Next Article in Journal
Modular Synthesis and Antiproliferative Activity of New Dihydro-1H-pyrazolo[1,3-b]pyridine Embelin Derivatives
Next Article in Special Issue
Combinatorial Regimen of Carbamazepine and Imipramine Exhibits Synergism against Grandmal Epilepsy in Rats: Inhibition of Pro-Inflammatory Cytokines and PI3K/Akt/mTOR Signaling Pathway
Previous Article in Journal
Identification of a Novel Neuropeptide S Receptor Antagonist Scaffold Based on the SHA-68 Core
Previous Article in Special Issue
The Effects of N-Acetylcysteine on the Rat Mesocorticolimbic Pathway: Role of mGluR5 Receptors and Interaction with Ethanol
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Repurposing Peroxisome Proliferator-Activated Receptor Agonists in Neurological and Psychiatric Disorders

1
Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, 09042 Monserrato, Italy
2
Neuroscience Institute, National Research Council of Italy (CNR), Section of Cagliari, 09042 Monserrato, Italy
*
Author to whom correspondence should be addressed.
Pharmaceuticals 2021, 14(10), 1025; https://doi.org/10.3390/ph14101025
Submission received: 16 September 2021 / Revised: 5 October 2021 / Accepted: 6 October 2021 / Published: 8 October 2021
(This article belongs to the Special Issue Repurposing Drug Strategies for CNS Disorders)

Abstract

:
Common pathophysiological mechanisms have emerged for different neurological and neuropsychiatric conditions. In particular, mechanisms of oxidative stress, immuno-inflammation, and altered metabolic pathways converge and cause neuronal and non-neuronal maladaptative phenomena, which underlie multifaceted brain disorders. The peroxisome proliferator-activated receptors (PPARs) are nuclear receptors modulating, among others, anti-inflammatory and neuroprotective genes in diverse tissues. Both endogenous and synthetic PPAR agonists are approved treatments for metabolic and systemic disorders, such as diabetes, fatty liver disease, and dyslipidemia(s), showing high tolerability and safety profiles. Considering that some PPAR-acting drugs permeate through the blood–brain barrier, the possibility to extend their scope from the periphery to central nervous system has gained interest in recent years. Here, we review preclinical and clinical evidence that PPARs possibly exert a neuroprotective role, thereby providing a rationale for repurposing PPAR-targeting drugs to counteract several diseases affecting the central nervous system.

1. Introduction

Over the past decade, evidence has shown converging mechanisms for different brain disorders whereby inflammation and cellular metabolic imbalance have emerged at different stages in neuropathological processes. Within the wide spectrum of neuropsychiatric disorders, finding overlapping substrates is key to managing the therapeutics and efficiently counteracting the clinical outcome. In this view, peroxisome proliferator-activated receptors (PPARs) are considered multi-purpose molecular targets. PPARs have been involved in medical conditions ranging from hepatocellular to myocardial diseases, and have been successfully targeted for drug development and pharmacological treatment. Evidence of the participation of PPARs in brain disorders is relatively recent, although remarkable. Genome-wide association studies, together with computational approach, suggest new therapeutic opportunities for PPAR agonists, which were already used for decades to treat different peripheral pathologies. Hence, the repurposing of existing drugs against central nervous system (CNS) diseases is prominent as a rapid and rationally “safe” approach, and is also promising for multiple organ dysfunction syndromes, as we will review and discuss in the following sections.

2. PPARs: From the Periphery to CNS

PPARs are intranuclear receptors, which serve as ligand-activated transcription factors [1,2]. Their name comes from the original observation that chronic activation of these receptors causes hepatic peroxisome proliferation in rodents [3]. Although this effect is not observed in humans and is limited to PPARα, the name remains. Like other nuclear receptors, PPARs, once activated, bind to consensus DNA sequences to regulate gene expression through transcriptional co-activation [4,5,6]. The PPAR subfamily comprises three isoforms: PPARα, PPARβ/δ and PPARγ, which share a structural homology [1,2,7,8]. PPARs are activated by small, lipophilic molecules and, in the cytoplasm, form heterodimers with the retinoid X receptor-α (RXR) to regulate gene expression [9].
The activated protein-DNA complex, PPAR/RXR, targets the peroxisome proliferator response element (PPRE) within the gene promoter [1,10], eliciting intracellular molecular cascades [10].
PPARs are recognized to regulate the expression of several target genes in both the periphery and the CNS. These are primarily implicated in energy regulation, lipid and glucose metabolism, and in anti-inflammatory processes. Considering their pleiotropic functions, the investigation of pathological deregulation of PPARs, together with their potential pharmacological activation, is a topic of increasing interest in both preclinical and clinical research. The implications of PPARs as lipid sensors are well known in whole-body adipogenesis and hepatic homeostasis, so that their impairment is associated with progressive liver fibrosis and steatosis [11,12,13]. PPARs have also been involved in regulating mitochondrial β-oxidation and cellular growth [14], with the β/δ isotype being particularly considered as a potential therapeutic target for metabolic syndromes [15,16,17].
Among the endogenous agonists, the fatty acid ethanolamides, also known as N-acylethanolamines (NAEs), show a high affinity. These molecules are long-chain fatty acids [18,19] that are ubiquitously found in animal tissues [20], in both the periphery and the brain. The most-studied NAEs are arachidonoylethanolamide (anandamide, AEA), palmitoylethanolamide (PEA) and oleoylethanolamide (OEA). Other, less-characterized NAEs are stearoylethanolamide (SEA) and linoleoylethanolamide (LEA) [21,22]. AEA, the first endocannabinoid to be identified [23], is the only NAE that binds to cannabinoid type 1 (CB1) and type 2 (CB2) receptors with high affinity. PEA and OEA were known for many years, but their role, as well as that of other NAEs, in the CNS, was only characterized in the last two decades [24].
PPARα agonists such as fibrates have long been clinically used to treat hypertriglyceridemia, whereas PPARγ agonists, such as thiazolidinediones, are approved for type-2 diabetes (T2D) [25,26,27]. Recent findings show the impact of PPARs in cancer development, although whether this is a direct influence or a consequence of their target gene transcription has not been elucidated [26,28,29].
In the CNS, PPAR mRNA and protein are ubiquitously expressed, suggesting that these receptors are involved in the regulation of neuron and glial cell metabolism and energy balance. Different brain disorders, including neurological, neuropsychiatric and neurodegenerative diseases, share general etiopathogenic factors such as an altered cellular metabolism, modifications in synaptic/electrical activity and concurrent inflammatory processes. Remarkably, PPARs regulate genes that are involved in all of these components. This suggests that the agonists for different isotypes of PPAR could be attractive pharmacological tools that might regulate different pathological mechanisms at once. Their potential contribution against inflammation, pain, demyelination, and epileptic seizures [30,31] is emerging; preventive neuroprotection in the early stages of degenerative disorders would also be desirable [32]. Increasing evidence shows the pivotal role of PPARs in the whole organism and overlapping physiopathological processes for different disorders; thus, the repositioning of the already marketed PPAR-acting medications in brain disorders appears attractive. Moreover, given the partial overlap in genes modulated by PPARα and -γ, newly developed co-activators, such as the dual-acting agonist tesaglitazar, deserve special mention for future clinical applications [33,34].

3. The Pleiotropic Mechanism of Action of PPARs: Transcriptional Effects and Rapid Synaptic Regulation

PPARs display isoform-specific tissue expression, as well as a plethora of endogenous ligands, which show different binding affinities. Inducing/suppressing control of several genes ultimately leads to a multifaceted impact, in both physiological and pathological conditions. For instance, pan-PPAR targeting would regulate the overall energy balance [13]. Nevertheless, PPARα would mostly influence the fatty acid metabolism and promote low triglyceride levels, while PPARγ would mainly influence lipid biosynthesis/adipogenesis, glucose metabolism and intracellular insulin signalling, and PPARβ/δ would operate on fatty acid oxidation and blood glucose [17,26,35].
In the brain, activation of PPARs occurs in genetic mechanisms at different levels, ranging from basal cellular neurobiology to complex cognitive processes. Hence, PPARs positively or negatively modulate the genes that are implicated in sleep/circadian rhythm, feeding behaviors, neuroinflammatory and degenerative processes, leading to overall neuroprotective effects [32,36]. In this regard, PPARγ, which is the most abundant isoform in microglia, suppresses the immunoinflammatory response by regulating genes for the biosynthesis of cytokines, prostaglandins and nitric oxide, as well as inducing the apoptosis of reactive microglial cells [37]. PPARα modulates the synthesis of proteins involved in neuroprotection and regulates synaptic signaling. At present, the -β/δ is the least-characterized isoform in the CNS, although it is only highly expressed in neuronal cell types [38]. However, a relevant recent study found PPARβ/δ to be repressed by the mutant form of huntingtin (HTT) in Huntington’s disease (HD), while its activation reverted transcriptional alterations associated with neurodegeneration, motor impairments and mitochondrial abnormalities in experimental models of HD [39,40]. According to the emerging evidence, the repurposing of PPAR agonists is increasingly promising in several human brain disorders.
In addition to the multiple, even opposing, genomic actions of PPARs, rapid effects have also been described. Non-liganded PPARβ/δ negatively regulates the -α and -γ downstream signaling, suggesting cross-talk amongst isoforms when expressed in the same cell type [41]. This mutual tuning prompts possible implications concerning PPAR-based therapeutical approaches.
PPARγ has been shown to affect, among others, platelet functionality. In different cells, PPARγ represses β-catenin signaling and/or triggers mitogen-activated protein kinase (MAPK) pathways. Specifically, PPARγ-dependent phosphorylation was observed over the classical extracellular signal-regulated kinase tipe1/2 (MAPK ERK1/2) in tumoral cell lines, in contrast with the dephosphorylation of myosin phosphatase in aortic smooth myocytes. Finally, PPARα also exhibits non-genomic effects. For instance, agonists for α isotype reduce glucose-induced intracellular Ca2+ concentration and insulin secretion in pancreatic β cells and regulate β subunit phosphorylation in nicotinic acetylcholine (ACh) receptors expressed by midbrain dopamine neurons, modulating their electrical activity in turn. This mechanism has particular relevance for several dopamine-associated neurological and neuropsychiatric conditions [24,42]. The peculiar dual-genomic and non-genomic pathway that follows the activation of PPARα warrants further investigation and suggests that these nuclear receptors are a promising therapeutic target in diverse psychiatric and neurological illnesses [43,44].
Neurodevelopmental disorders such as autism and schizophrenia [45,46,47,48], as well as mood disorders [43,49,50], have been shown to exhibit a disruption of neuroimmune functions. Thus, it is particularly tempting to repurpose PPAR agonists for neurological and psychiatric disorders, as detailed in the following sections.

4. PPAR Agonists in Psychiatric Disorders

PPARα seems to be critically involved in the pathophysiology of schizophrenia [44,51,52]. The evidence for this derives from preclinical studies with the maternal immune activation (MIA) model or the postnatal lesional model and from clinical investigations into the association of schizophrenia with PPARα genes. In the MIA model, an inflammatory insult is evoked at a precise stage of pregnancy. Consequently, this insult disrupts brain development in offspring, which shows behavioral, neurophysiological and neurochemical imbalances. The PPARα agonist fenofibrate, which is approved for the treatment of hypercholesterolemia and hypertriglyceridemia, was shown to attenuate behavioral disruption and dopaminergic dysfunction in MIA offspring [53,54]. Although the precise mechanism is still under investigation, it is hypothesized that PPARα activation negatively regulates the maternal inflammatory response by dampening nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB) signaling, activator protein 1 (AP-1) [55], tumor necrosis factor-α (TNF-α) [56], as well as inhibiting the production of cytokines and interferons. Fenofibrate was also evaluated in a neurodevelopmental rat model of schizophrenia consisting of postnatal kainic acid injection. In this model, fenofibrate attenuated the disruption of pre-pulse inhibition, a measure of sensorimotor gating functions whose deficits accompany a psychotic-like phenotype in young adulthood [57]. The gene encoding for PPARα (PPARA) was found to be downregulated in hair follicle cells from schizophrenia patients [51], further showing an association between PPARα and this psychiatric condition [52].
Consistent with the notion that schizophrenia displays a pro-inflammatory phenotype [45,46,47], genes involved in inflammation have been found to be affected, including decreased PPARα mRNA levels and increased IL-6 and TNFα. Additionally, a study on a Croatian population investigated whether a functional L162V polymorphism in the PPARα gene was also associated with schizophrenia risk [58]. Remarkably, the PPARα-L162V polymorphism impacts the clinical manifestation (i.e., severity) of the illness, plasma lipid concentrations, and the risk of addiction to tobacco (i.e., nicotine) in female patients. Nonetheless, an association with schizophrenia risk was not reported [58,59].
Rosiglitazone, a PPARγ agonist approved for T2D, has been proposed to improve the cognitive symptoms of schizophrenia due to its ability to induce the expression of BDNF [60]. However, in a pilot study by Yi et al. [61], rosiglitazone administration had no significant beneficial effect on the cognitive performance of patients with schizophrenia, who were also administered with clozapine. Conversely, results from Watson et al. [62] showed improved cognitive tasks, at least in Alzheimer’s Disease (AD)-affected patients treated with rosiglitazone. The contrasting results are not surprising, given the differences in study populations, dosing, and treatment duration [61]. On the other hand, pioglitazone (another PPARγ agonist approved for T2D) showed a positive effect. Smith et al. [63] demonstrated that pioglitazone is efficacious to treat glucose and lipid abnormalities in schizophrenic patients, and reduces their symptoms, as measured by the positive and negative syndrome scale (PANSS). Based on the high prevalence of diabetes in patients with schizophrenia [64], pioglitazone, concomitant with risperidone, appeared to be beneficial for counteracting negative symptoms in schizophrenia [65], such as lack of concentration, loss of interest, and social withdrawal.
The converging evidence suggests a reciprocal modulation of depressive states and inflammation [50,66,67,68]. Notably, stress plays a critical role in the expression of depressive symptoms, being associated with alterations in immunological and neuroendocrine responses, as well as circuit changes perpetuating depressive moods [68]. Furthermore, imbalances in cholinergic systems have long been hypothesized to be crucially involved in the pathophysiology of depression [69,70]. In this scenario, the observation that activation of PPARα is engaged by dopamine cells as a self-regulatory mechanism in response to a hypercholinergic drive [71] makes it a potential target in the treatment of depressive states. Accordingly, in rodents, the acute direct and indirect stimulation of PPARα results in an effective anti-depressant-like activity [71,72,73]. Chronic administration of either synthetic or natural PPARα ligands has been shown to prevent and relieve depressive behaviors resulting from chronic stress exposure [49,72,73,74], as well as restoring dopamine system function [49] and hippocampal BDNF signaling cascade [73], which are key to the pathogenesis of mood disorders [75,76]. Notably, not only is the PPARα-dependent enhancement of hippocampal BDNF signaling and neurogenesis involved in the pathophysiology of depression, it is also involved in the mechanism of action of the antidepressant fluoxetine [77]. These effects add to the many other actions triggered by these receptors, including the increased biosynthesis of neuroactive steroids, which exert antidepressant activity [78]. This is not unexpected, as PPARα is a nuclear receptor at the crossroads of many signaling pathways and, as mentioned above, acts via genomic and non-genomic actions. Hence, the clinical evidence suggesting that the activation of these receptors by synthetic agonists is effective in diverse depressive states [43,78,79], along with their safe pharmacological profile, further demonstrates that it may be a promising and feasible antidepressant target.
PPARγ agonists (i.e., pioglitazone, rosiglitazone) have proven their efficacy as antidepressants in animal models [80,81], as well as in patients that also presented insulin resistance [82,83,84]. Moreover, depressive symptom levels have been correlated with molecular alterations, such as the dysregulation of interleukin (IL)-6 levels [84], a further anti-inflammatory effect of PPARγ agonists.
The localization of PPAR isoforms in brain regions involved in the neurobiology of substance-use disorders (SUDs) [85], together with their broadly investigated influences on mesocorticolimbic dopamine system neurophysiology [24,42,86,87], highlights PPAR potential in this psychiatric condition [88,89]. Indeed, accumulating evidence suggests that neuroinflammation contributes to imbalanced reward/aversion circuits, leading to addictive behaviors. The stages of drug dependence, oversimplified as abuse/binge–high–withdrawal/compulsion for reuse–relapse (see [90] and [91]), suggest multiple therapeutical timepoints for successful treatment. The PPAR signaling cascade and their metabolic machinery are being considered as an innovative target, among others, for preclinical investigations into the development of therapies against alcohol [92,93,94,95,96,97], tobacco [98,99,100,101,102,103,104,105,106], opioid [107,108,109], and psychostimulant [110,111] dependence. Hence, the repurposing of PPAR-based medications is of increasing interest in this area of neuropsychiatry [111,112,113], although controversial results in humans [114,115] suggest that cautiousness is required. Table 1 summarizes the preclinical and clinical studies aiming to repurpose PPARα and PPARγ agonists in the treatment of neurological and psychiatric diseases.

5. PPAR Agonists in Neurological Disorders

Given their multifaceted pharmacological properties (anti-inflammatory, neurotrophic/neuroprotective), PPAR pathways have also attracted considerable attention as potential therapeutic targets, as well as risk factors, in various neurological disorders, including AD, multiple sclerosis (MS), Parkinson’s Disease (PD), and epilepsy [43,44,79,132,133] (Table 1). PPARs appear to play a direct role in the pathophysiology of AD [134,135,136], the leading cause of dementia in the elderly. From a neuropathological perspective, PPARα has been implicated in the etiopathogenesis of AD, particularly in the upstream homeostasis of amyloid precursor protein (APP) [135]. The expression levels of PPARα and β/δ are significantly reduced, while PPARγ is selectively increased in AD brains, suggesting that a dysfunctional PPAR system might contribute to AD’s onset and progression. Accordingly, not only have several studies reported promising effects of different PPAR agonists in experimental models, but these effects have also been reported in AD patients [79,134,135,136]. Among the three receptor isotypes, the PPARγ is the first and the most extensively studied. Hence, initial preclinical and clinical evidence showed that PPARγ modulators improve learning and memory by reducing microglial activation and Aβ plaques, in both humans [62,126,137,138,139] and an AD mouse model [43,125]. However, in clinical trials, neither rosiglitazone or pioglitazone yielded meaningful outcomes [79,128,129]. Several in vitro and in vivo studies point to PPARα as an emerging therapeutic target for AD due to the ability of both natural (e.g., PEA) and synthetic agonists (e.g., fibrates) to modulate AD pathogenetic mechanisms and progression [136,140]. Scuderi et al. revealed that PEA’s anti-inflammatory properties accounted for a PPARα-dependent reduction in astrogliosis, pro-inflammatory signals and neuronal loss [141,142,143,144,145].
PPARα has been shown in preclinical models to be key for the neuroprotective and memory-rescuing effects of PEA [144,146]. It has consistently been shown that administration of gemfibrozil or permafibrate in a well-characterized model of familial AD (5XFAD transgenic mice co-expressing five mutations of familial AD and characterized by rapid brain amyloidosis) decreases amyloid plaque deposition, microgliosis and astrogliosis in the hippocampus and cortex, and is associated with a significant improvement in spatial learning, memory and hippocampal plasticity [122,123]. Luo and colleagues demonstrated that either gemfibrozil or Wy14643 reverse not only memory deficits, but also amyloid plaque pathology and anxiety in the APP-PSEN1ΔE9 model (co-expressing the Swedish APP mutation and exon 9 deletion of the PSEN1 gene) via a PPARα-dependent enhancement of autophagosome biogenesis [124]. Investigation of the β/δ isoform in the pathophysiology of AD is in the early stages [36]. However, a phase II clinical trial for the T3D-959 dual-agonist for the -β/δ and -γ isoforms [147] showed promising results for further studies in AD patients. Finally, the co-activator-1α (PGC1α), a PPARs’ transcriptional regulator, represents a possible therapeutic target. PGC1α protein levels were found to be reduced in the Tg2576 mouse model (that overexpresses a mutant form of APP with the Swedish mutation KM670/671NL, which increases Aβ levels and amyloid plaques) [148], and the mRNA in the brain of AD patients correlated with the progression of clinical dementia [149]. Accordingly, the polyphenol resveratrol has proven to be neuroprotective through the modulation of PGC1α [150,151]. Collectively, these findings support that, in AD, repurposing the thiazolidinediones as PGC1α/PPARγ-activating drugs warrants further investigations (see [152] for an extensive review).
Due to their role in the regulation of neuroinflammation and immune responses, PPAR dysfunction might also be involved in the mechanisms underlying MS [153,154], the most common disabling neurological condition among young adults. MS patients show decreased PPARγ expression [155] as well as reduced levels of NAEs (i.e., OEA, PEA, AEA) [156]. Of note, the latter tend to increase during MS clinical exacerbation, probably because NAEs act as endogenous neuroprotective molecules [156]. This is in accordance with the possible compensatory mechanism induced by increased levels of PEA in the CNS, as indexed by the reduced progression of neurodegenerative markers in two different mouse models of chronic MS
In the same MS chronic models, PEA administration reduced spasticity and motor disability, together with an anti-inflammatory effect [157,158]. PEA also reduced the expression of inflammatory cytokines in the myelin oligodendrocyte glycoprotein-induced experimental autoimmune encephalomyelitis (MOG-EAE) mice, an effect accompanied by decreased demyelination and axonal damage [159]. Similarly, the PPARγ agonist troglitazone attenuates inflammation and ameliorates EAE-related signs in mice [131]. Unfortunately, clinical data on the potential therapeutic use of PPARγ agonists are limited [43,79], although both drugs have proven efficacy in reducing lesion burden. The need to upscale these trials is, therefore, critically relevant.
Concerning PD, the most prevalent neurodegenerative movement disorder [160], a large body of evidence supports the protective/neurotrophic properties of different PPAR ligands [44]. OEA attenuates behavioral symptoms and exerts neuroprotection on the nigrostriatal system in an experimental model via a PPARα-dependent mechanism [161,162]. In addition, chronic treatment with PEA counteracts 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced glial cell activation, loss of nigral dopamine neurons, behavioral impairments, and motor dysfunctions, while the genetic ablation of PPARα exacerbates MPTP systemic toxicity [163]. Consistently, Barbiero et al. demonstrated the ability of the synthetic PPARα agonist fenofibrate to protect against the detrimental effects of MPTP in a rat model of PD [116,117]. The observation that both PEA and fenofibrate exert a neuroprotective effect after the onset of the pathology is remarkable, as disease-modifying drugs administered during disease evolution may delay the progression.
This is particularly significant in PD, as possible pharmacotherapeutic interventions are limited once the dopamine system has degenerated and subsequent symptoms have appeared [44,163].
Similarly, PPARγ ligands have been reported to mitigate PD progression in preclinical settings [43,164,165,166]. For instance, in different MPTP models, pioglitazone reduced microglial activation and iNOS-positive cells and expression of the monoamine oxidase B gene, which are considered protective against neurotoxicity, whereas rosiglitazone counteracted dopamine neuron loss and prevented olfactory and motor dysfunctions [127,130,166]. Accordingly, the PPARγ agonist, MDG548, and the PPARα/γ dual agonist, MHY908, exert neuroprotective effects by boosting phagocytosis and anti-inflammatory cytokines production, and by reducing microglial activation and neuroinflammation in MPTP mouse models of PD, thus attenuating neuronal damage and motor deficit [167,168]. When combined, these findings further support the involvement of activated PPARγ in microglial function, phagocytosis and neuroinflammation [43]. It is also worth mentioning the interaction between PPARγ and PGC-1α, which has recently proven to be neuroprotective. On the other hand, the results of a study [169] in which the PPARγ agonist pioglitazone was used in an attempt to modify disease progression in PD did not support the initiation of further trials with this drug (see also [44,170]).
PPAR modulation also represents a promising pharmacological approach against a common and prevalent neurological disorder, that is epilepsy, which, in its different manifestations, affects up to 2% of the global population [171].
Selective PPARα agonists have been proven to raise seizure thresholds in animal models of epilepsy, suggesting their possible repurposing in seizure management in patients [172]. PEA, for example, displayed anticonvulsant effects in mice [173] and showed antiepileptic actions in kindled rats [174] as well as in a genetic model of absence epilepsy [175]. Next, a study by Saha et al. [121] reported bezafibrate as being effective in the pentylentetrazole (PTZ)-induced kindling seizure model, and in reducing hippocampal cell loss in rat brain [121]. Likewise, the synthetic PPARα ligand fenofibrate and the ketogenic diet (KD) displayed the same efficacy as an anticonvulsant in adult rats, by decreasing PTZ-induced seizures and by extending the latency to the onset of epileptic symptoms induced by lithium-pilocarpine [118].
Moreover, acute and chronic fenofibrate is protective against nicotine-evoked epileptic manifestation and synchronization in the frontal cortex [119]. Lately, PPARα agonists have been proposed as a novel disease-modifying target for sleep-related hypermotor epilepsy (SHE), formerly referred to as nocturnal frontal lobe epilepsy (NFLE), idiopathic epilepsy with an autosomal inherited component, based on the effectiveness of fenofibrate in an NFLE mouse model and as an adjunctive therapy in pharmacoresistant NFLE patients [120]. The evidence also supports the repurposing of PPARγ agonists in epilepsy, given their neuroprotective and anti-seizure properties, with this isoform being involved in the KD mechanism of action [176], suggesting the repositioning of pioglitazone [177] and rosiglitazone [178,179,180] as therapeutic adjuvants for severe, refractory epilepsy.

6. Concluding Remarks

Since the advent of endocannabinoid system research, additional lipid signaling molecules have been found to play a role in brain functions. As they are ubiquitously expressed, PPARs are the new widgets in the pharmacologists’ toolkit. PPAR agonists are not miracle drugs, but their effects on lipid and glucose metabolism might affect the pathophysiological mechanisms of multifaceted psychiatric and neurological diseases. It is intriguing to speculate that one possible common denominator of all the disorders is the disruption of the neuronal and glial metabolism [181], which can be effectively modulated by PPARs.
Repurposing drugs by reprofiling their initial therapeutic indication, along with designing novel compounds with the same biological target, is pivotal to better understanding the pathophysiological underpinnings of brain disorders, but it is also paving new roads for their treatment.

Author Contributions

Writing—original draft preparation, C.S., M.M., A.L.M., M.P.; writing—review and editing, C.S., M.M., A.L.M., M.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by “FSC 2014–2020—Patto per lo Sviluppo della Regione Sardegna, Legge Regionale n. 7 del 7 agosto 2007 (Bando 2017)” and by “Progetti di Rilevante Interesse Nazionale” (PRIN) 2017 (2017YH3SXK).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Desvergne, B.; Wahli, W. Peroxisome proliferator-activated receptors: Nuclear control of metabolism. Endocr. Rev. 1999, 20, 649–688. [Google Scholar] [CrossRef] [Green Version]
  2. Straus, D.S.; Glass, C.K. Cyclopentenone prostaglandins: New insights on biological activities and cellular targets. Med. Res. Rev. 2001, 21, 185–210. [Google Scholar] [CrossRef]
  3. Green, S. PPAR: A mediator of peroxisome proliferator action. Mutat. Res. 1995, 333, 101–109. [Google Scholar] [CrossRef]
  4. Nolte, R.T.; Wisely, G.B.; Westin, S.; Cobb, J.E.; Lambert, M.H.; Kurokawa, R.; Rosenfeld, M.G.; Willson, T.M.; Glass, C.K.; Milburn, M.V. Ligand binding and co-activator assembly of the peroxisome proliferator-activated receptor-gamma. Nature 1998, 395, 137–143. [Google Scholar] [CrossRef]
  5. Berger, J.; Moller, D.E. The mechanisms of action of PPARs. Annu. Rev. Med. 2002, 53, 409–435. [Google Scholar] [CrossRef] [Green Version]
  6. Castrillo, A.; Tontonoz, P. Nuclear receptors in macrophage biology: At the crossroads of lipid metabolism and inflammation. Annu. Rev. Cell Dev. Biol. 2004, 20, 455–480. [Google Scholar] [CrossRef]
  7. Bishop-Bailey, D. Peroxisome proliferator-activated receptors in the cardiovascular system. Br. J. Pharmacol. 2000, 129, 823–834. [Google Scholar] [CrossRef] [Green Version]
  8. Buchan, K.W.; Hassall, D.G. PPAR agonists as direct modulators of the vessel wall in cardiovascular disease. Med. Res. Rev. 2000, 20, 350–366. [Google Scholar] [CrossRef]
  9. van Neerven, S.; Mey, J. RAR/RXR and PPAR/RXR Signaling in Spinal Cord Injury. PPAR Res. 2007, 2007, 29275. [Google Scholar] [CrossRef] [Green Version]
  10. Qi, C.; Zhu, Y.; Reddy, J.K. Peroxisome proliferator-activated receptors, coactivators, and downstream targets. Cell Biochem. Biophys. 2000, 32, 187–204. [Google Scholar] [CrossRef]
  11. Zardi, E.M.; Navarini, L.; Sambataro, G.; Piccinni, P.; Sambataro, F.M.; Spina, C.; Dobrina, A. Hepatic PPARs: Their role in liver physiology, fibrosis and treatment. Curr. Med. Chem. 2013, 20, 3370–3396. [Google Scholar] [CrossRef]
  12. Everett, L.; Galli, A.; Crabb, D. The role of hepatic peroxisome proliferator-activated receptors (PPARs) in health and disease. Liver 2000, 20, 191–199. [Google Scholar] [CrossRef]
  13. Evans, R.M.; Barish, G.D.; Wang, Y.X. PPARs and the complex journey to obesity. Nat. Med. 2004, 10, 355–361. [Google Scholar] [CrossRef]
  14. Tugwood, J.D.; Issemann, I.; Anderson, R.G.; Bundell, K.R.; McPheat, W.L.; Green, S. The mouse peroxisome proliferator activated receptor recognizes a response element in the 5’ flanking sequence of the rat acyl CoA oxidase gene. Embo. J. 1992, 11, 433–439. [Google Scholar]
  15. Reilly, S.M.; Lee, C.H. PPAR delta as a therapeutic target in metabolic disease. FEBS Lett. 2008, 582, 26–31. [Google Scholar] [CrossRef] [Green Version]
  16. Coll, T.; Rodriguez-Calvo, R.; Barroso, E.; Serrano, L.; Eyre, E.; Palomer, X.; Vazquez-Carrera, M. Peroxisome proliferator-activated receptor (PPAR) beta/delta: A new potential therapeutic target for the treatment of metabolic syndrome. Curr. Mol. Pharmacol. 2009, 2, 46–55. [Google Scholar] [CrossRef]
  17. Benetti, E.; Patel, N.S.; Collino, M. The role of PPARbeta/delta in the management of metabolic syndrome and its associated cardiovascular complications. Endocr. Metab. Immune Disord. Drug Targets 2011, 11, 273–284. [Google Scholar] [CrossRef]
  18. Schmid, H.H.; Schmid, P.C.; Natarajan, V. N-acylated glycerophospholipids and their derivatives. Prog. Lipid Res. 1990, 29, 1–43. [Google Scholar]
  19. Hansen, H.S.; Moesgaard, B.; Hansen, H.H.; Petersen, G. N-Acylethanolamines and precursor phospholipids—Relation to cell injury. Chem. Phys. Lipids 2000, 108, 135–150. [Google Scholar]
  20. Bachur, N.R.; Masek, K.; Melmon, K.L.; Udenfriend, S. Fatty Acid Amides of Ethanolamine in Mammalian Tissues. J. Biol. Chem. 1965, 240, 1019–1024. [Google Scholar]
  21. Hansen, H.S. Palmitoylethanolamide and other anandamide congeners. Proposed role in the diseased brain. Exp. Neurol. 2010, 224, 48–55. [Google Scholar] [CrossRef]
  22. Rahman, I.A.; Tsuboi, K.; Uyama, T.; Ueda, N. New players in the fatty acyl ethanolamide metabolism. Pharmacol. Res. 2014, 86, 1–10. [Google Scholar] [CrossRef]
  23. Devane, W.A.; Hanus, L.; Breuer, A.; Pertwee, R.G.; Stevenson, L.A.; Griffin, G.; Gibson, D.; Mandelbaum, A.; Etinger, A.; Mechoulam, R. Isolation and structure of a brain constituent that binds to the cannabinoid receptor. Science 1992, 258, 1946–1949. [Google Scholar]
  24. Sagheddu, C.; Torres, L.H.; Marcourakis, T.; Pistis, M. Endocannabinoid-Like Lipid Neuromodulators in the Regulation of Dopamine Signaling: Relevance for Drug Addiction. Front. Synaptic Neurosci. 2020, 12, 588660. [Google Scholar] [CrossRef]
  25. Fruchart, J.C.; Staels, B.; Duriez, P. PPARS, metabolic disease and atherosclerosis. Pharmacol. Res. 2001, 44, 345–352. [Google Scholar] [CrossRef]
  26. Lehrke, M.; Lazar, M.A. The many faces of PPARgamma. Cell 2005, 123, 993–999. [Google Scholar] [CrossRef] [Green Version]
  27. de la Rosa Rodriguez, M.A.; Kersten, S. Regulation of lipid droplet-associated proteins by peroxisome proliferator-activated receptors. Biochim. Biophys. Acta Mol. Cell Biol. Lipids 2017, 1862, 1212–1220. [Google Scholar] [CrossRef]
  28. Wagner, N.; Wagner, K.D. PPAR Beta/Delta and the Hallmarks of Cancer. Cells 2020, 9, 1133. [Google Scholar] [CrossRef]
  29. Mrowka, P.; Glodkowska-Mrowka, E. PPARgamma Agonists in Combination Cancer Therapies. Curr. Cancer Drug Targets 2020, 20, 197–215. [Google Scholar] [CrossRef]
  30. Bougarne, N.; Weyers, B.; Desmet, S.J.; Deckers, J.; Ray, D.W.; Staels, B.; De Bosscher, K. Molecular Actions of PPARalpha in Lipid Metabolism and Inflammation. Endocr. Rev. 2018, 39, 760–802. [Google Scholar] [CrossRef] [Green Version]
  31. Wagner, N.; Wagner, K.D. The Role of PPARs in Disease. Cells 2020, 9, 2367. [Google Scholar] [CrossRef]
  32. Esmaeili, M.A.; Yadav, S.; Gupta, R.K.; Waggoner, G.R.; Deloach, A.; Calingasan, N.Y.; Beal, M.F.; Kiaei, M. Preferential PPAR-alpha activation reduces neuroinflammation, and blocks neurodegeneration in vivo. Hum. Mol. Genet. 2016, 25, 317–327. [Google Scholar] [CrossRef] [Green Version]
  33. Ericsson, H.; Hamren, B.; Bergstrand, S.; Elebring, M.; Fryklund, L.; Heijer, M.; Ohman, K.P. Pharmacokinetics and metabolism of tesaglitazar, a novel dual-acting peroxisome proliferator-activated receptor alpha/gamma agonist, after a single oral and intravenous dose in humans. Drug Metab. Dispos. 2004, 32, 923–929. [Google Scholar]
  34. Mirza, A.Z.; Althagafi, I.I.; Shamshad, H. Role of PPAR receptor in different diseases and their ligands: Physiological importance and clinical implications. Eur. J. Med. Chem. 2019, 166, 502–513. [Google Scholar] [CrossRef]
  35. Lagana, A.S.; Vitale, S.G.; Nigro, A.; Sofo, V.; Salmeri, F.M.; Rossetti, P.; Rapisarda, A.M.; La Vignera, S.; Condorelli, R.A.; Rizzo, G.; et al. Pleiotropic Actions of Peroxisome Proliferator-Activated Receptors (PPARs) in Dysregulated Metabolic Homeostasis, Inflammation and Cancer: Current Evidence and Future Perspectives. Int. J. Mol. Sci. 2016, 17, 999. [Google Scholar] [CrossRef] [Green Version]
  36. Strosznajder, A.K.; Wojtowicz, S.; Jezyna, M.J.; Sun, G.Y.; Strosznajder, J.B. Recent Insights on the Role of PPAR-beta/delta in Neuroinflammation and Neurodegeneration, and Its Potential Target for Therapy. Neuromolecular. Med. 2021, 23, 86–98. [Google Scholar] [CrossRef]
  37. Christofides, A.; Konstantinidou, E.; Jani, C.; Boussiotis, V.A. The role of peroxisome proliferator-activated receptors (PPAR) in immune responses. Metabolism 2020, 154338. [Google Scholar] [CrossRef]
  38. Warden, A.; Truitt, J.; Merriman, M.; Ponomareva, O.; Jameson, K.; Ferguson, L.B.; Mayfield, R.D.; Harris, R.A. Localization of PPAR isotypes in the adult mouse and human brain. Sci. Rep. 2016, 6, 27618. [Google Scholar] [CrossRef]
  39. Chandra, A.; Sharma, A.; Calingasan, N.Y.; White, J.M.; Shurubor, Y.; Yang, X.W.; Beal, M.F.; Johri, A. Enhanced mitochondrial biogenesis ameliorates disease phenotype in a full-length mouse model of Huntington’s disease. Hum. Mol. Genet. 2016, 25, 2269–2282. [Google Scholar] [CrossRef] [Green Version]
  40. Dickey, A.S.; Pineda, V.V.; Tsunemi, T.; Liu, P.P.; Miranda, H.C.; Gilmore-Hall, S.K.; Lomas, N.; Sampat, K.R.; Buttgereit, A.; Torres, M.J.; et al. PPAR-delta is repressed in Huntington’s disease, is required for normal neuronal function and can be targeted therapeutically. Nat. Med. 2016, 22, 37–45. [Google Scholar] [CrossRef]
  41. Aleshin, S.; Strokin, M.; Sergeeva, M.; Reiser, G. Peroxisome proliferator-activated receptor (PPAR)beta/delta, a possible nexus of PPARalpha- and PPARgamma-dependent molecular pathways in neurodegenerative diseases: Review and novel hypotheses. Neurochem. Int. 2013, 63, 322–330. [Google Scholar] [CrossRef]
  42. Melis, M.; Carta, S.; Fattore, L.; Tolu, S.; Yasar, S.; Goldberg, S.R.; Fratta, W.; Maskos, U.; Pistis, M. Peroxisome proliferator-activated receptors-alpha modulate dopamine cell activity through nicotinic receptors. Biol. Psychiatry 2010, 68, 256–264. [Google Scholar] [CrossRef] [Green Version]
  43. Tufano, M.; Pinna, G. Is There a Future for PPARs in the Treatment of Neuropsychiatric Disorders? Molecules 2020, 25, 1062. [Google Scholar] [CrossRef] [Green Version]
  44. Pistis, M.; Muntoni, A. Roles of N-acylethanolamines in brain functions and neuropsychiatric diseases. In Endocannabinoids and Lipid Mediators in Brain Functions; Springer, Ed.: Berlin, Germany, 2017; pp. 319–346. [Google Scholar]
  45. Chase, K.A.; Rosen, C.; Gin, H.; Bjorkquist, O.; Feiner, B.; Marvin, R.; Conrin, S.; Sharma, R.P. Metabolic and inflammatory genes in schizophrenia. Psychiatry Res. 2015, 225, 208–211. [Google Scholar] [CrossRef] [Green Version]
  46. Martínez-Gras, I.; Pérez-Nievas, B.G.; García-Bueno, B.; Madrigal, J.L.M.; Andrés-Esteban, E.; Rodríguez-Jiménez, R.; Hoenicka, J.; Palomo, T.; Rubio, G.; Leza, J.C. The anti-inflammatory prostaglandin 15d-PGJ2 and its nuclear receptor PPARgamma are decreased in schizophrenia. Schizophr. Res. 2011, 128, 15–22. [Google Scholar] [CrossRef]
  47. Müller, N. Inflammation in Schizophrenia: Pathogenetic Aspects and Therapeutic Considerations. Schizophr. Bull. 2018, 44, 973–982. [Google Scholar] [CrossRef] [Green Version]
  48. Gottfried, C.; Bambini-Junior, V. Insights into the Relationship of the Immune System with Neurodevelopmental and Psychiatric Disorders. Neuroimmunomodulation 2018, 25, 243–245. [Google Scholar] [CrossRef]
  49. Scheggi, S.; Melis, M.; De Felice, M.; Aroni, S.; Muntoni, A.L.; Pelliccia, T.; Gambarana, C.; De Montis, M.G.; Pistis, M. PPARalpha modulation of mesolimbic dopamine transmission rescues depression-related behaviors. Neuropharmacology 2016, 110, 251–259. [Google Scholar] [CrossRef]
  50. Pfau, M.L.; Menard, C.; Russo, S.J. Inflammatory Mediators in Mood Disorders: Therapeutic Opportunities. Annu. Rev. Pharmacol. Toxicol. 2018, 58, 411–428. [Google Scholar] [CrossRef]
  51. Maekawa, M.; Watanabe, A.; Iwayama, Y.; Kimura, T.; Hamazaki, K.; Balan, S.; Ohba, H.; Hisano, Y.; Nozaki, Y.; Ohnishi, T.; et al. Polyunsaturated fatty acid deficiency during neurodevelopment in mice models the prodromal state of schizophrenia through epigenetic changes in nuclear receptor genes. Transl. Psychiatry 2017, 7, e1229. [Google Scholar] [CrossRef] [Green Version]
  52. Costa, M.; Squassina, A.; Congiu, D.; Chillotti, C.; Niola, P.; Galderisi, S.; Pistis, M.; Del Zompo, M. Investigation of endocannabinoid system genes suggests association between peroxisome proliferator activator receptor-alpha gene (PPARA) and schizophrenia. Eur. Neuropsychopharmacol. J. Eur. Coll. Neuropsychopharmacol. 2013, 23, 749–759. [Google Scholar] [CrossRef]
  53. Luchicchi, A.; Lecca, S.; Melis, M.; De Felice, M.; Cadeddu, F.; Frau, R.; Muntoni, A.L.; Fadda, P.; Devoto, P.; Pistis, M. Maternal Immune Activation Disrupts Dopamine System in the Offspring. Int. J. Neuropsychopharmacol. 2016, 19. [Google Scholar] [CrossRef] [Green Version]
  54. De Felice, M.; Melis, M.; Aroni, S.; Muntoni, A.L.; Fanni, S.; Frau, R.; Devoto, P.; Pistis, M. The PPARalpha agonist fenofibrate attenuates disruption of dopamine function in a maternal immune activation rat model of schizophrenia. CNS Neurosci. Ther. 2019, 25, 549–561. [Google Scholar] [CrossRef] [Green Version]
  55. Ramanan, S.; Kooshki, M.; Zhao, W.; Hsu, F.C.; Robbins, M.E. PPARalpha ligands inhibit radiation-induced microglial inflammatory responses by negatively regulating NF-kappaB and AP-1 pathways. Free Radic. Biol. Med. 2008, 45, 1695–1704. [Google Scholar] [CrossRef] [Green Version]
  56. Hill, M.R.; Clarke, S.; Rodgers, K.; Thornhill, B.; Peters, J.M.; Gonzalez, F.J.; Gimble, J.M. Effect of peroxisome proliferator-activated receptor alpha activators on tumor necrosis factor expression in mice during endotoxemia. Infect. Immun. 1999, 67, 3488–3493. [Google Scholar]
  57. Rolland, B.; Marche, K.; Cottencin, O.; Bordet, R. The PPARalpha Agonist Fenofibrate Reduces Prepulse Inhibition Disruption in a Neurodevelopmental Model of Schizophrenia. Schizophr. Res. Treat. 2012, 2012, 839853. [Google Scholar] [CrossRef] [Green Version]
  58. Nadalin, S.; Giacometti, J.; Buretic-Tomljanovic, A. PPARalpha-L162V polymorphism is not associated with schizophrenia risk in a Croatian population. Prostaglandins Leukot. Essent. Fat. Acids 2014, 91, 221–225. [Google Scholar] [CrossRef]
  59. Nadalin, S.; Buretic-Tomljanovic, A.; Rebic, J.; Plesa, I.; Sendula Jengic, V. An association between the PPARalpha-L162V polymorphism and nicotine dependency among patients with schizophrenia. Compr. Psychiatry 2016, 70, 118–124. [Google Scholar] [CrossRef]
  60. Kariharan, T.; Nanayakkara, G.; Parameshwaran, K.; Bagasrawala, I.; Ahuja, M.; Abdel-Rahman, E.; Amin, A.T.; Dhanasekaran, M.; Suppiramaniam, V.; Amin, R.H. Central activation of PPAR-gamma ameliorates diabetes induced cognitive dysfunction and improves BDNF expression. Neurobiol. Aging 2015, 36, 1451–1461. [Google Scholar] [CrossRef]
  61. Yi, Z.; Fan, X.; Wang, J.; Liu, D.; Freudenreich, O.; Goff, D.; Henderson, D.C. Rosiglitazone and cognitive function in clozapine-treated patients with schizophrenia: A pilot study. Psychiatry Res. 2012, 200, 79–82. [Google Scholar] [CrossRef]
  62. Watson, G.S.; Cholerton, B.A.; Reger, M.A.; Baker, L.D.; Plymate, S.R.; Asthana, S.; Fishel, M.A.; Kulstad, J.J.; Green, P.S.; Cook, D.G.; et al. Preserved cognition in patients with early Alzheimer disease and amnestic mild cognitive impairment during treatment with rosiglitazone: A preliminary study. Am. J. Geriatr. Psychiatry 2005, 13, 950–958. [Google Scholar] [CrossRef]
  63. Smith, R.C.; Jin, H.; Li, C.; Bark, N.; Shekhar, A.; Dwivedi, S.; Mortiere, C.; Lohr, J.; Hu, Q.; Davis, J.M. Effects of pioglitazone on metabolic abnormalities, psychopathology, and cognitive function in schizophrenic patients treated with antipsychotic medication: A randomized double-blind study. Schizophr. Res. 2013, 143, 18–24. [Google Scholar] [CrossRef]
  64. Schoepf, D.; Potluri, R.; Uppal, H.; Natalwala, A.; Narendran, P.; Heun, R. Type-2 diabetes mellitus in schizophrenia: Increased prevalence and major risk factor of excess mortality in a naturalistic 7-year follow-up. Eur. Psychiatry 2012, 27, 33–42. [Google Scholar] [CrossRef]
  65. Iranpour, N.; Zandifar, A.; Farokhnia, M.; Goguol, A.; Yekehtaz, H.; Khodaie-Ardakani, M.R.; Salehi, B.; Esalatmanesh, S.; Zeionoddini, A.; Mohammadinejad, P.; et al. The effects of pioglitazone adjuvant therapy on negative symptoms of patients with chronic schizophrenia: A double-blind and placebo-controlled trial. Hum. Psychopharmacol. 2016, 31, 103–112. [Google Scholar] [CrossRef]
  66. Hodes, G.E.; Kana, V.; Menard, C.; Merad, M.; Russo, S.J. Neuroimmune mechanisms of depression. Nat. Neurosci. 2015, 18, 1386–1393. [Google Scholar] [CrossRef]
  67. Menard, C.; Pfau, M.L.; Hodes, G.E.; Russo, S.J. Immune and Neuroendocrine Mechanisms of Stress Vulnerability and Resilience. Neuropsychopharmacology 2017, 42, 62–80. [Google Scholar] [CrossRef] [Green Version]
  68. Jeon, S.W.; Kim, Y.K. Neuroinflammation and cytokine abnormality in major depression: Cause or consequence in that illness? World J. Psychiatry 2016, 6, 283–293. [Google Scholar] [CrossRef]
  69. Janowsky, D.S.; el-Yousef, M.K.; Davis, J.M.; Sekerke, H.J. A cholinergic-adrenergic hypothesis of mania and depression. Lancet 1972, 2, 632–635. [Google Scholar] [CrossRef]
  70. Dulawa, S.C.; Janowsky, D.S. Cholinergic regulation of mood: From basic and clinical studies to emerging therapeutics. Mol. Psychiatry 2019, 24, 694–709. [Google Scholar] [CrossRef]
  71. Melis, M.; Scheggi, S.; Carta, G.; Madeddu, C.; Lecca, S.; Luchicchi, A.; Cadeddu, F.; Frau, R.; Fattore, L.; Fadda, P.; et al. PPARalpha Regulates Cholinergic-Driven Activity of Midbrain Dopamine Neurons via a Novel Mechanism Involving alpha7 Nicotinic Acetylcholine Receptors. J. Neurosci. 2013, 33, 6203–6211. [Google Scholar] [CrossRef] [Green Version]
  72. Yu, H.L.; Deng, X.Q.; Li, Y.J.; Li, Y.C.; Quan, Z.S.; Sun, X.Y. N-palmitoylethanolamide, an endocannabinoid, exhibits antidepressant effects in the forced swim test and the tail suspension test in mice. Pharmacol. Rep. 2011, 63, 834–839. [Google Scholar]
  73. Jiang, B.; Wang, Y.J.; Wang, H.; Song, L.; Huang, C.; Zhu, Q.; Wu, F.; Zhang, W. Antidepressant-like effects of fenofibrate in mice via the hippocampal brain-derived neurotrophic factor signalling pathway. Br. J. Pharmacol. 2017, 174, 177–194. [Google Scholar] [CrossRef]
  74. Jin, P.; Yu, H.L.; Tian, L.; Zhang, F.; Quan, Z.S. Antidepressant-like effects of oleoylethanolamide in a mouse model of chronic unpredictable mild stress. Pharmacol. Biochem. Behav. 2015, 133, 146–154. [Google Scholar] [CrossRef]
  75. Krishnan, V.; Nestler, E.J. Linking molecules to mood: New insight into the biology of depression. Am. J. Psychiatry 2010, 167, 1305–1320. [Google Scholar] [CrossRef] [Green Version]
  76. Krishnan, V.; Nestler, E.J. Animal models of depression: Molecular perspectives. Curr. Top Behav. Neurosci. 2011, 7, 121–147. [Google Scholar] [CrossRef] [Green Version]
  77. Song, L.; Wang, H.; Wang, Y.J.; Wang, J.L.; Zhu, Q.; Wu, F.; Zhang, W.; Jiang, B. Hippocampal PPARalpha is a novel therapeutic target for depression and mediates the antidepressant actions of fluoxetine in mice. Br. J. Pharmacol. 2018, 175, 2968–2987. [Google Scholar] [CrossRef] [Green Version]
  78. Matrisciano, F.; Pinna, G. PPAR and functional foods: Rationale for natural neurosteroid-based interventions for postpartum depression. Neurobiol. Stress 2020, 12, 100222. [Google Scholar] [CrossRef]
  79. Cheng, H.S.; Tan, W.R.; Low, Z.S.; Marvalim, C.; Lee, J.Y.H.; Tan, N.S. Exploration and Development of PPAR Modulators in Health and Disease: An Update of Clinical Evidence. Int. J. Mol. Sci. 2019, 20, 5055. [Google Scholar] [CrossRef] [Green Version]
  80. Eissa Ahmed, A.A.; Al-Rasheed, N.M. Antidepressant-like effects of rosiglitazone, a PPARgamma agonist, in the rat forced swim and mouse tail suspension tests. Behav. Pharmacol. 2009, 20, 635–642. [Google Scholar] [CrossRef]
  81. Sadaghiani, M.S.; Javadi-Paydar, M.; Gharedaghi, M.H.; Fard, Y.Y.; Dehpour, A.R. Antidepressant-like effect of pioglitazone in the forced swimming test in mice: The role of PPAR-gamma receptor and nitric oxide pathway. Behav. Brain Res. 2011, 224, 336–343. [Google Scholar] [CrossRef]
  82. Rasgon, N.L.; Kenna, H.A.; Williams, K.E.; Powers, B.; Wroolie, T.; Schatzberg, A.F. Rosiglitazone add-on in treatment of depressed patients with insulin resistance: A pilot study. Sci. World J. 2010, 10, 321–328. [Google Scholar] [CrossRef] [Green Version]
  83. Sepanjnia, K.; Modabbernia, A.; Ashrafi, M.; Modabbernia, M.J.; Akhondzadeh, S. Pioglitazone adjunctive therapy for moderate-to-severe major depressive disorder: Randomized double-blind placebo-controlled trial. Neuropsychopharmacol. Off. Publ. Am. Coll. Neuropsychopharmacol. 2012, 37, 2093–2100. [Google Scholar] [CrossRef] [Green Version]
  84. Kemp, D.E.; Schinagle, M.; Gao, K.; Conroy, C.; Ganocy, S.J.; Ismail-Beigi, F.; Calabrese, J.R. PPAR-gamma agonism as a modulator of mood: Proof-of-concept for pioglitazone in bipolar depression. CNS Drugs 2014, 28, 571–581. [Google Scholar] [CrossRef] [Green Version]
  85. Moreno, S.; Farioli-Vecchioli, S.; Ceru, M.P. Immunolocalization of peroxisome proliferator-activated receptors and retinoid X receptors in the adult rat CNS. Neuroscience 2004, 123, 131–145. [Google Scholar]
  86. Sagheddu, C.; Muntoni, A.L.; Pistis, M.; Melis, M. Endocannabinoid Signaling in Motivation, Reward, and Addiction: Influences on Mesocorticolimbic Dopamine Function. Int. Rev. Neurobiol. 2015, 125, 257–302. [Google Scholar] [CrossRef]
  87. Sagheddu, C.; Melis, M. Individual differences and vulnerability to drug addiction: A focus on the endocannabinoid system. CNS Neurol. Disord. Drug Targets 2015, 14, 502–517. [Google Scholar] [CrossRef]
  88. Ray, L.A.; Roche, D.J.; Heinzerling, K.; Shoptaw, S. Opportunities for the development of neuroimmune therapies in addiction. Int. Rev. Neurobiol. 2014, 118, 381–401. [Google Scholar] [CrossRef] [Green Version]
  89. Le Foll, B.; Di Ciano, P.; Panlilio, L.V.; Goldberg, S.R.; Ciccocioppo, R. Peroxisome proliferator-activated receptor (PPAR) agonists as promising new medications for drug addiction: Preclinical evidence. Curr. Drug Targets 2013, 14, 768–776. [Google Scholar] [CrossRef] [Green Version]
  90. Koob, G.F.; Le Moal, M. Drug addiction, dysregulation of reward, and allostasis. Neuropsychopharmacol. Off. Publ. Am. Coll. Neuropsychopharmacol. 2001, 24, 97–129. [Google Scholar] [CrossRef]
  91. Koob, G.F.; Le Moal, M. Drug abuse: Hedonic homeostatic dysregulation. Science 1997, 278, 52–58. [Google Scholar] [CrossRef]
  92. Meredith, L.R.; Burnette, E.M.; Grodin, E.N.; Irwin, M.R.; Ray, L.A. Immune Treatments for Alcohol Use Disorder: A Translational Framework. Brain Behav. Immun. 2021. [Google Scholar] [CrossRef]
  93. Erickson, E.K.; Grantham, E.K.; Warden, A.S.; Harris, R.A. Neuroimmune signaling in alcohol use disorder. Pharmacol. Biochem. Behav. 2019, 177, 34–60. [Google Scholar] [CrossRef]
  94. Domi, E.; Domi, A.; Ubaldi, M.; Somaini, L.; Demopulos, G.; Gaitanaris, G.; Ciccocioppo, R. Further evidence for the involvement of the PPARgamma system on alcohol intake and sensitivity in rodents. Psychopharmacology 2020, 237, 2983–2992. [Google Scholar] [CrossRef]
  95. Fotio, Y.; Borruto, A.M.; Benvenuti, F.; Demopulos, G.; Gaitanaris, G.; Roberto, M.; Ciccocioppo, R. Activation of peroxisome proliferator-activated receptor gamma reduces alcohol drinking and seeking by modulating multiple mesocorticolimbic regions in rats. Neuropsychopharmacology 2021, 46, 360–367. [Google Scholar] [CrossRef]
  96. Cippitelli, A.; Domi, E.; Ubaldi, M.; Douglas, J.C.; Li, H.W.; Demopulos, G.; Gaitanaris, G.; Roberto, M.; Drew, P.D.; Kane, C.J.M.; et al. Protection against alcohol-induced neuronal and cognitive damage by the PPARgamma receptor agonist pioglitazone. Brain Behav. Immun. 2017, 64, 320–329. [Google Scholar] [CrossRef]
  97. Stopponi, S.; de Guglielmo, G.; Somaini, L.; Cippitelli, A.; Cannella, N.; Kallupi, M.; Ubaldi, M.; Heilig, M.; Demopulos, G.; Gaitanaris, G.; et al. Activation of PPARgamma by pioglitazone potentiates the effects of naltrexone on alcohol drinking and relapse in msP rats. Alcohol. Clin. Exp. Res. 2013, 37, 1351–1360. [Google Scholar] [CrossRef]
  98. Mascia, P.; Pistis, M.; Justinova, Z.; Panlilio, L.V.; Luchicchi, A.; Lecca, S.; Scherma, M.; Fratta, W.; Fadda, P.; Barnes, C.; et al. Blockade of nicotine reward and reinstatement by activation of alpha-type peroxisome proliferator-activated receptors. Biol. Psychiatry 2011, 69, 633–641. [Google Scholar] [CrossRef] [Green Version]
  99. Jackson, A.; Bagdas, D.; Muldoon, P.P.; Lichtman, A.H.; Carroll, F.I.; Greenwald, M.; Miles, M.F.; Damaj, M.I. In vivo interactions between alpha7 nicotinic acetylcholine receptor and nuclear peroxisome proliferator-activated receptor-alpha: Implication for nicotine dependence. Neuropharmacology 2017, 118, 38–45. [Google Scholar] [CrossRef] [Green Version]
  100. Melis, M.; Pistis, M. Targeting the interaction between fatty acid ethanolamides and nicotinic receptors: Therapeutic perspectives. Pharmacol. Res. 2014, 86, 42–49. [Google Scholar] [CrossRef]
  101. Sagheddu, C.; Scherma, M.; Congiu, M.; Fadda, P.; Carta, G.; Banni, S.; Wood, J.T.; Makriyannis, A.; Malamas, M.S.; Pistis, M. Inhibition of N-acylethanolamine acid amidase reduces nicotine-induced dopamine activation and reward. Neuropharmacology 2019, 144, 327–336. [Google Scholar] [CrossRef]
  102. Domi, E.; Caputi, F.F.; Romualdi, P.; Domi, A.; Scuppa, G.; Candeletti, S.; Atkins, A.; Heilig, M.; Demopulos, G.; Gaitanaris, G.; et al. Activation of PPARgamma Attenuates the Expression of Physical and Affective Nicotine Withdrawal Symptoms through Mechanisms Involving Amygdala and Hippocampus Neurotransmission. J. Neurosci. Off. J. Soc. Neurosci. 2019, 39, 9864–9875. [Google Scholar] [CrossRef]
  103. Scherma, M.; Muntoni, A.L.; Melis, M.; Fattore, L.; Fadda, P.; Fratta, W.; Pistis, M. Interactions between the endocannabinoid and nicotinic cholinergic systems: Preclinical evidence and therapeutic perspectives. Psychopharmacology 2016, 233, 1765–1777. [Google Scholar] [CrossRef]
  104. Melis, M.; Pillolla, G.; Luchicchi, A.; Muntoni, A.L.; Yasar, S.; Goldberg, S.R.; Pistis, M. Endogenous fatty acid ethanolamides suppress nicotine-induced activation of mesolimbic dopamine neurons through nuclear receptors. J. Neurosci. Off. J. Soc. Neurosci. 2008, 28, 13985–13994. [Google Scholar] [CrossRef] [Green Version]
  105. Panlilio, L.V.; Justinova, Z.; Mascia, P.; Pistis, M.; Luchicchi, A.; Lecca, S.; Barnes, C.; Redhi, G.H.; Adair, J.; Heishman, S.J.; et al. Novel use of a lipid-lowering fibrate medication to prevent nicotine reward and relapse: Preclinical findings. Neuropsychopharmacology 2012, 37, 1838–1847. [Google Scholar] [CrossRef] [Green Version]
  106. Jones, J.D.; Comer, S.D.; Metz, V.E.; Manubay, J.M.; Mogali, S.; Ciccocioppo, R.; Martinez, S.; Mumtaz, M.; Bisaga, A. Pioglitazone, a PPARgamma agonist, reduces nicotine craving in humans, with marginal effects on abuse potential. Pharmacol. Biochem. Behav. 2017, 163, 90–100. [Google Scholar] [CrossRef]
  107. de Guglielmo, G.; Melis, M.; De Luca, M.A.; Kallupi, M.; Li, H.W.; Niswender, K.; Giordano, A.; Senzacqua, M.; Somaini, L.; Cippitelli, A.; et al. PPARgamma activation attenuates opioid consumption and modulates mesolimbic dopamine transmission. Neuropsychopharmacol. Off. Publ. Am. Coll. Neuropsychopharmacol. 2015, 40, 927–937. [Google Scholar] [CrossRef] [Green Version]
  108. Jones, J.D.; Bisaga, A.; Metz, V.E.; Manubay, J.M.; Mogali, S.; Ciccocioppo, R.; Madera, G.; Doernberg, M.; Comer, S.D. The PPARgamma Agonist Pioglitazone Fails to Alter the Abuse Potential of Heroin, But Does Reduce Heroin Craving and Anxiety. J. Psychoact. Drugs 2018, 50, 390–401. [Google Scholar] [CrossRef]
  109. Jones, J.D.; Sullivan, M.A.; Manubay, J.M.; Mogali, S.; Metz, V.E.; Ciccocioppo, R.; Comer, S.D. The effects of pioglitazone, a PPARgamma receptor agonist, on the abuse liability of oxycodone among nondependent opioid users. Physiol. Behav. 2016, 159, 33–39. [Google Scholar] [CrossRef] [Green Version]
  110. Schmitz, J.M.; Green, C.E.; Hasan, K.M.; Vincent, J.; Suchting, R.; Weaver, M.F.; Moeller, F.G.; Narayana, P.A.; Cunningham, K.A.; Dineley, K.T.; et al. PPAR-gamma agonist pioglitazone modifies craving intensity and brain white matter integrity in patients with primary cocaine use disorder: A double-blind randomized controlled pilot trial. Addiction 2017, 112, 1861–1868. [Google Scholar] [CrossRef]
  111. Miller, W.R.; Fox, R.G.; Stutz, S.J.; Lane, S.D.; Denner, L.; Cunningham, K.A.; Dineley, K.T. PPARgamma agonism attenuates cocaine cue reactivity. Addict. Biol. 2018, 23, 55–68. [Google Scholar] [CrossRef]
  112. Quiroga, C.; Barberena, J.J.; Alcaraz-Silva, J.; Machado, S.; Imperatori, C.; Yadollahpour, A.; Budde, H.; Yamamoto, T.; Telles-Correia, D.; Murillo-Rodriguez, E. The Role of Peroxisome Proliferator-Activated Receptor in Addiction: A Novel Drug Target. Curr. Top Med. Chem. 2021. [Google Scholar] [CrossRef]
  113. Lujan, M.A.; Cheer, J.F.; Melis, M. Choosing the right drug: Status and future of endocannabinoid research for the prevention of drug-seeking reinstatement. Curr. Opin. Pharmacol. 2021, 56, 29–38. [Google Scholar] [CrossRef]
  114. Schwandt, M.L.; Diazgranados, N.; Umhau, J.C.; Kwako, L.E.; George, D.T.; Heilig, M. PPARgamma activation by pioglitazone does not suppress cravings for alcohol, and is associated with a risk of myopathy in treatment seeking alcohol dependent patients: A randomized controlled proof of principle study. Psychopharmacology 2020, 237, 2367–2380. [Google Scholar] [CrossRef]
  115. Matheson, J.; Le Foll, B. Therapeutic Potential of Peroxisome Proliferator-Activated Receptor (PPAR) Agonists in Substance Use Disorders: A Synthesis of Preclinical and Human Evidence. Cells 2020, 9, 1196. [Google Scholar] [CrossRef]
  116. Barbiero, J.K.; Santiago, R.M.; Persike, D.S.; da Silva Fernandes, M.J.; Tonin, F.S.; da Cunha, C.; Lucio Boschen, S.; Lima, M.M.; Vital, M.A. Neuroprotective effects of peroxisome proliferator-activated receptor alpha and gamma agonists in model of parkinsonism induced by intranigral 1-methyl-4-phenyl-1,2,3,6-tetrahyropyridine. Behav. Brain Res. 2014, 274, 390–399. [Google Scholar] [CrossRef]
  117. Barbiero, J.K.; Santiago, R.; Tonin, F.S.; Boschen, S.; da Silva, L.M.; Werner, M.F.; da Cunha, C.; Lima, M.M.; Vital, M.A. PPAR-alpha agonist fenofibrate protects against the damaging effects of MPTP in a rat model of Parkinson’s disease. Prog. Neuropsychopharmacol. Biol. Psychiatry 2014, 53, 35–44. [Google Scholar] [CrossRef]
  118. Porta, N.; Vallee, L.; Lecointe, C.; Bouchaert, E.; Staels, B.; Bordet, R.; Auvin, S. Fenofibrate, a peroxisome proliferator-activated receptor-alpha agonist, exerts anticonvulsive properties. Epilepsia 2009, 50, 943–948. [Google Scholar] [CrossRef]
  119. Puligheddu, M.; Pillolla, G.; Melis, M.; Lecca, S.; Marrosu, F.; De Montis, M.G.; Scheggi, S.; Carta, G.; Murru, E.; Aroni, S.; et al. PPAR-alpha agonists as novel antiepileptic drugs: Preclinical findings. PLoS ONE 2013, 8, e64541. [Google Scholar] [CrossRef]
  120. Puligheddu, M.; Melis, M.; Pillolla, G.; Milioli, G.; Parrino, L.; Terzano, G.M.; Aroni, S.; Sagheddu, C.; Marrosu, F.; Pistis, M.; et al. Rationale for an adjunctive therapy with fenofibrate in pharmacoresistant nocturnal frontal lobe epilepsy. Epilepsia 2017, 58, 1762–1770. [Google Scholar] [CrossRef] [Green Version]
  121. Saha, L.; Bhandari, S.; Bhatia, A.; Banerjee, D.; Chakrabarti, A. Anti-kindling Effect of Bezafibrate, a Peroxisome Proliferator-activated Receptors Alpha Agonist, in Pentylenetetrazole Induced Kindling Seizure Model. J. Epilepsy Res. 2014, 4, 45–54. [Google Scholar]
  122. Chandra, S.; Roy, A.; Jana, M.; Pahan, K. Cinnamic acid activates PPARalpha to stimulate Lysosomal biogenesis and lower Amyloid plaque pathology in an Alzheimer’s disease mouse model. Neurobiol. Dis. 2019, 124, 379–395. [Google Scholar] [CrossRef]
  123. Pierrot, N.; Ris, L.; Stancu, I.C.; Doshina, A.; Ribeiro, F.; Tyteca, D.; Bauge, E.; Lalloyer, F.; Malong, L.; Schakman, O.; et al. Sex-regulated gene dosage effect of PPARalpha on synaptic plasticity. Life Sci. Alliance 2019, 2. [Google Scholar] [CrossRef]
  124. Luo, R.; Su, L.Y.; Li, G.; Yang, J.; Liu, Q.; Yang, L.X.; Zhang, D.F.; Zhou, H.; Xu, M.; Fan, Y.; et al. Activation of PPARA-mediated autophagy reduces Alzheimer disease-like pathology and cognitive decline in a murine model. Autophagy 2020, 16, 52–69. [Google Scholar] [CrossRef]
  125. Papadopoulos, P.; Rosa-Neto, P.; Rochford, J.; Hamel, E. Pioglitazone improves reversal learning and exerts mixed cerebrovascular effects in a mouse model of Alzheimer’s disease with combined amyloid-beta and cerebrovascular pathology. PLoS ONE 2013, 8, e68612. [Google Scholar] [CrossRef]
  126. Risner, M.E.; Saunders, A.M.; Altman, J.F.; Ormandy, G.C.; Craft, S.; Foley, I.M.; Zvartau-Hind, M.E.; Hosford, D.A.; Roses, A.D. Efficacy of rosiglitazone in a genetically defined population with mild-to-moderate Alzheimer’s disease. Pharmacogenomics J. 2006, 6, 246–254. [Google Scholar] [CrossRef] [Green Version]
  127. Quinn, L.P.; Crook, B.; Hows, M.E.; Vidgeon-Hart, M.; Chapman, H.; Upton, N.; Medhurst, A.D.; Virley, D.J. The PPARgamma agonist pioglitazone is effective in the MPTP mouse model of Parkinson’s disease through inhibition of monoamine oxidase B. Br. J. Pharmacol. 2008, 154, 226–233. [Google Scholar] [CrossRef] [Green Version]
  128. Geldmacher, D.S.; Fritsch, T.; McClendon, M.J.; Landreth, G. A randomized pilot clinical trial of the safety of pioglitazone in treatment of patients with Alzheimer disease. Arch. Neurol. 2011, 68, 45–50. [Google Scholar] [CrossRef]
  129. Galimberti, D.; Scarpini, E. Pioglitazone for the treatment of Alzheimer’s disease. Expert Opin. Investig. Drugs 2017, 26, 97–101. [Google Scholar] [CrossRef]
  130. Dehmer, T.; Heneka, M.T.; Sastre, M.; Dichgans, J.; Schulz, J.B. Protection by pioglitazone in the MPTP model of Parkinson’s disease correlates with I kappa B alpha induction and block of NF kappa B and iNOS activation. J. Neurochem. 2004, 88, 494–501. [Google Scholar] [CrossRef]
  131. Niino, M.; Iwabuchi, K.; Kikuchi, S.; Ato, M.; Morohashi, T.; Ogata, A.; Tashiro, K.; Onoe, K. Amelioration of experimental autoimmune encephalomyelitis in C57BL/6 mice by an agonist of peroxisome proliferator-activated receptor-gamma. J. Neuroimmunol. 2001, 116, 40–48. [Google Scholar] [CrossRef]
  132. Pistis, M.; Melis, M. From surface to nuclear receptors: The endocannabinoid family extends its assets. Curr. Med. Chem. 2010, 17, 1450–1467. [Google Scholar]
  133. Fidaleo, M.; Fanelli, F.; Ceru, M.P.; Moreno, S. Neuroprotective properties of peroxisome proliferator-activated receptor alpha (PPARalpha) and its lipid ligands. Curr. Med. Chem. 2014, 21, 2803–2821. [Google Scholar]
  134. Heneka, M.T.; Reyes-Irisarri, E.; Hull, M.; Kummer, M.P. Impact and Therapeutic Potential of PPARs in Alzheimer’s Disease. Curr. Neuropharmacol. 2011, 9, 643–650. [Google Scholar] [CrossRef]
  135. Wojtowicz, S.; Strosznajder, A.K.; Jezyna, M.; Strosznajder, J.B. The Novel Role of PPAR Alpha in the Brain: Promising Target in Therapy of Alzheimer’s Disease and Other Neurodegenerative Disorders. Neurochem. Res. 2020, 45, 972–988. [Google Scholar] [CrossRef] [Green Version]
  136. Saez-Orellana, F.; Octave, J.N.; Pierrot, N. Alzheimer’s Disease, a Lipid Story: Involvement of Peroxisome Proliferator-Activated Receptor alpha. Cells 2020, 9, 1215. [Google Scholar] [CrossRef]
  137. Heneka, M.T.; Sastre, M.; Dumitrescu-Ozimek, L.; Hanke, A.; Dewachter, I.; Kuiperi, C.; O’Banion, K.; Klockgether, T.; Van Leuven, F.; Landreth, G.E. Acute treatment with the PPARgamma agonist pioglitazone and ibuprofen reduces glial inflammation and Abeta1-42 levels in APPV717I transgenic mice. Brain 2005, 128, 1442–1453. [Google Scholar] [CrossRef] [Green Version]
  138. Landreth, G.; Jiang, Q.; Mandrekar, S.; Heneka, M. PPARgamma agonists as therapeutics for the treatment of Alzheimer’s disease. Neurotherapeutics 2008, 5, 481–489. [Google Scholar] [CrossRef] [Green Version]
  139. Jiang, Q.; Heneka, M.; Landreth, G.E. The role of peroxisome proliferator-activated receptor-gamma (PPARgamma) in Alzheimer’s disease: Therapeutic implications. CNS Drugs 2008, 22, 1–14. [Google Scholar] [CrossRef]
  140. D’Orio, B.; Fracassi, A.; Ceru, M.P.; Moreno, S. Targeting PPARalpha in Alzheimer’s Disease. Curr. Alzheimer Res. 2018, 15, 345–354. [Google Scholar] [CrossRef]
  141. Scuderi, C.; Bronzuoli, M.R.; Facchinetti, R.; Pace, L.; Ferraro, L.; Broad, K.D.; Serviddio, G.; Bellanti, F.; Palombelli, G.; Carpinelli, G.; et al. Ultramicronized palmitoylethanolamide rescues learning and memory impairments in a triple transgenic mouse model of Alzheimer’s disease by exerting anti-inflammatory and neuroprotective effects. Transl. Psychiatry 2018, 8, 32. [Google Scholar] [CrossRef]
  142. Cipriano, M.; Esposito, G.; Negro, L.; Capoccia, E.; Sarnelli, G.; Scuderi, C.; De Filippis, D.; Steardo, L.; Iuvone, T. Palmitoylethanolamide Regulates Production of Pro-Angiogenic Mediators in a Model of beta Amyloid-Induced Astrogliosis In Vitro. CNS Neurol. Disord. Drug Targets 2015, 14, 828–837. [Google Scholar] [CrossRef]
  143. Scuderi, C.; Stecca, C.; Valenza, M.; Ratano, P.; Bronzuoli, M.R.; Bartoli, S.; Steardo, L.; Pompili, E.; Fumagalli, L.; Campolongo, P. Palmitoylethanolamide controls reactive gliosis and exerts neuroprotective functions in a rat model of Alzheimer’s disease. Cell Death Dis. 2014, 5, e1419. [Google Scholar] [CrossRef] [Green Version]
  144. Scuderi, C.; Steardo, L. Neuroglial roots of neurodegenerative diseases: Therapeutic potential of palmitoylethanolamide in models of Alzheimer’s disease. CNS Neurol. Disord. Drug Targets 2013, 12, 62–69. [Google Scholar]
  145. Scuderi, C.; Esposito, G.; Blasio, A.; Valenza, M.; Arietti, P.; Steardo, L., Jr.; Carnuccio, R.; De Filippis, D.; Petrosino, S.; Iuvone, T.; et al. Palmitoylethanolamide counteracts reactive astrogliosis induced by beta-amyloid peptide. J. Cell Mol. Med. 2011, 15, 2664–2674. [Google Scholar] [CrossRef] [Green Version]
  146. D’Agostino, G.; Russo, R.; Avagliano, C.; Cristiano, C.; Meli, R.; Calignano, A. Palmitoylethanolamide protects against the amyloid-beta25-35-induced learning and memory impairment in mice, an experimental model of Alzheimer disease. Neuropsychopharmacol. Off. Publ. Am. Coll. Neuropsychopharmacol. 2012, 37, 1784–1792. [Google Scholar] [CrossRef] [Green Version]
  147. Chamberlain, S.; Gabriel, H.; Strittmatter, W.; Didsbury, J. An Exploratory Phase IIa Study of the PPAR delta/gamma Agonist T3D-959 Assessing Metabolic and Cognitive Function in Subjects with Mild to Moderate Alzheimer’s Disease. J. Alzheimers Dis. 2020, 73, 1085–1103. [Google Scholar] [CrossRef] [Green Version]
  148. Wang, J.; Guo, M.N.; Liu, Z.Z.; Ma, S.F.; Liu, W.J.; Qian, J.J.; Zhang, W.N. PGC-1alpha reduces Amyloid-beta deposition in Alzheimer’s disease: Effect of increased VDR expression. Neurosci. Lett. 2021, 744, 135598. [Google Scholar] [CrossRef]
  149. Qin, W.; Haroutunian, V.; Katsel, P.; Cardozo, C.P.; Ho, L.; Buxbaum, J.D.; Pasinetti, G.M. PGC-1alpha expression decreases in the Alzheimer disease brain as a function of dementia. Arch. Neurol. 2009, 66, 352–361. [Google Scholar] [CrossRef]
  150. Lagouge, M.; Argmann, C.; Gerhart-Hines, Z.; Meziane, H.; Lerin, C.; Daussin, F.; Messadeq, N.; Milne, J.; Lambert, P.; Elliott, P.; et al. Resveratrol improves mitochondrial function and protects against metabolic disease by activating SIRT1 and PGC-1alpha. Cell 2006, 127, 1109–1122. [Google Scholar] [CrossRef]
  151. Marambaud, P.; Zhao, H.; Davies, P. Resveratrol promotes clearance of Alzheimer’s disease amyloid-beta peptides. J. Biol. Chem. 2005, 280, 37377–37382. [Google Scholar] [CrossRef] [Green Version]
  152. Mota, B.C.; Sastre, M. The Role of PGC1alpha in Alzheimer’s Disease and Therapeutic Interventions. Int. J. Mol. Sci. 2021, 22, 5769. [Google Scholar] [CrossRef]
  153. Szalardy, L.; Zadori, D.; Tanczos, E.; Simu, M.; Bencsik, K.; Vecsei, L.; Klivenyi, P. Elevated levels of PPAR-gamma in the cerebrospinal fluid of patients with multiple sclerosis. Neurosci. Lett. 2013, 554, 131–134. [Google Scholar] [CrossRef] [Green Version]
  154. Szalardy, L.; Zadori, D.; Bencsik, K.; Vecsei, L.; Klivenyi, P. Unlike PPARgamma, neither other PPARs nor PGC-1alpha is elevated in the cerebrospinal fluid of patients with multiple sclerosis. Neurosci. Lett. 2017, 651, 128–133. [Google Scholar] [CrossRef]
  155. Ferret-Sena, V.; Capela, C.; Sena, A. Metabolic Dysfunction and Peroxisome Proliferator-Activated Receptors (PPAR) in Multiple Sclerosis. Int. J. Mol. Sci. 2018, 19, 1639. [Google Scholar] [CrossRef] [Green Version]
  156. Di Filippo, M.; Pini, L.A.; Pelliccioli, G.P.; Calabresi, P.; Sarchielli, P. Abnormalities in the cerebrospinal fluid levels of endocannabinoids in multiple sclerosis. J. Neurol. Neurosurg. Psychiatry 2008, 79, 1224–1229. [Google Scholar] [CrossRef]
  157. Baker, D.; Pryce, G.; Croxford, J.L.; Brown, P.; Pertwee, R.G.; Makriyannis, A.; Khanolkar, A.; Layward, L.; Fezza, F.; Bisogno, T.; et al. Endocannabinoids control spasticity in a multiple sclerosis model. Faseb. J. 2001, 15, 300–302. [Google Scholar] [CrossRef] [Green Version]
  158. Loria, F.; Petrosino, S.; Mestre, L.; Spagnolo, A.; Correa, F.; Hernangomez, M.; Guaza, C.; Di Marzo, V.; Docagne, F. Study of the regulation of the endocannabinoid system in a virus model of multiple sclerosis reveals a therapeutic effect of palmitoylethanolamide. Eur. J. Neurosci. 2008, 28, 633–641. [Google Scholar] [CrossRef] [Green Version]
  159. Rahimi, A.; Faizi, M.; Talebi, F.; Noorbakhsh, F.; Kahrizi, F.; Naderi, N. Interaction between the protective effects of cannabidiol and palmitoylethanolamide in experimental model of multiple sclerosis in C57BL/6 mice. Neuroscience 2015, 290, 279–287. [Google Scholar] [CrossRef]
  160. Pringsheim, T.; Jette, N.; Frolkis, A.; Steeves, T.D. The prevalence of Parkinson’s disease: A systematic review and meta-analysis. Mov. Disord. 2014, 29, 1583–1590. [Google Scholar] [CrossRef]
  161. Galan-Rodriguez, B.; Suarez, J.; Gonzalez-Aparicio, R.; Bermudez-Silva, F.J.; Maldonado, R.; Robledo, P.; Rodriguez de Fonseca, F.; Fernandez-Espejo, E. Oleoylethanolamide exerts partial and dose-dependent neuroprotection of substantia nigra dopamine neurons. Neuropharmacology 2009, 56, 653–664. [Google Scholar]
  162. Gonzalez-Aparicio, R.; Moratalla, R. Oleoylethanolamide reduces L-DOPA-induced dyskinesia via TRPV1 receptor in a mouse model of Parkinson s disease. Neurobiol. Dis. 2014, 62, 416–425. [Google Scholar] [CrossRef] [Green Version]
  163. Esposito, E.; Impellizzeri, D.; Mazzon, E.; Paterniti, I.; Cuzzocrea, S. Neuroprotective activities of palmitoylethanolamide in an animal model of Parkinson’s disease. PLoS ONE 2012, 7, e41880. [Google Scholar] [CrossRef] [Green Version]
  164. Randy, L.H.; Guoying, B. Agonism of Peroxisome Proliferator Receptor-Gamma may have Therapeutic Potential for Neuroinflammation and Parkinson’s Disease. Curr. Neuropharmacol. 2007, 5, 35–46. [Google Scholar]
  165. Carta, A.R. PPAR-gamma: Therapeutic prospects in Parkinson’s disease. Curr. Drug Targets 2013, 14, 743–751. [Google Scholar]
  166. Schintu, N.; Frau, L.; Ibba, M.; Caboni, P.; Garau, A.; Carboni, E.; Carta, A.R. PPAR-gamma-mediated neuroprotection in a chronic mouse model of Parkinson’s disease. Eur. J. Neurosci. 2009, 29, 954–963. [Google Scholar] [CrossRef]
  167. Lecca, D.; Janda, E.; Mulas, G.; Diana, A.; Martino, C.; Angius, F.; Spolitu, S.; Casu, M.A.; Simbula, G.; Boi, L.; et al. Boosting phagocytosis and anti-inflammatory phenotype in microglia mediates neuroprotection by PPARgamma agonist MDG548 in Parkinson’s disease models. Br. J. Pharmacol. 2018, 175, 3298–3314. [Google Scholar] [CrossRef] [Green Version]
  168. Lee, Y.; Cho, J.H.; Lee, S.; Lee, W.; Chang, S.C.; Chung, H.Y.; Moon, H.R.; Lee, J. Neuroprotective effects of MHY908, a PPAR alpha/gamma dual agonist, in a MPTP-induced Parkinson’s disease model. Brain Res. 2019, 1704, 47–58. [Google Scholar] [CrossRef]
  169. Pioglitazone in early Parkinson’s disease: A phase 2, multicentre, double-blind, randomised trial. Lancet Neurol. 2015, 14, 795–803. [CrossRef] [Green Version]
  170. Brundin, P.; Wyse, R. Parkinson disease: Laying the foundations for disease-modifying therapies in PD. Nat. Rev. Neurol. 2015, 11, 553–555. [Google Scholar] [CrossRef]
  171. Thurman, D.J.; Beghi, E.; Begley, C.E.; Berg, A.T.; Buchhalter, J.R.; Ding, D.; Hesdorffer, D.C.; Hauser, W.A.; Kazis, L.; Kobau, R.; et al. Standards for epidemiologic studies and surveillance of epilepsy. Epilepsia 2011, 52 (Suppl. 7), 2–26. [Google Scholar] [CrossRef]
  172. Auvin, S. Fatty acid oxidation and epilepsy. Epilepsy Res 2012, 100, 224–228. [Google Scholar] [CrossRef]
  173. Lambert, D.M.; Vandevoorde, S.; Diependaele, G.; Govaerts, S.J.; Robert, A.R. Anticonvulsant activity of N-palmitoylethanolamide, a putative endocannabinoid, in mice. Epilepsia 2001, 42, 321–327. [Google Scholar] [CrossRef] [Green Version]
  174. Sheerin, A.H.; Zhang, X.; Saucier, D.M.; Corcoran, M.E. Selective antiepileptic effects of N-palmitoylethanolamide, a putative endocannabinoid. Epilepsia 2004, 45, 1184–1188. [Google Scholar] [CrossRef]
  175. Citraro, R.; Russo, E.; Scicchitano, F.; van Rijn, C.M.; Cosco, D.; Avagliano, C.; Russo, R.; D’Agostino, G.; Petrosino, S.; Guida, F.; et al. Antiepileptic action of N-palmitoylethanolamine through CB1 and PPAR-alpha receptor activation in a genetic model of absence epilepsy. Neuropharmacology 2013, 69, 115–126. [Google Scholar] [CrossRef]
  176. Simeone, T.A.; Matthews, S.A.; Samson, K.K.; Simeone, K.A. Regulation of brain PPARgamma2 contributes to ketogenic diet anti-seizure efficacy. Exp. Neurol. 2017, 287, 54–64. [Google Scholar] [CrossRef] [Green Version]
  177. Simeone, T.A.; Matthews, S.A.; Simeone, K.A. Synergistic protection against acute flurothyl-induced seizures by adjuvant treatment of the ketogenic diet with the type 2 diabetes drug pioglitazone. Epilepsia 2017, 58, 1440–1450. [Google Scholar] [CrossRef]
  178. Wong, S.B.; Cheng, S.J.; Hung, W.C.; Lee, W.T.; Min, M.Y. Rosiglitazone Suppresses In Vitro Seizures in Hippocampal Slice by Inhibiting Presynaptic Glutamate Release in a Model of Temporal Lobe Epilepsy. PLoS ONE 2015, 10, e0144806. [Google Scholar] [CrossRef] [Green Version]
  179. Peng, J.; Wang, K.; Xiang, W.; Li, Y.; Hao, Y.; Guan, Y. Rosiglitazone polarizes microglia and protects against pilocarpine-induced status epilepticus. CNS Neurosci. Ther. 2019, 25, 1363–1372. [Google Scholar] [CrossRef]
  180. Hong, S.; Xin, Y.; HaiQin, W.; GuiLian, Z.; Ru, Z.; ShuQin, Z.; HuQing, W.; Li, Y.; Ning, B.; YongNan, L. The PPARgamma agonist rosiglitazone prevents neuronal loss and attenuates development of spontaneous recurrent seizures through BDNF/TrkB signaling following pilocarpine-induced status epilepticus. Neurochem. Int. 2013, 63, 405–412. [Google Scholar] [CrossRef]
  181. Roosterman, D.; Cottrell, G.S. The two-cell model of glucose metabolism: A hypothesis of schizophrenia. Mol. Psychiatry 2021, 26, 1738–1747. [Google Scholar] [CrossRef]
Table 1. This table summarizes the preclinical and clinical studies aiming to repurpose PPARα and PARγ agonists in psychiatric and neurological disorders. Further details can be found in the text.
Table 1. This table summarizes the preclinical and clinical studies aiming to repurpose PPARα and PARγ agonists in psychiatric and neurological disorders. Further details can be found in the text.
Preclinical or Clinical StudyDiseaseAnimal Model or PatientsOutcomeReferences
PPARα agonist
FenofibratepreclinicalSchizophreniaRat: maternal immune activationAttenuation of behavioral disruption and dopaminergic dysfunction[53,54]
Rat: postnatal kainic lesionAttenuation of disruption of pre-pulse inhibition[57]
DepressionRat: chronic stressAntidepressant-like effect[49]
Parkinson’s DiseaseRat: MPTP modelProtection from neurotoxicity[116,117]
EpilepsyPharmacologically induced seizures or genetic modelsReduction in seizures[118,118]
clinicalEpilepsySleep-related hypermotor epilepsyReduction in seizures[119]
BezafibratepreclinicalEpilepsyPentylenetetrazole-Induced Kindling SeizureReduction in seizures[121]
ClofibratepreclinicalNicotine DependenceNicotine self-administration in rats and monkeysBlockade of nicotine self-administration and conditioned place preference[105]
GemfibrozilpreclinicalAlzheimer’s DiseaseTransgenic AD miceDecreases amyloid plaque deposition, astrogliosis; improves spatial learning, memory, and hippocampal plasticity[122,123,124]
clinicalNicotine DependenceSmokersNo efficacy[115]
PPARγ agonist
RosiglitazonepreclinicalDiabetes-Induced Cognitive DeclineMice model (diabetic mice)Induction of BDNF expression[60]
clinicalSchizophreniaSchizophrenic patientsno significant benefits on cognition[61]
preclinicalDepressionRat: forced swimMouse: tail suspension testsAntidepressant-like effect[80]
clinicalDepressed patients with insulin resistanceEffective as adjunctive treatment[82]
preclinicalAlzheimer’s DiseaseRat and mice models of Alzheimer’s diseaseImprovement in learning and memory, Reduction in microglial activation and Aβ plaques[43,125]
clinicalPatients with Alzheimer’s diseaseImprovement in cognitive abilities or no efficacy[62,126]
preclinicalParkinson’s DiseaseRodent: MPTP modelProtection from neurotoxicity[127]
PioglitazoneclinicalSchizophreniaSchizophrenic patients with metabolic syndromeTreatment of glucose and lipid abnormalities in schizophrenic patients, and symptom reductions[65]
preclinicalDepressionMouse: forced swimming testAntidepressant-like effect[81]
clinicalPatients with moderate-to-severe major depressive disorderEffective as adjunctive short-term treatment[83]
preclinicalAlzheimer’ s DiseaseMouse models of Alzheimer’s diseaseImprovement in learning and memory; reduction in microglial activation and Aβ plaques[43,125]
clinicalAlzheimer’s disease patientsNo efficacy[128,129]
preclinicalAlcohol Use DisorderRat models of alcoholism (alcohol preferring rats, binge alcohol drinking)Reduction in alcohol seeking. Protection against neuronal and cognitive degeneration elicited by binge alcohol exposure[95,96,97]
clinicalAlcohol-dependent patientsNo efficacy[114]
preclinicalOpioid Use DisorderMorphine-dependent miceAttenuation of morphine withdrawal symptoms, of reinstatement of heroin seeking and of heroin-induced reinstatement.[107]
clinicalHeroin users or nondependent prescription opioid abusersFailure to alter the reinforcing or positive subjective effects of heroin. Reduction in heroin craving and anxiety. Failure to alter the abuse liability of oxycodone[108,109]
preclinicalParkinson’s DiseaseRodent: MPTP modelProtection from neurotoxicity[127,130]
TroglitazonepreclinicalMultiple SclerosisMouse: experimental autoimmune encephalitisAnti-inflammatory effects[131]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Sagheddu, C.; Melis, M.; Muntoni, A.L.; Pistis, M. Repurposing Peroxisome Proliferator-Activated Receptor Agonists in Neurological and Psychiatric Disorders. Pharmaceuticals 2021, 14, 1025. https://doi.org/10.3390/ph14101025

AMA Style

Sagheddu C, Melis M, Muntoni AL, Pistis M. Repurposing Peroxisome Proliferator-Activated Receptor Agonists in Neurological and Psychiatric Disorders. Pharmaceuticals. 2021; 14(10):1025. https://doi.org/10.3390/ph14101025

Chicago/Turabian Style

Sagheddu, Claudia, Miriam Melis, Anna Lisa Muntoni, and Marco Pistis. 2021. "Repurposing Peroxisome Proliferator-Activated Receptor Agonists in Neurological and Psychiatric Disorders" Pharmaceuticals 14, no. 10: 1025. https://doi.org/10.3390/ph14101025

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