Next Article in Journal
Current Approaches to the Management of Sentinel Node Procedures in Early Vulvar Cancer in Germany: A Web-Based Nationwide Analysis of Practices
Previous Article in Journal
Spiradenocarcinoma: SEER Study of Epidemiology, Survival, and Treatment Options
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Perspective

Cure of Alzheimer’s Dementia Requires Addressing All of the Affected Brain Cell Types

Department of Medicine, University of California, San Francisco, CA 94143, USA
J. Clin. Med. 2023, 12(5), 2049; https://doi.org/10.3390/jcm12052049
Submission received: 1 February 2023 / Revised: 27 February 2023 / Accepted: 2 March 2023 / Published: 4 March 2023
(This article belongs to the Section Mental Health)

Abstract

:
Multiple genetic, metabolic, and environmental abnormalities are known to contribute to the pathogenesis of Alzheimer’s dementia (AD). If all of those abnormalities were addressed it should be possible to reverse the dementia; however, that would require a suffocating volume of drugs. Nevertheless, the problem may be simplified by using available data to address, instead, the brain cells whose functions become changed as a result of the abnormalities, because at least eleven drugs are available from which to formulate a rational therapy to correct those changes. The affected brain cell types are astrocytes, oligodendrocytes, neurons, endothelial cells/pericytes, and microglia. The available drugs include clemastine, dantrolene, erythropoietin, fingolimod, fluoxetine, lithium, memantine, minocycline, pioglitazone, piracetam, and riluzole. This article describes the ways by which the individual cell types contribute to AD’s pathogenesis and how each of the drugs corrects the changes in the cell types. All five of the cell types may be involved in the pathogenesis of AD; of the 11 drugs, fingolimod, fluoxetine, lithium, memantine, and pioglitazone, each address all five of the cell types. Fingolimod only slightly addresses endothelial cells, and memantine is the weakest of the remaining four. Low doses of either two or three drugs are suggested in order to minimize the likelihood of toxicity and drug–drug interactions (including drugs used for co-morbidities). Suggested two-drug combinations are pioglitazone plus lithium and pioglitazone plus fluoxetine; a three-drug combination could add either clemastine or memantine. Clinical trials are required to validate that the suggest combinations may reverse AD.

1. Introduction

The last five decades have seen a deluge of data concerning the genetic, metabolic, and environmental factors that contribute to the pathogenesis of Alzheimer’s dementia (AD). Birle et al. pointed out that cognitive dysfunction represents a consequence of a global imbalance within three levels of brain organization: the cellular and molecular level, circuitry level, and large-scale network level [1]. The central nervous system is bidirectionally related with other fundamental systems such as the immune, endocrine, and autonomic systems and the microbiota, all of which have been shown to change adversely in patients with cognitive dysfunctions. If all of the known abnormalities were addressed it should be possible to cure the dementia, which should be the goal of treatment. However, that would require a suffocating number of drugs. Nevertheless, the problem may be simplified by addressing, instead, the brain cell types whose functions become changed as a result of all the above abnormalities because it is these cells, whose number and functions are changed, that are ultimately responsible for AD pathogenesis. This approach to formulating pharmacotherapies for psychiatric conditions in general, was described in an earlier article [2]; here, it is described in detail for AD in particular. Fortunately, at least eleven drugs are available from which to formulate a rational therapy to correct those changes. After a very brief summary of the multiple metabolic and genetic factors that participate in the pathogenesis of AD, this article will describe the brain cell types that are affected in AD, and the available drugs that can correct those changes.

2. Altered Metabolism and Genetics in AD

Reactive oxygen species (ROS) have a major role in the pathogenesis of AD; their formation has several causes including mitochondrial damage. ROS link to multiple down-stream damages: to mitochondria themselves, due to reduced levels of NAD/NADH; diminished glucose uptake and response rates to insulin/IGF-1 signaling (due to decreased levels of the insulin receptor and glucose transporter 1 densities); aberrant calcium regulation; disturbed lipid metabolism; impairments of autophagic pathways affecting proteostasis; and reducing water diffusion into brain cells which impacts drug delivery.
Mutations in the APP, PS1, and PS2 genes are causal for familial AD (FAD). Many other genes participate in the pathogenesis of late onset AD (LOAD). The ε4 allele of the APOE gene is frequently present. Patients with both FAD and LOAD who are APOε4 carriers, have abnormally low rates of glucose metabolism in the neocortex that may also be present in carriers of APOε4 without dementia. How APOε4 affects different brain cell types is discussed below. Brain cells from subjects with LOAD have down-regulation of many genes affecting multiple metabolic pathways: abnormalities in glucose metabolism via the pentose phosphate pathway, glycolysis, and tricarboxylic/citric acid pathways; oxidative phosphorylation; and genes responsible for subunits of cytochrome p450 that are involved in fatty acid and cholesterol oxidation. Other down-regulated genes include those responsible for metabolism of glucose [GPD1, GPDHL, and GPDH2, IGF-1 and 2, insulin receptor gene (INSRR), insulin receptor substrate 2 (IRS2), glycogen phosphorylase (PYGL), phosphoglucomutase (PGM1, PGM2L1), hexokinase (HK1 and 2), phosphofructokinase (PFKFB3, PFKP)], lactate dehydrogenase A (LDHA), isocitrate dehydrogenase 1 (IDH1), pyruvate dehydrogenase kinase (PDK3)] (see Ref. [3] for full descriptions). In both Parkinson’s disease and diabetes mellitus, each of which has genetic underpinnings, there are high rates of dementia. Individuals with trisomy 21 have an extra copy of the amyloid precursor protein (APP) gene on chromosome 21, leading to excessive cerebral amyloid.
As is obvious from this highly abbreviated description, even were it possible to correct the innumerable metabolic and genetic abnormalities seen in AD, the task would be impossible to achieve because correction of all of them would require an intolerable number of drugs. It would be far simpler is to correct the changed functions of the affected brain cell types.

3. Brain Cell Types That Are Changed in AD

Changes that are in the direction of decreased functions affect astrocytes, oligodendrocytes, neurons, and endothelial cells/pericytes; changes that result in gain of function involve microglia. The following discusses the affected cell types.
Astrocytes in AD. Emphasizing their importance for cognition, astrocyte numbers in the dentate gyrus were more reduced in Braak stages 3–4 than in stages 0–2 [4]. Because astrocyte processes wrap around the cerebral microvasculature, the morphological modifications of astrocytes affect micro-cerebral blood flow and, therefore, the nutrients available to neurons [5,6]; in addition, the processes of astrocytes contain aquaporin-4 (AQ4), which regulates water entry into endothelial cells [7]. The consequences of the decreased numbers of astrocytes in AD include decreases in neural function and the passage of drugs into the brain.
Oligodendrocytes in AD. Mature oligodendrocytes myelinate the naked axons of neurons; if their numbers are decreased, e.g., from impaired maturation of oligodendrocyte precursor cells (OPCs), so that myelination becomes inadequate, then neural tracts suffer and cognition may be disturbed [6]. Exposure of oligodendrocyte cultures to 1 μM of Aβ1-42 induced cell death, morphological changes with shrunken cell bodies and a breakdown of their processes, and an 3.2-fold increase of lactate dehydrogenase activity that was released into the culture media by oligodendrocytes [8].
Neurons in AD. AD patients with severe tau pathologies had a decreased number of newly generated neurons in the dentate gyrus [4]. Leng et al. found a selectively vulnerable subpopulation of excitatory neurons in the entorhinal cortex (EC) and showed a depletion of this subpopulation during AD progression [9]. In AD, Whitehouse et al. showed a 75% reduction in cholinergic neurons in the nucleus basalis of Meynert [10]. Tse et al. analyzed MRI data from 507 subjects including healthy controls, and persons with mild cognitive impairment {MCI} or AD; they found that, with the progress of dementia, the gray matter volume of the subjects was strongly reduced and correlated with both white matter volume and MMSE scores [11].
Endothelial cells in AD. In MCI, there are already decreased levels of VEGF [12], and in 55 patients with established AD, there were reduced numbers of endothelial cell precursors in the peripheral blood that were associated with lower MMSE scores and higher (i.e., worse) Clinical Dementia Rating (CDR) scores [13]. Microvascular diseases affect the progression of cognitive deficits in AD [14]. Endothelial cells contain the scavenge receptor, CD36, whose interaction with Aβ activates NADPH oxidase, producing both vascular oxidative stress and neurovascular dysregulation [15]. String vessels, the remnants of destroyed microcapillaries, are seen throughout the brain of AD patients [16]. In addition, microinfarcts, with minute foci of neuronal loss as small as 50 µm, were seen in 43% of AD brains [17].
Microglia in AD. Several reports concerning the role of microglia in AD are conflicting, the likely reason being the need to distinguish between proinflammatory (M1) and anti-inflammatory (M2) microglia, because a skewed M1 activation over M2 has been related to disease progression in AD [18], and although this is the dominant change in AD, it is not present in all cases. Olah et al. obtained microglia from the brains of 14 participants in a study of cognitive aging and 3 subjects with intractable epilepsy, and recognized seven clusters of microglia, only one of which was reduced in patients with AD [19]. Grubman et al. found that the APOE risk gene is up-regulated in an AD-specific subpopulation of microglia [20].

4. In Summary, the Affected Brain Cell Types in AD Are Astrocytes, Oligodendrocytes, Neurons, Endothelial Cells/Pericytes, and Microglia

Affecting the functions of all of the above brain cell types is the APOE4 variant of the APOE gene product, that renders those who carry it susceptible to AD.
The APOE4 variant of the APOE gene product, renders its carriers susceptible to AD. Relevant to the thesis of this article, one mechanism for this may be its effect on the five affected brain cell types. Blanchard et al. reported several such effects: cholesterol transport was defective in oligodendrocytes that were derived from induced pluripotent stem cells (iPSCs) carrying APOE4; these oligodendrocytes accumulated cholesteryl ester species this was associated with down-regulation of genes linked to myelination, so that the oligodendrocytes had decreased production of Myelin Basic Protein (MBP) [21]. Besides affecting oligodendrocytes, Lin et al. showed that the APOE4 variant can also lead to extensive gene expression alterations in neurons, astrocytes, and microglia; APOE4 astrocytes had cholesterol accumulation, and APOE4 microglia-like cells had increased up-regulation of genes associated with immune responses and inflammation [22]. Victor et al. saw lipid accumulation in APOE4+ microglia, and found that medium taken from cultures of APOE4 microglia, exacerbated the pro-inflammatory activity compared with that taken from cultures of APOE3 microglia [23]. Regarding endothelial cells, those derived from APOE4+ pluripotent stem cells had dysfunctions in pathways leading to effects on blood clotting factors and inflammation [24].
Summary: the dominant changes in brain cell-type in AD are decreases in astrocytes, oligodendrocytes, neurons and endothelial cells, and increases in microglia; those changes may in part be caused by the presence of APOE4.

5. Available Drugs That Address the Changes in Brain Cell Types That Underpin the Pathogenesis of AD

Fortunately, there are several drugs that correct the changes in the affected brain cell types, and from which we can formulate a rational treatment regimen. It is recommended to choose a combination of two drugs from the following list, in order to both reduce the dosages and minimize the likelihood of adverse events.
Highly abbreviated descriptions for likely therapeutic effects are provided here: the drugs either induce an increase in numbers or activation of astrocytes, oligodendrocytes, synapses and neurons, and endothelial cells; or a decrease in numbers or activation of microglia.
Clemastine increased astrocytes [25], postsynaptic proteins [25], muscarinic receptor, synapsin 1 and Homer 1, and oligodendrocytes survival/function [26,27]. It also improved myelin repair [28] and neuronal function [29] and enhanced myelination in the prefrontal cortex (PFC) [30] and enhanced visual function [26,31]. Clemastine provided mitochondrial protection [32,33] and suppressed microglial M1 activation [34].
Dantrolene, by antagonizing ryanodine receptors and blocking Ca2+ release, prevented the detriment to astrocytes caused by excessive glutamate [35] It also prevented oligodendrocyte death [33,36], and preserved synaptic and neural function. References [37,38,39,40,41,42,43,44,45,46,47,48] showed several other mechanisms for neuroprotection by dantrolene. A potential adverse effect is via the cerebral microvasculature, because release of brain derived neurotrophic factor (BDNF) by endothelial cells requires Ca2+ mobilization [36].
Erythropoietin increased differentiation of neuronal stem cells (NSCs) into astrocytes; increased oligodendrocytes [49] and neurons and dendritic spines [50] via increased production of BDNF and its receptor [51]; and it also did so by enhancing differentiation of NSCs [52]. Erythropoietin improved the functioning of synapses [53,54,55], endothelial cells [56,57], and microglia [58].
Fingolimod, which is a pro-drug for an agonist of sphingomyelin phosphate (S1P), induced cell proliferation [59]. In astrocytes, fingolimod activated neurotrophic genes [60], and reduced formation of ceramides that cause apoptosis [61,62]. It activated myelinating oligodendrocyte [63], increased OPCs, myelination, and neurological function, and ameliorated brain demyelination [64]. It enriched synaptic genes [65], prevented synaptic toxicity [66], and reversed synaptic hypersensitivities [66]. It produced myelin in brains that had been demyelinated [67], and prevented neural death from N-methyl-D-aspartate (NMDA) [68]. In multiple sclerosis, it improved axonal and myelin integrity [69]; and in mice it prevented demyelination [70]. It increased dendritic spines [71,72], reduced neuronal death from reactive oxygen species (ROS) [73], and improved mitochondrial production of ATP [74]. Weak evidence shows some benefit to the cerebral microcirculation: fingolimod prevented BBB disruption induced by sera from patients with multiple sclerosis [75].
In microglia, fingolimod shifted M1 polarization toward M2 [76,77].
Fluoxetine induced increases in: astrocytes [78], oligodendrocytes [79,80,81], neurons [82,83,84], and endothelial cells [85], and decreased microglial activation [81,86].
Fluoxetine activated astrocytes to produce BDNF [78,87,88] and promoted clearance of astrocytes with damaged mitochondria [89]. It up-regulated oligodendrocyte precursor cells (OPCs) and oligodendrocyte markers [79], and reduced oligodendrocyte senescence [90]. Fluoxetine increased neurogenesis [79,82] and neuronal circuits [84]. Neurons deprived of glucose and oxygen had increased survival with fluoxetine [83]. It increased vasodilatation via differentiation and proliferation of endothelial cells and decreased arteriolar tone [85,91]. In microglia, it attenuated NADPH oxidase activation, production of ROS and reactive nitrogen species, and down-regulated M1 and up-regulated M2 activation [92,93].
Lithium doubled astrocytic numbers and their VEGF secretion [94]. It increased oligodendrocyte expression of proteolipid protein (PLP) and myelin basic protein (MBP), improving synaptic and neuronal function [95,96,97,98,99]. By negating activation of GSK-3β, synaptic expression of post synaptic density (PSD)-95 and gephyrin were enhanced [100]. Lithium promoted neurogenesis [99,101,102,103], doubled BDNF levels, increased dendritic length, increased anti-apoptotic Bcl2 and Bcl-XL, and prevented neuronal death caused by excessive glutamate, pro-apoptotic BAD and BAX, and caspases [104]. It increased the number and size of neuronal mitochondria [105], increased antioxidants [97,105], minimized neurotoxicity from cytochrome c, and promoted mitochondrial biogenesis [106,107]. Lithium benefitted the microvasculature by increasing endothelial cell survival [108], enhancing VEGF secretion [109], and increasing BBB integrity [110]. Lithium’s inhibition of GSK-3β reduced production of microglial [111,112] and activation of pro-inflammatory mediators [113].
Memantine induced activation of astrocytes [114] and it prevented losses of oligodendrocytes [114], synapses and neurons [115,116,117], and endothelial cells [118]. It is a NMDAR antagonist, enters the NMDAR’s Ca2+ channel and decreases its permeability, so prevents neuronal excitotoxicity and death [115]. Memantine provides synaptic protection via several mechanisms [119,120,121], preventing cytotoxicity by blocking inhibition of a guanosine triphosphatase involved in multiple cellular processes, thus protecting against cell death caused by mitochondrial dysfunction that leads to cytochrome c release, and ROS and peroxide production [122,123,124,125]. Blocking the ion channel of the acetylcholine receptors also prevents neurotoxicity [126]. Memantine benefitted brain endothelial cells and blocked disruption of the BBB. Microglial function improves because memantine up-regulates the M2 isotype [127].
Minocycline prevented oligodendrocyte toxicity caused by deprivation of oxygen and glucose [128], by microglial-induced apoptosis [129]. Minocycline also prevented Aβ inhibition of cAMP-responsive element-binding protein (CREB) up-regulation [119,130]. Synaptic function improved from increased levels of PSD-95 and dendritic spines [121]. It prevented cognitive decline caused by an antagonist of the NMDAR [120]. Neuronal benefit was also derived from its increased expression of BDNF, CREB, and phospho-CREB [119], proliferation of neuronal precursor cells (NPC) [127], and potentiation of neurite outgrowth [131]. In a transgenic mouse model of Down’s syndrome, minocycline prevented the decline of cholinergic neurons [132]. In a list of 1040 drugs that prevent the release of cytochrome c (which induces a series of biochemical reactions that result in caspase activation and subsequent cell death), minocycline was the second most potent [133]; the mechanism involves inhibiting mitochondrial increases in Ca2+ concentration plus inhibition of NADH-cytochrome c reductase and cytochrome c oxidase [133,134]. Finally, minocycline prevented microglial activation [135,136], production of pro-inflammatory cytokines [137], and promoted M2 microglial polarization [127].
Pioglitazone, a peroxisome proliferator-activated receptor gamma (PPARγ) agonist, prevents phosphorylation of JAK–STAT in astrocytes and thereby induces increases in neurons [138,139], oligodendrocytes [139,140], endothelial cells [141], and decreases in microglia [142]. Other PPARγ agonists, ciglitazone and curcumin, were also cytoprotective for astrocytes and reversed their decreased expression of PPARγ receptor after exposure to Aβ25–35 [143,144]. Ciglitazone also increased formation of oligodendrocyte progenitors [139]. Pioglitazone rescued the demyelination caused by anti-MOG autoantibodies [145]. The promotion of OPC differentiation into mature oligodendrocytes by IL-4 was mediated by PPARγ or curcumin [140,142]. PPARγ agonism increases the neurogenic differentiation gene NeyroD1 [139] and protected cortical neurons and axons against toxicity induced by NO or KCl [138,146].
PPARγ also induces endothelial cell proliferation and angiogenesis [141,147]. In microglia, PPARγ agonists inhibit cytokine production by down-regulating pro-inflammatory genes [148]. In addition, the PPARγ agonist rosiglitazone up-regulates M2 microglia [149].
Piracetam increased the numbers and functioning of astrocytes [150]. It also increased the function of synapses [151,152,153] and neurons [151]: it decreased neurotoxicity from deprivation of oxygen and glucose, hypoperfusion, ethanol feeding, or ethanol withdrawal [152,154,155,156]. It also caused longer neurites [151]. 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 [157]. By decreasing mitochondrial swelling and permeability caused by excessive Ca2+ opening the mitochondrial permeability transition pore (MPTP), it improved mitochondrial membrane potential and ATP levels, shifted the balance of mitochondrial fission or fusion towards fusion, and reversed the adverse effects of pro-oxidants [158]. Additionally, it reversed the cytotoxic effects of p53 and BAX [152].
Riluzole increased both the gene and protein expression of the excitatory amino acid transporter (EAAT2), thus enhancing glutamate uptake by astrocytes, which protects against excitotoxicity of neurons [159,160]. It benefits synapses as shown by its inhibition of voltage-activated sodium currents which prevents reverse operation of the Na+/Ca2+ exchanger [161], and by increasing Long-Term Potentiation (LTP) [162]. It caused a 40-fold increase in hippocampal BDNF, inducing neurogenesis [163] and protected against neural degeneration caused by ischemia [164]. N-acetylaspartate is exclusively expressed in neurons and was increased in the cerebral cortex due to exposure to riluzole [165]. In neurons, it also decreased oxidative stress, lipid peroxidation, and ATP depletion [166,167]. Microglial activation was ameliorated by riluzole [168] which also upregulated the mRNA levels of M2 markers and downregulated those of M1 markers [169].

6. Adverse Effects

The choice of a medication is partly determined by its safety profile, particularly for serious adverse events (SAEs). The following are summaries of reports of the SAEs for the three drugs that are suggested here for use in clinical trials.
Fluoxetine: Beasely et al. obtained data from 25 double-blind clinical trials involving 4016 patients with MDD randomized to treatment that included fluoxetine 20 to 80 mg/d [170]. At a dose of 20 mg/d, fluoxetine-treated patients had a discontinuation rate due to adverse events that was not significantly different from that in placebo recipients. In this article, the suggested dose is 10 mg/d in combination with lithium.
Lithium: SAEs from lithium include hypercalcemia, hypothyroidism [171], nephrogenic diabetes insipidus, and renal insufficiency [172]. Low dosage (<100 mg daily) benefited Alzheimer’s dementia [173]; thus, 75 mg daily might be an appropriate dose in BD, MDD, SCZ, AD, or PTSD. It should not be used if the glomerular filtration rate is <60 mL/min.
Pioglitazone. The main adverse effects reported with pioglitazone are congestive heart failure, pedal edema and weight gain, and bone loss. Lincoff et al. performed a meta-analysis of 19 trials enrolling 16,390 patients receiving pioglitazone or comparator for 4 months to 3.5 years. Death, myocardial infarction, or stroke occurred in 4.4% of participants receiving pioglitazone and in 5.7% receiving the control therapy [174]. The number of neoplasms was similar in the pioglitazone and placebo arms of the trials. In a review, Betteridge noted sustained improvements in serum levels of triglycerides and HDL cholesterol and favorable effects on the particle size of LDL cholesterol [175]. The PROactive trial saw manageable increases in edema (26.4% vs. 15.1% for placebo) and weight gain (+3.8 kg vs. −0.6 kg for placebo); and, despite more heart failure in the pioglitazone group (5.7% vs. 4.1% for placebo), there was a proportional improvement in macrovascular outcomes among the patients who developed heart failure, and whose absolute rates of macrovascular events and mortality were similar to those in the placebo group. The incidence of malignancies were similar in the pioglitazone versus placebo arms of the study, but a higher rate of bone fractures was observed among pioglitazone-treated female patients (5.1% vs. 2.5%) [176]. This profile suggests that adverse events are outweighed by favorable outcomes.

7. Discussion

The cure of established AD requires addressing the cerebral cell types that are responsible for its pathogenesis. Those cell types are astrocytes, oligodendrocytes, neurons, endothelial cells/pericytes, and microglia. While it is not necessarily the case that, for individual subjects with AD, all five cell types are involved, for practical purposes it is prudent to assume that they are. Therefore, drugs that address all five cell types are the best candidates for a regimen that has the best chance for successful reversal of the dementia. It is recommended to use a combination of two drugs in order to minimize dosages and the risk of both serious adverse reactions (SAEs) and drug–drug interactions (including with drugs used for co-morbid conditions). The best chances would come from use of the drugs that address all five cell types. Those are erythropoietin, fingolimod, fluoxetine, lithium, memantine, and pioglitazone. However, unless there is compromise of the BBB, erythropoietin will not cross it; and fingolimod only weakly benefits the cerebral microcirculation. Therefore, the choice of a two-drug combination must be chosen from fluoxetine, lithium, and pioglitazone; however, fluoxetine and lithium potentially cause the most SAEs. Therefore, a clinical trial to test whether two of the suggested drugs will, in fact, cure AD should administer pioglitazone plus either fluoxetine or lithium, using low dosages: pioglitazone 15 mg/day, lithium 75 mg/day, or fluoxetine 10 mg/day. Clemastine (benefits four cell types, and causes no SAEs), fingolimod, and memantine could be added 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 clinical trials are required to establish their efficacy for reversal of AD.
In a clinical trial that randomly assigns participants to receive active drugs or placebo, calculation of the required number of participants would be based upon the primary objective showing ≥30% more cure of AD in those using active drugs versus zero for placebo. Cure is defined as the reversion of cognition to normal levels in standard tests such as Consortium to Establish a Registry for Alzheimer’s Disease (CERAD) and the Mini-Mental State Examination (MMSE). The number in each group would be calculated on the basis of ≥30% more patients experiencing reversal of AD from active drugs than from placebo, and an annual drop-out rate of 10%. The planned duration of treatment would be three years with an annual review of data by a study board (DSMB). Participants would be aged ≥70, with a diagnosis of AD made according to standard criteria [177]. Exclusions would be individuals with particular risk factors for AD, viz., type-1 diabetes, untreated hypertension (BP ≥ 130/85), and untreated hepatitis C.

8. Conclusions and Summary

Curative treatment of AD should address the decreases in astrocytes, oligodendrocytes, neurons, and endothelial cells/pericytes, and increases in microglia, that occur in AD.
A two-drug combination might comprise pioglitazone with fluoxetine or pioglitazone with lithium; a three-drug combination might comprise those two drugs plus clemastine, fingolimod, or erythropoietin.
Low doses of those drugs will reduce SAEs and drug–drug interactions.
Clinical trial will demonstrate the validity of the proposed treatment.

Funding

No funds from public or private sources were used in the preparation of this article.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

There are no conflict of interest.

References

  1. Birle, C.; Slavoaca, D.; Balea, M.; Livint Popa, L.; Muresanu, I.; Stefanescu, E.; Vacaras, V.; Dina, C.; Strilciuc, S.; Popescu, B.O.; et al. Cognitive function: Holarchy or holacracy? Neurol. Sci. 2021, 42, 89–99. [Google Scholar] [CrossRef] [PubMed]
  2. Fessel, J. Fluoxetine plus lithium for treatment of mental health impairment in Long Covid. Discov. Ment. Health 2023, 3, 1–12. [Google Scholar] [CrossRef] [PubMed]
  3. Ryu, W.-I.; Bormann, M.K.; Shen, M.; Kim, D.; Forester, B.; Park, Y.; So, J.; Seo, H.; Sonntag, K.-C.; Cohen, B.M. Brain cells derived from Alzheimer’s disease patients have multiple specific innate abnormalities in energy metabolism. Mol. Psychiatry 2021, 26, 5702–5714. [Google Scholar] [CrossRef] [PubMed]
  4. Ekonomou, A.; Savva, G.; Brayne, C.; Forster, G.; Francis, P.; Johnson, M.; Perry, E.; Attems, J.; Somani, A.; Minger, S. Medical Research Council Cognitive Function and Ageing Neuropathology Study. Stage-specific changes in neurogenic and glial markers in Alzheimer s disease. Biol. Psychiatry 2015, 77, 711–719. [Google Scholar] [CrossRef] [Green Version]
  5. Marina, N.; Christie, I.N.; Korsak, A.; Doronin, M.; Brazhe, A.; Hosford, P.S.; Wells, J.A.; Sheikhbahaei, S.; Humoud, I.; Paton, J.F. Astrocytes monitor cerebral perfusion and control systemic circulation to maintain brain blood flow. Nat. Commun. 2020, 11, 1–9. [Google Scholar] [CrossRef] [Green Version]
  6. Kapogiannis, D.; Mattson, M.P. Disrupted energy metabolism and neuronal circuit dysfunction in cognitive impairment and Alzheimer’s disease. Lancet Neurol. 2011, 10, 187–198. [Google Scholar] [CrossRef] [Green Version]
  7. Lan, Y.-L.; Zou, S.; Chen, J.-J.; Zhao, J.; Li, S. The neuroprotective effect of the association of aquaporin-4/glutamate transporter-1 against Alzheimer’s disease. Neural Plast. 2016, 2016. [Google Scholar] [CrossRef] [Green Version]
  8. Roth, A.D.; Ramírez, G.; Alarcón, R.; Von Bernhardi, R. Oligodendrocytes damage in Alzheimer’s disease: Beta amyloid toxicity and inflammation. Biol. Res. 2005, 38, 381–387. [Google Scholar] [CrossRef] [Green Version]
  9. Leng, K.; Li, E.; Eser, R.; Piergies, A.; Sit, R.; Tan, M.; Neff, N.; Li, S.H.; Rodriguez, R.D.; Suemoto, C.K. Molecular characterization of selectively vulnerable neurons in Alzheimer’s disease. Nat. Neurosci. 2021, 24, 276–287. [Google Scholar] [CrossRef]
  10. Whitehouse, P.J.; Price, D.L.; Struble, R.G.; Clark, A.W.; Coyle, J.T.; Delon, M.R.J.S. Alzheimer’s disease and senile dementia: Loss of neurons in the basal forebrain. Science 1982, 215, 1237–1239. [Google Scholar] [CrossRef]
  11. Tse, K.H.; Cheng, A.; Chow, H.M.; Herrup, K. White Matter Degeneration and Aberrant Oligodendrocyte Differentiation during the Progress of Alzheimer’s Disease. In Proceedings of the 45th Annual Meeting of the Society for Neuroscience (Neuroscience 2015), Chicago, IL, USA, 17–21 October 2015. [Google Scholar]
  12. Huang, L.; Jia, J.; Liu, R. Decreased serum levels of the angiogenic factors VEGF and TGF-β1 in Alzheimer’s disease and amnestic mild cognitive impairment. Neurosci. Lett. 2013, 550, 60–63. [Google Scholar] [CrossRef]
  13. Lee, S.; Chu, K.; Jung, K.; Park, H.; Kim, D.; Bahn, J.; Kim, J.; Oh, M.; Lee, S.; Kim, M. Reduced circulating angiogenic cells in Alzheimer disease. Neurology 2009, 72, 1858–1863. [Google Scholar] [CrossRef]
  14. Pimentel-Coelho, P.M.; Rivest, S. The early contribution of cerebrovascular factors to the pathogenesis of Alzheimer’s disease. Eur. J. Neurosci. 2012, 35, 1917–1937. [Google Scholar] [CrossRef]
  15. Park, L.; Wang, G.; Zhou, P.; Zhou, J.; Pitstick, R.; Previti, M.L.; Younkin, L.; Younkin, S.G.; Van Nostrand, W.E.; Cho, S. Scavenger receptor CD36 is essential for the cerebrovascular oxidative stress and neurovascular dysfunction induced by amyloid-β. Proc. Natl. Acad. Sci. USA 2011, 108, 5063–5068. [Google Scholar] [CrossRef] [Green Version]
  16. Fessel, W. Concordance of several subcellular interactions initiates Alzheimer’s dementia: Their reversal requires combination treatment. Am. J. Alzheimer’s Disease Other Dement. 2017, 32, 166–181. [Google Scholar] [CrossRef]
  17. Brundel, M.; De Bresser, J.; Van Dillen, J.J.; Kappelle, L.J.; Biessels, G.J. Cerebral microinfarcts: A systematic review of neuropathological studies. J. Cereb. Blood Flow Metab. 2012, 32, 425–436. [Google Scholar] [CrossRef] [Green Version]
  18. Sarlus, H.; Heneka, M.T. Microglia in Alzheimer’s disease. J. Clin. Investig. 2017, 127, 3240–3249. [Google Scholar] [CrossRef]
  19. Olah, M.; Menon, V.; Habib, N.; Taga, M.F.; Ma, Y.; Yung, C.J.; Cimpean, M.; Khairallah, A.; Coronas-Samano, G.; Sankowski, R. Single cell RNA sequencing of human microglia uncovers a subset associated with Alzheimer’s disease. Nat. Commun. 2020, 11, 1–18. [Google Scholar] [CrossRef]
  20. Grubman, A.; Chew, G.; Ouyang, J.F.; Sun, G.; Choo, X.Y.; McLean, C.; Simmons, R.K.; Buckberry, S.; Vargas-Landin, D.B.; Poppe, D. A single-cell atlas of entorhinal cortex from individuals with Alzheimer’s disease reveals cell-type-specific gene expression regulation. Nat. Neurosci. 2019, 22, 2087–2097. [Google Scholar] [CrossRef]
  21. Blanchard, J.W.; Akay, L.A.; Davila-Velderrain, J.; von Maydell, D.; Mathys, H.; Davidson, S.M.; Effenberger, A.; Chen, C.-Y.; Maner-Smith, K.; Hajjar, I. APOE4 impairs myelination via cholesterol dysregulation in oligodendrocytes. Nature 2022, 611, 1–11. [Google Scholar] [CrossRef]
  22. Lin, Y.-T.; Seo, J.; Gao, F.; Feldman, H.M.; Wen, H.-L.; Penney, J.; Cam, H.P.; Gjoneska, E.; Raja, W.K.; Cheng, J. APOE4 causes widespread molecular and cellular alterations associated with Alzheimer’s disease phenotypes in human iPSC-derived brain cell types. Neuron 2018, 98, 1141–1154. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Victor, M.B.; Leary, N.; Luna, X.; Meharena, H.S.; Scannail, A.N.; Bozzelli, P.L.; Samaan, G.; Murdock, M.H.; von Maydell, D.; Effenberger, A.H. Lipid accumulation induced by APOE4 impairs microglial surveillance of neuronal-network activity. Cell Stem Cell 2022, 29, 1197–1212. [Google Scholar] [CrossRef] [PubMed]
  24. Rieker, C.; Migliavacca, E.; Vaucher, A.; Mayer, F.C.; Baud, G.; Marquis, J.; Charpagne, A.; Hegde, N.; Guignard, L.; McLachlan, M. Apolipoprotein E4 expression causes gain of toxic function in isogenic human induced pluripotent stem cell-derived endothelial cells. Arterioscler. Thromb. Vasc. Biol. 2019, 39, e195–e207. [Google Scholar] [CrossRef] [PubMed]
  25. 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, 2207. [Google Scholar] [CrossRef] [PubMed]
  26. Green, A.J.; Gelfand, J.M.; Cree, B.A.; Bevan, C.; Boscardin, W.J.; Mei, F.; Inman, J.; Arnow, S.; Devereux, M.; Abounasr, A. 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] [Green Version]
  27. 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, 1–13. [Google Scholar] [CrossRef]
  28. 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]
  29. Wang, F.; Yang, Y.-J.; Yang, N.; Chen, X.-J.; Huang, N.-X.; Zhang, J.; Wu, Y.; Liu, Z.; Gao, X.; Li, T. Enhancing oligodendrocyte myelination rescues synaptic loss and improves functional recovery after chronic hypoxia. Neuron 2018, 99, 689–701.e685. [Google Scholar] [CrossRef] [Green Version]
  30. Chen, J.-F.; Liu, K.; Hu, B.; Li, R.-R.; Xin, W.; Chen, H.; Wang, F.; Chen, L.; Li, R.-X.; Ren, S.-Y. Enhancing myelin renewal reverses cognitive dysfunction in a murine model of Alzheimer’s disease. Neuron 2021, 109, 2292–2307. [Google Scholar] [CrossRef]
  31. 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.A. Clemastine rescues myelination defects and promotes functional recovery in hypoxic brain injury. Brain 2018, 141, 85–98. [Google Scholar] [CrossRef] [Green Version]
  32. 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]
  33. Alberdi, E.; Sánchez-Gómez, M.V.; Matute, C. Calcium and glial cell death. J. Clin. Psychopharmacol. 2005, 38, 417–425. [Google Scholar] [CrossRef] [Green Version]
  34. 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] [Green Version]
  35. Yuan, F.; Wang, T. Glutamate-induced swelling of cultured astrocytes is mediated by metabotropic glutamate receptor. Sci. China Ser. C Life Sci.-Engl. Ed.- 1996, 39, 517–522. [Google Scholar]
  36. Wang, H.; Ward, N.; Boswell, M.; Katz, D.M. Secretion of brain-derived neurotrophic factor from brain microvascular endothelial cells. Eur. J. Neurosci. 2006, 23, 1665–1670. [Google Scholar] [CrossRef]
  37. Salinska, E.; Sobczuk, A.; Lazarewicz, J.W. Dantrolene antagonizes the glycineB site of the NMDA receptor. Neurosci. Lett. 2008, 432, 137–140. [Google Scholar] [CrossRef]
  38. Lei, S.Z.; Zhang, D.; Abele, A.E.; Lipton, S.A. Blockade of NMDA receptor-mediated mobilization of intracellular Ca2+ prevents neurotoxicity. Brain Res. 1992, 598, 196–202. [Google Scholar] [CrossRef]
  39. Kelliher, M.; Fastbom, J.; Cowburn, R.; Bonkale, W.; Ohm, T.; Ravid, R.; Sorrentino, V.; O’Neill, C. Alterations in the ryanodine receptor calcium release channel correlate with Alzheimer’s disease neurofibrillary and β-amyloid pathologies. Neuroscience 1999, 92, 499–513. [Google Scholar] [CrossRef]
  40. Chakroborty, S.; Goussakov, I.; Miller, M.B.; Stutzmann, G.E. Deviant ryanodine receptor-mediated calcium release resets synaptic homeostasis in presymptomatic 3xTg-AD mice. J. Neurosci. 2009, 29, 9458–9470. [Google Scholar] [CrossRef] [Green Version]
  41. Muehlschlegel, S.; Sims, J.R. Dantrolene: Mechanisms of neuroprotection and possible clinical applications in the neurointensive care unit. Neurocrit. Care 2009, 10, 103–115. [Google Scholar] [CrossRef] [Green Version]
  42. Liou, B.; Peng, Y.; Li, R.; Inskeep, V.; Zhang, W.; Quinn, B.; Dasgupta, N.; Blackwood, R.; Setchell, K.D.; Fleming, S. Modulating ryanodine receptors with dantrolene attenuates neuronopathic phenotype in Gaucher disease mice. Hum. Mol. Genet. 2016, 25, 5126–5141. [Google Scholar] [CrossRef] [PubMed]
  43. Keles, I.; Bozkurt, M.F.; Aglamis, E.; Fidan, A.F.; Ceylan, C.; Karalar, M.; Coban, S.; Denk, B.; Büyükokuroğlu, M.E. Protective effects of dantrolene and methylprednisolone against spinal cord injury-induced early oxidative damage in rabbit bladder: A comparative experimental study. Adv. Clin. Exp. Med. 2019, 28, 1697–1704. [Google Scholar] [CrossRef] [PubMed]
  44. Todorova, V.K.; Siegel, E.R.; Kaufmann, Y.; Kumarapeli, A.; Owen, A.; Wei, J.Y.; Makhoul, I.; Klimberg, V.S. Dantrolene attenuates cardiotoxicity of doxorubicin without reducing its antitumor efficacy in a breast cancer model. Transl. Oncol. 2020, 13, 471–480. [Google Scholar] [CrossRef] [PubMed]
  45. Zhang, L.; Andou, Y.; Masuda, S.; Mitani, A.; Kataoka, K. Dantrolene protects against ischemic, delayed neuronal death in gerbil brain. Neurosci. Lett. 1993, 158, 105–108. [Google Scholar] [CrossRef] [PubMed]
  46. Rowley, H.L.; Marsden, C.A.; Martin, K.F. Generalised seizure-induced changes in rat hippocampal glutamate but not GABA release are potentiated by repeated seizures. Neurosci. Lett. 1997, 234, 143–146. [Google Scholar] [CrossRef] [PubMed]
  47. Gursoy, I.D.; Barun, S.; Erdem, R.; Keskin, U.; Kiziltas, M.; Atilla, P.; Muftuoglu, S.; Yuce, D.; Narin, F.; Ertunc, M. Investigation of the Possible Protective Effects of Ketamine and Dantrolene on the Hippocampal Apoptosis and Spatial Learning in Rats Exposed to Repeated Electroconvulsive Seizures as a Model of Status Epilepticus. Turk Neurosurg. 2020, 30, 871–884. [Google Scholar] [CrossRef]
  48. Chen, X.; Wu, J.; Lvovskaya, S.; Herndon, E.; Supnet, C.; Bezprozvanny, I. Dantrolene is neuroprotective in Huntington’s disease transgenic mouse model. Mol. Neurodegener. 2011, 6, 1–12. [Google Scholar] [CrossRef] [Green Version]
  49. 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]
  50. Wakhloo, D.; Scharkowski, F.; Curto, Y.; Butt, U.J.; Bansal, V.; Steixner-Kumar, A.A.; Wüstefeld, L.; Rajput, A.; Arinrad, S.; Zillmann, M.R. Functional hypoxia drives neuroplasticity and neurogenesis via brain erythropoietin. Nat. Commun. 2020, 11, 1–12. [Google Scholar] [CrossRef] [Green Version]
  51. 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]
  52. 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]
  53. Adamcio, B.; Sargin, D.; Stradomska, A.; Medrihan, L.; Gertler, C.; Theis, F.; Zhang, M.; Müller, M.; Hassouna, I.; Hannke, K. Erythropoietin enhances hippocampal long-term potentiation and memory. BMC Biol. 2008, 6, 37. [Google Scholar] [CrossRef] [Green Version]
  54. 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]
  55. 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]
  56. 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]
  57. 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] [Green Version]
  58. 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]
  59. 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. FTY720 (fingolimod) efficacy in an animal model of multiple sclerosis requires astrocyte sphingosine 1-phosphate receptor 1 (S1P1) modulation. Proc. Natl. Acad. Sci. USA 2011, 108, 751–756. [Google Scholar] [CrossRef] [Green Version]
  60. Hoffmann, F.S.; Hofereiter, J.; Rübsamen, H.; Melms, J.; Schwarz, S.; Faber, H.; Weber, P.; Pütz, B.; Loleit, V.; Weber, F. Fingolimod induces neuroprotective factors in human astrocytes. J. Neuroinflamm. 2015, 12, 1–13. [Google Scholar] [CrossRef] [Green Version]
  61. 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. Fingolimod attenuates ceramide-induced blood–brain barrier dysfunction in multiple sclerosis by targeting reactive astrocytes. Acta Neuropathol. 2012, 124, 397–410. [Google Scholar] [CrossRef]
  62. Spiegel, S.; Milstien, S. Sphingosine-1-phosphate: Signaling inside and out. FEBS Lett. 2000, 476, 55–57. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Cruz, V.T.; Fonseca, J. Central effects of fingolimod. Rev. Neurol. 2014, 59, 121–128. [Google Scholar] [PubMed]
  64. 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. Effect of fingolimod on oligodendrocyte maturation under prolonged cerebral hypoperfusion. Brain Res. 2019, 1720, 146294. [Google Scholar] [CrossRef] [PubMed]
  65. 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]
  66. Joshi, P.; Gabrielli, M.; Ponzoni, L.; Pelucchi, S.; Stravalaci, M.; Beeg, M.; Mazzitelli, S.; Braida, D.; Sala, M.; Boda, E. Fingolimod limits acute Aβ neurotoxicity and promotes synaptic versus extrasynaptic NMDA receptor functionality in hippocampal neurons. Sci. Rep. 2017, 7, 1–15. [Google Scholar] [CrossRef] [Green Version]
  67. 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]
  68. Di Menna, L.; Molinaro, G.; Di Nuzzo, L.; Riozzi, B.; Zappulla, C.; Pozzilli, C.; Turrini, R.; Caraci, F.; Copani, A.; Battaglia, G. Fingolimod protects cultured cortical neurons against excitotoxic death. Pharmacol. Res. 2013, 67, 1–9. [Google Scholar] [CrossRef]
  69. Gurevich, I.; Tamir, H.; Arango, V.; Dwork, A.J.; Mann, J.J.; Schmauss, C. Altered Editing of Serotonin 2C Receptor Pre-mRNA in the Prefrontal Cortex of Depressed Suicide Victims. Neuron 2002, 34, 349–356. [Google Scholar] [CrossRef] [Green Version]
  70. 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]
  71. 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]
  72. Rossi, S.; Lo Giudice, T.; De Chiara, V.; Musella, A.; Studer, V.; Motta, C.; Bernardi, G.; Martino, G.; Furlan, R.; Martorana, A. Oral fingolimod rescues the functional deficits of synapses in experimental autoimmune encephalomyelitis. Br. J. Pharmacol. 2012, 165, 861–869. [Google Scholar] [CrossRef] [Green Version]
  73. 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]
  74. Bai, B.; Lunn, S.; Avila, R.; Wang, J.; Chmura, D.; Benson, E.; Kidd, G.; Medicetty, S.; Trapp, B. Fingolimod Reduces Axonal Transection during Demyelination (P1. 153); AAN Enterprises: Philadelphia, PA, USA, 2015. [Google Scholar]
  75. Nishihara, H.; Shimizu, F.; Sano, Y.; Takeshita, Y.; Maeda, T.; Abe, M.; Koga, M.; Kanda, T. Fingolimod prevents blood-brain barrier disruption induced by the sera from patients with multiple sclerosis. PLoS ONE 2015, 10, e0121488. [Google Scholar] [CrossRef]
  76. 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]
  77. 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]
  78. Kinoshita, M.; Hirayama, Y.; Fujishita, K.; Shibata, K.; Shinozaki, Y.; Shigetomi, E.; Takeda, A.; Le, H.; Hayashi, H.; Hiasa, M. Anti-Depressant Fluoxetine Reveals its Therapeutic Effect Via Astrocytes. Ebiomedicine 2018, 32, 72–83. [Google Scholar] [CrossRef]
  79. 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]
  80. 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]
  81. 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]
  82. 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]
  83. 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]
  84. 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]
  85. 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] [PubMed]
  86. 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] [PubMed]
  87. 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] [Green Version]
  88. 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]
  89. 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, 1–16. [Google Scholar] [CrossRef] [Green Version]
  90. 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, 494. [Google Scholar] [CrossRef]
  91. 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]
  92. 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. Fluoxetine prevents MPTP-induced loss of dopaminergic neurons by inhibiting microglial activation. Neuropharmacology 2011, 60, 963–974. [Google Scholar] [CrossRef]
  93. Zhang, F.; Zhou, H.; Wilson, B.C.; Shi, J.-S.; Hong, J.-S.; Gao, H.-M. Fluoxetine protects neurons against microglial activation-mediated neurotoxicity. Parkinsonism Relat. Disord. 2012, 18, S213–S217. [Google Scholar] [CrossRef] [Green Version]
  94. 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, 1–14. [Google Scholar] [CrossRef] [Green Version]
  95. 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] [Green Version]
  96. Green, M.V.; Pengo, T.; Raybuck, J.D.; Naqvi, T.; McMullan, H.M.; Hawkinson, J.E.; Marron Fernandez de Velasco, E.; Muntean, B.S.; Martemyanov, K.A.; Satterfield, R. Automated live-cell imaging of synapses in rat and human neuronal cultures. Front. Cell. Neurosci. 2019, 13, 467. [Google Scholar] [CrossRef] [Green Version]
  97. 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]
  98. 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]
  99. 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]
  100. Tyagarajan, S.K.; Ghosh, H.; Yévenes, G.E.; Nikonenko, I.; Ebeling, C.; Schwerdel, C.; Sidler, C.; Zeilhofer, H.U.; Gerrits, B.; Muller, D. Regulation of GABAergic synapse formation and plasticity by GSK3β-dependent phosphorylation of gephyrin. Proc. Natl. Acad. Sci. USA 2011, 108, 379–384. [Google Scholar] [CrossRef] [Green Version]
  101. 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. [Google Scholar]
  102. 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]
  103. 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]
  104. 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] [PubMed] [Green Version]
  105. Fornai, F.; Longone, P.; Cafaro, L.; Kastsiuchenka, O.; Ferrucci, M.; Manca, M.L.; Lazzeri, G.; Spalloni, A.; Bellio, N.; Lenzi, P. Lithium delays progression of amyotrophic lateral sclerosis. Proc. Natl. Acad. Sci. USA 2008, 105, 2052–2057. [Google Scholar] [CrossRef] [Green Version]
  106. 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] [PubMed]
  107. 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] [PubMed] [Green Version]
  108. Kim, E.C.; Meng, H.; Jun, A.S. Lithium treatment increases endothelial cell survival and autophagy in a mouse model of Fuchs endothelial corneal dystrophy. Br. J. Ophthalmol. 2013, 97, 1068–1073. [Google Scholar] [CrossRef] [Green Version]
  109. 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] [Green Version]
  110. 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. 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]
  111. Nassar, A.; Azab, A.N. Effects of lithium on inflammation. ACS Chem. Neurosci. 2014, 5, 451–458. [Google Scholar] [CrossRef] [Green Version]
  112. 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. Neuroinflammation 2014, 11, 140. [Google Scholar] [CrossRef] [Green Version]
  113. 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]
  114. 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]
  115. Chen, H.S.; Lipton, S.A. The chemical biology of clinically tolerated NMDA receptor antagonists. Neurochem 2006, 97, 1611–1626. [Google Scholar] [CrossRef] [PubMed]
  116. Dimpfel, W. Effects of memantine on synaptic transmission in the hippocampus in vitro. Arzneimittelforschung 1995, 45, 1–5. [Google Scholar] [PubMed]
  117. 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]
  118. 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]
  119. Zhao, Y.; Xiao, M.; He, W.; Cai, Z. 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. [Google Scholar]
  120. 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 2008, 32, 336–339. [Google Scholar] [CrossRef]
  121. 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. Mem. 2015, 121, 20–29. [Google Scholar] [CrossRef]
  122. 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]
  123. Kushnareva, Y.; Gerencser, A.; Bossy, B.; Ju, W.; White, A.; Waggoner, J.; Ellisman, M.; Perkins, G.; Bossy-Wetzel, E. Loss of OPA1 disturbs cellular calcium homeostasis and sensitizes for excitotoxicity. Cell Death Differ. 2013, 20, 353–365. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. 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] [Green Version]
  125. 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] [Green Version]
  126. 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]
  127. 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]
  128. 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]
  129. 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] [Green Version]
  130. 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]
  131. 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]
  132. Hunter, C.L.; Bachman, D.; Granholm, A.C. Minocycline prevents cholinergic loss in a mouse model of Down’s syndrome. Ann. Neurol. Off. J. Am. Neurol. Assoc. Child Neurol. Soc. 2004, 56, 675–688. [Google Scholar] [CrossRef]
  133. 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] [PubMed] [Green Version]
  134. Fernandez-Gomez, F.; Galindo, M.; Gomez-Lazaro, M.; González-García, C.; Ceña, V.; Aguirre, N.; Jordán, J. Involvement of Mitochondrial Potential and Calcium Buffering Capacity in Minocycline Cytoprotective Actions. Neuroscience 2005, 133, 959–967. [Google Scholar] [CrossRef] [PubMed]
  135. 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]
  136. 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]
  137. Schmidtner, A.K.; Slattery, D.A.; Gläsner, J.; Hiergeist, A.; Gryksa, K.; Malik, V.A.; Hellmann-Regen, J.; Heuser, I.; Baghai, T.C.; Gessner, A. Minocycline alters behavior, microglia and the gut microbiome in a trait-anxiety-dependent manner. Transl. Psychiatry 2019, 9, 1–12. [Google Scholar] [CrossRef] [Green Version]
  138. 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] [Green Version]
  139. 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]
  140. Zhang, Q.; Zhu, W.; Xu, F.; Dai, X.; Shi, L.; Cai, W.; Mu, H.; Hitchens, T.K.; Foley, L.M.; Liu, X. The interleukin-4/PPARγ signaling axis promotes oligodendrocyte differentiation and remyelination after brain injury. PLoS Biol. 2019, 17, e3000330. [Google Scholar] [CrossRef] [Green Version]
  141. Huang, W.; Shao, M.; Liu, H.; Chen, J.; Hu, J.; Zhu, L.; Liu, F.; Wang, D.; Zou, Y.; Xiong, Y. 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]
  142. 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]
  143. 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. [Google Scholar] [CrossRef] [Green Version]
  144. 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]
  145. 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]
  146. 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]
  147. Fukunaga, Y.; Itoh, H.; Doi, K.; Tanaka, T.; Yamashita, J.; Chun, T.-H.; Inoue, M.; Masatsugu, K.; Sawada, N.; Saito, T. Thiazolidinediones, peroxisome proliferator-activated receptor γ agonists, regulate endothelial cell growth and secretion of vasoactive peptides. Atherosclerosis 2001, 158, 113–119. [Google Scholar] [CrossRef]
  148. Wang, D.; Shi, L.; Xin, W.; Xu, J.; Xu, J.; Li, Q.; Xu, Z.; Wang, J.; Wang, G.; Yao, W. Activation of PPARγ inhibits pro-inflammatory cytokines production by upregulation of miR-124 in vitro and in vivo. Biochem. Biophys. Res. Commun. 2017, 486, 726–731. [Google Scholar] [CrossRef]
  149. 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]
  150. 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, 23, 19–31. [Google Scholar] [CrossRef]
  151. 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. [Google Scholar] [CrossRef] [Green Version]
  152. 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. 2008, 28, 613–627. [Google Scholar] [CrossRef]
  153. 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]
  154. 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]
  155. Brandao, F.; Paula-Barbosa, M.M.; Cadete-Leite, A. Piracetam impedes hippocampal neuronal loss during withdrawal after chronic alcohol intake. Alcohol 1995, 12, 279–288. [Google Scholar] [CrossRef] [PubMed]
  156. 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] [PubMed]
  157. Winblad, B. Piracetam: A review of pharmacological properties and clinical uses. CNS Drug Rev. 2005, 11, 169–182. [Google Scholar] [CrossRef]
  158. 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]
  159. 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]
  160. Pereira, A.C.; Gray, J.D.; Kogan, J.F.; Davidson, R.L.; 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 2017, 22, 296. [Google Scholar] [CrossRef]
  161. Urbani, A.; Belluzzi, O. Riluzole inhibits the persistent sodium current in mammalian CNS neurons. Eur. J. Neurosci. 2000, 12, 3567–3574. [Google Scholar] [CrossRef]
  162. Lesuis, S.; Kaplick, P.; Lucassen, P.; Krugers, H. 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]
  163. Katoh-Semba, 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]
  164. Malgouris, C.; Bardot, F.; Daniel, M.; Pellis, F.; Rataud, J.; Uzan, A.; Blanchard, J.-C.; Laduron, P.M. Riluzole, a novel antiglutamate, prevents memory loss and hippocampal neuronal damage in ischemic gerbils. J. Neurosci. 1989, 9, 3720–3727. [Google Scholar] [CrossRef]
  165. Kalra, S.; Cashman, N.; Genge, A.; Arnold, D. Recovery of N-acetylaspartate in Corticomotor Neurons of Patients With ALS After Riluzole Therapy. Neuroreport 1998, 9, 1757–1761. [Google Scholar] [CrossRef]
  166. Storch, A.; Burkhardt, K.; Ludolph, A.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]
  167. Umemiya, M.; Berger, A.J. Inhibition by riluzole of glycinergic postsynaptic currents in rat hypoglossal motoneurones. Br. J. Pharmacol. 1995, 116, 3227. [Google Scholar] [CrossRef] [Green Version]
  168. 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]
  169. 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]
  170. Beasley, C.M., Jr.; Koke, S.C.; Nilsson, M.E.; Gonzales, J.S. Adverse events and treatment discontinuations in clinical trials of fluoxetine in major depressive disorder: An updated meta-analysis. Clin. Ther. 2000, 22, 1319–1330. [Google Scholar] [CrossRef]
  171. Grandjean, E.M.; Aubry, J.-M. Lithium: Updated human knowledge using an evidence-based approach. CNS Drugs 2009, 23, 397–418. [Google Scholar] [CrossRef]
  172. Schoot, T.S.; Molmans, T.H.; Grootens, K.P.; Kerckhoffs, A.P. Systematic review and practical guideline for the prevention and management of the renal side effects of lithium therapy. Eur. Neuropsychopharmacol. 2020, 31, 16–32. [Google Scholar] [CrossRef]
  173. Macdonald, A.; Briggs, K.; Poppe, M.; Higgins, A.; Velayudhan, L.; Lovestone, S. A feasibility and tolerability study of lithium in Alzheimer’s disease. Int. J. Geriatr. Psychiatry A J. Psychiatry Late Life Allied Sci. 2008, 23, 704–711. [Google Scholar] [CrossRef] [PubMed]
  174. Lincoff, A.M.; Wolski, K.; Nicholls, S.J.; Nissen, S.E. Pioglitazone and risk of cardiovascular events in patients with type 2 diabetes mellitus: A meta-analysis of randomized trials. J. Am. Med. Assoc. 2007, 298, 1180–1188. [Google Scholar] [CrossRef] [PubMed]
  175. Betteridge, D.J. Effects of pioglitazone on lipid and lipoprotein metabolism. Diabetes Obes. Metab. 2007, 9, 640–647. [Google Scholar] [CrossRef]
  176. Dormandy, J.; Bhattacharya, M.; de Bruyn, A.-R.v.T. Safety and tolerability of pioglitazone in high-risk patients with type 2 diabetes. Drug Saf. 2009, 32, 187–202. [Google Scholar] [CrossRef] [PubMed]
  177. Dubois, B.; Feldman, H.H.; Jacova, C.; Hampel, H.; Molinuevo, J.L.; Blennow, K.; DeKosky, S.T.; Gauthier, S.; Selkoe, D.; Bateman, R. Advancing research diagnostic criteria for Alzheimer’s disease: The IWG-2 criteria. Lancet Neurol. 2014, 13, 614–629. [Google Scholar] [CrossRef]
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. Cure of Alzheimer’s Dementia Requires Addressing All of the Affected Brain Cell Types. J. Clin. Med. 2023, 12, 2049. https://doi.org/10.3390/jcm12052049

AMA Style

Fessel J. Cure of Alzheimer’s Dementia Requires Addressing All of the Affected Brain Cell Types. Journal of Clinical Medicine. 2023; 12(5):2049. https://doi.org/10.3390/jcm12052049

Chicago/Turabian Style

Fessel, Jeffrey. 2023. "Cure of Alzheimer’s Dementia Requires Addressing All of the Affected Brain Cell Types" Journal of Clinical Medicine 12, no. 5: 2049. https://doi.org/10.3390/jcm12052049

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