Next Article in Journal
Prognostic Factors Affecting the Outcome of Surgical Root Canal Treatment—A Retrospective Cone-Beam Computed Tomography Cohort Study
Next Article in Special Issue
The Cyclical Battle of Insomnia and Mental Health Impairment in Firefighters: A Narrative Review
Previous Article in Journal
The Effect of the Direction of Primary Lateral Spinal Curvature on Postural Stability in Children with Scoliosis
Previous Article in Special Issue
Are Sleep Aids Associated with the Severity of Attention Deficit Hyperactivity Disorder Symptoms in Adults Screened for Insomnia? A Cross-Sectional Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Insomnia in Forensic Detainees: Is Salience Network the Common Pathway for Sleep, Neuropsychiatric, and Neurodegenerative Disorders?

by
Adonis Sfera
1,2,*,
Kyle A. Thomas
1,
Isaac A. Ogunjale
1,
Nyla Jafri
1 and
Peter G. Bota
2
1
Department of Psychiatry, Patton State Hospital, University of California, Riverside, CA 92521, USA
2
School of Medicine, California University of Science and Medicine, Colton, CA 92324, USA
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2024, 13(6), 1691; https://doi.org/10.3390/jcm13061691
Submission received: 24 February 2024 / Revised: 10 March 2024 / Accepted: 12 March 2024 / Published: 15 March 2024
(This article belongs to the Special Issue Effect of Long-Term Insomnia on Mental Health)

Abstract

:

Highlights

What are the main findings?
  • SN dysfunction is the common denominator of insomnia, schizophrenia (SCZ), and frontotemporal dementia behavioral variant (bvFTD).
What is the implication of the main findings?
  • The diagnosis of bvFTD is often missed or misdiagnosed in forensic institutions.
  • To ensure adequate placement and treatment planning, courts and clinicians require education to differentiate bvFTD from SCZ.

Abstract

Forensic hospitals throughout the country house individuals with severe mental illness and history of criminal violations. Insomnia affects 67.4% of hospitalized patients with chronic neuropsychiatric disorders, indicating that these conditions may hijack human somnogenic pathways. Conversely, somnolence is a common adverse effect of many antipsychotic drugs, further highlighting a common etiopathogenesis. Since the brain salience network is likely the common denominator for insomnia, neuropsychiatric and neurodegenerative disorders, here, we focus on the pathology of this neuronal assembly and its likely driver, the dysfunctional neuronal and mitochondrial membrane. We also discuss potential treatment strategies ranging from membrane lipid replacement to mitochondrial transplantation. The aims of this review are threefold: 1. Examining the causes of insomnia in forensic detainees with severe mental illness, as well as its role in predisposing them to neurodegenerative disorders. 2. Educating State hospital and prison clinicians on frontotemporal dementia behavioral variant, a condition increasingly diagnosed in older first offenders which is often missed due to the absence of memory impairment. 3. Introducing clinicians to natural compounds that are potentially beneficial for insomnia and severe mental illness.

Graphical Abstract

1. Introduction

One of the most common sleep disorders in the United States, primary insomnia, is usually defined as long sleep latency, difficulty staying asleep, prolonged nighttime wakefulness, and/or early morning awakening [1]. In prison, approximately 60% of inmates experience insomnia, a prevalence 6–10 times higher than in the population at large [2]. Moreover, insomnia is present in 67.4% of hospitalized patients with severe mental illness, suggesting that the pathways of sleep and neuropathology are highly intertwined [3].
Forensic psychiatric hospitals admit patients with schizophrenia (SCZ) or schizophrenia-like disorders (SLDs) and criminal violations. Insomnia is common in this population and failure to address this condition may increase healthcare expenditure due to medical complications, including metabolic, cardiovascular, and neurodegenerative disorders.
The salience network (SN), comprised of the anterior insular cortex (AIC), anterior cingulate cortex (ACC) and several subcortical nodes, has recently been implicated in the etiopathogenesis of insomnia, SCZ, and neurodegenerative disorders [4,5,6,7,8,9]. SN is comprised of Von Economo neurons (VENs), a special class of large, spindle-shaped cells found only in humans and superior mammals that are believed to drive empathy, social awareness, and emotional intelligence [10].
At the molecular level, incarceration, insomnia, and severe mental illness have been associated with premature cellular senescence, a phenotype marked by increased intracellular iron and mitochondrial damage [11,12,13,14,15,16,17,18]. Premature cellular senescence is driven by the aryl hydrocarbon receptor (AhR), expressed in neuronal cytosol and mitochondria [19,20,21]. Senescent cells upregulate intracellular iron which, in the proximity of cytosolic fats, increases the risk of lipid peroxidation and neuronal demise by ferroptosis [22,23,24]. Ferroptosis is a programmed cell death induced by iron in the context of antioxidant failure marked by the depletion of glutathione peroxidase-4 (GPX-4) [25,26]. GPX-4 is a mitochondrial enzyme which averts ferroptosis by repairing the oxidized phospholipids and cholesterol in mitochondrial and neuronal membranes [27]. Ferroptosis causes mitochondrial swelling, loss of cristae, dissipation of the membrane potential, as well as an increase in membrane permeability, changes that ultimately lead to mitochondrial loss [28]. Mitochondrial dysfunction and loss drive cellular senescence, a phenotype found in insomnia, severe mental illness and frontotemporal lobar degeneration (FTLD) [29,30,31,32]. In addition, insomnia, SCZ, and frontotemporal dementia (FTD) have been connected to impaired phagocytosis of senescent cells by natural killer cells (NKCs) [33,34,35]. Accumulation of senescent cells due to accelerated aging and impaired removal leads to inflammation, a pathology encountered in sleep deprivation, severe mental illness and FTD [36,37,38]. Since mitochondria is a key driver of inflammation, dysfunction or loss of these organelles likely account for these pathologies [39,40].
To compensate for dysfunctional mitochondria, neurons import these organelles from glial cells, especially the astrocyte [41,42]. In large cells, such as VENs, mitochondria are more vulnerable to damage and autophagic elimination as they undergo more wear and tear during their journey through the long axons of these neurons [42]. Due to their small number (around 193,000) and their large sizes, VENs are more susceptible to plasma membrane oxidative stress, which may trigger significant pathology even after a limited neuronal loss, a pathology encountered in frontotemporal dementia behavioral variant (bvFTD) [43].
Since mitochondria are crucial for neuronal function, preserving the integrity of these organelles via membrane lipid replacement (MLR) and other natural strategies is of utmost importance. Microbial phenazines and the novel antioxidant phenothiazine derivatives offer new opportunities to combat insomnia, psychosis, and neurodegeneration at the level of cell and mitochondrial membranes.

1.1. Salienve Network in Sleep and Neuropathology

The SN is comprised of ACC and AIC which, along with subcortical nodes in the hypothalamus, thalamus, striatum, and midbrain, process salient stimuli [44,45]. SN functions as a switch between exteroception and interoception or central executive network (CEN) and default mode network (DMN), depending on stimulus relevance [46]. Switching from CEN to DMN and vice versa is impaired in severe mental illness, insomnia, and neurodegenerative disorders [47]. Several antipsychotic drugs are known to lower the salience assignment to objects and events, likely restoring SN function, which, in turn, may ameliorate insomnia and psychosis [48].
The SN harbors VENs, which are large, corkscrew neurons located in layer V of the AIC and ACC. These non-telencephalic cells are believed to drive prosocial cognition, empathy, and emotional intelligence. As parts of the SN, VENs respond to endogenous or exogenous stimuli in the order of priority. VENs are selectively eliminated in bvFTD, a disorder marked by criminal violations, lack of empathy, poor insight, and sleep impairment [49,50,51,52,53]. In forensic institutions, bvFTD is increasingly diagnosed in older first offenders with no previous criminal history and often coexists with insomnia and altered eating habits.
Under physiological circumstances, sleep is driven by the ventrolateral preoptic nucleus (VLPO) of the anterior hypothalamus which releases inhibitory neurotransmitters, including γ-aminobutyric acid (GABA), and galanin [54]. The opposing system, orexin (hypocretin) neurons in the lateral hypothalamus, inhibits VLPO [55,56,57]. In addition, orexin/hypocretin neurons induce wakefulness by blocking melanin concentrating hormone (MCH), a somnogen released by the hypothalamus and zona incerta [58,59]. Orexin and DA, the key players of saliency, have been implicated in the neuropsychiatric disorders associated with sleep disturbances, including narcolepsy, attention-deficit/hyperactivity disorder (ADHD), and Parkinson’s disease (PD) [60]. Histamine is another wakefulness-promoting neurotransmitter implicated in SCZ and a novel target for treating negative and cognitive symptoms [61].
To better comprehend the pathogenesis of insomnia, it is necessary to study the pathways of wakefulness, a brain state driving self-awareness and probably consciousness [62]. Early studies on this subject have focused on the locus coeruleus, midbrain tegmentum, pons, and parabrachial nucleus, as neurons in these regions are active during wakefulness [63,64]. In the early 1900s, while studying encephalitis lethargica, Constantin von Economo found that lesions in the posterior hypothalamus were associated with sleep, hypothesizing that this area contained the “center of wakefulness” [65,66,67].
Fatal familial insomnia (FFI), a rare autosomal dominant disease, is marked by hypometabolism and neuronal loss in the thalamus and ACC, linking this condition to the SN [68,69,70,71,72]. The role of SN in sleep physiology and pathology is further highlighted by the anesthetics, especially propofol, which lower salience processing, inducing sleep [68,69,70,71,72,73,74,75,76,77,78]. Moreover, recent studies on sleep-deprived human volunteers and patients with primary insomnia demonstrated altered connectivity in AIC, further linking SN to sleep and wakefulness [79,80]. Furthermore, several preclinical studies are in line with the findings in humans, implicating the SN in slumber homeostasis [74,81].
Aside from insomnia and neuropsychiatric pathology, the SN connectivity is disrupted in neurodegenerative disorders, including Alzheimer’s disease (AD), Parkinson’s disease (PD), and bvFTD, suggesting that insomnia and neuropathology are highly intertwined [82,83,84,85,86]. Indeed, dysfunctional AIC and ACC connectivity may account for the criminal violations in patients with bvFTD, in which breaking the law may often be the initial dementia symptom [87,88].

1.2. Salience Network in Frontotemporal Dementia Behavioral Variant

The second most common neurodegenerative disorder after AD, bvFTD, is marked by inappropriate emotional responses and disinhibited behaviors, often leading to criminal violations, as this pathology targets VENs selectively [52,89]. In forensic institutions, individuals with first incarceration after the age of 55 may suffer from bvFTD, an entity difficult to diagnose as the memory may remain intact for longer periods of time. As a result, bvFTD is often missed or misdiagnosed as antisocial personality disorder (APD), SCZ, or even major depressive disorder [90].
Over the past two decades, the number of senior first offenders has grown in parallel with the prevalence of young-onset dementia (YOD, emergence of symptoms before age 65), a subgroup of neurodegenerative disorders, which may include bvFTD [91,92]. Indeed, recent studies have revealed that the prevalence of bvFTD has increased from 15/100,000 in 2013 to 119 per 100,000 in 2021, mirroring the growing number of forensic detainees with this diagnosis [92,93].
Compared to AD, in which 12% of patients exhibit criminal behavior, bvFTD is associated with a crime rate of 54%, suggesting an acquired psychopathy [94]. Frontotemporal lobar degeneration (FTLD), the pathology driving bvFTD, is associated with impulsivity and criminal violations due to the paucity of “honesty cells”, VENs [95]. The latter is likely due to the autophagy of damaged organelles traveling through the long VENs axons. Indeed, lysosomal aggregates, hallmarks of hyperactive autophagy, were demonstrated in the VENs derived from patients with bvFTD and SCZ, suggesting excessive mitophagy [95,96,97]. Depletion of VENs has been associated with a lack of empathy, aggressive behavior, and criminal violations documented in bvFTD and severe mental illness [51,52]. For example, homicide or attempted homicide have been documented in bvFTD, indicating that criminal behavior and murder can sometimes be the earliest manifestation of this disorder [98,99]. Since VENs are only present in large mammals, including humans, great apes, macaques, cetaceans, and elephants, but not in rodents, these cells are difficult to study in vivo [10]. VENs are larger than pyramidal neurons and drive interoceptive awareness, which is the ability to detect and process internal cues such as heartbeat, respiration and the overall visceral state [100,101]. VENs are components of the SN, an attention-shifting large neuronal assembly that can activate or silence CEN to DMN [102,103].
Recent transcriptomic studies found that VENs express monoaminergic proteins, including vesicular monoamine transporter 2 (VMAT2) and adrenergic receptor α-1A (ADRA1A), suggesting involvement in autonomic functions, including the circadian rhythm [104,105,106]. Indeed, impaired monoaminergic signaling has been documented in insomnia, bvFTD, SCZ, and SLDs, implicating VENs in these pathologies [107,108,109,110,111].

1.3. Sleep and Glial Cells

Astrocytes, the most numerous brain cells, communicate with each other via calcium waves, attaining synchronization with neurons and supporting slow-wave sleep [112,113]. Moreover, astrocytes release molecules, including adenosine, lactate, glutamate, GABA, and interleukin-1 (IL-1), which may indirectly influence the status of neuronal cells, inducing sleep [114].
Astrocytes are central to the neurovascular unit (NVU) and bridge the gap between the neuron and brain microvessels, regulating the flow of interstitial fluid through the aquaporin 4 (AQP-4) receptors [115] (Figure 1). The volume of the brain interstitial fluid (ISF) fluctuates in a circadian manner as it flows through the glymphatic system, a mechanism for clearing misfolded proteins during sleep [116]. The glymphatic system can also carry extracellular vesicles containing mitochondria from astrocytes to neurons [117]. Astrocytes support the neurons by generating GPX-4 to avert neuronal death by ferroptosis. GPX-4 functions to repair oxidized lipids and oxysterols, including 7-ketocholesterol (7KCl), toxins that disrupt plasma and mitochondrial membranes, triggering neuronal death [118]. Ferroptosis has been associated with sleep deprivation, indicating that neurons likely import GPX-4 during sleep [119]. As mitochondria play a key role in sleep homeostasis, insomnia may be the result of plasma or mitochondrial membrane oxidation. Indeed, it has been suggested that sleep is necessary for abrogating neuronal oxidative stress [120].
Intracellular iron is stored in ferritin and released for intracellular needs via ferritinophagy (ferritin autophagy) in lysosomes. Several antipsychotic drugs, including haloperidol, accumulate in lysosomes disrupting ferritinophagy, which, in turn, lowers intracellular iron, averting ferroptosis [121,122] (Figure 2). This may highlight a DA-independent, antipsychotic action of haloperidol, suggesting that dopaminergic blockade is not the only psychosis-deterring mechanism of this drug. Indeed, ferroptosis of hippocampal neurons, documented in AD and severe mental illness, is the likely cause of cognitive impairment and negative symptoms in these conditions [123,124]. Prolonged insomnia has been demonstrated to damage the astrocyte which, in turn, may trigger neuronal demise [125]. Moreover, chronic sleep loss was demonstrated to activate both astrocytes and microglia, turning these cells into neurotoxic phenotypes capable of eliminating healthy neurons and synapses [126,127,128].

2. Mitochondria and Aryl Hydrocarbon Receptor

Recent studies have implicated mitochondria in the pathophysiology of sleep and neurodegenerative disorders, while the role of these organelles in severe mental illness, including SCZ and SLDs, has been previously established [129,130]. AhR is the master regulator of cellular senescence, a phenotype conducive to aging and neurodegeneration and is expressed by the mitochondrion [19,20,21]. Oxidized lipids in the mitochondrial membrane are AhR ligands, which in conjunction with senescence-upregulated intracellular iron, can trigger ferroptosis and organelle demise [131,132,133,134]. Indeed, lipid peroxides and oxysterols, such as 7KCl, are mitoAhR ligands, contributing to mitochondrial dysfunction and autophagic elimination [135].
AhR is a xenobiotic sensor which regulates cytochrome p450 and binds the environmental toxin, dioxin (2,3,7,8-tetrachlorodibenzo-p-dioxin). Other AhR ligands include somnogens, such as phenazines, melatonin, and tryptophan derivatives, which participate in the physiology of sleep, wakefulness, and the circadian rhythm [136,137,138]. In addition, reactive oxygen species (ROS), known to induce sleep via a redox-sensitive potassium channel, are AhR ligands, bringing this transcription factor in the arena of slumber, mental illness, and neurodegeneration [131,139]. Indeed, microbial phenazines, including pyocyanin and 1-hydroxyphenazine, activate AhR, influencing the transcription of many genes, including those involved in sleep regulation [140,141].
The importance of mitochondria in sleep physiology is further substantiated by the organelle involvement in FFI, as well as in general anesthesia [142,143]. Indeed, general anesthetics are known to inhibit N-methyl-d-aspartate (NMDA) and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) glutamate receptors while stimulating GABA. NMDA and AMPA upregulate intracellular and mitochondrial calcium, inducing cell and organelle demise [144]. Interestingly, elevated mitochondrial calcium, a characteristic of prion diseases, may link these organelles to FFI [145,146]. Indeed, the prion peptide causes calcium inflow via L-type calcium channels, triggering neuronal damage and apoptosis [147]. In contrast, the typical antipsychotic, chlorpromazine, not only induces sleep, but also exerts anti-prion properties, probably by promoting autophagy of the misfolded protein [148,149,150].
Mitochondrial trafficking from astrocytes to neurons supports neuronal bioenergetic needs, especially in large pyramidal cells or VENs. Mitochondria can be imported via cell–cell fusion, tunneling nanotubes (cytoskeletal protrusions reaching to other cells), as well as transported by extracellular vesicles [151,152] (Figure 2). Moreover, astrocytes generate GPX-4 from cysteine obtained via the cystine/glutamate antiporter system (Xc−) or by transmethylation of methionine. Glutathione is generated from cysteine and glutathione disulfide (GSSC) [153] (Figure 2).
Mitochondrial trafficking as well as autophagy (mitophagy) occur during sleep, probably explaining the reason most living beings require rest [154]. Interestingly, serotonin (5-HT) promotes mitochondrial transport in hippocampal neurons, suggesting that antidepressant drugs, serotonin reuptake inhibitors (SSRIs), may “exert their action by supplying healthy mitochondria to stressed neurons [155]. This may imply that ROS accumulation during wakefulness may induce slumber to repair oxidized lipids and import mitochondria from glial cells [120,131,139]. In addition, the accumulation of intracellular microtubule-associated protein tau (MAPT) in VENs likely impairs mitochondrial transport, contributing to bvFTD pathogenesis [156].

2.1. Mitochondria-Protective Treatments

The key role of mitochondria in sleep disorders, SCZ, SLDs, and neurodegeneration, highlights the importance of mitoprotective approaches to resuscitate, replace, or increase the import of mitochondria from glial cells [157]. For example, treatment with SSRIs during the early stages of dementia may delay the onset of cognitive decline. Along this line, a recent study found that treatment with SSRIs slowed the conversion of mild cognitive impairment to frank dementia, suggesting that prophylactic treatment with these agents may be beneficial [158]. In addition, natural anti-ferroptosis drugs and iron chelators, such as halogenated phenazines, may improve the course of neurodegenerative disorders, suggesting novel therapeutic strategies [159,160].

2.2. Membrane Lipid Replacement (MLR)

MLR refers to the oral supplementation with natural cell membrane glycerophospholipids and kaempferol (3,4′,5,7-tetrahydroxyflavone), a natural flavonoid found in tea, broccoli, cabbage, kale, beans, endive, leek, tomato, strawberries, and grapes [161]. Kaempferol is a glycogen synthase kinase-3β (GSK-3β) inhibitor which prevents sleep deprivation-induced cognitive decline [162,163]. Like lithium and several antipsychotic drugs, kaempferol blocks GSK-3β, an enzyme previously implicated in SCZ and circadian rhythm disorders, suggesting that this natural compound may exert antipsychotic properties without the adverse effects of conventional therapeutics [164,165,166,167].
The aim of MLR + kaempferol is the gradual replacement of damaged phospholipids and oxysterols from neuronal and/or mitochondrial membranes with natural glycerophospholipids and a polyphenol. Indeed, oxidized membrane lipids have been implicated in SCZ, SLDs, insomnia, and neurodegeneration, while MLR and kaempferol offer a dual mechanism of action: (1) elimination of lipid peroxides and (2) GSK-3β inhibition [168]. Replacing oxidized plasma and/or mitochondrial membrane fats with healthy natural lipids averts deformation of the neuronal membrane and misalignment of neuroreceptors. Conversely, oxidized membrane lipids and ferroptosis alter the biophysical properties of membranes, disrupting neuronal functions [169].

2.3. Phenazines and Phenothiazine Derivatives

Several natural phenazines and phenothiazines are neuroprotective, improve sleep, and delay neurodegenerative processes. For example, geranyl-phenazine is a naural acetylcholinesterase inhibitor which exerts antipsychotic effects via muscarinic receptors. Indeed, a new class of antipsychotic drugs is currently being developed for SCZ and a patent exists for treating sleep disorders by upregulating acetylcholine [170,171,172] (WO2005016327A2). Other natural phenazines with neuroprotective functions include baraphenazines A–G fused compounds derived from Streptomyces sp. PU-10A which likely possess antipsychotic properties [173]. Moreover, several natural phenazines, including baraphenazines, leucanicidin and endophenasides, exert antimicrobial, anticancer activity, and very likely possess antipsychotic properties [173,174,175].
Natural antipsychotic and phytotherapeutic compounds are not only devoid of extrapyramidal adverse effects but more accepted by many patients who often dread or distrust pharmaceuticals.
Synthetic phenazine derivatives consist of over 6000 compounds, exerting antimicrobial, antiparasitic, neuroprotective, anti-inflammatory, and anticancer activities [176,177,178]. To the best of our knowledge, natural or synthetic phenazines have not been tested for SCZ, insomnia, or neurodegeneration. Pontemazines A and B are neuroprotective phenazine derivatives that, in animal studies, have rescued hippocampal neurons from glutamate cytotoxicity, highlighting their pro-cognitive properties which could benefit patients with negative symptoms of SCZ or neurodegenerative disorders [176].
Synthetic phenazines exert antioxidant and radical-scavenging properties, and inhibit lipid peroxidation, suggesting beneficial effects in severe insomnia, mental illness and neurodegeneration [179,180] (Figure 3). Moreover, halogenated phenazines act as iron chelators, likely preventing neuronal ferroptosis [181]. We believe that pontemazines and halogenated phenazines should be assessed for antipsychotic/anti-neurodegenerative properties.
From the biochemical standpoint, phenazines are almost identical to phenothiazine antipsychotics and likely possess similar properties (Figure 4). Phenothiazines are typical antipsychotic drugs utilized primarily for SCZ and SLDs, which block dopaminergic transmission at the level of postsynaptic neuron. Several phenothiazines influence other receptors, including adrenergic, histaminergic, and cholinergic, exerting various clinical effects as well as adverse reactions. Aside from psychotic disorders, phenothiazines are also used for the treatment of migraine headaches, hiccups, nausea, vomiting, and cancer [182]. Like phenazines, phenothiazines intercalate themselves into the lipid bilayer of plasma and mitochondrial membranes, disrupting the curvature and receptor alignment on neuronal/mitochondrial surfaces [183] (Figure 3). In contrast, oxidized lipids, including 7-ketocholesterol (7KCl), form looped structures, generating membrane curvatures and pores that may trigger cell death [184].
Antioxidant phenothiazine and their derivatives have recently been developed for cancer, cardiovascular disease (CVD), Mycobacterium leprae and other antibiotic-resistant microbes [185,186].
Phenothiazine derivatives exert anti-peroxidation properties and protect against lipid pathology and ferroptosis, suggesting efficacy as antipsychotic drugs [187]. In addition, antioxidant phenothiazines are likely beneficial for insomnia and neurodegenerative disorders, suggesting that these compounds should be tested for neuropsychiatric pathology [186].
Propenylphenothiazine is a potent antioxidant with electron-donor capability that could prevent gray matter loss, a hallmark of SCZ and SLDs [188,189]. Electron-donating psychotropic drugs have been known to preserve the brain volume, suggesting that propenylphenothiazine may treat psychosis without reducing the gray matter volume. The majority of conventional antipsychotic drugs are electron-acceptors which often lower the brain volume as documented by many neuroimaging studies [190]. An even newer category of tetracyclic and pentacyclic phenothiazines with antioxidant properties has recently been developed, suggesting likely efficacy for cognitive impairment and negative SCZ symptoms. Moreover, the N10-carbonyl-substituted phenothiazines were demonstrated to inhibit lipid peroxidation, suggesting superior antipsychotic efficacy [191].
Natural and some synthetic phenazines and novel antioxidant phenothiazines have not been tested for SCZ, insomnia or neurodegenerative disorders but are likely efficient somnogens and antipsychotics. For example, synthetic phenazines, known as pontemazines A and B, rescued hippocampal neurons from glutamate cytotoxicity in rodents, highlighting their pro-cognitive properties which could benefit patients with negative symptoms of SCZ [192].

2.4. Natural Antioxidants

SCZ and SLDs have been associated with premature cellular senescence, a phenotype marked by shortened telomeres, accumulation of macromolecular aggregates, increased level of senescence-associated β-galactosidase (SA-β-gal) and a detrimental secretome known as senescence-associated secretory phenotype (SASP).

Natural Antioxidant Foods

Antioxidants are major players in repairing damages to macromolecules, opposing pathological events associated with cellular senescence (Table 1). Since AhR is the master regulator of cellular senescence and responds to external pollutants (such as polycyclic aromatic hydrocarbons (PAHs) as well as internal toxins, including oxidized lipids, antioxidants likely have the opposite effect.

2.5. Mitochondrial Transfer and Transplantation

Early studies on mitochondrial transplantation from the 1980s utilized co-incubation of various cell types with naked mitochondria, hoping that cells would internalize the organelles from the extracellular environment [202,203,204]. Later, HeLa cells and mesenchymal stem cells were used as mitochondrial sources and found that successful organelle uptake occurred in a short time interval of 1–2 h [205,206,207]. At present, mitochondrial transplantation into cardiomyocytes has been accomplished successfully and confirmed by mitochondrial DNA (mtDNA) detected in host cells [208,209].
Mitochondrial transplantation and neuronal rescue from ferroptosis have been performed successfully in both animals and humans, suggesting a novel strategy for neurometabolic disorders [210]. To our knowledge, mitochondrial transplantation has not been attempted in sleep disorders, while in mental illness, it has been tried in animal models only [132]. Trafficking mitochondria from astrocytes and microglia to neurons can take place spontaneously after brain injuries, reflecting a likely compensatory mechanism to preserve neuronal viability [211]. In addition, it has been established that SSRIs, GJA1-20K, and CD38 signaling can facilitate mitochondrial transfer, emphasizing potential strategies for insomnia, severe mental illness, and neurodegeneration [210,211].

3. Conclusions

Forensic detainees with severe mental illness and comorbid insomnia age at an accelerated pace, suggesting that premature cellular senescence, a characteristic of SCZ, may comprise the common pathway where sleep and mental illness intersect. Loss of neurons due to impaired sleep may trigger the premature development of dementia and other age-related conditions, known to occur earlier in life compared to the general population. These comorbidities increase healthcare expenditures and shorten patients’ lifespan; thus, identifying and treating these conditions early is crucial.
YOD, a category of neurodegenerative disorders which include bvFTD, has been on the rise over the past few decades, as evidenced by the increased number of first offenders before the age of 65. Selective loss of VENs in bvFTD is likely due to the large size of these cells, predisposed to peroxidation of plasma membrane lipids and mitochondrial loss by dysfunctional autophagy.
At the molecular level, AhR is the equivalent of VENs, as this protein responds to both endogenous and exogenous ligands, including lipid peroxides and other insomnia and psychosis-related molecules.
Antioxidants and phenazine and phenothiazine derivatives are AhR ligands, highlighting potential natural treatment strategies against psychosis, insomnia, and neurodegeneration.

Author Contributions

Conceptualization, A.S. and P.G.B.; methodology, N.J.; validation, K.A.T. and I.A.O.; writing—review and editing, P.G.B. 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.

References

  1. Sateia, M.J.; Doghramji, K.; Hauri, P.J.; Morin, C.M. Evaluation of chronic insomnia. An American Academy of Sleep Medicine review. Sleep 2000, 23, 243–308. [Google Scholar] [CrossRef]
  2. Dewa, L.H.; Thibaut, B.; Pattison, N.; Campbell, S.J.; Woodcock, T.; Aylin, P.; Archer, S. Treating insomnia in people who are incarcerated: A feasibility study of a multi-component treatment pathway. Sleep Adv. 2024, 5, zpae003. [Google Scholar] [CrossRef]
  3. Talih, F.; Ajaltouni, J.; Ghandour, H.; Abu-Mohammad, A.S.; Kobeissy, F. Insomnia in hospitalized psychiatric patients: Prevalence and associated factors. Neuropsychiatr. Dis. Treat. 2018, 14, 969–975. [Google Scholar] [CrossRef]
  4. Levichkina, E.V.; Busygina, I.I.; Pigareva, M.L.; Pigarev, I.N. The Mysterious Island: Insula and Its Dual Function in Sleep and Wakefulness. Front. Syst. Neurosci. 2021, 14, 592660. [Google Scholar] [CrossRef] [PubMed]
  5. Xu, H.; Shen, H.; Wang, L.; Zhong, Q.; Lei, Y.; Yang, L.; Zeng, L.L.; Zhou, Z.; Hu, D.; Yang, Z. Impact of 36 h of total sleep depri-vation on resting-state dynamic functional connectivity. Brain Res. 2018, 1688, 22–32. [Google Scholar] [CrossRef] [PubMed]
  6. Wylie, K.P.; Tregellas, J.R. The role of the insula in schizophrenia. Schizophr. Res. 2010, 123, 93–104. [Google Scholar] [CrossRef] [PubMed]
  7. Fathy, Y.Y.; Hoogers, S.E.; Berendse, H.W.; van der Werf, Y.D.; Visser, P.J.; de Jong, F.J.; van de Berg, W.D. Differential insular cortex sub-regional atrophy in neurodegenerative diseases: A systematic review and meta-analysis. Brain Imaging Behav. 2019, 14, 2799–2816. [Google Scholar] [CrossRef] [PubMed]
  8. Koutsouleris, N.; Pantelis, C.; Velakoulis, D.; McGuire, P.; Dwyer, D.B.; Urquijo-Castro, M.-F.; Paul, R.; Dong, S.; Popovic, D.; Oeztuerk, O.; et al. Exploring Links Between Psychosis and Frontotemporal Dementia Using Multimodal Machine Learning: Dementia Praecox Revisited. JAMA Psychiatry 2022, 79, 907–919. [Google Scholar] [CrossRef]
  9. Triarhou, L.C. The percipient observations of Constantin von Economo on encephalitis lethargica and sleep disruption and their lasting impact on contemporary sleep research. Brain Res. Bull. 2006, 69, 244–258. [Google Scholar] [CrossRef]
  10. Allman, J.M.; Tetreault, N.A.; Hakeem, A.Y.; Manaye, K.F.; Semendeferi, K.; Erwin, J.M.; Park, S.; Goubert, V.; Hof, P.R. The von Economo neurons in frontoinsular and anterior cingulate cortex in great apes and humans. Brain Struct. Funct. 2010, 214, 495–517. [Google Scholar] [CrossRef]
  11. Berg, M.T.; Rogers, E.M.; Lei, M.-K.; Simons, R.L. Losing Years Doing Time: Incarceration Exposure and Accelerated Biological Aging among African American Adults. J. Health Soc. Behav. 2021, 62, 460–476. [Google Scholar] [CrossRef]
  12. Kaiksow, F.A.; Brown, L.; Merss, K.B. Caring for the Rapidly Aging Incarcerated Population: The Role of Policy. J. Gerontol. Nurs. 2023, 49, 7–11. [Google Scholar] [CrossRef] [PubMed]
  13. Papanastasiou, E.; Gaughran, F.; Smith, S. Schizophrenia as segmental progeria. J. R. Soc. Med. 2011, 104, 475–484. [Google Scholar] [CrossRef] [PubMed]
  14. Killilea, D.W.; Wong, S.L.; Cahaya, H.S.; Atamna, H.; Ames, B.N. Iron accumulation during cellular senescence. Ann. N. Y. Acad. Sci. 2004, 1019, 365–367. [Google Scholar] [CrossRef] [PubMed]
  15. Urrutia, P.J.; Mena, N.P.; Núñez, M.T. The interplay between iron accumulation, mitochondrial dysfunction, and inflammation during the execution step of neurodegenerative disorders. Front. Pharmacol. 2014, 5, 38. [Google Scholar] [CrossRef] [PubMed]
  16. Carvalhas-Almeida, C.; Cavadas, C.; Álvaro, A.R. The impact of insomnia on frailty and the hallmarks of aging. Aging Clin. Exp. Res. 2022, 35, 253–269. [Google Scholar] [CrossRef] [PubMed]
  17. Carroll, J.E.; Prather, A.A. Sleep and biological aging: A short review. Curr. Opin. Endocr. Metab. Res. 2021, 18, 159–164. [Google Scholar] [CrossRef] [PubMed]
  18. Skonieczna-Żydecka, K.; Jamioł-Milc, D.; Borecki, K.; Stachowska, E.; Zabielska, P.; Kamińska, M.; Karakiewicz, B. The Prevalence of Insomnia and the Link between Iron Metabolism Genes Polymorphisms, TF rs1049296 C>T, TF rs3811647 G>A, TFR rs7385804 A>C, HAMP rs10421768 A>G and Sleep Disorders in Polish Individuals with ASD. Int. J. Environ. Res. Public Health 2020, 17, 400. [Google Scholar] [CrossRef]
  19. Nacarino-Palma, A.; Rico-Leo, E.M.; Campisi, J.; Ramanathan, A.; González-Rico, F.J.; Rejano-Gordillo, C.M.; Ordiales-Talavero, A.; Merino, J.M.; Fernández-Salguero, P.M. Aryl hydrocarbon receptor blocks aging-induced senescence in the liver and fibroblast cells. Aging 2022, 14, 4281–4304. [Google Scholar] [CrossRef]
  20. Panda, S.K.; Peng, V.; Sudan, R.; Antonova, A.U.; Di Luccia, B.; Ohara, T.E.; Fachi, J.L.; Grajales-Reyes, G.E.; Jaeger, N.; Trsan, T.; et al. Repression of the aryl-hydrocarbon receptor prevents oxidative stress and ferroptosis of intestinal intraepithelial lymphocytes. Immunity 2023, 56, 797–812.e4. [Google Scholar] [CrossRef]
  21. Hwang, H.J.; Dornbos, P.; Steidemann, M.; Dunivin, T.K.; Rizzo, M.; LaPres, J.J. Mitochondrial-targeted aryl hydrocarbon receptor and the impact of 2,3,7,8-tetrachlorodibenzo-p-dioxin on cellular respiration and the mitochondrial proteome. Toxicol. Appl. Pharmacol. 2016, 304, 121–132. [Google Scholar] [CrossRef]
  22. Dietrich-Muszalska, A.; Kontek, B. Lipid peroxidation in patients with schizophrenia. Psychiatry Clin. Neurosci. 2010, 64, 469–475. [Google Scholar] [CrossRef] [PubMed]
  23. 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] [PubMed]
  24. Gulec, M.; Ozkol, H.; Selvi, Y.; Tuluce, Y.; Aydin, A.; Besiroglu, L.; Ozdemir, P.G. Oxidative stress in patients with primary insomnia. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2012, 37, 247–251. [Google Scholar] [CrossRef]
  25. Liang, H.; Van Remmen, H.; Frohlich, V.; Lechleiter, J.; Richardson, A.; Ran, Q. Gpx4 protects mitochondrial ATP generation against oxidative damage. Biochem. Biophys. Res. Commun. 2007, 356, 893–898. [Google Scholar] [CrossRef]
  26. Seibt, T.M.; Proneth, B.; Conrad, M. Role of GPX4 in ferroptosis and its pharmacological implication. Free Radic. Biol. Med. 2018, 133, 144–152. [Google Scholar] [CrossRef] [PubMed]
  27. Azuma, K.; Koumura, T.; Iwamoto, R.; Matsuoka, M.; Terauchi, R.; Yasuda, S.; Shiraya, T.; Watanabe, S.; Aihara, M.; Imai, H.; et al. Mitochondrial glutathione peroxidase 4 is indispensable for photoreceptor development and survival in mice. J. Biol. Chem. 2022, 298, 101824. [Google Scholar] [CrossRef] [PubMed]
  28. Dixon, S.J.; Lemberg, K.M.; Lamprecht, M.R.; Skouta, R.; Zaitsev, E.M.; Gleason, C.E.; Patel, D.N.; Bauer, A.J.; Cantley, A.M.; Yang, W.S.; et al. Fer-roptosis: An iron-dependent form of nonapoptotic cell death. Cell 2012, 149, 1060–1072. [Google Scholar] [CrossRef]
  29. 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]
  30. Carroll, J.E.; Esquivel, S.; Goldberg, A.; Seeman, T.E.; Effros, R.B.; Dock, J.; Olmstead, R.; Breen, E.C.; Irwin, M.R. Insomnia and Telomere Length in Older Adults. Sleep 2016, 39, 559–564. [Google Scholar] [CrossRef]
  31. 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]
  32. Porterfield, V.; Khan, S.S.; Foff, E.P.; Koseoglu, M.M.; Blanco, I.K.; Jayaraman, S.; Lien, E.; McConnell, M.J.; Bloom, G.S.; Lazo, J.S.; et al. A three-dimensional dementia model reveals spontaneous cell cycle re-entry and a senescence-associated secretory phenotype. Neurobiol. Aging 2020, 90, 125–134. [Google Scholar] [CrossRef]
  33. De Lorenzo, B.H.; Marchioro, L.d.O.; Greco, C.R.; Suchecki, D. Sleep-deprivation reduces NK cell number and function mediated by β-adrenergic signalling. Psychoneuroendocrinology 2015, 57, 134–143. [Google Scholar] [CrossRef] [PubMed]
  34. Tarantino, N.; Leboyer, M.; Bouleau, A.; Hamdani, N.; Richard, J.R.; Boukouaci, W.; Ching-Lien, W.; Godin, O.; Bengoufa, D.; Le Corvoisier, P.; et al. Natural killer cells in first-episode psychosis: An innate immune signature? Mol. Psychiatry 2021, 26, 5297–5306. [Google Scholar] [CrossRef]
  35. Huang, A.; Shinde, P.V.; Huang, J.; Senff, T.; Xu, H.C.; Margotta, C.; Häussinger, D.; Willnow, T.E.; Zhang, J.; Pandyra, A.A.; et al. Progranulin prevents regulatory NK cell cytotoxicity against antiviral T cells. J. Clin. Investig. 2019, 4, e129856. [Google Scholar] [CrossRef] [PubMed]
  36. Irwin, M.R. Sleep disruption induces activation of inflammation and heightens risk for infectious disease: Role of impairments in thermoregulation and elevated ambient temperature. Temperature 2022, 10, 198–234. [Google Scholar] [CrossRef]
  37. Bright, F.; Werry, E.L.; Dobson-Stone, C.; Piguet, O.; Ittner, L.M.; Halliday, G.M.; Hodges, J.R.; Kiernan, M.C.; Loy, C.T.; Kassiou, M.; et al. Neuroinflammation in frontotemporal dementia. Nat. Rev. Neurol. 2019, 15, 540–555. [Google Scholar] [CrossRef]
  38. Vallée, A. Neuroinflammation in Schizophrenia: The Key Role of the WNT/β-Catenin Pathway. Int. J. Mol. Sci. 2022, 23, 2810. [Google Scholar] [CrossRef]
  39. Nesci, S.; Spagnoletta, A.; Oppedisano, F. Inflammation, Mitochondria and Natural Compounds Together in the Circle of Trust. Int. J. Mol. Sci. 2023, 24, 6106. [Google Scholar] [CrossRef] [PubMed]
  40. Andrieux, P.; Chevillard, C.; Cunha-Neto, E.; Nunes, J.P.S. Mitochondria as a Cellular Hub in Infection and Inflammation. Int. J. Mol. Sci. 2021, 22, 11338. [Google Scholar] [CrossRef]
  41. 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. [Google Scholar] [CrossRef]
  42. Gollihue, J.; Norris, C. Astrocyte mitochondria: Central players and potential therapeutic targets for neurodegenerative diseases and injury. Ageing Res. Rev. 2020, 59, 101039. [Google Scholar] [CrossRef] [PubMed]
  43. Boas, S.M.; Joyce, K.L.; Cowell, R.M. The NRF2-Dependent Transcriptional Regulation of Antioxidant Defense Pathways: Relevance for Cell Type-Specific Vulnerability to Neurodegeneration and Therapeutic Intervention. Antioxidants 2021, 11, 8. [Google Scholar] [CrossRef] [PubMed]
  44. Downar, J.; Crawley, A.P.; Mikulis, D.J.; Davis, K.D. A multimodal cortical network for the detection of changes in the sensory environment. Nat. Neurosci. 2000, 3, 277–283. [Google Scholar] [CrossRef] [PubMed]
  45. Wolff, M.; Vann, S.D. The cognitive thalamus as a gateway to mental representations. J. Neurosci. 2018, 39, 3–14. [Google Scholar] [CrossRef]
  46. Sridharan, D.; Levitin, D.J.; Menon, V. A critical role for the right fronto-insular cortex in switching between central-executive and default-mode networks. Proc. Natl. Acad. Sci. USA 2008, 105, 12569–12574. [Google Scholar] [CrossRef]
  47. Ueno, D.; Matsuoka, T.; Kato, Y.; Ayani, N.; Maeda, S.; Takeda, M.; Narumoto, J. Individual Differences in Interoceptive Accuracy Are Correlated with Salience Network Connectivity in Older Adults. Front. Aging Neurosci. 2020, 12, 592002. [Google Scholar] [CrossRef]
  48. Blessing, W.W.; Blessing, E.M.; Mohammed, M.; Ootsuka, Y. Clozapine, chlorpromazine and risperidone dose-dependently reduce emotional hyperthermia, a biological marker of salience. Psychopharmacology 2017, 234, 3259–3269. [Google Scholar] [CrossRef]
  49. Seeley, W.W. The Salience Network: A Neural System for Perceiving and Responding to Homeostatic Demands. J. Neurosci. 2019, 39, 9878–9882. [Google Scholar] [CrossRef] [PubMed]
  50. Pasquini, L.; Nana, A.L.; Toller, G.; Brown, J.A.; Deng, J.; Staffaroni, A.; Kim, E.-J.; Hwang, J.-H.L.; Li, L.; Park, Y.; et al. Salience Network Atrophy Links Neuron Type-Specific Pathobiology to Loss of Empathy in Frontotemporal Dementia. Cereb. Cortex 2020, 30, 5387–5399. [Google Scholar] [CrossRef] [PubMed]
  51. Mendez, M.F.; Anderson, E.; Shapira, J.S. An investigate von of moral judgement in frontotemporal dementia. Cogn. Behav. Neurol. 2005, 18, 193–197. [Google Scholar] [CrossRef]
  52. Mendez, M.F. The neurobiology of moral behavior: Review and neuropsychiatric implications. CNS Spectr. 2009, 14, 608–620. [Google Scholar] [CrossRef] [PubMed]
  53. Boeve, B.F. Behavioral Variant Frontotemporal Dementia. Continuum 2022, 28, 702–725. [Google Scholar] [CrossRef] [PubMed]
  54. Arrigoni, E.; Fuller, P.M. The Sleep-Promoting Ventrolateral Preoptic Nucleus: What Have We Learned over the Past 25 Years? Int. J. Mol. Sci. 2022, 23, 2905. [Google Scholar] [CrossRef] [PubMed]
  55. De Luca, R.; Nardone, S.; Grace, K.P.; Venner, A.; Cristofolini, M.; Bandaru, S.S.; Sohn, L.T.; Kong, D.; Mochizuki, T.; Viberti, B.; et al. Orexin neurons inhibit sleep to promote arousal. Nat. Commun. 2022, 13, 4163. [Google Scholar] [CrossRef] [PubMed]
  56. Inutsuka, A.; Yamanaka, A. The regulation of sleep and wakefulness by the hypothalamic neuropeptide orexin/hypocretin. Nagoya J. Med. Sci. 2013, 75, 29–36. [Google Scholar]
  57. Yin, D.; Dong, H.; Wang, T.-X.; Hu, Z.-Z.; Cheng, N.-N.; Qu, W.-M.; Huang, Z.-L. Glutamate Activates the Histaminergic Tuberomammillary Nucleus and Increases Wakefulness in Rats. Neuroscience 2019, 413, 86–98. [Google Scholar] [CrossRef] [PubMed]
  58. Konadhode, R.R.; Pelluru, D.; Shiromani, P.J. Neurons containing orexin or melanin concentrating hormone reciprocally regulate wake and sleep. Front. Syst. Neurosci. 2015, 8, 244. [Google Scholar] [CrossRef]
  59. Chung, S.; Weber, F.; Zhong, P.; Tan, C.L.; Nguyen, T.N.; Beier, K.T.; Hörmann, N.; Chang, W.-C.; Zhang, Z.; Do, J.P.; et al. Identification of preoptic sleep neurons using retrograde labelling and gene profiling. Nature 2017, 545, 477–481. [Google Scholar] [CrossRef]
  60. Bandarabadi, M.; Li, S.; Aeschlimann, L.; Colombo, G.; Tzanoulinou, S.; Tafti, M.; Becchetti, A.; Boutrel, B.; Vassalli, A. Inactivation of hypocretin receptor-2 signaling in dopaminergic neurons induces hyperarousal and enhanced cognition but impaired inhibitory control. Mol. Psychiatry 2023. online ahead of print. [Google Scholar] [CrossRef]
  61. Wu, S.; Gao, C.; Han, F.; Cheng, H. Histamine H1 receptor in basal forebrain cholinergic circuit: A novel target for the negative symptoms of schizophrenia? Neurosci. Bull. 2022, 38, 558–560. [Google Scholar] [CrossRef]
  62. Grady, F.S.; Boes, A.D.; Geerling, J.C. A Century Searching for the Neurons Necessary for Wakefulness. Front. Neurosci. 2022, 16, 930514. [Google Scholar] [CrossRef]
  63. Kerkhofs, M.; Lavie, P. Frédéric Bremer 1892–1982: A pioneer in sleep research. Sleep Med. Rev. 2000, 4, 505–514. [Google Scholar] [CrossRef]
  64. Fuller, P.; Sherman, D.; Pedersen, N.P.; Saper, C.B.; Lu, J. Reassessment of the structural basis of the ascending arousal system. J. Comp. Neurol. 2010, 519, 933–956, Erratum in J. Comp. Neurol. 2011, 519, 3817. [Google Scholar] [CrossRef] [PubMed]
  65. Lavie, P. The sleep theory of Constantin von Economo. J. Sleep Res. 1993, 2, 175–178. [Google Scholar] [CrossRef]
  66. Vyas, A.; De Jesus, O. Von Economo Encephalitis. In StatPearls [Internet]; StatPearls Publishing: Treasure Island, FL, USA, 2024. [Google Scholar]
  67. Rosen, D. Asleep: The Forgotten Epidemic That Remains One of Medicine’s Greatest Mysteries. J. Clin. Sleep Med. 2010, 6, 299. [Google Scholar] [CrossRef]
  68. Cortelli, P.; Perani, D.; Parchi, P.; Grassi, F.; Montagna, P.; De Martin, M.; Castellani, R.; Tinuper, P.; Gambetti, P.; Lugaresi, E.; et al. Cerebral metabolism in fatal familial insomnia: Relation to duration, neuropathology, and distribution of protease-resistent prion protein. Neurology 1997, 49, 126–133. [Google Scholar] [CrossRef]
  69. Gallassi, R.; Morreale, A.; Montagna, P.; Cortelli, P.; Avoni, P.; Castellani, R.; Gambetti, R.; Lugaresi, E. Fatal familial insomnia: Behavioral and cognitive features. Neurology 1996, 46, 935–939. [Google Scholar] [CrossRef] [PubMed]
  70. Sturm, V.E.; Brown, J.A.; Hua, A.Y.; Lwi, S.J.; Zhou, J.; Kurth, F.; Eickhoff, S.B.; Rosen, H.J.; Kramer, J.H.; Miller, B.L.; et al. Network Architecture Underlying Basal Autonomic Outflow: Evidence from Frontotemporal Dementia. J. Neurosci. 2018, 38, 8943–8955. [Google Scholar] [CrossRef] [PubMed]
  71. Mallikarjun, P.K.; Lalousis, P.A.; Dunne, T.F.; Heinze, K.; Reniers, R.L.; Broome, M.R.; Farmah, B.; Oyebode, F.; Wood, S.J.; Upthegrove, R. Aberrant salience network functional connectivity in auditory verbal hallucinations: A first episode psychosis sample. Transl. Psychiatry 2018, 8, 69. [Google Scholar] [CrossRef] [PubMed]
  72. Cracco, L.; Appleby, B.S.; Gambetti, P. Fatal familial insomnia and sporadic fatal insomnia. In Handbook of Clinical Neurology; Elsevier: Amsterdam, The Netherlands, 2018; Volume 153, pp. 271–299. [Google Scholar] [CrossRef]
  73. Wang, Y.; Li, M.; Li, W.; Xiao, L.; Huo, X.; Ding, J.; Sun, T. Is the insula linked to sleep? A systematic review and narrative synthesis. Heliyon 2022, 8, e11406. [Google Scholar] [CrossRef]
  74. Hehr, A.; Huntley, E.D.; Marusak, H.A. Getting a Good Night′s Sleep: Associations Between Sleep Duration and Par-ent-Reported Sleep Quality on Default Mode Network Connectivity in Youth. J. Adolesc. Health 2023, 72, 933–942. [Google Scholar] [CrossRef] [PubMed]
  75. Guo, Y.; Zou, G.; Shao, Y.; Chen, J.; Li, Y.; Liu, J.; Yao, P.; Zhou, S.; Xu, J.; Hu, S.; et al. Increased connectivity of the anterior cingulate cortex is associated with the tendency to awakening during N2 sleep in patients with insomnia disorder. Sleep 2022, 46, zsac290. [Google Scholar] [CrossRef] [PubMed]
  76. Guldenmund, P.; Demertzi, A.; Boveroux, P.; Boly, M.; Vanhaudenhuyse, A.; Bruno, M.-A.; Gosseries, O.; Noirhomme, Q.; Brichant, J.-F.; Bonhomme, V.; et al. Thalamus, brainstem and salience network connectivity changes during propofol-induced sedation and unconsciousness. Brain Connect. 2013, 3, 273–285. [Google Scholar] [CrossRef] [PubMed]
  77. Zhang, L.; Luo, L.; Zhou, Z.; Xu, K.; Zhang, L.; Liu, X.; Tan, X.; Zhang, J.; Ye, X.; Gao, J.; et al. Functional Connectivity of Anterior Insula Predicts Recovery of Patients with Disorders of Consciousness. Front. Neurol. 2018, 9, 1024. [Google Scholar] [CrossRef]
  78. Mashour, G.A. Anesthetizing the Self: The Neurobiology of Humbug. Anesthesiology 2016, 124, 747–749. [Google Scholar] [CrossRef] [PubMed]
  79. Qi, J.; Li, B.-Z.; Zhang, Y.; Pan, B.; Gao, Y.-H.; Zhan, H.; Liu, Y.; Shao, Y.-C.; Zhang, X. Altered insula-prefrontal functional connectivity correlates to decreased vigilant attention after total sleep deprivation. Sleep Med. 2021, 84, 187–194. [Google Scholar] [CrossRef]
  80. Li, C.; Dong, M.; Yin, Y.; Hua, K.; Fu, S.; Jiang, G. Aberrant Effective Connectivity of the Right Anterior Insula in Primary Insomnia. Front. Neurol. 2018, 9, 317. [Google Scholar] [CrossRef]
  81. Chen, M.C.; Chiang, W.-Y.; Yugay, T.; Patxot, M.; Ozcivit, I.B.; Hu, K.; Lu, J. Anterior Insula Regulates Multiscale Temporal Organization of Sleep and Wake Activity. J. Biol. Rhythm. 2016, 31, 182–193. [Google Scholar] [CrossRef]
  82. Palaniyappan, L.; White, T.; Liddle, P. The concept of salience network dysfunction in schizophrenia: From neuroimaging observations to therapeutic opportunities. Curr. Top. Med. Chem. 2012, 12, 2324–2338. [Google Scholar] [CrossRef]
  83. Huang, H.; Chen, C.; Rong, B.; Wan, Q.; Chen, J.; Liu, Z.; Zhou, Y.; Wang, G.; Wang, H. Resting-state functional connectivity of salience network in schizophrenia and depression. Sci. Rep. 2022, 12, 11204. [Google Scholar] [CrossRef]
  84. He, X.; Qin, W.; Liu, Y.; Zhang, X.; Duan, Y.; Song, J.; Li, K.; Jiang, T.; Yu, C. Abnormal salience network in normal aging and in amnestic mild cognitive impairment and Alzheimer’s disease. Hum. Brain Mapp. 2013, 35, 3446–3464. [Google Scholar] [CrossRef] [PubMed]
  85. Putcha, D.; Ross, R.S.; Cronin-Golomb, A.; Janes, A.C.; Stern, C.E. Salience and Default Mode Network Coupling Predicts Cognition in Aging and Parkinson’s Disease. J. Int. Neuropsychol. Soc. 2016, 22, 205–215. [Google Scholar] [CrossRef] [PubMed]
  86. Day, G.S.; Farb, N.A.S.; Tang-Wai, D.F.; Masellis, M.; Black, S.E.; Freedman, M.; Pollock, B.G.; Chow, T.W. Salience Network Resting-State Activity: Prediction of Frontotemporal Dementia Pro-gression. JAMA Neurol. 2013, 70, 1249–1253. [Google Scholar] [CrossRef] [PubMed]
  87. Sheffield, J.M.; Rogers, B.P.; Blackford, J.U.; Heckers, S.; Woodward, N.D. Insula functional connectivity in schizophrenia. Schizophr. Res. 2020, 220, 69–77. [Google Scholar] [CrossRef]
  88. Adams, R.; David, A.S. Patterns of anterior cingulate activation in schizophrenia: A selective review. Neuropsychiatr. Dis. Treat. 2007, 3, 87–101. [Google Scholar] [CrossRef]
  89. Nana, A.L.; Sidhu, M.; Gaus, S.E.; Hwang, J.-H.L.; Li, L.; Park, Y.; Kim, E.-J.; Pasquini, L.; Allen, I.E.; Rankin, K.P.; et al. Neurons selectively targeted in frontotemporal dementia reveal early stage TDP-43 pathobiology. Acta Neuropathol. 2018, 137, 27–46. [Google Scholar] [CrossRef] [PubMed]
  90. Zago, S.; Scarpazza, C.; Difonzo, T.; Arighi, A.; Hajhajate, D.; Torrente, Y.; Sartori, G. Behavioral Variant of Frontotemporal Dementia and Homicide in a Historical Case. J. Am. Acad. Psychiatry Law 2021, 49, 219–227. [Google Scholar] [PubMed]
  91. Nilsson, C.; Waldö, M.L.; Nilsson, K.; Santillo, A.; Vestberg, S. Age-related incidence and family history in frontotemporal dementia: Data from the swedish dementia registry. PLoS ONE 2014, 9, e94901. [Google Scholar] [CrossRef]
  92. Hendriks, S.; Peetoom, K.; Bakker, C.; van der Flier, W.M.; Papma, J.M.; Koopmans, R.; Verhey, F.R.J.; de Vugt, M.; Köhler, S.; Withall, A.; et al. Global Prevalence of Young-Onset Dementia: A Systematic Review and Meta-analysis. JAMA Neurol. 2021, 78, 1080–1090. [Google Scholar] [CrossRef]
  93. Onyike, C.U.; Diehl-Schmid, J. The epidemiology of frontotemporal dementia. Int. Rev. Psychiatry 2013, 25, 130–137. [Google Scholar] [CrossRef]
  94. Diehl-Schmid, J.; Perneczky, R.; Koch, J.; Nedopil, N.; Kurz, A. Guilty by suspicion? Criminal behavior in frontotemporal lobar degeneration. Cogn. Behav. Neurol. 2013, 26, 73–77. [Google Scholar] [CrossRef]
  95. Krause, M.; Theiss, C.; Brüne, M. Ultrastructural Alterations of Von Economo Neurons in the Anterior Cingulate Cortex in Schizophrenia. Anat. Rec. 2017, 300, 2017–2024. [Google Scholar] [CrossRef]
  96. Kim, S.H.; Kim, Y.J.; Lee, B.H.; Lee, P.; Park, J.H.; Seo, S.W.; Jeong, Y. Behavioral Reserve in Behavioral Variant Frontotemporal Dementia. Front. Aging Neurosci. 2022, 14, 875589. [Google Scholar] [CrossRef]
  97. Vohryzek, J.; Cabral, J.; Vuust, P.; Deco, G.; Kringelbach, M.L. Understanding brain states across spacetime informed by whole-brain modelling. Philos. Trans. R. Soc. A Math. Phys. Eng. Sci. 2022, 380, 20210247. [Google Scholar] [CrossRef]
  98. Nathani, M.; Jaleel, V.; Turner, A.; Dirvonas, C.; Suryadevara, U.; Tandon, R. When you hear hoofbeats, think horses and zebras: The importance of a wide differential when it comes to frontotemporal lobar degeneration. Asian J. Psychiatry 2019, 47, 101875. [Google Scholar] [CrossRef] [PubMed]
  99. Herbert, B.M.; Herbert, C.; Pollatos, O. On the Relationship Between Interoceptive Awareness and Alexithymia: Is Interoceptive Awareness Related to Emotional Awareness? J. Personal. 2011, 79, 1149–1175. [Google Scholar] [CrossRef]
  100. Quadt, L.; Critchley, H.D.; Garfinkel, S.N. The neurobiology of interoception in health and disease. Ann. N. Y. Acad. Sci. 2018, 1428, 112–128. [Google Scholar] [CrossRef] [PubMed]
  101. Cauda, F.; Geminiani, G.C.; Vercelli, A. Evolutionary appearance of von Economo’s neurons in the mammalian cerebral cortex. Front. Hum. Neurosci. 2014, 8, 104. [Google Scholar] [CrossRef] [PubMed]
  102. Menon, V.; Uddin, L.Q. Saliency, switching, attention and control: A network model of insula function. Brain Struct. Funct. 2010, 214, 655–667. [Google Scholar] [CrossRef]
  103. López-Ojeda, W.; Hurley, R.A. Von Economo Neuron Involvement in Social Cognitive and Emotional Impairments in Neuropsychiatric Disorders. J. Neuropsychiatry Clin. Neurosci. 2022, 34, 302–306. [Google Scholar] [CrossRef]
  104. Hodge, R.D.; Miller, J.A.; Novotny, M.; Kalmbach, B.E.; Ting, J.T.; Bakken, T.E.; Aevermann, B.D.; Barkan, E.R.; Berkowitz-Cerasano, M.L.; Cobbs, C.; et al. Transcriptomic evidence that von Economo neurons are regionally specialized extratelencephalic-projecting excitatory neurons. Nat. Commun. 2020, 11, 1172. [Google Scholar] [CrossRef] [PubMed]
  105. Dijkstra, A.A.; Lin, L.-C.; Nana, A.L.; Gaus, S.E.; Seeley, W.W. Von Economo Neurons and Fork Cells: A Neurochemical Signature Linked to Monoaminergic Function. Cereb. Cortex 2016, 28, 131–144. [Google Scholar] [CrossRef] [PubMed]
  106. Azizi, S.A. Monoamines: Dopamine, Norepinephrine, and Serotonin, Beyond Modulation, “Switches” That Alter the State of Target Networks. Neuroscientist 2020, 28, 121–143. [Google Scholar] [CrossRef] [PubMed]
  107. Valli, M.; Cho, S.S.; Uribe, C.; Masellis, M.; Chen, R.; Mihaescu, A.; Strafella, A.P. VMAT2 availability in Parkinson’s disease with probable REM sleep behaviour disorder. Mol. Brain 2021, 14, 165. [Google Scholar] [CrossRef] [PubMed]
  108. Broese, M.; Riemann, D.; Hein, L.; Nissen, C. α-Adrenergic Receptor Function, Arousal and Sleep: Mechanisms and Therapeutic Implications. Pharmacopsychiatry 2012, 45, 209–216. [Google Scholar] [CrossRef] [PubMed]
  109. Gaus, R.; Popal, M.; Heinsen, H.; Schmitt, A.; Falkai, P.; Hof, P.R.; Schmitz, C.; Vollhardt, A. Reduced cortical neuron number and neuron density in schizophrenia with focus on area 24: A post-mortem case–control study. Eur. Arch. Psychiatry Clin. Neurosci. 2022, 273, 1209–1223. [Google Scholar] [CrossRef] [PubMed]
  110. Sjögren, M.; Minthon, L.; Passant, U.; Blennow, K.; Wallin, A. Decreased monoamine metabolites in frontotemporal dementia and Alzheimer’s disease. Neurobiol. Aging 1998, 19, 379–384. [Google Scholar] [CrossRef]
  111. Levenson, J.C.; Kay, D.B.; Buysse, D.J. The Pathophysiology of Insomnia. Chest 2015, 147, 1179–1192. [Google Scholar] [CrossRef]
  112. Zhou, M.; Kiyoshi, C.M. Astrocyte syncytium: A functional reticular system in the brain. Neural Regen. Res. 2019, 14, 595–596. [Google Scholar] [CrossRef]
  113. Garofalo, S.; Picard, K.; Limatola, C.; Nadjar, A.; Pascual, O.; Tremblay, M.E. Role of Glia in the Regulation of Sleep in Health and Disease. Compr. Physiol. 2020, 10, 687–712. [Google Scholar] [CrossRef] [PubMed]
  114. Que, M.; Li, Y.; Wang, X.; Zhan, G.; Luo, X.; Zhou, Z. Role of astrocytes in sleep deprivation: Accomplices, resisters, or bystanders? Front. Cell. Neurosci. 2023, 17, 1188306. [Google Scholar] [CrossRef] [PubMed]
  115. Mader, S.; Brimberg, L. Aquaporin-4 Water Channel in the Brain and Its Implication for Health and Disease. Cells 2019, 8, 90. [Google Scholar] [CrossRef] [PubMed]
  116. Jessen, N.A.; Munk, A.S.F.; Lundgaard, I.; Nedergaard, M. The Glymphatic System: A Beginner’s Guide. Neurochem. Res. 2015, 40, 2583–2599. [Google Scholar] [CrossRef] [PubMed]
  117. Valenti, D.; Vacca, R.A.; Moro, L.; Atlante, A. Mitochondria Can Cross Cell Boundaries: An Overview of the Biological Relevance, Pathophysiological Implications and Therapeutic Perspectives of Intercellular Mitochondrial Transfer. Int. J. Mol. Sci. 2021, 22, 8312. [Google Scholar] [CrossRef] [PubMed]
  118. Leow, D.M.-K.; Cheah, I.K.-M.; Fong, Z.W.-J.; Halliwell, B.; Ong, W.-Y. Protective Effect of Ergothioneine against 7-Ketocholesterol-Induced Mitochondrial Damage in hCMEC/D3 Human Brain Endothelial Cells. Int. J. Mol. Sci. 2023, 24, 5498. [Google Scholar] [CrossRef] [PubMed]
  119. Asami, T.; Bouix, S.; Whitford, T.J.; Shenton, M.E.; Salisbury, D.F.; McCarley, R.W. Longitudinal loss of gray matter volume in patients with first-episode schizophrenia: DARTEL automated analysis and ROI validation. Neuroimage 2012, 59, 986–996. [Google Scholar] [CrossRef]
  120. Hill, V.M.; O’connor, R.M.; Sissoko, G.B.; Irobunda, I.S.; Leong, S.; Canman, J.C.; Stavropoulos, N.; Shirasu-Hiza, M. A bidirectional relationship between sleep and oxidative stress in Drosophila. PLoS Biol. 2018, 16, e2005206. [Google Scholar] [CrossRef]
  121. Patergnani, S.; Bonora, M.; Ingusci, S.; Previati, M.; Marchi, S.; Zucchini, S.; Perrone, M.; Wieckowski, M.R.; Castellazzi, M.; Pugliatti, M.; et al. Antipsychotic drugs counteract autophagy and mitophagy in multiple sclerosis. Proc. Natl. Acad. Sci. USA 2021, 118, e2020078118. [Google Scholar] [CrossRef]
  122. Hirata, Y.; Cai, R.; Volchuk, A.; Steinberg, B.E.; Saito, Y.; Matsuzawa, A.; Grinstein, S.; Freeman, S.A. Lipid peroxidation increases membrane tension, Piezo1 gating, and cation permeability to execute ferroptosis. Curr. Biol. 2023, 33, 1282–1294.e5. [Google Scholar] [CrossRef]
  123. Heckers, S.; Konradi, C. Hippocampal neurons in schizophrenia. J. Neural Transm. 2002, 109, 891–905. [Google Scholar] [CrossRef] [PubMed]
  124. Padurariu, M.; Ciobica, A.; Mavroudis, I.; Fotiou, D.; Baloyannis, S. Hippocampal neuronal loss in the CA1 and CA3 areas of Alz-heimer’s disease patients. Psychiatr. Danub. 2012, 24, 152–158. [Google Scholar] [PubMed]
  125. Zhang, P.; Li, Y.-X.; Zhang, Z.-Z.; Yang, Y.; Rao, J.-X.; Xia, L.; Li, X.-Y.; Chen, G.-H.; Wang, F. Astroglial Mechanisms Underlying Chronic Insomnia Disorder: A Clinical Study. Nat. Sci. Sleep 2020, 12, 693–704. [Google Scholar] [CrossRef]
  126. Bellesi, M.; de Vivo, L.; Chini, M.; Gilli, F.; Tononi, G.; Cirelli, C. Sleep Loss Promotes Astrocytic Phagocytosis and Microglial Activation in Mouse Cerebral Cortex. J. Neurosci. 2017, 37, 5263–5273. [Google Scholar] [CrossRef]
  127. Vilalta, A.; Brown, G.C. Neurophagy, the phagocytosis of live neurons and synapses by glia, contributes to brain development and disease. FEBS J. 2017, 285, 3566–3575. [Google Scholar] [CrossRef] [PubMed]
  128. Liddelow, S.A.; Guttenplan, K.A.; Clarke, L.E.; Bennett, F.C.; Bohlen, C.J.; Schirmer, L.; Bennett, M.L.; Münch, A.E.; Chung, W.-S.; Peterson, T.C.; et al. Neurotoxic reactive astrocytes are induced by activated microglia. Nature 2017, 541, 481–487. [Google Scholar] [CrossRef]
  129. Whitehurst, T.; Howes, O. The role of mitochondria in the pathophysiology of schizophrenia: A critical review of the evidence focusing on mitochondrial complex one. Neurosci. Biobehav. Rev. 2021, 132, 449–464. [Google Scholar] [CrossRef]
  130. Beaupre, L.M.M.; Brown, G.M.; Braganza, N.A.; Kennedy, J.L.; Gonçalves, V.F. Mitochondria’s role in sleep: Novel insights from sleep deprivation and restriction studies. World J. Biol. Psychiatry 2021, 23, 1–13. [Google Scholar] [CrossRef]
  131. Richardson, R.B.; Mailloux, R.J. Mitochondria Need Their Sleep: Redox, Bioenergetics, and Temperature Regulation of Circadian Rhythms and the Role of Cysteine-Mediated Redox Signaling, Uncoupling Proteins, and Substrate Cycles. Antioxidants 2023, 12, 674. [Google Scholar] [CrossRef]
  132. Ene, H.M.; Karry, R.; Farfara, D.; Ben-Shachar, D. Mitochondria play an essential role in the trajectory of adolescent neurodevelopment and behavior in adulthood: Evidence from a schizophrenia rat model. Mol. Psychiatry 2022, 28, 1170–1181. [Google Scholar] [CrossRef]
  133. Wang, X.; Wang, W.; Li, L.; Perry, G.; Lee, H.-G.; Zhu, X. Oxidative stress and mitochondrial dysfunction in Alzheimer’s disease. Biochim. Biophys. Acta 2014, 1842, 1240–1247. [Google Scholar] [CrossRef]
  134. Heo, M.J.; Suh, J.H.; Lee, S.H.; Poulsen, K.L.; An, Y.A.; Moorthy, B.; Hartig, S.M.; Moore, D.D.; Kim, K.H. Aryl hydrocarbon receptor maintains hepatic mitochondrial homeostasis in mice. Mol. Metab. 2023, 72, 101717. [Google Scholar] [CrossRef] [PubMed]
  135. Duarte-Hospital, C.; Tête, A.; Brial, F.; Benoit, L.; Koual, M.; Tomkiewicz, C.; Kim, M.J.; Blanc, E.B.; Coumoul, X.; Bortoli, S. Mitochondrial Dysfunction as a Hallmark of Environmental Injury. Cells 2021, 11, 110. [Google Scholar] [CrossRef] [PubMed]
  136. Heath-Pagliuso, S.; Rogers, W.J.; Tullis, K.; Seidel, S.D.; Cenijn, P.H.; Brouwer, A.; Denison, M.S. Activation of the Ah receptor by tryptophan and tryptophan metabolites. Biochemistry 1998, 37, 11508–11515. [Google Scholar] [CrossRef] [PubMed]
  137. Slominski, A.T.; Kim, T.-K.; Slominski, R.M.; Song, Y.; Qayyum, S.; Placha, W.; Janjetovic, Z.; Kleszczyński, K.; Atigadda, V.; Song, Y.; et al. Melatonin and Its Metabolites Can Serve as Agonists on the Aryl Hydrocarbon Receptor and Peroxisome Proliferator-Activated Receptor Gamma. Int. J. Mol. Sci. 2023, 24, 15496. [Google Scholar] [CrossRef] [PubMed]
  138. Xu, C.-X.; Wang, C.; Krager, S.L.; Bottum, K.M.; Tischkau, S.A. Aryl hydrocarbon receptor activation attenuates Per1 gene induction and influences circadian clock resetting. Toxicol. Sci. 2013, 132, 368–378. [Google Scholar] [CrossRef] [PubMed]
  139. Hartmann, C.; Kempf, A. Mitochondrial control of sleep. Curr. Opin. Neurobiol. 2023, 81, 102733. [Google Scholar] [CrossRef] [PubMed]
  140. Wei, Y.-D.; Helleberg, H.; Rannug, U.; Rannug, A. Rapid and transient induction of CYP1A1 gene expression in human cells by the tryptophan photoproduct 6-formylindolo(3,2-b)carbazole. Chem. Biol. Interact. 1998, 110, 39–55. [Google Scholar] [CrossRef]
  141. Ziv-Gal, A.; Flaws, J.A.; Mahoney, M.M.; Miller, S.R.; Zacur, H.A.; Gallicchio, L. Genetic polymorphisms in the aryl hydrocarbon receptor-signaling pathway and sleep disturbances in middle-aged women. Sleep Med. 2013, 14, 883–887. [Google Scholar] [CrossRef]
  142. Frau-Méndez, M.A.; Fernández-Vega, I.; Ansoleaga, B.; Tech, R.B.; Tech, M.C.; del Rio, J.A.; Zerr, I.; Llorens, F.; Zarranz, J.J.; Ferrer, I. Fatal familial insomnia: Mitochondrial and protein synthesis machinery decline in the mediodorsal thalamus. Brain Pathol. 2016, 27, 95–106. [Google Scholar] [CrossRef]
  143. Kishikawa, J.-I.; Inoue, Y.; Fujikawa, M.; Nishimura, K.; Nakanishi, A.; Tanabe, T.; Imamura, H.; Yokoyama, K. General anesthetics cause mitochondrial dysfunction and reduction of intracellular ATP levels. PLoS ONE 2018, 13, e0190213. [Google Scholar] [CrossRef]
  144. Wei, H. The role of calcium dysregulation in anesthetic-mediated neurotoxicity. Anesth. Analg. 2011, 113, 972–974. [Google Scholar] [CrossRef]
  145. Lee, H.-G.; Choi, S.-I.; Park, S.-K.; Park, S.-J.; Kim, N.-H.; Choi, E.-K. Alteration of glutathione metabolism and abnormal calcium accumulation in the mitochondria of hamster brain infected with scrapie agent. Neurobiol. Aging 2000, 21, 151. [Google Scholar] [CrossRef]
  146. Glatzel, M.; Sepulveda-Falla, D. Losing sleep over mitochondria: A new player in the pathophysiology of fatal familial insomnia. Brain Pathol. 2016, 27, 107–108. [Google Scholar] [CrossRef] [PubMed]
  147. Moon, J.-H.; Park, S.-Y. Prion peptide-mediated calcium level alteration governs neuronal cell damage through AMPK-autophagy flux. Cell Commun. Signal. 2020, 18, 109. [Google Scholar] [CrossRef] [PubMed]
  148. Matteoni, S.; Matarrese, P.; Ascione, B.; Ricci-Vitiani, L.; Pallini, R.; Villani, V.; Pace, A.; Paggi, M.G.; Abbruzzese, C. Chlorpromazine induces cytotoxic autophagy in glioblastoma cells via endoplasmic reticulum stress and unfolded protein response. J. Exp. Clin. Cancer Res. 2021, 40, 347. [Google Scholar] [CrossRef] [PubMed]
  149. Barreca, M.L.; Iraci, N.; Biggi, S.; Cecchetti, V.; Biasini, E. Pharmacological Agents Targeting the Cellular Prion Protein. Pathogens 2018, 7, 27. [Google Scholar] [CrossRef] [PubMed]
  150. Korth, C.; May, B.C.H.; Cohen, F.E.; Prusiner, S.B. Acridine and phenothiazine derivatives as pharmacotherapeutics for prion disease. Proc. Natl. Acad. Sci. USA 2001, 98, 9836–9841. [Google Scholar] [CrossRef] [PubMed]
  151. Khattar, K.E.; Safi, J.; Rodriguez, A.-M.; Vignais, M.-L. Intercellular Communication in the Brain through Tunneling Nanotubes. Cancers 2022, 14, 1207. [Google Scholar] [CrossRef] [PubMed]
  152. Wang, X.; Gerdes, H.-H. Transfer of mitochondria via tunneling nanotubes rescues apoptotic PC12 cells. Cell Death Differ. 2015, 22, 1181–1191. [Google Scholar] [CrossRef]
  153. Savaskan, N.E.; Borchert, A.; Bräuer, A.U.; Kuhn, H. Role for glutathione peroxidase-4 in brain development and neuronal apoptosis: Specific induction of enzyme expression in reactive astrocytes following brain injury. Free Radic. Biol. Med. 2007, 43, 191–201. [Google Scholar] [CrossRef] [PubMed]
  154. Mauri, S.; Favaro, M.; Bernardo, G.; Mazzotta, G.M.; Ziviani, E. Mitochondrial autophagy in the sleeping brain. Front. Cell Dev. Biol. 2022, 10, 956394. [Google Scholar] [CrossRef] [PubMed]
  155. Chen, S.; Owens, G.C.; Crossin, K.L.; Edelman, D.B. Serotonin stimulates mitochondrial transport in hippocampal neurons. Mol. Cell. Neurosci. 2007, 36, 472–483. [Google Scholar] [CrossRef] [PubMed]
  156. Lin, L.-C.; Nana, A.L.; Hepker, M.; Hwang, J.-H.L.; Gaus, S.E.; Spina, S.; Cosme, C.G.; Gan, L.; Grinberg, L.T.; Geschwind, D.H.; et al. Preferential tau aggregation in von Economo neurons and fork cells in frontotemporal lobar degeneration with specific MAPT variants. Acta Neuropathol. Commun. 2019, 7, 159. [Google Scholar] [CrossRef] [PubMed]
  157. Kato, T. The other, forgotten genome: Mitochondrial DNA and mental disorders. Mol. Psychiatry 2001, 6, 625–633. [Google Scholar] [CrossRef] [PubMed]
  158. Course, M.M.; Wang, X. Transporting mitochondria in neurons. F1000Research 2016, 5, 1735. [Google Scholar] [CrossRef]
  159. 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]
  160. Kupershmidt, L.; Youdim, M.B.H. The Neuroprotective Activities of the Novel Multi-Target Iron-Chelators in Models of Alzheimer’s Disease, Amyotrophic Lateral Sclerosis and Aging. Cells 2023, 12, 763. [Google Scholar] [CrossRef]
  161. Calderon-Montaño, J.M.; Burgos-Morón, E.; Perez-Guerrero, C.; Lopez-Lazaro, M. A Review on the Dietary Flavonoid Kaempferol. Mini-Rev. Med. Chem. 2011, 11, 298–344. [Google Scholar] [CrossRef]
  162. Du, Y.-Y.; Sun, T.; Yang, Q.; Liu, Q.-Q.; Li, J.-M.; Yang, L.; Luo, L.-X. Therapeutic Potential of Kaempferol against Sleep Deprivation-Induced Cognitive Impairment: Modulation of Neuroinflammation and Synaptic Plasticity Disruption in Mice. ACS Pharmacol. Transl. Sci. 2023, 6, 1934–1944. [Google Scholar] [CrossRef]
  163. Saleem, A.; Ain, Q.U.; Akhtar, M.F. Alternative Therapy of Psychosis: Potential Phytochemicals and Drug Targets in the Management of Schizophrenia. Front. Pharmacol. 2022, 13, 895668. [Google Scholar] [CrossRef]
  164. 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. Oxidative Med. Cell. Longev. 2015, 2015, 481405. [Google Scholar] [CrossRef] [PubMed]
  165. 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]
  166. Mohammad, M.K.; Al-Masri, I.M.; Taha, M.O.; Al-Ghussein, M.A.; AlKhatib, H.S.; Najjar, S.; Bustanji, Y. Olanzapine inhibits glycogen synthase kinase-3beta: An investigation by docking simulation and experimental validation. Eur. J. Pharmacol. 2008, 584, 185–191. [Google Scholar] [CrossRef] [PubMed]
  167. Shilovsky, G.A.; Putyatina, T.S.; Morgunova, G.V.; Seliverstov, A.V.; Ashapkin, V.V.; Sorokina, E.V.; Markov, A.V.; Skulachev, V.P. A Crosstalk between the Biorhythms and Gatekeepers of Longevity: Dual Role of Glycogen Synthase Kinase-3. Biochemistry 2021, 86, 433–448. [Google Scholar] [CrossRef] [PubMed]
  168. Dozza, B.; Smith, M.A.; Perry, G.; Tabaton, M.; Strocchi, P. Regulation of glycogen synthase kinase-3beta by products of lipid peroxidation in human neuroblastoma cells. J. Neurochem. 2004, 89, 1224–1232. [Google Scholar] [CrossRef]
  169. Agmon, E.; Solon, J.; Bassereau, P.; Stockwell, B.R. Modeling the effects of lipid peroxidation during ferroptosis on membrane properties. Sci. Rep. 2018, 8, 5155. [Google Scholar] [CrossRef]
  170. 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]
  171. Foster, D.J.; Bryant, Z.K.; Conn, P.J. Targeting muscarinic receptors to treat schizophrenia. Behav. Brain Res. 2021, 405, 113201. [Google Scholar] [CrossRef]
  172. 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. Baraphenazines A–G, Divergent Fused Phenazine-Based Metabolites from a Himalayan Streptomyces. J. Nat. Prod. 2019, 82, 1686–1693. [Google Scholar] [CrossRef]
  173. van Wezel, G.P.; McKenzie, N.L.; Nodwell, J.R. Chapter 5 Applying the genetics of secondary metabolism in model actino-mycetes to the discovery of new antibiotics. In Methods in Enzymology, 1st ed.; Elsevier Inc.: Amsterdam, The Netherlands, 2009; Volume 458. [Google Scholar]
  174. Cha, J.W.; Lee, S.I.; 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]
  175. Krishnaiah, M.; Rodriguez de Almeida, N.; Udumula, V.; Song, Z.; Chhonker, Y.S.; Abdelmoaty, M.M.; Aragao do Nascimento, V.; Murry, D.J.; Conda-Sheridan, M. Synthesis, biological evaluation, and metabolic stability of phenazine derivatives as antibacterial agents. Eur. J. Med. Chem. 2018, 143, 936–947. [Google Scholar] [CrossRef] [PubMed]
  176. Lavaggi, M.L.; Aguirre, G.; Boiani, L.; Orelli, L.; García, B.; Cerecetto, H.; González, M. Pyrimido[1,2-a]quinoxaline 6-oxide and phenazine 5,10-dioxide derivatives and related compounds as growth inhibitors of Trypanosoma cruzi. Eur. J. Med. Chem. 2008, 43, 1737–1741. [Google Scholar] [CrossRef] [PubMed]
  177. 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]
  178. Laxmi, M.; Bhat, S.G. Characterization of pyocyanin with radical scavenging and antibiofilm properties isolated from Pseudomonas aeruginosa strain BTRY1. 3 Biotech 2016, 6, 27. [Google Scholar] [CrossRef]
  179. Alatawneh, N.; Meijler, M.M. Unraveling the Antibacterial and Iron Chelating Activity ofN-Oxide Hydroxy-Phenazine natural Products and Synthetic Analogs againstStaphylococcus aureus. Isr. J. Chem. 2023, 63, e202200112. [Google Scholar] [CrossRef]
  180. Edinoff, A.N.; Armistead, G.; Rosa, C.A.; Anderson, A.; Patil, R.; Cornett, E.M.; Murnane, K.S.; Kaye, A.M.; Kaye, A.D. Phenothiazines and their Evolving Roles in Clinical Practice: A Narrative Review. Health Psychol. Res. 2022, 10, 38930. [Google Scholar] [CrossRef] [PubMed]
  181. 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]
  182. 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]
  183. Wu, C.-H.; Bai, L.-Y.; Tsai, M.-H.; Chu, P.-C.; Chiu, C.-F.; Chen, M.Y.; Chiu, S.-J.; Chiang, J.-H.; Weng, J.-R. Pharmacological exploitation of the phenothiazine antipsychotics to develop novel antitumor agents–A drug repurposing strategy. Sci. Rep. 2016, 6, 27540. [Google Scholar] [CrossRef]
  184. Voronova, O.; Zhuravkov, S.; Korotkova, E.; Artamonov, A.; Plotnikov, E. Antioxidant Properties of New Phenothiazine Derivatives. Antioxidants 2022, 11, 1371. [Google Scholar] [CrossRef]
  185. 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]
  186. Iuga, C.; Campero, A.; Vivier-Bunge, A. Antioxidant vs. prooxidant action of phenothiazine in a biological environment in the presence of hydroxyl and hydroperoxyl radicals: A quantum chemistry study. RSC Adv. 2015, 5, 14678–14689. [Google Scholar] [CrossRef]
  187. Yue, Y.; Kong, L.; Wang, J.; Li, C.; Tan, L.; Su, H.; Xu, Y. Regional Abnormality of Grey Matter in Schizophrenia: Effect from the Illness or Treatment? PLoS ONE 2016, 11, e0147204. [Google Scholar] [CrossRef]
  188. Martínez, A.; Ibarra, I.A.; Vargas, R. A quantum chemical approach representing a new perspective concerning agonist and antagonist drugs in the context of schizophrenia and Parkinson’s disease. PLoS ONE 2019, 14, e0224691. [Google Scholar] [CrossRef]
  189. Goode-Romero, G.; Dominguez, L.; Martínez, A. Electron Donor–Acceptor Properties of Different Muscarinic Ligands: On the Road to Control Schizophrenia. J. Chem. Inf. Model. 2021, 61, 5117–5124. [Google Scholar] [CrossRef] [PubMed]
  190. Ho, B.C.; Andreasen, N.C.; Ziebell, S.; Pierson, R.; Magnotta, V. Long-term antipsychotic treatment and brain volumes: A longitudinal study of first-episode schizophrenia. Arch. Gen. Psychiatry 2011, 68, 128–137. [Google Scholar] [CrossRef]
  191. 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]
  192. Liang, Z.; Currais, A.; Soriano-Castell, D.; Schubert, D.; Maher, P. Natural products targeting mitochondria: Emerging therapeutics for age-associated neurological disorders. Pharmacol. Ther. 2021, 221, 107749. [Google Scholar] [CrossRef] [PubMed]
  193. Imran, M.; Ghorat, F.; Ul-Haq, I.; Ur-Rehman, H.; Aslam, F.; Heydari, M.; Shariati, M.A.; Okuskhanova, E.; Yessimbekov, Z.; Thiruvengadam, M.; et al. Lycopene as a Natural Antioxidant Used to Prevent Human Health Disorders. Antioxidants 2020, 9, 706. [Google Scholar] [CrossRef] [PubMed]
  194. Urquiaga, I.; Leighton, F. Plant polyphenol antioxidants and oxidative stress. Biol. Res. 2000, 33, 55–64. [Google Scholar] [CrossRef] [PubMed]
  195. Kumar, N.; Goel, N. Phenolic acids: Natural versatile molecules with promising therapeutic applications. Biotechnol. Rep. 2019, 24, e00370. [Google Scholar] [CrossRef] [PubMed]
  196. Jakubczyk, K.; Drużga, A.; Katarzyna, J.; Skonieczna-Żydecka, K. Antioxidant Potential of Curcumin—A Meta-Analysis of Randomized Clinical Trials. Antioxidants 2020, 9, 1092. [Google Scholar] [CrossRef] [PubMed]
  197. Nagle, D.G.; Ferreira, D.; Zhou, Y.-D. Epigallocatechin-3-gallate (EGCG): Chemical and biomedical perspectives. Phytochemistry 2006, 67, 1849–1855. [Google Scholar] [CrossRef] [PubMed]
  198. Li, Z.; Geng, Y.-N.; Jiang, J.-D.; Kong, W.-J. Antioxidant and anti-inflammatory activities of berberine in the treatment of diabetes mellitus. Evid. Based Complement. Altern. Med. 2014, 2014, 289264. [Google Scholar] [CrossRef]
  199. Deepika; Maurya, P.K. Health Benefits of Quercetin in Age-Related Diseases. Molecules 2022, 27, 2498. [Google Scholar] [CrossRef] [PubMed]
  200. Chen, A.Y.; Chen, Y.C. A review of the dietary flavonoid, kaempferol on human health and cancer chemoprevention. Food Chem. 2013, 138, 2099–2107. [Google Scholar] [CrossRef]
  201. Frankel, E.N. The antioxidant and nutritional effects of tocopherols, ascorbic acid and beta-carotene in relation to processing of edible oils. Bibl. Nutr. Dieta 1989, 43, 297–312. [Google Scholar] [CrossRef]
  202. Liu, D.; Gao, Y.; Liu, J.; Huang, Y.; Yin, J.; Feng, Y.; Shi, L.; Meloni, B.P.; Zhang, C.; Zheng, M.; et al. Intercellular mitochondrial transfer as a means of tissue revitalization. Signal Transduct. Target. Ther. 2021, 6, 65. [Google Scholar] [CrossRef]
  203. 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]
  204. Pacak, C.A.; 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] [PubMed]
  205. Berridge, M.V.; Schneider, R.T.; McConnell, M.J. Mitochondrial Transfer from Astrocytes to Neurons following Ischemic Insult: Guilt by Association? Cell Metab. 2016, 24, 376–378. [Google Scholar] [CrossRef] [PubMed]
  206. Pour, P.A.; 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]
  207. Cowan, D.B.; Yao, R.; Thedsanamoorthy, J.K.; Zurakowski, D.; del Nido, P.J.; McCully, J.D. Transit and fusion of exogenous mitochondria in human heart cells. Nat. Sci. Rep. 2017, 7, 17450. [Google Scholar] [CrossRef]
  208. Zhang, T.-G.; Miao, C.-Y. Mitochondrial transplantation as a promising therapy for mitochondrial diseases. Acta Pharm. Sin. B 2023, 13, 1028–1035. [Google Scholar] [CrossRef]
  209. Scheiblich, H.; Dansokho, C.; Mercan, D.; Schmidt, S.V.; Bousset, L.; Wischhof, L.; Eikens, F.; Odainic, A.; Spitzer, J.; Griep, A.; et al. Microglia jointly degrade fibrillar alpha-synuclein cargo by distribution through tunneling nanotubes. Cell 2021, 184, 5089–5106.e21. [Google Scholar] [CrossRef]
  210. Geng, Z.; Guan, S.; Wang, S.; Yu, Z.; Liu, T.; Du, S.; Zhu, C. Intercellular mitochondrial transfer in the brain, a new perspec-tive for targeted treatment of central nervous system diseases. CNS Neurosci. Ther. 2023, 29, 3121–3135. [Google Scholar] [CrossRef]
  211. Ren, D.; Zheng, P.; Zou, S.; Gong, Y.; Wang, Y.; Duan, J.; Deng, J.; Chen, H.; Feng, J.; Zhong, C.; et al. GJA1-20K Enhances Mitochondria Transfer from Astrocytes to Neurons via Cx43-TnTs After Traumatic Brain Injury. Cell. Mol. Neurobiol. 2021, 42, 1887–1895. [Google Scholar] [CrossRef]
Figure 1. Astrocytes contact cerebral microvessels with their end-feet processes, delineating a pathway for the flow of extracellular fluid, known as the glymphatic system. The volume of interstitial fluid (ISF) in the brain parenchyma varies with the brain work. During high intensity work, AQP-4 water receptors are upregulated in the end-feet, pumping the ISF into astrocytes. This results in low ISF (hypovolemia). During sleep (low-intensity brain work), less ISF enters the astrocyte. The circulation of ISF clears the molecular debris (including beta amyloid) from the extracellular space.
Figure 1. Astrocytes contact cerebral microvessels with their end-feet processes, delineating a pathway for the flow of extracellular fluid, known as the glymphatic system. The volume of interstitial fluid (ISF) in the brain parenchyma varies with the brain work. During high intensity work, AQP-4 water receptors are upregulated in the end-feet, pumping the ISF into astrocytes. This results in low ISF (hypovolemia). During sleep (low-intensity brain work), less ISF enters the astrocyte. The circulation of ISF clears the molecular debris (including beta amyloid) from the extracellular space.
Jcm 13 01691 g001
Figure 2. Astrocytes support the postmitotic, long-lived neurons by helping them avert death by ferroptosis and loss of mitochondria. The former is accomplished by exporting GPX-4 to neurons (to repair oxidized lipids), while the latter by exporting healthy mitochondria to neuronal cells (via tunneling nanotubules, extracellular vesicles, or cell–cell fusion). Astrocytes import cystine via cystine/glutamate antiporter (Xc-). Cystine is reduced to cysteine and generates glutathione and GPX-4 (which is transferred to neurons). Cysteine can also be derived from methionine, while glutathione can be generated from cysteine and glutathione disulfide (GSSC). In neurons, iron is stored in ferritin and, when needed, ferritin undergoes ferritinophagy (autophagy) in lysosomes, releasing free iron. Iron ingresses the neuron via transferrin receptor 1 (TRF-1), while the excess intracellular iron is eliminated via ferroportin.
Figure 2. Astrocytes support the postmitotic, long-lived neurons by helping them avert death by ferroptosis and loss of mitochondria. The former is accomplished by exporting GPX-4 to neurons (to repair oxidized lipids), while the latter by exporting healthy mitochondria to neuronal cells (via tunneling nanotubules, extracellular vesicles, or cell–cell fusion). Astrocytes import cystine via cystine/glutamate antiporter (Xc-). Cystine is reduced to cysteine and generates glutathione and GPX-4 (which is transferred to neurons). Cysteine can also be derived from methionine, while glutathione can be generated from cysteine and glutathione disulfide (GSSC). In neurons, iron is stored in ferritin and, when needed, ferritin undergoes ferritinophagy (autophagy) in lysosomes, releasing free iron. Iron ingresses the neuron via transferrin receptor 1 (TRF-1), while the excess intracellular iron is eliminated via ferroportin.
Jcm 13 01691 g002
Figure 3. The lipid bilayer of neuronal membrane is easily oxidated when intracellular iron is upregulated. Oxysterols, including 7-Ketocholesterol (a toxic oxide), and oxidated phospholipids alter the biophysical properties of cell membranes, disrupting neurotransmission. In addition, oxidized lipids activate 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 3. The lipid bilayer of neuronal membrane is easily oxidated when intracellular iron is upregulated. Oxysterols, including 7-Ketocholesterol (a toxic oxide), and oxidated phospholipids alter the biophysical properties of cell membranes, disrupting neurotransmission. In addition, oxidized lipids activate 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.
Jcm 13 01691 g003
Figure 4. Phenazine vs. phenothiazine: similarities and differences.
Figure 4. Phenazine vs. phenothiazine: similarities and differences.
Jcm 13 01691 g004
Table 1. SCZ-relevant antioxidants and sources.
Table 1. SCZ-relevant antioxidants and sources.
AntioxidantsSourceReferences
LycopeneGrape skin, guava, grapefruit, blueberries, tomatoes[193]
ApigeninCabbage, blueberries, acai berries[194]
Phenolic acidOilseeds, cereals, grains[195]
Curcuminchicken, beef, tofu, vegetables[196]
Epigallocatechin gallateApples, blackberries, broad beans, cherries, black grapes, pears, raspberries, and chocolate[197]
BerberineOregon grape, phellodendron, and tree turmeric.[198]
QuercetinFruits, apples, onions, parsley, sage, tea, and red wine[199]
KempferolFruits and vegetables.[200]
TocopherolsOilseed, cereals, eggs, deary products[201]
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.; Thomas, K.A.; Ogunjale, I.A.; Jafri, N.; Bota, P.G. Insomnia in Forensic Detainees: Is Salience Network the Common Pathway for Sleep, Neuropsychiatric, and Neurodegenerative Disorders? J. Clin. Med. 2024, 13, 1691. https://doi.org/10.3390/jcm13061691

AMA Style

Sfera A, Thomas KA, Ogunjale IA, Jafri N, Bota PG. Insomnia in Forensic Detainees: Is Salience Network the Common Pathway for Sleep, Neuropsychiatric, and Neurodegenerative Disorders? Journal of Clinical Medicine. 2024; 13(6):1691. https://doi.org/10.3390/jcm13061691

Chicago/Turabian Style

Sfera, Adonis, Kyle A. Thomas, Isaac A. Ogunjale, Nyla Jafri, and Peter G. Bota. 2024. "Insomnia in Forensic Detainees: Is Salience Network the Common Pathway for Sleep, Neuropsychiatric, and Neurodegenerative Disorders?" Journal of Clinical Medicine 13, no. 6: 1691. https://doi.org/10.3390/jcm13061691

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