Next Article in Journal
Maternal LPS Exposure Enhances the 5-HT Level in the Prefrontal Cortex of Autism-like Young Offspring
Next Article in Special Issue
Novel Compounds in the Treatment of Schizophrenia—A Selective Review
Previous Article in Journal
Effects of Maternal Separation and Subsequent Stress on Behaviors and Brain Monoamines in Rats
Previous Article in Special Issue
Effectiveness of SGA-LAIs on Clinical, Cognitive, and Social Domains in Schizophrenia: Results from a Prospective Naturalistic Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Is It Time for a Paradigm Shift in the Treatment of Schizophrenia? The Use of Inflammation-Reducing and Neuroprotective Drugs—A Review

by
Antonino Messina
1,*,
Carmen Concerto
1,
Alessandro Rodolico
1,
Antonino Petralia
1,
Filippo Caraci
2,3 and
Maria Salvina Signorelli
1
1
Department of Clinical and Experimental Medicine, Institute of Psychiatry, University of Catania, 95123 Catania, Italy
2
Department of Drug and Health Sciences, University of Catania, 95123 Catania, Italy
3
Unit of Translational Neuropharmacology and Translational Neurosciences, Oasi Research Institute-IRCCS, 94018 Troina, Italy
*
Author to whom correspondence should be addressed.
Brain Sci. 2023, 13(6), 957; https://doi.org/10.3390/brainsci13060957
Submission received: 20 May 2023 / Revised: 3 June 2023 / Accepted: 13 June 2023 / Published: 15 June 2023
(This article belongs to the Special Issue New Insights in Psychiatric Disorder Psychopharmacology)

Abstract

:
Comprehending the pathogenesis of schizophrenia represents a challenge for global mental health. To date, although it is evident that alterations in dopaminergic, serotonergic, and glutamatergic neurotransmission underlie the clinical expressiveness of the disease, neuronal disconnections represent only an epiphenomenon. In recent years, several clinical studies have converged on the hypothesis of microglia hyperactivation and a consequent neuroinflammatory state as a pathogenic substrate of schizophrenia. Prenatal, perinatal, and postnatal factors can cause microglia to switch from M2 anti-inflammatory to M1 pro-inflammatory states. A continuous mild neuroinflammatory state progressively leads to neuronal loss, a reduction in dendritic spines, and myelin degeneration. The augmentation of drugs that reduce neuroinflammation to antipsychotics could be an effective therapeutic modality in managing schizophrenia. This review will consider studies in which drugs with anti-inflammatory and neuroprotective properties have been used in addition to antipsychotic treatment in patients with schizophrenia.

1. Introduction

Within psychotic disorders, schizophrenia represents a chronic illness with a poor outcome. Identifying the causes and treatment of severe psychiatric illnesses, such as schizophrenia, is a challenge for healthcare systems worldwide, as patients with severe mental disorders have a higher mortality rate than the general population [1]. Although atypical antipsychotics are effective in controlling the symptomatology of schizophrenia, there are no drugs to date that can impact the pathogenetic core of schizophrenia, which appears uncertain. Above all, it is a challenge to treat patients with resistant schizophrenia, i.e., the condition in which two antipsychotic drug trials have failed to cause remission. In these cases, the only effective and available drug remains clozapine [2]. Schizophrenia presents positive symptoms such as delusions and hallucinations, and negative symptoms such as poor thoughts, flat affect, apathy, social withdrawal, and cognitive and disorganized symptoms [3]. Although the causes of schizophrenia remain unclear, there is a growing interest in exploring the neuroinflammatory and immune hypothesis as a potential contributor to the disorder’s pathophysiology [4]. Mediators of neuroinflammation are cytokines that are also implicated in neurons’ generation, differentiation, and maturation. Cytokine levels under physiological conditions fluctuate at specific periods when significant changes occur in the prefrontal cortex. Specifically, a peak of Interleukins (IL) occurs in pre-school age and another peak of tumor necrosis factor-α (TNF-α) and Interleukin-6 (IL-6) in adolescence [5].
Several noxious stimuli can trigger cytokines production by microglia (Table 1), which can switch from an anti-inflammatory M2 phenotype to an M1 phenotype that fuels the neuroinflammatory process [6]. Various risk factors are correlated with neuroinflammation (Table 1). Prenatal (i.e., maternal immune activation, MIA, caused by infections during pregnancy), perinatal (i.e., hypoxia at birth) [7], and postnatal stimuli (trauma, stress, infections) can increase the immune system’s reactivity, which, through cytokines production, produces hyperactivation of microglia and, in the long run, neuronal damage, neurotransmission abnormalities, and neurodegeneration [8]. Patients with schizophrenia show higher levels of proinflammatory cytokines such as Interleukin-1β (IL-1β), IL-6 in the blood and cerebrospinal fluid of individuals with the disorder, TNF-α, and as well as increased activation of microglia and astrocytes and an unspecific inflammatory blood marker, the C-reactive protein (CRP) [9,10]. Particularly at the onset of schizophrenia and during the recurrence of psychotic episodes, blood levels of proinflammatory cytokines such as IL-1β, IL-6, and TNF-α tend to increase [11], and levels of IL-6 are associated with poor schizophrenia prognosis [12]. In fact, microglia activation during psychotic relapses results in an increased production of proinflammatory cytokines [11]. Intriguing preclinical research found that animals exposed to MIA showed an increased expression of the nod-like receptor pyrin domain-containing protein 3 (NLRP3) inflammasome. The high expression of NLPR3, which is involved in the inflammatory pathway, is associated with schizophrenia-like behavior [13]. Moreover, the activation of microglia induces increased oxidative stress related to the increase in quinolinic acid produced by the kynurenic acid pathway [14]. Indeed, tryptophan metabolism, in which the kynurenine pathway is involved, is impaired in schizophrenia [15]. Thus, high levels of kynurenic acid can damage dopaminergic and glutamatergic neurotransmission and lead to psychotic symptoms and cognitive impairment [16,17]. Furthermore, the neuroinflammation-related state of microglia activation leads to a reduction in brain-derived neurotrophic factor (BDNF), with neuron loss, reduced synaptic plasticity, and consequent neurodegeneration [18].
The gene expression of DNA sequences coding for proteins involved in TNF-α and IL-17 signaling processes appears more pronounced in schizophrenia patients than in healthy ones [19]. Multiple genes, such as FOS, IL1B, CXCL8, CASP1, CFL1, CAMP, ITPR2, and ACTG1, implicated in immune response and inflammation, are more highly expressed in schizophrenia than in the general population [19].
In light of this, a paradigm shift has been taking place in recent years regarding psychiatric disorders. Emerging evidence brings schizophrenia closer to multiple sclerosis relative to the pathogenetic basis, albeit with different anatomopathological and clinical manifestations [20]. Whether the pathogenetic processes of schizophrenia are similar to the neuroinflammation observed in multiple sclerosis, several clinical trials have investigated the role of anti-inflammatory and immunomodulatory therapies in treating schizophrenia. This finding is consistent with risperidone’s efficacy in reducing the blood concentration of IL-6, which appears higher in patients with schizophrenia than in controls [21]. Recently, lumateperone, an atypical antipsychotic, which modulates dopaminergic, serotoninergic, and glutamatergic neurotransmission, has been shown to have anti-inflammatory activity, reducing the levels of IL-1β, IL-6, and TNF-α and promoting the restoring of blood–brain barrier (BBB) integrity [22]. In a study conducted by Fitton [23], the researchers examined the potential use of anti-inflammatory medication to treat mental disorders. Their review involved analyzing existing literature, specifically emphasizing controlled trials and systematic reviews.
The treatment of schizophrenia is a challenge for the clinician. A meta-analysis of 62 double-blind, randomized studies showed that different molecules with anti-inflammatory action improved both positive and negative symptoms of schizophrenia [24]. Given these findings, researchers have investigated the use of anti-inflammatory and monoclonal antibody drugs as promising add-on treatments for schizophrenia. Due to their anti-inflammatory properties and neuroprotective action [25], these drugs belong to different pharmacological categories and can be defined as neuroinflammatory-reducing and neuroprotective drugs (NRNDs). These drugs alleviate neuroinflammation, show a demonstrated neuroprotective effect, and improve symptoms in patients with schizophrenia. As much as treatments with second- and third-generation atypical antipsychotics are valuable tools in terms of efficacy and tolerability, they do not affect the pathogenetic mechanisms of schizophrenia, but rather the epiphenomena represented by neurotransmission abnormalities.
This systematic search followed the PRISMA guidelines. Two authors independently searched the MEDLINE, Cochrane Central Register, EMBASE, and Mendeley databases for the following entries: schizophrenia or patients with schizophrenia and celecoxib and PUFA and omega-3-fatty acids and acetylsalicylic acid and minocycline and statins and PPAR agonist and pioglitazone and rosiglitazone and ace-inhibitors and prednisolone and immunomodulators and fingolimod and monoclonal antibody and rituximab. Only English-written papers were considered (Figure 1).

2. Polyunsaturated Fatty Acids

Omega-3 and omega-6 fatty acids belong to polyunsaturated fatty acids (PUFAs). PUFAs are essential constituents of neuronal membranes and provide proper membrane function. A large cross-national study showed a correlation between low levels of PUFAs and an increased risk of schizophrenia [26]. PUFAs reduce neuroinflammation and, in patients with schizophrenia, result in a reduction in proinflammatory cytokines, such as IL-6 and TNF-α; a reduction in CRP; and an increase in BDNF, which, due to its neurotrophic action, has positive effects on cognition function [27]. Given omega-3 fatty acids’ neuroprotective and antioxidant effects, they have been proposed as a treatment in first-episode psychotic patients or in ultra-high-risk state subjects (UHR), i.e., those with subthreshold psychotic symptoms at risk of developing full-blown psychosis [28,29,30,31]. In addition, some authors found that the reduction in omega-3 fatty acid in the erythrocyte membrane (omega-3 index) could be a biomarker of risk in UHR individuals [32] and a risk factor for drug treatment resistance [33]. The efficacy and safety of PUFAs augmentation to antipsychotic therapy have been demonstrated in a meta-analysis of randomized controlled trials [34]. Another meta-analysis of RCTs reported that the assumption of 1 g/day of omega-3 fatty acid improved positive symptomatology [35]. Some authors in a randomized clinical trial (RCT) reported a reduction in violent behavior in patients with schizophrenia treated with PUFAs at twelve weeks [36]. However, another meta-analysis concluded that although there are some efficacy data, these are of poor quality, and further studies would be needed [37].

3. Statins

Like other molecules, statins, drugs that inhibit 3-hydroxy-3-methyl-glutaryl-coenzyme A (HMG-CoA) reductase by inducing a lowering of cholesterol levels, used in hypercholesterolemia, also possess anti-inflammatory activity [38]. Statins can be distinguished into hydrophilic: pravastatin and rosuvastatin; and lipophilic: atorvastatin, fluvastatin, lovastatin, pitavastatin, and simvastatin [39]. Two meta-analyses, which included six randomized clinical trials (RCTs), observed in patients taking statins in addition to antipsychotics, showed a reduction in positive and negative symptoms, compared with the control group not taking them [40,41]. At a daily dose of 40 mg, simvastatin added to risperidone proved effective in reducing negative symptoms of schizophrenia at eight weeks but did not show the same effectiveness in controlling positive symptoms [42]. Nevertheless, not all studies agree: some authors came to opposite conclusions of no efficacy [43,44], and in a meta-analysis, statins were not reported to be effective in controlling the severity of schizophrenia symptoms, regardless of the molecule’s tendency to pass the BBB [18]. In a large retrospective study performed on veterans with schizophrenia, the authors observed that the risk of incurring hospitalization was lower in patients taking statins [45]. The effect of statins may be due to their effect in reducing neuroinflammation [46], and decreasing blood values of IL-1β, IL-6, TNF-α, and C-reactive protein (CRP) [46,47]. Within the prefrontal cortex of patients with schizophrenia, the gene expression of the Toll-like receptors 4 (TLR4), pivotal in the proinflammatory pathway, is altered [48]. In schizophrenia, statins have been shown to effectively modulate both NLRP3 inflammasome and TLR pathways involved in neuroinflammation [47]. However, it is necessary to remember that not all statins are the same: some are lipophilic, while others are hydrophilic. Lipophilicity ensures their passage through the blood–brain barrier (BBB); thus, in studies involving these drugs, one must keep this in mind, as some may be biased by the ineffectiveness of statins that do not pass the BBB.

4. Peroxisome Proliferator-Activated Receptors’ Agonists

The peroxisome proliferator-activated receptors (PPARs) are intranuclear receptors, which act as transcription factors, binding to DNA and thus regulating gene expression [49]. In inflammatory processes, a key role is played by the nuclear factor kappa-light-chain-enhancer of activated B cells (NFkB), a transcription factor that stimulates the expression of enzymes involved in the prostaglandin pathway by inducing COX-2 gene expression [50]. The pro-inflammatory action of NFkB is inhibited by the PPARs, which comprise three isoforms, PPAR-α, PPAR-β/δ, and PPAR-γ [50]. PPAR-γ is widely expressed in microglia and exhibits a potent anti-inflammatory activity, influencing multiple pathways through inhibiting cytokine gene expression and prostaglandins and inducing apoptosis in activated microglia cells [51]. On the other hand, the main effect of PPAR-α is to facilitate neurotransmission processes and have a neuroprotective effect, while the action of PPAR-β/δ is unknown [52].
Because NFkB and PPARs are dysregulated in schizophrenia and are associated with higher levels of neuroinflammation [50], the agonist of PPARs can reduce inflammatory processes, reducing TNF-α and IL-6 levels [50,53]. PPARs not only inhibit NFkB gene expression, but also modulate the action of TLRs, which, as already mentioned, play a key role in the production of proinflammatory cytokines and the triggering of the neuroinflammatory process [54]. The PPARs agonists approved to date for the treatment of diabetes are rosiglitazone and pioglitazone. In preclinical studies, rosiglitazone improved memory because of its positive effect on BDNF gene expression [55].
The use of pioglitazone has been studied in patients with schizophrenia, and at a dosage of 30 mg per day for eight weeks resulted in a reduction in the severity of symptoms of the disorder [18]. It would also appear that pioglitazone, in addition to antipsychotics, improves negative symptomatology [56]. In view of the broad action of PPARs in neurons, it would be opportune to investigate PPARs agonists extensively, as also suggested in a recent review on the potential use of PPARs agonists in psychopharmacology [57].

5. AT1 Antagonists and ACE Inhibitors

Interestingly, the renin–angiotensin system (RAS) and angiotensin-converting enzyme (ACE), primarily involved in blood pressure regulation, appear to modulate PPARs and neuroinflammation and regulate GABAergic and dopaminergic neurotransmission, which are involved in schizophrenia [58,59]. According to recent evidence, RAS and ACE appear to be linked to neurodegenerative diseases and schizophrenia [60], and reduced ACE levels have been found in patients with schizophrenia [61]. Thus, using drugs that modulate RAS, such as angiotensin 1 receptor (AT1) antagonists and angiotensin-converting enzyme (ACE) inhibitors, could help treat the neuroinflammatory processes underlying the pathogenesis of schizophrenia. The pleiotropic activity of AT1 antagonists, which contributes to reducing neuroinflammation, modulating the immune response and the coagulation cascade, and protecting endothelial cells and mitochondria, can explain the role of AT1 antagonists in preventing neurodegeneration observed in schizophrenia [62]. The anti-inflammatory properties of AT1 antagonists are likely to be related to the decrease in pro-inflammatory cytokines, mediated by the reduction in gene expression of NLPR3 and NF-κB [62]. The disruption of the BBB, which is related to neuroinflammation, is increased by AT1 receptors, so the use of AT1 antagonists lowers the permeability of the BBB, thereby preventing harmful agents from penetrating the brain [62,63].
Telmisartan, an AT1 antagonist, has been shown to effectively reduce the neurotoxic effect of IL-1β that can result in neurodegeneration [64]. Moreover, the use of telmisartan appears to be efficacious, in addition to clozapine or olanzapine, in improving the symptomatology of schizophrenia [65].
Preclinical studies observed that AT1 antagonists, particularly irbesartan, losartan, and telmisartan, reduce levels of kynurenic acid, which at high levels results in the blockade of NMDA glutamate receptors, associated to the onset of psychotic symptoms [66]. In mice models, candesartan reduces hippocampal microglia activation [67].
Moreover, ACE inhibitors alter the metabolism of kynurenic acid. In vitro studies on rat cortex showed that among the various ace inhibitors, while lisinopril tends to increase kynurenic acid levels, ramipril conversely reduces them. In contrast, perindopril appears to have a neutral action on kynurenic acid levels [68].

6. Acetylsalicylic Acid and Other Nonsteroidal Anti-Inflammatory Drugs

Acetylsalicylic acid, a non-selective COX inhibitor, modulates cy-cyclooxygenase-2 (COX-2) and inhibits cyclooxygenase-1 (COX-1) irreversibly. The anti-inflammatory action of acetylsalicylic acid is achieved by inhibiting the production of thromboxanes and prostaglandins [69] and has proven effective in addition to antipsychotic therapy in reducing both positive and negative symptoms of schizophrenia [18,70,71]. The dosage of acetylsalicylic acid used in patients with schizophrenia ranged from 325 mg up to 1000 mg daily. Acetylsalicylic acid is effective in reducing the production of IL-6 and TNF-α and protecting against oxidative stress damage [72]. In a meta-analysis that considered different nonsteroidal anti-inflammatory drugs (NSAIDs), such as ibuprofen, diclofenac, naproxen sodium, and acetylsalicylic acid, it was observed that the augmentation of NSAIDs to antipsychotics was effective in reducing the severity of symptoms of schizophrenia [73].

7. Celecoxib

Celecoxib, a drug that inhibits the enzyme cyclooxygenase-2 (COX-2), has been investigated as an additional treatment option for schizophrenia. COX-2, unlike the other isoform of the enzyme COX-1, plays a specific role in the pathogenesis of inflammation [74]. COX-2 is also expressed in nervous tissue, and through the production of prostaglandin E2 modulates immune action in the central nervous system (CNS) and plays a crucial role in neuroinflammatory processes [75], with specific involvement of the hippocampus as well [76]. Various researchers have reviewed randomized clinical trials that assessed using celecoxib as an add-on treatment for schizophrenia [77]. The action of celecoxib manifests through its neuroprotective and immunomodulatory effects [78]. In a double-blind study, the combination of 400 mg/day of celecoxib with risperidone at standard dosages (2–6 mg/day), regardless of sex, age, and duration of illness, was more effective in improving positive and negative symptomatology in schizophrenia [79,80]; the same effect was observed with the association of celecoxib and amisulpride [81]. The cognitive function of patients with schizophrenia also improved following the addition of celecoxib [82]. However, other authors using 400 mg/day of celecoxib combined with an antipsychotic found no difference from using an antipsychotic [83]. It is likely that the effectiveness of celecoxib would depend on the stage of schizophrenia, being more useful in the early rather than later stages [77,84]. This datum is confirmed by a meta-analysis that concluded that further use of celecoxib is more effective in the first episode of schizophrenia [85].

8. Minocycline

Minocycline, a tetracycline antibiotic, has been investigated for its potential anti-inflammatory effects in treating schizophrenia. Preclinical studies in mice have shown that minocycline can reduce microglia activation at the hippocampal and prefrontal levels [18,86]. Many authors have conducted meta-analyses of randomized controlled trials [87,88,89,90]. They found that minocycline significantly improved negative symptoms of schizophrenia and general psychopathology and reduced inflammation markers, especially in studies where the treatment lasted longer. However, the authors did not report differences regarding positive symptoms. Minocycline combined with clozapine was an optimal treatment strategy in resistant schizophrenia. The treatment’s efficacy in the add-on can also be an effect of increased clozapine plasma levels caused by minocycline [91]. Specifically, in resistant patients with schizophrenia, improvement was mostly observed in cognitive function and in reducing avolition [92]. These findings suggest that minocycline may be a promising additional treatment for schizophrenia, particularly for patients experiencing cognitive impairment and negative symptoms [93,94,95,96]. The improvement of cognitive function was associated with a reduction in a marker of neuroinflammation interleukin-6 [97], and greater efficacy of minocycline appears to be related to higher neuroinflammation [98]. Due to its neuroprotective and anti-inflammatory properties, minocycline reduces microglia activation observed in patients with schizophrenia [99]. Minocycline may facilitate the transition from M1 to M2 by inhibiting microglia hyperactivation and related neuroinflammation [100]. The inactivation of microglia, and consequently the reduction in IL-1β, IL-6, and TNF-α levels, is mediated by the suppression of TLR4 signaling [101].
In addition to its effects on microglia, minocycline exerts neuroprotective and antiapoptotic actions [102]. The hippocampus, a formation involved in schizophrenia, is one of the targets of minocycline, which stimulates neurogenesis and reduces microglia activation [103]. Exciting speculation hypothesizes that minocycline acts on microglia and regulates the remodeling synapses and circuits involved in the “social brain” [104]. Several pieces of evidence have shown that the NMDA glutamate receptor plays a key role in the pathogenesis of schizophrenia [105]; in fact, molecules that antagonize the NMDA receptor cause the onset of psychotic symptoms. Minocycline inhibits the neurotoxicity of NMDA receptor antagonists [106].
As with other molecules considered in add on, there are conflicting studies for minocycline. In RCTs, the authors found no difference between patients taking an antipsychotic and minocycline at 200 mg/day and the group taking a placebo [107,108,109]. Nevertheless, given the amount of positive data on the use of minocycline and considering some conflicting data to date, it cannot be completely ruled out that minocycline may find efficacy in patients with the positivity of inflammatory biomarkers. Some studies do not consider patients with more severe symptoms, relapses, and the presence of negative symptoms before the start of minocycline treatment [110]. More homogeneous studies by disease duration and severity, differentiated by symptom cluster, and considering neuroinflammatory profile would be needed to clarify the usefulness of minocycline in the treatment of schizophrenia in combination with antipsychotics.

9. Prednisolone

Prednisolone, a corticosteroid, has also been studied as an adjunctive treatment for schizophrenia. Nitta [111] conducted a meta-analysis of randomized controlled trials investigating prednisolone use in schizophrenia and found some evidence for its effectiveness. However, the study had methodological limitations, and the overall effect size was small. Nevertheless, it must be considered that high cortisol levels are associated with psychotic symptoms, so the use of prednisolone may be risky [112].

10. Immunomodulator Drugs

The new frontier in treatment studies of schizophrenia is the use of immunomodulators. Among them, fingolimod, used in treating multiple sclerosis, which possesses marked anti-inflammatory and neuroprotective activity, appears effective in improving the cognitive symptoms of schizophrenia. Preclinical studies showed that fingolimod reduces microglial activation and levels of proinflammatory cytokines such as IL-6, while increasing BDNF [113]. The effect of fingolimod is expressed in the increase in white matter at the level of the corpus callosum and superior longitudinal fasciculus, and the reduction in lymphocyte counts [114]. The protective action of fingolimod would be related to the direct effect of the molecule on oligodendrocytes [115]. In an RCT, some authors found a significative improvement in negative symptomatology and global functioning in 80 patients with schizophrenia taking fingolimod, compared with as many patients taking a placebo [116].
Another drug used to treat rheumatoid arthritis is methotrexate. This drug, used once weekly at 10 mg, effectively reduces positive symptoms while remaining ineffective on negative ones [117]. However, methotrexate, which has antagonistic effects on folic acid synthesis, is burdened by severe side effects on the immune system that make it hardly usable.

11. Monoclonal Antibodies

According to emerging studies, monoclonal antibodies may also play a role in treating some psychopathological domains of schizophrenia. Monoclonal antibodies are a class of molecules that act by antagonizing the cytokines. The main field of use of this category of drugs is oncological disease. Several monoclonal antibodies exist, among which adalimumab has proven to be significantly superior to placebo in combination with risperidone in treating the negative symptoms of schizophrenia [118]. Among the many molecules, adalimumab selectively binds to TNF-α by preventing its action on the receptor [119]. Another piece of research showed an improvement in the general symptomatology of schizophrenia with efficacy in improving global functioning in a small group of resistant patients treated with rituximab [120], which targets CD20, a transmembrane protein present on B lymphocytes whose proliferation it inhibits. Cognitive improvement was observed with the administration of tocilizumab, an IL-6 antagonist [121]; this datum was not confirmed in trial, however, which did not attribute the ineffectiveness to the molecule itself, but to the fact that tocilizumab passes BBB with difficulty [122]. In a recent review, the investigation of the efficacy of rituximab and ocrelizumab on the cognitive function of patients with schizophrenia yielded controversial results. However, the use of adalimumab has been shown to be effective in controlling negative and positive symptoms of schizophrenia [123].

12. Conclusions

Although the use of anti-inflammatory drugs as supplementary treatments for schizophrenia shows potential, more research is necessary to determine their ideal usage and safety. According to the neuroinflammatory hypothesis of schizophrenia, inflammation plays a critical role in the etiology and neuro-progression of the disorder. Thus, neuroinflammation-reducing and neuroprotective drugs (NRNDs) hold promise as a potential treatment option. However, the complexity of schizophrenia and the interaction between inflammation and other biological and psychosocial factors make it challenging to identify patient groups that could benefit from NRNDs (Table 2). Therefore, future research should strive to identify biomarkers that could aid in predicting treatment response and explore the optimal dosing and duration. NRNDs could be used as an add-on to antipsychotics in some forms of schizophrenia in which the neuroinflammatory component is more significant, or in predominantly negative or cognitively impaired schizophrenia, in resistant form, and specific internist comorbidity (Table 2 and Table 3). In this regard, in patients with schizophrenia, it would be desirable for neuroinflammatory screening to be carried out, allowing patients with neuroinflammatory schizophrenia to be identified and treated appropriately.
In this regard, to improve and individualize the pharmacological treatment of schizophrenia, some authors have proposed using pro-inflammatory cytokines as a biomarker to stage schizophrenia from the prodromal stages, the first episode, to chronic forms in relation also to the predominance of negative or positive symptoms [124]. Patients with treatment-resistant forms of schizophrenia, who account for 30% of patients with schizophrenia, could benefit from a staging involving pro-inflammatory cytokines dosage to tailor therapy, using drugs that act on neuroinflammatory mechanisms [125].
NRNDs represent a new therapeutic option for patients with schizophrenia. Future research should involve case-control studies differentiated by the subtype of schizophrenia, evaluating the presence of forms with a high neuroinflammatory component versus forms of schizophrenia with a low neuroinflammatory component, as inferred from serological biomarkers. In addition, the use of NRNDs should be investigated in schizophrenia variants with the prevalence of negative or positive symptomatology and a possible impact on cognitive function.

Author Contributions

Conceptualization, A.M. and M.S.S.; methodology, A.R. and C.C.; validation, F.C., A.P. and M.S.S.; formal analysis, A.M. and A.R.; data curation, A.M.; writing—original draft preparation, A.M.; writing—review and editing, A.M., F.C. and M.S.S.; supervision, M.S.S. and F.C.; project administration, A.M. and M.S.S. 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

Suggested Data Availability Statements are available in section “MDPI Research Data Policies” at https://www.mdpi.com/ethics, accessed on 1 July 2023.

Conflicts of Interest

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

References

  1. Giannouli, V. Ethnicity, mortality, and severe mental illness. Lancet Psychiatry 2017, 4, 517. [Google Scholar] [CrossRef]
  2. Correll, C.U.; Howes, O.D. Treatment-Resistant Schizophrenia: Definition, Predictors, and Therapy Options. J. Clin. Psychiatry 2021, 82, 36608. [Google Scholar] [CrossRef]
  3. Fond, G.; Lançon, C.; Korchia, T.; Auquier, P.; Boyer, L. The Role of Inflammation in the Treatment of Schizophrenia. Front. Psychiatry 2020, 11, 160. [Google Scholar] [CrossRef]
  4. 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]
  5. Sager, R.E.H.; Walker, A.K.; Middleton, F.A.; Robinson, K.; Webster, M.J.; Gentile, K.; Wong, M.L.; Shannon Weickert, C. Changes in cytokine and cytokine receptor levels during postnatal development of the human dorsolateral prefrontal cortex. Brain Behav. Immun. 2023, 111, 186–201. [Google Scholar] [CrossRef] [PubMed]
  6. Köhler-Forsberg, O.; Müller, N.; Lennox, B.R. Editorial: The Role of Inflammation in the Etiology and Treatment of Schizophrenia. Front. Psychiatry 2020, 11, 603296. [Google Scholar] [CrossRef]
  7. Jenkins, T.A. Perinatal complications and schizophrenia: Involvement of the immune system. Front. Neurosci. 2013, 7, 110. [Google Scholar] [CrossRef] [PubMed]
  8. Müller, N. Inflammation in Schizophrenia: Pathogenetic Aspects and Therapeutic Considerations. Schizophr. Bull. 2018, 44, 973–982. [Google Scholar] [CrossRef]
  9. Girgis, R.R.; Kumar, S.S.; Brown, A.S. The Cytokine Model of Schizophrenia: Emerging Therapeutic Strategies. Biol. Psychiatry 2014, 75, 292–299. [Google Scholar] [CrossRef]
  10. Luo, Y.; He, H.; Zhang, M.; Huang, X.; Zhang, J.; Zhou, Y.; Liu, X.; Fan, N. Elevated Serum Levels of TNF-α, IL-6 and IL-18 in Chronic Schizophrenic Patients. Schizophr. Res. 2014, 159, 556–557. [Google Scholar] [CrossRef]
  11. Hong, J.; Bang, M. Anti-inflammatory Strategies for Schizophrenia: A Review of Evidence for Therapeutic Applications and Drug Repurposing. Clin. Psychopharmacol. Neurosci. 2020, 18, 10–24. [Google Scholar] [CrossRef]
  12. Lin, A.; Kenis, G.; Bignotti, S.; Tura, G.J.; De Jong, R.; Bosmans, E.; Pioli, R.; Altamura, C.; Scharpe, S.; Maes, M. The inflammatory response system in treatment-resistant schizophrenia: Increased serum interleukin-6. Schizophr. Res. 1998, 32, 9–15. [Google Scholar] [CrossRef]
  13. Ventura, L.; Freiberger, V.; Thiesen, V.B.; Dias, P.; Dutra, M.L.; Silva, B.B.; Schlindwein, A.D.; Comim, C.M. Involvement of NLRP3 inflammasome in schizophrenia-like behaviour in young animals after maternal immune activation. Acta Neuropsychiatr. 2020, 32, 321–327. [Google Scholar] [CrossRef] [PubMed]
  14. Block, M.L.; Hong, J.S. Microglia and inflammation-mediated neurodegeneration: Multiple triggers with a common mechanism. Prog. Neurobiol. 2005, 76, 77–98. [Google Scholar] [CrossRef] [PubMed]
  15. Muneer, A. Kynurenine Pathway of Tryptophan Metabolism in Neuropsychiatric Disorders: Pathophysiologic and Therapeutic Considerations. Clin. Psychopharmacol. Neurosci. 2020, 18, 507–526. [Google Scholar] [CrossRef]
  16. Erhardt, S.; Schwieler, L.; Imbeault, S.; Engberg, G. The kynurenine pathway in schizophrenia and bipolar disorder. Neuropharmacology 2017, 112, 297–306. [Google Scholar] [CrossRef] [PubMed]
  17. Tóth, F.; Cseh, E.K.; Vécsei, L. Natural Molecules and Neuroprotection: Kynurenic Acid, Pantethine and α-Lipoic Acid. Int. J. Mol. Sci. 2021, 22, 403. [Google Scholar] [CrossRef]
  18. Çakici, N.; van Beveren, N.J.M.; Judge-Hundal, G.; Koola, M.M.; Sommer, I.E.C. An update on the efficacy of anti-inflammatory agents for patients with schizophrenia: A meta-analysis. Psychol. Med. 2019, 49, 2307–2319. [Google Scholar] [CrossRef]
  19. Zhang, Y.; You, X.; Li, S.; Long, Q.; Zhu, Y.; Teng, Z.; Zeng, Y. Peripheral Blood Leukocyte RNA-Seq Identifies a Set of Genes Related to Abnormal Psychomotor Behavior Characteristics in Patients with Schizophrenia. Med. Sci. Monit. 2020, 10, 26. [Google Scholar] [CrossRef]
  20. Pape, K.; Tamouza, R.; Leboyer, M.; Zipp, F. Immunoneuropsychiatry—Novel Perspectives on Brain Disorders. Nat. Rev. Neurol. 2019, 15, 317–328. [Google Scholar] [CrossRef]
  21. Feng, Z.; Zhang, Y.; You, X.; Zhang, W.; Ma, Y.; Long, Q.; Liu, Z.; Hao, W.; Zeng, Y.; Teng, Z. Effects of risperidone on blood levels of interleukin-6 in schizophrenia: A meta-analysis. Medicine 2020, 99, e19694. [Google Scholar] [CrossRef] [PubMed]
  22. Dutheil, S.; Watson, L.S.; Davis, R.E.; Snyder, G.L. Lumateperone Normalizes Pathological Levels of Acute Inflammation through Important Pathways Known to Be Involved in Mood Regulation. J. Neurosci. 2023, 43, 863–877. [Google Scholar] [CrossRef]
  23. Fitton, R.; Sweetman, J.; Heseltine-Carp, W.; van der Feltz-Cornelis, C. Anti-Inflammatory Medications for the Treatment of Mental Disorders: A Scoping Review. Brain Behav. Immun. Health 2022, 26, 100518. [Google Scholar] [CrossRef]
  24. 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]
  25. Subbanna, M.; Shivakumar, V.; Venugopal, D.; Narayanaswamy, J.C.; Berk, M.; Varambally, S.; Venkatasubramanian, G.; Debnath, M. Impact of Antipsychotic Medication on IL-6/STAT3 Signaling Axis in Peripheral Blood Mononuclear Cells of Drug-Naive Schizophrenia Patients. Psychiatry Clin. Neurosci. 2019, 74, 64–69. [Google Scholar] [CrossRef]
  26. Gao, Y.; Hu, X.; Wang, D.; Jiang, J.; Li, M.; Qing, Y.; Yang, X.; Zhang, J.; Zhang, Y.; Wan, C. Association between Arachidonic Acid and the Risk of Schizophrenia: A Cross-National Study and Mendelian Randomization Analysis. Nutrients 2023, 15, 1195. [Google Scholar] [CrossRef]
  27. Tang, W.; Wang, Y.; Xu, F.; Fan, W.; Zhang, Y.; Fan, K.; Wang, W.; Zhang, Y.; Zhang, C. Omega-3 Fatty Acids Ameliorate Cognitive Dysfunction in Schizophrenia Patients with Metabolic Syndrome. Brain Behav. Immun. 2020, 88, 529–534. [Google Scholar] [CrossRef]
  28. Chen, A.T.; Chibnall, J.T.; Nasrallah, H.A. A Meta-Analysis of Placebo-Controlled Trials of Omega-3 Fatty Acid Augmentation in Schizophrenia: Possible Stage-Specific Effects. Ann. Clin. Psychiatry Off. J. Am. Acad. Clin. Psychiatr. 2015, 27, 289–296. [Google Scholar]
  29. Casquero-Veiga, M.; Romero-Miguel, D.; MacDowell, K.S.; Torres-Sanchez, S.; Garcia-Partida, J.A.; Lamanna-Rama, N.; Gómez-Rangel, V.; Romero-Miranda, A.; Berrocoso, E.; Leza, J.C.; et al. Omega-3 Fatty Acids during Adolescence Prevent Schizophrenia-Related Behavioural Deficits: Neurophysiological Evidences from the Prenatal Viral Infection with PolyI:C. Eur. Neuropsychopharmacol. 2021, 46, 14–27. [Google Scholar] [CrossRef] [PubMed]
  30. Frajerman, A.; Scoriels, L.; Kebir, O.; Chaumette, B. Shared Biological Pathways between Antipsychotics and Omega-3 Fatty Acids: A Key Feature for Schizophrenia Preventive Treatment? Int. J. Mol. Sci. 2021, 22, 6881. [Google Scholar] [CrossRef] [PubMed]
  31. Jones, H.J.; Borges, M.C.; Carnegie, R.; Mongan, D.; Rogers, P.J.; Lewis, S.J.; Thompson, A.D.; Zammit, S. Associations between Plasma Fatty Acid Concentrations and Schizophrenia: A Two-Sample Mendelian Randomisation Study. Lancet Psychiatry 2021, 8, 1062–1070. [Google Scholar] [CrossRef] [PubMed]
  32. Alqarni, A.; Mitchell, T.W.; McGorry, P.D.; Nelson, B.; Markulev, C.; Yuen, H.P.; Schäfer, M.R.; Berger, M.; Mossaheb, N.; Schlögelhofer, M.; et al. Comparison of Erythrocyte Omega-3 Index, Fatty Acids and Molecular Phospholipid Species in People at Ultra-High Risk of Developing Psychosis and Healthy People. Schizophr. Res. 2020, 226, 44–51. [Google Scholar] [CrossRef]
  33. Li, N.; Yang, P.; Tang, M.; Liu, Y.; Guo, W.; Lang, B.; Wang, J.; Wu, H.; Tang, H.; Yu, Y.; et al. Reduced Erythrocyte Membrane Polyunsaturated Fatty Acid Levels Indicate Diminished Treatment Response in Patients with Multi- versus First-Episode Schizophrenia. Schizophrenia 2022, 8, 7. [Google Scholar] [CrossRef] [PubMed]
  34. Pawełczyk, T.; Grancow-Grabka, M.; Kotlicka-Antczak, M.; Trafalska, E.; Pawełczyk, A. A randomized controlled study of the efficacy of six-month supplementation with concentrated fish oil rich in omega-3 polyunsaturated fatty acids in first episode schizophrenia. J. Psychiatr. Res. 2016, 73, 34–44. [Google Scholar] [CrossRef]
  35. Goh, K.K.; Chen, C.Y.-A.; Chen, C.-H.; Lu, M.-L. Effects of Omega-3 Polyunsaturated Fatty Acids Supplements on Psychopathology and Metabolic Parameters in Schizophrenia: A Meta-Analysis of Randomized Controlled Trials. J. Psychopharmacol. 2021, 35, 221–235. [Google Scholar] [CrossRef]
  36. Qiao, Y.; Mei, Y.; Han, H.; Liu, F.; Yang, X.M.; Shao, Y.; Xie, B.; Long, B. Effects of Omega-3 in the Treatment of Violent Schizophrenia Patients. Schizophr. Res. 2018, 195, 283–285. [Google Scholar] [CrossRef]
  37. Bosnjak Kuharic, D.; Kekin, I.; Hew, J.; Rojnic Kuzman, M.; Puljak, L. Interventions for Prodromal Stage of Psychosis. Cochrane Database Syst. Rev. 2019, 2019, CD012236. [Google Scholar] [CrossRef]
  38. Francesconi, L.P.; Victorino, A.T.; Salah, I.A.; Cordova, V.H.S.; Dias da Rosa, E.; Oliveira, L.; Jacobus, R.V.M.; Belmonte-de-Abreu, P.S.; Ceresér, K.M. Proinflammatory and Anti-Inflammatory Biomarkers in Schizophrenia and Influence of Simvastatin on the Interleukin-6. Int. Clin. Psychopharmacol. 2019, 34, 84–88. [Google Scholar] [CrossRef]
  39. Climent, E.; Benaiges, D.; Pedro-Botet, J. Hydrophilic or Lipophilic Statins? Front. Cardiovasc. Med. 2021, 8, 687585. [Google Scholar] [CrossRef]
  40. Nomura, I.; Kishi, T.; Ikuta, T.; Iwata, N. Statin Add-on Therapy in the Antipsychotic Treatment of Schizophrenia: A Meta-Analysis. Psychiatry Res. 2018, 260, 41–47. [Google Scholar] [CrossRef] [PubMed]
  41. 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]
  42. Tajik-Esmaeeli, S.; Moazen-Zadeh, E.; Abbasi, N.; Shariat, S.V.; Rezaei, F.; Salehi, B.; Akhondzadeh, S. Simvastatin adjunct therapy for negative symptoms of schizophrenia: A randomized double-blind placebo-controlled trial. Int. Clin. Psychopharmacol. 2017, 32, 87–94. [Google Scholar] [CrossRef]
  43. Sommer, I.E.; Gangadin, S.S.; de Witte, L.D.; Koops, S.; van Baal, C.; Bahn, S.; Drexhage, H.; van Haren, N.E.M.; Veling, W.; Bruggeman, R.; et al. Simvastatin Augmentation for Patients with Early-Phase Schizophrenia-Spectrum Disorders: A Double-Blind, Randomized Placebo-Controlled Trial. Schizophr. Bull. 2021, 47, 1108–1115. [Google Scholar] [CrossRef]
  44. Aichholzer, M.; Gangadin, S.S.; Sommer, I.E.C.; Wijkhuis, A.; de Witte, L.D.; Kahn, R.S.; Bahn, S.; Drexhage, H.A.; Schiweck, C. Inflammatory Monocyte Gene Signature Predicts Beneficial within Group Effect of Simvastatin in Patients with Schizophrenia Spectrum Disorders in a Secondary Analysis of a Randomized Controlled Trial. Brain Behav. Immun. Health 2022, 26, 100551. [Google Scholar] [CrossRef] [PubMed]
  45. Postolache, T.T.; Medoff, D.R.; Brown, C.H.; Fang, L.J.; Upadhyaya, S.K.; Lowry, C.A.; Miller, M.; Kreyenbuhl, J.A. Lipophilic vs. Hydrophilic Statins and Psychiatric Hospitalizations and Emergency Room Visits in US Veterans with Schizophrenia and Bipolar Disorder. Pteridines 2021, 32, 48–69. [Google Scholar] [CrossRef]
  46. Kim, S.W.; Kang, H.J.; Jhon, M.; Kim, J.W.; Lee, J.Y.; Walker, A.J.; Agustini, B.; Kim, J.M.; Berk, M. Statins and Inflammation: New Therapeutic Opportunities in Psychiatry. Front. Psychiatry 2019, 10, 103. [Google Scholar] [CrossRef] [PubMed]
  47. Koushki, K.; Shahbaz, S.K.; Mashayekhi, K.; Sadeghi, M.; Zayeri, Z.D.; Taba, M.Y.; Banach, M.; Al-Rasadi, K.; Johnston, T.P.; Sahebkar, A. Anti-inflammatory Action of Statins in Cardiovascular Disease: The Role of Inflammasome and Toll-Like Receptor Pathways. Clin. Rev. Allergy Immunol. 2021, 60, 175–199. [Google Scholar] [CrossRef] [PubMed]
  48. García-Bueno, B.; Gassó, P.; MacDowell, K.S.; Callado, L.F.; Mas, S.; Bernardo, M.; Lafuente, A.; Meana, J.J.; Leza, J.C. Evidence of activation of the Toll-like receptor-4 proinflammatory pathway in patients with schizophrenia. J. Psychiatry Neurosci. 2016, 41, E46–E55. [Google Scholar] [CrossRef]
  49. Berger, J.; Moller, D.E. The mechanisms of action of PPARs. Annu. Rev. Med. 2002, 53, 409–435. [Google Scholar] [CrossRef]
  50. Korbecki, J.; Bobiński, R.; Dutka, M. Self-Regulation of the Inflammatory Response by Peroxisome Proliferator-Activated Receptors. Inflamm. Res. 2019, 68, 443–458. [Google Scholar] [CrossRef]
  51. Christofides, A.; Konstantinidou, E.; Jani, C.; Boussiotis, V.A. The role of peroxisome proliferator-activated receptors (PPAR) in immune responses. Metabolism 2020, 114, 154338. [Google Scholar] [CrossRef]
  52. Warden, A.; Truitt, J.; Merriman, M.; Ponomareva, O.; Jameson, K.; Ferguson, L.B.; Mayfield, R.D.; Harris, R.A. Localization of PPAR isotypes in the adult mouse and human brain. Sci. Rep. 2016, 6, 7618. [Google Scholar] [CrossRef]
  53. Han, L.; Shen, W.-J.; Bittner, S.; Kraemer, F.B.; Azhar, S. PPARs: Regulators of Metabolism and as Therapeutic Targets in Cardiovascular Disease. Part I: PPAR-α. Future Cardiol. 2017, 13, 259–278. [Google Scholar] [CrossRef]
  54. Dana, N.; Vaseghi, G.; Haghjooy Javanmard, S. Crosstalk between Peroxisome Proliferator-Activated Receptors and Toll-Like Receptors: A Systematic Review. Adv. Pharm. Bull. 2019, 9, 12–21. [Google Scholar] [CrossRef]
  55. Kariharan, T.; Nanayakkara, G.; Parameshwaran, K.; Bagasrawala, I.; Ahuja, M.; Abdel-Rahman, E.; Amin, A.T.; Dhanasekaran, M.; Suppiramaniam, V.; Amin, R.H. Central activation of PPAR-gamma ameliorates diabetes induced cognitive dysfunction and improves BDNF expression. Neurobiol. Aging 2015, 36, 1451–1461. [Google Scholar] [CrossRef]
  56. Iranpour, N.; Zandifar, A.; Farokhnia, M.; Goguol, A.; Yekehtaz, H.; Khodaie-Ardakani, M.-R.; Salehi, B.; Esalatmanesh, S.; Zeionoddini, A.; Mohammadinejad, P.; et al. The Effects of Pioglitazone Adjuvant Therapy on Negative Symptoms of Patients with Chronic Schizophrenia: A Double-Blind and Placebo-Controlled Trial. Hum. Psychopharmacol. Clin. Exp. 2016, 31, 103–112. [Google Scholar] [CrossRef] [PubMed]
  57. Sagheddu, C.; Melis, M.; Muntoni, A.L.; Pistis, M. Repurposing Peroxisome Proliferator-Activated Receptor Agonists in Neurological and Psychiatric Disorders. Pharmaceuticals 2021, 14, 1025. [Google Scholar] [CrossRef]
  58. Oh, S.J.; Fan, X. The Possible Role of the Angiotensin System in the Pathophysiology of Schizophrenia: Implications for Pharmacotherapy. CNS Drugs 2019, 33, 539–547. [Google Scholar] [CrossRef]
  59. Teixeira, A.L.; de Miranda, A.S.; Macedo, D.S.; Rocha, N.P. Targeting the Renin-Angiotensin System (RAS) for Neuropsychiatric Disorders. Curr. Neuropharmacol. 2022. online ahead of print. [Google Scholar] [CrossRef] [PubMed]
  60. Santiago, T.C.; Parra, L.; Nani, J.V.; Fidalgo, T.M.; Bradshaw, N.J.; Hayashi, M.A.F. Angiotensin-converting enzymes as druggable features of psychiatric and neurodegenerative disorders. J. Neurochem. 2023. early view. [Google Scholar] [CrossRef] [PubMed]
  61. Mohite, S.; de Campos-Carli, S.M.; Rocha, N.P.; Sharma, S.; Miranda, A.S.; Barbosa, I.G.; Salgado, J.V.; Simoes-E-Silva, A.C.; Teixeira, A.L. Lower circulating levels of angiotensin-converting enzyme (ACE) in patients with schizophrenia. Schizophr. Res. 2018, 202, 50–54. [Google Scholar] [CrossRef]
  62. Saavedra, J.M. Angiotensin Receptor Blockers Are Not Just for Hypertension Anymore. Physiology 2021, 36, 160–173. [Google Scholar] [CrossRef] [PubMed]
  63. Biancardi, V.C.; Son, S.J.; Ahmadi, S.; Filosa, J.A.; Stern, J.E. Circulating angiotensin II gains access to the hypothalamus and brain stem during hypertension via breakdown of the blood-brain barrier. Hypertension 2014, 63, 572–579. [Google Scholar] [CrossRef] [PubMed]
  64. Pang, T.; Wang, J.; Benicky, J.; Sánchez-Lemus, E.; Saavedra, J.M. Telmisartan directly ameliorates the neuronal inflammatory response to IL-1β partly through the JNK/c-Jun and NADPH oxidase pathways. J. Neuroinflammation 2012, 9, 102. [Google Scholar] [CrossRef] [PubMed]
  65. Fan, X.; Song, X.; Zhao, M.; Jarskog, L.F.; Natarajan, R.; Shukair, N.; Freudenreich, O.; Henderson, D.C.; Goff, D.C. The Effect of Adjunctive Telmisartan Treatment on Psychopathology and Cognition in Patients with Schizophrenia. Acta Psychiatr. Scand. 2017, 136, 465–472. [Google Scholar] [CrossRef]
  66. Zakrocka, I.; Targowska-Duda, K.M.; Wnorowski, A.; Kocki, T.; Jóźwiak, K.; Turski, W.A. Angiotensin II Type 1 Receptor Blockers Inhibit KAT II Activity in the Brain—Its Possible Clinical Applications. Neurotox. Res. 2017, 32, 639–648. [Google Scholar] [CrossRef]
  67. Vasconcelos, G.S.; dos Santos Júnior, M.A.; Monte, A.S.; da Silva, F.E.R.; de Carvalho Lima, C.N.; Moreira Lima Neto, A.B.; da Silva Medeiros, I.; Teixeira, A.L.; de Lucena, D.F.; Vasconcelos, S.M.M.; et al. Low-Dose Candesartan Prevents Schizophrenia-like Behavioral Alterations in a Neurodevelopmental Two-Hit Model of Schizophrenia. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2021, 111, 110348. [Google Scholar] [CrossRef]
  68. Zakrocka, I.; Turski, W.A.; Kocki, T. Angiotensin-converting enzyme inhibitors modulate kynurenic acid production in rat brain cortex in vitro. Eur. J. Pharmacol. 2016, 789, 308–312. [Google Scholar] [CrossRef]
  69. Vane, J.R.; Bakhle, Y.S.; Botting, R.M. Cyclooxygenases 1 and 2. Annu. Rev. Pharmacol. Toxicol. 1998, 138, 97–120. [Google Scholar] [CrossRef]
  70. 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]
  71. Attari, A.; Mojdeh, A.; Khalifeh Soltani, F.A.S.; Najarzadegan, M.R. Aspirin inclusion in antipsychotic treatment on severity of symptoms in schizophrenia: A randomized clinical trial. Iran. J. Psychiatry Behav. Sci. 2017, 11, e5848. [Google Scholar]
  72. Berk, M.; Dean, O.; Drexhage, H.; McNeil, J.J.; Moylan, S.; O’Neil, A.; Davey, C.G.; Sanna, L.; Maes, M. Aspirin: A review of its neurobiological properties and therapeutic potential for mental illness. BMC Med. 2013, 11, 74. [Google Scholar] [CrossRef]
  73. Sommer, I.E.; de Witte, L.; Begemann, M.; Kahn, R.S. Nonsteroidal anti-inflammatory drugs in schizophrenia: Ready for practice or a good start? A meta-analysis. J. Clin. Psychiatry 2012, 73, 414–419. [Google Scholar] [CrossRef] [PubMed]
  74. Cruz, J.V.; Rosa, J.M.C.; Kimani, N.M.; Giuliatti, S.; dos Santos, C.B.R. The Role of Celecoxib as a Potential Inhibitor in the Treatment of Inflammatory Diseases—A Review. Curr. Med. Chem. 2022, 29, 3028–3049. [Google Scholar] [CrossRef]
  75. Müller, N.; Strassnig, M.; Schwarz, M.J.; Ulmschneider, M.; Riedel, M. COX-2 Inhibitors as Adjunctive Therapy in Schizophrenia. Expert Opin. Investig. Drugs 2004, 13, 1033–1044. [Google Scholar] [CrossRef] [PubMed]
  76. Yokota, O.; Terada, S.; Ishihara, T.; Nakashima, H.; Kugo, A.; Ujike, H.; Tsuchiya, K.; Ikeda, K.; Saito, Y.; Murayama, S.; et al. Neuronal Expression of Cyclooxygenase-2, a Pro-Inflammatory Protein, in the Hippocampus of Patients with Schizophrenia. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2004, 28, 715–721. [Google Scholar] [CrossRef] [PubMed]
  77. Marini, S.; De Berardis, D.; Vellante, F.; Santacroce, R.; Orsolini, L.; Valchera, A.; Girinelli, G.; Carano, A.; Fornaro, M.; Gambi, F.; et al. Celecoxib Adjunctive Treatment to Antipsychotics in Schizophrenia: A Review of Randomized Clinical Add-on Trials. Mediat. Inflamm. 2016, 2016, 3476240. [Google Scholar] [CrossRef]
  78. Krebs, M.; Leopold, K.; Hinzpeter, A.; Schaefer, M. Neuroprotective Agents in Schizophrenia and Affective Disorders. Expert Opin. Pharmacother. 2006, 7, 837–848. [Google Scholar] [CrossRef]
  79. 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]
  80. Akhondzadeh, S.; Tabatabaee, M.; Amini, H.; Ahmadiabhari, S.; Abbasi, S.; 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]
  81. Müller, N.; Krause, D.; Dehning, S.; Musil, R.; Schennach-Wolff, R.; Obermeier, M.; Möller, H.-J.; Klauss, V.; Schwarz, M.J.; Riedel, M. Celecoxib Treatment in an Early Stage of Schizophrenia: Results of a Randomized, Double-Blind, Placebo-Controlled Trial of Celecoxib Augmentation of Amisulpride Treatment. Schizophr. Res. 2010, 121, 118–124. [Google Scholar] [CrossRef] [PubMed]
  82. Müller, N.; Riedel, M.; Schwarz, M.J.; Engel, R.R. Clinical Effects of COX-2 Inhibitors on Cognition in Schizophrenia. Eur. Arch. Psychiatry Clin. Neurosci. 2004, 255, 149–151. [Google Scholar] [CrossRef] [PubMed]
  83. Bresee, C.J.; Delrahim, K.; Maddux, R.E.; Dolnak, D.; Ahmadpour, O.; Rapaport, M.H. The Effects of Celecoxib Augmentation on Cytokine Levels in Schizophrenia. Int. J. Neuropsychopharmacol. 2005, 9, 343. [Google Scholar] [CrossRef] [PubMed]
  84. Müller, N. COX-2 Inhibitors, Aspirin, and Other Potential Anti-Inflammatory Treatments for Psychiatric Disorders. Front. Psychiatry 2019, 10, 375. [Google Scholar] [CrossRef]
  85. Zheng, W.; Cai, D.-B.; Yang, X.-H.; Ungvari, G.S.; Ng, C.H.; Müller, N.; Ning, Y.-P.; Xiang, Y.-T. Adjunctive Celecoxib for Schizophrenia: A Meta-Analysis of Randomized, Double-Blind, Placebo-Controlled Trials. J. Psychiatr. Res. 2017, 92, 139–146. [Google Scholar] [CrossRef]
  86. Giovanoli, S.; Engler, H.; Engler, A.; Richetto, J.; Feldon, J.; Riva, M.A.; Schedlowski, M.; Meyer, U. Preventive Effects of Minocycline in a Neurodevelopmental Two-Hit Model with Relevance to Schizophrenia. Transl. Psychiatry 2016, 6, e772. [Google Scholar] [CrossRef]
  87. Oya, K.; Kishi, T.; Iwata, N. Efficacy and Tolerability of Minocycline Augmentation Therapy in Schizophrenia: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Hum. Psychopharmacol. Clin. Exp. 2014, 29, 483–491. [Google Scholar] [CrossRef]
  88. Solmi, M.; Veronese, N.; Thapa, N.; Facchini, S.; Stubbs, B.; Fornaro, M.; Carvalho, A.F.; Correll, C.U. Systematic Review and Meta-Analysis of the Efficacy and Safety of Minocycline in Schizophrenia. CNS Spectr. 2017, 22, 415–426. [Google Scholar] [CrossRef]
  89. Xiang, Y.-Q.; Zheng, W.; Wang, S.-B.; Yang, X.-H.; Cai, D.-B.; Ng, C.H.; Ungvari, G.S.; Kelly, D.L.; Xu, W.-Y.; Xiang, Y.-T. Adjunctive Minocycline for Schizophrenia: A Meta-Analysis of Randomized Controlled Trials. Eur. Neuropsychopharmacol. 2017, 27, 8–18. [Google Scholar] [CrossRef]
  90. Zheng, W.; Zhu, X.-M.; Zhang, Q.-E.; Cheng, G.; Cai, D.-B.; He, J.; Ng, C.H.; Ungvari, G.S.; Peng, X.-J.; Ning, Y.-P.; et al. Adjunctive Minocycline for Major Mental Disorders: A Systematic Review. J. Psychopharmacol. 2019, 33, 1215–1226. [Google Scholar] [CrossRef]
  91. Wehring, H.J.; Elsobky, T.; McEvoy, J.P.; Vyas, G.; Richardson, C.M.; McMahon, R.P.; DiPaula, B.A.; Liu, F.; Sullivan, K.; Buchanan, R.W.; et al. Adjunctive Minocycline in Clozapine-Treated Patients with Schizophrenia: Analyzing the Effects of Minocycline on Clozapine Plasma Levels. Psychiatr. Q. 2017, 89, 73–80. [Google Scholar] [CrossRef]
  92. Kelly, D.L.; Sullivan, K.M.; McEvoy, J.P.; McMahon, R.P.; Wehring, H.J.; Gold, J.M.; Liu, F.; Warfel, D.; Vyas, G.; Richardson, C.M.; et al. Adjunctive Minocycline in Clozapine-Treated Schizophrenia Patients with Persistent Symptoms. J. Clin. Psychopharmacol. 2015, 35, 374. [Google Scholar] [CrossRef] [PubMed]
  93. Ghanizadeh, A.; Dehbozorgi, S.; OmraniSigaroodi, M.; Rezaei, Z. Minocycline as Add-on Treatment Decreases the Negative Symptoms of Schizophrenia; a Randomized Placebo-Controlled Clinical Trial. Recent Pat. Inflamm. Allergy Drug Discov. 2014, 8, 211–215. [Google Scholar] [CrossRef] [PubMed]
  94. Liu, F.; Guo, X.; Wu, R.; Ou, J.; Zheng, Y.; Zhang, B.; Xie, L.; Zhang, L.; Yang, L.; Yang, S.; et al. Minocycline Supplementation for Treatment of Negative Symptoms in Early-Phase Schizophrenia: A Double Blind, Randomized, Controlled Trial. Schizophr. Res. 2014, 153, 169–176. [Google Scholar] [CrossRef] [PubMed]
  95. Chen, X.; Xiong, Z.; Li, Z.; Yang, Y.; Zheng, Z.; Li, Y.; Xie, Y.; Li, Z. Minocycline as Adjunct Therapy for a Male Patient with Deficit Schizophrenia. Neuropsychiatr. Dis. Treat. 2018, 14, 2697–2701. [Google Scholar] [CrossRef] [PubMed]
  96. Zhang, L.; Zheng, H.; Wu, R.; Zhu, F.; Kosten, T.R.; Zhang, X.-Y.; Zhao, J. Minocycline Adjunctive Treatment to Risperidone for Negative Symptoms in Schizophrenia: Association with Pro-Inflammatory Cytokine Levels. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2018, 85, 69–76. [Google Scholar] [CrossRef]
  97. Zhang, L.; Zheng, H.; Wu, R.; Kosten, T.R.; Zhang, X.-Y.; Zhao, J. The Effect of Minocycline on Amelioration of Cognitive Deficits and Pro-Inflammatory Cytokines Levels in Patients with Schizophrenia. Schizophr. Res. 2019, 212, 92–98. [Google Scholar] [CrossRef]
  98. Tendilla-Beltrán, H.; Flores, G. Due to Their Anti-Inflammatory, Antioxidant and Neurotrophic Properties, Second-Generation Antipsychotics Are Suitable in Patients with Schizophrenia and COVID-19. Gen. Hosp. Psychiatry 2021, 71, 137–139. [Google Scholar] [CrossRef]
  99. Kato, T.A.; Hyodo, F.; Yamato, M.; Utsumi, H.; Kanba, S. Redox and Microglia in the Pathophysiology of Schizophrenia. Yakugaku Zasshi 2015, 135, 739–743. [Google Scholar] [CrossRef]
  100. Kobayashi, K.; Imagama, S.; Ohgomori, T.; Hirano, K.; Uchimura, K.; Sakamoto, K.; Hirakawa, A.; Takeuchi, H.; Suzumura, A.; Ishiguro, N.; et al. Minocycline Selectively Inhibits M1 Polarization of Microglia. Cell Death Dis. 2013, 4, e525. [Google Scholar] [CrossRef]
  101. Yuan, Z.; Chen, X.; Yang, W.; Lou, B.; Ye, N.; Liu, Y. The anti-inflammatory effect of minocycline on endotoxin-induced uveitis and retinal inflammation in rats. Mol. Vis. 2019, 25, 359–372. [Google Scholar]
  102. Blecharz-Lang, K.G.; Patsouris, V.; Nieminen-Kelhä, M.; Seiffert, S.; Schneider, U.C.; Vajkoczy, P. Minocycline Attenuates Microglia/Macrophage Phagocytic Activity and Inhibits SAH-Induced Neuronal Cell Death and Inflammation. Neurocritical Care 2022, 37, 410–423. [Google Scholar] [CrossRef]
  103. Inta, D.; Guzman, R.; Gass, P. Microglia Activation and Adult Neurogenesis in the Hippocampus: New Clues about the Antidepressant Effect of Minocycline. Brain Behav. Immun. 2021, 94, 27–28. [Google Scholar] [CrossRef]
  104. Jones, M.C.; Koh, J.M.; Cheong, K.H. Synaptic Pruning in Schizophrenia: Does Minocycline Modulate Psychosocial Brain Development? BioEssays 2020, 42, 2000046. [Google Scholar] [CrossRef] [PubMed]
  105. Balu, D.T. The NMDA Receptor and Schizophrenia. Neuropsychopharmacol. Tribut. Joseph T. Coyle 2016, 76, 351–382. [Google Scholar] [CrossRef]
  106. Chaves, C.; Marque, C.R.; Trzesniak, C.; Machado de Sousa, J.P.; Zuardi, A.W.; Crippa, J.a.S.; Dursun, S.M.; Hallak, J.E. Glutamate-N-Methyl-D-Aspartate Receptor Modulation and Minocycline for the Treatment of Patients with Schizophrenia: An Update. Braz. J. Med. Biol. Res.—Rev. Bras. Pesqui. Med. E Biol. 2009, 42, 1002–1014. [Google Scholar] [CrossRef] [PubMed]
  107. Deakin, B.; Suckling, J.; Barnes, T.R.E.; Byrne, K.; Chaudhry, I.B.; Dazzan, P.; Drake, R.J.; Giordano, A.; Husain, N.; Jones, P.B.; et al. The Benefit of Minocycline on Negative Symptoms of Schizophrenia in Patients with Recent-Onset Psychosis (BeneMin): A Randomised, Double-Blind, Placebo-Controlled Trial. Lancet Psychiatry 2018, 5, 885–894. [Google Scholar] [CrossRef]
  108. Weiser, M.; Levi, L.; Burshtein, S.; Chiriță, R.; Cirjaliu, D.; Gonen, I.; Yolken, R.; Davidson, M.; Zamora, D.; Davis, J.M. The Effect of Minocycline on Symptoms in Schizophrenia: Results from a Randomized Controlled Trial. Schizophr. Res. 2019, 206, 325–332. [Google Scholar] [CrossRef]
  109. Krynicki, C.R.; Dazzan, P.; Pariante, C.M.; Barnes, N.M.; Vincent, R.C.; Roberts, A.; Giordano, A.; Watson, A.; Suckling, J.; Barnes, T.R.E.; et al. Deconstructing Depression and Negative Symptoms of Schizophrenia; Differential and Longitudinal Immune Correlates, and Response to Minocycline Treatment. Brain Behav. Immun. 2021, 91, 498–504. [Google Scholar] [CrossRef] [PubMed]
  110. Kishimoto, T.; Horigome, T.; Takamiya, A. Minocycline as a Treatment for Schizophrenia: Is the Discussion Truly Finished? Lancet Psychiatry 2018, 5, 856–857. [Google Scholar] [CrossRef]
  111. Nitta, M.; Kishimoto, T.; Müller, N.; Weiser, M.; Davidson, M.; Kane, J.M.; Correll, C.U. Adjunctive Use of Nonsteroidal Anti-Inflammatory Drugs for Schizophrenia: A Meta-Analytic Investigation of Randomized Controlled Trials. Schizophr. Bull. 2013, 39, 1230–1241. [Google Scholar] [CrossRef]
  112. Garner, B.; Phillips, L.J.; Bendall, S.; Hetrick, S.E. Antiglucocorticoid and related treatments for psychosis. Cochrane Database Syst. Rev. 2016, 2016, CD006995. [Google Scholar] [CrossRef] [PubMed]
  113. Yu, X.; Qi, X.; Wei, L.; Zhao, L.; Deng, W.; Guo, W.; Wang, Q.; Ma, X.; Hu, X.; Ni, P.; et al. Fingolimod Ameliorates Schizophrenia-like Cognitive Impairments Induced by Phencyclidine in Male Rats. Br. J. Pharmacol. 2022, 180, 161–173. [Google Scholar] [CrossRef] [PubMed]
  114. Francis, M.M.; Hummer, T.A.; Liffick, E.; Vohs, J.L.; Mehdiyoun, N.F.; Visco, A.C.; Yang, Z.; Kovacs, R.J.; Zhang, Y.; Breier, A. Effects of Fingolimod, a Sphingosine-1-Phosphate (S1P) Receptor Agonist, on White Matter Microstructure, Cognition and Symptoms in Schizophrenia. Brain Imaging Behav. 2020, 15, 1802–1814. [Google Scholar] [CrossRef] [PubMed]
  115. Fessel, J. Abnormal Oligodendrocyte Function in Schizophrenia Explains the Long Latent Interval in Some Patients. Transl. Psychiatry 2022, 12, 120. [Google Scholar] [CrossRef] [PubMed]
  116. Karbalaee, M.; Jameie, M.; Amanollahi, M.; TaghaviZanjani, F.; Parsaei, M.; Basti, F.A.; Mokhtari, S.; Moradi, K.; Ardakani, M.-R.K.; Akhondzadeh, S. Efficacy and Safety of Adjunctive Therapy with Fingolimod in Patients with Schizophrenia: A Randomized, Double-Blind, Placebo-Controlled Clinical Trial. Schizophr. Res. 2023, 254, 92–98. [Google Scholar] [CrossRef]
  117. Chaudhry, I.B.; Husain, M.O.; Khoso, A.B.; Husain, M.I.; Buch, M.H.; Kiran, T.; Fu, B.; Bassett, P.; Qurashi, I.; ur Rahman, R.; et al. A Randomised Clinical Trial of Methotrexate Points to Possible Efficacy and Adaptive Immune Dysfunction in Psychosis. Transl. Psychiatry 2020, 10, 415. [Google Scholar] [CrossRef]
  118. Motamed, M.; Karimi, H.; Sanjari Moghaddam, H.; Taherzadeh Boroujeni, S.; Sanatian, Z.; Hasanzadeh, A.; Khodaei Ardakani, M.-R.; Akhondzadeh, S. Risperidone Combination Therapy with Adalimumab for Treatment of Chronic Schizophrenia. Int. Clin. Psychopharmacol. 2022, 37, 92–101. [Google Scholar] [CrossRef]
  119. Lin, C.; Chen, K.; Yu, J.; Feng, W.; Fu, W.; Yang, F.; Zhang, X.; Chen, D. Relationship between TNF-α Levels and Psychiatric Symptoms in First-Episode Drug-Naïve Patients with Schizophrenia before and after Risperidone Treatment and in Chronic Patients. BMC Psychiatry 2021, 21, 561. [Google Scholar] [CrossRef]
  120. Bejerot, S.; Sigra Stein, S.; Welin, E.; Eklund, D.; Hylén, U.; Humble, M.B. Rituximab as an Adjunctive Treatment for Schizophrenia Spectrum Disorder or Obsessive-Compulsive Disorder: Two Open-Label Pilot Studies on Treatment-Resistant Patients. J. Psychiatr. Res. 2023, 158, 319–329. [Google Scholar] [CrossRef]
  121. 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, 13353. [Google Scholar] [CrossRef] [PubMed]
  122. 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. Neuropsychopharmacology 2017, 43, 1317–1323. [Google Scholar] [CrossRef]
  123. Hansen, N.; Malchow, B. Monoclonal Antibody Therapy in Autoantibody-Associated Psychotic Disorders and Schizophrenia: Narrative Review of Past and Current Clinical Trials. Psychiatr. Danub. 2023, 35, 8–15. [Google Scholar] [CrossRef] [PubMed]
  124. Pandurangi, A.K.; Buckley, P.F. Inflammation, Antipsychotic Drugs, and Evidence for Effectiveness of Anti-inflammatory Agents in Schizophrenia. Curr. Top. Behav. Neurosci. 2020, 44, 227–244. [Google Scholar] [CrossRef] [PubMed]
  125. 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. [Google Scholar] [CrossRef]
Figure 1. Prisma Flow Diagram.
Figure 1. Prisma Flow Diagram.
Brainsci 13 00957 g001
Table 1. Risk factor of neuroinflammation in schizophrenia.
Table 1. Risk factor of neuroinflammation in schizophrenia.
Risk Factor of Neuroinflammation in Schizophrenia (Hong, 2020 [11])
Infectious agents (i.e., toxoplasma gondii)
Maternal immune activation caused by prenatal infections
Pro-inflammatory genes (FOS, IL1B, CXCL8)
Stress inflammation correlated (psychosocial stress)
Table 2. Possible indications to use NRNDs in schizophrenia.
Table 2. Possible indications to use NRNDs in schizophrenia.
Early state of psychosis
Resistant schizophrenia
Schizophrenia with neuroinflammation (CRP, cytokines, L/N ratio)
Schizophrenia with prevalent negative and cognitive symptoms
PET neuroimaging signs of microgliosis
Table 3. Utilization of NRNDs in different cluster symptoms of the schizophrenia and in relation to the comorbidity.
Table 3. Utilization of NRNDs in different cluster symptoms of the schizophrenia and in relation to the comorbidity.
Positive SymptomsNegative SymptomsCognitive SymptomsFEP/UHRGFRS DiabetesCh/Tri.AHRD
PUFAs
Statins
PPARs agonists
Minocycline
Celecoxib
MAb
Fingolimod
Prednisolone
AT1 antagonists/ACE inhibitors
MAb: monoclonal antibodies; FEP/UHR: first episode of psychosis/ultra-high risk; GF: global functioning; RS: resistant schizophrenia; Ch/Tri: cholesterol and triglycerides levels; AH: arterial hypertension; RD: rheumatologic diseases. √: drug action on specific symptoms of diseases.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Messina, A.; Concerto, C.; Rodolico, A.; Petralia, A.; Caraci, F.; Signorelli, M.S. Is It Time for a Paradigm Shift in the Treatment of Schizophrenia? The Use of Inflammation-Reducing and Neuroprotective Drugs—A Review. Brain Sci. 2023, 13, 957. https://doi.org/10.3390/brainsci13060957

AMA Style

Messina A, Concerto C, Rodolico A, Petralia A, Caraci F, Signorelli MS. Is It Time for a Paradigm Shift in the Treatment of Schizophrenia? The Use of Inflammation-Reducing and Neuroprotective Drugs—A Review. Brain Sciences. 2023; 13(6):957. https://doi.org/10.3390/brainsci13060957

Chicago/Turabian Style

Messina, Antonino, Carmen Concerto, Alessandro Rodolico, Antonino Petralia, Filippo Caraci, and Maria Salvina Signorelli. 2023. "Is It Time for a Paradigm Shift in the Treatment of Schizophrenia? The Use of Inflammation-Reducing and Neuroprotective Drugs—A Review" Brain Sciences 13, no. 6: 957. https://doi.org/10.3390/brainsci13060957

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