Next Article in Journal
Evidence Supporting a Role of Alternative Splicing Participates in Melon (Cucumis melo L.) Fruit Ripening
Previous Article in Journal
Substance P’s Impact on Chronic Pain and Psychiatric Conditions—A Narrative Review
Previous Article in Special Issue
Molecular Mechanism Biomarkers Predict Diagnosis in Schizophrenia and Schizoaffective Psychosis, with Implications for Treatment
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Novel Insights into Psychosis and Antipsychotic Interventions: From Managing Symptoms to Improving Outcomes

by
Adonis Sfera
1,2,3,*,
Hassan Imran
1,2,3,
Dan O. Sfera
1,2,3,
Jacob J. Anton
4,
Zisis Kozlakidis
5 and
Sabine Hazan
6
1
Patton State Hospital, 3102 Highland Ave., Patton, CA 92369, USA
2
University of California Riverside, Riverside 900 University Ave., Riverside, CA 92521, USA
3
Loma Linda University, 11139 Anderson St., Loma Linda, CA 92350, USA
4
California Baptist University, Riverside, CA 92521, USA
5
International Agency for Research on Cancer, 69372 Lyon, France
6
ProgenaBiome, Ventura, CA 93003, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(11), 5904; https://doi.org/10.3390/ijms25115904
Submission received: 26 March 2024 / Revised: 20 May 2024 / Accepted: 24 May 2024 / Published: 28 May 2024

Abstract

:
For the past 70 years, the dopamine hypothesis has been the key working model in schizophrenia. This has contributed to the development of numerous inhibitors of dopaminergic signaling and antipsychotic drugs, which led to rapid symptom resolution but only marginal outcome improvement. Over the past decades, there has been limited research on the quantifiable pathological changes in schizophrenia, including premature cellular/neuronal senescence, brain volume loss, the attenuation of gamma oscillations in electroencephalograms, and the oxidation of lipids in the plasma and mitochondrial membranes. We surmise that the aberrant activation of the aryl hydrocarbon receptor by toxins derived from gut microbes or the environment drives premature cellular and neuronal senescence, a hallmark of schizophrenia. Early brain aging promotes secondary changes, including the impairment and loss of mitochondria, gray matter depletion, decreased gamma oscillations, and a compensatory metabolic shift to lactate and lactylation. The aim of this narrative review is twofold: (1) to summarize what is known about premature cellular/neuronal senescence in schizophrenia or schizophrenia-like disorders, and (2) to discuss novel strategies for improving long-term outcomes in severe mental illness with natural senotherapeutics, membrane lipid replacement, mitochondrial transplantation, microbial phenazines, novel antioxidant phenothiazines, inhibitors of glycogen synthase kinase-3 beta, and aryl hydrocarbon receptor antagonists.

1. Introduction

The discovery of chlorpromazine in the 1950s revolutionized psychiatry and contributed to the deinstitutionalization of people with severe mental illness. Subsequently, the homelessness and incarceration of individuals with schizophrenia (SCZ) and schizophrenia-like disorders (SLDs) dramatically increased, suggesting that symptomatic relief in these conditions rarely translates into sustained recovery [1,2].
Although most patients treated with antipsychotic drugs attain partial remission or amelioration of symptoms, few return to premorbid levels of functioning, measured by stable employment, attending school, raising a family, and being independent in all activities of daily living (ADLs) [3]. For this reason, large public institutions for the treatment of mental illness, such as state hospitals, are still in existence, while sanatoria for tuberculosis or leprosy were closed almost a century ago.
The early antipsychotic drugs were derived from methylene blue (MB), a phenothiazine synthesized in 1876 in Germany. The interest in this agent dramatically surged after the realization that it exerts antidepressant actions by inhibiting monoamine oxidase A (MAO-A), a surreptitious discovery that commenced the era of modern psychopharmacology [4]. Upon chlorpromazine’s approval in the US, over 40 dopamine-blocking antipsychotic drugs were developed, aiming at restoring premorbid functioning by targeting major symptoms.
Although antipsychotic drugs are extremely efficacious for acute psychosis as the symptoms are often cleared within hours or days, sustained recovery is achieved by only 13.5% of patients after an initial psychotic episode [5]. Moreover, looking at the entire 20th century, in the early decades, long-term recovery was at 20%, not differing from the end of the century when antipsychotic drugs were being widely utilized [6]. At present, 33% of patients with SCZ relapse within 12 months after an initial psychotic episode, 26% remain homeless at the 2-year follow-up, and 5 years after the first psychotic outbreak, only 10% are employed [7,8,9]. Together, these data indicate that the blockade of dopamine (DA) receptors seldom improves the outcomes of SCZ or SLDs. Moreover, some antipsychotic drugs, including clozapine and aripiprazole, upregulate DA, suggesting that this neurotransmitter may play an indirect role in the etiopathogenesis of severe mental illness [10,11].
The aims of this narrative review are as follows:
  • To summarize what is known about the role of premature cellular/neuronal senescence in the pathogenesis of SCZ and SLDs.
  • To discuss potential strategies for improving sustained recovery in SCZ and SLDs via natural senotherapeutics, microbial phenazines, aryl hydrocarbon receptor (AhR) antagonists, membrane lipid replacement (MLR), and mitochondrial transplantation.

2. Premature Cellular Senescence in Schizophrenia

Patients with SCZ and SLDs live, on average, 15–20 years less than the general population, exhibit shortened telomeres, and develop age-related diseases earlier in life, suggesting that premature cellular senescence plays an important role in this pathology [12,13,14,15]. Indeed, many researchers and clinicians refer to SCZ as a “segmental progeria” to highlight the accelerated aging of tissues and organs, including the brain, in this disorder [12].
Cellular senescence is a program of permanent cell cycle arrest with an active metabolism, shortened telomeres, accumulation of macromolecular aggregates, increased levels of senescence-associated β-galactosidase (SA-β-gal), and a toxic secretome, known as the senescence-associated secretory phenotype (SASP), which can spread senescence to neighboring healthy cells [16]. It is believed that cellular senescence defends against tumorigenesis by preventing oncogene-driven malignant transformation. However, the accumulation of aged cells and the subsequent inflammation may paradoxically promote cancer and disrupt biological barriers, facilitating the dissemination of metastases [16,17]. Inflammation and senescent cells increase the permeability of the gut barrier, facilitating the translocation of the gastrointestinal (GI) tract bacteria (or their molecules) into the systemic circulation, a phenomenon encountered in neuropsychiatric and neurodegenerative disorders [18]. In another example, microbiota-derived gallic acid converts p53, the key anticancer protein, into an oncogene that drives tumorigenesis [19,20,21]. As p53, an SCZ risk gene, also promotes cellular senescence, it likely connects microbial translocation to severe mental illness [22,23]. Indeed, bacterial molecules were demonstrated to induce cellular senescence in neurons and microglia, as documented in SCZ [24,25,26,27]. For example, Escherichia coli (E. coli)-induced psychosis was reported in epidemics as well as in urinary tract infections (UTI), connecting bacteria to SCZ and SLDs [28].
Recent studies have found that downregulated DA receptors and transporters induce premature neuronal senescence, suggesting that dopaminergic signaling is required to avert early brain aging [29]. These findings were further substantiated by virus-induced senescence (VIS), a phenotype documented during the COVID-19 pandemic, marked by brain aging due to the infection of DA neurons [29,30]. Clostridium sp. are gut microbes also known for interfering with dopaminergic signaling, further linking cellular senescence to insufficient DA [31,32]. This is significant since both treated and untreated SCZ patients were found to exhibit body-wide premature cellular/neuronal aging, linking this condition to abnormal intestinal permeability [33,34]. Indeed, SCZ has been associated with increased microbial migration into the host’s systemic circulation [35,36,37]. Furthermore, premature cellular senescence may contribute to the other SCZ markers, including gray matter loss, decreased mitochondrial abundance, the attenuation of gamma (γ)-oscillations in EEGs, and the peroxidation of cell membrane lipids.

2.1. Ferrosenescence vs. Ferroptosis

Fe2+ is an essential nutrient for both hosts and pathogens. It is also a cofactor in the biosynthesis of tyrosine hydroxylase and tryptophan hydroxylase, enzymes involved in the synthesis of DA and serotonin (5-HT), respectively. In the gut, DA acts as a microbial siderophore, which clears Fe2+ from the microenvironment, lowering the risk of ferroptosis [38,39].
Senescent cells, including neurons, upregulate intracellular Fe2+, which in the vicinity of cytosolic lipids, may increase the risk of peroxidation and neuronal death by ferroptosis. In addition, senescence-upregulated lactate may also increase intracellular Fe2+ levels further, predisposing to neuronal demise [40]. Moreover, Fe2+ and lactate are known to enhance the biosynthesis of ceramide, a cell membrane lipid, which, in excess, can cause neurotoxicity [41,42]. Ferroptosis has been documented in SCZ; however, senescent cells are often resistant to programmed cell death, suggesting that impaired autophagy may drive iron-mediated brain aging [43,44,45]. Conversely, inducing ferroptosis in senescent cells precipitates their clearance by the immune system, indicating that this mechanism may be compensatory [46].
In our previous work, we introduced the concept of ferrosenescence, senescent cells, including neurons, with damaged DNA and dysfunctional p53-mediated genomic repair, as well as defective NKCs that are incapable of clearing senescent cells [47,48,49]. Ferrosenescent cells are resistant to ferroptosis due to impaired ferritin autophagy as well as the upregulation of ferroptosis inhibitors [glutathione peroxidase 4 (GPX-4) and apolipoprotein E (APOE)] [50,51,52,53]. For example, in SCZ, upregulated APOE, low ferritin levels, and increased intracellular Fe2+ levels likely reflect ferrosenescence [43,54]. Consequently, we believe that ferrosenescence may be more prevalent in SCZ than ferroptosis. Moreover, ferrosenescence may account for the other SCZ markers, including a decreased brain volume, γ-oscillations in EEGs, and mitochondrial dysfunction [55,56,57,58].

2.2. Senescent Gut Barrier

The relationship between the gut microbiome and senescent intestinal epithelial cells (IECs) is an emerging field that likely plays a major role in SCZ and SLDs [59,60]. For example, antibodies against translocated microbes, such as Hafnei alvei, Pseudomonas aeruginosa, Pseudomonas putida, and Klebsiella pneumoniae, were demonstrated in SCZ with negative symptoms, connecting microbial translocation to this pathology [61]. Moreover, translocated gut microbes may trigger nutritional immunity and iron sequestration in macrophages to withhold it from microbes, decreasing total circulatory Fe2+ levels [62]. In conditions of low circulatory Fe2+ levels, translocated bacteria may adopt a dormant state in human tissues, including the brain, awaiting increased Fe2+ availability to be reactivated [63,64].
The gut microbiota is immunologically tolerated in the gut lumen; however, upon translocation into the systemic circulation, the immune system is activated, triggering inflammation and, often, antibodies against microbial molecules. Since the microorganisms populating the GI tract express receptors identical or structurally related to human proteins, antibodies against these molecules may be construed as autoantibodies. Moreover, translocated microbes elicit inflammation, promoting cellular senescence, engendering a vicious circle in which senescent IECs facilitate the translocation of bacteria across the lamina propria, while inflammation triggered by these pathogens promotes further senescence [65,66,67].
Senescent cells were recently found to play a major role in the pathogenesis of inflammatory bowel disease (IBD), a condition marked by the translocation of microbes from the GI tract into the host tissues, including the brain [68]. This is further enhanced by the increased prevalence of SCZ in patients with IBD, emphasizing the role of the GI tract in severe mental illness [68,69]. Microbial translocation has been extensively studied in human immunodeficiency virus (HIV) infection, a condition marked by the massive exit of microbes from the GI tract due to interleukin-22 (IL-22) deficiency [70,71]. For this reason, we believe that recombinant IL-22 may comprise a new SCZ treatment [72].

3. Aryl Hydrocarbon, the Master Regulator of Cellular Senescence

The AhR is a ligand-activated transcription factor originally characterized as the receptor for dioxin (2,3,7,8-tetrachlorodibenzo-p-dioxin). Later, it was revealed that the AhR is also activated by various endogenous and exogenous ligands, driving multiple physiological processes and pathologies, likely including SCZ [73,74]. Aside from being the master regulator of cellular senescence, the AhR negatively regulates lactate and, by extension, the posttranslational modification, lactylation. As such, a dysfunctional AhR may drive both premature neuronal and glial aging as well as the excessive lactylation documented in SCZ and SLDs [75,76].
Several AhR ligands are molecules of interest in neuropsychiatry, including DA, phenazines, phenothiazines, serotonin, melatonin, and clozapine, suggesting that this transcription factor may play a significant role in the etiopathogenesis of SCZ (Figure 1) [77,78,79]. In prokaryotes, DA exerts iron-chelating properties; therefore, depletion in this neurotransmitter may upregulate intracellular Fe2+, leading to ferroptosis or ferrosenescence [39,43]. In addition, DA enhances the phagocytic properties of NKCs, facilitating the elimination of senescent and damaged cells [80]. Moreover, DA, via the DA 1 receptor (D1R), enhances the generation of acetylcholine (ACh), a neurotransmitter required for maintaining both the gray matter volume and rapid brain oscillations [81]. Conversely, lowering DA levels may promote inflammation by the accumulation of aged cells.
Compared with young neurons, senescent neuronal cells downregulate most surface receptors, including the dopaminergic and cholinergic ones, contain fewer mitochondria, and undergo metabolic and epigenetic reprogramming via lactate and lactylation, respectively [82]. Indeed, the lactylation of histone 3 (H3) lysine opposes neuronal senescence and restores the pre-senescent cellular status, suggesting that harnessing lactylation may comprise a potential neuropsychiatric therapy [83]. However, dysfunctional lactylation was shown to induce cell cycle reentry in senescent neurons, as demonstrated in Alzheimer’s disease (AD) and other neurodegenerative disorders [84]. Moreover, senescent microglia with histone 3 (H3K18) lactylation were demonstrated to adopt a neurotoxic phenotype, engaging in the elimination of healthy synapses and neurons, a phenomenon documented in SCZ and neurodegeneration [85,86,87,88,89]. Following this line of research, preclinical studies have revealed that neuronal excitation and social stress enhance the lactylation of histones, further linking this posttranslational modification to the pathogenesis of SCZ and SLDs [90]. Furthermore, previous studies have associated SCZ with increased brain lactate levels and the alteration of histone proteins, suggesting that lactylated neurotoxic microglia may be more detrimental to severe mental illness than previously thought [91,92,93,94].
Figure 1. In the cytosol, the AhR is stabilized by two HSP90 molecules. DA, oxidized lipids (and toxic ceramide), clozapine, serotonin, melatonin, and vitamin D3 are AhR ligands [95,96]. Pollutants, such as phthalate and bisphenol A (BPA), are also AhR ligands. In contrast, aripiprazole binds to the AhR chaperone, HSP90. HSP90 prevents AhR’s entry into the nucleus where it drives the transcription of genes, including those for cellular senescence.
Figure 1. In the cytosol, the AhR is stabilized by two HSP90 molecules. DA, oxidized lipids (and toxic ceramide), clozapine, serotonin, melatonin, and vitamin D3 are AhR ligands [95,96]. Pollutants, such as phthalate and bisphenol A (BPA), are also AhR ligands. In contrast, aripiprazole binds to the AhR chaperone, HSP90. HSP90 prevents AhR’s entry into the nucleus where it drives the transcription of genes, including those for cellular senescence.
Ijms 25 05904 g001

3.1. Gray and White Matter Loss

The data on sustained remission in SCZ match with neuroimaging studies, showing life-long gray matter loss in both medicated and unmedicated patients. This is significant, as preventing or restoring brain volume homeostasis likely improves outcomes in severe mental illness [97,98,99,100,101]. Indeed, preclinical studies have shown that AhR homeostasis is directly correlated with brain volume, suggesting that DA/AhR binding is critical for averting parenchymal loss [95]. The AhR also preserves the gray matter volume by regulating the availability of ACh, which, like DA, acts as a brain volume guardian [102]. Low brain DA levels also induce white matter loss, but to a lesser extent in SCZ compared with demyelinating disorders [103,104]. This is significant, as microbial toxins, including lipopolysaccharide (LPS), as well as environmental pollutants, such as plasticizers, are AhR ligands associated with SCZ. For example, the high comorbidity between SCZ and IBD may be the result of bacterial molecules “escaping” through the hyperpermeable intestinal barrier, a hallmark of IBD [105,106,107]. In addition, the higher prevalence of SCZ in northern regions of the world compared with the equatorial countries may be explained by AhR binding to vitamin D3 [96].
Taken together, DA is an indispensable neurotransmitter that prevents gray matter depletion by acting as an AhR ligand. Conversely, DA-blocking antipsychotic drugs may induce iatrogenic gray matter loss. This is further substantiated by the fact that clozapine, the most effective antipsychotic drug, is an AhR ligand that also upregulates DA levels [77,78,108].

3.2. Dopamine-Sparing Antipsychotics

Like SCZ itself, treatment with first- and second-generation antipsychotic drugs is associated with gray matter loss, suggesting that brain DA levels are strictly regulated so that minimal fluctuations in this neurotransmitter may cause brain volume depletion [99,100,109,110,111,112,113,114].
Novel studies have found that antipsychotic drugs transfer positive or negative electrical charges to their substrates, donating or accepting electrons. For example, DA, lithium, clozapine, novel phenothiazines, and aripiprazole give away electrons, preserving the gray matter volume, while most other antipsychotics are electron acceptors and associated with brain volume loss [115,116,117]. More studies are needed to identify and develop DA-sparing, electron-donor antipsychotics.

4. Mitochondrial Dysfunction and Loss of Gamma-Band Oscillations

According to the endosymbiotic hypothesis, mitochondria are derived from ancestral bacteria and can communicate with the gut microbes via chemical messengers, such as ROS and sphingolipids [118,119,120]. Ceramide, one of the sphingolipids, is secreted by the microbiome and, in excess, can be toxic to mitochondria [121]. Senescent cells, including neurons, upregulate ceramide, probably accounting for the paucity of mitochondria in the aging brain [122,123,124]. Moreover, senescence-upregulated intracellular Fe2+ disrupts ceramide metabolism, further contributing to mitochondrial loss [125]. Indeed, ceramide-induced mitochondrial damage has been associated with atherosclerosis and SCZ, disorders associated with premature cellular senescence [126,127].
Acid sphingomyelinase (ASM), the enzyme catalyzing the conversion of sphingomyelin into ceramide, has been identified as an SCZ target, and ASM inhibitors, such as fluvoxamine and rosuvastatin, appear to ameliorate clinical outcomes in this disorder [128]. In addition, a natural alkaloid, berberine, decreases ceramide levels as well as inflammation, indicating a potentially beneficial effect on SCZ and SLDs [129,130].
Neurons have the capability of replacing defective mitochondria by importing them from microglia and astrocytes via tunneling nanotubules or extracellular vesicles (EVs) (Figure 2) [131,132]. Indeed, preventing neuronal loss by supplying mitochondria to neurons is one of the main functions of astrocytes.
Interestingly, antidepressant drugs in the class of serotonin reuptake inhibitors (SSRIs), including fluvoxamine, facilitate mitochondrial export to neuronal cells, emphasizing the neuroprotective role of these agents (Figure 3) [133]. Indeed, SSRIs were demonstrated to delay the conversion of mild cognitive impairment (MCI) into dementia, indicating that mitochondrial import contributes to neuronal rescue [134]. In addition, ferroptosis-blocking drugs and iron chelators, including halogenated phenazines, may delay or prevent neurodegenerative disorders, suggesting a novel therapeutic approach (see the section on phenazines and phenothiazines) [135,136].
Figure 2. Glial cells, including astrocytes, supply neurons with healthy mitochondria via tunneling nanotubules, preventing apoptosis [137,138]. In addition, astrocytes prevent neuronal ferroptosis by transferring antioxidants, including GPX-4. Astrocytes uptake cystine via the cystine/glutamate antiporter (Xc). Cysteine can also be obtained from methionine via glutathione. Fe2+ enters neurons through the transferrin receptor-1 (TRF-1), which is stored in ferritin and requires ferritinophagy to be released. Excess Fe2+ exits the neurons via ferroportin (FPT) channels.
Figure 2. Glial cells, including astrocytes, supply neurons with healthy mitochondria via tunneling nanotubules, preventing apoptosis [137,138]. In addition, astrocytes prevent neuronal ferroptosis by transferring antioxidants, including GPX-4. Astrocytes uptake cystine via the cystine/glutamate antiporter (Xc). Cysteine can also be obtained from methionine via glutathione. Fe2+ enters neurons through the transferrin receptor-1 (TRF-1), which is stored in ferritin and requires ferritinophagy to be released. Excess Fe2+ exits the neurons via ferroportin (FPT) channels.
Ijms 25 05904 g002
Aside from supplying healthy mitochondria to neurons, astrocytes also export antioxidants, including GPX-4, an enzyme involved in rescuing neuronal cells from ferroptosis. Additionally, ASM inhibitors, such as fluvoxamine, facilitate GPX-4 biosynthesis, averting ferroptosis. Indeed, fluvoxamine and other SSRIs preserve neuronal cells by both inhibiting ferroptosis and facilitating mitochondrial transfer [137,138]. GPX-4 is derived from cysteine, which enters astrocytes via the cystine/glutamate antiporter (Xc) and enables the biosynthesis of glutathione and GPX-4 (Figure 2).
Given that γ-band frequencies are energy-consuming, mitochondria are essential for the generation of these rapid brain oscillations [139]. In this regard, preclinical studies have shown that lost γ-oscillations can be restored by inhibiting GSK-3β, an enzyme previously implicated in SCZ and SLDs [140,141]. Several antipsychotic drugs, including lithium, and natural compounds, kaempferol, are GSK-3β inhibitors and, therefore, capable of restoring the γ-rhythm (Figure 3).

Entrainment of Gamma-Band Oscillations in Schizophrenia

Oscillations in the γ-range are rhythmic patterns of high-frequency (25 Hz to 100 Hz) EEG waves, playing a key role in cognition, attention, perception, and movement [142]. Under normal circumstances, this neuronal activity is synchronized across numerous brain regions, while in SCZ, there are γ-rhythm abnormalities, especially those elicited by auditory stimuli [143,144].
Interneurons, including parvalbumin (PV), vasoactive intestinal polypeptide (VIP), and somatostatin (SST), are the main drivers of γ-rhythms, indicating that the defective inhibition of pyramidal cells may trigger the loss of rapid rhythms [145,146]. Regarding neurotransmitters, γ-oscillations are dependent on ACh, an AhR-regulated biomolecule controlled by acetylcholinesterase (AChE), a direct AhR ligand [147].
Several studies have connected γ-oscillations to the microbiome-derived ACh, linking rapid EEG rhythms to intestinal microbes [148,149]. This is significant, as decreased brain ACh levels in SCZ have been associated with gray matter loss, emphasizing the key role of the AhR and AChE in maintaining both brain volume and rapid oscillations [150,151]. Given the important role of the cholinergic system in SCZ, it is not surprising that novel ACh-based antipsychotics have been developed [130,152].
Aside from ACh and mitochondrial import, lost γ-oscillations may be restored by entrainment with sensory stimuli, such as ultrasound or transcutaneous vagal nerve stimulation (tVNS) at 40 Hz, emphasizing a potential strategy for SCZ and SLDs [149,153]. Interestingly, tVNS improves not only neuronal function but also optimizes gut permeability, decreasing microbial translocation [149,154].

5. Senotherapeutics

It is currently established that severe mental illness is associated with cellular/neuronal senescence, indicating that endogenous or exogenous toxins may play a key role in this pathology [155,156,157]. For example, viral or bacterial infections induce premature aging in brain cells by eliciting immune responses, likely triggering psychosis [158,159]. Indeed, new-onset psychosis was documented in infections with senescence-inducing viruses including HIV and SARS-CoV-2 [160,161].
Senotherapeutics are natural or synthetic compounds that can delay, prevent, or reverse cellular/neuronal senescence. Senotherapeutics comprise senolytic agents that facilitate the elimination of senescent cells, and senomorphic compounds capable of deleting senescence markers, including SASP and SA-β-gal [162]. While, in the past, it was thought that cellular senescence could not be reversed, newer studies have found that inhibiting 3-phosphoinositide-dependent protein kinase-1 (PDK1) can revert cellular senescence in humans [163]. Interestingly, PDK1 is an upstream modulator of SCZ-linked GSK-3β.
Senolytic antibiotics belong to a distinct class of agents that include azithromycin, minocycline, and roxithromycin and possess neuroprotective, anti-inflammatory, and senolytic properties [164]. For example, it has been known for some time that minocycline may be beneficial for SCZ, suggesting that senolytics have a place in the treatment of SCZ and SLDs, probably by clearing neurotoxic glial cells [165].
Other senolytic agents relevant to SCZ and SLDs are summarized in Table 1.
A senolytic vaccine recently tested in progeroid mice may usher in a new era of neuropsychiatry, raising the possibility of vaccination or serum treatment for SCZ and SLDs [174]. Another immunological intervention, an antibody–drug conjugate against a membrane senescence marker, was demonstrated to clear senescent, damaged, or infected cells, emphasizing a new therapeutic strategy [175].

6. Membrane Lipid Replacement (MLR)

MLR refers to the oral administration of natural cell membrane glycerophospholipids, along with kaempferol (3,4′,5,7-tetrahydroxyflavone), a flavonoid found in tea, broccoli, cabbage, kale, beans, endive, leek, tomato, strawberries, and grapes [176]. Like lithium and some antipsychotics, kaempferol is an inhibitor of GSK-3β, suggesting that it may exert antipsychotic properties without the typical adverse effects of psychotropic drugs [177,178].
The administration of MLR + kaempferol gradually replaces damaged phospholipids, ceramides, and oxysterols from neuronal and/or mitochondrial membranes with natural glycerophospholipids and a polyphenol.
Oxidized membrane lipids are AhR ligands that have previously been implicated in the pathogenesis of SCZ (Figure 1). MLR and kaempferol exert a dual mechanism of action: the elimination of lipid peroxides and GSK-3β inhibition [179]. Replacing oxidized plasma and/or mitochondrial membrane fats with healthy natural lipids averts ferroptosis and optimizes neurotransmission by correcting membrane distortion. Conversely, oxidized membrane lipids can trigger neuronal demise by ferroptosis [180]. Indeed, MLR reverses biophysical changes in the plasma and mitochondrial membrane induced by oxidized lipids. This action is not different from that of phenothiazines, which insert themselves into the lipid bilayer, lowering the curvature of cell membranes (Figure 4). In contrast, oxidized lipids form looped structures and generate membrane curvatures and pores, which leads to cell death [181].

7. Phenazines and Antioxidant Phenothiazines

Phenazines are microbial metabolites produced by various soil and water microorganisms that exert antibacterial, anticancer, antimalarial, and antipsychotic properties [184].

7.1. Natural Phenazines

Natural phenazines are synthesized by bacteria, including Streptococcus species and Pseudomonas aeruginosa, the latter known for generating pyocyanin (5-N-methyl-1-hydroxyphenazine), a compound with electron-shuttling properties [185,186].
Neuroprotective natural phenazines such as geranyl-phenazine, an AChE inhibitor, upregulate ACh, exerting antipsychotic effects via muscarinic receptors [187,188]. Another natural phenazine with neuroprotective functions, bara-phenazines A–G, are fused molecules with antipsychotic properties derived from Streptomyces sp. PU-10A [189].

7.2. Synthetic Phenazine Derivatives

Synthetic phenazine derivatives consist of over 6000 compounds, exerting antimicrobial, antiparasitic, neuroprotective, anti-inflammatory, and anticancer activities. Pontemazines A and B are neuroprotective phenazines derived from Streptomyces sp. UT1123, which, in animal studies, rescued hippocampal neurons from glutamate cytotoxicity, highlighting their pro-cognitive properties that could benefit patients with negative SCZ symptoms [190]. Pontemazines exert antioxidant, radical-scavenging properties and inhibit lipid peroxidation, suggesting beneficial effects on SCZ [191]. Halogenated phenazines act as iron chelators and are probably helpful against ferroptosis [192,193]. We believe that pontemazines and halogenated phenazines should be assessed for their antipsychotic and anti-neurodegenerative properties (Figure 4).
From a biochemical standpoint, phenazines are almost identical to phenothiazine antipsychotics and likely possess similar properties (Figure 5). Phenothiazines are typical antipsychotic drugs utilized primarily for SCZ and SLDs that block dopaminergic transmission at the level of postsynaptic neurons. They also correct the curvature and receptor alignment on neuronal/mitochondrial surfaces, restoring signaling homeostasis (Figure 4) [182]. In contrast, oxidized lipids, toxic ceramides, and 7-ketocholesterol (7KCl) form looped structures, generating membrane curvatures and pores that may lead to neuronal death [183].

7.3. Antioxidant Phenothiazines and Their Derivatives

Antioxidant phenothiazines and their derivatives have recently been developed for cancer, cardiovascular disease (CVD), Mycobacterium leprae, and other antibiotic-resistant microbes [194].
Phenothiazine derivatives exert anti-peroxidation properties and protect against lipid pathology and ferroptosis, suggesting their efficacy as antipsychotic drugs [195]. Phenothiazine nuclei possess hydrophobic properties that facilitate their insertion into the plasma or mitochondrial membranes [196].
Propenyl-phenothiazine is a potent antioxidant with electron-donor capabilities that likely prevents gray matter loss in patients with SCZ or SLDs. Moreover, a new category of tetracyclic and pentacyclic phenothiazines with antioxidant properties have recently been developed, suggesting their likely efficacy for cognitive impairment and negative SCZ symptoms [197,198]. Furthermore, N10-carbonyl-substituted phenothiazines were demonstrated to inhibit lipid peroxidation, suggesting their enhanced antipsychotic efficacy [100].

8. Mitochondrial Transfer and Transplantation

Mitochondrial transplantation experiments started in the 1980s, wherein naked organelles were co-incubated with various cell types, attempting to facilitate internalization [199]. Using HeLa cells and mesenchymal stem cells as mitochondrial sources, this transplantation technique takes only 1–2 h to supply organelles to mitochondria-depleted cells [200,201,202]. At present, mitochondrial transplantation in cardiomyocytes is possible and can be confirmed by the presence of mitochondrial DNA (mtDNA) in the heart [203,204].
Mitochondrial transplantation to rescue neurons from ferroptosis is currently possible and has been successfully performed in both animals and humans; however, to the best of our knowledge, it has not been attempted as a treatment for mental illness [205].
Rescuing the mitochondria with MLR, kaempferol, and berberine (Figure 6) is a strategy for averting GSK-3β overactivation by toxic ceramides, oxysterols, or oxidized phospholipids [130,206]. In addition, SSRIs, GJA1-20K, and CD38 signaling were demonstrated to facilitate mitochondrial transfer, emphasizing potential strategies for restoring neurometabolic homeostasis in severe mental illness and neurodegeneration [207,208].

9. Using AhR Antagonists as Antipsychotics

Aberrant AhR overactivation has been associated with psychosis, while several antagonists of this receptor exert antipsychotic properties. The following natural and synthetic AhR inhibitors were found to be therapeutic for SCZ:
  • Quercetin is a natural flavonoid and plant pigment that exerts antioxidant and anticancer properties. In the CNS, quercetin is a negative allosteric modulator of GABARs as well as an enhancer of glutamatergic neurotransmission, a signaling pathway deficient in SCZ [209]. In addition, quercetin inhibits the apoptosis of cortical neurons, likely preventing gray matter loss.
  • Apigenin is a plant-based remedy extract from Elsholtzia rugulosa used by traditional practitioners from Africa for treating mental illness. Aside from antagonizing the AhR, apigenin exhibits vasorelaxant, antioxidant, and antipsychotic properties [209].
  • Alstonine is an indole alkaloid with antipsychotic effects that increases serotonergic, but not dopaminergic, signaling, possibly facilitating mitochondrial transfer [209,210].
  • Luteolin is a natural antipsychotic that exerts its beneficial actions by reducing microglial inflammation [202]. Luteolin is currently under clinical trials for SCZ treatment (NCT05204407).

10. Synthetic AhR Antagonists

Synthetic AhR antagonists are anti-inflammatory and anticancer compounds that likely exert antipsychotic properties.
IK-175 (structure undisclosed) was shown to block ligand-mediated AhR activation in preclinical studies. IK-175 was recently approved for cancer treatment, and it may possess antipsychotic properties [211].
HBU651 is a novel synthetic AhR antagonist developed primarily for cancer treatment that appears to be a suitable candidate for SCZ treatment [212]. Figure 7 summarizes AhR agonists/antagonists relevant for neuropsychiatry.

11. Recombinant Interleukin-22

Recombinant interleukin-22 (IL-22) is a pleiotropic cytokine known for facilitating tissue regeneration and protecting the GI tract barrier. Recombinant IL-22, comprising two molecules connected by a fusion protein, exerts better efficacy with reduced systemic side effects [223].
In our previous work, we hypothesized that SCZ and SLDs may be initiated by aberrant AhR hyperactivation by endogenous or exogenous ligands, including intestinal or environmental toxins, such as LPS or plasticizers, respectively [224].
This hypothesis is supported by the following findings:
  • SCZ is often comorbid with IBD, conditions associated with increased gut barrier permeability and microbial translocation from the GI tract into host tissues, including the brain.
  • Translocation markers, including soluble CD14 (sCD14) and lipopolysaccharide-binding protein (LBP), are elevated in SCZ, suggesting bacterial translocation.
  • Increased BBB permeability in SCZ enables translocated gut microbes to reach the brain.
The following are examples of pathogens triggering psychosis:
  • The Escherichia coli (E. coli) outbreak in 2011 in Germany has been associated with cases of new-onset psychosis.
  • New-onset psychosis, or its exacerbation, has been identified in E. coli-associated UTIs.
IL-22 has been successfully used to restore the integrity of the gut barrier in various conditions, including IBD, HIV, and nonalcoholic fatty liver disease [225,226]. We construe that recombinant IL-22 would be effective for SCZ by limiting the translocation of bacteria and/or their molecules.
Table 2 summarizes the major representatives from the categories discussed above with potential therapeutic properties for SCZ and SLDs.

12. Vehicles: Lipid Nanoparticles

The COVID-19 pandemic has accelerated the development of lipid nanoparticles (LNPs), vehicles for drug delivery. As LNPs are liposoluble, they can access specific body niches, including the brain [227].
The COVID-19 messenger RNA (mRNA) vaccines, Pfizer-BioNTech and Moderna, are incorporated in LNPs comprising four lipids: 1,2-distearoyl-sn-glycero-3-phosphocholine (DSPC), PEG, an alternative cholesterol, and ionizable lipids such as SM-102 or ALC-0315 [228]. SM-102, ALC-0315, and the alternative cholesterol are proprietary molecules and have not been revealed. However, looking at the previous LNP research, the ionizable lipids likely resemble DLin-MC3-DMA, which was approved by the Food and Drug Administration (FDA) for transthyretin-mediated amyloidosis [229,230,231]. Like phenothiazines, LNPs enter cells, including neurons, through the endocytic pathway (EP). Subsequently, LNPs travel from the early to late endosomes, but not to lysosomes because the organelle pH of 4.5–5.0 could degrade the nanoparticles. Therefore, “lysosomal escape” into the cytosol must occur from the late endosomes without interfering with autophagy, as most antipsychotic drugs do.
Utilizing LNPs not for vaccination, but as a vehicle for transporting psychotropic drugs directly to the neuronal networks could revolutionize psychopharmacology. As nano-doses of antipsychotics or mood stabilizers would be utilized for the treatment of psychotic symptoms or affective disorders, systemic adverse effects would be avoided. We surmise that LNPs would be extremely efficacious as vehicles for psychotropic drugs.

13. Conclusions

Antipsychotic drugs are extremely helpful for the acute symptoms of SCZ or SLDs; however, sustained recovery (measured by the ability to hold a job, go to school, raise a family, or be independent in ADLs) is rarely achieved. For this reason, the next chapter in neuropsychopharmacology will have to improve functionality rather than symptom resolution.
Over the past 70 years, it has become obvious that lowering dopaminergic transmission does not restore premorbid functions in patients with SCZ and SLDs. DA is an indispensable neurotransmitter that maintains the integrity of the brain parenchyma, a physiological function disrupted by both SCZ and its treatment.
The study of neuronal senescence-induced neuropsychiatric disorders is in its infancy but is rapidly developing, especially after the advent of age-accelerating viruses, such as HIV and COVID-19. Aberrantly activated AhR, the master regulator of cellular senescence, explains not only how gut microbes and/or their molecules trigger psychosis but also how environmental pollutants precipitate SCZ or SLDs. AhR hyperactivation likely accounts for gray matter reduction, the loss of rapid brain oscillations, and oxidized lipids in the plasma and mitochondrial membranes. The recent discovery that DA, serotonin, vitamin D3, clozapine, and melatonin are AhR ligands has opened new horizons in the management of chronic psychosis. Novel strategies, such as natural senotherapeutics, MLR, GSK-3β, ceramide inhibitors, recombinant IL-22, mitochondrial transplantation or transfer, and AhR antagonists, could improve long-term recovery in SCZ or SLDs. Senescence-associated downregulation of ACh and DA likely reduces brain volume and rapid oscillations, contributing to deficit symptoms.
The utilization of natural compounds, such as kaempferol or berberine, alone or in conjunction with LNP-delivered DA-sparing antipsychotic drugs, may improve sustained recovery in patients with severe mental illness.

Author Contributions

A.S. conceptualization, S.H. writing—review and editing, D.O.S. data curation, H.I. methodology and software, J.J.A. draft preparation, Z.K. supervision. 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.

Conflicts of Interest

The authors declare no conflicts of interest. Where authors are identified as personnel of the International Agency for Research on Cancer/WHO, the authors alone are responsible for the views expressed in this article and they do not necessarily represent the decisions, policy or views of the International Agency for Research on Cancer/WHO.

References

  1. Lamb, H.R. Deinstitutionalization and the homeless mentally ill. Hosp. Community Psychiatry 1984, 35, 899–907. [Google Scholar] [CrossRef] [PubMed]
  2. Scott, J. Homelessness and mental illness. Br. J. Psychiatry 1993, 162, 314–324. [Google Scholar] [CrossRef] [PubMed]
  3. Insel, T. Rethinking schizophrenia. Nature 2010, 468, 187–193. [Google Scholar] [CrossRef] [PubMed]
  4. Delport, A.; Harvey, B.H.; Petzer, A.; Petzer, J.P. The monoamine oxidase inhibition properties of selected structural analogues of methylene blue. Toxicol. Appl. Pharmacol. 2017, 325, 1–8. [Google Scholar] [CrossRef]
  5. Jääskeläinen, E.; Juola, P.; Hirvonen, N.; McGrath, J.J.; Saha, S.; Isohanni, M.; Veijola, J.; Miettunen, J. A Systematic Review and Meta-Analysis of Recovery in Schizophrenia. Schizophr. Bull. 2012, 39, 1296–1306. [Google Scholar] [CrossRef]
  6. Warner, R. Recovery from Schizophrenia Psychiatry and Political Economy, 3rd ed.; Brunner-Routledge: Hove, UK; New York, NY, USA, 1997; p. 74. [Google Scholar]
  7. Üçok, A.; Polat, A.; Çakır, S.; Genç, A. One year outcome in first episode schizophrenia: Predictors of relapse. Eur. Arch. Psychiatry Clin. Neurosci. 2005, 256, 37–43. [Google Scholar] [CrossRef]
  8. Holm, M.; Taipale, H.; Tanskanen, A.; Tiihonen, J.; Mitterdorfer-Rutz, E. Employment among people with schizophrenia or bipolar disorder: A population-based study using nationwide registers. Acta Psychiatr. Scand. 2020, 143, 61–71. [Google Scholar] [CrossRef] [PubMed]
  9. Lévesque, I.S.; Abdel-Baki, A. Homeless youth with first-episode psychosis: A 2-year outcome study. Schizophr. Res. 2019, 216, 460–469. [Google Scholar] [CrossRef]
  10. Lai, C.H.; Wu, Y.T.; Chen, C.Y.; Hou, Y.C. Gray matter increases in fronto-parietal regions of depression patients with aripiprazole monotherapy: An exploratory study. Medicine 2016, 95, e4654. [Google Scholar] [CrossRef]
  11. Rollema, H.; Lu, Y.; Schmidt, A.W.; Zorn, S.H. Clozapine increases dopamine release in prefrontal cortex by 5-HT1A receptor activation. Eur. J. Pharmacol. 1997, 338, R3–R5. [Google Scholar] [CrossRef]
  12. Papanastasiou, E.; Gaughran, F.; Smith, S. Schizophrenia as segmental progeria. J. R. Soc. Med. 2011, 104, 475–484. [Google Scholar] [CrossRef] [PubMed]
  13. Russo, P.; Prinzi, G.; Proietti, S.; Lamonaca, P.; Frustaci, A.; Boccia, S.; Amore, R.; Lorenzi, M.; Onder, G.; Marzetti, E.; et al. Shorter telomere length in schizophrenia: Evidence from a real-world population and meta-analysis of Most recent literature. Schizophr. Res. 2018, 202, 37–45. [Google Scholar] [CrossRef] [PubMed]
  14. Dada, O.; Adanty, C.; Dai, N.; Jeremian, R.; Alli, S.; Gerretsen, P.; Graff, A.; Strauss, J.; De Luca, V. Biological aging in schizophrenia and psychosis severity: DNA methylation analysis. Psychiatry Res. 2021, 296, 113646. [Google Scholar] [CrossRef] [PubMed]
  15. Schnack, H.G.; van Haren, N.E.; Nieuwenhuis, M.; Hulshoff Pol, H.E.; Cahn, W.; Kahn, R.S. Accelerated Brain Aging in Schizophrenia: A Longitudinal Pattern Recognition Study. Am. J. Psychiatry 2016, 173, 607–616. [Google Scholar] [CrossRef] [PubMed]
  16. Langhi Prata, L.G.P.; Tchkonia, T.; Kirkland, J.L. Cell senescence, the senescence-associated secretory phenotype, and cancers. PLoS Biol. 2023, 21, e3002326. [Google Scholar] [CrossRef] [PubMed]
  17. Huang, W.; Hickson, L.J.; Eirin, A.; Kirkland, J.L.; Lerman, L.O. Cellular senescence: The good, the bad and the unknown. Nat. Rev. Nephrol. 2022, 18, 611–627. [Google Scholar] [CrossRef] [PubMed]
  18. Stec, A.; Maciejewska, M.; Zaremba, M.; Paralusz-Stec, K.; Michalska, M.; Rudnicka, L.; Sikora, M. The Clinical Significance of Serum Biomarkers of the Intestinal Barrier in Systemic Sclerosis: A Cross-Sectional Study. J. Pers. Med. 2023, 13, 678. [Google Scholar] [CrossRef]
  19. Sepich-Poore, G.D.; Zitvogel, L.; Straussman, R.; Hasty, J.; Wargo, J.A.; Knight, R. The microbiome and human cancer. Science 2021, 371, eabc4552. [Google Scholar] [CrossRef]
  20. White, M.G.; Wargo, J.A. Gut Microbes’ Impact on Oncogenic Drivers: Location Matters. Mol. Cell 2020, 79, 878–880. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  21. Ağagündüz, D.; Cocozza, E.; Cemali, Ö.; Bayazıt, A.D.; Nanì, M.F.; Cerqua, I.; Morgillo, F.; Saygılı, S.K.; Berni Canani, R.; Amero, P.; et al. Understanding the role of the gut microbiome in gastrointestinal cancer: A review. Front. Pharmacol. 2023, 14, 1130562. [Google Scholar] [CrossRef]
  22. Mijit, M.; Caracciolo, V.; Melillo, A.; Amicarelli, F.; Giordano, A. Role of p53 in the Regulation of Cellular Senescence. Biomolecules 2020, 10, 420. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  23. Ni, X.; Trakalo, J.; Valente, J.; Azevedo, M.H.; Pato, M.T.; Pato, C.N.; Kennedy, J.L. Human p53 tumor suppressor gene (TP53) and schizophrenia: Case-control and family studies. Neurosci. Lett. 2005, 388, 173–178. [Google Scholar] [CrossRef] [PubMed]
  24. Dono, A.; Nickles, J.; Rodriguez-Armendariz, A.G.; McFarland, B.C.; Ajami, N.J.; Ballester, L.Y.; Wargo, J.A.; Esquenazi, Y. Glioma and the gut-brain axis: Opportunities and future perspectives. Neurooncol. Adv. 2022, 4, vdac054. [Google Scholar] [CrossRef] [PubMed]
  25. Palacios, E.; Lobos-González, L.; Guerrero, S.; Kogan, M.J.; Shao, B.; Heinecke, J.W.; Quest, A.F.G.; Leyton, L.; Valenzuela-Valderrama, M. Helicobacter pylori outer membrane vesicles induce astrocyte reactivity through nuclear factor-κappa B activation and cause neuronal damage in vivo in a murine model. J. Neuroinflamm. 2023, 20, 66. [Google Scholar] [CrossRef] [PubMed]
  26. Zhuo, C.; Tian, H.; Song, X.; Jiang, D.; Chen, G.; Cai, Z.; Ping, J.; Cheng, L.; Zhou, C.; Chen, C.; et al. Microglia and cognitive impairment in schizophrenia: Translating scientific progress into novel therapeutic interventions. Schizophrenia 2023, 9, 42. [Google Scholar] [CrossRef] [PubMed]
  27. Niraula, A.; Sheridan, J.F.; Godbout, J.P. Microglia Priming with Aging and Stress. Neuropsychopharmacol. Rev. 2017, 42, 318–333. [Google Scholar] [CrossRef] [PubMed]
  28. Wiwanitkit, V. Hemolysis in E. coli O104, H4 Infection. Indian J. Hematol. Blood Transfus. 2012, 28, 127. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  29. Karrer, T.M.; Josef, A.K.; Mata, R.; Morris, E.D.; Samanez-Larkin, G.R. Reduced dopamine receptors and transporters but not synthesis capacity in normal aging adults: A meta-analysis. Neurobiol. Aging 2017, 57, 36–46. [Google Scholar] [CrossRef] [PubMed]
  30. Yang, L.; Kim, T.W.; Han, Y.; Nair, M.S.; Harschnitz, O.; Zhu, J.; Wang, P.; Koo, S.Y.; Lacko, L.A.; Chandar, V.; et al. SARS-CoV-2 infection causes dopaminergic neuron senescence. Cell Stem Cell 2024, 31, 196–211.e6. [Google Scholar] [CrossRef] [PubMed]
  31. Fettucciari, K.; Fruganti, A.; Stracci, F.; Spaterna, A.; Marconi, P.; Bassotti, G. Clostridioides difficile Toxin B Induced Senescence: A New Pathologic Player for Colorectal Cancer? Int. J. Mol. Sci. 2023, 24, 8155. [Google Scholar] [CrossRef]
  32. Vinithakumari, A.A.; Padhi, P.; Hernandez, B.; Lin, S.J.; Dunkerson-Kurzhumov, A.; Showman, L.; Breitzman, M.; Stokes, C.; Sulaiman, Y.; Tangudu, C.; et al. Clostridioides difficile Infection Dysregulates Brain Dopamine Metabolism. Microbiol. Spectr. 2022, 10, e0007322. [Google Scholar] [CrossRef]
  33. Feng, Y.; Shen, J.; He, J.; Lu, M. Schizophrenia and cell senescence candidate genes screening, machine learning, diagnostic models, and drug prediction. Front. Psychiatry 2023, 14, 1105987. [Google Scholar] [CrossRef] [PubMed]
  34. Ling, E.; Nemesh, J.; Goldman, M.; Kamitaki, N.; Reed, N.; Handsaker, R.E.; Genovese, G.; Vogelgsang, J.S.; Gerges, S.; Kashin, S.; et al. A concerted neuron–astrocyte program declines in ageing and schizophrenia. Nature 2024, 627, 604–611. [Google Scholar] [CrossRef] [PubMed]
  35. Ishida, I.; Ogura, J.; Aizawa, E.; Ota, M.; Hidese, S.; Yomogida, Y.; Matsuo, J.; Yoshida, S.; Kunugi, H. Gut permeability and its clinical relevance in schizophrenia. Neuropsychopharmacol. Rep. 2022, 42, 70–76. [Google Scholar] [CrossRef] [PubMed]
  36. Scheurink, T.A.W.; Borkent, J.; Gangadin, S.S.; El Aidy, S.; Mandl, R.; Sommer, I.E.C. Association between gut permeability, brain volume, and cognition in healthy participants and patients with schizophrenia spectrum disorder. Brain Behav. 2023, 13, e3011. [Google Scholar] [CrossRef] [PubMed]
  37. Wasiak, J.; Gawlik-Kotelnicka, O. Intestinal permeability and its significance in psychiatric disorders—A narrative review and future perspectives. Behav. Brain Res. 2023, 448, 114459. [Google Scholar] [CrossRef] [PubMed]
  38. Lotan, A.; Luza, S.; Opazo, C.M.; Ayton, S.; Lane, D.J.R.; Mancuso, S.; Pereira, A.; Sundram, S.; Weickert, C.S.; Bousman, C.; et al. Perturbed iron biology in the prefrontal cortex of people with schizophrenia. Mol. Psychiatry 2023, 28, 2058–2070. [Google Scholar] [CrossRef]
  39. Dichtl, S.; Demetz, E.; Haschka, D.; Tymoszuk, P.; Petzer, V.; Nairz, M.; Seifert, M.; Hoffmann, A.; Brigo, N.; Würzner, R.; et al. Dopamine Is a Siderophore-Like Iron Chelator That Promotes Salmonella enterica Serovar Typhimurium Virulence in Mice. mBio 2019, 10, e02624-18. [Google Scholar] [CrossRef] [PubMed]
  40. Anderson, G.J.; Frazer, D.M. Lactate as a regulator of iron homeostasis. Life Metab. 2023, 2, load033. [Google Scholar] [CrossRef]
  41. Cai, Z.; Deng, L.; Fan, Y.; Ren, Y.; Ling, Y.; Tu, J.; Cai, Y.; Xu, X.; Chen, M. Dysregulation of Ceramide Metabolism Is Linked to Iron Deposition and Activation of Related Pathways in the Aorta of Atherosclerotic Miniature Pigs. Antioxidants 2024, 13, 4. [Google Scholar] [CrossRef]
  42. de la Monte, S.M. Triangulated mal-signaling in Alzheimer’s disease: Roles of neurotoxic ceramides, ER stress, and insulin resistance reviewed. J. Alzheimers Dis. 2012, 30 (Suppl. S2), S231–S249. [Google Scholar] [CrossRef] [PubMed]
  43. Ravanfar, P.; Syeda, W.T.; Jayaram, M.; Rushmore, R.J.; Moffat, B.; Lin, A.P.; Lyall, A.E.; Merritt, A.H.; Yaghmaie, N.; Laskaris, L.; et al. In Vivo 7-Tesla MRI Investigation of Brain Iron and Its Metabolic Correlates in Chronic Schizophrenia. Schizophrenia 2022, 8, 86. [Google Scholar] [CrossRef] [PubMed]
  44. Feng, S.; Chen, J.; Qu, C.; Yang, L.; Wu, X.; Wang, S.; Yang, T.; Liu, H.; Fang, Y.; Sun, P. Identification of Ferroptosis-Related Genes in Schizophrenia Based on Bioinformatic Analysis. Genes 2022, 13, 2168. [Google Scholar] [CrossRef]
  45. Lian, K.; Li, Y.; Yang, W.; Ye, J.; Liu, H.; Wang, T.; Yang, G.; Cheng, Y.; Xu, X. Hub genes, a diagnostic model, and immune infiltration based on ferroptosis-linked genes in schizophrenia. IBRO Neurosci. Rep. 2024, 16, 317–328. [Google Scholar] [CrossRef] [PubMed]
  46. Go, S.; Kang, M.; Kwon, S.P.; Jung, M.; Jeon, O.H.; Kim, B.S. The Senolytic Drug JQ1 Removes Senescent Cells via Ferroptosis. Tissue Eng. Regen. Med. 2021, 18, 841–850. [Google Scholar] [CrossRef] [PubMed]
  47. Masaldan, S.; Clatworthy, S.A.S.; Gamell, C.; Meggyesy, P.M.; Rigopoulos, A.T.; Haupt, S.; Haupt, Y.; Denoyer, D.; Adlard, P.A.; Bush, A.I.; et al. Iron accumulation in senescent cells is coupled with impaired ferritinophagy and inhibition of ferroptosis. Redox Biol. 2018, 14, 100–115. [Google Scholar] [CrossRef] [PubMed]
  48. Sfera, A.; Osorio, C.; Maguire, G.; Rahman, L.; Afzaal, J.; Cummings, M.; Maldonado, J.C. COVID-19, ferrosenescence and neurodegeneration, a mini-review. Prog. Neuropsychopharmacol. Biol. Psychiatry 2021, 109, 110230. [Google Scholar] [CrossRef]
  49. Sfera, A.; Bullock, K.; Price, A.; Inderias, L.; Osorio, C. Ferrosenescence: The iron age of neurodegeneration? Mech. Ageing Dev. 2018, 174, 63–75. [Google Scholar] [CrossRef] [PubMed]
  50. Boshkovski, T.; Cohen-Adad, J.; Misic, B.; Arnulf, I.; Corvol, J.C.; Vidailhet, M.; Lehéricy, S.; Stikov, N.; Mancini, M. The Myelin-Weighted Connectome in Parkinson’s Disease. Mov. Disord. 2022, 37, 724–733. [Google Scholar] [CrossRef]
  51. Belaidi, A.A.; Masaldan, S.; Southon, A.; Kalinowski, P.; Acevedo, K.; Appukuttan, A.T.; Portbury, S.; Lei, P.; Agarwal, P.; Leurgans, S.E.; et al. Apolipoprotein E potently inhibits ferroptosis by blocking ferritinophagy. Mol. Psychiatry 2022, 29, 211–220. [Google Scholar] [CrossRef]
  52. Arnold, S.E.; Joo, E.; Martinoli, M.G.; Roy, N.; Trojanowski, J.Q.; Gur, R.E.; Cannon, T.; Price, R.A. Apolipoprotein E genotype in schizophrenia: Frequency, age of onset, and neuropathologic features. Neuroreport. 1997, 8, 1523–1526. [Google Scholar] [CrossRef] [PubMed]
  53. Kampman, O.; Anttila, S.; Illi, A.; Mattila, K.M.; Rontu, R.; Leinonen, E.; Lehtimäki, T. Apolipoprotein E polymorphism is associated with age of onset in schizophrenia. J. Hum. Genet. 2004, 49, 355–359. [Google Scholar] [CrossRef] [PubMed]
  54. Xu, M.; Guo, Y.; Cheng, J.; Xue, K.; Yang, M.; Song, X.; Feng, Y.; Cheng, J. Brain iron assessment in patients with First-episode schizophrenia using quantitative susceptibility mapping. Neuroimage Clin. 2021, 31, 102736. [Google Scholar] [CrossRef] [PubMed]
  55. Sabbatinelli, J.; Prattichizzo, F.; Olivieri, F.; Procopio, A.D.; Rippo, M.R.; Giuliani, A. Where Metabolism Meets Senescence: Focus on Endothelial Cells. Front. Physiol. 2019, 10, 1523. [Google Scholar] [CrossRef] [PubMed]
  56. Pruett, B.S.; Meador-Woodruff, J.H. Evidence for altered energy metabolism, increased lactate, and decreased pH in schizophrenia brain: A focused review and meta-analysis of human postmortem and magnetic resonance spectroscopy studies. Schizophr. Res. 2020, 223, 29–42. [Google Scholar] [CrossRef] [PubMed]
  57. Chou, S.M.; Yen, Y.H.; Yuan, F.; Zhang, S.C.; Chong, C.M. Neuronal Senescence in the Aged Brain. Aging Dis. 2023, 14, 1618–1632. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  58. Murty, D.V.P.S.; Manikandan, K.; Kumar, W.S.; Ramesh, R.G.; Purokayastha, S.; Javali, M.; Rao, N.P.; Ray, S. Gamma oscillations weaken with age in healthy elderly in human EEG. Neuroimage 2020, 215, 116826. [Google Scholar] [CrossRef] [PubMed]
  59. Sharma, R. Emerging Interrelationship between the Gut Microbiome and Cellular Senescence in the Context of Aging and Disease: Perspectives and Therapeutic Opportunities. Probiotics Antimicrob. Proteins 2022, 14, 648–663. [Google Scholar] [CrossRef] [PubMed]
  60. Frey, N.; Venturelli, S.; Zender, L.; Bitzer, M. Cellular senescence in gastrointestinal diseases: From pathogenesis to therapeutics. Nat. Rev. Gastroenterol. Hepatol. 2018, 15, 81–95. [Google Scholar] [CrossRef]
  61. Maes, M.; Kanchanatawan, B.; Sirivichayakul, S.; Carvalho, A.F. In Schizophrenia, Increased Plasma IgM/IgA Responses to Gut Commensal Bacteria Are Associated with Negative Symptoms, Neurocognitive Impairments, and the Deficit Phenotype. Neurotox. Res. 2019, 35, 684–698. [Google Scholar] [CrossRef]
  62. Hennigar, S.R.; McClung, J.P. Nutritional Immunity: Starving Pathogens of Trace Minerals. Am. J. Lifestyle Med. 2016, 10, 170–173. [Google Scholar] [CrossRef] [PubMed]
  63. Pretorius, L.; Kell, D.B.; Pretorius, E. Iron Dysregulation and Dormant Microbes as Causative Agents for Impaired Blood Rheology and Pathological Clotting in Alzheimer’s Type Dementia. Front. Neurosci. 2018, 12, 851. [Google Scholar] [CrossRef] [PubMed]
  64. Link, C.D. Is There a Brain Microbiome? Neurosci. Insights 2021, 16, 26331055211018709. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  65. Dworkin, J.; Shah, I.M. Exit from dormancy in microbial organisms. Nat. Rev. Microbiol. 2010, 8, 890–896. [Google Scholar] [CrossRef]
  66. Peyrusson, F.; Nguyen, T.K.; Najdovski, T.; Van Bambeke, F. Host Cell Oxidative Stress Induces Dormant Staphylococcus aureus Persisters. Microbiol. Spectr. 2022, 10, e0231321. [Google Scholar] [CrossRef]
  67. Berthelot, J.M.; de la Cochetière, M.F.; Potel, G.; Le Goff, B.; Maugars, Y. Evidence supporting a role for dormant bacteria in the pathogenesis of spondylarthritis. Jt. Bone Spine 2013, 80, 135–140. [Google Scholar] [CrossRef] [PubMed]
  68. Sienkiewicz, M.; Sroka, K.; Binienda, A.; Jurk, D.; Fichna, J. A new face of old cells: An overview about the role of senescence and telomeres in inflammatory bowel diseases. Ageing Res. Rev. 2023, 91, 102083. [Google Scholar] [CrossRef]
  69. Qian, L.; He, X.; Gao, F.; Fan, Y.; Zhao, B.; Ma, Q.; Yan, B.; Wang, W.; Ma, X.; Yang, J. Estimation of the bidirectional relationship between schizophrenia and inflammatory bowel disease using the mendelian randomization approach. Schizophrenia 2022, 8, 31. [Google Scholar] [CrossRef]
  70. Bartocci, B.; Dal Buono, A.; Gabbiadini, R.; Busacca, A.; Quadarella, A.; Repici, A.; Mencaglia, E.; Gasparini, L.; Armuzzi, A. Mental Illnesses in Inflammatory Bowel Diseases: Mens sana in corpore sano. Medicina 2023, 59, 682. [Google Scholar] [CrossRef]
  71. Zevin, A.S.; McKinnon, L.; Burgener, A.; Klatt, N.R. Microbial translocation and microbiome dysbiosis in HIV-associated immune activation. Curr. Opin. HIV AIDS 2016, 11, 182–190. [Google Scholar] [CrossRef]
  72. Sfera, A.; Jafri, N.; Rahman, L. F-652 (Recombinant Human Interleukin-22) For Schizophrenia. Arch. Pharmacal Res. 2023, 3, 1–6. [Google Scholar]
  73. Schubert, K.O.; Föcking, M.; Cotter, D.R. Proteomic pathway analysis of the hippocampus in schizophrenia and bipolar affective disorder implicates 14-3-3 signaling, aryl hydrocarbon receptor signaling, and glucose metabolism: Potential roles in GABAergic interneuron pathology. Schizophr. Res. 2015, 167, 64–72. [Google Scholar] [CrossRef] [PubMed]
  74. Juricek, L.; Coumoul, X. The Aryl Hydrocarbon Receptor and the Nervous System. Int. J. Mol. Sci. 2018, 19, 2504. [Google Scholar] [CrossRef] [PubMed]
  75. Lopez-Otin, C.; Blasco, M.A.; Partridge, L.; Serrano, M.; Kroemer, G. The hallmarks of aging. Cell 2013, 153, 1194–1217. [Google Scholar] [CrossRef] [PubMed]
  76. Murray, I.; Nichols, R.; Zhang, L.; Patterson, A.D.; Perdew, G.H. Expression of the aryl hydrocarbon receptor contributes to the establishment of intestinal microbial community structure in mice. Sci. Rep. 2016, 6, 33969. [Google Scholar] [CrossRef] [PubMed]
  77. Park, H.; Jin, U.H.; Karki, K.; Jayaraman, A.; Allred, C.; Michelhaugh, S.K.; Mittal, S.; Chapkin, R.S.; Safe, S. Dopamine is an aryl hydrocarbon receptor agonist. Biochem. J. 2020, 477, 3899–3910. [Google Scholar] [CrossRef] [PubMed]
  78. Fehsel, K.; Schwanke, K.; Kappel, B.A.; Fahimi, E.; Meisenzahl-Lechner, E.; Esser, C.; Hemmrich, K.; Haarmann-Stemmann, T.; Kojda, G.; Lange-Asschenfeldt, C. Activation of the aryl hydrocarbon receptor by clozapine induces preadipocyte differentiation and contributes to endothelial dysfunction. J. Psychopharmacol. 2022, 36, 191–201. [Google Scholar] [CrossRef] [PubMed]
  79. Moura-Alves, P.; Faé, K.; Houthuys, E.; Dorhoi, A.; Kreuchwig, A.; Furkert, J.; Barison, N.; Diehl, A.; Munder, A.; Constant, P.; et al. AhR sensing of bacterial pigments regulates antibacterial defence. Nature 2014, 512, 387–392. [Google Scholar] [CrossRef]
  80. Bai, Z.; Yang, P.; Yu, F.; Li, Z.; Yao, Z.; Martinez, J.; Li, M.; Xu, H. Combining adoptive NK cell infusion with a dopamine-releasing peptide reduces senescent cells in aged mice. Cell Death Dis. 2022, 13, 305. [Google Scholar] [CrossRef]
  81. Yurchenko, O.P.; Grigoriev, N.G.; Turpaev, T.M.; Konjević, D.; Rakić, L. Intracellular injection of dopamine enhances acetylcholine responses of neuron R2 in the Aplysia abdominal ganglion. Comp. Biochem. Physiol. C Comp. Pharmacol. Toxicol. 1987, 87, 389–391. [Google Scholar] [CrossRef] [PubMed]
  82. Nakao, M.; Tanaka, H.; Koga, T. Cellular Senescence Variation by Metabolic and Epigenomic Remodeling. Trends Cell Biol. 2020, 30, 919–922. [Google Scholar] [CrossRef] [PubMed]
  83. Zhang, X.; Meng, F.; Lyu, W.; He, J.; Wei, R.; Du, Z.; Zhang, C. Histone lactylation antagonizes senescence and skeletal muscle aging via facilitating gene expression reprogramming. bioRxiv 2023. [Google Scholar] [CrossRef]
  84. Xie, Y.; Hu, H.; Liu, M.; Zhou, T.; Cheng, X.; Huang, W.; Cao, L. The role and mechanism of histone lactylation in health and diseases. Front. Genet. 2022, 13, 949252. [Google Scholar] [CrossRef] [PubMed]
  85. Wei, L.; Yang, X.; Wang, J.; Wang, Z.; Wang, Q.; Ding, Y.; Yu, A. H3K18 lactylation of senescent microglia potentiates brain aging and Alzheimer’s disease through the NFκB signaling pathway. J. Neuroinflamm. 2023, 20, 208. [Google Scholar] [CrossRef] [PubMed]
  86. Ng, P.Y.; McNeely, T.L.; Baker, D.J. Untangling senescent and damage-associated microglia in the aging and diseased brain. FEBS J. 2023, 290, 1326–1339. [Google Scholar] [CrossRef] [PubMed]
  87. Tay, T.L.; Béchade, C.; D‘Andrea, I.; St-Pierre, M.K.; Henry, M.S.; Roumier, A.; Tremblay, M.E. Microglia Gone Rogue: Impacts on Psychiatric Disorders across the Lifespan. Front. Mol. Neurosci. 2018, 10, 421. [Google Scholar] [CrossRef]
  88. Zhu, H.; Guan, A.; Liu, J.; Peng, L.; Zhang, Z.; Wang, S. Noteworthy perspectives on microglia in neuropsychiatric disorders. J. Neuroinflamm. 2023, 20, 223. [Google Scholar] [CrossRef]
  89. Galle, E.; Wong, C.W.; Ghosh, A.; Desgeorges, T.; Melrose, K.; Hinte, L.C.; Castellano-Castillo, D.; Engl, M.; de Sousa, J.A.; Ruiz-Ojeda, F.J.; et al. H3K18 lactylation marks tissue-specific active enhancers. Genome Biol. 2022, 23, 207. [Google Scholar] [CrossRef]
  90. Hagihara, H.; Shoji, H.; Otabi, H.; Toyoda, A.; Katoh, K.; Namihira, M.; Miyakawa, T. Protein lactylation induced by neural excitation. Cell Rep. 2021, 37, 109820. [Google Scholar] [CrossRef]
  91. Föcking, M.; Doyle, B.; Munawar, N.; Dillon, E.T.; Cotter, D.; Cagney, G. Epigenetic Factors in Schizophrenia: Mechanisms and Experimental Approaches. Mol. Neuropsychiatry 2019, 5, 6–12. [Google Scholar] [CrossRef]
  92. Miwa, S.; Kashyap, S.; Chini, E.; von Zglinicki, T. Mitochondrial dysfunction in cell senescence and aging. J. Clin. Investig. 2022, 132, e158447. [Google Scholar] [CrossRef] [PubMed]
  93. Wiley, C.D.; Campisi, J. From Ancient Pathways to Aging Cells-Connecting Metabolism and Cellular Senescence. Cell Metab. 2016, 23, 1013–1021. [Google Scholar] [CrossRef]
  94. Chen, A.N.; Luo, Y.; Yang, Y.H.; Fu, J.T.; Geng, X.M.; Shi, J.P.; Yang, J. Lactylation, a Novel Metabolic Reprogramming Code: Current Status and Prospects. Front. Immunol. 2021, 12, 688910. [Google Scholar] [CrossRef] [PubMed]
  95. Nishijo, M.; Tawara, K.; Nakagawa, H.; Honda, R.; Kido, T.; Nishijo, H.; Saito, S. 2,3,7,8-Tetrachlorodibenzo-p-dioxin in maternal breast milk newborn head circumference. J. Expo. Sci. Environ. Epidemiol. 2008, 18, 246–251. [Google Scholar] [CrossRef] [PubMed]
  96. Kinney, D.K.; Teixeira, P.; Hsu, D.; Napoleon, S.C.; Crowley, D.J.; Miller, A.; Hyman, W.; Huang, E. Relation of schizophrenia prevalence to latitude, climate, fish consumption, infant mortality, and skin color: A role for prenatal vitamin d deficiency and infections? Schizophr. Bull. 2009, 35, 582–595. [Google Scholar] [CrossRef] [PubMed]
  97. Howes, O.D.; Cummings, C.; Chapman, G.E.; Shatalina, E. Neuroimaging in schizophrenia: An overview of findings and their implications for synaptic changes. Neuropsychopharmacology 2022, 48, 151–167. [Google Scholar] [CrossRef] [PubMed]
  98. Leung, M.; Cheung, C.; Yu, K.; Yip, B.; Sham, P.; Li, Q.; Chua, S.; McAlonan, G. Gray Matter in First-Episode Schizophrenia Before and After Antipsychotic Drug Treatment. Anatomical Likelihood Estimation Meta-analyses with Sample Size Weighting. Schizophr. Bull. 2009, 37, 199–211. [Google Scholar] [CrossRef] [PubMed]
  99. Fusar-Poli, P.; Smieskova, R.; Kempton, M.; Ho, B.; Andreasen, N.; Borgwardt, S. Progressive brain changes in schizophrenia related to antipsychotic treatment? A meta-analysis of longitudinal MRI studies. Neurosci. Biobehav. Rev. 2013, 37, 1680–1691. [Google Scholar] [CrossRef] [PubMed]
  100. Veijola, J.; Guo, J.Y.; Moilanen, J.S.; Jääskeläinen, E.; Miettunen, J.; Kyllönen, M.; Haapea, M.; Huhtaniska, S.; Alaräisänen, A.; Mäki, P.; et al. Longitudinal changes in total brain volume in schizophrenia: Relation to symptom severity, cognition and antipsychotic medication. PLoS ONE. 2014, 18, e101689. [Google Scholar] [CrossRef]
  101. Cahn, W.; Pol, H.E.H.; Lems, E.B.; van Haren, N.E.; Schnack, H.G.; van der Linden, J.A.; Schothorst, P.F.; van Engeland, H.; Kahn, R.S. Brain volume changes in first-episode schizophrenia: A 1-year follow-up study. Arch. Gen. Psychiatry 2002, 59, 1002–1010. [Google Scholar] [CrossRef]
  102. Banwinkler, M.; Dzialas, V. Parkinson’s Progression Markers Initiative; Hoenig MC, van Eimeren T. Gray Matter Volume Loss in Proposed Brain-First and Body-First Parkinson’s Disease Subtypes. Mov Disord. 2022, 37, 2066–2074. [Google Scholar] [CrossRef] [PubMed]
  103. Chung, S.J.; Kim, Y.J.; Kim, Y.J.; Lee, H.S.; Jeong, S.H.; Hong, J.M.; Sohn, Y.H.; Yun, M.; Jeong, Y.; Lee, P.H. Association between White Matter Networks and the Pattern of Striatal Dopamine Depletion in Patients with Parkinson Disease. Neurology 2022, 99, e2672–e2682. [Google Scholar] [CrossRef]
  104. Dean, D.C., 3rd; Sojkova, J.; Hurley, S.; Kecskemeti, S.; Okonkwo, O.; Bendlin, B.B.; Theisen, F.; Johnson, S.C.; Alexander, A.L.; Gallagher, C.L. Alterations of Myelin Content in Parkinson’s Disease: A Cross-Sectional Neuroimaging Study. PLoS ONE 2016, 11, e0163774. [Google Scholar] [CrossRef]
  105. Brown, J.S., Jr. Effects of bisphenol-A and other endocrine disruptors compared with abnormalities of schizophrenia: An endocrine-disruption theory of schizophrenia. Schizophr. Bull. 2009, 35, 256–278. [Google Scholar] [CrossRef] [PubMed]
  106. Newbury, J.B.; Stewart, R.; Fisher, H.L.; Beevers, S.; Dajnak, D.; Broadbent, M.; Pritchard, M.; Shiode, N.; Heslin, M.; Hammoud, R.; et al. Association between air pollution exposure and mental health service use among individuals with first presentations of psychotic and mood disorders: Retrospective cohort study. Br. J. Psychiatry 2021, 219, 678–685. [Google Scholar] [CrossRef]
  107. Domínguez-Acosta, O.; Vega, L.; Estrada-Muñiz, E.; Rodríguez, M.S.; Gonzalez, F.J.; Elizondo, G. Activation of aryl hydrocarbon receptor regulates the LPS/IFNγ-induced inflammatory response by inducing ubiquitin-proteosomal and lysosomal degradation of RelA/p65. Biochem. Pharmacol. 2018, 155, 141–149. [Google Scholar] [CrossRef] [PubMed]
  108. Youngren, K.; Inglis, F.; Pivirotto, P.; Jedema, H.P.; Bradberry, C.W.; Goldman-Rakic, P.S.; Roth, R.H.; Moghaddam, B. Clozapine Preferentially Increases Dopamine Release in the Rhesus Monkey Prefrontal Cortex Compared with the Caudate Nucleus. Neuropsychopharmacology 1999, 20, 403–412. [Google Scholar] [CrossRef]
  109. Tronchin, G.; Akudjedu, T.N.; Ahmed, M.; Holleran, L.; Hallahan, B.; Cannon, D.M.; McDonald, C. Progressive subcortical volume loss in treatment-resistant schizophrenia patients after commencing clozapine treatment. Neuropsychopharmacology 2020, 45, 1353–1361, Erratum in Neuropsychopharmacology 2021, 46, 1857–1858. [Google Scholar] [CrossRef]
  110. van Erp, T.G.M.; Walton, E.; Hibar, D.P.; Schmaal, L.; Jiang, W.; Glahn, D.C.; Pearlson, G.D.; Yao, N.; Fukunaga, M.; Hashimoto, R.; et al. Cortical brain abnormalities in 4474 individuals with schizophrenia and 5098 control subjects via the enhancing neuro imaging genetics through meta analysis (ENIGMA) consortium. Biol. Psychiatry 2018, 84, 644–654. [Google Scholar] [CrossRef]
  111. Zhang, X.; Zhang, Y.; Liao, J.; Jiang, S.; Yan, J.; Yue, W.; Zhang, D.; Yan, H. Progressive Grey Matter Volume Changes in Patients with Schizophrenia over 6 Weeks of Antipsychotic Treatment and Their Relationship to Clinical Improvement. Neurosci. Bull. 2018, 34, 816–826. [Google Scholar] [CrossRef]
  112. Liu, N.; Xiao, Y.; Zhang, W.; Tang, B.; Zeng, J.; Hu, N.; Chandan, S.; Gong, Q.; Lui, S. Characteristics of gray matter alterations in never-treated and treated chronic schizophrenia patients. Transl. Psychiatry 2020, 10, 136. [Google Scholar] [CrossRef]
  113. Van Haren, N.E.; Hulshoff Pol, H.E.; Schnack, H.G.; Cahn, W.; Mandl, R.C.; Collins, D.L.; Evans, A.C.; Kahn, R.S. Focal gray matter changes in schizophrenia across the course of the illness: A 5-year follow-up study. Neuropsychopharmacology 2007, 32, 2057–2066. [Google Scholar] [CrossRef]
  114. Alamri, A.S.; Alhomrani, M.; Alsanie, W.F.; Alyami, H.; Shakya, S.; Habeeballah, H.; Abdulaziz, O.; Alamri, A.; Alkhatabi, H.A.; Felimban, R.I.; et al. Spectroscopic and Molecular Docking Analysis of π-Acceptor Complexes with the Drug Barbital. Appl. Sci. 2022, 12, 10130. [Google Scholar] [CrossRef]
  115. Refat, M.S.; Gaber, A.; Althobaiti, Y.S.; Alyami, H.; Alsanie, W.F.; Shakya, S.; Adam, A.M.A.; Kobeasy, M.I.; Asla, K.A. Spectroscopic and Molecular Docking Studies of Cu(II), Ni(II), Co(II), and Mn(II) Complexes with Anticonvulsant Therapeutic Agent Gabapentin. Molecules 2022, 27, 4311. [Google Scholar] [CrossRef]
  116. Hulshoff Pol, H.E.; Schnack, H.G.; Mandl, R.C.; van Haren, N.E.; Koning, H.; Collins, D.L.; Evans, A.C.; Kahn, R.S. Focal gray matter density changes in schizophrenia. Arch. Gen. Psychiatry 2001, 58, 1118–1125. [Google Scholar] [CrossRef]
  117. Winkler, T.E.; Lederer, S.L.; Kim, E.; Ben-Yoav, H.; Kelly, D.L.; Payne, G.F.; Ghodssi, R. Molecular processes in an electrochemical clozapine sensor. Biointerphases 2017, 12, 02B401. [Google Scholar] [CrossRef] [PubMed]
  118. Adam, A.M.A.; Saad, H.A.; Refat, M.S.; Hegab, M.S. Charge-transfer complexes of antipsychotic drug sulpiride with inorganic and organic acceptors generated through two different approaches: Spectral characterization. J. Mol. Liq. 2022, 357, 119092. [Google Scholar] [CrossRef]
  119. Chartoumpekis, D.V.; Zaravinos, A.; Apidianakis, Y.; Lagoumintzis, G. Editorial: Microbiota and mitochondria: Impact on cell signaling, physiology, and disease. Front. Microbiol. 2022, 13, 1056499. [Google Scholar] [CrossRef]
  120. Boguszewska, K.; Szewczuk, M.; Kaźmierczak-Barańska, J.; Karwowski, B.T. The Similarities between Human Mitochondria and Bacteria in the Context of Structure, Genome, and Base Excision Repair System. Molecules 2020, 25, 2857. [Google Scholar] [CrossRef]
  121. Liesa, M.; Shirihai, O.S. Mitochondrial dynamics in the regulation of nutrient utilization and energy expenditure. Cell Metab. 2013, 17, 491–506. [Google Scholar] [CrossRef]
  122. Johnson, E.L.; Heaver, S.L.; Waters, J.L.; Kim, B.I.; Bretin, A.; Goodman, A.L.; Gewirtz, A.T.; Worgall, T.S.; Ley, R.E. Sphingolipids produced by gut bacteria enter host metabolic pathways impacting ceramide levels. Nat. Commun. 2020, 11, 2471. [Google Scholar] [CrossRef]
  123. Venable, M.E.; Lee, J.Y.; Smyth, M.J.; Bielawska, A.; Obeid, L.M. Role of ceramide in cellular senescence. J. Biol. Chem. 1995, 270, 30701–30708. [Google Scholar] [CrossRef]
  124. Dadsena, S.; Bockelmann, S.; Mina, J.G.M.; Hassan, D.G.; Korneev, S.; Razzera, G.; Jahn, H.; Niekamp, P.; Müller, D.; Schneider, M.; et al. Ceramides bind VDAC2 to trigger mitochondrial apoptosis. Nat. Commun. 2019, 10, 1832. [Google Scholar] [CrossRef]
  125. Colombini, M. Ceramide channels and their role in mitochondria-mediated apoptosis. Biochim. Biophys. Acta 2010, 1797, 1239–1244. [Google Scholar] [CrossRef]
  126. Dutta, D.; Kanca, O.; Byeon, S.K.; Marcogliese, P.C.; Zuo, Z.; Shridharan, R.V.; Park, J.H.; Undiagnosed Diseases Network; Lin, G.; Ge, M.; et al. A defect in mitochondrial fatty acid synthesis impairs iron metabolism and causes elevated ceramide levels. Nat. Metab. 2023, 5, 1595–1614. [Google Scholar] [CrossRef]
  127. Zietzer, A.; Düsing, P.; Reese, L.; Nickenig, G.; Jansen, F. Ceramide Metabolism in Cardiovascular Disease: A Network with High Therapeutic Potential. Arterioscler. Thromb. Vasc. Biol. 2022, 42, 1220–1228. [Google Scholar] [CrossRef]
  128. Smesny, S.; Schmelzer, C.E.; Hinder, A.; Köhler, A.; Schneider, C.; Rudzok, M.; Schmidt, U.; Milleit, B.; Milleit, C.; Nenadic, I.; et al. Skin ceramide alterations in first-episode schizophrenia indicate abnormal sphingolipid metabolism. Schizophr. Bull. 2013, 39, 933–941. [Google Scholar] [CrossRef]
  129. Zhuo, C.; Zhao, F.; Tian, H.; Chen, J.; Li, Q.; Yang, L.; Ping, J.; Li, R.; Wang, L.; Xu, Y.; et al. Acid sphingomyelinase/ceramide system in schizophrenia: Implications for therapeutic intervention as a potential novel target. Transl. Psychiatry 2022, 12, 260. [Google Scholar] [CrossRef]
  130. Yuan, X.; Bhat, O.M.; Zou, Y.; Li, X.; Zhang, Y.; Li, P.L. Endothelial Acid Sphingomyelinase Promotes NLRP3 Inflammasome and Neointima Formation During Hypercholesterolemia. J. Lipid Res. 2022, 63, 100298. [Google Scholar] [CrossRef]
  131. Xia, Q.S.; Wu, F.; Wu, W.B.; Dong, H.; Huang, Z.Y.; Xu, L.; Lu, F.E.; Gong, J. Berberine reduces hepatic ceramide levels to improve insulin resistance in HFD-fed mice by inhibiting HIF-2α. Biomed. Pharmacother. 2022, 150, 112955. [Google Scholar] [CrossRef]
  132. Torralba, D.; Baixauli, F.; Sánchez-Madrid, F. Mitochondria know no boundaries: Mechanisms and functions of intercellular mitochondrial transfer. Front. Cell. Dev. Biol. 2016, 4, 107. [Google Scholar] [CrossRef] [PubMed]
  133. Fairley, L.H.; Grimm, A.; Eckert, A. Mitochondria transfer in brain injury and disease. Cells 2022, 11, 3603. [Google Scholar] [CrossRef]
  134. Jackson, J.G.; Robinson, M.B. Regulation of mitochondrial dynamics in astrocytes: Mechanisms, consequences, and unknowns. Glia 2018, 66, 1213–1234. [Google Scholar] [CrossRef] [PubMed]
  135. Hogan, D.B.; Jetté, N.; Fiest, K.M.; Roberts, J.I.; Pearson, D.; Smith, E.E.; Roach, P.; Kirk, A.; Pringsheim, T.; Maxwell, C.J. The Prevalence and Incidence of Frontotemporal Dementia: A Systematic Review. Can. J. Neurol. Sci. 2016, 43 (Suppl. S1), S96–S109. [Google Scholar] [CrossRef] [PubMed]
  136. Course, M.M.; Wang, X. Transporting mitochondria in neurons. F1000Research 2016, 5, 1735. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  137. Nuñez, M.T.; Chana-Cuevas, P. New Perspectives in Iron Chelation Therapy for the Treatment of Neurodegenerative Diseases. Pharmaceuticals 2018, 11, 109. [Google Scholar] [CrossRef] [PubMed]
  138. Thayyullathil, F.; Cheratta, A.R.; Alakkal, A.; Subburayan, K.; Pallichankandy, S.; Hannun, Y.A.; Galadari, S. Acid sphingomyelinase-dependent autophagic degradation of GPX4 is critical for the execution of ferroptosis. Cell Death Dis. 2021, 12, 26. [Google Scholar] [CrossRef] [PubMed]
  139. Abdel-Salam, O.M.E.; Morsy, S.M.Y.; Sleem, A.A. The effect of different antidepressant drugs on oxidative stress after lipopolysaccharide administration in mice. EXCLI J. 2011, 10, 290–302. [Google Scholar] [PubMed]
  140. Kann, O. The energy demand of fast neuronal network oscillations: Insights from brain slice preparations. Front. Pharmacol. 2012, 2, 90. [Google Scholar] [CrossRef]
  141. Nakao, K.; Singh, M.; Sapkota, K.; Hagler, B.C.; Hunter, R.N.; Raman, C.; Hablitz, J.J.; Nakazawa, K. GSK3β inhibition restores cortical gamma oscillation and cognitive behavior in a mouse model of NMDA receptor hypofunction relevant to schizophrenia. Neuropsychopharmacology 2020, 45, 2207–2218. [Google Scholar] [CrossRef]
  142. Linseman, D.A.; Butts, B.D.; Precht, T.A.; Phelps, R.A.; Le, S.S.; Laessig, T.A.; Bouchard, R.J.; Florez-McClure, M.L.; Heidenreich, K.A. Glycogen synthase kinase-3beta phosphorylates Bax and promotes its mitochondrial localization during neuronal apoptosis. J. Neurosci. 2004, 24, 9993–10002. [Google Scholar] [CrossRef] [PubMed]
  143. McNally, J.M.; McCarley, R.W. Gamma band oscillations: A key to understanding schizophrenia symptoms and neural circuit abnormalities. Curr. Opin. Psychiatry 2016, 29, 202–210. [Google Scholar] [CrossRef]
  144. Tada, M.; Kirihara, K.; Koshiyama, D.; Nagai, T.; Fujiouka, M.; Usui, K.; Satomura, Y.; Koike, S.; Sawada, K.; Matsuoka, J.; et al. Alterations of auditory-evoked gamma oscillations are more pronounced than alterations of spontaneous power of gamma oscillation in early stages of schizophrenia. Transl. Psychiatry 2023, 13, 218. [Google Scholar] [CrossRef]
  145. Williams, S.; Boksa, P. Gamma oscillations and schizophrenia. J. Psychiatry Neurosci. 2010, 35, 75–77. [Google Scholar] [CrossRef]
  146. Veit, J.; Handy, G.; Mossing, D.P.; Doiron, B.; Adesnik, H. Cortical VIP neurons locally control the gain but globally control the coherence of gamma band rhythms. Neuron 2023, 111, 405–417.e5. [Google Scholar] [CrossRef] [PubMed]
  147. Antonoudiou, P.; Tan, Y.L.; Kontou, G.; Upton, A.L.; Mann, E.O. Parvalbumin and Somatostatin Interneurons Contribute to the Generation of Hippocampal Gamma Oscillations. J. Neurosci. 2020, 40, 7668–7687. [Google Scholar] [CrossRef]
  148. Betterton, R.; Mellor, J.; Tsaneva-Atanasova, K. Modulation of hippocampal gamma oscillations by acetylcholine: Insights from mathematical and in vitro optogenetic models. BMC Neurosci. 2015, 16 (Suppl. S1), P267. [Google Scholar] [CrossRef]
  149. Çalışkan, G.; French, T.; Enrile Lacalle, S.; Del Angel, M.; Steffen, J.; Heimesaat, M.M.; Rita Dunay, I.; Stork, O. Antibiotic-induced gut dysbiosis leads to activation of microglia and impairment of cholinergic gamma oscillations in the hippocampus. Brain Behav. Immun. 2022, 99, 203–217. [Google Scholar] [CrossRef]
  150. Chen, Y.; Xu, J.; Chen, Y. Regulation of Neurotransmitters by the Gut Microbiota and Effects on Cognition in Neurological Disorders. Nutrients 2021, 13, 2099. [Google Scholar] [CrossRef]
  151. Klaver, R.; De Vries, H.E.; Schenk, G.J.; Geurts, J.J. Grey matter damage in multiple sclerosis: A pathology perspective. Prion 2013, 7, 66–75. [Google Scholar] [CrossRef]
  152. Keo, A.; Dzyubachyk, O.; van der Grond, J.; Hafkemeijer, A.; van de Berg, W.D.J.; van Hilten, J.J.; Reinders, M.J.T.; Mahfouz, A. Cingulate networks associated with gray matter loss in Parkinson’s disease show high expression of cholinergic genes in the healthy brain. Eur. J. Neurosci. 2021, 53, 3727–3739. [Google Scholar] [CrossRef]
  153. Yohn, S.E.; Weiden, P.J.; Felder, C.C.; Stahl, S.M. Muscarinic acetylcholine receptors for psychotic disorders: Bench-side to clinic. Trends Pharmacol. Sci. 2022, 43, 1098–1112. [Google Scholar] [CrossRef]
  154. Sahu, P.P.; Tseng, P. Gamma sensory entrainment for cognitive improvement in neurodegenerative diseases: Opportunities and challenges ahead. Front. Integr. Neurosci. 2023, 17, 1146687. [Google Scholar] [CrossRef]
  155. Yan, L.; Li, H.; Qian, Y.; Zhang, J.; Cong, S.; Zhang, X.; Wu, L.; Wang, Y.; Wang, M.; Yu, T. Transcutaneous vagus nerve stimulation: A new strategy for Alzheimer’s disease intervention through the brain-gut-microbiota axis? Front. Aging Neurosci. 2024, 16, 1334887. [Google Scholar] [CrossRef]
  156. Karpiński, P.; Żebrowska-Różańska, P.; Kujawa, D.; Łaczmański, Ł.; Samochowiec, J.; Jabłoński, M.; Plichta, P.; Piotrowski, P.; Bielawski, T.; Misiak, B. Gut microbiota alterations in schizophrenia might be related to stress exposure: Findings from the machine learning analysis. Psychoneuroendocrinology 2023, 155, 106335. [Google Scholar] [CrossRef]
  157. Attademo, L.; Bernardini, F.; Garinella, R.; Compton, M.T. Environmental pollution and risk of psychotic disorders: A review of the science to date. Schizophr. Res. 2017, 181, 55–59. [Google Scholar] [CrossRef] [PubMed]
  158. Breno SDiniz, B.S.; Seitz-Holland, J.; Sehgal, R.; Kasamoto, J.; Higgins-Chen, A.T.; Lenze, E. Geroscience-Centric Perspective for Geriatric Psychiatry: Integrating Aging Biology with Geriatric Mental Health Research. Geriatr. Psychiatry 2024, 32, 1–16. [Google Scholar]
  159. Seeman, M.V. Subjective Overview of Accelerated Aging in Schizophrenia. Int. J. Environ. Res. Public Health 2023, 20, 737. [Google Scholar] [CrossRef] [PubMed]
  160. Marin, I.; Serrano, M.; Pietrocola, F. Recent insights into the crosstalk between senescent cells and CD8 T lymphocytes. NPJ Aging 2023, 9, 8. [Google Scholar] [CrossRef]
  161. Harris, M.J.; Jeste, D.V.; Gleghorn, A.; Sewell, D.D. New-onset psychosis in HIV-infected patients. J. Clin. Psychiatry 1991, 52, 369–376. [Google Scholar]
  162. Kozato, N.; Mishra, M.; Firdosi, M. New-onset psychosis due to COVID-19. BMJ Case Rep. 2021, 14, e242538. [Google Scholar] [CrossRef] [PubMed]
  163. Luís, C.; Maduro, A.T.; Pereira, P.; Mendes, J.J.; Soares, R.; Ramalho, R. Nutritional senolytics and senomorphics: Implications to immune cells metabolism and aging—From theory to practice. Front. Nutr. 2022, 9, 958563. [Google Scholar] [CrossRef]
  164. An, S.; Cho, S.Y.; Kang, J.; Lee, S.; Kim, H.S.; Min, D.J.; Son, E.; Cho, K.H. Inhibition of 3-phosphoinositide-dependent protein kinase 1 (PDK1) can revert cellular senescence in human dermal fibroblasts. Proc. Natl. Acad. Sci. USA 2020, 117, 31535–31546. [Google Scholar] [CrossRef] [PubMed]
  165. Solis, G.M.; Kardakaris, R.; Valentine, E.R.; Bar-Peled, L.; Chen, A.L.; Blewett, M.M.; McCormick, M.A.; Williamson, J.R.; Kennedy, B.; Cravatt, B.F.; et al. Translation attenuation by minocycline enhances longevity and proteostasis in old post-stress-responsive organisms. eLife 2018, 7, e40314. [Google Scholar] [CrossRef]
  166. Deakin, B.; Suckling, J.; Dazzan, P.; Joyce, E.; Lawrie, S.M.; Upthegrove, R.; Husain, N.; Chaudhry, I.B.; Dunn, G.; Jones, P.B.; et al. Minocycline for Negative Symptoms of Schizophrenia and Possible Mechanistic Actions: The BeneMin RCT; NIHR Journals Library: Southampton, UK, 2019. [Google Scholar]
  167. Abir, M.H.; Mahamud, A.G.M.S.U.; Tonny, S.H.; Anu, M.S.; Hossain, K.H.S.; Protic, I.A.; Khan, M.S.U.; Baroi, A.; Moni, A.; Uddin, M.J. Pharmacological potentials of lycopene against aging and aging-related disorders: A review. Food Sci. Nutr. 2023, 11, 5701–5735. [Google Scholar] [CrossRef] [PubMed]
  168. Perrott, K.M.; Wiley, C.D.; Desprez, P.Y.; Campisi, J. Apigenin suppresses the senescence-associated secretory phenotype and paracrine effects on breast cancer cells. Geroscience 2017, 39, 161–173. [Google Scholar] [CrossRef] [PubMed]
  169. Elsallabi, O.; Patruno, A.; Pesce, M.; Cataldi, A.; Carradori, S.; Gallorini, M. Fisetin as a Senotherapeutic Agent: Biopharmaceutical Properties and Crosstalk between Cell Senescence and Neuroprotection. Molecules 2022, 27, 738. [Google Scholar] [CrossRef] [PubMed]
  170. Kumar, R.; Sharma, A.; Kumari, A.; Gulati, A.; Padwad, Y.; Sharma, R. Epigallocatechin gallate suppresses premature senescence of preadipocytes by inhibition of PI3K/Akt/mTOR pathway and induces senescent cell death by regulation of Bax/Bcl-2 pathway. Biogerontology 2019, 20, 171–189. [Google Scholar] [CrossRef]
  171. Li, W.; He, Y.; Zhang, R.; Zheng, G.; Zhou, D. The curcumin analog EF24 is a novel senolytic agent. Aging 2019, 11, 771–782. [Google Scholar] [CrossRef]
  172. Dang, Y.; An, Y.; He, J.; Huang, B.; Zhu, J.; Gao, M.; Zhang, S.; Wang, X.; Yang, B.; Xie, Z. Berberine ameliorates cellular senescence and extends the lifespan of mice via regulating p16 and cyclin protein expression. Aging Cell 2020, 19, e13060. [Google Scholar] [CrossRef]
  173. Islam, M.T.; Tuday, E.; Allen, S.; Kim, J.; Trott, D.W.; Holland, W.L.; Donato, A.J.; Lesniewski, L.A. Senolytic drugs, dasatinib and quercetin, attenuate adipose tissue inflammation, and ameliorate metabolic function in old age. Aging Cell 2023, 22, e13767. [Google Scholar] [CrossRef] [PubMed]
  174. von Kobbe, C. Targeting senescent cells: Approaches, opportunities, challenges. Aging 2019, 11, 12844–12861. [Google Scholar] [CrossRef] [PubMed]
  175. Suda, M.; Shimizu, I.; Katsuumi, G.; Yoshida, Y.; Hayashi, Y.; Ikegami, R.; Matsumoto, N.; Yoshida, Y.; Mikawa, R.; Katayama, A.; et al. Senolytic vaccination improves normal and pathological age-related phenotypes and increases lifespan in progeroid mice. Nat. Aging 2021, 1, 1117–1126. [Google Scholar] [CrossRef] [PubMed]
  176. Poblocka, M.; Bassey, A.L.; Smith, V.M.; Falcicchio, M.; Manso, A.S.; Althubiti, M.; Sheng, X.; Kyle, A.; Barber, R.; Frigerio, M.; et al. Targeted clearance of senescent cells using an antibody-drug conjugate against a specific membrane marker. Sci. Rep. 2021, 11, 20358. [Google Scholar] [CrossRef] [PubMed]
  177. Nicolson, G.L.; Ferreira de Mattos, G.; Ash, M.; Settineri, R.; Escribá, P.V. Fundamentals of Membrane Lipid Replacement: A Natural Medicine Approach to Repairing Cellular Membranes and Reducing Fatigue, Pain, and Other Symptoms While Restoring Function in Chronic Illnesses and Aging. Membranes 2021, 11, 944. [Google Scholar] [CrossRef] [PubMed]
  178. Zhou, M.; Ren, H.; Han, J.; Wang, W.; Zheng, Q.; Wang, D. Protective Effects of Kaempferol against Myocardial Ischemia/Reperfusion Injury in Isolated Rat Heart via Antioxidant Activity and Inhibition of Glycogen Synthase Kinase-3β. Oxid. Med. Cell. Longev. 2015, 2015, 481405. [Google Scholar] [CrossRef] [PubMed]
  179. Jope, R.S.; Roh, M.S. Glycogen synthase kinase-3 (GSK3) in psychiatric diseases and therapeutic interventions. Curr. Drug Targets 2006, 7, 1421–1434. [Google Scholar] [CrossRef] [PubMed]
  180. Jin, S.; Zhang, L.; Wang, L. Kaempferol, a potential neuroprotective agent in neurodegenerative diseases: From chemistry to medicine. Biomed. Pharmacother. 2023, 165, 115215. [Google Scholar] [CrossRef]
  181. Nicolson, G.L.; Ash, M.E. Membrane Lipid Replacement for chronic illnesses, aging and cancer using oral glycerolphospholipid formulations with fructooligosaccharides to restore phospholipid function in cellular membranes, organelles, cells and tissues. Biochim. Biophys. Acta Biomembr. 2017, 1859 Pt B, 1704–1724. [Google Scholar] [CrossRef]
  182. Nadeem, A.; Meijler, M.M. Unraveling the Antibacterial and Iron Chelating Activity of N-Oxide Hydroxy-Phenazine natural Products and Synthetic Analogs against Staphylococcus aureus. Isr. J. Chem. 2023, 63, 5–6. [Google Scholar] [CrossRef]
  183. Heitmann, A.S.B.; Zanjani, A.A.H.; Klenow, M.B.; Mularski, A.; Sønder, S.L.; Lund, F.W.; Boye, T.L.; Dias, C.; Bendix, P.M.; Simonsen, A.C.; et al. Phenothiazines alter plasma membrane properties and sensitize cancer cells to injury by inhibiting annexin-mediated repair. J. Biol. Chem. 2021, 297, 101012. [Google Scholar] [CrossRef] [PubMed]
  184. Schmidt, N.W.; Mishra, A.; Lai, G.H.; Davis, M.; Sanders, L.K.; Dat, T.; Garcia, A.; Tai, K.P.; McCray, J.; Paul, B.; et al. Criterion for amino acid composition of defensins and antimicrobial peptides based on geometry of membrane destabilization. J. Am. Chem. Soc. 2011, 133, 6720–6727. [Google Scholar] [CrossRef] [PubMed]
  185. Blankenfeldt, W.; Parsons, J.F. The structural biology of phenazine biosynthesis. Curr. Opin. Struct. Biol. 2014, 29, 26–33. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  186. Pierson, L.S., 3rd; Pierson, E.A. Metabolism and function of phenazines in bacteria: Impacts on the behavior of bacteria in the environment and biotechnological processes. Appl. Microbiol. Biotechnol. 2010, 86, 1659–1670. [Google Scholar] [CrossRef] [PubMed]
  187. Abdelaziz, A.A.; Kamer, A.M.A.; Al-Monofy, K.B.; Al-Madboly, L.A. Pseudomonas aeruginosa’s greenish-blue pigment pyocyanin: Its production and biological activities. Microb. Cell Fact. 2023, 22, 110. [Google Scholar] [CrossRef] [PubMed]
  188. Ohlendorf, B.; Schulz, D.; Erhard, A.; Nagel, K.; Imhoff, J.F. Geranylphenazinediol, an acetylcholinesterase inhibitor produced by a Streptomyces species. J. Nat. Prod. 2012, 75, 1400–1404. [Google Scholar] [CrossRef] [PubMed]
  189. Paul, S.M.; Yohn, S.E.; Popiolek, M.; Miller, A.C.; Felder, C.C. Muscarinic Acetylcholine Receptor Agonists as Novel Treatments for Schizophrenia. Am. J. Psychiatry 2022, 179, 611–627. [Google Scholar] [CrossRef] [PubMed]
  190. Wang, X.; Abbas, M.; Zhang, Y.; Elshahawi, S.I.; Ponomareva, L.V.; Cui, Z.; Van Lanen, S.G.; Sajid, I.; Voss, S.R.; Shaaban, K.A.; et al. Divergent Fused Phenazine-Based Metabolites from a Himalayan Streptomyces. J. Nat. Prod. 2019, 82, 1686–1693. [Google Scholar] [CrossRef]
  191. Cha, J.W.; Lee, S.; Kim, M.C.; Thida, M.; Lee, J.W.; Park, J.S.; Kwon, H.C. Pontemazines a and B, phenazine derivatives containing a methylamine linkage from Streptomyces sp. UT1123 and their protective effect to HT-22 neuronal cells. Bioorganic Med. Chem. Lett. 2015, 25, 5083–5086. [Google Scholar] [CrossRef]
  192. Kim, W.G.; Ryoo, I.J.; Yun, B.S.; Shin-ya, K.; Seto, H.; Yoo, I.D. Phenazostatin C, a new diphenazine with neuronal cell protecting activity from Streptomyces sp. J. Antibiot. 1999, 52, 758–761. [Google Scholar] [CrossRef]
  193. Kato, S.; Shindo, K.; Yamagishi, Y.; Matsuoka, M.; Kawai, H.; Mochizuki, J. Phenazoviridin, a novel free radical scavenger from Streptomyces sp. taxonomy, fermentation, isolation, structure elucidation and biological properties. J. Antibiot. 1993, 46, 1485–1493. [Google Scholar] [CrossRef] [PubMed]
  194. Boonnoy, P.; Jarerattanachat, V.; Karttunen, M.; Wong-ekkabut, J. Bilayer Deformation, Pores, and Micellation Induced by Oxidized Lipids. J. Phys. Chem. Lett. 2015, 6, 4884–4888. [Google Scholar] [CrossRef] [PubMed]
  195. Voronova, O.; Zhuravkov, S.; Korotkova, E.; Artamonov, A.; Plotnikov, E. Antioxidant Properties of New Phenothiazine Derivatives. Antioxidants 2022, 11, 1371. [Google Scholar] [CrossRef]
  196. Keynes, R.G.; Karchevskaya, A.; Riddall, D.; Griffiths, C.H.; Bellamy, T.C.; Chan, A.W.E.; Selwood, D.L.; Garthwaite, J. N10-carbonyl-substituted phenothiazines inhibiting lipid peroxidation and associated nitric oxide consumption powerfully protect brain tissue against oxidative stress. Chem. Biol. Drug Des. 2019, 94, 1680–1693. [Google Scholar] [CrossRef] [PubMed]
  197. Philot, E.A.; da Mata Lopes, D.; de Souza, A.T.; Braz, A.S.; Nantes, I.L.; Rodrigues, T.; Perahia, D.; Miteva, M.A.; Scott, L.P. Binding of phenothiazines into allosteric hydrophobic pocket of human thioredoxin 1. Eur. Biophys. J. 2016, 45, 279–286. [Google Scholar] [CrossRef] [PubMed]
  198. Ho, B.C.; Andreasen, N.C.; Ziebell, S.; Pierson, S.; Magnotta, V. Long-term antipsychotic treatment and brain volumes: A longitudinal study of first-episode schizophrenia. Arch. Gen. Psychiatry 2011, 68, 128–137. [Google Scholar] [CrossRef] [PubMed]
  199. Engwa, G.A.; Ayuk, E.L.; Igbojekwe, B.U.; Unaegbu, M. Potential Antioxidant Activity of New Tetracyclic and Pentacyclic Nonlinear Phenothiazine Derivatives. Biochem. Res. Int. 2016, 2016, 9896575. [Google Scholar] [CrossRef] [PubMed]
  200. Clark, M.A.; Shay, J.W. Mitochondrial transformation of mammalian cells. Nature 1982, 295, 605–607. [Google Scholar] [CrossRef] [PubMed]
  201. Katrangi, E.; D’Souza, G.; Boddapati, S.V.; Kulawiec, M.; Singh, K.K.; Bigger, B.; Weissig, V. Xenogenic transfer of isolated murine mitochondria into human rho0 cells can improve respiratory function. Rejuvenation Res. 2007, 10, 561–570. [Google Scholar] [CrossRef]
  202. Pacak, A.C.; Preble, J.M.; Kondo, H.; Seibel, P.; Levitsky, S.; del Nido, P.J.; Cowan, D.B.; McCully, J.D. Actin-dependent mitochondrial internalization in cardiomyocytes: Evidence for rescue of mitochondrial function. Biol. Open 2015, 4, 622–626. [Google Scholar] [CrossRef]
  203. Sheng, Z.H. Mitochondrial trafficking and anchoring in neurons: New insight and implications. J. Cell Biol. 2014, 204, 1087–1098. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  204. Ali Pour, P.; Hosseinian, S.; Kheradvar, A. Mitochondrial transplantation in cardiomyocytes: Foundation, methods, and outcomes. Am. J. Physiol. Cell Physiol. 2021, 321, C489–C503. [Google Scholar] [CrossRef] [PubMed]
  205. Sasaki, D.; Abe, J.; Takeda, A.; Harashima, H.; Yamada, Y. Transplantation of MITO cells, mitochondria activated cardiac progenitor cells, to the ischemic myocardium of mouse enhances the therapeutic effect. Sci. Rep. 2022, 12, 4344. [Google Scholar] [CrossRef] [PubMed]
  206. Chen, T.; Majerníková, N.; Marmolejo-Garza, A.; Trombetta-Lima, M.; Sabogal-Guáqueta, A.M.; Zhang, Y.; Ten Kate, R.; Zuidema, M.; Mulder, P.P.M.F.A.; den Dunnen, W.; et al. Mitochondrial transplantation rescues neuronal cells from ferroptosis. Free Radic. Biol. Med. 2023, 208, 62–72. [Google Scholar] [CrossRef] [PubMed]
  207. Um, J.-H.; Lee, K.-M.; Kim, Y.-Y.; Lee, D.-Y.; Kim, E.; Kim, D.-H.; Yun, J. Berberine Induces Mitophagy through Adenosine Monophosphate-Activated Protein Kinase and Ameliorates Mitochondrial Dysfunction in PINK1 Knockout Mouse Embryonic Fibroblasts. Int. J. Mol. Sci. 2024, 25, 219. [Google Scholar] [CrossRef] [PubMed]
  208. Hayakawa, K.; Esposito, E.; Wang, X.; Terasaki, Y.; Liu, Y.; Xing, C.; Ji, X.; Lo, E.H. Transfer of mitochondria from astrocytes to neurons after stroke. Nature 2016, 535, 551–555, Erratum in Nature 2016, 539, 123. [Google Scholar] [CrossRef] [PubMed]
  209. Goya-Jorge, E.; Jorge Rodríguez, M.E.; Veitía, M.S.; Giner, R.M. Plant Occurring Flavonoids as Modulators of the Aryl Hydrocarbon Receptor. Molecules 2021, 26, 2315. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  210. Linck, V.M.; Ganzella, M.; Herrmann, A.P.; Okunji, C.O.; Souza, D.O.; Antonelli, M.C.; Elisabetsky, E. Original mechanisms of antipsychotic action by the indole alkaloid alstonine (Picralima nitida). Phytomedicine 2015, 22, 52–55. [Google Scholar] [CrossRef] [PubMed]
  211. Cordaro, M.; Cuzzocrea, S.; Crupi, R. An Update of Palmitoylethanolamide and Luteolin Effects in Preclinical and Clinical Studies of Neuroinflammatory Events. Antioxidants 2020, 9, 216. [Google Scholar] [CrossRef]
  212. McGovern, K.; Castro, A.C.; Cavanaugh, J.; Coma, S.; Walsh, M.; Tchaicha, J.; Syed, S.; Natarajan, P.; Manfredi, M.; Zhang, X.M.; et al. Discovery and Characterization of a Novel Aryl Hydrocarbon Receptor Inhibitor, IK-175, and Its Inhibitory Activity on Tumor Immune Suppression. Mol. Cancer Ther. 2022, 21, 1261–1272. [Google Scholar] [CrossRef]
  213. Zhang, S.; Qin, C.; Safe, S.H. Flavonoids as aryl hydrocarbon receptor agonists/antagonists: Effects of structure and cell context. Environ. Health Perspect. 2003, 111, 1877–1882. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  214. Avancini, D.; Testori, A.; Fresolone, L.; Andolfi, G.; Vuono, M.; Martinelli, V.; Santoni de Sio, F.R.; Gregori, S. Aryl hydrocarbon receptor activity downstream of IL-10 signaling is required to promote regulatory functions in human dendritic cells. Cell Rep. 2023, 42, 112193. [Google Scholar] [CrossRef] [PubMed]
  215. Monteleone, I.; Rizzo, A.; Sarra, M.; Sica, G.; Sileri, P.; Biancone, L.; MacDonald, T.T.; Pallone, F.; Monteleone, G. Aryl hydrocarbon receptor-induced signals up-regulate IL-22 production and inhibit inflammation in the gastrointestinal tract. Gastroenterology 2011, 141, 237–248.e1. [Google Scholar] [CrossRef] [PubMed]
  216. Anderson, G. Clozapine and the aryl hydrocarbon receptor. J. Psychopharmacol. 2022, 36, 516. [Google Scholar] [CrossRef] [PubMed]
  217. Korac, K.; Rajasekaran, D.; Sniegowski, T.; Schniers, B.K.; Ibrahim, A.F.; Bhutia, Y.D. Carbidopa, an activator of aryl hydrocarbon receptor, suppresses IDO1 expression in pancreatic cancer and decreases tumor growth. Biochem. J. 2022, 479, 1807–1824. [Google Scholar] [CrossRef] [PubMed]
  218. Alexander, D.L.; Ganem, L.G.; Fernandez-Salguero, P.; Gonzalez, F.; Jefcoate, C.R. Aryl-hydrocarbon receptor is an inhibitory regulator of lipid synthesis and of commitment to adipogenesis. J. Cell Sci. 1998, 111 Pt 22, 3311–3322. [Google Scholar] [CrossRef]
  219. Matsunawa, M.; Amano, Y.; Endo, K.; Uno, S.; Sakaki, T.; Yamada, S.; Makishima, M. The aryl hydrocarbon receptor activator benzo[a]pyrene enhances vitamin D3 catabolism in macrophages. Toxicol. Sci. 2009, 109, 50–58. [Google Scholar] [CrossRef]
  220. Manzella, C.R.; Ackerman, M.; Singhal, M.; Ticho, A.L.; Ceh, J.; Alrefai, W.A.; Saksena, S.; Dudeja, P.K.; Gill, R.K. Serotonin Modulates AhR Activation by Interfering with CYP1A1-Mediated Clearance of AhR Ligands. Cell. Physiol. Biochem. 2020, 54, 126–141. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  221. Guillon, C.; Meziani, M.; Abdelli, S.; Sigaudo Roussel, D.; Bonod, C.; Nosbaum, A. The aryl hydrocarbon receptor pathway plays a central role in the cutaneous response to pollutants. Eur. J. Dermatol. 2022, 32, 305–311. (In English) [Google Scholar] [CrossRef] [PubMed]
  222. Grycová, A.; Joo, H.; Maier, V.; Illés, P.; Vyhlídalová, B.; Poulíková, K.; Sládeková, L.; Nádvorník, P.; Vrzal, R.; Zemánková, L.; et al. Targeting the Aryl Hydrocarbon Receptor with Microbial Metabolite Mimics Alleviates Experimental Colitis in Mice. J. Med. Chem. 2022, 65, 6859–6868. [Google Scholar] [CrossRef]
  223. Kang, S.; Lee, A.G.; Im, S.; Oh, S.J.; Yoon, H.J.; Park, J.H.; Pak, Y.K. A Novel Aryl Hydrocarbon Receptor Antagonist HBU651 Ameliorates Peripheral and Hypothalamic Inflammation in High-Fat Diet-Induced Obese Mice. Int. J. Mol. Sci. 2022, 23, 14871. [Google Scholar] [CrossRef] [PubMed]
  224. Zai, W.; Chen, W.; Liu, H.; Ju, D. Therapeutic Opportunities of IL-22 in Non-Alcoholic Fatty Liver Disease: From Molecular Mechanisms to Clinical Applications. Biomedicines 2021, 9, 1912. [Google Scholar] [CrossRef] [PubMed]
  225. Sfera, A. Six Decades of Dopamine Hypothesis: Is Aryl Hydrocarbon Receptor the New D2? Reports 2023, 6, 36. [Google Scholar] [CrossRef]
  226. Shi, C.; Su, C.; Cen, L.; Han, L.; Tang, J.; Wang, Z.; Shi, X.; Ju, D.; Cao, Y.; Zhu, H. Vunakizumab-IL22, a Novel Fusion Protein, Promotes Intestinal Epithelial Repair and Protects against Gut Injury Induced by the Influenza Virus. Biomedicines 2023, 11, 1160. [Google Scholar] [CrossRef]
  227. Kim, C.J.; Nazli, A.; Rojas, O.L.; Chege, D.; Alidina, Z.; Huibner, S.; Mujib, S.; Benko, E.; Kovacs, C.; Shin, L.Y.; et al. A role for mucosal IL-22 production and Th22 cells in HIV-associated mucosal immunopathogenesis. Mucosal Immunol. 2012, 5, 670–680. [Google Scholar] [CrossRef]
  228. Nsairat, H.; Khater, D.; Odeh, F.; Al-Adaileh, F.; Al-Taher, S.; Jaber, A.M.; Alshaer, W.; Al Bawab, A.; Mubarak, M.S. Lipid nanostructures for targeting brain cancer. Heliyon 2021, 7, e07994. [Google Scholar] [CrossRef]
  229. Aldosari, B.N.; Alfagih, I.M.; Almurshedi, A.S. Lipid Nanoparticles as Delivery Systems for RNA-Based Vaccines. Pharmaceutics 2021, 13, 206. [Google Scholar] [CrossRef] [PubMed]
  230. Sun, D.; Lu, Z.R. Structure and Function of Cationic and Ionizable Lipids for Nucleic Acid Delivery. Pharm. Res. 2023, 40, 27–46. [Google Scholar] [CrossRef]
  231. Brannagan, T.H., 3rd; Berk, J.L.; Gillmore, J.D.; Maurer, M.S.; Waddington-Cruz, M.; Fontana, M.; Masri, A.; Obici, L.; Brambatti, M.; Baker, B.F.; et al. Liver-directed drugs for transthyretin-mediated amyloidosis. J. Peripher. Nerv. Syst. 2022, 27, 228–237. [Google Scholar] [CrossRef]
Figure 3. The AhR is represented in the cytosol and mitochondria (mitoAhR). Akt negatively phosphorylates GSK-3β, inhibiting its function. Toxic ceramides and iron activate GSK-3β, resulting in excessive mitochondrial ROS (mtROS) levels, which activate the mitoAhR, triggering organelle death. mtROS can also cause mitochondrial demise by activating cytochrome-C and caspase-3. The natural compounds berberine and kaempferol inhibit GSK-3β, averting organelle death.
Figure 3. The AhR is represented in the cytosol and mitochondria (mitoAhR). Akt negatively phosphorylates GSK-3β, inhibiting its function. Toxic ceramides and iron activate GSK-3β, resulting in excessive mitochondrial ROS (mtROS) levels, which activate the mitoAhR, triggering organelle death. mtROS can also cause mitochondrial demise by activating cytochrome-C and caspase-3. The natural compounds berberine and kaempferol inhibit GSK-3β, averting organelle death.
Ijms 25 05904 g003
Figure 4. The lipid bilayer of neuronal membranes is easily oxidated when intracellular iron is upregulated [182]. Oxysterols, including 7-ketocholesterol (a toxic oxide), and oxidated phospholipids alter the biophysical properties of cell membranes, disrupting neurotransmission [183]. In addition, oxidized lipids activate the AhR, triggering premature neuronal senescence. Phenazines, phenothiazines, and their derivatives intercalate themselves into the lipid bilayer, repairing the lipids in cellular and/or mitochondrial membranes.
Figure 4. The lipid bilayer of neuronal membranes is easily oxidated when intracellular iron is upregulated [182]. Oxysterols, including 7-ketocholesterol (a toxic oxide), and oxidated phospholipids alter the biophysical properties of cell membranes, disrupting neurotransmission [183]. In addition, oxidized lipids activate the AhR, triggering premature neuronal senescence. Phenazines, phenothiazines, and their derivatives intercalate themselves into the lipid bilayer, repairing the lipids in cellular and/or mitochondrial membranes.
Ijms 25 05904 g004
Figure 5. Phenazine vs. phenothiazine: similarities and differences.
Figure 5. Phenazine vs. phenothiazine: similarities and differences.
Ijms 25 05904 g005
Figure 6. Membrane lipid replacement (MLR) replenishes oxidized lipids from the plasma and mitochondrial membrane, such as oxysterols, ceramide, and oxidized phospholipids (OxPLs) with natural glycerophospholipids. Oxidized lipids inhibit AKT (by serine-9 phosphorylation), activating GSK-3β, an enzyme associated with SCZ, SLDs, and cancer (by p53 inhibition). Berberine and kaempferol inhibit GSK-3β activation by different mechanisms, generating beneficial effects. Ceramide activates GSK-3β by the dephosphorylation of serine-9.
Figure 6. Membrane lipid replacement (MLR) replenishes oxidized lipids from the plasma and mitochondrial membrane, such as oxysterols, ceramide, and oxidized phospholipids (OxPLs) with natural glycerophospholipids. Oxidized lipids inhibit AKT (by serine-9 phosphorylation), activating GSK-3β, an enzyme associated with SCZ, SLDs, and cancer (by p53 inhibition). Berberine and kaempferol inhibit GSK-3β activation by different mechanisms, generating beneficial effects. Ceramide activates GSK-3β by the dephosphorylation of serine-9.
Ijms 25 05904 g006
Figure 7. AhR agonists and antagonists relevant for neuropsychiatry [74,213,214,215,216,217,218,219,220,221,222].
Figure 7. AhR agonists and antagonists relevant for neuropsychiatry [74,213,214,215,216,217,218,219,220,221,222].
Ijms 25 05904 g007
Table 1. Natural senolytics and their sources.
Table 1. Natural senolytics and their sources.
SenolyticsSourcesReference
LycopeneGrape skin, guava, grapefruit, blueberries, and tomatoes[166]
ApigeninCabbage, blueberries, and acai berries[167]
FisetinStrawberries, onions, apples, mangoes, persimmons, and kiwis[168]
Curcumin and EF24 analogChicken, beef, tofu, and vegetables[169]
Epigallocatechin gallateApples, blackberries, broad beans, cherries, black grapes, pears, raspberries, and chocolate[170]
BerberineOregon grape, phellodendron, and tree turmeric[171]
QuercetinFruits, apples, onions, parsley, sage, tea, and red wine[172]
KaempferolFruits and vegetables[173]
Table 2. Naturally occurring and synthetic compounds with potential benefits for SCZ and SLDs.
Table 2. Naturally occurring and synthetic compounds with potential benefits for SCZ and SLDs.
CompoundNaturally Occurring Synthetic
PhenazinesGeranyl-phenazine and bara-phenazines A–GPontemazines A and B and
halogenated phenazines
Phenothiazines Propenyl-phenothiazine and
N10-carbonyl-substituted phenothiazines
GSK-3β inhibitorsKaempferol and curcuminLithium, valproic acid, clozapine, and olanzapine
AhR inhibitorsQuercetin, apigenin, alstonine, and luteolinIK-175 and HBU651
Acid sphingomyelinase (ASM) inhibitors Fluvoxamine, rosuvastatin, and tricyclic antidepressants
Dopamine D1R agonists A68930, A77636, and dihydrexidine
Mitochondrial export SSRIs
ACh agonistsCatharanthus roseus and
Salvia spp. (Lamiaceae)
Cholinesterase inhibitors: donepezil, galantamine, and rivastigmine
SenotherapeuticsPlease see Table 1Senotherapeutic antibiotics
Ferroptosis inhibitorsNatural flavonoids and
berberine
Fluvoxamine, SSRIs, and N acetyl cysteine (NAC)
Recombinant IL-22
Membrane lipid replacement
Mitochondrial transplantation
40 HZ entrainment with sensory stimuli
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

Sfera, A.; Imran, H.; Sfera, D.O.; Anton, J.J.; Kozlakidis, Z.; Hazan, S. Novel Insights into Psychosis and Antipsychotic Interventions: From Managing Symptoms to Improving Outcomes. Int. J. Mol. Sci. 2024, 25, 5904. https://doi.org/10.3390/ijms25115904

AMA Style

Sfera A, Imran H, Sfera DO, Anton JJ, Kozlakidis Z, Hazan S. Novel Insights into Psychosis and Antipsychotic Interventions: From Managing Symptoms to Improving Outcomes. International Journal of Molecular Sciences. 2024; 25(11):5904. https://doi.org/10.3390/ijms25115904

Chicago/Turabian Style

Sfera, Adonis, Hassan Imran, Dan O. Sfera, Jacob J. Anton, Zisis Kozlakidis, and Sabine Hazan. 2024. "Novel Insights into Psychosis and Antipsychotic Interventions: From Managing Symptoms to Improving Outcomes" International Journal of Molecular Sciences 25, no. 11: 5904. https://doi.org/10.3390/ijms25115904

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