societies of Biological Psychiatry (WFSBP) Guidelines for Biological Treatment of Schizophrenia, part 1: Update 2012 on the acute treatment of schizophrenia and the management of treatment resistance. World J. Biol. Psychiatry Off. J. World Fed. Soc. Biol. Psychiatry 2012, 13, 318–378. [Google Scholar] [CrossRef] [PubMed]
  13. National Collaborating Centre for Mental Health (UK). Psychosis and Schizophrenia in Adults: Treatment and Management; National Institute for Health and Care Excellence (UK): London, UK, 2014. [Google Scholar]
  14. Howes, O.D.; McCutcheon, R.; Agid, O.; de Bartolomeis, A.; van Beveren, N.J.; Birnbaum, M.L.; Bloomfield, M.A.; Bressan, R.A.; Buchanan, R.W.; Carpenter, W.T.; et al. Treatment-Resistant Schizophrenia: Treatment Response and Resistance in Psychosis (TRRIP) Working Group Consensus Guidelines on Diagnosis and Terminology. Am. J. Psychiatry 2017, 174, 216–229. [Google Scholar] [CrossRef] [PubMed]
  15. Moore, T.A.; Buchanan, R.W.; Buckley, P.F.; Chiles, J.A.; Conley, R.R.; Crismon, M.L.; Essock, S.M.; Finnerty, M.; Marder, S.R.; Miller, D.D.; et al. The Texas Medication Algorithm Project antipsychotic algorithm for schizophrenia: 2006 update. J. Clin. Psychiatry 2007, 68, 1751–1762. [Google Scholar] [CrossRef] [PubMed]
  16. Buchanan, R.W.; Kreyenbuhl, J.; Kelly, D.L.; Noel, J.M.; Boggs, D.L.; Fischer, B.A.; Himelhoch, S.; Fang, B.; Peterson, E.; Aquino, P.R.; et al. The 2009 schizophrenia PORT psychopharmacological treatment recommendations and summary statements. Schizophr. Bull. 2010, 36, 71–93. [Google Scholar] [CrossRef]
  17. The International Psychopharmacology Algorithm Project. Available online: http://www.ipap.org (accessed on 15 August 2022).
  18. Mørup, M.F.; Kymes, S.M.; Oudin Åström, D. A modelling approach to estimate the prevalence of treatment-resistant schizophrenia in the United States. PLoS ONE 2020, 15, e0234121. [Google Scholar] [CrossRef]
  19. Drugs@FDA: FDA-Approved Drugs. Available online: https://www.accessdata.fda.gov/scripts/cder/daf/index.cfm (accessed on 15 August 2022).
  20. Leung, C.C.; Gadelrab, R.; Ntephe, C.U.; McGuire, P.K.; Demjaha, A. Clinical Course, Neurobiology and Therapeutic Approaches to Treatment Resistant Schizophrenia. Toward an Integrated View. Front. Psychiatry 2019, 10, 601. [Google Scholar] [CrossRef]
  21. Kinon, B.J. The Group of Treatment Resistant Schizophrenias. Heterogeneity in Treatment Resistant Schizophrenia (TRS). Front. Psychiatry 2019, 9, 757. [Google Scholar] [CrossRef]
  22. Pisanu, C.; Squassina, A. Treatment-Resistant Schizophrenia: Insights From Genetic Studies and Machine Learning Approaches. Front. Pharmacol. 2019, 10, 617. [Google Scholar] [CrossRef]
  23. Vita, A.; Minelli, A.; Barlati, S.; Deste, G.; Giacopuzzi, E.; Valsecchi, P.; Turrina, C.; Gennarelli, M. Treatment-Resistant Schizophrenia: Genetic and Neuroimaging Correlates. Front. Pharmacol. 2019, 10, 402. [Google Scholar] [CrossRef]
  24. Fatemi, S.H.; Folsom, T.D. The neurodevelopmental hypothesis of schizophrenia, revisited. Schizophr. Bull. 2009, 35, 528–548. [Google Scholar] [CrossRef]
  25. Potkin, S.G.; Kane, J.M.; Correll, C.U.; Lindenmayer, J.P.; Agid, O.; Marder, S.R.; Olfson, M.; Howes, O.D. The neurobiology of treatment-resistant schizophrenia: Paths to antipsychotic resistance and a roadmap for future research. NPJ Schizophr. 2020, 6, 1. [Google Scholar] [CrossRef] [PubMed]
  26. Takao, N.; Murai, T.; Fujiwara, H. Treatment-resistant schizophrenia characterised by dopamine supersensitivity psychosis and efficacy of asenapine. BMJ Case Rep. 2021, 14, e242495. [Google Scholar] [CrossRef] [PubMed]
  27. Lowe, P.; Krivoy, A.; Porffy, L.; Henriksdottir, E.; Eromona, W.; Shergill, S.S. When the drugs don’t work: Treatment-resistant schizophrenia, serotonin and serendipity. Ther. Adv. Psychopharmacol. 2018, 8, 63–70. [Google Scholar] [CrossRef] [PubMed]
  28. Huang, L.C.; Lin, S.H.; Tseng, H.H.; Chen, K.C.; Yang, Y.K. The integrated model of glutamate and dopamine hypothesis for schizophrenia: Prediction and personalized medicine for prevent potential treatment-resistant patients. Med. Hypotheses 2020, 143, 110159. [Google Scholar] [CrossRef] [PubMed]
  29. Shnayder, N.A.; Abdyrakhmanova, A.K.; Nasyrova, R.F. Oxidation of antipsychotics. Encyclopedia 2022, 2, 974–989. [Google Scholar] [CrossRef]
  30. Abdyrakhmanova, A.K.; Nasyrova, R.F. Pharmacogenetic testing of cytochrome P450 metabolizing enzymes in 28-year-old man with treatment-resistant schizophrenia. Pers. Psychiatry Neurol. 2022, 2, 81–88. [Google Scholar] [CrossRef]
  31. Linova, L.P.; Torgovtsev, A.A.; Limankin, O.V.; Nasyrova, R.F. Clinical case of a 36-year-old man with treatment-resistant paranoid shizophrenia: Personalized therapy selection. Pers. Psychiatry Neurol. 2022, 2, 89–97. [Google Scholar]
  32. Moons, T.; de Roo, M.; Claes, S.; Dom, G. Relationship between P-glycoprotein and second-generation antipsychotics. Pharmacogenomics 2011, 12, 1193–1211. [Google Scholar] [CrossRef]
  33. Osipova, S.M.; Shnayder, N.A. Pharmacogenetic testing of antipsychotic transporter proteins: A case report in a 32-year-old woman with treatment-resistant schizophrenia. Pers. Psychiatry Neurol. 2022, 2, 98–106. [Google Scholar] [CrossRef]
  34. Hoosain, F.G.; Choonara, Y.E.; Tomar, L.K.; Kumar, P.; Tyagi, C.; du Toit, L.C.; Pillay, V. Bypassing P-Glycoprotein drug efflux mechanisms: Possible applications in pharmacoresistant schizophrenia therapy. BioMed Res. Int. 2015, 2015, 84963. [Google Scholar] [CrossRef]
  35. Bošković, M.; Vovk, T.; Kores Plesničar, B.; Grabnar, I. Oxidative stress in schizophrenia. Curr. Neuropharmacol. 2011, 9, 301–312. [Google Scholar] [PubMed] [Green Version]
  36. Buosi, P.; Borghi, F.A.; Lopes, A.M.; Facincani, I.; Fernandes-Ferreira, R.; Oliveira-Brancati, C.; do Carmo, T.S.; Souza, D.; da Silva, D.; de Almeida, E.A.; et al. Oxidative stress biomarkers in treatment-responsive and treatment-resistant schizophrenia patients. Trends Psychiatry Psychother. 2021, 43, 278–285. [Google Scholar] [CrossRef] [PubMed]
  37. Ermakov, E.A.; Dmitrieva, E.M.; Parshukova, D.A.; Kazantseva, D.V.; Vasilieva, A.R.; Smirnova, L.P. Oxidative stress-related mechanisms in schizophrenia pathogenesis and new treatment perspectives. Oxid. Med. Cell. Longev. 2021, 2021, 8881770. [Google Scholar] [CrossRef] [PubMed]
  38. Leboyer, M.; Godin, O.; Terro, E.; Boukouaci, W.; Lu, C.L.; Andre, M.; Aouizerate, B.; Berna, F.; Barau, C.; Capdevielle, D.; et al. Immune signatures of treatment-resistant schizophrenia: A FondaMental Academic Centers of Expertise for Schizophrenia (FACE-SZ) Study. Schizophr. Bull. Open 2021, 2, sgab012. [Google Scholar] [CrossRef]
  39. Miller, B.J.; Goldsmith, D.R. Evaluating the hypothesis that schizophrenia is an inflammatory disorder. Focus (Am. Psychiatr. Publ.) 2020, 18, 391–401. [Google Scholar] [CrossRef]
  40. Labonté, C.; Zhand, N.; Park, A.; Harvey, P.D. Complete blood count inflammatory markers in treatment-resistant schizophrenia: Evidence of association between treatment responsiveness and levels of inflammation. Psychiatry Res. 2022, 308, 114382. [Google Scholar] [CrossRef]
  41. Manchia, M.; Fontana, A.; Panebianco, C.; Paribello, P.; Arzedi, C.; Cossu, E.; Garzilli, M.; Montis, M.A.; Mura, A.; Pisanu, C.; et al. Involvement of Gut Microbiota in Schizophrenia and Treatment Resistance to Antipsychotics. Biomedicines 2021, 9, 875. [Google Scholar] [CrossRef]
  42. Seeman, M.V. The Gut Microbiome and Treatment-Resistance in Schizophrenia. Psychiatr. Q. 2020, 91, 127–136. [Google Scholar] [CrossRef]
  43. Manchia, M.; Squassina, A.; Tozzi, F.; Antoniades, A.; Carpiniello, B. Gut microbiota and treatment-resistant schizophrenia: Many questions, fewer answers. Pharmacogenomics 2022, 23, 277–280. [Google Scholar] [CrossRef]
  44. Teasdale, S.; Mörkl, S.; Müller-Stierlin, A.S. Nutritional Psychiatry in the treatment of psychotic disorders: Current hypotheses and research challenges. Brain Behav. Immun. Health 2020, 5, 100070. [Google Scholar] [CrossRef]
  45. Marx, W.; Moseley, G.; Berk, M.; Jacka, F. Nutritional psychiatry: The present state of the evidence. Proc. Nutr. Soc. 2017, 76, 427–436. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Onaolapo, O.J.; Onaolapo, A.Y. Nutrition, nutritional deficiencies, and schizophrenia: An association worthy of constant reassessment. World J. Clin. Cases 2021, 9, 8295–8311. [Google Scholar] [CrossRef] [PubMed]
  47. Borst, K.; Dumas, A.A.; Prinz, M. Microglia: Immune and non-immune functions. Immunity 2021, 54, 2194–2208. [Google Scholar] [CrossRef] [PubMed]
  48. Lasselin, J. Back to the future of psychoneuroimmunology: Studying inflammation-induced sickness behavior. Brain Behav. Immun. Health 2021, 18, 100379. [Google Scholar] [CrossRef] [PubMed]
  49. Martínez-Cué, C.; Rueda, N. Cellular Senescence in Neurodegenerative Diseases. Front. Cell. Neurosci. 2020, 14, 16. [Google Scholar] [CrossRef] [PubMed]
  50. Lecours, C.; Bordeleau, M.; Cantin, L.; Parent, M.; Paolo, T.D.; Tremblay, M.È. Microglial implication in Parkinson’s disease: Loss of beneficial physiological roles or gain of inflammatory functions? Front. Cell. Neurosci. 2018, 12, 282. [Google Scholar] [CrossRef]
  51. Fonken, L.K.; Frank, M.G.; Gaudet, A.D.; Maier, S.F. Stress and aging act through common mechanisms to elicit neuroinflammatory priming. Brain Behav. Immun. 2018, 73, 133–148. [Google Scholar] [CrossRef]
  52. Woodburn, S.C.; Bollinger, J.L.; Wohleb, E.S. The semantics of microglia activation: Neuroinflammation, homeostasis, and stress. J. Neuroinflamm. 2021, 18, 258. [Google Scholar] [CrossRef]
  53. Dantzer, R. Neuroimmune interactions: From the brain to the immune system and vice versa. Physiol. Rev. 2018, 98, 477–504. [Google Scholar] [CrossRef]
  54. Mondelli, V.; Vernon, A.C.; Turkheimer, F.; Dazzan, P.; Pariante, C.M. Brain microglia in psychiatric disorders. Lancet Psychiatry 2017, 4, 563–572. [Google Scholar] [CrossRef]
  55. Amato, D.; Beasley, C.L.; Hahn, M.K.; Vernon, A.C. Neuroadaptations to antipsychotic drugs: Insights from pre-clinical and human post-mortem studies. Neurosci. Biobehav. Rev. 2017, 76, 317–335. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Stapel, B.; Sieve, I.; Falk, C.S.; Bleich, S.; Hilfiker-Kleiner, D.; Kahl, K.G. Second generation atypical antipsychotics olanzapine and aripiprazole reduce expression and secretion of inflammatory cytokines in human immune cells. J. Psychiatr. Res. 2018, 105, 95–102. [Google Scholar] [CrossRef] [PubMed]
  57. Frank, M.G.; Fonken, L.K.; Annis, J.L.; Watkins, L.R.; Maier, S.F. Stress disinhibits microglia via down-regulation of CD200R: A mechanism of neuroinflammatory priming. Brain Behav. Immun. 2018, 69, 62–73. [Google Scholar] [CrossRef] [PubMed]
  58. Jeon, S.W.; Kim, Y.K. The role of neuroinflammation and neurovascular dysfunction in major depressive disorder. J. Inflamm. Res. 2018, 11, 179. [Google Scholar] [CrossRef] [PubMed]
  59. Schwarz, M.J. Cytokines, neurophysiology, neuropsychology, and psychiatric symptoms. Dialogues Clin. Neurosci. 2022, 5, 139–153. [Google Scholar] [CrossRef]
  60. Réus, G.Z.; Abelaira, H.M.; Coutellier, L.D.; Manosso, L.M.; Pavlovic, Z.M. Role of Glutamatergic Neurotransmission in the Pathophysiology of Stress-Related Disorders and Chronic Stress Response. In Glutamate and Neuropsychiatric Disorders; Springer International Publishing: Berlin/Heidelberg, Germany, 2022; pp. 65–112. [Google Scholar]
  61. Kumar, V.; Manchegowda, S.; Jacob, A.; Rao, N.P. Glutamate metabolites in treatment resistant schizophrenia: A meta-analysis and systematic review of 1H-MRS studies. Psychiatry Res. Neuroimaging 2020, 300, 111080. [Google Scholar] [CrossRef]
  62. Zubin, J.; Spring, B. Vulnerability-a new view of schizophrenia. J. Abnorm. Psychol. 1977, 86, 103–126. [Google Scholar] [CrossRef]
  63. Müller, N. Inflammation in Schizophrenia: Pathogenetic Aspects and Therapeutic Considerations. Schizophr. Bull. 2018, 44, 973–982. [Google Scholar] [CrossRef]
  64. Lumertz, F.S.; Kestering-Ferreira, E.; Orso, R.; Creutzberg, K.C.; Tractenberg, S.G.; Stocchero, B.A.; Viola, T.W.; Grassi-Oliveira, R. Effects of early life stress on brain cytokines: A systematic review and meta-analysis of rodent studies. Neurosci. Biobehav. Rev. 2022, 139, 104746. [Google Scholar] [CrossRef]
  65. Cheslack-Postava, K.; Brown, A.S. Prenatal infection and schizophrenia: A decade of further progress. Schizophr. Res. 2022, 247, 7–15. [Google Scholar] [CrossRef]
  66. Rovira, P.; Gutiérrez, B.; Sorlózano-Puerto, A.; Gutiérrez-Fernández, J.; Molina, E.; Rivera, M.; Martínez-Leal, R.; Ibanez-Casas, I.; Martín-Laguna, M.V.; Rosa, A.; et al. Toxoplasma gondii Seropositivity Interacts with Catechol-O-methyltransferase Val105/158Met Variation Increasing the Risk of Schizophrenia. Genes 2022, 13, 1088. [Google Scholar] [CrossRef] [PubMed]
  67. Stilo, S.A.; Murray, R.M. Non-genetic factors in schizophrenia. Curr. Psychiatry Rep. 2019, 21, 100. [Google Scholar] [CrossRef] [PubMed]
  68. Cavalcante, M.B.; Cavalcante, C.T.D.M.B.; Sarno, M.; Barini, R.; Kwak-Kim, J. Maternal immune responses and obstetrical outcomes of pregnant women with COVID-19 and possible health risks of offspring. J. Reprod. Immunol. 2021, 143, 103250. [Google Scholar] [CrossRef] [PubMed]
  69. Minakova, E.; Warner, B.B. Maternal immune activation, central nervous system development and behavioral phenotypes. Birth Defects Res. 2018, 110, 1539–1550. [Google Scholar] [CrossRef]
  70. Benros, M.E.; Mortensen, P.B. Role of Infection, Autoimmunity, Atopic Disorders, and the Immune System in Schizophrenia: Evidence from Epidemiological and Genetic Studies. Curr. Top. Behav. Neurosci. 2020, 44, 141–159. [Google Scholar]
  71. Lee, Y.H.; Cherkerzian, S.; Seidman, L.J.; Papandonatos, G.D.; Savitz, D.A.; Tsuang, M.T.; Goldstein, J.M.; Buka, S.L. Maternal Bacterial Infection During Pregnancy and Offspring Risk of Psychotic Disorders: Variation by Severity of Infection and Offspring Sex. Am. J. Psychiatry 2020, 177, 66–75. [Google Scholar] [CrossRef]
  72. Vasistha, N.A.; Pardo-Navarro, M.; Gasthaus, J.; Weijers, D.; Müller, M.K.; García-González, D.; Malwade, S.; Korshunova, I.; Pfisterer, U.; von Engelhardt, J.; et al. Maternal inflammation has a profound effect on cortical interneuron development in a stage and subtype-specific manner. Mol. Psychiatry 2020, 25, 2313–2329. [Google Scholar] [CrossRef]
  73. Lydholm, C.N.; Köhler-Forsberg, O.; Nordentoft, M.; Yolken, R.H.; Mortensen, P.B.; Petersen, L.; Benros, M.E. Parental Infections Before, During, and After Pregnancy as Risk Factors for Mental Disorders in Childhood and Adolescence: A Nationwide Danish Study. Biol. Psychiatry 2019, 85, 317–325. [Google Scholar] [CrossRef]
  74. Chaves Filho, A.; Mottin, M.; Lós, D.B.; Andrade, C.H.; Macedo, D.S. The tetrapartite synapse in neuropsychiatric disorders: Matrix metalloproteinases (MMPs) as promising targets for treatment and rational drug design. Biochimie 2022, 201, 79–99. [Google Scholar] [CrossRef]
  75. Meehan, C.; Harms, L.; Frost, J.D.; Barreto, R.; Todd, J.; Schall, U.; Shannon Weickert, C.; Zavitsanou, K.; Michie, P.T.; Hodgson, D.M. Effects of immune activation during early or late gestation on schizophrenia-related behaviour in adult rat offspring. Brain Behav. Immun. 2017, 63, 8–20. [Google Scholar] [CrossRef]
  76. Müller, N. Inflammation and Immunity in Schizophrenia. Immuno-Psychiatry 2021, 2, 227–240. [Google Scholar]
  77. Metcalf, S.A.; Jones, P.B.; Nordstrom, T.; Timonen, M.; Mäki, P.; Miettunen, J.; Jääskeläinen, E.; Järvelin, M.R.; Stochl, J.; Murray, G.K.; et al. Serum C-reactive protein in adolescence and risk of schizophrenia in adulthood: A prospective birth cohort study. Brain Behav. Immun. 2017, 59, 253–259. [Google Scholar] [CrossRef] [PubMed]
  78. Orlovska-Waast, S.; Benros, M.E. Autoimmune Diseases and Infections as Risk Factors for Mental Disorders. In Immuno-Psychiatry; Springer: Berlin/Heidelberg, Germany, 2021; pp. 3–16. [Google Scholar]
  79. Momtazmanesh, S.; Zare-Shahabadi, A.; Rezaei, N. Cytokine alterations in schizophrenia: An updated review. Front. Psychiatry 2019, 10, 892. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Goldsmith, D.R.; Rapaport, M.H.; Miller, B.J. A meta-analysis of blood cytokine network alterations in psychiatric patients: Comparisons between schizophrenia, bipolar disorder and depression. Mol. Psychiatry 2016, 21, 1696–1709. [Google Scholar] [CrossRef] [PubMed]
  81. Miller, B.J.; Buckley, P.; Seabolt, W.; Mellor, A.; Kirkpatrick, B. Meta-analysis of cytokine alterations in schizophrenia: Clinical status and antipsychotic effects. Biol. Psychiatry 2011, 70, 663–671. [Google Scholar] [CrossRef]
  82. Orlovska-Waast, S.; Köhler-Forsberg, O.; Brix, S.W.; Nordentoft, M.; Kondziella, D.; Krogh, J.; Benros, M.E. Cerebrospinal fluid markers of inflammation and infections in schizophrenia and affective disorders: A systematic review and meta-analysis. Mol. Psychiatry 2019, 24, 869–887. [Google Scholar] [CrossRef]
  83. Wang, A.K.; Miller, B.J. Meta-analysis of Cerebrospinal Fluid Cytokine and Tryptophan Catabolite Alterations in Psychiatric Patients: Comparisons Between Schizophrenia, Bipolar Disorder, and Depression. Schizophr. Bull. 2018, 44, 75–83. [Google Scholar] [CrossRef]
  84. Brandon, A.; Cui, X.; Luan, W.; Ali, A.A.; Pertile, R.A.N.; Alexander, S.A.; Eyles, D.W. Prenatal hypoxia alters the early ontogeny of dopamine neurons. Transl. Psychiatry 2022, 12, 238. [Google Scholar] [CrossRef]
  85. Wang, M.; Ling, K.H.; Tan, J.J.; Lu, C.B. Development and differentiation of midbrain dopaminergic neuron: From bench to bedside. Cells 2020, 9, 1489. [Google Scholar] [CrossRef]
  86. Jarskog, L.F.; Xiao, H.; Wilkie, M.B.; Lauder, J.M.; Gilmore, J.H. Cytokine regulation of embryonic rat dopamine and serotonin neuronal survival in vitro. Int. J. Dev. Neurosci. Off. J. Int. Soc. Dev. Neurosci. 1997, 15, 711–716. [Google Scholar] [CrossRef]
  87. Romeo, B.; Brunet-Lecomte, M.; Martelli, C.; Benyamina, A. Kinetics of cytokine levels during antipsychotic treatment in schizophrenia: A meta-analysis. Int. J. Neuropsychopharmacol. 2018, 21, 828–836. [Google Scholar] [CrossRef] [PubMed]
  88. Scheiber, C.; Schulz, T.; Schneider, J.M.; Bechter, K.; Schneider, E.M. Old and New Biomarkers for Infection, Inflammation, and Autoimmunity in Treatment-Resistant Affective and Schizophrenic Spectrum Disorders. Pharmaceuticals 2022, 15, 299. [Google Scholar] [CrossRef] [PubMed]
  89. Dziurkowska, E.; Wesolowski, M. Cortisol as a Biomarker of Mental Disorder Severity. J. Clin. Med. 2021, 10, 5204. [Google Scholar] [CrossRef] [PubMed]
  90. Zhang, X.Y.; Zhou, D.F.; Cao, L.Y.; Wu, G.Y.; Shen, Y.C. Cortisol and cytokines in chronic and treatment-resistant patients with schizophrenia: Association with psychopathology and response to antipsychotics. Neuropsychopharmacol. Off. Publ. Am. Coll. Neuropsychopharmacol. 2005, 30, 1532–1538. [Google Scholar] [CrossRef]
  91. Kany, S.; Vollrath, J.T.; Relja, B. Cytokines in Inflammatory Disease. Int. J. Mol. Sci. 2019, 20, 6008. [Google Scholar] [CrossRef]
  92. Dawidowski, B.; Górniak, A.; Podwalski, P.; Lebiecka, Z.; Misiak, B.; Samochowiec, J. The Role of Cytokines in the Pathogenesis of Schizophrenia. J. Clin. Med. 2021, 10, 3849. [Google Scholar] [CrossRef]
  93. Kravtsov, V.V.; Shnayder, N.A.; Neznanov, N.G.; Krivopalov, A.A.; Yanov, Y.K.; Nasyrova, R.F.; Shamkina, P.A.; Gavrilyuk, O.A. Genetic predictors of cytokine response in ENT-associated encephalitis. Pers. Psychiatry Neurol. 2021, 1, 18–36. [Google Scholar] [CrossRef]
  94. Velazquez-Salinas, L.; Verdugo-Rodriguez, A.; Rodriguez, L.L.; Borca, M.V. The role of interleukin 6 during viral infections. Front. Microbiol. 2019, 10, 1057. [Google Scholar] [CrossRef]
  95. Chauhan, P.; Nair, A.; Patidar, A.; Dandapat, J.; Sarkar, A.; Saha, B. A primer on cytokines. Cytokine 2021, 145, 155458. [Google Scholar] [CrossRef]
  96. Zádor, F.; Joca, S.; Nagy-Grócz, G.; Dvorácskó, S.; Szűcs, E.; Tömböly, C.; Benyhe, S.; Vécsei, L. Pro-Inflammatory Cytokines: Potential Links between the Endocannabinoid System and the Kynurenine Pathway in Depression. Int. J. Mol. Sci. 2021, 22, 5903. [Google Scholar] [CrossRef]
  97. Vallée, A. Neuroinflammation in Schizophrenia: The Key Role of the WNT/β-Catenin Pathway. Int. J. Mol. Sci. 2022, 23, 2810. [Google Scholar] [CrossRef] [PubMed]
  98. Boiko, A.S.; Mednova, I.A.; Kornetova, E.G.; Gerasimova, V.I.; Kornetov, A.N.; Loonen, A.J.M.; Bokhan, N.A.; Ivanova, S.A. Cytokine level changes in schizophrenia patients with and without metabolic syndrome treated with atypical antipsychotics. Pharmaceuticals 2021, 14, 446. [Google Scholar] [CrossRef] [PubMed]
  99. Mendiola, A.S.; Cardona, A.E. The IL-1β phenomena in neuroinflammatory diseases. J. Neural Transm. 2018, 125, 781–795. [Google Scholar] [CrossRef]
  100. Wooff, Y.; Man, S.M.; Aggio-Bruce, R.; Natoli, R.; Fernando, N. IL-1 family members mediate cell death, inflammation and angiogenesis in retinal degenerative diseases. Front. Immunol. 2019, 10, 1618. [Google Scholar] [CrossRef] [PubMed]
  101. Becher, B.; Spath, S.; Goverman, J. Cytokine networks in neuroinflammation. Nat. Rev. Immunol. 2017, 17, 49–59. [Google Scholar] [CrossRef] [PubMed]
  102. Enache, D.; Nikkheslat, N.; Fathalla, D.; Morgan, B.P.; Lewis, S.; Drake, R.; Deakin, B.; Walters, J.; Lawrie, S.M.; Egerton, A.; et al. Peripheral immune markers and antipsychotic non-response in psychosis. Schizophr. Res. 2021, 230, 1–8. [Google Scholar] [CrossRef] [PubMed]
  103. Moustafa, S.R.; Al-Rawi, K.F.; Stoyanov, D.; Al-Dujaili, A.H.; Supasitthumrong, T.; Al-Hakeim, H.K.; Maes, M. The endogenous opioid system in schizophrenia and treatment resistant schizophrenia: Increased plasma endomorphin 2, and κ and μ opioid receptors are associated with interleukin-6. Diagnostics 2020, 10, 633. [Google Scholar] [CrossRef]
  104. Subedi, L.; Lee, S.E.; Madiha, S.; Gaire, B.P.; Jin, M.; Yumnam, S.; Kim, S.Y. Phytochemicals against TNFα-Mediated Neuroinflammatory Diseases. Int. J. Mol. Sci. 2020, 21, 764. [Google Scholar] [CrossRef]
  105. Desu, H.L.; Illiano, P.; Choi, J.S.; Ascona, M.C.; Gao, H.; Lee, J.K.; Brambilla, R. TNFR2 signaling regulates the immunomodulatory function of oligodendrocyte precursor cells. Cells 2021, 10, 1785. [Google Scholar] [CrossRef]
  106. Inoubli, O.; Jemli, A.; Ben Fredj, S.; Mechri, A.; Gaha, L.; Bel Hadj Jrad, B. Haplotypes of TNFα/β Genes Associated with Sex-Specific Paranoid Schizophrenic Risk in Tunisian Population. Dis. Markers 2018, 2018, 3502564. [Google Scholar] [CrossRef]
  107. Aytac, H.M.; Ozdilli, K.; Tuncel, F.C.; Pehlivan, M.; Pehlivan, S. Tumor Necrosis Factor-alpha (TNF-α) -238 G/A Polymorphism Is Associated with the Treatment Resistance and Attempted Suicide in Schizophrenia. Immunol. Investig. 2022, 51, 368–380. [Google Scholar] [CrossRef] [PubMed]
  108. Noto, C.; Maes, M.; Ota, V.K.; Teixeira, A.L.; Bressan, R.A.; Gadelha, A.; Brietzke, E. High predictive value of immune-inflammatory biomarkers for schizophrenia diagnosis and association with treatment resistance. World J. Biol. Psychiatry Off. J. World Fed. Soc. Biol. Psychiatry 2015, 16, 422–429. [Google Scholar] [CrossRef] [PubMed]
  109. Ta, T.T.; Dikmen, H.O.; Schilling, S.; Chausse, B.; Lewen, A.; Hollnagel, J.O.; Kann, O. Priming of microglia with IFN-γ slows neuronal gamma oscillations in situ. Proc. Natl. Acad. Sci. USA 2019, 116, 4637–4642. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Upthegrove, R.; Khandaker, G.M. Cytokines, Oxidative Stress and Cellular Markers of Inflammation in Schizophrenia. Curr. Top. Behav. Neurosci. 2020, 44, 49–66. [Google Scholar] [PubMed]
  111. Kaplanski, G. Interleukin-18: Biological properties and role in disease pathogenesis. Immunol. Rev. 2018, 281, 138–153. [Google Scholar] [CrossRef]
  112. Syed, A.A.S.; He, L.; Shi, Y.; Mahmood, S. Elevated levels of IL-18 associated with schizophrenia and first episode psychosis: A systematic review and meta-analysis. Early Interv. Psychiatry 2021, 15, 896–905. [Google Scholar] [CrossRef]
  113. Mantovani, A.; Dinarello, C.A.; Molgora, M.; Garlanda, C. Interleukin-1 and Related Cytokines in the Regulation of Inflammation and Immunity. Immunity 2019, 50, 778–795. [Google Scholar] [CrossRef]
  114. Dunleavy, C.; Elsworthy, R.J.; Upthegrove, R.; Wood, S.J.; Aldred, S. Inflammation in first-episode psychosis: The contribution of inflammatory biomarkers to the emergence of negative symptoms, a systematic review and meta-analysis. Acta Psychiatr. Scand. 2022, 146, 6–20. [Google Scholar] [CrossRef]
  115. Fang, X.; Zhang, Y.; Fan, W.; Tang, W.; Zhang, C. Interleukin-17 Alteration in First-Episode Psychosis: A Meta-Analysis. Mol. Neuropsychiatry 2017, 3, 135–140. [Google Scholar] [CrossRef]
  116. Marcinowicz, P.; Więdłocha, M.; Zborowska, N.; Dębowska, W.; Podwalski, P.; Misiak, B.; Tyburski, E.; Szulc, A. A Meta-Analysis of the Influence of Antipsychotics on Cytokines Levels in First Episode Psychosis. J. Clin. Med. 2021, 10, 2488. [Google Scholar] [CrossRef]
  117. Pandolfo, G.; Genovese, G.; Casciaro, M.; Muscatello, M.; Bruno, A.; Pioggia, G.; Gangemi, S. IL-33 in Mental Disorders. Medicina 2021, 57, 315. [Google Scholar] [CrossRef] [PubMed]
  118. Liu, J.Y.; Chen, H.Y.; Lin, J.J.; Lu, M.K.; Tan, H.P.; Jang, F.L.; Lin, S.H. Alterations of plasma cytokine biomarkers for identifying age at onset of schizophrenia with neurological soft signs. Int. J. Med. Sci. 2020, 17, 255–262. [Google Scholar] [CrossRef] [PubMed]
  119. Lin, A.; Kenis, G.; Bignotti, S.; Tura, G.J.; De Jong, R.; Bosmans, E.; Pioli, R.; Altamura, C.; Scharpé, S.; Maes, M. The inflammatory response system in treatment-resistant schizophrenia: Increased serum interleukin-6. Schizophr. Res. 1998, 32, 9–15. [Google Scholar] [CrossRef]
  120. Kartalcı, Ş.; Gönenir Erbay, L.; Porgalı Zayman, E.; Otlu, Ö.; Bay Karabulut, A.; Kartalcı, G. IL-4, TGF-β, NF-κB and MPO levels in Patients with Treatment Resistant Schizophrenia. Tedaviye Dirençli Şizofreni Hastalarında IL-4, TGF-β, NF-κB ve MPO Düzeyleri. Turk psikiyatri dergisi = Turk. J. Psychiatry 2016, 27, 170–175. [Google Scholar]
  121. Luo, Y.; He, H.; Zhang, J.; Ou, Y.; Fan, N. Changes in serum TNF-α, IL-18, and IL-6 concentrations in patients with chronic schizophrenia at admission and at discharge. Compr. Psychiatry 2019, 90, 82–87. [Google Scholar] [CrossRef]
  122. Mongan, D.; Ramesar, M.; Föcking, M.; Cannon, M.; Cotter, D. Role of inflammation in the pathogenesis of schizophrenia: A review of the evidence, proposed mechanisms and implications for treatment. Early Interv. Psychiatry 2020, 14, 385–397. [Google Scholar] [CrossRef]
  123. Schwieler, L.; Larsson, M.K.; Skogh, E.; Kegel, M.E.; Orhan, F.; Abdelmoaty, S.; Finn, A.; Bhat, M.; Samuelsson, M.; Lundberg, K.; et al. Increased levels of IL-6 in the cerebrospinal fluid of patients with chronic schizophrenia--significance for activation of the kynurenine pathway. J. Psychiatry Neurosci. JPN 2015, 40, 126–133. [Google Scholar] [CrossRef]
  124. Wang, J.; Zhao, Y.; Li, C.; Zhou, C.; Tang, X.; Zhu, Q. Analysis of interleukin levels in patients with treatment-resistant schizophrenia. J. Clin. Med. Pract. 2020, 24, 96–98. [Google Scholar]
  125. Reale, M.; Costantini, E.; Greig, N.H. Cytokine Imbalance in Schizophrenia. From Research to Clinic: Potential Implications for Treatment. Front. Psychiatry 2021, 12, 536257. [Google Scholar] [CrossRef]
  126. Wilbers, R.; van Raaij, D.R.; Westerhof, L.B.; Bakker, J.; Smant, G.; Schots, A. Re-evaluation of IL-10 signaling reveals novel insights on the contribution of the intracellular domain of the IL-10R2 chain. PLoS ONE 2017, 12, e0186317. [Google Scholar] [CrossRef]
  127. Saxton, R.A.; Tsutsumi, N.; Su, L.L.; Abhiraman, G.C.; Mohan, K.; Henneberg, L.T.; Aduri, N.G.; Gati, C.; Garcia, K.C. Structure-based decoupling of the pro- and anti-inflammatory functions of interleukin-10. Science 2021, 371, eabc8433. [Google Scholar] [CrossRef] [PubMed]
  128. Patel, K.R.; Cherian, J.; Gohil, K.; Atkinson, D. Schizophrenia: Overview and treatment options. P T: A Peer-Rev. J. Formul. Manag. 2014, 39, 638–645. [Google Scholar]
  129. Zubov, D.S.; Dorofeikova, M.V.; Petrova, N.N.; Dorofeykov, V.V.; Ivanov, M.V. Changes in levels of neuromarkers and cognitive functioning of patients with treatment-resistant schizophrenia. V.M. Bekhterev Rev. Psychiatry Med. Psychol. 2016, 3, 51–56. [Google Scholar]
  130. Melbourne, J.K.; Feiner, B.; Rosen, C.; Sharma, R.P. Targeting the Immune System with Pharmacotherapy in Schizophrenia. Curr. Treat. Options Psychiatry 2017, 4, 139–151. [Google Scholar] [CrossRef] [PubMed]
  131. Kroken, R.A.; Sommer, I.E.; Steen, V.M.; Dieset, I.; Johnsen, E. Constructing the Immune Signature of Schizophrenia for Clinical Use and Research; An Integrative Review Translating Descriptives Into Diagnostics. Front. Psychiatry 2019, 9, 753. [Google Scholar] [CrossRef]
  132. Sommer, I.E.; van Westrhenen, R.; Begemann, M.J.; de Witte, L.D.; Leucht, S.; Kahn, R.S. Efficacy of anti-inflammatory agents to improve symptoms in patients with schizophrenia: An update. Schizophr. Bull. 2014, 40, 181–191. [Google Scholar] [CrossRef]
  133. Akhondzadeh, S.; Tabatabaee, M.; Amini, H.; Ahmadi Abhari, S.A.; Abbasi, S.H.; Behnam, B. Celecoxib as adjunctive therapy in schizophrenia: A double-blind, randomized and placebo-controlled trial. Schizophr. Res. 2007, 90, 179–185. [Google Scholar] [CrossRef]
  134. Müller, N.; Riedel, M.; Scheppach, C.; Brandstätter, B.; Sokullu, S.; Krampe, K.; Ulmschneider, M.; Engel, R.R.; Möller, H.J.; Schwarz, M.J. Beneficial antipsychotic effects of celecoxib add-on therapy compared to risperidone alone in schizophrenia. Am. J. Psychiatry 2002, 159, 1029–1034. [Google Scholar] [CrossRef]
  135. Attari, A.; Mojdeh, A.; Soltani, F.A.S.K.; Najarzadegan, M.R. Aspirin inclusion in antipsychotic treatment on severity of symptoms in schizophrenia: A randimized clinical trial. Iran. J. Psychiatry Behav. Sci. 2017, 11, e5848. [Google Scholar]
  136. Laan, W.; Grobbee, D.E.; Selten, J.P.; Heijnen, C.J.; Kahn, R.S.; Burger, H. Adjuvant aspirin therapy reduces symptoms of schizophrenia spectrum disorders: Results from a randomized, double-blind, placebo-controlled trial. J. Clin. Psychiatry 2010, 71, 520–527. [Google Scholar] [CrossRef]
  137. Shen, H.; Li, R.; Yan, R.; Zhou, X.; Feng, X.; Zhao, M.; Xiao, H. Adjunctive therapy with statins in schizophrenia patients: A meta-analysis and implications. Psychiatry Res. 2018, 262, 84–93. [Google Scholar] [CrossRef] [PubMed]
  138. Vincenzi, B.; Stock, S.; Borba, C.P.; Cleary, S.M.; Oppenheim, C.E.; Petruzzi, L.J.; Fan, X.; Copeland, P.M.; Freudenreich, O.; Cather, C.; et al. A randomized placebo-controlled pilot study of pravastatin as an adjunctive therapy in schizophrenia patients: Effect on inflammation, psychopathology, cognition and lipid metabolism. Schizophr. Res. 2014, 159, 395–403. [Google Scholar] [CrossRef] [PubMed]
  139. Rees, L.; King, G.M. Intensive Cortisone Therapy in Schizophrenia. J. Ment. Sci. 1956, 102, 155–159. [Google Scholar] [CrossRef] [PubMed]
  140. Nasib, L.G.; Sommer, I.E.; Winter-van Rossum, I.; de Vries, J.; Gangadin, S.S.; Oomen, P.P.; Judge, G.; Blom, R.E.; Luykx, J.J.; van Beveren, N.; et al. Prednisolone versus placebo addition in the treatment of patients with recent-onset psychotic disorder: A trial design. Trials 2020, 21, 492. [Google Scholar] [CrossRef] [PubMed]
  141. Miller, B.J.; Dias, J.K.; Lemos, H.P.; Buckley, P.F. An open-label, pilot trial of adjunctive tocilizumab in schizophrenia. J. Clin. Psychiatry 2016, 77, 275–276. [Google Scholar] [CrossRef]
  142. Girgis, R.R.; Ciarleglio, A.; Choo, T.; Haynes, G.; Bathon, J.M.; Cremers, S.; Kantrowitz, J.T.; Lieberman, J.A.; Brown, A.S. A Randomized, Double-Blind, Placebo-Controlled Clinical Trial of Tocilizumab, An Interleukin-6 Receptor Antibody, For Residual Symptoms in Schizophrenia. Neuropsychopharmacol. Off. Publ. Am. Coll. Neuropsychopharmacol. 2018, 43, 1317–1323. [Google Scholar] [CrossRef]
  143. Grüber, L.; Bunse, T.; Weidinger, E.; Reichard, H.; Müller, N. Adjunctive recombinant human interferon gamma-1b for treatment-resistant schizophrenia in 2 patients. J. Clin. Psychiatry 2014, 75, 1266–1267. [Google Scholar] [CrossRef]
  144. Lennox, B.R.; Tomei, G.; Vincent, S.A.; Yeeles, K.; Pollard, R.; Palmer-Cooper, E.; Jones, P.; Zandi, M.S.; Coles, A. Study of immunotherapy in antibody positive psychosis: Feasibility and acceptability (SINAPPS1). J. Neurol. Neurosurg. Psychiatry 2019, 90, 365–367. [Google Scholar] [CrossRef]
  145. Zandi, M.S.; Deakin, J.B.; Morris, K.; Buckley, C.; Jacobson, L.; Scoriels, L.; Cox, A.L.; Coles, A.J.; Jones, P.B.; Vincent, A.; et al. Immunotherapy for patients with acute psychosis and serum N-Methyl D-Aspartate receptor (NMDAR) antibodies: A description of a treated case series. Schizophr. Res. 2014, 160, 193–195. [Google Scholar] [CrossRef]
  146. Berk, M.; Copolov, D.; Dean, O.; Lu, K.; Jeavons, S.; Schapkaitz, I.; Anderson-Hunt, M.; Judd, F.; Katz, F.; Katz, P.; et al. N-acetyl cysteine as a glutathione precursor for schizophrenia--a double-blind, randomized, placebo-controlled trial. Biol. Psychiatry 2008, 64, 361–368. [Google Scholar] [CrossRef]
  147. Farokhnia, M.; Azarkolah, A.; Adinehfar, F.; Khodaie-Ardakani, M.R.; Hosseini, S.M.; Yekehtaz, H.; Tabrizi, M.; Rezaei, F.; Salehi, B.; Sadeghi, S.M.; et al. N-acetylcysteine as an adjunct to risperidone for treatment of negative symptoms in patients with chronic schizophrenia: A randomized, double-blind, placebo-controlled study. Clin. Neuropharmacol. 2013, 36, 185–192. [Google Scholar] [CrossRef] [PubMed]
  148. 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. J. Eur. Coll. Neuropsychopharmacol. 2017, 27, 8–18. [Google Scholar] [CrossRef] [PubMed]
  149. Cho, M.; Lee, T.Y.; Kwak, Y.B.; Yoon, Y.B.; Kim, M.; Kwon, J.S. Adjunctive use of anti-inflammatory drugs for schizophrenia: A meta-analytic investigation of randomized controlled trials. Aust. N. Z. J. Psychiatry 2019, 53, 742–759. [Google Scholar] [CrossRef] [PubMed]
  150. Hong, J.; Bang, M. Anti-inflammatory Strategies for Schizophrenia: A Review of Evidence for Therapeutic Applications and Drug Repurposing. Clin. Psychopharmacol. Neurosci. Off. Sci. J. Korean Coll. Neuropsychopharmacol. 2020, 18, 10–24. [Google Scholar] [CrossRef] [PubMed]
  151. Liu, M.; Saredy, J.; Zhang, R.; Shao, Y.; Sun, Y.; Yang, W.Y.; Wang, J.; Liu, L.; Drummer, C., 4th; Johnson, C.; et al. Approaching Inflammation Paradoxes-Proinflammatory Cytokine Blockages Induce Inflammatory Regulators. Front. Immunol. 2020, 11, 554301. [Google Scholar] [CrossRef]
  152. de Lange, K.M.; Moutsianas, L.; Lee, J.C.; Lamb, C.A.; Luo, Y.; Kennedy, N.A.; Jostins, L.; Rice, D.L.; Gutierrez-Achury, J.; Ji, S.G.; et al. Genome-wide association study implicates immune activation of multiple integrin genes in inflammatory bowel disease. Nat. Genet. 2017, 49, 256–261. [Google Scholar] [CrossRef]
  153. Fuster, J.J.; Walsh, K. Somatic Mutations and Clonal Hematopoiesis: Unexpected Potential New Drivers of Age-Related Cardiovascular Disease. Circ. Res. 2018, 122, 523–532. [Google Scholar] [CrossRef]
  154. Ferrucci, L.; Fabbri, E. Inflammageing: Chronic inflammation in ageing, cardiovascular disease, and frailty. Nat. Rev. Cardiol. 2018, 15, 505–522. [Google Scholar] [CrossRef]
  155. Bennett, J.M.; Reeves, G.; Billman, G.E.; Sturmberg, J.P. Inflammation-Nature’s Way to Efficiently Respond to All Types of Challenges: Implications for Understanding and Managing “the Epidemic” of Chronic Diseases. Front. Med. 2018, 5, 316. [Google Scholar] [CrossRef]
  156. Johnson, C.; Drummer, C., 4th; Virtue, A.; Gao, T.; Wu, S.; Hernandez, M.; Singh, L.; Wang, H.; Yang, X.F. Increased Expression of Resistin in MicroRNA-155-Deficient White Adipose Tissues May Be a Possible Driver of Metabolically Healthy Obesity Transition to Classical Obesity. Front. Physiol. 2018, 9, 1297. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Timeline of antipsychotics (APs) approved by the Food and Drug Administration (FDA) [19].
Figure 1. Timeline of antipsychotics (APs) approved by the Food and Drug Administration (FDA) [19].
Ijms 23 11324 g001
Figure 2. Hypotheses of treatment-resistant schizophrenia (TRS).
Figure 2. Hypotheses of treatment-resistant schizophrenia (TRS).
Ijms 23 11324 g002
Figure 3. Mechanisms of neuroinflammation that are associated with the treatment-resistant schizophrenia (TRS).
Figure 3. Mechanisms of neuroinflammation that are associated with the treatment-resistant schizophrenia (TRS).
Ijms 23 11324 g003
Figure 4. Potential role of normal and abnormal cytokine levels in the cytokine imbalance as a biomarker of treatment-resistant schizophrenia (TRS). Note: green—low risk; yellow—middle risk; red—high risk; red + attention sign—very high risk.
Figure 4. Potential role of normal and abnormal cytokine levels in the cytokine imbalance as a biomarker of treatment-resistant schizophrenia (TRS). Note: green—low risk; yellow—middle risk; red—high risk; red + attention sign—very high risk.
Ijms 23 11324 g004
Table 1. Hypotheses for the development of treatment-resistant schizophrenia.
Table 1. Hypotheses for the development of treatment-resistant schizophrenia.
HypothesisMechanismReferences
GeneticGenetic predisposition to low affinity of targets (dopaminergic receptors) to APs of the first and new generations.[22,23]
NeurodevelopmentalCongenital minor anomalies of brain development (microdysgenesis) in brain regions critical for Sch development.[24,25]
NeurotransmitterViolation of the synthesis, release, or breakdown of dopamine and other neurotransmitters (serotonin, melatonin, etc.).[19,26,27,28]
MetabolicPrimary (genetically determined) and secondary metabolic disorders of APs of the first and new generation in the liver.[29,30,31]
TransportPrimary (genetically determined) and secondary impairment of expression and/or functional activity of APs transporter proteins of the first and new generations across the blood–brain barrier.[32,33,34]
Oxidative stressViolation of the prooxidant-antioxidant balance in favor of the former, which leads to oxidative damage to cellular lipids, proteins, enzymes, carbohydrates and DNA, which contributes to a worsening of the course and an unfavorable outcome of Sch.[35,36,37]
InflammatoryPrimary (genetically determined) and secondary violation of the cytokine status (absolute or relative hyperproduction of pro-inflammatory cytokines).[38,39,40]
MicrobiomeMicrobiota through the gut–brain axis is associated with the development and severity of Sch, intestinal microbiota is associated with the response to APs.[41,42,43]
NutritionalDeficiency or excess of nutrients (vitamins, minerals, amino acids) necessary for the functioning of the dopaminergic system of the brain.[44,45,46]
Note: APs—antipsychotics; DNA—deoxyribonucleic acid; Sch—schizophrenia.
Table 2. Studies of the role of neuroinflammation in the development of treatment-resistant schizophrenia.
Table 2. Studies of the role of neuroinflammation in the development of treatment-resistant schizophrenia.
Author, YearMechanismPathogenesisReference
Meehan et al.,
2017
Prenatal, perinatal and postnatal infectionImmune activation. Violation of neurogenesis processes, including dopaminergic and glutamatergic neurotransmission.[75]
Frank et al.,
2018
Sensitization or kindlingStimulation of the immune response. Activation of cell proliferation, increased production and release of pro-inflammatory cytokines.[57]
Momtazmanesh et al.,
2019
Cytokine imbalance Increased serum levels of pro-inflammatory cytokines IL-1β, IL-6 and TGF-β.[79]
Wang et al.,
2020
Cytokine imbalanceInteractions between cytokines and neurotransmitters in certain areas of the brain, as well as during brain development. Induction of IL-1β conversion of mesencephalic progenitor cells into a dopaminergic phenotype. Reduced survival of serotonergic neurons through IL-6.[83]
Kumar et al.,
2020
Sensitization or kindlingStimulation of the glutamatergic system, ionotropic and metabotropic glutamate receptors that excite amino acid transporters. Increased levels of glutamate in the anterior cingulate cortex.[61]
Woodburn et al.,
2021
Changes in the functional activity of microgliaPriming of microglia causes an exaggerated immune response. Proliferation and increased production of pro-inflammatory cytokines. [52]
Müller et al.,
2021
Prenatal, perinatal and postnatal infectionIncreased levels of CRP and pro-inflammatory cytokines in childhood.[76]
Dziurkowska et al.,
2021
Cytokine imbalanceIncreased plasma levels of IL-2 and IL-6, activation of IRS. Positive correlation of IL-2, IL-6 and cortisol, hypercortisolemia.[89]
Woodburn et al.,
2021
Sensitization or kindlingPro-inflammatory immune response in the CNS. Activation and proliferation of microglia. Mediated neurotransmitter disorders.[52]
Rovira et al.,
2022
Prenatal, perinatal and postnatal infectionViolation of the structure, exposure to inflammatory factors, neurochemical changes. Increased dopamine levels, impaired COMT activity.[66]
Note: CNS—central nervous system; COMT—catechol-O-methyl transferase; CRP—C-reactive protein; IL-1β—interleukin 1 β; IL-2—interleukin 2; IL-6—interleukin 6; IRS—inflammatory response system; TGF-β—transforming growth factor beta.
Table 3. Pro-inflammatory and anti-inflammatory cytokines.
Table 3. Pro-inflammatory and anti-inflammatory cytokines.
Pro-Inflammatory CytokinesAnti-Inflammatory Cytokines
Ciliary neurotrophic factor (CNTF)
Granulocytic-macrophage colony-stimulating factor (GM-CSF)
Interferon gamma (IFN-γ)
Interleukin 20 (IL-20)
Interleukin 1 alpha (IL1-α)
Interleukin 1 β (IL1-β)
Interleukin 11 (IL-11)
Interleukin 12 (IL-12)
Interleukin 17 (IL-17)
Interleukin 18 (IL-18)
Interleukin 18 (IL-8)
Interleukin 33 (IL-33)
Interleukin 6 (IL-6)
Leukemia inhibitory factor (LIF)
Oncostatin M (OSM)
Transforming growth factor beta (TGF-β)
Tumor necrosis factor alpha (TNF-α)
Interleukin 1 receptor antagonist (IL-1Ra)
Interleukin 10 (IL-10)
Interleukin 11 (IL-11)
Interleukin 13 (IL-13)
Interleukin 4 (IL-4)
Interleukin 6 (IL-6)
Interleukin-18-binding protein (IL-18BP)
Transforming growth factor beta (TGF-β)
Table 4. Role of pro-inflammatory cytokines in treatment-resistant schizophrenia.
Table 4. Role of pro-inflammatory cytokines in treatment-resistant schizophrenia.
CytokineGene:
OMIM
Role in NeuroinflammationRole in TRS References
IL-1βIL1B:
147720
Stimulation of the synthesis of other pro-inflammatory and chemotactic mediators in the CNS.
Stimulation of aberrant release and accumulation of glutamate, which subsequently leads to neuronal death in most neurodegenerative diseases.
+/− or +[99,100,101,102]
TNF-αTNFA:
191160
Regulation of several processes including sleep, learning and memory, synaptic plasticity and astrocytic-induced synaptic strengthening. Initiation of inflammatory, apoptotic and neurodegenerative cascades, while TNF-α signaling via TNFR2 is anti-inflammatory and cytoprotective, resulting in induction of proliferation, differentiation, angiogenesis and tissue repair.+++[38,104,105,106,107,108]
IFN-γIFNG: 147570Priming of microglia, which is associated with various cellular adaptations, including changes in morphology, upregulation of receptors and an increase in pro-inflammatory cytokines.+/−[102,109,110]
IL-12AIL12A: 161560Stimulation of proliferation. Activation and increase in the cytotoxicity of NK cells and T cells.
Stimulation of differentiation in Th1. Induction of IFN-γ and TNF-α secretion, synergism with pro-inflammatory cytokines with IL-18.
+++[38,92]
IL-18IL18:
600953
Potentiation of the development of the relationship between the immune and nervous systems, since IL-18 and its receptors in the CNS mediate neuroinflammation of the brain, modulating homeostasis and behavior.++[111,112]
IL-8CXCL8: 146930Increased migration of neutrophils, T cells and monocytes, whose enzymes produce free oxygen radicals Indirect increase in oxidative stress, which can lead to neuronal death.+++[102,113,114]
IL-17IL17A: 603149Stimulation of macrophages and microglia to secrete pro-inflammatory cytokines in the CNS.+++[38,109,115]
Note: (+/−)—questionable prognostic role in the development of TRS; (+)—mild prognostic role in the development of TRS; (++)—moderate prognostic role in the development of TRS; (+++)—significant prognostic role in the development of TRS; CNS—central nervous system; IFN-γ—interferon gamma; IL-12—interleukin 12; IL-17—interleukin 17; IL-18—interleukin 18; IL-1β—interleukin 1 β; IL-8—interleukin 8; NK cells—natural killer cells; T cells—T-lymphocytes; Th1—type 1 helper T cells; TNFR2—tumor necrosis factor receptor 2; TNF-α—tumor necrosis factor alpha.
Table 5. Role of anti-inflammatory cytokines in treatment-resistant schizophrenia.
Table 5. Role of anti-inflammatory cytokines in treatment-resistant schizophrenia.
CytokineGene:
OMIM
Role in NeuroinflammationRole in TRS References
IL-4IL4: 147780Initiation of T-helper differentiation into T-helper 2 lymphocytes.
Increased Th2 cytotoxicity.
Modulation of the function of macrophages and microglial cells.
Decreased cytotoxicity.
+/−[118,119]
IL-6IL6: 147620A key role in the processes associated with immunity and neuroinflammation.
Modulation of the sensitivity of neurons to neurotransmitters.
+++[120,121,122,123,124]
IL-10IL10: 124092Initiation of cellular effects through canonical JAK/ STAT, which includes JAK1 and STAT3.
Induction of expression of genes associated with immunosuppression.
+++[38,116,125,126,127]
Note: +/−—mild prognostic role in the development of TRS; +++—significant prognostic role in the development of TRS; IL-10—interleukin 10; IL-10R1—inter-leukin-10 receptor 1; IL-10R2—interleukin-10 receptor 2; IL-4—interleukin 4; IL-6—interleukin 6; JAK—Janus kinase; JAK1—Janus kinase 1; STAT—signal transducer and activator of transcription; STAT3—signal transducer and activator of transcription 3.
Table 6. Perspective strategies for anti-inflammatory therapy in treatment-resistant schizophrenia.
Table 6. Perspective strategies for anti-inflammatory therapy in treatment-resistant schizophrenia.
DrugMechanismResultsReferences
Non-steroidal anti-inflammatory drugs
CelecoxibSelective inhibition of COX-2.Significant reduction in PANSS positive TRS symptom scores and overall PANSS score, but no significant change in negative TRS symptoms. Improvement in conceptual disorganization and abstract thinking by PANSS in patients with TRS.[133,134]
Acetylsalicylic acidInhibition of COX-1 and c COX-2.Improvement in PANSS symptoms.[135,136]
Statins
SimvastatinInhibition of HMG-CoA reductase, anti-inflammatory effect, reduction of pro-inflammatory cytokines (IL-1β, IL-6, TNF-α) and CRP.Decrease in negative symptom scores on the PANSS scale in patients with TRS, decrease in the total score on the PANSS scale.[137]
PravastatinInhibition of HMG-CoA reductase, anti-inflammatory effect, reduction of pro-inflammatory cytokines (IL-1β, IL-6, TNF-α) and CRP.Marked decrease in scores
positive symptoms on the PANSS scale.
[138]
Corticosteroids
CortisoneInfluence on carbohydrate and electrolyte metabolism, anti-inflammatory (inhibition of phospholipase A2), desensitizing and anti-allergic, immunosuppressive effects.Most patients with Sch did not show significant changes in Sch symptoms.[139]
PrednisoloneSuppression of the function of leukocytes and tissue macrophages. Limitation of migration of leukocytes to the area of inflammation, impairment of the ability of macrophages to phagocytosis, as well as to the formation of IL-1, inhibition of the activity of phospholipase A2, suppression of the release of COX-1 and COX-2, etc.There was no significant difference in improvement in the severity of Sch symptoms with the placebo group in patients with Sch.[140]
Monoclonal antibody
Tocilizumab Selective binding and suppression of expression and functional activity of IL-6 receptors.No significant change in scores for positive and negative TRS symptoms, but improvement in BACS cognition.[141,142]
Cytokines
- IFN-γ-1bActivation of macrophages and induction of expression of the class II major histocompatibility complex molecule, inhibition of virus replication.A pronounced decrease in the total PANSS score in patients with TRS.[143]
Intravenous immunoglobulins
- IgGIncreasing the content of antibodies in the blood to a physiological level, creating passive immunity.A pronounced decrease in the total PANSS score in patients with antibody positive psychosis. Most patients gave a clinical response to therapy.[144,145]
Other groups of drugs
Mucolytics/antioxidants:
- N-acetylcysteine
Precursor of the biological antioxidant glutathione, anti-inflammatory and antioxidant effect.A decrease in scores on all three PANSS scales, an improvement on the CGI-S, CGI-I scales in patients with TRS. The reduction in negative symptom scores on the PANSS scale was more significant in patients with TRS.[146,147]
Antibiotics:
- Minocycline
Bacteriostatic action due to the suppression of protein synthesis by reversible binding to the 30S ribosomal subunit of sensitive microorganisms.Decrease in scores on all three PANSS scales, improvement in BPRS scores, no changes in cognitive function in patients with TRS.[148]
Polyunsaturated fatty acids:
- Omega-3 fatty acids
Antioxidant, anti-inflammatory and neuroprotective effect.Significant improvement on the three PANSS scales, as well as improvement in cognitive functions, was not revealed.[149,150]
Note: BACS—Brief Assessment of Cognition in Schizophrenia; BPRS—Brief Psychiatric Rating Scale; CGI-I—Clinical Global Impression—Improvement; CGI-S—Clinical Global Impression—Severity; COX-1—cyclooxygenase-1; COX-2—cyclooxygenase-2; CRP—C-reactive protein; HMG-CoA—3-hydroxy-3-methyl-glutaryl-coenzyme A reductase; IFN-γ-1b—interferon-γ-1b; IgG—immunoglobulins G; PANSS—Positive and Negative Syndrome Scale.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Shnayder, N.A.; Khasanova, A.K.; Strelnik, A.I.; Al-Zamil, M.; Otmakhov, A.P.; Neznanov, N.G.; Shipulin, G.A.; Petrova, M.M.; Garganeeva, N.P.; Nasyrova, R.F. Cytokine Imbalance as a Biomarker of Treatment-Resistant Schizophrenia. Int. J. Mol. Sci. 2022, 23, 11324. https://doi.org/10.3390/ijms231911324

AMA Style

Shnayder NA, Khasanova AK, Strelnik AI, Al-Zamil M, Otmakhov AP, Neznanov NG, Shipulin GA, Petrova MM, Garganeeva NP, Nasyrova RF. Cytokine Imbalance as a Biomarker of Treatment-Resistant Schizophrenia. International Journal of Molecular Sciences. 2022; 23(19):11324. https://doi.org/10.3390/ijms231911324

Chicago/Turabian Style

Shnayder, Natalia A., Aiperi K. Khasanova, Anna I. Strelnik, Mustafa Al-Zamil, Andrey P. Otmakhov, Nikolay G. Neznanov, German A. Shipulin, Marina M. Petrova, Natalia P. Garganeeva, and Regina F. Nasyrova. 2022. "Cytokine Imbalance as a Biomarker of Treatment-Resistant Schizophrenia" International Journal of Molecular Sciences 23, no. 19: 11324. https://doi.org/10.3390/ijms231911324

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