Next Article in Journal
Causal Inference of Central Nervous System-Regulated Hormones in COVID-19: A Bidirectional Two-Sample Mendelian Randomization Study
Previous Article in Journal
Endometriosis in Adolescents: Diagnostics, Clinical and Laparoscopic Features
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Perspective

Supplementary Pharmacotherapy for the Behavioral Abnormalities Caused by Stressors in Humans, Focused on Post-Traumatic Stress Disorder (PTSD)

Department of Medicine, University of California, 2069 Filbert Street, San Francisco, CA 94123, USA
J. Clin. Med. 2023, 12(4), 1680; https://doi.org/10.3390/jcm12041680
Submission received: 14 January 2023 / Revised: 17 February 2023 / Accepted: 18 February 2023 / Published: 20 February 2023
(This article belongs to the Section Mental Health)

Abstract

:
Used as a supplement to psychotherapy, pharmacotherapy that addresses all of the known metabolic and genetic contributions to the pathogenesis of psychiatric conditions caused by stressors would require an inordinate number of drugs. Far simpler is to address the abnormalities caused by those metabolic and genetic changes in the cell types of the brain that mediate the behavioral abnormality. Relevant data regarding the changed brain cell types are described in this article and are derived from subjects with the paradigmatic behavioral abnormality of PTSD and from subjects with traumatic brain injury or chronic traumatic encephalopathy. If this analysis is correct, then therapy is required that benefits all of the affected brain cell types; those are astrocytes, oligodendrocytes, synapses and neurons, endothelial cells, and microglia (the pro-inflammatory (M1) subtype requires switching to the anti-inflammatory (M2) subtype). Combinations are advocated using several drugs, erythropoietin, fluoxetine, lithium, and pioglitazone, that benefit all of the five cell types, and that should be used to form a two-drug combination, suggested as pioglitazone with either fluoxetine or lithium. Clemastine, fingolimod, and memantine benefit four of the cell types, and one chosen from those could be added to the two-drug combination to form a three-drug combination. Using low doses of chosen drugs will limit both toxicity and drug-drug interactions. A clinical trial is required to validate both the advocated concept and the choice of drugs.

1. Introduction

Stressors are widely recognized as causing such behavioral abnormalities as excessive irritability, anger, impatience, fear, depression, and anhedonia. Although psychotherapy would be the usual approach to treatment, it is not always successful; yet, there is no generally-approved pharmacotherapy for use as a supplement to psychotherapy. The myriad genetic and metabolic abnormalities that are either associated with or result from stress would require a suffocating, impractical number of drugs for their reversal. However, the task of formulating pharmacotherapy for stress-related behavioral abnormalities may be simplified if one considers that any psychiatric condition ultimately must reflect changes in the function within the brain of one or more of its cell types, i.e., astrocytes, oligodendrocytes, neurons, endothelial cells, and microglia [1], some of which (neurons and astrocytes) also affect the hypothalamus-pituitary-adrenal axis (HPA) [2]. In another article, the formulation of pharmacotherapy in general for patients with major psychiatric conditions is discussed [1]; here, the focus is on those patients whose disturbed behavior results from stressors. Post-Traumatic Stress Disorder (PTSD) is a convenient surrogate for approaching treatment for the behavioral abnormalities caused in humans by stressors because PTSD has been extensively studied so that its underlying molecular biology is known and provides a rational approach to pharmacotherapy that may provide benefit as a supplement psychotherapy. In addition to PTSD, there are useful data in humans with chronic traumatic encephalopathy (CTE) or traumatic brain injury (TBI) that may be taken into account. Based upon data obtained in humans with PTSD, CTE, and TBI, this article will examine the formulation of pharmacotherapy that may be useful for those patients with aberrant behavior caused by stressors and whose response to psychotherapy is inadequate. After a brief summary of the multiple metabolic and genetic factors that participate in the pathogenesis of PTSD, this article will describe the roles of brain cell types. Results from rodent studies are avoided for reasons discussed in another article, but when mentioned, they will have human correlates [1].

2. Metabolic and Genetic Changes in PTSD, TBI and CTE

Peripheral and central levels of corticotropin-releasing hormone, which represents the initiating step in the activation of the hypothalamic/pituitary/adrenal (HPA) axis, are elevated, and enhanced glucocorticoid negative feedback on the HPA axis is consistently reported; as a result, individuals with PTSD have elevated levels of the glucocorticoid receptor (GR), and enhanced glucocorticoid sensitivity [3]. The FK506 binding protein 5 (FKBP5) is a cochaperone of the steroid receptor complex, inhibits GR ligand binding and nuclear translocation of GRs, and thus regulates the sensitivity of the GR. FKBP5 is decreased in PTSD. Binder et al. found that child abuse and other childhood traumas predicted the level of adult PTSD symptomatology, and four SNPs in the FKBP5 locus significantly interacted with the severity of childhood trauma to predict the level of adult PTSD symptoms [4].
PTSD also has an associated, significantly elevated risk for metabolic syndrome and its individual components, i.e., obesity, insulin resistance and diabetes, hypertension, and dyslipidemia.
Swanberg et al. reviewed 32 studies using proton magnetic resonance spectroscopy (1H MRS) in subjects with PTSD, which showed the hippocampus and anterior cingulate cortex as having significant decreases in N-acetyl aspartate (NAA); since NAA is the second most abundant amino acid in the brain and is involved in protein synthesis, its decrement in PTSD is deleterious [5].
Multiple reports describe a genetic background for PTSD. That may be via several pathways, including the N-methyl-D-aspartate receptor (NMDAR), the dopaminergic pathway, the serotoninergic system, the adrenocorticotropic axis, and pathways affecting the number or functions of brain cell types. Because of these multiple effects, the overall significance of genetic participation in the pathogenesis of PTSD is difficult to summarize. For a comprehensive review, see Ref. [6]; some illustrative examples follow.
The NMDAR ligates glutamate, which is an excitatory neurotransmitter that, in excess, causes neurotoxicity. This receptor is composed of several subunits, and it has as a co-receptor a glycine binding site that modulates the permeability of the NMDAR. The gene GRIN3B expresses the NMDAR subunit 3B. Lori et al. studied 377 patients who had been seen in an Emergency Department immediately after their exposure to trauma which, for 70.3%, was a road accident [7]. During 1, 3, 6, and 12 months after the trauma, blood levels of the mRNA transcribed from GRIN3B were increased in those who developed a future, non-remitting PTSD as compared with levels seen in those who did not develop PTSD. GRIN3B is found, particularly in the neocortex and hippocampus.
Considering the dopaminergic pathway, the A1 allele of the type-2 dopaminergic receptor is associated with comorbidities between PTSD and anxiety, social change and depression [8].
Regarding noradrenergic neuromodulation, there is an interaction between the polymorphism of GABRA2 and the occurrence of PTSD; and there was an association between the number of traumatic events and the V158M allele of the gene coding for catecholamine-o-methyltransferase [8].
With respect to the serotonergic pathway, several studies have found that a high risk of developing PTSD is associated with a specific polymorphism in the promoter region of the serotonin transporter gene SLC6A4 [6].
As a final example, the V66M mutation in the BDNF gene was seen in 33.3% of individuals with PTSD as compared to 17.5% in non-PTSD controls [6]; the V66M mutation affects localization of brain-derived neurotrophic factor (BDNF) and thus affects neuronal cell function [9].
A notable difficulty is that the results in published studies are often conflicting, exemplified by reports from Logue et al., who analyzed blood samples for differentially expressed genes from 115 cases of PTSD and 28 controls, using 10,264 probes for genes or gene transcripts [10]. They found 41 genes that were differentially expressed by the PTSD cases; they then used two replication cohorts to reanalyze for those 41 and now could demonstrate only seven as being significantly associated (p < 0.05): but of those seven, only one survived correction for multiple testing. That gene was ATP6AP1L, which is not one of three (BDNF, NA3C1, TXNRD1) that Logue et al. indicated as having been reported by other investigators. Six years later, Logue et al. reported further studies, this time involving 73 cases of PTSD alone, 32 with PTSD plus depression, 32 with depression alone, and 44 controls [11]. They performed 190,346 single-gene analyses. After strict correction for multiple testing, they identified 21 genes differentially expressed by PTSD. Strikingly, ATP6AP1L was not among the 21. Those 21 included three down-regulated genes for oligodendrocytes (MBP, MOBP, ERMN) and two dysregulated genes for microglia. They used two different methods to find correlations of cell types with PTSD: the first method showed high correlations (r > 0.90) for oligodendrocytes and astrocytes; the second method showed zero correlations. It is interesting that the gene for endothelin 1 was up-regulated in the dorsolateral (dl) prefrontal cortex (PFC) of PTSD cases because endothelin 1 causes vasoconstriction, which would be deleterious but it has also been reported to correlate positively, which would be beneficial, with numbers of oligodendrocyte precursor cells (OPC) in humans with strokes and leukoencephalopathy [12].
In sum, there is poor reproducibility of the multiple genetic findings in PTSD, but even were therapies available to modify the effects of those genes, that would require too many drugs and would be impractical. As by pointed out by Willsey et al., neuronal circuits exhibit emergent behavior that reflects the collective properties of multiple cell types [13]. A simpler and more available solution to the problem of pharmacotherapy for PTSD is to address whatever effect these genes exert on the changes seen in brain cell types of persons with PTSD.

3. Changes in Brain Cell Types in PTSD, CTE, and TBI

The processes of astrocytes wrap around and affect axons and cerebral microvessels, and they also supply growth factors for oligodendrocytes that promote the proliferation of OPCs. Thinner myelin ensues when contact between astrocytes and oligodendrocytes is lost because myelination derives from oligodendrocytes [14]. There are several ways by which astrocytes are crucial for neurons: (1) they control glutamate clearance via the excitatory amino acid transporter (EEAT) and prevent excitatory neurotoxicity; (2) they synthesize and release BDNF; (3) they control ion (K+, Ca2+, Cl) homeostasis in the neuropil; (4) they produce a Ca2+ binding protein, S100B, which controls neuron firing [15]; (5) via the water channel aquaporin-4 (AQP4) in the end feet of their processes, astrocytes control water transport in the brain and, therefore, entry of drugs into the brain. Astrocytes also produce vascular-derived growth factor (VEGF), through which they benefit cerebral microcirculation [16].
MAO-B is a monoamine oxidase-B enzyme that is primarily located in astrocytes. Gill et al. used positron emission tomography with the MAO-B probe [11C]SL25.1188 to assess astrocyte levels, and coincidentally of MAO-B, and found both of them decreased in subjects with PTSD as compared with controls [17].
Wu et al. found that single, prolonged stress (SPS) caused prolonged extinction of contextual fear memory that correlated with increased hippocampal autophagy; and also found that preventing autophagy by astrocytes promoted the extinction of contextual fear memory, which for PTSD would be beneficial [18]. Lei et al. used SPS in rats as a model for PTSD and also saw that it increased autophagy by astrocytes [19], which provides benefits by clearing disease-related proteins in astrocytes and maintaining astrocyte function [20]. After unilateral TBI, an analog of PTSD, activated astrocytes were seen not only ipsilaterally but also in the contralateral neocortex and cingulate cortex (CC), which is important insofar as it shows that a localized TBI may cause a diffuse brain reaction [21].
Thus, data show that astrocytes have substantial importance in PTSD, which should benefit from inducing their increase.

3.1. Oligodendrocytes

While under optimal conditions, oligodendrocytes use glycolysis, under metabolic stress, as in PTSD, they have reduced glycolysis and impaired function [22]. Among seven genes identified by Wingo et al., that contribute to the pathogenesis of PTSD, three were expressed by oligodendrocyte precursor cells (OPC) [23]. The fibroblast growth factor 17 (Fgf17) is involved in this: Fgf17 infusion induced OPC proliferation [24]; its receptor is FGFR3, and FGFR3-deficient mice had reduced numbers of differentiated oligodendrocytes in the forebrain, cerebellum, hindbrain, and spinal cord and, in parallel, myelination was delayed [25]. Further, the knock-out of the Fgf17 gene in mice created deficits in the results of tests for social recognition [26], which would predispose to PTSD because social recognition protects against the development of PTSD [27]. Another linkage is because the Fgf17 gene controls the size of the dorsal prefrontal cortex (dlPFC), which is crucially relevant to behaviors that are seen in PTSD [28]. In brief, the data show the relationship between PTSD, oligodendrocytes, and FGF17.
From eight individuals with neuropathologically confirmed CTE and eight age- and sex-matched controls, Chancellor et al. studied postmortem white matter from the dlPFC [29]. Using the marker genes PLP1, MBP, and CLDN11 to identify mature oligodendrocytes and VCAN and PDGFRα to identify OPC, they found that the total number of oligodendrocyte lineage cells in the CTE white matter was decreased by 41%. Those were chronic cases; acute cases have shown loss of mature oligodendrocytes but a reactive increase in their precursors. Thus, Flygt et al. used brain tissue from ten patients with severe, acute TBI and from autopsies of five age-matched controls; the TBI brains had increased apoptosis of oligodendrocytes (p < 0.05) and a striking, inverse correlation with the time since injury (r = −0.8, p < 0.05) [30]. Another report showed that TBI brains had reduced myelin basic protein (MBP) [31]. Lotocki et al. confirmed this by showing that TBI in rats led to a reduced number and function of oligodendrocytes [32]. One may infer that persistently reduced axonal myelin due to TBI must be due to a defective response from myelinating oligodendrocytes since oligodendrocyte formation leading to myelination is expected during brain repair [33]. Illustrating this, the cortical brain levels of MBP decreased initially in 157 persons with TBI, then gradually increased during survival [34]. A reasonable interpretation is that the trauma caused initial axonal damage and myelin loss and that oligodendrocyte formation during the period of recovery caused myelination and increased MBP.
Changes in axonal myelin and changes in oligodendrocytes are concurrent; however, increased oligodendrocyte formation is expected during brain repair, so one may infer that persistently reduced axonal myelin due to TBI must be due to a defective response from myelinating oligodendrocytes [33].
Finally, it is worth noting that, for various reasons, oligodendrocytes in different brain regions may have different properties, as discussed by Power et al. in an interesting and relevant article [35].

3.2. Neurons

Imaging studies in PTSD show lowered volumes in various regions of the brain that, in part, reflects decrements in either neuronal somata or their processes. In PTSD, volume reduction in the hippocampus and ventromedial (vm) PFC may reflect a reduced cortical capacity to inhibit the fear and negative emotional responses seen in that condition. The Enigma consortium used data for 68 cortical regions across both hemispheres from 1379 PTSD patients and 2192 controls; in prefrontal regulatory regions of PTSD, they saw reduced volumes in orbitofrontal gyri (OFG), the superior temporal gyrus (STG), and the right insular, lingual and superior parietal gyri (all p values < 0.039) and those were negatively correlated with PTSD severity [36]. Studies reviewed by Karl et al. confirmed that PTSD is associated with abnormalities in multiple fronto-limbic structures and also noted that hippocampal volumetric differences co-vary with PTSD severity [37]. Hayes et al. reviewed data from 26 studies that, among other findings, showed robust hyperactivity in the dorsal anterior cingulate cortex (aCC) and that hypo-activity in the vmPFC was associated with hyperactivity in the amygdala [38]. Kasai et al. studied monozygotic twins having various combinations of combat exposure in Vietnam and the presence or absence of PTSD [39]. Compared to the combat-exposed twins without PTSD, the combat-exposed monozygotic twins with PTSD showed significant reductions of gray matter density in the right hippocampus, pregenual aCC, and bilateral insulae, suggesting that the gray matter reduction in this region represents an acquired finding in PTSD that is consistent with stress-induced loss, and that the pathogenesis of PTSD involves both genetic factors and environmental trauma.
Girgenti et al. reported differential gene expression in four PFC subregions from postmortem tissue of 52 people with PTSD and 46 matched, neurotypical comparison subjects [40]. Integration of this data set with genotype data from a large PTSD genome-wide association study identified down-regulation of the interneuron synaptic gene ELFN1 as conferring a significant liability for PTSD. ELFN1 recruits presynaptic GRM7 (metabotropic glutamate receptor 7) and GRIK2 (glutamate receptor kainate 2) [41]; both of these receptors are expressed by OPC [42]; thus, reduced ELFN1 would interfere with OPC functions and would impact myelinating, mature oligodendrocytes.
Results from the many imaging studies in PTSD showed marked variation that probably has numerous causes: genetic differences; the nature of the provoking trauma, e.g., childhood neglect and abuse, torture, and war combat; the duration of symptoms; and the severity of symptoms.

3.3. Endothelial Cells (ECs)

ECs have prime importance because they form the cerebral microvasculature that supplies oxygen and nutrients to all other brain cells. Logue et al. reported down-regulation of ECs in the ventromedial (vm) PFC, which would adversely affect cerebral microcirculation [11]. EC precursor cells in the brain increased after TBI, and microvessel density in the injured brain increased >4-fold [43]. ECs contain gene transcripts called erythropoietin-producing hepatocellular (Eph) that induce apoptosis; Assis-Nascimento et al. created Eph−/− mice and, using them, demonstrated that after TBI, EphB3 induced decreased endothelial cell survival and, thus, decreased density of blood vessels, and increased blood-brain barrier (BBB) permeability [44].
Clearly, any treatment that dilates cerebral microvessels would be beneficial. Peripheral vasodilatation can be measured easily by ultrasound, but the dilatation of cerebral microvessels cannot be so-assessed. Therefore, investigators turned to the renin-angiotensin system. Renin converts angiotensinogen to angiotensin-1 (AT-1), which is physiologically inactive but becomes converted to angiotensin-2 (AT-2) by one of the angiotensin-converting enzymes (ACE). AT-2 is the major effector peptide of the renin-angiotensin system and causes vasoconstriction by very complex mechanisms (see Haspula and Clark [45]). A highly-simplified description of those mechanisms is that AT-2 stimulates the adrenal release of vasoconstrictive catecholamines, and in the brain, it stimulates the release of vasopressin [which is antidiuretic hormone (ADH)] by the posterior pituitary. The vasopressin produces increased plasma volume, but since another effect of AT-2 is to decrease the sensitivity of the baroreceptor reflex, there is inadequate vasodilation in response to the increased plasma volume, and this produces a local rise in pressure. The antihypertensive drug, losartan, is an AT1 receptor antagonist, so Seligowski et al. examined the medications used by a large hypertensive population. Among the 397 persons using losartan for hypertension, the occurrence of PTSD was reduced by 6.9% in users versus 9.1% in non-users of losartan (p < 0.001) [46]. Concordant with this, Pacak et al. showed that exposure of animals to the stress of immobilization markedly and rapidly increased rates of synthesis, release, and metabolism of norepinephrine in the hypothalamus and amygdala [47]. Nevertheless, a placebo-controlled, randomized study in 149 patients with PTSD, reported by Stein et al., showed no benefit from using losartan at a maximum tolerated dosage for ten weeks [48].

3.4. Microglia

It is important to note that differing results for microglia may be seen in different brain regions; thus, Li et al. saw the numbers of microglia increased in the hippocampus and PFC but not in the amygdala [49]. While there are few studies in humans that are relevant to the role of microglia in PTSD’s many studies in rodents, reviewed in detail by Enomoto and Kato, affirm that activation of microglia is important for the pathogenesis of PTSD [50]. In humans with PTSD, the only relevant studies are those involving PET scans using the translocator protein (TSPO) and those involving the glucocorticoid (GC) signaling pathway; but because TSPO is expressed by both microglia and astrocytes, and that expression varies according to the time following the event causing a brain reaction, the interpretation of PET studies in humans using TSPO ligands, is difficult (see Ref. [51]). That leaves investigations of the GC signaling pathway to show microglial participation.

3.5. The Overall Changes in Brain Cell Types in PTSD, CTE and TBI

In brief, the brain cell types that are changed in PTSD are decreases in astrocytes, oligodendrocytes, neurons and synapses, and endothelial cells, and increases in microglia.

3.6. The Hypothalamus-Pituitary-Adrenal (HPA) Pathway in PTSD

The hypothalamus-pituitary-adrenal (HPA) pathway in PTSD has been extensively investigated because stress involves activations of the adrenal gland. For a complete discussion, the reader is referred to reviews by Herman et al., Dunlop and Wong, and Szeszko et al. [52,53,54]. Several brain pathways modulate HPA axis activity. The paraventricular nucleus (PVN) of the hypothalamus is one of the most important autonomic control centers in the brain and has important hormonal receptors, including those for mineralocorticoids, glucocorticoids, and thyroid; those receptors regulate the pituitary and adrenal production of their relevant hormones. The PVN is influenced by signaling molecules (orexins, adiponectin, glutamate, and GABA) but, relevant in the present context, also by neuronal afferents from multiple sources, including the brainstem (the nucleus tractus solitarius and the nucleus of the vagus nerve), PFC, hippocampus, and amygdala, and from within the hypothalamus itself. In the PVN, there are neurons that cause ACTH production by the pituitary; the hippocampus and PFC inhibit them, and the amygdala and brain stem neurons stimulate them; in addition, glucocorticoids exert negative feedback control of the HPA axis and, as shown below, that control is disturbed in PTSD. Changes in these neural circuits have a direct link to the development of PTSD. However, addressed here is the likelihood that involvement of the HPA axis occurs primarily from the antecedent effect of functional changes in the brain cells that are the present topic: clearly, neuronal circuits control the PVN, and their importance is emphasized by studies showing that inactivation of the basolateral amygdala during stress prevented the expected increase in glucocorticoid receptors in the PFC [55], and it also prevented, a reduction of astrocyte numbers that otherwise would occur during stress [56]. It may be concluded that the disturbed HPA pathway in PTSD results from changes in neuronal cells and neuronal tracts that, in turn, result from impaired function of myelinating oligodendrocytes. Besides, with respect to PTSD, chronic stress reduced the length and volume of astrocytic processes by 40.6% and 56%, respectively, and the number of their branch points was reduced by 57.8% [57], and shown above, cerebral endothelial cells are down-regulated, and activated microglia are not inhibited. All evidence shows that pharmacotherapy for PTSD should be directed to brain cell types.
Since both hydrocortisone and dexamethasone cause profound inhibition of microglial actions, their deficient action in PTSD would allow undisturbed effects of microglia [58,59]. Brain cortisol can be assessed by levels of 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1), which catalyzes the conversion of inert 11-dehydrocorticosterone to active corticosterone; higher levels of 11β-HSD reflect higher brain cortisol and thus, correlate with enhanced suppression of the hypothalamus-pituitary-adrenal (HPA) axis. Observations in PTSD consistently show enhanced sensitivity of the HPA axis negative feedback loop, such that greater cortical and limbic 11β-HSD1 expression is associated with HPA axis suppression. Bhatt et al. found that greater overall PTSD severity was associated with lower pre-frontal-limbic 11β-HSD1 availability, i.e., lower active corticosterone, which would result in reduced suppression of activated microglia [60]. Van Zuiden et al. noted that vulnerability to PTSD includes various dysregulations of the glucocorticoid signaling cascade, including low FKB5 levels in peripheral blood mononuclear cells (PBMC) prior to trauma and single nucleotide polymorphisms (SNP) in genes associated with glucocorticoid signaling such as FKBP5 [61]. Further, a meta-analysis involving 47 studies and 6008 subjects showed that those with PTSD had a lower daily cortisol output [62]. The receptor for the FK506 binding protein (FKBP5) is a co-receptor for the glucocorticoid receptor (GR); its increased levels would cause resistance to glucocorticoids which is seen in PTSD [61]. Thus, all of the factors mentioned in this paragraph show an overall decrease in the availability in the brain of glucocorticoids which associate with less inhibition of activated microglia and greater vulnerability to PTSD.

3.7. Available Drugs That Address the Changes in Brain Cell-Types That Underpin the Pathogenesis of PTSD, CTE and TBI

Fortunately, there are several drugs that correct the changes in the affected brain cell types and from which to choose a rational formulation of treatment. From the following list, it is recommended to choose combinations of drugs, using them in reduced dosages, that will minimize the likelihood of adverse events, including those from drug-drug interactions.
Highly abbreviated descriptions for likely therapeutic effects are provided here, showing either an increase that the drugs induce in the number or activation of astrocytes, oligodendrocytes, synapses and neurons, and endothelial cells; or a decrease in the number or activation of microglia.
Clemastine decreased loss of astrocytes [63]; increased postsynaptic proteins [63], muscarinic receptor, synapsin 1 and Homer 1, and improved oligodendrocyte survival/function [64,65]; it enhanced myelin repair [66] and neuronal function [67]; it enhanced myelination in the PFC [68,69]; it also enhanced visual function [65,70]. Clemastine provided mitochondrial protection [71,72]; and suppressed microglial M1 activation [73].
Erythropoietin (EPO) increased the differentiation of neuronal stem cells (NSC) into astrocytes [74], increased oligodendrocytes [74], and neurons and dendritic spines [75] via increased production of brain-derived neurotrophic factor (BDNF) and its receptor [76]; and by enhancing differentiation of NSC [77]. EPO improved the function of synapses [78,79,80], endothelial cells [81,82] and microglia [83].
Fingolimod, an agonist of sphingomyelin phosphate (S1P), produces cell proliferation [84]. In astrocytes, fingolimod activated neurotrophic genes [85] and reduced the formation of ceramide that causes apoptosis [86,87]. It activated myelinating oligodendrocytes [88], increased OPCs, myelination, and neurological function [89], and ameliorated brain demyelination [90]. It enriched synaptic genes [91], prevented synaptic toxicity [92], and reversed synaptic hypersensitivities. It produced myelin in the demyelinated brain [93] and prevented neural death from NMDA [94]. In multiple sclerosis, it improved axonal and myelin integrity [95] and, in mice, prevented demyelination [96]. It increased dendritic spines [97,98], reduced neuronal death from ROS [99], and improved mitochondrial production of ATP [100]. In microglia, fingolimod shifted M1 polarization toward M2 [101,102].
Fluoxetine induced increases in astrocytes [103], oligodendrocytes [104,105,106], neurons [104,105,106], and endothelial cells [107] and decreased microglial activation [106,108,109].
Fluoxetine activates astrocytes to produce BDNF and VEGF [16,103,110] and promotes the clearance of astrocytes with damaged mitochondria [111]. It up-regulated OPC and oligodendrocyte markers [104] and reduced oligodendrocyte senescence [112]. Fluoxetine increased neurogenesis [104,113], neuronal circuits [114], and spatial learning [115]. Neurons deprived of glucose and oxygen had increased survival with fluoxetine [116]. It increased vasodilatation via differentiation and proliferation of endothelial cells and decreased arteriolar tone, and attenuated EC death and disruption of the BBB [107,109,117]. In microglia, it attenuated NADPH oxidase activation, production of ROS and reactive nitrogen species, and down-regulated M1 and up-regulated M2 activation [108,118,119].
Several clinical trials in patients with PTSD have shown the therapeutic benefits of fluoxetine. Martenyi et al. reported a double-blind, randomized, placebo-controlled study conducted primarily in war-torn countries [120]. Among its 301 participants, 48% were exposed to a combat-related traumatic episode. Patients were randomly assigned to 12 weeks of acute treatment with fluoxetine, 20 to 80 mg/day (n = 226), or placebo (n = 75). Compared with placebo, fluoxetine was associated with a greater improvement from baseline in the primary efficacy measurement, which was the Treatment Outcome PTSD rating scale (p = 0.006), and also in significantly greater improvements of the scores in the Clinician-Administered PTSD Scale (CAPS), Clinical Global Impressions (CGI)-Severity of Illness scale, the CGI-Improvement scale, the Montgomery-Asberg Depression Rating Scale, and the Hamilton Rating Scale for Anxiety. The mean fluoxetine dose at the endpoint was 57 mg. There were no clinically significant safety differences. In a subsequent article, Martenyi and Soldatenkova reported on the efficacy of maintenance treatment with fluoxetine as compared with a placebo in 144 veterans of the war in Yugoslavia who had been diagnosed with PTSD [121]. In addition to the rating scales mentioned above, this trial used the Davidson Trauma Scale, which is a standard, clinical global rating scale to which are added separate measures for the three main PTSD symptom clusters (intrusion, avoidance/numbing, and hyperarousal). The risk of relapse in the placebo arm was significantly greater than in the fluoxetine arm (p = 0.048). Fluoxetine was well tolerated at a mean daily dose of 65 mg.
Smaller studies have been reported. Connor et al. randomized a civilian sample of PTSD subjects, 27 to receive fluoxetine and 27 to receive a placebo, for 12 weeks [122]. If PTSD symptoms had been present for <6 years, the response rate from fluoxetine was 93% versus 42% from placebo (p = 0.008). Two open-label trials without placebo controls have reported successful results from fluoxetine [123,124]. On the contrary, a small trial gave negative results: Hertzberg et al. enrolled twelve male veterans with PTSD in a 12 week double-blind evaluation of fluoxetine and placebo; the mean fluoxetine dose at the endpoint was 48 mg/day [125]. Only one of the six patients taking fluoxetine responded versus two of the six using a placebo.
Lithium doubled astrocytic numbers and their VEGF secretion [126]. Oligodendrocyte expression of PLP and MBP increased, improving synaptic and neuronal function [127,128,129,130,131]. By negating activation of GSK-3β, synaptic expression of PSD-95 and gephyrin were enhanced. Lithium promoted neurogenesis [131,132,133,134]; doubled BDNF levels, increased dendritic length, increased anti-apoptotic Bcl2 and Bcl-XL, and prevented neuronal death from glutamate and pro-apoptotic BAD, BAX, and caspase 3 [135]. It increased the numbers and size of neural mitochondria [136], increased antioxidants [129,136], minimized neurotoxicity from cytochrome c, and promoted mitochondrial biogenesis [137,138]. Lithium benefitted microvascularity by increasing VEGF secretion [139] and BBB integrity [140]. Lithium’s inhibition of GSK-3β reduced the production and activation of pro-inflammatory mediators [141,142,143].
Memantine induced activation of astrocytes [144] and prevented losses of oligodendrocytes [145], synapses and neurons [146,147,148], and endothelial cells [149]. It is an NMDAR antagonist, enters the NMDAR’s Ca2+ channel and decreases its permeability, so it prevents neuronal excitotoxicity and death [146]. Memantine provides synaptic protection via several mechanisms: preventing cytotoxicity by blocking inhibition of a guanosine triphosphatase involved in multiple cellular processes, thus protecting against mitochondrial dysfunction causing cell death via cytochrome c release, ROS and peroxide production [150,151,152,153]. Blocking the ion channel of the acetylcholine receptors also prevents neurotoxicity [154]. Memantine benefitted brain endothelial cells and blocked disruption of the BBB [155].
Minocycline prevented oligodendrocyte toxicity caused by the deprivation of oxygen and glucose [156] and microglial-induced apoptosis [157] by minocycline preventing the inhibition of CREB’s up-regulation [158], and caused by Aβ [159]. Synaptic function improved from increased levels of PSD-95 and dendritic spines [160]. It prevented cognitive decline caused by an antagonist of the NMDAR [161]. The neuronal benefit was also from its increased expression of BDNF, CREB, and phospho-CREB [158] and the proliferation of NPCs [162]. Minocycline potentiated neurite outgrowth [163]. In a transgenic mouse model of Down’s syndrome, minocycline prevented the decline of cholinergic neurons [164]. In a list of 1040 drugs that prevent the release of cytochrome c, minocycline was the second most potent [165]; the mechanism involves inhibiting mitochondrial increases in Ca2+ concentration plus inhibition of NADH-cytochrome c reductase and cytochrome c oxidase [165,166]. Finally, minocycline prevented microglial activation [167,168].
Pioglitazone, a PPARγ agonist, prevents phosphorylation of JAK-STAT in astrocytes and thereby induces increases in neurons [169,170], oligodendrocytes [119,170], endothelial cells [171] and decreases in microglia [172]. Ciglitazone and curcumin, also PPARγ agonists, were cytoprotective for astrocytes and reversed their decreased expression of PPARγ receptor after exposure to Aβ25-35 [173,174]; and ciglitazone increased the formation of oligodendrocyte progenitors [170]. Pioglitazone rescued the demyelination caused by anti-MOG autoantibody [175]. The promotion of OPC differentiation into mature oligodendrocytes by IL-4 was mediated by PPARγ or curcumin [176,177]. PPARγ agonism increases the neurogenic differentiation gene, NeyroD1 [170] and protects cortical neurons and axons against toxicity induced by NO or KCl-induced toxicity [169,178].
PPARγ also produces endothelial cell proliferation and angiogenesis [171,179]. In microglia, PPARγ agonists inhibit cytokine production by down-regulating proinflammatory genes [180]. In addition, rosiglitazone up-regulated M2 microglia [180].
Piracetam increased the numbers and function of astrocytes [181]. It also increased the function of synapses [182,183,184,185] and neurons [182]: it decreased neurotoxicity from deprivation of oxygen and glucose, hypoperfusion, ethanol feeding or ethanol withdrawal [183,186,187,188]. It also caused longer neurites [182]. The effects of piracetam may derive in part from the restoration of cell membrane fluidity induced by a conformational change in the phospholipids of the liposomal membrane [185].
By decreasing mitochondrial swelling and permeability caused by excessive Ca2+ opening the MPTP, piracetam improved mitochondrial membrane potential and ATP levels, shifted the balance of mitochondrial fission or fusion towards fusion, and reversed the adverse effect of pro-oxidants [189]. Additionally, it reversed the cytotoxic effects of p53 and BAX [183].
Riluzole increased both the gene for the excitatory amino acid transporter (EAAT2) and its expression, thus enhancing glutamate uptake by astrocytes and protecting against excitotoxicity of neurons [190,191]. It benefits synapses, as shown by the inhibition of voltage-activated sodium currents, which prevents the reverse operation of the Na+/Ca2+ exchanger [192], and by increasing LTP [193]. It caused a 40-fold increase in hippocampal BDNF, creating neurogenesis [194] and protecting against neural degeneration caused by ischemia [195]. N-acetylaspartate is exclusively expressed in neurons, was decreased in PTSD (see above) and was increased in the cerebral cortex by exposure to riluzole [196]. In neurons, riluzole also decreased oxidative stress, lipid peroxidation, and ATP depletion [197,198]. Microglial activation was ameliorated by riluzole [199], which also upregulated the mRNA levels of M2 markers (anti-inflammatory) and downregulated those of M1 markers (pro-inflammatory) [200].

4. Discussion

This article argues that because in behavioral reactions caused by stressors (the paradigm for which is PTSD), there are such numerous and complex metabolic and genetic abnormalities that addressing the changes that those abnormalities mediate in brain cell types is the optimum approach to formulating a rational pharmacotherapy that may be used to supplement psychotherapy. Data show that those stress reactions are associated with decreases in astrocytes, oligodendrocytes, synapses and neurons, and endothelial cells/cerebral microcirculation, and increases in microglia, particularly M2 microglia that are pro-inflammatory. Drugs that beneficially address the changes in those brain cell types include clemastine, dantrolene, erythropoietin, fingolimod, fluoxetine, lithium, memantine, minocycline, pioglitazone, piracetam, and riluzole.
The cure for PTSD requires addressing the cerebral cell types that are responsible for its pathogenesis. While it is not necessarily the case that, for individual subjects with PTSD, all five cell types are involved, for practical purposes, it is prudent to assume that they are; so, drugs that address all five cell types are the best candidates for a regimen that has the best chance for successful reversal of the behavioral problem caused by stressors. It is recommended to use combinations of two or three drugs in the above list in order to minimize dosages and the risk of both serious adverse reactions (SAE) and drug-drug interactions, including with drugs used for co-morbid conditions. The best chances would come from the use of drugs that address all five cell types. Those are erythropoietin, fluoxetine, lithium, and pioglitazone. However, unless there is a compromise of the blood-brain barrier (BBB), erythropoietin will not cross it. So, the choice of a two-drug combination must be chosen from fluoxetine, lithium and pioglitazone; however, fluoxetine and lithium potentially cause the most SAE. Therefore, a clinical trial to test whether two of the suggested drugs will, in fact, cure PTSD should administer pioglitazone plus either fluoxetine or lithium, using low dosages: pioglitazone 15 mg/day, lithium 75 mg/day, or fluoxetine 10 mg/day. Four cell types benefit from clemastine (which causes no SAE), fingolimod, and memantine; one drug from this group could be added to the two-drug combination to form a three-drug combination. Clinical trials would demonstrate the validity of both the proposed concept and the choice of drugs for treatment and would also show the percentage of patients benefitting from the chosen combination of drugs. It is important to note that this use of the suggested drugs would be off-label and that a clinical trial is required to establish their efficacy for the reversal of PTSD.
Table 1 provides an abbreviated summary of some of the pharmacodynamic and pharmacokinetic issues regarding the suggested drugs.
It must also be emphasized that urine tests should be obtained in appropriate patients if there is any suspicion of self-treatment by the use of alcohol, cannabis, or illegally-obtained drugs, and if other providers have been involved, they should be contacted for whatever useful information that they had obtained.
In a clinical trial that randomly assigns participants to receive active drugs or a placebo, the calculation of the required number of participants would be based upon the primary objective showing ≥50% more reversal of PTSD in those using active drugs than those participants randomized to take a placebo. The number in each group would be calculated on the basis of an annual drop-out rate of 10%. The planned duration of treatment would be 48 weeks with an option to extend for a further 48 weeks if recommended by the study board (DSMB), which will meet every 24 weeks to review the data. Participants would be aged ≥18, with a diagnosis of PTSD made according to the current diagnostic and statistical manual of mental disorders (DSM). Exclusions would be individuals with other active psychiatric diagnoses, particularly MDD and substance abuse.

5. Conclusions

  • Data show many metabolic and genetic associates of behavioral conditions due to stressors, for which PTSD is paradigmatic. Addressing all of them would require an unacceptable number of drugs. However, the effects of those metabolic and genetic associates affect brain cell types, whose changed functions result in behavioral disturbance;
  • In PTSD, there is an impairment in the numbers and functions of all of the five brain cell types. Therefore, optimum pharmacotherapy for PTSD must benefit astrocytes, oligodendrocytes, neurons, endothelial cells, and microglia;
  • Each of fluoxetine, lithium, and pioglitazone corrects the changed functions of all five cell types. Administering two of them together should benefit all patients with PTSD, including those having psychotherapy. For a two-drug regimen, it is recommended to use low dosages of pioglitazone paired with low doses of either lithium or fluoxetine. One additional drug, taken from among clemastine, fingolimod, and memantine., would form a three-drug regimen;
  • The cure of PTSD by the above groups of drugs in patients with an inadequate response to psychotherapy should be validated by clinical trials.

Funding

This research received no external funding.

Conflicts of Interest

The author declares no conflict of interest.

References

  1. Fessel, J. Formulating treatment of major psychiatric disorders: Algorithm targets the dominantly affected brain cell-types. Discov. Ment. Health 2023, 3, 3. [Google Scholar] [CrossRef]
  2. Smith, S.M.; Vale, W.W. The role of the hypothalamic-pituitary-adrenal axis in neuroendocrine responses to stress. Dialogues Clin. Neurosci. 2006, 8, 383–395. [Google Scholar] [CrossRef] [PubMed]
  3. Michopoulos, V.; Vester, A.; Neigh, G. Posttraumatic stress disorder: A metabolic disorder in disguise? Exp. Neurol. 2016, 284, 220–229. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Binder, E.B.; Bradley, R.G.; Liu, W.; Epstein, M.P.; Deveau, T.C.; Mercer, K.B.; Tang, Y.; Gillespie, C.F.; Heim, C.M.; Nemeroff, C.B.; et al. Association of FKBP5 polymorphisms and childhood abuse with risk of posttraumatic stress disorder symptoms in adults. JAMA 2008, 299, 1291–1305. [Google Scholar] [CrossRef] [Green Version]
  5. Swanberg, K.M.; Campos, L.; Abdallah, C.G.; Juchem, C. Proton Magnetic Resonance Spectroscopy in Post-Traumatic Stress Disorder—Updated Systematic Review and Meta-Analysis. Chronic Stress. 2022, 6, 24705470221128004. [Google Scholar] [CrossRef]
  6. Banerjee, S.B.; Morrison, F.G.; Ressler, K.J. Genetic approaches for the study of PTSD: Advances and challenges. Neurosci. Lett. 2017, 649, 139–146. [Google Scholar] [CrossRef] [Green Version]
  7. Lori, A.; Schultebraucks, K.; Galatzer-Levy, I.; Daskalakis, N.P.; Katrinli, S.; Smith, A.K.; Myers, A.J.; Richholt, R.; Huentelman, M.; Guffanti, G.; et al. Transcriptome-wide association study of post-trauma symptom trajectories identified GRIN3B as a potential biomarker for PTSD development. Neuropsychopharmacology 2021, 46, 1811–1820. [Google Scholar] [CrossRef]
  8. Auxéméry, Y. Posttraumatic stress disorder (PTSD) as a consequence of the interaction between an individual genetic susceptibility, a traumatogenic event and a social context. Encephale 2012, 38, 373. [Google Scholar] [CrossRef]
  9. Reichardt, L.F. Neurotrophin-regulated signalling pathways. Philos. Trans. R. Soc. B Biol. Sci. 2006, 361, 1545–1564. [Google Scholar] [CrossRef] [Green Version]
  10. Logue, M.W.; Smith, A.K.; Baldwin, C.; Wolf, E.J.; Guffanti, G.; Ratanatharathorn, A.; Stone, A.; Schichman, S.A.; Humphries, D.; Binder, E.B.; et al. An analysis of gene expression in PTSD implicates genes involved in the glucocorticoid receptor pathway and neural responses to stress. Psychoneuroendocrinology 2015, 57, 1–13. [Google Scholar] [CrossRef] [Green Version]
  11. Logue, M.W.; Zhou, Z.; Morrison, F.G.; Wolf, E.J.; Daskalakis, N.P.; Chatzinakos, C.; Georgiadis, F.; Labadorf, A.T.; Girgenti, M.J.; Young, K.A.; et al. Gene expression in the dorsolateral and ventromedial prefrontal cortices implicates immune-related gene networks in PTSD. Neurobiol. Stress 2021, 15, 100398. [Google Scholar] [CrossRef] [PubMed]
  12. Adams, K.L.; Riparini, G.; Banerjee, P.; Breur, M.; Bugiani, M.; Gallo, V. Endothelin-1 signaling maintains glial progenitor proliferation in the postnatal subventricular zone. Nat. Commun. 2020, 11, 2138. [Google Scholar] [CrossRef]
  13. Willsey, A.J.; Morris, M.T.; Wang, S.; Willsey, H.R.; Sun, N.; Teerikorpi, N.; Baum, T.B.; Cagney, G.; Bender, K.J.; Desai, T.A.; et al. The psychiatric cell map initiative: A convergent systems biological approach to illuminating key molecular pathways in neuropsychiatric disorders. Cell 2018, 174, 505–520. [Google Scholar] [CrossRef] [Green Version]
  14. Tognatta, R.; Karl, M.T.; Fyffe-Maricich, S.L.; Popratiloff, A.; Garrison, E.D.; Schenck, J.K.; Abu-Rub, M.; Miller, R.H. Astrocytes are required for oligodendrocyte survival and maintenance of myelin compaction and integrity. Front. Cell. Neurosci. 2020, 14, 74. [Google Scholar] [CrossRef] [Green Version]
  15. Li, B.; Zhang, D.; Verkhratsky, A. Astrocytes in Post-traumatic Stress Disorder. Neurosci. Bull. 2022, 38, 953–965. [Google Scholar] [CrossRef] [PubMed]
  16. Allaman, I.; Fiumelli, H.; Magistretti, P.J.; Martin, J.-L. Fluoxetine regulates the expression of neurotrophic/growth factors and glucose metabolism in astrocytes. Psychopharmacology 2011, 216, 75–84. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Gill, T.; Watling, S.E.; Richardson, J.D.; McCluskey, T.; Tong, J.; Meyer, J.H.; Warsh, J.; Jetly, R.; Hutchison, M.G.; Rhind, S.G.; et al. Imaging of astrocytes in posttraumatic stress disorder: A PET study with the monoamine oxidase B radioligand [11C] SL25. 1188. Eur. Neuropsychopharmacol. 2022, 54, 54–61. [Google Scholar] [CrossRef]
  18. Wu, Z.-M.; Yang, L.-H.; Cui, R.; Ni, G.-L.; Wu, F.-T.; Liang, Y. Contribution of hippocampal 5-HT3 receptors in hippocampal autophagy and extinction of conditioned fear responses after a single prolonged stress exposure in rats. Cell. Mol. Neurobiol. 2017, 37, 595–606. [Google Scholar] [CrossRef]
  19. Lei, X.; Yuan, Y.; Zou, Q. The role and mechanism of gastrodin in the medial prefrontal cortex autophagy of PTSD rats. Int. J. Clin. Exp. Pathol. 2020, 13, 989–994. [Google Scholar]
  20. Sung, K.; Jimenez-Sanchez, M. Autophagy in astrocytes and its implications in neurodegeneration. J. Mol. Biol. 2020, 432, 2605–2621. [Google Scholar] [CrossRef]
  21. Zhao, J.; Wang, B.; Huang, T.; Guo, X.; Yang, Z.; Song, J.; Zhang, M. Glial response in early stages of traumatic brain injury. Neurosci. Lett. 2019, 708, 134335. [Google Scholar] [CrossRef] [PubMed]
  22. Rao, V.T.S.; Khan, D.; Cui, Q.-L.; Fuh, S.-C.; Hossain, S.; Almazan, G.; Multhaup, G.; Healy, L.M.; Kennedy, T.E.; Antel, J.P. Distinct age and differentiation-state dependent metabolic profiles of oligodendrocytes under optimal and stress conditions. PLoS ONE 2017, 12, e0182372. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Wingo, T.S.; Gerasimov, E.S.; Liu, Y.; Duong, D.M.; Vattathil, S.M.; Lori, A.; Gockley, J.; Breen, M.S.; Maihofer, A.X.; Nievergelt, C.M.; et al. Integrating human brain proteomes with genome-wide association data implicates novel proteins in post-traumatic stress disorder. Mol. Psychiatry 2022, 27, 3075–3084. [Google Scholar] [CrossRef] [PubMed]
  24. Iram, T.; Kern, F.; Kaur, A.; Myneni, S.; Morningstar, A.R.; Shin, H.; Garcia, M.A.; Yerra, L.; Palovics, R.; Yang, A.C.; et al. Young CSF restores oligodendrogenesis and memory in aged mice via Fgf17. Nature 2022, 605, 509–515. [Google Scholar] [CrossRef] [PubMed]
  25. Oh, L.Y.S.; Denninger, A.; Colvin, J.S.; Vyas, A.; Tole, S.; Ornitz, D.; Bansal, R. Fibroblast growth factor receptor 3 signaling regulates the onset of oligodendrocyte terminal differentiation. J. Neurosci. 2003, 23, 883–894. [Google Scholar] [CrossRef] [Green Version]
  26. Scearce-Levie, K.; Roberson, E.D.; Gerstein, H.; Cholfin, J.A.; Mandiyan, V.S.; Shah, N.M.; Rubenstein, J.L.R.; Mucke, L. Abnormal social behaviors in mice lacking Fgf17. Genes Brain Behav. 2008, 7, 344–354. [Google Scholar] [CrossRef]
  27. Van der Velden, P.G.; Oudejans, M.; Das, M.; Bosmans, M.W.; Maercker, A. The longitudinal effect of social recognition on PTSD symptomatology and vice versa: Evidence from a population-based study. Psychiatry Res. 2019, 279, 287–294. [Google Scholar] [CrossRef] [PubMed]
  28. Cholfin, J.A.; Rubenstein, J.L.R. Patterning of frontal cortex subdivisions by Fgf17. Proc. Natl. Acad. Sci. USA 2007, 104, 7652–7657. [Google Scholar] [CrossRef] [Green Version]
  29. Chancellor, K.B.; Chancellor, S.E.; Duke-Cohan, J.E.; Huber, B.R.; Stein, T.D.; Alvarez, V.E.; Okaty, B.W.; Dymecki, S.M.; McKee, A.C. Altered oligodendroglia and astroglia in chronic traumatic encephalopathy. Acta Neuropathol. 2021, 142, 295–321. [Google Scholar] [CrossRef]
  30. Flygt, J.; Gumucio, A.; Ingelsson, M.; Skoglund, K.; Holm, J.; Alafuzoff, I.; Marklund, N. Human traumatic brain injury results in oligodendrocyte death and increases the number of oligodendrocyte progenitor cells. J. Neuropathol. Exp. Neurol. 2016, 75, 503–515. [Google Scholar] [CrossRef] [Green Version]
  31. Bohnert, S.; Wirth, C.; Schmitz, W.; Trella, S.; Monoranu, C.-M.; Ondruschka, B.; Bohnert, M. Myelin basic protein and neurofilament H in postmortem cerebrospinal fluid as surrogate markers of fatal traumatic brain injury. Int. J. Leg. Med. 2021, 135, 1525–1535. [Google Scholar] [CrossRef] [PubMed]
  32. Lotocki, G.; Vaccari, J.D.R.; Alonso, O.; Molano, J.S.; Nixon, R.; Safavi, P.; Dietrich, W.D.; Bramlett, H.M. Oligodendrocyte vulnerability following traumatic brain injury in rats. Neurosci. Lett. 2011, 499, 143–148. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Zhang, R.; Chopp, M.; Zhang, Z.G. Oligodendrogenesis after cerebral ischemia. Front. Cell. Neurosci. 2013, 7, 201. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Shang, Y.; Wang, Y.; Guo, Y.; Ren, L.; Zhang, X.; Wang, S.; Zhang, C.; Cai, J. Analysis of the risk of traumatic brain injury and evaluation neurogranin and myelin basic protein as potential biomarkers of traumatic brain injury in postmortem examination. Forensic Sci. Med. Pathol. 2022, 18, 288–298. [Google Scholar] [CrossRef]
  35. Power, J.; Mayer-Pröschel, M.; Smith, J.; Noble, M. Oligodendrocyte precursor cells from different brain regions express divergent properties consistent with the differing time courses of myelination in these regions. Dev. Biol. 2002, 245, 362–375. [Google Scholar] [CrossRef] [Green Version]
  36. Wang, X.; Xie, H.; Chen, T.; Cotton, A.S.; Salminen, L.E.; Logue, M.W.; Clarke-Rubright, E.K.; Wall, J.; Dennis, E.L.; O’Leary, B.M.; et al. Cortical volume abnormalities in posttraumatic stress disorder: An ENIGMA-psychiatric genomics consortium PTSD workgroup mega-analysis. Mol. Psychiatry 2020, 26, 4331–4343. [Google Scholar] [CrossRef]
  37. Karl, A.; Schaefer, M.; Malta, L.S.; Dörfel, D.; Rohleder, N.; Werner, A. A meta-analysis of structural brain abnormalities in PTSD. Neurosci. Biobehav. Rev. 2006, 30, 1004–1031. [Google Scholar] [CrossRef]
  38. Hayes, J.P.; Hayes, S.M.; Mikedis, A.M. Quantitative meta-analysis of neural activity in posttraumatic stress disorder. Biol. Mood Anxiety Disord. 2012, 2, 9. [Google Scholar] [CrossRef] [Green Version]
  39. Kasai, K.; Yamasue, H.; Gilbertson, M.W.; Shenton, M.E.; Rauch, S.L.; Pitman, R.K. Evidence for acquired pregenual anterior cingulate gray matter loss from a twin study of combat-related posttraumatic stress disorder. Biol. Psychiatry 2008, 63, 550–556. [Google Scholar] [CrossRef] [Green Version]
  40. Girgenti, M.J.; Traumatic Stress Brain Research Group; Wang, J.; Ji, D.; Cruz, D.A.; Stein, M.B.; Gelernter, J.; Young, K.A.; Huber, B.R.; Williamson, D.E.; et al. Transcriptomic organization of the human brain in post-traumatic stress disorder. Nat. Neurosci. 2020, 24, 24–33. [Google Scholar] [CrossRef]
  41. Stachniak, T.J.; Sylwestrak, E.L.; Scheiffele, P.; Hall, B.J.; Ghosh, A. Elfn1-Induced constitutive activation of mGluR7 determines frequency-dependent recruitment of somatostatin interneurons. J. Neurosci. 2019, 39, 4461–4474. [Google Scholar] [CrossRef] [Green Version]
  42. Luyt, K.; Varadi, A.; Molnar, E. Functional metabotropic glutamate receptors are expressed in oligodendrocyte progenitor cells. J. Neurochem. 2003, 84, 1452–1464. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Guo, X.; Liu, L.; Zhang, M.; Bergeron, A.; Cui, Z.; Dong, J.F.; Zhang, J. Correlation of CD34+ cells with tissue angiogenesis after traumatic brain injury in a rat model. J. Neurotrauma 2009, 26, 1337–1344. [Google Scholar] [CrossRef] [PubMed]
  44. Assis-Nascimento, P.; Tsenkina, Y.; Liebl, D.J. EphB3 signaling induces cortical endothelial cell death and disrupts the blood–brain barrier after traumatic brain injury. Cell Death Dis. 2018, 9, 7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Haspula, D.; Clark, M.A. Molecular basis of the brain renin angiotensin system in cardiovascular and neurologic disorders: Uncovering a key role for the astroglial angiotensin type 1 receptor AT1R. J. Pharmacol. Exp. Ther. 2018, 366, 251–264. [Google Scholar] [CrossRef] [Green Version]
  46. Seligowski, A.V.; Duffy, L.A.; Merker, J.B.; Michopoulos, V.; Gillespie, C.F.; Marvar, P.J.; Stein, M.B.; Ressler, K.J. The renin–angiotensin system in PTSD: A replication and extension. Neuropsychopharmacology 2020, 46, 750–755. [Google Scholar] [CrossRef]
  47. Pacak, K.; Palkovits, M.; Kopin, I.J.; Goldstein, D.S. Stress-induced norepinephrine release in the hypothalamic paraventricular nucleus and pituitary-adrenocortical and sympathoadrenal activity: In vivo microdialysis studies. Front. Neuroendocrinol. 1995, 16, 89–150. [Google Scholar] [CrossRef]
  48. Stein, M.B.; Jain, S.; Simon, N.M.; West, J.C.; Marvar, P.J.; Bui, E.; He, F.; Benedek, D.M.; Cassano, P.; Griffith, J.L.; et al. Randomized, placebo-controlled trial of the angiotensin receptor antagonist losartan for posttraumatic stress disorder. Biol. Psychiatry 2021, 90, 473–481. [Google Scholar] [CrossRef]
  49. Li, S.; Liao, Y.; Dong, Y.; Li, X.; Li, J.; Cheng, Y.; Cheng, J.; Yuan, Z. Microglial deletion and inhibition alleviate behavior of post-traumatic stress disorder in mice. J. Neuroinflamm. 2021, 18, 7. [Google Scholar] [CrossRef]
  50. Enomoto, S.; Kato, T.A. Involvement of microglia in disturbed fear memory regulation: Possible microglial contribution to the pathophysiology of posttraumatic stress disorder. Neurochem. Int. 2021, 142, 104921. [Google Scholar] [CrossRef]
  51. Guilarte, T.R. TSPO in diverse CNS pathologies and psychiatric disease: A critical review and a way forward. Pharmacol. Ther. 2018, 194, 44–58. [Google Scholar] [CrossRef] [PubMed]
  52. Herman, J.P.; Nawreen, N.; Smail, M.A.; Cotella, E.M. Brain mechanisms of HPA axis regulation: Neurocircuitry and feedback in context Richard Kvetnansky lecture. Stress 2020, 23, 617–632. [Google Scholar] [CrossRef]
  53. Dunlop, B.W.; Wong, A. The hypothalamic-pituitary-adrenal axis in PTSD: Pathophysiology and treatment interventions. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2018, 89, 361–379. [Google Scholar] [CrossRef]
  54. Szeszko, P.R.; Lehrner, A.; Yehuda, R. Glucocorticoids and hippocampal structure and function in PTSD. Harv. Rev. Psychiatry 2018, 26, 142–157. [Google Scholar] [CrossRef] [PubMed]
  55. Tripathi, S.J.; Chakraborty, S.; Srikumar, B.; Raju, T.; Rao, B.S. Prevention of chronic immobilization stress-induced enhanced expression of glucocorticoid receptors in the prefrontal cortex by inactivation of basolateral amygdala. J. Chem. Neuroanat. 2019, 95, 134–145. [Google Scholar] [CrossRef]
  56. Tripathi, S.J.; Chakraborty, S.; Srikumar, B.; Raju, T.; Rao, S. Inactivation of basolateral amygdala prevents stress-induced astroglial loss in the prefrontal cortex. Mol. Neurobiol. 2019, 56, 350–366. [Google Scholar] [CrossRef] [PubMed]
  57. Tynan, R.J.; Beynon, S.B.; Hinwood, M.; Johnson, S.J.; Nilsson, M.; Woods, J.J.; Walker, F.R. Chronic stress-induced disruption of the astrocyte network is driven by structural atrophy and not loss of astrocytes. Acta Neuropathol. 2013, 126, 75–91. [Google Scholar] [CrossRef]
  58. Ganter, S.; Northoff, H.; Männel, D.N.; Gebicke-Härter, P.J. Growth control of cultured microglia. J. Neurosci. Res. 1992, 33, 218–230. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Park, M.-J.; Park, H.-S.; You, M.-J.; Yoo, J.; Kim, S.H.; Kwon, M.-S. Dexamethasone Induces a Specific Form of Ramified Dysfunctional Microglia. Mol. Neurobiol. 2018, 56, 1421–1436. [Google Scholar] [CrossRef] [PubMed]
  60. Xu, B.; Lang, L.M.; Li, S.Z.; Guo, J.R.; Wang, J.F.; Yang, H.M.; Lian, S. Microglia activated by excess cortisol induce HMGB1 acetylation and neuroinflammation in the hippocampal DG region of mice following cold exposure. Biomolecules 2019, 9, 426. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Van Zuiden, M.; Kavelaars, A.; Geuze, E.; Olff, M.; Heijnen, C.J. Predicting PTSD: Pre-existing vulnerabilities in glucocorticoid-signaling and implications for preventive interventions. Brain Behav. Immun. 2013, 30, 12–21. [Google Scholar] [CrossRef] [PubMed]
  62. Morris, M.C.; Compas, B.E.; Garber, J. Relations among posttraumatic stress disorder, comorbid major depression, and HPA function: A systematic review and meta-analysis. Clin. Psychol. Rev. 2012, 32, 301–315. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Wu, W.; Zhang, X.; Zhou, J.; Yang, H.; Chen, J.; Zhao, L.; Zhong, J.; Lin, W.-J.; Wang, Z. Clemastine Ameliorates Perioperative Neurocognitive Disorder in Aged Mice Caused by Anesthesia and Surgery. Front. Pharmacol. 2021, 12, 738590. [Google Scholar] [CrossRef]
  64. Carlström, K.E.; Zhu, K.; Ewing, E.; Krabbendam, I.E.; Harris, R.A.; Falcão, A.M.; Jagodic, M.; Castelo-Branco, G.; Piehl, F. Gsta4 controls apoptosis of differentiating adult oligodendrocytes during homeostasis and remyelination via the mitochondria-associated Fas-Casp8-Bid-axis. Nat. Commun. 2020, 11, 4071. [Google Scholar] [CrossRef]
  65. Green, A.J.; Gelfand, J.M.; Cree, B.A.; Bevan, C.; Boscardin, W.J.; Mei, F.; Inman, J.; Arnow, S.; Devereux, M.; Abounasr, A.; et al. Clemastine fumarate as a remyelinating therapy for multiple sclerosis (ReBUILD): A randomised, controlled, double-blind, crossover trial. Lancet 2017, 390, 2481–2489. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Li, Z.; He, Y.; Fan, S.; Sun, B. Clemastine rescues behavioral changes and enhances remyelination in the cuprizone mouse model of demyelination. Neurosci. Bull. 2015, 31, 617–625. [Google Scholar] [CrossRef]
  67. Wang, F.; Yang, Y.-J.; Yang, N.; Chen, X.-J.; Huang, N.-X.; Zhang, J.; Wu, Y.; Liu, Z.; Gao, X.; Li, T.; et al. Enhancing Oligodendrocyte Myelination Rescues Synaptic Loss and Improves Functional Recovery after Chronic Hypoxia. Neuron 2018, 99, 689–701.e5. [Google Scholar] [CrossRef] [Green Version]
  68. Chen, J.-F.; Liu, K.; Hu, B.; Li, R.-R.; Xin, W.; Chen, H.; Wang, F.; Chen, L.; Li, R.-X.; Ren, S.-Y.; et al. Enhancing myelin renewal reverses cognitive dysfunction in a murine model of Alzheimer’s disease. Neuron 2021, 109, 2292–2307.e5. [Google Scholar] [CrossRef]
  69. Liu, J.; Dietz, K.C.; DeLoyht, J.M.; Pedreiturria, X.; Kelkar, D.; Kaur, J.; Vialou, V.; Lobo, M.K.; Dietz, D.; Nestler, E.J.; et al. Impaired adult myelination in the prefrontal cortex of socially isolated mice. Nat. Neurosci. 2012, 15, 1621–1623. [Google Scholar] [CrossRef] [Green Version]
  70. Cree, B.A.; Niu, J.; Hoi, K.K.; Zhao, C.; Caganap, S.D.; Henry, R.G.; Dao, D.Q.; Zollinger, D.R.; Mei, F.; Shen, Y.A.; et al. Clemastine rescues myelination defects and promotes functional recovery in hypoxic brain injury. Brain 2018, 141, 85–98. [Google Scholar] [CrossRef] [Green Version]
  71. Liu, N.; Wang, W.; Zhang, R.; Han, H. Effects of Clemastine Fumarate on Expression of Toll Like Receptor 4 in Lung Ischemia-Reperfusion. J. Pract. Med. 2016, 32, 2988–2991. [Google Scholar]
  72. Alberdi, E.; Sánchez-Gómez, M.V.; Matute, C. Calcium and glial cell death. J. Clin. Psychopharmacol. 2005, 38, 417–425. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Su, W.-J.; Zhang, T.; Jiang, C.-L.; Wang, W. Clemastine Alleviates Depressive-Like Behavior Through Reversing the Imbalance of Microglia-Related Pro-inflammatory State in Mouse Hippocampus. Front. Cell. Neurosci. 2018, 12, 412. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Sugawa, M.; Sakurai, Y.; Ishikawa-Ieda, Y.; Suzuki, H.; Asou, H. Effects of erythropoietin on glial cell development; oligodendrocyte maturation and astrocyte proliferation. Neurosci. Res. 2002, 4, 391–403. [Google Scholar] [CrossRef] [PubMed]
  75. Wakhloo, D.; Scharkowski, F.; Curto, Y.; Butt, U.J.; Bansal, V.; Steixner-Kumar, A.A.; Wüstefeld, L.; Rajput, A.; Arinrad, S.; Zillmann, M.R.; et al. Functional hypoxia drives neuroplasticity and neurogenesis via brain erythropoietin. Nat. Commun. 2020, 11, 1313. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Viviani, B.; Bartesaghi, S.; Corsini, E.; Villa, P.; Ghezzi, P.; Garau, A.; Galli, C.L.; Marinovich, M. Erythropoietin protects primary hippocampal neurons increasing the expression of brain-derived neurotrophic factor. J. Neurochem. 2005, 93, 412–421. [Google Scholar] [CrossRef]
  77. Osredkar, D.; Sall, J.W.; Bickler, P.E.; Ferriero, D.M. Erythropoietin promotes hippocampal neurogenesis in in vitro models of neonatal stroke. Neurobiol. Dis. 2010, 38, 259–265. [Google Scholar] [CrossRef] [Green Version]
  78. Adamcio, B.; Sargin, D.; Stradomska, A.; Medrihan, L.; Gertler, C.; Theis, F.; Zhang, M.; Müller, M.; Hassouna, I.; Hannke, K.; et al. Erythropoietin enhances hippocampal long-term potentiation and memory. BMC Biol. 2008, 6, 37. [Google Scholar] [CrossRef] [Green Version]
  79. Tazangi, P.E.; Moosavi, S.M.S.; Shabani, M.; Haghani, M. Erythropoietin improves synaptic plasticity and memory deficits by decrease of the neurotransmitter release probability in the rat model of Alzheimer’s disease. Pharmacol. Biochem. Behav. 2015, 130, 15–21. [Google Scholar] [CrossRef]
  80. Weber, A.; Maier, R.F.; Hoffmann, U.; Grips, M.; Hoppenz, M.; Aktas, A.G.; Heinemann, U.; Obladen, M.; Schuchmann, S. Erythropoietin improves synaptic transmission during and following ischemia in rat hippocampal slice cultures. Brain Res. 2002, 958, 305–311. [Google Scholar] [CrossRef]
  81. Xiong, Y.; Chopp, M.; Lee, C.-P. Erythropoietin improves brain mitochondrial function in rats after traumatic brain injury. Neurol. Res. 2009, 31, 496–502. [Google Scholar] [CrossRef] [PubMed]
  82. Chong, Z.Z.; Kang, J.-Q.; Maiese, K. Erythropoietin Is a Novel Vascular Protectant Through Activation of Akt1 and Mitochondrial Modulation of Cysteine Proteases. Circulation 2002, 106, 2973–2979. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Tamura, T.; Aoyama, M.; Ukai, S.; Kakita, H.; Sobue, K.; Asai, K. Neuroprotective erythropoietin attenuates microglial activation, including morphological changes, phagocytosis, and cytokine production. Brain Res. 2017, 1662, 65–74. [Google Scholar] [CrossRef]
  84. Choi, J.W.; Gardell, S.E.; Herr, D.R.; Rivera, R.; Lee, C.-W.; Noguchi, K.; Teo, S.T.; Yung, Y.C.; Lu, M.; Kennedy, G.; et al. FTY720 (fingolimod) efficacy in an animal model of multiple sclerosis requires astrocyte sphingosine 1-phosphate receptor 1 (S1P1) modulation. Proc. Natl. Acad. Sci. USA 2010, 108, 751–756. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Hoffmann, F.S.; Hofereiter, J.; Rübsamen, H.; Melms, J.; Schwarz, S.; Faber, H.; Weber, P.; Pütz, B.; Loleit, V.; Weber, F.; et al. Fingolimod induces neuroprotective factors in human astrocytes. J. Neuroinflamm. 2015, 12, 184. [Google Scholar] [CrossRef] [Green Version]
  86. Van Doorn, R.; Nijland, P.G.; Dekker, N.; Witte, M.E.; Lopes-Pinheiro, M.A.; van het Hof, B.; Kooij, G.; Reijerkerk, A.; Dijkstra, C.; van van der Valk, P.; et al. Fingolimod attenuates ceramide-induced blood–brain barrier dysfunction in multiple sclerosis by targeting reactive astrocytes. Acta Neuropathol. 2012, 124, 397–410. [Google Scholar] [CrossRef] [PubMed]
  87. Spiegel, S.; Milstien, S. Sphingosine-1-phosphate: Signaling inside and out. FEBS Lett. 2000, 476, 55–57. [Google Scholar] [CrossRef] [Green Version]
  88. Cruz, V.T.; Fonseca, J. Central effects of fingolimod. Rev. Neurol. 2014, 59, 121–128. [Google Scholar]
  89. Zhang, J.; Zhang, Z.G.; Li, Y.; Ding, X.; Shang, X.; Lu, M.; Elias, S.B.; Chopp, M. Fingolimod treatment promotes proliferation and differentiation of oligodendrocyte progenitor cells in mice with experimental autoimmune encephalomyelitis. Neurobiol. Dis. 2015, 76, 57–66. [Google Scholar] [CrossRef]
  90. Yasuda, K.; Maki, T.; Saito, S.; Yamamoto, Y.; Kinoshita, H.; Choi, Y.K.; Arumugam, T.V.; Lim, Y.-A.; Chen, C.L.H.; Wong, P.T.-H.; et al. Effect of fingolimod on oligodendrocyte maturation under prolonged cerebral hypoperfusion. Brain Res. 2019, 1720, 146294. [Google Scholar] [CrossRef]
  91. Yin, P.; Xue, Y.; Wang, T.; Zhong, D.; Li, G. The Therapeutic Targets of Fingolimod (FTY720) Are Involved in Pathological Processes in the Frontal Cortex of Alzheimer’s Disease Patients: A Network Pharmacology Study. Front. Aging Neurosci. 2021, 13, 6. [Google Scholar] [CrossRef]
  92. Joshi, P.; Gabrielli, M.; Ponzoni, L.; Pelucchi, S.; Stravalaci, M.; Beeg, M.; Mazzitelli, S.; Braida, D.; Sala, M.; Boda, E.; et al. Fingolimod limits acute Aβ neurotoxicity and promotes synaptic versus extrasynaptic NMDA receptor functionality in hippocampal neurons. Sci. Rep. 2017, 7, 41734. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Miron, V.E.; Ludwin, S.K.; Darlington, P.J.; Jarjour, A.A.; Soliven, B.; Kennedy, T.E.; Antel, J.P. Fingolimod (FTY720) enhances remyelination following demyelination of organotypic cerebellar slices. Am. J. Pathol. 2010, 176, 2682–2694. [Google Scholar] [CrossRef] [PubMed]
  94. Di Menna, L.; Molinaro, G.; Di Nuzzo, L.; Riozzi, B.; Zappulla, C.; Pozzilli, C.; Turrini, R.; Caraci, F.; Copani, A.; Battaglia, G.; et al. Fingolimod protects cultured cortical neurons against excitotoxic death. Pharmacol. Res. 2013, 67, 1–9. [Google Scholar] [CrossRef] [PubMed]
  95. Gurevich, M.; Waknin, R.; Stone, E.; Achiron, A. Fingolimod-improved axonal and myelin integrity of white matter tracts associated with multiple sclerosis-related functional impairments. CNS Neurosci. Ther. 2018, 24, 412–419. [Google Scholar] [CrossRef] [Green Version]
  96. Gol, M.; Ghorbanian, D.; Hassanzadeh, S.; Javan, M.; Mirnajafi-Zadeh, J.; Ghasemi-Kasman, M. Fingolimod enhances myelin repair of hippocampus in pentylenetetrazol-induced kindling model. Eur. J. Pharm. Sci. 2017, 96, 72–83. [Google Scholar] [CrossRef]
  97. Patnaik, A.; Spiombi, E.; Frasca, A.; Landsberger, N.; Zagrebelsky, M.; Korte, M. Fingolimod Modulates Dendritic Architecture in a BDNF-Dependent Manner. Int. J. Mol. Sci. 2020, 21, 3079. [Google Scholar] [CrossRef] [PubMed]
  98. Rossi, S.; Giudice, T.L.; De Chiara, V.; Musella, A.; Studer, V.; Motta, C.; Bernardi, G.; Martino, G.; Furlan, R.; Martorana, A.; et al. Oral fingolimod rescues the functional deficits of synapses in experimental autoimmune encephalomyelitis. Br. J. Pharmacol. 2011, 165, 861–869. [Google Scholar] [CrossRef] [Green Version]
  99. Candadai, A.A.; Liu, F.; Verma, A.; Adil, M.S.; Alfarhan, M.; Fagan, S.C.; Somanath, P.R.; Narayanan, S.P. Neuroprotective Effects of Fingolimod in a Cellular Model of Optic Neuritis. Cells 2021, 10, 2938. [Google Scholar] [CrossRef]
  100. Bai, B.; Lunn, S.; Avila, R.; Wang, J.; Chmura, D.; Benson, E.; Kidd, G. Fingolimod reduces axonal transection during demyelination (P1. 153). Neurology 2015, 84. [Google Scholar]
  101. Noda, H.; Takeuchi, H.; Mizuno, T.; Suzumura, A. Fingolimod phosphate promotes the neuroprotective effects of microglia. J. Neuroimmunol. 2013, 256, 13–18. [Google Scholar] [CrossRef] [PubMed]
  102. Qin, C.; Fan, W.-H.; Liu, Q.; Shang, K.; Murugan, M.; Wu, L.-J.; Wang, W.; Tian, D.-S. Fingolimod Protects Against Ischemic White Matter Damage by Modulating Microglia Toward M2 Polarization via STAT3 Pathway. Stroke 2017, 48, 3336–3346. [Google Scholar] [CrossRef] [PubMed]
  103. Kinoshita, M.; Hirayama, Y.; Fujishita, K.; Shibata, K.; Shinozaki, Y.; Shigetomi, E.; Takeda, A.; Le, H.P.N.; Hayashi, H.; Hiasa, M.; et al. Anti-Depressant Fluoxetine Reveals its Therapeutic Effect Via Astrocytes. Ebiomedicine 2018, 32, 72–83. [Google Scholar] [CrossRef] [PubMed]
  104. Czéh, B.; Müller-Keuker, J.I.; Rygula, R.; Abumaria, N.; Hiemke, C.; Domenici, E.; Fuchs, E. Chronic social stress inhibits cell proliferation in the adult medial prefrontal cortex: Hemispheric asymmetry and reversal by fluoxetine treatment. Neuropsychopharmacology 2007, 32, 1490–1503. [Google Scholar] [CrossRef] [Green Version]
  105. de Leeuw, V.C.; Hessel, E.V.; Pennings, J.L.; Hodemaekers, H.M.; Wackers, P.F.; van Oostrom, C.T.; Piersma, A.H. Differential effects of fluoxetine and venlafaxine in the neural embryonic stem cell test (ESTn) revealed by a cell lineage map. Neurotoxicology 2020, 76, 1–9. [Google Scholar] [CrossRef]
  106. Lee, J.Y.; Kang, S.R.; Yune, T.Y. Fluoxetine prevents oligodendrocyte cell death by inhibiting microglia activation after spinal cord injury. J. Neurotrauma 2015, 32, 633–644. [Google Scholar] [CrossRef] [Green Version]
  107. Ofek, K.; Schoknecht, K.; Melamed-Book, N.; Heinemann, U.; Friedman, A.; Soreq, H. Fluoxetine induces vasodilatation of cerebral arterioles by co-modulating NO/muscarinic signalling. J. Cell. Mol. Med. 2012, 16, 2736–2744. [Google Scholar] [CrossRef]
  108. Chung, Y.C.; Kim, S.R.; Park, J.Y.; Chung, E.S.; Park, K.W.; Won, S.Y.; Bok, E.; Jin, M.; Park, E.S.; Yoon, S.H.; et al. Fluoxetine prevents MPTP-induced loss of dopaminergic neurons by inhibiting microglial activation. Neuropharmacology 2011, 60, 963–974. [Google Scholar] [CrossRef]
  109. Lee, J.Y.; Lee, H.E.; Kang, S.R.; Choi, H.Y.; Ryu, J.H.; Yune, T.Y. Fluoxetine inhibits transient global ischemia-induced hippocampal neuronal death and memory impairment by preventing blood–brain barrier disruption. Neuropharmacology 2014, 79, 161–171. [Google Scholar] [CrossRef]
  110. Mercier, G.; Lennon, A.M.; Renouf, B.; Dessouroux, A.; Ramaugé, M.; Courtin, F.; Pierre, M. MAP Kinase Activation by Fluoxetine and Its Relation to Gene Expression in Cultured Rat Astrocytes. J. Mol. Neurosci. 2004, 24, 207–216. [Google Scholar] [CrossRef]
  111. Shu, X.; Sun, Y.; Sun, X.; Zhou, Y.; Bian, Y.; Shu, Z.; Ding, J.; Lu, M.; Hu, G. The effect of fluoxetine on astrocyte autophagy flux and injured mitochondria clearance in a mouse model of depression. Cell Death Dis. 2019, 10, 577. [Google Scholar] [CrossRef] [Green Version]
  112. Chao, F.L.; Zhang, Y.; Zhang, L.; Jiang, L.; Zhou, C.N.; Tang, J.; Liang, X.; Fan, J.H.; Dou, X.Y.; Tang, Y. Fluoxetine promotes hippocampal oligodendrocyte maturation and delays learning and memory decline in APP/PS1 mice. Front. Aging Neurosci. 2021, 12, 1–18. [Google Scholar] [CrossRef]
  113. Bianchi, M.; Shah, A.J.; Fone, K.C.; Atkins, A.R.; Dawson, L.A.; Heidbreder, C.A.; Hows, M.E.; Hagan, J.J.; Marsden, C.A. Fluoxetine administration modulates the cytoskeletal microtubular system in the rat hippocampus. Synapse 2009, 63, 359–364. [Google Scholar] [CrossRef]
  114. Stanisavljević, A.; Perić, I.; Gass, P.; Inta, D.; Lang, U.E.; Borgwardt, S.; Filipović, D. Fluoxetine modulates neuronal activity in stress-related limbic areas of adult rats subjected to the chronic social isolation. Brain Res. Bull. 2020, 163, 95–108. [Google Scholar] [CrossRef] [PubMed]
  115. Ma, J.; Gao, Y.; Jiang, L.; Chao, F.L.; Huang, W.; Zhou, C.N.; Tang, W.; Zhang, L.; Huang, C.X.; Zhang, Y.; et al. Fluoxetine attenuates the impairment of spatial learning ability and prevents neuron loss in middle-aged APPswe/PSEN1dE9 double transgenic Alzheimer’s disease mice. Oncotarget 2017, 8, 27676. [Google Scholar] [CrossRef] [Green Version]
  116. Dhami, K.; Churchward, M.; Baker, G.; Todd, K. Fluoxetine and citalopram decrease microglial release of glutamate and D-serine to promote cortical neuronal viability following ischemic insult. Mol. Cell. Neurosci. 2013, 56, 365–374. [Google Scholar] [CrossRef] [PubMed]
  117. Hu, Q.; Liu, L.; Zhou, L.; Lu, H.; Wang, J.; Chen, X.; Wang, Q. Effect of fluoxetine on HIF-1α-Netrin/VEGF cascade, angiogenesis and neuroprotection in a rat model of transient middle cerebral artery occlusion. Exp. Neurol. 2020, 329, 113312. [Google Scholar] [CrossRef]
  118. Su, F.; Yi, H.; Xu, L.; Zhang, Z. Fluoxetine and S-citalopram inhibit M1 activation and promote M2 activation of microglia in vitro. Neuroscience 2015, 294, 60–68. [Google Scholar] [CrossRef] [PubMed]
  119. Zhang, F.; Zhou, H.; Wilson, B.C.; Shi, J.-S.; Hong, J.-S.; Gao, H.-M. Fluoxetine protects neurons against microglial activation-mediated neurotoxicity. Park. Relat. Disord. 2012, 18, S213–S217. [Google Scholar] [CrossRef] [Green Version]
  120. Martenyi, F.; Brown, E.B.; Zhang, H.; Prakash, A.; Koke, S.C. Fluoxetine versus placebo in posttraumatic stress disorder. J. Clin. Psychiatry 2002, 63, 199–206. [Google Scholar] [CrossRef]
  121. Martenyi, F.; Soldatenkova, V. Fluoxetine in the acute treatment and relapse prevention of combat-related post-traumatic stress disorder: Analysis of the veteran group of a placebo-controlled, randomized clinical trial. Eur. Neuropsychopharmacol. 2006, 16, 340–349. [Google Scholar] [CrossRef] [PubMed]
  122. Connor, K.M.; Sutherland, S.M.; Tupler, L.A.; Malik, M.L.; Jonathan, R.; Davidson, T. Fluoxetine in post-traumatic stress disorder: Randomised, double-blind study. Br. J. Psychiatry 1999, 175, 17–22. [Google Scholar] [CrossRef] [PubMed]
  123. McDougle, C.J.; Southwick, S.M.; Charney, D.S.; St James, R.L. An open trial of fluoxetine in the treatment of posttraumatic stress disorder. J. Clin. Psychopharmacol. 1991, 11, 325–327. [Google Scholar]
  124. Nagy, L.M.; Morgan, C.A.; Southwick, S.M.; Charney, D.S. Open prospective trial of fluoxetine for posttraumatic stress disorder. J. Clin. Psychopharmacol. 1993, 13, 107–113. [Google Scholar] [CrossRef] [PubMed]
  125. Hertzberg, M.A.; Feldman, M.E.; Beckham, J.C.; Kudler, H.S.; Davidson, J.R. Lack of efficacy for fluoxetine in PTSD: A placebo con-trolled trial in combat veterans. Ann. Clin. Psychiatry 2000, 12, 101–105. [Google Scholar] [CrossRef]
  126. Rivera, A.D.; Butt, A.M. Astrocytes are direct cellular targets of lithium treatment: Novel roles for lysyl oxidase and peroxisome-proliferator activated receptor-γ as astroglial targets of lithium. Transl. Psychiatry 2019, 9, 211. [Google Scholar] [CrossRef] [Green Version]
  127. Kim, H.J.; Thayer, S.A. Lithium Increases Synapse Formation between Hippocampal Neurons by Depleting Phosphoinositides. Mol. Pharmacol. 2009, 75, 1021–1030. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Green, M.V.; Pengo, T.; Raybuck, J.D.; Naqvi, T.; McMullan, H.M.; Hawkinson, J.E.; De Velasco, E.M.F.; Muntean, B.S.; Martemyanov, K.A.; Satterfield, R.; et al. Automated Live-Cell Imaging of Synapses in Rat and Human Neuronal Cultures. Front. Cell. Neurosci. 2019, 13, 467. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Kerr, F.; Bjedov, I.; Sofola-Adesakin, O. Molecular Mechanisms of Lithium Action: Switching the Light on Multiple Targets for Dementia Using Animal Models. Front. Mol. Neurosci. 2018, 11, 297. [Google Scholar] [CrossRef] [Green Version]
  130. Voytovych, H.; Kriváneková, L.; Ziemann, U. Lithium: A switch from LTD- to LTP-like plasticity in human cortex. Neuropharmacology 2012, 63, 274–279. [Google Scholar] [CrossRef]
  131. Son, H.; Yu, I.T.; Hwang, S.J.; Kim, J.S.; Lee, S.H.; Lee, Y.S.; Kaang, B.K. Lithium enhances long-term potentiation independently of hippocampal neurogenesis in the rat dentate gyrus. J. Neurochem. 2003, 85, 872–881. [Google Scholar] [CrossRef]
  132. Emamghoreishi, M.; Keshavarz, M.; Nekooeian, A.A. Acute and chronic effects of lithium on BDNF and GDNF mRNA and protein levels in rat primary neuronal, astroglial and neuroastroglia cultures. Iran. J. Basic Med. Sci. 2015, 18, 240–246. [Google Scholar] [CrossRef] [PubMed]
  133. Chen, G.; Rajkowska, G.; Du, F.; Seraji-Bozorgzad, N.; Manji, H.K. Enhancement of hippocampal neurogenesis by lithium. J Neurochem. 2000, 75, 1729–1734. [Google Scholar] [CrossRef] [PubMed]
  134. Fiorentini, A.; Rosi, M.C.; Grossi, C.; Luccarini, I.; Casamenti, F. Lithium Improves Hippocampal Neurogenesis, Neuropathology and Cognitive Functions in APP Mutant Mice. PLoS ONE 2010, 5, e14382. [Google Scholar] [CrossRef] [Green Version]
  135. Dwivedi, T.; Zhang, H. Lithium-induced neuroprotection is associated with epigenetic modification of specific BDNF gene promoter and altered expression of apoptotic-regulatory proteins. Front. Neurosci. 2015, 8, 457. [Google Scholar] [CrossRef] [Green Version]
  136. Fornai, F.; Longone, P.; Cafaro, L.; Kastsiuchenka, O.; Ferrucci, M.; Manca, M.L.; Lazzeri, G.; Spalloni, A.; Bellio, N.; Lenzi, P.; et al. Lithium delays progression of amyotrophic lateral sclerosis. Proc. Natl. Acad. Sci. USA 2008, 105, 2052–2057. [Google Scholar] [CrossRef] [Green Version]
  137. Maurer, I.C.; Schippel, P.; Volz, H.-P. Lithium-induced enhancement of mitochondrial oxidative phosphorylation in human brain tissue. Bipolar Disord. 2009, 11, 515–522. [Google Scholar] [CrossRef]
  138. Shalbuyeva, N.; Brustovetsky, T.; Brustovetsky, N. Lithium Desensitizes Brain Mitochondria to Calcium, Antagonizes Permeability Transition, and Diminishes Cytochrome c Release. J. Biol. Chem. 2007, 282, 18057–18068. [Google Scholar] [CrossRef] [Green Version]
  139. Guo, S.; Arai, K.; Stins, M.F.; Chuang, D.-M.; Lo, E.H. Lithium Upregulates Vascular Endothelial Growth Factor in Brain Endothelial Cells and Astrocytes. Stroke 2009, 40, 652–655. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  140. Ji, Y.-B.; Gao, Q.; Tan, X.-X.; Huang, X.-W.; Ma, Y.-Z.; Fang, C.; Wang, S.-N.; Qiu, L.-H.; Cheng, Y.-X.; Guo, F.-Y.; et al. Lithium alleviates blood-brain barrier breakdown after cerebral ischemia and reperfusion by upregulating endothelial Wnt/β-catenin signaling in mice. Neuropharmacology 2021, 186, 108474. [Google Scholar] [CrossRef] [PubMed]
  141. Nassar, A.; Azab, A.N. Effects of Lithium on Inflammation. ACS Chem. Neurosci. 2014, 5, 451–458. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Dong, H.; Zhang, X.; Dai, X.; Lu, S.; Gui, B.; Jin, W.; Zhang, S.; Zhang, S.; Qian, Y. Lithium ameliorates lipopolysaccharide-induced microglial activation via inhibition of toll-like receptor 4 expression by activating the PI3K/Akt/FoxO1 pathway. J. Neuroinflamm. 2014, 11, 140. [Google Scholar] [CrossRef] [Green Version]
  143. Li, N.; Zhang, X.; Dong, H.; Zhang, S.; Sun, J.; Qian, Y. Lithium Ameliorates LPS-Induced Astrocytes Activation Partly via Inhibition of Toll-Like Receptor 4 Expression. Cell. Physiol. Biochem. 2016, 38, 714–725. [Google Scholar] [CrossRef]
  144. Torrez, V.R.; Zimmer, E.R.; Kalinine, E.; Haas, C.B.; Zenki, K.C.; Muller, A.P.; de Souza, D.O.; Portela, L.V. Memantine mediates astrocytic activity in response to excitotoxicity induced by PP2A inhibition. Neurosci. Lett. 2019, 696, 179–183. [Google Scholar] [CrossRef] [PubMed]
  145. Ma, G.; Liu, C.; Hashim, J.; Conley, G.; Morriss, N.; Meehan, W.P.; Qiu, J.; Mannix, R. Memantine mitigates oligodendrocyte damage after repetitive mild traumatic brain injury. Neuroscience 2019, 421, 152–161. [Google Scholar] [CrossRef] [PubMed]
  146. Chen, H.-S.V.; Lipton, S.A. The chemical biology of clinically tolerated NMDA receptor antagonists. J. Neurochem. 2006, 97, 1611–1626. [Google Scholar] [CrossRef] [PubMed]
  147. Dimpfel, W. Effects of memantine on synaptic transmission in the hippocampus in vitro. Arzneimittelforschung 1995, 45, 1–5. [Google Scholar]
  148. Volbracht, C.; Van Beek, J.; Zhu, C.; Blomgren, K.; Leist, M. Neuroprotective properties of memantine in different in vitro and in vivo models of excitotoxicity. Eur. J. Neurosci. 2006, 23, 2611–2622. [Google Scholar] [CrossRef] [Green Version]
  149. Liu, Y.; Huang, Y.; Xu, Y.; Qu, P.; Wang, M. Memantine protects against ischemia/reperfusion-induced brain endothelial permeability. IUBMB Life 2018, 70, 336–343. [Google Scholar] [CrossRef] [Green Version]
  150. Davies, V.J.; Hollins, A.J.; Piechota, M.J.; Yip, W.; Davies, J.R.; White, K.E.; Nicols, P.P.; Boulton, M.E.; Votruba, M. Opa1 deficiency in a mouse model of autosomal dominant optic atrophy impairs mitochondrial morphology, optic nerve structure and visual function. Hum. Mol. Genet. 2007, 16, 1307–1318. [Google Scholar] [CrossRef] [Green Version]
  151. Kushnareva, Y.E.; Gerencser, A.A.; Bossy, B.; Ju, W.-K.; White, A.D.; Waggoner, J.; Ellisman, M.H.; Perkins, G.; Bossy-Wetzel, E. Loss of OPA1 disturbs cellular calcium homeostasis and sensitizes for excitotoxicity. Cell Death Differ. 2012, 20, 353–365. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Ju, W.K.; Kim, K.Y.; Angert, M.; Duong-Polk, K.X.; Lindsey, J.D.; Ellisman, M.H.; Weinreb, R.N. Memantine blocks mitochondrial OPA1 and cytochrome c release and subsequent apoptotic cell death in glaucomatous retina. Investig Ophthalmol. Vis. Sci. 2009, 50, 707–716. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. McAllister, J.; Ghosh, S.; Berry, D.; Park, M.; Sadeghi, S.; Wang, K.X.; Parker, W.D.; Swerdlow, R.H. Effects of memantine on mitochondrial function. Biochem. Pharmacol. 2008, 75, 956–964. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Maskell, P.D.; Speder, P.; Newberry, N.R.; Bermudez, I. Inhibition of human α7 nicotinic acetylcholine receptors by open channel blockers of N-methyl-D-aspartate receptors. Br. J. Pharmacol. 2003, 140, 1313–1319. [Google Scholar] [CrossRef] [Green Version]
  155. Wang, F.; Zou, Z.; Gong, Y.; Yuan, D.; Chen, X.; Sun, T. Regulation of Human Brain Microvascular Endothelial Cell Adhesion and Barrier Functions by Memantine. J. Mol. Neurosci. 2017, 62, 123–129. [Google Scholar] [CrossRef]
  156. Schmitz, T.; Endesfelder, S.; Chew, L.-J.; Zaak, I.; Bührer, C. Minocycline protects oligodendroglial precursor cells against injury caused by oxygen-glucose deprivation. J. Neurosci. Res. 2012, 90, 933–944. [Google Scholar] [CrossRef]
  157. Yune, T.Y.; Lee, J.Y.; Jung, G.Y.; Kim, S.J.; Jiang, M.H.; Kim, Y.C.; Oh, Y.J.; Markelonis, G.J.; Oh, T.H. Minocycline Alleviates Death of Oligodendrocytes by Inhibiting Pro-Nerve Growth Factor Production in Microglia after Spinal Cord Injury. J. Neurosci. 2007, 27, 7751–7761. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Cai, Z.; Xiao, M.; He, W.; Zhao, Y. Minocycline upregulates cyclic AMP response element binding protein and brain-derived neurotrophic factor in the hippocampus of cerebral ischemia rats and improves behavioral deficits. Neuropsychiatr. Dis. Treat. 2015, 11, 507–516. [Google Scholar] [CrossRef] [Green Version]
  159. Burgos-Ramos, E.; Puebla-Jiménez, L.; Arilla-Ferreiro, E. Minocycline provides protection against β-amyloid(25-35)-induced alterations of the somatostatin signaling pathway in the rat temporal cortex. Neuroscience 2008, 154, 1458–1466. [Google Scholar] [CrossRef]
  160. Jiang, Y.; Liu, Y.; Zhu, C.; Ma, X.; Ma, L.; Zhou, L.; Huang, Q.; Cen, L.; Pi, R.; Chen, X. Minocycline enhances hippocampal memory, neuroplasticity and synapse-associated proteins in aged C57 BL/6 mice. Neurobiol. Learn. Memory 2015, 121, 20–29. [Google Scholar] [CrossRef]
  161. Fujita, Y.; Ishima, T.; Kunitachi, S.; Hagiwara, H.; Zhang, L.; Iyo, M.; Hashimoto, K. Phencyclidine-induced cognitive deficits in mice are improved by subsequent subchronic administration of the antibiotic drug minocycline. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2007, 32, 336–339. [Google Scholar] [CrossRef]
  162. Miao, H.; Li, R.; Han, C.; Lu, X.; Zhang, H. Minocycline promotes posthemorrhagic neurogenesis via M2 microglia polarization via upregulation of the TrkB/BDNF pathway in rats. J. Neurophysiol. 2018, 120, 1307–1317. [Google Scholar] [CrossRef]
  163. Hashimoto, K.; Ishima, T. A novel target of action of minocycline in NGF-induced neurite outgrowth in PC12 cells: Translation initiation factor eIF4AI. PLoS ONE 2010, 5, e15430. [Google Scholar] [CrossRef]
  164. Hunter, C.L.; Bachman, D.; Granholm, A.C. Minocycline prevents cholinergic loss in a mouse model of Down’s syndrome. Ann. Neurol. 2004, 56, 675–688. [Google Scholar] [CrossRef]
  165. Wang, X.; Zhu, S.; Pei, Z.; Drozda, M.; Stavrovskaya, I.G.; Del Signore, S.J.; Cormier, K.; Shimony, E.M.; Wang, H.; Ferrante, R.J. Inhibitors of cytochrome c release with therapeutic potential for Huntington’s disease. J. Neurosci. 2008, 28, 9473–9485. [Google Scholar] [CrossRef] [Green Version]
  166. Fernandez-Gomez, F.J.; Galindo, M.F.; Gomez-Lazaro, M.; Gonzalez-Garcia, C.; Cena, V.; Aguirre, N.; Jordan, J. Involvement of Mitochondrial Potential and Calcium Buffering Capacity in Minocycline Cytoprotective Actions. Neuroscience 2005, 133, 959–967. [Google Scholar] [CrossRef]
  167. Yoon, S.-Y.; Patel, D.; Dougherty, P. Minocycline blocks lipopolysaccharide induced hyperalgesia by suppression of microglia but not astrocytes. Neuroscience 2012, 221, 214–224. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  168. Lai, A.Y.; Todd, K.G. Hypoxia-activated microglial mediators of neuronal survival are differentially regulated by tetracyclines. Glia 2006, 53, 809–816. [Google Scholar] [CrossRef]
  169. Gray, E.; Ginty, M.; Kemp, K.; Scolding, N.; Wilkins, A. The PPAR-γ agonist pioglitazone protects cortical neurons from inflammatory mediators via improvement in peroxisomal function. J Neuroinflamm. 2012, 9, 63. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  170. Kanakasabai, S.; Pestereva, E.; Chearwae, W.; Gupta, S.K.; Ansari, S.; Bright, J.J. PPARγ agonists promote oligodendrocyte differentiation of neural stem cells by modulating stemness and differentiation genes. PLoS ONE 2012, 7, e50500. [Google Scholar] [CrossRef] [PubMed]
  171. Huang, W.; Shao, M.; Liu, H.; Chen, J.; Hu, J.; Zhu, L.; Liu, F.; Wang, D.; Zou, Y.; Xiong, Y.; et al. Fibroblast growth factor 21 enhances angiogenesis and wound healing of human brain microvascular endothelial cells by activating PPARγ. J. Pharmacol. Sci. 2019, 140, 120–127. [Google Scholar] [CrossRef] [PubMed]
  172. Bernardo, A.; Levi, G.; Minghetti, L. Role of the peroxisome proliferator-activated receptor-γ (PPAR-γ) and its natural ligand 15-deoxy-Δ12, 14-prostaglandin J2 in the regulation of microglial functions. Eur. J. Neurosci. 2000, 12, 2215–2223. [Google Scholar] [CrossRef]
  173. Pérez-Ortiz, J.M.; Tranque, P.; Vaquero, C.F.; Domingo, B.; Molina, F.; Calvo, S.; Jordán, J.; Ceña, V.; Llopis, J. Glitazones differentially regulate primary astrocyte and glioma cell survival: Involvement of reactive oxygen species and peroxisome proliferator-activated receptor-γ. J. Biol. Chem. 2004, 279, 8976–8985. [Google Scholar] [CrossRef] [Green Version]
  174. Wang, Y.; Zhao, W.; Li, G.; Chen, J.; Guan, X.; Chen, X.; Guan, Z. Neuroprotective effect and mechanism of thiazolidinedione on dopaminergic neurons in vivo and in vitro in Parkinson’s disease. PPAR Res. 2017, 2017, 4089214. [Google Scholar] [CrossRef] [Green Version]
  175. Duvanel, C.B.; Honegger, P.; Pershadsingh, H.; Feinstein, D.; Matthieu, J.M. Inhibition of glial cell proinflammatory activities by peroxisome proliferator-activated receptor gamma agonist confers partial protection during antimyelin oligodendrocyte glycoprotein demyelination in vitro. J. Neurosci. Res. 2003, 71, 246–255. [Google Scholar] [CrossRef]
  176. Zhang, Q.; Zhu, W.; Xu, F.; Dai, X.; Shi, L.; Cai, W.; Mu, H.; Hitchens, T.K.; Foley, L.M.; Liu, X.; et al. The interleukin-4/PPARγ signaling axis promotes oligodendrocyte differentiation and remyelination after brain injury. PLoS Biol. 2019, 17, e3000330. [Google Scholar] [CrossRef] [Green Version]
  177. Bernardo, A.; Plumitallo, C.; De Nuccio, C.; Visentin, S.; Minghetti, L. Curcumin promotes oligodendrocyte differentiation and their protection against TNF-α through the activation of the nuclear receptor PPAR-γ. Sci. Rep. 2021, 11, 4952. [Google Scholar] [CrossRef]
  178. Smith, S.A.; Monteith, G.R.; Holman, N.A.; Robinson, J.A.; May, F.J.; Roberts-Thomson, S.J. Effects of peroxisome proliferator-activated receptor γ ligands ciglitazone and 15-deoxy-Δ12, 14-prostaglandin J2 on rat cultured cerebellar granule neuronal viability. J. Neurosci. Res. 2003, 72, 747–755. [Google Scholar] [CrossRef] [PubMed]
  179. Fukunaga, Y.; Itoh, H.; Doi, K.; Tanaka, T.; Yamashita, J.; Chun, T.-H.; Inoue, M.; Masatsugu, K.; Sawada, N.; Saito, T.; et al. Thiazolidinediones, peroxisome proliferator-activated receptor γ agonists, regulate endothelial cell growth and secretion of vasoactive peptides. Atherosclerosis 2001, 158, 113–119. [Google Scholar] [CrossRef] [PubMed]
  180. Pisanu, A.; Lecca, D.; Mulas, G.; Wardas, J.; Simbula, G.; Spiga, S.; Carta, A.R. Dynamic changes in pro- and anti-inflammatory cytokines in microglia after PPAR-γ agonist neuroprotective treatment in the MPTPp mouse model of progressive Parkinson’s disease. Neurobiol. Dis. 2014, 71, 280–291. [Google Scholar] [CrossRef]
  181. Gabryel, B.; Adamek, M.; Pudełko, A.; Małecki, A.; Trzeciak, H.I. Piracetam and Vinpocetine Exert Cytoprotective Activity and Prevent Apoptosis of Astrocytes In Vitro in Hypoxia and Reoxygenation. Neurotoxicology 2002, 1, 19–31. [Google Scholar] [CrossRef] [PubMed]
  182. Stockburger, C.; Miano, D.; Pallas, T.; Friedland, K.; Müller, W.E. Enhanced Neuroplasticity by the Metabolic Enhancer Piracetam Associated with Improved Mitochondrial Dynamics and Altered Permeability Transition Pore Function. Neural Plast. 2016, 2016, 8075903. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. He, Z.; Liao, Y.; Zheng, M.; Zeng, F.-D.; Guo, L.-J. Piracetam Improves Cognitive Deficits Caused by Chronic Cerebral Hypoperfusion in Rats. Cell. Mol. Neurobiol. 2007, 28, 613–627. [Google Scholar] [CrossRef]
  184. Yang, Y.; Feng, J.; Xu, F.; Wang, J. Piracetam inhibits ethanol (EtOH)-induced memory deficit by mediating multiple pathways. Brain Res. 2017, 1676, 83–90. [Google Scholar] [CrossRef] [PubMed]
  185. Winblad, B. Piracetam: A Review of Pharmacological Properties and Clinical Uses. CNS Drug Rev. 2006, 11, 169–182. [Google Scholar] [CrossRef]
  186. He, Z.; Hu, M.; Zha, Y.-H.; Li, Z.-C.; Zhao, B.; Yu, L.L.; Yu, M.; Qian, Y. Piracetam Ameliorated Oxygen and Glucose Deprivation-Induced Injury in Rat Cortical Neurons Via Inhibition of Oxidative Stress, Excitatory Amino Acids Release and P53/Bax. Cell. Mol. Neurobiol. 2014, 4, 539–547. [Google Scholar] [CrossRef] [PubMed]
  187. Brandão, F.; Paula-Barbosa, M.; Cadete-Leite, A. Piracetam impedes hippocampal neuronal loss during withdrawal after chronic alcohol intake. Alcohol 1995, 12, 279–288. [Google Scholar] [CrossRef]
  188. Paula-Barbosa, M.; Brandao, F.; Pinho, M.; Andrade, J.; Madeira, M.; Cadete-Leite, A. The effects of piracetam on lipofuscin of the rat cerebellar and hippocampal neurons after long-term alcohol treatment and withdrawal: A quantitative study. Alcohol. Clin. Exp. Res. 1991, 15, 834–838. [Google Scholar] [CrossRef]
  189. Gupta, S.; Verma, D.K.; Biswas, J.; Raju, K.S.R.; Joshi, N.; Singh, S. The metabolic enhancer piracetam attenuates mitochondrion-specific endonuclease G translocation and oxidative DNA fragmentation. Free Radic. Biol. Med. 2014, 73, 278–290. [Google Scholar] [CrossRef]
  190. Frizzo, M.E.d.S.; Dall’Onder, L.P.; Dalcin, K.B.; Souza, D.O. Riluzole enhances glutamate uptake in rat astrocyte cultures. Cell. Mol. Neurobiol. 2004, 24, 123–128. [Google Scholar] [CrossRef]
  191. Pereira, A.C.; Gray, J.D.; Kogan, J.F.; Davidson, R.; Rubin, T.G.; Okamoto, M.; Morrison, J.H.; McEwen, B.S. Age and Alzheimer’s disease gene expression profiles reversed by the glutamate modulator riluzole. Mol. Psychiatry 2016, 22, 296–305. [Google Scholar] [CrossRef]
  192. Urbani, A.; Belluzzi, O. Riluzole inhibits the persistent sodium current in mammalian CNS neurons. Eur. J. Neurosci. 2000, 12, 3567–3574. [Google Scholar] [CrossRef]
  193. Lesuis, S.L.; Kaplick, P.M.; Lucassen, P.J.; Krugers, H.J. Treatment with the glutamate modulator riluzole prevents early life stress-induced cognitive deficits and impairments in synaptic plasticity in APPswe/PS1dE9 mice. Neuropharmacology 2019, 150, 175–183. [Google Scholar] [CrossRef]
  194. Katoh, R.; Asano, T.; Ueda, H.; Morishita, R.; Takeuchi, I.K.; Inaguma, Y.; Kato, K. Riluzole enhances expression of brain-derived neurotrophic factor with consequent proliferation of granule precursor cells in the rat hippocampus. FASEB J. 2002, 16, 1328–1330. [Google Scholar] [CrossRef] [PubMed]
  195. Malgouris, C.; Bardot, F.; Daniel, M.; Pellis, F.; Rataud, J.; Uzan, A.; Blanchard, J.; Laduron, P. Riluzole, a novel antiglutamate, prevents memory loss and hippocampal neuronal damage in ischemic gerbils. J. Neurosci. 1989, 9, 3720–3727. [Google Scholar] [CrossRef] [PubMed]
  196. Kalra, S.; Cashman, N.R.; Genge, A.; Arnold, D.L. Recovery of N-acetylaspartate in corticomotor neurons of patients with ALS after riluzole therapy. Neuroreport 1998, 9, 1757–1761. [Google Scholar] [CrossRef] [PubMed]
  197. Storch, A.; Burkhardt, K.; Ludolpha, C.; Schwarz, J. Protective effects of Riluzole on dopamine neurons: Involvement of oxidative stress and cellular energy metabolism. J. Neurochem. 2000, 75, 2259–2269. [Google Scholar] [CrossRef]
  198. Umemiya, M.; Berger, A.J. Inhibition by riluzole of glycinergic postsynaptic currents in rat hypoglossal motoneurones. Br. J. Pharmacol. 1995, 116, 3227–3230. [Google Scholar] [CrossRef] [Green Version]
  199. Liu, B.-S.; Ferreira, R.; Lively, S.; Schlichter, L. Microglial SK3 and SK4 currents and activation state are modulated by the neuroprotective drug, riluzole. J. Neuroimmune Pharmacol. 2013, 8, 227–237. [Google Scholar] [CrossRef]
  200. Wu, Q.; Zhang, Y.; Zhang, Y.; Zhang, W.; Zhang, W.; Liu, Y.; Xu, S.; Guan, Y.; Chen, X. Riluzole improves functional recovery after acute spinal cord injury in rats and may be associated with changes in spinal microglia/macrophages polarization. Neurosci. Lett. 2020, 723, 134829. [Google Scholar] [CrossRef]
Table 1. Suggested drugs: their dosages, pharmacodynamics, and pharmacokinetics.
Table 1. Suggested drugs: their dosages, pharmacodynamics, and pharmacokinetics.
DrugSuggested DoseSerious Adverse Effects (SAE)Drug-Drug InteractionsEffects of ComorbiditiesEffects of AgeEffects of Pregnancy
clemastine5.36 mg bid (a) None
erythropoietin10,000 IU IM once/monthSAE only if high dosage.
fingolimod0.5 mg/day(b)
fluoxetine10 mg/day (c), (d). No effect
lithium75 mg/day(e), (f), (g), (h), (i), (j). Avoid if GFR < 60 mL/min. Avoid in pregnancy
memantine5 mg qd ×7d, then bid (j).(k). Avoid if GFR < 60 mL/min.
minocycline100 mg/day(l), (m), (n), (o).
piracetam1.2 G/dayNo SAENone
pioglitazone15 mg/dayNo SAE
riluzole100 mg/day(p).
See text for descriptions of therapeutic effects. Suggested two-drug combinations are pioglitazone plus fluoxetine or pioglitazone plus lithium. Suggested three-drug combinations would add one drug from clemastine, fingolimod, or memantine. (a) this high dose for a study was approved by FDA. (b) fingolimod may cause transient 20 heart block. (c), fluoxetine + lithium may cause serotonergic syndrome. (d), fluoxetine may increase blood levels of antipsychotic drugs. Lithium may cause (e) hypercalcemia, (f) hypothyroidism, (g) rarely hyperthyroidism, (h) GFR may fall; do not use if GFR < 60 mL/min, (i) ~20% patients In clinical trials, quit Rx due to adverse events, (j) NB suggested very low dosage is unlikely to cause problems. (k) Use a lower dose of memantine in (j) elderly patients. (k) do not use if GFR < 60 mL/min. (l) vestibular symptoms are the commonest side effect but infrequent. (m) systemic lupus erythematosus as SAE is serious but rare; ANA testing before commencing Rx is mandatory, (n) other hypersensitivity syndromes may also occur, (o) cutaneous and dental hyperpigmentation occurs with prolonged Rx, (p) Liver enzymes abnormal in ~10% Rx’ed with riluzole.
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

Fessel, J. Supplementary Pharmacotherapy for the Behavioral Abnormalities Caused by Stressors in Humans, Focused on Post-Traumatic Stress Disorder (PTSD). J. Clin. Med. 2023, 12, 1680. https://doi.org/10.3390/jcm12041680

AMA Style

Fessel J. Supplementary Pharmacotherapy for the Behavioral Abnormalities Caused by Stressors in Humans, Focused on Post-Traumatic Stress Disorder (PTSD). Journal of Clinical Medicine. 2023; 12(4):1680. https://doi.org/10.3390/jcm12041680

Chicago/Turabian Style

Fessel, Jeffrey. 2023. "Supplementary Pharmacotherapy for the Behavioral Abnormalities Caused by Stressors in Humans, Focused on Post-Traumatic Stress Disorder (PTSD)" Journal of Clinical Medicine 12, no. 4: 1680. https://doi.org/10.3390/jcm12041680

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